1
|
Zamel D, Pan X, Ye ZL. Ethanol-mediated Anaerobic Digestion: Functional Bacteria and Metabolic Pathways. CHEMOSPHERE 2024; 367:143560. [PMID: 39426748 DOI: 10.1016/j.chemosphere.2024.143560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/04/2024] [Accepted: 10/16/2024] [Indexed: 10/21/2024]
Abstract
Ethanol-mediated Anaerobic digestion (Ethanol-AD) is a biological process that converts organic waste into biogas, predominantly composed of methane (CH₄), hydrogen (H₂), and carbon dioxide (CO₂), through the breakdown of complex organic materials while ethanol is an intermediate metabolite. Ethanol improves the digestion of complex organic waste by serving as an electron precursor for interspecies electron transfer, leading to enhanced biogas production. It further serves as a substrate for acetogens or syntrophic bacteria, while mean its oxidation leads to acetate formation, which methanogens can then consume to generate methane. Methanogenesis, the final and crucial step in the anaerobic digestion in which methanogens produce methane through various metabolic routes, most notably via the hydrogenotrophic and syntrophic pathways. In hydrogenotrophic methanogenesis, methanogens consume hydrogen as an electron precursor and carbon dioxide as an electron acceptor, leading to methane generation. Alternatively, syntrophic methanogenesis, which is increasingly recognized for its efficiency, is dominated by DIET between syntrophic partners, bypassing the need for hydrogen as a mediator. This mode of electron transfer enhances the metabolic cooperation between microbes, facilitating a more efficient methanogenesis process. As research continues to explore the mechanisms underlying DIET and the role of (semi) conductive materials, there is potential for optimizing AD systems for renewable energy production by advancing the methanogenesis process, and enhancing biogas quality. The novelty of this review lies in its dual exploration of direct and indirect interspecies electron transfer (DIET and IIET) within ethanol-mediated anaerobic digestion. While DIET in ethanol-driven systems has been previously discussed, this review is the first to comprehensively examine the interplay between both direct and indirect electron transfer mechanisms, offering new insights into optimizing microbial interactions and improving methane production efficiency.
Collapse
Affiliation(s)
- Doaa Zamel
- Key Laboratory of Urban Pollutant Conversion, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, 361021, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaofang Pan
- Key Laboratory of Urban Pollutant Conversion, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, 361021, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Zhi-Long Ye
- Key Laboratory of Urban Pollutant Conversion, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, 361021, China.
| |
Collapse
|
2
|
Gao W, Chen X, Wu S, Jin L, Chen X, Mao G, Wan X, Xing W. Monascus red pigments alleviate high-fat and high-sugar diet-induced NAFLD in mice by modulating the gut microbiota and metabolites. Food Sci Nutr 2024; 12:5762-5775. [PMID: 39139961 PMCID: PMC11317676 DOI: 10.1002/fsn3.4208] [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: 12/18/2023] [Revised: 03/21/2024] [Accepted: 04/27/2024] [Indexed: 08/15/2024] Open
Abstract
Monascus red pigments (MRP) may have benefits against NAFLD with an unclear mechanism. This study aimed to explore the protective effect of MRP supplementation against NAFLD through regulation of gut microbiota and metabolites. The C57BL/6 mice animals were randomly allocated into the normal diet (NC), HFHS diet-induced NAFLD model, and MRP intervention group fed with HFHS diet. Serum lipid profiles and liver function parameters were measured. Liver and colon histopathology analysis was conducted to determine the injury in the liver and colon. 16S rRNA gene sequencing was employed to analyze gut microbial composition from fecal samples. Untargeted metabonomics was performed to analyze changes in metabolites in serum and fecal samples. MRP supplementation significantly improved the HFHS-induced alterations in body weight, lipid profiles, and liver function (p < .01). MRP supplementation decreased the abundance of Akkermansia, Candidatus saccharimonas, Dubosiella, and Oscillibacter, while increasing Lactobacillus, Lachnospiraceae NK4A136 group, and Rikenella in mice fed the HFHS diet. Furthermore, MRP supplementation improved the serum and fecal metabolic profiles induced by the HFHS diet, primarily involving the arachidonic acid metabolism, unsaturated fatty acid biosynthesis, and adipocyte lipolysis pathways. Liver function and lipid profiles were closely associated with the abundance of Lactobacillus, Streptococcus, Oscillibacter, Akkemansia, and Desulfovibrio (p < .01). These findings revealed that MRP supplementation may help restore gut microbiota composition and balance its metabolites, thereby improving NAFLD. This study presents a novel outlook on the potential benefits of MRP supplementation in ameliorating NAFLD and supports the application of MRP as a new functional food.
Collapse
Affiliation(s)
- Wenyan Gao
- School of PharmacyHangzhou Medical CollegeHangzhouChina
| | - Xinghao Chen
- School of PharmacyHangzhou Medical CollegeHangzhouChina
| | - Shaokang Wu
- Department of PharmacyQingdao Sixth People's HospitalQingdaoChina
| | - Lu Jin
- School of PharmacyHangzhou Medical CollegeHangzhouChina
| | - Xu Chen
- School of PharmacyHangzhou Medical CollegeHangzhouChina
| | - Genxiang Mao
- Zhejiang Provincial Key Lab of GeriatricsZhejiang HospitalHangzhouChina
| | - Xiaoqing Wan
- Zhejiang Provincial Key Lab of GeriatricsZhejiang HospitalHangzhouChina
| | - Wenmin Xing
- Zhejiang Provincial Key Lab of GeriatricsZhejiang HospitalHangzhouChina
| |
Collapse
|
3
|
Meijnikman AS, Nieuwdorp M, Schnabl B. Endogenous ethanol production in health and disease. Nat Rev Gastroenterol Hepatol 2024; 21:556-571. [PMID: 38831008 DOI: 10.1038/s41575-024-00937-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/23/2024] [Indexed: 06/05/2024]
Abstract
The gut microbiome exerts metabolic actions on distal tissues and organs outside the intestine, partly through microbial metabolites that diffuse into the circulation. The disruption of gut homeostasis results in changes to microbial metabolites, and more than half of the variance in the plasma metabolome can be explained by the gut microbiome. Ethanol is a major microbial metabolite that is produced in the intestine of nearly all individuals; however, elevated ethanol production is associated with pathological conditions such as metabolic dysfunction-associated steatotic liver disease and auto-brewery syndrome, in which the liver's capacity to metabolize ethanol is surpassed. In this Review, we describe the mechanisms underlying excessive ethanol production in the gut and the role of ethanol catabolism in mediating pathogenic effects of ethanol on the liver and host metabolism. We conclude by discussing approaches to target excessive ethanol production by gut bacteria.
Collapse
Affiliation(s)
| | - Max Nieuwdorp
- Department of Internal Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, Netherlands
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, Netherlands
- Diabeter Centrum Amsterdam, Amsterdam, Netherlands
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.
- Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA.
- Center for Innovative Phage Applications and Therapeutics, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
4
|
Rajendran S, Khomenko I, Silcock P, Betta E, Biasioli F, Bremer P. Impact of Different Carbon Sources on Volatile Organic Compounds (VOCs) Produced during Fermentation by Levilactobacillus brevis WLP672 Measured Using Proton Transfer Reaction Time-of-Flight Mass Spectrometry (PTR-ToF-MS). Molecules 2024; 29:3275. [PMID: 39064855 PMCID: PMC11279293 DOI: 10.3390/molecules29143275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Bacterial fermentation is considered to be a cost-effective means of generating desired flavour compounds from plant-based substrates. However, the wide range of substrates present in plants makes it challenging to understand how individual components impact on flavour volatile organic compound (VOC) production. To simplify this, a defined medium can be used to better understand VOCs production with regard to individual compounds. In the current study, the VOCs produced by the lactic acid bacterium, Levilactobacillus brevis WLP672, growing in a defined medium containing different carbon sources (either glucose (DM), fructose (DMFr) or citrate (DMCi)) under a range of fermentation conditions (time: 0, 7, and 14 days; and temperature: 25 and 35 °C) were assessed using proton transfer reaction time-of-flight mass spectrometry (PTR-ToF-MS). Among the detected mass peaks (m/z), after 7 days of fermentation, the concentrations of m/z 45.033 (t.i. acetaldehyde), m/z 49.011 (t.i. methanethiol), and m/z 89.060 (t.i. ethyl acetate) were significantly (p < 0.05) higher in DM at 35 °C than all other treatments at either temperature. The knowledge obtained will help to produce desirable LAB fermentation flavour VOCs or VOC mixtures that could be used in developing plant-based analogues with acceptable sensory properties.
Collapse
Affiliation(s)
- Sarathadevi Rajendran
- Department of Food Science, University of Otago, P.O. Box 56, Dunedin 9054, New Zealand; (S.R.); (P.B.)
- Sensory Quality Unit, Research and Innovation Centre, Fondazione Edmund Mach, 38098 Trento, Italy; (I.K.); (E.B.); (F.B.)
- Department of Agricultural Chemistry, Faculty of Agriculture, University of Jaffna, Kilinochchi 44000, Sri Lanka
| | - Iuliia Khomenko
- Sensory Quality Unit, Research and Innovation Centre, Fondazione Edmund Mach, 38098 Trento, Italy; (I.K.); (E.B.); (F.B.)
- ONFoods-Research and Innovation Network on Food and Nutrition Sustainability, Safety and Security-Working ON Foods, 43121 Parma, Italy
| | - Patrick Silcock
- Department of Food Science, University of Otago, P.O. Box 56, Dunedin 9054, New Zealand; (S.R.); (P.B.)
| | - Emanuela Betta
- Sensory Quality Unit, Research and Innovation Centre, Fondazione Edmund Mach, 38098 Trento, Italy; (I.K.); (E.B.); (F.B.)
- ONFoods-Research and Innovation Network on Food and Nutrition Sustainability, Safety and Security-Working ON Foods, 43121 Parma, Italy
| | - Franco Biasioli
- Sensory Quality Unit, Research and Innovation Centre, Fondazione Edmund Mach, 38098 Trento, Italy; (I.K.); (E.B.); (F.B.)
- ONFoods-Research and Innovation Network on Food and Nutrition Sustainability, Safety and Security-Working ON Foods, 43121 Parma, Italy
| | - Phil Bremer
- Department of Food Science, University of Otago, P.O. Box 56, Dunedin 9054, New Zealand; (S.R.); (P.B.)
| |
Collapse
|
5
|
Martin-Grau M, Monleón D. The Role of Microbiota-Related Co-Metabolites in MASLD Progression: A Narrative Review. Curr Issues Mol Biol 2024; 46:6377-6389. [PMID: 39057023 PMCID: PMC11276081 DOI: 10.3390/cimb46070381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/17/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) represents a growing health concern due to its increasing prevalence worldwide. Metabolic homeostasis encompasses the stable internal conditions vital for efficient metabolism. This equilibrium extends to the intestinal microbiota, whose metabolic activities profoundly influence overall metabolic balance and organ health. The metabolites derived from the gut microbiota metabolism can be defined as microbiota-related co-metabolites. They serve as mediators between the gut microbiota and the host, influencing various physiological processes. The recent redefinition of the term MASLD has highlighted the metabolic dysfunction that characterize the disease. Metabolic dysfunction encompasses a spectrum of abnormalities, including impaired glucose regulation, dyslipidemia, mitochondrial dysfunction, inflammation, and accumulation of toxic byproducts. In addition, MASLD progression has been linked to dysregulation in the gut microbiota and associated co-metabolites. Short-chain fatty acids (SCFAs), hippurate, indole derivatives, branched-chain amino acids (BCAAs), and bile acids (BAs) are among the key co-metabolites implicated in MASLD progression. In this review, we will unravel the relationship between the microbiota-related metabolites which have been associated with MASLD and that could play an important role for developing effective therapeutic interventions for MASLD and related metabolic disorders.
Collapse
Affiliation(s)
- Maria Martin-Grau
- Department of Pathology, University of Valencia, 46010 Valencia, Spain
- University Clinical Hospital of Valencia Research Foundation (INCLIVA), 46010 Valencia, Spain
| | - Daniel Monleón
- Department of Pathology, University of Valencia, 46010 Valencia, Spain
- University Clinical Hospital of Valencia Research Foundation (INCLIVA), 46010 Valencia, Spain
| |
Collapse
|
6
|
Mbaye B, Wasfy RM, Alou MT, Borentain P, Gerolami R, Dufour JC, Million M. A catalog of ethanol-producing microbes in humans. Future Microbiol 2024; 19:697-714. [PMID: 38700288 PMCID: PMC11259083 DOI: 10.2217/fmb-2023-0250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 02/13/2024] [Indexed: 05/05/2024] Open
Abstract
Aim: Endogenous ethanol production emerges as a mechanism of nonalcoholic steatohepatitis, obesity, diabetes and auto-brewery syndrome. Methods: To identify ethanol-producing microbes in humans, we used the NCBI taxonomy browser and the PubMed database with an automatic query and manual verification. Results: 85 ethanol-producing microbes in human were identified. Saccharomyces cerevisiae, Candida and Pichia were the most represented fungi. Enterobacteriaceae was the most represented bacterial family with mainly Escherichia coli and Klebsiella pneumoniae. Species of the Lachnospiraceae and Clostridiaceae family, of the Lactobacillales order and of the Bifidobacterium genus were also identified. Conclusion: This catalog will help the study of ethanol-producing microbes in human in the pathophysiology, diagnosis, prevention and management of human diseases associated with endogenous ethanol production.
