1
|
Basnet J, Eissa MA, Cardozo LLY, Romero DG, Rezq S. Impact of Probiotics and Prebiotics on Gut Microbiome and Hormonal Regulation. GASTROINTESTINAL DISORDERS 2024; 6:801-815. [PMID: 39649015 PMCID: PMC11623347 DOI: 10.3390/gidisord6040056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2024] Open
Abstract
The gut microbiome plays a crucial role in human health by influencing various physiological functions through complex interactions with the endocrine system. These interactions involve the production of metabolites, signaling molecules, and direct communication with endocrine cells, which modulate hormone secretion and activity. As a result, the microbiome can exert neuroendocrine effects and contribute to metabolic regulation, adiposity, and appetite control. Additionally, the gut microbiome influences reproductive health by altering levels of sex hormones such as estrogen and testosterone, potentially contributing to conditions like polycystic ovary syndrome (PCOS) and hypogonadism. Given these roles, targeting the gut microbiome offers researchers and clinicians novel opportunities to improve overall health and well-being. Probiotics, such as Lactobacillus and Bifidobacterium, are live beneficial microbes that help maintain gut health by balancing the microbiota. Prebiotics, non-digestible fibers, nourish these beneficial bacteria, promoting their growth and activity. When combined, probiotics and prebiotics form synbiotics, which work synergistically to enhance the gut microbiota balance and improve metabolic, immune, and hormonal health. This integrated approach shows promising potential for managing conditions related to hormonal imbalances, though further research is needed to fully understand their specific mechanisms and therapeutic potential.
Collapse
Affiliation(s)
- Jelina Basnet
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Manar A. Eissa
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Licy L. Yanes Cardozo
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Department of Medicine, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS 39216, USA
| | - Damian G. Romero
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Samar Rezq
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
2
|
Jia X, Chen Q, Zhang Y, Asakawa T. Multidirectional associations between the gut microbiota and Parkinson's disease, updated information from the perspectives of humoral pathway, cellular immune pathway and neuronal pathway. Front Cell Infect Microbiol 2023; 13:1296713. [PMID: 38173790 PMCID: PMC10762314 DOI: 10.3389/fcimb.2023.1296713] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024] Open
Abstract
The human gastrointestinal tract is inhabited by a diverse range of microorganisms, collectively known as the gut microbiota, which form a vast and complex ecosystem. It has been reported that the microbiota-gut-brain axis plays a crucial role in regulating host neuroprotective function. Studies have shown that patients with Parkinson's disease (PD) have dysbiosis of the gut microbiota, and experiments involving germ-free mice and fecal microbiota transplantation from PD patients have revealed the pathogenic role of the gut microbiota in PD. Interventions targeting the gut microbiota in PD, including the use of prebiotics, probiotics, and fecal microbiota transplantation, have also shown efficacy in treating PD. However, the causal relationship between the gut microbiota and Parkinson's disease remains intricate. This study reviewed the association between the microbiota-gut-brain axis and PD from the perspectives of humoral pathway, cellular immune pathway and neuronal pathway. We found that the interactions among gut microbiota and PD are very complex, which should be "multidirectional", rather than conventionally regarded "bidirectional". To realize application of the gut microbiota-related mechanisms in the clinical setting, we propose several problems which should be addressed in the future study.
Collapse
Affiliation(s)
- Xiaokang Jia
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Qiliang Chen
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yuanyuan Zhang
- Department of Acupuncture and Moxibustion, The Affiliated Traditional Chinese Medicine (TCM) Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Tetsuya Asakawa
- Institute of Neurology, National Clinical Research Center for Infectious Diseases, the Third People’s Hospital of Shenzhen, Shenzhen, Guangdong, China
| |
Collapse
|
3
|
van der Toorn M, Chatziioannou AC, Pellis L, Haandrikman A, van der Zee L, Dijkhuizen L. Biological Relevance of Goat Milk Oligosaccharides to Infant Health. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:13935-13949. [PMID: 37691562 PMCID: PMC10540210 DOI: 10.1021/acs.jafc.3c02194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/12/2023]
Abstract
Milk is often regarded as the gold standard for the nourishment of all mammalian offspring. The World Health Organization (WHO) recommends exclusive breastfeeding for the first 6 months of the life of the infant, followed by a slow introduction of complementary foods to the breastfeeding routine for a period of approximately 2 years, whenever this is possible ( Global Strategy for Infant and Young Child Feeding; WHO, 2003). One of the most abundant components in all mammals' milk, which is associated with important health benefits, is the oligosaccharides. The milk oligosaccharides (MOS) of humans and other mammals differ in terms of their concentration and diversity. Among those, goat milk contains more oligosaccharides (gMOS) than other domesticated dairy animals, as well as a greater range of structures. This review summarizes the biological functions of MOS found in both human and goat milk to identify the possible biological relevance of gMOS in human health and development. Based on the existing literature, seven biological functions of gMOS were identified, namely, MOS action as prebiotics, immune modulators, and pathogen traps; their modulation of intestinal cells; protective effect against necrotizing enterocolitis; improved brain development; and positive effects on stressor exposure. Overall, goat milk is a viable alternate supply of functional MOS that could be employed in a newborn formula.
Collapse
Affiliation(s)
| | - Anastasia Chrysovalantou Chatziioannou
- CarbExplore
Research BV, Groningen, 9747 AN The Netherlands
- Department
of Chemistry, Laboratory of Analytical Biochemistry, University of Crete, Heraklion, 70013, Greece
| | | | | | | | - Lubbert Dijkhuizen
- CarbExplore
Research BV, Groningen, 9747 AN The Netherlands
- Microbial
Physiology, Groningen Biomolecular Sciences and Biotechnology Institute
(GBB), University of Groningen, Groningen, 9747 AG, The Netherlands
| |
Collapse
|
4
|
Wang Y, Zhu J, Zou N, Zhang L, Wang Y, Zhang M, Wang C, Yang L. Pathogenesis from the microbial-gut-brain axis in white matter injury in preterm infants: A review. Front Integr Neurosci 2023; 17:1051689. [PMID: 37006416 PMCID: PMC10060642 DOI: 10.3389/fnint.2023.1051689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
White matter injury (WMI) in premature infants is a unique form of brain injury and a common cause of chronic nervous system conditions such as cerebral palsy and neurobehavioral disorders. Very preterm infants who survive are at high risk of WMI. With developing research regarding the pathogenesis of premature WMI, the role of gut microbiota has attracted increasing attention in this field. As premature infants are a special group, early microbial colonization of the microbiome can affect brain development, and microbiome optimization can improve outcomes regarding nervous system development. As an important communication medium between the gut and the nervous system, intestinal microbes form a microbial-gut-brain axis. This axis affects the occurrence of WMI in premature infants via the metabolites produced by intestinal microorganisms, while also regulating cytokines and mediating oxidative stress. At the same time, deficiencies in the microbiota and their metabolites may exacerbate WMI in premature infants. This confers promise for probiotics and prebiotics as treatments for improving neurodevelopmental outcomes. Therefore, this review attempted to elucidate the potential mechanisms behind the communication of gut bacteria and the immature brain through the gut-brain axis, so as to provide a reference for further prevention and treatment of premature WMI.
Collapse
|
5
|
Abstract
Abnormalities in gut microbiota have been suggested to be involved in the pathophysiology and progression of Parkinson's disease (PD). Gastrointestinal nonmotor symptoms often precede the onset of motor features in PD, suggesting a role for gut dysbiosis in neuroinflammation and α-synuclein (α-syn) aggregation. In the first part of this chapter, we analyze critical features of healthy gut microbiota and factors (environmental and genetic) that modify its composition. In the second part, we focus on the mechanisms underlying the gut dysbiosis and how it alters anatomically and functionally the mucosal barrier, triggering neuroinflammation and subsequently α-syn aggregation. In the third part, we describe the most common alterations in the gut microbiota of PD patients, dividing the gastrointestinal system in higher and lower tract to examine the association between microbiota abnormalities and clinical features. In the final section, we report on current and future therapeutic approaches to gut dysbiosis aiming to either reduce the risk for PD, modify the disease course, or improve the pharmacokinetic profile of dopaminergic therapies. We also suggest that further studies will be needed to clarify the role of the microbiome in PD subtyping and of pharmacological and nonpharmacological interventions in modifying specific microbiota profiles in individualizing disease-modifying treatments in PD.
Collapse
Affiliation(s)
- Salvatore Bonvegna
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Department of Clinical Neurosciences, Parkinson and Movement Disorders Unit, Milan, Italy
| | - Roberto Cilia
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Department of Clinical Neurosciences, Parkinson and Movement Disorders Unit, Milan, Italy.
| |
Collapse
|
6
|
Can breastfeeding promote an antibiotic-resilient microbiome? MED 2023; 4:67-68. [PMID: 36773598 DOI: 10.1016/j.medj.2023.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
In this issue of Med, Dai, Petersen, and colleagues report that the antibiotic-associated risk of asthma in children may be mitigated by breastfeeding in early life. They find that breastfeeding promotes a specific microbiome composition responsible for this resilience, mainly driven by the bacterium Bifidobacterium longum subspecies infantis.
Collapse
|
7
|
Huang Y, Lu W, Zeng M, Hu X, Su Z, Liu Y, Liu Z, Yuan J, Li L, Zhang X, Huang L, Hu W, Wang X, Li S, Zhang H. Mapping the early life gut microbiome in neonates with critical congenital heart disease: multiomics insights and implications for host metabolic and immunological health. MICROBIOME 2022; 10:245. [PMID: 36581858 PMCID: PMC9801562 DOI: 10.1186/s40168-022-01437-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 11/25/2022] [Indexed: 05/25/2023]
Abstract
BACKGROUND The early life gut microbiome is crucial in maintaining host metabolic and immune homeostasis. Though neonates with critical congenital heart disease (CCHD) are at substantial risks of malnutrition and immune imbalance, the microbial links to CCHD pathophysiology remain poorly understood. In this study, we aimed to investigate the gut microbiome in neonates with CCHD in association with metabolomic traits. Moreover, we explored the clinical implications of the host-microbe interactions in CCHD. METHODS Deep metagenomic sequencing and metabolomic profiling of paired fecal samples from 45 neonates with CCHD and 50 healthy controls were performed. The characteristics of gut microbiome were investigated in three dimensions (microbial abundance, functionality, and genetic variation). An in-depth analysis of gut virome was conducted to elucidate the ecological interaction between gut viral and bacterial communities. Correlations between multilevel microbial features and fecal metabolites were determined using integrated association analysis. Finally, we conducted a subgroup analysis to examine whether the interactions between gut microbiota and metabolites could mediate inflammatory responses and poor surgical prognosis. RESULTS Gut microbiota dysbiosis was observed in neonates with CCHD, characterized by the depletion of Bifidobacterium and overgrowth of Enterococcus, which was highly correlated with metabolomic perturbations. Genetic variations of Bifidobacterium and Enterococcus orchestrate the metabolomic perturbations in CCHD. A temperate core virome represented by Siphoviridae was identified to be implicated in shaping the gut bacterial composition by modifying microbial adaptation. The overgrowth of Enterococcus was correlated with systemic inflammation and poor surgical prognosis in subgroup analysis. Mediation analysis indicated that the overgrowth of Enterococcus could mediate gut barrier impairment and inflammatory responses in CCHD. CONCLUSIONS We demonstrate for the first time that an aberrant gut microbiome associated with metabolomic perturbations is implicated in immune imbalance and adverse clinical outcomes in neonates with CCHD. Our data support the importance of reconstituting optimal gut microbiome in maintaining host metabolic and immunological homeostasis in CCHD. Video Abstract.
