51
|
Isidori M, Corbee RJ, Trabalza-Marinucci M. Nonpharmacological Treatment Strategies for the Management of Canine Chronic Inflammatory Enteropathy—A Narrative Review. Vet Sci 2022; 9:vetsci9020037. [PMID: 35202290 PMCID: PMC8878421 DOI: 10.3390/vetsci9020037] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/15/2022] [Accepted: 01/17/2022] [Indexed: 02/04/2023] Open
Abstract
Chronic inflammatory enteropathy (CIE) refers to a heterogeneous group of idiopathic diseases of the dog characterised by persistent gastrointestinal (GI) clinical signs. If conventional dietary treatment alone would be unsuccessful, management of CIE is traditionally attained by the use of pharmaceuticals, such as antibiotics and immunosuppressive drugs. While being rather effective, however, these drugs are endowed with side effects, which may impact negatively on the animal’s quality of life. Therefore, novel, safe and effective therapies for CIE are highly sought after. As gut microbiota imbalances are often associated with GI disorders, a compelling rationale exists for the use of nonpharmacological methods of microbial manipulation in CIE, such as faecal microbiota transplantation and administration of pre-, pro-, syn- and postbiotics. In addition to providing direct health benefits to the host via a gentle modulation of the intestinal microbiota composition and function, these treatments may also possess immunomodulatory and epithelial barrier-enhancing actions. Likewise, intestinal barrier integrity, along with mucosal inflammation, are deemed to be two chief therapeutic targets of mesenchymal stem cells and selected vegetable-derived bioactive compounds. Although pioneering studies have revealed encouraging findings regarding the use of novel treatment agents in CIE, a larger body of research is needed to address fully their mode of action, efficacy and safety.
Collapse
Affiliation(s)
- Marco Isidori
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy;
- Correspondence:
| | - Ronald Jan Corbee
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Yalelaan 108, 3584 CM Utrecht, The Netherlands;
| | | |
Collapse
|
52
|
Melim C, Lauro MR, Pires IM, Oliveira PJ, Cabral C. The Role of Glucosinolates from Cruciferous Vegetables (Brassicaceae) in Gastrointestinal Cancers: From Prevention to Therapeutics. Pharmaceutics 2022; 14:pharmaceutics14010190. [PMID: 35057085 PMCID: PMC8777706 DOI: 10.3390/pharmaceutics14010190] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/31/2021] [Accepted: 01/10/2022] [Indexed: 02/04/2023] Open
Abstract
The gastrointestinal (GI) tract is composed of rapidly renewing cells, which increase the likelihood of cancer. Colorectal cancer is one of the most frequently diagnosed GI cancers and currently stands in second place regarding cancer-related mortality. Unfortunately, the treatment of GI is limited, and few developments have occurred in the field over the years. With this in mind, new therapeutic strategies involving biologically active phytocompounds are being evaluated as anti-cancer agents. Vegetables such as broccoli, brussels sprouts, cabbage, cauliflower, and radish, all belonging to the Brassicaceae family, are high in dietary fibre, minerals, vitamins, carotenoids, polyphenols, and glucosinolates. The latter compound is a secondary metabolite characteristic of this family and, when biologically active, has demonstrated anti-cancer properties. This article reviews the literature regarding the potential of Cruciferous vegetables in the prevention and/or treatment of GI cancers and the relevance of appropriate compound formulations for improving the stability and bioaccessibility of the major Cruciferous compounds, with a particular focus on glucosinolates.
Collapse
Affiliation(s)
- Catarina Melim
- Faculty of Medicine, Clinic Academic Center of Coimbra (CACC), Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, 3000-548 Coimbra, Portugal;
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal;
| | - Maria R. Lauro
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 84084 Fisciano, Italy;
| | - Isabel M. Pires
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Hull, Hull HU6 7RX, UK;
| | - Paulo J. Oliveira
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal;
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Célia Cabral
- Faculty of Medicine, Clinic Academic Center of Coimbra (CACC), Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, 3000-548 Coimbra, Portugal;
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal;
- Center for Functional Ecology, Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- Correspondence: ; Tel.: +351-239-480-066
| |
Collapse
|
53
|
Mortaz E, Nomani M, Adcock I, Folkerts G, Garssen J. Galactooligosaccharides (GOS) and 2′-fucosyllactose (2′-FL) can directly suppress growth of specific pathogenic microbes and impact phagocytosis of neutrophils. Nutrition 2022; 96:111601. [DOI: 10.1016/j.nut.2022.111601] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 01/03/2022] [Accepted: 01/09/2022] [Indexed: 11/16/2022]
|
54
|
Ho LH, Tan TC, Chong LC. Designer foods as an effective approach to enhance disease preventative properties of food through its health functionalities. FUTURE FOODS 2022. [DOI: 10.1016/b978-0-323-91001-9.00031-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
55
|
Ambrogi V, Bottacini F, Mac Sharry J, van Breen J, O'Keeffe E, Walsh D, Schoemaker B, Cao L, Kuipers B, Lindner C, Jimeno ML, Doyagüez EG, Hernandez-Hernandez O, Moreno FJ, Schoterman M, van Sinderen D. Bifidobacterial β-Galactosidase-Mediated Production of Galacto-Oligosaccharides: Structural and Preliminary Functional Assessments. Front Microbiol 2021; 12:750635. [PMID: 34777303 PMCID: PMC8581567 DOI: 10.3389/fmicb.2021.750635] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/27/2021] [Indexed: 11/13/2022] Open
Abstract
In the current study the ability of four previously characterized bifidobacterial β-galactosidases (designated here as BgaA, BgaC, BgaD, and BgaE) to produce galacto-oligosaccharides (GOS) was optimized. Of these enzymes, BgaA and BgaE were found to be promising candidates for GOS production (and the corresponding GOS mixtures were called GOS-A and GOS-E, respectively) with a GOS concentration of 19.0 and 40.3% (of the initial lactose), respectively. GOS-A and GOS-E were partially purified and structurally characterized. NMR analysis revealed that the predominant (non-lactose) disaccharide was allo-lactose in both purified GOS preparations. The predominant trisaccharide in GOS-A and GOS-E was shown to be 3′-galactosyllactose, with lower levels of 6′-galactosyllactose and 4′-galactosyllactose. These three oligosaccharides have also been reported to occur in human milk. Purified GOS-A and GOS-E were shown to be able to support bifidobacterial growth similar to a commercially available GOS. In addition, GOS-E and the commercially available GOS were shown to be capable of reducing Escherichia coli adhesion to a C2BBe1 cell line. Both in vitro bifidogenic activity and reduced E. coli adhesion support the prebiotic potential of GOS-E and GOS-A.
Collapse
Affiliation(s)
- Valentina Ambrogi
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,School of Microbiology, University College Cork, Cork, Ireland
| | - Francesca Bottacini
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Biological Sciences, Munster Technological University, Cork, Ireland
| | - John Mac Sharry
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,School of Microbiology, University College Cork, Cork, Ireland.,School of Medicine, University College Cork, Cork, Ireland
| | | | - Ellen O'Keeffe
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Dan Walsh
- School of Microbiology, University College Cork, Cork, Ireland
| | | | - Linqiu Cao
- FrieslandCampina, Amersfoort, Netherlands
| | | | | | | | | | - Oswaldo Hernandez-Hernandez
- Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - F Javier Moreno
- Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | | | - Douwe van Sinderen
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,School of Microbiology, University College Cork, Cork, Ireland
| |
Collapse
|
56
|
The role of dietary proteins and carbohydrates in gut microbiome composition and activity: A review. Food Hydrocoll 2021. [DOI: 10.1016/j.foodhyd.2021.106911] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
57
|
Ambrogi V, Bottacini F, Cao L, Kuipers B, Schoterman M, van Sinderen D. Galacto-oligosaccharides as infant prebiotics: production, application, bioactive activities and future perspectives. Crit Rev Food Sci Nutr 2021; 63:753-766. [PMID: 34477457 DOI: 10.1080/10408398.2021.1953437] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Galacto-oligosaccharides (GOS) are non-digestible oligosaccharides characterized by a mix of structures that vary in their degree of polymerization (DP) and glycosidic linkage between the galactose moieties or between galactose and glucose. They have enjoyed extensive scientific scrutiny, and their health-promoting effects are supported by a large number of scientific and clinical studies. A variety of GOS-associated health-promoting effects have been reported, such as growth promotion of beneficial bacteria, in particular bifidobacteria and lactobacilli, inhibition of pathogen adhesion and improvement of gut barrier function. GOS have attracted significant interest from food industries for their versatility as a bioactive ingredient and in particular as a functional component of infant formulations. These oligosaccharides are produced in a kinetically-controlled reaction involving lactose transgalactosylation, being catalyzed by particular β-galactosidases of bacterial or fungal origin. Despite the well-established technology applied for GOS production, this process may still meet with technological challenges when employed at an industrial scale. The current review will cover relevant scientific literature on the beneficial physiological properties of GOS as a prebiotic for the infant gut microbiota, details of GOS structures, the associated reaction mechanism of β-galactosidase, and its (large-scale) production.
Collapse
Affiliation(s)
- Valentina Ambrogi
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Francesca Bottacini
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Biological Sciences, Munster Technological University, Cork, Ireland
| | - Linqiu Cao
- FrieslandCampina, Amersfoort, The Netherlands
| | - Bas Kuipers
- FrieslandCampina, Amersfoort, The Netherlands
| | | | - Douwe van Sinderen
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
58
|
Application of chia (Salvia hispanica) mucilage as an ingredient replacer in foods. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.06.039] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
59
|
Xue Y, Zhu MJ. Unraveling enterohemorrhagic Escherichia coli infection: the promising role of dietary compounds and probiotics in bacterial elimination and host innate immunity boosting. Crit Rev Food Sci Nutr 2021; 63:1551-1563. [PMID: 34404306 DOI: 10.1080/10408398.2021.1965538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The innate immune system has developed sophisticated strategies to defense against infections. Host cells utilize the recognition machineries such as toll-like receptors and nucleotide binding and oligomerization domain-like receptors to identify the pathogens and alert immune system. However, some pathogens have developed tactics to evade host defenses, including manipulation of host inflammatory response, interference with cell death pathway, and highjack of phagocytosis signaling for a better survival and colonization in host. Enterohemorrhagic Escherichia coli (EHEC) is a notorious foodborne pathogen that causes severe tissue damages and gastrointestinal diseases, which has been reported to disturb host immune responses. Diverse bioactive compounds such as flavonoids, phenolic acids, alkaloids, saccharides, and terpenoids derived from food varieties and probiotics have been discovered and investigated for their capability of combating bacterial infections. Some of them serve as novel antimicrobial agents and act as immune boosters that harness host immune system. In this review, we will discuss how EHEC, specifically E. coli O157:H7, hijacks the host immune system and interferes with host signaling pathway; and highlight the promising role of food-derived bioactive compounds and probiotics in harnessing host innate immunity and eliminating E. coli O157:H7 infection with multiple strategies.
Collapse
Affiliation(s)
- Yansong Xue
- Key Laboratory of Functional Dairy, Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Mei-Jun Zhu
- School of Food Science, Washington State University, Pullman, WA, USA
| |
Collapse
|
60
|
Sauvaitre T, Etienne-Mesmin L, Sivignon A, Mosoni P, Courtin CM, Van de Wiele T, Blanquet-Diot S. Tripartite relationship between gut microbiota, intestinal mucus and dietary fibers: towards preventive strategies against enteric infections. FEMS Microbiol Rev 2021; 45:5918835. [PMID: 33026073 DOI: 10.1093/femsre/fuaa052] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023] Open
Abstract
The human gut is inhabited by a large variety of microorganims involved in many physiological processes and collectively referred as to gut microbiota. Disrupted microbiome has been associated with negative health outcomes and especially could promote the onset of enteric infections. To sustain their growth and persistence within the human digestive tract, gut microbes and enteric pathogens rely on two main polysaccharide compartments, namely dietary fibers and mucus carbohydrates. Several evidences suggest that the three-way relationship between gut microbiota, dietary fibers and mucus layer could unravel the capacity of enteric pathogens to colonise the human digestive tract and ultimately lead to infection. The review starts by shedding light on similarities and differences between dietary fibers and mucus carbohydrates structures and functions. Next, we provide an overview of the interactions of these two components with the third partner, namely, the gut microbiota, under health and disease situations. The review will then provide insights into the relevance of using dietary fibers interventions to prevent enteric infections with a focus on gut microbial imbalance and impaired-mucus integrity. Facing the numerous challenges in studying microbiota-pathogen-dietary fiber-mucus interactions, we lastly describe the characteristics and potentialities of currently available in vitro models of the human gut.