Collapse
Affiliation(s)
- Babacar Mbaye
- Aix Marseille Université, AP-HM, MEPHI, Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Reham Magdy Wasfy
- Aix Marseille Université, AP-HM, MEPHI, Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Maryam Tidjani Alou
- Aix Marseille Université, AP-HM, MEPHI, Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| | - Patrick Borentain
- Department of Hepatology, La Timone University Hospital, AP-HM, Marseille, France
| | - Rene Gerolami
- Aix Marseille Université, AP-HM, MEPHI, Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
- Department of Hepatology, La Timone University Hospital, AP-HM, Marseille, France
| | - Jean-Charles Dufour
- INSERM, IRD, SESSTIM, Sciences Economiques & Sociales de la Santé & Traitement de l'Information Médicale, ISSPAM, Aix Marseille University, 13385 Marseille, France
- APHM, Hôpital de la Timone, Service Biostatistique et Technologies de l'Information et de la Communication, 13385 Marseille, France
| | - Matthieu Million
- Aix Marseille Université, AP-HM, MEPHI, Institut Hospitalo-Universitaire Méditerranée Infection, Marseille, France
| |
Collapse
|
7
|
Kouroumalis E, Tsomidis I, Voumvouraki A. Viral Liver Disease and Intestinal Gut–Liver Axis. GASTROINTESTINAL DISORDERS 2024; 6:64-93. [DOI: 10.3390/gidisord6010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
The intestinal microbiota is closely related to liver diseases via the intestinal barrier and bile secretion to the gut. Impairment of the barrier can translocate microbes or their components to the liver where they can contribute to liver damage and fibrosis. The components of the barrier are discussed in this review along with the other elements of the so-called gut–liver axis. This bidirectional relation has been widely studied in alcoholic and non-alcoholic liver disease. However, the involvement of microbiota in the pathogenesis and treatment of viral liver diseases have not been extensively studied, and controversial data have been published. Therefore, we reviewed data regarding the integrity and function of the intestinal barrier and the changes of the intestinal microbioma that contribute to progression of Hepatitis B (HBV) and Hepatitis C (HCV) infection. Their consequences, such as cirrhosis and hepatic encephalopathy, were also discussed in connection with therapeutic interventions such as the effects of antiviral eradication and the use of probiotics that may influence the outcome of liver disease. Profound alterations of the microbioma with significant reduction in microbial diversity and changes in the abundance of both beneficial and pathogenic bacteria were found.
Collapse
Affiliation(s)
- Elias Kouroumalis
- Department of Gastroenterology, Medical School, University of Crete, 71500 Heraklion, Greece
| | - Ioannis Tsomidis
- Department of Gastroenterology, Medical School, University of Crete, 71500 Heraklion, Greece
| | - Argyro Voumvouraki
- 1st Department of Internal Medicine, AHEPA University Hospital, 54621 Thessaloniki, Greece
| |
Collapse
|
8
|
Seki D, Kirkegaard R, Osvatic J, Hausmann B, Séneca J, Pjevac P, Berger A, J Hall L, Wisgrill L, Berry D. Gut microbiota genome features associated with brain injury in extremely premature infants. Gut Microbes 2024; 16:2410479. [PMID: 39679616 DOI: 10.1080/19490976.2024.2410479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/19/2024] [Accepted: 09/24/2024] [Indexed: 12/17/2024] Open
Abstract
Severe brain damage is common among premature infants, and the gut microbiota has been implicated in its pathology. Although the order of colonizing bacteria is well described, the mechanisms underlying aberrant assembly of the gut microbiota remain elusive. Here, we employed long-read nanopore sequencing to assess abundances of microbial species and their functional genomic potential in stool samples from a cohort of 30 extremely premature infants. We identify several key microbial traits significantly associated with severe brain damage, such as the genomic potential for nitrate respiration and iron scavenging. Members of the Enterobacteriaceae were prevalent across the cohort and displayed a versatile metabolic potential, including pathogenic and nonpathogenic traits. Predominance of Enterobacter hormaechei and Klebsiella pneumoniae were associated with an overall loss of genomic functional redundancy as well as poor neurophysiological outcome. These findings reveal microbial traits that may be involved in exacerbating brain injury in extremely premature infants and provide suitable targets for therapeutic interventions.
Collapse
Affiliation(s)
- David Seki
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria
- Intestinal Microbiome, School of Life Sciences, ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Rasmus Kirkegaard
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria
| | - Jay Osvatic
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Bela Hausmann
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Joana Séneca
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria
| | - Petra Pjevac
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria
| | - Angelika Berger
- Department of Pediatrics and Adolescent Medicine, Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Comprehensive Center for Pediatrics. Medical University of Vienna, Vienna, Austria
| | - Lindsay J Hall
- Intestinal Microbiome, School of Life Sciences, ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
- Food Microbiomes and Health, Quadram Institute Bioscience, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
- Microbes, Infections, and Microbiomes, Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Lukas Wisgrill
- Department of Pediatrics and Adolescent Medicine, Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Comprehensive Center for Pediatrics. Medical University of Vienna, Vienna, Austria
| | - David Berry
- Centre for Microbiology and Environmental Systems Science, Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, University of Vienna, Vienna, Austria
- Intestinal Microbiome, School of Life Sciences, ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| |
Collapse
|
9
|
Mbaye B, Magdy Wasfy R, Borentain P, Tidjani Alou M, Mottola G, Bossi V, Caputo A, Gerolami R, Million M. Increased fecal ethanol and enriched ethanol-producing gut bacteria Limosilactobacillus fermentum, Enterocloster bolteae, Mediterraneibacter gnavus and Streptococcus mutans in nonalcoholic steatohepatitis. Front Cell Infect Microbiol 2023; 13:1279354. [PMID: 38035329 PMCID: PMC10687429 DOI: 10.3389/fcimb.2023.1279354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
Background Non-alcoholic steatohepatitis (NASH) has become a major public health issue as one of the leading causes of liver disease and transplantation worldwide. The instrumental role of the gut microbiota is emerging but still under investigation. Endogenous ethanol (EtOH) production by gut bacteria and yeasts is an emerging putative mechanism. Microbial metagenomics and culture studies targeting enterobacteria or yeasts have been reported, but no culturomics studies have been conducted so far. Aim To assess fecal EtOH and other biochemical parameters, characterize NASH-associated dysbiosis and identify EtOH-producing gut microbes associated with the disease, fecal samples from 41 NASH patients and 24 controls were analyzed. High-performance liquid chromatography (HPLC) was used for EtOH, glucose, total proteins, triglyceride and total cholesterol. Viable bacteria were assessed with microbial culturomics. Microbial genetic material was assessed using 16S metagenomics targeting the hypervariable V3V4 region. Results Fecal EtOH and glucose was elevated in the stools of NASH patients (p < 0.05) but not triglyceride, total cholesterol or proteins. In culturomics, EtOH-producing Enterocloster bolteae and Limosilactobacillus fermentum were enriched in NASH. V3V4 16S rRNA amplicon sequencing confirmed the enrichment in EtOH-producing bacteria including L. fermentum, Mediterraneibacter gnavus and Streptococcus mutans, species previously associated with NASH and other dysbiosis-associated diseases. Strikingly, E. bolteae was identified only by culturomics. The well-known Lacticaseibacillus casei was identified in controls but never isolated in patients with NASH (p < 0.05). Conclusion Elevated fecal EtOH and glucose is a feature of NASH. Several different EtOH-producing gut bacteria may play an instrumental role in the disease. Culturomics and metagenomics, two complementary methods, will be critical to identify EtOH-producing bacteria for future diagnostic markers and therapeutic targets for NASH. Suppression of EtOH-producing gut microbes and L. casei administration are options to be tested in NASH treatment.
Collapse
Affiliation(s)
- Babacar Mbaye
- Aix-Marseille Université, IRD, APHM, MEPHI, IHU Méditerranée Infection, Marseille, France
| | - Reham Magdy Wasfy
- Aix-Marseille Université, IRD, APHM, MEPHI, IHU Méditerranée Infection, Marseille, France
| | - Patrick Borentain
- Assistance Publique-Hôpitaux de Marseille (AP-HM), Hôpital de la Timone, Unité d’hépatologie, Marseille, France
| | - Maryam Tidjani Alou
- Aix-Marseille Université, IRD, APHM, MEPHI, IHU Méditerranée Infection, Marseille, France
| | - Giovanna Mottola
- Assistance Publique-Hôpitaux de Marseille (AP-HM), Hôpital de la Timone, Laboratoire de Biochimie, Marseille, France
- C2VN, INSERM 1263, INRAE 1260, Team 5, Aix-Marseille Université, Marseille, France
| | - Vincent Bossi
- Aix-Marseille Université, IRD, APHM, MEPHI, IHU Méditerranée Infection, Marseille, France
| | - Aurelia Caputo
- Aix-Marseille Université, IRD, APHM, MEPHI, IHU Méditerranée Infection, Marseille, France
| | - Rene Gerolami
- Aix-Marseille Université, IRD, APHM, MEPHI, IHU Méditerranée Infection, Marseille, France
- Assistance Publique-Hôpitaux de Marseille (AP-HM), Hôpital de la Timone, Unité d’hépatologie, Marseille, France
| | - Matthieu Million
- Aix-Marseille Université, IRD, APHM, MEPHI, IHU Méditerranée Infection, Marseille, France
| |
Collapse
|
10
|
Roques S, Deborde C, Skiba-Cassy S, Médale F, Dupont-Nivet M, Lefevre F, Bugeon J, Labbé L, Marchand Y, Moing A, Fauconneau B. New alternative ingredients and genetic selection are the next game changers in rainbow trout nutrition: a metabolomics appraisal. Sci Rep 2023; 13:19634. [PMID: 37949954 PMCID: PMC10638236 DOI: 10.1038/s41598-023-46809-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 11/05/2023] [Indexed: 11/12/2023] Open
Abstract
The formulation of sustainable fish feeds based on plant ingredients supplemented by alternative ingredients to plant (insect, micro-algae, yeast) and genetic selection of fish for plant-based diets were tested on rainbow trout in two separate experiments. Plant-based diets and corresponding diets supplemented with an ingredient mix: insect, micro-algae and yeast in Experiment A, and insect and yeast in Experiment B were compared to commercial-like diets. In experiment A, the mix-supplemented diet was successful in compensating the altered growth performance of fish fed their respective plant-based diet compared to those fed the commercial diet, by restoring feed conversion. In experiment B, the selected line demonstrated improved growth performances of fish fed mix-supplemented and plant-based diets compared to the non-selected line. Metabolomics demonstrated a plasma compositional stability in fish fed mix-supplemented and basal plant-based diets comprising an amino acid accumulation and a glucose depletion, compared to those fed commercial diets. The selected line fed mix-supplemented and commercial diets showed changes in inositol, ethanol and methanol compared to the non-selected line, suggesting an involvement of microbiota. Changes in plasma glycine-betaine content in fish fed the mix-supplemented diet suggest the ability of the selected line to adapt to alternative ingredients.
Collapse
Affiliation(s)
- Simon Roques
- INRAE, Univ. Pau & Pays Adour, E2S UPPA, Nutrition, Métabolisme et Aquaculture, UMR 1419, 64310, Saint Pée sur Nivelle, France
- Phileo by Lesaffre, 59700, Marcq-en-Barœul, France
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMR Herbivores, 63122, Saint-Genes-Champanelle, France
| | - Catherine Deborde
- Bordeaux Metabolome, MetaboHUB, Centre INRAE de Nouvelle-Aquitaine Bordeaux, 33140, Villenave d'Ornon, France
- Centre INRAE de Nouvelle-Aquitaine Bordeaux, INRAE, Univ. Bordeaux, Biologie du Fruit et Pathologie, UMR 1332, 33140, Villenave d'Ornon, France
- INRAE, Biopolymères Interactions Assemblages, UR1268, 44300, Nantes, France
- INRAE, BIBS Facility, Centre INRAE Pays de Loire - Nantes, 44000, Nantes, France
| | - Sandrine Skiba-Cassy
- INRAE, Univ. Pau & Pays Adour, E2S UPPA, Nutrition, Métabolisme et Aquaculture, UMR 1419, 64310, Saint Pée sur Nivelle, France
| | - Françoise Médale
- INRAE, Univ. Pau & Pays Adour, E2S UPPA, Nutrition, Métabolisme et Aquaculture, UMR 1419, 64310, Saint Pée sur Nivelle, France
| | - Mathilde Dupont-Nivet
- Université Paris-Saclay, INRAE, AgroParisTech, Génétique Animale et Biologie Intégrative, UMR 1313, 78350, Jouy-en-Josas, France
| | - Florence Lefevre
- INRAE, Laboratoire de Physiologie et Génomique des Poissons, UR 1037, 35000, Rennes, France
| | - Jérome Bugeon
- INRAE, Laboratoire de Physiologie et Génomique des Poissons, UR 1037, 35000, Rennes, France
| | | | | | - Annick Moing
- Bordeaux Metabolome, MetaboHUB, Centre INRAE de Nouvelle-Aquitaine Bordeaux, 33140, Villenave d'Ornon, France
- Centre INRAE de Nouvelle-Aquitaine Bordeaux, INRAE, Univ. Bordeaux, Biologie du Fruit et Pathologie, UMR 1332, 33140, Villenave d'Ornon, France
| | - Benoit Fauconneau
- INRAE, Univ. Pau & Pays Adour, E2S UPPA, Nutrition, Métabolisme et Aquaculture, UMR 1419, 64310, Saint Pée sur Nivelle, France.
| |
Collapse
|
11
|
Ralli T, Saifi Z, Tyagi N, Vidyadhari A, Aeri V, Kohli K. Deciphering the role of gut metabolites in non-alcoholic fatty liver disease. Crit Rev Microbiol 2023; 49:815-833. [PMID: 36394607 DOI: 10.1080/1040841x.2022.2142091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/30/2022] [Accepted: 10/26/2022] [Indexed: 11/18/2022]
Abstract
Perturbations in microbial abundance or diversity in the intestinal lumen leads to intestinal inflammation and disruption of intestinal membrane which eventually facilitates the translocation of microbial metabolites or whole microbes to the liver and other organs through portal vein. This process of translocation finally leads to multitude of health disorders. In this review, we are going to focus on the mechanisms by which gut metabolites like SCFAs, tryptophan (Trp) metabolites, bile acids (BAs), ethanol, and choline can either cause the development/progression of non-alcoholic fatty liver disease (NAFLD) or serves as a therapeutic treatment for the disease. Alterations in some metabolites like SCFAs, Trp metabolites, etc., can serve as biomarker molecules whereas presence of specific metabolites like ethanol definitely leads to disease progression. Thus, proper understanding of these mechanisms will subsequently help in designing of microbiome-based therapeutic approaches. Furthermore, we have also focussed on the role of dysbiosis on the mucosal immune system. In addition, we would also compile up the microbiome-based clinical trials which are currently undergoing for the treatment of NAFLD and non-alcoholic steatohepatitis (NASH). It has been observed that the use of microbiome-based approaches like prebiotics, probiotics, symbiotics, etc., can act as a beneficial treatment option but more research needs to be done to know how to manipulate the composition of gut microbes.