Collapse
Affiliation(s)
- Yuan Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Pediatric Cardiac Surgery Center, Fuwai Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College, Beijing, China
| | - Wenlong Lu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Pediatric Cardiac Surgery Center, Fuwai Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College, Beijing, China
| | - Min Zeng
- PICU, Pediatric Cardiac Center, Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xiaoyue Hu
- Department of Neonatology, Affiliated Children's Hospital of Capital Institute of Pediatrics, Beijing, China
| | - Zhanhao Su
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Pediatric Cardiac Surgery Center, Fuwai Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College, Beijing, China
| | - Yiwei Liu
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zeye Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Pediatric Cardiac Surgery Center, Fuwai Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College, Beijing, China
| | - Jianhui Yuan
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Pediatric Cardiac Surgery Center, Fuwai Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College, Beijing, China
| | - Li Li
- Department of Neonatology, Affiliated Children's Hospital of Capital Institute of Pediatrics, Beijing, China
| | - Xiaoling Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Pediatric Cardiac Surgery Center, Fuwai Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College, Beijing, China
| | - Long Huang
- Shanghai Majorbio Bio-Pharm Technology Co, Shanghai, China
| | - Wanjin Hu
- Shanghai Majorbio Bio-Pharm Technology Co, Shanghai, China
| | - Xu Wang
- PICU, Pediatric Cardiac Center, Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Shoujun Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Pediatric Cardiac Surgery Center, Fuwai Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College, Beijing, China.
| | - Hao Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Pediatric Cardiac Surgery Center, Fuwai Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College, Beijing, China.
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
8
|
A Randomized, Double-Blind, Placebo-Controlled Trial to Evaluate the Effects of Multi-Strain Synbiotic in Patients with Functional Diarrhea and High Fecal Calprotectin Levels: A Pilot Study. Nutrients 2022; 14:nu14235017. [PMID: 36501047 PMCID: PMC9735760 DOI: 10.3390/nu14235017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Synbiotics, including probiotics and prebiotics, are useful for patients with functional bowel disorders. However, which synbiotics are beneficial for patients with which diseases, especially those with functional diarrhea (FDr) with high fecal calprotectin levels, is currently unknown. FDr is an extension of irritable bowel syndrome with diarrhea (IBS-D). Although fewer studies have been conducted on FDr compared to IBS-D, its importance is increasing as its prevalence increases. The aim of this study was to evaluate the effects of a synbiotic containing a mixture of Lactobacillus and Bifidobacterium and its substrate, fructooligosaccharide, on bowel symptoms, fecal calprotectin levels, fecal microbiota, and safety in FDr patients with high fecal calprotectin levels. Forty patients were randomly assigned to either a synbiotic group or a placebo group. A total of 20 subjects in the synbiotic group and 19 subjects in the placebo group completed the study (8 weeks). Changes in FDr symptoms, fecal calprotectin levels, and gut microbiota were assessed during the intervention period. At 4 and 8 weeks, the number of bowel movements tended to increase in the synbiotic group, with a significant increase in the number of formed stools rather than loose stools (p < 0.05). Bowel movement satisfaction was significantly increased in the synbiotic group, but not in the placebo group. Intestinal flora analysis revealed that Lactobacillales at the order level was increased only in the synbiotic group at the end of the intervention. In contrast, at week 8 of the intervention, log-transformed fecal calprotectin levels were significantly decreased in the synbiotic group, although the change was not significantly different from that of the placebo group. These findings suggest that the intake of a multi-strain-containing synbiotic for 8 weeks could improve gut symptoms and the intestinal microenvironment of FDr patients with high fecal calprotectin levels.
Collapse
|
9
|
He Y, Zhang Y, Li F, Shi Y. White Matter Injury in Preterm Infants: Pathogenesis and Potential Therapy From the Aspect of the Gut–Brain Axis. Front Neurosci 2022; 16:849372. [PMID: 35573292 PMCID: PMC9099073 DOI: 10.3389/fnins.2022.849372] [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: 01/06/2022] [Accepted: 04/04/2022] [Indexed: 12/12/2022] Open
Abstract
Very preterm infants who survive are at high risk of white matter injury (WMI). With a greater understanding of the pathogenesis of WMI, the gut microbiota has recently drawn increasing attention in this field. This review tries to clarify the possible mechanisms behind the communication of the gut bacteria and the immature brain via the gut–brain axis. The gut microbiota releases signals, such as microbial metabolites. These metabolites regulate inflammatory and immune responses characterized by microglial activation, which ultimately impact the differentiation of pre-myelinating oligodendrocytes (pre-OLs) and lead to WMI. Moreover, probiotics and prebiotics emerge as a promising therapy to improve the neurodevelopmental outcome. However, future studies are required to clarify the function of these above products and the optimal time for their administration within a larger population. Based on the existing evidence, it is still too early to recommend probiotics and prebiotics as effective treatments for WMI.
Collapse
Affiliation(s)
- Yu He
- Department of Neonatology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Yuni Zhang
- Department of Neonatology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Fang Li
- Department of Neonatology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- *Correspondence: Fang Li,
| | - Yuan Shi
- Department of Neonatology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Yuan Shi,
| |
Collapse
|
10
|
Zavistanaviciute P, Zokaityte E, Starkute V, Ruzauskas M, Viskelis P, Bartkiene E. Berry By-Products in Combination with Antimicrobial Lactic Acid Bacteria Strains for the Sustainable Formulation of Chewing Candies. Foods 2022; 11:foods11091177. [PMID: 35563900 PMCID: PMC9102268 DOI: 10.3390/foods11091177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/15/2022] [Accepted: 04/16/2022] [Indexed: 11/16/2022] Open
Abstract
The purpose of this research was to develop formulations of chewing candies (CCs) in a sustainable manner by using berry by-products in combination with antimicrobial lactic acid bacteria (LAB) strains. To implement this aim, the optimal quantities of by-products from lyophilised raspberry (Rasp) and blackcurrant (Bcur) from the juice production industry were selected. Prior to use, Lactiplantibacillus plantarum LUHS135, Liquorilactobacillusuvarum LUHS245, Lacticaseibacillusparacasei LUHS244, and Pediococcus acidilactici LUHS29 strains were multiplied in a dairy industry by-product-milk permeate (MP). The antimicrobial activity of the selected ingredients (berry by-products and LAB) was evaluated. Two texture-forming agents were tested for the CC formulations: gelatin (Gl) and agar (Ag). In addition, sugar was replaced with xylitol. The most appropriate formulation of the developed CCs according to the product's texture, colour, total phenolic compound (TPC) content, antioxidant activity, viable LAB count during storage, overall acceptability (OA), and emotions (EMs) induced in consumers was selected. It was established that the tested LAB inhibited three pathogens out of the 11 tested, while the blackcurrant by-products inhibited all 11 tested pathogens. The highest OA was shown for the CC prepared with gelatin in addition to 5 g of Rasp and 5 g of Bcur by-products. The Rasp and LUHS135 formulation showed the highest TPC content (147.16 mg 100 g-1 d.m.), antioxidant activity (88.2%), and LAB count after 24 days of storage (6.79 log10 CFU g-1). Finally, it was concluded that Gl, Rasp and Bcur by-products, and L. plantarum LUHS135 multiplied in MP are promising ingredients for preparing CCs in a sustainable manner; the best CC formula consisted of Gl, Rasp by-products, and LUHS135 and showed the highest OA (score 9.52) and induced the highest intensity of the EM 'happy' (0.231).
Collapse
Affiliation(s)
- Paulina Zavistanaviciute
- Department of Food Safety and Quality, Veterinary Academy, Lithuanian University of Health Sciences, Tilzes Str. 18, LT-47181 Kaunas, Lithuania; (E.Z.); (V.S.); (E.B.)
- Institute of Animal Rearing Technologies, Faculty of Animal Sciences, Lithuanian University of Health Sciences, Tilzes Str. 18, LT-47181 Kaunas, Lithuania
- Correspondence: ; Tel.: +370-655-06461
| | - Egle Zokaityte
- Department of Food Safety and Quality, Veterinary Academy, Lithuanian University of Health Sciences, Tilzes Str. 18, LT-47181 Kaunas, Lithuania; (E.Z.); (V.S.); (E.B.)
- Institute of Animal Rearing Technologies, Faculty of Animal Sciences, Lithuanian University of Health Sciences, Tilzes Str. 18, LT-47181 Kaunas, Lithuania
| | - Vytaute Starkute
- Department of Food Safety and Quality, Veterinary Academy, Lithuanian University of Health Sciences, Tilzes Str. 18, LT-47181 Kaunas, Lithuania; (E.Z.); (V.S.); (E.B.)
- Institute of Animal Rearing Technologies, Faculty of Animal Sciences, Lithuanian University of Health Sciences, Tilzes Str. 18, LT-47181 Kaunas, Lithuania
| | - Modestas Ruzauskas
- Department of Anatomy and Physiology, Faculty of Veterinary, Lithuanian University of Health Sciences, Tilzes Str. 18, LT-47181 Kaunas, Lithuania;
- Institute of Microbiology and Virology, Faculty of Veterinary, Lithuanian University of Health Sciences, Mickeviciaus Str. 9, LT-44307 Kaunas, Lithuania
| | - Pranas Viskelis
- Institute of Horticulture, Lithuanian Research Centre for Agriculture and Forestry, Kauno Str. 30, LT-54333 Babtai, Lithuania;
| | - Elena Bartkiene
- Department of Food Safety and Quality, Veterinary Academy, Lithuanian University of Health Sciences, Tilzes Str. 18, LT-47181 Kaunas, Lithuania; (E.Z.); (V.S.); (E.B.)
- Institute of Animal Rearing Technologies, Faculty of Animal Sciences, Lithuanian University of Health Sciences, Tilzes Str. 18, LT-47181 Kaunas, Lithuania
| |
Collapse
|
11
|
Jang KB, Kim SW. Role of milk carbohydrates in intestinal health of nursery pigs: a review. J Anim Sci Biotechnol 2022; 13:6. [PMID: 34983676 PMCID: PMC8729129 DOI: 10.1186/s40104-021-00650-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 11/16/2021] [Indexed: 12/14/2022] Open
Abstract
Intestinal health is essential for the resistance to enteric diseases and for nutrient digestion and absorption to support growth. The intestine of nursery pigs are immature and vulnerable to external challenges, which cause negative impacts on the structure and function of the intestine. Among nutritional interventions, the benefits of milk are significant for the intestinal health of pigs. Milk coproducts have traditionally been used in starter feeds to improve the growth of nursery pigs, but their use is somewhat limited due to the high costs and potential risks of excessive lactose on the intestine. Thus, understanding a proper feeding level of milk carbohydrates is an important start of the feeding strategy. For nursery pigs, lactose is considered a highly digestible energy source compared with plant-based starch, whereas milk oligosaccharides are considered bioactive compounds modulating intestinal immunity and microbiota. Therefore, milk carbohydrates, mainly composed of lactose and oligosaccharides, have essential roles in the intestinal development and functions of nursery pigs. The proper feeding levels of lactose in starter feeds could be variable by weaning age, body weight, or genetic lines. Effects of lactose and milk oligosaccharides have been broadly studied in human health and animal production. Therefore, this review focuses on the mechanisms of lactose and milk oligosaccharides affecting intestinal maturation and functions through modulation of enterocyte proliferation, intestinal immunity, and intestinal microbiota of nursery pigs.
