51
|
Amato KR, Mallott EK, D’Almeida Maia P, Savo Sardaro ML. Predigestion as an Evolutionary Impetus for Human Use of Fermented Food. CURRENT ANTHROPOLOGY 2021. [DOI: 10.1086/715238] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
52
|
Craig OE. Prehistoric Fermentation, Delayed-Return Economies, and the Adoption of Pottery Technology. CURRENT ANTHROPOLOGY 2021. [DOI: 10.1086/716610] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
53
|
Some Important Metabolites Produced by Lactic Acid Bacteria Originated from Kimchi. Foods 2021; 10:foods10092148. [PMID: 34574257 PMCID: PMC8465840 DOI: 10.3390/foods10092148] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/03/2021] [Accepted: 09/07/2021] [Indexed: 01/16/2023] Open
Abstract
Lactic acid bacteria (LAB) have been used for various food fermentations for thousands of years. Recently, LAB are receiving increased attention due to their great potential as probiotics for man and animals, and also as cell factories for producing enzymes, antibodies, vitamins, exopolysaccharides, and various feedstocks. LAB are safe organisms with GRAS (generally recognized as safe) status and possess relatively simple metabolic pathways easily subjected to modifications. However, relatively few studies have been carried out on LAB inhabiting plants compared to dairy LAB. Kimchi is a Korean traditional fermented vegetable, and its fermentation is carried out by LAB inhabiting plant raw materials of kimchi. Kimchi represents a model food with low pH and is fermented at low temperatures and in anaerobic environments. LAB have been adjusting to kimchi environments, and produce various metabolites such as bacteriocins, γ-aminobutyric acid, ornithine, exopolysaccharides, mannitol, etc. as products of metabolic efforts to adjust to the environments. The metabolites also contribute to the known health-promoting effects of kimchi. Due to the recent progress in multi-omics technologies, identification of genes and gene products responsible for the synthesis of functional metabolites becomes easier than before. With the aid of tools of metabolic engineering and synthetic biology, it can be envisioned that LAB strains producing valuable metabolites in large quantities will be constructed and used as starters for foods and probiotics for improving human health. Such LAB strains can also be useful as production hosts for value-added products for food, feed, and pharmaceutical industries. In this review, recent findings on the selected metabolites produced by kimchi LAB are discussed, and the potentials of metabolites will be mentioned.
Collapse
|
54
|
Abstract
Recent human activity has profoundly transformed Earth biomes on a scale and at rates that are unprecedented. Given the central role of symbioses in ecosystem processes, functions, and services throughout the Earth biosphere, the impacts of human-driven change on symbioses are critical to understand. Symbioses are not merely collections of organisms, but co-evolved partners that arise from the synergistic combination and action of different genetic programs. They function with varying degrees of permanence and selection as emergent units with substantial potential for combinatorial and evolutionary innovation in both structure and function. Following an articulation of operational definitions of symbiosis and related concepts and characteristics of the Anthropocene, we outline a basic typology of anthropogenic change (AC) and a conceptual framework for how AC might mechanistically impact symbioses with select case examples to highlight our perspective. We discuss surprising connections between symbiosis and the Anthropocene, suggesting ways in which new symbioses could arise due to AC, how symbioses could be agents of ecosystem change, and how symbioses, broadly defined, of humans and "farmed" organisms may have launched the Anthropocene. We conclude with reflections on the robustness of symbioses to AC and our perspective on the importance of symbioses as ecosystem keystones and the need to tackle anthropogenic challenges as wise and humble stewards embedded within the system.
Collapse
Affiliation(s)
- Erik F. Y. Hom
- Department of Biology and Center for Biodiversity and Conservation Research, University of Mississippi, University, MS 38677 USA
| | - Alexandra S. Penn
- Department of Sociology and Centre for Evaluation of Complexity Across the Nexus, University of Surrey, Guildford, Surrey, GU2 7XH UK
| |
Collapse
|
55
|
Laursen MF. Gut Microbiota Development: Influence of Diet from Infancy to Toddlerhood. ANNALS OF NUTRITION & METABOLISM 2021; 77:1-14. [PMID: 34461613 DOI: 10.1159/000517912] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/15/2021] [Indexed: 11/19/2022]
Abstract
Early life is a critical period as our gut microbiota establishes here and may impact both current and future health. Thus, it is of importance to understand how different factors govern the complex microbial colonization patterns in this period. The gut microbiota changes substantially during infancy and toddlerhood in terms of both taxonomic composition and diversity. This developmental trajectory differs by a variety of factors, including term of birth, mode of birth, intake of antibiotics, presence of furred pets, siblings and family members, host genetics, local environment, geographical location, and maternal and infant/toddler diet. The type of milk feeding and complementary feeding is particularly important in early and late infancy/toddlerhood, respectively. Breastfeeding, due to the supply of human milk oligosaccharide into the gut, promotes the growth of specific human milk oligosaccharide (HMO)-utilizing Bifidobacterium species that dominate the ecosystem as long as the infant is primarily breastfed. These species perform saccharolytic fermentation in the gut and produce metabolites with physiological effects that may contribute to protection against infectious and immune-related diseases. Formula feeding, due to its lack of HMOs and higher protein content, give rise to a more diverse gut microbiota that contains more opportunistic pathogens and results in a more proteolytic metabolism in the gut. Complementary feeding, due to the introduction of dietary fibers and new protein sources, induces a shift in the gut microbiota and metabolism away from the milk-adapted and toward a more mature and diverse adult-like community with increased abundances of short chain fatty acid-producing bacterial taxa. While the physiological implication of these complementary diet-induced changes remains to be established, a few recent studies indicate that an inadequately matured gut microbiota may be causally related to poor growth and development. Further studies are required to expand our knowledge on interactions between diet, gut microbiota, and health in the early life setting.
Collapse
|
56
|
Yang X, Wei W, Tan S, Guo L, Qiao S, Yao B, Wang Z. Identification and verification of HCAR3 and INSL5 as new potential therapeutic targets of colorectal cancer. World J Surg Oncol 2021; 19:248. [PMID: 34419055 PMCID: PMC8380340 DOI: 10.1186/s12957-021-02335-x] [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: 03/04/2021] [Accepted: 07/16/2021] [Indexed: 01/05/2023] Open
Abstract
Background Colorectal cancer (CRC) is one of the most common cancers of the gastrointestinal tract and ranks third in cancer-related deaths worldwide. This study was conducted to identify novel biomarkers related to the pathogenesis of CRC based upon a bioinformatics analysis, and further verify the biomarkers in clinical tumor samples and CRC cell lines. Methods A series of bioinformatics analyses were performed using datasets from NCBI-GEO and constructed a protein–protein interaction (PPI) network. This analysis enabled the identification of Hub genes, for which the mRNA expression and overall survival of CRC patients data distribution was explored in The Cancer Genome Atlas (TCGA) colon cancer and rectal cancer (COADREAD) database. Furthermore, the differential expression of HCAR3 and INLS5 was validated in clinical tumor samples by Real-time quantitative PCR analysis, western blotting analysis, and immunohistochemistry analysis. Finally, CRC cells over-expressing INSL5 were constructed and used for CCK8, cell cycle, and cell apoptosis validation assays in vitro. Results A total of 286 differentially expressed genes (DEGs) were screened, including 64 genes with increased expression and 143 genes with decreased expression in 2 CRC database, from which 10 key genes were identified: CXCL1, HCAR3, CXCL6, CXCL8, CXCL2, CXCL5, PPY, SST, INSL5, and NPY1R. Among these genes, HCAR3 and INSL5 had not previously been explored and were further verified in vitro. Conclusions HCAR3 expression was higher in CRC tissues and associated with better overall survival of CRC patients. INSL5 expression in normal tissue was higher than that in tumor tissue and its high expression was associated with a better prognosis for CRC. The overexpression of INSL5 significantly inhibited the proliferation and promoted the shearing of PARP of CRC cells. This integrated bioinformatics study presented 10 key hub genes associated with CRC. HCAR3 and INSL5 were expressed in tumor tissue and these were associated with poor survival and warrant further studies as potential therapeutic targets. Supplementary Information The online version contains supplementary material available at 10.1186/s12957-021-02335-x.
