1
|
Liu Y, Dai J, Zhou G, Chen R, Bai C, Shi F. Innovative Therapeutic Strategies for Asthma: The Role of Gut Microbiome in Airway Immunity. J Asthma Allergy 2025; 18:257-267. [PMID: 39996012 PMCID: PMC11849427 DOI: 10.2147/jaa.s504571] [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: 11/04/2024] [Accepted: 02/13/2025] [Indexed: 02/26/2025] Open
Abstract
There is a growing acknowledgment of the gut microbiome's impact on widespread immune responses, which holds considerable importance for comprehending and addressing asthma. Recent research has clarified the complex interactions between gut microbiota and airway immune systems, demonstrating that microbial diversity and composition can affect both the initiation and advancement of asthma. Gut microbial species and metabolites primarily short-chain fatty acids (SCFAs) may either worsen or reduce airway inflammation by regulating the balance of helper T cell 1 (Th1) / helper T cell 2 (Th2) and other immune mediators. This interaction presents innovative therapeutic possibilities, including modulation of gut microbiome during early life through breastfeeding and control of antibiotic use, particularly with prebiotics, which could selectively stimulate the growth of beneficial bacteria, promote immune maturation, reducing susceptibility to asthma and allergic airway inflammation. Besides, investigating the gut-lung axis reveals new opportunities for personalized medicine in asthma treatment, emphasizing the necessity for integrated strategies that take individual microbiome profiles into account. This paper examines the latest developments in comprehending the mechanisms by which gut microbiota affect airway inflammation and hypersensitivity, especially focusing on treatment strategies.
Collapse
Affiliation(s)
- Yaqin Liu
- The Second Clinical Medical College, Jinan University, Shenzhen, Guangdong, 518020, People’s Republic of China
| | - Junjie Dai
- The Second Clinical Medical College, Jinan University, Shenzhen, Guangdong, 518020, People’s Republic of China
| | - Guibao Zhou
- Department of Pharmacy, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, 518020, People’s Republic of China
| | - Rongchang Chen
- Key Laboratory of Shenzhen Respiratory Diseases, Institute of Shenzhen Respiratory Diseases, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, 518020, People’s Republic of China
| | - Chengwen Bai
- Emergency Department, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, 518020, People’s Republic of China
| | - Fei Shi
- Department of Infectious Diseases, Institute of Shenzhen Respiratory Diseases, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, 518020, People’s Republic of China
| |
Collapse
|
2
|
Savova MV, Zhu P, Harms AC, van der Molen RG, Belzer C, Hendrickx DM. Current insights into cow's milk allergy in children: Microbiome, metabolome, and immune response-A systematic review. Pediatr Allergy Immunol 2024; 35:e14084. [PMID: 38363041 DOI: 10.1111/pai.14084] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/17/2024]
Abstract
The increasing prevalence of IgE-mediated cow's milk allergy (CMA) in childhood is a worldwide health concern. There is a growing awareness that the gut microbiome (GM) might play an important role in CMA development. Therefore, treatment with probiotics and prebiotics has gained popularity. This systematic review provides an overview of the alterations of the GM, metabolome, and immune response in CMA children and animal models, including post-treatment modifications. MEDLINE, PubMed, Scopus, and Web of Science were searched for studies on GM in CMA-diagnosed children, published before 1 March 2023. A total of 21 articles (13 on children and 8 on animal models) were included. The studies suggest that the GM, characterized by an enrichment of the Clostridia class and reductions in the Lactobacillales order and Bifidobacterium genus, is associated with CMA in early life. Additionally, reduced levels of short-chain fatty acids (SCFAs) and altered amino acid metabolism were reported in CMA children. Commonly used probiotic strains belong to the Bifidobacterium and Lactobacillus genera. However, only Bifidobacterium levels were consistently upregulated after the intervention, while alterations of other bacteria taxa remain inconclusive. These interventions appear to contribute to the restoration of SCFAs and amino acid metabolism balance. Mouse models indicate that these interventions tend to restore the Th 2/Th 1 balance, increase the Treg response, and/or silence the overall pro- and anti-inflammatory cytokine response. Overall, this systematic review highlights the need for multi-omics-related research in CMA children to gain a mechanistic understanding of this disease and to develop effective treatments and preventive strategies.
