1
|
Shibata R, Nakanishi Y, Suda W, Nakano T, Sato N, Inaba Y, Kawasaki Y, Hattori M, Shimojo N, Ohno H. Neonatal gut microbiota and risk of developing food sensitization and allergy. J Allergy Clin Immunol 2025; 155:932-946. [PMID: 39692676 DOI: 10.1016/j.jaci.2024.10.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 09/20/2024] [Accepted: 10/24/2024] [Indexed: 12/19/2024]
Abstract
BACKGROUND Food sensitization (FS) develops in early infancy and is a risk factor for subsequent food allergy (FA). Recent evidence suggests relationships of gut microbiota with FS and FA. However, little is known about the role of neonatal gut microbiota in the pathobiology of these manifestations. OBJECTIVES We sought to characterize gut microbiota in children using an enterotyping approach and determine the association of gut microbiota and the enterotypes with the development of FS and FA. METHODS We combined gut microbiome and fecal short-chain fatty acid data from 2 longitudinal birth-cohort studies in Japan, clustered the microbiome data from children who were 1 week to 7 years old and their mothers and identified enterotypes. We also determined the associations of gut microbiota and enterotypes with risks of developing FS and FA across the 2 studies using multivariable regression models. RESULTS Data from the 2563 microbiomes identified 6 enterotypes. More gut bacteria (eg, Bifidobacterium) in 1-month-old children showed significant relationships with the development of FS and FA than in 1-week-old children. Enterotypes at 1 month old consisted of Bacteroides-dominant, Klebsiella-dominant, and Bifidobacterium-dominant enterotypes. Bifidobacterium-dominant enterotypes with the highest fecal propionate concentration had the lowest risks of developing FS and FA, especially of hen egg white sensitization. Bifidobacterium-dominant enterotypes had lower risks at 2 years old in one study (vs Bacteroides-dominant enterotype, adjusted odds ratio [adjOR]: 0.10, 95% CI: 0.01-0.78; vs Klebsiella-dominant enterotype, adjOR: 0.10, 95% CI: 0.01-0.77) and at 9 months old in the other study (vs Bacteroides-dominant enterotype, adjOR: 0.33, 95% CI: 0.11-0.91). CONCLUSIONS In these birth-cohort studies, gut microbiome clustering identified distinct neonatal enterotypes with differential risks of developing FS and FA.
Collapse
Affiliation(s)
- Ryohei Shibata
- Laboratorie for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan; Department of Pediatric Surgery, Graduate School of Medicine, Chiba University, Chiba City, Japan.
| | - Yumiko Nakanishi
- Laboratorie for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Wataru Suda
- Laboratorie for Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Taiji Nakano
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba City, Japan
| | - Noriko Sato
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba City, Japan
| | - Yosuke Inaba
- Clinical Research Center, Chiba University Hospital, Chiba City, Japan
| | - Yohei Kawasaki
- Faculty of Nursing, Japanese Red Cross College of Nursing, Tokyo, Japan
| | - Masahira Hattori
- Laboratorie for Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Naoki Shimojo
- Center for Preventive Medical Sciences, Chiba University, Chiba City, Japan
| | - Hiroshi Ohno
- Laboratorie for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan; Laboratorie for Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Japan.
| |
Collapse
|
2
|
Bai J, Zeng Q, Den W, Huang L, Wu Z, Li X, Tong P, Chen H, Yang A. Synergistic Synbiotic-Containing Lactiplantibacillus plantarum and Fructo-Oligosaccharide Alleviate the Allergenicity of Mice Induced by Soy Protein. Foods 2025; 14:109. [PMID: 39796399 PMCID: PMC11720218 DOI: 10.3390/foods14010109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 11/30/2024] [Accepted: 12/27/2024] [Indexed: 01/13/2025] Open
Abstract
Prebiotics and probiotics have key roles in the intervention and treatment of food allergies. This study assesses the effect of Lactiplantibacillus plantarum synergistic fructo-oligosaccharide (Lp-FOS) intervention using an allergic mouse model induced by soy protein. The results showed that Lp synergistic FOS significantly decreased clinical allergy scores, inhibited specific antibodies (IgE, IgG, and IgG1), IL-4, IL-6, and IL-17A levels, and increased IFN-γ and IL-10 levels. Meanwhile, flow cytometry showed that Lp-FOS intervention inhibited the percentage of dendritic cell (DC) subsets in splenocytes and increased the Th1/Th2 and Treg/Th17 ratios. Furthermore, Lp-FOS intervention upregulated the mRNA levels of T-bet and Foxp3 and downregulated the mRNA levels of GATA3. Finally, non-targeted metabolomic analysis showed that Lp-FOS improved serum metabolic disorders caused by food allergies through regulating glycine, serine, and threonine metabolism, butanoate metabolism, glyoxylate and dicarboxylate metabolism, the biosynthesis of cofactors, and glycerophospholipid metabolism. These data showed that the combination formulation Lp-FOS could be a promising adjuvant treatment for food allergies.
Collapse
Affiliation(s)
- Jing Bai
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanjing Dong Lu 235, Nanchang 330047, China; (J.B.); (Q.Z.); (W.D.); (L.H.); (Z.W.); (X.L.); (P.T.); (H.C.)
- School of Food Science and Technology, Nanchang University, Nanchang 330047, China
- Sino-German Joint Research Institute, Nanchang University, Nanjing Dong Lu 235, Nanchang 330047, China
| | - Qian Zeng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanjing Dong Lu 235, Nanchang 330047, China; (J.B.); (Q.Z.); (W.D.); (L.H.); (Z.W.); (X.L.); (P.T.); (H.C.)
- School of Food Science and Technology, Nanchang University, Nanchang 330047, China
- Sino-German Joint Research Institute, Nanchang University, Nanjing Dong Lu 235, Nanchang 330047, China
| | - Wen Den
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanjing Dong Lu 235, Nanchang 330047, China; (J.B.); (Q.Z.); (W.D.); (L.H.); (Z.W.); (X.L.); (P.T.); (H.C.)
- School of Food Science and Technology, Nanchang University, Nanchang 330047, China
- Sino-German Joint Research Institute, Nanchang University, Nanjing Dong Lu 235, Nanchang 330047, China
| | - Liheng Huang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanjing Dong Lu 235, Nanchang 330047, China; (J.B.); (Q.Z.); (W.D.); (L.H.); (Z.W.); (X.L.); (P.T.); (H.C.)
- School of Food Science and Technology, Nanchang University, Nanchang 330047, China
- Sino-German Joint Research Institute, Nanchang University, Nanjing Dong Lu 235, Nanchang 330047, China
| | - Zhihua Wu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanjing Dong Lu 235, Nanchang 330047, China; (J.B.); (Q.Z.); (W.D.); (L.H.); (Z.W.); (X.L.); (P.T.); (H.C.)
- Sino-German Joint Research Institute, Nanchang University, Nanjing Dong Lu 235, Nanchang 330047, China
| | - Xin Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanjing Dong Lu 235, Nanchang 330047, China; (J.B.); (Q.Z.); (W.D.); (L.H.); (Z.W.); (X.L.); (P.T.); (H.C.)
- School of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Ping Tong
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanjing Dong Lu 235, Nanchang 330047, China; (J.B.); (Q.Z.); (W.D.); (L.H.); (Z.W.); (X.L.); (P.T.); (H.C.)
| | - Hongbing Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanjing Dong Lu 235, Nanchang 330047, China; (J.B.); (Q.Z.); (W.D.); (L.H.); (Z.W.); (X.L.); (P.T.); (H.C.)
- Sino-German Joint Research Institute, Nanchang University, Nanjing Dong Lu 235, Nanchang 330047, China
| | - Anshu Yang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanjing Dong Lu 235, Nanchang 330047, China; (J.B.); (Q.Z.); (W.D.); (L.H.); (Z.W.); (X.L.); (P.T.); (H.C.)
- Sino-German Joint Research Institute, Nanchang University, Nanjing Dong Lu 235, Nanchang 330047, China
| |
Collapse
|
3
|
Wu Y, Chen B, Wu H, Gao J, Meng X, Chen H. How maternal factors shape the immune system of breastfed infants to alleviate food allergy: A systematic and updated review. Immunology 2025; 174:1-16. [PMID: 39344356 DOI: 10.1111/imm.13864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 09/06/2024] [Indexed: 10/01/2024] Open
Abstract
What infants eat early in life may shape the immune system and have long-standing consequences on the health of the host during later life. In the early months post-birth, breast milk serves as the exclusive and optimal nourishment for infants, facilitating crucial molecular exchanges between mother and infant. Recent advances have uncovered that some maternal factors influence breastfed infant outcomes, including the risk of food allergy (FA). To date, accumulated data show that breastfed infants have a lower risk of FA. However, the issue remains disputed, some reported preventive allergy effects, while others did not confirm such effects, or if identified, protective effects were limited to early childhood. The disputed outcomes may be attributed to the maternal status, as it determines the compounds of the breast milk that breastfed infants are exposed to. In this review, we first detail the compounds in breast milk and their roles in infant FA. Then, we present maternal factors resulting in alterations in breast milk compounds, such as maternal health status, maternal diet intake, and maternal food allergen intake, which subsequently impact FA in breastfed infants. Finally, we analyze how these compounds in breast milk alleviated the infant FA by mother-to-infant transmission. Altogether, the mechanisms are primarily linked to the synergetic and direct effects of compounds in breast milk, via promoting the colonization of gut microbiota and the development of the immune system in infants.
Collapse
Affiliation(s)
- Yuhong Wu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- College of Food Science and Technology, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, China
| | - Bihua Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- College of Food Science and Technology, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, China
| | - Huan Wu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- College of Food Science and Technology, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, China
| | - Jinyan Gao
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- College of Food Science and Technology, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, China
| | - Xuanyi Meng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, China
- Jiangxi-OAI Joint Research Institute, Nanchang University, Nanchang, China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang, China
- Jiangxi-OAI Joint Research Institute, Nanchang University, Nanchang, China
| |
Collapse
|
4
|
Pei Z, Qian L, Miao T, Wang H, Lu W, Chen Y, Zhuang Q. Uncovering the mechanisms underlying the efficacy of probiotic strains in mitigating food allergies: an emphasis on gut microbiota and indoleacrylic acid. Front Nutr 2024; 11:1523842. [PMID: 39726866 PMCID: PMC11670748 DOI: 10.3389/fnut.2024.1523842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024] Open
Abstract
Food allergies manifest as systemic or digestive allergic responses induced by food allergens, and their progression has been demonstrated to be intimately associated with the host's gut microbiota. Our preceding investigation has revealed that the probiotic strains Lactiplantibacillus plantarum CCFM1189 and Limosilactobacillus reuteri CCFM1190 possess the capability to mitigate the symptoms of food allergy in mice. However, the underlying mechanisms and material foundations through which these probiotic strains exert their effects remain enigmatic. Here, we initially compared the ameliorative effects of these two probiotic strains on food allergy mice subjected to antibiotic cocktail (ABX) treatment. It is indicated that ABX treatment was ineffective in alleviating weight loss, diarrhea, and allergic symptoms in mice, and it also inhibited the reduction of histamine and T helper cell 2 (Th2) cytokines mediated by effective strains, suggesting that effective strains must operate through the gut microbiota. Then, building upon the outcomes of prior non-targeted metabolomics studies, by quantifying the content of indoleacrylic acid (IA) in single-strain fermentation of probiotic strains and mouse feces, it was ascertained that effective strains do not synthesize IA themselves but can augment the concentration of IA in the gut by modulating the gut microbiota. Ultimately, we discovered that direct intervention with IA could mitigate diarrhea, allergic symptoms, and intestinal damage by modulating immunoglobulin E (IgE) levels, histamine, Th2 cytokines, and tight junction proteins, thereby corroborating that IA is a pivotal metabolite for the alleviation of food allergies. These observations underscore the significance of gut microbiota and metabolites like IA in the management of food allergies and hold potential implications for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Zhangming Pei
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Li Qian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Taolin Miao
- Children's ENT Department, Affiliated Children’s Hospital of Jiangnan University (Wuxi Children’s Hospital), Wuxi, China
| | - Hongchao Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wenwei Lu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| | - Yuqing Chen
- Children's ENT Department, Affiliated Women’s Hospital of Jiangnan University (Wuxi Maternal and Child Healthcare Hospital), Wuxi, China
| | - Qianger Zhuang
- Children's ENT Department, Affiliated Children’s Hospital of Jiangnan University (Wuxi Children’s Hospital), Wuxi, China
| |
Collapse
|
5
|
Moriki D, León ED, García-Gamero G, Jiménez-Hernández N, Artacho A, Pons X, Koumpagioti D, Dinopoulos A, Papaevangelou V, Priftis KN, Douros K, Francino MP. Specific Gut Microbiome Signatures in Children with Cow's Milk Allergy. Nutrients 2024; 16:2752. [PMID: 39203888 PMCID: PMC11357501 DOI: 10.3390/nu16162752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 09/03/2024] Open
Abstract
Although gut dysbiosis is associated with cow's milk allergy (CMA), causality remains uncertain. This study aimed to identify specific bacterial signatures that influence the development and outcome of the disease. We also investigated the effect of hypoallergenic formula (HF) consumption on the gut microbiome of milk-allergic children. 16S rRNA amplicon sequencing was applied to characterize the gut microbiome of 32 milk-allergic children aged 5-12 years and 36 age-matched healthy controls. We showed that the gut microbiome of children with CMA differed significantly from that of healthy children, regardless of whether they consumed cow's milk. Compared to that of healthy cow's milk consumers, it was depleted in Bifidobacterium, Coprococcus catus, Monoglobus, and Lachnospiraceae GCA-900066575, while being enriched in Oscillibacter valericigenes, Negativibacillus massiliensis, and three genera of the Ruminococcaceae family. Of these, only the Ruminococcaceae taxa were also enriched in healthy children not consuming cow's milk. Furthermore, the gut microbiome of children who developed tolerance and had received an HF was similar to that of healthy children, whereas that of children who had not received an HF was significantly different. Our results demonstrate that specific gut microbiome signatures are associated with CMA, which differ from those of dietary milk elimination. Moreover, HF consumption affects the gut microbiome of children who develop tolerance.