Collapse
Affiliation(s)
- Thomas Sauvaitre
- Université Clermont Auvergne, UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Clermont-Ferrand, France.,Ghent University, Faculty of Bioscience Engineering, Center for Microbial Ecology and Technology (CMET), Ghent, Belgium
| | - Lucie Etienne-Mesmin
- Université Clermont Auvergne, UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Clermont-Ferrand, France
| | - Adeline Sivignon
- Université Clermont Auvergne, UMR 1071 Inserm, USC-INRAe 2018, Microbes, Intestin, Inflammation et Susceptibilité de l'Hôte (M2iSH), Clermont-Ferrand, France
| | - Pascale Mosoni
- Université Clermont Auvergne, UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Clermont-Ferrand, France
| | - Christophe M Courtin
- KU Leuven, Faculty of Bioscience Engineering, Laboratory of Food Chemistry and Biochemistry & Leuven Food Science and Nutrition Research Centre (LFoRCe), Leuven, Belgium
| | - Tom Van de Wiele
- Ghent University, Faculty of Bioscience Engineering, Center for Microbial Ecology and Technology (CMET), Ghent, Belgium
| | - Stéphanie Blanquet-Diot
- Université Clermont Auvergne, UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Clermont-Ferrand, France
| |
Collapse
|
61
|
Abdul Rahman R, Lamarca A, Hubner RA, Valle JW, McNamara MG. The Microbiome as a Potential Target for Therapeutic Manipulation in Pancreatic Cancer. Cancers (Basel) 2021; 13:cancers13153779. [PMID: 34359684 PMCID: PMC8345056 DOI: 10.3390/cancers13153779] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/19/2021] [Accepted: 07/22/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Pancreatic cancer is one of the most lethal cancers. It is a difficult cancer to treat, and the complexity surrounding the pancreatic tumour is one of the contributing factors. The microbiome is the collection of microorganisms within an environment and its genetic material. They reside on body surfaces and most abundantly within the human gut in symbiotic balance with their human host. Disturbance in the balance can lead to many diseases, including cancers. Significant advances have been made in cancer treatment since the introduction of immunotherapy, and the microbiome may play a part in the outcome and survival of patients with cancer, especially those treated with immunotherapy. Immunotherapy use in pancreatic cancer remains challenging. This review will focus on the potential interaction of the microbiome with pancreas cancer and how this could be manipulated. Abstract Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers and is projected to be the second most common cause of cancer-related death by 2030, with an overall 5-year survival rate between 7% and 9%. Despite recent advances in surgical, chemotherapy, and radiotherapy techniques, the outcome for patients with PDAC remains poor. Poor prognosis is multifactorial, including the likelihood of sub-clinical metastatic disease at presentation, late-stage at presentation, absence of early and reliable diagnostic biomarkers, and complex biology surrounding the extensive desmoplastic PDAC tumour micro-environment. Microbiota refers to all the microorganisms found in an environment, whereas microbiome is the collection of microbiota and their genome within an environment. These organisms reside on body surfaces and within mucosal layers, but are most abundantly found within the gut. The commensal microbiome resides in symbiosis in healthy individuals and contributes to nutritive, metabolic and immune-modulation to maintain normal health. Dysbiosis is the perturbation of the microbiome that can lead to a diseased state, including inflammatory bowel conditions and aetiology of cancer, such as colorectal and PDAC. Microbes have been linked to approximately 10% to 20% of human cancers, and they can induce carcinogenesis by affecting a number of the cancer hallmarks, such as promoting inflammation, avoiding immune destruction, and microbial metabolites can deregulate host genome stability preceding cancer development. Significant advances have been made in cancer treatment since the advent of immunotherapy. The microbiome signature has been linked to response to immunotherapy and survival in many solid tumours. However, progress with immunotherapy in PDAC has been challenging. Therefore, this review will focus on the available published evidence of the microbiome association with PDAC and explore its potential as a target for therapeutic manipulation.
Collapse
Affiliation(s)
- Rozana Abdul Rahman
- Experimental Cancer Medicine Team, The Christie NHS Foundation Trust, Manchester M20 4BX, UK;
| | - Angela Lamarca
- Department of Medical Oncology, The Christie NHS Foundation Trust/Division of Cancer Sciences, University of Manchester, Manchester M20 4BX, UK; (A.L.); (R.A.H.)
| | - Richard A. Hubner
- Department of Medical Oncology, The Christie NHS Foundation Trust/Division of Cancer Sciences, University of Manchester, Manchester M20 4BX, UK; (A.L.); (R.A.H.)
| | - Juan W. Valle
- Division of Cancer Sciences, University of Manchester/Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4BX, UK;
| | - Mairéad G. McNamara
- Division of Cancer Sciences, University of Manchester/Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4BX, UK;
- Correspondence:
| |
Collapse
|
62
|
Isolation of Prebiotics from Artocarpus integer's Seed. INTERNATIONAL JOURNAL OF FOOD SCIENCE 2021; 2021:9940078. [PMID: 34336997 PMCID: PMC8318755 DOI: 10.1155/2021/9940078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/25/2021] [Accepted: 07/01/2021] [Indexed: 11/18/2022]
Abstract
There has been a high amount of attention given to prebiotics due to their significant physiological function and health benefits. Prebiotics contain nondigestible compounds that allow specific changes, both in the growth and in the activity of bacteria in the host gastrointestinal tract, that provide benefits upon the host by promoting a healthy digestive system and preventing disease. This study aims at investigating the potential prebiotic activity of bioactive compounds extracted from the seeds of an underutilized indigenous plant Artocarpus integer (A. integer). The optimum microwave-assisted extraction conditions were a microwave power of 1500 W, extraction time of 180 s, and solvent-to-sample ratio of 1000 : 1. The maximum amount of the total carbohydrate content extracted from A. integer was 787 mg/L. The percentage hydrolysis levels of A. integer extract in gastric juice at pH 1, 2, 3, and 4 were 6.14%, 7.12%, 8.98%, and 10.23%, respectively. For enzymatic digestion, the percentage of hydrolysis was 0.16% at pH 7. A. integer extract was found to support the growth of probiotics such as L. acidophilus and L. casei. After 72 hours of incubation, L. acidophilus achieved 6.96 log10 CFU, whereas L. casei reached 8.33 log10 CFU. The study makes an important contribution to the development of the use of Sarawakian underutilized plants and to the identification of new sources of prebiotic materials to be used in food.
Collapse
|
63
|
Yaqoob MU, El-Hack MEA, Hassan F, El-Saadony MT, Khafaga AF, Batiha GE, Yehia N, Elnesr SS, Alagawany M, El-Tarabily KA, Wang M. The potential mechanistic insights and future implications for the effect of prebiotics on poultry performance, gut microbiome, and intestinal morphology. Poult Sci 2021; 100:101143. [PMID: 34062442 PMCID: PMC8170421 DOI: 10.1016/j.psj.2021.101143] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/12/2021] [Accepted: 03/14/2021] [Indexed: 12/02/2022] Open
Abstract
Prebiotics may modify the biological processes in the chickens' gastrointestinal tract to improve poultry performance and health. Prebiotics are natural feed additives that offer many economic advantages by decreasing mortality rates, increasing growth rates, and improving birds' feed efficiency. Prebiotic action potentially affects the degradation of indigestible dietary compounds, the synthesis of nitrogen components and vitamins, and simplifies the removal of undesirable elements in the diet. Prebiotics could also induce desirable gut microbiome modifications and affect host metabolism and immune health. It is worth mentioning that gut bacteria metabolize the prebiotic compounds into organic compounds that the host can subsequently use. It is important to limit the concept of prebiotics to compounds that influence the metabolism of resident microorganisms. Any medicinal component or feed ingredient beneficial to the intestinal microecosystem can be considered a prebiotic. In this review, the impacts of prebiotics on the gut microbiome and physiological structure are discussed, emphasizing the poultry's growth performance. The current review will highlight the knowledge gaps in this area and future research directions.
Collapse
Affiliation(s)
- M U Yaqoob
- College of Animal Science, Zhejiang University, Hangzhou 310058, PR China
| | - M E Abd El-Hack
- Department of Poultry, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt
| | - F Hassan
- Institute of Animal and Dairy Sciences, University of Agriculture, Faisalabad 38040, Pakistan
| | - M T El-Saadony
- Agricultural Microbiology Department, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt
| | - A F Khafaga
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Edfina 22758, Egypt
| | - G E Batiha
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, 080-8555, Obihiro, Hokkaido, Japan; Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, AlBeheira, Egypt
| | - N Yehia
- Reference Laboratory for Veterinary Quality Control on Poultry Production, Animal Health Research Institute, Agricultural Research center, Cairo, Egypt
| | - S S Elnesr
- Poultry Production Department, Faculty of Agriculture, Fayoum University, Fayoum 63514, Egypt
| | - M Alagawany
- Department of Poultry, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt
| | - K A El-Tarabily
- Department of Biology, College of Science, United Arab Emirates University, 15551, Al-Ain, United Arab Emirates; Biosecurity and One Health Research Centre, Harry Butler Institute, Murdoch University, Murdoch, Western Australia, 6150, Australia
| | - M Wang
- College of Animal Science, Zhejiang University, Hangzhou 310058, PR China.
| |
Collapse
|
64
|
Preventing Colorectal Cancer through Prebiotics. Microorganisms 2021; 9:microorganisms9061325. [PMID: 34207094 PMCID: PMC8234836 DOI: 10.3390/microorganisms9061325] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/09/2021] [Accepted: 06/14/2021] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC), the third most common cancer in the world, has been recently rising in emerging countries due to environmental and lifestyle factors. Many of these factors are brought up by industrialization, which includes lack of physical activity, poor diet, circadian rhythm disruption, and increase in alcohol consumption. They can increase the risk of CRC by changing the colonic environment and by altering gut microbiota composition, a state referred to as gut dysbiosis. Prebiotics, which are nutrients that can help maintain intestinal microbial homeostasis and mitigate dysbiosis, could be beneficial in preventing inflammation and CRC. These nutrients can hinder the effects of dysbiosis by encouraging the growth of beneficial bacteria involved in short-chain fatty acids (SCFA) production, anti-inflammatory immunity, maintenance of the intestinal epithelial barrier, pro-apoptotic mechanisms, and other cellular mechanisms. This review aims to summarize recent reports about the implication of prebiotics, and probable mechanisms, in the prevention and treatment of CRC. Various experimental studies, specifically in gut microbiome, have effectively demonstrated the protective effect of prebiotics in the progress of CRC. Hence, comprehensive knowledge is urgent to understand the clinical applications of prebiotics in the prevention or treatment of CRC.
Collapse
|
65
|
A review on enzyme-producing lactobacilli associated with the human digestive process: From metabolism to application. Enzyme Microb Technol 2021; 149:109836. [PMID: 34311881 DOI: 10.1016/j.enzmictec.2021.109836] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/30/2021] [Accepted: 05/27/2021] [Indexed: 12/12/2022]
Abstract
Complex carbohydrates, proteins, and other food components require a longer digestion process to be absorbed by the lining of the alimentary canal. In addition to the enzymes of the gastrointestinal tract, gut microbiota, comprising a large range of bacteria and fungi, has complementary action on the production of digestive enzymes. Within this universe of "hidden soldiers", lactobacilli are extensively studied because of their ability to produce lactase, proteases, peptidases, fructanases, amylases, bile salt hydrolases, phytases, and esterases. The administration of living lactobacilli cells has been shown to increase nutrient digestibility. However, it is still little known how these microbial-derived enzymes act in the human body. Enzyme secretion may be affected by variations in temperature, pH, and other extreme conditions faced by the bacterial cells in the human body. Besides, lactobacilli administration cannot itself be considered the only factor interfering with enzyme secretion, human diet (microbial substrate) being determinant in their metabolism. This review highlights the potential of lactobacilli to release functional enzymes associated with the digestive process and how this complex metabolism can be explored to contribute to the human diet. Enzymatic activity of lactobacilli is exerted in a strain-dependent manner, i.e., within the same lactobacilli species, there are different enzyme contents, leading to a large variety of enzymatic activities. Thus, we report current methods to select the most promising lactobacilli strains as sources of bioactive enzymes. Finally, a patent landscape and commercial products are described to provide the state of art of the transfer of knowledge from the scientific sphere to the industrial application.