Collapse
Affiliation(s)
- Xuan Yang
- Guizhou University Medical College, Guiyang, 550025, Guizhou, China
| | - Wangao Wei
- Tongren Municipal People's Hospital, Guizhou, 554300, Tongren, China
| | - Shisheng Tan
- Guizhou University Medical College, Guiyang, 550025, Guizhou, China.,Department of Oncology, Guizhou Provincial People's Hospital, Guizhou, 550002, Guiyang, China
| | - Linrui Guo
- Tongren Municipal People's Hospital, Guizhou, 554300, Tongren, China
| | - Song Qiao
- Tongren Municipal People's Hospital, Guizhou, 554300, Tongren, China
| | - Biao Yao
- Tongren Municipal People's Hospital, Guizhou, 554300, Tongren, China.
| | - Zi Wang
- Guizhou University Medical College, Guiyang, 550025, Guizhou, China. .,Department of Oncology, Guizhou Provincial People's Hospital, Guizhou, 550002, Guiyang, China.
| |
Collapse
|
57
|
Integrative biology defines novel biomarkers of resistance to strongylid infection in horses. Sci Rep 2021; 11:14278. [PMID: 34253752 PMCID: PMC8275762 DOI: 10.1038/s41598-021-93468-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/16/2021] [Indexed: 02/06/2023] Open
Abstract
The widespread failure of anthelmintic drugs against nematodes of veterinary interest requires novel control strategies. Selective treatment of the most susceptible individuals could reduce drug selection pressure but requires appropriate biomarkers of the intrinsic susceptibility potential. To date, this has been missing in livestock species. Here, we selected Welsh ponies with divergent intrinsic susceptibility (measured by their egg excretion levels) to cyathostomin infection and found that their divergence was sustained across a 10-year time window. Using this unique set of individuals, we monitored variations in their blood cell populations, plasma metabolites and faecal microbiota over a grazing season to isolate core differences between their respective responses under worm-free or natural infection conditions. Our analyses identified the concomitant rise in plasma phenylalanine level and faecal Prevotella abundance and the reduction in circulating monocyte counts as biomarkers of the need for drug treatment (egg excretion above 200 eggs/g). This biological signal was replicated in other independent populations. We also unravelled an immunometabolic network encompassing plasma beta-hydroxybutyrate level, short-chain fatty acid producing bacteria and circulating neutrophils that forms the discriminant baseline between susceptible and resistant individuals. Altogether our observations open new perspectives on the susceptibility of equids to strongylid infection and leave scope for both new biomarkers of infection and nutritional intervention.
Collapse
|
58
|
Varsha KK, Maheshwari AP, Nampoothiri KM. Accomplishment of probiotics in human health pertaining to immunoregulation and disease control. Clin Nutr ESPEN 2021; 44:26-37. [PMID: 34330476 DOI: 10.1016/j.clnesp.2021.06.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 06/23/2021] [Indexed: 12/17/2022]
Abstract
It is a well-established fact that the microbiome harboring the human body plays a critical role in maintaining human health and can influence treatments against various ailments. Human microbiome-based research contemplates the possibility of selecting and administering specific commensal bacterial strains to modulate the gut microbiota to attain favorable outcomes to the therapies. Consumption of probiotics and probiotic-based dietary supplements as functional foods has been a promising treatment strategy against various diseases. Clinical studies demonstrate that probiotic administration alters gut microbiota composition and instigates immune modulation in the host. The benefits of probiotics are reported to be strain-specific and depend on the host's baseline immune competence. This review explores the role of probiotics in alleviating symptoms of allergy, cancer, cardio vascular (CV) diseases, diabetes mellitus (DM), bowel diseases (IBD and IBS), periodontal disease, diseases affecting liver and kidney, neuroinflammatory diseases, and viral infections. Also, it surveyed the broad spectrum bioactive compounds produced by probiotics and possible mechanisms that trigger the immune system.
Collapse
Affiliation(s)
- Kontham Kulangara Varsha
- Microbial Processes and Technology Division (MPTD), CSIR- National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram, 695 019, Kerala, India
| | - Arun Padmakumar Maheshwari
- Microbial Processes and Technology Division (MPTD), CSIR- National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram, 695 019, Kerala, India
| | - Kesavan Madhavan Nampoothiri
- Microbial Processes and Technology Division (MPTD), CSIR- National Institute for Interdisciplinary Science and Technology (NIIST), Thiruvananthapuram, 695 019, Kerala, India.
| |
Collapse
|
59
|
Allwood JG, Wakeling LT, Bean DC. Fermentation and the microbial community of Japanese koji and miso: A review. J Food Sci 2021; 86:2194-2207. [PMID: 34056716 DOI: 10.1111/1750-3841.15773] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 04/12/2021] [Accepted: 04/14/2021] [Indexed: 12/26/2022]
Abstract
Miso is a well-known traditional Japanese fermented food, with a characteristic savory flavor and aroma, known predominately as the seasoning in miso soup. Miso production involves a two-stage fermentation, where first a mold, such as Aspergillus oryzae, is inoculated onto a substrate to make koji. A subsequent fermentation, this time by bacteria and yeast, occurs when the koji is added to a salt and soybean mash, with the miso left to ferment for up to 2 years. The microbial community of miso is considered essential to the development of the unique taste, texture, and nutritional profile of miso. Despite the importance of microorganisms in the production of miso, very little research has been undertaken to characterize and describe the microbial process. In this review, we provide an overview of the two-stage fermentation process, describe what is currently known about the microbial communities involved and consider any potential health benefits associated with the consumption of miso, along with food safety concerns. As the popularity of miso continues to expand globally and is produced under new environmental conditions, understanding the microbiological processes involved will assist to ensure that global production of miso is safe as well as delicious.
Collapse
Affiliation(s)
- Joanne G Allwood
- School of Science, Psychology and Sport, Federation University Australia, Mount Helen Campus, Ballarat, Victoria, Australia
| | - Lara T Wakeling
- School of Science, Psychology and Sport, Federation University Australia, Mount Helen Campus, Ballarat, Victoria, Australia
| | - David C Bean
- School of Science, Psychology and Sport, Federation University Australia, Mount Helen Campus, Ballarat, Victoria, Australia
| |
Collapse
|
60
|
Shahbazi R, Sharifzad F, Bagheri R, Alsadi N, Yasavoli-Sharahi H, Matar C. Anti-Inflammatory and Immunomodulatory Properties of Fermented Plant Foods. Nutrients 2021; 13:1516. [PMID: 33946303 PMCID: PMC8147091 DOI: 10.3390/nu13051516] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 12/11/2022] Open
Abstract
Fermented plant foods are gaining wide interest worldwide as healthy foods due to their unique sensory features and their health-promoting potentials, such as antiobesity, antidiabetic, antihypertensive, and anticarcinogenic activities. Many fermented foods are a rich source of nutrients, phytochemicals, bioactive compounds, and probiotic microbes. The excellent biological activities of these functional foods, such as anti-inflammatory and immunomodulatory functions, are widely attributable to their high antioxidant content and lactic acid-producing bacteria (LAB). LAB contribute to the maintenance of a healthy gut microbiota composition and improvement of local and systemic immunity. Besides, antioxidant compounds are involved in several functional properties of fermented plant products by neutralizing free radicals, regulating antioxidant enzyme activities, reducing oxidative stress, ameliorating inflammatory responses, and enhancing immune system performance. Therefore, these products may protect against chronic inflammatory diseases, which are known as the leading cause of mortality worldwide. Given that a large body of evidence supports the role of fermented plant foods in health promotion and disease prevention, we aim to discuss the potential anti-inflammatory and immunomodulatory properties of selected fermented plant foods, including berries, cabbage, and soybean products, and their effects on gut microbiota.
Collapse
Affiliation(s)
- Roghayeh Shahbazi
- Cellular and Molecular Medicine Department, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (R.S.); (F.S.); (N.A.); (H.Y.-S.)
| | - Farzaneh Sharifzad
- Cellular and Molecular Medicine Department, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (R.S.); (F.S.); (N.A.); (H.Y.-S.)
| | - Rana Bagheri
- College of Liberal Art and Sciences, Portland State University, Portland, OR 97201, USA;
| | - Nawal Alsadi
- Cellular and Molecular Medicine Department, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (R.S.); (F.S.); (N.A.); (H.Y.-S.)
| | - Hamed Yasavoli-Sharahi
- Cellular and Molecular Medicine Department, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (R.S.); (F.S.); (N.A.); (H.Y.-S.)
| | - Chantal Matar
- Cellular and Molecular Medicine Department, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (R.S.); (F.S.); (N.A.); (H.Y.-S.)
- School of Nutrition, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
61
|
Penrose HM, Iftikhar R, Collins ME, Toraih E, Ruiz E, Ungerleider N, Nakhoul H, Flemington EF, Kandil E, Shah SB, Savkovic SD. Ulcerative colitis immune cell landscapes and differentially expressed gene signatures determine novel regulators and predict clinical response to biologic therapy. Sci Rep 2021; 11:9010. [PMID: 33907256 PMCID: PMC8079702 DOI: 10.1038/s41598-021-88489-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 04/08/2021] [Indexed: 12/27/2022] Open
Abstract
The heterogeneous pathobiology underlying Ulcerative Colitis (UC) is not fully understood. Using publicly available transcriptomes from adult UC patients, we identified the immune cell landscape, molecular pathways, and differentially expressed genes (DEGs) across patient cohorts and their association with treatment outcomes. The global immune cell landscape of UC tissue included increased neutrophils, T CD4 memory activated cells, active dendritic cells (DC), and M0 macrophages, as well as reduced trends in T CD8, Tregs, B memory, resting DC, and M2 macrophages. Pathway analysis of DEGs across UC cohorts demonstrated activated bacterial, inflammatory, growth, and cellular signaling. We identified a specific transcriptional signature of one hundred DEGs (UC100) that distinctly separated UC inflamed from uninflamed transcriptomes. Several UC100 DEGs, with unidentified roles in UC, were validated in primary tissue. Additionally, non-responders to anti-TNFα and anti-α4β7 therapy displayed distinct profiles of immune cells and pathways pertaining to inflammation, growth, and metabolism. We identified twenty resistant DEGs in UC non-responders to both therapies of which four had significant predictive power to treatment outcome. We demonstrated the global immune landscape and pathways in UC tissue, highlighting a unique UC signature across cohorts and a UC resistant signature with predictive performance to biologic therapy outcome.
