1
|
Schöneberg T. Modulating vertebrate physiology by genomic fine-tuning of GPCR functions. Physiol Rev 2025; 105:383-439. [PMID: 39052017 DOI: 10.1152/physrev.00017.2024] [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: 04/22/2024] [Revised: 07/08/2024] [Accepted: 07/20/2024] [Indexed: 07/27/2024] Open
Abstract
G protein-coupled receptors (GPCRs) play a crucial role as membrane receptors, facilitating the communication of eukaryotic species with their environment and regulating cellular and organ interactions. Consequently, GPCRs hold immense potential in contributing to adaptation to ecological niches and responding to environmental shifts. Comparative analyses of vertebrate genomes reveal patterns of GPCR gene loss, expansion, and signatures of selection. Integrating these genomic data with insights from functional analyses of gene variants enables the interpretation of genotype-phenotype correlations. This review underscores the involvement of GPCRs in adaptive processes, presenting numerous examples of how alterations in GPCR functionality influence vertebrate physiology or, conversely, how environmental changes impact GPCR functions. The findings demonstrate that modifications in GPCR function contribute to adapting to aquatic, arid, and nocturnal habitats, influencing camouflage strategies, and specializing in particular dietary preferences. Furthermore, the adaptability of GPCR functions provides an effective mechanism in facilitating past, recent, or ongoing adaptations in animal domestication and human evolution and should be considered in therapeutic strategies and drug development.
Collapse
Affiliation(s)
- Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, University of Leipzig, Leipzig, Germany
- School of Medicine, University of Global Health Equity, Kigali, Rwanda
| |
Collapse
|
2
|
Kozakai T, Nakajima A, Miyazawa K, Sasaki Y, Odamaki T, Katoh T, Fukuma T, Xiao JZ, Suzuki T, Katayama T, Sakanaka M. An improved temperature-sensitive shuttle vector system for scarless gene deletion in human-gut-associated Bifidobacterium species. iScience 2024; 27:111080. [PMID: 39502284 PMCID: PMC11536034 DOI: 10.1016/j.isci.2024.111080] [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: 04/01/2024] [Revised: 08/27/2024] [Accepted: 09/27/2024] [Indexed: 11/08/2024] Open
Abstract
Bifidobacterium is a prevalent bacterial taxon in the human gut that comprises over 10 (sub)species. Previous studies suggest that these species use evolutionarily distinct strategies for symbiosis with their hosts. However, the underlying species-specific mechanisms remain unclear due to the lack of efficient gene knockout systems applicable across different species. Here, we developed improved temperature-sensitive shuttle vectors by introducing Ser139Trp into the replication protein RepB. We then used temperature-sensitive plasmids to construct a double-crossover-mediated scarless gene deletion system. The system was employed for targeted gene deletion in Bifidobacterium longum subsp. longum, B. longum subsp. infantis, Bifidobacterium breve, Bifidobacterium adolescentis, Bifidobacterium kashiwanohense, and Bifidobacterium pseudocatenulatum. Deletion of genes involved in capsular polysaccharide biosynthesis, aromatic lactic acid production, and sugar utilization resulted in the expected phenotypic changes in the respective (sub)species. The temperature-sensitive plasmids developed in this study will aid in deciphering the evolutionary traits of the human-gut-associated Bifidobacterium species.
Collapse
Affiliation(s)
- Tomoya Kozakai
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Aruto Nakajima
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Keisuke Miyazawa
- Faculty of Frontier Engineering, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
- WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Yuki Sasaki
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Toshitaka Odamaki
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
- Innovative Research Institute, R&D Division, Morinaga Milk Industry Co. Ltd., Zama, Kanagawa 252-8583, Japan
| | - Toshihiko Katoh
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Takeshi Fukuma
- Faculty of Frontier Engineering, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
- WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Jin-zhong Xiao
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
- Innovative Research Institute, R&D Division, Morinaga Milk Industry Co. Ltd., Zama, Kanagawa 252-8583, Japan
| | - Tohru Suzuki
- Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193, Japan
| | - Takane Katayama
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Mikiyasu Sakanaka
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| |
Collapse
|
3
|
Kwoji ID, Okpeku M, Aiyegoro OA, Adeleke MA. Metabolic interactions of Limosilactobacillus reuteri ZJ625 and Ligilactobacillus salivarius ZJ614 in co-culture: implications for multi-strain probiotics. J Appl Microbiol 2024; 135:lxae264. [PMID: 39510973 DOI: 10.1093/jambio/lxae264] [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: 02/18/2024] [Revised: 08/31/2024] [Accepted: 11/05/2024] [Indexed: 11/15/2024]
Abstract
AIMS Limosilactobacillus reuteri ZJ625 and Ligilactobacillus salivarius ZJ614 are potential probiotic bacteria. The mechanisms of enhanced benefits by muti-strain probiotics are yet fully understood. We elucidated the influence of co-culturing on the metabolite profiles of Limosilactobacillus reuteri ZJ625 and Ligilactobacillus salivarius ZJ614 to decipher the impacts of co-culturing on metabolic interactions between the strains. METHODS AND RESULTS Limosilactobacillus reuteri ZJ625 and Ligilactobacillus salivarius ZJ614 were grown in single and co-cultures in defined media. Bacterial cell metabolites were extracted at the mid-stationary growth phase and analysed using two-dimensional gas chromatography-time-of-flight mass spectrometry (GC × GC-TOFMS). Mass-spectral data were preprocessed and analysed using unsupervised and supervised methods based on the group allocations. A total of 1387 metabolites were identified, with 18.31% significant metabolites (P < 0.05) and 10.17% differential metabolites (P < 0.05, variable importance on projection > 1). The differential metabolites identified include arabinofuranose, methyl-galactoside, N-acetylglutamic acid, phosphoric acid, and decanoic acid. The metabolites impacted carbohydrate and amino-sugar metabolism. CONCLUSION Co-culturing of Limosilactobacillus reuteri ZJ625 and Ligilactobacillus salivarius ZJ614 influenced the metabolite profiles of the strains and impacted metabolic/biosynthetic pathways, indicating cell-to-cell interactions between the strains.
Collapse
Affiliation(s)
- Iliya Dauda Kwoji
- Discipline of Genetics, School of Life Sciences, College of Agriculture, Engineering and Sciences, University of KwaZulu-Natal, 4090 Durban, South Africa
| | - Moses Okpeku
- Discipline of Genetics, School of Life Sciences, College of Agriculture, Engineering and Sciences, University of KwaZulu-Natal, 4090 Durban, South Africa
| | - Olayinka Ayobami Aiyegoro
- Unit for Environmental Sciences and Management, Northwest University, Potchefstroom, Northwest 2520, South Africa
| | - Matthew Adekunle Adeleke
- Discipline of Genetics, School of Life Sciences, College of Agriculture, Engineering and Sciences, University of KwaZulu-Natal, 4090 Durban, South Africa
| |
Collapse
|
4
|
Kasperek MC, Velasquez Galeas A, Caetano-Silva ME, Xie Z, Ulanov A, La Frano M, Devkota S, Miller MJ, Allen JM. Microbial aromatic amino acid metabolism is modifiable in fermented food matrices to promote bioactivity. Food Chem 2024; 454:139798. [PMID: 38823201 DOI: 10.1016/j.foodchem.2024.139798] [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: 02/09/2024] [Revised: 04/17/2024] [Accepted: 05/20/2024] [Indexed: 06/03/2024]
Abstract
Ingestion of fermented foods impacts human immune function, yet the bioactive food components underlying these effects are not understood. Here, we interrogated whether fermented food bioactivity relates to microbial metabolites derived from aromatic amino acids, termed aryl-lactates. Using targeted metabolomics, we established the presence of aryl-lactates in commercially available fermented foods. After pinpointing fermented food-associated lactic acid bacteria that produce high levels of aryl-lactates, we identified fermentation conditions to increase aryl-lactate production in food matrices up to 5 × 103 fold vs. standard fermentation conditions. Using ex vivo reporter assays, we found that food matrix conditions optimized for aryl-lactate production exhibited enhanced agonist activity for the human aryl-hydrocarbon receptor (AhR) as compared to standard fermentation conditions and commercial products. Reduced microbial-induced AhR activity has emerged as a hallmark of many chronic inflammatory diseases, thus we envision strategies to enhance AhR bioactivity of fermented foods to be leveraged to improve human health.
Collapse
Affiliation(s)
- Mikaela C Kasperek
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| | - Adriana Velasquez Galeas
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| | - Maria Elisa Caetano-Silva
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Department of Health and Kinesiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| | - Zifan Xie
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| | - Alexander Ulanov
- Carver Metabolomics Core, Roy J. Carver Biotechnology Center, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| | - Michael La Frano
- Carver Metabolomics Core, Roy J. Carver Biotechnology Center, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| | - Suzanne Devkota
- Human Microbiome Research Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA.
| | - Michael J Miller
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| | - Jacob M Allen
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Department of Health and Kinesiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
5
|
Chen M, Xia L, Wu C, Wang Z, Ding L, Xie Y, Feng W, Chen Y. Microbe-material hybrids for therapeutic applications. Chem Soc Rev 2024; 53:8306-8378. [PMID: 39005165 DOI: 10.1039/d3cs00655g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
As natural living substances, microorganisms have emerged as useful resources in medicine for creating microbe-material hybrids ranging from nano to macro dimensions. The engineering of microbe-involved nanomedicine capitalizes on the distinctive physiological attributes of microbes, particularly their intrinsic "living" properties such as hypoxia tendency and oxygen production capabilities. Exploiting these remarkable characteristics in combination with other functional materials or molecules enables synergistic enhancements that hold tremendous promise for improved drug delivery, site-specific therapy, and enhanced monitoring of treatment outcomes, presenting substantial opportunities for amplifying the efficacy of disease treatments. This comprehensive review outlines the microorganisms and microbial derivatives used in biomedicine and their specific advantages for therapeutic application. In addition, we delineate the fundamental strategies and mechanisms employed for constructing microbe-material hybrids. The diverse biomedical applications of the constructed microbe-material hybrids, encompassing bioimaging, anti-tumor, anti-bacteria, anti-inflammation and other diseases therapy are exhaustively illustrated. We also discuss the current challenges and prospects associated with the clinical translation of microbe-material hybrid platforms. Therefore, the unique versatility and potential exhibited by microbe-material hybrids position them as promising candidates for the development of next-generation nanomedicine and biomaterials with unique theranostic properties and functionalities.
Collapse
Affiliation(s)
- Meng Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai 200444, P. R. China.
| | - Lili Xia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Chenyao Wu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Zeyu Wang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Li Ding
- Department of Medical Ultrasound, National Clinical Research Center of Interventional Medicine, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Tongji University, Shanghai, 200072, P. R. China.
| | - Yujie Xie
- School of Medicine, Shanghai University, Shanghai 200444, P. R. China.
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
- Shanghai Institute of Materdicine, Shanghai 200051, P. R. China
| |
Collapse
|
6
|
Crossley P, Sutar Y, Tsoy I, Mukkirwar S, Łaniewski P, Herbst-Kralovetz MM, Date AA. Development of phenyllactic acid ionic liquids and evaluation of cytotoxicity to human cervical epithelial cells. RSC Adv 2024; 14:16083-16092. [PMID: 38765482 PMCID: PMC11100303 DOI: 10.1039/d4ra01812e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 04/24/2024] [Indexed: 05/22/2024] Open
Abstract
Phenyllactic acid (PLA), is a naturally produced, broad-spectrum antimicrobial compound with activity against bacteria and fungi. PLA can be produced by a variety of lactic acid bacteria, including vaginal Lactobacillus species, which are healthy constituents of the vaginal microbiome with a protective role against invading pathogenic bacteria and/or fungi. Additionally, PLA has been shown to exhibit anti-inflammatory and immunomodulatory properties, overall indicating its therapeutic potential as an intravaginally delivered compound for modulation of the vaginal microbiome. However, PLA has low kinetic solubility in water. Hence, strategies to improve the solubility of PLA are necessary to facilitate its intravaginal delivery. Using biocompatible cations, choline and carnitine, we successfully transformed both d- and l-enantiomers of crystalline PLA into amorphous low-melting ionic liquids (ILs) with high water solubility. We further evaluated the in vitro cytotoxicity of PLA ILs to human cervical epithelial cells. Microscopic visualisation of cellular morphology using crystal violet staining and MTT cell proliferation assay revealed that PLA ILs result in minimal morphological changes and low cytotoxicity to human cervical epithelial cells. Overall, we successfully demonstrated that transforming PLA into ILs efficiently enhances its solubility in water and these formulations are not toxic to human epithelial cells. This investigation lays the groundwork for future testing of PLA ILs for their antimicrobial properties and metabolic activity within the cervicovaginal microenvironment.