Collapse
Affiliation(s)
- Tanya Ralli
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, New Delhi, India
| | - Zoya Saifi
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, New Delhi, India
| | - Neha Tyagi
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, New Delhi, India
| | - Arya Vidyadhari
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, New Delhi, India
| | - Vidhu Aeri
- Department of Pharmacognosy, School of Pharmaceutical Education and Research, New Delhi, India
| | - Kanchan Kohli
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, New Delhi, India
- Research and Publications, Llyod Institute of Management and Technology, Greater Noida, India
| |
Collapse
|
12
|
Han SG, Kwon HC, Kim DH, Hong SJ, Han SG. In Vitro Synergistic Antibacterial and Anti-Inflammatory Effects of Nisin and Lactic Acid in Yogurt against Helicobacter pylori and Human Gastric Cells. Food Sci Anim Resour 2023; 43:751-766. [PMID: 37701745 PMCID: PMC10493562 DOI: 10.5851/kosfa.2023.e34] [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: 06/13/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 09/14/2023] Open
Abstract
Helicobacter pylori is a bacterium that naturally thrives in acidic environments and has the potential to induce various gastrointestinal disorders in humans. The antibiotic therapy utilized for treating H. pylori can lead to undesired side effects, such as dysbiosis in the gut microbiota. The objective of our study was to explore the potential antibacterial effects of nisin and lactic acid (LA) in yogurt against H. pylori. Additionally, we investigated the anti-inflammatory effects of nisin and LA in human gastric (AGS) cells infected with H. pylori. Nisin and LA combination showed the strongest inhibitory activity, with confirmed synergy at 0.375 fractional inhibitory concentration index. Also, post-fermented yogurt with incorporation of nisin exhibited antibacterial effect against H. pylori. The combination of nisin and LA resulted in a significant reduction of mRNA levels of bacterial toxins of H. pylori and pro-inflammatory cytokines in AGS cells infected with H. pylori. Furthermore, this also increased bacterial membrane damage, which led to DNA and protein leakage in H. pylori. Overall, the combination of nisin and LA shows promise as an alternative therapy for H. pylori infection. Additionally, the incorporation of nisin into foods containing LA presents a potential application. Further studies, including animal research, are needed to validate these findings and explore clinical applications.
Collapse
Affiliation(s)
- Seo Gu Han
- Department of Food Science and
Biotechnology of Animal Resources, Konkuk University, Seoul
05029, Korea
| | - Hyuk Cheol Kwon
- Department of Food Science and
Biotechnology of Animal Resources, Konkuk University, Seoul
05029, Korea
| | - Do Hyun Kim
- Department of Food Science and
Biotechnology of Animal Resources, Konkuk University, Seoul
05029, Korea
| | - Seong Joon Hong
- Department of Food Science and
Biotechnology of Animal Resources, Konkuk University, Seoul
05029, Korea
| | - Sung Gu Han
- Department of Food Science and
Biotechnology of Animal Resources, Konkuk University, Seoul
05029, Korea
| |
Collapse
|
13
|
Schropp N, Stanislas V, Michels KB, Thriene K. How Do Prebiotics Affect Human Intestinal Bacteria?-Assessment of Bacterial Growth with Inulin and XOS In Vitro. Int J Mol Sci 2023; 24:12796. [PMID: 37628977 PMCID: PMC10454692 DOI: 10.3390/ijms241612796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/07/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Prebiotics are believed to exhibit high specificity in stimulating the growth or activity of a limited number of commensal microorganisms, thereby conferring health benefits to the host. However, the mechanism of action of prebiotics depends on multiple factors, including the composition of an individual's gut microbiota, and is therefore difficult to predict. It is known that different bacteria can utilize inulin and xylooligosaccharides (XOS), but an overview of which bacteria in the human gut may be affected is lacking. Detailed knowledge of how bacterial growth is affected by prebiotics is furthermore useful for the development of new synbiotics, which combine a living microorganism with a selective substrate to confer a health benefit to the host. Hence, we developed a statistical model to compare growth in vitro among typical human gut bacteria from different phylogenetic lineages. Based on continuous observation of the optical density (OD600), we compare maximal growth rates (rmax), maximal attained OD600 (ODmax), and area under the growth curve (AUC) of bacteria grown on inulin or XOS. The consideration of these three parameters suggests strain-specific preferences for inulin or XOS and reveals previously unknown preferences such as Streptococcus salivarius growth on XOS.
Collapse
Affiliation(s)
| | | | | | - Kerstin Thriene
- Institute for Prevention and Cancer Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, 79110 Freiburg, Germany; (N.S.); (V.S.); (K.B.M.)
| |
Collapse
|
14
|
Laue C, Papazova E, Pannenbeckers A, Schrezenmeir J. Effect of a Probiotic and a Synbiotic on Body Fat Mass, Body Weight and Traits of Metabolic Syndrome in Individuals with Abdominal Overweight: A Human, Double-Blind, Randomised, Controlled Clinical Study. Nutrients 2023; 15:3039. [PMID: 37447365 DOI: 10.3390/nu15133039] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 06/28/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
L. fermentum strains K7-Lb1, K8-Lb1 and K11-Lb3 were found to suppress Th1 and Th2 response and to enhance defensin release by enterocytes, respectively. Based on these anti-inflammatory actions, we investigated the effect of these strains on traits of metabolic syndrome, which is driven by low-grade inflammation. In a double-blind, randomised, placebo-controlled clinical trial with three parallel arms, 180 individuals with abdominal overweight were administered for 3 months with (1) placebo; (2) probiotic, comprising L. fermentum strains; or (3) synbiotic, comprising the strains + acacia gum (10 g daily). The effects were evaluated using Kruskal-Wallis one-way analysis of variance on ranks and post hoc tests (Holm-Sidak and Dunn's tests). The alteration (∆) in body fat mass (kg) (primary parameter) during intervention was significantly (p = 0.039) more pronounced in the Probiotic group (-0.61 ± 1.94; mean ± SD) compared with the Placebo group (+0.13 ± 1.64). Accordingly, differences were found in ∆ body weight (p = 0.012), BMI (p = 0.011), waist circumference (p = 0.03), waist-to-height ratio (p = 0.033), visceral adipose tissue (SAD) (p < 0.001) and liver steatosis grade (LSG) (p < 0.001), as assessed using sonography. In the Synbiotic group, ∆SAD (p = 0.002), ∆LSG (p < 0.001) and ∆constipation score (p = 0.009) were improved compared with Placebo. The probiotic mixture and the synbiotic improved the parameters associated with overweight.
Collapse
Affiliation(s)
- Christiane Laue
- Clinical Research Center Kiel, Kiel Center of Innovation and Technology, Schauenburgstraße 116, D-24118 Kiel, Germany
| | - Ekaterina Papazova
- Clinical Research Center Kiel, Kiel Center of Innovation and Technology, Schauenburgstraße 116, D-24118 Kiel, Germany
| | - Angelika Pannenbeckers
- Clinical Research Center Kiel, Kiel Center of Innovation and Technology, Schauenburgstraße 116, D-24118 Kiel, Germany
| | - Jürgen Schrezenmeir
- Clinical Research Center Kiel, Kiel Center of Innovation and Technology, Schauenburgstraße 116, D-24118 Kiel, Germany
- University Medicine, Johannes-Gutenberg University, D-55131 Mainz, Germany
| |
Collapse
|
15
|
Maccioni L, Fu Y, Horsmans Y, Leclercq I, Stärkel P, Kunos G, Gao B. Alcohol-associated bowel disease: new insights into pathogenesis. EGASTROENTEROLOGY 2023; 1:e100013. [PMID: 37662449 PMCID: PMC10472976 DOI: 10.1136/egastro-2023-100013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Excessive alcohol drinking can cause pathological changes including carcinogenesis in the digestive tract from mouth to large intestine, but the underlying mechanisms are not fully understood. In this review, we discuss the effects of alcohol on small and large intestinal functions, such as leaky gut, dysbiosis and alterations of intestinal epithelium and gut immune dysfunctions, commonly referred to as alcohol-associated bowel disease (ABD). To date, detailed mechanistic insights into ABD are lacking. Accumulating evidence suggests a pathogenic role of ethanol metabolism in dysfunctions of the intestinal tract. Ethanol metabolism generates acetaldehyde and acetate, which could potentially promote functional disruptions of microbial and host components of the intestinal barrier along the gastrointestinal tract. The potential involvement of acetaldehyde and acetate in the pathogenesis of the underlying ABD, including cancer, is discussed. We also highlight some gaps in knowledge existing in the field of ABD. Finally, we discuss future directions in exploring the role of acetaldehyde and acetate generated during chronic alcohol intake in various pathologies affecting different sites of the intestinal tract.
Collapse
Affiliation(s)
- Luca Maccioni
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Yaojie Fu
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Yves Horsmans
- Department of Hepato-Gastroenterology, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Isabelle Leclercq
- Laboratory of Hepato-Gastroenterology, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| | - Peter Stärkel
- Department of Hepato-Gastroenterology, Cliniques universitaires Saint-Luc, Brussels, Belgium
- Laboratory of Hepato-Gastroenterology, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| | - George Kunos
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
16
|
Cordell BJ. Shifting microbiomes: pathobionts hiding in our guts. EBioMedicine 2023; 92:104592. [PMID: 37119669 PMCID: PMC10165201 DOI: 10.1016/j.ebiom.2023.104592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 04/14/2023] [Indexed: 05/01/2023] Open
|
17
|
Mbaye B, Wasfy RM, Alou MT, Borentain P, Andrieu C, Caputo A, Raoult D, Gerolami R, Million M. Limosilactobacillus fermentum, Lactococcus lactis and Thomasclavelia ramosa are enriched and Methanobrevibacter smithii is depleted in patients with non-alcoholic steatohepatitis. Microb Pathog 2023; 180:106160. [PMID: 37217120 DOI: 10.1016/j.micpath.2023.106160] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/28/2023] [Accepted: 05/15/2023] [Indexed: 05/24/2023]
Abstract
Non-alcoholic fatty liver (NAFLD), and its complicated form, non-alcoholic steatohepatitis (NASH), have been associated with gut dysbiosis with specific signatures. Endogenous ethanol production by Klebsiella pneumoniae or yeasts has been identified as a potential physio-pathological mechanism. A species-specific association between Lactobacillus and obesity and metabolic diseases has been reported. In this study, the microbial composition of ten cases of NASH and ten controls was determined using v3v4 16S amplicon sequencing as well as quantitative PCR (qPCR). Using different statistical approaches, we found an association of Lactobacillus and Lactoccocus with NASH, and an association of Methanobrevibacter, Faecalibacterium and Romboutsia with controls. At the species level, Limosilactobacillus fermentum and Lactococcus lactis, two species producing ethanol, and Thomasclavelia ramosa, a species already associated with dysbiosis, were associated with NASH. Using qPCR, we observed a decreased frequency of Methanobrevibacter smithii and confirmed the high prevalence of L. fermentum in NASH samples (5/10), while all control samples were negative (p = 0.02). In contrast, Ligilactobacillus ruminis was associated with controls. This supports the critical importance of taxonomic resolution at the species level, notably with the recent taxonomic reclassification of the Lactobacillus genus. Our results point towards the potential instrumental role of ethanol-producing gut microbes in NASH patients, notably lactic acid bacteria, opening new avenues for prevention and treatment.
Collapse
Affiliation(s)
- Babacar Mbaye
- IHU Méditerranée Infection, Marseille, France; Microbes Evolution Phylogeny and Infections (MEPHI), Institut de Recherche pour le Développement, Aix-Marseille Université, Marseille, France
| | - Reham Magdy Wasfy
- IHU Méditerranée Infection, Marseille, France; Microbes Evolution Phylogeny and Infections (MEPHI), Institut de Recherche pour le Développement, Aix-Marseille Université, Marseille, France
| | - Maryam Tidjani Alou
- IHU Méditerranée Infection, Marseille, France; Microbes Evolution Phylogeny and Infections (MEPHI), Institut de Recherche pour le Développement, Aix-Marseille Université, Marseille, France
| | | | - Claudia Andrieu
- IHU Méditerranée Infection, Marseille, France; Microbes Evolution Phylogeny and Infections (MEPHI), Institut de Recherche pour le Développement, Aix-Marseille Université, Marseille, France; Assistance Publique-Hôpitaux de Marseille, Marseille, France
| | - Aurelia Caputo
- IHU Méditerranée Infection, Marseille, France; Microbes Evolution Phylogeny and Infections (MEPHI), Institut de Recherche pour le Développement, Aix-Marseille Université, Marseille, France; Assistance Publique-Hôpitaux de Marseille, Marseille, France
| | - Didier Raoult
- IHU Méditerranée Infection, Marseille, France; Microbes Evolution Phylogeny and Infections (MEPHI), Institut de Recherche pour le Développement, Aix-Marseille Université, Marseille, France; Assistance Publique-Hôpitaux de Marseille, Marseille, France
| | - Rene Gerolami
- Microbes Evolution Phylogeny and Infections (MEPHI), Institut de Recherche pour le Développement, Aix-Marseille Université, Marseille, France; Assistance Publique-Hôpitaux de Marseille, Marseille, France; Unité hépatologie, Hôpital de la Timone, Marseille, France
| | - Matthieu Million
- IHU Méditerranée Infection, Marseille, France; Microbes Evolution Phylogeny and Infections (MEPHI), Institut de Recherche pour le Développement, Aix-Marseille Université, Marseille, France; Assistance Publique-Hôpitaux de Marseille, Marseille, France.
| |
Collapse
|
18
|
Trius-Soler M, Praticò G, Gürdeniz G, Garcia-Aloy M, Canali R, Fausta N, Brouwer-Brolsma EM, Andrés-Lacueva C, Dragsted LO. Biomarkers of moderate alcohol intake and alcoholic beverages: a systematic literature review. GENES & NUTRITION 2023; 18:7. [PMID: 37076809 PMCID: PMC10114415 DOI: 10.1186/s12263-023-00726-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 04/04/2023] [Indexed: 04/21/2023]
Abstract
The predominant source of alcohol in the diet is alcoholic beverages, including beer, wine, spirits and liquors, sweet wine, and ciders. Self-reported alcohol intakes are likely to be influenced by measurement error, thus affecting the accuracy and precision of currently established epidemiological associations between alcohol itself, alcoholic beverage consumption, and health or disease. Therefore, a more objective assessment of alcohol intake would be very valuable, which may be established through biomarkers of food intake (BFIs). Several direct and indirect alcohol intake biomarkers have been proposed in forensic and clinical contexts to assess recent or longer-term intakes. Protocols for performing systematic reviews in this field, as well as for assessing the validity of candidate BFIs, have been developed within the Food Biomarker Alliance (FoodBAll) project. The aim of this systematic review is to list and validate biomarkers of ethanol intake per se excluding markers of abuse, but including biomarkers related to common categories of alcoholic beverages. Validation of the proposed candidate biomarker(s) for alcohol itself and for each alcoholic beverage was done according to the published guideline for biomarker reviews. In conclusion, common biomarkers of alcohol intake, e.g., as ethyl glucuronide, ethyl sulfate, fatty acid ethyl esters, and phosphatidyl ethanol, show considerable inter-individual response, especially at low to moderate intakes, and need further development and improved validation, while BFIs for beer and wine are highly promising and may help in more accurate intake assessments for these specific beverages.
