151
|
Zhang P, Lan X, Fan B, Chen Y, Wei X, Li X, Fan N, Tang C, Lu L. A protocol for the integration of multi-omics bioinformatics: Mechanism of acupuncture as an adjunctive therapy for alcohol use disorder. Front Neurol 2023; 13:977487. [PMID: 36686540 PMCID: PMC9849375 DOI: 10.3389/fneur.2022.977487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 11/28/2022] [Indexed: 01/06/2023] Open
Abstract
Background Alcohol use disorder (AUD) has become a significant global factor in various diseases. As a non-pharmacological therapy, certain therapeutic potential has been found in acupuncture; however, in-depth mechanistic studies related to acupuncture for patients with AUD are still insufficient. Methods Based on a randomized control design and a multi-omics analysis plan, this protocol details the recruitment (42 AUD patients), group allocation (21 in acupuncture group vs. 21 in sham acupuncture group), intervention and follow-up (replacement drugs as a normal treatment, 2 weeks acupuncture duration, and 3 month follow-up), and data collection and analytical processes. For the clinical outcomes, in addition to the time required for alcohol withdrawal symptoms to subside as the primary outcome, changes in the alcohol withdrawal symptoms, alcohol craving, mood dysfunction, sleep disorder, fatigue, self-efficacy, gastrointestinal symptoms, the quality of life, and the relapse outcomes will be compared between the groups to confirm the acupuncture clinical effectiveness on alcohol withdraw. The gut microbiome and the fecal metabolomics will also be assessed to explore the association of the structure and the function of gut microflora and the mediation of acupuncture effect on AUD fully utilizing gut microflora multi-modal data and clinical information, via the combination of multi-omics methods, feature screening algorithms and appropriate models. Discussion The results of this study may help to strengthen clinical evidence of the mechanism of acupuncture intervention in patients with AUD, through understanding of the regulatory mechanism of acupuncture in the gut microbiome and its metabolism as well as AUD-related clinical manifestations. Trial registration Chinese Clinical Trial Registry ChiCTR2200058120. Registered on 24 Mar 2022.
Collapse
Affiliation(s)
- Peiming Zhang
- Clinical Research and Big Data Laboratory, South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Xiaochang Lan
- Department of Substance Dependence, The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, Guangdong Province, China
| | - Baochao Fan
- Clinical Research and Big Data Laboratory, South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Yiming Chen
- Clinical Research and Big Data Laboratory, South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Xiaojing Wei
- Clinical Research and Big Data Laboratory, South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Xiangli Li
- Department of Chinese Medicine, Guangzhou First People's Hospital, Guangzhou, Guangdong Province, China
| | - Ni Fan
- Department of Substance Dependence, The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, Guangdong Province, China
| | - Chunzhi Tang
- Clinical Research and Big Data Laboratory, South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Liming Lu
- Clinical Research and Big Data Laboratory, South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| |
Collapse
|
152
|
Zhang Y, Xie Z, Zhou J, Li Y, Ning C, Su Q, Ye L, Ai S, Lai J, Pan P, Liu N, Liao Y, Su Q, Li Z, Liang H, Cui P, Huang J. The altered metabolites contributed by dysbiosis of gut microbiota are associated with microbial translocation and immune activation during HIV infection. Front Immunol 2023; 13:1020822. [PMID: 36685491 PMCID: PMC9845923 DOI: 10.3389/fimmu.2022.1020822] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 11/28/2022] [Indexed: 01/05/2023] Open
Abstract
Background The immune activation caused by microbial translocation has been considered to be a major driver of HIV infection progression. The dysbiosis of gut microbiota has been demonstrated in HIV infection, but the interplay between gut microbiota and its metabolites in the pathogenesis of HIV is seldom reported. Methods We conducted a case-controlled study including 41 AIDS patients, 39 pre-AIDS patients and 34 healthy controls. Both AIDS group and pre-AIDS group were divided according to clinical manifestations and CD4 + T cell count. We collected stool samples for 16S rDNA sequencing and untargeted metabolomics analysis, and examined immune activation and microbial translocation for blood samples. Results The pre-AIDS and AIDS groups had higher levels of microbial translocation and immune activation. There were significant differences in gut microbiota and metabolites at different stages of HIV infection. Higher abundances of pathogenic bacteria or opportunistic pathogen, as well as lower abundances of butyrate-producing bacteria and bacteria with anti-inflammatory potential were associated with HIV severity. The metabolism of tryptophan was disordered after HIV infection. Lower level of anti-inflammatory metabolites and phosphonoacetate, and higher level of phenylethylamine and polyamines were observed in HIV infection. And microbial metabolic pathways related to altered metabolites differed. Moreover, disrupted metabolites contributed by altered microbiota were found to be correlated to microbial translocation and immune activation. Conclusions Metabolites caused by dysbiosis of gut microbiota and related metabolic function are correlated to immune activation and microbial translocation, suggesting that the effect of microbiota on metabolites is related to intestinal barrier disruption in HIV infection.
Collapse
Affiliation(s)
- Yu Zhang
- Guangxi Key Laboratory of AIDS Prevention and Treatment and Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Nanning, China
- School of Public Health, Guangxi Medical University, Nanning, China
- The Tenth Affiliated Hospital of Guangxi Medical University, Qinzhou, China
| | - Zhiman Xie
- Department of Infectious Diseases, The Fourth People's Hospital of Nanning, Nanning, China
| | - Jie Zhou
- Guangxi Key Laboratory of AIDS Prevention and Treatment and Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Nanning, China
- School of Public Health, Guangxi Medical University, Nanning, China
| | - Yanjun Li
- Department of Infectious Diseases, The Fourth People's Hospital of Nanning, Nanning, China
| | - Chuanyi Ning
- Guangxi Key Laboratory of AIDS Prevention and Treatment and Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Nanning, China
| | - Qisi Su
- Department of Infectious Diseases, The Fourth People's Hospital of Nanning, Nanning, China
| | - Li Ye
- Guangxi Key Laboratory of AIDS Prevention and Treatment and Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Nanning, China
- School of Public Health, Guangxi Medical University, Nanning, China
| | - Sufang Ai
- Department of Infectious Diseases, The Fourth People's Hospital of Nanning, Nanning, China
| | - Jingzhen Lai
- Guangxi Key Laboratory of AIDS Prevention and Treatment and Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Nanning, China
- Life Science Institute, Guangxi Medical University, Nanning, China
| | - Peijiang Pan
- Guangxi Key Laboratory of AIDS Prevention and Treatment and Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Nanning, China
- Life Science Institute, Guangxi Medical University, Nanning, China
| | - Ningmei Liu
- Department of Infectious Diseases, The Fourth People's Hospital of Nanning, Nanning, China
| | - Yanyan Liao
- Guangxi Key Laboratory of AIDS Prevention and Treatment and Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Nanning, China
- Life Science Institute, Guangxi Medical University, Nanning, China
| | - Qijian Su
- The Tenth Affiliated Hospital of Guangxi Medical University, Qinzhou, China
| | - Zhuoxin Li
- Guangxi Key Laboratory of AIDS Prevention and Treatment and Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Nanning, China
- School of Public Health, Guangxi Medical University, Nanning, China
| | - Hao Liang
- Guangxi Key Laboratory of AIDS Prevention and Treatment and Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Nanning, China
- School of Public Health, Guangxi Medical University, Nanning, China
- Life Science Institute, Guangxi Medical University, Nanning, China
| | - Ping Cui
- Guangxi Key Laboratory of AIDS Prevention and Treatment and Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Nanning, China
- Life Science Institute, Guangxi Medical University, Nanning, China
| | - Jiegang Huang
- Guangxi Key Laboratory of AIDS Prevention and Treatment and Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, Nanning, China
- School of Public Health, Guangxi Medical University, Nanning, China
| |
Collapse
|
153
|
Abstract
Nuclear Magnetic Resonance (NMR) spectroscopy is one of the two major analytical platforms in the field of metabolomics, the other being mass spectrometry (MS). NMR is less sensitive than MS and hence it detects a relatively small number of metabolites. However, NMR exhibits numerous unique characteristics including its high reproducibility and non-destructive nature, its ability to identify unknown metabolites definitively, and its capabilities to obtain absolute concentrations of all detected metabolites, sometimes even without an internal standard. These characteristics outweigh the relatively low sensitivity and resolution of NMR in metabolomics applications. Since biological mixtures are highly complex, increased demand for new methods to improve detection, better identify unknown metabolites, and provide more accurate quantitation continues unabated. Technological and methodological advances to date have helped to improve the resolution and sensitivity and detection of a larger number of metabolite signals. Efforts focused on measuring unknown metabolite signals have resulted in the identification and quantitation of an expanded pool of metabolites including labile metabolites such as cellular redox coenzymes, energy coenzymes, and antioxidants. This chapter describes quantitative NMR methods in metabolomics with an emphasis on recent methodological developments, while highlighting the benefits and challenges of NMR-based metabolomics.
Collapse
Affiliation(s)
- G A Nagana Gowda
- Northwest Metabolomics Research Center, University of Washington, Seattle, WA, USA.
- Mitochondria and Metabolism Center, Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA.
| | - Daniel Raftery
- Northwest Metabolomics Research Center, University of Washington, Seattle, WA, USA.
- Mitochondria and Metabolism Center, Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA.
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
| |
Collapse
|
154
|
Shen Y, Wang P, Yang X, Chen M, Dong Y, Li J. Untargeted metabolomics unravel serum metabolic alterations in smokers with hypertension. Front Physiol 2023; 14:1127294. [PMID: 36935758 PMCID: PMC10018148 DOI: 10.3389/fphys.2023.1127294] [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: 12/19/2022] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
Background: Cigarette smoking is an important environmental risk factor for cardiovascular events of hypertension (HTN). Existing studies have provided evidence supporting altered gut microbiota by cigarette smoking, especially in hypertensive patients. Metabolic biomarkers play a central role in the functional potentials of the gut microbiome but are poorly characterized in hypertensive smokers. To explore whether serum metabolomics signatures and compositions of HTN patients were varied in smokers, and investigate their connecting relationship to gut microbiota, the serum metabolites were examined in untreated hypertensive patients using untargeted liquid chromatography-mass spectrometry (LC/MS) analysis. Results: A dramatic difference and clear separation in community features of circulating metabolomics members were seen in smoking HTN patients compared with the non-smoking controls, according to partial least squares discrimination analysis (PLS-DA) and orthogonal partial least squares discrimination analysis (OPLS-DA). Serum metabolic profiles and compositions of smoking patients with HTN were significantly distinct from the controls, and were characterized by enrichment of 12-HETE, 7-Ketodeoxycholic acid, Serotonin, N-Stearoyl tyrosine and Deoxycholic acid glycine conjugate, and the depletion of Tetradecanedioic acid, Hippuric acid, Glyceric acid, 20-Hydroxyeicosatetraenoic acid, Phenylpyruvic acid and Capric acid. Additionally, the metabolome displayed prominent functional signatures, with a majority proportion of the metabolites identified to be discriminating between groups distributed in Starch and sucrose metabolism, Caffeine metabolism, Pyruvate metabolism, Glycine, serine and threonine metabolism, and Phenylalanine metabolic pathways. Furthermore, the observation of alterations in metabolites associated with intestinal microbial taxonomy indicated that these metabolic members might mediate the effects of gut microbiome on the smoking host. Indeed, the metabolites specific to smoking HTNs were strongly organized into co-abundance networks, interacting with an array of clinical parameters, including uric acid (UA), low-denstiy lipoprotein cholesterol (LDLC) and smoking index. Conclusions: In conclusion, we demonstrated disparate circulating blood metabolome composition and functional potentials in hypertensive smokers, showing a linkage between specific metabolites in blood and the gut microbiome.
Collapse
Affiliation(s)
- Yang Shen
- Department of Nephrology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Pan Wang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xinchun Yang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Mulei Chen
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Ying Dong
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- *Correspondence: Ying Dong, ; Jing Li,
| | - Jing Li
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- *Correspondence: Ying Dong, ; Jing Li,
| |
Collapse
|
155
|
Wang L, Gou X, Ding Y, Liu J, Wang Y, Wang Y, Zhang J, Du L, Peng W, Fan G. The interplay between herbal medicines and gut microbiota in metabolic diseases. Front Pharmacol 2023; 14:1105405. [PMID: 37033634 PMCID: PMC10079915 DOI: 10.3389/fphar.2023.1105405] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 03/09/2023] [Indexed: 04/11/2023] Open
Abstract
Globally, metabolic diseases are becoming a major public health problem. Herbal medicines are medicinal materials or preparations derived from plants and are widely used in the treatment of metabolic diseases due to their good curative effects and minimal side effects. Recent studies have shown that gut microbiota plays an important role in the herbal treatment of metabolic diseases. However, the mechanisms involved are still not fully understood. This review provides a timely and comprehensive summary of the interactions between herbal medicines and gut microbiota in metabolic diseases. Mechanisms by which herbal medicines treat metabolic diseases include their effects on the gut microbial composition, the intestinal barrier, inflammation, and microbial metabolites (e.g., short-chain fatty acids and bile acids). Herbal medicines can increase the abundance of beneficial bacteria (e.g., Akkermansia and Blautia), reduce the abundance of harmful bacteria (e.g., Escherichia-Shigella), protect the intestinal barrier, and alleviate inflammation. In turn, gut microbes can metabolize herbal compounds and thereby increase their bioavailability and bioactivity, in addition to reducing their toxicity. These findings suggest that the therapeutic effects of herbal medicines on metabolic diseases are closely related to their interactions with the gut microbiota. In addition, some methods, and techniques for studying the bidirectional interaction between herbal medicines and gut microbiota are proposed and discussed. The information presented in this review will help with a better understanding of the therapeutic mechanisms of herbal medicines and the key role of gut microbiota.