Collapse
Affiliation(s)
- Phoebe Crossley
- Department of Life Sciences, University of Bath Bath UK
- Department of Obstetrics and Gynecology, College of Medicine - Phoenix, University of Arizona Phoenix AZ USA
| | - Yogesh Sutar
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, University of Arizona Tucson AZ USA
| | - Irina Tsoy
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, University of Arizona Tucson AZ USA
| | - Srushti Mukkirwar
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, University of Arizona Tucson AZ USA
| | - Paweł Łaniewski
- Department of Basic Medical Sciences, College of Medicine - Phoenix, University of Arizona Phoenix AZ USA
| | - Melissa M Herbst-Kralovetz
- Department of Basic Medical Sciences, College of Medicine - Phoenix, University of Arizona Phoenix AZ USA
- Department of Obstetrics and Gynecology, College of Medicine - Phoenix, University of Arizona Phoenix AZ USA
- University of Arizona Cancer Center, University of Arizona Tucson AZ USA
| | - Abhijit A Date
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, University of Arizona Tucson AZ USA
- University of Arizona Cancer Center, University of Arizona Tucson AZ USA
- Department of Ophthalmology and Visual Sciences, University of Arizona College of Medicine Tucson AZ USA
| |
Collapse
|
7
|
Zhou J, Hou HT, Song Y, Zhou XL, Chen HX, Zhang LL, Xue HM, Yang Q, He GW. Metabolomics Analysis Identifies Differential Metabolites as Biomarkers for Acute Myocardial Infarction. Biomolecules 2024; 14:532. [PMID: 38785939 PMCID: PMC11117998 DOI: 10.3390/biom14050532] [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: 02/13/2024] [Revised: 04/07/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024] Open
Abstract
Myocardial infarction (MI), including ST-segment elevation MI (STEMI) and non-ST-segment elevation MI (NSTEMI), is still a leading cause of death worldwide. Metabolomics technology was used to explore differential metabolites (DMs) as potential biomarkers for early diagnosis of STEMI and NSTEMI. In the study, 2531 metabolites, including 1925 DMs, were discovered. In the selected 27 DMs, 14 were successfully verified in a new cohort, and the AUC values were all above 0.8. There were 10 in STEMI group, namely L-aspartic acid, L-acetylcarnitine, acetylglycine, decanoylcarnitine, hydroxyphenyllactic acid, ferulic acid, itaconic acid, lauroylcarnitine, myristoylcarnitine, and cis-4-hydroxy-D-proline, and 5 in NSTEMI group, namely L-aspartic acid, arachidonic acid, palmitoleic acid, D-aspartic acid, and palmitelaidic acid. These 14 DMs may be developed as biomarkers for the early diagnosis of MI with high sensitivity and specificity. These findings have particularly important clinical significance for NSTEMI patients because these patients have no typical ECG changes.
Collapse
Affiliation(s)
- Jie Zhou
- Department of Cardiac Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Tianjin University, Tianjin 300457, China; (J.Z.); (H.-T.H.); (H.-X.C.); (L.-L.Z.); (H.-M.X.); (Q.Y.)
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin 300457, China; (Y.S.); (X.-L.Z.)
| | - Hai-Tao Hou
- Department of Cardiac Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Tianjin University, Tianjin 300457, China; (J.Z.); (H.-T.H.); (H.-X.C.); (L.-L.Z.); (H.-M.X.); (Q.Y.)
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin 300457, China; (Y.S.); (X.-L.Z.)
- Department of Cardiac Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Tianjin 300457, China
| | - Yu Song
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin 300457, China; (Y.S.); (X.-L.Z.)
- Department of Cardiology & The Institute of Cardiovascular Diseases and the Critical Care Unit, TEDA International Cardiovascular Hospital, Tianjin University, Chinese Academy of Medical Sciences, Tianjin 300457, China
| | - Xiao-Lin Zhou
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin 300457, China; (Y.S.); (X.-L.Z.)
- Department of Cardiology & The Institute of Cardiovascular Diseases and the Critical Care Unit, TEDA International Cardiovascular Hospital, Tianjin University, Chinese Academy of Medical Sciences, Tianjin 300457, China
| | - Huan-Xin Chen
- Department of Cardiac Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Tianjin University, Tianjin 300457, China; (J.Z.); (H.-T.H.); (H.-X.C.); (L.-L.Z.); (H.-M.X.); (Q.Y.)
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin 300457, China; (Y.S.); (X.-L.Z.)
- Department of Cardiac Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Tianjin 300457, China
| | - Li-Li Zhang
- Department of Cardiac Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Tianjin University, Tianjin 300457, China; (J.Z.); (H.-T.H.); (H.-X.C.); (L.-L.Z.); (H.-M.X.); (Q.Y.)
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin 300457, China; (Y.S.); (X.-L.Z.)
- Department of Cardiac Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Tianjin 300457, China
| | - Hong-Mei Xue
- Department of Cardiac Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Tianjin University, Tianjin 300457, China; (J.Z.); (H.-T.H.); (H.-X.C.); (L.-L.Z.); (H.-M.X.); (Q.Y.)
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin 300457, China; (Y.S.); (X.-L.Z.)
- Department of Cardiac Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Tianjin 300457, China
| | - Qin Yang
- Department of Cardiac Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Tianjin University, Tianjin 300457, China; (J.Z.); (H.-T.H.); (H.-X.C.); (L.-L.Z.); (H.-M.X.); (Q.Y.)
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin 300457, China; (Y.S.); (X.-L.Z.)
- Department of Cardiac Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Tianjin 300457, China
| | - Guo-Wei He
- Department of Cardiac Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Tianjin University, Tianjin 300457, China; (J.Z.); (H.-T.H.); (H.-X.C.); (L.-L.Z.); (H.-M.X.); (Q.Y.)
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin 300457, China; (Y.S.); (X.-L.Z.)
- Department of Cardiac Surgery & The Institute of Cardiovascular Diseases, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Tianjin 300457, China
| |
Collapse
|
8
|
Mukherjee A, Breselge S, Dimidi E, Marco ML, Cotter PD. Fermented foods and gastrointestinal health: underlying mechanisms. Nat Rev Gastroenterol Hepatol 2024; 21:248-266. [PMID: 38081933 DOI: 10.1038/s41575-023-00869-x] [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: 11/01/2023] [Indexed: 12/20/2023]
Abstract
Although fermentation probably originally developed as a means of preserving food substrates, many fermented foods (FFs), and components therein, are thought to have a beneficial effect on various aspects of human health, and gastrointestinal health in particular. It is important that any such perceived benefits are underpinned by rigorous scientific research to understand the associated mechanisms of action. Here, we review in vitro, ex vivo and in vivo studies that have provided insights into the ways in which the specific food components, including FF microorganisms and a variety of bioactives, can contribute to health-promoting activities. More specifically, we draw on representative examples of FFs to discuss the mechanisms through which functional components are produced or enriched during fermentation (such as bioactive peptides and exopolysaccharides), potentially toxic or harmful compounds (such as phytic acid, mycotoxins and lactose) are removed from the food substrate, and how the introduction of fermentation-associated live or dead microorganisms, or components thereof, to the gut can convey health benefits. These studies, combined with a deeper understanding of the microbial composition of a wider variety of modern and traditional FFs, can facilitate the future optimization of FFs, and associated microorganisms, to retain and maximize beneficial effects in the gut.
Collapse
Affiliation(s)
| | - Samuel Breselge
- Teagasc Food Research Centre, Moorepark, Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - Eirini Dimidi
- Department of Nutritional Sciences, King's College London, London, UK
| | - Maria L Marco
- Department of Food Science & Technology, University of California, Davis, CA, USA
| | - Paul D Cotter
- Teagasc Food Research Centre, Moorepark, Cork, Ireland.
- APC Microbiome Ireland, Cork, Ireland.
- VistaMilk, Cork, Ireland.
| |
Collapse
|
9
|
Sung J, Rajendraprasad SS, Philbrick KL, Bauer BA, Gajic O, Shah A, Laudanski K, Bakken JS, Skalski J, Karnatovskaia LV. The human gut microbiome in critical illness: disruptions, consequences, and therapeutic frontiers. J Crit Care 2024; 79:154436. [PMID: 37769422 PMCID: PMC11034825 DOI: 10.1016/j.jcrc.2023.154436] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/23/2023] [Accepted: 09/18/2023] [Indexed: 09/30/2023]
Abstract
With approximately 39 trillion cells and over 20 million genes, the human gut microbiome plays an integral role in both health and disease. Modern living has brought a widespread use of processed food and beverages, antimicrobial and immunomodulatory drugs, and invasive procedures, all of which profoundly disrupt the delicate homeostasis between the host and its microbiome. Of particular interest is the human gut microbiome, which is progressively being recognized as an important contributing factor in many aspects of critical illness, from predisposition to recovery. Herein, we describe the current understanding of the adverse impacts of standard intensive care interventions on the human gut microbiome and delve into how these microbial alterations can influence patient outcomes. Additionally, we explore the potential association between the gut microbiome and post-intensive care syndrome, shedding light on a previously underappreciated avenue that may enhance patient recuperation following critical illness. There is an impending need for future epidemiological studies to encompass detailed phenotypic analyses of gut microbiome perturbations. Interventions aimed at restoring the gut microbiome represent a promising therapeutic frontier in the quest to prevent and treat critical illnesses.
Collapse
Affiliation(s)
- Jaeyun Sung
- Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | | | - Kemuel L Philbrick
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Brent A Bauer
- Department of General Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Ognjen Gajic
- Department of Pulmonary & Critical Care, Mayo Clinic, Rochester, MN, USA
| | - Aditya Shah
- Division of Public Health, Infectious Diseases and Occupational Medicine, Mayo Clinic, Rochester, MN, USA
| | - Krzysztof Laudanski
- Department of Anesthesiology and Perioperative Care, Mayo Clinic, Rochester, MN, USA
| | - Johan S Bakken
- Department of Infectious Diseases, St Luke's Hospital, Duluth, MN, United States of America
| | - Joseph Skalski
- Department of Pulmonary & Critical Care, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
10
|
Lin Y, Fang M, Liu J, Zhang Y, Yu Y. Transcriptomic analyses of Vibrio parahaemolyticus under the phenyllactic acid stress. Appl Microbiol Biotechnol 2024; 108:180. [PMID: 38285117 PMCID: PMC10824802 DOI: 10.1007/s00253-024-13024-6] [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: 08/08/2023] [Revised: 12/19/2023] [Accepted: 01/18/2024] [Indexed: 01/30/2024]
Abstract
Phenyllactic acid (PLA) generally recognized as a natural organic acid shows against Vibrio parahaemolyticus activity. In this study, V. parahaemolyticus ATCC17802 (Vp17802) was cultured under the stress of 1/2MIC PLA, and then the antibacterial mechanisms were explored via transcriptomics. The minimum inhibitory concentration (MIC) of PLA against Vp17802 was 3.2 mg/mL, and the time-kill analysis resulted that Vp17802 was inhibited. PLA was able to destroy the bacterial membrane, leading to the leakage of intracellular substances and decline of ATP levels. The RNA-sequencing analysis results indicated that 1616 significantly differentially expressed genes were identified, among which 190 were up-regulated and 1426 were down-regulated. Down-regulation of the icd2 gene in the TCA cycle mediates blockage of tyrosine metabolic, arginine biosynthesis, and oxidative phosphorylation, causing insufficient energy supply of Vp17802. Moreover, PLA could cause amino acids, metal ions, and phosphate transporters to be blocked, affecting the acquisition of nutrients. The treatment by PLA altered the expression of genes encoding functions involved in quorum sensing, flagellar assembly, and cell chemotaxis pathway, which may be interfering with the biofilm formation in Vp17802, reducing cell motility. Overall, 1.6 mg/mL PLA inhibited the growth of Vp17802 by disrupting to uptake of nutrients, cell metabolism, and the formation of biofilms. The results suggested a new direction for exploring the activity of PLA against Vp17802 and provided a theoretical basis for bacterial pathogen control in the food industry. KEY POINTS: •RNA sequencing was carried out to indicate the antibacterial mechanism of Vp17802. •The icd2 gene in the TCA cycle mediates blockage of metabolic of Vp17802. •The biofilm formation has interfered with 1.6 mg/mL PLA, which could reduce cell motility and virulence.
Collapse
Affiliation(s)
- Yilin Lin
- South China University of Technology, School of Food Sciences and Engineering, Guangzhou, 510640, China
| | - Meimei Fang
- South China University of Technology, School of Food Sciences and Engineering, Guangzhou, 510640, China
| | - Jun Liu
- Sericulture & Agri-Food Research Institute Guangdong Academy of Agricultural Sciences, Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou, 510610, China
| | - Yehui Zhang
- Sericulture & Agri-Food Research Institute Guangdong Academy of Agricultural Sciences, Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou, 510610, China.
| | - Yigang Yu
- South China University of Technology, School of Food Sciences and Engineering, Guangzhou, 510640, China.
- South China University of Technology, Research Center of Food Safety and Detection, Guangzhou, 510640, China.
| |
Collapse
|
11
|
Bryant KL, Hansen C, Hecht EE. Fermentation technology as a driver of human brain expansion. Commun Biol 2023; 6:1190. [PMID: 37996482 PMCID: PMC10667226 DOI: 10.1038/s42003-023-05517-3] [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: 03/08/2023] [Accepted: 10/27/2023] [Indexed: 11/25/2023] Open
Abstract
Brain tissue is metabolically expensive. Consequently, the evolution of humans' large brains must have occurred via concomitant shifts in energy expenditure and intake. Proposed mechanisms include dietary shifts such as cooking. Importantly, though, any new food source must have been exploitable by hominids with brains a third the size of modern humans'. Here, we propose the initial metabolic trigger of hominid brain expansion was the consumption of externally fermented foods. We define "external fermentation" as occurring outside the body, as opposed to the internal fermentation in the gut. External fermentation could increase the bioavailability of macro- and micronutrients while reducing digestive energy expenditure and is supported by the relative reduction of the human colon. We discuss the explanatory power of our hypothesis and survey external fermentation practices across human cultures to demonstrate its viability across a range of environments and food sources. We close with suggestions for empirical tests.