Collapse
|
66
|
Asadpoor M, Varasteh S, Pieters RJ, Folkerts G, Braber S. Differential effects of oligosaccharides on the effectiveness of ampicillin against Escherichia coli in vitro. PHARMANUTRITION 2021. [DOI: 10.1016/j.phanu.2021.100264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
67
|
Sakr EA, Massoud MI. Impact of prebiotic potential of stevia sweeteners-sugar used as synbiotic preparation on antimicrobial, antibiofilm, and antioxidant activities. Lebensm Wiss Technol 2021. [DOI: 10.1016/j.lwt.2021.111260] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
68
|
Morrin ST, Buck RH, Farrow M, Hickey RM. Milk-derived anti-infectives and their potential to combat bacterial and viral infection. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104442] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
69
|
Kong C, Faas MM, de Vos P, Akkerman R. Impact of dietary fibers in infant formulas on gut microbiota and the intestinal immune barrier. Food Funct 2021; 11:9445-9467. [PMID: 33150902 DOI: 10.1039/d0fo01700k] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human milk (HM) is the gold standard for the nutrition of infants. An important component of HM is human milk oligosaccharides (hMOs), which play an important role in gut microbiota colonization and gut immune barrier establishment, and thereby contribute to the maturation of the immune system in early life. Guiding these processes is important as disturbances have life-long health effects and can lead to the development of allergic diseases. Unfortunately, not all infants can be exclusively fed with HM. These infants are routinely fed with infant formulas that contain hMO analogs and other non-digestible carbohydrates (NDCs) to mimic the effects of hMOs. Currently, the hMO analogs 2'-fucosyllactose (2'-FL), galacto-oligosaccharides (GOS), fructo-oligosaccharides (FOS), and pectins are added to infant formulas; however, these NDCs cannot mimic all hMO functions and therefore new NDCs and NDC mixtures need to become available for specific groups of neonates like preterm and disease-prone neonates. In this review, we discuss human data on the beneficial effects of infant formula supplements such as the specific hMO analog 2'-FL and NDCs as well as their mechanism of effects like stimulation of microbiota development, maturation of different parts of the gut immune barrier and anti-pathogenic effects. Insights into the structure-specific mechanisms by which hMOs and NDCs exert their beneficial functions might contribute to the development of new tailored NDCs and NDC mixtures. We also describe the needs for new in vitro systems that can be used for research on hMOs and NDCs. The current data suggest that "tailored infant formulas" for infants of different ages and healthy statuses are needed to ensure a healthy development of the microbiota and the gut immune system of infants.
Collapse
Affiliation(s)
- Chunli Kong
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB Groningen, The Netherlands.
| | | | | | | |
Collapse
|
70
|
Rodríguez-Sorrento A, Castillejos L, López-Colom P, Cifuentes-Orjuela G, Rodríguez-Palmero M, Moreno-Muñoz JA, Luise D, Trevisi P, Martín-Orúe SM. Effects of the Administration of Bifidobacterium longum subsp. infantis CECT 7210 and Lactobacillus rhamnosus HN001 and Their Synbiotic Combination With Galacto-Oligosaccharides Against Enterotoxigenic Escherichia coli F4 in an Early Weaned Piglet Model. Front Microbiol 2021; 12:642549. [PMID: 33935999 PMCID: PMC8086512 DOI: 10.3389/fmicb.2021.642549] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/23/2021] [Indexed: 01/09/2023] Open
Abstract
We evaluated the potential of multi-strain probiotic (Bifidobacterium longum subsp. infantis CECT 7210 and Lactobacillus rhamnosus HN001) with or without galacto-oligosaccharides against enterotoxigenic Escherichia coli (ETEC) F4 infection in post-weaning pigs. Ninety-six piglets were distributed into 32 pens assigned to five treatments: one non-challenged (CTR+) and four challenged: control diet (CTR-), with probiotics (>3 × 1010 CFU/kg body weight each, PRO), prebiotic (5%, PRE), or their combination (SYN). After 1 week, animals were orally inoculated with ETEC F4. Feed intake, weight, and clinical signs were recorded. On days 4 and 8 post-inoculation (PI), one animal per pen was euthanized and samples from blood, digesta, and tissues collected. Microbiological counts, ETEC F4 real-time PCR (qPCR) quantification, fermentation products, serum biomarkers, ileal histomorphometry, and genotype for mucin 4 (MUC4) polymorphism were determined. Animals in the PRO group had similar enterobacteria and coliform numbers to the CTR+ group, and the ETEC F4 prevalence, the number of mitotic cells at day 4 PI, and villus height at day 8 PI were between that observed in the CTR+ and CTR- groups. The PRO group exhibited reduced pig major acute-phase protein (Pig-MAP) levels on day 4 PI. The PRE diet group presented similar reductions in ETEC F4 and Pig-MAP, but there was no effect on microbial groups. The SYN group showed reduced fecal enterobacteria and coliform counts after the adaptation week but, after the inoculation, the SYN group showed lower performance and more animals with high ETEC F4 counts at day 8 PI. SYN treatment modified the colonic fermentation differently depending on the MUC4 polymorphism. These results confirm the potential of the probiotic strains and the prebiotic to fight ETEC F4, but do not show any synergy when administered together, at least in this animal model.
Collapse
Affiliation(s)
- Agustina Rodríguez-Sorrento
- Servicio de Nutrición y Bienestar Animal, Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Lorena Castillejos
- Servicio de Nutrición y Bienestar Animal, Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Paola López-Colom
- Servicio de Nutrición y Bienestar Animal, Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | | | | | | | - Diana Luise
- Department of Agricultural and Food Science, University of Bologna, Bologna, Italy
| | - Paolo Trevisi
- Department of Agricultural and Food Science, University of Bologna, Bologna, Italy
| | - Susana María Martín-Orúe
- Servicio de Nutrición y Bienestar Animal, Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| |
Collapse
|
71
|
Identification of Synbiotics Conducive to Probiotics Adherence to Intestinal Mucosa Using an In Vitro Caco-2 and HT29-MTX Cell Model. Processes (Basel) 2021. [DOI: 10.3390/pr9040569] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The ability of bacteria to adhere to the intestinal mucosa is a critical property necessary for the long-term colonization of the intestinal tract. This ability can be highly sensitive to the presence of prebiotics. However, limited data are available in this respect for beneficial bacteria such as probiotics or resident gut microbiota. We previously demonstrated that the presence of prebiotics may decrease adherence in several pre- and probiotic combinations. Thus, characterizing the interactions between numerous combinations involving different classes of pre- and probiotics can be crucial in identifying new synbiotics. Accordingly, here, we extend our prior analyses to evaluate the adhesion of five lactobacilli, six bifidobacteria, and one probiotic Escherichia coli strains, as commercial probiotics or promising probiotic candidates, together with the cariogenic Bifidobacterium dentium strain. As an in vitro intestinal mucosa model, Caco-2 and mucin-secreting HT29-MTX cells were co-cultured at 9:1 in the presence or absence of prebiotics. Commercial inulin-type fructooligosaccharide prebiotics Orafti® GR, Orafti® P95, and galactooligosaccharide-based prebiotic formula Vivinal®, including purified human milk oligosaccharides (HMOs) were added into the cultivation media as the sole sugar source (2.5% each). Adherence was tested using microtiter plates and was evaluated as the percentage of fluorescently labeled bacteria present in the wells after three washes. Consistent prebiotics-mediated enhanced adherence was observed only for the commercial probiotic strain E. coli O83. For the remaining strains, the presence of HMO or prebiotics Orafti® P95 or Orafti® GR decreased adherence, reaching statistical significance (p < 0.05) for three of out of eight (HMO) or five of out of 11 strains tested, respectively. Conversely, Vivinal® enhanced adhesion in six out of the 12 strains tested, and notably, it significantly attenuated the adherence of the cariogenic Bifidobacterium dentium Culture Collection of Dairy Microorganisms (CCDM) 318. To our knowledge, this represents the first report on the influence of commercial prebiotics and HMOs on the adhesion of the cariogenic Bifidobacterium sp. Vivinal® seems to be a promising prebiotic to be used in the formulation of synbiotics, supporting the adhesion of a wide range of probiotics, especially the strains B. bifidum BBV and BBM and the probiotic Escherichia coli O83.
Collapse
|
72
|
Mohamed A, Menon H, Chulkina M, Yee NS, Pinchuk IV. Drug-Microbiota Interaction in Colon Cancer Therapy: Impact of Antibiotics. Biomedicines 2021; 9:259. [PMID: 33807878 PMCID: PMC7999677 DOI: 10.3390/biomedicines9030259] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 12/14/2022] Open
Abstract
Colon adenocarcinoma is one of the most common malignancies, and it is highly lethal. Chemotherapy plays an important role in the treatment of colon cancer at various stages of the disease. The gut microbiome has emerged as a key player in colon cancer development and progression, and it can also alter the therapeutic agent's efficacy and toxicities. Antibiotics can directly and/or indirectly affect the balance of the gut microbiome and, therefore, the clinical outcomes. In this article, we provided an overview of the composition of the gut microbiome under homeostasis and the mechanistic links between gut microbiota and colon cancer. The relationship between the use of oral antibiotics and colon cancer, as well as the impact of the gut microbiome on the efficacy and toxicities of chemotherapy in colon cancer, are discussed. Potential interventions to modulate microbiota and improve chemotherapy outcomes are discussed. Further studies are indicated to address these key gaps in the field and provide a scientific basis for the design of novel microbiota-based approaches for prevention/use as adjuvant therapeutics for patients with colon cancer.
Collapse
Affiliation(s)
- Ali Mohamed
- Division of Hematology-Oncology, Department of Medicine, Penn State Health Milton S. Hershey Medical Center, Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (A.M.); (H.M.)
| | - Harry Menon
- Division of Hematology-Oncology, Department of Medicine, Penn State Health Milton S. Hershey Medical Center, Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (A.M.); (H.M.)
| | - Marina Chulkina
- Mechanisms of Carcinogenesis Program, Division of Gastroenterology, Department of Medicine, Penn State Health Milton S. Hershey Medical Center, Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA;
| | - Nelson S. Yee
- Next-Generation Therapies Program, Division of Hematology-Oncology, Department of Medicine, Penn State Health Milton S. Hershey Medical Center, Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Irina V. Pinchuk
- Mechanisms of Carcinogenesis Program, Division of Gastroenterology, Department of Medicine, Penn State Health Milton S. Hershey Medical Center, Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA;
| |
Collapse
|
73
|
Asadpoor M, Ithakisiou GN, Henricks PAJ, Pieters R, Folkerts G, Braber S. Non-Digestible Oligosaccharides and Short Chain Fatty Acids as Therapeutic Targets against Enterotoxin-Producing Bacteria and Their Toxins. Toxins (Basel) 2021; 13:175. [PMID: 33668708 PMCID: PMC7996226 DOI: 10.3390/toxins13030175] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/08/2021] [Accepted: 02/17/2021] [Indexed: 02/07/2023] Open
Abstract
Enterotoxin-producing bacteria (EPB) have developed multiple mechanisms to disrupt gut homeostasis, and provoke various pathologies. A major part of bacterial cytotoxicity is attributed to the secretion of virulence factors, including enterotoxins. Depending on their structure and mode of action, enterotoxins intrude the intestinal epithelium causing long-term consequences such as hemorrhagic colitis. Multiple non-digestible oligosaccharides (NDOs), and short chain fatty acids (SCFA), as their metabolites produced by the gut microbiota, interact with enteropathogens and their toxins, which may result in the inhibition of the bacterial pathogenicity. NDOs characterized by diverse structural characteristics, block the pathogenicity of EPB either directly, by inhibiting bacterial adherence and growth, or biofilm formation or indirectly, by promoting gut microbiota. Apart from these abilities, NDOs and SCFA can interact with enterotoxins and reduce their cytotoxicity. These anti-virulent effects mostly rely on their ability to mimic the structure of toxin receptors and thus inhibiting toxin adherence to host cells. This review focuses on the strategies of EPB and related enterotoxins to impair host cell immunity, discusses the anti-pathogenic properties of NDOs and SCFA on EPB functions and provides insight into the potential use of NDOs and SCFA as effective agents to fight against enterotoxins.
Collapse
Affiliation(s)
- Mostafa Asadpoor
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands; (M.A.); (G.-N.I.); (P.A.J.H.); (G.F.)
| | - Georgia-Nefeli Ithakisiou
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands; (M.A.); (G.-N.I.); (P.A.J.H.); (G.F.)
| | - Paul A. J. Henricks
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands; (M.A.); (G.-N.I.); (P.A.J.H.); (G.F.)
| | - Roland Pieters
- Division of Medicinal Chemistry and Chemical Biology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands;
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands; (M.A.); (G.-N.I.); (P.A.J.H.); (G.F.)