Collapse
Affiliation(s)
- Lijie Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoling Gou
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yin Ding
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jingye Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yue Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yaqian Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jing Zhang
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Leilei Du
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Gang Fan, ; Wei Peng, ; Leilei Du,
| | - Wei Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Gang Fan, ; Wei Peng, ; Leilei Du,
| | - Gang Fan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Gang Fan, ; Wei Peng, ; Leilei Du,
| |
Collapse
|
156
|
Luo Z, Yong K, Luo Q, Du Z, Ma L, Huang Y, Zhou T, Yao X, Shen L, Yu S, Deng J, Ren Z, Zhang Y, Yan Z, Zuo Z, Cao S. Altered Fecal Microbiome and Correlations of the Metabolome with Plasma Metabolites in Dairy Cows with Left Displaced Abomasum. Microbiol Spectr 2022; 10:e0197222. [PMID: 36222683 PMCID: PMC9769586 DOI: 10.1128/spectrum.01972-22] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/18/2022] [Indexed: 01/06/2023] Open
Abstract
Left displaced abomasum (LDA) in postpartum dairy cows contributes to significant economic losses. Dairy cows with LDA undergo excessive lipid mobilization and insulin resistance. Although gut dysbiosis is implicated, little is known about the role of the gut microbiota in the abnormal metabolic processes of LDA. To investigate the functional links among microbiota, metabolites, and disease phenotypes in LDA, we performed 16S rDNA gene amplicon sequencing and liquid chromatography-tandem mass spectrometry (LC-MS/MS) of fecal samples from cows with LDA (n = 10) and healthy cows (n = 10). Plasma marker profiling was synchronously analyzed. In the LDA event, gut microbiota composition and fecal metabolome were shifted in circulation with an amino acid pool deficit in dairy cows. Compared with the healthy cows, salicylic acid derived from microbiota catabolism was decreased in the LDA cows, which negatively correlated with Akkermansia, Prevotella, non-esterified fatty acid (NEFA), and β-hydroxybutyric acid (BHBA) levels. Conversely, fecal taurolithocholic acid levels were increased in cows with LDA. Based on integrated analysis with the plasma metabolome, eight genera and eight metabolites were associated with LDA. Of note, the increases in Akkermansia and Oscillospira abundances were negatively correlated with the decreases in 4-pyridoxic acid and cytidine levels, and positively correlated with the increases in NEFA and BHBA levels in amino acid deficit, indicating pyridoxal metabolism-associated gut dysbiosis and lipolysis. Changes in branched-chain amino acids implicated novel host-microbial metabolic pathways involving lipolysis and insulin resistance in cows with LDA. Overall, these results suggest an interplay between host and gut microbes contributing to LDA pathogenesis. IMPORTANCE LDA is a major contributor to economic losses in the dairy industry worldwide; however, the mechanisms associated with the metabolic changes in LDA remain unclear. Most previous studies have focused on the rumen microbiota in terms of understanding the contributors to the productivity and health of dairy cows; this study further sheds light on the relevance of the lower gut microbiota and its associated metabolites in mediating the development of LDA. This study is the first to characterize the correlation between gut microbes and metabolic phenotypes in dairy cows with LDA by leveraging multi-omics data, highlighting that the gut microbe may be involved in the regulation of lipolysis and insulin resistance by modulating the amino acid composition. Moreover, this study provides new markers for further research to understand the pathogenesis of the disease as well as to develop effective treatment and prevention strategies.
Collapse
Affiliation(s)
- Zhengzhong Luo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Kang Yong
- Department of Animal Husbandry & Veterinary Medicine, College of Animal Science and Technology, Chongqing Three Gorges Vocational College, Chongqing, China
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Qiao Luo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhenlong Du
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Li Ma
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yixin Huang
- Institute of Biodiversity, Animal Health & Comparative Medicine, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Tao Zhou
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Lanzhou Institute of Animal Husbandry and Veterinary Pharmaceutical, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xueping Yao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Liuhong Shen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Shumin Yu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Junliang Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhihua Ren
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yong Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Zuoting Yan
- Lanzhou Institute of Animal Husbandry and Veterinary Pharmaceutical, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Zhicai Zuo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Suizhong Cao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
157
|
Haffner JJ, Katemauswa M, Kagone TS, Hossain E, Jacobson D, Flores K, Parab AR, Obregon-Tito AJ, Tito RY, Reyes LM, Troncoso-Corzo L, Guija-Poma E, Meda N, Carabin H, Honap TP, Sankaranarayanan K, Lewis CM, McCall LI. Untargeted Fecal Metabolomic Analyses across an Industrialization Gradient Reveal Shared Metabolites and Impact of Industrialization on Fecal Microbiome-Metabolome Interactions. mSystems 2022; 7:e0071022. [PMID: 36416540 PMCID: PMC9765122 DOI: 10.1128/msystems.00710-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 11/01/2022] [Indexed: 11/24/2022] Open
Abstract
The metabolome is a central determinant of human phenotypes and includes the plethora of small molecules produced by host and microbiome or taken up from exogenous sources. However, studies of the metabolome have so far focused predominantly on urban, industrialized populations. Through an untargeted metabolomic analysis of 90 fecal samples from human individuals from Africa and the Americas-the birthplace and the last continental expansion of our species, respectively-we characterized a shared human fecal metabolome. The majority of detected metabolite features were ubiquitous across populations, despite any geographic, dietary, or behavioral differences. Such shared metabolite features included hyocholic acid and cholesterol. However, any characterization of the shared human fecal metabolome is insufficient without exploring the influence of industrialization. Here, we show chemical differences along an industrialization gradient, where the degree of industrialization correlates with metabolomic changes. We identified differential metabolite features such as amino acid-conjugated bile acids and urobilin as major metabolic correlates of these behavioral shifts. Additionally, coanalyses with over 5,000 publicly available human fecal samples and cooccurrence probability analyses with the gut microbiome highlight connections between the human fecal metabolome and gut microbiome. Our results indicate that industrialization significantly influences the human fecal metabolome, but diverse human lifestyles and behavior still maintain a shared human fecal metabolome. This study represents the first characterization of the shared human fecal metabolome through untargeted analyses of populations along an industrialization gradient. IMPORTANCE As the world becomes increasingly industrialized, understanding the biological consequences of these lifestyle shifts and what it means for past, present, and future human health is critical. Indeed, industrialization is associated with rises in allergic and autoimmune health conditions and reduced microbial diversity. Exploring these health effects on a chemical level requires consideration of human lifestyle diversity, but understanding the significance of any differences also requires knowledge of what molecular components are shared between human groups. Our study reveals the key chemistry of the human gut as defined by varied industrialization-based differences and ubiquitous shared features. Ultimately, these novel findings extend our knowledge of human molecular biology, especially as it is influenced by lifestyle and behavior, and provide steps toward understanding how human biology has changed over our species' history.
Collapse
Affiliation(s)
- Jacob J. Haffner
- Department of Anthropology, University of Oklahoma, Norman, Oklahoma, USA
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, Oklahoma, USA
| | - Mitchelle Katemauswa
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, Oklahoma, USA
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, USA
| | - Thérèse S. Kagone
- Burkina Faso Ministry of Health, Ouagadougou, Kadiogo, Burkina Faso
- Centre MURAZ Research Institute, Bobo-Dioulasso, Burkina Faso
| | - Ekram Hossain
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, Oklahoma, USA
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, USA
| | - David Jacobson
- Department of Anthropology, University of Oklahoma, Norman, Oklahoma, USA
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, Oklahoma, USA
| | - Karina Flores
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, Oklahoma, USA
- Department of Biology, University of Oklahoma, Norman, Oklahoma, USA
| | - Adwaita R. Parab
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, Oklahoma, USA
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma, USA
| | - Alexandra J. Obregon-Tito
- Department of Anthropology, University of Oklahoma, Norman, Oklahoma, USA
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, Oklahoma, USA
| | - Raul Y. Tito
- Department of Anthropology, University of Oklahoma, Norman, Oklahoma, USA
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, Oklahoma, USA
| | | | | | - Emilio Guija-Poma
- Centro de Investigación de Bioquímica y Nutrición, Facultad de Medicina Humana, Universidad de San Martín de Porres, Lima, Perú
| | - Nicolas Meda
- Burkina Faso Ministry of Health, Ouagadougou, Kadiogo, Burkina Faso
| | - Hélène Carabin
- Department of Biostatistics and Epidemiology, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
- Département de Médecine Sociale et Préventive, École de Santé Publique de l’Université de Montréal, Montréal, Quebec, Canada
- Centre de Recherche en Santé Publique (CReSP) de l’Université de Montréal et du CIUSS du Centre Sud de Montréal, Montréal, Quebec, Canada
| | - Tanvi P. Honap
- Department of Anthropology, University of Oklahoma, Norman, Oklahoma, USA
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, Oklahoma, USA
| | - Krithivasan Sankaranarayanan
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, Oklahoma, USA
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma, USA
| | - Cecil M. Lewis
- Department of Anthropology, University of Oklahoma, Norman, Oklahoma, USA
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, Oklahoma, USA
| | - Laura-Isobel McCall
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, Oklahoma, USA
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, USA
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma, USA
| |
Collapse
|
158
|
Najmanová L, Vídeňská P, Cahová M. Healthy microbiome – a mere idea or a sound concept? Physiol Res 2022. [PMID: 36426891 DOI: 10.33549/physiolres.934967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Hundreds of studies in last decades have aimed to compare the microbiome of patients suffering from diverse diseases with that of healthy controls. The microbiome-related component was additionally identified in pathophysiology of many diseases formerly considered to depend only on the host physiology. This, however, opens important questions like: “What is the healthy microbiome?” or “Is it possible to define it unequivocally?”. In this review, we describe the main hindrances complicating the definition of “healthy microbiome” in terms of microbiota composition. We discuss the human microbiome from the perspective of classical ecology and we advocate for the shift from the stress on microbiota composition to the functions that microbiome ensures for the host. Finally, we propose to leave the concept of ideal healthy microbiome and replace it by focus on microbiome advantageous for the host, which always depends on the specific context like the age, genetics, dietary habits, body site or physiological state.
Collapse
Affiliation(s)
| | | | - M Cahová
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| |
Collapse
|
159
|
Global Metabolomics Discovers Two Novel Biomarkers in Pyridoxine-Dependent Epilepsy Caused by ALDH7A1 Deficiency. Int J Mol Sci 2022; 23:ijms232416061. [PMID: 36555701 PMCID: PMC9784804 DOI: 10.3390/ijms232416061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/13/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Pyridoxine-dependent epilepsy (PDE) is a rare autosomal recessive developmental and epileptic encephalopathy caused by pathogenic variants in the ALDH7A1 gene (PDE-ALDH7A1), which mainly has its onset in neonates and infants. Early diagnosis and treatment are crucial to prevent severe neurological sequelae or death. Sensitive, specific, and stable biomarkers for diagnostic evaluations and follow-up examinations are essential to optimize outcomes. However, most of the known biomarkers for PDE lack these criteria. Additionally, there is little discussion regarding the interdependence of biomarkers in the PDE-ALDH7A1 metabolite profile. Therefore, the aim of this study was to understand the underlying mechanisms in PDE-ALDH7A1 and to discover new biomarkers in the plasma of patients using global metabolomics. Plasma samples from 9 patients with genetically confirmed PDE-ALDH7A1 and 22 carefully selected control individuals were analyzed by ultra high performance liquid chromatography-high-resolution mass spectrometry (UHPLC-HRMS). Two novel and reliable pyridoxine-independent diagnostic markers, 6-hydroxy-2-aminocaproic acid (HACA) and an isomer of C9H11NO4, were identified. Furthermore, a possible reaction mechanism is proposed for HACA. This study demonstrates the capability of global metabolomics in disease screening to detect established and novel biomarkers.
Collapse
|
160
|
Wang H, Qi S, Mu X, Yuan L, Li Y, Qiu J. Bisphenol F induces liver-gut alteration in zebrafish. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 851:157974. [PMID: 35963407 DOI: 10.1016/j.scitotenv.2022.157974] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/23/2022] [Accepted: 08/07/2022] [Indexed: 06/15/2023]
Abstract
The unease of consumers with bisphenol A has led to the increased industrial usage of bisphenol F (BPF), which is a new hazard to environmental health. Here, zebrafish were exposed to three BPF concentrations (0.5, 5, and 50 μg/L) from the embryonic stage for 180 days. Results showed that zebrafish body length and weight decreased and hepatosomatic index values increased, even at environmentally relevant concentration. Histological analysis identified the occurrence of hepatic fibrosis and steatosis in 5 and 50 μg/L groups, which indicated the liver injury caused by BPF. Based on the untargeted metabolomics results, a dose-dependent variation in the effects of BPF on liver metabolism was found, and amino acids, purines and one carbon metabolism were the main affected processes in the 0.5, 5, and 50 μg/L treatments, respectively. At the same time, BPF induced a shift in intestinal microbiome composition, including decreased abundance of Erysipelotrichaceae, Rhodobacteraceae and Gemmobacter. In addition, the correlation analysis suggested an association between gut microbiome changes and affected hepatic metabolites after BPF exposure. These findings indicate that a liver-gut alteration is induced by long-term BPF exposure.