Collapse
Affiliation(s)
- Katherine L Bryant
- Laboratoire de Psychologie Cognitive, Aix-Marseille Université, Marseille, France.
| | - Christi Hansen
- Hungry Heart Farm and Dietary Consulting, Conley, GA, USA
| | - Erin E Hecht
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
12
|
Gupta SK, Vyavahare S, Duchesne Blanes IL, Berger F, Isales C, Fulzele S. Microbiota-derived tryptophan metabolism: Impacts on health, aging, and disease. Exp Gerontol 2023; 183:112319. [PMID: 37898179 DOI: 10.1016/j.exger.2023.112319] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/05/2023] [Accepted: 10/25/2023] [Indexed: 10/30/2023]
Abstract
The intricate interplay between gut microbiota and the host is pivotal in maintaining homeostasis and health. Dietary tryptophan (TRP) metabolism initiates a cascade of essential endogenous metabolites, including kynurenine, kynurenic acid, serotonin, and melatonin, as well as microbiota-derived Trp metabolites like tryptamine, indole propionic acid (IPA), and other indole derivatives. Notably, tryptamine and IPA, among the indole metabolites, exert crucial roles in modulating immune, metabolic, and neuronal responses at both local and distant sites. Additionally, these metabolites demonstrate potent antioxidant and anti-inflammatory activities. The levels of microbiota-derived TRP metabolites are intricately linked to the gut microbiota's health, which, in turn, can be influenced by age-related changes. This review aims to comprehensively summarize the cellular and molecular impacts of tryptamine and IPA on health and aging-related complications. Furthermore, we explore the levels of tryptamine and IPA and their corresponding bacteria in select diseased conditions, shedding light on their potential significance as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Sonu Kumar Gupta
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Sagar Vyavahare
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Ian L Duchesne Blanes
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Ford Berger
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Carlos Isales
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA; Centre for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Sadanand Fulzele
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA; Centre for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, USA; Department of Cell Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA; Department of Orthopedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
13
|
Derakhshan Nazari MH, Shahrokh S, Ghanbari-Maman L, Maleknia S, Ghorbaninejad M, Meyfour A. Prediction of anti-TNF therapy failure in ulcerative colitis patients by ensemble machine learning: A prospective study. Heliyon 2023; 9:e21154. [PMID: 37928018 PMCID: PMC10623293 DOI: 10.1016/j.heliyon.2023.e21154] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/05/2023] [Accepted: 10/17/2023] [Indexed: 11/07/2023] Open
Abstract
Nowadays, anti-TNF therapy remarkably improves the medical management of ulcerative colitis (UC), but approximately 40 % of patients do not respond to this treatment. In this study, we used 79 anti-TNF-naive patients with moderate-to-severe UC from four cohorts to discover alternative therapeutic targets and develop a personalized medicine approach that can diagnose UC non-responders (UCN) prior to receiving anti-TNF therapy. To this end, two microarray data series were integrated to create a discovery cohort with 35 UC samples. A comprehensive gene expression and functional analysis was performed and identified 313 significantly altered genes, among which IL6 and INHBA were highlighted as overexpressed genes in the baseline mucosal biopsies of UCN, whose cooperation may lead to a decrease in the Tregs population. Besides, screening the abundances of immune cell subpopulations showed neutrophils' accumulation increasing the inflammation. Furthermore, the correlation of KRAS signaling activation with unresponsiveness to anti-TNF mAb was observed using network analysis. Using 50x repeated 10-fold cross-validation LASSO feature selection and a stack ensemble machine learning algorithm, a five-mRNA prognostic panel including IL13RA2, HCAR3, CSF3, INHBA, and MMP1 was introduced that could predict the response of UC patients to anti-TNF antibodies with an average accuracy of 95.3 %. The predictive capacity of the introduced biomarker panel was also validated in two independent cohorts (44 UC patients). Moreover, we presented a distinct immune cell landscape and gene signature for UCN to anti-TNF drugs and further studies should be considered to make this predictive biomarker panel and therapeutic targets applicable in the clinical setting.
Collapse
Affiliation(s)
- Mohammad Hossein Derakhshan Nazari
- Basic and Molecular Epidemiology of Gastrointestinal Disorders, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shabnam Shahrokh
- Research Center for Gastroenterology and Liver Diseases, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Ghanbari-Maman
- Basic and Molecular Epidemiology of Gastrointestinal Disorders, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Computer Science, Faculty of Mathematical Sciences, University of Kashan, Kashan, Iran
| | - Samaneh Maleknia
- Basic and Molecular Epidemiology of Gastrointestinal Disorders, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahsa Ghorbaninejad
- Basic and Molecular Epidemiology of Gastrointestinal Disorders, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Anna Meyfour
- Basic and Molecular Epidemiology of Gastrointestinal Disorders, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Suzuki S, Tanaka K, Nishikawa K, Suzuki H, Oshima A, Fujiyoshi Y. Structural basis of hydroxycarboxylic acid receptor signaling mechanisms through ligand binding. Nat Commun 2023; 14:5899. [PMID: 37736747 PMCID: PMC10516952 DOI: 10.1038/s41467-023-41650-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 09/12/2023] [Indexed: 09/23/2023] Open
Abstract
Hydroxycarboxylic acid receptors (HCA) are expressed in various tissues and immune cells. HCA2 and its agonist are thus important targets for treating inflammatory and metabolic disorders. Only limited information is available, however, on the active-state binding of HCAs with agonists. Here, we present cryo-EM structures of human HCA2-Gi and HCA3-Gi signaling complexes binding with multiple compounds bound. Agonists were revealed to form a salt bridge with arginine, which is conserved in the HCA family, to activate these receptors. Extracellular regions of the receptors form a lid-like structure that covers the ligand-binding pocket. Although transmembrane (TM) 6 in HCAs undergoes dynamic conformational changes, ligands do not directly interact with amino acids in TM6, suggesting that indirect signaling induces a slight shift in TM6 to activate Gi proteins. Structural analyses of agonist-bound HCA2 and HCA3 together with mutagenesis and molecular dynamics simulation provide molecular insights into HCA ligand recognition and activation mechanisms.
Collapse
Affiliation(s)
- Shota Suzuki
- TMDU Advanced Research Institute, Tokyo Medical and Dental University Bunkyo-ku, Tokyo, Japan
| | - Kotaro Tanaka
- Cellular and Structural Physiology Institute (CeSPI), Nagoya University, Nagoya, Japan
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan
| | - Kouki Nishikawa
- Joint Research Course for Advanced Biomolecular Characterization, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Hiroshi Suzuki
- TMDU Advanced Research Institute, Tokyo Medical and Dental University Bunkyo-ku, Tokyo, Japan
| | - Atsunori Oshima
- Cellular and Structural Physiology Institute (CeSPI), Nagoya University, Nagoya, Japan
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan
- Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan
- Center for One Medicine Innovative Translational Research, Gifu University Institute for Advanced Study, Gifu City, Japan
| | - Yoshinori Fujiyoshi
- TMDU Advanced Research Institute, Tokyo Medical and Dental University Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
15
|
Shiver AL, Sun J, Culver R, Violette A, Wynter C, Nieckarz M, Mattiello SP, Sekhon PK, Friess L, Carlson HK, Wong D, Higginbottom S, Weglarz M, Wang W, Knapp BD, Guiberson E, Sanchez J, Huang PH, Garcia PA, Buie CR, Good B, DeFelice B, Cava F, Scaria J, Sonnenburg J, Sinderen DV, Deutschbauer AM, Huang KC. A mutant fitness compendium in Bifidobacteria reveals molecular determinants of colonization and host-microbe interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.29.555234. [PMID: 37693407 PMCID: PMC10491234 DOI: 10.1101/2023.08.29.555234] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Bifidobacteria commonly represent a dominant constituent of human gut microbiomes during infancy, influencing nutrition, immune development, and resistance to infection. Despite interest as a probiotic therapy, predicting the nutritional requirements and health-promoting effects of Bifidobacteria is challenging due to major knowledge gaps. To overcome these deficiencies, we used large-scale genetics to create a compendium of mutant fitness in Bifidobacterium breve (Bb). We generated a high density, randomly barcoded transposon insertion pool in Bb, and used this pool to determine Bb fitness requirements during colonization of germ-free mice and chickens with multiple diets and in response to hundreds of in vitro perturbations. To enable mechanistic investigation, we constructed an ordered collection of insertion strains covering 1462 genes. We leveraged these tools to improve models of metabolic pathways, reveal unexpected host- and diet-specific requirements for colonization, and connect the production of immunomodulatory molecules to growth benefits. These resources will greatly reduce the barrier to future investigations of this important beneficial microbe.
Collapse
Affiliation(s)
- Anthony L. Shiver
- Department of Bioengineering, Stanford University, Stanford CA 94305, USA
| | - Jiawei Sun
- Department of Bioengineering, Stanford University, Stanford CA 94305, USA
| | - Rebecca Culver
- Department of Genetics, Stanford University, Stanford CA 94305, USA
| | - Arvie Violette
- Department of Bioengineering, Stanford University, Stanford CA 94305, USA
| | - Charles Wynter
- Department of Bioengineering, Stanford University, Stanford CA 94305, USA
| | - Marta Nieckarz
- Department of Molecular Biology and Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, SE-90187, Sweden
| | - Samara Paula Mattiello
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, 57007, USA
- College of Mathematics and Science, The University of Tennessee Southern, Pulaski TN 38478, USA
| | - Prabhjot Kaur Sekhon
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, 57007, USA
- Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, OK, 74074, USA
| | - Lisa Friess
- School of Microbiology, University College Cork, Ireland
- APC Microbiome Ireland, University College Cork, Ireland
| | - Hans K. Carlson
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Daniel Wong
- Department of Applied Physics, Stanford University, Stanford CA 94305, USA
| | - Steven Higginbottom
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Meredith Weglarz
- Stanford Shared FACS Facility, Center for Molecular and Genetic Medicine, Stanford University, Stanford, California, USA
| | - Weigao Wang
- Department of Chemical Engineering, Stanford University, Stanford CA 94305, USA
| | | | - Emma Guiberson
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Po-Hsun Huang
- Department of Mechanical Engineering, Laboratory for Energy and Microsystems Innovation, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, 02139, MA, USA
| | - Paulo A. Garcia
- Department of Mechanical Engineering, Laboratory for Energy and Microsystems Innovation, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, 02139, MA, USA
| | - Cullen R. Buie
- Department of Mechanical Engineering, Laboratory for Energy and Microsystems Innovation, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, 02139, MA, USA
| | - Benjamin Good
- Department of Applied Physics, Stanford University, Stanford CA 94305, USA
| | | | - Felipe Cava
- Department of Molecular Biology and Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, SE-90187, Sweden
| | - Joy Scaria
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, 57007, USA
- Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, OK, 74074, USA
| | - Justin Sonnenburg
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Douwe Van Sinderen
- School of Microbiology, University College Cork, Ireland
- APC Microbiome Ireland, University College Cork, Ireland
| | - Adam M. Deutschbauer
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- Department of Plant and Microbial Biology, University of California, Berkeley, CA 94720, USA
| | - Kerwyn Casey Huang
- Department of Bioengineering, Stanford University, Stanford CA 94305, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Chan-Zuckerberg Biohub, San Francisco, CA 94158
| |
Collapse
|
16
|
Duncan EM, Vita L, Dibnah B, Hudson BD. Metabolite-sensing GPCRs controlling interactions between adipose tissue and inflammation. Front Endocrinol (Lausanne) 2023; 14:1197102. [PMID: 37484963 PMCID: PMC10357040 DOI: 10.3389/fendo.2023.1197102] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/13/2023] [Indexed: 07/25/2023] Open
Abstract
Metabolic disorders including obesity, diabetes and non-alcoholic steatohepatitis are a group of conditions characterised by chronic low-grade inflammation of metabolic tissues. There is now a growing appreciation that various metabolites released from adipose tissue serve as key signalling mediators, influencing this interaction with inflammation. G protein-coupled receptors (GPCRs) are the largest family of signal transduction proteins and most historically successful drug targets. The signalling pathways for several key adipose metabolites are mediated through GPCRs expressed both on the adipocytes themselves and on infiltrating macrophages. These include three main groups of GPCRs: the FFA4 receptor, which is activated by long chain free fatty acids; the HCA2 and HCA3 receptors, activated by hydroxy carboxylic acids; and the succinate receptor. Understanding the roles these metabolites and their receptors play in metabolic-immune interactions is critical to establishing how these GPCRs may be exploited for the treatment of metabolic disorders.
Collapse
|
17
|
Guse K, Sharma A, Weyenberg E, Davison S, Ma Y, Choi Y, Johnson AJ, Chen C, Gomez A. Regular consumption of lacto-fermented vegetables has greater effects on the gut metabolome compared with the microbiome. GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2023; 4:e11. [PMID: 39295909 PMCID: PMC11406409 DOI: 10.1017/gmb.2023.9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/02/2023] [Accepted: 05/27/2023] [Indexed: 09/21/2024]
Abstract
The industrialisation of Western food systems has reduced the regular consumption of lacto-fermented vegetables (LFV). Consuming LFV may exert health benefits through the alteration of the gut microbiome, but the mechanisms involved remain unclear. To start understanding the possible benefits of LFV, we compared faecal microbial diversity and composition, as well as dietary habits between individuals who regularly consume LFV (n = 23) and those who do not (n = 24). We utilised microbial DNA amplicon sequencing (16S rRNA and ITS2) and untargeted metabolomics (LC-MS) to analyse stool samples. Study participants also provided three consecutive days of dietary data. Results show minor effects on microbiome composition; with the enrichment of a few microorganisms potentially associated with vegetable ferments, such as Leuconostoc mesenteroides and Rhodotorula mucilaginosa (P < 0.05), in LFV consumers. However, LFV consumption had greater effects on the faecal metabolome, with higher abundances of butyrate, acetate, and valerate (P < 0.05) and significantly greater metabolome diversity (P < 0.001). Overall, the observations of minor changes in the faecal microbiome and greater effects on the faecal metabolome from LFV consumption warrant further investigations on the health significance of LFV as regular components of the daily diet in humans.
Collapse
Affiliation(s)
- Kylene Guse
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN, USA
| | - Ashok Sharma
- Department of Animal Science, University of Minnesota, St. Paul, MN, USA
| | - Emily Weyenberg
- Department of Animal Science, University of Minnesota, St. Paul, MN, USA
| | - Sam Davison
- Department of Animal Science, University of Minnesota, St. Paul, MN, USA
| | - Yiwei Ma
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN, USA
| | - Yuni Choi
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Abigail J Johnson
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Twin Cities, Minneapolis, MN, USA
| | - Chi Chen
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN, USA
- Department of Animal Science, University of Minnesota, St. Paul, MN, USA
| | - Andres Gomez
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN, USA
- Department of Animal Science, University of Minnesota, St. Paul, MN, USA
| |
Collapse
|
18
|
Zhou Y, Liu M, Liu K, Wu G, Tan Y. Lung microbiota and potential treatment of respiratory diseases. Microb Pathog 2023:106197. [PMID: 37321423 DOI: 10.1016/j.micpath.2023.106197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/21/2023] [Accepted: 06/06/2023] [Indexed: 06/17/2023]
Abstract
The unique microbiome found in the lungs has been studied and shown to be associated with both pulmonary homeostasis and lung diseases. The lung microbiome has the potential to produce metabolites that modulate host-microbe interactions. Specifically, short-chain fatty acids (SCFAs) produced by certain strains of the lung microbiota have been shown to regulate immune function and maintain gut mucosal health. In response, this review described the distribution and composition of the microbiota in lung diseases and discussed the impact of the lung microbiota on health and lung disease. In addition, the review further elaborated on the mechanism of microbial metabolites in microbial-host interaction and their application in the treatment of lung diseases. A better understanding of the interaction between the microbiota, metabolites, and host will provide potential strategies for the development of novel methods for the treatment of pulmonary microbial induced lung diseases.