Collapse
Affiliation(s)
- Dafni Moriki
- 3rd Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University Hospital, 12462 Athens, Greece
| | - E. Daniel León
- Department of Genomics and Health, Fundación Para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valencia (FISABIO), 46020 Valencia, Spain
| | - Gabriel García-Gamero
- Department of Genomics and Health, Fundación Para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valencia (FISABIO), 46020 Valencia, Spain
| | - Nuria Jiménez-Hernández
- Department of Genomics and Health, Fundación Para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valencia (FISABIO), 46020 Valencia, Spain
| | - Alejandro Artacho
- Department of Genomics and Health, Fundación Para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valencia (FISABIO), 46020 Valencia, Spain
| | - Xavier Pons
- Department of Genomics and Health, Fundación Para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valencia (FISABIO), 46020 Valencia, Spain
| | - Despoina Koumpagioti
- Department of Nursing, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Argirios Dinopoulos
- 3rd Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University Hospital, 12462 Athens, Greece
| | - Vassiliki Papaevangelou
- 3rd Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University Hospital, 12462 Athens, Greece
| | - Kostas N. Priftis
- 3rd Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University Hospital, 12462 Athens, Greece
| | - Konstantinos Douros
- 3rd Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University Hospital, 12462 Athens, Greece
| | - M. Pilar Francino
- Department of Genomics and Health, Fundación Para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valencia (FISABIO), 46020 Valencia, Spain
- CIBER en Epidemiología y Salud Pública, 28029 Madrid, Spain
| |
Collapse
|
6
|
Huang H, Lin X, Meng X, Liu Y, Fan J, Zhu L, Chen J, Zhang L, Mi H, Deng J. Effects of replacing wheat bran with palm kernel cake or fermented palm kernel cake on the growth performance, intestinal microbiota and intestinal health of tilapia (GIFT, Oreochromis niloticus). Front Nutr 2024; 11:1368251. [PMID: 38450228 PMCID: PMC10915778 DOI: 10.3389/fnut.2024.1368251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 02/12/2024] [Indexed: 03/08/2024] Open
Abstract
A nine-week feeding trial was conducted to evaluate the effects of replacing wheat bran (WB) with palm kernel cake (PKC) or fermented palm kernel cake (FPKC) on the growth performance, intestinal microbiota and intestinal health of genetically improved farmed tilapia (GIFT, Oreochromis niloticus) (initial weight 7.00 ± 0.01 g). Eleven isonitrogenous and isolipidic experimental diets were formulated by replacing 0, 20, 40, 60, 80, and 100% of dietary WB with PKC or FPKC. Replacement of WB with PKC concentrations up to 80% had no significant effect on the growth rate of tilapia or feed utilisation (p > 0.05). FPKC improved the growth performance of tilapia, with optimum growth achieved at 40% replacement level (p < 0.05). Complete replacement with PKC significantly decreased the activity of lipase and trypsin, and reduced the height of muscularis and the height of villus (p < 0.05). However, FPKC significantly increased amylase activity and villus height (p < 0.05). The apparent digestibility of dry matter and energy decreased linearly with increasing levels of PKC substitution, while FPKC showed the opposite trend (p < 0.05). PKC replacement of WB by 20% significantly reduced serum diamine oxidase activity and endothelin levels and increased intestinal tight junctions (p < 0.05). However, FPKC significantly decreased diamine oxidase activity and increased intestinal tight junctions (p < 0.05). PKC completely replaced WB, up-regulating the expression of pro-inflammatory factors (il-1β) (p < 0.05). When 40% of WB was replaced with FPKC, the expression of pro-inflammatory factors (il-1β and il-6) was decreased significantly (p < 0.05). Completely replacement of WB with PKC reduced the abundance of Firmicutes and Chloroflexi, while FPKC reduced the abundance of Fusobacteriota and increased the levels of Actinobacteriota. WB can be replaced with PKC up to 80% in tilapia feeds. However, the high percentage of gluten induced intestinal inflammation, impaired gut health, and reduced dietary nutrient utilisation and growth performance. Complete replacement of WB with FPKC promoted intestinal immunity. It also improved dietary nutrient utilisation and growth performance. However, the optimal growth was achieved at a 40% replacement level.
Collapse
Affiliation(s)
- Huajing Huang
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
- Aquatic Animals Precision Nutrition and High-Efficiency Feed Engineering Research Centre of Guangdong Province, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Xiangqin Lin
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
- Aquatic Animals Precision Nutrition and High-Efficiency Feed Engineering Research Centre of Guangdong Province, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Xiaoxue Meng
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
- Aquatic Animals Precision Nutrition and High-Efficiency Feed Engineering Research Centre of Guangdong Province, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Yu Liu
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
- Aquatic Animals Precision Nutrition and High-Efficiency Feed Engineering Research Centre of Guangdong Province, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Jiongting Fan
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
- Aquatic Animals Precision Nutrition and High-Efficiency Feed Engineering Research Centre of Guangdong Province, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Lulu Zhu
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
- Aquatic Animals Precision Nutrition and High-Efficiency Feed Engineering Research Centre of Guangdong Province, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Jiaxian Chen
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
- Aquatic Animals Precision Nutrition and High-Efficiency Feed Engineering Research Centre of Guangdong Province, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Lu Zhang
- Tongwei Agricultural Development Co., Ltd., Chengdu, Sichuan, China
| | - Haifeng Mi
- Tongwei Agricultural Development Co., Ltd., Chengdu, Sichuan, China
| | - Junming Deng
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
- Aquatic Animals Precision Nutrition and High-Efficiency Feed Engineering Research Centre of Guangdong Province, Guangdong Ocean University, Zhanjiang, Guangdong, China
| |
Collapse
|
7
|
Lin M, Yanjun C. Research progress on the mechanism of probiotics regulating cow milk allergy in early childhood and its application in hypoallergenic infant formula. Front Nutr 2024; 11:1254979. [PMID: 38419849 PMCID: PMC10900986 DOI: 10.3389/fnut.2024.1254979] [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: 07/08/2023] [Accepted: 01/22/2024] [Indexed: 03/02/2024] Open
Abstract
Some infants and young children suffer from cow's milk allergy (CMA), and have always mainly used hypoallergenic infant formula as a substitute for breast milk, but some of these formulas can still cause allergic reactions. In recent years, it has been found that probiotic nutritional interventions can regulate CMA in children. Scientific and reasonable application of probiotics to hypoallergenic infant formula is the key research direction in the future. This paper discusses the mechanism and clinical symptoms of CMA in children. This review critically ex- amines the issue of how probiotics use intestinal flora as the main vector to combine with the immune system to exert physiological functions to intervene CMA in children, with a particular focus on four mechanisms: promoting the early establishment of intestinal microecological balance, regulating the body's immunity and alleviating allergic response, enhancing the intestinal mucosal barrier function, and destroying allergen epitopes. Additionally, it overviews the development process of hypoallergenic infant formula and the research progress of probiotics in hypoallergenic infant formula. The article also offers suggestions and outlines potential future research directions and ideas in this field.
Collapse
Affiliation(s)
| | - Cong Yanjun
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, College of Food and Health, Beijing Technology and Business University, Beijing, China
| |
Collapse
|
8
|
Gu K, Wu A, Yu B, Zhang T, Lai X, Chen J, Yan H, Zheng P, Luo Y, Luo J, Pu J, Wang Q, Wang H, Chen D. Iron overload induces colitis by modulating ferroptosis and interfering gut microbiota in mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 905:167043. [PMID: 37717771 DOI: 10.1016/j.scitotenv.2023.167043] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/19/2023]
Abstract
BACKGROUND Iron plays a pivotal role in various physiological processes, including intestinal inflammation, ferroptosis, and the modulation of the gut microbiome. However, the way these factors interact with each other is unclear. METHODS Mice models were fed with low, normal and high iron diets to assess their impacts on colitis, ferroptosis and gut microbiota. Untargeted fecal metabolomics analysis, 16S rRNA sequencing, histopathology analysis, real-time quantitative PCR and western blot were performed to analyze the differences in the intestinal inflammatory response and understanding its regulatory mechanisms between low, normal and high iron groups. RESULTS The iron overload changed the serum iron, colon iron and fecal iron. In addition, the iron overload induced the colitis, induced the ferroptosis and altered the microbiome composition in the fecal of mice. By using untargeted fecal metabolomics analysis to screen of metabolites in the fecal, we found that different metabolomics profiles in the fecal samples between iron deficiency, normal iron and iron overload groups. The correlation analysis showed that both of iron deficiency and overload were closely related to Dubosiella. The relationship between microbial communities (e.g., Akkermansia, Alistipes, and Dubosiella) and colitis-related parameters was highly significant. Additionally, Alistipes and Bacteroides microbial communities displayed a close association with ferroptosis-related parameters. Iron overload reduced the concentration of metabolites, which exert the anti-inflammatory effects (e.g., (+)-.alpha.-tocopherol) in mice. The nucleotide metabolism, enzyme metabolism and metabolic diseases were decreased and the lipid metabolism was increased in iron deficiency and iron overload groups compared with normal iron group. CONCLUSION Iron overload exacerbated colitis in mice by modulating ferroptosis and perturbing the gut microbiota. Iron overload-induced ferroptosis was associated with NRF2/GPX-4 signaling pathway. Specific microbial taxa and their associated metabolites were closely intertwined with both colitis and ferroptosis markers.
Collapse
Affiliation(s)
- Ke Gu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Aimin Wu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Tingting Zhang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Xin Lai
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Junzhou Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Hui Yan
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Ping Zheng
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuheng Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Junqiu Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Junning Pu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Quyuan Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Huifen Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
9
|
Borbet TC, Pawline MB, Li J, Ho ML, Yin YS, Zhang X, Novikova E, Jackson K, Mullins BJ, Ruiz VE, Hines MJ, Zhang XS, Müller A, Koralov SB, Blaser MJ. Disruption of the early-life microbiota alters Peyer's patch development and germinal center formation in gastrointestinal-associated lymphoid tissue. iScience 2023; 26:106810. [PMID: 37235047 PMCID: PMC10206152 DOI: 10.1016/j.isci.2023.106810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 03/17/2023] [Accepted: 05/01/2023] [Indexed: 05/28/2023] Open
Abstract
During postnatal development, both the maturing microbiome and the host immune system are susceptible to environmental perturbations such as antibiotic use. The impact of timing in which antibiotic exposure occurs was investigated by treating mice from days 5-9 with amoxicillin or azithromycin, two of the most commonly prescribed medications in children. Both early-life antibiotic regimens disrupted Peyer's patch development and immune cell abundance, with a sustained decrease in germinal center formation and diminished intestinal immunoglobulin A (IgA) production. These effects were less pronounced in adult mice. Through comparative analysis of microbial taxa, Bifidobacterium longum abundance was found to be associated with germinal center frequency. When re-introduced to antibiotic-exposed mice, B. longum partially rescued the immunological deficits. These findings suggest that early-life antibiotic use affects the development of intestinal IgA-producing B cell functions and that probiotic strains could be used to restore normal development after antibiotic exposure.