Collapse
Affiliation(s)
- Hui Wang
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing, China; Fishery Resource and Environment Research Center, Chinese Academy of Fishery Sciences, Beijing, China
| | - Suzhen Qi
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiyan Mu
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing, China; Fishery Resource and Environment Research Center, Chinese Academy of Fishery Sciences, Beijing, China.
| | - Lilai Yuan
- Fishery Resource and Environment Research Center, Chinese Academy of Fishery Sciences, Beijing, China
| | - Yingren Li
- Fishery Resource and Environment Research Center, Chinese Academy of Fishery Sciences, Beijing, China
| | - Jing Qiu
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing, China.
| |
Collapse
|
161
|
Cheong YE, Kim J, Jin YS, Kim KH. Elucidation of the fucose metabolism of probiotic Lactobacillus rhamnosus GG by metabolomic and flux balance analyses. J Biotechnol 2022; 360:110-116. [PMID: 36336085 DOI: 10.1016/j.jbiotec.2022.11.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 10/08/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022]
Abstract
Lactobacillus rhamnosus GG (LGG) is one of the most widely used probiotics because of its health benefits and safety. Fucose is among the most abundant hexoses in the human intestine, and LGG consumes fucose to produce energy or proliferate. However, no study has elucidated the metabolism by which LGG metabolizes fucose to produce energy, biomass, and extracellular metabolites. We used metabolomics and flux balance analysis to elucidate these mechanisms and highlight how they might affect the host. We found three different metabolic flux modes by which LGG anaerobically metabolizes fucose to produce energy and biomass. These metabolic flux modes differ from homolactic or heterolactic fermentation and account for the production of lactic acid, 1,2-propanediol, acetic acid, formic acid, and carbon dioxide as a result of fucose metabolism in LGG. We also used gas chromatography/time-of-flight mass spectrometry to identify a variety of short-chain fatty acids and organic acids secreted during fucose metabolism by LGG. Our study is the first to elucidate the unique fucose metabolism of LGG in anaerobic condition.
Collapse
Affiliation(s)
- Yu Eun Cheong
- Department of Biotechnology, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Jungyeon Kim
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Yong-Su Jin
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Kyoung Heon Kim
- Department of Biotechnology, Graduate School, Korea University, Seoul 02841, Republic of Korea; Department of Food Bioscience and Technology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
162
|
Chen Z, Sun Y, Chen L, Zhang Y, Wang J, Li H, Yan X, Xia L, Yao G. Differences in meat quality between Angus cattle and Xinjiang brown cattle in association with gut microbiota and its lipid metabolism. Front Microbiol 2022; 13:988984. [PMID: 36560955 PMCID: PMC9763702 DOI: 10.3389/fmicb.2022.988984] [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: 07/07/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022] Open
Abstract
Gut microbiota plays important roles in mediating fat metabolic events in humans and animals. However, the differences of meat quality traits related to the lipid metabolism (MQT-LM) in association with gut microbiota involving in lipid metabolism have not been well explored between Angus cattle (AG) and Xinjiang brown cattle (BC). Ten heads of 18-month-old uncastrated male AG and BC (5 in each group) raised under the identical conditions were selected to test MQT-LM, i.e., the backfat thickness (BFT), the intramuscular fat (IMF) content, the intramuscular adipocyte areas (IAA), the eye muscle area (EMA), the muscle fiber sectional area (MFSA) and the muscle shear force after sacrifice. The gut microbiota composition and structure with its metabolic function were analyzed by means of metagenomics and metabolomics with rectal feces. The correlation of MQT-LM with the gut microbiota and its metabolites was analyzed. In comparison with AG, BC had significant lower EMA, IMF content and IAA but higher BFT and MFSA. Chao1 and ACE indexes of α-diversity were lower. β-diversity between AG and BC were significantly different. The relative abundance of Bacteroidetes, Prevotella and Blautia and Prevotella copri, Blautia wexlerae, and Ruminococcus gnavus was lower. The lipid metabolism related metabolites, i.e., succinate, oxoglutaric acid, L-aspartic acid and L-glutamic acid were lower, while GABA, L-asparagine and fumaric acid were higher. IMF was positively correlated with Prevotella copri, Blautia wexlerae and Ruminococcus gnavus, and the metabolites succinate, oxoglutaric acid, L-aspartic acid and L-glutamic acid, while negatively with GABA, L-asparagine and fumaric acid. BFT was negatively correlated with Blautia wexlerae and the metabolites succinate, L-aspartic acid and L-glutamic acid, while positively with GABA, L-asparagine and fumaric acid. Prevotella Copri, Blautia wexlerae, and Ruminococcus gnavus was all positively correlated with succinate, oxoglutaric acid, while negatively with L-asparagine and fumaric acid. In conclusion, Prevotella copri, Prevotella intermedia, Blautia wexlerae, and Ruminococcus gnavus may serve as the potential differentiated bacterial species in association with MQT-LM via their metabolites of oxoglutaric acid, succinate, fumaric acid, L-aspartic acid, L-asparagine, L-glutamic acid and GABA between BC and AG.
Collapse
Affiliation(s)
- Zhuo Chen
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Yawei Sun
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Lijing Chen
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Yang Zhang
- Institute of Animal Science, Xinjiang Academy of Animal Sciences, Urumqi, China
| | - Jinquan Wang
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Hongbo Li
- Institute of Animal Science, Xinjiang Academy of Animal Sciences, Urumqi, China
| | - Xiangming Yan
- Institute of Animal Science, Xinjiang Academy of Animal Sciences, Urumqi, China
| | - Lining Xia
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China,Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animals (XJKLNDSCHA), Xinjiang Agricultural University, Urumqi, China,*Correspondence: Lining Xia,
| | - Gang Yao
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China,Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animals (XJKLNDSCHA), Xinjiang Agricultural University, Urumqi, China,Gang Yao,
| |
Collapse
|
163
|
The Role of the Gut Microbiome in Pediatric Obesity and Bariatric Surgery. Int J Mol Sci 2022; 23:ijms232315421. [PMID: 36499739 PMCID: PMC9740713 DOI: 10.3390/ijms232315421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
Obesity affects 42.4% of adults and 19.3% of children in the United States. Childhood obesity drives many comorbidities including hypertension, fatty liver disease, and type 2 diabetes mellitus. Prior research suggests that aberrant compositional development of the gut microbiome, with low-grade inflammation, precedes being overweight. Therefore, childhood may provide opportunities for interventions that shape the microbiome to mitigate obesity-related diseases. Children with obesity have gut microbiota compositional and functional differences, including increased proinflammatory bacterial taxa, compared to lean controls. Restoration of the gut microbiota to a healthy state may ameliorate conditions associated with obesity and help maintain a healthy weight. Pediatric bariatric (weight-loss) surgery is an effective treatment for childhood obesity; however, there is limited research into the role of the gut microbiome after weight-loss surgery in children. This review will discuss the magnitude of childhood obesity, the importance of the developing microbiome in establishing metabolic pathways, interventions such as bariatric surgery that may modulate the gut microbiome, and future directions for the potential development of microbiome-based therapeutics to treat obesity.
Collapse
|
164
|
Chen J, Zhou D, Miao J, Zhang C, Li X, Feng H, Xing Y, Zhang Z, Bao C, Lin Z, Chen Y, Yuan JXJ, Sun D, Yang K, Wang J. Microbiome and metabolome dysbiosis of the gut-lung axis in pulmonary hypertension. Microbiol Res 2022; 265:127205. [DOI: 10.1016/j.micres.2022.127205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 09/13/2022] [Accepted: 09/16/2022] [Indexed: 10/14/2022]
|
165
|
Loganathan T, Priya Doss C G. The influence of machine learning technologies in gut microbiome research and cancer studies - A review. Life Sci 2022; 311:121118. [DOI: 10.1016/j.lfs.2022.121118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/19/2022] [Accepted: 10/19/2022] [Indexed: 11/18/2022]
|
166
|
Su L, Zeng Y, Li G, Chen J, Chen X. Quercetin improves high-fat diet-induced obesity by modulating gut microbiota and metabolites in C57BL/6J mice. Phytother Res 2022; 36:4558-4572. [PMID: 35906097 DOI: 10.1002/ptr.7575] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 07/09/2022] [Accepted: 07/12/2022] [Indexed: 12/13/2022]
Abstract
High-fat diet-induced obesity is characterized by low-grade inflammation, which has been linked to gut microbiota dysbiosis. We hypothesized that quercetin supplementation would alter gut microbiota and reduce inflammation in obese mice. Male C57BL/6J mice, 4 weeks of age, were divided into 3 groups, including a low-fat diet group, a high-fat diet (HFD) group, and a high-fat diet plus quercetin (HFD+Q) group. The mice in HFD+Q group were given 50 mg per kg BW quercetin by gavage for 20 weeks. The body weight, fat accumulation, gut barrier function, glucose tolerance, and adipose tissue inflammation were determined in mice. 16 s rRNA amplicon sequence and non-targeted metabolomics analysis were used to explore the alteration of gut microbiota and metabolites. We found that quercetin significantly alleviated HFD-induced obesity, improved glucose tolerance, recovered gut barrier function, and reduced adipose tissue inflammation. Moreover, quercetin ameliorated HFD-induced gut microbiota disorder by regulating the abundance of gut microbiota, such as Adlercreutzia, Allobaculum, Coprococcus_1, Lactococcus, and Akkermansia. Quercetin influenced the production of metabolites that were linked to alterations in obesity-related inflammation and oxidative stress, such as Glycerophospho-N-palmitoyl ethanolamine, sanguisorbic acid dilactone, O-Phospho-L-serine, and P-benzoquinone. Our results demonstrate that the anti-obesity effects of quercetin may be mediated through regulation in gut microbiota and metabolites.
Collapse
Affiliation(s)
- Lijie Su
- Department of Nutrition and Food Hygiene, School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Yupeng Zeng
- Department of Nutrition and Food Hygiene, School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Guokun Li
- Department of Nutrition and Food Hygiene, School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Jing Chen
- Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Xiaoyi Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
167
|
Fu Y, Chen B, Liu Z, Wang H, Zhang F, Zhao Q, Zhu Y, Yong X, Shen Q. Effects of different foxtail millet addition amounts on the cognitive ability of mice. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.102286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
168
|
Bhosle A, Wang Y, Franzosa EA, Huttenhower C. Progress and opportunities in microbial community metabolomics. Curr Opin Microbiol 2022; 70:102195. [PMID: 36063685 DOI: 10.1016/j.mib.2022.102195] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 01/25/2023]
Abstract
The metabolome lies at the interface of host-microbiome crosstalk. Previous work has established links between chemically diverse microbial metabolites and a myriad of host physiological processes and diseases. Coupled with scalable and cost-effective technologies, metabolomics is thus gaining popularity as a tool for characterization of microbial communities, particularly when combined with metagenomics as a window into microbiome function. A systematic interrogation of microbial community metabolomes can uncover key microbial compounds, metabolic capabilities of the microbiome, and also provide critical mechanistic insights into microbiome-linked host phenotypes. In this review, we discuss methods and accompanying resources that have been developed for these purposes. The accomplishments of these methods demonstrate that metabolomes can be used to functionally characterize microbial communities, and that microbial properties can be used to identify and investigate chemical compounds.
Collapse
Affiliation(s)
- Amrisha Bhosle
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Ya Wang
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Eric A Franzosa
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Curtis Huttenhower
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
169
|
Intestinal Ecology Changes in Diarrheic Père David's Deer Revealed by Gut Microbiota and Fecal Metabolites Analysis. Animals (Basel) 2022; 12:ani12233366. [PMID: 36496887 PMCID: PMC9737761 DOI: 10.3390/ani12233366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 12/02/2022] Open
Abstract
Diarrhea is one of the most common diseases affecting the health of Père David's deer (Elaphurus davidianus). It is believed that an imbalanced intestinal ecology contributes to the etiology of the condition. However, little is known about how the intestinal ecology changes in these diarrheic animals. In this study, 16S rRNA gene sequencing and ultra-high performance liquid chromatography combined with tandem mass spectrometry (UPLC-MS/MS) were used to investigate the gut microbiota and fecal metabolites in five Père David's deer with diarrhea. The results showed that when compared with healthy individuals, considerable changes in the gut microbiome were observed in diarrheic animals, including a significant reduction in microbial diversity and gut microbiota composition alterations. Furthermore, the profiles of numerous fecal metabolites were altered in diarrheic individuals, showing large-scale metabolite dysregulation. Among metabolites, acylcarnitines, lysophosphatidylcholine, bile acids, and oxidized lipids were elevated significantly. Constantly, several metabolic pathways were significantly altered. Interestingly, predicted metabolic pathways based on 16S rRNA gene sequence and differential metabolite analysis showed that lipid metabolism, cofactor, and vitamin metabolism were altered in sick animals, indicating microbiota-host crosstalk in these deer. When combined, the results provide the first comprehensive description of an intestinal microbiome and metabolic imbalance in diarrheic Père David's deer, which advances our understanding and potential future treatment of diarrheic animals.