Collapse
Affiliation(s)
- Marta Trius-Soler
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 1958, Frederiksberg C, Denmark
- Polyphenol Research Laboratory, Department of Nutrition, Food Sciences and Gastronomy, XIA School of Pharmacy and Food Sciences, University of Barcelona, 08028, Barcelona, Spain
- INSA-UB, Nutrition and Food Safety Research Institute, University of Barcelona, 08921, Santa Coloma de Gramanet, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Giulia Praticò
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 1958, Frederiksberg C, Denmark
| | - Gözde Gürdeniz
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 1958, Frederiksberg C, Denmark
| | - Mar Garcia-Aloy
- Biomarker & Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028, Barcelona, Spain
- Metabolomics Unit, Research and Innovation Centre, Fondazione Edmund Mach, San Michele All'Adige, Italy
| | - Raffaella Canali
- Consiglio Per La Ricerca in Agricoltura E L'analisi Dell'economia Agraria (CREA) Research Centre for Food and Nutrition, Rome, Italy
| | - Natella Fausta
- Consiglio Per La Ricerca in Agricoltura E L'analisi Dell'economia Agraria (CREA) Research Centre for Food and Nutrition, Rome, Italy
| | - Elske M Brouwer-Brolsma
- Division of Human Nutrition and Health, Department Agrotechnology and Food Sciences, Wageningen University and Research, P.O. Box 17, 6700 AA, Wageningen, The Netherlands
| | - Cristina Andrés-Lacueva
- INSA-UB, Nutrition and Food Safety Research Institute, University of Barcelona, 08921, Santa Coloma de Gramanet, Spain
- Biomarker & Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fragilidad Y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Lars Ove Dragsted
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 1958, Frederiksberg C, Denmark.
| |
Collapse
|
19
|
Kato I, Sun J. Microbiome and Diet in Colon Cancer Development and Treatment. Cancer J 2023; 29:89-97. [PMID: 36957979 PMCID: PMC10037538 DOI: 10.1097/ppo.0000000000000649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2023]
Abstract
ABSTRACT Diet plays critical roles in defining our immune responses, microbiome, and progression of human diseases. With recent progress in sequencing and bioinformatic techniques, increasing evidence indicates the importance of diet-microbial interactions in cancer development and therapeutic outcome. Here, we focus on the epidemiological studies on diet-bacterial interactions in the colon cancer. We also review the progress of mechanistic studies using the experimental models. Finally, we discuss the limits and future directions in the research of microbiome and diet in cancer development and therapeutic outcome. Now, it is clear that microbes can influence the efficacy of cancer therapies. These research results open new possibilities for the diagnosis, prevention, and treatment of cancer. However, there are still big gaps to apply these new findings to the clinical practice.
Collapse
Affiliation(s)
- Ikuko Kato
- Department of Oncology, Wayne State University, Detroit Michigan, USA
- Department of Pathology, Wayne State University, Detroit Michigan, USA
| | - Jun Sun
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, 840 S Wood Street, Room 704 CSB, MC716, Chicago, IL 60612, USA
- Department of Microbiology/Immunology, University of Illinois Chicago, Chicago, IL 60612, USA
- University of Illinois Cancer Center, 818 S Wolcott Avenue, Chicago, IL 60612, USA
- Jesse Brown VA Medical Center, 820 S. Damen Avenue, Chicago, IL 60612, USA
| |
Collapse
|
20
|
Yao H, Flanagan BM, Williams BA, Mikkelsen D, Gidley MJ. Lactate and buyrate proportions, methanogen growth and gas production during in vitro dietary fibre fermentation all depend on fibre concentration. Food Hydrocoll 2023. [DOI: 10.1016/j.foodhyd.2022.108061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
21
|
Koning M, Herrema H, Nieuwdorp M, Meijnikman AS. Targeting nonalcoholic fatty liver disease via gut microbiome-centered therapies. Gut Microbes 2023; 15:2226922. [PMID: 37610978 PMCID: PMC10305510 DOI: 10.1080/19490976.2023.2226922] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 08/25/2023] Open
Abstract
Humans possess abundant amounts of microorganisms, including bacteria, fungi, viruses, and archaea, in their gut. Patients with nonalcoholic fatty liver disease (NAFLD) exhibit alterations in their gut microbiome and an impaired gut barrier function. Preclinical studies emphasize the significance of the gut microbiome in the pathogenesis of NAFLD. In this overview, we explore how adjusting the gut microbiome could serve as an innovative therapeutic strategy for NAFLD. We provide a summary of current information on untargeted techniques such as probiotics and fecal microbiota transplantation, as well as targeted microbiome-focused therapies including engineered bacteria, prebiotics, postbiotics, and phages for the treatment of NAFLD.
Collapse
Affiliation(s)
- Mijra Koning
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Diabetes, Amsterdam, The Netherlands
| | - Hilde Herrema
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
- Amsterdam Gastroenterology and Metabolism, Amsterdam, The Netherlands
| | - Max Nieuwdorp
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Diabetes, Amsterdam, The Netherlands
| | - Abraham S. Meijnikman
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Diabetes, Amsterdam, The Netherlands
| |
Collapse
|
22
|
Enespa, Chandra P. Tool and techniques study to plant microbiome current understanding and future needs: an overview. Commun Integr Biol 2022; 15:209-225. [PMID: 35967908 PMCID: PMC9367660 DOI: 10.1080/19420889.2022.2082736] [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] [Indexed: 12/02/2022] Open
Abstract
Microorganisms are present in the universe and they play role in beneficial and harmful to human life, society, and environments. Plant microbiome is a broad term in which microbes are present in the rhizo, phyllo, or endophytic region and play several beneficial and harmful roles with the plant. To know of these microorganisms, it is essential to be able to isolate purification and identify them quickly under laboratory conditions. So, to improve the microbial study, several tools and techniques such as microscopy, rRNA, or rDNA sequencing, fingerprinting, probing, clone libraries, chips, and metagenomics have been developed. The major benefits of these techniques are the identification of microbial community through direct analysis as well as it can apply in situ. Without tools and techniques, we cannot understand the roles of microbiomes. This review explains the tools and their roles in the understanding of microbiomes and their ecological diversity in environments.
Collapse
Affiliation(s)
- Enespa
- Department of Plant Pathology, School of Agriculture, SMPDC, University of Lucknow, Lucknow, India
| | - Prem Chandra
- Department of Environmental Microbiology, Babasaheb Bhimrao Ambedkar (A Central) University, Lucknow, India
| |
Collapse
|
23
|
Fatty Liver Disease-Alcoholic and Non-Alcoholic: Similar but Different. Int J Mol Sci 2022; 23:ijms232416226. [PMID: 36555867 PMCID: PMC9783455 DOI: 10.3390/ijms232416226] [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: 10/17/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
In alcohol-induced liver disease (ALD) and in non-alcoholic fatty liver disease (NAFLD), there are abnormal accumulations of fat in the liver. This phenomenon may be related to excessive alcohol consumption, as well as the combination of alcohol consumption and medications. There is an evolution from simple steatosis to steatohepatitis, fibrosis and cirrhosis leading to hepatocellular carcinoma (HCC). Hepatic pathology is very similar regarding non-alcoholic fatty liver disease (NAFLD) and ALD. Initially, there is lipid accumulation in parenchyma and progression to lobular inflammation. The morphological changes in the liver mitochondria, perivenular and perisinusoidal fibrosis, and hepatocellular ballooning, apoptosis and necrosis and accumulation of fibrosis may lead to the development of cirrhosis and HCC. Medical history of ethanol consumption, laboratory markers of chronic ethanol intake, AST/ALT ratio on the one hand and features of the metabolic syndrome on the other hand, may help in estimating the contribution of alcohol intake and the metabolic syndrome, respectively, to liver steatosis.
Collapse
|
24
|
Pezzino S, Sofia M, Faletra G, Mazzone C, Litrico G, La Greca G, Latteri S. Gut-Liver Axis and Non-Alcoholic Fatty Liver Disease: A Vicious Circle of Dysfunctions Orchestrated by the Gut Microbiome. BIOLOGY 2022; 11:1622. [PMID: 36358323 PMCID: PMC9687983 DOI: 10.3390/biology11111622] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 09/24/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a prevalent, multifactorial, and poorly understood liver disease with an increasing incidence worldwide. NAFLD is typically asymptomatic and coupled with other symptoms of metabolic syndrome. The prevalence of NAFLD is rising in tandem with the prevalence of obesity. In the Western hemisphere, NAFLD is one of the most prevalent causes of liver disease and liver transplantation. Recent research suggests that gut microbiome dysbiosis may play a significant role in the pathogenesis of NAFLD by dysregulating the gut-liver axis. The so-called "gut-liver axis" refers to the communication and feedback loop between the digestive system and the liver. Several pathological mechanisms characterized the alteration of the gut-liver axis, such as the impairment of the gut barrier and the increase of the intestinal permeability which result in endotoxemia and inflammation, and changes in bile acid profiles and metabolite levels produced by the gut microbiome. This review will explore the role of gut-liver axis disruption, mediated by gut microbiome dysbiosis, on NAFLD development.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Saverio Latteri
- Department of Surgical Sciences and Advanced Technologies “G. F. Ingrassia”, Cannizzaro Hospital, University of Catania, 95126 Catania, Italy
| |
Collapse
|
25
|
Guman MSS, Hoozemans JB, Haal S, de Jonge PA, Aydin Ö, Lappa D, Meijnikman AS, Westerink F, Acherman Y, Bäckhed F, de Brauw M, Nielsen J, Nieuwdorp M, Groen AK, Gerdes VEA. Adipose Tissue, Bile Acids, and Gut Microbiome Species Associated With Gallstones After Bariatric Surgery. J Lipid Res 2022; 63:100280. [PMID: 36115596 PMCID: PMC9672443 DOI: 10.1016/j.jlr.2022.100280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 08/09/2022] [Accepted: 08/27/2022] [Indexed: 11/29/2022] Open
Abstract
Several risk factors are associated with gallstone disease after bariatric surgery, but the underlying pathophysiological mechanisms of gallstone formation are unclear. We hypothesize that gallstone formation after bariatric surgery is induced by different pathways compared with gallstone formation in the general population, since postoperative formation occurs rapidly in patients who did not develop gallstones in preceding years. To identify both pathophysiological and potentially protective mechanisms against postoperative gallstone formation, we compared the preoperative fasting metabolome, fecal microbiome, and liver and adipose tissue transcriptome obtained before or during bariatric surgery of obese patients with and without postoperative gallstones. In total, 88 patients were selected from the BARIA longitudinal cohort study. Within this group, 32 patients had postoperative gallstones within 2 years. Gut microbiota metagenomic analyses showed group differences in abundance of 41 bacterial species, particularly abundance of Lactobacillaceae and Enterobacteriaceae in patients without gallstones. Subcutaneous adipose tissue transcriptomic analyses revealed four genes that were suppressed in gallstone patients compared with patients without gallstones. These baseline gene expression and gut microbiota composition differences might relate to protective mechanisms against gallstone formation after bariatric surgery. Moreover, baseline fasting blood samples of patients with postoperative gallstones showed increased levels of several bile acids. Overall, we revealed different genes and bacteria associated with gallstones than those previously reported in the general population, supporting the hypothesis that gallstone formation after bariatric surgery follows a different trajectory. Further research is necessary to confirm the involvement of the bile acids, adipose tissue activity, and microbial species observed here.
Collapse
Affiliation(s)
- M S S Guman
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands; Department of Internal Medicine, Spaarne Gasthuis, Hoofddorp, the Netherlands.
| | - J B Hoozemans
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands; Department of Internal Medicine, Spaarne Gasthuis, Hoofddorp, the Netherlands
| | - S Haal
- Department of Internal Medicine, Spaarne Gasthuis, Hoofddorp, the Netherlands; Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Location AMC, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, the Netherlands
| | - P A de Jonge
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
| | - Ö Aydin
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
| | - D Lappa
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - A S Meijnikman
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
| | - F Westerink
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
| | - Y Acherman
- Department of Surgery, Spaarne Gasthuis, Hoofddorp, the Netherlands
| | - F Bäckhed
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Goteborg, Sweden; Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Kobenhavn, Denmark; Department of Clinical Physiology, Region Västtra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - M de Brauw
- Department of Surgery, Spaarne Gasthuis, Hoofddorp, the Netherlands
| | - J Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - M Nieuwdorp
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
| | - A K Groen
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
| | - V E A Gerdes
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands; Department of Internal Medicine, Spaarne Gasthuis, Hoofddorp, the Netherlands
| |
Collapse
|
26
|
Mbaye B, Borentain P, Magdy Wasfy R, Alou MT, Armstrong N, Mottola G, Meddeb L, Ranque S, Gérolami R, Million M, Raoult D. Endogenous Ethanol and Triglyceride Production by Gut Pichia kudriavzevii, Candida albicans and Candida glabrata Yeasts in Non-Alcoholic Steatohepatitis. Cells 2022; 11:cells11213390. [PMID: 36359786 PMCID: PMC9654979 DOI: 10.3390/cells11213390] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/23/2022] [Accepted: 10/24/2022] [Indexed: 11/23/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH) increases with fructose consumption and metabolic syndrome and has been recently linked with endogenous ethanol production, notably by high alcohol-producing Klebsiella pneumoniae (HiAlc Kpn). Candida yeasts are the main causes of auto-brewery syndromes but have been neglected in NASH. Here, the fecal ethanol and microbial content of 10 cases and 10 controls were compared. Ethanol was measured by gas chromatography-mass spectrometry. Species identification was performed by MALDI-TOF MS, and triglyceride production was assessed by a colorimetric enzymatic assay. The fecal ethanol concentration was four times higher in patients with NASH (median [interquartile range]: 0.13 [0.05–1.43] vs. 0.034 [0.008–0.57], p = 0.037). Yeasts were isolated from almost all cases but not from controls (9/10 vs. 0/10, p = 0.0001). Pichia kudriavzevii was the most frequent (four patients), while Candida glabrata, Candida albicans, and Galactomyces geotrichum were identified in two cases each. The concentration of ethanol produced by yeasts was 10 times higher than that produced by bacteria (median, 3.36 [0.49–5.60] vs. 0.32 [0.009–0.43], p = 0.0029). Using a 10% D-fructose restricted medium, we showed that NASH-associated yeasts transformed fructose in ethanol. Unexpectedly, yeasts isolated from NASH patients produced a substantial amount of triglycerides. Pichia kudriavzevii strains produced the maximal ethanol and triglyceride levels in vitro. Our preliminary human descriptive and in vitro experimental results suggest that yeasts have been neglected. In addition to K. pneumoniae, gut Pichia and Candida yeasts could be linked with NASH pathophysiology in a species- and strain-specific manner through fructose-dependent endogenous alcohol and triglyceride production.