Collapse
Affiliation(s)
- Timothy C. Borbet
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Miranda B. Pawline
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Jackie Li
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Melody L. Ho
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Yue Sandra Yin
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
- Center for Advanced Biotechnology and Medicine, Rutgers University, New Brunswick, NJ 08854, USA
| | - Xiaozhou Zhang
- Institute of Molecular Cancer Research, University of Zurich, Zurich 8057, Switzerland
| | - Ekaterina Novikova
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Katelyn Jackson
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
- Department of Biological Sciences, Mississippi State University, Mississippi State, MS 39762, USA
| | - Briana J. Mullins
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Victoria E. Ruiz
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Marcus J. Hines
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Xue-Song Zhang
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
- Center for Advanced Biotechnology and Medicine, Rutgers University, New Brunswick, NJ 08854, USA
| | - Anne Müller
- Institute of Molecular Cancer Research, University of Zurich, Zurich 8057, Switzerland
| | - Sergei B. Koralov
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Martin J. Blaser
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
- Center for Advanced Biotechnology and Medicine, Rutgers University, New Brunswick, NJ 08854, USA
| |
Collapse
|
10
|
Yang Y, Wang Y, Zhao L, Wang F, Li M, Wang Q, Luo H, Zhao Q, Zeng J, Zhao Y, Du F, Chen Y, Shen J, Wei S, Xiao Z, Wu X. Chinese herbal medicines for treating ulcerative colitis via regulating gut microbiota-intestinal immunity axis. CHINESE HERBAL MEDICINES 2023; 15:181-200. [PMID: 37265772 PMCID: PMC10230642 DOI: 10.1016/j.chmed.2023.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/05/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Ulcerative colitis (UC) is one of types of inflammatory bowel disease with high recurrence. Recent studies have highlighted that microbial dysbiosis as well as abnormal gut immunity are crucial factors that initiate a series of inflammatory responses in the UC. Modulating the gut microbiota-intestinal immunity loop has been suggested as one of key strategies for relieving UC. Many Chinese herbal medicines including some of single herb, herbal formulas and the derived constituents have been reported with protective effect against UC through modulating gut microbiome and intestinal immunity. Some clinical trials have shown promising results. This review thus focused on the current knowledge on using Chinese herbal medicines for treating UC from the mechanism aspects of regulating intestinal homeostasis involving microbiota and gut immunity. The existing clinical trials are also summarized.
Collapse
Affiliation(s)
- Yifei Yang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Luzhou Key Laboratory of Cell Therapy & Cell Drugs, Luzhou 646000, China
| | - Yi Wang
- Sichuan Fifth People’s Hospital, Chengdu 610015, China
| | - Long Zhao
- Department of Spleen and Stomach Diseases, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China
| | - Fang Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Luzhou Key Laboratory of Cell Therapy & Cell Drugs, Luzhou 646000, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Luzhou Key Laboratory of Cell Therapy & Cell Drugs, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Qin Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Luzhou Key Laboratory of Cell Therapy & Cell Drugs, Luzhou 646000, China
| | - Haoming Luo
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Luzhou Key Laboratory of Cell Therapy & Cell Drugs, Luzhou 646000, China
| | - Qianyun Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Luzhou Key Laboratory of Cell Therapy & Cell Drugs, Luzhou 646000, China
| | - Jiuping Zeng
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Luzhou Key Laboratory of Cell Therapy & Cell Drugs, Luzhou 646000, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Luzhou Key Laboratory of Cell Therapy & Cell Drugs, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Luzhou Key Laboratory of Cell Therapy & Cell Drugs, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Yu Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Luzhou Key Laboratory of Cell Therapy & Cell Drugs, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Luzhou Key Laboratory of Cell Therapy & Cell Drugs, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Shulin Wei
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Luzhou Key Laboratory of Cell Therapy & Cell Drugs, Luzhou 646000, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Luzhou Key Laboratory of Cell Therapy & Cell Drugs, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Luzhou Key Laboratory of Cell Therapy & Cell Drugs, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao 999078, China
| |
Collapse
|
11
|
Oral Administration of Lotus-Seed Resistant Starch Protects against Food Allergy. Foods 2023; 12:foods12040737. [PMID: 36832810 PMCID: PMC9956242 DOI: 10.3390/foods12040737] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/28/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023] Open
Abstract
Food allergy is a serious food safety and public health issue. However, the medical interventions for allergy treatment are still suboptimal. Recently, the gut microbiome-immune axis has been considered as a promising target to reduce the symptoms of food allergy. In this study, we explore the oral administration of lotus-seed resistant starch as a means to protect against food allergy using an ovalbumin (OVA) sensitization and challenge rodent model. The results obtained showed that lotus-seed resistant starch intervention alleviated the food allergy symptoms (such as reductions in body temperature and allergic diarrhea). Furthermore, lotus-seed resistant starch also attenuated the increase in OVA-specific immunoglobulins and improved Th1/Th2 imbalance in OVA-sensitized mice. These anti-allergic effects might be associated with the actions of lotus-seed resistant starch on intestinal microbiota. Taken together, our findings suggest that daily ingestion of lotus-seed resistant starch might be effective for the alleviation of food allergy.
Collapse
|
12
|
Bifidobacterium longum subsp. longum 5 1A Attenuates Signs of Inflammation in a Murine Model of Food Allergy. Probiotics Antimicrob Proteins 2023; 15:63-73. [PMID: 34558015 DOI: 10.1007/s12602-021-09846-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2021] [Indexed: 01/18/2023]
Abstract
Food allergy is a pathological condition that can lead to hives, swelling, gastrointestinal distress, cardiovascular and respiratory compromise, and even anaphylaxis. The lack of treatment resources emphasizes the necessity for new therapeutic strategies, and in this way, probiotics has been pointed out as an alternative, especially because of its immunomodulatory properties. The goal of this study was to evaluate the probiotic effect of Bifidobacterium longum subsp. longum 51A (BL51A) in a murine model of ovalbumin (OVA) food allergy, as well as to investigate the effect of the dose and viability of the bacteria on the proposed model. For this purpose, the probiotic effect was assessed by clinical, immunological, and histological parameters in mice treated or not with the BL51A and sensitized or not with OVA. Oral administration of BL51A prevented weight loss and reduced serum levels of IgE anti-OVA and of sIgA in the intestinal fluid. Also, it reduced the intestinal permeability, proximal jejunum damage, recruitment of eosinophils and neutrophils, and levels of eotaxin-1, CXCL1/KC, IL4, IL5, IL6, IL13, and TNF. Furthermore, the treatment was able to increase the levels of IL10. Investigating different doses administered, the level of 108 CFU showed the best results in terms of protective effect. In addition, the administration of the inactivated bacteria did not present any beneficial effect. Results demonstrate that BL51A promotes a systemic immunomodulatory protective effect in a murine model of food allergy that depends on the dose and viability of the bacteria, suggesting its use as probiotic in such disease.
Collapse
|
13
|
Liu P, Hu T, Kang C, Liu J, Zhang J, Ran H, Zeng X, Qiu S. Research Advances in the Treatment of Allergic Rhinitis by Probiotics. J Asthma Allergy 2022; 15:1413-1428. [PMID: 36238950 PMCID: PMC9552798 DOI: 10.2147/jaa.s382978] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/11/2022] [Indexed: 11/23/2022] Open
Abstract
Allergic rhinitis (AR) impairs the quality of life of patients and reduces the efficiency of social work, it is an increasingly serious public medical and economic problem in the world. Conventional anti-allergic drugs for the treatment of allergic rhinitis (AR) can cause certain side effects, which limit the quality of life of patients. Therefore, it makes sense to look for other forms of treatment. Several studies in recent years have shown that probiotics have shown anti-allergic effects in various mouse and human studies. For example, the application of certain probiotic strains can effectively relieve the typical nasal and ocular symptoms of allergic rhinitis in children and adults, thereby improving the quality of life and work efficiency. At the same time, previous studies in humans and mice have found that probiotics can produce multiple effects, such as reduction of Th2 cell inflammatory factors and/or increase of Th1 cell inflammatory factors, changes in allergy-related immunoglobulins and cell migration, regulate Th1/Th2 balance or restore intestinal microbiota disturbance. For patients with limited activity or allergic rhinitis with more attacks and longer attack duration, oral probiotics have positive effects. The efficacy of probiotics in the prevention and treatment of allergic rhinitis is remarkable, but its specific mechanism needs further study. This review summarizes the research progress of probiotics in the treatment of allergic rhinitis in recent years.
Collapse
Affiliation(s)
- Peng Liu
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zunyi, People’s Republic of China
| | - Tianyong Hu
- Department of Otolaryngology, Longgang E.N.T Hospital & Shenzhen Key Laboratory of E.N.T, Institute of E.N.T Shenzhen, Shenzhen, People’s Republic of China
| | - Chenglin Kang
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zunyi, People’s Republic of China
| | - Jiangqi Liu
- Department of Otolaryngology, Longgang E.N.T Hospital & Shenzhen Key Laboratory of E.N.T, Institute of E.N.T Shenzhen, Shenzhen, People’s Republic of China
| | - Jin Zhang
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zunyi, People’s Republic of China
| | - Hong Ran
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zunyi, People’s Republic of China
| | - Xianhai Zeng
- Department of Otolaryngology, Longgang E.N.T Hospital & Shenzhen Key Laboratory of E.N.T, Institute of E.N.T Shenzhen, Shenzhen, People’s Republic of China
| | - Shuqi Qiu
- Department of Otolaryngology, Longgang E.N.T Hospital & Shenzhen Key Laboratory of E.N.T, Institute of E.N.T Shenzhen, Shenzhen, People’s Republic of China
| |
Collapse
|
14
|
Lv XC, Wu Q, Yuan YJ, Li L, Guo WL, Lin XB, Huang ZR, Rao PF, Ai LZ, Ni L. Organic chromium derived from the chelation of Ganoderma lucidum polysaccharide and chromium (III) alleviates metabolic syndromes and intestinal microbiota dysbiosis induced by high-fat and high-fructose diet. Int J Biol Macromol 2022; 219:964-979. [DOI: 10.1016/j.ijbiomac.2022.07.211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/10/2022] [Accepted: 07/26/2022] [Indexed: 11/05/2022]
|
15
|
Fong FLY, El-Nezami H, Mykkänen O, Kirjavainen PV. The Effects of Single Strains and Mixtures of Probiotic Bacteria on Immune Profile in Liver, Spleen, and Peripheral Blood. Front Nutr 2022; 9:773298. [PMID: 35495948 PMCID: PMC9039324 DOI: 10.3389/fnut.2022.773298] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 03/04/2022] [Indexed: 12/12/2022] Open
Abstract
Probiotic bacteria have potential use as immunomodulators but comparative data on their immunological effects are very limited. The aim of this study was to characterize the effect of oral administration of probiotic strains, alone or as mixtures, on systemic and organ-specific immune responses. For this purpose, healthy C57BL/6 mice were perorally administered probiotics for 3 weeks. A total of five common probiotic strains, Lactobacillus rhamnosus species GG (LGG) and LC705, Bifidobacterium breve 99 (Bb99), Propionibacterium freudenreichii Shermanii JS (PJS), and Escherichia coli Nissle 1917 (EcN), and two of their mixtures, were tested. Livers, spleens, and blood were collected for investigation. A number of five treatments increased the abundance of the natural killer (NK) cells. Bb99 had the most prominent effect on hepatic NK cells (20.0 ± 1.8%). LGG (liver: 5.8 ± 1.0%; spleen: 1.6 ± 0.4%), Bb99 (liver: 13.9 ± 4.3%; spleen: 10.3 ± 3.7%), and EcN (liver: 8.5 ± 3.2%; spleen: 1.0 ± 0.2%) increased the percentage of both the hepatic and splenic T-helper 17 cells. Moreover, LGG (85.5 ± 3.0%) and EcN (89.6 ± 1.2%) increased the percentage of splenic regulatory T-cells. The tested mixtures of the probiotics had different immunological effects from their individual components on cell-mediated responses and cytokine production. In conclusion, our results confirm that the immunomodulatory potential of the probiotics is strain- and organ/tissue-specific, and the effects of probiotic mixtures cannot be predicted based on their single constituents.