Collapse
Affiliation(s)
- Harrison M Penrose
- Department of Pathology and Laboratory Medicine, Tulane University, 1430 Tulane Ave SL-79, New Orleans, LA, 70112, USA
| | - Rida Iftikhar
- Department of Pathology and Laboratory Medicine, Tulane University, 1430 Tulane Ave SL-79, New Orleans, LA, 70112, USA
| | - Morgan E Collins
- Department of Pathology and Laboratory Medicine, Tulane University, 1430 Tulane Ave SL-79, New Orleans, LA, 70112, USA
| | - Eman Toraih
- Division of Endocrine and Oncologic Surgery, Department of Surgery, Tulane University, New Orleans, LA, 70112, USA
| | - Emmanuelle Ruiz
- Division of Endocrine and Oncologic Surgery, Department of Surgery, Tulane University, New Orleans, LA, 70112, USA
| | - Nathan Ungerleider
- Department of Pathology and Laboratory Medicine, Tulane University, 1430 Tulane Ave SL-79, New Orleans, LA, 70112, USA
| | - Hani Nakhoul
- Department of Pathology and Laboratory Medicine, Tulane University, 1430 Tulane Ave SL-79, New Orleans, LA, 70112, USA
| | - Erik F Flemington
- Department of Pathology and Laboratory Medicine, Tulane University, 1430 Tulane Ave SL-79, New Orleans, LA, 70112, USA
| | - Emad Kandil
- Division of Endocrine and Oncologic Surgery, Department of Surgery, Tulane University, New Orleans, LA, 70112, USA
| | - Shamita B Shah
- Division of Gastroenterology, Ochsner Clinic Foundation, New Orleans, LA, 70121, USA
| | - Suzana D Savkovic
- Department of Pathology and Laboratory Medicine, Tulane University, 1430 Tulane Ave SL-79, New Orleans, LA, 70112, USA.
| |
Collapse
|
62
|
Li KJ, Brouwer-Brolsma EM, Burton-Pimentel KJ, Vergères G, Feskens EJM. A systematic review to identify biomarkers of intake for fermented food products. GENES AND NUTRITION 2021; 16:5. [PMID: 33882831 PMCID: PMC8058972 DOI: 10.1186/s12263-021-00686-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 03/30/2021] [Indexed: 12/22/2022]
Abstract
Background Fermented foods are ubiquitous in human diets and often lauded for their sensory, nutritious, and health-promoting qualities. However, precise associations between the intake of fermented foods and health have not been well-established. This is in part due to the limitations of current dietary assessment tools that rely on subjective reporting, making them prone to memory-related errors and reporting bias. The identification of food intake biomarkers (FIBs) bypasses this challenge by providing an objective measure of intake. Despite numerous studies reporting on FIBs for various types of fermented foods and drinks, unique biomarkers associated with the fermentation process (“fermentation-dependent” biomarkers) have not been well documented. We therefore conducted a comprehensive, systematic review of the literature to identify biomarkers of fermented foods commonly consumed in diets across the world. Results After title, abstract, and full-text screening, extraction of data from 301 articles resulted in an extensive list of compounds that were detected in human biofluids following the consumption of various fermented foods, with the majority of articles focusing on coffee (69), wine (69 articles), cocoa (62), beer (34), and bread (29). The identified compounds from all included papers were consolidated and sorted into FIBs proposed for a specific food, for a food group, or for the fermentation process. Alongside food-specific markers (e.g., trigonelline for coffee), and food-group markers (e.g., pentadecanoic acid for dairy intake), several fermentation-dependent markers were revealed. These comprised compounds related to the fermentation process of a particular food, such as mannitol (wine), 2-ethylmalate (beer), methionine (sourdough bread, cheese), theabrownins (tea), and gallic acid (tea, wine), while others were indicative of more general fermentation processes (e.g., ethanol from alcoholic fermentation, 3-phenyllactic acid from lactic fermentation). Conclusions Fermented foods comprise a heterogeneous group of foods. While many of the candidate FIBs identified were found to be non-specific, greater specificity may be observed when considering a combination of compounds identified for individual fermented foods, food groups, and from fermentation processes. Future studies that focus on how fermentation impacts the composition and nutritional quality of food substrates could help to identify novel biomarkers of fermented food intake. Supplementary Information The online version contains supplementary material available at 10.1186/s12263-021-00686-4.
Collapse
Affiliation(s)
- Katherine J Li
- Division of Human Nutrition and Health, Department of Agrotechnology and Food Science, Wageningen University & Research, Wageningen, Netherlands. .,Food Microbial Systems Research Division, Federal Department of Economic Affairs, Education and Research (EAER), Federal Office for Agriculture (FOAG), Agroscope, Bern, Switzerland.
| | - Elske M Brouwer-Brolsma
- Division of Human Nutrition and Health, Department of Agrotechnology and Food Science, Wageningen University & Research, Wageningen, Netherlands
| | - Kathryn J Burton-Pimentel
- Food Microbial Systems Research Division, Federal Department of Economic Affairs, Education and Research (EAER), Federal Office for Agriculture (FOAG), Agroscope, Bern, Switzerland
| | - Guy Vergères
- Food Microbial Systems Research Division, Federal Department of Economic Affairs, Education and Research (EAER), Federal Office for Agriculture (FOAG), Agroscope, Bern, Switzerland
| | - Edith J M Feskens
- Division of Human Nutrition and Health, Department of Agrotechnology and Food Science, Wageningen University & Research, Wageningen, Netherlands
| |
Collapse
|
63
|
Amato KR, Chaves ÓM, Mallott EK, Eppley TM, Abreu F, Baden AL, Barnett AA, Bicca-Marques JC, Boyle SA, Campbell CJ, Chapman CA, De la Fuente MF, Fan P, Fashing PJ, Felton A, Fruth B, Fortes VB, Grueter CC, Hohmann G, Irwin M, Matthews JK, Mekonnen A, Melin AD, Morgan DB, Ostner J, Nguyen N, Piel AK, Pinacho-Guendulain B, Quintino-Arêdes EP, Razanaparany PT, Schiel N, Sanz CM, Schülke O, Shanee S, Souto A, Souza-Alves JP, Stewart F, Stewart KM, Stone A, Sun B, Tecot S, Valenta K, Vogel ER, Wich S, Zeng Y. Fermented food consumption in wild nonhuman primates and its ecological drivers. AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2021; 175:513-530. [PMID: 33650680 DOI: 10.1002/ajpa.24257] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 02/03/2021] [Accepted: 02/10/2021] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Although fermented food use is ubiquitous in humans, the ecological and evolutionary factors contributing to its emergence are unclear. Here we investigated the ecological contexts surrounding the consumption of fruits in the late stages of fermentation by wild primates to provide insight into its adaptive function. We hypothesized that climate, socioecological traits, and habitat patch size would influence the occurrence of this behavior due to effects on the environmental prevalence of late-stage fermented foods, the ability of primates to detect them, and potential nutritional benefits. MATERIALS AND METHODS We compiled data from field studies lasting at least 9 months to describe the contexts in which primates were observed consuming fruits in the late stages of fermentation. Using generalized linear mixed-effects models, we assessed the effects of 18 predictor variables on the occurrence of fermented food use in primates. RESULTS Late-stage fermented foods were consumed by a wide taxonomic breadth of primates. However, they generally made up 0.01%-3% of the annual diet and were limited to a subset of fruit species, many of which are reported to have mechanical and chemical defenses against herbivores when not fermented. Additionally, late-stage fermented food consumption was best predicted by climate and habitat patch size. It was more likely to occur in larger habitat patches with lower annual mean rainfall and higher annual mean maximum temperatures. DISCUSSION We posit that primates capitalize on the natural fermentation of some fruits as part of a nutritional strategy to maximize periods of fruit exploitation and/or access a wider range of plant species. We speculate that these factors contributed to the evolutionary emergence of the human propensity for fermented foods.