Collapse
Affiliation(s)
- Ki Beom Jang
- Department of Animal Science, North Carolina State University, Raleigh, NC, 27695, USA
| | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, Raleigh, NC, 27695, USA.
| |
Collapse
|
12
|
Liu D, Liu DC, Fan H, Wang Y. Lactobacillus fermentum CQPC08 Attenuates Exercise-Induced Fatigue in Mice Through Its Antioxidant Effects and Effective Intervention of Galactooligosaccharide. Drug Des Devel Ther 2021; 15:5151-5164. [PMID: 34992351 PMCID: PMC8714972 DOI: 10.2147/dddt.s317456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/18/2021] [Indexed: 12/02/2022] Open
Abstract
Aim The purpose of this study is to study the antioxidant effect of Lactobacillus fermentum CQPC08 (CQPC08) on exercise-induced fatigue, and the beneficial intervention of GOS on CQPC08. Methods We use the treadmill to establish a fatigue model caused by exercise, and perform drug treatment after exercise. We tested the exhaustive exercise time of mice; investigated the changes of mice body weight, liver index, histopathology, serum biochemical indicators and mRNA expression levels of oxidative and inflammation-related genes; and assessed the potential fatigue inhibitory effect of CQPC08, and the anti-oxidation effect of the combination of GOS and CQPC08. Results The results suggest that CQPC08 and combination with GOS reduces fatigue-induced oxidative damage of the liver, and it decreases blood urea nitrogen (BUN), lactic acid (LA), glutamic-oxaloacetic transaminase (GOT), glutamic-pyruvic transaminase (GPT), malonaldehyde (MDA), inducible nitric oxide synthase (iNOS), tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6 in serum. Higher levels of serum catalase (CAT), glutathione peroxidase (GSH-Px) and superoxide dismutase (SOD) were found. Treatment with the CQPC08 and combination with GOS correlates with lower relative mRNA expression levels of neuronal NOS (nNOS), iNOS, and TNF-α, and with higher mRNA expression levels of catalase and copper/zinc (Cu/Zn) and manganese (Mn) SOD enzymes in the liver and muscles. Conclusion These results suggest that CQPC08 can resolve exercise-induced fatigue by improving antioxidant ability in mice, and the combination of GOS and CQPC08 enhances this ability of CQPC08.
Collapse
Affiliation(s)
- Dong Liu
- Development Chongqing University of Education, Chongqing, People’s Republic of China
- Education Major in Physical Education, University of Perpetual Help System DALTA Las Pinas, Manila, Philippines
| | - Da Chuan Liu
- Student Affairs Department, Jiangmen Preschool Education College, Jiangmen, Guangdong, People’s Republic of China
| | - Hao Fan
- School of Tourism and Service Management, Chongqing University of Education, Chongqing, People’s Republic of China
- Cultural Industries and Cultural Policy, Yuan Ze University, Taoyuan, Taiwan
| | - Yu Wang
- Orthopedics Department, General Hospital of Northern Theatre Command, Liaoning Province, People’s Republic of China
- Correspondence: Yu Wang Orthopedics Department, General Hospital of Northern Theatre Command, No. 83 Wenhua Road, Shenhe District, Shenyang, Liaoning Province, 110016, People’s Republic of ChinaTel +86-18609886338 Email
| |
Collapse
|
13
|
Zheng SY, Li HX, Xu RC, Miao WT, Dai MY, Ding ST, Liu HD. Potential roles of gut microbiota and microbial metabolites in Parkinson's disease. Ageing Res Rev 2021; 69:101347. [PMID: 33905953 DOI: 10.1016/j.arr.2021.101347] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 04/06/2021] [Accepted: 04/22/2021] [Indexed: 02/07/2023]
Abstract
Parkinson's disease (PD) is a complicated neurodegenerative disease attributed to multifactorial changes. However, its pathological mechanism remains undetermined. Accumulating evidence has revealed the emerging functions of gut microbiota and microbial metabolites, which can affect both the enteric nervous system and the central nervous system via the microbiota-gut-brain axis. Accordingly, intestinal dysbiosis might be closely associated with PD. This review explores alterations to gut microbiota, correlations with clinical manifestations of PD, and briefly probes the underlying mechanisms. Next, the highly controversial roles of microbial metabolites including short-chain fatty acids (SCFAs), H2 and H2S are discussed. Finally, the pros and cons of the current treatments for PD, including those targeting microbiota, are assessed. Advancements in research techniques, further studies on levels of specific strains and longitudinal prospective clinical trials are urgently needed for the identification of early diagnostic markers and the development of novel therapeutic approaches for PD.
Collapse
|
14
|
Cunningham M, Vinderola G, Charalampopoulos D, Lebeer S, Sanders ME, Grimaldi R. Applying probiotics and prebiotics in new delivery formats – is the clinical evidence transferable? Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.04.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
15
|
Fleming SA, Hauser J, Yan J, Donovan SM, Wang M, Dilger RN. A Mediation Analysis to Identify Links between Gut Bacteria and Memory in Context of Human Milk Oligosaccharides. Microorganisms 2021; 9:846. [PMID: 33920826 PMCID: PMC8071191 DOI: 10.3390/microorganisms9040846] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/12/2021] [Accepted: 04/14/2021] [Indexed: 12/23/2022] Open
Abstract
Elucidating relationships between the gut and brain is of intense research focus. Multiple studies have demonstrated that modulation of the intestinal environment via prebiotics or probiotics can induce cognitively beneficial effects, such as improved memory or reduced anxiety. However, the mechanisms by which either act remain largely unknown. We previously demonstrated that different types of oligosaccharides affected short- and long-term memory in distinct ways. Given that the oligosaccharide content of human milk is highly variable, and that formula-fed infants typically do not consume similar amounts or types of oligosaccharides, their potential effects on brain development warrant investigation. Herein, a mediation analysis was performed on existing datasets, including relative abundance of bacterial genera, gene expression, brain volume, and cognition in young pigs. Analyses revealed that numerous bacterial genera in both the colon and feces were related to short- and/or long-term memory. Relationships between genera and memory appeared to differ between diets. Mediating variables frequently included GABAergic and glutamatergic hippocampal gene expression. Other mediating variables included genes related to myelination, transcription factors, brain volume, and exploratory behavior. Overall, this analysis identified multiple pathways between the gut and brain, with a focus on genes related to excitatory/inhibitory neurotransmission.
Collapse
Affiliation(s)
| | - Jonas Hauser
- Société des Produits Nestlé SA, 1000 Lausanne, Switzerland;
| | - Jian Yan
- Nestlé Product Technology Center Nutrition, CH-1800 Vevey, Switzerland;
| | - Sharon M. Donovan
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL 61801, USA; (S.M.D.); (M.W.)
- Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA
| | - Mei Wang
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL 61801, USA; (S.M.D.); (M.W.)
| | - Ryan N. Dilger
- Traverse Science, Inc., Champaign, IL 61820, USA;
- Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA
- Piglet Nutrition and Cognition Laboratory, Department of Animal Sciences, University of Illinois, Urbana, IL 61801, USA
- Neuroscience Program, University of Illinois, Urbana, IL 61801, USA
| |
Collapse
|
16
|
Castro-Alves VC, Nascimento JROD. Size matters: TLR4-mediated effects of α-(1,5)-linear arabino-oligosaccharides in macrophage-like cells depend on their degree of polymerization. Food Res Int 2021; 141:110093. [PMID: 33641969 DOI: 10.1016/j.foodres.2020.110093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 12/09/2020] [Accepted: 12/27/2020] [Indexed: 02/08/2023]
Abstract
Linear arabino-oligosaccharides (LAOS) produced from controlled enzymatic hydrolysis of arabinans from sugar beet are well-known because of their chain-length dependent prebiotic effects. However, it is not clear if these α-(1,5)-linked arabinose oligosaccharides can interact directly with immune system cells, as well as if its degree of polymerization (DP) influences possible biological effects. Four high purity LAOS with distinct DP were tested in macrophage-like cells exposed or not to LPS. Results shown that LAOS interact with Toll-like receptor (TLR) 4 in a chain length-dependent manner. LAOS with higher DP induce stimulatory effects mainly through the TLR4/MyD88 pathway, thereby enhancing the release of tumor necrosis factor alpha (TNF-α), interleukin (IL-) 1β, 6, 12, and chemokines including MCP-1, RANTES, IL-8, and IP-10. Notably, LAOS with lower DP appears to have an opposite effect to those counterparts with higher DP, as they does not induce the secretion of cytokines and chemokines in macrophages-like cells, while also inhibit TLR4-mediated effects induced by both lipopolysaccharide and LAOS with higher DP. These findings provide not only insights into potential biological effects of LAOS, but also reveal that controlled enzymatic hydrolysis of sugar beet arabinans may lead to dietary oligosaccharides with desired biological properties.
Collapse
Affiliation(s)
- Victor Costa Castro-Alves
- Department of Food Science and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil; Food Research Center (FoRC), Research, Innovation and Dissemination Centers, São Paulo Research Foundation (CEPID-FAPESP), São Paulo, Brazil.
| | - João Roberto Oliveira do Nascimento
- Department of Food Science and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil; Food Research Center (FoRC), Research, Innovation and Dissemination Centers, São Paulo Research Foundation (CEPID-FAPESP), São Paulo, Brazil; Food and Nutrition Research Center (NAPAN), University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
17
|
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
|
18
|
Zhang N, Li C, Niu Z, Kang H, Wang M, Zhang B, Tian H. Colonization and immunoregulation of Lactobacillus plantarum BF_15, a novel probiotic strain from the feces of breast-fed infants. Food Funct 2021; 11:3156-3166. [PMID: 32207765 DOI: 10.1039/c9fo02745a] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Immunosuppression is a manifestation imbalance in the immune system, often during unhealthy states. In recent years, lactic acid bacteria (LAB) have been found to be important components of the body's innate immune system, and indispensable to maintaining normal immune function. Lactobacillus plantarum BF_15, a novel strain isolated from the feces of breast-fed infants, which has shown potential as an immunomodulator in vitro. In the present study, with the Polymerase Chain Reaction-Denaturing Gradient Gel Electrophoresis (PCR-DGGE) based on RNA-polymerase beta subunit encoding gene (rpoB) to analyze the colonization of L. plantarum BF_15 in the intestine of mice. In addition, Lactobacillus rhamnosus GG (LGG) as a positive control strain, by measuring immune-related indexes and the diversity of intestinal microbiota, the effects of BF_15 on immunoregulation and intestinal microbiota dysbiosis were investigated to elucidate whether the attenuation of immunosuppression is related to the modulation of intestinal microbiota. Results did indeed support this notion that BF_15 did colonize murine intestines well, in which it could still be detected in mice feces 14 days after stopping the probiotic administration. Moreover, BF_15 found to protect mice against reduction in the levels of several immune-related indicators, including the thymus and spleen indexes, splenic lymphocyte proliferation, toe swelling degree, serum hemolysin-antibody level, and macrophage phagocytosis index, triggered by high-dose (200 mg kg-1) intraperitoneal administration of cyclophosphamide (CTX). In addition, the strain was also found to effectively balance intestinal microbiota dysbiosis in the mice. Collectively, these results indicated that L. plantarum BF_15 can not only successfully colonize murine intestines, but also can effectively alleviate CTX-induced immunosuppression, once established, by rebalancing the intestinal microbiota. This, therefore, provides strong evidence for the view that BF_15 has the potential to become a highly effective immunomodulating probiotic in human microbiota as well.
Collapse
Affiliation(s)
- Na Zhang
- College of Food Science and Technology, Agricultural University of Hebei, Baoding, Hebei 071000, China. and College of Biochemistry and Environmental Engineering, Baoding University, Baoding, Hebei 071000, China
| | - Chen Li
- College of Food Science and Technology, Agricultural University of Hebei, Baoding, Hebei 071000, China.
| | - Zhihua Niu
- College of Food Science and Technology, Agricultural University of Hebei, Baoding, Hebei 071000, China.
| | - Hongyan Kang
- New Hope Tensun (Hebei) Dairy Co., Ltd, Baoding, Hebei 071000, China
| | - Miaoshu Wang
- New Hope Tensun (Hebei) Dairy Co., Ltd, Baoding, Hebei 071000, China
| | - Bo Zhang
- College of Food Science and Technology, Agricultural University of Hebei, Baoding, Hebei 071000, China.
| | - Hongtao Tian
- College of Food Science and Technology, Agricultural University of Hebei, Baoding, Hebei 071000, China. and National Engineering Research Center for Agriculture in Northern Mountainous Areas, Baoding, Hebei 071000, China
| |
Collapse
|
19
|
Yan X, Yan J, Xiang Q, Wang F, Dai H, Huang K, Fang L, Yao H, Wang L, Zhang W. Fructooligosaccharides protect against OVA-induced food allergy in mice by regulating the Th17/Treg cell balance using tryptophan metabolites. Food Funct 2021; 12:3191-3205. [PMID: 33735338 DOI: 10.1039/d0fo03371e] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Fructooligosaccharides (FOS) can change gut microbiota composition and play a protective role in food allergy (FA).