Collapse
Affiliation(s)
- Yaxuan Zhou
- Department of Psychiatry, Department of Medicine, Xiangya School of Medical, Central South University, Changsha, 410083, Hunan, China
| | - Mengjun Liu
- Department of Clinical Medicine, Xiangya School of Medicine, Central South University, Changsha, 410083, Hunan, China
| | - Kaixuan Liu
- Department of Excellent Doctor Training, Xiangya School of Medicine, Central South University, Changsha, 410083, Hunan, China
| | - Guojun Wu
- Department of Medical Microbiology, School of Basic Medicine, Central South University, Changsha, 410083, Hunan, China.
| | - Yurong Tan
- Department of Medical Microbiology, School of Basic Medicine, Central South University, Changsha, 410083, Hunan, China.
| |
Collapse
|
19
|
Crowder SL, Jim HSL, Hogue S, Carson TL, Byrd DA. Gut microbiome and cancer implications: Potential opportunities for fermented foods. Biochim Biophys Acta Rev Cancer 2023; 1878:188897. [PMID: 37086870 DOI: 10.1016/j.bbcan.2023.188897] [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: 02/07/2023] [Revised: 04/17/2023] [Accepted: 04/17/2023] [Indexed: 04/24/2023]
Abstract
There is a critical opportunity to improve response to immunotherapies and overall cancer survivorship via dietary interventions targeted to modify the gut microbiome, and in turn, potentially enhance anti-cancer immunity. A promising dietary intervention is fermented foods, which may alter gut microbiome composition and, in turn, improve immunity. In this article, we summarize the state of the literature pertaining to the gut microbiome and response to immunotherapy and other cancer treatments, potential clinical implications of utilizing a fermented foods dietary approach to improve cancer treatment outcomes, and existing gaps in the literature regarding the implementation of fermented food interventions among individuals with cancer or with a history of cancer. This review synthesizes a compelling rationale across different disciplines to lay a roadmap for future fermented food dietary intervention research aimed at modulating the gut microbiome to reduce cancer burden.
Collapse
Affiliation(s)
- Sylvia L Crowder
- Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| | - Heather S L Jim
- Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Stephanie Hogue
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Tiffany L Carson
- Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Doratha A Byrd
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
20
|
Liebing AD, Krumbholz P, Stäubert C. Protocol to characterize G i/o and G s protein-coupled receptors in transiently transfected cells using ELISA and cAMP measurements. STAR Protoc 2023; 4:102120. [PMID: 36853674 PMCID: PMC9958081 DOI: 10.1016/j.xpro.2023.102120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/03/2023] [Accepted: 01/31/2023] [Indexed: 02/16/2023] Open
Abstract
Activation of Gs or Gi/o protein-coupled receptors (GPCRs) leads to changes of intracellular cyclic adenosine monophosphate (cAMP) levels. This protocol describes steps for cloning HA- and FLAG-tagged GPCRs, transient transfection of CHO-K1 or HEK293-T cells, and determination of basal and ligand-induced changes in intracellular cAMP levels. We detail enzyme-linked immunosorbent assays to determine relative GPCR plasma membrane and total expression levels. For complete details on the use and execution of this protocol, please refer to Schulze et al. (2022).1.
Collapse
Affiliation(s)
- Aenne-Dorothea Liebing
- Rudolf-Schönheimer Institute for Biochemistry, Faculty of Medicine, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany
| | - Petra Krumbholz
- Rudolf-Schönheimer Institute for Biochemistry, Faculty of Medicine, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany
| | - Claudia Stäubert
- Rudolf-Schönheimer Institute for Biochemistry, Faculty of Medicine, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany.
| |
Collapse
|
21
|
Identification of Specific Biomarkers and Pathways in the Treatment Response of Infliximab for Inflammatory Bowel Disease: In-Silico Analysis. Life (Basel) 2023; 13:life13030680. [PMID: 36983834 PMCID: PMC10057676 DOI: 10.3390/life13030680] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/19/2023] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
Background: Inflammatory bowel disease (IBD) is characterized by chronic inflammation of the gastrointestinal tract. In biological therapy, infliximab became the first anti-tumor necrosis factor (TNF) agent approved for IBD. Despite this success, infliximab is expensive, often ineffective, and associated with adverse events. Prediction of infliximab resistance would improve overall potential outcomes. Therefore, there is a pressing need to widen the scope of investigating the role of genetics in IBD to their association with therapy response. Methods: In the current study, an in-silico analysis of publicly available IBD patient transcriptomics datasets from Gene Expression Omnibus (GEO) are used to identify subsets of differentially expressed genes (DEGs) involved in the pathogenesis of IBD and may serve as potential biomarkers for Infliximab response. Five datasets were found that met the inclusion criteria. The DEGs for datasets were identified using limma R packages through the GEOR2 tool. The probes’ annotated genes in each dataset intersected with DGEs from all other datasets. Enriched gene Ontology Clustering for the identified genes was performed using Metascape to explore the possible connections or interactions between the genes. Results: 174 DEGs between IBD and healthy controls were found from analyzing two datasets (GSE14580 and GSE73661), indicating a possible role in the pathogenesis of IBD. Of the 174 DEGs, five genes (SELE, TREM1, AQP9, FPR2, and HCAR3) were shared between all five datasets. Moreover, these five genes were identified as downregulated in the infliximab responder group compared to the non-responder group. Conclusions: We hypothesize that alteration in the expression of these genes leads to an impaired response to infliximab in IBD patients. Thus, these genes can serve as potential biomarkers for the early detection of compromised infliximab response in IBD patients.
Collapse
|
22
|
Sjödin KS, Sjödin A, Ruszczyński M, Kristensen MB, Hernell O, Szajewska H, West CE. Targeting the gut-lung axis by synbiotic feeding to infants in a randomized controlled trial. BMC Biol 2023; 21:38. [PMID: 36803508 PMCID: PMC9940374 DOI: 10.1186/s12915-023-01531-3] [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: 04/12/2022] [Accepted: 01/27/2023] [Indexed: 02/21/2023] Open
Abstract
BACKGROUND Formula-fed infants are at increased risk of infections. Due to the cross-talk between the mucosal systems of the gastrointestinal and respiratory tracts, adding synbiotics (prebiotics and probiotics) to infant formula may prevent infections even at distant sites. Infants that were born full term and weaned from breast milk were randomized to prebiotic formula (fructo- and galactooligosaccharides) or the same prebiotic formula with Lactobacillus paracasei ssp. paracasei F19 (synbiotics) from 1 to 6 months of age. The objective was to examine the synbiotic effects on gut microbiota development. RESULTS Fecal samples collected at ages 1, 4, 6, and 12 months were analyzed using 16S rRNA gene sequencing and a combination of untargeted gas chromatography-mass spectrometry/liquid chromatography-mass spectrometry. These analyses revealed that the synbiotic group had a lower abundance of Klebsiella, a higher abundance of Bifidobacterium breve compared to the prebiotic group, and increases in the anti-microbial metabolite d-3-phenyllactic acid. We also analyzed the fecal metagenome and antibiotic resistome in the 11 infants that had been diagnosed with lower respiratory tract infection (cases) and 11 matched controls using deep metagenomic sequencing. Cases with lower respiratory tract infection had a higher abundance of Klebsiella species and antimicrobial resistance genes related to Klebsiella pneumoniae, compared to controls. The results obtained using 16S rRNA gene amplicon and metagenomic sequencing were confirmed in silico by successful recovery of the metagenome-assembled genomes of the bacteria of interest. CONCLUSIONS This study demonstrates the additional benefit of feeding specific synbiotics to formula-fed infants over prebiotics only. Synbiotic feeding led to the underrepresentation of Klebsiella, enrichment of bifidobacteria, and increases in microbial degradation metabolites implicated in immune signaling and in the gut-lung and gut-skin axes. Our findings support future clinical evaluation of synbiotic formula in the prevention of infections and associated antibiotic treatment as a primary outcome when breastfeeding is not feasible. TRIAL REGISTRATION ClinicalTrials.gov NCT01625273 . Retrospectively registered on 21 June 2012.
Collapse
Affiliation(s)
- Kotryna Simonyté Sjödin
- grid.12650.300000 0001 1034 3451Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, 901 85 Sweden
| | - Andreas Sjödin
- grid.12650.300000 0001 1034 3451Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, 901 85 Sweden ,Division of CBRN Security and Defense, FOI – Swedish Defense Research Agency, Umeå, Sweden
| | - Marek Ruszczyński
- grid.13339.3b0000000113287408Department of Paediatrics, The Medical University of Warsaw, Warsaw, Poland
| | | | - Olle Hernell
- grid.12650.300000 0001 1034 3451Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, 901 85 Sweden
| | - Hania Szajewska
- grid.13339.3b0000000113287408Department of Paediatrics, The Medical University of Warsaw, Warsaw, Poland
| | - Christina E. West
- grid.12650.300000 0001 1034 3451Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, 901 85 Sweden
| |
Collapse
|
23
|
Lee M, Yun YR, Choi EJ, Song JH, Kang JY, Kim D, Lee KW, Chang JY. Anti-obesity effect of vegetable juice fermented with lactic acid bacteria isolated from kimchi in C57BL/6J mice and human mesenchymal stem cells. Food Funct 2023; 14:1349-1356. [PMID: 36630124 DOI: 10.1039/d2fo02998g] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
This study aimed to investigate the effect of fermented vegetable juice (VJ) obtained from a blend of four crops (Brassica oleracea var. capitata, B. oleracea var. italica, Daucus carota L., and Beta vulgaris) on adipogenesis along with the identification of active compounds. Two lactic acid bacteria (LAB) (Companilactobacillus allii WiKim39 and Lactococcus lactis WiKim0124), isolated from kimchi, were used to ferment the VJ and their effectiveness was evaluated in differentiated human mesenchymal stem cells and obese mice. In vitro antibody array analysis was done to understand signaling proteins in adipogenesis. Gene Ontology enrichment analysis showed that differentially expressed proteins are related to biological processes including immunological processes. These were effectively regulated by LAB and fermented VJ. Supplementation of fermented VJ reduced the weight gain, blood biochemical indicators, and liver fat accumulation in mice. Oil Red O staining indicated that the fermentation metabolites of VJ (indole-3-lactic acid, leucic acid, and phenyllactic acid) had an inhibitory effect on lipid accumulation in vitro. Therefore, it can be concluded that LAB-fermented VJ and its metabolites have the potential to counter obesity, and thus can be therapeutically effective.
Collapse
Affiliation(s)
- Moeun Lee
- Research and Development Division, World Institute of Kimchi, Gwangju 61755, Korea. .,Biomodulation Major, Department of Agricultural Biotechnology, Seoul National University, Seoul, 08826, Korea.
| | - Ye-Rang Yun
- Research and Development Division, World Institute of Kimchi, Gwangju 61755, Korea.
| | - Eun Ji Choi
- Research and Development Division, World Institute of Kimchi, Gwangju 61755, Korea.
| | - Jung Hee Song
- Research and Development Division, World Institute of Kimchi, Gwangju 61755, Korea.
| | - Jin Yong Kang
- Research and Development Division, World Institute of Kimchi, Gwangju 61755, Korea.
| | - Daun Kim
- Research and Development Division, World Institute of Kimchi, Gwangju 61755, Korea.
| | - Ki Won Lee
- Biomodulation Major, Department of Agricultural Biotechnology, Seoul National University, Seoul, 08826, Korea. .,Advanced Institutes of Convergence Technology, Seoul National University, Suwon 16229, Korea
| | - Ji Yoon Chang
- Research and Development Division, World Institute of Kimchi, Gwangju 61755, Korea.
| |
Collapse
|
24
|
Jahn LJ, Rekdal VM, Sommer MOA. Microbial foods for improving human and planetary health. Cell 2023; 186:469-478. [PMID: 36657442 DOI: 10.1016/j.cell.2022.12.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/18/2022] [Accepted: 12/01/2022] [Indexed: 01/19/2023]
Abstract
The current food production system is negatively impacting planetary and human health. A transition to a sustainable and fair food system is urgently needed. Microorganisms are likely enablers of this process, as they can produce delicious and healthy microbial foods with low environmental footprints. We review traditional and current approaches to microbial foods, such as fermented foods, microbial biomass, and food ingredients derived from microbial fermentations. We discuss how future advances in science-driven fermentation, synthetic biology, and sustainable feedstocks enable a new generation of microbial foods, potentially impacting the sustainability, resilience, and health effects of our food system.