Collapse
|
170
|
Shi T, Feng Y, Liu W, Liu H, Li T, Wang M, Li Z, Lu J, Abudurexiti A, Maimaitireyimu A, Hu J, Gao F. Characteristics of gut microbiota and fecal metabolomes in patients with celiac disease in Northwest China. Front Microbiol 2022; 13:1020977. [PMID: 36519162 PMCID: PMC9742481 DOI: 10.3389/fmicb.2022.1020977] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/14/2022] [Indexed: 06/30/2024] Open
Abstract
Celiac disease (CD) is an autoimmune small bowel disease. The pattern of gut microbiota is closely related to dietary habits, genetic background, and geographical factors. There is a lack of research on CD-related gut microbiota in China. This study aimed to use 16S rDNA sequencing and metabolomics to analyze the fecal microbial composition and metabolome characteristics in patients diagnosed with CD in Northwest China, and to screen potential biomarkers that could be used for its diagnosis. A significant difference in the gut microbiota composition was observed between the CD and healthy controls groups. At the genus level, the abundance of Streptococcus, Lactobacillus, Veillonella, and Allisonella communities in the CD group were increased (Q < 0.05). Furthermore, the abundance of Ruminococcus, Faecalibacterium, Blautia, Gemmiger, and Anaerostipes community in this group were decreased (Q < 0.05). A total of 222 different fecal metabolites were identified in the two groups, suggesting that CD patients have a one-carbon metabolism defect. Four species of bacteria and six metabolites were selected as potential biomarkers using a random forest model. Correlation analysis showed that changes in the gut microbiota were significantly correlated with changes in fecal metabolite levels. In conclusion, the patterns of distribution of gut microbiota and metabolomics in patients with CD in Northwest China were found to be unique to these individuals. This has opened up a new way to explore potential beneficial effects of supplementing specific nutrients and potential diagnostic and therapeutic targets in the future.
Collapse
Affiliation(s)
- Tian Shi
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
| | - Yan Feng
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
| | - Weidong Liu
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
| | - Huan Liu
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
| | - Ting Li
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
| | - Man Wang
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
| | - Ziqiong Li
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
| | - Jiajie Lu
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
| | - Adilai Abudurexiti
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
| | - Ayinuer Maimaitireyimu
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
| | - Jiali Hu
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
| | - Feng Gao
- Department of Gastroenterology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
| |
Collapse
|
171
|
Caffeine-Induced Sleep Restriction Alters the Gut Microbiome and Fecal Metabolic Profiles in Mice. Int J Mol Sci 2022; 23:ijms232314837. [PMID: 36499163 PMCID: PMC9737546 DOI: 10.3390/ijms232314837] [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: 10/07/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 12/02/2022] Open
Abstract
Insufficient sleep is becoming increasingly common and contributes to many health issues. To combat sleepiness, caffeine is consumed daily worldwide. Thus, caffeine consumption and sleep restriction often occur in succession. The gut microbiome can be rapidly affected by either one's sleep status or caffeine intake, whereas the synergistic effects of a persistent caffeine-induced sleep restriction remain unclear. In this study, we investigated the impact of a chronic caffeine-induced sleep restriction on the gut microbiome and its metabolic profiles in mice. Our results revealed that the proportion of Firmicutes and Bacteroidetes was not altered, while the abundance of Proteobacteria and Actinobacteria was significantly decreased. In addition, the content of the lipids was abundant and significantly increased. A pathway analysis of the differential metabolites suggested that numerous metabolic pathways were affected, and the glycerophospholipid metabolism was most significantly altered. Combined analysis revealed that the metabolism was significantly affected by variations in the abundance and function of the intestinal microorganisms and was closely relevant to Proteobacteria and Actinobacteria. In conclusion, a long-term caffeine-induced sleep restriction affected the diversity and composition of the intestinal microbiota in mice, and substantially altered the metabolic profiles of the gut microbiome. This may represent a novel mechanism by which an unhealthy lifestyle such as mistimed coffee breaks lead to or exacerbates disease.
Collapse
|
172
|
Wang XL, Yu N, Wang C, Zhou HR, Wu C, Yang L, Wei S, Miao AJ. Changes in Gut Microbiota Structure: A Potential Pathway for Silver Nanoparticles to Affect the Host Metabolism. ACS NANO 2022; 16:19002-19012. [PMID: 36315867 DOI: 10.1021/acsnano.2c07924] [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] [Indexed: 06/16/2023]
Abstract
Silver nanoparticles (AgNPs) are one of the most widely used NPs. Their adverse effects on either the host or its gut microbiota (GM) have been examined. Nevertheless, whether the GM plays any role in AgNP toxicity to the host remains unclear. In the present study, AgNPs were administered to mice by oral gavage once a day for 120 days. A significant dose-dependent accumulation of Ag in the liver was observed, with a steady state reached within 21 days. The AgNPs changed the structure of the GM, mainly with respect to microorganisms involved in the metabolism of energy, amino acids, organic acids, and lipids, as predicted in a PICRUST analysis. Effects of the AgNPs on liver metabolism were also demonstrated, as a KEGG pathway analysis showed the enrichment of pathways responsible for the metabolism of amino acids, purines and pyrimidine, lipids, and energy. More interestingly, the changes in GM structure and liver metabolism were highly correlated, evidenced by the correlation between ∼23% of the differential microorganisms at the genus level and ∼60% of the differential metabolites. This implies that the metabolic variations in liver as affected by AgNPs were partly attributable to NP-induced changes of GM structure. Therefore, our results demonstrate the importance of considering the roles of GM in the toxicity of NPs to the host in evaluations of the health risks of NPs.
Collapse
Affiliation(s)
- Xin-Lei Wang
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu Province 210023, China
| | - Nanyang Yu
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu Province 210023, China
| | - Chuan Wang
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu Province 210023, China
| | - Hao-Ran Zhou
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu Province 210023, China
| | - Chao Wu
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu Province 210023, China
| | - Liuyan Yang
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu Province 210023, China
| | - Si Wei
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu Province 210023, China
| | - Ai-Jun Miao
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu Province 210023, China
| |
Collapse
|
173
|
Ding J, Ouyang R, Zheng S, Wang Y, Huang Y, Ma X, Zou Y, Chen R, Zhuo Z, Li Z, Xin Q, Zhou L, Mei S, Yan J, Lu X, Ren Z, Liu X, Xu G. Effect of Breastmilk Microbiota and Sialylated Oligosaccharides on the Colonization of Infant Gut Microbial Community and Fecal Metabolome. Metabolites 2022; 12:1136. [PMID: 36422276 PMCID: PMC9698434 DOI: 10.3390/metabo12111136] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/09/2022] [Accepted: 11/16/2022] [Indexed: 07/30/2023] Open
Abstract
The complex microbiota and sialylated oligosaccharides in breastmilk are important bioactive components that affect the gut microbiota. However, the effect of breastmilk microbiota and sialylated oligosaccharides on the gut microbiota during the neonatal period has been largely overlooked. Here, 16S rRNA gene sequencing and metabolomics analysis were applied to the breastmilk and feces of 69 newborns to clarify the link between breastmilk components and the newborn gut. Results showed that Staphylococcus, Enterococcus, and Bacteroides were commonly shared and positively correlated between breastmilk and the neonatal intestine and they were the main bacteria of breastmilk that interacted with the newborn fecal metabolome. Breastmilk Staphylococcus mainly interacted with amino acids, whereas Bacteroides was involved in the tryptophan, nucleotide, and vitamin metabolism. Breastmilk sialylated oligosaccharides were related to Bacteroides and amino acids of the newborn fecal metabolites. Moreover, Bacteroides was related to the interaction between breastmilk 3'-sialyllactose and newborn fecal metabolites in the mediation effect models. Finally, we pointed out that breastmilk Bacteroides was important in the milk-gut interaction, and it was negatively associated with waist circumference in infants aged 1 year. Our study provides a scientific basis for understanding the role of breastmilk in the development of newborn gut microbiota and metabolome.
Collapse
Affiliation(s)
- Juan Ding
- Department of Quality Control, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Runze Ouyang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian 116023, China
| | - Sijia Zheng
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian 116023, China
| | - Yanfeng Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian 116023, China
| | - Yan Huang
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiao Ma
- Department of Nursing, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yuxin Zou
- Liaocheng People’s Hospital, Liaocheng 252000, China
| | - Rong Chen
- Dalian Municipal Women and Children’s Medical Center (Group), Dalian 116011, China
| | - Zhihong Zhuo
- Department of Pediatric, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhen Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Qi Xin
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Lina Zhou
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian 116023, China
| | - Surong Mei
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jingyu Yan
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Xin Lu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian 116023, China
| | - Zhigang Ren
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xinyu Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian 116023, China
| | - Guowang Xu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Liaoning Province Key Laboratory of Metabolomics, Dalian 116023, China
| |
Collapse
|
174
|
Kelly PE, Ng HJ, Farrell G, McKirdy S, Russell RK, Hansen R, Rattray Z, Gerasimidis K, Rattray NJW. An Optimised Monophasic Faecal Extraction Method for LC-MS Analysis and Its Application in Gastrointestinal Disease. Metabolites 2022; 12:1110. [PMID: 36422250 PMCID: PMC9698041 DOI: 10.3390/metabo12111110] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/06/2022] [Accepted: 11/07/2022] [Indexed: 12/28/2023] Open
Abstract
Liquid chromatography coupled with mass spectrometry (LC-MS) metabolomic approaches are widely used to investigate underlying pathogenesis of gastrointestinal disease and mechanism of action of treatments. However, there is an unmet requirement to assess faecal metabolite extraction methods for large-scale metabolomics studies. Current methods often rely on biphasic extractions using harmful halogenated solvents, making automation and large-scale studies challenging. The present study reports an optimised monophasic faecal extraction protocol that is suitable for untargeted and targeted LC-MS analyses. The impact of several experimental parameters, including sample weight, extraction solvent, cellular disruption method, and sample-to-solvent ratio, were investigated. It is suggested that a 50 mg freeze-dried faecal sample should be used in a methanol extraction (1:20) using bead beating as the means of cell disruption. This is revealed by a significant increase in number of metabolites detected, improved signal intensity, and wide metabolic coverage given by each of the above extraction parameters. Finally, we addressed the applicability of the method on faecal samples from patients with Crohn's disease (CD) and coeliac disease (CoD), two distinct chronic gastrointestinal diseases involving metabolic perturbations. Untargeted and targeted metabolomic analysis demonstrated the ability of the developed method to detect and stratify metabolites extracted from patient groups and healthy controls (HC), highlighting characteristic changes in the faecal metabolome according to disease. The method developed is, therefore, suitable for the analysis of patients with gastrointestinal disease and can be used to detect and distinguish differences in the metabolomes of CD, CoD, and HC.
Collapse
Affiliation(s)
- Patricia E. Kelly
- Strathclyde Institute of Pharmacy and Biomedical Sciences (SIPBS), University of Strathclyde, Glasgow G4 0RE, UK
- Bacteria, Immunology, Nutrition, Gastroenterology and Omics (BINGO) Group, University of Glasgow, Glasgow G12 8QQ, UK
| | - H Jene Ng
- School of Medicine, Dentistry & Nursing, University of Glasgow, Glasgow Royal Infirmary, Glasgow G12 8QQ, UK
| | - Gillian Farrell
- Strathclyde Institute of Pharmacy and Biomedical Sciences (SIPBS), University of Strathclyde, Glasgow G4 0RE, UK
| | - Shona McKirdy
- Bacteria, Immunology, Nutrition, Gastroenterology and Omics (BINGO) Group, University of Glasgow, Glasgow G12 8QQ, UK
- School of Medicine, Dentistry & Nursing, University of Glasgow, Glasgow Royal Infirmary, Glasgow G12 8QQ, UK
| | - Richard K. Russell
- Bacteria, Immunology, Nutrition, Gastroenterology and Omics (BINGO) Group, University of Glasgow, Glasgow G12 8QQ, UK
- Royal Hospital for Children and Young People, 50 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Richard Hansen
- Bacteria, Immunology, Nutrition, Gastroenterology and Omics (BINGO) Group, University of Glasgow, Glasgow G12 8QQ, UK
- Royal Hospital for Children, 1345 Govan Road, Glasgow G52 4TF, UK
| | - Zahra Rattray
- Strathclyde Institute of Pharmacy and Biomedical Sciences (SIPBS), University of Strathclyde, Glasgow G4 0RE, UK
| | - Konstantinos Gerasimidis
- Bacteria, Immunology, Nutrition, Gastroenterology and Omics (BINGO) Group, University of Glasgow, Glasgow G12 8QQ, UK
- School of Medicine, Dentistry & Nursing, University of Glasgow, Glasgow Royal Infirmary, Glasgow G12 8QQ, UK
| | - Nicholas J. W. Rattray
- Strathclyde Institute of Pharmacy and Biomedical Sciences (SIPBS), University of Strathclyde, Glasgow G4 0RE, UK
- Bacteria, Immunology, Nutrition, Gastroenterology and Omics (BINGO) Group, University of Glasgow, Glasgow G12 8QQ, UK
| |
Collapse
|
175
|
Karl JP, Armstrong NJ, Player RA, Rood JC, Soares JW, McClung HL. The Fecal Metabolome Links Diet Composition, Foacidic positive ion conditions, chromatographicallyod Processing, and the Gut Microbiota to Gastrointestinal Health in a Randomized Trial of Adults Consuming a Processed Diet. J Nutr 2022; 152:2343-2357. [PMID: 36774101 DOI: 10.1093/jn/nxac161] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/17/2022] [Accepted: 07/15/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Food processing alters diet digestibility and composition, thereby influencing interactions between host biology, diet, and the gut microbiota. The fecal metabolome offers insight into those relations by providing a readout of diet-microbiota interactions impacting host health. OBJECTIVES The aims were to determine the effects of consuming a processed diet on the fecal metabolome and to explore relations between changes in the fecal metabolome with fecal microbiota composition and gastrointestinal health markers. METHODS This was a secondary analysis of a randomized controlled trial wherein healthy adults [94% male; 18-61 y; BMI (kg/m2): 26 ± 3] consumed their usual diet [control (CON), n = 27] or a Meal, Ready-to-EatTM (Ameriqual Packaging) military ration diet composed of processed, shelf-stable, ready-to-eat items for 21 d (MRE; n = 27). Fecal metabolite profiles, fecal microbiota composition, biomarkers of intestinal barrier function, and gastrointestinal symptoms were measured before and after the intervention. Between-group differences and associations were assessed using nonparametric t tests, partial least-squares discriminant analysis, correlation, and redundancy analysis. RESULTS Fecal concentrations of multiple dipeptides [Mann-Whitney effect size (ES) = 0.27-0.50] and long-chain SFAs (ES = 0.35-0.58) increased, whereas plant-derived compounds (ES = 0.31-0.60) decreased in MRE versus CON (P < 0.05; q < 0.20). Changes in dipeptides correlated positively with changes in fecal concentrations of Maillard-reaction products (ρ = 0.29-0.70; P < 0.05) and inversely with changes in serum prealbumin (ρ = -0.30 to -0.48; P ≤ 0.03). Multiple bile acids, coffee and caffeine metabolites, and plant-derived compounds were associated with both fecal microbiota composition and gastrointestinal health markers, with changes in fecal microbiota composition explaining 26% of the variability within changes in gastrointestinal health-associated fecal metabolites (P = 0.001). CONCLUSIONS Changes in the fecal metabolomes of adults consuming a Meal, Ready-to-EatTM diet implicate interactions between diet composition, diet digestibility, and the gut microbiota as contributing to variability within gastrointestinal responses to the diet. Findings underscore the need to consider both food processing and nutrient composition when investigating the impact of diet-gut microbiota interactions on health outcomes. This trial was registered at www. CLINICALTRIALS gov as NCT02423551.