Collapse
Affiliation(s)
- Xiumei Yan
- Department of Pediatrics
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University
- Wenzhou 325000
- China
| | - Jingbin Yan
- Department of Ultrasonography
- Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine
- Wenzhou 325000
- China
| | - Qiangwei Xiang
- Department of Pediatrics
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University
- Wenzhou 325000
- China
| | - Fanyan Wang
- Department of Pathophysiology
- School of Basic Medical Sciences
- Wenzhou Medical University
- Wenzhou 325000
- China
| | - Huan Dai
- Department of Pediatrics
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University
- Wenzhou 325000
- China
| | - Kaiyu Huang
- Department of Pediatrics
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University
- Wenzhou 325000
- China
| | - Lingjuan Fang
- Department of Pediatrics
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University
- Wenzhou 325000
- China
| | - Hao Yao
- Department of Pediatrics
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University
- Wenzhou 325000
- China
| | - Lingya Wang
- Department of Pediatrics
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University
- Wenzhou 325000
- China
| | - Weixi Zhang
- Department of Pediatrics
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University
- Wenzhou 325000
- China
| |
Collapse
|
20
|
Lockyer S. Effects of diets, foods and nutrients on immunity: Implications for COVID‐19? NUTR BULL 2020. [DOI: 10.1111/nbu.12470] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
21
|
Laigaard A, Krych L, Zachariassen LF, Ellegaard-Jensen L, Nielsen DS, Hansen AK, Hansen CHF. Dietary prebiotics promote intestinal Prevotella in association with a low-responding phenotype in a murine oxazolone-induced model of atopic dermatitis. Sci Rep 2020; 10:21204. [PMID: 33273678 PMCID: PMC7713185 DOI: 10.1038/s41598-020-78404-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 11/24/2020] [Indexed: 01/08/2023] Open
Abstract
Atopic dermatitis is a chronic eczema commonly observed among children in Western countries. The gut microbiota is a significant factor in the pathogenesis, and ways to promote intestinal colonizers with anti-inflammatory capabilities are therefore favorable. The present study addressed the effects of a prebiotic, xylooligosaccharide (XOS), on the gut microbiota and ear inflammation in an oxazolone-induced dermatitis model in BALB/c mice. Mice were fed a XOS supplemented or a control diet throughout the experiment. Ear thickness and clinical skin inflammation were scored blindly after three weeks topical challenge with 0.4% oxazolone. The mice were divided into high and low responders to oxazolone-induced dermatitis based on clinical inflammation and histological evaluation of ear biopsies, and significantly fewer high responders were present in the XOS fed group. In addition, XOS fed mice had higher abundance of Prevotella spp. in their gut microbiota compared to the control fed mice. Serum IgE and ear tissue cytokine levels correlated significantly with the clinical scores, and with the abundance of Prevotella spp. The strong association between the low-responding phenotype and high abundance of Prevotella spp., indicates an alleviating effect of this intestinal colonizer in allergic sensitization. Prevotella should be considered as a relevant target for future microbiota-directed treatment strategies in atopic patients.
Collapse
Affiliation(s)
- Ann Laigaard
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Lukasz Krych
- Department of Food Science, Faculty of Science, University of Copenhagen, Frederiksberg C, Denmark
| | - Line F Zachariassen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | | | - Dennis S Nielsen
- Department of Food Science, Faculty of Science, University of Copenhagen, Frederiksberg C, Denmark
| | - Axel K Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Camilla H F Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark. .,Section of Experimental Animal Models, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 9, 1870, Frederiksberg C, Denmark.
| |
Collapse
|
22
|
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
|
23
|
Kerperien J, Veening-Griffioen D, Oja A, Wehkamp T, Jeurink PV, Garssen J, Knippels LMJ, Willemsen LEM. Dietary Vitamin D Supplementation Is Ineffective in Preventing Murine Cow's Milk Allergy, Irrespective of the Presence of Nondigestible Oligosaccharides. Int Arch Allergy Immunol 2020; 181:908-918. [PMID: 32814335 DOI: 10.1159/000509750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 06/09/2020] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION Cow's milk allergy (CMA) is one of the most common food allergies especially early in life. A mixture of nondigestible short-chain galacto-oligosaccharides, long-chain fructo-oligosaccharides, and pectin-derived acidic-oligosaccharides (GFA) may reduce allergy development and allergic symptoms in murine CMA. Recently, vitamin D (VitD) has been suggested to have beneficial effects in reducing allergy as well. OBJECTIVE In this study, the immune modulatory effect on allergy prevention using the combination of GFA and VitD was investigated. METHODS Female C3H/HeOuJ mice were fed a control or GFA-containing diet with depleted, standard (1,000 IU/kg), or supplemented (5,000 IU/kg) VitD content for 2 weeks before and during whey sensitization (n = 10-15). Mice were sensitized 5 times intragastrically with PBS as a control, whey as cow's milk allergen, and/or cholera toxin as adjuvant on a weekly interval. One week after the last sensitization, mice were intradermally challenged in both ear pinnae and orally with whey, subsequently the acute allergic skin response and shock symptoms were measured. After 18 h, terminal blood samples, mesenteric lymph nodes, and spleens were collected. Whey-specific immunoglobulin (Ig) E and IgG1 levels were measured by means of ELISA. T cell subsets and dendritic cells (DCs) were studied using flow cytometry. RESULTS Additional VitD supplementation did not lower the allergic symptoms compared to the standard VitD diet. CMA mice fed the GFA diet supplemented with VitD (GFA VitD+) significantly decreased the acute allergic skin response of whey sensitized mice when compared to the CMA mice fed VitD (VitD+) group (p < 0.05). The effect of GFA was not improved by extra VitD supplementation even though the CMA mice fed the GFA VitD+ diet had a significantly increased percentage of CD103+ DCs compared to the VitD+ group (p < 0.05). The VitD-deprived mice showed a high percentage of severe shock and many reached the humane endpoint; therefore, these groups were not further analyzed. CONCLUSIONS High-dose VitD supplementation in mice does not protect against CMA development in the presence or absence of GFA.
Collapse
Affiliation(s)
- JoAnn Kerperien
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Désirée Veening-Griffioen
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands.,Nutricia Research B.V, Utrecht, The Netherlands
| | - Anna Oja
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | | | - Prescilla V Jeurink
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands.,Nutricia Research B.V, Utrecht, The Netherlands
| | - Johan Garssen
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands.,Nutricia Research B.V, Utrecht, The Netherlands
| | - Leon M J Knippels
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands.,Nutricia Research B.V, Utrecht, The Netherlands
| | - Linette E M Willemsen
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands,
| |
Collapse
|
24
|
Akkerman R, Logtenberg MJ, An R, Van Den Berg MA, de Haan BJ, Faas MM, Zoetendal E, de Vos P, Schols HA. Endo-1,3(4)-β-Glucanase-Treatment of Oat β-Glucan Enhances Fermentability by Infant Fecal Microbiota, Stimulates Dectin-1 Activation and Attenuates Inflammatory Responses in Immature Dendritic Cells. Nutrients 2020; 12:nu12061660. [PMID: 32503178 PMCID: PMC7352905 DOI: 10.3390/nu12061660] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 05/29/2020] [Accepted: 06/01/2020] [Indexed: 12/12/2022] Open
Abstract
Background: Non-digestible carbohydrates are added to infant formula to mimic the effects of human milk oligosaccharide by acting as prebiotics and stimulating the immune system. Although not yet used in infant formulas, β-glucans are known to have beneficial health effects, and are therefore of potential interest for supplementation. Methods and results: We investigated the in vitro fermentation of native and endo-1,3(4)-β-glucanase-treated oat β-glucan using pooled fecal inocula of 2- and 8-week-old infants. While native oat β-glucan was not utilized, both inocula specifically utilized oat β-glucan oligomers containing β(1→4)-linkages formed upon enzyme treatment. The fermentation rate was highest in the fecal microbiota of 2-week-old infants, and correlated with a high lactate production. Fermentation of media supplemented with native and enzyme-treated oat β-glucans increased the relative abundance of Enterococcus and attenuated pro-inflammatory cytokine production (IL-1β, IL-6, TNFα) in immature dendritic cells. This attenuating effect was more pronounced after enzyme treatment. This attenuation might result from the enhanced ability of fermented oat β-glucan to stimulate Dectin-1 receptors. Conclusion: Our findings demonstrate that endo-1,3(4)-β-glucanase treatment enhances the fermentability of oat β-glucan and attenuates pro-inflammatory responses. Hence, this study shows that especially enzyme-treated oat β-glucans have a high potential for supplementation of infant formula.
Collapse
Affiliation(s)
- Renate Akkerman
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Centre Groningen, Groningen, Hanzeplein 1, 9700 RB Groningen, The Netherlands; (B.J.d.H.); (M.M.F.); (P.d.V.)
- Correspondence: (R.A.); (M.J.L.)
| | - Madelon J. Logtenberg
- Laboratory of Food Chemistry, Wageningen University & Research, Bornse Weilanden 9, 6708 WG Wageningen, The Netherlands;
- Correspondence: (R.A.); (M.J.L.)
| | - Ran An
- Laboratory of Microbiology, Wageningen University & Research, Stippeneng 4, 6708 WE Wageningen, The Netherlands; (R.A.); (E.Z.)
| | | | - Bart J. de Haan
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Centre Groningen, Groningen, Hanzeplein 1, 9700 RB Groningen, The Netherlands; (B.J.d.H.); (M.M.F.); (P.d.V.)
| | - Marijke M. Faas
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Centre Groningen, Groningen, Hanzeplein 1, 9700 RB Groningen, The Netherlands; (B.J.d.H.); (M.M.F.); (P.d.V.)
| | - Erwin Zoetendal
- Laboratory of Microbiology, Wageningen University & Research, Stippeneng 4, 6708 WE Wageningen, The Netherlands; (R.A.); (E.Z.)
| | - Paul de Vos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Centre Groningen, Groningen, Hanzeplein 1, 9700 RB Groningen, The Netherlands; (B.J.d.H.); (M.M.F.); (P.d.V.)
| | - Henk A. Schols
- Laboratory of Food Chemistry, Wageningen University & Research, Bornse Weilanden 9, 6708 WG Wageningen, The Netherlands;
| |
Collapse
|
25
|
Myhill LJ, Stolzenbach S, Mejer H, Jakobsen SR, Hansen TVA, Andersen D, Brix S, Hansen LH, Krych L, Nielsen DS, Nejsum P, Thamsborg SM, Williams AR. Fermentable Dietary Fiber Promotes Helminth Infection and Exacerbates Host Inflammatory Responses. THE JOURNAL OF IMMUNOLOGY 2020; 204:3042-3055. [PMID: 32284331 DOI: 10.4049/jimmunol.1901149] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 03/24/2020] [Indexed: 01/01/2023]
Abstract
Fermentable dietary fibers promote the growth of beneficial bacteria, can enhance mucosal barrier integrity, and reduce chronic inflammation. However, effects on intestinal type 2 immune function remain unclear. In this study, we used the murine whipworm Trichuris muris to investigate the effect of the fermentable fiber inulin on host responses to infection regimes that promote distinct Th1 and Th2 responses in C57BL/6 mice. In uninfected mice, dietary inulin stimulated the growth of beneficial bacteria, such as Bifidobacterium (Actinobacteria) and Akkermansia (Verrucomicrobia). Despite this, inulin prevented worm expulsion in normally resistant mice, instead resulting in chronic infection, whereas mice fed an equivalent amount of nonfermentable fiber (cellulose) expelled worms normally. Lack of expulsion in the mice fed inulin was accompanied by a significantly Th1-skewed immune profile characterized by increased T-bet+ T cells and IFN-γ production in mesenteric lymph nodes, increased expression of Ido1 in the cecum, and a complete absence of mast cell and IgE production. Furthermore, the combination of dietary inulin and high-dose T. muris infection caused marked dysbiosis, with expansion of the Firmicutes and Proteobacteria phyla, near elimination of Bacteroidetes, and marked reductions in cecal short-chain fatty acids. Neutralization of IFN-γ during infection abrogated Ido1 expression and was sufficient to restore IgE production and worm expulsion in inulin-fed mice. Our results indicate that, whereas inulin promoted gut health in otherwise healthy mice, during T. muris infection, it exacerbated inflammatory responses and dysbiosis. Thus, the positive effects of fermentable fiber on gut inflammation appear to be context dependent, revealing a novel interaction between diet and infection.