Collapse
Affiliation(s)
- Leonie J Jahn
- Novo Nordisk Foundation Center for Biosustainability, DTU Biosustain, Kgs. Lyngby, Denmark
| | - Vayu M Rekdal
- Novo Nordisk Foundation Center for Biosustainability, DTU Biosustain, Kgs. Lyngby, Denmark; Joint BioEnergy Institute, Emeryville, CA 94608, USA; Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Morten O A Sommer
- Novo Nordisk Foundation Center for Biosustainability, DTU Biosustain, Kgs. Lyngby, Denmark.
| |
Collapse
|
25
|
Shvabskaia OB, Karamnova NS, Izmailova OV, Drapkina OM. Healthy Eating in Population Models of Nutrition: Asian Diet Style Summary. RATIONAL PHARMACOTHERAPY IN CARDIOLOGY 2023. [DOI: 10.20996/1819-6446-2022-12-08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The population of Japan and Okinawa is known for the longest life expectancy, which many researchers rightly associate with the nature of nutrition existing in these territories. The Japanese diet and Okinawan diet, along with other traditional diets, are real examples of historically established sustainable patterns of healthy eating. Asian eating styles have marked differences from European eating patterns, not only in differences in food sources, but also in eating habits. The article presents the historical, climatic and cultural features of these diets; the issues of food composition, energy and nutritional value of these models of nutrition are considered in detail with an analysis of the differences existing between them; highlights the benefits of products grown mainly in Japan, which are ration-forming for the population of this country; as well as the results of scientific studies on the protective effect of the Japanese and Okinawan diets on human health and disease prevention.
Collapse
Affiliation(s)
- O. B. Shvabskaia
- National Medical Research Center for Therapy and Preventive Medicine
| | - N. S. Karamnova
- National Medical Research Center for Therapy and Preventive Medicine
| | - O. V. Izmailova
- National Medical Research Center for Therapy and Preventive Medicine
| | - O. M. Drapkina
- National Medical Research Center for Therapy and Preventive Medicine
| |
Collapse
|
26
|
Cho S, Yang X, Won KJ, Leone VA, Chang EB, Guzman G, Ko Y, Bae ON, Lee H, Jeong H. Phenylpropionic acid produced by gut microbiota alleviates acetaminophen-induced hepatotoxicity. Gut Microbes 2023; 15:2231590. [PMID: 37431867 PMCID: PMC10337503 DOI: 10.1080/19490976.2023.2231590] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 06/13/2023] [Accepted: 06/27/2023] [Indexed: 07/12/2023] Open
Abstract
The gut microbiota affects hepatic drug metabolism. However, gut microbial factors modulating hepatic drug metabolism are largely unknown. In this study, using a mouse model of acetaminophen (APAP)-induced hepatotoxicity, we identified a gut bacterial metabolite that controls the hepatic expression of CYP2E1 that catalyzes the conversion of APAP to a reactive, toxic metabolite. By comparing C57BL/6 substrain mice from two different vendors, Jackson (6J) and Taconic (6N), which are genetically similar but harbor different gut microbiotas, we established that the differences in the gut microbiotas result in differential susceptibility to APAP-induced hepatotoxicity. 6J mice exhibited lower susceptibility to APAP-induced hepatotoxicity than 6N mice, and such phenotypic difference was recapitulated in germ-free mice by microbiota transplantation. Comparative untargeted metabolomic analysis of portal vein sera and liver tissues between conventional and conventionalized 6J and 6N mice led to the identification of phenylpropionic acid (PPA), the levels of which were higher in 6J mice. PPA supplementation alleviated APAP-induced hepatotoxicity in 6N mice by lowering hepatic CYP2E1 levels. Moreover, PPA supplementation also reduced carbon tetrachloride-induced liver injury mediated by CYP2E1. Our data showed that previously known PPA biosynthetic pathway is responsible for PPA production. Surprisingly, while PPA in 6N mouse cecum contents is almost undetectable, 6N cecal microbiota produces PPA as well as 6J cecal microbiota in vitro, suggesting that PPA production in the 6N gut microbiota is suppressed in vivo. However, previously known gut bacteria harboring the PPA biosynthetic pathway were not detected in either 6J or 6N microbiota, suggesting the presence of as-yet-unidentified PPA-producing gut microbes. Collectively, our study reveals a novel biological function of the gut bacterial metabolite PPA in the gut-liver axis and presents a critical basis for investigating PPA as a modulator of CYP2E1-mediated liver injury and metabolic diseases.
Collapse
Affiliation(s)
- Sungjoon Cho
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
| | - Xiaotong Yang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
| | - Kyoung-Jae Won
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, IN, USA
| | - Vanessa A Leone
- Department of Animal & Dairy Sciences, College of Agriculture & Life Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Eugene B Chang
- Section of Gastroenterology, Knapp Center for Biomedical Discovery, University of Chicago, Chicago, IL, USA
| | - Grace Guzman
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Yeonju Ko
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Republic of Korea
| | - Ok-Nam Bae
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Republic of Korea
| | - Hyunwoo Lee
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, IN, USA
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Hyunyoung Jeong
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, IN, USA
- Department of Pharmacy Practice, College of Pharmacy, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
27
|
Sen A, Nishimura T, Yoshimoto S, Yoshida K, Gotoh A, Katoh T, Yoneda Y, Hashimoto T, Xiao JZ, Katayama T, Odamaki T. Comprehensive analysis of metabolites produced by co-cultivation of Bifidobacterium breve MCC1274 with human iPS-derived intestinal epithelial cells. Front Microbiol 2023; 14:1155438. [PMID: 37125172 PMCID: PMC10133457 DOI: 10.3389/fmicb.2023.1155438] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/23/2023] [Indexed: 05/02/2023] Open
Abstract
Examining how host cells affect metabolic behaviors of probiotics is pivotal to better understand the mechanisms underlying the probiotic efficacy in vivo. However, studies to elucidate the interaction between probiotics and host cells, such as intestinal epithelial cells, remain limited. Therefore, in this study, we performed a comprehensive metabolome analysis of a co-culture containing Bifidobacterium breve MCC1274 and induced pluripotent stem cells (iPS)-derived small intestinal-like cells. In the co-culture, we observed a significant increase in several amino acid metabolites, including indole-3-lactic acid (ILA) and phenyllactic acid (PLA). In accordance with the metabolic shift, the expression of genes involved in ILA synthesis, such as transaminase and tryptophan synthesis-related genes, was also elevated in B. breve MCC1274 cells. ILA production was enhanced in the presence of purines, which were possibly produced by intestinal epithelial cells (IECs). These findings suggest a synergistic action of probiotics and IECs, which may represent a molecular basis of host-probiotic interaction in vivo.
Collapse
Affiliation(s)
- Akira Sen
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Kanagawa, Japan
- *Correspondence: Akira Sen,
| | - Tatsuki Nishimura
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Kanagawa, Japan
| | - Shin Yoshimoto
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Kanagawa, Japan
| | - Keisuke Yoshida
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Kanagawa, Japan
| | - Aina Gotoh
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Toshihiko Katoh
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Yasuko Yoneda
- Technology Research Laboratory, Shimadzu Corp., Kyoto, Japan
| | | | - Jin-Zhong Xiao
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Kanagawa, Japan
| | - Takane Katayama
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Toshitaka Odamaki
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Kanagawa, Japan
| |
Collapse
|
28
|
Siddeeg A, Afzaal M, Saeed F, Ali R, Shah YA, Shehzadi U, Ateeq H, Waris N, Hussain M, Raza MA, Al-Farga A. Recent updates and perspectives of fermented healthy super food sauerkraut: a review. INTERNATIONAL JOURNAL OF FOOD PROPERTIES 2022. [DOI: 10.1080/10942912.2022.2135531] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Azhari Siddeeg
- Department of Food Sciences, Government College University Faisalabad, Pakistan
| | - Muhammad Afzaal
- Department of Food Engineering and Technology, Faculty of Engineering and Technology, University of Gezira, Wad Medani, Sudan
| | - Farhan Saeed
- Department of Food Engineering and Technology, Faculty of Engineering and Technology, University of Gezira, Wad Medani, Sudan
| | - Rehman Ali
- Department of Food Engineering and Technology, Faculty of Engineering and Technology, University of Gezira, Wad Medani, Sudan
| | - Yasir Abbas Shah
- Department of Food Engineering and Technology, Faculty of Engineering and Technology, University of Gezira, Wad Medani, Sudan
| | - Umber Shehzadi
- Department of Food Engineering and Technology, Faculty of Engineering and Technology, University of Gezira, Wad Medani, Sudan
| | - Huda Ateeq
- Department of Food Engineering and Technology, Faculty of Engineering and Technology, University of Gezira, Wad Medani, Sudan
| | - Numra Waris
- Department of Food Engineering and Technology, Faculty of Engineering and Technology, University of Gezira, Wad Medani, Sudan
| | - Muzzamal Hussain
- Department of Food Engineering and Technology, Faculty of Engineering and Technology, University of Gezira, Wad Medani, Sudan
| | - Muhammad Ahtisham Raza
- Department of Food Engineering and Technology, Faculty of Engineering and Technology, University of Gezira, Wad Medani, Sudan
| | - Ammar Al-Farga
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| |
Collapse
|
29
|
Shin M, Truong VL, Lee M, Kim D, Kim MS, Cho H, Jung YH, Yang J, Jeong WS, Kim Y. Investigation of phenyllactic acid as a potent tyrosinase inhibitor produced by probiotics. Curr Res Food Sci 2022; 6:100413. [PMID: 36569188 PMCID: PMC9772785 DOI: 10.1016/j.crfs.2022.100413] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 12/13/2022] Open
Abstract
Melanogenesis is responsible for skin pigmentation and the enzymatic browning of foods. Tyrosinases play a major role in melanin synthesis, and many attempts have been made to identify new natural tyrosinase inhibitors, but few have sought to do in microbes. Postbiotics are bioactive compounds produced by the metabolism of probiotics and have been reported to be safe and effective. In this study, we evaluated the tyrosinase inhibitory effects of culture supernatants of probiotics and discovered novel bacterial metabolites that can be used as a potent tyrosinase inhibitor based on metabolomics. Cultures of Bifidobacterium bifidum IDCC 4201 and Lactiplantibacillus plantarum IDCC 3501 showed effective anti-tyrosinase, reduced melanin synthesis, and altered protein expression associated with the melanogenesis pathway. Comparative metabolomics analyses conducted by GC-MS identified metabolites commonly produced by B. bifidum and L. plantarum. Of eight selected metabolites, phenyllactic acid exhibited significant tyrosinase-inhibitory activity. Our findings suggest that applications of probiotic culture supernatants containing high amounts of phenyllactic acid have potential use as anti-melanogenesis agents in food and medicines.
Collapse
Affiliation(s)
- Minhye Shin
- Department of Microbiology, College of Medicine, Inha University, Incheon, 22212, Republic of Korea
- Department of Biomedical Science, Program in Biomedical Science and Engineering, Inha University, Incheon, 22212, Republic of Korea
| | - Van-Long Truong
- Food and Bio-industry Research Institute, School of Food Science & Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Minjee Lee
- Ildong Bioscience, Pyeongtaek-si, Gyeonggi-do, 17957, Republic of Korea
| | - Donggyu Kim
- Department of Microbiology, College of Medicine, Inha University, Incheon, 22212, Republic of Korea
- Department of Biomedical Science, Program in Biomedical Science and Engineering, Inha University, Incheon, 22212, Republic of Korea
| | - Myun Soo Kim
- ICBIO, Cheonan-si, Chungchengnam-do, 31027, Republic of Korea
| | - Hana Cho
- ICBIO, Cheonan-si, Chungchengnam-do, 31027, Republic of Korea
| | - Young Hoon Jung
- Food and Bio-industry Research Institute, School of Food Science & Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Jungwoo Yang
- Ildong Bioscience, Pyeongtaek-si, Gyeonggi-do, 17957, Republic of Korea
- Corresponding author.
| | - Woo Sik Jeong
- Food and Bio-industry Research Institute, School of Food Science & Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
- Corresponding author.
| | - Younghoon Kim
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Republic of Korea
- Corresponding author.
| |
Collapse
|
30
|
Oezguen N, Yılmaz V, Horvath TD, Akbayir E, Haidacher SJ, Hoch KM, Thapa S, Palacio J, Türkoğlu R, Kürtüncü M, Engevik MA, Versalovic J, Haag AM, Tüzün E. Serum 3-phenyllactic acid level is reduced in benign multiple sclerosis and is associated with effector B cell ratios. Mult Scler Relat Disord 2022; 68:104239. [PMID: 36279598 DOI: 10.1016/j.msard.2022.104239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/24/2022] [Accepted: 10/09/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND 3-phenyllactic acid (PLA) is produced by both intestinal bacteria and the human host. PLA exists in its D- and L- chiral forms. It modulates human immune functions, thereby acting as a mediator of bacterial-host interactions. We aim to determine the amount and potential influence of PLA on clinical and immunological features of MS. METHODS We measured D- and L-PLA levels in bacterial supernatants and in sera of 60 MS patients and 25 healthy controls. We investigated potential associations between PLA levels, clinical features of MS, serum cytokine levels and ratios of peripheral blood lymphocyte subsets. RESULTS Multiple gut commensal bacteria possessed the capacity to generate D- and L-PLA. MS patients with benign phenotype showed markedly lower PLA levels than healthy controls or other MS patients. Fingolimod resistant patients had higher PLA levels at baseline. Furthermore, MS patients with higher PLA levels tended to display increased memory B and plasma cell ratios, elevated IL-4 levels and increased ratios of IL-4 and IL-10 producing T cell subsets. CONCLUSION Collectively, our work indicates that reduced serum levels of PLA could be associated with a favorable clinical course in MS and possibly be used as a biomarker.