Collapse
Affiliation(s)
- J Philip Karl
- Military Nutrition Division, US Army Research Institute of Environmental Medicine, Natick, MA, USA.
| | - Nicholes J Armstrong
- Military Nutrition Division, US Army Research Institute of Environmental Medicine, Natick, MA, USA
| | - Robert A Player
- Johns Hopkins University Applied Physics Laboratory, Laurel, MD, USA
| | | | - Jason W Soares
- Soldier Effectiveness Directorate, US Army Combat Capabilities Development Command Soldier Center, Natick, MA, USA
| | - Holly L McClung
- Military Nutrition Division, US Army Research Institute of Environmental Medicine, Natick, MA, USA
| |
Collapse
|
176
|
Moutinho TJ, Powers DA, Hanson GF, Levy S, Baveja R, Hefner I, Mohamed M, Abdelghani A, Baker RL, Papin JA, Moore SR, Hourigan SK. Fecal sphingolipids predict parenteral nutrition-associated cholestasis in the neonatal intensive care unit. JPEN J Parenter Enteral Nutr 2022; 46:1903-1913. [PMID: 35285019 PMCID: PMC9468188 DOI: 10.1002/jpen.2374] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/23/2022] [Accepted: 03/06/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND Parenteral nutrition-associated cholestasis (PNAC) in the neonatal intensive care unit (NICU) causes significant morbidity and associated healthcare costs. Laboratory detection of PNAC currently relies on elevated serum conjugated bilirubin levels in the aftermath of impaired bile flow. Here, we sought to identify fecal biomarkers, which when integrated with clinical data, would better predict risk for developing PNAC. METHODS Using untargeted metabolomics in 200 serial stool samples from 60 infants, we applied statistical and machine learning approaches to identify clinical features and metabolic biomarkers with the greatest associative potential for risk of developing PNAC. Stools were collected prospectively from infants receiving PN with soybean oil-based lipid emulsion at a level IV NICU. RESULTS Low birth weight, extreme prematurity, longer duration of PN, and greater number of antibiotic courses were all risk factors for PNAC (P < 0.05). We identified 78 stool biomarkers with early predictive potential (P < 0.05). From these 78 biomarkers, we further identified 12 sphingomyelin lipids with high association for the development of PNAC in precholestasis stool samples when combined with birth anthropometry. CONCLUSION We demonstrate the potential for stool metabolomics to enhance early identification of PNAC risk. Earlier detection of high-risk infants would empower proactive mitigation with alterations to PN for at-risk infants and optimization of energy nutrition with PN for infants at lower risk.
Collapse
Affiliation(s)
- Thomas J. Moutinho
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Deborah A. Powers
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Gabriel F. Hanson
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Shira Levy
- Inova Children's HospitalFalls ChurchVirginiaUSA
| | - Rajiv Baveja
- Fairfax Neonatal AssociatesFalls ChurchVirginiaUSA
| | | | | | | | | | - Jason A. Papin
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Sean R. Moore
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of PediatricsUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Suchitra K. Hourigan
- Inova Children's HospitalFalls ChurchVirginiaUSA,Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of PediatricsUniversity of VirginiaCharlottesvilleVirginiaUSA,Division of Pediatric GastroenterologyPediatric Specialists of VirginiaFairfaxVirginiaUSA,Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious DiseasesNational Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
177
|
Alrahawy M, Javed S, Atif H, Elsanhoury K, Mekhaeil K, Eskander G. Microbiome and Colorectal Cancer Management. Cureus 2022; 14:e30720. [DOI: 10.7759/cureus.30720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2022] [Indexed: 11/06/2022] Open
|
178
|
Jiang CH, Fang X, Huang W, Guo JY, Chen JY, Wu HY, Li ZS, Zou WB, Liao Z. Alterations in the Gut Microbiota and Metabolomics of Seafarers after a Six-Month Sea Voyage. Microbiol Spectr 2022; 10:e0189922. [PMID: 36197290 PMCID: PMC9603232 DOI: 10.1128/spectrum.01899-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 09/15/2022] [Indexed: 12/31/2022] Open
Abstract
Maintaining the health of seafarers is a difficult task during long-term voyages. Little is known about the corresponding changes in the gut microbiome-host interaction. This study recruited 30 seafarers undertaking a 6-month voyage and analyzed their gut microbiota using 16S rRNA gene sequencing. Fecal untargeted metabolomics analysis was performed using liquid chromatography-mass spectrometry. Significant changes in the composition of the gut microbiota and an increased ratio of Firmicutes/Bacteroidetes at the end (day 180) of the 6-month voyage, relative to the start (day 0), were observed. At the genus level, the abundances of Holdemanella and Plesiomonas were significantly increased, while the abundance of Bacteroides was decreased. Predicted microbial functional analysis revealed significant decreases in folate biosynthesis and biotin metabolism. Furthermore, 20 differential metabolites within six differentially enriched human metabolic pathways (including arginine biosynthesis, lysine degradation, phenylalanine metabolism, sphingolipid metabolism, pentose and glucuronate interconversions, and glycine, serine, and threonine metabolism) were identified by comparing the fecal metabolites at day 0 and day 180. Spearman correlation analysis revealed close relationships between the 14 differential microbiota members and the six differential fecal metabolites that might affect specific human metabolic pathways. This study adopted a multi-omics approach and provides potential targets for maintaining the health of seafarers during long-term voyages. These findings are worthy of more in-depth exploration in future studies. IMPORTANCE Maintaining the health of seafarers undertaking long-term voyages is a difficult task. Apart from the alterations in the gut microbiome and fecal metabolites after a long-term voyage, our study also revealed that 20 differential metabolites within six differentially enriched human metabolic pathways are worthy of attention. Moreover, we found close relationships between the 14 differential microbiota members and the six differential fecal metabolites that might impact specific human metabolic pathways. Accordingly, preventative measures, such as adjusting the gut microbiota by decreasing potential pathobionts or increasing potential probiotics as well as offsetting the decrease in B vitamins and beneficial metabolites (e.g., d-glucuronic acid and citrulline) via dietary adjustment or nutritional supplements, might improve the health of seafarers during long-term sea voyages. These findings provide valuable clues about gut microbiome-host interactions and propose potential targets for maintaining the health of seafarers engaged in long-term sea voyages.
Collapse
Affiliation(s)
- Chun-Hui Jiang
- Department of Gastroenterology, Digestive Endoscopy Center, Changhai Hospital, Shanghai, China
- Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Xue Fang
- Department of Gastroenterology, Digestive Endoscopy Center, Changhai Hospital, Shanghai, China
| | - Wen Huang
- Department of Gastroenterology, Digestive Endoscopy Center, Changhai Hospital, Shanghai, China
| | - Ji-Yao Guo
- Department of Gastroenterology, Digestive Endoscopy Center, Changhai Hospital, Shanghai, China
| | - Jia-Yun Chen
- Department of Gastroenterology, Digestive Endoscopy Center, Changhai Hospital, Shanghai, China
| | - Hong-Yu Wu
- Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Zhao-Shen Li
- Department of Gastroenterology, Digestive Endoscopy Center, Changhai Hospital, Shanghai, China
- Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Wen-Bin Zou
- Department of Gastroenterology, Digestive Endoscopy Center, Changhai Hospital, Shanghai, China
- Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Zhuan Liao
- Department of Gastroenterology, Digestive Endoscopy Center, Changhai Hospital, Shanghai, China
- Shanghai Institute of Pancreatic Diseases, Shanghai, China
| |
Collapse
|
179
|
Jian T, Zhou L, Chen Y, Tian Y, Wu R, Tong B, Niu G, Gai Y, Li W, Chen J. Total Sesquiterpenoids of Loquat Leaves Alleviated High-Fat Diet-Induced Obesity by Targeting Fecal Metabolic Profiling and Gut Microbiota Composition. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:13279-13288. [PMID: 36198678 DOI: 10.1021/acs.jafc.2c04900] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
In the present study, we demonstrated that whether the gut microbiota and related metabolites contribute to the therapeutic effect of total sesquiterpenoids (TSs) from loquat leaves on obesity. A 4-week high fat diet was used to induce obesity which was then treated with TSs for another 4 weeks. TSs remarkedly reduced the weight of body and white adipose and the levels of total cholesterol (TC) and triglyceride (TG) in serum. We also found that TSs restored the diversity and richness of gut microbiota. In addition, TSs administration affected the relative abundance of seven key genera. Meanwhile, TSs were determined to affect the metabolism of the host through detecting the metabolites in feces. By applying KEGG and the correlation analysis with gut microbiota, 10 differential metabolites were identified to be the key. The results in this work proved that TSs inhibited obesity by remodeling gut microbiota and related metabolites.
Collapse
Affiliation(s)
- Tunyu Jian
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
| | - Lina Zhou
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
- Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yan Chen
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
- Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yuwen Tian
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
- Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ruoyun Wu
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
| | - Bei Tong
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
| | - Guanting Niu
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
| | - Yanan Gai
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
| | - Weilin Li
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
- Co-Innovation Center for Sustainable Forestry in Southern China, Forestry College, Nanjing Forestry University, Nanjing 210037, China
| | - Jian Chen
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
- Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
180
|
Jiang T, Li Y, Li L, Liang T, Du M, Yang L, Yang J, Yang R, Zhao H, Chen M, Ding Y, Zhang J, Wang J, Xie X, Wu Q. Bifidobacterium longum 070103 Fermented Milk Improve Glucose and Lipid Metabolism Disorders by Regulating Gut Microbiota in Mice. Nutrients 2022; 14:nu14194050. [PMID: 36235706 PMCID: PMC9573661 DOI: 10.3390/nu14194050] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/21/2022] [Accepted: 09/23/2022] [Indexed: 12/08/2022] Open
Abstract
Background: Fermented milk is beneficial for metabolic disorders, while the underlying mechanisms of action remain unclear. This study explored the benefits and underlying mechanisms of Bifidobacterium longum 070103 fermented milk (BLFM) in thirteen-week high-fat and high-sugar (HFHS) fed mice using omics techniques. Methods and results: BLFM with activated glucokinase (GK) was screened by a double-enzyme coupling method. After supplementing BLFM with 10 mL/kg BW per day, fasting blood glucose, total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), and leptin were significantly reduced compared with the HFHS group. Among them, the final body weight (BW), epididymal fat, perirenal fat, and brown fat in BLFM group had better change trends than Lacticaseibacillus rhamnosus GG fermented milk (LGGFM) group. The amplicon and metabolomic data analysis identified Bifibacterium as a key gut microbiota at regulating glycolipid metabolism. BLFM reverses HFHS-induced reduction in bifidobacteria abundance. Further studies showed that BLFM significantly reduces the content of 3-indoxyl sulofphate associated with intestinal barrier damage. In addition, mice treated with BLFM improved BW, glucose tolerance, insulin resistance, and hepatic steatosis. Conclusion: BLFM consumption attenuates obesity and related symptoms in HFHS-fed mice probably via the modulation of gut microbes and metabolites.