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands; (M.A.); (G.-N.I.); (P.A.J.H.); (G.F.)
| |
Collapse
|
74
|
Figueroa-Lozano S, Ren C, Yin H, Pham H, van Leeuwen S, Dijkhuizen L, de Vos P. The impact of oligosaccharide content, glycosidic linkages and lactose content of galacto-oligosaccharides (GOS) on the expression of mucus-related genes in goblet cells. Food Funct 2021; 11:3506-3515. [PMID: 32253406 DOI: 10.1039/d0fo00064g] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Galacto-oligosaccharides (GOS) have been reported to modulate the function of intestinal goblet cells and to improve mucus barrier function. However, GOS is available in many structurally different compositions and it is unknown how GOS structural diversity impacts this modulation of goblet cells. This study aims to investigate the effects of oligosaccharide content and glycosidic linkages of GOS on expression of genes associated with the secretory function of goblet cells. To investigate the effect of oligosaccharide content, LS174T cells were incubated with (β1 → 4)GOS of variable transgalactosylated oligosaccharides and lactose (Lac) composition. To investigate the effect of glycosidic linkages, we compared the effects of (β1 → 4)GOS with (β1 → 3)GOS, and with a mixture of α-linked oligosaccharides (lactose-derived oligosaccharides-LDO). The changes in mRNA expression of mucus-related genes were assessed by RT-PCR. GOS containing Lac significantly enhanced the expression of MUC2, TFF3 and RETNLB but not of Golgi sulfotransferases genes. In contrast, GOS without Lac did not impact these genes. Lac alone significantly enhanced MUC2, TFF3, RETNLB, CHST5, and GAL3ST2 genes suggesting that Lac might be responsible for goblet cell modulation in (β1 → 4)GOS preparations. (β1 → 3)GOS induced the expression of MUC2 and TFF3, and downregulated the RETNLB gene. Compared with the (β1 → 3) and GOS (β1 → 4)GOS, the α-linked LDO significantly upregulated the expression MUC2, TFF3, RETNLB and the Golgi sulfotransferases genes. We identify structural features of GOS that contribute to enhanced mucus integrity. Our study might lead to better GOS formulations for foods to prevent or treat different types of intestinal disorders.
Collapse
Affiliation(s)
- Susana Figueroa-Lozano
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen (RUG-UMCG), Hanzeplein 1, 9700 RB Groningen, The Netherlands.
| | - Chengcheng Ren
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen (RUG-UMCG), Hanzeplein 1, 9700 RB Groningen, The Netherlands.
| | - Huifang Yin
- Microbial Physiology, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - Hien Pham
- Microbial Physiology, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - Sander van Leeuwen
- Microbial Physiology, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), Nijenborgh 7, 9747 AG Groningen, The Netherlands and Department of Laboratory Medicine, RUG-UMCG, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Lubbert Dijkhuizen
- Microbial Physiology, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), Nijenborgh 7, 9747 AG Groningen, The Netherlands and CarbExplore Research BV, Zernikepark 12, 9747 AN Groningen, The Netherlands
| | - Paul de Vos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen (RUG-UMCG), Hanzeplein 1, 9700 RB Groningen, The Netherlands.
| |
Collapse
|
75
|
Oliero M, Calvé A, Fragoso G, Cuisiniere T, Hajjar R, Dobrindt U, Santos MM. Oligosaccharides increase the genotoxic effect of colibactin produced by pks+ Escherichia coli strains. BMC Cancer 2021; 21:172. [PMID: 33596864 PMCID: PMC7890614 DOI: 10.1186/s12885-021-07876-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 02/03/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Colibactin is a genotoxin that induces DNA double-strand breaks that may lead to carcinogenesis and is produced by Escherichia coli strains harboring the pks island. Human and animal studies have shown that colibactin-producing gut bacteria promote carcinogenesis and enhance the progression of colorectal cancer through cellular senescence and chromosomal abnormalities. In this study, we investigated the impact of prebiotics on the genotoxicity of colibactin-producing E. coli strains Nissle 1917 and NC101. METHODS Bacteria were grown in medium supplemented with 20, 30 and 40 mg/mL of prebiotics inulin or galacto-oligosaccharide, and with or without 5 μM, 25 μM and 125 μM of ferrous sulfate. Colibactin expression was assessed by luciferase reporter assay for the clbA gene, essential for colibactin production, in E. coli Nissle 1917 and by RT-PCR in E. coli NC101. The human epithelial colorectal adenocarcinoma cell line, Caco-2, was used to assess colibactin-induced megalocytosis by methylene blue binding assay and genotoxicity by γ-H2AX immunofluorescence analysis. RESULTS Inulin and galacto-oligosaccharide enhanced the expression of clbA in pks+ E. coli. However, the addition of 125 μM of ferrous sulfate inhibited the expression of clbA triggered by oligosaccharides. In the presence of either oligosaccharide, E. coli NC101 increased dysplasia and DNA double-strand breaks in Caco-2 cells compared to untreated cells. CONCLUSION Our results suggest that, in vitro, prebiotic oligosaccharides exacerbate DNA damage induced by colibactin-producing bacteria. Further studies are necessary to establish whether oligosaccharide supplementation may lead to increased colorectal tumorigenesis in animal models colonized with pks+ E. coli.
Collapse
Affiliation(s)
- Manon Oliero
- Nutrition and Microbiome Laboratory, Institut du cancer de Montréal, Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), 900 Rue Saint Denis, Montreal, QC H2X 0A9 Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, 2900 boulevard Édouard-Montpetit, Montréal, QC H3T 1J4 Canada
| | - Annie Calvé
- Nutrition and Microbiome Laboratory, Institut du cancer de Montréal, Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), 900 Rue Saint Denis, Montreal, QC H2X 0A9 Canada
| | - Gabriela Fragoso
- Nutrition and Microbiome Laboratory, Institut du cancer de Montréal, Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), 900 Rue Saint Denis, Montreal, QC H2X 0A9 Canada
| | - Thibault Cuisiniere
- Nutrition and Microbiome Laboratory, Institut du cancer de Montréal, Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), 900 Rue Saint Denis, Montreal, QC H2X 0A9 Canada
| | - Roy Hajjar
- Nutrition and Microbiome Laboratory, Institut du cancer de Montréal, Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), 900 Rue Saint Denis, Montreal, QC H2X 0A9 Canada
- Department of Surgery, Faculty of Medicine, Université de Montréal, 2900 boulevard Édouard-Montpetit, Montréal, QC H3T 1J4 Canada
| | - Ulrich Dobrindt
- Institute of Hygiene, Section Microbial Genome Plasticity, University of Münster, Mendelstraße 7, 48149 Münster, Germany
| | - Manuela M. Santos
- Nutrition and Microbiome Laboratory, Institut du cancer de Montréal, Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), 900 Rue Saint Denis, Montreal, QC H2X 0A9 Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, 2900 boulevard Édouard-Montpetit, Montréal, QC H3T 1J4 Canada
| |
Collapse
|
76
|
Arnold JW, Roach J, Fabela S, Moorfield E, Ding S, Blue E, Dagher S, Magness S, Tamayo R, Bruno-Barcena JM, Azcarate-Peril MA. The pleiotropic effects of prebiotic galacto-oligosaccharides on the aging gut. MICROBIOME 2021; 9:31. [PMID: 33509277 PMCID: PMC7845053 DOI: 10.1186/s40168-020-00980-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 12/16/2020] [Indexed: 05/05/2023]
Abstract
BACKGROUND Prebiotic galacto-oligosaccharides (GOS) have an extensively demonstrated beneficial impact on intestinal health. In this study, we determined the impact of GOS diets on hallmarks of gut aging: microbiome dysbiosis, inflammation, and intestinal barrier defects ("leaky gut"). We also evaluated if short-term GOS feeding influenced how the aging gut responded to antibiotic challenges in a mouse model of Clostridioides difficile infection. Finally, we assessed if colonic organoids could reproduce the GOS responder-non-responder phenotypes observed in vivo. RESULTS Old animals had a distinct microbiome characterized by increased ratios of non-saccharolytic versus saccharolytic bacteria and, correspondingly, a lower abundance of β-galactosidases compared to young animals. GOS reduced the overall diversity, increased the abundance of specific saccharolytic bacteria (species of Bacteroides and Lactobacillus), increased the abundance of β-galactosidases in young and old animals, and increased the non-saccharolytic organisms; however, a robust, homogeneous bifidogenic effect was not observed. GOS reduced age-associated increased intestinal permeability and increased MUC2 expression and mucus thickness in old mice. Clyndamicin reduced the abundance Bifidobacterium while increasing Akkermansia, Clostridium, Coprococcus, Bacillus, Bacteroides, and Ruminococcus in old mice. The antibiotics were more impactful than GOS on modulating serum markers of inflammation. Higher serum levels of IL-17 and IL-6 were observed in control and GOS diets in the antibiotic groups, and within those groups, levels of IL-6 were higher in the GOS groups, regardless of age, and higher in the old compared to young animals in the control diet groups. RTqPCR revealed significantly increased gene expression of TNFα in distal colon tissue of old mice, which was decreased by the GOS diet. Colon transcriptomics analysis of mice fed GOS showed increased expression of genes involved in small-molecule metabolic processes and specifically the respirasome in old animals, which could indicate an increased oxidative metabolism and energetic efficiency. In young mice, GOS induced the expression of binding-related genes. The galectin gene Lgals1, a β-galactosyl-binding lectin that bridges molecules by their sugar moieties and is an important modulator of the immune response, and the PI3K-Akt and ECM-receptor interaction pathways were also induced in young mice. Stools from mice exhibiting variable bifidogenic response to GOS injected into colon organoids in the presence of prebiotics reproduced the response and non-response phenotypes observed in vivo suggesting that the composition and functionality of the microbiota are the main contributors to the phenotype. CONCLUSIONS Dietary GOS modulated homeostasis of the aging gut by promoting changes in microbiome composition and host gene expression, which was translated into decreased intestinal permeability and increased mucus production. Age was a determining factor on how prebiotics impacted the microbiome and expression of intestinal epithelial cells, especially apparent from the induction of galectin-1 in young but not old mice. Video abstract.
Collapse
Affiliation(s)
- Jason W Arnold
- Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- UNC Microbiome Core, Center for Gastrointestinal Biology and Disease (CGIBD), School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Jeffery Roach
- UNC Microbiome Core, Center for Gastrointestinal Biology and Disease (CGIBD), School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- UNC Information Technology Services and Research Computing, University of North Carolina, Chapel Hill, NC, USA
| | - Salvador Fabela
- Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- UNC Microbiome Core, Center for Gastrointestinal Biology and Disease (CGIBD), School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Current affiliation: Programa de Inmunología Molecular Microbiana. Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de Mexico, Mexico City, Mexico
| | - Emily Moorfield
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| | - Shengli Ding
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| | - Eric Blue
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| | - Suzanne Dagher
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| | - Scott Magness
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, USA
| | - Rita Tamayo
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Jose M Bruno-Barcena
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| | - M Andrea Azcarate-Peril
- Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA.
- UNC Microbiome Core, Center for Gastrointestinal Biology and Disease (CGIBD), School of Medicine, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
77
|
Gut Microbiota Influence in Hematological Malignancies: From Genesis to Cure. Int J Mol Sci 2021; 22:ijms22031026. [PMID: 33498529 PMCID: PMC7864170 DOI: 10.3390/ijms22031026] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 12/13/2022] Open
Abstract
Hematological malignancies, including multiple myeloma, lymphoma, and leukemia, are a heterogeneous group of neoplasms that affect the blood, bone marrow, and lymph nodes. They originate from uncontrolled growth of hematopoietic and lymphoid cells from different stages in their maturation/differentiation and account for 6.5% of all cancers around the world. During the last decade, it has been proven that the gut microbiota, more specifically the gastrointestinal commensal bacteria, is implicated in the genesis and progression of many diseases. The immune-modulating effects of the human microbiota extend well beyond the gut, mostly through the small molecules they produce. This review aims to summarize the current knowledge of the role of the microbiota in modulating the immune system, its role in hematological malignancies, and its influence on different therapies for these diseases, including autologous and allogeneic stem cell transplantation, chemotherapy, and chimeric antigen receptor T cells.