Collapse
Affiliation(s)
- Numan Oezguen
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA; Department of Pathology, Texas Children's Hospital, Houston, TX, USA.
| | - Vuslat Yılmaz
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Thomas D Horvath
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA; Department of Pathology, Texas Children's Hospital, Houston, TX, USA
| | - Ece Akbayir
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Sigmund J Haidacher
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA; Department of Pathology, Texas Children's Hospital, Houston, TX, USA
| | - Kathleen M Hoch
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA; Department of Pathology, Texas Children's Hospital, Houston, TX, USA
| | - Santosh Thapa
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA; Department of Pathology, Texas Children's Hospital, Houston, TX, USA
| | - Jeremy Palacio
- Department of Forensic Science, Saint Louis University, St. Louis, MO, USA
| | - Recai Türkoğlu
- Department of Neurology, Istanbul Haydarpasa Numune Training and Research Hospital, Istanbul, Turkey
| | - Murat Kürtüncü
- Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Melinda A Engevik
- Department of Regenerative Medicine & Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - James Versalovic
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA; Department of Pathology, Texas Children's Hospital, Houston, TX, USA
| | - Anthony M Haag
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA; Department of Pathology, Texas Children's Hospital, Houston, TX, USA
| | - Erdem Tüzün
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
31
|
Zhu H, Guo L, Yu D, Du X. New insights into immunomodulatory properties of lactic acid bacteria fermented herbal medicines. Front Microbiol 2022; 13:1073922. [DOI: 10.3389/fmicb.2022.1073922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 11/10/2022] [Indexed: 11/29/2022] Open
Abstract
The COVID-19 pandemic has brought more attention to the immune system, the body’s defense against infectious diseases. The immunomodulatory ability of traditional herbal medicine has been confirmed through clinical trial research, and has obvious advantages over prescription drugs due to its high number of potential targets and low toxicity. The active compounds of herbal drugs primarily include polysaccharides, saponins, flavonoids, and phenolics and can be modified to produce new active compounds after lactic acid bacteria (LAB) fermentation. LAB, primary source of probiotics, can produce additional immunomodulatory metabolites such as exopolysaccharides, short-chain fatty acids, and bacteriocins. Moreover, several compounds from herbal medicines can promote the growth and production of LAB-based immune active metabolites. Thus, LAB-mediated fermentation of herbal medicines has become a novel strategy for regulating human immune responses. The current review discusses the immunomodulatory properties and active compounds of LAB fermented herbal drugs, the interaction between LAB and herbal medicines, and changes in immunoregulatory components that occur during fermentation. This study also discusses the mechanisms by which LAB-fermented herbal medicines regulate the immune response, including activation of the innate or adaptive immune system and the maintenance of intestinal immune homeostasis.
Collapse
|
32
|
Schulze AS, Kleinau G, Krakowsky R, Rochmann D, Das R, Worth CL, Krumbholz P, Scheerer P, Stäubert C. Evolutionary analyses reveal immune cell receptor GPR84 as a conserved receptor for bacteria-derived molecules. iScience 2022; 25:105087. [PMID: 36164652 PMCID: PMC9508565 DOI: 10.1016/j.isci.2022.105087] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/26/2022] [Accepted: 08/31/2022] [Indexed: 10/31/2022] Open
Abstract
The G protein-coupled receptor 84 (GPR84) is found in immune cells and its expression is increased under inflammatory conditions. Activation of GPR84 by medium-chain fatty acids results in pro-inflammatory responses. Here, we screened available vertebrate genome data and found that GPR84 is present in vertebrates for more than 500 million years but absent in birds and a pseudogene in bats. Cloning and functional characterization of several mammalian GPR84 orthologs in combination with evolutionary and model-based structural analyses revealed evidence for positive selection of bear GPR84 orthologs. Naturally occurring human GPR84 variants are most frequent in Asian populations causing a loss of function. Further, we identified cis- and trans-2-decenoic acid, both known to mediate bacterial communication, as evolutionary highly conserved ligands. Our integrated set of approaches contributes to a comprehensive understanding of GPR84 in terms of evolutionary and structural aspects, highlighting GPR84 as a conserved immune cell receptor for bacteria-derived molecules.
Collapse
Affiliation(s)
- Amadeus Samuel Schulze
- Rudolf Schönheimer Institute of Biochemistry, Faculty of Medicine, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany
| | - Gunnar Kleinau
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Physics and Biophysics, Group Protein X-ray Crystallography and Signal Transduction, Charitéplatz 1, 10117 Berlin, Germany
| | - Rosanna Krakowsky
- Rudolf Schönheimer Institute of Biochemistry, Faculty of Medicine, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany
| | - David Rochmann
- Rudolf Schönheimer Institute of Biochemistry, Faculty of Medicine, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany
| | - Ranajit Das
- Yenepoya Research Centre, Yenepoya University, Mangalore, Karnataka, India
| | - Catherine L Worth
- Independent Data Lab UG, Frauenmantelanger 31, 80937 Munich, Germany
| | - Petra Krumbholz
- Rudolf Schönheimer Institute of Biochemistry, Faculty of Medicine, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany
| | - Patrick Scheerer
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Physics and Biophysics, Group Protein X-ray Crystallography and Signal Transduction, Charitéplatz 1, 10117 Berlin, Germany
| | - Claudia Stäubert
- Rudolf Schönheimer Institute of Biochemistry, Faculty of Medicine, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany
| |
Collapse
|
33
|
Galena AE, Chai J, Zhang J, Bednarzyk M, Perez D, Ochrietor JD, Jahan-Mihan A, Arikawa AY. The effects of fermented vegetable consumption on the composition of the intestinal microbiota and levels of inflammatory markers in women: A pilot and feasibility study. PLoS One 2022; 17:e0275275. [PMID: 36201455 PMCID: PMC9536613 DOI: 10.1371/journal.pone.0275275] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 08/02/2022] [Indexed: 11/18/2022] Open
Abstract
The primary objective of this pilot study was to investigate the feasibility of regular consumption of fermented vegetables for six weeks on markers of inflammation and the composition of the gut microflora in women (clinical trials ID: NTC03407794). Thirty-one women were randomized into one of three groups: 100 g/day of fermented vegetables (group A), 100 g/day pickled vegetables (group B), or no vegetables (group C) for six weeks. Dietary intake was assessed by a food frequency questionnaire and blood and stool samples were provided before and after the intervention for measurement of C-reactive protein (CRP), tumor necrosis factor alpha (TNF-α), and lipopolysaccharide binding protein (LBP). Next-generation sequencing of the V4 region of the 16S rRNA gene was performed on the Illumina MiSeq platform. Participants' ages ranged between 18 and 69 years. Both groups A and B had a mean daily consumption of 91g of vegetables for 32 and 36 days, respectively. Serum CRP ranged between 0.9 and 265 ng/mL (SD = 92.4) at baseline, while TNF-α and LBP concentrations ranged between 0 and 9 pg/mL (SD = 2.3), and 7 and 29 μg/mL (SD = 4.4), respectively. There were no significant changes in levels of inflammatory markers among groups. At timepoint 2, group A showed an increase in Faecalibacterium prausnitzii (P = 0.022), a decrease in Ruminococcus torques (P<0.05), and a trend towards greater alpha diversity measured by the Shannon index (P = 0.074). The findings indicate that consumption of ~100 g/day of fermented vegetables for six weeks is feasible and may result in beneficial changes in the composition of the gut microbiota. Future trials should determine whether consumption of fermented vegetables is an effective strategy against gut dysbiosis.
Collapse
Affiliation(s)
- Amy E. Galena
- Department of Nutrition and Dietetics, University of North Florida, Jacksonville, FL, United States of America
| | - Jianmin Chai
- Division of Agriculture, Department of Animal Science, University of Arkansas, Fayetteville, AR, United States of America
| | - Jiangchao Zhang
- Division of Agriculture, Department of Animal Science, University of Arkansas, Fayetteville, AR, United States of America
| | - Michele Bednarzyk
- School of Nursing, University of North Florida, Jacksonville, FL, United States of America
| | - Doreen Perez
- School of Nursing, University of North Florida, Jacksonville, FL, United States of America
| | - Judith D. Ochrietor
- Department of Biology, University of North Florida, Jacksonville, FL, United States of America
| | - Alireza Jahan-Mihan
- Department of Nutrition and Dietetics, University of North Florida, Jacksonville, FL, United States of America
| | - Andrea Y. Arikawa
- Department of Nutrition and Dietetics, University of North Florida, Jacksonville, FL, United States of America
| |
Collapse
|
34
|
Li KJ, Burton-Pimentel KJ, Vergères G, Feskens EJM, Brouwer-Brolsma EM. Fermented foods and cardiometabolic health: Definitions, current evidence, and future perspectives. Front Nutr 2022; 9:976020. [PMID: 36204374 PMCID: PMC9530890 DOI: 10.3389/fnut.2022.976020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/30/2022] [Indexed: 11/30/2022] Open
Abstract
Unhealthy diets contribute to the increasing burden of non-communicable diseases. Annually, over 11 million deaths worldwide are attributed to dietary risk factors, with the vast majority of deaths resulting from cardiometabolic diseases (CMDs) including cardiovascular disease (∼10 million) and type II diabetes (∼339,000). As such, defining diets and dietary patterns that mitigate CMD risk is of great public health importance. Recently, the consumption of fermented foods has emerged as an important dietary strategy for improving cardiometabolic health. Fermented foods have been present in the human diet for over 10,000 years, but knowledge on whether their consumption benefits human health, and the molecular and microbiological mechanisms underpinning their purported health benefits, is relatively nascent. This review provides an overview of the definitions of fermented foods, types and qualities of fermented foods consumed in Europe and globally, possible mechanisms between the consumption of fermented foods and cardiometabolic health, as well as the current state of the epidemiological evidence on fermented food intake and cardiometabolic health. Finally, we outline future perspectives and opportunities for improving the role of fermented foods in human diets.
Collapse
Affiliation(s)
- Katherine J. Li
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, Netherlands
- Agroscope, Bern, Switzerland
| | | | | | - Edith J. M. Feskens
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, Netherlands
| | - Elske M. Brouwer-Brolsma
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, Netherlands
| |
Collapse
|
35
|
Navale VD, Vamkudoth K. Toxicity and preventive approaches of Fusarium derived mycotoxins using lactic acid bacteria: state of the art. Biotechnol Lett 2022; 44:1111-1126. [PMID: 36006577 DOI: 10.1007/s10529-022-03293-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 08/15/2022] [Indexed: 11/02/2022]
Abstract
Mycotoxin contamination of food and feed is a serious food safety issue and causes acute and chronic diseases in humans and livestock. Climatic and agronomic changes helps in the proliferation of fungal growth and mycotoxin production in food commodities. Mycotoxin contamination has attracted global attention due to its wide range of toxicity to humans and animals. However, physical and chemical management approaches in practice are unsafe for well-being due to their health-hazardous nature. Various antibiotics and preservatives are in use to reduce the microbial load and improve the shelf life of food products. In addition, the use of antibiotic growth promotors in livestock production may increase the risk of antimicrobial resistance, which is a global health concern. Due to their many uses, probiotics are helpful microbes that have a significant impact on food and nutrition. Furthermore, the probiotic potential of lactic acid bacteria (LAB) is employed in various food and feed preparations to neutralize mycotoxins, antimicrobial activities, balance the gut microbiome, and various immunomodulatory activities in both humans and livestock. In addition, LAB produces various antimicrobials, flavouring agents, peptides, and proteins linked to various food and health care applications. The LAB-based processes for mycotoxin management are more effective, eco-friendly, and low-cost than physical and chemical approaches. The toxicity, novel preventive measures, binding nature, and molecular mechanisms of mycotoxins' detoxification using LAB have been highlighted in this review.
Collapse
Affiliation(s)
- Vishwambar D Navale
- Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune, 411008, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, New Delhi, India
| | - KoteswaraRao Vamkudoth
- Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune, 411008, India. .,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, New Delhi, India.
| |
Collapse
|
36
|
Cheng YY, Park TH, Seong H, Kim TJ, Han NS. Biological characterization of D-lactate dehydrogenase responsible for high-yield production of D-phenyllactic acid in Sporolactobacillus inulinus. Microb Biotechnol 2022; 15:2717-2729. [PMID: 35921426 PMCID: PMC9618312 DOI: 10.1111/1751-7915.14125] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/12/2022] [Accepted: 07/21/2022] [Indexed: 01/05/2023] Open
Abstract
PLA (3‐D‐phenyllactic acid) is an ideal antimicrobial and immune regulatory compound present in honey and fermented foods. Sporolactobacillus inulinus is regarded as a potent D‐PLA producer that reduces phenylpyruvate (PPA) with D‐lactate dehydrogenases. In this study, PLA was produced by whole‐cell bioconversion of S. inulinus ATCC 15538. Three genes encoding D‐lactate dehydrogenase (d‐ldh1, d‐ldh2, and d‐ldh3) were cloned and expressed in Escherichia coli BL21 (DE3), and their biochemical and structural properties were characterized. Consequently, a high concentration of pure D‐PLA (47 mM) was produced with a high conversion yield of 88%. Among the three enzymes, D‐LDH1 was responsible for the efficient conversion of PPA to PLA with kinetic parameters of Km (0.36 mM), kcat (481.10 s−1), and kcat/Km (1336.39 mM−1 s−1). In silico structural analysis and site‐directed mutagenesis revealed that the Ile307 in D‐LDH1 is a key residue for excellent PPA reduction with low steric hindrance at the substrate entrance. This study highlights that S. inulinus ATCC 15538 is an excellent PLA producer, equipped with a highly specific and efficient D‐LDH1 enzyme.
Collapse
Affiliation(s)
- Ya-Yun Cheng
- Brain Korea 21 Center for Bio-Health Industry, Development, Division of Animal, Horticultural, and Food Sciences, Chungbuk National University, Cheongju, Korea
| | - Tae Hyeon Park
- Brain Korea 21 Center for Bio-Health Industry, Development, Division of Animal, Horticultural, and Food Sciences, Chungbuk National University, Cheongju, Korea
| | - Hyunbin Seong
- Brain Korea 21 Center for Bio-Health Industry, Development, Division of Animal, Horticultural, and Food Sciences, Chungbuk National University, Cheongju, Korea
| | - Tae-Jip Kim
- Brain Korea 21 Center for Bio-Health Industry, Development, Division of Animal, Horticultural, and Food Sciences, Chungbuk National University, Cheongju, Korea
| | - Nam Soo Han
- Brain Korea 21 Center for Bio-Health Industry, Development, Division of Animal, Horticultural, and Food Sciences, Chungbuk National University, Cheongju, Korea
| |
Collapse
|
37
|
Varsha KK, Narisetty V, Brar KK, Madhavan A, Alphy MP, Sindhu R, Awasthi MK, Varjani S, Binod P. Bioactive metabolites in functional and fermented foods and their role as immunity booster and anti-viral innate mechanisms. JOURNAL OF FOOD SCIENCE AND TECHNOLOGY 2022; 60:1-10. [PMID: 35789583 PMCID: PMC9243801 DOI: 10.1007/s13197-022-05528-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Revised: 04/19/2022] [Accepted: 05/18/2022] [Indexed: 01/08/2023]
Abstract
Live microorganisms in the fermented foods termed probiotics and their secondary metabolites with bioactive potential were considered as potential anti-viral capabilities through various mechanisms. Given the importance of functional and fermented foods in disease prevention, there is a need to discuss the contextualization and deep understanding of the mechanism of action of these foods, particularly considering the appearance of coronavirus (COVID-19) pandemic, which is causing health concerns and increased social services globally. The mechanism of probiotic strains or their bioactive metabolites is due to stimulation of immune response through boosting T-lymphocytes, cytokines, and cell toxicity of natural killer cells. Proper consumption of these functional and fermented foods may provide additional antiviral approaches for public benefit by modulating the immune functions in the hosts. Supplementary Information The online version contains supplementary material available at 10.1007/s13197-022-05528-8.