Collapse
Affiliation(s)
- Babacar Mbaye
- IHU Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13005 Marseille, France
- Microbes Evolution Phylogeny and Infections (MEPHI), Institut de Recherche Pour le Développement, Aix-Marseille Université, 13005 Marseille, France
| | - Patrick Borentain
- Unité Hépatologie, Hôpital de la Timone, APHM, 13005 Marseille, France
| | - Reham Magdy Wasfy
- IHU Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13005 Marseille, France
- Microbes Evolution Phylogeny and Infections (MEPHI), Institut de Recherche Pour le Développement, Aix-Marseille Université, 13005 Marseille, France
| | - Maryam Tidjani Alou
- IHU Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13005 Marseille, France
- Microbes Evolution Phylogeny and Infections (MEPHI), Institut de Recherche Pour le Développement, Aix-Marseille Université, 13005 Marseille, France
| | - Nicholas Armstrong
- IHU Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13005 Marseille, France
- Microbes Evolution Phylogeny and Infections (MEPHI), Institut de Recherche Pour le Développement, Aix-Marseille Université, 13005 Marseille, France
- Assistance Publique-Hôpitaux de Marseille, 13005 Marseille, France
| | - Giovanna Mottola
- Laboratoire de Biochimie, Hôpital de la Timone, APHM, 13005 Marseille, France
- C2VN, INSERM 1263, INRAE 1260, Team 5, Aix-Marseille Université, 13005 Marseille, France
| | - Line Meddeb
- IHU Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13005 Marseille, France
- Assistance Publique-Hôpitaux de Marseille, 13005 Marseille, France
| | - Stéphane Ranque
- IHU Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13005 Marseille, France
- VITROME: Vecteurs-Infections Tropicales et Méditerranéennes, Institut de Recherche Pour le Développement, Assistance Publique-Hôpitaux de Marseille, Service de Santé des Armées, Aix Marseille Université, 13385 Marseille, France
| | - René Gérolami
- IHU Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13005 Marseille, France
- Microbes Evolution Phylogeny and Infections (MEPHI), Institut de Recherche Pour le Développement, Aix-Marseille Université, 13005 Marseille, France
- Unité Hépatologie, Hôpital de la Timone, APHM, 13005 Marseille, France
| | - Matthieu Million
- IHU Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13005 Marseille, France
- Microbes Evolution Phylogeny and Infections (MEPHI), Institut de Recherche Pour le Développement, Aix-Marseille Université, 13005 Marseille, France
- Assistance Publique-Hôpitaux de Marseille, 13005 Marseille, France
- Correspondence: ; Tel.: +33-413-732-401; Fax: +33-413-732-402
| | - Didier Raoult
- IHU Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13005 Marseille, France
- Microbes Evolution Phylogeny and Infections (MEPHI), Institut de Recherche Pour le Développement, Aix-Marseille Université, 13005 Marseille, France
| |
Collapse
|
27
|
Remy R, Kemnitz N, Trefz P, Fuchs P, Bartels J, Klemenz AC, Rührmund L, Sukul P, Miekisch W, Schubert JK. Profiling of exhaled volatile organics in the screening scenario of a COVID-19 test center. iScience 2022; 25:105195. [PMID: 36168390 PMCID: PMC9502439 DOI: 10.1016/j.isci.2022.105195] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 07/23/2022] [Accepted: 09/20/2022] [Indexed: 11/29/2022] Open
Abstract
Breath volatile organics (VOCs) may provide immediate information on infection mechanisms and host response. We conducted real-time mass spectrometry-based breath profiling in 708 non-preselected consecutive subjects in the screening scenario of a COVID-19 test center. Recruited subjects were grouped based on PCR-confirmed infection status and presence or absence of flu-like symptoms. Exhaled VOC profiles of SARS-CoV-2-positive cases (n = 36) differed from healthy (n = 256) and those with other respiratory infections (n = 416). Concentrations of most VOCs were suppressed in COVID-19. VOC concentrations also differed between symptomatic and asymptomatic cases. Breath markers mirror effects of infections onto host's cellular metabolism and microbiome. Downregulation of specific VOCs was attributed to suppressive effects of SARS-CoV-2 onto gut or pulmonary microbial metabolism. Breath analysis holds potential for monitoring SARS-CoV-2 infections rather than for primary diagnosis. Breath profiling offers unconventional insight into host-virus cross-talk and infection microbiology and enables non-invasive assessment of disease manifestation.
Collapse
Affiliation(s)
- Rasmus Remy
- Rostock Medical Breath Research Analytics and Technologies (ROMBAT), Department of Anaesthesiology and Intensive Care, University Medicine Rostock, 18057 Rostock, Germany
| | - Nele Kemnitz
- Rostock Medical Breath Research Analytics and Technologies (ROMBAT), Department of Anaesthesiology and Intensive Care, University Medicine Rostock, 18057 Rostock, Germany
| | - Phillip Trefz
- Rostock Medical Breath Research Analytics and Technologies (ROMBAT), Department of Anaesthesiology and Intensive Care, University Medicine Rostock, 18057 Rostock, Germany
| | - Patricia Fuchs
- Rostock Medical Breath Research Analytics and Technologies (ROMBAT), Department of Anaesthesiology and Intensive Care, University Medicine Rostock, 18057 Rostock, Germany
| | - Julia Bartels
- Rostock Medical Breath Research Analytics and Technologies (ROMBAT), Department of Anaesthesiology and Intensive Care, University Medicine Rostock, 18057 Rostock, Germany
| | - Ann-Christin Klemenz
- Rostock Medical Breath Research Analytics and Technologies (ROMBAT), Department of Anaesthesiology and Intensive Care, University Medicine Rostock, 18057 Rostock, Germany
| | - Leo Rührmund
- Rostock Medical Breath Research Analytics and Technologies (ROMBAT), Department of Anaesthesiology and Intensive Care, University Medicine Rostock, 18057 Rostock, Germany
| | - Pritam Sukul
- Rostock Medical Breath Research Analytics and Technologies (ROMBAT), Department of Anaesthesiology and Intensive Care, University Medicine Rostock, 18057 Rostock, Germany
| | - Wolfram Miekisch
- Rostock Medical Breath Research Analytics and Technologies (ROMBAT), Department of Anaesthesiology and Intensive Care, University Medicine Rostock, 18057 Rostock, Germany
| | - Jochen K. Schubert
- Rostock Medical Breath Research Analytics and Technologies (ROMBAT), Department of Anaesthesiology and Intensive Care, University Medicine Rostock, 18057 Rostock, Germany
| |
Collapse
|
28
|
Kumar A, Sakhare K, Bhattacharya D, Chattopadhyay R, Parikh P, Narayan KP, Mukherjee A. Communication in non-communicable diseases (NCDs) and role of immunomodulatory nutraceuticals in their management. Front Nutr 2022; 9:966152. [PMID: 36211513 PMCID: PMC9532975 DOI: 10.3389/fnut.2022.966152] [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: 06/13/2022] [Accepted: 08/11/2022] [Indexed: 12/24/2022] Open
Abstract
Conveyance of pathogens between organisms causes communicable diseases. On the other hand, a non-communicable disease (NCD) was always thought to have no causative transmissible infective agents. Today, this clear distinction is increasingly getting blurred and NCDs are found to be associated with some transmissible components. The human microbiota carries a congregation of microbes, the majority and the most widely studied being bacteria in the gut. The adult human gut harbors ginormous inhabitant microbes, and the microbiome accommodates 150-fold more genes than the host genome. Microbial communities share a mutually beneficial relationship with the host, especially with respect to host physiology including digestion, immune responses, and metabolism. This review delineates the connection between environmental factors such as infections leading to gut dysbiosis and NCDs and explores the evidence regarding possible causal link between them. We also discuss the evidence regarding the value of appropriate therapeutic immunomodulatory nutritional interventions to reduce the development of such diseases. We behold such immunomodulatory effects have the potential to influence in various NCDs and restore homeostasis. We believe that the beginning of the era of microbiota-oriented personalized treatment modalities is not far away.
Collapse
Affiliation(s)
- Abhiram Kumar
- Esperer Onco Nutrition Pvt. Ltd., Mumbai, India
- Department of Biological Sciences, Birla Institute of Technology and Science – Pilani, Hyderabad, India
| | - Kalyani Sakhare
- Department of Biological Sciences, Birla Institute of Technology and Science – Pilani, Hyderabad, India
| | - Dwaipayan Bhattacharya
- Department of Biological Sciences, Birla Institute of Technology and Science – Pilani, Hyderabad, India
| | | | - Purvish Parikh
- Department of Clinical Haematology, Mahatma Gandhi Medical College and Hospital, Jaipur, India
| | - Kumar P. Narayan
- Department of Biological Sciences, Birla Institute of Technology and Science – Pilani, Hyderabad, India
- *Correspondence: Kumar P. Narayan,
| | | |
Collapse
|
29
|
Lapiere A, Richard ML. Bacterial-fungal metabolic interactions within the microbiota and their potential relevance in human health and disease: a short review. Gut Microbes 2022; 14:2105610. [PMID: 35903007 PMCID: PMC9341359 DOI: 10.1080/19490976.2022.2105610] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [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
The composition of the microbiota is the focus of many recent publications describing the effects of the microbiota on host health. In recent years, research has progressed further, investigating not only the diversity of genes and functions but also metabolites produced by microorganisms composing the microbiota of various niches and how these metabolites affect and shape the microbial community. While an abundance of data has been published on bacterial interactions, much less data are available on the interactions of bacteria with another component of the microbiota: the fungal community. Although present in smaller numbers, fungi are essential to the balance of this complex microbial ecosystem. Both bacterial and fungal communities produce metabolites that influence their own population but also that of the other. However, to date, interkingdom interactions occurring through metabolites produced by bacteria and fungi have rarely been described. In this review, we describe the major metabolites produced by both kingdoms and discuss how they influence each other, by what mechanisms and with what consequences for the host.
Collapse
Affiliation(s)
- Alexia Lapiere
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France,Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, France
| | - Mathias L Richard
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France,Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, France,CONTACT Mathias L Richard INRAE, Micalis Institute, Probihote Team, Domaine de Vilvert, 78352, Jouy en Josas, France
| |
Collapse
|
30
|
Bo B, Seong H, Kim G, Han NS. Antioxidant and prebiotic activities of Laphet, fermented tea leaves in Myanmar, during in vitro gastrointestinal digestion and colonic fermentation. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022] Open
|
31
|
Zhou J, Duan M, Wang X, Zhang F, Zhou H, Ma T, Yin Q, Zhang J, Tian F, Wang G, Yang C. A feedback loop engaging propionate catabolism intermediates controls mitochondrial morphology. Nat Cell Biol 2022; 24:526-537. [PMID: 35418624 DOI: 10.1038/s41556-022-00883-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 02/28/2022] [Indexed: 12/17/2022]
Abstract
D-2-Hydroxyglutarate (D-2HG) is an α-ketoglutarate-derived mitochondrial metabolite that causes D-2-hydroxyglutaric aciduria, a devastating developmental disorder. How D-2HG adversely affects mitochondria is largely unknown. Here, we report that in Caenorhabditis elegans, loss of the D-2HG dehydrogenase DHGD-1 causes D-2HG accumulation and mitochondrial damage. The excess D-2HG leads to a build-up of 3-hydroxypropionate (3-HP), a toxic metabolite in mitochondrial propionate oxidation, by inhibiting the 3-HP dehydrogenase HPHD-1. We demonstrate that 3-HP binds the MICOS subunit MIC60 (encoded by immt-1) and inhibits its membrane-binding and membrane-shaping activities. We further reveal that dietary and gut bacteria affect mitochondrial health by modulating the host production of 3-HP. These findings identify a feedback loop that links the toxic effects of D-2HG and 3-HP on mitochondria, thus providing important mechanistic insights into human diseases related to D-2HG and 3-HP.
Collapse
Affiliation(s)
- Junxiang Zhou
- State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Mei Duan
- State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China.
| | - Xin Wang
- State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Fengxia Zhang
- State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Hejiang Zhou
- State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Tengfei Ma
- State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Qiuyuan Yin
- State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Jie Zhang
- State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Fei Tian
- State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Guodong Wang
- State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Chonglin Yang
- State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China.
| |
Collapse
|
32
|
Effect of β-Estradiol on Mono- and Mixed-Species Biofilms of Human Commensal Bacteria Lactobacillus paracasei AK508 and Micrococcus luteus C01 on Different Model Surfaces. COATINGS 2022. [DOI: 10.3390/coatings12040436] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The impact of steroid hormones, and particularly estradiol, on human microbiota could be recognized as a substantial part of human-microbiota interactions. However, an area that remains poorly investigated is that of the skin and vaginal microbial communities and biofilms, which contain non-pathogenic bacteria of phyla Firmicutes and Actinobacteria, especially probiotic bacteria of the genus Lactobacillus and the widespread, safe skin genus, Micrococcus. Experiments with Lactobacillus paracasei AK508 and Micrococcus luteus C01 biofilms on PTFE cubes showed dose-dependent effects of estradiol at concentrations of 0.22 nM and 22 nM. The hormone mostly inhibits L. paracasei growth and stimulates M. luteus. The presented studies of colony-forming unit (CFU) amountsand cell aggregation in biofilms on glass fiber filters showed the same general tendencies. Estradiol generally increased the aggregation of cells in monospecies communities and potentially changed the synthesis of antibacterial metabolites in L. paracasei. The balance between two bacteria in mixed-species biofilms depended on the initial adhesion stage, and when this stage was reduced, micrococci were more resistant to the antagonistic action of L. paracasei. Moreover, in mixed-species biofilms, the effect of estradiol on lactobacilli altered from inhibition to stimulation, potentially due to the presence of M. luteus. At the same time, ethanol as a solvent for estradiol at the concentration 0.6% acted mostly as an antagonist of the hormone and had an opposite effect on bacteria; nevertheless, the overlapping of ethanol and estradiol effects was shown to be minimal. The data obtained prove the complexity of microbial interactions and the regulatory effect of estradiol on commensal bacteria biofilms.