Collapse
Affiliation(s)
- Fiona Long Yan Fong
- School of Biological Sciences, The University of Hong Kong, Pokfulam, Hong Kong, SAR China.,Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Hani El-Nezami
- School of Biological Sciences, The University of Hong Kong, Pokfulam, Hong Kong, SAR China.,Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Otto Mykkänen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Pirkka V Kirjavainen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland.,Department of Environmental Health, Finnish Institute for Health and Welfare, Kuopio, Finland
| |
Collapse
|
16
|
Chen J, Chen X, Ho CL. Recent Development of Probiotic Bifidobacteria for Treating Human Diseases. Front Bioeng Biotechnol 2022; 9:770248. [PMID: 35004640 PMCID: PMC8727868 DOI: 10.3389/fbioe.2021.770248] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/08/2021] [Indexed: 12/12/2022] Open
Abstract
Bifidobacterium is a non-spore-forming, Gram-positive, anaerobic probiotic actinobacterium and commonly found in the gut of infants and the uterine region of pregnant mothers. Like all probiotics, Bifidobacteria confer health benefits on the host when administered in adequate amounts, showing multifaceted probiotic effects. Examples include B. bifidum, B. breve, and B. longum, common Bifidobacterium strains employed to prevent and treat gastrointestinal disorders, including intestinal infections and cancers. Herein, we review the latest development in probiotic Bifidobacteria research, including studies on the therapeutic impact of Bifidobacterial species on human health and recent efforts in engineering Bifidobacterium. This review article would provide readers with a wholesome understanding of Bifidobacteria and its potentials to improve human health.
Collapse
Affiliation(s)
- Jun Chen
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, China
| | - Xinyi Chen
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, China
| | - Chun Loong Ho
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, China
| |
Collapse
|
17
|
Ma X, Bi Q, Kong Y, Xu H, Liang M, Mai K, Zhang Y. Dietary lipid levels affected antioxidative status, inflammation response, apoptosis and microbial community in the intestine of juvenile turbot (Scophthalmus maximus L.). Comp Biochem Physiol A Mol Integr Physiol 2021; 264:111118. [PMID: 34793954 DOI: 10.1016/j.cbpa.2021.111118] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/11/2021] [Accepted: 11/11/2021] [Indexed: 02/08/2023]
Abstract
A nine-week feeding trial was conducted to comprehensively investigate the effects of different levels of dietary lipid on intestinal physiology of juvenile turbot. Three diets with different lipid levels (8%, 12% and 16%) were formulated, which were designated as the low-lipid group (LL), medium-lipid group (ML) and high-lipid group (HL), respectively. Each diet was fed to six replicate tanks, and each tank was stocked with 35 fish. The results revealed that medium dietary lipid (12%) increased the activities of intestinal digestive enzymes and brush border enzymes. Excessive dietary lipid (16%) decreased the intestinal antioxidative enzyme levels and increased the lipid peroxidation pressure. In addition, HL stimulated the occurrence of intestinal inflammation and significantly up-regulated the mRNA expression level of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interferon-γ (IFN-γ) and transforming growth factor-β (TGF-β). Dietary LL and HL induced the apoptosis of intestinal epithelial cells. Sequencing of bacterial 16 s rRNA V4 region indicated that the abundance and diversity of intestinal microflora in fish fed with medium lipid diet (12%) were significantly higher than those in other groups, indicating the intestinal microflora ecology in group ML was more balanced. MetaStat analysis indicated that both low- and high-lipid diets significantly reduced the relative abundance of intestinal beneficial bacteria. In conclusion, results of this study demonstrated the sensitivity of intestinal health and microbiota to dietary lipid levels. From the perspective of microecological balance, medium dietary lipid (12%) was more conducive to maintaining the intestinal microflora stability of turbot.
Collapse
Affiliation(s)
- Xiuhua Ma
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture, Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, 5, Yushan Road, Qingdao 266003, China
| | - Qingzhu Bi
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106, Nanjing Road, Qingdao 266071, China
| | - Yaoyao Kong
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture, Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, 5, Yushan Road, Qingdao 266003, China
| | - Houguo Xu
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106, Nanjing Road, Qingdao 266071, China
| | - Mengqing Liang
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106, Nanjing Road, Qingdao 266071, China; Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao 266237, China.
| | - Kangsen Mai
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture, Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, 5, Yushan Road, Qingdao 266003, China; Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao 266237, China
| | - Yanjiao Zhang
- Key Laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture, Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, 5, Yushan Road, Qingdao 266003, China; Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao 266237, China.
| |
Collapse
|
18
|
Lei W, Liu C, Pan L, Peng C, Wang J, Zhou H. Screening of probiotic Lactobacilli with potential anti-allergic activity based on hyaluronidase inhibition and degranulation of RBL-2H3 cells in vitro. Lebensm Wiss Technol 2021. [DOI: 10.1016/j.lwt.2020.110707] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
19
|
Zheng Y, Fang Z, Xue Y, Zhang J, Zhu J, Gao R, Yao S, Ye Y, Wang S, Lin C, Chen S, Huang H, Hu L, Jiang GN, Qin H, Zhang P, Chen J, Ji H. Specific gut microbiome signature predicts the early-stage lung cancer. Gut Microbes 2020; 11:1030-1042. [PMID: 32240032 PMCID: PMC7524275 DOI: 10.1080/19490976.2020.1737487] [Citation(s) in RCA: 160] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Alterations of gut microbiota have been implicated in multiple diseases including cancer. However, the gut microbiota spectrum in lung cancer remains largely unknown. Here we profiled the gut microbiota composition in a discovery cohort containing 42 early-stage lung cancer patients and 65 healthy individuals through the 16S ribosomal RNA (rRNA) gene sequencing analysis. We found that lung cancer patients displayed a significant shift of microbiota composition in contrast to the healthy populations. To identify the optimal microbiota signature for noninvasive diagnosis purpose, we took advantage of Support-Vector Machine (SVM) and found that the predictive model with 13 operational taxonomic unit (OTU)-based biomarkers achieved a high accuracy in lung cancer prediction (area under curve, AUC = 97.6%). This signature performed reasonably well in the validation cohort (AUC = 76.4%), which contained 34 lung cancer patients and 40 healthy individuals. To facilitate potential clinical practice, we further constructed a 'patient discrimination index' (PDI), which largely retained the prediction efficiency in both the discovery cohort (AUC = 92.4%) and the validation cohort (AUC = 67.7%). Together, our study uncovered the microbiota spectrum of lung cancer patients and established the specific gut microbial signature for the potential prediction of the early-stage lung cancer.
Collapse
Affiliation(s)
- Yajuan Zheng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Zhaoyuan Fang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yun Xue
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Jian Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Junjie Zhu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Renyuan Gao
- Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shun Yao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yi Ye
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Shihui Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Changdong Lin
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Shiyang Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Hsinyi Huang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Liang Hu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Ge-Ning Jiang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Huanlong Qin
- Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Peng Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China,Peng Zhang Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai200433, China
| | - Jianfeng Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China,CONTACT Hongbin Ji ; JianFeng Chen Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 YueYang Road, Shanghai200031, China
| | - Hongbin Ji
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China,School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| |
Collapse
|
20
|
Yang X, Lu D, Zhuo J, Lin Z, Yang M, Xu X. The Gut-liver Axis in Immune Remodeling: New insight into Liver Diseases. Int J Biol Sci 2020; 16:2357-2366. [PMID: 32760203 PMCID: PMC7378637 DOI: 10.7150/ijbs.46405] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/12/2020] [Indexed: 02/07/2023] Open
Abstract
The gut microbiota consists of a dynamic multispecies community of bacteria, fungi, archaea, and protozoans, playing a fundamental role in the induction, training, and function of the host immune system. The liver is anatomically and physiologically linked to the gut microbiota via enterohepatic circulation, specifically receiving intestine-derived blood through the portal vein. The gut microbiota is crucial for maintaining immune homeostasis of the gut-liver axis. A shift in gut microbiota composition can result in activation of the mucosal immune response causing homeostasis imbalance. This imbalance results in translocation of bacteria and migration of immune cells to the liver, which is related to inflammation-mediated liver injury and tumor progression. In this review, we outline the role of the gut microbiota in modulating host immunity and summarize novel findings and recent advances in immune-based therapeutics associated with the gut-liver axis. Moving forward, a deep understanding of the microbiome-immune-liver axis will provide insight into the basic mechanisms of gut microbiota dysbiosis affecting liver diseases.
Collapse
Affiliation(s)
- Xinyu Yang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China.,NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Di Lu
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China.,NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Jianyong Zhuo
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China.,NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Zuyuan Lin
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China.,NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Modan Yang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China.,NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Xiao Xu
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China.,NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| |
Collapse
|
21
|
Fu L, Xie M, Wang C, Qian Y, Huang J, Sun Z, Zhang H, Wang Y. Lactobacillus Casei Zhang Alleviates Shrimp Tropomyosin-Induced Food Allergy by Switching Antibody Isotypes through the NF-κB-Dependent Immune Tolerance. Mol Nutr Food Res 2020; 64:e1900496. [PMID: 32243079 DOI: 10.1002/mnfr.201900496] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 02/29/2020] [Indexed: 12/28/2022]
Abstract
SCOPE Shellfish allergy is an important cause of food allergy, and tropomyosin (TM) is the major allergen within shellfish. Probiotics are safe bacteria that benefit host health and nutrition and is proposed as a novel approach for treating immunological diseases, including food allergies. METHODS AND RESULTS The probiotic strain Lactobacillus casei Zhang (LcZ) isolated from koumiss is investigated for its capacity to modulate food allergy induced by TM in BALB/c mice. Oral administration of LcZ attenuated allergy symptoms and intestinal epithelial damage. Furthermore, flow cytometry, real-time quantitative PCR, and ELISA demonstrated that LcZ administration altered the development and function of dendritic cells (DCs), T cells, and B cells, finally resulting in the change of TM-specific antibody isotypes into a tolerogenic pattern. Moreover, an in vitro spleen cell culture model reveals that LcZ directly modulates regulatory tolerogenic DC and T cell development, dependent on the activation of the nuclear factor kappa B (NF-κB) signaling pathway. CONCLUSION This work indicates the ability of LcZ to alleviate TM-induced food allergy and demonstrates the involvement of the tolerogenic immune cells and NF-κB signaling pathway, indicating LcZ to be a potential immunomodulator and immunotherapy assistor.
Collapse
Affiliation(s)
- Linglin Fu
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, P. R. China
| | - Menghua Xie
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, P. R. China
| | - Chong Wang
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, P. R. China
| | - Yi Qian
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, P. R. China
| | - Jianjian Huang
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, P. R. China
| | - Zhihong Sun
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, and Key Laboratory of Dairy Products Processing, Ministry of Agriculture, Inner Mongolia Agricultural University, Huhhot, 010018, P. R. China
| | - Heping Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, and Key Laboratory of Dairy Products Processing, Ministry of Agriculture, Inner Mongolia Agricultural University, Huhhot, 010018, P. R. China
| | - Yanbo Wang
- Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, 310018, P. R. China
| |
Collapse
|
22
|
Yong W, Zhang L, Chen Y, Li J, Liu Y, Zhang Z. Jianpi Huatan Tongfu granule alleviates inflammation and improves intestinal flora in patients with acute exacerbation of chronic obstructive pulmonary disease. J Int Med Res 2020; 48:300060520909235. [PMID: 32295442 PMCID: PMC7163244 DOI: 10.1177/0300060520909235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Objectives To investigate the clinical efficacy and mechanism of Jianpi Huatan Tongfu
granule in treating acute exacerbation of chronic obstructive pulmonary
disease (AECOPD). Methods Sixty patients with AECOPD were enrolled in either of two groups: integrative
treatment (Western medicine combined with Jianpi Huatan Tongfu granule)
(n = 30) and Western medical treatment (n = 30). Thirty healthy individuals
were included in the control group. Results Compared with healthy participants, patients with AECOPD had elevated
clinical symptom and dyspnea severity scores. Patients with AECOPD had
worsened lung function, compared with healthy participants. The therapeutic
efficacy for integrative treatment was superior to Western medical
treatment. Inflammatory proteins and cytokines were significantly elevated
in patients with AECOPD, including C-reactive protein, interleukin-6,
interleukin-8, and tumor necrosis factor-α; these were alleviated by both
treatments, with more obvious effects for integrative treatment. Integrative
treatment significantly changed the intestinal flora in patients with
AECOPD, reaching levels comparable with those of healthy participants.
Firmicutes abundance was significantly higher in
healthy participants, whereas Bacteroidetes abundance was
significantly higher in patients with AECOPD. After treatment,
Verrucomicrobia abundance was significantly reduced in
patients with AECOPD. Conclusion Jianpi Huatan Tongfu granule could alleviate inflammatory responses and
improve clinical therapeutic efficacy in patients with AECOPD.