Collapse
|
78
|
Cao Y, Liu J, Zhu W, Qin N, Ren X, Zhu B, Xia X. Impact of dietary components on enteric infectious disease. Crit Rev Food Sci Nutr 2021; 62:4010-4035. [PMID: 33455435 DOI: 10.1080/10408398.2021.1871587] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Diets impact host health in multiple ways and an unbalanced diet could contribute to the initiation or progression of a variety of diseases. Although a wealth of information exists on the connections between diet and chronic metabolic diseases such as cardiovascular disease, diabetes mellitus, etc., how diet influences enteric infectious disease still remain underexplored. The review summarizes the current findings on the link between various dietary components and diverse enteric infectious diseases. Dietary ingredients discussed include macronutrients (carbohydrates, lipids, proteins), micronutrients (vitamins, minerals), and other dietary ingredients (phytonutrients and probiotic supplements). We first describe the importance of enteric infectious diseases and the direct and indirect relationship between diet and enteric infectious diseases. Then we discuss the effects of different dietary components on the susceptibility to or progression of enteric infectious disease. Finally, we delineate current knowledge gap and highlighted future research directions. The literature review revealed that different dietary components affect host resistance to enteric infections through a variety of mechanisms. Dietary components may directly inhibit or bind to enteric pathogens, or indirectly influence enteric infections through modulating immune function and gut microbiota. Elucidating the unique repercussions of different diets on enteric infections in this review may help provide dietary guidelines or design dietary interventions to prevent or alleviate enteric infectious diseases.
Collapse
Affiliation(s)
- Yu Cao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.,National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning, China
| | - Jiaxiu Liu
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning, China
| | - Wenxiu Zhu
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning, China
| | - Ningbo Qin
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning, China
| | - Xiaomeng Ren
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning, China
| | - Beiwei Zhu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.,National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning, China
| | - Xiaodong Xia
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning, China
| |
Collapse
|
79
|
Effects of Dietary Fiber on Nutrients Utilization and Gut Health of Poultry: A Review of Challenges and Opportunities. Animals (Basel) 2021; 11:ani11010181. [PMID: 33466662 PMCID: PMC7828824 DOI: 10.3390/ani11010181] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/08/2021] [Accepted: 01/11/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary The inclusion of agricultural co-products has been increased to utilize the nutrients in these products available at low cost, but inherently, it adds a high dietary fiber content in the poultry diets. The use of exogenous feed enzymes along with advancements in feed milling, feed formulation, and processing of these non-conventional ingredients to improve their digestibility and utilization have played an emphatic role in boosting their use globally. Despite such developments, the presence of a high level of dietary fibers (DF) acting in an anti-nutritive manner still poses challenges in poultry feeding. Various isolated forms of fiber or feed enzymes to break DF into fermentable substrates are being used extensively to provide potential prebiotics to support beneficial gut microbiota or probiotics to improve the gut health of poultry raised without antibiotic growth promoters (AGP). This review reports and discusses the existing challenges in feeding high-DF feed ingredients to poultry and the opportunities that are available to improve the nutritive value of such non-conventional feed ingredients by adopting various technologies. Abstract Many fibrous ingredients incorporated in poultry feed to reduce production costs have low digestibility and cause poor growth in poultry. However, all plant-based fibers are not equal, and thus exert variable physiological effects on the birds, including but not limited to, digestibility, growth performance, and microbial fermentation. Several types of fibers, especially oligosaccharides, when supplemented in poultry diets in isolated form, exhibit prebiotic effects by enhancing beneficial gut microbiota, modulating gut immunity, boosting intestinal mucosal health, and increasing the production of short-chain fatty acids (SCFA) in the gut. Recently, poultry producers are also facing the challenge of limiting the use of antibiotic growth promoters (AGP) in poultry feed. In addition to other alternatives in use, exogenous non-starch polysaccharides digesting enzymes (NSPase) and prebiotics are being used to provide substrates to support the gut microbiome. We also conducted a meta-analysis of different studies conducted in similar experimental conditions to evaluate the variability and conclusiveness in effects of NSPase on growth performance of broilers fed fibrous ingredients. This review presents a holistic approach in discussing the existing challenges of incorporating high-fiber ingredients in poultry feed, as well as strategies to fully utilize the potential of such ingredients in improving feed efficiency and gut health of poultry.
Collapse
|
80
|
Salli K, Hirvonen J, Siitonen J, Ahonen I, Anglenius H, Maukonen J. Selective Utilization of the Human Milk Oligosaccharides 2'-Fucosyllactose, 3-Fucosyllactose, and Difucosyllactose by Various Probiotic and Pathogenic Bacteria. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:170-182. [PMID: 33382612 DOI: 10.1021/acs.jafc.0c06041] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Prebiotic human milk oligosaccharides (HMOs) are found in human milk, which are not digested by infants but are metabolized by beneficial gut bacteria. We determined the ability of 57 bacterial strains within the Family Lactobacillaceae and genera Bifidobacterium and Bacteroides and potentially pathogenic bacteria to ferment the HMOs 2'-fucosyllactose, 3-fucosyllactose, and difucosyllactose. In addition, prebiotic galacto-oligosaccharides (GOS), lactose, fucose, and glucose were evaluated as carbon sources for these bacterial strains. Bacterial growth was monitored using the automatic Bioscreen C system. Only certain bifidobacteria, such as Bifidobacterium longum subsp. infantis and Bifidobacterium bifidum, as well as Bacteroides fragilis, Bacteroides vulgatus, and Bacteroides thetaiotaomicron utilized the studied HMOs as their sole carbon source, whereas almost all studied bacterial strains were able to utilize GOS, lactose, and glucose. The selectivity in utilization of HMOs by only certain bacteria can be advantageous by promoting beneficial microbes but not supporting the harmful pathogens in contrast to other less selective prebiotics.
Collapse
Affiliation(s)
- Krista Salli
- Global Health and Nutrition Science, DuPont Nutrition & Biosciences, 02460 Kantvik, Finland
| | - Johanna Hirvonen
- Global Health and Nutrition Science, DuPont Nutrition & Biosciences, 02460 Kantvik, Finland
| | - Jani Siitonen
- Manufacturing Technology, DuPont Nutrition & Biosciences, 02460 Kantvik, Finland
| | | | - Heli Anglenius
- Global Health and Nutrition Science, DuPont Nutrition & Biosciences, 02460 Kantvik, Finland
| | - Johanna Maukonen
- Global Health and Nutrition Science, DuPont Nutrition & Biosciences, 02460 Kantvik, Finland
| |
Collapse
|
81
|
Lafond M, Tauzin AS, Bruel L, Laville E, Lombard V, Esque J, André I, Vidal N, Pompeo F, Quinson N, Perrier J, Fons M, Potocki-Veronese G, Giardina T. α-Galactosidase and Sucrose-Kinase Relationships in a Bi-functional AgaSK Enzyme Produced by the Human Gut Symbiont Ruminococcus gnavus E1. Front Microbiol 2020; 11:579521. [PMID: 33281771 PMCID: PMC7688924 DOI: 10.3389/fmicb.2020.579521] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 10/19/2020] [Indexed: 12/31/2022] Open
Abstract
Plant α-galactosides belonging to the raffinose family oligosaccharides (RFOs) and considered as prebiotics, are commonly degraded by α-galactosidases produced by the human gut microbiome. In this environment, the Ruminococcus gnavus E1 symbiont-well-known for various benefit-is able to produce an original RgAgaSK bifunctional enzyme. This enzyme contains an hydrolytic α-galactosidase domain linked to an ATP dependent extra-domain, specifically involved in the α-galactoside hydrolysis and the phosphorylation of the glucose, respectively. However, the multi-modular relationships between both catalytic domains remained hitherto unexplored and has been, consequently, herein investigated. Biochemical characterization of heterologously expressed enzymes either in full-form or in separated domains revealed similar kinetic parameters. These results were supported by molecular modeling studies performed on the whole enzyme in complex with different RFOs. Further enzymatic analysis associated with kinetic degradation of various substrates followed by high pressure anionic exchange chromatography revealed that catalytic efficiency decreased as the number of D-galactosyl moieties branched onto the oligosaccharide increased, suggesting a preference of RgAgaSK for RFO's short chains. A wide prevalence and abundance study on a human metagenomic library showed a high prevalence of the RgAgaSK encoding gene whatever the health status of the individuals. Finally, phylogeny and synteny studies suggested a limited spread by horizontal transfer of the clusters' containing RgAgaSK to only few species of Firmicutes, highlighting the importance of these undispersed tandem activities in the human gut microbiome.
Collapse
Affiliation(s)
- Mickael Lafond
- Aix-Marseille Université, CNRS, Centrale Marseille, iSm2, Marseille, France
| | | | - Laetitia Bruel
- Aix-Marseille Université, CNRS, Centrale Marseille, iSm2, Marseille, France
| | | | | | - Jérémy Esque
- TBI, Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France
| | - Isabelle André
- TBI, Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France
| | - Nicolas Vidal
- Yelen Analytics, Aix-Marseille Université, ICR, Marseille, France
| | | | - Nathalie Quinson
- Aix-Marseille Université, CNRS, Centrale Marseille, iSm2, Marseille, France
| | - Josette Perrier
- Aix-Marseille Université, CNRS, Centrale Marseille, iSm2, Marseille, France
| | - Michel Fons
- Aix-Marseille Université, CNRS, IMM, BIP, Marseille, France
| | | | - Thierry Giardina
- Aix-Marseille Université, CNRS, Centrale Marseille, iSm2, Marseille, France
| |
Collapse
|
82
|
Dong L, Ariëns RMC, Tomassen MM, Wichers HJ, Govers C. In Vitro Studies Toward the Use of Chitin as Nutraceutical: Impact on the Intestinal Epithelium, Macrophages, and Microbiota. Mol Nutr Food Res 2020; 64:e2000324. [PMID: 33067879 PMCID: PMC7757189 DOI: 10.1002/mnfr.202000324] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 09/17/2020] [Indexed: 12/11/2022]
Abstract
SCOPE Chitin, the most abundant polysaccharide found in nature after cellulose, is known for its ability to support wound healing and to lower plasma-oxidized low-density lipoprotein (LDL) levels. Studies have also revealed immunomodulatory potential but contradicting results are often impossible to coalesce through usage of chitin of different or unknown physicochemical consistency. In addition, only a limited set of cellular models have been used to test the bioactivity of chitin. METHODS AND RESULTS Chitin is investigated with well-defined physicochemical consistency for its immunomodulatory potency using THP-1 macrophages, impact on intestinal epithelial barrier using Caco-2 cells, and fermentation by fecal-derived microbiota. Results show that chitin with a degree of acetylation (DA) of ≈83%, regardless of size, does not affect the intestinal epithelial barrier integrity. Large-sized chitin significantly increases acetic acid production by gut microbiota without altering the composition. Exposure of small-sized chitin to THP-1 macrophages lead to significantly increased secretion of IL-1β, IL-8, IL-10, and CXCL10 in a multi-receptor and clathrin-mediated endocytosis dependent manner. CONCLUSIONS These findings indicate that small-sized chitin does not harm the intestinal barrier nor affects SCFA secretion and microbiota composition, but does impact immune activity which could be beneficial to subjects in need of immune support or activation.
Collapse
Affiliation(s)
- Liyou Dong
- Wageningen Food and Biobased ResearchWageningen URBornse Weilanden 96708WGWageningenThe Netherlands
- Laboratory of Food ChemistryWageningen URBornse Weilanden 96708WGWageningenThe Netherlands
| | - Renata M. C. Ariëns
- Wageningen Food and Biobased ResearchWageningen URBornse Weilanden 96708WGWageningenThe Netherlands
| | - Monic M. Tomassen
- Wageningen Food and Biobased ResearchWageningen URBornse Weilanden 96708WGWageningenThe Netherlands
| | - Harry J. Wichers
- Wageningen Food and Biobased ResearchWageningen URBornse Weilanden 96708WGWageningenThe Netherlands
- Laboratory of Food ChemistryWageningen URBornse Weilanden 96708WGWageningenThe Netherlands
| | - Coen Govers
- Wageningen Food and Biobased ResearchWageningen URBornse Weilanden 96708WGWageningenThe Netherlands
| |
Collapse
|
83
|
de Oliveira EB, Xisto MIDDS, Rollin-Pinheiro R, Rochetti VP, Barreto-Bergter E. Peptidorhamnomannans From Scedosporium and Lomentospora Species Display Microbicidal Activity Against Bacteria Commonly Present in Cystic Fibrosis Patients. Front Cell Infect Microbiol 2020; 10:598823. [PMID: 33251161 PMCID: PMC7673444 DOI: 10.3389/fcimb.2020.598823] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/07/2020] [Indexed: 11/13/2022] Open
Abstract
Scedosporium and Lomentospora species are filamentous fungi that cause a wide range of infections in humans. They are usually found in the lungs of cystic fibrosis (CF) patients and are the second most frequent fungal genus after Aspergillus species. Several studies have been recently performed in order to understand how fungi and bacteria interact in CF lungs, since both can be isolated simultaneously from patients. In this context, many bacterial molecules were shown to inhibit fungal growth, but little is known about how fungi could interfere in bacterial development in CF lungs. Scedosporium and Lomentospora species present peptidorhamnomannans (PRMs) in their cell wall that play crucial roles in fungal adhesion and interaction with host epithelial cells and the immune system. The present study aimed to analyze whether PRMs extracted from Lomentospora prolificans, Scedosporium apiospermum, Scedosporium boydii, and Scedosporium aurantiacum block bacterial growth and biofilm formation in vitro. PRM from L. prolificans and S. boydii displayed the best bactericidal effect against methicillin resistant Staphylococcus aureus (MRSA), Burkholderia cepacia, and Escherichia coli, but not Pseudomonas aeruginosa, all of which are the most frequently found bacteria in CF lungs. In addition, biofilm formation was inhibited in all bacteria tested using PRMs at minimal inhibitory concentration (MIC). These results suggest that PRMs from the Scedosporium and Lomentospora surface seem to play an important role in Scedosporium colonization in CF patients, helping to clarify how these pathogens interact to each other in CF lungs.