Collapse
Affiliation(s)
- Mariyana V Savova
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Pingping Zhu
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Amy C Harms
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Renate G van der Molen
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboudumc, Nijmegen, The Netherlands
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Diana M Hendrickx
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| |
Collapse
|
3
|
Xu Z, Bai H, Ma X, Wu Y, Wu Z, Yang A, Mao W, Li X, Chen H. Cytological evaluation by Caco-2 and KU812 of non-allergenic peptides from simulated digestion of infant formula in vitro. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2022.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
4
|
Liu M, Thijssen S, Hennink WE, Garssen J, van Nostrum CF, Willemsen LM. Oral pretreatment with β-lactoglobulin derived peptide and CpG co-encapsulated in PLGA nanoparticles prior to sensitizations attenuates cow's milk allergy development in mice. Front Immunol 2023; 13:1053107. [PMID: 36703973 PMCID: PMC9872660 DOI: 10.3389/fimmu.2022.1053107] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 12/19/2022] [Indexed: 01/07/2023] Open
Abstract
Cow's milk allergy is a common food allergy among infants. Improved hygiene conditions and loss of microbial diversity are associated with increased risk of allergy development. The intestinal immune system is essential for oral tolerance induction. In this respect, bacterial CpG DNA is known to drive Th1 and regulatory T-cell (Treg) development via Toll-Like-Receptor 9 (TLR-9) signaling, skewing away from the allergic Th2 phenotype. We aimed to induce allergen specific tolerance via oral delivery of poly (lactic-co-glycolic acid) nanoparticles (NP) co-encapsulated with a selected β-lactoglobulin derived peptide (BLG-Pep) and TLR-9 ligand CpG oligodeoxynucleotide (CpG). In vivo, 3-4-week-old female C3H/HeOuJ mice housed in individually ventilated cages received 6-consecutive-daily gavages of either PBS, whey, BLG-Pep/NP, CpG/NP, a mixture of BLG-Pep/NP plus CpG/NP or co-encapsulated BLG-Pep+CpG/NP, before 5-weekly oral sensitizations with whey plus cholera toxin (CT) or only CT (sham) and were challenged with whey 5 days after the last sensitization. The co-encapsulated BLG-Pep+CpG/NP pretreatment, but not BLG-Pep/NP, CpG/NP or the mixture of BLG-Pep/NP plus CpG/NP, prevented the whey-induced allergic skin reactivity and prevented rise in serum BLG-specific IgE compared to whey-sensitized mice. Importantly, co-encapsulated BLG-Pep+CpG/NP pretreatment reduced dendritic cell (DC) activation and lowered the frequencies of PD-L1+ DC in the mesenteric lymph nodes compared to whey-sensitized mice. By contrast, co-encapsulated BLG-Pep+CpG/NP pretreatment increased the frequency of splenic PD-L1+ DC compared to the BLG-Pep/NP plus CpG/NP recipients, in association with lower Th2 development and increased Treg/Th2 and Th1/Th2 ratios in the spleen. Oral administration of PLGA NP co-encapsulated with BLG-Pep and CpG prevented rise in serum BLG-specific IgE and symptom development while lowering splenic Th2 cell frequency in these mice which were kept under strict hygienic conditions.