Collapse
Affiliation(s)
- Wenxing Yong
- Department of Emergency, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Liying Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China.,Key Laboratory for Transfer of Dunhuang Medicine at the Provincial and Ministerial Level, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yuexuan Chen
- Department of Emergency, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China.,Department of Emergency, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Juan Li
- Department of Emergency, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Yongqi Liu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China.,Key Laboratory for Transfer of Dunhuang Medicine at the Provincial and Ministerial Level, Gansu University of Chinese Medicine, Lanzhou, China
| | - Zhiming Zhang
- Department of Emergency, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
23
|
Fu P, Bai L, Cai Z, Li R, Yung KKL. Fine particulate matter aggravates intestinal and brain injury and affects bacterial community structure of intestine and feces in Alzheimer's disease transgenic mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 192:110325. [PMID: 32078839 DOI: 10.1016/j.ecoenv.2020.110325] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 02/09/2020] [Accepted: 02/10/2020] [Indexed: 06/10/2023]
Abstract
Fine particulate matter (PM2.5) was a risk factor for neurological disorders when emerging studies revealed that PM2.5 affected the bacterial community structure of gut in Alzheimer's disease (AD) patients. The purpose of this study was to explore the effects of PM2.5 on intestinal and brain injury and on bacterial community structure in the intestine and feces of APP/PS1 transgenic mice exposed to PM2.5 for eight weeks with a real-world whole-body inhalation exposure system in Taiyuan, China. The brain and intestinal tissues were collected to evaluate histopathological changes by HE staining. TNF-α and IL-6 levels in intestines, brains, and serums, and Aβ-42 levels in brains were detected. Intestinal and fecal samples were subjected to 16S rRNA gene sequencing. Results showed that PM2.5 significantly aggravated the pathological injury in intestines and brains in AD mice with elevated pro-inflammatory cytokine levels. The estimators of Shannon, Simpson, Chao1, and ACE indexes reflected the diversity and richness of the bacterial community. Compared with the FA-WT group, the FA-AD group had lower diversity and richness when the PM2.5-AD group had the highest ones. PCA and NMDS revealed the specific influence of PM2.5 on the bacterial community of intestine and feces because that the PM2.5-FA and PM2.5-AD group clumped visibly closer than the other groups in both bacterial communities of intestine and feces. The KEGG pathway analysis predicted the vital functional genes and metabolic pathways in the bacterial community of PM2.5-AD mice. This study indicated the histopathological changes and inflammation in the intestine and brain were seriously caused in PM2.5-AD mice when the α-diversity of the bacterial community in intestine and feces was visibly changed.
Collapse
Affiliation(s)
- Pengfei Fu
- Department of Biology, Golden Meditech Center for NeuroRegeneration Sciences, Hong Kong Baptist University, Hong Kong Special Administrative Region
| | - Lirong Bai
- Institute of Environmental Science, Shanxi University, Taiyuan, PR China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong Special Administrative Region
| | - Ruijin Li
- Institute of Environmental Science, Shanxi University, Taiyuan, PR China.
| | - Ken Kin Lam Yung
- Department of Biology, Golden Meditech Center for NeuroRegeneration Sciences, Hong Kong Baptist University, Hong Kong Special Administrative Region; Institute of Environmental Science, Shanxi University, Taiyuan, PR China; State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong Special Administrative Region.
| |
Collapse
|
24
|
Turroni F, Milani C, Duranti S, Lugli GA, Bernasconi S, Margolles A, Di Pierro F, van Sinderen D, Ventura M. The infant gut microbiome as a microbial organ influencing host well-being. Ital J Pediatr 2020; 46:16. [PMID: 32024556 PMCID: PMC7003403 DOI: 10.1186/s13052-020-0781-0] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 01/29/2020] [Indexed: 12/16/2022] Open
Abstract
Initial establishment of the human gut microbiota is generally believed to occur immediately following birth, involving key gut commensals such as bifidobacteria that are acquired from the mother. The subsequent development of this early gut microbiota is driven and modulated by specific dietary compounds present in human milk that support selective colonization. This represents a very intriguing example of host-microbe co-evolution, where both partners are believed to benefit. In recent years, various publications have focused on dissecting microbial infant gut communities and their interaction with their human host, being a determining factor in host physiology and metabolic activities. Such studies have highlighted a reduction of microbial diversity and/or an aberrant microbiota composition, sometimes referred to as dysbiosis, which may manifest itself during the early stage of life, i.e., in infants, or later stages of life. There are growing experimental data that may explain how the early human gut microbiota affects risk factors related to adult health conditions. This concept has fueled the development of various nutritional strategies, many of which are based on probiotics and/or prebiotics, to shape the infant microbiota. In this review, we will present the current state of the art regarding the infant gut microbiota and the role of key commensal microorganisms like bifidobacteria in the establishment of the first microbial communities in the human gut.
Collapse
Affiliation(s)
- Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11a, 43124, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11a, 43124, Parma, Italy
| | - Sabrina Duranti
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11a, 43124, Parma, Italy
| | - Gabriele Andrea Lugli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11a, 43124, Parma, Italy
| | | | - Abelardo Margolles
- Departamento de Microbiologia y Bioquimica de Productos Lacteos, IPLA - CSIC, Villaviciosa, Asturias, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias-ISPA, Oviedo, Spain
| | | | - Douwe van Sinderen
- School of Microbiology & APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11a, 43124, Parma, Italy.
- Microbiome Research Hub, University of Parma, Parma, Italy.
| |
Collapse
|
25
|
Cheng R, Guo J, Pu F, Wan C, Shi L, Li H, Yang Y, Huang C, Li M, He F. Loading ceftriaxone, vancomycin, and Bifidobacteria bifidum TMC3115 to neonatal mice could differently and consequently affect intestinal microbiota and immunity in adulthood. Sci Rep 2019; 9:3254. [PMID: 30824845 PMCID: PMC6397183 DOI: 10.1038/s41598-018-35737-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 09/18/2018] [Indexed: 02/05/2023] Open
Abstract
Recent studies have demonstrated that antibiotics/or probiotics administration in early life play key roles on modulating intestinal microbiota and the alterations might cause long-lasting consequences both physiologically and immunologically. We investigated the effects of early life ceftriaxone, vancomycin and Bifidobacterium bifidum TMC3115 (TMC3115) treatment on intestinal microbiota and immunity both in neonates and adults even after termination of antibiotics exposure. We found that ceftriaxone and vancomycin, but not TMC3115, significantly altered the intestinal microbiota, serum total IgE level, and the morphology and function of the intestinal epithelium in the neonatal mice. In the adult stages, the diversity and composition of the intestinal microbiota were significantly different in the antibiotic-treated mice, and ceftriaxone-treated mice exhibited significantly higher serum total IgE and OVA-specific IgE levels. TMC3115 significantly mitigated the alteration of intestinal microbiota caused by ceftriaxone not vancomycin. Antibiotics and TMC3115 can differently modulate intestinal microbiota and SCFAs metabolism, affecting the development and function of the immunity and intestinal epithelium to different degrees in neonatal mice. Neonatal ceftriaxone-induced abnormal intestinal microbiota, immunity and epithelium could last to adulthood partly, which might be associated with the enhancement of host susceptibility to IgE-mediated allergies and related immune responses, TMC3115 may protect against the side effects of antibiotic treatment, at least partly.
Collapse
Affiliation(s)
- RuYue Cheng
- Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health and Healthy Food Evaluation Research Center, Sichuan University, 610041, Chengdu, Sichuan, PR China
| | - JiaWen Guo
- Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health and Healthy Food Evaluation Research Center, Sichuan University, 610041, Chengdu, Sichuan, PR China
| | - FangFang Pu
- Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health and Healthy Food Evaluation Research Center, Sichuan University, 610041, Chengdu, Sichuan, PR China
| | - ChaoMin Wan
- Department of Pediatrics of Western China Second Hospital of Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children, 610041, Chengdu, Sichuan, PR China
| | - Lei Shi
- Department of Clinical Nutrition, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, PR China
| | - HuaWen Li
- Hebei Inatural Biotech Co., Ltd, 050000, Shijiazhuang, Hebei, PR China
| | - YuHong Yang
- Hebei Inatural Biotech Co., Ltd, 050000, Shijiazhuang, Hebei, PR China
| | - ChengYu Huang
- Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health and Healthy Food Evaluation Research Center, Sichuan University, 610041, Chengdu, Sichuan, PR China
| | - Ming Li
- Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health and Healthy Food Evaluation Research Center, Sichuan University, 610041, Chengdu, Sichuan, PR China.
| | - Fang He
- Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health and Healthy Food Evaluation Research Center, Sichuan University, 610041, Chengdu, Sichuan, PR China.
| |
Collapse
|
26
|
Immunomodulatory effects of probiotics: Can they be used to treat allergies and autoimmune diseases? Maturitas 2018; 119:25-38. [PMID: 30502748 DOI: 10.1016/j.maturitas.2018.11.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 11/06/2018] [Accepted: 11/08/2018] [Indexed: 12/12/2022]
Abstract
As a person ages, physiological, immunological and gut microbiome changes collectively result in an array of chronic conditions. According to the 'hygiene hypothesis' the increasing prevalence of immune-mediated disorders may be related to intestinal dysbiosis, leading to immune dysfunction and associated conditions such as eczema, asthma, allergies and autoimmune diseases. Beneficial probiotic bacteria can be utilized by increasing their abundance within the gastrointestinal lumen, which in turn will modulate immune cells, such as, T helper (Th)-1, Th2, Th17, regulatory T (Treg) cells and B cells, which have direct relevance to human health and the pathogenesis of immune disorders. Here, we describe the cross-talk between probiotics and the gastrointestinal immune system, and their effects in relation to inflammatory bowel disease, multiple sclerosis, allergies and atopic dermatitis.
Collapse
|
27
|
Role of novel synbiotic supplementation on ovalbumin induced allergy model: Impact on immunemodulation. ACTA ACUST UNITED AC 2018. [DOI: 10.1016/j.bcdf.2018.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
28
|
Ren J, Zhao Y, Huang S, Lv D, Yang F, Lou L, Zheng Y, Zhang J, Liu S, Zhang N, Bachert C. Immunomodulatory effect of Bifidobacterium breve on experimental allergic rhinitis in BALB/c mice. Exp Ther Med 2018; 16:3996-4004. [PMID: 30344677 PMCID: PMC6176161 DOI: 10.3892/etm.2018.6704] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 05/17/2018] [Indexed: 02/05/2023] Open
Abstract
Bifidobacterium breve (B. breve) may have a beneficial effect on allergic rhinitis (AR). The aim of the present study was to investigate whether microbial induction of regulatory T cells (Tregs) and adjustment of Th1 and Th2 responses by B. breve are associated with protection against allergic inflammation, and to identify a dose-response association in a murine AR model. Ovalbumin (OVA)-sensitized BALB/c mice were orally treated with different doses of B. breve [1010, 109, 107 and 105 colony forming units (CFU)]. Following nasal challenge with OVA, sneeze frequency, serum OVA-specific immunoglobulin E (IgE) and cytokine concentrations [interleukin (IL)-4, IL-10, IL-13 and interferon-γ], splenic percentage of cluster of differentiation (CD)4+CD25+ Tregs, and morphology of the nasal mucosa were examined. Oral treatment with live B. breve at doses of 107 CFU or higher alleviated nasal mucosal injury and suppressed sneezing upon repeated administration over a 6-week period. Furthermore, treatment with B. breve at these higher doses reduced the concentrations of serum OVA-specific IgE, IL-4 and IL-10, and increased the splenic percentage of CD4+CD25+ Tregs in rhinitic mice compared with those who did not receive probiotics. In contrast, treatment with B. breve at a lower dose did not indicate any effect on sneezing frequency or mucosal morphology in this animal model, even though the splenic percentage of CD4+CD25+ Tregs increased and the concentrations of serum OVA-specific IgE and IL-10 declined. B. breve exerts its anti-allergic effects by inhibiting type 2 helper T cell immune responses and enhancing CD4+CD25+ Treg activity. Sneezing was also reduced at a dose of 107 CFU or higher. The current study investigated the role of B. breve and aided in identifying the optimal dose of B. breve administration in the treatment of AR.