Collapse
|
33
|
Sukul P, Grzegorzewski S, Broderius C, Trefz P, Mittlmeier T, Fischer DC, Miekisch W, Schubert JK. Physiological and metabolic effects of healthy female aging on exhaled breath biomarkers. iScience 2022; 25:103739. [PMID: 35141500 PMCID: PMC8810402 DOI: 10.1016/j.isci.2022.103739] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 11/12/2021] [Accepted: 01/05/2022] [Indexed: 11/30/2022] Open
Abstract
Healthy aging driven physio-metabolic events in females hold the key to complex in vivo mechanistic links and systemic cross talks. Effects from basic changes at genome, proteome, metabolome, and lipidome levels are often reflected at the upstream phenome (e.g., breath volatome) cascades. Here, we have analyzed exhaled volatile metabolites (measured via real time mass spectrometry based breathomics) data from 204 healthy females, aged between 07 and 80 years. Age related substance-specific differences were observed in breath biomarkers. Exhalation of blood-borne endogenous organosulfur, short-chain fatty acids, alcohols, aldehydes, alkene, ketones and exogenous nitriles, terpenes, and aromatics have denominated interplay between endocrine differences, energy homeostasis, systemic microbial diversity, oxidative stress, and lifestyle. Overall marker expressions were suppressed under daily oral contraception. Young homosexual/lesbian adults turned out as breathomic outliers. Previously proposed disease-specific breath biomarkers should be reevaluated upon aging effects. Breathomics offers a noninvasive window toward system-wide understanding and personalized monitoring of aging i.e., translatable to gerontology.
Collapse
Affiliation(s)
- Pritam Sukul
- Rostock Medical Breath Research Analytics and Technologies (ROMBAT), Department of Anesthesiology and Intensive Care, University Medicine Rostock, Schillingallee 35, 18057 Rostock, Germany
| | - Simon Grzegorzewski
- Department of Traumatology, Hand and Reconstructive Surgery, University Medicine Rostock, Schillingallee 35, 18057 Rostock, Germany
| | - Celine Broderius
- Department of Traumatology, Hand and Reconstructive Surgery, University Medicine Rostock, Schillingallee 35, 18057 Rostock, Germany
| | - Phillip Trefz
- Rostock Medical Breath Research Analytics and Technologies (ROMBAT), Department of Anesthesiology and Intensive Care, University Medicine Rostock, Schillingallee 35, 18057 Rostock, Germany
| | - Thomas Mittlmeier
- Department of Traumatology, Hand and Reconstructive Surgery, University Medicine Rostock, Schillingallee 35, 18057 Rostock, Germany
| | - Dagmar-Christiane Fischer
- Department of Pediatrics, University Medicine Rostock, Ernst-Heydemann-Str. 8, 18057 Rostock, Germany
| | - Wolfram Miekisch
- Rostock Medical Breath Research Analytics and Technologies (ROMBAT), Department of Anesthesiology and Intensive Care, University Medicine Rostock, Schillingallee 35, 18057 Rostock, Germany
| | - Jochen K. Schubert
- Rostock Medical Breath Research Analytics and Technologies (ROMBAT), Department of Anesthesiology and Intensive Care, University Medicine Rostock, Schillingallee 35, 18057 Rostock, Germany
| |
Collapse
|
34
|
Sukul P, Bartels J, Fuchs P, Trefz P, Remy R, Rührmund L, Kamysek S, Schubert JK, Miekisch W. Effects of COVID-19 protective face-masks and wearing durations onto respiratory-haemodynamic physiology and exhaled breath constituents. Eur Respir J 2022; 60:13993003.00009-2022. [PMID: 35169028 PMCID: PMC9492982 DOI: 10.1183/13993003.00009-2022] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 02/03/2022] [Indexed: 11/28/2022]
Abstract
Background While assumed to protect against coronavirus transmission, face masks may have effects on respiratory–haemodynamic parameters. Within this pilot study, we investigated immediate and progressive effects of FFP2 and surgical masks on exhaled breath constituents and physiological attributes in 30 adults at rest. Methods We continuously monitored exhaled breath profiles within mask space in older (age 60–80 years) and young to middle-aged (age 20–59 years) adults over the period of 15 and 30 min by high-resolution real-time mass-spectrometry. Peripheral oxygen saturation (SpO2) and respiratory and haemodynamic parameters were measured (noninvasively) simultaneously. Results Profound, consistent and significant (p≤0.001) changes in SpO2 (≥60_FFP2-15 min: 5.8±1.3%↓, ≥60_surgical-15 min: 3.6±0.9%↓, <60_FFP2-30 min: 1.9±1.0%↓, <60_surgical-30 min: 0.9±0.6%↓) and end-tidal carbon dioxide tension (PETCO2) (≥60_FFP2-15 min: 19.1±8.0%↑, ≥60_surgical-15 min: 11.6±7.6%↑, <60_FFP2- 30 min: 12.1±4.5%↑, <60_surgical- 30 min: 9.3±4.1%↑) indicate ascending deoxygenation and hypercarbia. Secondary changes (p≤0.005) to haemodynamic parameters (e.g. mean arterial pressure (MAP) ≥60_FFP2-15 min: 9.8±10.4%↑) were found. Exhalation of bloodborne volatile metabolites, e.g. aldehydes, hemiterpene, organosulfur, short-chain fatty acids, alcohols, ketone, aromatics, nitrile and monoterpene mirrored behaviour of cardiac output, MAP, SpO2, respiratory rate and PETCO2. Exhaled humidity (e.g. ≥60_FFP2-15 min: 7.1±5.8%↑) and exhaled oxygen (e.g. ≥60_FFP2-15 min: 6.1±10.0%↓) changed significantly (p≤0.005) over time. Conclusions Breathomics allows unique physiometabolic insights into immediate and transient effects of face mask wearing. Physiological parameters and breath profiles of endogenous and/or exogenous volatile metabolites indicated putative cross-talk between transient hypoxaemia, oxidative stress, hypercarbia, vasoconstriction, altered systemic microbial activity, energy homeostasis, compartmental storage and washout. FFP2 masks had a more pronounced effect than surgical masks. Older adults were more vulnerable to FFP2 mask-induced hypercarbia, arterial oxygen decline, blood pressure fluctuations and concomitant physiological and metabolic effects. While assumed to protect against SARS-CoV-2 transmission, face masks cause various physiometabolic side-effects and changes in exhaled VOC profiles. Effects are more pronounced in FFP2 masks and are profound at age ≥60 years.https://bit.ly/33fzOMA
Collapse
Affiliation(s)
- Pritam Sukul
- Rostock Medical Breath Research Analytics and Technologies (ROMBAT), Dept. of Anaesthesiology and Intensive Care, University Medicine Rostock, Rostock, Germany
| | - Julia Bartels
- Rostock Medical Breath Research Analytics and Technologies (ROMBAT), Dept. of Anaesthesiology and Intensive Care, University Medicine Rostock, Rostock, Germany
| | - Patricia Fuchs
- Rostock Medical Breath Research Analytics and Technologies (ROMBAT), Dept. of Anaesthesiology and Intensive Care, University Medicine Rostock, Rostock, Germany
| | - Phillip Trefz
- Rostock Medical Breath Research Analytics and Technologies (ROMBAT), Dept. of Anaesthesiology and Intensive Care, University Medicine Rostock, Rostock, Germany
| | - Rasmus Remy
- Rostock Medical Breath Research Analytics and Technologies (ROMBAT), Dept. of Anaesthesiology and Intensive Care, University Medicine Rostock, Rostock, Germany
| | - Leo Rührmund
- Rostock Medical Breath Research Analytics and Technologies (ROMBAT), Dept. of Anaesthesiology and Intensive Care, University Medicine Rostock, Rostock, Germany
| | - Svend Kamysek
- Rostock Medical Breath Research Analytics and Technologies (ROMBAT), Dept. of Anaesthesiology and Intensive Care, University Medicine Rostock, Rostock, Germany
| | - Jochen K Schubert
- Rostock Medical Breath Research Analytics and Technologies (ROMBAT), Dept. of Anaesthesiology and Intensive Care, University Medicine Rostock, Rostock, Germany
| | - Wolfram Miekisch
- Rostock Medical Breath Research Analytics and Technologies (ROMBAT), Dept. of Anaesthesiology and Intensive Care, University Medicine Rostock, Rostock, Germany
| |
Collapse
|
35
|
Gbaoui L, Fachet M, Lüno M, Meyer-Lotz G, Frodl T, Hoeschen C. Breathomics profiling of metabolic pathways affected by major depression: Possibilities and limitations. Front Psychiatry 2022; 13:1061326. [PMID: 36590606 PMCID: PMC9795849 DOI: 10.3389/fpsyt.2022.1061326] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/24/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Major depressive disorder (MDD) is one of the most common psychiatric disorders with multifactorial etiologies. Metabolomics has recently emerged as a particularly potential quantitative tool that provides a multi-parametric signature specific to several mechanisms underlying the heterogeneous pathophysiology of MDD. The main purpose of the present study was to investigate possibilities and limitations of breath-based metabolomics, breathomics patterns to discriminate MDD patients from healthy controls (HCs) and identify the altered metabolic pathways in MDD. METHODS Breath samples were collected in Tedlar bags at awakening, 30 and 60 min after awakening from 26 patients with MDD and 25 HCs. The non-targeted breathomics analysis was carried out by proton transfer reaction mass spectrometry. The univariate analysis was first performed by T-test to rank potential biomarkers. The metabolomic pathway analysis and hierarchical clustering analysis (HCA) were performed to group the significant metabolites involved in the same metabolic pathways or networks. Moreover, a support vector machine (SVM) predictive model was built to identify the potential metabolites in the altered pathways and clusters. The accuracy of the SVM model was evaluated by receiver operating characteristics (ROC) analysis. RESULTS A total of 23 differential exhaled breath metabolites were significantly altered in patients with MDD compared with HCs and mapped in five significant metabolic pathways including aminoacyl-tRNA biosynthesis (p = 0.0055), branched chain amino acids valine, leucine and isoleucine biosynthesis (p = 0.0060), glycolysis and gluconeogenesis (p = 0.0067), nicotinate and nicotinamide metabolism (p = 0.0213) and pyruvate metabolism (p = 0.0440). Moreover, the SVM predictive model showed that butylamine (p = 0.0005, pFDR=0.0006), 3-methylpyridine (p = 0.0002, pFDR = 0.0012), endogenous aliphatic ethanol isotope (p = 0.0073, pFDR = 0.0174), valeric acid (p = 0.005, pFDR = 0.0162) and isoprene (p = 0.038, pFDR = 0.045) were potential metabolites within identified clusters with HCA and altered pathways, and discriminated between patients with MDD and non-depressed ones with high sensitivity (0.88), specificity (0.96) and area under curve of ROC (0.96). CONCLUSION According to the results of this study, the non-targeted breathomics analysis with high-throughput sensitive analytical technologies coupled to advanced computational tools approaches offer completely new insights into peripheral biochemical changes in MDD.
Collapse
Affiliation(s)
- Laila Gbaoui
- Chair of Medical Systems Technology, Institute for Medical Technology, Otto von Guericke University, Magdeburg, Germany
| | - Melanie Fachet
- Chair of Medical Systems Technology, Institute for Medical Technology, Otto von Guericke University, Magdeburg, Germany
| | - Marian Lüno
- Department for Psychiatry and Psychotherapy, Medical Faculty, Otto von Guericke University, Magdeburg, Germany
| | - Gabriele Meyer-Lotz
- Department for Psychiatry and Psychotherapy, Medical Faculty, Otto von Guericke University, Magdeburg, Germany
| | - Thomas Frodl
- Department for Psychiatry and Psychotherapy, Medical Faculty, Otto von Guericke University, Magdeburg, Germany.,Department of Psychiatry, Psychotherapy and Psychosomatics, University Hospital, RWTH Aachen, Aachen, Germany
| | - Christoph Hoeschen
- Chair of Medical Systems Technology, Institute for Medical Technology, Otto von Guericke University, Magdeburg, Germany
| |
Collapse
|
36
|
Roussel C, De Paepe K, Galia W, de Bodt J, Chalancon S, Denis S, Leriche F, Vandekerkove P, Ballet N, Blanquet-Diot S, Van de Wiele T. Multi-targeted properties of the probiotic saccharomyces cerevisiae CNCM I-3856 against enterotoxigenic escherichia coli (ETEC) H10407 pathogenesis across human gut models. Gut Microbes 2021; 13:1953246. [PMID: 34432600 PMCID: PMC8405159 DOI: 10.1080/19490976.2021.1953246] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [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
Enterotoxigenic Escherichia coli (ETEC) is one of the most common causes of acute traveler's diarrhea. Adhesins and enterotoxins constitute the major ETEC virulence traits. With the dramatic increase in antibiotic resistance, probiotics are considered a wholesome alternative to prevent or treat ETEC infections. Here, we examined the antimicrobial properties of the probiotic Saccharomyces cerevisiae CNCM I-3856 against ETEC H10407 pathogenesis upon co-administration in the TNO gastrointestinal Model (TIM-1), simulating the physicochemical and enzymatic conditions of the human upper digestive tract and preventive treatment in the Mucosal Simulator of the Human Intestinal Microbial Ecosystem (M-SHIME), integrating microbial populations of the ileum and ascending colon. Interindividual variability was assessed by separate M-SHIME experiments with microbiota from six human individuals. The probiotic did not affect ETEC survival along the digestive tract. However, ETEC pathogenicity was significantly reduced: enterotoxin encoding virulence genes were repressed, especially in the TIM-1 system, and a lower enterotoxin production was noted. M-SHIME experiments revealed that 18-days probiotic treatment stimulate the growth of Bifidobacterium and Lactobacillus in different gut regions (mucosal and luminal, ileum and ascending colon) while a stronger metabolic activity was noted in terms of short-chain fatty acids (acetate, propionate, and butyrate) and ethanol production. Moreover, the probiotic pre-treated microbiota displayed a higher robustness in composition following ETEC challenge compared to the control condition. We thus demonstrated the multi-inhibitory properties of the probiotic S. cerevisiae CNCM I-3856 against ETEC in the overall simulated human digestive tract, regardless of the inherent variability across individuals in the M-SHIME.