Collapse
Affiliation(s)
- Tong Jiang
- College of Food Science, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, 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 510070, China
| | - Ying Li
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, 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 510070, China
| | - Longyan Li
- College of Food Science, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, 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 510070, China
| | - Tingting Liang
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, 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 510070, China
| | - Mingzhu Du
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, 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 510070, China
| | - Lingshuang Yang
- College of Food Science, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, 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 510070, China
| | - Juan Yang
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, 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 510070, China
| | - Runshi Yang
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, 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 510070, China
| | - Hui Zhao
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, 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 510070, China
| | - Moutong Chen
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, 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 510070, China
| | - Yu Ding
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, 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 510070, China
| | - Jumei Zhang
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, 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 510070, China
| | - Juan Wang
- College of Food Science, South China Agricultural University, Guangzhou 510642, China
- Correspondence: (J.W.); (X.X.); (Q.W.)
| | - Xinqiang Xie
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, 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 510070, China
- Correspondence: (J.W.); (X.X.); (Q.W.)
| | - Qingping Wu
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, 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 510070, China
- Correspondence: (J.W.); (X.X.); (Q.W.)
| |
Collapse
|
181
|
Chen Z, Chen L, Sun Y, Li N, Chen R, Ma Y, Song W, Shi H, Xia L, Yao G. Association of differential meat quality traits with gut microbiota
in Angus cattle and Xinjiang Brown cattle. JOURNAL OF ANIMAL AND FEED SCIENCES 2022. [DOI: 10.22358/jafs/153077/2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
|
182
|
Chen X, Li S, Lin C, Zhang Z, Liu X, Wang C, Chen J, Yang B, Yuan J, Zhang Z. Isomaltooligosaccharides inhibit early colorectal carcinogenesis in a 1,2-dimethylhydrazine-induced rat model. Front Nutr 2022; 9:995126. [PMID: 36185671 PMCID: PMC9521046 DOI: 10.3389/fnut.2022.995126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Colon cancer (CC) is a multistage disease and one of the most common cancers worldwide. Establishing an effective treatment strategies of early colon cancer is of great significance for preventing its development and reducing mortality. The occurrence of colon cancer is closely related to changes in the intestinal flora structure. Therefore, remodelling the intestinal flora structure through prebiotics is a powerful approach for preventing and treating the occurrence and development of colon cancer. Isomaltooligosaccharides (IMOs) are often found in fermented foods and can directly reach the gut for use by microorganisms. In this study, a rat model of early colon cancer (DMH) was established by subcutaneous injection of 1,2-dimethylhydrazine, and the model rats were fed IMOs as a dietary intervention (DI). The untargeted faecal metabolomics, gut metabolome and intestinal function of the model rats were investigated. The results showed that DMH, DI and IMOs alone (IMOs) groups exhibited gut microbial community changes. In the DI group, there was an increased abundance of probiotics (Lactobacillus) and decreased abundance of CC marker bacteria (Fusobacterium). The key variations in the faecal metabolites of the DI group included decreased levels of glucose, bile acids (including deoxycholic acid and chenodeoxycholic acid) and amino acids (including L-glutamic acid and L-alanine). In addition, dietary intake of IMOs attenuated the intestinal inflammatory response, improved the intestinal microecological environment, and slowed the development of DMH-induced early CC in rats. This work provides a theoretical basis and technical support for the clinical prevention or treatment of CC with prebiotics.
Collapse
|
183
|
Activity fingerprinting of polysaccharides on oral, gut, pancreas and lung microbiota in diabetic rats. Biomed Pharmacother 2022; 155:113681. [PMID: 36108392 DOI: 10.1016/j.biopha.2022.113681] [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: 06/25/2022] [Revised: 09/01/2022] [Accepted: 09/05/2022] [Indexed: 11/23/2022] Open
Abstract
The modern rise in type 2 diabetes mellitus (T2DM) and its correlation to commensal microbiota have elicited global concern about the patterns of microbial action in the host. With the exception of that linked to gut, microbiota were also colonized in pancreas, oral, and lung, contributing to the physiopathology of T2DM. In this study, we aimed to explore the protective effects of Ganoderma atrum polysaccharide (PSG) and White Hyacinth Bean polysaccharide (WHBP) on the intestine, pancreas, oral, and lung microbiota in T2DM rats. Here we showed that, despite capacities of polysaccharides that exerted similar protective effects on hyperglycemia, dyslipidemia, insulin resistance and dysbacteriosis in T2DM rats, PSG and WHBP were able to be characterized by their own "target" bacteria, which could be proposed for activity-fingerprinting of polysaccharide species. Furthermore, we found a mutual bacteria spectrum in the pancreas and lung, and most bacteria could be tracked to oral or gut samples. Notably, the overlapping areas of the microbiota profile between organs (pancreas, lung) and saliva were more than in the gut, suggesting that a saliva sample was also of interest to serve as a "telltale sign" for judging pancreatic injury. Together, these microbiota interactions provided a new potential to harvest alternative samples for disease surveillance. Meanwhile, polysaccharides had anti-T2DM abilities, which could be distinguished by their own characteristic bacteria.
Collapse
|
184
|
Microbial Tryptophan Metabolism Tunes Host Immunity, Metabolism, and Extraintestinal Disorders. Metabolites 2022; 12:metabo12090834. [PMID: 36144238 PMCID: PMC9505266 DOI: 10.3390/metabo12090834] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 11/17/2022] Open
Abstract
The trillions of commensal microorganisms comprising the gut microbiota have received growing attention owing to their impact on host physiology. Recent advances in our understandings of the host–microbiota crosstalk support a pivotal role of microbiota-derived metabolites in various physiological processes, as they serve as messengers in the complex dialogue between commensals and host immune and endocrine cells. In this review, we highlight the importance of tryptophan-derived metabolites in host physiology, and summarize the recent findings on the role of tryptophan catabolites in preserving intestinal homeostasis and fine-tuning immune and metabolic responses. Furthermore, we discuss the latest evidence on the effects of microbial tryptophan catabolites, describe their mechanisms of action, and discuss how perturbations of microbial tryptophan metabolism may affect the course of intestinal and extraintestinal disorders, including inflammatory bowel diseases, metabolic disorders, chronic kidney diseases, and cardiovascular diseases.
Collapse
|
185
|
Preethy S, Ikewaki N, Levy GA, Raghavan K, Dedeepiya VD, Yamamoto N, Srinivasan S, Ranganathan N, Iwasaki M, Senthilkumar R, Abraham SJK. Two unique biological response-modifier glucans beneficially regulating gut microbiota and faecal metabolome in a non-alcoholic steatohepatitis animal model, with potential applications in human health and disease. BMJ Open Gastroenterol 2022; 9:e000985. [PMID: 36167455 PMCID: PMC9516208 DOI: 10.1136/bmjgast-2022-000985] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 09/04/2022] [Indexed: 11/20/2022] Open
Abstract
OBJECTIVE The gut microbiome and its metabolites are influenced by age and stress and reflect the metabolism and health of the immune system. We assessed the gut microbiota and faecal metabolome in a static animal model of non-alcoholic steatohepatitis (NASH). DESIGN This model was subjected to the following treatments: reverse osmosis water, AFO-202, N-163, AFO-202+N-163 and telmisartan treatment. Faecal samples were collected at 6 and 9 weeks of age. The gut microbiome was analysed using 16S ribosomal RNA sequences acquired by next-generation sequencing, and the faecal metabolome was analysed using gas chromatography-mass spectrometry. RESULTS Gut microbial diversity increased greatly in the AFO-202+N-163 group. Postintervention, the abundance of Firmicutes decreased, whereas that of Bacteroides increased and was the highest in the AFO-202+N-163 group. The decrease in the abundance of Enterobacteriaceae and other Firmicutes and the abundance of Turicibacter and Bilophila were the highest in the AFO-202 and N-163 groups, respectively. Lactobacillus abundance was highest in the AFO-202+N-163 group. The faecal metabolite spermidine, which is beneficial against inflammation and NASH, was significantly decreased (p=0.012) in the N-163 group. Succinic acid, which is beneficial in neurodevelopmental and neurodegenerative diseases, was increased in the AFO-202 group (p=0.06). The decrease in fructose was the highest in the N-163 group (p=0.0007). Isoleucine and Leucine decreased with statistical significance (p=0.004 and 0.012, respectively), and tryptophan also decreased (p=0.99), whereas ornithine, which is beneficial against chronic immune-metabolic-inflammatory pathologies, increased in the AFO-202+N-163 group. CONCLUSION AFO-202 treatment in mice is beneficial against neurodevelopmental and neurodegenerative diseases, and has prophylactic potential against metabolic conditions. N-163 treatment exerts anti-inflammatory effects against organ fibrosis and neuroinflammation. In combination, these compounds exhibit anticancer activity.
Collapse
Affiliation(s)
- Senthilkumar Preethy
- Fujio-Eiji Academic Terrain (FEAT), Nichi-In Centre for Regenerative Medicine (NCRM), Chennai, India
| | - Nobunao Ikewaki
- Dept. of Medical Life Science, Kyushu University of Health and Welfare, Nobeoka, Japan
- Institute of Immunology, Junsei Educational Institute, Nobeoka, Japan
| | - Gary A Levy
- Medicine and Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Kadalraja Raghavan
- Dept of Paediatric Neurology, Jesuit Antonyraj memorial Inter-disciplinary Centre for Advanced Recovery and Education (JAICARE), Madurai, India
| | | | - Naoki Yamamoto
- Genome Medical Sciences Project, National Center for Global Health and Medicine (NCGM), Kohnodai, Chiba, Japan
| | - Subramaniam Srinivasan
- Mary-Yoshio Translational Hexagon (MYTH), Nichi-In Centre for Regenerative Medicine (NCRM), Chennai, India
| | | | - Masaru Iwasaki
- Centre for Advancing Clinical Research (CACR), University of Yamanashi - School of Medicine, Chuo, Japan
| | - Rajappa Senthilkumar
- Fujio-Eiji Academic Terrain (FEAT), Nichi-In Centre for Regenerative Medicine (NCRM), Chennai, India
| | - Samuel J K Abraham
- Mary-Yoshio Translational Hexagon (MYTH), Nichi-In Centre for Regenerative Medicine (NCRM), Chennai, India
- Centre for Advancing Clinical Research (CACR), University of Yamanashi - School of Medicine, Chuo, Japan
- Antony- Xavier Interdisciplinary Scholastics (AXIS), GN Corporation Co. Ltd, Kofu, Japan
| |
Collapse
|
186
|
Chen L, Xu R, McDonald JD, Bruno RS, Choueiry F, Zhu J. Dairy Milk Casein and Whey Proteins Differentially Alter the Postprandial Lipidome in Persons with Prediabetes: A Comparative Lipidomics Study. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:10209-10220. [PMID: 35948437 PMCID: PMC10352119 DOI: 10.1021/acs.jafc.2c03662] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Dairy milk, likely through its bioactive proteins, has been reported to attenuate postprandial hyperglycemia-induced oxidative stress responses implicated in cardiovascular diseases (CVDs). However, how its major proteins, whey and casein, alter metabolic excursions of the lipidome in persons with prediabetes is unclear. Therefore, the objective of this study was to examine whey or casein protein ingestion on glucose-induced alternations in lipidomic responses in adults (17 males and 6 females) with prediabetes. In this clinical study, participants consumed glucose alone, glucose + nonfat milk (NFM), or glucose with either whey (WHEY) or casein (CASEIN) protein, and plasma samples were collected at multiple time points. Lipidomics data from plasma samples was acquired using an ultra-high-performance liquid chromatography-high-resolution mass spectrometry-based platform. Our results indicated that glucose ingestion alone induced the largest number of changes in plasma lipids. WHEY showed an earlier and stronger impact to maintain the stability of the lipidome compared with CASEIN. WHEY protected against glucose-induced changes in glycerophospholipid and sphingolipid (SP) metabolism, while ether lipid metabolism and SP metabolism were the pathways most greatly impacted in CASEIN. Meanwhile, the decreased acyl carnitines and fatty acid (FA) 16:0 levels could attenuate lipid peroxidation after protein intervention to protect insulin secretory capacity. Diabetes-associated lipids, the increased phosphatidylethanolamine (PE) 34:2 and decreased phosphatidylcholine (PC) 34:3 in the NFM-T90 min, elevated PC 35:4 and decreased CE 18:1 to a CE 18:2 ratio in the WHEY-T180 min, decreased lysophosphatidylcholine (LPC) 22:6 and LPC 22:0/0:0 in the CASEIN-T90 min, and decreased PE 36:1 in the CASEIN-T180 min, indicating a decreased risk for prediabetes. Collectively, our study suggested that dairy milk proteins are responsible for the protective effect of non-fat milk on glucose-induced changes in the lipidome, which may potentially influence long-term CVD risk.
Collapse
Affiliation(s)
- Li Chen
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
- James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Rui Xu
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Joshua D. McDonald
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Richard S. Bruno
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Fouad Choueiry
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Jiangjiang Zhu
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
- James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
187
|
Beale DJ, Nguyen TV, Shah RM, Bissett A, Nahar A, Smith M, Gonzalez-Astudillo V, Braun C, Baddiley B, Vardy S. Host-Gut Microbiome Metabolic Interactions in PFAS-Impacted Freshwater Turtles ( Emydura macquarii macquarii). Metabolites 2022; 12:747. [PMID: 36005619 PMCID: PMC9415956 DOI: 10.3390/metabo12080747] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/27/2022] [Accepted: 08/03/2022] [Indexed: 01/09/2023] Open
Abstract
Per-and polyfluoroalkyl substances (PFAS) are a growing concern for humans, wildlife, and more broadly, ecosystem health. Previously, we characterised the microbial and biochemical impact of elevated PFAS on the gut microbiome of freshwater turtles (Emydura macquarii macquarii) within a contaminated catchment in Queensland, Australia. However, the understanding of PFAS impacts on this species and other aquatic organisms is still very limited, especially at the host-gut microbiome molecular interaction level. To this end, the present study aimed to apply these leading-edge omics technologies within an integrated framework that provides biological insight into the host turtle-turtle gut microbiome interactions of PFAS-impacted wild-caught freshwater turtles. For this purpose, faecal samples from PFAS-impacted turtles (n = 5) and suitable PFAS-free reference turtles (n = 5) were collected and analysed. Data from 16S rRNA gene amplicon sequencing and metabolomic profiling of the turtle faeces were integrated using MetOrigin to assign host, microbiome, and co-metabolism activities. Significant variation in microbial composition was observed between the two turtle groups. The PFAS-impacted turtles showed a higher relative abundance of Firmicutes and a lower relative abundance of Bacteroidota than the reference turtles. The faecal metabolome showed several metabolites and pathways significantly affected by PFAS exposure. Turtles exposed to PFAS displayed altered amino acid and butanoate metabolisms, as well as altered purine and pyrimidine metabolism. It is predicted from this study that PFAS-impacted both the metabolism of the host turtle and its gut microbiota which in turn has the potential to influence the host's physiology and health.