Collapse
Affiliation(s)
- Evely Bertulino de Oliveira
- Laboratório de Química Biológica de Microrganismos, Instituto de Microbiologia Paulo de Góes, Departamento de Microbiologia Geral, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Mariana Ingrid Dutra da Silva Xisto
- Laboratório de Química Biológica de Microrganismos, Instituto de Microbiologia Paulo de Góes, Departamento de Microbiologia Geral, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Rodrigo Rollin-Pinheiro
- Laboratório de Química Biológica de Microrganismos, Instituto de Microbiologia Paulo de Góes, Departamento de Microbiologia Geral, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Victor Pereira Rochetti
- Laboratório de Química Biológica de Microrganismos, Instituto de Microbiologia Paulo de Góes, Departamento de Microbiologia Geral, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Eliana Barreto-Bergter
- Laboratório de Química Biológica de Microrganismos, Instituto de Microbiologia Paulo de Góes, Departamento de Microbiologia Geral, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| |
Collapse
|
84
|
Pujari R, Banerjee G. Impact of prebiotics on immune response: from the bench to the clinic. Immunol Cell Biol 2020; 99:255-273. [PMID: 32996638 DOI: 10.1111/imcb.12409] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 08/31/2020] [Accepted: 09/29/2020] [Indexed: 12/23/2022]
Abstract
Several preclinical and clinical studies have shown the immunomodulatory role exerted by prebiotics in regulating the immune response. In this review, we describe the mechanistic and clinical studies that decipher the cell signaling pathways implicated in the process. Prebiotic fibers are conventionally known to serve as substrate for probiotic commensal bacteria that release of short-chain fatty acids in the intestinal tract along with several other metabolites. Subsequently, they then act on the local as well as the systemic immune cells and the gut-associated epithelial cells, primarily through G-protein-coupled receptor-mediated pathways. However, other pathways including histone deacetylase inhibition and inflammasome pathway have also been implicated in regulating the immunomodulatory effect. The prebiotics can also induce a microbiota-independent effect by directly acting on the gut-associated epithelial and innate immune cells through the Toll-like receptors. The cumulative effect results in the maintenance of the epithelial barrier integrity and modulation of innate immunity through secretion of pro- and anti-inflammatory cytokines, switches in macrophage polarization and function, neutrophil recruitment and migration, dendritic cell and regulatory T-cell differentiation. Extending these in vitro and ex vivo observations, some prebiotics have been well investigated, with successful human and animal trials demonstrating the association between gut microbes and immunity biomarkers leading to improvement in health endpoints across populations. This review discusses scientific insights into the association between prebiotics, innate immunity and gut microbiome from in vitro to human oral intervention.
Collapse
Affiliation(s)
- Radha Pujari
- Innovation Centre, Tata Chemicals Ltd, Pune, Maharashtra, India
| | - Gautam Banerjee
- Innovation Centre, Tata Chemicals Ltd, Pune, Maharashtra, India
| |
Collapse
|
85
|
Kruschitz A, Nidetzky B. Downstream processing technologies in the biocatalytic production of oligosaccharides. Biotechnol Adv 2020; 43:107568. [DOI: 10.1016/j.biotechadv.2020.107568] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 04/27/2020] [Accepted: 05/17/2020] [Indexed: 12/22/2022]
|
86
|
Addai FP, Lin F, Wang T, Kosiba AA, Sheng P, Yu F, Gu J, Zhou Y, Shi H. Technical integrative approaches to cheese whey valorization towards sustainable environment. Food Funct 2020; 11:8407-8423. [PMID: 32955061 DOI: 10.1039/d0fo01484b] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Whey, a byproduct of cheese production, is often treated as an industrial dairy waste. A large volume of this product is disposed of annually due to inadequate bioconversion approaches. With its high pollutant load, disposal without pretreatment has raised a lot of environmental concerns alerting the need to seek optimal methods for adequately extracting and utilizing its organic content. In recent years, several techniques for whey valorization have emerged which may serve as interventionary measures against its environmental effects after disposal. In this review, we discuss five major approaches, by which whey can be converted into eco-friendly products, to significantly cut whey wastage. The approaches to whey valorization are therefore examined under the following perspectives: whey as a raw material for the production of bioethanol and prebiotic oligosaccharides via β-galactosidase and microbe catalyzed reactions, for the production of refined lactose as an excipient for pharmaceutical purposes, and the clinical significance of whey hydrolysates and their antifungal activity in food processing.
Collapse
Affiliation(s)
- Frank Peprah Addai
- Institute of Life Sciences, Jiangsu University, No. 301 Xuefu Road, Zhenjiang 212013, P. R. China.
| | - Feng Lin
- Key Laboratory of Healthy Freshwater Aquaculture, Ministry of Agriculture, Zhejiang Institute of Freshwater Fisheries, Huzhou 313001, P. R. China
| | - Taotao Wang
- Institute of Life Sciences, Jiangsu University, No. 301 Xuefu Road, Zhenjiang 212013, P. R. China.
| | - Anthony A Kosiba
- Institute of Life Sciences, Jiangsu University, No. 301 Xuefu Road, Zhenjiang 212013, P. R. China.
| | - Pengcheng Sheng
- Key Laboratory of Healthy Freshwater Aquaculture, Ministry of Agriculture, Zhejiang Institute of Freshwater Fisheries, Huzhou 313001, P. R. China
| | - Feng Yu
- Institute of Life Sciences, Jiangsu University, No. 301 Xuefu Road, Zhenjiang 212013, P. R. China.
| | - Jie Gu
- Institute of Life Sciences, Jiangsu University, No. 301 Xuefu Road, Zhenjiang 212013, P. R. China.
| | - Yang Zhou
- Institute of Life Sciences, Jiangsu University, No. 301 Xuefu Road, Zhenjiang 212013, P. R. China.
| | - Haifeng Shi
- Institute of Life Sciences, Jiangsu University, No. 301 Xuefu Road, Zhenjiang 212013, P. R. China.
| |
Collapse
|
87
|
Kim MG, Jo K, Chang YB, Suh HJ, Hong KB. Changes in the Gut Microbiome after Galacto-Oligosaccharide Administration in Loperamide-Induced Constipation. J Pers Med 2020; 10:jpm10040161. [PMID: 33050434 PMCID: PMC7711924 DOI: 10.3390/jpm10040161] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/07/2020] [Accepted: 10/08/2020] [Indexed: 01/02/2023] Open
Abstract
Unbalanced dietary habits and the consumption of high protein and instant foods cause an increase in constipation. Here, we evaluated the effects of galacto-oligosaccharide (GOS) on a rat model of loperamide-induced constipation by measuring various biological markers and cecal microbiota. The fecal water content and intestinal transit ratio significantly increased in the GOS-administered (GL and GH) groups than in the control group (p < 0.05, p < 0.01, and p < 0.001, respectively). The length of intestinal mucosa (p < 0.05 and p < 0.01, respectively) and area of crypt cells were (p < 0.01, both) significantly increased in the GOS-administered groups compared to the control group. The distribution of interstitial cells of Cajal, which is related to the intestinal movement, showed a significant increase in GOS-administered groups than in the control group (p < 0.01, both). The relative abundance of lactic acid bacteria (LAB), especially Lactobacillus and Lactococcus, significantly increased in the GL group than in the control group. Furthermore, there was a positive correlation between short chain fatty acids (SCFAs) and the gut microbiota in the GL groups. These results demonstrated that GOS administration effectively alleviates constipation by increasing LAB proliferation in the intestinal microbiota and SCFA production.
Collapse
Affiliation(s)
| | | | | | | | - Ki-Bae Hong
- Correspondence: ; Tel.: +82-2-940-2853; Fax: +82-2-921-7207
| |
Collapse
|
88
|
Froebel LK, Froebel LE, Duong T. Refined functional carbohydrates reduce adhesion of Salmonella and Campylobacter to poultry epithelial cells in vitro. Poult Sci 2020; 99:7027-7034. [PMID: 33248619 PMCID: PMC7704970 DOI: 10.1016/j.psj.2020.09.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 09/05/2020] [Accepted: 09/11/2020] [Indexed: 10/28/2022] Open
Abstract
The development of interventions to reduce human foodborne pathogens in the gastrointestinal (GI) tract of chickens will be important for improving the microbial food safety of poultry. Saccharomyces-derived prebiotic refined functional carbohydrates (RFC), composed primarily of β-glucans, mannanoligosaccharides (MOS), and D-mannose have been demonstrated to reduce GI colonization of Salmonella and Campylobacter when administered to poultry. Although they are presumed to inhibit adhesion of pathogens to the GI epithelium, this functionality of RFC has not been well characterized. In this study, we investigated the effects of RFC and other prebiotics on the adhesion of Salmonella Typhimurium and Campylobacter jejuni to the LMH chicken epithelial cell line in vitro. The reduction of adherent pathogens was observed to be dose-dependent with C. jejuni being more sensitive than Salmonella to inhibition by RFC. Comparison of the primary constituent carbohydrates of RFC found D-mannose to inhibit both pathogens less effectively than β-glucan and MOS, suggesting that it contributes less to inhibition of pathogen adhesion than the other carbohydrates. Finally, the reduction of adherent pathogens by RFC was compared with that of fructooligosaccharides (FOS), galactooligosaccharides (GOS), and raffinose. All 4 prebiotics inhibited adhesion of both pathogens to chicken epithelial cells. Reduction of adherent Salmonella was greatest with FOS and lowest with GOS, whereas reduction of adherent C. jejuni was greater with RFC and raffinose than with FOS and GOS. These results will inform future research elucidating mechanisms important to adhesion inhibition of pathogens by RFC and other prebiotics.
Collapse
Affiliation(s)
- L K Froebel
- Department of Poultry Science, Texas A&M University, College Station, USA
| | - L E Froebel
- Department of Poultry Science, Texas A&M University, College Station, USA
| | - T Duong
- Department of Poultry Science, Texas A&M University, College Station, USA.
| |
Collapse
|
89
|
Zokaityte E, Cernauskas D, Klupsaite D, Lele V, Starkute V, Zavistanaviciute P, Ruzauskas M, Gruzauskas R, Juodeikiene G, Rocha JM, Bliznikas S, Viskelis P, Ruibys R, Bartkiene E. Bioconversion of Milk Permeate with Selected Lactic Acid Bacteria Strains and Apple By-Products into Beverages with Antimicrobial Properties and Enriched with Galactooligosaccharides. Microorganisms 2020; 8:E1182. [PMID: 32756465 PMCID: PMC7463965 DOI: 10.3390/microorganisms8081182] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 07/30/2020] [Accepted: 08/01/2020] [Indexed: 12/12/2022] Open
Abstract
The present research study aims to prepare prototypes of beverages from milk permeate (MP) using fermentation with 10 different strains of lactic acid bacteria (LAB) showing antimicrobial properties (L. uvarum LUHS245, L. casei LUHS210, L. curvatus LUHS51, L. plantarum LUHS135, P. acidilactici LUHS29, L. plantarum LUHS122, L. coryniformins LUHS71, L. paracasei LUHS244, P. pentosaceus LUHS183, L. faraginis LUHS206) and MP with (AppMP) or without (MP) the addition of 8% (w/w) apple by-products (App). Two groups of prototypes of beverages were prepared: fermented MP and fermented MP with App (AppMP). Acidity parameters, LAB viable counts, lactose and galactooligosaccharides (GOSs) content, antimicrobial properties against 15 pathogenic and opportunistic bacterial strains, overall acceptability and emotions induced of the final fermented beverages for consumers were evaluated. Results showed that all LAB grew well in MP and LAB strain exhibited a significant (p ≤ 0.05) influence on galactobiose and galactotriose synthesis in the fermentable MP substrate. The highest total content of GOS (26.80 mg/100 mL) was found in MPLUHS29 fermented beverage. In addition, MPLUHS245, MPLUHS210 and AppMPLUHS71 fermented beverages showed high antimicrobial activity, inhibiting 13 out of 15 tested microbial pathogens. The overall acceptability of AppMP fermented beverages was 26.8% higher when compared with fermented beverages without App (MP), and the most intensive "happy" emotion was induced by MPLUHS71, MPLUHS24, MPLUHS183 and MPLUHS206 samples. Finally, very promising results were also attained by the bioconversion of MP with selected LAB and App addition into the prototypes of antimicrobial beverages enriched with GOS.