Collapse
Affiliation(s)
| | - Vivek Narisetty
- Moolec Science, Innovation Centre, Gallows Hill, CV34 6UW, Warwick, UK
| | - Kamalpreet Kaur Brar
- Department of Civil Engineering, Lassonde School of Engineering, York University, North York, Toronto, Ontario M3J 1P3 Canada
- Centre Technologique des Résidus Industriels en Abitibi Témiscamingue, 433 Boulevard du collège, J9X0E1, Rouyn-Noranda, Canada
| | - Aravind Madhavan
- Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, 695 014 India
| | - Maria Paul Alphy
- Microbial Processes and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Trivandrum, Kerala 695 019 India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002 India
| | - Raveendran Sindhu
- Department of Food Technology, T K M Institute of Technology, Kollam, Kerala 691505 India
| | - Mukesh Kumar Awasthi
- College of Natural Resources and Environment, Northwest A & F University, Yangling, 712 100 Shaanxi China
| | - Sunita Varjani
- Gujarat Pollution Control Board, Paryavaran Bhavan, CHH Road, Sector 10 A, Gujarat Gandhinagar, 382010 India
| | - Parameswaran Binod
- Microbial Processes and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Trivandrum, Kerala 695 019 India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002 India
| |
Collapse
|
38
|
Rabe P, Gehmlich M, Peters A, Krumbholz P, Nordström A, Stäubert C. Combining metabolic phenotype determination with metabolomics and transcriptional analyses to reveal pathways regulated by hydroxycarboxylic acid receptor 2. Discov Oncol 2022; 13:47. [PMID: 35697980 PMCID: PMC9192902 DOI: 10.1007/s12672-022-00503-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/20/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The adaptation of cellular metabolism is considered a hallmark of cancer. Oncogenic signaling pathways support tumorigenesis and cancer progression through the induction of certain metabolic phenotypes associated with altered regulation of key metabolic enzymes. Hydroxycarboxylic acid receptor 2 (HCA2) is a G protein-coupled receptor previously shown to act as a tumor suppressor. Here, we aimed to unveil the connection between cellular metabolism and HCA2 in BT-474 cells. Moreover, we intend to clarify how well this metabolic phenotype is reflected in transcriptional changes and metabolite levels as determined by global metabolomics analyses. METHODS We performed both, siRNA mediated knockdown of HCA2 and stimulation with the HCA2-specific agonist monomethyl fumarate. Seahorse technology was used to determine the role of HCA2 in BT-474 breast cancer cell metabolism and its potential to induce a switch in the metabolic phenotype in the presence of different energy substrates. Changes in the mRNA expression of metabolic enzymes were detected with real-time quantitative PCR (RT-qPCR). Untargeted liquid chromatography-mass spectrometry (LC-MS) metabolic profiling was used to determine changes in metabolite levels. RESULTS Knockdown or stimulation of HCA2 induced changes in the metabolic phenotype of BT474 cells dependent on the availability of energy substrates. The presence of HCA2 was associated with increased glycolytic flux with no fatty acids available. This was reflected in the increased mRNA expression of the glycolytic enzymes PFKFB4 and PKM2, which are known to promote the Warburg effect and have been described as prognostic markers in different types of cancer. With exogenous palmitate present, HCA2 caused elevated fatty acid oxidation and likely lipolysis. The increase in lipolysis was also detectable at the transcriptional level of ATGL and the metabolite levels of palmitic and stearic acid. CONCLUSIONS We combined metabolic phenotype determination with metabolomics and transcriptional analyses and identified HCA2 as a regulator of glycolytic flux and fatty acid metabolism in BT-474 breast cancer cells. Thus, HCA2, for which agonists are already widely used to treat diseases such as psoriasis or hyperlipidemia, may prove useful as a target in combination cancer therapy.
Collapse
Affiliation(s)
- Philipp Rabe
- Rudolf Schönheimer Institute of Biochemistry, Faculty of Medicine, Leipzig University, Johannisallee 30, 04103, Leipzig, Germany
| | - Mareike Gehmlich
- Rudolf Schönheimer Institute of Biochemistry, Faculty of Medicine, Leipzig University, Johannisallee 30, 04103, Leipzig, Germany
| | - Anna Peters
- Rudolf Schönheimer Institute of Biochemistry, Faculty of Medicine, Leipzig University, Johannisallee 30, 04103, Leipzig, Germany
| | - Petra Krumbholz
- Rudolf Schönheimer Institute of Biochemistry, Faculty of Medicine, Leipzig University, Johannisallee 30, 04103, Leipzig, Germany
| | - Anders Nordström
- Swedish Metabolomics Centre, Department of Forest Genetics and Plant Physiology, Swedish University of Agricultural Sciences, Linnaeus väg 6, 901 87, Umeå, Sweden
| | - Claudia Stäubert
- Rudolf Schönheimer Institute of Biochemistry, Faculty of Medicine, Leipzig University, Johannisallee 30, 04103, Leipzig, Germany.
| |
Collapse
|
39
|
Hitch TCA, Hall LJ, Walsh SK, Leventhal GE, Slack E, de Wouters T, Walter J, Clavel T. Microbiome-based interventions to modulate gut ecology and the immune system. Mucosal Immunol 2022; 15:1095-1113. [PMID: 36180583 PMCID: PMC9705255 DOI: 10.1038/s41385-022-00564-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/12/2022] [Accepted: 08/22/2022] [Indexed: 02/04/2023]
Abstract
The gut microbiome lies at the intersection between the environment and the host, with the ability to modify host responses to disease-relevant exposures and stimuli. This is evident in how enteric microbes interact with the immune system, e.g., supporting immune maturation in early life, affecting drug efficacy via modulation of immune responses, or influencing development of immune cell populations and their mediators. Many factors modulate gut ecosystem dynamics during daily life and we are just beginning to realise the therapeutic and prophylactic potential of microbiome-based interventions. These approaches vary in application, goal, and mechanisms of action. Some modify the entire community, such as nutritional approaches or faecal microbiota transplantation, while others, such as phage therapy, probiotics, and prebiotics, target specific taxa or strains. In this review, we assessed the experimental evidence for microbiome-based interventions, with a particular focus on their clinical relevance, ecological effects, and modulation of the immune system.
Collapse
Affiliation(s)
- Thomas C A Hitch
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Lindsay J Hall
- Gut Microbes & Health, Quadram Institute Biosciences, Norwich, UK
- Intestinal Microbiome, School of Life Sciences, ZIEL-Institute for Food & Health, Technical University of Munich, Freising, Germany
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Sarah Kate Walsh
- School of Food and Nutritional Sciences, University College Cork, Cork, Ireland
- APC Microbiome Ireland, School of Microbiology and Department of Medicine, University College Cork, Cork, Ireland
| | | | - Emma Slack
- Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | | | - Jens Walter
- APC Microbiome Ireland, School of Microbiology and Department of Medicine, University College Cork, Cork, Ireland
| | - Thomas Clavel
- Functional Microbiome Research Group, Institute of Medical Microbiology, University Hospital of RWTH Aachen, Aachen, Germany.
| |
Collapse
|
40
|
Frank HER, Amato K, Trautwein M, Maia P, Liman ER, Nichols LM, Schwenk K, Breslin PAS, Dunn RR. The evolution of sour taste. Proc Biol Sci 2022; 289:20211918. [PMID: 35135352 PMCID: PMC8826303 DOI: 10.1098/rspb.2021.1918] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 01/05/2022] [Indexed: 01/05/2023] Open
Abstract
The evolutionary history of sour taste has been little studied. Through a combination of literature review and trait mapping on the vertebrate phylogenetic tree, we consider the origin of sour taste, potential cases of the loss of sour taste, and those factors that might have favoured changes in the valence of sour taste-from aversive to appealing. We reconstruct sour taste as having evolved in ancient fish. By contrast to other tastes, sour taste does not appear to have been lost in any major vertebrate taxa. For most species, sour taste is aversive. Animals, including humans, that enjoy the sour taste triggered by acidic foods are exceptional. We conclude by considering why sour taste evolved, why it might have persisted as vertebrates made the transition to land and what factors might have favoured the preference for sour-tasting, acidic foods, particularly in hominins, such as humans.
Collapse
Affiliation(s)
- Hannah E. R. Frank
- Department of Crop and Soil Sciences North Carolina State University, Raleigh, USA
| | - Katie Amato
- Department of Anthropology, Northwestern University, Evanston, IL, USA
| | - Michelle Trautwein
- Entomology Department, Institute for Biodiversity Science and Sustainability, California Academy of Sciences, San Francisco, USA
| | - Paula Maia
- Department of Anthropology, Northwestern University, Evanston, IL, USA
| | - Emily R. Liman
- Department of Biological Sciences, Section of Neurobiology, University of Southern California, Los Angeles, CA, USA
| | - Lauren M. Nichols
- Department of Applied Ecology, North Carolina State University, Raleigh, USA
| | - Kurt Schwenk
- Department of Ecology and Evolutionary Biology, University of Connecticut, Storrs, CT, USA
| | - Paul A. S. Breslin
- Department of Nutritional Sciences, Rutgers The State University of New Jersey, New Brunswick, NJ, USA
- Monell Chemical Senses Center, Philadelphia, PA, USA
| | - Robert R. Dunn
- Department of Applied Ecology, North Carolina State University, Raleigh, USA
- Center for Evolutionary Hologenomics, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
41
|
Antibacterial effect of phenyllactic acid against Vibrio parahaemolyticus and its application on raw salmon fillets. Lebensm Wiss Technol 2022. [DOI: 10.1016/j.lwt.2021.112586] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
42
|
Coffee brews as food matrices for delivering probiotics: Opportunities, challenges, and potential health benefits. Trends Food Sci Technol 2022. [DOI: 10.1016/j.tifs.2021.11.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
43
|
Zhang Q, Guo WL, Chen GM, Qian M, Han JZ, Lv XC, Chen LJ, Rao PF, Ai LZ, Ni L. Pediococcus acidilactici FZU106 alleviates high-fat diet-induced lipid metabolism disorder in association with the modulation of intestinal microbiota in hyperlipidemic rats. Curr Res Food Sci 2022; 5:775-788. [PMID: 35520273 PMCID: PMC9064835 DOI: 10.1016/j.crfs.2022.04.009] [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: 12/31/2021] [Revised: 04/15/2022] [Accepted: 04/20/2022] [Indexed: 12/03/2022] Open
Abstract
Probiotics have been proved to have beneficial effects in improving hyperlipidemia. The purpose of the current research was to investigate the ameliorative effects of Pediococcus acidilactici FZU106, isolated from the traditional brewing of Hongqu rice wine, on lipid metabolism and intestinal microbiota in high-fat diet (HFD)-induced hyperlipidemic rats. Results showed that P. acidilactici FZU106 intervention obviously inhibited the abnormal increase of body weight, ameliorated serum and liver biochemical parameters related to lipid metabolism and oxidative stress. Histopathological evaluation also showed that P. acidilactici FZU106 could significantly reduce the excessive lipid accumulation in liver caused by HFD-feeding. Furthermore, P. acidilactici FZU106 intervention significantly increased the short-chain fatty acids (SCFAs) levels in HFD-fed rats, which was closely related to the changes of intestinal microbial composition and metabolism. Intestinal microbiota profiling by high-throughput sequencing demonstrated that P. acidilactici FZU106 intervention evidently increased the proportion of Butyricicoccus, Pediococcus, Rothia, Globicatella and [Eubacterium]_coprostanoligenes_group, and decreased the proportion of Corynebacterium_1, Psychrobacter, Oscillospira, Facklamia, Pseudogracilibacillus, Clostridium_innocuum_group, Enteractinococcus and Erysipelothrix in HFD-fed rats. Additionally, P. acidilactici FZU106 significantly regulated the mRNA levels of liver genes (including CD36, CYP7A1, SREBP-1c, BSEP, LDLr and HMGCR) involved in lipid metabolism and bile acid homeostasis. Therefore, these findings support the possibility that P. acidilactici FZU106 has the potential to reduce the disturbance of lipid metabolism by regulating intestinal microflora and liver gene expression profiles. Pediococcus acidilactici FZU106 protects against hyperlipidemia. Pediococcus acidilactici FZU106 regulates serum and liver lipid levels. Pediococcus acidilactici FZU106 regulates intestinal microbial composition. Pediococcus acidilactici FZU106 regulates lipid metabolism related genes.
Collapse
|
44
|
Metaproteomics insights into fermented fish and vegetable products and associated microbes. FOOD CHEMISTRY. MOLECULAR SCIENCES 2021; 3:100045. [PMID: 35415649 PMCID: PMC8991600 DOI: 10.1016/j.fochms.2021.100045] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 10/11/2021] [Accepted: 10/15/2021] [Indexed: 12/13/2022]
Abstract
Increasing global population means higher demand for healthy food. Fish and vegetables are healthy foods, but overproduction leads to spoilage. Fermentation of fish/vegetables elongate their shelf lives, improved flavour and functions. Microbes associated with Fish/vegetable fermentation produce health conferring peptides. There is little review on peptides elicited during fish/vegetable fermentations.