Collapse
Affiliation(s)
- Laura J Myhill
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C DK-1870, Denmark;
| | - Sophie Stolzenbach
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C DK-1870, Denmark
| | - Helena Mejer
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C DK-1870, Denmark
| | - Simon R Jakobsen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C DK-1870, Denmark
| | - Tina V A Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C DK-1870, Denmark
| | - Daniel Andersen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby DK-2800, Denmark
| | - Susanne Brix
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby DK-2800, Denmark
| | - Lars H Hansen
- Department of Plant and Environmental Sciences, Faculty of Science, University of Copenhagen, Frederiksberg C DK-1871, Denmark
| | - Lukasz Krych
- Department of Food Sciences, Faculty of Science, University of Copenhagen, Frederiksberg C DK-1958, Denmark; and
| | - Dennis S Nielsen
- Department of Food Sciences, Faculty of Science, University of Copenhagen, Frederiksberg C DK-1958, Denmark; and
| | - Peter Nejsum
- Department of Clinical Medicine, Aarhus University, Aarhus DK-8200, Denmark
| | - Stig M Thamsborg
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C DK-1870, Denmark
| | - Andrew R Williams
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C DK-1870, Denmark;
| |
Collapse
|
26
|
Beyond Heat Stress: Intestinal Integrity Disruption and Mechanism-Based Intervention Strategies. Nutrients 2020; 12:nu12030734. [PMID: 32168808 PMCID: PMC7146479 DOI: 10.3390/nu12030734] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 12/11/2022] Open
Abstract
The current climate changes have increased the prevalence and intensity of heat stress (HS) conditions. One of the initial consequences of HS is the impairment of the intestinal epithelial barrier integrity due to hyperthermia and hypoxia following blood repartition, which often results in a leaky gut followed by penetration and transfer of luminal antigens, endotoxins, and pathogenic bacteria. Under extreme conditions, HS may culminate in the onset of “heat stroke”, a potential lethal condition if remaining untreated. HS-induced alterations of the gastrointestinal epithelium, which is associated with a leaky gut, are due to cellular oxidative stress, disruption of intestinal integrity, and increased production of pro-inflammatory cytokines. This review summarizes the possible resilience mechanisms based on in vitro and in vivo data and the potential interventions with a group of nutritional supplements, which may increase the resilience to HS-induced intestinal integrity disruption and maintain intestinal homeostasis.
Collapse
|
27
|
Xing YY, Li KN, Xu YQ, Wu YZ, Shi LL, Guo SW, Yan SM, Jin X, Shi BL. Effects of galacto-oligosaccharide on growth performance, feacal microbiota, immune response and antioxidant capability in weaned piglets. JOURNAL OF APPLIED ANIMAL RESEARCH 2020. [DOI: 10.1080/09712119.2020.1732394] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Yuan-yuan Xing
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, People’s Republic of China
| | - Ke-nan Li
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, People’s Republic of China
| | - Yuan-qing Xu
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, People’s Republic of China
| | - Ying-zhao Wu
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, People’s Republic of China
| | - Lu-lu Shi
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, People’s Republic of China
| | - Shi-wei Guo
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, People’s Republic of China
| | - Su-mei Yan
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, People’s Republic of China
| | - Xiao Jin
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, People’s Republic of China
| | - Bin-lin Shi
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, People’s Republic of China
| |
Collapse
|
28
|
Janbazacyabar H, van Bergenhenegouwen J, Verheijden KA, Leusink-Muis T, van Helvoort A, Garssen J, Folkerts G, Braber S. Non-digestible oligosaccharides partially prevent the development of LPS-induced lung emphysema in mice. PHARMANUTRITION 2019. [DOI: 10.1016/j.phanu.2019.100163] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
29
|
Mine Y, Jin Y, Zhang H, Rupa P, Majumder K, Sakurai T, Taniguchi Y, Takagaki R, Watanabe H, Mitsuzumi H. Prophylactic effects of isomaltodextrin in a Balb/c mouse model of egg allergy. NPJ Sci Food 2019; 3:23. [PMID: 31728412 PMCID: PMC6834578 DOI: 10.1038/s41538-019-0057-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 08/23/2019] [Indexed: 12/21/2022] Open
Abstract
The aim of this study was to evaluate the potential effects of isomaltodextrin (IMD), a dietary saccharide polymer derived from enzymatically produced from starch, on the ability to alter immune response (IR) bias to hen egg ovalbumin (Ova) induced allergic inflammation in mice. Groups of Balb/c mice were pre-treated with various doses of IMD in drinking water (1.0, 2.5, and 5.0% w/v) for 6 weeks and subsequently sensitized to the Ova together with continuous administration of IMD. To evaluate changes in immune response bias, immunoglobulin isotype-associated antibody activity, concentrations of type 1 and 2 cytokines and the percentage of T-regulatory cells (T-regs) in blood were measured. Clinical signs of allergy were assessed after oral challenge with Ova. Treatment with IMD did not significantly alter the frequency of clinical signs, however there was a trend in the overall reduction of clinical signs. Effect on IR bias was observed in the treatment groups as reflected by reduction in a type 1-biased phenotype as evident by decrease in isotype-specific IgE, IgG and increase in IL-12 cytokine production and a high proportion of T-regs. This study revealed that IMD could be a useful prophylactic candidate for alteration of allergic IR bias in mice and an immune-stimulator for reducing egg induced allergic reactions.
Collapse
Affiliation(s)
- Yoshinori Mine
- Department of Food Science, University of Guelph, Guelph, ON N1G2W1 Canada
| | - Yan Jin
- Department of Food Science, University of Guelph, Guelph, ON N1G2W1 Canada
- Present Address: College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, 300457 Tianjin, People’s Republic of China
| | - Hua Zhang
- Department of Food Science, University of Guelph, Guelph, ON N1G2W1 Canada
| | - Prithy Rupa
- Department of Food Science, University of Guelph, Guelph, ON N1G2W1 Canada
| | - Kaustav Majumder
- Department of Food Science, University of Guelph, Guelph, ON N1G2W1 Canada
- Present Address: Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE 68588-6205 USA
| | - Takeo Sakurai
- R&D Center, Hayashibara CO., LTD., 675-1 Fujisaki, Naka-ku, Okayama 702-8006 Japan
| | - Yoshifumi Taniguchi
- R&D Center, Hayashibara CO., LTD., 675-1 Fujisaki, Naka-ku, Okayama 702-8006 Japan
| | - Ryodai Takagaki
- R&D Center, Hayashibara CO., LTD., 675-1 Fujisaki, Naka-ku, Okayama 702-8006 Japan
| | - Hikaru Watanabe
- R&D Center, Hayashibara CO., LTD., 675-1 Fujisaki, Naka-ku, Okayama 702-8006 Japan
| | - Hitoshi Mitsuzumi
- R&D Center, Hayashibara CO., LTD., 675-1 Fujisaki, Naka-ku, Okayama 702-8006 Japan
| |
Collapse
|
30
|
Wu C, Pan L, Luo Y, Niu W, Fang X, Liang W, Li J, Li H, Pan X, Yang G, Chen W, Zhang H, Lakey JRT, Agerberth B, Vos P, Sun J. Low Methoxyl Pectin Protects against Autoimmune Diabetes and Associated Caecal Dysfunction. Mol Nutr Food Res 2019; 63:e1900307. [DOI: 10.1002/mnfr.201900307] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 07/29/2019] [Indexed: 02/06/2023]
Affiliation(s)
- Chengfei Wu
- State Key Laboratory of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
- School of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
| | - Li‐Long Pan
- School of MedicineJiangnan University Wuxi 214122 P. R. China
| | - Yang Luo
- State Key Laboratory of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
- School of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
| | - Wenying Niu
- State Key Laboratory of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
- School of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
| | - Xin Fang
- State Key Laboratory of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
- School of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
| | - Wenjie Liang
- State Key Laboratory of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
- School of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
| | - Jiahong Li
- State Key Laboratory of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
- School of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
| | - Hongli Li
- State Key Laboratory of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
- School of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
| | - Xiaohua Pan
- State Key Laboratory of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
- School of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
| | - Guilian Yang
- College of Animal Science and TechnologyJilin Agricultural University Changchun 130118 P. R. China
| | - Wei Chen
- State Key Laboratory of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
- School of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
| | - Hao Zhang
- State Key Laboratory of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
- School of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
| | | | - Birgitta Agerberth
- Division of Clinical MicrobiologyDepartment of Laboratory MedicineKarolinska InstituteKarolinska University Hospital 17177 Stockholm Sweden
| | - Paul Vos
- Division of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of GroningenUniversity Medical Center Groningen 9713 GZ Groningen The Netherlands
| | - Jia Sun
- State Key Laboratory of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
- School of Food Science and TechnologyJiangnan University Wuxi 214122 P. R. China
| |
Collapse
|
31
|
Kalampokini S, Becker A, Fassbender K, Lyros E, Unger MM. Nonpharmacological Modulation of Chronic Inflammation in Parkinson's Disease: Role of Diet Interventions. PARKINSON'S DISEASE 2019; 2019:7535472. [PMID: 31534664 PMCID: PMC6732577 DOI: 10.1155/2019/7535472] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 07/22/2019] [Accepted: 08/12/2019] [Indexed: 12/30/2022]
Abstract
Neuroinflammation is increasingly recognized as an important pathophysiological feature of neurodegenerative diseases such as Parkinson's disease (PD). Recent evidence suggests that neuroinflammation in PD might originate in the intestine and the bidirectional communication between the central and enteric nervous system, the so-called "gut-brain axis," has received growing attention due to its contribution to the pathogenesis of neurological disorders. Diet targets mediators of inflammation with various mechanisms and combined with dopaminergic treatment can exert various beneficial effects in PD. Food-based therapies may favorably modulate gut microbiota composition and enhance the intestinal epithelial integrity or decrease the proinflammatory response by direct effects on immune cells. Diets rich in pre- and probiotics, polyunsaturated fatty acids, phenols including flavonoids, and vitamins, such as the Mediterranean diet or a plant-based diet, may attenuate chronic inflammation and positively influence PD symptoms and even progression of the disease. Dietary strategies should be encouraged in the context of a healthy lifestyle with physical activity, which also has neuroimmune-modifying properties. Thus, diet adaptation appears to be an effective additive, nonpharmacological therapeutic strategy that can attenuate the chronic inflammation implicated in PD, potentially slow down degeneration, and thereby modify the course of the disease. PD patients should be highly encouraged to adopt corresponding lifestyle modifications, in order to improve not only PD symptoms, but also general quality of life. Future research should focus on planning larger clinical trials with dietary interventions in PD in order to obtain hard evidence for the hypothesized beneficial effects.