Collapse
Affiliation(s)
- Egle Zokaityte
- Institute of Animal Rearing Technologies, Faculty of Animal Sciences, Lithuanian University of Health Sciences, Mickeviciaus str. 9, LT-44307 Kaunas, Lithuania; (E.Z.); (D.C.); (D.K.); (V.L.); (V.S.); (P.Z.); (S.B.)
- Department of Food Safety and Quality, Faculty of Veterinary, Lithuanian University of Health Sciences, Mickeviciaus str. 9, LT-44307 Kaunas, Lithuania
| | - Darius Cernauskas
- Institute of Animal Rearing Technologies, Faculty of Animal Sciences, Lithuanian University of Health Sciences, Mickeviciaus str. 9, LT-44307 Kaunas, Lithuania; (E.Z.); (D.C.); (D.K.); (V.L.); (V.S.); (P.Z.); (S.B.)
- Food Institute, Kaunas University of Technology, Radvilenu rd. 19, LT-50254 Kaunas, Lithuania
| | - Dovile Klupsaite
- Institute of Animal Rearing Technologies, Faculty of Animal Sciences, Lithuanian University of Health Sciences, Mickeviciaus str. 9, LT-44307 Kaunas, Lithuania; (E.Z.); (D.C.); (D.K.); (V.L.); (V.S.); (P.Z.); (S.B.)
| | - Vita Lele
- Institute of Animal Rearing Technologies, Faculty of Animal Sciences, Lithuanian University of Health Sciences, Mickeviciaus str. 9, LT-44307 Kaunas, Lithuania; (E.Z.); (D.C.); (D.K.); (V.L.); (V.S.); (P.Z.); (S.B.)
- Department of Food Safety and Quality, Faculty of Veterinary, Lithuanian University of Health Sciences, Mickeviciaus str. 9, LT-44307 Kaunas, Lithuania
| | - Vytaute Starkute
- Institute of Animal Rearing Technologies, Faculty of Animal Sciences, Lithuanian University of Health Sciences, Mickeviciaus str. 9, LT-44307 Kaunas, Lithuania; (E.Z.); (D.C.); (D.K.); (V.L.); (V.S.); (P.Z.); (S.B.)
- Department of Food Safety and Quality, Faculty of Veterinary, Lithuanian University of Health Sciences, Mickeviciaus str. 9, LT-44307 Kaunas, Lithuania
| | - Paulina Zavistanaviciute
- Institute of Animal Rearing Technologies, Faculty of Animal Sciences, Lithuanian University of Health Sciences, Mickeviciaus str. 9, LT-44307 Kaunas, Lithuania; (E.Z.); (D.C.); (D.K.); (V.L.); (V.S.); (P.Z.); (S.B.)
- Department of Food Safety and Quality, Faculty of Veterinary, Lithuanian University of Health Sciences, Mickeviciaus str. 9, LT-44307 Kaunas, Lithuania
| | - Modestas Ruzauskas
- Department of Anatomy and Physiology, Faculty of Veterinary, Lithuanian University of Health Sciences, Mickeviciaus str. 9, LT-44307 Kaunas, Lithuania;
- Institute of Microbiology and Virology, Faculty of Veterinary, Lithuanian University of Health Sciences, Mickeviciaus str. 9, LT-44307 Kaunas, Lithuania
| | - Romas Gruzauskas
- Department of Food Science and Technology, Kaunas University of Technology, Radvilenu rd. 19, LT-50254 Kaunas, Lithuania; (R.G.); (G.J.)
| | - Grazina Juodeikiene
- Department of Food Science and Technology, Kaunas University of Technology, Radvilenu rd. 19, LT-50254 Kaunas, Lithuania; (R.G.); (G.J.)
| | - João Miguel Rocha
- REQUIMTE–Rede de Química e Tecnologia, Laboratório de Química Verde (LAQV), Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto (FCUP), Rua do Campo Alegre, s/n., P-4169-007 Porto, Portugal
| | - Saulius Bliznikas
- Institute of Animal Rearing Technologies, Faculty of Animal Sciences, Lithuanian University of Health Sciences, Mickeviciaus str. 9, LT-44307 Kaunas, Lithuania; (E.Z.); (D.C.); (D.K.); (V.L.); (V.S.); (P.Z.); (S.B.)
| | - Pranas Viskelis
- Institute of Horticulture, Lithuanian Research Centre for Agriculture and Forestry, Kauno str. 30, LT-54333 Babtai, Lithuania;
| | - Romas Ruibys
- Institute of Agricultural and Food Sciences, Agriculture Academy, Vytautas Magnus University, K. Donelaicio str. 58, LT-44244 Kaunas, Lithuania;
| | - Elena Bartkiene
- Institute of Animal Rearing Technologies, Faculty of Animal Sciences, Lithuanian University of Health Sciences, Mickeviciaus str. 9, LT-44307 Kaunas, Lithuania; (E.Z.); (D.C.); (D.K.); (V.L.); (V.S.); (P.Z.); (S.B.)
- Department of Food Safety and Quality, Faculty of Veterinary, Lithuanian University of Health Sciences, Mickeviciaus str. 9, LT-44307 Kaunas, Lithuania
| |
Collapse
|
90
|
Božić A, Anderson RC, Crippen TL, Swaggerty CL, Hume ME, Beier RC, He H, Genovese KJ, Poole TL, Harvey RB, Nisbet DJ. Inhibition of Salmonella Binding to Porcine Intestinal Cells by a Wood-Derived Prebiotic. Microorganisms 2020; 8:microorganisms8071051. [PMID: 32679904 PMCID: PMC7409177 DOI: 10.3390/microorganisms8071051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/07/2020] [Accepted: 07/14/2020] [Indexed: 01/21/2023] Open
Abstract
Numerous Salmonellaenterica serovars can cause disease and contamination of animal-produced foods. Oligosaccharide-rich products capable of blocking pathogen adherence to intestinal mucosa are attractive alternatives to antibiotics as these have potential to prevent enteric infections. Presently, a wood-derived prebiotic composed mainly of glucose-galactose-mannose-xylose oligomers was found to inhibit mannose-sensitive binding of select SalmonellaTyphimurium and Escherichia coli strains when reacted with Saccharomyces boulardii. Tests for the ability of the prebiotic to prevent binding of a green fluorescent protein (GFP)-labeled S.Typhimurium to intestinal porcine epithelial cells (IPEC-J2) cultured in vitro revealed that prebiotic-exposed GFP-labeled S.Typhimurium bound > 30% fewer individual IPEC-J2 cells than did GFP-labeled S.Typhimurium having no prebiotic exposure. Quantitatively, 90% fewer prebiotic-exposed GFP-labeled S.Typhimurium cells were bound per individual IPEC-J2 cell compared to non-prebiotic exposed GFP-labeled S.Typhimurium. Comparison of invasiveness of S.Typhimurium DT104 against IPEC-J2 cells revealed greater than a 90% decrease in intracellular recovery of prebiotic-exposed S.Typhimurium DT104 compared to non-exposed controls (averaging 4.4 ± 0.2 log10 CFU/well). These results suggest compounds within the wood-derived prebiotic bound to E. coli and S.Typhimurium-produced adhesions and in the case of S.Typhimurium, this adhesion-binding activity inhibited the binding and invasion of IPEC-J2 cells.
Collapse
Affiliation(s)
- Aleksandar Božić
- Faculty of Agriculture, University of Novi Sad, Novi Sad 21000, Serbia;
- Food and Feed Safety Research Unit, United States Department of Agriculture/Agricultural Research Service, College Station, TX 77845, USA; (T.L.C.); (C.L.S.); (M.E.H.); (R.C.B.); (H.H.); (K.J.G.); (T.L.P.); (R.B.H.); (D.J.N.)
| | - Robin C. Anderson
- Food and Feed Safety Research Unit, United States Department of Agriculture/Agricultural Research Service, College Station, TX 77845, USA; (T.L.C.); (C.L.S.); (M.E.H.); (R.C.B.); (H.H.); (K.J.G.); (T.L.P.); (R.B.H.); (D.J.N.)
- Correspondence:
| | - Tawni L. Crippen
- Food and Feed Safety Research Unit, United States Department of Agriculture/Agricultural Research Service, College Station, TX 77845, USA; (T.L.C.); (C.L.S.); (M.E.H.); (R.C.B.); (H.H.); (K.J.G.); (T.L.P.); (R.B.H.); (D.J.N.)
| | - Christina L. Swaggerty
- Food and Feed Safety Research Unit, United States Department of Agriculture/Agricultural Research Service, College Station, TX 77845, USA; (T.L.C.); (C.L.S.); (M.E.H.); (R.C.B.); (H.H.); (K.J.G.); (T.L.P.); (R.B.H.); (D.J.N.)
| | - Michael E. Hume
- Food and Feed Safety Research Unit, United States Department of Agriculture/Agricultural Research Service, College Station, TX 77845, USA; (T.L.C.); (C.L.S.); (M.E.H.); (R.C.B.); (H.H.); (K.J.G.); (T.L.P.); (R.B.H.); (D.J.N.)
| | - Ross C. Beier
- Food and Feed Safety Research Unit, United States Department of Agriculture/Agricultural Research Service, College Station, TX 77845, USA; (T.L.C.); (C.L.S.); (M.E.H.); (R.C.B.); (H.H.); (K.J.G.); (T.L.P.); (R.B.H.); (D.J.N.)
| | - Haiqi He
- Food and Feed Safety Research Unit, United States Department of Agriculture/Agricultural Research Service, College Station, TX 77845, USA; (T.L.C.); (C.L.S.); (M.E.H.); (R.C.B.); (H.H.); (K.J.G.); (T.L.P.); (R.B.H.); (D.J.N.)
| | - Kenneth J. Genovese
- Food and Feed Safety Research Unit, United States Department of Agriculture/Agricultural Research Service, College Station, TX 77845, USA; (T.L.C.); (C.L.S.); (M.E.H.); (R.C.B.); (H.H.); (K.J.G.); (T.L.P.); (R.B.H.); (D.J.N.)
| | - Toni L. Poole
- Food and Feed Safety Research Unit, United States Department of Agriculture/Agricultural Research Service, College Station, TX 77845, USA; (T.L.C.); (C.L.S.); (M.E.H.); (R.C.B.); (H.H.); (K.J.G.); (T.L.P.); (R.B.H.); (D.J.N.)
| | - Roger B. Harvey
- Food and Feed Safety Research Unit, United States Department of Agriculture/Agricultural Research Service, College Station, TX 77845, USA; (T.L.C.); (C.L.S.); (M.E.H.); (R.C.B.); (H.H.); (K.J.G.); (T.L.P.); (R.B.H.); (D.J.N.)
| | - David J. Nisbet
- Food and Feed Safety Research Unit, United States Department of Agriculture/Agricultural Research Service, College Station, TX 77845, USA; (T.L.C.); (C.L.S.); (M.E.H.); (R.C.B.); (H.H.); (K.J.G.); (T.L.P.); (R.B.H.); (D.J.N.)
| |
Collapse
|
91
|
Anti-Pathogenic Functions of Non-Digestible Oligosaccharides In Vitro. Nutrients 2020; 12:nu12061789. [PMID: 32560186 PMCID: PMC7353314 DOI: 10.3390/nu12061789] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/10/2020] [Accepted: 06/12/2020] [Indexed: 12/11/2022] Open
Abstract
Non-digestible oligosaccharides (NDOs), complex carbohydrates that resist hydrolysis by salivary and intestinal digestive enzymes, fulfill a diversity of important biological roles. A lot of NDOs are known for their prebiotic properties by stimulating beneficial bacteria in the intestinal microbiota. Human milk oligosaccharides (HMOs) represent the first prebiotics that humans encounter in life. Inspired by these HMO structures, chemically-produced NDO structures (e.g., galacto-oligosaccharides and chito-oligosaccharides) have been recognized as valuable food additives and exert promising health effects. Besides their apparent ability to stimulate beneficial microbial species, oligosaccharides have shown to be important inhibitors of the development of pathogenic infections. Depending on the type and structural characteristics, oligosaccharides can exert a number of anti-pathogenic effects. The most described effect is their ability to act as a decoy receptor, thereby inhibiting adhesion of pathogens. Other ways of pathogenic inhibition, such as interference with pathogenic cell membrane and biofilm integrity and DNA transcription, are less investigated, but could be equally impactful. In this review, a comprehensive overview of In vitro anti-pathogenic properties of different NDOs and associated pathways are discussed. A framework is created categorizing all anti-pathogenic effects and providing insight into structural necessities for an oligosaccharide to exert one of these effects.