Collapse
Affiliation(s)
- David J. Beale
- Land and Water, Commonwealth Scientific and Industrial Research Organisation, Ecosciences Precinct, Dutton Park, QLD 4102, Australia
| | - Thao V. Nguyen
- Land and Water, Commonwealth Scientific and Industrial Research Organisation, Ecosciences Precinct, Dutton Park, QLD 4102, Australia
| | - Rohan M. Shah
- Land and Water, Commonwealth Scientific and Industrial Research Organisation, Ecosciences Precinct, Dutton Park, QLD 4102, Australia
| | - Andrew Bissett
- Oceans and Atmosphere, Commonwealth Scientific and Industrial Research Organisation, Battery Point, TAS 7004, Australia
| | - Akhikun Nahar
- Land and Water, Commonwealth Scientific and Industrial Research Organisation, Research and Innovation Park, Black Mountain, ACT 2601, Australia
| | - Matthew Smith
- NCMI, Commonwealth Scientific and Industrial Research Organisation, Battery Point, TAS 7004, Australia
| | | | - Christoph Braun
- Water Quality and Investigation, Science and Technology Division, Department of Environment and Science, Queensland Government, Dutton Park, QLD 4102, Australia
| | - Brenda Baddiley
- Water Quality and Investigation, Science and Technology Division, Department of Environment and Science, Queensland Government, Dutton Park, QLD 4102, Australia
| | - Suzanne Vardy
- Water Quality and Investigation, Science and Technology Division, Department of Environment and Science, Queensland Government, Dutton Park, QLD 4102, Australia
| |
Collapse
|
188
|
Huang Y, Lu J, Zhao Q, Chen J, Dong W, Lin M, Zheng H. Potential Therapeutic Mechanism of Traditional Chinese Medicine on Diabetes in Rodents: A Review from an NMR-Based Metabolomics Perspective. Molecules 2022; 27:molecules27165109. [PMID: 36014349 PMCID: PMC9414875 DOI: 10.3390/molecules27165109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/05/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
Traditional Chinese medicine (TCM) has been used to treat diabetes for a long time, but its application has not been widely accepted due to unstandardized product quality and complex pharmacological mechanisms. The modernization of TCM is crucial for its further development, and in recent years the metabolomics technique has largely driven its modernization. This review focuses on the application of NMR-based metabolomics in diabetic therapy using TCM. We identified a series of metabolic pathways that altered significantly after TCM treatment, providing a better understanding of the metabolic mechanisms of TCM for diabetes care.
Collapse
Affiliation(s)
- Yinli Huang
- Department of Endocrinology, Pingyang Affiliated Hospital of Wenzhou Medical University, Wenzhou 325400, China
| | - Jiahui Lu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Qihui Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Junli Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Wei Dong
- Department of Endocrinology, Pingyang Affiliated Hospital of Wenzhou Medical University, Wenzhou 325400, China
| | - Minjie Lin
- Department of Endocrinology, Pingyang Affiliated Hospital of Wenzhou Medical University, Wenzhou 325400, China
| | - Hong Zheng
- Department of Endocrinology, Pingyang Affiliated Hospital of Wenzhou Medical University, Wenzhou 325400, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Correspondence:
| |
Collapse
|
189
|
Miao Z, Chen GD, Huo S, Fu Y, Wu MY, Xu F, Jiang Z, Tang J, Gou W, Xiao C, Liu YP, Wu YY, Sun TY, Sun L, Shen LR, Lin X, Chen YM, Zheng JS. Interaction of n-3 polyunsaturated fatty acids with host CD36 genetic variant for gut microbiome and blood lipids in human cohorts. Clin Nutr 2022; 41:1724-1734. [DOI: 10.1016/j.clnu.2022.05.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/24/2022] [Accepted: 05/30/2022] [Indexed: 11/27/2022]
|
190
|
Cao C, Wang L, Ai C, Gong G, Wang Z, Huang L, Song S, Zhu B. Impact of Lycium barbarum arabinogalactan on the fecal metabolome in a DSS-induced chronic colitis mouse model. Food Funct 2022; 13:8703-8716. [PMID: 35912853 DOI: 10.1039/d2fo01283a] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Ulcerative colitis (UC) is often accompanied by the dysbiosis of gut microbiota and metabolism. Our previous study indicated that arabinogalactan from Lycium barbarum (LBP-3) could markedly attenuate the symptoms of chronic UC in mice by modulating the structure of gut microbiota. This study explored the impact of LBP-3 on the fecal metabolomic profiling of the same cohort of mice by HPLC-TripleTOF/MS. Untargeted metabolomic analyses indicated that supplementation with LBP-3 markedly reversed 18 of the 48 differential metabolites (mainly belonging to amino acids and organic acids) disturbed by DSS. Targeted metabolomics revealed that the lower levels of tryptophan, lysine, diiodothyronine, kynurenine, and betaine and higher levels of phenylalanine, leucine, glutamine, isoleucine, homoserine, (S)-2-hydroxyglutarate, 2-isopropylmalic acid, ascorbic acid, gluconic acid, and taurine, which were caused by DSS induction, were reversed by LBP-3 treatment. In addition, pathway analysis showed that the pentose phosphate pathway, phenylalanine metabolism, ascorbate and aldarate metabolism, and phenylalanine, tyrosine and tryptophan biosynthesis were strongly affected by LBP-3. More importantly, the above amino acids, organic acids, and metabolic pathways changed by LBP-3 were correlated with the abundance of gut microbiota such as Turicibacter, Lactobacillus, Parasutterella, Odoribacter, Veillonella, Faecalibacterium, and Ruminococcaceae. This study advances our understanding of the interaction between the microbiome and metabolomics in DSS-induced chronic colitis after LBP-3 treatment.
Collapse
Affiliation(s)
- Cui Cao
- Collaborative Innovation Center of Seafood Deep Processing, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China. .,National and Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian 116034, P. R. China.,Shaanxi Natural Carbohydrate Resource Engineering Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, P. R. China.
| | - Linlin Wang
- Collaborative Innovation Center of Seafood Deep Processing, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China. .,National and Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Chunqing Ai
- Collaborative Innovation Center of Seafood Deep Processing, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China. .,National and Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Guiping Gong
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, P. R. China.
| | - Zhongfu Wang
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, P. R. China.
| | - Linjuan Huang
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, P. R. China.
| | - Shuang Song
- Collaborative Innovation Center of Seafood Deep Processing, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China. .,National and Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Beiwei Zhu
- Collaborative Innovation Center of Seafood Deep Processing, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China. .,National and Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian 116034, P. R. China
| |
Collapse
|
191
|
Lee-Sarwar K, Dedrick S, Momeni B, Kelly RS, Zeiger RS, O'Connor GT, Sandel MT, Bacharier LB, Beigelman A, Laranjo N, Gold DR, Lasky-Su J, Litonjua AA, Liu YY, Weiss ST. Association of the gut microbiome and metabolome with wheeze frequency in childhood asthma. J Allergy Clin Immunol 2022; 150:325-336. [PMID: 35196534 PMCID: PMC9359927 DOI: 10.1016/j.jaci.2022.02.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 01/23/2022] [Accepted: 02/01/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND While the microbiome has an established role in asthma development, less is known about its contribution to morbidity in children with asthma. OBJECTIVE In this ancillary study of the Vitamin D Antenatal Asthma Reduction Trial (VDAART), we analyzed the gut microbiome and metabolome of wheeze frequency in children with asthma. METHODS Bacterial 16S ribosomal RNA microbiome and untargeted metabolomic profiling were performed on fecal samples collected from 3-year-old children with parent-reported physician-diagnosed asthma. We analyzed wheeze frequency by calculating the proportion of quarterly questionnaires administered between ages 3 and 5 years in which parents reported the child had wheezed (wheeze proportion). Taxa and metabolites associated with wheeze were analyzed by identifying log fold changes with respect to wheeze frequency and correlation/linear regression analyses, respectively. Microbe-metabolite and microbe-microbe correlation networks were compared between subjects with high and low wheeze proportion. RESULTS Specific taxa, including the genus Veillonella and histidine pathway metabolites, were enriched in subjects with high wheeze proportion. Among wheeze-associated taxa, Veillonella and Oscillospiraceae UCG-005, which was inversely associated with wheeze, were correlated with the greatest number of fecal metabolites. Microbial networks were similar between subjects with low versus high wheeze frequency. CONCLUSION Gut microbiome features are associated with wheeze frequency in children with asthma, suggesting an impact of the gut microbiome on morbidity in childhood asthma.
Collapse
Affiliation(s)
- Kathleen Lee-Sarwar
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass; Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Sandra Dedrick
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass; Department of Biology, Boston College, Chestnut Hill, Mass
| | - Babak Momeni
- Department of Biology, Boston College, Chestnut Hill, Mass
| | - Rachel S Kelly
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Robert S Zeiger
- Departments of Clinical Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, Calif
| | - George T O'Connor
- Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Mass
| | - Megan T Sandel
- Department of Pediatrics, Boston Medical Center, Boston, Mass
| | - Leonard B Bacharier
- Department of Pediatric Allergy, Immunology, and Pulmonary, Vanderbilt Children's Hospital, Vanderbilt University Medical Center, Nashville, Tenn
| | - Avraham Beigelman
- Division of Pediatric Allergy, Immunology and Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine, and St Louis Children's Hospital, St Louis, Mo; Kipper Institute of Allergy and Immunology, Schneider Children's Medical Center of Israel, Tel Aviv University, Tel Aviv, Israel
| | - Nancy Laranjo
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Diane R Gold
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass; Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Mass
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Augusto A Litonjua
- Division of Pediatric Pulmonary Medicine, Golisano Children's Hospital at Strong, University of Rochester Medical Center, Rochester, NY
| | - Yang-Yu Liu
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass.
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass.
| |
Collapse
|
192
|
Modoux M, Rolhion N, Lefevre JH, Oeuvray C, Nádvorník P, Illes P, Emond P, Parc Y, Mani S, Dvorak Z, Sokol H. Butyrate acts through HDAC inhibition to enhance aryl hydrocarbon receptor activation by gut microbiota-derived ligands. Gut Microbes 2022; 14:2105637. [PMID: 35895845 PMCID: PMC9336500 DOI: 10.1080/19490976.2022.2105637] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Aryl hydrocarbon receptor (AhR) is a critical player in the crosstalk between the gut microbiota and its host. However, factors regulating AhR within the gut, which is a complex metabolomic environment, are poorly understood. This study investigates the effect of a combination of metabolites on the activation mechanism of AhR. AhR activity was evaluated using both a luciferase reporter system and mRNA levels of AhR target genes on human cell lines and human colonic explants. AhR activation was studied by radioligand-binding assay, nuclear translocation of AhR by immuofluorescence and protein co-immunoprecipitation of AhR with ARNT. Indirect activation of AhR was evaluated using several tests and inhibitors. The promoter of the target gene CYP1A1 was studied both by chromatin immunoprecipitation and by using an histone deacetylase HDAC inhibitor (iHDAC). Short-chain fatty acids, and butyrate in particular, enhance AhR activity mediated by endogenous tryptophan metabolites without binding to the receptor. This effect was confirmed in human intestinal explants and did not rely on activation of receptors targeted by SCFAs, inhibition of AhR degradation or clearance of its ligands. Butyrate acted directly on AhR target gene promoter to reshape chromatin through iHDAC activity. Our findings revealed that butyrate is not an AhR ligand but acts as iHDAC leading to an increase recruitment of AhR to the target gene promoter in the presence of tryptophan-derived AhR agonists. These data contribute to a novel understanding of the complex regulation of AhR activation by gut microbiota-derived metabolites.
Collapse
Affiliation(s)
- Morgane Modoux
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology Department, Paris, France,Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Nathalie Rolhion
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology Department, Paris, France,Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Jeremie H. Lefevre
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France,Sorbonne Université, Department of Digestive Surgery, AP-HP, Hôpital Saint Antoine, Paris, France
| | - Cyriane Oeuvray
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology Department, Paris, France,Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Petr Nádvorník
- Departments of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic
| | - Peter Illes
- Departments of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic
| | - Patrick Emond
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, Centre-Val de Loire, France
| | - Yann Parc
- Sorbonne Université, Department of Digestive Surgery, AP-HP, Hôpital Saint Antoine, Paris, France
| | - Sridhar Mani
- Departments of Molecular Pharmacology, Genetics and Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Zdenek Dvorak
- Departments of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic
| | - Harry Sokol
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology Department, Paris, France,Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France,INRAe, UMR1319 Micalis & AgroParisTech, Jouy en Josas, France,CONTACT Harry Sokol Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology Department, ParisF-75012, France
| |
Collapse
|
193
|
Pant A, Maiti TK, Mahajan D, Das B. Human Gut Microbiota and Drug Metabolism. MICROBIAL ECOLOGY 2022:1-15. [PMID: 35869999 PMCID: PMC9308113 DOI: 10.1007/s00248-022-02081-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 07/18/2022] [Indexed: 05/31/2023]
Abstract
The efficacy of drugs widely varies in individuals, and the gut microbiota plays an important role in this variability. The commensal microbiota living in the human gut encodes several enzymes that chemically modify systemic and orally administered drugs, and such modifications can lead to activation, inactivation, toxification, altered stability, poor bioavailability, and rapid excretion. Our knowledge of the role of the human gut microbiome in therapeutic outcomes continues to evolve. Recent studies suggest the existence of complex interactions between microbial functions and therapeutic drugs across the human body. Therapeutic drugs or xenobiotics can influence the composition of the gut microbiome and the microbial encoded functions. Both these deviations can alter the chemical transformations of the drugs and hence treatment outcomes. In this review, we provide an overview of (i) the genetic ecology of microbially encoded functions linked with xenobiotic degradation; (ii) the effect of drugs on the composition and function of the gut microbiome; and (iii) the importance of the gut microbiota in drug metabolism.