Collapse
Affiliation(s)
- Stefania Kalampokini
- Department of Neurology, University Hospital of Saarland, Kirrberger Straße, 66421 Homburg, Germany
| | - Anouck Becker
- Department of Neurology, University Hospital of Saarland, Kirrberger Straße, 66421 Homburg, Germany
| | - Klaus Fassbender
- Department of Neurology, University Hospital of Saarland, Kirrberger Straße, 66421 Homburg, Germany
| | - Epameinondas Lyros
- Department of Neurology, University Hospital of Saarland, Kirrberger Straße, 66421 Homburg, Germany
| | - Marcus M. Unger
- Department of Neurology, University Hospital of Saarland, Kirrberger Straße, 66421 Homburg, Germany
| |
Collapse
|
32
|
Gazerani P. Probiotics for Parkinson's Disease. Int J Mol Sci 2019; 20:E4121. [PMID: 31450864 PMCID: PMC6747430 DOI: 10.3390/ijms20174121] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 08/14/2019] [Accepted: 08/21/2019] [Indexed: 02/08/2023] Open
Abstract
Parkinson's disease (PD) is a complex neurological disorder classically characterized by impairments in motor system function associated with loss of dopaminergic neurons in the substantia nigra. After almost 200 years since the first description of PD by James Parkinson, unraveling the complexity of PD continues to evolve. It is now recognized that an interplay between genetic and environmental factors influences a diverse range of cellular processes, reflecting on other clinical features including non-motor symptoms. This has consequently highlighted the extensive value of early clinical diagnosis to reduce difficulties of later stage management of PD. Advancement in understanding of PD has made remarkable progress in introducing new tools and strategies such as stem cell therapy and deep brain stimulation. A link between alterations in gut microbiota and PD has also opened a new line. Evidence exists of a bidirectional pathway between the gastrointestinal tract and the central nervous system. Probiotics, prebiotics and synbiotics are being examined that might influence gut-brain axis by altering gut microbiota composition, enteric nervous system, and CNS. This review provides status on use of probiotics for PD. Limitations and future directions will also be addressed to promote further research considering use of probiotics for PD.
Collapse
Affiliation(s)
- Parisa Gazerani
- Biomedicine: Department of Health Science and Technology, Faculty of Medicine, Aalborg University,Frederik Bajers Vej 3B, 9220 Aalborg East, Denmark.
| |
Collapse
|
33
|
Gao X, Lu Y, Wei M, Yang M, Zheng C, Wang C, Zhang Y, Huang L, Wang Z. Matrix-Assisted Laser Desorption/Ionization Time-of-Flight Mass Spectrometry Analysis of Human Milk Neutral and Sialylated Free Oligosaccharides Using Girard's Reagent P On-Target Derivatization. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:8958-8966. [PMID: 31334644 DOI: 10.1021/acs.jafc.9b02635] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The functional role of human milk oligosaccharides (HMOs) is closely associated with their type, composition, and structure. However, a detailed analysis of HMOs is difficult because neutral oligosaccharides (NHMOs) are mixed with sialylated oligosaccharides (SHMOs) in milk. Here, NHMOs were separated from SHMOs by DEAE-52 anion chromatography, and lactose was removed by graphite carbon solid-phase extraction. Lactose-free NHMOs were analyzed by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS) based on Girard's reagent P on-target derivatization (GPOD), and SHMOs were analyzed by MALDI-TOF-MS following selective sialic acid derivatization and GPOD. Sixty-four oligosaccharides were detected: 36 NHMOs, of which 28 were fucosylated, and 28 SHMOs, of which 8 with α-2,3-linked monosialic acid, 2 with α-2,3-linked disialic acid, 10 with α-2,6-linked monosialic acid, 2 with α-2,6-linked disialic acid, and 5 with both α-2,3- and α-2,6-linked disialic acid. These findings provide the groundwork for further characterization of the structure and activity of HMOs.
Collapse
Affiliation(s)
| | | | | | | | - CaiXia Zheng
- The Second Affiliated Hospital of Xi'an Jiaotong University , Xi'an 710069 , China
| | | | | | | | | |
Collapse
|
34
|
Wu C, Pan LL, Niu W, Fang X, Liang W, Li J, Li H, Pan X, Chen W, Zhang H, Lakey JRT, Agerberth B, de Vos P, Sun J. Modulation of Gut Microbiota by Low Methoxyl Pectin Attenuates Type 1 Diabetes in Non-obese Diabetic Mice. Front Immunol 2019; 10:1733. [PMID: 31417546 PMCID: PMC6682655 DOI: 10.3389/fimmu.2019.01733] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 07/09/2019] [Indexed: 01/09/2023] Open
Abstract
Intestinal homeostasis underpins the development of type 1 diabetes (T1D), and dietary manipulations to enhance intestinal homeostasis have been proposed to prevent T1D. The current study aimed to investigate the efficacy of supplementing a novel specific low-methoxyl pectin (LMP) dietary fiber in preventing T1D development. Female NOD mice were weaned onto control or 5% (wt/wt) LMP supplemented diets for up to 40 weeks of age, overt diabetes incidence and blood glucose were monitored. Then broad-spectrum antibiotics (ABX) treatment per os for 7 days followed by gut microbiota transfer was performed to demonstrate gut microbiota-dependent effects. Next-generation sequencing was used for analyzing the composition of microbiota in caecum. Concentration of short chain fatty acids were determined by GC-MS. The barrier reinforcing tight junction proteins zonula occludens-2 (ZO-2), claudin-1 and NOD like receptor protein 3 (NLRP3) inflammasome activation were determined by Western blot. The proportion of CD25+Foxp3+CD4+ regulatory T cell (Foxp3+ Treg) in the pancreas, pancreatic and mesenteric lymph nodes was analyzed by flow cytometry. We found that LMP supplementation ameliorated T1D development in non-obese diabetic (NOD) mice, as evidenced by decreasing diabetes incidence and fasting glucose levels in LMP fed NOD mice. Further microbiota analysis revealed that LMP supplementation prevented T1D-associated caecal dysbiosis and selectively enriched caecal bacterial species to produce more SCFAs. The LMP-mediated microbial balance further enhanced caecal barrier function and shaped gut-pancreatic immune environment, as characterized by higher expression of tight junction proteins claudin-1, ZO-2 in caecum, increased Foxp3+ Treg population and decreased NLRP3 inflammasome activation in both caecum and pancreas. The microbiota-dependent beneficial effect of LMP on T1D was further proven by the fact that aberration of caecal microbiota by ABX treatment worsened T1D autoimmunity and could be restored with transfer of feces of LMP-fed NOD mice. These data demonstrate that this novel LMP limits T1D development by inducing caecal homeostasis to shape pancreatic immune environment. This finding opens a realistic option for gut microbiota manipulation and prevention of T1D in humans.
Collapse
Affiliation(s)
- Chengfei Wu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Li-Long Pan
- School of Medicine, Jiangnan University, Wuxi, China
| | - Wenying Niu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Xin Fang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wenjie Liang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jiahong Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Hongli Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Xiaohua Pan
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jonathan R T Lakey
- Department of Surgery, University of California, Irvine, Orange, CA, United States
| | - Birgitta Agerberth
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Karolinska University Hospital, Huddinge, Sweden
| | - Paul de Vos
- Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Jia Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
35
|
Tavakoli A, Flanagan JL. The Case for a More Holistic Approach to Dry Eye Disease: Is It Time to Move beyond Antibiotics? Antibiotics (Basel) 2019; 8:E88. [PMID: 31262073 PMCID: PMC6783892 DOI: 10.3390/antibiotics8030088] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 06/27/2019] [Indexed: 12/28/2022] Open
Abstract
Dry eye disease (DED) is one of the most frequent presentations to optometrists with over 16 million US adults (6.8% of adult population) diagnosed as having this disorder. The majority of associated marketed products offer relief from symptomatology but do not address aetiology. DED harbours many distinguishing features of a chronic inflammatory disorder. The recent explosion in human microbiome research has sparked interest in the ocular microbiome and its role in the preservation and extension of ocular surface health and in the contribution of the gut microbiome to chronic systemic inflammation and associated "Western life-style" diseases. With a significant lack of success for many patients using currently available DED treatments, in this era of the microbiome, we are interested in exploring potential novel therapies that aim to reconstitute healthy bacterial communities both locally and distally (in the gut) as a treatment for DED. Although this direction of investigation is in its infancy, burgeoning interest makes such a review timely. This paper considers a number of studies into the use functional foods and associated products to ameliorate dry eye.
Collapse
Affiliation(s)
- Azadeh Tavakoli
- School of Optometry and Vision Science, University of New South Wales, Sydney, 2052, Australia
| | - Judith Louise Flanagan
- School of Optometry and Vision Science, University of New South Wales, Sydney, 2052, Australia.
- Brien Holden Vision Institute, Sydney, 2052, Australia.
| |
Collapse
|
36
|
Zhang YF, Liu QM, Gao YY, Liu B, Liu H, Cao MJ, Yang XW, Liu GM. Attenuation of allergic responses following treatment with resveratrol in anaphylactic models and IgE-mediated mast cells. Food Funct 2019; 10:2030-2039. [PMID: 30907398 DOI: 10.1039/c9fo00077a] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Resveratrol exists widely in plant species and has a variety of anti-oxidant, anti-inflammatory, and immunomodulatory properties. However, there have been few reports regarding its anti-food allergic activity. In this study, we demonstrated that resveratrol (isolated from Abies georgei) could decrease the release of β-hexosaminidase and histamine in rat basophilic leukemia-2H3 cells. Resveratrol was not only found to suppress the development of diarrhea, up-regulate the rectal temperature of ovalbumin-allergic mice, and decrease the serum level of specific immunoglobulin E, mouse mast cell protease-1 and histamine, but also found to decrease the population of dendritic cells, B cells and mast cells of ovalbumin -allergic mice in the spleen or mesenteric lymph node. Furthermore, resveratrol inhibited the release of β-hexosaminidase and histamine in bone marrow-derived cells and alleviated mast cell-mediated passive cutaneous anaphylaxis reactions. These findings indicated that resveratrol isolated from Abies georgei might have the potential to alleviate food hypersensitivity or allergic disease.
Collapse
Affiliation(s)
- Ya-Fen Zhang
- College of Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Jimei University, 43 Yindou Road, Xiamen, 361021, Fujian, P.R. China.
| | - Qing-Mei Liu
- College of Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Jimei University, 43 Yindou Road, Xiamen, 361021, Fujian, P.R. China.
| | - Yuan-Yuan Gao
- College of Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Jimei University, 43 Yindou Road, Xiamen, 361021, Fujian, P.R. China.
| | - Bo Liu
- College of Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Jimei University, 43 Yindou Road, Xiamen, 361021, Fujian, P.R. China.
| | - Hong Liu
- College of Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Jimei University, 43 Yindou Road, Xiamen, 361021, Fujian, P.R. China.
| | - Min-Jie Cao
- College of Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Jimei University, 43 Yindou Road, Xiamen, 361021, Fujian, P.R. China.
| | - Xian-Wen Yang
- Key Laboratory of Marine Biogenetic Resources, South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Third Institute of Oceanography, State Oceanic Administration, Daxue Road, Xiamen, 361005, Fujian, P.R. China
| | - Guang-Ming Liu
- College of Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Jimei University, 43 Yindou Road, Xiamen, 361021, Fujian, P.R. China.
| |
Collapse
|
37
|
McKeen S, Young W, Mullaney J, Fraser K, McNabb WC, Roy NC. Infant Complementary Feeding of Prebiotics for theMicrobiome and Immunity. Nutrients 2019; 11:nu11020364. [PMID: 30744134 PMCID: PMC6412789 DOI: 10.3390/nu11020364] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/01/2019] [Accepted: 02/06/2019] [Indexed: 02/06/2023] Open
Abstract
Complementary feeding transitions infants from a milk-based diet to solid foods, providing essential nutrients to the infant and the developing gut microbiome while influencing immune development. Some of the earliest microbial colonisers readily ferment select oligosaccharides, influencing the ongoing establishment of the microbiome. Non-digestible oligosaccharides in prebiotic-supplemented formula and human milk oligosaccharides promote commensal immune-modulating bacteria such as Bifidobacterium, which decrease in abundance during weaning. Incorporating complex, bifidogenic, non-digestible carbohydrates during the transition to solid foods may present an opportunity to feed commensal bacteria and promote balanced concentrations of beneficial short chain fatty acid concentrations and vitamins that support gut barrier maturation and immunity throughout the complementary feeding window.
Collapse
Affiliation(s)
- Starin McKeen
- AgResearch, Food Nutrition & Health, Grasslands Research Centre, Private Bag 11008, Palmerston north4442, New Zealand.