Collapse
|
92
|
Gut microbiota modulation: a novel strategy for prevention and treatment of colorectal cancer. Oncogene 2020; 39:4925-4943. [PMID: 32514151 PMCID: PMC7314664 DOI: 10.1038/s41388-020-1341-1] [Citation(s) in RCA: 366] [Impact Index Per Article: 73.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/17/2020] [Accepted: 05/27/2020] [Indexed: 02/08/2023]
Abstract
Research about the role of gut microbiome in colorectal cancer (CRC) is a newly emerging field of study. Gut microbiota modulation, with the aim to reverse established microbial dysbiosis, is a novel strategy for prevention and treatment of CRC. Different strategies including probiotics, prebiotics, postbiotics, antibiotics, and fecal microbiota transplantation (FMT) have been employed. Although these strategies show promising results, mechanistically by correcting microbiota composition, modulating innate immune system, enhancing gut barrier function, preventing pathogen colonization and exerting selective cytotoxicity against tumor cells, it should be noted that they are accompanied by risks and controversies that can potentially introduce clinical complications. During bench-to-bedside translation, evaluation of risk-and-benefit ratio, as well as patient selection, should be carefully performed. In view of the individualized host response to gut microbiome intervention, developing personalized microbiome therapy may be the key to successful clinical treatment.
Collapse
|
93
|
Ayuso M, Michiels J, Wuyts S, Yan H, Degroote J, Lebeer S, Le Bourgot C, Apper E, Majdeddin M, Van Noten N, Vanden Hole C, Van Cruchten S, Van Poucke M, Peelman L, Van Ginneken C. Short-chain fructo-oligosaccharides supplementation to suckling piglets: Assessment of pre- and post-weaning performance and gut health. PLoS One 2020; 15:e0233910. [PMID: 32502215 PMCID: PMC7274435 DOI: 10.1371/journal.pone.0233910] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 05/14/2020] [Indexed: 01/04/2023] Open
Abstract
Farmers face difficulties in redeeming their investment in larger litter sizes since this comes with larger litter heterogenicity, lower litter resilience and risk of higher mortality. Dietary oligosaccharides, given to the sow, proved beneficial for the offspring’s performance. However, giving oligosaccharides to the suckling piglet is poorly explored. Therefore, this field trial studied the effect of dietary short-chain fructo-oligosaccharides (scFOS; 1g/day; drenched) supplementation to low (LBW, lower quartile), normal (NBW, two intermediate quartiles) and high (HBW, upper quartile) birth weight piglets from birth until 7 or 21 days of age. Performance parameters, gut microbiome and short-chain fatty acids profile of feces and digesta were assessed at birth (d 0), d 7, weaning (d 21.5) and 2 weeks post-weaning (d 36.5). Additional parameters reflecting gut health (intestinal integrity and morphology, mucosal immune system) were analysed at d 36.5. Most parameters changed with age or differed with the piglet’s birth weight. Drenching with scFOS increased body weight by 1 kg in NBW suckling piglets and reduced the post-weaning mortality rate by a 100%. No clear difference in the IgG level, the microbiota composition and fermentative activity between the treatment groups was observed. Additionnally, intestinal integrity, determined by measuring intestinal permeability and regenerative capacity, was similar between the treatment groups. Also, intestinal architecture (villus lenght, crypt depth) was not affected by scFOS supplementation. The density of intra-epithelial lymphocytes and the expression profiles (real-time qPCR) for immune system-related genes (IL-10, IL-1ß, IL-6, TNFα and IFNγ) were used to assess mucosal immunity. Only IFNγ expression, was upregulated in piglets that received scFOS for 7 days. The improved body weight and the reduced post-weaning mortality seen in piglets supplemented with scFOS support the view that scFOS positively impact piglet’s health and resilience. However, the modes of action for these effects are not yet fully elucidated and its potential to improve other performance parameters needs further investigation.
Collapse
Affiliation(s)
- Miriam Ayuso
- Department of Veterinary Medicine, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
- * E-mail:
| | - Joris Michiels
- Department of Animal Sciences and Aquatic Ecology, Laboratory for Animal Production and Animal Product Quality, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Sander Wuyts
- Department of Bioengineering, Faculty of Sciences, University of Antwerp, Wilrijk, Belgium
| | - Honglin Yan
- Department of Animal Sciences and Aquatic Ecology, Laboratory for Animal Production and Animal Product Quality, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Jeroen Degroote
- Department of Animal Sciences and Aquatic Ecology, Laboratory for Animal Production and Animal Product Quality, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Sarah Lebeer
- Department of Bioengineering, Faculty of Sciences, University of Antwerp, Wilrijk, Belgium
| | | | | | - Maryam Majdeddin
- Department of Animal Sciences and Aquatic Ecology, Laboratory for Animal Production and Animal Product Quality, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Noémie Van Noten
- Department of Animal Sciences and Aquatic Ecology, Laboratory for Animal Production and Animal Product Quality, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Charlotte Vanden Hole
- Department of Veterinary Medicine, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Steven Van Cruchten
- Department of Veterinary Medicine, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Mario Van Poucke
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Luc Peelman
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Chris Van Ginneken
- Department of Veterinary Medicine, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
94
|
Gerbino E, Ghibaudo F, Tymczyszyn EE, Gomez-Zavaglia A, Hugo AA. Probiotics, Galacto-oligosaccharides, and zinc antagonize biological effects of enterohaemorrhagic Escherichia coli on cultured cells and brine shrimp model. Lebensm Wiss Technol 2020. [DOI: 10.1016/j.lwt.2020.109435] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
95
|
Zhou JC, Zhang XW. Akkermansia muciniphila: a promising target for the therapy of metabolic syndrome and related diseases. Chin J Nat Med 2020; 17:835-841. [PMID: 31831130 DOI: 10.1016/s1875-5364(19)30101-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Indexed: 12/18/2022]
Abstract
The probiotic Akkermansia muciniphila (A. muciniphila) is an intestinal bacterium that was first identified in human feces in 2004. Its specialization in mucin degradation makes it a key microorganism that maintains intestinal mucosal barrier function. As an unique representative strain of the phylum Verrucomicrobia that can be cultured in vitro, A. muciniphila is much easier to detect by metagenomic analysis of intestinal flora. In the past few years, A. muciniphila has been getting increasing attention for the positive correlation between its intestinal colonization and host homeostatic metabolism. In this review, we summarize the relationship between A. muciniphila and host health and diseases, especially focusing on metabolic diseases and related mechanisms, as well as the natural food and drug-derived substrates affecting its colonization in the host, expecting to provide evidence and clues for the development of drugs targeting A. muciniphila.
Collapse
Affiliation(s)
- Ji-Chao Zhou
- Molecular Immunology and Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100000, China
| | - Xiao-Wei Zhang
- Molecular Immunology and Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100000, China.
| |
Collapse
|
96
|
Wojciechowska A, Klewicki R, Klewicka E. The potential of new bionic acids as prebiotics and antimicrobials. Lebensm Wiss Technol 2020. [DOI: 10.1016/j.lwt.2020.109246] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
97
|
Lafontaine GMF, Fish NM, Connerton IF. In Vitro Evaluation of the Effects of Commercial Prebiotic GOS and FOS Products on Human Colonic Caco-2 Cells. Nutrients 2020; 12:nu12051281. [PMID: 32366023 PMCID: PMC7282019 DOI: 10.3390/nu12051281] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/21/2020] [Accepted: 04/28/2020] [Indexed: 01/04/2023] Open
Abstract
Prebiotic oligosaccharides are widely used as human and animal feed additives for their beneficial effects on the gut microbiota. However, there are limited data to assess the direct effect of such functional foods on the transcriptome of intestinal epithelial cells. The purpose of this study is to describe the differential transcriptomes and cellular pathways of colonic cells directly exposed to galacto-oligosaccharides (GOS) and fructo-oligosaccharides (FOS). We have examined the differential gene expression of polarized Caco–2 cells treated with GOS or FOS products and their respective mock-treated cells using mRNA sequencing (RNA-seq). A total of 89 significant differentially expressed genes were identified between GOS and mock-treated groups. For FOS treatment, a reduced number of 12 significant genes were observed to be differentially expressed relative to the control group. KEGG and gene ontology functional analysis revealed that genes up-regulated in the presence of GOS were involved in digestion and absorption processes, fatty acids and steroids metabolism, potential antimicrobial proteins, energy-dependent and -independent transmembrane trafficking of solutes and amino acids. Using our data, we have established complementary non-prebiotic modes of action for these frequently used dietary fibers.
Collapse
Affiliation(s)
- Geraldine M. Flaujac Lafontaine
- Division of Microbiology, Brewing and Biotechnology, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, UK;
| | - Neville M. Fish
- Saputo Dairy UK, Innovation Centre, Harper Adams University, Newport TF10 8NB, UK;
| | - Ian F. Connerton
- Division of Microbiology, Brewing and Biotechnology, School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, UK;
- Correspondence:
| |
Collapse
|
98
|
Differential effects of inulin or its fermentation metabolites on gut barrier and immune function of porcine intestinal epithelial cells. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.103855] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
99
|
Optimization for galactooligosaccharides synthesis: A potential alternative for gut health and immunity. Life Sci 2020; 245:117353. [DOI: 10.1016/j.lfs.2020.117353] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 01/20/2020] [Accepted: 01/22/2020] [Indexed: 11/23/2022]
|
100
|
Kariyawasam KMGMM, Yang SJ, Lee NK, Paik HD. Probiotic Properties of Lactobacillus brevis KU200019 and Synergistic Activity with Fructooligosaccharides in Antagonistic Activity against Foodborne Pathogens. Food Sci Anim Resour 2020; 40:297-310. [PMID: 32161924 PMCID: PMC7057033 DOI: 10.5851/kosfa.2020.e15] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/11/2020] [Accepted: 02/16/2020] [Indexed: 12/15/2022] Open
Abstract
This study aims to evaluate the probiotic properties of Lactobacillus
brevis (L. brevis) KU200019 and the synergistic activity with
prebiotics on antimicrobial activity, and the potential application as an
adjunct culture in fermented dairy products. The commercial strain, L.
brevis ATCC 14869 was used as reference strain. L.
brevis KU200019 was showed higher viability in simulated gastric
(99.38±0.21%) and bile (115.10±0.13%) conditions
compared to reference strain. L. brevis KU200019 exhibited
antimicrobial activity against various foodborne pathogens. The supplementation
of fructooligosaccharides (FOS) enhanced viability of lactic acid bacteria
(>8 Log CFU/mL) and antioxidant activity [2,2-diphenyl-2-picrylhydrazyl
radical assay (DPPH) assay, 31.23±1.14%; 2,2-azinobis
(3-ethylbenzothiazoline-6-sulfonic acid (ABTS) assay, 38.82±1.46%]
in fermented skim milk during refrigerated storage. L. brevis
KU200019 was distinguished from the reference strain by its higher probiotic
potential, antimicrobial activity, and higher antioxidant activity in fermented
milk. Therefore, L. brevis KU200019 with FOS was demonstrated
promising properties for further application in fermented dairy products with
enhanced safety and quality.
Collapse
Affiliation(s)
| | - Seo Jin Yang
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Korea
| | - Na-Kyoung Lee
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Korea
| | - Hyun-Dong Paik
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Korea
| |
Collapse
|