Collapse
Affiliation(s)
- Jianjun Ren
- Department of Otorhinolaryngology, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yu Zhao
- Department of Otorhinolaryngology, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Shi Huang
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610000, P.R. China
| | - Dan Lv
- Department of Otorhinolaryngology, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Fengling Yang
- Department of Otorhinolaryngology, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Lin Lou
- Department of Otorhinolaryngology, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yongbo Zheng
- Department of Otorhinolaryngology, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jie Zhang
- Upper Airways Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Shixi Liu
- Upper Airways Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Nan Zhang
- Upper Airways Research Laboratory, Department of Otorhinolaryngology, Ghent University, 9000 Ghent, Belgium
| | - Claus Bachert
- Upper Airways Research Laboratory, Department of Otorhinolaryngology, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
29
|
Killed Bifidobacterium longum enhanced stress tolerance and prolonged life span of Caenorhabditis elegans via DAF-16. Br J Nutr 2018; 120:872-880. [PMID: 30178731 DOI: 10.1017/s0007114518001563] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Probiotics are bacteria among the intestinal flora that are beneficial for human health. Bifidobacterium longum (BL) is a prototypical probiotic that is widely used in yogurt making, supplements and others. Although various physiological effects of BL have been reported, those associated with longevity and anti-ageing still remain elusive. Here we aimed to elucidate the physiological effects of killed BL (BR-108) on stress tolerance and longevity of Caenorhabditis elegans and their mechanisms. Worms fed killed BL in addition to Escherichia coli (OP50) displayed reduced body length in a BL dose-dependent manner. When compared with those fed E. coli alone, these worms had a higher survival rate following heat stress at 35°C and hydrogen peroxide-induced oxidative stress. A general decrease in motility was observed over time in all worms; however, killed BL-fed ageing worms displayed increased movement and longer life span than those fed E. coli alone. However, the longevity effect was suppressed in sir-2.1, daf-16 and skn-1-deficient worms. Killed BL induced DAF-16 nuclear localisation and increased the expression of the DAF-16 target gene hsp-12.6. These results revealed that the physiological effects of killed BL in C. elegans were mediated by DAF-16 activation. These findings contradict previous observations with different Bifidobacterium and Lactobacillus strains, which showed the role for SKN-1 independently of DAF-16.
Collapse
|
30
|
Cheng RY, Yao JR, Wan Q, Guo JW, Pu FF, Shi L, Hu W, Yang YH, Li L, Li M, He F. Oral administration of Bifidobacterium bifidum TMC3115 to neonatal mice may alleviate IgE-mediated allergic risk in adulthood. Benef Microbes 2018; 9:815-828. [PMID: 29888657 DOI: 10.3920/bm2018.0005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
This study aimed to demonstrate whether exposure to bifidobacteria during early life influences immunity and alleviates the risk of immunoglobulin E (IgE)-mediated allergies in adulthood. BALB/c neonatal mice (n=54) were administered with a lyophilised cell preparation of Bifidobacterium bifidum TMC3115 (TMC3115) for 3 weeks. Following the intervention, the mice were immunised with intraperitoneal ovalbumin (OVA). The morphology and function of the intestinal epithelium were determined using histopathological examinations. Intestinal microbiota was detected using quantitative PCR and characterised using next-generation sequencing of 16S rRNA genes from faecal DNA. Caecal short-chain fatty acids (SCFAs) were measured using gas chromatography-mass spectrometry. Serum levels of tumour necrosis factor (TNF)-α, interleukin (IL)-6, IL-10, and immunoglobulin E (IgE) and the percentage of splenic CD4+ T cells were examined using enzyme-linked immunosorbent assay and flow cytometry, respectively. TMC3115 did not significantly affect body weight, and cause any severe systemic inflammation or other clinical symptoms among the neonatal or adult mice, although the crypt depths and Muc2-positive cells in some intestinal segments of neonatal mice were significantly lower than control. Oral TMC3115 administration significantly increased faecal microbial diversity, relative abundance of Bacteroidetes and caecal SCFAs production in neonatal mice. Following the intervention, neonatal mice treated with TMC3115 exhibited less increase in serum IgE levels induced by OVA in adults and significantly higher TNF-α and IL-10 levels than in control. Our findings indicate that the oral administration of bifidobacteria, particularly certain strains, such as TMC3115, during early life could alleviate the risk of IgE-mediated allergies in adult host animals. Modifications of intestinal microbiota, SCFAs metabolism and anti-inflammatory cytokine IL-10 production by bifidobacteria may at least in part be a key mechanism underlying the effect of bifidobacteria on the IgE-mediated immune sensitivity of hosts to attacks by allergens at both neonatal and adult stages.
Collapse
Affiliation(s)
- R Y Cheng
- 1 Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health, Sichuan University and Healthy Food Evaluation Research Center, Chengdu 610041, Sichuan, China P.R
| | - J R Yao
- 2 West China Second University Hospital/Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Sichuan University, Chengdu 610041, Sichuan, China P.R
| | - Q Wan
- 1 Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health, Sichuan University and Healthy Food Evaluation Research Center, Chengdu 610041, Sichuan, China P.R
| | - J W Guo
- 1 Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health, Sichuan University and Healthy Food Evaluation Research Center, Chengdu 610041, Sichuan, China P.R
| | - F F Pu
- 1 Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health, Sichuan University and Healthy Food Evaluation Research Center, Chengdu 610041, Sichuan, China P.R
| | - L Shi
- 3 Department of Clinical Nutrition, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China P.R
| | - W Hu
- 3 Department of Clinical Nutrition, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China P.R
| | - Y H Yang
- 4 Department of Planning, Hebei Inatural Biotech Co., Ltd. Shijiazhuang 050000, Hebei, China P.R
| | - L Li
- 5 Department of Research and Development, Hebei Inatural Biotech Co., Ltd. Shijiazhuang 050000, Hebei, China P.R
| | - M Li
- 1 Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health, Sichuan University and Healthy Food Evaluation Research Center, Chengdu 610041, Sichuan, China P.R
| | - F He
- 1 Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health, Sichuan University and Healthy Food Evaluation Research Center, Chengdu 610041, Sichuan, China P.R
| |
Collapse
|
31
|
Human Milk and Allergic Diseases: An Unsolved Puzzle. Nutrients 2017; 9:nu9080894. [PMID: 28817095 PMCID: PMC5579687 DOI: 10.3390/nu9080894] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 08/01/2017] [Accepted: 08/01/2017] [Indexed: 02/07/2023] Open
Abstract
There is conflicting evidence on the protective role of breastfeeding in relation to the development of allergic sensitisation and allergic disease. Studies vary in methodology and definition of outcomes, which lead to considerable heterogeneity. Human milk composition varies both within and between individuals, which may partially explain conflicting data. It is known that human milk composition is very complex and contains variable levels of immune active molecules, oligosaccharides, metabolites, vitamins and other nutrients and microbial content. Existing evidence suggests that modulation of human breast milk composition has potential for preventing allergic diseases in early life. In this review, we discuss associations between breastfeeding/human milk composition and allergy development.
Collapse
|
32
|
Strzępa A, Majewska-Szczepanik M, Lobo FM, Wen L, Szczepanik M. Broad spectrum antibiotic enrofloxacin modulates contact sensitivity through gut microbiota in a murine model. J Allergy Clin Immunol 2017; 140:121-133.e3. [DOI: 10.1016/j.jaci.2016.11.052] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 11/10/2016] [Accepted: 11/23/2016] [Indexed: 12/24/2022]
|
33
|
Huang CH, Wang CC, Lin YC, Hori M, Jan TR. Oral administration with diosgenin enhances the induction of intestinal T helper 1-like regulatory T cells in a murine model of food allergy. Int Immunopharmacol 2016; 42:59-66. [PMID: 27886644 DOI: 10.1016/j.intimp.2016.11.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 11/08/2016] [Accepted: 11/21/2016] [Indexed: 02/02/2023]
Abstract
Although the development of T helper (Th)1-like regulatory T (Treg) cells under Th1 inflammatory conditions has been reported, the role of Th1-like Treg cells in Th2 allergic responses remains mostly unclear. We previously demonstrated that diosgenin, the major sapogenin contained in the Chinese yam, attenuated food allergy and augmented Th1 and Treg immune responses. In this study, we hypothesized that diosgenin may enhance the induction of Th1-like Treg cells in the gut of mice with food allergy. Ovalbumin (OVA)-sensitized BALB/c mice were gavaged daily with diosgenin and received repeatedly intragastric ovalbumin challenges to induce intestinal allergic responses. The induction of Foxp3+ Treg cells co-expressing Th1-type transcription factors, cytokines and chemokines in the intestine was examined, and the mRNA expression of the chemokines corresponding to Th1-like Treg cells was measured. Diosgenin administration increased the number of Foxp3+ Treg cells co-expressing Th1 markers, including CCR5, CXCR3, IFN-γ and T-bet in the intestine, and enhanced populations of Foxp3+IFN-γ+ and Foxp3+T-bet+ cells that expressed the regulatory cytokine IL-10 in the Peyer's patches. Diosgenin also augmented the intestinal expression of CXCR3, CCL3, and CXCL10. Concordantly, diosgenin increased the number of CXCR3+Foxp3+IL-10 cells in the Peyer's patches. Our data demonstrated the enhanced induction of Th1-like Treg cells in allergic mice treated with diosgenin, providing evidence to suggest a role for Th1-like Treg cells in diosgenin-mediated anti-allergic effects against Th2-type allergy.
Collapse
Affiliation(s)
- Chung-Hsiung Huang
- Department and Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan; National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Chia-Chi Wang
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu-Chin Lin
- Department of Medicinal Botanicals and Health Applications, College of Biotechnology & Bio-resources, Dayeh University, Changhua, Taiwan
| | - Masatoshi Hori
- Department of Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Tong-Rong Jan
- Department and Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
34
|
Milani C, Ferrario C, Turroni F, Duranti S, Mangifesta M, van Sinderen D, Ventura M. The human gut microbiota and its interactive connections to diet. J Hum Nutr Diet 2016; 29:539-46. [PMID: 27161433 DOI: 10.1111/jhn.12371] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The microbiota of the gastrointestinal tract plays an important role in human health. In addition to their metabolic interactions with dietary constituents, gut bacteria may also be involved in more complex host interactions, such as modulation of the immune system. Furthermore, the composition of the gut microbiota may be important in reducing the risk of contracting particular gut infections. Changes in the microbiota during an individual's lifespan are accompanied by modifications in multiple health parameters, and such observations have prompted intense scientific efforts aiming to understand the complex interactions between the microbiota and its human host, as well as how this may be influenced by diet.
Collapse
Affiliation(s)
- C Milani
- Laboratory of Probiogenomics, Department of Life Sciences, University of Parma, Parma, Italy
| | - C Ferrario
- Laboratory of Probiogenomics, Department of Life Sciences, University of Parma, Parma, Italy
| | - F Turroni
- Laboratory of Probiogenomics, Department of Life Sciences, University of Parma, Parma, Italy
| | - S Duranti
- Laboratory of Probiogenomics, Department of Life Sciences, University of Parma, Parma, Italy
| | | | - D van Sinderen
- APC Microbiome Institute and School of Microbiology, National University of Ireland, Cork, Ireland
| | - M Ventura
- Laboratory of Probiogenomics, Department of Life Sciences, University of Parma, Parma, Italy.
| |
Collapse
|
35
|
Keely S, Walker MM, Marks E, Talley NJ. Immune dysregulation in the functional gastrointestinal disorders. Eur J Clin Invest 2015; 45:1350-9. [PMID: 26444549 DOI: 10.1111/eci.12548] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 10/03/2015] [Indexed: 12/14/2022]
Abstract
Gastrointestinal conditions may be broadly classified into two: organic and functional disease, with functional disorders accounting for the majority of patients with chronic gastrointestinal symptoms. Functional gastrointestinal disorders (FGIDs) present with no obvious pathology or well-accepted biochemical mechanism and, as such, treatment strategies are limited and focus on symptoms rather than cure. Irritable bowel syndrome and functional dyspepsia are the most widely recognised FGIDs, and there is a growing body of evidence to suggest an underlying inflammatory phenotype in subsets with these conditions. Here, we discuss the current knowledge of immune involvement in FGIDs and the commonalities between the different manifestations of FGIDs and propose a new hypothesis, potentially defining an underlying immunopathological basis of these conditions.