Collapse
Affiliation(s)
- Mengshan Liu
- Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands,Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Suzan Thijssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Wim E. Hennink
- Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands,Department of Immunology, Nutricia Research B.V., Utrecht, Netherlands
| | - Cornelus F. van Nostrum
- Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Linette E. M. Willemsen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands,*Correspondence: Linette E. M. Willemsen,
| |
Collapse
|
5
|
Shi J, Wang Y, Cheng L, Wang J, Raghavan V. Gut microbiome modulation by probiotics, prebiotics, synbiotics and postbiotics: a novel strategy in food allergy prevention and treatment. Crit Rev Food Sci Nutr 2022; 64:5984-6000. [PMID: 36576159 DOI: 10.1080/10408398.2022.2160962] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Food allergy has caused lots of global public health issues, particularly in developed countries. Presently, gut microbiota has been widely studied on allergy, while the role of dysbiosis in food allergy remains unknown. Scientists found that changes in gut microbial compositions and functions are strongly associated with a dramatic increase in the prevalence of food allergy. Altering microbial composition is crucial in modulating food antigens' immunogenicity. Thus, the potential roles of probiotics, prebiotics, synbiotics, and postbiotics in affecting gut bacteria communities and the immune system, as innovative strategies against food allergy, begins to attract high attention of scientists. This review briefly summarized the mechanisms of food allergy and discussed the role of the gut microbiota and the use of probiotics, prebiotics, synbiotics, and postbiotics as novel therapies for the prevention and treatment of food allergy. The perspective studies on the development of novel immunotherapy in food allergy were also described. A better understanding of these mechanisms will facilitate the development of preventive and therapeutic strategies for food allergy.
Collapse
Affiliation(s)
- Jialu Shi
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, China
| | - Youfa Wang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, China
| | - Lei Cheng
- Department of Otorhinolaryngology and Clinical Allergy Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jin Wang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, China
| | - Vijaya Raghavan
- Department of Bioresource Engineering, Faculty of Agricultural and Environmental Sciences, McGill University, Montreal, Canada
| |
Collapse
|
6
|
Keulers L, Dehghani A, Knippels L, Garssen J, Papadopoulos N, Folkerts G, Braber S, van Bergenhenegouwen J. Probiotics, prebiotics, and synbiotics to prevent or combat air pollution consequences: The gut-lung axis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 302:119066. [PMID: 35240267 DOI: 10.1016/j.envpol.2022.119066] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 05/26/2023]
Abstract
Air pollution exposure is a public health emergency, which attributes globally to an estimated seven million deaths on a yearly basis We are all exposed to air pollutants, varying from ambient air pollution hanging over cities to dust inside the home. It is a mixture of airborne particulate matter and gases that can be subdivided into three categories based on particle diameter. The smallest category called PM0.1 is the most abundant. A fraction of the particles included in this category might enter the blood stream spreading to other parts of the body. As air pollutants can enter the body via the lungs and gut, growing evidence links its exposure to gastrointestinal and respiratory impairments and diseases, like asthma, rhinitis, respiratory tract infections, Crohn's disease, ulcerative colitis, and abdominal pain. It has become evident that there exists a crosstalk between the respiratory and gastrointestinal tracts, commonly referred to as the gut-lung axis. Via microbial secretions, metabolites, immune mediators and lipid profiles, these two separate organ systems can influence each other. Well-known immunomodulators and gut health stimulators are probiotics, prebiotics, together called synbiotics. They might combat air pollution-induced systemic inflammation and oxidative stress by optimizing the microbiota composition and microbial metabolites, thereby stimulating anti-inflammatory pathways and strengthening mucosal and epithelial barriers. Although clinical studies investigating the role of probiotics, prebiotics, and synbiotics in an air pollution setting are lacking, these interventions show promising health promoting effects by affecting the gastrointestinal- and respiratory tract. This review summarizes the current data on how air pollution can affect the gut-lung axis and might impact gut and lung health. It will further elaborate on the potential role of probiotics, prebiotics and synbiotics on the gut-lung axis, and gut and lung health.