Collapse
Affiliation(s)
- Katherine R Amato
- Department of Anthropology, Northwestern University, Evanston, Illinois, USA
| | - Óscar M Chaves
- Escuela de Biología, Universidad de Costa Rica, UCR, San José, Costa Rica
| | - Elizabeth K Mallott
- Department of Anthropology, Northwestern University, Evanston, Illinois, USA
| | - Timothy M Eppley
- Institute for Conservation Research, San Diego Zoo Global, San Diego, California, USA.,Department of Anthropology, Portland State University, Portland, Oregon, USA
| | - Filipa Abreu
- Department of Biology, Federal Rural University of Pernambuco, Recife, Pernambuco, Brazil
| | - Andrea L Baden
- Department of Anthropology, Hunter College of the City University of New York, New York, New York, USA.,The New York Consortium in Evolutionary Primatology (NYCEP), City University of New York, New York, New York, USA
| | - Adrian A Barnett
- Amazon Mammals Research Group, National Amazon Research Institute (INPA), Manaus, AM, Brazil & Department of. Zoology, Federal University of Pernambuco, Recife, Prince Edward Island, Brazil
| | - Julio Cesar Bicca-Marques
- Laboratório de Primatologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, PUCRS, Porto Alegre, RS, Brazil
| | - Sarah A Boyle
- Department of Biology, Rhodes College, Memphis, Tennessee, USA
| | - Christina J Campbell
- Department of Anthropology, California State University Northridge, Northridge, California, USA
| | - Colin A Chapman
- Department of Anthropology, Center for the Advanced Study of Human Paleobiology, George Washington University, Washington, District of Columbia, USA.,School of Life Sciences, University of KwaZulu-Natal, Pietermaritzburg, South Africa.,Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi'an, China
| | | | - Pengfei Fan
- School of Life Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Peter J Fashing
- Department of Anthropology and Environmental Studies Program, California State University Fullerton, Fullerton, California, USA.,Centre for Ecological and Evolutionary Synthesis (CEES), University of Oslo, Oslo, Norway
| | - Annika Felton
- Southern Swedish Forest Research Centre, Swedish University of Agricultural Sciences (SLU), Alnarp, Sweden
| | - Barbara Fruth
- Department of Human Behavior, Ecology and Culture, Max-Planck-Institute for Evolutionary Anthropology, Leipzig, Germany.,School of Biological and Environmental Sciences, Liverpool John Moores University, Liverpool, United Kingdom.,Centre for Research and Conservation, Royal Zoological Society of Antwerp, Antwerp, Belgium
| | - Vanessa B Fortes
- Laboratório de Primatologia, Departamento de Zootecnia e Ciências Biológicas, Universidade Federal de Santa Maria, Palmeira das Missões, RS, Brazil
| | - Cyril C Grueter
- School of Human Sciences, The University of Western Australia, Perth, Australia.,Centre for Evolutionary Biology, School of Biological Sciences, The University of Western Australia, Perth, Australia
| | - Gottfried Hohmann
- Department of Human Evolution, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Mitchell Irwin
- Department of Anthropology, Northern Illinois University, DeKalb, Illinois, USA
| | - Jaya K Matthews
- Centre for Evolutionary Biology, School of Biological Sciences, The University of Western Australia, Perth, Australia.,Africa Research & Engagement Centre, The University of Western Australia, Crawley, Western Australia, Australia
| | - Addisu Mekonnen
- Centre for Ecological and Evolutionary Synthesis (CEES), University of Oslo, Oslo, Norway
| | - Amanda D Melin
- Department of Anthropology and Archaeology, University of Calgary, Calgary, Canada
| | - David B Morgan
- Lester E. Fisher Center for the Study and Conservation of Apes, Lincoln Park Zoo, Chicago, Illinois, USA
| | - Julia Ostner
- Department of Behavioral Ecology, University of Goettingen, Goettingen, Germany.,Research Group Primate Social Evolution, German Primate Center, Leibniz Institute for Primate Research, Goettingen, Germany
| | - Nga Nguyen
- Department of Anthropology and Environmental Studies Program, California State University Fullerton, Fullerton, California, USA.,Centre for Ecological and Evolutionary Synthesis (CEES), University of Oslo, Oslo, Norway
| | - Alex K Piel
- Department of Anthropology, University College London, London, United Kingdom
| | - Braulio Pinacho-Guendulain
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana (UAM), Lerma, Mexico.,Centro Interdisciplinario de Investigación para el Desarrollo Integral Regional (CIIDIR), Unidad Oaxaca, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Erika Patricia Quintino-Arêdes
- Laboratório de Primatologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, PUCRS, Porto Alegre, RS, Brazil
| | - Patrick Tojotanjona Razanaparany
- Graduate School of Asian and African Area Studies, Kyoto University, Kyoto, Japan.,Department of Zoology and Animal Biodiversity, University of Antananarivo, Antananarivo, Madagascar
| | - Nicola Schiel
- Department of Biology, Federal Rural University of Pernambuco, Recife, Pernambuco, Brazil
| | - Crickette M Sanz
- Department of Anthropology, Washington University in St. Louis, St. Louis, Missouri, USA.,Congo Program, Wildlife Conservation Society, Brazzaville, Congo
| | - Oliver Schülke
- Department of Behavioral Ecology, University of Goettingen, Goettingen, Germany.,Research Group Primate Social Evolution, German Primate Center, Leibniz Institute for Primate Research, Goettingen, Germany
| | - Sam Shanee
- Neotropical Primate Conservation, Cornwall, United Kingdom
| | - Antonio Souto
- Departamento de Zoologia, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - João Pedro Souza-Alves
- Departamento de Zoologia, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - Fiona Stewart
- School of Biological and Environmental Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Kathrine M Stewart
- Centre for Research and Conservation, Royal Zoological Society of Antwerp, Antwerp, Belgium
| | - Anita Stone
- Biology Department, California Lutheran University, Thousand Oaks, California, USA
| | - Binghua Sun
- School of Resource and Environmental Engineering, Anhui University, Hefei, China
| | - Stacey Tecot
- School of Anthropology, University of Arizona, Tucson, Arizona, USA
| | - Kim Valenta
- Department of Anthropology, University of Florida, Gainesville, Florida, USA
| | - Erin R Vogel
- Department of Anthropology, Rutgers University, New Brunswick, New Jersey, USA
| | - Serge Wich
- School of Biological and Environmental Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Yan Zeng
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Ya'an, China
| |
Collapse
|
64
|
Marco ML, Sanders ME, Gänzle M, Arrieta MC, Cotter PD, De Vuyst L, Hill C, Holzapfel W, Lebeer S, Merenstein D, Reid G, Wolfe BE, Hutkins R. The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on fermented foods. Nat Rev Gastroenterol Hepatol 2021; 18:196-208. [PMID: 33398112 PMCID: PMC7925329 DOI: 10.1038/s41575-020-00390-5] [Citation(s) in RCA: 293] [Impact Index Per Article: 73.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/11/2020] [Indexed: 02/07/2023]
Abstract
An expert panel was convened in September 2019 by The International Scientific Association for Probiotics and Prebiotics (ISAPP) to develop a definition for fermented foods and to describe their role in the human diet. Although these foods have been consumed for thousands of years, they are receiving increased attention among biologists, nutritionists, technologists, clinicians and consumers. Despite this interest, inconsistencies related to the use of the term 'fermented' led the panel to define fermented foods and beverages as "foods made through desired microbial growth and enzymatic conversions of food components". This definition, encompassing the many varieties of fermented foods, is intended to clarify what is (and is not) a fermented food. The distinction between fermented foods and probiotics is further clarified. The panel also addressed the current state of knowledge on the safety, risks and health benefits, including an assessment of the nutritional attributes and a mechanistic rationale for how fermented foods could improve gastrointestinal and general health. The latest advancements in our understanding of the microbial ecology and systems biology of these foods were discussed. Finally, the panel reviewed how fermented foods are regulated and discussed efforts to include them as a separate category in national dietary guidelines.
Collapse
Affiliation(s)
- Maria L Marco
- Department of Food Science and Technology, University of California-Davis, Davis, CA, USA
| | - Mary Ellen Sanders
- International Scientific Association for Probiotics and Prebiotics, Centennial, CO, USA
| | - Michael Gänzle
- University of Alberta, Department of Agricultural, Food and Nutritional Science, Edmonton, Canada
| | - Marie Claire Arrieta
- Department of Physiology and Pharmacology, International Microbiome Center, University of Calgary, Calgary, Canada
| | - Paul D Cotter
- Teagasc Food Research Centre, Moorepark, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- VistaMilk, Cork, Ireland
| | - Luc De Vuyst
- Research Group of Industrial Microbiology and Food Biotechnology, Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Colin Hill
- APC Microbiome Ireland and School of Microbiology, University College Cork, Cork, Ireland
| | - Wilhelm Holzapfel
- Advanced Green Energy and Environment Institute, Handong Global University, Pohang, Gyeongbuk, South Korea
| | - Sarah Lebeer
- Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Dan Merenstein
- Department of Family Medicine, Georgetown University, Washington, DC, USA
| | - Gregor Reid
- Lawson Health Research Institute, and Departments of Microbiology & Immunology and Surgery, University of Western Ontario, London, Ontario, Canada
| | | | - Robert Hutkins
- Department of Food Science and Technology, University of Nebraska - Lincoln, Lincoln, NE, USA.