Collapse
Affiliation(s)
- Archana Pant
- Molecular Genetics Lab, National Institute of Immunology, New Delhi, Delhi-110067, India
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad-121001, India
- Molecular Genetics Laboratory, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, PO box, Gurgaon Expressway, #04 Faridabad-121001, Haryana, India
| | - Tushar K Maiti
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad-121001, India
| | - Dinesh Mahajan
- Chemistry and Pharmacology Lab, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad, India
| | - Bhabatosh Das
- Molecular Genetics Laboratory, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, PO box, Gurgaon Expressway, #04 Faridabad-121001, Haryana, India.
| |
Collapse
|
194
|
Intestinal homeostasis in autoimmune liver diseases. Chin Med J (Engl) 2022; 135:1642-1652. [PMID: 36193976 PMCID: PMC9509077 DOI: 10.1097/cm9.0000000000002291] [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] [Indexed: 02/04/2023] Open
Abstract
ABSTRACT Intestinal homeostasis depends on complex interactions between the gut microbiota and host immune system. Emerging evidence indicates that the intestinal microbiota is a key player in autoimmune liver disease (AILD). Autoimmune hepatitis, primary biliary cholangitis, primary sclerosing cholangitis, and IgG4-related sclerosing cholangitis have been linked to gut dysbiosis. Diverse mechanisms contribute to disturbances in intestinal homeostasis in AILD. Bacterial translocation and molecular mimicry can lead to hepatic inflammation and immune activation. Additionally, the gut and liver are continuously exposed to microbial metabolic products, mediating variable effects on liver immune pathologies. Importantly, microbiota-specific or associated immune responses, either hepatic or systemic, are abnormal in AILD. Comprehensive knowledge about host-microbiota interactions, included but not limited to this review, facilitates novel clinical practice from a microbiome-based perspective. However, many challenges and controversies remain in the microbiota field of AILD, and there is an urgent need for future investigations.
Collapse
|
195
|
Qi B, Ren D, Li T, Niu P, Zhang X, Yang X, Xiao J. Fu Brick Tea Manages HFD/STZ-Induced Type 2 Diabetes by Regulating the Gut Microbiota and Activating the IRS1/PI3K/Akt Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:8274-8287. [PMID: 35767631 DOI: 10.1021/acs.jafc.2c02400] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The antidiabetic effects of Fu brick tea aqueous extract (FTE) and its underlying molecular mechanism in type 2 diabetes mellitus (T2DM) mice were investigated. FTE treatment significantly relieved dyslipidemia, insulin resistance (IR), and hepatic oxidative stress caused by T2DM. FTE also ameliorated the T2DM-induced gut dysbiosis by decreasing the Firmicutes/Bacteroidota (F/B) ratio at the phylum level and promoting the proliferation of Bifidobacterium, Parabacteroides, and Roseburia at the genus level. Besides, FTE significantly improved colonic short-chain fatty acid levels of T2DM mice. Furthermore, the antidiabetic effects of FTE were proved to be mediated by the IRS1/PI3K/Akt and AMPK-mediated gluconeogenesis signaling pathways. Metabolomics analysis illustrated that FTE recovered the levels of 28 metabolites associated with T2DM to the levels of normal mice. Taken together, these findings suggest that FTE can alleviate T2DM by reshaping the gut microbiota, activating the IRS1/PI3K/Akt pathway, and regulating intestinal metabolites.
Collapse
Affiliation(s)
- Bangran Qi
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Daoyuan Ren
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Ting Li
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Pengfei Niu
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Xiangnan Zhang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Xingbin Yang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Jianbo Xiao
- Nutrition and Bromatology Group, Department of Analytical and Food Chemistry, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain
| |
Collapse
|
196
|
Xie Y, Xie F, Zhou X, Zhang L, Yang B, Huang J, Wang F, Yan H, Zeng L, Zhang L, Zhou F. Microbiota in Tumors: From Understanding to Application. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200470. [PMID: 35603968 PMCID: PMC9313476 DOI: 10.1002/advs.202200470] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 04/30/2022] [Indexed: 05/09/2023]
Abstract
Microbes with complex functions have been found to be a potential component in tumor microenvironments. Due to their low biomass and other obstacles, intratumor microbiota is poorly understood. Mucosal sites and normal adjacent tissues are important sources of intratumor microbiota, while hematogenous spread also leads to the invasion of microbes. Intratumor microbiota affects the progression of tumors through several mechanisms, such as DNA damage, activation of oncogenic pathways, induction of immunosuppression, and metabolization of drugs. Notably, in different types of tumors, the composition and abundance of intratumor microbiota are highly heterogeneous and may play different roles in the progression of tumors. Because of the concern in this field, several techniques such as omics and immunological methods have been used to study intratumor microbiota. Here, recent progress in this field is reviewed, including the potential sources of intratumor microbiota, their functions and related mechanisms, and their heterogeneity. Techniques that can be used to study intratumor microbiota are also discussed. Moreover, research is summarized into the development of strategies that can be used in antitumor treatment and prospects for possible future research in this field.
Collapse
Affiliation(s)
- Yifan Xie
- School of MedicineZhejiang University City CollegeSuzhou215123P. R. China
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Feng Xie
- Institutes of Biology and Medical ScienceSoochow UniversitySuzhou215123P. R. China
| | - Xiaoxue Zhou
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Lei Zhang
- Department of Orthopaedic Surgery WenzhouThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhou32500P. R. China
| | - Bing Yang
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Jun Huang
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Fangwei Wang
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Haiyan Yan
- School of MedicineZhejiang University City CollegeSuzhou215123P. R. China
| | - Linghui Zeng
- School of MedicineZhejiang University City CollegeSuzhou215123P. R. China
| | - Long Zhang
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Fangfang Zhou
- Institutes of Biology and Medical ScienceSoochow UniversitySuzhou215123P. R. China
| |
Collapse
|
197
|
Cope H, Willis CR, MacKay MJ, Rutter LA, Toh LS, Williams PM, Herranz R, Borg J, Bezdan D, Giacomello S, Muratani M, Mason CE, Etheridge T, Szewczyk NJ. Routine omics collection is a golden opportunity for European human research in space and analog environments. PATTERNS 2022; 3:100550. [PMID: 36277820 PMCID: PMC9583032 DOI: 10.1016/j.patter.2022.100550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
198
|
Wang W, Xu Y, Wang X, Chu Y, Zhang H, Zhou L, Zhu H, Li J, Kuai R, Zhou F, Yang D, Peng H. Swimming Impedes Intestinal Microbiota and Lipid Metabolites of Tumorigenesis in Colitis-Associated Cancer. Front Oncol 2022; 12:929092. [PMID: 35847876 PMCID: PMC9285133 DOI: 10.3389/fonc.2022.929092] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/03/2022] [Indexed: 12/09/2022] Open
Abstract
Background Accumulating data support that regular physical activity potentially inhibits chronic colitis, a risk factor for colitis-associated cancer (CAC). However, possible effects of physical activity on CAC and the underlying mechanisms remain poorly understood. Methods A pretreatment of swimming on azoxymethane/dextran sodium sulfate (AOM/DSS)-induced CAC mice was implemented to determine its protective effect. Inflammation and tumorigenesis were assessed using colorectums from C57BL/6 mice. In order to determine how swimming alters colonic lipid metabolism and gene expression, a comparative analysis was conducted. Meanwhile, alterations in intestinal microbiota and short-chain fatty acids (SCFAs) were detected and analyzed. Finally, an integration analysis of colonic lipid metabolism with gene expression and intestinal microbiota was performed respectively. Result Swimming pretreatment relieved bowel inflammation and minimized tumor formation. We demonstrated that prostaglandin E2 (PGE2)/PGE2 receptor 2 subtype (EP2) signaling as a potential regulatory target for swimming induces colonic lipid metabolites. Swimming-induced genera, Erysipelatoclostridium, Parabacteroides, Bacteroides, and Rikenellaceae_RC9_gut_group, induced intestinal SCFAs and affected the function of colonic lipid metabolites enriched in glycerophospholipid metabolism and choline metabolism in cancer. Conclusion According to our experiments, swimming pretreatment can protect mice from CAC by intervention in the possible link between colonic lipid metabolites and PGE2/EP2 signaling. Further, swimming-induced genera and probiotics promoted glycerophospholipid metabolism and choline metabolism in cancer, the major constituents of colonic lipid metabolites, and increased SCFAs, which were also important mechanisms for the anti-inflammatory and anti-tumorigenic effects of swimming.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Daming Yang
- *Correspondence: Haixia Peng, ; Daming Yang,
| | - Haixia Peng
- *Correspondence: Haixia Peng, ; Daming Yang,
| |
Collapse
|
199
|
Lindell AE, Zimmermann-Kogadeeva M, Patil KR. Multimodal interactions of drugs, natural compounds and pollutants with the gut microbiota. Nat Rev Microbiol 2022; 20:431-443. [PMID: 35102308 PMCID: PMC7615390 DOI: 10.1038/s41579-022-00681-5] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2022] [Indexed: 02/08/2023]
Abstract
The gut microbiota contributes to diverse aspects of host physiology, ranging from immunomodulation to drug metabolism. Changes in the gut microbiota composition are associated with various diseases as well as with the response to medications. It is therefore important to understand how different lifestyle and environmental factors shape gut microbiota composition. Beyond the commonly considered factor of diet, small-molecule drugs have recently been identified as major effectors of the microbiota composition. Other xenobiotics, such as environmental or chemical pollutants, can also impact gut bacterial communities. Here, we review the mechanisms of interactions between gut bacteria and antibiotics, host-targeted drugs, natural food compounds, food additives and environmental pollutants. While xenobiotics can impact bacterial growth and metabolism, bacteria in turn can bioaccumulate or chemically modify these compounds. These reciprocal interactions can manifest in complex xenobiotic-microbiota-host relationships. Our Review highlights the need to study mechanisms underlying interactions with pollutants and food additives towards deciphering the dynamics and evolution of the gut microbiota.
Collapse
Affiliation(s)
- Anna E Lindell
- The Medical Research Council Toxicology Unit, University of Cambridge, Cambridge, UK
| | | | - Kiran R Patil
- The Medical Research Council Toxicology Unit, University of Cambridge, Cambridge, UK.
| |
Collapse
|
200
|
Zhu M, Liu X, Ye Y, Yan X, Cheng Y, Zhao L, Chen F, Ling Z. Gut Microbiota: A Novel Therapeutic Target for Parkinson’s Disease. Front Immunol 2022; 13:937555. [PMID: 35812394 PMCID: PMC9263276 DOI: 10.3389/fimmu.2022.937555] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 05/26/2022] [Indexed: 12/16/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease characterized by motor dysfunction. Growing evidence has demonstrated that gut dysbiosis is involved in the occurrence, development and progression of PD. Numerous clinical trials have identified the characteristics of the changed gut microbiota profiles, and preclinical studies in PD animal models have indicated that gut dysbiosis can influence the progression and onset of PD via increasing intestinal permeability, aggravating neuroinflammation, aggregating abnormal levels of α-synuclein fibrils, increasing oxidative stress, and decreasing neurotransmitter production. The gut microbiota can be considered promising diagnostic and therapeutic targets for PD, which can be regulated by probiotics, psychobiotics, prebiotics, synbiotics, postbiotics, fecal microbiota transplantation, diet modifications, and Chinese medicine. This review summarizes the recent studies in PD-associated gut microbiota profiles and functions, the potential roles, and mechanisms of gut microbiota in PD, and gut microbiota-targeted interventions for PD. Deciphering the underlying roles and mechanisms of the PD-associated gut microbiota will help interpret the pathogenesis of PD from new perspectives and elucidate novel therapeutic strategies for PD.
Collapse
Affiliation(s)
- Manlian Zhu
- Department of Geriatrics, Lishui Second People’s Hospital, Lishui, China
| | - Xia Liu
- Department of Intensive Care Unit, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yiru Ye
- Department of Respiratory Medicine, Lishui Central Hospital, Lishui, China
| | - Xiumei Yan
- Department of Laboratory Medicine, Lishui Second People’s Hospital, Lishui, China
| | - Yiwen Cheng
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Longyou Zhao
- Department of Laboratory Medicine, Lishui Second People’s Hospital, Lishui, China
| | - Feng Chen
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Zongxin Ling, ; ; Feng Chen,
| | - Zongxin Ling
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Zongxin Ling, ; ; Feng Chen,
| |
Collapse
|