Collapse
Affiliation(s)
- Simon Keely
- School of Biomedical Sciences & Pharmacy, University of Newcastle & Vaccine and Asthma (VIVA) Program, Hunter Medical Research Institute, Callaghan, NSW, Australia
| | - Marjorie M Walker
- School of Medicine & Public Health, University of Newcastle, Callaghan, NSW, Australia
| | - Ellen Marks
- School of Biomedical Sciences & Pharmacy, University of Newcastle & Vaccine and Asthma (VIVA) Program, Hunter Medical Research Institute, Callaghan, NSW, Australia
| | - Nicholas J Talley
- School of Medicine & Public Health, University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
36
|
Fong FLY, Shah NP, Kirjavainen P, El-Nezami H. Mechanism of Action of Probiotic Bacteria on Intestinal and Systemic Immunities and Antigen-Presenting Cells. Int Rev Immunol 2015; 35:179-88. [PMID: 26606641 DOI: 10.3109/08830185.2015.1096937] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Immunomodulation has been shown to be one of the major functions of probiotic bacteria. This review is presented to provide detailed information on the immunomodulatory properties of probiotics in various animal models and clinical practices. Probiotics can regulate helper T (Th) responses and release of cytokines in a strain-specific manner. For example, Lactobacillus rhamnosus GG can induce beneficial Th1 immunomodulatory effect in infants with cow's milk allergy and relieve intestinal inflammation in atopic children by promoting IL-10 generation. Mechanism of action of probiotics on antigen-presenting cells at gastrointestinal tract is also postulated in this review. Probiotic bacterial cells and their soluble factors may activate dendritic cells, macrophages, and to certain extent monocytes via toll-like-receptor recognition and may further provoke specific Th responses. They are speculated to elicit immunomodulatory effects on intestinal and systemic immunities.
Collapse
Affiliation(s)
- Fiona Long Yan Fong
- a School of Biological Sciences , The University of Hong Kong , Hong Kong.,b Present address: School of Science and Technology , The Open University of Hong Kong >, Hong Kong
| | - Nagendra P Shah
- a School of Biological Sciences , The University of Hong Kong , Hong Kong
| | - Pirkka Kirjavainen
- b Present address: School of Science and Technology , The Open University of Hong Kong >, Hong Kong.,c Present address: Department of Environmental Health , National Institute for Health and Welfare , Kuopio , Finland
| | - Hani El-Nezami
- a School of Biological Sciences , The University of Hong Kong , Hong Kong.,d Institute of Public Health and Clinical Nutrition , University of Easter Finland , Kuopio , Finland
| |
Collapse
|
37
|
Oberg TS, Ward RE, Steele JL, Broadbent JR. Transcriptome analysis of Bifidobacterium longum strains that show a differential response to hydrogen peroxide stress. J Biotechnol 2015; 212:58-64. [PMID: 26299205 DOI: 10.1016/j.jbiotec.2015.06.405] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Revised: 06/16/2015] [Accepted: 06/19/2015] [Indexed: 01/02/2023]
Abstract
Consumer and commercial interest in foods containing probiotic bifidobacteria is increasing. However, because bifidobacteria are anaerobic, oxidative stress can diminish cell viability during production and storage of bioactive foods. We previously found Bifidobacterium longum strain NCC2705 had significantly greater intrinsic and inducible resistance to hydrogen peroxide (H2O2) than strain D2957. Here, we explored the basis for these differences by examining the transcriptional responses of both strains to sub-lethal H2O2 exposure for 5- or 60-min. Strain NCC2705 had 288 genes that were differentially expressed after the 5-min treatment and 114 differentially expressed genes after the 60-min treatment. In contrast, strain D2957 had only 21 and 90 differentially expressed genes after the 5- and 60-min treatments, respectively. Both strains showed up-regulation of genes coding enzymes implicated in oxidative stress resistance, such as thioredoxin, thioredoxin reductase, peroxiredoxin, ferredoxin, glutaredoxin, and anaerobic ribonucleotide reductase, but induction levels were typically highest in NCC2705. Compared to D2957, NCC2705 also had more up-regulated genes involved in transcriptional regulation and more down-regulated genes involved in sugar transport and metabolism. These results provide a greater understanding of the molecular basis for oxidative stress resistance in B. longum and the factors that contribute to strain-to-strain variability in survival in bioactive food products.
Collapse
Affiliation(s)
- Taylor S Oberg
- Department of Nutrition, Dietetics and Food Sciences, Utah State University, 8700 Old Main Hill, Logan, UT 84322-8700, USA.
| | - Robert E Ward
- Department of Nutrition, Dietetics and Food Sciences, Utah State University, 8700 Old Main Hill, Logan, UT 84322-8700, USA
| | - James L Steele
- Department of Food Science, University of Wisconsin, 1605 Linden Drive, Madison, WI 53706-1565, USA
| | - Jeff R Broadbent
- Department of Nutrition, Dietetics and Food Sciences, Utah State University, 8700 Old Main Hill, Logan, UT 84322-8700, USA
| |
Collapse
|
38
|
Tumor-specific Th2 responses inhibit growth of CT26 colon-cancer cells in mice via converting intratumor regulatory T cells to Th9 cells. Sci Rep 2015; 5:10665. [PMID: 26035423 PMCID: PMC4451845 DOI: 10.1038/srep10665] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 04/22/2015] [Indexed: 02/01/2023] Open
Abstract
The abnormality of immune regulation plays a critical role in the pathogenesis of cancer; the underlying mechanism has not been fully understood yet. This study aims to investigate the role of cancer specific T helper (Th)2 response in the inhibition of colon cancer (Cca) cell growth. The results showed that with Cca cell (CT26 cell) extracts as an antigen, the Cca-extract specific Th2 response was induced in the Cca-bearing mice. The Cca mass size was significantly reduced, or radically disappeared (5 out of 10; or 50%); the survival rate was markedly improved in mice immunized with Cca-extract, but not in those immunized with another tumor cell (U87 cell) extracts or to bovine serum albumin. The immunization with Cca-extract also induced Cca cell apoptosis and converted the intra-Cca Tregs to T helper (Th) 9 cells. In conclusion, Cca-specific Th2 responses inhibit Cca growth in a mouse model via inducing Cca cell apoptosis and converting intra-Cca Tregs to Th9 cells.
Collapse
|
39
|
Abstract
The science of food allergy has been rapidly evolving before our eyes in the past half century. Like other allergic disorders, the prevalence of food allergies has dramatically increased, and coupled with the increased public awareness of anaphylaxis due to food allergy, this has driven an explosion in basic and clinical research in this extremely broad subject. Treatment of food allergies has evolved and practices such as food challenges have become an integral part of an allergy practice. The impact of the increase of food allergy has driven package labeling laws, legislation on emergency treatment availability in schools and other public places, and school policy. But to this day, our knowledge of the pathogenesis of food allergy is still incomplete. There are the most obvious IgE-mediated immediate hypersensitivity reactions, but then multiple previously unidentified conditions such as eosinophilic esophagitis, food protein-induced enterocolitis syndrome, milk protein allergy, food-induced atopic dermatitis, oral allergy syndrome, and others have complicated the diagnosis and management of many of our patients who are unable to tolerate certain foods. Many of these conditions are not IgE-mediated, but may be T cell-driven diseases. The role of T regulatory cells and immune tolerance and the newly discovered immunological role of vitamin D have shed light on the variable clinical presentation of food allergy and the development of new methods of immunotherapy in an example of bench-to-bedside research. Component-resolved diagnostic techniques have already begun to allow us to more precisely define the epitopes that are targeted in food allergic patients. The development of biological modulators, research on genomics and proteomics, and epigenetic techniques all offer promising avenues for new modes of therapy of food allergy in the twenty-first century.
Collapse
Affiliation(s)
- Patrick S C Leung
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, 451 Health Sciences Drive, Suite 6510, Davis, CA, 95616, USA,
| | | | | |
Collapse
|
40
|
Abstract
The human fetus lives in a germ-free intrauterine environment and enters the outside world containing microorganisms from several sources, resulting in gut colonization. Full-term, vaginally born infants are completely colonized with a diverse array of bacterial families in clusters (Phyla) and species (>1000) by the first year of life. Colonizing bacteria communicating with the gut epithelium and underlying lymphoid tissues ('bacterial-epithelial crosstalk') result in a functional immune phenotype and no expression of disease (immune homeostasis). Appropriate colonization is influenced by the prebiotic effect of breast milk oligosaccharides. Adequate colonization results in an innate and adaptive mucosal immune phenotype via communication between molecular patterns on colonizing bacteria and pattern-recognition receptors (e.g., toll-like receptors) on epithelial and lymphoid cells. This ontogeny affects the immune system's capacity to develop oral tolerance to innocuous bacteria and benign antigens. Inadequate intestinal colonization with premature delivery, delivery by Cesarean section and excessive use of perinatal antibiotics results in the absence of adequate bacterial-epithelial crosstalk and an increased incidence of immune-mediated diseases [e.g., asthma, allergy in general and necrotizing enterocolitis (NEC)]. Fortunately, infants with inadequate intestinal colonization can be restored to a bacterial balance with the intake of probiotics. This has been shown to prevent debilitating diseases such as NEC. Thus, understanding the role of gut microbiota in programming of the immune phenotype may be important in preventing disease expression in later childhood and adulthood.
Collapse
|
41
|
Kemgang TS, Kapila S, Shanmugam VP, Kapila R. Cross-talk between probiotic lactobacilli and host immune system. J Appl Microbiol 2014; 117:303-19. [PMID: 24738909 DOI: 10.1111/jam.12521] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 04/02/2014] [Accepted: 04/08/2014] [Indexed: 12/14/2022]
Abstract
The mechanism by which probiotic lactobacilli affect the immune system is strain specific. As the immune system is a multicompartmental system, each strain has its way to interact with it and induce a visible and quantifiable effect. This review summarizes the interplay existing between the host immune system and probiotic lactobacilli, that is, with emphasis on lactobacilli as a prototype probiotic genus. Several aspects including the bacterial-host cross-talk with the mucosal and systemic immune system are presented, as well as short sections on the competing effect towards pathogenic bacteria and their uses as delivery vehicle for antigens.
Collapse
Affiliation(s)
- T S Kemgang
- Department of Animal Biochemistry, National Dairy Research Institute, Karnal, Haryana, India; Department of Food Science/Nutrition, National School of AgroIndustrial Sciences, University of Ngaoundere, Ngaoundere, Adamaoua, Cameroon
| | | | | | | |
Collapse
|
42
|
de Kivit S, Tobin MC, Forsyth CB, Keshavarzian A, Landay AL. Regulation of Intestinal Immune Responses through TLR Activation: Implications for Pro- and Prebiotics. Front Immunol 2014; 5:60. [PMID: 24600450 PMCID: PMC3927311 DOI: 10.3389/fimmu.2014.00060] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 02/03/2014] [Indexed: 12/12/2022] Open
Abstract
The intestinal mucosa is constantly facing a high load of antigens including bacterial antigens derived from the microbiota and food. Despite this, the immune cells present in the gastrointestinal tract do not initiate a pro-inflammatory immune response. Toll-like receptors (TLRs) are pattern recognition receptors expressed by various cells in the gastrointestinal tract, including intestinal epithelial cells (IEC) and resident immune cells in the lamina propria. Many diseases, including chronic intestinal inflammation (e.g., inflammatory bowel disease), irritable bowel syndrome (IBS), allergic gastroenteritis (e.g., eosinophilic gastroenteritis and allergic IBS), and infections are nowadays associated with a deregulated microbiota. The microbiota may directly interact with TLR. In addition, differences in intestinal TLR expression in health and disease may suggest that TLRs play an essential role in disease pathogenesis and may be novel targets for therapy. TLR signaling in the gut is involved in either maintaining intestinal homeostasis or the induction of an inflammatory response. This mini review provides an overview of the current knowledge regarding the contribution of intestinal epithelial TLR signaling in both tolerance induction or promoting intestinal inflammation, with a focus on food allergy. We will also highlight a potential role of the microbiota in regulating gut immune responses, especially through TLR activation.