| |
Collapse
|
65
|
Bousquet J, Anto JM, Czarlewski W, Haahtela T, Fonseca SC, Iaccarino G, Blain H, Vidal A, Sheikh A, Akdis CA, Zuberbier T, Hamzah Abdul Latiff A, Abdullah B, Aberer W, Abusada N, Adcock I, Afani A, Agache I, Aggelidis X, Agustin J, Akdis M, Al‐Ahmad M, Al‐Zahab Bassam A, Alburdan H, Aldrey‐Palacios O, Alvarez Cuesta E, Alwan Salman H, Alzaabi A, Amade S, Ambrocio G, Angles R, Annesi‐Maesano I, Ansotegui IJ, Anto J, Ara Bardajo P, Arasi S, Arshad H, Cristina Artesani M, Asayag E, Avolio F, Azhari K, Bachert C, Bagnasco D, Baiardini I, Bajrović N, Bakakos P, Bakeyala Mongono S, Balotro‐Torres C, Barba S, Barbara C, Barbosa E, Barreto B, Bartra J, Bateman ED, Battur L, Bedbrook A, Bedolla Barajas M, Beghé B, Bekere A, Bel E, Ben Kheder A, Benson M, Berghea EC, Bergmann K, Bernardini R, Bernstein D, Bewick M, Bialek S, Białoszewski A, Bieber T, Billo NE, Bilo MB, Bindslev‐Jensen C, Bjermer L, Bobolea I, Bochenska Marciniak M, Bond C, Boner A, Bonini M, Bonini S, Bosnic‐Anticevich S, Bosse I, Botskariova S, Bouchard J, Boulet L, Bourret R, Bousquet P, Braido F, Briggs A, Brightling CE, Brozek J, Brussino L, Buhl R, Bumbacea R, Buquicchio R, Burguete Cabañas M, Bush A, Busse WW, Buters J, Caballero‐Fonseca F, Calderon MA, Calvo M, Camargos P, Camuzat T, Canevari F, Cano A, Canonica GW, Capriles‐Hulett A, Caraballo L, Cardona V, Carlsen K, Carmon Pirez J, Caro J, Carr W, Carreiro‐Martins P, Carreon‐Asuncion F, Carriazo A, Casale T, Castor M, Castro E, Caviglia A, Cecchi L, Cepeda Sarabia A, Chandrasekharan R, Chang Y, Chato‐Andeza V, Chatzi L, Chatzidaki C, Chavannes NH, Chaves Loureiro C, Chelninska M, Chen Y, Cheng L, Chinthrajah S, Chivato T, Chkhartishvili E, Christoff G, Chrystyn H, Chu DK, Chua A, Chuchalin A, Chung KF, Cicerán A, Cingi C, Ciprandi G, Cirule I, Coelho AC, Compalati E, Constantinidis J, Correia de Sousa J, Costa EM, Costa D, Costa Domínguez MDC, Coste A, Cottini M, Cox L, Crisci C, Crivellaro MA, Cruz AA, Cullen J, Custovic A, Cvetkovski B, Czarlewski W, D'Amato G, Silva J, Dahl R, Dahlen S, Daniilidis V, DarjaziniNahhas L, Darsow U, Davies J, Blay F, De Feo G, De Guia E, los Santos C, De Manuel Keenoy E, De Vries G, Deleanu D, Demoly P, Denburg J, Devillier P, Didier A, Dimic Janjic S, Dimou M, Dinh‐Xuan AT, Djukanovic R, Do Ceu Texeira M, Dokic D, Dominguez Silva MG, Douagui H, Douladiris N, Doulaptsi M, Dray G, Dubakiene R, Dupas E, Durham S, Duse M, Dykewicz M, Ebo D, Edelbaher N, Eiwegger T, Eklund P, El‐Gamal Y, El‐Sayed ZA, El‐Sayed SS, El‐Seify M, Emuzyte R, Enecilla L, Erhola M, Espinoza H, Espinoza Contreras JG, Farrell J, Fernandez L, Fink Wagner A, Fiocchi A, Fokkens WJ, Lenia F, Fonseca JA, Fontaine J, Forastiere F, Fuentes Pèrez JM, Gaerlan–Resureccion E, Gaga M, Gálvez Romero JL, Gamkrelidze A, Garcia A, García Cobas CY, García Cruz MDLLH, Gayraud J, Gelardi M, Gemicioglu B, Gennimata D, Genova S, Gereda J, Gerth van Wijk R, Giuliano A, Gomez M, González Diaz S, Gotua M, Grigoreas C, Grisle I, Gualteiro L, Guidacci M, Guldemond N, Gutter Z, Guzmán A, Halloum R, Halpin D, Hamelmann E, Hammadi S, Harvey R, Heffler E, Heinrich J, Hejjaoui A, Hellquist‐Dahl B, Hernández Velázquez L, Hew M, Hossny E, Howarth P, Hrubiško M, Huerta Villalobos YR, Humbert M, Salina H, Hyland M, Ibrahim M, Ilina N, Illario M, Incorvaia C, Infantino A, Irani C, Ispayeva Z, Ivancevich J, E.J. Jares E, Jarvis D, Jassem E, Jenko K, Jiméneracruz Uscanga RD, Johnston SL, Joos G, Jošt M, Julge K, Jung K, Just J, Jutel M, Kaidashev I, Kalayci O, Kalyoncu F, Kapsali J, Kardas P, Karjalainen J, Kasala CA, Katotomichelakis M, Kavaliukaite L, Kazi BS, Keil T, Keith P, Khaitov M, Khaltaev N, Kim Y, Kirenga B, Kleine‐Tebbe J, Klimek L, Koffi N’Goran B, Kompoti E, Kopač P, Koppelman G, KorenJeverica A, Koskinen S, Košnik M, Kostov KV, Kowalski ML, Kralimarkova T, Kramer Vrščaj K, Kraxner H, Kreft S, Kritikos V, Kudlay D, Kuitunen M, Kull I, Kuna P, Kupczyk M, Kvedariene V, Kyriakakou M, Lalek N, Landi M, Lane S, Larenas‐Linnemann D, Lau S, Laune D, Lavrut J, Le L, Lenzenhuber M, Lessa M, Levin M, Li J, Lieberman P, Liotta G, Lipworth B, Liu X, Lobo R, Lodrup Carlsen KC, Lombardi C, Louis R, Loukidis S, Lourenço O, Luna Pech JA, Madjar B, Maggi E, Magnan A, Mahboub B, Mair A, Mais Y, Maitland van der Zee A, Makela M, Makris M, Malling H, Mandajieva M, Manning P, Manousakis M, Maragoudakis P, Marseglia G, Marshall G, Reza Masjedi M, Máspero JF, Matta Campos JJ, Maurer M, Mavale‐Manuel S, Meço C, Melén E, Melioli G, Melo‐Gomes E, Meltzer EO, Menditto E, Menzies‐Gow A, Merk H, Michel J, Micheli Y, Miculinic N, Midão L, Mihaltan F, Mikos N, Milanese M, Milenkovic B, Mitsias D, Moalla B, Moda G, Mogica Martínez MD, Mohammad Y, Moin M, Molimard M, Momas I, Mommers M, Monaco A, Montefort S, Mora D, Morais‐Almeida M, Mösges R, Mostafa B, Mullol J, Münter L, Muraro A, Murray R, Musarra A, Mustakov T, Naclerio R, Nadeau KC, Nadif R, Nakonechna A, Namazova‐Baranova L, Navarro‐Locsin G, Neffen H, Nekam K, Neou A, Nettis E, Neuberger D, Nicod L, Nicola S, Niederberger‐Leppin V, Niedoszytko M, Nieto A, Novellino E, Nunes E, Nyembue D, O’Hehir R, Odjakova C, Ohta K, Okamoto Y, Okubo K, Oliver B, Onorato GL, Pia Orru M, Ouédraogo S, Ouoba K, Paggiaro PL, Pagkalos A, Pajno G, Pala G, Palaniappan S, Pali‐Schöll I, Palkonen S, Palmer S, Panaitescu Bunu C, Panzner P, Papadopoulos NG, Papanikolaou V, Papi A, Paralchev B, Paraskevopoulos G, Park H, Passalacqua G, Patella V, Pavord I, Pawankar R, Pedersen S, Peleve S, Pellegino S, Pereira A, Pérez T, Perna A, Peroni D, Pfaar O, Pham‐Thi N, Pigearias B, Pin I, Piskou K, Pitsios C, Plavec D, Poethig D, Pohl W, Poplas Susic A, Popov TA, Portejoie F, Potter P, Poulsen L, Prados‐Torres A, Prarros F, Price D, Prokopakis E, Puggioni F, Puig‐Domenech E, Puy R, Rabe K, Raciborski F, Ramos J, Recto MT, Reda SM, Regateiro FS, Reider N, Reitsma S, Repka‐Ramirez S, Ridolo E, Rimmer J, Rivero Yeverino D, Angelo Rizzo J, Robalo‐Cordeiro C, Roberts G, Roche N, Rodríguez González M, Rodríguez Zagal E, Rolla G, Rolland C, Roller‐Wirnsberger R, Roman Rodriguez M, Romano A, Romantowski J, Rombaux P, Romualdez J, Rosado‐Pinto J, Rosario N, Rosenwasser L, Rossi O, Rottem M, Rouadi P, Rovina N, Rozman Sinur I, Ruiz M, Ruiz Segura LT, Ryan D, Sagara H, Sakai D, Sakurai D, Saleh W, Salimaki J, Samitas K, Samolinski B, Sánchez Coronel MG, Sanchez‐Borges M, Sanchez‐Lopez J, Sarafoleanu C, Sarquis Serpa F, Sastre‐Dominguez J, Savi E, Sawaf B, Scadding GK, Scheire S, Schmid‐Grendelmeier P, Schuhl JF, Schunemann H, Schvalbová M, Schwarze J, Scichilone N, Senna G, Sepúlveda C, Serrano E, Shields M, Shishkov V, Siafakas N, Simeonov A, FER Simons E, Carlos Sisul J, Sitkauskiene B, Skrindo I, SokličKošak T, Solé D, Sooronbaev T, Soto‐Martinez M, Soto‐Quiros M, Sousa Pinto B, Sova M, Soyka M, Specjalski K, Spranger O, Stamataki S, Stefanaki L, Stellato C, Stelmach R, Strandberg T, Stute P, Subramaniam A, Suppli Ulrik C, Sutherland M, Sylvestre S, Syrigou A, Taborda Barata L, Takovska N, Tan R, Tan F, Tan V, Ping Tang I, Taniguchi M, Tannert L, Tantilipikorn P, Tattersall J, Tesi F, Thijs C, Thomas M, To T, Todo‐Bom A, Togias A, Tomazic P, Tomic‐Spiric V, Toppila‐Salmi S, Toskala E, Triggiani M, Triller N, Triller K, Tsiligianni I, Uberti M, Ulmeanu R, Urbancic J, Urrutia Pereira M, Vachova M, Valdés F, Valenta R, Valentin Rostan M, Valero A, Valiulis A, Vallianatou M, Valovirta E, Van Eerd M, Van Ganse E, Hage M, Vandenplas O, Vasankari T, Vassileva D, Velasco Munoz C, Ventura MT, Vera‐Munoz C, Vicheva D, Vichyanond P, Vidgren P, Viegi G, Vogelmeier C, Von Hertzen L, Vontetsianos T, Vourdas D, Tran Thien Quan V, Wagenmann M, Walker S, Wallace D, Wang DY, Waserman S, Wickman M, Williams S, Williams D, Wilson N, Wong G, Woo K, Wright J, Wroczynski P, Xepapadaki P, Yakovliev P, Yamaguchi M, Yan K, Yeow Yap Y, Yawn B, Yiallouros P, Yorgancioglu A, Yoshihara S, Young I, Yusuf OB, Zaidi A, Zaitoun F, Zar H, Zedda M, Zernotti ME, Zhang L, Zhong N, Zidarn M, Zubrinich C. Cabbage and fermented vegetables: From death rate heterogeneity in countries to candidates for mitigation strategies of severe COVID-19. Allergy 2021; 76:735-750. [PMID: 32762135 PMCID: PMC7436771 DOI: 10.1111/all.14549] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 07/31/2020] [Accepted: 08/04/2020] [Indexed: 12/20/2022]