Collapse
Affiliation(s)
- Charlène Roussel
- Université Clermont Auvergne, UMR UCA-INRA 454 MEDIS, Microbiology Digestive Environment and Health, Clermont-Ferrand, France,CMET, Center for Microbial Ecology and Technology, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Kim De Paepe
- CMET, Center for Microbial Ecology and Technology, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Wessam Galia
- UMR 5557 Microbial Ecology, Research Group On Bacterial Opportunistic Pathogens And Environment, CNRS, VetAgro Sup, Lyon, France
| | - Jana de Bodt
- CMET, Center for Microbial Ecology and Technology, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Sandrine Chalancon
- Université Clermont Auvergne, UMR UCA-INRA 454 MEDIS, Microbiology Digestive Environment and Health, Clermont-Ferrand, France
| | - Sylvain Denis
- Université Clermont Auvergne, UMR UCA-INRA 454 MEDIS, Microbiology Digestive Environment and Health, Clermont-Ferrand, France
| | | | | | - Nathalie Ballet
- Lesaffre International, Lesaffre Group, Marcq-en-Baroeul, France
| | - Stéphanie Blanquet-Diot
- CONTACT Stéphanie Blanquet-Diot Université Clermont Auvergne, UMR UCA-INRA 454 MEDIS, Microbiology Digestive Environment and Health, Clermont-Ferrand, France
| | - Tom Van de Wiele
- CMET, Center for Microbial Ecology and Technology, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| |
Collapse
|
37
|
Lactic Acid Bacteria Bacteriocin, an Antimicrobial Peptide Effective Against Multidrug Resistance: a Comprehensive Review. Int J Pept Res Ther 2021. [DOI: 10.1007/s10989-021-10317-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
38
|
Terburgh K, Lindeque JZ, van der Westhuizen FH, Louw R. Cross-comparison of systemic and tissue-specific metabolomes in a mouse model of Leigh syndrome. Metabolomics 2021; 17:101. [PMID: 34792662 DOI: 10.1007/s11306-021-01854-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/03/2021] [Indexed: 01/03/2023]
Abstract
INTRODUCTION The value of metabolomics in multi-systemic mitochondrial disease research has been increasingly recognized, with the ability to investigate a variety of biofluids and tissues considered a particular advantage. Although minimally invasive biofluids are the generally favored sample type, it remains unknown whether systemic metabolomes provide a clear reflection of tissue-specific metabolic alterations. OBJECTIVES Here we cross-compare urine and tissue-specific metabolomes in the Ndufs4 knockout mouse model of Leigh syndrome-a complex neurometabolic MD defined by progressive focal lesions in specific brain regions-to identify and evaluate the extent of common and unique metabolic alterations on a systemic and brain regional level. METHODS Untargeted and semi-targeted multi-platform metabolomics were performed on urine, four brain regions, and two muscle types of Ndufs4 KO (n≥19) vs wildtype (n≥20) mice. RESULTS Widespread alterations were evident in alanine, aspartate, glutamate, and arginine metabolism in Ndufs4 KO mice; while brain-region specific metabolic signatures include the accumulation of branched-chain amino acids, proline, and glycolytic intermediates. Furthermore, we describe a systemic dysregulation in one-carbon metabolism and the tricarboxylic acid cycle, which was not clearly reflected in the Ndufs4 KO brain. CONCLUSION Our results confirm the value of urinary metabolomics when evaluating MD-associated metabolites, while cautioning against mechanistic studies relying solely on systemic biofluids.
Collapse
Affiliation(s)
- Karin Terburgh
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University (Potchefstroom Campus), Private Bag X6001, Potchefstroom, South Africa
| | - Jeremie Z Lindeque
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University (Potchefstroom Campus), Private Bag X6001, Potchefstroom, South Africa
| | - Francois H van der Westhuizen
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University (Potchefstroom Campus), Private Bag X6001, Potchefstroom, South Africa
| | - Roan Louw
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University (Potchefstroom Campus), Private Bag X6001, Potchefstroom, South Africa.
| |
Collapse
|
39
|
Engevik M, Ruan W, Visuthranukul C, Shi Z, Engevik KA, Engevik AC, Fultz R, Schady DA, Spinler JK, Versalovic J. Limosilactobacillus reuteri ATCC 6475 metabolites upregulate the serotonin transporter in the intestinal epithelium. Benef Microbes 2021; 12:583-599. [PMID: 34550056 DOI: 10.3920/bm2020.0216] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The serotonin transporter (SERT) readily takes up serotonin (5-HT), thereby regulating the availability of 5-HT within the intestine. In the absence of SERT, 5-HT remains in the interstitial space and has the potential to aberrantly activate the many 5-HT receptors distributed on the epithelium, immune cells and enteric neurons. Perturbation of SERT is common in many gastrointestinal disorders as well as mouse models of colitis. Select commensal microbes regulate intestinal SERT levels, but the mechanism of this regulation is poorly understood. Additionally, ethanol upregulates SERT in the brain and dendritic cells, but its effects in the intestine have never been examined. We report that the intestinal commensal microbe Limosilactobacillus (previously classified as Lactobacillus) reuteri ATCC PTA 6475 secretes 83.4 mM ethanol. Consistent with the activity of L. reuteri alcohol dehydrogenases, we found that L. reuteri tolerated various levels of ethanol. Application of L. reuteri conditioned media or exogenous ethanol to human colonic T84 cells was found to upregulate SERT at the level of mRNA. A 4-(4-(dimethylamino) phenyl)-1-methylpyridinium (APP+) uptake assay confirmed the functional activity of SERT. These findings were mirrored in mouse colonic organoids, where L. reuteri metabolites and ethanol were found to upregulate SERT at the apical membrane. Finally, in a trinitrobenzene sulphonic acid model of acute colitis, we observed that mice treated with L. reuteri maintained SERT at the colon membrane compared with mice receiving phosphate buffered saline vehicle control. These data suggest that L. reuteri metabolites, including ethanol, can upregulate SERT and may be beneficial for maintaining intestinal homeostasis with respect to serotonin signalling.
Collapse
Affiliation(s)
- M Engevik
- Department of Pathology & Immunology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
- Department of Regenerative Medicine & Cell Biology, Medical University of South Carolina, 173 Ashely Ave, BSB 626, Charleston, SC 29425, USA
| | - W Ruan
- Department of Pediatrics, Baylor College of Medicine, 6701 Fannin Street, Houston, TX 77030, USA
- Section of Gastroenterology, Hepatology, and Nutrition, Texas Children's Hospital, 6701 Fannin St, Houston, TX 77030, USA
| | - C Visuthranukul
- Department of Pathology & Immunology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
- Pediatric Nutrition Research Unit, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Z Shi
- Department of Pathology & Immunology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
- Department of Pathology, Texas Children's Hospital, 6621 Fannin St, Houston, TX 77030, USA
| | - K A Engevik
- Department of Molecular Virology & Microbiology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 7703, USA
| | - A C Engevik
- Departments of Surgery, Vanderbilt University Medical Center, 1211 Medical Center Dr, Nashville, TN 37232, USA
| | - R Fultz
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0625, USA
| | - D A Schady
- Department of Pathology & Immunology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
- Department of Pathology, Texas Children's Hospital, 6621 Fannin St, Houston, TX 77030, USA
| | - J K Spinler
- Department of Pathology & Immunology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
- Department of Pathology, Texas Children's Hospital, 6621 Fannin St, Houston, TX 77030, USA
| | - J Versalovic
- Department of Pathology & Immunology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
- Department of Pathology, Texas Children's Hospital, 6621 Fannin St, Houston, TX 77030, USA
| |
Collapse
|
40
|
Sturgess C, Montgomery H. Selection pressure at altitude for genes related to alcohol metabolism: A role for endogenous enteric ethanol synthesis? Exp Physiol 2021; 106:2155-2167. [PMID: 34487385 DOI: 10.1113/ep089628] [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: 04/16/2021] [Accepted: 08/31/2021] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the topic of this review? Highland natives have undergone natural selection for genetic variants advantageous in adaptation to the hypobaric hypoxia experienced at high altitude. Why genes related to alcohol metabolism appear consistently selected for has not been greatly considered. We hypothesize that altitude-related changes in the gut microbiome offer one possible explanation. What advances does it highlight? Low intestinal oxygen tension might favour the production of ethanol through anaerobic fermentation by the gut microbiome. Subsequent increases in endogenous ethanol absorption could therefore provide a selection pressure for gene variants favouring its increased degradation, or perhaps reduced degradation if endogenously synthesized ethanol acts as a metabolic signalling molecule. ABSTRACT Reduced tissue availability of oxygen results from ascent to high altitude, where atmospheric pressure, and thus the partial pressure of inspired oxygen, fall (hypobaric hypoxia). In humans, adaptation to such hypoxia is necessary for survival. These functional changes remain incompletely characterized, although metabolic adaptation (rather than simple increases in convective oxygen delivery) appears to play a fundamental role. Those populations that have remained native to high altitude have undergone natural selection for genetic variants associated with advantageous phenotypic traits. Interestingly, a consistent genetic signal has implicated alcohol metabolism in the human adaptive response to hypobaric hypoxia. The reasons for this remain unclear. One possibility is that increased alcohol synthesis occurs through fermentation by gut bacteria in response to enteric hypoxia. There is growing evidence that anaerobes capable of producing ethanol become increasingly prevalent with high-altitude exposure. We hypothesize that: (1) ascent to high altitude renders the gut luminal environment increasingly hypoxic, favouring (2) an increase in the population of enteric fermenting anaerobes, hence (3) the synthesis of alcohol which, through systemic absorption, leads to (4) selection pressure on genes relating to alcohol metabolism. In theory, alcohol could be viewed as a toxic product, leading to selection of gene variants favouring its metabolism. On the contrary, alcohol is a metabolic substrate that might be beneficial. This mechanism could also account for some of the interindividual differences of lowlanders in acclimatization to altitude. Future research should be aimed at determining any shifts to favour ethanol-producing anaerobes after ascent to altitude.
Collapse
Affiliation(s)
- Connie Sturgess
- Institute for Human Health and Performance, Department of Medicine, University College London, London, UK
| | - Hugh Montgomery
- Institute for Human Health and Performance, Department of Medicine, University College London, London, UK
| |
Collapse
|
41
|
Ansari A, Bose S, You Y, Park S, Kim Y. Molecular Mechanism of Microbiota Metabolites in Preterm Birth: Pathological and Therapeutic Insights. Int J Mol Sci 2021; 22:8145. [PMID: 34360908 PMCID: PMC8347546 DOI: 10.3390/ijms22158145] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 12/11/2022] Open
Abstract
Preterm birth (PTB) refers to the birth of infants before 37 weeks of gestation and is a challenging issue worldwide. Evidence reveals that PTB is a multifactorial dysregulation mediated by a complex molecular mechanism. Thus, a better understanding of the complex molecular mechanisms underlying PTB is a prerequisite to explore effective therapeutic approaches. During early pregnancy, various physiological and metabolic changes occur as a result of endocrine and immune metabolism. The microbiota controls the physiological and metabolic mechanism of the host homeostasis, and dysbiosis of maternal microbial homeostasis dysregulates the mechanistic of fetal developmental processes and directly affects the birth outcome. Accumulating evidence indicates that metabolic dysregulation in the maternal or fetal membranes stimulates the inflammatory cytokines, which may positively progress the PTB. Although labour is regarded as an inflammatory process, it is still unclear how microbial dysbiosis could regulate the molecular mechanism of PTB. In this review based on recent research, we focused on both the pathological and therapeutic contribution of microbiota-generated metabolites to PTB and the possible molecular mechanisms.
Collapse
Affiliation(s)
- AbuZar Ansari
- Department of Obstetrics and Gynecology, Ewha Medical Research Institute, College of Medicine, Ewha Womans University, Mokdong Hospital, Seoul 07985, Korea; (A.A.); (Y.Y.); (S.P.)
| | - Shambhunath Bose
- Department of Bioscience, Sri Sathya Sai University for Human Excellence, Navanihal, Okali Post, Kamalapur, Kalaburagi, Karnataka 585313, India;
| | - Youngah You
- Department of Obstetrics and Gynecology, Ewha Medical Research Institute, College of Medicine, Ewha Womans University, Mokdong Hospital, Seoul 07985, Korea; (A.A.); (Y.Y.); (S.P.)
| | - Sunwha Park
- Department of Obstetrics and Gynecology, Ewha Medical Research Institute, College of Medicine, Ewha Womans University, Mokdong Hospital, Seoul 07985, Korea; (A.A.); (Y.Y.); (S.P.)
| | - Youngju Kim
- Department of Obstetrics and Gynecology, Ewha Medical Research Institute, College of Medicine, Ewha Womans University, Mokdong Hospital, Seoul 07985, Korea; (A.A.); (Y.Y.); (S.P.)
| |
Collapse
|
42
|
Wu LL, Huang TS, Shyu YC, Wang CL, Wang HY, Chen PJ. Gut microbiota in the innate immunity against hepatitis B virus - implication in age-dependent HBV clearance. Curr Opin Virol 2021; 49:194-202. [PMID: 34242953 DOI: 10.1016/j.coviro.2021.06.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 02/08/2023]
Abstract
Hepatitis B virus (HBV) chronically infects 257 million people and is one of the most important liver diseases worldwide. A unique feature of HBV infection in humans is that viral clearance heavily depends on the age at exposure. Recent studies demonstrated that the virus takes advantage of immature innate immunity, especially hepatic macrophages, and not-yet-stabilized gut microbiota in early life to establish a chronic infection. The liver contains resident and infiltrating myeloid cells involved in immune responses to pathogens. They influence both innate and adaptive sectors of the immune system and their interplay with HBV has only been noticed recently. Here, we discuss how interactions between gut microbiota and hepatic macrophages influence the outcomes of HBV infection. Understanding the underlying mechanism would pave the way for the treatment of chronic HBV infection.