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| | - Wayne Young
- AgResearch, Food Nutrition & Health, Grasslands Research Centre, Private Bag 11008, Palmerston north4442, New Zealand.
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| | - Jane Mullaney
- AgResearch, Food Nutrition & Health, Grasslands Research Centre, Private Bag 11008, Palmerston north4442, New Zealand.
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| | - Karl Fraser
- AgResearch, Food Nutrition & Health, Grasslands Research Centre, Private Bag 11008, Palmerston north4442, New Zealand.
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| | - Warren C McNabb
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| | - Nicole C Roy
- AgResearch, Food Nutrition & Health, Grasslands Research Centre, Private Bag 11008, Palmerston north4442, New Zealand.
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| |
Collapse
|
38
|
Young ID, Montilla A, Olano A, Wittmann A, Kawasaki N, Villamiel M. Effect of purification of galactooligosaccharides derived from lactulose with Saccharomyces cerevisiae on their capacity to bind immune cell receptor Dectin-2. Food Res Int 2019; 115:10-15. [DOI: 10.1016/j.foodres.2018.07.039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 07/19/2018] [Accepted: 07/28/2018] [Indexed: 10/28/2022]
|
39
|
Guney Varal I, Koksal N, Ozkan H, Bagci O, Dogan P. Potential use of multi-strain synbiotics for improving postnatal head circumference. Pak J Med Sci 2018; 34:1502-1506. [PMID: 30559812 PMCID: PMC6290199 DOI: 10.12669/pjms.346.16107] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Background & Objective Preterm infants need nutritional and medical requirements in accordance with the physiologic maturity at birth and maintaining optimal postnatal corporal and cerebral growth is one of the main targets of medical caregivers. However, only a few strategies exist to improve the outcomes of infants in a pathogen-rich and nutrient-poor neonatal intensive care unit environment. In this pilot study, we hypothesize that synbiotics may enhance brain growth, which is reflected indirectly by an increase in head circumference through several signalling molecules. Methods A pilot study was conducted in preterm infants with a gestational age of ≤32 weeks and a birth weight of ≤1500 grams at neonatal intensive care unit of Uludag Univeristy Medical Faculty (NICU) for one-year period. Following the randomization of the infants, a prepared commercial synbiotic solution containing multi-combined probiotics and prebiotics was administered enterally to the study group. Results The odds of a patient having a lower body weight and head circumference below the 10th percentile were significantly lower in the probiotic group (p=0.001, p=0.03, respectively). Moreover, the infants in the synbiotics group had a more optimal head circumference (between the 50th and 90th percentiles, p=0.001). Conclusions Our results show that if we can maintain optimal gut microbiota, we might achieve better neuro-development via the beneficial effects of synbiotics on cytokines, neurotransmitters, and the cellular immunity of the nervous system. Further investigational models are needed to demonstrate the beneficial effects of synbiotics on the central nervous system.
Collapse
Affiliation(s)
- Ipek Guney Varal
- Ipek Guney-Varal, M.D. Department of Pediatrics, Division of Neonatology, Uludag University Faculty of Medicine, Bursa, Turkey
| | - Nilgun Koksal
- Nilgun Koksal, M.D, Professor of Pediatrics, Department of Pediatrics, Division of Neonatology, Uludag University Faculty of Medicine, Bursa, Turkey
| | - Hilal Ozkan
- Hilal Ozkan, M.D, Associate Professor of Neonatology, Department of Pediatrics, Division of Neonatology, Uludag University Faculty of Medicine, Bursa, Turkey
| | - Onur Bagci
- Onur Bagci, M.D. Department of Pediatrics, Division of Neonatology, Uludag University Faculty of Medicine, Bursa, Turkey
| | - Pelin Dogan
- Pelin Dogan, M.D. Department of Pediatrics, Division of Neonatology, Uludag University Faculty of Medicine, Bursa, Turkey
| |
Collapse
|
40
|
Wagenaar L, Bol‐Schoenmakers M, Giustarini G, Vonk MM, van Esch BC, Knippels LM, Garssen J, Smit JJ, Pieters RH. Dietary Supplementation with Nondigestible Oligosaccharides Reduces Allergic Symptoms and Supports Low Dose Oral Immunotherapy in a Peanut Allergy Mouse Model. Mol Nutr Food Res 2018; 62:e1800369. [PMID: 30102006 PMCID: PMC6766954 DOI: 10.1002/mnfr.201800369] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 06/15/2018] [Indexed: 12/31/2022]
Abstract
SCOPE A major downside of oral immunotherapy (OIT) for food allergy is the risk of severe side effects. Non-digestible short- and long-chain fructo-oligosaccharides (scFOS/lcFOS) reduce allergy development in murine models. Therefore, it is hypothesized that scFOS/lcFOS can also support the efficacy of OIT in a peanut allergy model. METHODS AND RESULTS After sensitization to peanut extract (PE) using cholera toxin, C3H/HeOuJ mice are fed a 1% scFOS/lcFOS or control diet and receive OIT (1.5 or 15 mg PE). Hereafter, mice are exposed to PE via different routes to determine the safety and efficacy of treatment in clinical outcomes, PE-specific antibody production, and numbers of various immune cells. scFOS/lcFOS increases short-chain fatty acid levels in the caecum and reduce the acute allergic skin response and drop in body temperature after PE exposure. Interestingly, 15 mg and 1.5 mg OIT with scFOS/lcFOS induce protection against anaphylaxis, whereas 1.5 mg OIT alone does not. OIT, with or without scFOS/lcFOS, induces PE-specific immunoglobulin (Ig) IgG and IgA levels and increases CD103+ dendritic cells in the mesenteric lymph nodes. CONCLUSIONS scFOS/lcFOS and scFOS/lcFOS combined with low dose OIT are able to protect against a peanut-allergic anaphylactic response.
Collapse
Affiliation(s)
- Laura Wagenaar
- Department of ImmunotoxicologyInstitute for Risk Assessment SciencesUtrecht UniversityYalelaan 104, NL‐3584 CMUtrechtThe Netherlands
| | - Marianne Bol‐Schoenmakers
- Department of ImmunotoxicologyInstitute for Risk Assessment SciencesUtrecht UniversityYalelaan 104, NL‐3584 CMUtrechtThe Netherlands
| | - Giulio Giustarini
- Department of ImmunotoxicologyInstitute for Risk Assessment SciencesUtrecht UniversityYalelaan 104, NL‐3584 CMUtrechtThe Netherlands
| | - Marlotte M. Vonk
- Division of PharmacologyUtrecht Institute for Pharmaceutical SciencesUtrecht UniversityUniversiteitsweg 99, NL‐3584 CGUtrechtThe Netherlands
- Immunology PlatformNutricia ResearchUppsalalaan 12, NL‐3584 CTUtrechtThe Netherlands
| | - Betty C.A.M. van Esch
- Division of PharmacologyUtrecht Institute for Pharmaceutical SciencesUtrecht UniversityUniversiteitsweg 99, NL‐3584 CGUtrechtThe Netherlands
- Immunology PlatformNutricia ResearchUppsalalaan 12, NL‐3584 CTUtrechtThe Netherlands
| | - Leon M.J. Knippels
- Division of PharmacologyUtrecht Institute for Pharmaceutical SciencesUtrecht UniversityUniversiteitsweg 99, NL‐3584 CGUtrechtThe Netherlands
- Immunology PlatformNutricia ResearchUppsalalaan 12, NL‐3584 CTUtrechtThe Netherlands
| | - Johan Garssen
- Division of PharmacologyUtrecht Institute for Pharmaceutical SciencesUtrecht UniversityUniversiteitsweg 99, NL‐3584 CGUtrechtThe Netherlands
- Immunology PlatformNutricia ResearchUppsalalaan 12, NL‐3584 CTUtrechtThe Netherlands
| | - Joost J. Smit
- Department of ImmunotoxicologyInstitute for Risk Assessment SciencesUtrecht UniversityYalelaan 104, NL‐3584 CMUtrechtThe Netherlands
| | - Raymond H.H. Pieters
- Department of ImmunotoxicologyInstitute for Risk Assessment SciencesUtrecht UniversityYalelaan 104, NL‐3584 CMUtrechtThe Netherlands
| |
Collapse
|
41
|
Dai Z, Lyu W, Xiang X, Tang Y, Hu B, Ou S, Zeng X. Immunomodulatory Effects of Enzymatic-Synthesized α-Galactooligosaccharides and Evaluation of the Structure-Activity Relationship. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:9070-9079. [PMID: 30086236 DOI: 10.1021/acs.jafc.8b01939] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
In this study, α-galactooligosaccharides (α-GOSs) were synthesized using galactose as the substrate and α-galactosidase from Aspergillus niger as the catalyst. In the reaction, synthesized products of U1, U2, U3, and U4 were detected by high-performance liquid chromatography. By mass spectrometry, nuclear magnetic resonance, and 1-phenyl-3-methyl-5-pyrazolone derivatization, U1 was the mixture of disaccharides of α-d-Gal p-(1→1)-α-d-Gal, α-d-Gal p-(1→2)-α-d-Gal, α-d-Gal p-(1→3)-α-d-Gal, α-d-Gal p-(1→4)-α-d-Gal, U2 was identified to be α-d-Gal p-(1→6)-α-d-Gal, U3 was the mixture of galactotrisaccharides linked by one α-(1→6)-glycosidic linkage and one other α-glycosidic linkage, and U4 was identified as α-d-Gal p-(1→6)-α-d-Gal p-(1→6)-α-d-Gal. Afterward, the synthesized α-GOSs (U1, U2, U3, U4, and their mixture) as well as α-GOSs (manninotriose, stachyose, ciceritol, and verbascose) obtained from natural materials were used as subjects to evaluate their immunomodulatory effects in vitro by culturing mouse macrophage RAW264.7 cells. The results showed that α-GOS with a higher degree of polymerization had better immunomodulatory activity, while to a certain extent, α-GOS linked with α-(1→6)-galactosidic linkage showed a better immunomodulatory effect.
Collapse
Affiliation(s)
- Zhuqing Dai
- College of Food Science and Technology , Nanjing Agricultural University , Nanjing , Jiangsu 210095 , People's Republic of China
- Institute of Farm Product Processing , Jiangsu Academy of Agricultural Sciences , Nanjing , Jiangsu 210014 , People's Republic of China
| | - Wanyong Lyu
- Nutrition and Food Branch of China Association of Gerontology and Geriatrics , Beijing 100050 , People's Republic of China
| | - Xiaoli Xiang
- College of Food Science and Technology , Nanjing Agricultural University , Nanjing , Jiangsu 210095 , People's Republic of China
| | - Yuhong Tang
- College of Food Science and Technology , Nanjing Agricultural University , Nanjing , Jiangsu 210095 , People's Republic of China
| | - Bing Hu
- College of Food Science and Technology , Nanjing Agricultural University , Nanjing , Jiangsu 210095 , People's Republic of China
| | - Shiyi Ou
- Department of Food Science and Engineering , Jinan University , Guangzhou , Guangdong 510632 , People's Republic of China
| | - Xiaoxiong Zeng
- College of Food Science and Technology , Nanjing Agricultural University , Nanjing , Jiangsu 210095 , People's Republic of China
| |
Collapse
|
42
|
Kumar A, Vlasova AN, Deblais L, Huang HC, Wijeratne A, Kandasamy S, Fischer DD, Langel SN, Paim FC, Alhamo MA, Shao L, Saif LJ, Rajashekara G. Impact of nutrition and rotavirus infection on the infant gut microbiota in a humanized pig model. BMC Gastroenterol 2018; 18:93. [PMID: 29929472 PMCID: PMC6013989 DOI: 10.1186/s12876-018-0810-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 05/30/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Human rotavirus (HRV) is a major cause of viral gastroenteritis in infants; particularly in developing countries where malnutrition is prevalent. Malnutrition perturbs the infant gut microbiota leading to sub-optimal functioning of the immune system and further predisposing infants to enteric infections. Therefore, we hypothesized that malnutrition exacerbates rotavirus disease severity in infants. METHODS In the present study, we used a neonatal germ free (GF) piglets transplanted with a two-month-old human infant's fecal microbiota (HIFM) on protein deficient and sufficient diets. We report the effects of malnourishment on the HRV infection and the HIFM pig microbiota in feces, intestinal and systemic tissues, using MiSeq 16S gene sequencing (V4-V5 region). RESULTS Microbiota analysis indicated that the HIFM transplantation resulted in a microbial composition in pigs similar to that of the original infant feces. This model was then used to understand the interconnections between microbiota diversity, diet, and HRV infection. Post HRV infection, HIFM pigs on the deficient diet had lower body weights, developed more severe diarrhea and increased virus shedding compared to HIFM pigs on sufficient diet. However, HRV induced diarrhea and shedding was more pronounced in non-colonized GF pigs compared to HIFM pigs on either sufficient or deficient diet, suggesting that the microbiota alone moderated HRV infection. HRV infected pigs on sufficient diet showed increased microbiota diversity in intestinal tissues; whereas, greater diversity was observed in systemic tissues of HRV infected pigs fed with deficient diet. CONCLUSIONS These results suggest that proper nourishment improves the microbiota quality in the intestines, alleviates HRV disease and lower probability of systemic translocation of potential opportunistic pathogens/pathobionts. In conclusion, our findings further support the role for microbiota and proper nutrition in limiting enteric diseases.