Abstract
Large differences in COVID‐19 death rates exist between countries and between regions of the same country. Some very low death rate countries such as Eastern Asia, Central Europe, or the Balkans have a common feature of eating large quantities of fermented foods. Although biases exist when examining ecological studies, fermented vegetables or cabbage have been associated with low death rates in European countries. SARS‐CoV‐2 binds to its receptor, the angiotensin‐converting enzyme 2 (ACE2). As a result of SARS‐CoV‐2 binding, ACE2 downregulation enhances the angiotensin II receptor type 1 (AT1R) axis associated with oxidative stress. This leads to insulin resistance as well as lung and endothelial damage, two severe outcomes of COVID‐19. The nuclear factor (erythroid‐derived 2)‐like 2 (Nrf2) is the most potent antioxidant in humans and can block in particular the AT1R axis. Cabbage contains precursors of sulforaphane, the most active natural activator of Nrf2. Fermented vegetables contain many lactobacilli, which are also potent Nrf2 activators. Three examples are: kimchi in Korea, westernized foods, and the slum paradox. It is proposed that fermented cabbage is a proof‐of‐concept of dietary manipulations that may enhance Nrf2‐associated antioxidant effects, helpful in mitigating COVID‐19 severity.
Collapse
Affiliation(s)
- Jean Bousquet
- Charité Universitätsmedizin BerlinHumboldt‐Universität zu Berlin Berlin Germany
- Department of Dermatology and Allergy Berlin Institute of HealthComprehensive Allergy Center Berlin Germany
- MACVIA‐France and CHU Montpellier France
| | - Josep M. Anto
- Centre for Research in Environmental Epidemiology (CREAL) ISGlobAL Barcelona Spain
- IMIM (Hospital del Mar Research Institute) Barcelona Spain
- Universitat Pompeu Fabra (UPF) Barcelona Spain
- CIBER Epidemiología y Salud Pública (CIBERESP) Barcelona Spain
| | | | - Tari Haahtela
- Skin and Allergy Hospital Helsinki University Hospital University of Helsinki Finland
| | - Susana C. Fonseca
- Faculty of Sciences GreenUPorto ‐ Sustainable Agrifood Production Research Centre DGAOTUniversity of Porto Porto Portugal
| | - Guido Iaccarino
- Department of Advanced Biomedical Sciences Federico II University Napoli Italy
| | - Hubert Blain
- Department of Geriatrics Montpellier University hospital and MUSE Montpellier France
| | - Alain Vidal
- World Business Council for Sustainable Development (WBCSD) Geneva Switzerland
- AgroParisTech ‐ Paris Institute of Technology for Life, Food and Environmental Sciences Paris France
| | - Aziz Sheikh
- Usher Institute University of Edinburgh Scotland, UK
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Torsten Zuberbier
- Charité Universitätsmedizin BerlinHumboldt‐Universität zu Berlin Berlin Germany
- Department of Dermatology and Allergy Berlin Institute of HealthComprehensive Allergy Center Berlin Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Saboktakin-Rizi M, Alizadeh Behbahani B, Hojjati M, Noshad M. Identification of Lactobacillus plantarum TW29-1 isolated from Iranian fermented cereal-dairy product (Yellow Zabol Kashk): probiotic characteristics, antimicrobial activity and safety evaluation. JOURNAL OF FOOD MEASUREMENT AND CHARACTERIZATION 2021. [DOI: 10.1007/s11694-021-00846-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
67
|
Marco ML. Defining how microorganisms benefit human health. Microb Biotechnol 2021; 14:35-40. [PMID: 33099885 PMCID: PMC7888441 DOI: 10.1111/1751-7915.13685] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 10/05/2020] [Indexed: 12/19/2022] Open
Abstract
An appreciation for how microorganisms can benefit human health has grown over the past century. The future of this research will be to identify the specific microbial enzymatic pathways and molecules necessary for health promotion. Some of these 'beneficial factors' are already known for probiotics and species in the human microbiome, however, precise descriptions of the mechanistic details for their effects remain to be discovered. The need for this research is elevated by the potential use of microorganisms for preventing and treating the non-communicable diseases which are now the leading causes of death worldwide.
Collapse
Affiliation(s)
- Maria L. Marco
- Department of Food Science and TechnologyUniversity of California, DavisDavisCA95616USA
| |
Collapse
|
68
|
Leech J, Cabrera-Rubio R, Walsh AM, Macori G, Walsh CJ, Barton W, Finnegan L, Crispie F, O'Sullivan O, Claesson MJ, Cotter PD. Fermented-Food Metagenomics Reveals Substrate-Associated Differences in Taxonomy and Health-Associated and Antibiotic Resistance Determinants. mSystems 2020; 5:e00522-20. [PMID: 33172966 PMCID: PMC7657593 DOI: 10.1128/msystems.00522-20] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/21/2020] [Indexed: 02/07/2023] Open
Abstract
Fermented foods have been the focus of ever greater interest as a consequence of purported health benefits. Indeed, it has been suggested that consumption of these foods helps to address the negative consequences of "industrialization" of the human gut microbiota in Western society. However, as the mechanisms via which the microbes in fermented foods improve health are not understood, it is necessary to develop an understanding of the composition and functionality of the fermented-food microbiota to better harness desirable traits. Here, we considerably expand the understanding of fermented-food microbiomes by employing shotgun metagenomic sequencing to provide a comprehensive insight into the microbial composition, diversity, and functional potential (including antimicrobial resistance and carbohydrate-degrading and health-associated gene content) of a diverse range of 58 fermented foods from artisanal producers from a number of countries. Food type, i.e., dairy-, sugar-, or brine-type fermented foods, was the primary driver of microbial composition, with dairy foods found to have the lowest microbial diversity. From the combined data set, 127 high-quality metagenome-assembled genomes (MAGs), including 10 MAGs representing putatively novel species of Acetobacter, Acidisphaera, Gluconobacter, Companilactobacillus, Leuconostoc, and Rouxiella, were generated. Potential health promoting attributes were more common in fermented foods than nonfermented equivalents, with water kefirs, sauerkrauts, and kvasses containing the greatest numbers of potentially health-associated gene clusters. Ultimately, this study provides the most comprehensive insight into the microbiomes of fermented foods to date and yields novel information regarding their relative health-promoting potential.IMPORTANCE Fermented foods are regaining popularity worldwide due in part to a greater appreciation of the health benefits of these foods and the associated microorganisms. Here, we use state-of-the-art approaches to explore the microbiomes of 58 of these foods, identifying the factors that drive the microbial composition of these foods and potential functional benefits associated with these populations. Food type, i.e., dairy-, sugar-, or brine-type fermented foods, was the primary driver of microbial composition, with dairy foods found to have the lowest microbial diversity and, notably, potential health promoting attributes were more common in fermented foods than nonfermented equivalents. The information provided here will provide significant opportunities for the further optimization of fermented-food production and the harnessing of their health-promoting potential.