Collapse
Affiliation(s)
- Li-Ling Wu
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taiwan
| | - Ting-Shuo Huang
- Department of General Surgery, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan; Community Medicine Research Center, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Yu-Chiau Shyu
- Community Medicine Research Center, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan; Department of Nursing, Chang Gung University of Science and Technology, Taoyuan City, Taiwan; Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Chih-Lin Wang
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine and National Taiwan University Hospital, Taipei, Taiwan
| | - Hurng-Yi Wang
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine and National Taiwan University Hospital, Taipei, Taiwan; Institute of Ecology and Evolutionary Biology, National Taiwan University College of Medicine and National Taiwan University Hospital, Taipei, Taiwan; Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine and National Taiwan University Hospital, Taipei, Taiwan.
| | - Pei-Jer Chen
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine and National Taiwan University Hospital, Taipei, Taiwan; Hepatitis Research Center, National Taiwan University College of Medicine and National Taiwan University Hospital, Taipei, Taiwan; Department of Microbiology, National Taiwan University College of Medicine and National Taiwan University Hospital, Taipei, Taiwan; Department of Internal Medicine, National Taiwan University College of Medicine and National Taiwan University Hospital, Taipei, Taiwan; Department of Medical Research, National Taiwan University College of Medicine and National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
43
|
Fernandez-Cantos MV, Garcia-Morena D, Iannone V, El-Nezami H, Kolehmainen M, Kuipers OP. Role of microbiota and related metabolites in gastrointestinal tract barrier function in NAFLD. Tissue Barriers 2021; 9:1879719. [PMID: 34280073 PMCID: PMC8489918 DOI: 10.1080/21688370.2021.1879719] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/13/2021] [Accepted: 01/18/2021] [Indexed: 11/06/2022] Open
Abstract
The Gastrointestinal (GI) tract is composed of four main barriers: microbiological, chemical, physical and immunological. These barriers play important roles in maintaining GI tract homeostasis. In the crosstalk between these barriers, microbiota and related metabolites have been shown to influence GI tract barrier integrity, and alterations of the gut microbiome might lead to an increase in intestinal permeability. As a consequence, translocation of bacteria and their products into the circulatory system increases, reaching proximal and distal tissues, such as the liver. One of the most prevalent chronic liver diseases, Nonalcoholic Fatty Liver Disease (NAFLD), has been associated with an altered gut microbiota and barrier integrity. However, the causal link between them has not been fully elucidated yet. In this review, we aim to highlight relevant bacterial taxa and their related metabolites affecting the GI tract barriers in the context of NAFLD, discussing their implications in gut homeostasis and in disease.
Collapse
Affiliation(s)
- Maria Victoria Fernandez-Cantos
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
| | - Diego Garcia-Morena
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
| | - Valeria Iannone
- Institute of Public Health and Clinical Nutrition, Department of Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Hani El-Nezami
- Molecular and Cell Biology Division, School of Biological Sciences, University of Hong Kong, Hong Kong SAR
| | - Marjukka Kolehmainen
- Institute of Public Health and Clinical Nutrition, Department of Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Oscar P. Kuipers
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
44
|
Elshaghabee FMF, Abd El-Maksoud AA, Alharbi SA, Alfarraj S, Mohamed MSM. Fortification of Acidophilus- bifidus- thermophilus (ABT) Fermented Milk with Heat-Treated Industrial Yeast Enhances Its Selected Properties. Molecules 2021; 26:3876. [PMID: 34201949 PMCID: PMC8271856 DOI: 10.3390/molecules26133876] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/18/2021] [Accepted: 06/19/2021] [Indexed: 02/03/2023] Open
Abstract
The improvement of milk dairy products' quality and nutritional value during shelf-life storage is the ultimate goal of many studies worldwide. Therefore, in the present study, prospective beneficial effects of adding two different industrial yeasts, Kluyveromyces lactis and Saccharomyces cerevisiae pretreated by heating at 85 °C for 10 min to be inactivated, before fermentation on some properties of ABT fermented milk were evaluated. The results of this study showed that the addition of 3% and 5% (w/v) heat-treated yeasts to the milk enhanced the growth of starter culture, Lactobacillus acidophilus, Bifidobacteria, and Streptococcus thermophilus, during the fermentation period as well as its viability after 20 days of cold storage at 5 ± 1 °C. Furthermore, levels of lactic and acetic acids were significantly increased from 120.45 ± 0.65 and 457.80 ± 0.70 µg/mL in the control without heat-treated yeast to 145.67 ± 0.77 and 488.32 ± 0.33 µg/mL with 5% supplementation of Sacch. cerevisiae respectively. Moreover, the addition of heat-treated yeasts to ABT fermented milk enhanced the antioxidant capacity by increasing the efficiency of free radical scavenging as well as the proteolytic activity. Taken together, these results suggest promising application of non-viable industrial yeasts as nutrients in the fermentation process of ABT milk to enhance the growth and viability of ABT starter cultures before and after a 20-day cold storage period by improving the fermented milk level of organic acids, antioxidant capacity, and proteolytic activities.
Collapse
Affiliation(s)
| | | | - Sulaiman Ali Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia;
| | - Saleh Alfarraj
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Mahmoud S. M. Mohamed
- Department of Botany and Microbiology, Faculty of Science, Cairo University, Giza 12613, Egypt
| |
Collapse
|
45
|
Translational Approaches with Antioxidant Phytochemicals against Alcohol-Mediated Oxidative Stress, Gut Dysbiosis, Intestinal Barrier Dysfunction, and Fatty Liver Disease. Antioxidants (Basel) 2021; 10:antiox10030384. [PMID: 33806556 PMCID: PMC8000766 DOI: 10.3390/antiox10030384] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/25/2021] [Accepted: 02/25/2021] [Indexed: 12/12/2022] Open
Abstract
Emerging data demonstrate the important roles of altered gut microbiomes (dysbiosis) in many disease states in the peripheral tissues and the central nervous system. Gut dysbiosis with decreased ratios of Bacteroidetes/Firmicutes and other changes are reported to be caused by many disease states and various environmental factors, such as ethanol (e.g., alcohol drinking), Western-style high-fat diets, high fructose, etc. It is also caused by genetic factors, including genetic polymorphisms and epigenetic changes in different individuals. Gut dysbiosis, impaired intestinal barrier function, and elevated serum endotoxin levels can be observed in human patients and/or experimental rodent models exposed to these factors or with certain disease states. However, gut dysbiosis and leaky gut can be normalized through lifestyle alterations such as increased consumption of healthy diets with various fruits and vegetables containing many different kinds of antioxidant phytochemicals. In this review, we describe the mechanisms of gut dysbiosis, leaky gut, endotoxemia, and fatty liver disease with a specific focus on the alcohol-associated pathways. We also mention translational approaches by discussing the benefits of many antioxidant phytochemicals and/or their metabolites against alcohol-mediated oxidative stress, gut dysbiosis, intestinal barrier dysfunction, and fatty liver disease.
Collapse
|
46
|
Yu SY, Xu L. The interplay between host cellular and gut microbial metabolism in NAFLD development and prevention. J Appl Microbiol 2021; 131:564-582. [PMID: 33411984 DOI: 10.1111/jam.14992] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/27/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022]
Abstract
Metabolism regulation centred on insulin resistance is increasingly important in nonalcoholic fatty liver disease (NAFLD). This review focuses on the interactions between the host cellular and gut microbial metabolism during the development of NAFLD. The cellular metabolism of essential nutrients, such as glucose, lipids and amino acids, is reconstructed with inflammation, immune mechanisms and oxidative stress, and these alterations modify the intestinal, hepatic and systemic environments, and regulate the composition and activity of gut microbes. Microbial metabolites, such as short-chain fatty acids, secondary bile acids, protein fermentation products, choline and ethanol and bacterial toxicants, such as lipopolysaccharides, peptidoglycans and bacterial DNA, play vital roles in NAFLD. The microbe-metabolite relationship is crucial for the modulation of intestinal microbial composition and metabolic activity. The intestinal microbiota and their metabolites participate in epithelial cell metabolism via a series of cell receptors and signalling pathways and remodel the metabolism of various cells in the liver via the gut-liver axis. Microbial metabolic manipulation is a promising strategy for NAFLD prevention, but larger-sampled clinical trials are required for future application.
Collapse
Affiliation(s)
- S-Y Yu
- Department of Gastroenterology, Ningbo First Hospital, Ningbo, China
| | - L Xu
- Department of Gastroenterology, Ningbo First Hospital, Ningbo, China
| |
Collapse
|
47
|
Xie X, Zhang L, Yuan S, Li H, Zheng C, Xie S, Sun Y, Zhang C, Wang R, Jin Y. Val-Val-Tyr-Pro protects against non-alcoholic steatohepatitis in mice by modulating the gut microbiota and gut-liver axis activation. J Cell Mol Med 2021; 25:1439-1455. [PMID: 33400402 PMCID: PMC7875918 DOI: 10.1111/jcmm.16229] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 12/06/2020] [Accepted: 12/11/2020] [Indexed: 12/13/2022] Open
Abstract
Val‐Val‐Tyr‐Pro (VVYP) peptide is one of the main active components of Globin digest (GD). Our previous studies indicated that VVYP could protect against acetaminophen and carbon tetrachloride‐induced acute liver failure in mice and decrease blood lipid level. However, the effects and underlying mechanisms of VVYP in the treatment of non‐alcoholic steatohepatitis (NASH) have not been discovered. Our present study was designed to investigate the preventive effect of VVYP on NASH and its underlying specific mechanisms. We found that VVYP inhibited the cytotoxicity and lipid accumulation in L‐02 cells that were exposed to a mixture of free fatty acid (FFA). VVYP effectively alleviated the liver injury induced by methionine‐choline‐deficient (MCD) diet, demonstrated by reducing the levels of serum alanine aminotransferase (ALT)/aspartate aminotransferase (AST)/triglycerides (TG)/non‐esterified fatty acids (NEFA) and improving liver histology. VVYP decreased expression levels of lipid synthesis‐related genes and reduced levels of the proinflammation cytokines in the liver of mice fed by MCD diet. Moreover, VVYP inhibited the increased level of LPS and reversed the liver mitochondria dysfunction induced by MCD diet. Meanwhile, VVYP significantly increased the abundance of beneficial bacteria such as Eubacteriaceae, coriobacteriacease, Desulfovibrionaceae, S24‐7 and Bacteroidia in high‐fat diet (HFD)‐fed mice, however, VVYP reduced the abundance of Lactobacillus. Moreover, VVYP conferred the protective effect of intestinal barrier via promoting the expression of the mucins and tight junction (TJ)‐associated genes and inhibited subsequent liver inflammatory responses. These results indicated that the protective role of VVYP on NASH is mediated by modulating gut microbiota imbalance and related gut‐liver axis activation. VVYP might be a promising drug candidate for NASH.
Collapse
Affiliation(s)
- Xinshu Xie
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Lang Zhang
- Jiangxi Provincial Children's Hospital, Nanchang, China
| | - Shun Yuan
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Huilan Li
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Chaojun Zheng
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Saisai Xie
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Yongbing Sun
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Changhua Zhang
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Rikang Wang
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China.,Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Basic Medical Sciences, Shenzhen University Health Science Centre, Shenzhen, China
| | - Yi Jin
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| |
Collapse
|
48
|
Kaur J, Singh BP, Chaudhary V, Elshaghabee FMF, Singh J, Singh A, Rokana N, Panwar H. Probiotics as Live Bio-therapeutics: Prospects and Perspectives. MICROORGANISMS FOR SUSTAINABILITY 2021:83-120. [DOI: 10.1007/978-981-15-6795-7_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
49
|
Moffett JR, Puthillathu N, Vengilote R, Jaworski DM, Namboodiri AM. Acetate Revisited: A Key Biomolecule at the Nexus of Metabolism, Epigenetics, and Oncogenesis - Part 2: Acetate and ACSS2 in Health and Disease. Front Physiol 2020; 11:580171. [PMID: 33304273 PMCID: PMC7693462 DOI: 10.3389/fphys.2020.580171] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 10/19/2020] [Indexed: 12/19/2022] Open
Abstract
Acetate, the shortest chain fatty acid, has been implicated in providing health benefits whether it is derived from the diet or is generated from microbial fermentation of fiber in the gut. These health benefits range widely from improved cardiac function to enhanced red blood cell generation and memory formation. Understanding how acetate could influence so many disparate biological functions is now an area of intensive research. Protein acetylation is one of the most common post-translational modifications and increased systemic acetate strongly drives protein acetylation. By virtue of acetylation impacting the activity of virtually every class of protein, acetate driven alterations in signaling and gene transcription have been associated with several common human diseases, including cancer. In part 2 of this review, we will focus on some of the roles that acetate plays in health and human disease. The acetate-activating enzyme acyl-CoA short-chain synthetase family member 2 (ACSS2) will be a major part of that focus due to its role in targeted protein acetylation reactions that can regulate central metabolism and stress responses. ACSS2 is the only known enzyme that can recycle acetate derived from deacetylation reactions in the cytoplasm and nucleus of cells, including both protein and metabolite deacetylation reactions. As such, ACSS2 can recycle acetate derived from histone deacetylase reactions as well as protein deacetylation reactions mediated by sirtuins, among many others. Notably, ACSS2 can activate acetate released from acetylated metabolites including N-acetylaspartate (NAA), the most concentrated acetylated metabolite in the human brain. NAA has been associated with the metabolic reprograming of cancer cells, where ACSS2 also plays a role. Here, we discuss the context-specific roles that acetate can play in health and disease.
Collapse
Affiliation(s)
- John R. Moffett
- Department of Anatomy, Physiology and Genetics, and Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Narayanan Puthillathu
- Department of Anatomy, Physiology and Genetics, and Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Ranjini Vengilote
- Department of Anatomy, Physiology and Genetics, and Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Diane M. Jaworski
- Department of Neurological Sciences, University of Vermont College of Medicine, Burlington, VT, United States
| | - Aryan M. Namboodiri
- Department of Anatomy, Physiology and Genetics, and Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
50
|
Mechanisms controlling bacterial infection in myeloid cells under hypoxic conditions. Cell Mol Life Sci 2020; 78:1887-1907. [PMID: 33125509 PMCID: PMC7966188 DOI: 10.1007/s00018-020-03684-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 09/08/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022]
Abstract
Various factors of the tissue microenvironment such as the oxygen concentration influence the host-pathogen interaction. During the past decade, hypoxia-driven signaling via hypoxia-inducible factors (HIF) has emerged as an important factor that affects both the pathogen and the host. In this chapter, we will review the current knowledge of this complex interplay, with a particular emphasis given to the impact of hypoxia and HIF on the inflammatory and antimicrobial activity of myeloid cells, the bacterial responses to hypoxia and the containment of bacterial infections under oxygen-limited conditions. We will also summarize how low oxygen concentrations influence the metabolism of neutrophils, macrophages and dendritic cells. Finally, we will discuss the consequences of hypoxia and HIFα activation for the invading pathogen, with a focus on Pseudomonas aeruginosa, Mycobacterium tuberculosis, Coxiella burnetii, Salmonella enterica and Staphylococcus aureus. This includes a description of the mechanisms and microbial factors, which the pathogens use to sense and react to hypoxic conditions.
Collapse
|