Collapse
Affiliation(s)
- Loret Keulers
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584, CG, Utrecht, the Netherlands; Danone Nutricia Research, Uppsalalaan 12, 3584, CT, Utrecht, the Netherlands.
| | - Ali Dehghani
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584, CG, Utrecht, the Netherlands
| | - Leon Knippels
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584, CG, Utrecht, the Netherlands; Danone Nutricia Research, Uppsalalaan 12, 3584, CT, Utrecht, the Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584, CG, Utrecht, the Netherlands; Danone Nutricia Research, Uppsalalaan 12, 3584, CT, Utrecht, the Netherlands
| | - Nikolaos Papadopoulos
- Centre for Paediatrics and Child Health, Institute of Human Development, University of Manchester, Oxford Road M13 9PL, Manchester, United Kingdom
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584, CG, Utrecht, the Netherlands
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584, CG, Utrecht, the Netherlands
| | - Jeroen van Bergenhenegouwen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584, CG, Utrecht, the Netherlands; Danone Nutricia Research, Uppsalalaan 12, 3584, CT, Utrecht, the Netherlands
| |
Collapse
|
7
|
Blanco-Pérez F, Steigerwald H, Schülke S, Vieths S, Toda M, Scheurer S. The Dietary Fiber Pectin: Health Benefits and Potential for the Treatment of Allergies by Modulation of Gut Microbiota. Curr Allergy Asthma Rep 2021; 21:43. [PMID: 34505973 PMCID: PMC8433104 DOI: 10.1007/s11882-021-01020-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/21/2021] [Indexed: 02/08/2023]
Abstract
Purpose of Review The incidence of allergies is increasing and has been associated with several environmental factors including westernized diets. Changes in environment and nutrition can result in dysbiosis of the skin, gut, and lung microbiota altering the production of microbial metabolites, which may in turn generate epigenetic modifications. The present review addresses studies on pectin-mediated effects on allergies, including the immune modulating mechanisms by bacterial metabolites. Recent Findings Recently, microbiota have gained attention as target for allergy intervention, especially with prebiotics, that are able to stimulate the growth and activity of certain microorganisms. Dietary fibers, which cannot be digested in the gastrointestinal tract, can alter the gut microbiota and lead to increased local and systemic concentrations of gut microbiota-derived short chain fatty acids (SCFAs). These can promote the generation of peripheral regulatory T cells (Treg) by epigenetic modulation and suppress the inflammatory function of dendritic cells (DCs) by transcriptional modulation. The dietary fiber pectin (a plant-derived polysaccharide commonly used as gelling agent and dietary supplement) can alter the ratio of Firmicutes to Bacteroidetes in gut and lung microbiota, increasing the concentrations of SCFAs in feces and sera, and reducing the development of airway inflammation by suppressing DC function. Summary Pectin has shown immunomodulatory effects on allergies, although the underlying mechanisms still need to be elucidated. It has been suggested that the different types of pectin may exert direct and/or indirect immunomodulatory effects through different mechanisms. However, little is known about the relation of certain pectin structures to allergies.
Collapse
Affiliation(s)
- Frank Blanco-Pérez
- Molecular Allergology, Federal Institute for Vaccines and Biomedicines, Paul-Ehrlich-Institut, Langen, Germany.
| | - Hanna Steigerwald
- Molecular Allergology, Federal Institute for Vaccines and Biomedicines, Paul-Ehrlich-Institut, Langen, Germany
| | - Stefan Schülke
- Molecular Allergology, Federal Institute for Vaccines and Biomedicines, Paul-Ehrlich-Institut, Langen, Germany
| | - Stefan Vieths
- Molecular Allergology, Federal Institute for Vaccines and Biomedicines, Paul-Ehrlich-Institut, Langen, Germany
| | - Masako Toda
- Laboratory of Food and Biomolecular Science, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Stephan Scheurer
- Molecular Allergology, Federal Institute for Vaccines and Biomedicines, Paul-Ehrlich-Institut, Langen, Germany
| |
Collapse
|
8
|
Szklany K, Kraneveld AD, Tiemessen MM, Garssen J, Knippels LMJ. Nutritional Interventions to Prevent the Development of Atopic Diseases: A Focus on Cow's Milk Allergy. Handb Exp Pharmacol 2021; 268:471-486. [PMID: 34085122 DOI: 10.1007/164_2021_480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
In the western world the prevalence of atopic diseases such as food allergies is increasing highly significantly. One of the earliest and most prevalent food allergies occurring in the first year of life is cow's milk allergy. No treatment is available and only avoidance of the cow's milk allergens prevents the occurrence of an allergic reaction. Since cow's milk allergic children have an increased risk of developing other allergies later in life, investigating nutritional strategies to prevent the development of cow's milk allergy by developing oral tolerance is of high interest. Nutritional components such as prebiotics, probiotics, synbiotics and long-chain polyunsaturated fatty acids possess potential to support the maturation of the immune system early in life that might prevent the development of cow's milk allergy. The available research, so far, shows promising results particularly on the development of eczema. However, the preventive effects of the nutritional interventions on the development of food allergy are inconclusive. Future research may benefit from the combination of various dietary components. To clarify the preventive effects of the nutritional components in food allergy more randomized clinical trials are needed.