Collapse
Affiliation(s)
- John Leech
- Teagasc Food Research Centre, Fermoy, Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Raul Cabrera-Rubio
- Teagasc Food Research Centre, Fermoy, Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | | | - Guerrino Macori
- Teagasc Food Research Centre, Fermoy, Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Calum J Walsh
- Teagasc Food Research Centre, Fermoy, Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Wiley Barton
- Teagasc Food Research Centre, Fermoy, Cork, Ireland
| | - Laura Finnegan
- Teagasc Food Research Centre, Fermoy, Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Fiona Crispie
- Teagasc Food Research Centre, Fermoy, Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Orla O'Sullivan
- Teagasc Food Research Centre, Fermoy, Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Marcus J Claesson
- Teagasc Food Research Centre, Fermoy, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Paul D Cotter
- Teagasc Food Research Centre, Fermoy, Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
| |
Collapse
|
69
|
Ning Y, Fu Y, Hou L, Ma M, Wang Z, Li X, Jia Y. iTRAQ-based quantitative proteomic analysis of synergistic antibacterial mechanism of phenyllactic acid and lactic acid against Bacillus cereus. Food Res Int 2020; 139:109562. [PMID: 33509445 DOI: 10.1016/j.foodres.2020.109562] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/14/2020] [Accepted: 07/17/2020] [Indexed: 11/15/2022]
Abstract
Phenyllactic acid (PLA) as a phenolic acid by lactic acid (LA) bacteria shows enhanced antibacterial activity when coexisting with LA, while the antibacterial mechanism of PLA combined with LA was unknown. Hence, the antibacterial mechanism of PLA and LA was investigated against Bacillus cereus. Flow cytometry and TEM analysis demonstrated that single PLA and LA disrupted the membrane integrity and the morphology, while combined PLA and LA synergistically enhanced the damage. iTRAQ-based proteomic analysis suggested that PLA down-regulated kdpB and inhibited K+ transport, disturbed the function of ribosome and expression of competence genes; LA down-regulated periplasmic phosphorus-binding proteins and inhibited phosphorus transport, disturbed the function of ribosome, TCA cycle, as well as purine and pyrimidine metabolism; and combined PLA and LA inhibited K+ and phosphorus transport, and influenced the synthesis of purine and pyrimidine metabolism. The investigation could provide some insights into the application of PLA in food preservation.
Collapse
Affiliation(s)
- Yawei Ning
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing 100048, China; College of Bioscience and Bioengineering, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Yunan Fu
- College of Bioscience and Bioengineering, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Linlin Hou
- College of Bioscience and Bioengineering, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Mengge Ma
- College of Bioscience and Bioengineering, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Zhixin Wang
- College of Bioscience and Bioengineering, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Xingfeng Li
- College of Bioscience and Bioengineering, Hebei University of Science and Technology, Shijiazhuang 050018, China.
| | - Yingmin Jia
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing 100048, China.
| |
Collapse
|
70
|
Correlation between the Antimicrobial Activity and Metabolic Profiles of Cell Free Supernatants and Membrane Vesicles Produced by Lactobacillus reuteri DSM 17938. Microorganisms 2020; 8:microorganisms8111653. [PMID: 33114410 PMCID: PMC7692313 DOI: 10.3390/microorganisms8111653] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/16/2020] [Accepted: 10/20/2020] [Indexed: 12/15/2022] Open
Abstract
The aim of the work is to assess the antimicrobial activities of Cell Free Supernatants (CFS) and Membrane Vesicles (MVs), produced by Lactobacillus reuteri DSM 17938, versus Gram-positive and Gram-negative bacteria and investigate their metabolic profiles. The Minimum Inhibitory Concentration was determined through the broth microdilution method and cell proliferation assay while the Minimum Bactericidal Concentration was determined by Colony Forming Units counts. The characteristics of the antimicrobial compounds were evaluated by pH adjustments, proteinase treatment, and size fractionation of the CFS. The cytotoxicity of CFS was tested on two human cell lines. A detailed snapshot of the L. reuteri metabolism was attained through an untargeted metabolic profiling by means of high resolution Fourier Transform Ion Cyclotron Resonance Mass Spectrometry (FT-ICR MS) coupled with Electrospray Ionization Source (ESI). The results showed (i) a greater efficacy of CFS and its fractions towards Gram-negative compared to Gram-positive bacteria; (ii) an antimicrobial effect related to pH-dependent compounds but not to MVs; (iii) a molecular weight < 3 KDa as well as an a non-proteinaceous nature of the antimicrobial compounds; and (iv) more than 200 and 500 putative metabolites annotated in MVs and supernatants, covering several classes of metabolites, including amino acids, lipids, fatty and organic acids, polyalcohols, nucleotides, and vitamins. Some putative compounds were proposed not only as characteristic of specific fractions, but also possibly involved in antimicrobial activity.
Collapse
|
71
|
Schirmer B, Giehl K, Kubatzky KF. Report of the 23rd Meeting on Signal Transduction 2019-Trends in Cancer and Infection. Int J Mol Sci 2020; 21:ijms21082728. [PMID: 32326408 PMCID: PMC7215334 DOI: 10.3390/ijms21082728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 04/06/2020] [Indexed: 11/16/2022] Open
Abstract
The annual meeting "Signal Transduction-Receptors, Mediators and Genes" of the Signal Transduction Society (STS) is an interdisciplinary conference open to all scientists sharing the common interest in elucidating the signalling pathways underlying the physiological or pathological processes in health and disease of humans, animals, plants, fungi, prokaryotes and protists. The 23rd meeting on signal transduction was held from 4-6 November 2019 in Weimar, Germany, and focused on "Trends in Cancer and Infection". As usual, keynote presentations by invited scientists introduced the respective workshops and were followed by speakers chosen from the submitted abstracts. Ample time had been reserved for discussion of the presented data during the workshops. In this report, we provide a concise summary of the various workshops and further aspects of the scientific program.
Collapse
Affiliation(s)
- Bastian Schirmer
- Institute of Pharmacology, Hannover Medical School, 30625 Hannover, Germany
- Correspondence: ; Tel.: +49-511-532-3875
| | - Klaudia Giehl
- Signal Transduction of Cellular Motility, Internal Medicine V, Justus Liebig University Giessen, 35392 Giessen, Germany;
| | - Katharina F. Kubatzky
- Department of Medical Microbiology and Hygiene, Heidelberg University Hospital, 69120 Heidelberg, Germany;
| |
Collapse
|
72
|
Taylor BC, Lejzerowicz F, Poirel M, Shaffer JP, Jiang L, Aksenov A, Litwin N, Humphrey G, Martino C, Miller-Montgomery S, Dorrestein PC, Veiga P, Song SJ, McDonald D, Derrien M, Knight R. Consumption of Fermented Foods Is Associated with Systematic Differences in the Gut Microbiome and Metabolome. mSystems 2020; 5:e00901-19. [PMID: 32184365 PMCID: PMC7380580 DOI: 10.1128/msystems.00901-19] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 02/19/2020] [Indexed: 12/22/2022] Open
Abstract
Lifestyle factors, such as diet, strongly influence the structure, diversity, and composition of the microbiome. While we have witnessed over the last several years a resurgence of interest in fermented foods, no study has specifically explored the effects of their consumption on gut microbiota in large cohorts. To assess whether the consumption of fermented foods is associated with a systematic signal in the gut microbiome and metabolome, we used a multi-omic approach (16S rRNA amplicon sequencing, metagenomic sequencing, and untargeted mass spectrometry) to analyze stool samples from 6,811 individuals from the American Gut Project, including 115 individuals specifically recruited for their frequency of fermented food consumption for a targeted 4-week longitudinal study. We observed subtle but statistically significant differences between consumers and nonconsumers in beta diversity as well as differential taxa between the two groups. We found that the metabolome of fermented food consumers was enriched with conjugated linoleic acid (CLA), a putatively health-promoting molecule. Cross-omic analyses between metagenomic sequencing and mass spectrometry suggest that CLA may be driven by taxa associated with fermented food consumers. Collectively, we found modest yet persistent signatures associated with fermented food consumption that appear present in multiple -omic types which motivate further investigation of how different types of fermented food impact the gut microbiome and overall health.IMPORTANCE Public interest in the effects of fermented food on the human gut microbiome is high, but limited studies have explored the association between fermented food consumption and the gut microbiome in large cohorts. Here, we used a combination of omics-based analyses to study the relationship between the microbiome and fermented food consumption in thousands of people using both cross-sectional and longitudinal data. We found that fermented food consumers have subtle differences in their gut microbiota structure, which is enriched in conjugated linoleic acid, thought to be beneficial. The results suggest that further studies of specific kinds of fermented food and their impacts on the microbiome and health will be useful.