Collapse
Affiliation(s)
- Anand Kumar
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
- Present address: Group B-10: Biosecurity and Public Health, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM USA
| | - Anastasia N. Vlasova
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - Loic Deblais
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - Huang-Chi Huang
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - Asela Wijeratne
- The Molecular and Cellular Imaging Center, The Ohio State University, Wooster, OH USA
| | - Sukumar Kandasamy
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - David D. Fischer
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - Stephanie N. Langel
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - Francine Chimelo Paim
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - Moyasar A. Alhamo
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - Lulu Shao
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
- Present address: Hillman Cancer Center, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA 15260 USA
| | - Linda J. Saif
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - Gireesh Rajashekara
- Food Animal Research Program, The Ohio Agricultural Research and Development Center,Veterinary Preventive Medicine Department, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| |
Collapse
|
43
|
Kurtys E, Casteels C, Real CC, Eisel ULM, Verkuyl JM, Broersen LM, Klein HC, Dierckx RAJO, Doorduin J, de Vries EFJ. Therapeutic effects of dietary intervention on neuroinflammation and brain metabolism in a rat model of photothrombotic stroke. CNS Neurosci Ther 2018; 25:36-46. [PMID: 29804326 PMCID: PMC6436598 DOI: 10.1111/cns.12976] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 04/23/2018] [Accepted: 04/23/2018] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION A possible target for stroke management is modulation of neuroinflammation. Evidence suggests that food components may exert anti-inflammatory properties and thus may reduce stroke-induced brain damage. AIM To investigate the efficacy of a diet, containing anti-inflammatory ingredients, as treatment for focal ischemic brain damage induced by photothrombotic stroke in the somatosensory cortex of rats. RESULTS Brain lesions were surrounded by strong astrogliosis on both day 7 and day 21 after stroke and were accompanied by a trend toward globally decreased glucose metabolism on day 7. The investigational diet applied 2 weeks before the ischemia did not affect astrocyte activation on day 7, but reduced it at day 21. The investigational diet applied immediately after the ischemia, increased astrocyte activation on day 7 and completely reversed this effect on day 21. Moreover, postischemic intervention increased glucose metabolism in somatosensory cortex ipsilateral to the lesion on day 7. CONCLUSION This study reveals potentially beneficial effects of a diet containing elevated amounts of anti-inflammatory nutrients on the recovery from ischemic brain damage. Therefore, dietary intervention can be considered as an adjuvant therapy for recovery from this brain pathology.
Collapse
Affiliation(s)
- Ewelina Kurtys
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Cindy Casteels
- Molecular Small Animal Imaging Center, Catholic University Leuven, Leuven, Belgium
| | - Caroline C Real
- Laboratory of Cellular Neurobiology, Department of Physiology and Biophysics, University of São Paulo, São Paulo, Brazil
| | - Ulrich L M Eisel
- Department of Molecular Neurobiology, University of Groningen, GELIFES, Groningen, The Netherlands
| | | | | | - Hans C Klein
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Rudi A J O Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Janine Doorduin
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Erik F J de Vries
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
44
|
Yan YL, Hu Y, Gänzle MG. Prebiotics, FODMAPs and dietary fiber — conflicting concepts in development of functional food products? Curr Opin Food Sci 2018. [DOI: 10.1016/j.cofs.2018.02.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
45
|
Kurtys E, Eisel ULM, Hageman RJJ, Verkuyl JM, Broersen LM, Dierckx RAJO, de Vries EFJ. Anti-inflammatory effects of rice bran components. Nutr Rev 2018. [DOI: 10.1093/nutrit/nuy011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Ewelina Kurtys
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, RB Groningen, The Netherlands
| | - Ulrich L M Eisel
- Department of Molecular Neurobiology, GELIFES, University of Groningen, Groningen, The Netherlands
| | | | | | | | - Rudi A J O Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, RB Groningen, The Netherlands
| | - Erik F J de Vries
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, RB Groningen, The Netherlands
| |
Collapse
|
46
|
Carlson JL, Erickson JM, Lloyd BB, Slavin JL. Health Effects and Sources of Prebiotic Dietary Fiber. Curr Dev Nutr 2018; 2:nzy005. [PMID: 30019028 PMCID: PMC6041804 DOI: 10.1093/cdn/nzy005] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/04/2017] [Accepted: 01/14/2018] [Indexed: 12/21/2022] Open
Abstract
Prebiotic dietary fibers act as carbon sources for primary and secondary fermentation pathways in the colon, and support digestive health in many ways. Fructooligosaccharides, inulin, and galactooligosaccharides are universally agreed-upon prebiotics. The objective of this paper is to summarize the 8 most prominent health benefits of prebiotic dietary fibers that are due to their fermentability by colonic microbiota, as well as summarize the 8 categories of prebiotic dietary fibers that support these health benefits. Although not all categories exhibit similar effects in human studies, all of these categories promote digestive health due to their fermentability. Scientific and regulatory definitions of prebiotics differ greatly, although health benefits of these compounds are uniformly agreed upon to be due to their fermentability by gut microbiota. Scientific evidence suggests that 8 categories of compounds all exhibit health benefits related to their metabolism by colonic taxa.
Collapse
Affiliation(s)
- Justin L Carlson
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN
| | - Jennifer M Erickson
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN
| | - Beate B Lloyd
- Global Scientific & Regulatory Affairs, The Coca-Cola Company, Atlanta, GA
| | - Joanne L Slavin
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN
| |
Collapse
|
47
|
Akkerman R, Faas MM, de Vos P. Non-digestible carbohydrates in infant formula as substitution for human milk oligosaccharide functions: Effects on microbiota and gut maturation. Crit Rev Food Sci Nutr 2018; 59:1486-1497. [PMID: 29333864 DOI: 10.1080/10408398.2017.1414030] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human milk (HM) is the golden standard for nutrition of newborn infants. Human milk oligosaccharides (HMOs) are abundantly present in HM and exert multiple beneficial functions, such as support of colonization of the gut microbiota, reduction of pathogenic infections and support of immune development. HMO-composition is during lactation continuously adapted by the mother to accommodate the needs of the neonate. Unfortunately, for many valid reasons not all neonates can be fed with HM and are either totally or partly fed with cow-milk derived infant formulas, which do not contain HMOs. These cow-milk formulas are supplemented with non-digestible carbohydrates (NDCs) that have functional effects similar to that of some HMOs, since production of synthetic HMOs is challenging and still very expensive. However, NDCs cannot substitute all HMO functions. More efficacious NDCs may be developed and customized for specific groups of neonates such as pre-matures and allergy prone infants. Here current knowledge of HMO functions in the neonate in view of possible replacement of HMOs by NDCs in infant formulas is reviewed. Furthermore, methods to expedite identification of suitable NDCs and structure/function relationships are reviewed as in vivo studies in babies are impossible.
Collapse
Affiliation(s)
- Renate Akkerman
- a Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology , University of Groningen and University Medical Center Groningen , Groningen , The Netherlands
| | - Marijke M Faas
- a Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology , University of Groningen and University Medical Center Groningen , Groningen , The Netherlands.,b Department of Obstetrics and Gynecology , University of Groningen and University Medical Center Groningen , Groningen , The Netherlands
| | - Paul de Vos
- a Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology , University of Groningen and University Medical Center Groningen , Groningen , The Netherlands
| |
Collapse
|
48
|
Lu E, Yeung M, Yeung CK. Comparative Analysis of Lactulose and Fructooligosaccharide on Growth Kinetics, Fermentation, and Antioxidant Activity of Common Probiotics. ACTA ACUST UNITED AC 2018. [DOI: 10.4236/fns.2018.93013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
49
|
Triantis V, Bode L, van Neerven RJJ. Immunological Effects of Human Milk Oligosaccharides. Front Pediatr 2018; 6:190. [PMID: 30013961 PMCID: PMC6036705 DOI: 10.3389/fped.2018.00190] [Citation(s) in RCA: 203] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 06/08/2018] [Indexed: 12/15/2022] Open
Abstract
Human milk oligosaccharides (HMOs) comprise a group of structurally complex, unconjugated glycans that are highly abundant in human milk. HMOs are minimally digested in the gastrointestinal tract and reach the colon intact, where they shape the microbiota. A small fraction of HMOs is absorbed, reaches the systemic circulation, and is excreted in urine. HMOs can bind to cell surface receptors expressed on epithelial cells and cells of the immune system and thus modulate neonatal immunity in the infant gut, and possibly also sites throughout the body. In addition, they have been shown to act as soluble decoy receptors to block the attachment of various microbial pathogens to cells. This review summarizes the current knowledge of the effects HMOs can have on infections, allergies, auto-immune diseases and inflammation, and will focus on the role of HMOs in altering immune responses through binding to immune-related receptors.
Collapse
Affiliation(s)
| | - Lars Bode
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
| | - R J Joost van Neerven
- FrieslandCampina, Amersfoort, Netherlands.,Wageningen University and Research, Cell Biology and Immunology, Wageningen, Netherlands
| |
Collapse
|
50
|
Prebiotic Oligosaccharides Potentiate Host Protective Responses against L. Monocytogenes Infection. Pathogens 2017; 6:pathogens6040068. [PMID: 29257110 PMCID: PMC5750592 DOI: 10.3390/pathogens6040068] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 12/05/2017] [Accepted: 12/15/2017] [Indexed: 12/22/2022] Open
Abstract
Prebiotic oligosaccharides are used to modulate enteric pathogens and reduce pathogen shedding. The interactions with prebiotics that alter Listeria monocytogenes infection are not yet clearly delineated. L. monocytogenes cellular invasion requires a concerted manipulation of host epithelial cell membrane receptors to initiate internalization and infection often via receptor glycosylation. Bacterial interactions with host glycans are intimately involved in modulating cellular responses through signaling cascades at the membrane and in intracellular compartments. Characterizing the mechanisms underpinning these modulations is essential for predictive use of dietary prebiotics to diminish pathogen association. We demonstrated that human milk oligosaccharide (HMO) pretreatment of colonic epithelial cells (Caco-2) led to a 50% decrease in Listeria association, while Biomos pretreatment increased host association by 150%. L. monocytogenes-induced gene expression changes due to oligosaccharide pretreatment revealed global alterations in host signaling pathways that resulted in differential subcellular localization of L. monocytogenes during early infection. Ultimately, HMO pretreatment led to bacterial clearance in Caco-2 cells via induction of the unfolded protein response and eIF2 signaling, while Biomos pretreatment resulted in the induction of host autophagy and L. monocytogenes vacuolar escape earlier in the infection progression. This study demonstrates the capacity of prebiotic oligosaccharides to minimize infection through induction of host-intrinsic protective responses.
Collapse
|