The interest in proteomic studies of fermented food is increasing; the role of proteins derived from fermentation extends beyond preservation, they also improve the organoleptic, anti-pathogenic, anti-cancer, anti-obesogenic properties, and other health conferring properties of fermented food. Traditional fermentation processes are still in use in certain cultures, but recently, the controlled process is gaining wider acceptance due to consistency and predictability. Scientists use modern biotechnological approaches to evaluate reactions and component yields from fermentation processes. Pieces of literature on fermented fish and vegetable end-products are scanty (compared to milk and meat), even though fish and vegetables are considered health conferring diets with high nutritional contents. Evaluations of peptides from fermented fish and vegetables show they have anti-obesity, anti-oxidative, anti-inflammatory, anti-pathogenic, anti-anti-nutrient, improves digestibility, taste, nutrient content, texture, aroma properties, etc. Despite challenges impeding the wider applications of the metaproteomic analysis of fermented fish and vegetables, their potential benefits cannot be underestimated.
Collapse
|
45
|
Alterations in Kynurenine and NAD + Salvage Pathways during the Successful Treatment of Inflammatory Bowel Disease Suggest HCAR3 and NNMT as Potential Drug Targets. Int J Mol Sci 2021; 22:ijms222413497. [PMID: 34948292 PMCID: PMC8705244 DOI: 10.3390/ijms222413497] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/03/2021] [Accepted: 12/14/2021] [Indexed: 02/08/2023] Open
Abstract
A meta-analysis of publicly available transcriptomic datasets was performed to identify metabolic pathways profoundly implicated in the progression and treatment of inflammatory bowel disease (IBD). The analysis revealed that genes involved in tryptophan (Trp) metabolism are upregulated in Crohn’s disease (CD) and ulcerative colitis (UC) and return to baseline after successful treatment with infliximab. Microarray and mRNAseq profiles from multiple experiments confirmed that enzymes responsible for Trp degradation via the kynurenine pathway (IDO1, KYNU, IL4I1, KMO, and TDO2), receptor of Trp metabolites (HCAR3), and enzymes catalyzing NAD+ turnover (NAMPT, NNMT, PARP9, CD38) were synchronously coregulated in IBD, but not in intestinal malignancies. The modeling of Trp metabolite fluxes in IBD indicated that changes in gene expression shifted intestinal Trp metabolism from the synthesis of 5-hydroxytryptamine (5HT, serotonin) towards the kynurenine pathway. Based on pathway modeling, this manifested in a decline in mucosal Trp and elevated kynurenine (Kyn) levels, and fueled the production of downstream metabolites, including quinolinate, a substrate for de novo NAD+ synthesis. Interestingly, IBD-dependent alterations in Trp metabolites were normalized in infliximab responders, but not in non-responders. Transcriptomic reconstruction of the NAD+ pathway revealed an increased salvage biosynthesis and utilization of NAD+ in IBD, which normalized in patients successfully treated with infliximab. Treatment-related changes in NAD+ levels correlated with shifts in nicotinamide N-methyltransferase (NNMT) expression. This enzyme helps to maintain a high level of NAD+-dependent proinflammatory signaling by removing excess inhibitory nicotinamide (Nam) from the system. Our analysis highlights the prevalent deregulation of kynurenine and NAD+ biosynthetic pathways in IBD and gives new impetus for conducting an in-depth examination of uncovered phenomena in clinical studies.
Collapse
|
46
|
Sakurai T, Horigome A, Odamaki T, Shimizu T, Xiao JZ. Production of Hydroxycarboxylic Acid Receptor 3 (HCA 3) Ligands by Bifidobacterium. Microorganisms 2021; 9:microorganisms9112397. [PMID: 34835522 PMCID: PMC8620054 DOI: 10.3390/microorganisms9112397] [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] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/31/2021] [Accepted: 11/18/2021] [Indexed: 12/18/2022] Open
Abstract
Hydroxycarboxylic acid receptor 3 (HCA3) was recently identified in the genomes of humans and other hominids but not in other mammals. We examined the production of HCA3 ligands by Bifidobacterium spp. In addition to 4-hydroxyphenyllactic acid, phenyllactic acid (PLA), and indole-3-lactic acid (ILA), we found that LeuA was produced by Bifidobacterium as an HCA3 ligand. The four ligands produced were the mixtures of enantiomers, and D-ILA, D-PLA, and D-LeuA showed stronger activity of the HCA3 ligand than their respective L-isomers. However, there was no difference in AhR activity between the two ILA enantiomers. These results provide new insights into the HCA3 ligands produced by Bifidobacterium and suggest the importance of investigating the absolute stereo structures of these metabolites.
Collapse
|
47
|
Kostner L. Über die hohe Lebenserwartung der japanischen Bevölkerung – Faktoren und mögliche Ursachen. DEUTSCHE ZEITSCHRIFT FÜR AKUPUNKTUR 2021. [PMCID: PMC8485572 DOI: 10.1007/s42212-021-00420-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
48
|
Laursen MF, Sakanaka M, von Burg N, Mörbe U, Andersen D, Moll JM, Pekmez CT, Rivollier A, Michaelsen KF, Mølgaard C, Lind MV, Dragsted LO, Katayama T, Frandsen HL, Vinggaard AM, Bahl MI, Brix S, Agace W, Licht TR, Roager HM. Bifidobacterium species associated with breastfeeding produce aromatic lactic acids in the infant gut. Nat Microbiol 2021; 6:1367-1382. [PMID: 34675385 PMCID: PMC8556157 DOI: 10.1038/s41564-021-00970-4] [Citation(s) in RCA: 233] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 08/26/2021] [Indexed: 12/12/2022]
Abstract
Breastfeeding profoundly shapes the infant gut microbiota, which is critical for early life immune development, and the gut microbiota can impact host physiology in various ways, such as through the production of metabolites. However, few breastmilk-dependent microbial metabolites mediating host-microbiota interactions are currently known. Here, we demonstrate that breastmilk-promoted Bifidobacterium species convert aromatic amino acids (tryptophan, phenylalanine and tyrosine) into their respective aromatic lactic acids (indolelactic acid, phenyllactic acid and 4-hydroxyphenyllactic acid) via a previously unrecognized aromatic lactate dehydrogenase (ALDH). The ability of Bifidobacterium species to convert aromatic amino acids to their lactic acid derivatives was confirmed using monocolonized mice. Longitudinal profiling of the faecal microbiota composition and metabolome of Danish infants (n = 25), from birth until 6 months of age, showed that faecal concentrations of aromatic lactic acids are correlated positively with the abundance of human milk oligosaccharide-degrading Bifidobacterium species containing the ALDH, including Bifidobacterium longum, B. breve and B. bifidum. We further demonstrate that faecal concentrations of Bifidobacterium-derived indolelactic acid are associated with the capacity of these samples to activate in vitro the aryl hydrocarbon receptor (AhR), a receptor important for controlling intestinal homoeostasis and immune responses. Finally, we show that indolelactic acid modulates ex vivo immune responses of human CD4+ T cells and monocytes in a dose-dependent manner by acting as an agonist of both the AhR and hydroxycarboxylic acid receptor 3 (HCA3). Our findings reveal that breastmilk-promoted Bifidobacterium species produce aromatic lactic acids in the gut of infants and suggest that these microbial metabolites may impact immune function in early life.
Collapse
Affiliation(s)
- Martin F Laursen
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Mikiyasu Sakanaka
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
- Faculty of Bioresources and Environmental Sciences, Ishikawa Prefectural University, Ishikawa, Japan
| | - Nicole von Burg
- Mucosal Immunology Group, Department of Health Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Urs Mörbe
- Mucosal Immunology Group, Department of Health Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Daniel Andersen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Janne Marie Moll
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Ceyda T Pekmez
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg C, Denmark
| | - Aymeric Rivollier
- Mucosal Immunology Group, Department of Health Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Kim F Michaelsen
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg C, Denmark
| | - Christian Mølgaard
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg C, Denmark
| | - Mads Vendelbo Lind
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg C, Denmark
| | - Lars O Dragsted
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg C, Denmark
| | - Takane Katayama
- Faculty of Bioresources and Environmental Sciences, Ishikawa Prefectural University, Ishikawa, Japan
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Henrik L Frandsen
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | | | - Martin I Bahl
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Susanne Brix
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - William Agace
- Mucosal Immunology Group, Department of Health Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
- Immunology Section, BMC D14, Department of Experimental Medicine, Lund University, Lund, Sweden
| | - Tine R Licht
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark.
| | - Henrik M Roager
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark.
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg C, Denmark.
| |
Collapse
|
49
|
Fitzgerald KC, Smith MD, Kim S, Sotirchos ES, Kornberg MD, Douglas M, Nourbakhsh B, Graves J, Rattan R, Poisson L, Cerghet M, Mowry EM, Waubant E, Giri S, Calabresi PA, Bhargava P. Multi-omic evaluation of metabolic alterations in multiple sclerosis identifies shifts in aromatic amino acid metabolism. Cell Rep Med 2021; 2:100424. [PMID: 34755135 PMCID: PMC8561319 DOI: 10.1016/j.xcrm.2021.100424] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 08/16/2021] [Accepted: 09/23/2021] [Indexed: 12/13/2022]
Abstract
The circulating metabolome provides unique insights into multiple sclerosis (MS) pathophysiology, but existing studies are relatively small or characterized limited metabolites. We test for differences in the metabolome between people with MS (PwMS; n = 637 samples) and healthy controls (HC; n = 317 samples) and assess the association between metabolomic profiles and disability in PwMS. We then assess whether metabolic differences correlate with changes in cellular gene expression using publicly available scRNA-seq data and whether identified metabolites affect human immune cell function. In PwMS, we identify striking abnormalities in aromatic amino acid (AAA) metabolites (p = 2.77E-18) that are also strongly associated with disability (p = 1.01E-4). Analysis of scRNA-seq data demonstrates altered AAA metabolism in CSF and blood-derived monocyte cell populations in PwMS. Treatment with AAA-derived metabolites in vitro alters monocytic endocytosis and pro-inflammatory cytokine production. We identify shifts in AAA metabolism resulting in the reduced production of immunomodulatory metabolites and increased production of metabotoxins in PwMS.
Collapse
Affiliation(s)
- Kathryn C. Fitzgerald
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins University School of Public Health, Baltimore, MD, USA
| | - Matthew D. Smith
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sol Kim
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elias S. Sotirchos
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael D. Kornberg
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Morgan Douglas
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bardia Nourbakhsh
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jennifer Graves
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Ramandeep Rattan
- Department of Neurology, Henry Ford Health System, Wayne State University School of Medicine, Detroit, MI, USA
| | - Laila Poisson
- Department of Neurology, Henry Ford Health System, Wayne State University School of Medicine, Detroit, MI, USA
| | - Mirela Cerghet
- Department of Neurology, Henry Ford Health System, Wayne State University School of Medicine, Detroit, MI, USA
| | - Ellen M. Mowry
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins University School of Public Health, Baltimore, MD, USA
| | - Emmanuelle Waubant
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Shailendra Giri
- Department of Neurology, Henry Ford Health System, Wayne State University School of Medicine, Detroit, MI, USA
| | - Peter A. Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Pavan Bhargava
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
50
|
Chan MZA, Lau H, Lim SY, Li SFY, Liu SQ. Untargeted LC-QTOF-MS/MS based metabolomics approach for revealing bioactive components in probiotic fermented coffee brews. Food Res Int 2021; 149:110656. [PMID: 34600658 DOI: 10.1016/j.foodres.2021.110656] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 10/20/2022]
Abstract
Amidst trends in non-dairy probiotic foods and functional coffees, we recently developed a fermented coffee brew containing high live counts of the probiotics Lacticaseibacillus rhamnosus GG and Saccharomyces boulardii CNCM-I745. However, probiotic fermentation did not alter levels of principal coffee bioactive components based on targeted analyses. Here, to provide therapeutic justification compared to other non-fermented coffee brews, we aimed to discover postbiotics in coffee brews fermented with L. rhamnosus GG and/or S. boulardii CNCM-I745. By using an untargeted LC-QTOF-MS/MS based metabolomics approach coupled with validated multivariate analyses, 37 differential metabolites between fermentation treatments were putatively annotated. These include the production of postbiotics such as 2-isopropylmalate by S. boulardii CNCM-I745, and aromatic amino acid catabolites (indole-3-lactate, p-hydroxyphenyllactate, 3-phenyllactate), and hydroxydodecanoic acid by L. rhamnosus GG. Overall, LC-QTOF based untargeted metabolomics can be an effective approach to uncover postbiotics, which may substantiate additional potential functionalities of probiotic fermented foods compared to their non-fermented counterparts.
Collapse
Affiliation(s)
- Mei Zhi Alcine Chan
- Department of Food Science & Technology, National University of Singapore, Science Drive 2, Singapore 117542, Singapore
| | - Hazel Lau
- Department of Chemistry, National University of Singapore, 3 Science Drive 3, 117543, Singapore; Institute of Materials Research and Engineering, Agency for Science, Technology and Research (A∗STAR), 2 Fusionopolis Way, Innovis, #08-03, Singapore 138634, Singapore
| | - Si Ying Lim
- Department of Chemistry, National University of Singapore, 3 Science Drive 3, 117543, Singapore
| | - Sam Fong Yau Li
- Department of Chemistry, National University of Singapore, 3 Science Drive 3, 117543, Singapore; NUS Environmental Research Institute (NERI), #02-01, T-Lab Building (TL), 5A Engineering Drive 1, Singapore 117411, Singapore
| | - Shao-Quan Liu
- Department of Food Science & Technology, National University of Singapore, Science Drive 2, Singapore 117542, Singapore; National University of Singapore (Suzhou) Research Institute, No. 377 Linquan Street, Suzhou Industrial Park, Suzhou 215123, Jiangsu, China.
| |
Collapse
|