Collapse
Affiliation(s)
- Bryn C Taylor
- Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, California, USA
| | - Franck Lejzerowicz
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, California, USA
| | - Marion Poirel
- IT&M Innovation on behalf of Danone Nutricia Research, Palaiseau, France
| | - Justin P Shaffer
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, California, USA
| | - Lingjing Jiang
- Division of Biostatistics, University of California San Diego, La Jolla, California, USA
| | - Alexander Aksenov
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA
| | - Nicole Litwin
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California, USA
| | - Gregory Humphrey
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, California, USA
| | - Cameron Martino
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, California, USA
| | - Sandrine Miller-Montgomery
- Department of Bioengineering, Jacobs School of Engineering, University of California San Diego, La Jolla, California, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California, USA
| | - Pieter C Dorrestein
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, California, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California, USA
| | | | - Se Jin Song
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California, USA
| | - Daniel McDonald
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, California, USA
| | | | - Rob Knight
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, California, USA
- Department of Bioengineering, Jacobs School of Engineering, University of California San Diego, La Jolla, California, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California, USA
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
73
|
Peters A, Rabe P, Krumbholz P, Kalwa H, Kraft R, Schöneberg T, Stäubert C. Natural biased signaling of hydroxycarboxylic acid receptor 3 and G protein-coupled receptor 84. Cell Commun Signal 2020; 18:31. [PMID: 32102673 PMCID: PMC7045412 DOI: 10.1186/s12964-020-0516-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/29/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Medium-chain fatty acids and their 3-hydroxy derivatives are metabolites endogenously produced in humans, food-derived or originating from bacteria. They activate G protein-coupled receptors, including GPR84 and HCA3, which regulate metabolism and immune functions. Although both receptors are coupled to Gi proteins, share at least one agonist and show overlapping tissue expression, GPR84 exerts pro-inflammatory effects whereas HCA3 is involved in anti-inflammatory responses. Here, we analyzed signaling kinetics of both HCA3 and GPR84, to unravel signal transduction components that may explain their physiological differences. METHODS To study the signaling kinetics and components involved in signal transduction of both receptors we applied the label-free dynamic mass redistribution technology in combination with classical cAMP, ERK signaling and β-arrestin-2 recruitment assays. For phenotypical analyses, we used spheroid cell culture models. RESULTS We present strong evidence for a natural biased signaling of structurally highly similar agonists at HCA3 and GPR84. We show that HCA3 signaling and trafficking depends on dynamin-2 function. Activation of HCA3 by 3-hydroxyoctanoic acid but not 3-hydroxydecanoic acid leads to β-arrestin-2 recruitment, which is relevant for cell-cell adhesion. GPR84 stimulation with 3-hydroxydecanoic acid causes a sustained ERK activation but activation of GPR84 is not followed by β-arrestin-2 recruitment. CONCLUSIONS In summary, our results highlight that biased agonism is a physiological property of HCA3 and GPR84 with relevance for innate immune functions potentially to differentiate between endogenous, non-pathogenic compounds and compounds originating from e.g. pathogenic bacteria. Video Abstract.
Collapse
Affiliation(s)
- Anna Peters
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Johannisallee 30, 04103, Leipzig, Germany
| | - Philipp Rabe
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Johannisallee 30, 04103, Leipzig, Germany
| | - Petra Krumbholz
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Johannisallee 30, 04103, Leipzig, Germany
| | - Hermann Kalwa
- Rudolf Boehm Institute of Pharmacology and Toxicology, Medical Faculty, Leipzig University, Härtelstraße 16-18, 04107, Leipzig, Germany
| | - Robert Kraft
- Carl Ludwig Institute for Physiology, Medical Faculty, Leipzig University, 04103, Leipzig, Germany
| | - Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Johannisallee 30, 04103, Leipzig, Germany
| | - Claudia Stäubert
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Johannisallee 30, 04103, Leipzig, Germany.
| |
Collapse
|
74
|
Amato KR, Mallott EK, McDonald D, Dominy NJ, Goldberg T, Lambert JE, Swedell L, Metcalf JL, Gomez A, Britton GAO, Stumpf RM, Leigh SR, Knight R. Convergence of human and Old World monkey gut microbiomes demonstrates the importance of human ecology over phylogeny. Genome Biol 2019; 20:201. [PMID: 31590679 PMCID: PMC6781418 DOI: 10.1186/s13059-019-1807-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 08/29/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Comparative data from non-human primates provide insight into the processes that shaped the evolution of the human gut microbiome and highlight microbiome traits that differentiate humans from other primates. Here, in an effort to improve our understanding of the human microbiome, we compare gut microbiome composition and functional potential in 14 populations of humans from ten nations and 18 species of wild, non-human primates. RESULTS Contrary to expectations from host phylogenetics, we find that human gut microbiome composition and functional potential are more similar to those of cercopithecines, a subfamily of Old World monkey, particularly baboons, than to those of African apes. Additionally, our data reveal more inter-individual variation in gut microbiome functional potential within the human species than across other primate species, suggesting that the human gut microbiome may exhibit more plasticity in response to environmental variation compared to that of other primates. CONCLUSIONS Given similarities of ancestral human habitats and dietary strategies to those of baboons, these findings suggest that convergent ecologies shaped the gut microbiomes of both humans and cercopithecines, perhaps through environmental exposure to microbes, diet, and/or associated physiological adaptations. Increased inter-individual variation in the human microbiome may be associated with human dietary diversity or the ability of humans to inhabit novel environments. Overall, these findings show that diet, ecology, and physiological adaptations are more important than host-microbe co-diversification in shaping the human microbiome, providing a key foundation for comparative analyses of the role of the microbiome in human biology and health.
Collapse
Affiliation(s)
- Katherine R. Amato
- Department of Anthropology, Northwestern University, 1810 Hinman Ave, Evanston, IL 60208 USA
| | - Elizabeth K. Mallott
- Department of Anthropology, Northwestern University, 1810 Hinman Ave, Evanston, IL 60208 USA
| | - Daniel McDonald
- Department of Pediatrics, University of California San Diego, San Diego, 92093 USA
- Center for Microbiome Innovation, University of California San Diego, San Diego, 92093 USA
| | | | - Tony Goldberg
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, 53706 USA
| | - Joanna E. Lambert
- Department of Ecology and Evolutionary Biology, University of Colorado Boulder, Boulder, 80302 USA
| | - Larissa Swedell
- Department of Anthropology, City University of New York - Queens College, New York, 11367 USA
| | - Jessica L. Metcalf
- Department of Animal Sciences, Colorado State University, Fort Collins, 80521 USA
| | - Andres Gomez
- Department of Animal Sciences, University of Minnesota, Minneapolis, 55108 USA
| | | | - Rebecca M. Stumpf
- Department of Anthropology, University of Illinois at Urbana-Champaign, Urbana, 61801 USA
| | - Steven R. Leigh
- Department of Anthropology, University of Colorado Boulder, Boulder, 80302 USA
| | - Rob Knight
- Department of Pediatrics, University of California San Diego, San Diego, 92093 USA
- Center for Microbiome Innovation, University of California San Diego, San Diego, 92093 USA
- Department of Computer Science and Engineering, University of California San Diego, San Diego, 92093 USA
- Department of Bioengineering, University of California San Diego, San Diego, 92093 USA
| |
Collapse
|
75
|
Production of Indole-3-Lactic Acid by Bifidobacterium Strains Isolated fromHuman Infants. Microorganisms 2019; 7:microorganisms7090340. [PMID: 31514325 PMCID: PMC6780619 DOI: 10.3390/microorganisms7090340] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/07/2019] [Accepted: 09/09/2019] [Indexed: 01/03/2023] Open
Abstract
Recent studies have shown that metabolites produced by microbes can be considered as mediators of host-microbial interactions. In this study, we examined the production of tryptophan metabolites by Bifidobacterium strains found in the gastrointestinal tracts of humans and other animals. Indole-3-lactic acid (ILA) was the only tryptophan metabolite produced in bifidobacteria culture supernatants. No others, including indole-3-propionic acid, indole-3-acetic acid, and indole-3-aldehyde, were produced. Strains of bifidobacterial species commonly isolated from the intestines of human infants, such as Bifidobacterium longum subsp. longum, Bifidobacterium longum subsp. infantis, Bifidobacterium breve, and Bifidobacterium bifidum, produced higher levels of ILA than did strains of other species. These results imply that infant-type bifidobacteria might play a specific role in host-microbial cross-talk by producing ILA in human infants.
Collapse
|
76
|
Peters A, Krumbholz P, Jäger E, Heintz-Buschart A, Çakir MV, Rothemund S, Gaudl A, Ceglarek U, Schöneberg T, Stäubert C. Correction: Metabolites of lactic acid bacteria present in fermented foods are highly potent agonists of human hydroxycarboxylic acid receptor 3. PLoS Genet 2019; 15:e1008283. [PMID: 31323042 PMCID: PMC6641466 DOI: 10.1371/journal.pgen.1008283] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|