Collapse
Affiliation(s)
- Sander de Kivit
- Division of Digestive Diseases and Nutrition, Rush University Medical Center , Chicago, IL , USA
| | - Mary C Tobin
- Department of Immunology/Microbiology, Rush University Medical Center , Chicago, IL , USA
| | - Christopher B Forsyth
- Division of Digestive Diseases and Nutrition, Rush University Medical Center , Chicago, IL , USA
| | - Ali Keshavarzian
- Division of Digestive Diseases and Nutrition, Rush University Medical Center , Chicago, IL , USA ; Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University , Utrecht , Netherlands
| | - Alan L Landay
- Department of Immunology/Microbiology, Rush University Medical Center , Chicago, IL , USA ; Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University , Utrecht , Netherlands
| |
Collapse
|
43
|
Kim HJ, Kim HY, Lee SY, Seo JH, Lee E, Hong SJ. Clinical efficacy and mechanism of probiotics in allergic diseases. KOREAN JOURNAL OF PEDIATRICS 2013; 56:369-76. [PMID: 24223597 PMCID: PMC3819679 DOI: 10.3345/kjp.2013.56.9.369] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 06/11/2013] [Indexed: 12/18/2022]
Abstract
A complex interplay between genetic and environmental factors partially contributes to the development of allergic diseases by affecting development during prenatal and early life. To explain the dramatic increase in the prevalence of allergic diseases, the hygiene hypothesis proposed that early exposure to infection prevented allergic diseases. The hygiene hypothesis has changed to the microbial hypothesis, in which exposure to microbes is closely linked to the development of the early immune system and allergic diseases. The intestinal flora may contribute to allergic disease through its substantial effect on mucosal immunity. Based on findings that exposure to microbial flora early in life can change the Th1/Th2 balance, thus favoring a Th1 cell response, probiotics may be beneficial in preventing allergic diseases. However, evidence from clinical and basic research to prove the efficacy of probiotics in preventing allergy is lacking. To date, studies have yielded inconsistent findings on the usefulness of probiotics in allergic diseases. It is difficult to demonstrate an exact effect of probiotics on asthma, allergic rhinitis, and food allergy because of study limitations, such as different first supplementation period, duration, different strains, short follow-up period, and host factors. However, many studies have demonstrated a significant clinical improvement in atopic dermatitis with the use of probiotics. An accurate understanding of the development of human immunity, intestinal barrier function, intestinal microbiota, and systemic immunity is required to comprehend the effects of probiotics on allergic diseases.
Collapse
Affiliation(s)
- Ha-Jung Kim
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
44
|
Schwarzer M, Srutkova D, Schabussova I, Hudcovic T, Akgün J, Wiedermann U, Kozakova H. Neonatal colonization of germ-free mice with Bifidobacterium longum prevents allergic sensitization to major birch pollen allergen Bet v 1. Vaccine 2013; 31:5405-12. [PMID: 24055352 DOI: 10.1016/j.vaccine.2013.09.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 08/19/2013] [Accepted: 09/06/2013] [Indexed: 12/19/2022]
Abstract
The main goal in reversing the allergy epidemic is the development of effective prophylactic strategies. We investigated the prophylactic effect of neonatal mother-to-offspring mono-colonization with Bifidobacterium longum ssp. longum CCM 7952 on subsequent allergic sensitization. Adult male and female germ-free (GF) mice were mono-colonized with B. longum, mated and their offspring, as well as age-matched GF controls, were sensitized with the major birch pollen allergen Bet v 1. Furthermore, signaling pathways involved in the recognition of B. longum were investigated in vitro. Neonatal mono-colonization of GF mice with B. longum suppressed Bet v 1-specific IgE-dependent β-hexosaminidase release as well as levels of total IgE and allergen-specific IgG2a in serum compared to sensitized GF controls. Accordingly, Bet v 1-induced production of both Th1- and Th2-associated cytokines in spleen cell cultures was significantly reduced in these mice. The general suppression of Bet v 1-specific immune responses in B. longum-colonized mice was associated with increased levels of regulatory cytokines IL-10 and TGF-β in serum. In vitro, B. longum induced low maturation status of bone marrow-derived dendritic cells and production of IL-10 in TLR2-, MyD88-, and MAPK-dependent manner. Our data demonstrate that neonatal mono-colonization with B. longum reduces allergic sensitization, likely by activation of regulatory responses via TLR2, MyD88, and MAPK signaling pathways. Thus, B. longum might be a promising candidate for perinatal intervention strategies against the onset of allergic diseases in humans.
Collapse
Affiliation(s)
- Martin Schwarzer
- Department of Immunology and Gnotobiology, Institute of Microbiology of the Academy of Sciences of the Czech Republic, v. v. i., Novy Hradek, Czech Republic.
| | | | | | | | | | | | | |
Collapse
|
45
|
Genetic and physiological responses of Bifidobacterium animalis subsp. lactis to hydrogen peroxide stress. J Bacteriol 2013; 195:3743-51. [PMID: 23772066 DOI: 10.1128/jb.00279-13] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Consumer interest in probiotic bifidobacteria is increasing, but industry efforts to secure high cell viability in foods is undermined by these anaerobes' sensitivity to oxidative stress. To address this limitation, we investigated genetic and physiological responses of two fully sequenced Bifidobacterium animalis subsp. lactis strains, BL-04 and DSM 10140, to hydrogen peroxide (H₂O₂) stress. Although the genome sequences for these strains are highly clonal, prior work showed that they differ in both intrinsic and inducible H₂O₂ resistance. Transcriptome analysis of early-stationary-phase cells exposed to a sublethal H₂O₂ concentration detected significant (P < 0.05) changes in expression of 138 genes in strain BL-04 after 5 min and 27 genes after 20 min. Surprisingly, no significant changes in gene expression were detected in DSM 10140 at either time. Genomic data suggested that differences in H₂O₂ stress resistance might be due to a mutation in a BL-04 gene encoding long-chain fatty acid coenzyme A (CoA) ligase. To explore this possibility, membrane fatty acids were isolated and analyzed by gas chromatography-mass spectrometry (GC-MS). Results confirmed that the strains had significantly different lipid profiles: the BL-04 membrane contained higher percentages of C(14:0) and C(16:0) and lower percentages of C(18:1n9). Alteration of the DSM 10140 membrane lipid composition using modified growth medium to more closely mimic that of BL-04 yielded cells that showed increased intrinsic resistance to lethal H₂O₂ challenge but did not display an inducible H₂O₂ stress response. The results show that deliberate stress induction or membrane lipid modification can be employed to significantly improve H₂O₂ resistance in B. animalis subsp. lactis strains.
Collapse
|
46
|
Corazza N, Kaufmann T. Novel insights into mechanisms of food allergy and allergic airway inflammation using experimental mouse models. Allergy 2012; 67:1483-90. [PMID: 23106364 DOI: 10.1111/all.12065] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2012] [Indexed: 11/28/2022]
Abstract
Over the last decades, considerable efforts have been undertaken in the development of animal models mimicking the pathogenesis of allergic diseases occurring in humans. The mouse has rapidly emerged as the animal model of choice, due to considerations of handling and costs and, importantly, due to the availability of a large and increasing arsenal of genetically modified mouse strains and molecular tools facilitating the analysis of complex disease models. Here, we review latest developments in allergy research that have arisen from in vivo experimentation in the mouse, with a focus on models of food allergy and allergic asthma, which constitute major health problems with increasing incidence in industrialized countries. We highlight recent novel findings and controversies in the field, most of which were obtained through the use of gene-deficient or germ-free mice, and discuss new potential therapeutic approaches that have emerged from animal studies and that aim at attenuating allergic reactions in human patients.
Collapse
Affiliation(s)
- N. Corazza
- Institute of Pathology; University of Bern; Bern; Switzerland
| | - T. Kaufmann
- Institute of Pharmacology; University of Bern; Bern; Switzerland
| |
Collapse
|
47
|
Lin WH, Wu CR, Lee HZ, Kuo YH, Wen HS, Lin TY, Lee CY, Huang SY, Lin CY. Induced apoptosis of Th2 lymphocytes and inhibition of airway hyperresponsiveness and inflammation by combined lactic acid bacteria treatment. Int Immunopharmacol 2012; 15:703-11. [PMID: 23142092 DOI: 10.1016/j.intimp.2012.10.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 10/10/2012] [Accepted: 10/26/2012] [Indexed: 12/24/2022]
Abstract
Several lactic acid bacteria (LAB) demonstrably regulate the immune system and inhibit allergic disease. This study examined whether oral feeding of either Lactobacillus paracasei (L. paracasei) BB5 and/or Lactobacillus rhamnosus (L. rhamnosus) BB1 suppresses ovalbumin (OVA)-induced airway hyperresponsiveness (AHR) and inflammation in a murine model. OVA-specific immune responses, cell profile of bronchoalveolar lavage fluid (BALF), and airway AHR were assessed following OVA and methacholine challenge. We investigated whether LAB can enhance CD4(+)FoxP3(+) and CD8(+)FoxP3(+) regulatory T (Treg) cells in splenic cells and apoptosis of CD4(+)IL-4(+) T cells. Results found oral administration of combined LAB better than single L. paracasei or L. rhamnosus strain, improving Penh ratio after challenge with methacholine. High-dose combined LAB starkly decreased synthesis of OVA-specific IgE and IgG2a levels, as well as eosinophils infiltration in BALF. In addition, CD4(+)IL-4(+) T cells decreased while CD4(+)FoxP3(+) and CD8(+)FoxP3(+) Treg cells increased significantly in splenic mononuclear cells of high-dose combined LAB group. Findings indicate allergen-induced AHR and airway allergic inflammation suppressed by enhances CD4(+)FoxP3(+) and CD8(+)FoxP3(+) Treg populations as well as Th1 cell response after treating with combined LAB. This study may provide a basis for developing a novel therapeutic or protective method for airway allergic disease.
Collapse
Affiliation(s)
- Wen-Hsin Lin
- School of Pharmacy, China Medical University, Taichung, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Bifidobacteria may be beneficial to intestinal microbiota and reduction of bacterial translocation in mice following ischaemia and reperfusion injury. Br J Nutr 2012; 109:1990-8. [PMID: 23122253 DOI: 10.1017/s0007114512004308] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The aim of the present study was to determine the effect of peroral bifidobacteria on the intestinal microbiota, barrier function and bacterial translocation (BT) in a mouse model of ischaemia and reperfusion (I/R) injury. A total of twenty-four male BALB/c mice were randomly allocated into three groups: (1) sham-operated, (2) I/R and (3) I/R injury and bifidobacteria pretreatment (109 colony-forming units/d). Bifidobacteria were administered daily intragastrically for 2 weeks before induction of I/R. Subsequently, samples of caecal content, intestinal mucosa, ileal segments, blood, mesenteric lymph nodes (MLN) and distant organs (liver, spleen and kidney) were prepared for examination. In the I/R model, barrier dysfunction (caecal microbiota dysbiosis, disruption of tight junction (TJ), increased epithelial cell apoptosis, disruption of mucosa and multiple erosions) in the intestine was observed, associated with increased BT to extraintestinal sites. The ratio of BT to MLN and distant organs in mice exposed to I/R injury was 62·5 %, which was significantly higher than the sham-operated group. However, pretreatment of animals with bifidobacteria prevented I/R-induced BT, reduced pro-inflammatory cytokine release, the levels of endotoxin, intestinal epithelial cell apoptosis, disruption of TJ and increased the concentration of SCFA, resulting in recovered microbiota and mucosal integrity. Bifidobacteria may be beneficial in reducing BT in I/R injury of mice. Therefore, peroral administration of bifidobacteria is a potential strategy to prevent I/R-induced BT and intestinal barrier dysfunction.
Collapse
|
49
|
Liu M, Huang YK. Progress in understanding the relationship between food allergy and intestinal microflora. Shijie Huaren Xiaohua Zazhi 2012; 20:1421-1427. [DOI: 10.11569/wcjd.v20.i16.1421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A food allergy is defined as a harmful immunological reaction to ingested food protein. According to 2007 CDC statistics, more than 3 million (3.9%) of children under 18 years old suffered from food allergy, and the morbidity is especially higher among children under 5 years old. Healthy intestinal microflora is very important for the development of mature human immune system. The composition of intestinal microflora differs significantly between children with and without food allergy. Food allergy in children may be closely associated with the immature development and damage of the intestinal mucosal barrier, intestinal dysbacteriosis and microflora disorder. Probiotics, as the balancer of intestinal microflora and regulator of intestinal mucosal immunity, can be used to prevent and treat allergic diseases; however, more larger randomized, controlled clinical studies are needed to verify its efficacy.
Collapse
|
50
|
Walton SF, Weir C. The interplay between diet and emerging allergy: what can we learn from Indigenous Australians? Int Rev Immunol 2012; 31:184-201. [PMID: 22587020 DOI: 10.3109/08830185.2012.667180] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The pathophysiology of atopic diseases, including asthma and allergy, is the result of complex gene-environment interactions. Since European colonization the Indigenous population of Australia has undergone significant changes with respect to their lifestyle as hunter-gatherers. These changes have had a detrimental effect on Aboriginal health, in part due to immunological modification. This review provides a comparative look at both the traditional Aboriginal/Indigenous diet and modern Western diets, examines some common allergies increasingly reported in contemporary Indigenous populations, and reviews concepts such the effect of vitamin deficiencies and changes in gut microbiota on immune function.
Collapse
Affiliation(s)
- Shelley F Walton
- School of Health and Sport Sciences, University of the Sunshine Coast, Sippy Downs, Queensland, Australia.
| | | |
Collapse
|