Collapse
Affiliation(s)
- Kirsten Szklany
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of science, Utrecht University, Utrecht, The Netherlands
| | - Aletta D Kraneveld
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of science, Utrecht University, Utrecht, The Netherlands.,Veterinary Pharmacology and Therapeutics, Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Machteld M Tiemessen
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of science, Utrecht University, Utrecht, The Netherlands.,Department of Immunology, Danone Nutricia Research, Utrecht, The Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of science, Utrecht University, Utrecht, The Netherlands.,Department of Immunology, Danone Nutricia Research, Utrecht, The Netherlands
| | - Leon M J Knippels
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of science, Utrecht University, Utrecht, The Netherlands. .,Department of Immunology, Danone Nutricia Research, Utrecht, The Netherlands.
| |
Collapse
|
9
|
Adel-Patient K, Guinot M, Guillon B, Bernard H, Chikhi A, Hazebrouck S, Junot C. Administration of Extensive Hydrolysates From Caseins and Lactobacillus rhamnosus GG Probiotic Does Not Prevent Cow's Milk Proteins Allergy in a Mouse Model. Front Immunol 2020; 11:1700. [PMID: 33042105 PMCID: PMC7516991 DOI: 10.3389/fimmu.2020.01700] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 06/25/2020] [Indexed: 11/18/2022] Open
Abstract
Background Early nutrition may influence the development of food allergies later in life. In the absence of breastfeeding, hydrolysates from cow's milk proteins (CMP) were indicated as a prevention strategy in at risk infants, but their proof of effectiveness in clinical and pre-clinical studies is still insufficient. Thanks to a validated mouse model, we then assessed specific and nonspecific preventive effects of administration of extensive hydrolysates from caseins (eHC) on the development of food allergy to CMP. The additional nonspecific effect of the probiotic Lactobacillus GG (LGG), commonly used in infant formula, was also assessed. Methods Groups of young BALB/cByJ female mice were pretreated by repeated gavage either with PBS (control mice), or with PBS solution containing non-hydrolyzed milk protein isolate (MPI), eHC or eHC+LGG (eq. of 10 mg of protein/gavage). All mice were then experimentally sensitized to CMP by gavage with whole CM mixed with the Th2 mucosal adjuvant Cholera toxin. All mice were further chronically exposed to cow's milk. A group of mice was kept naïve. Sensitization to both caseins and to the non-related whey protein β-lactoglobulin (BLG) was evaluated by measuring specific antibodies in plasma and specific ex vivo Th2/Th1/Th17 cytokine secretion. Elicitation of the allergic reaction was assessed by measuring mMCP1 in plasma obtained after oral food challenge (OFC) with CMP. Th/Treg cell frequencies in gut-associated lymphoid tissue and spleen were analyzed by flow cytometry at the end of the protocol. Robust statistical procedure combining non-supervised and supervised multivariate analyses and univariate analyses, was conducted to reveal any effect of the pretreatments. Results PBS pretreated mice were efficiently sensitized and demonstrated elicitation of allergic reaction after OFC, whereas mice pretreated with MPI were durably protected from allergy to CMP. eHC+/-LGG pretreatments had no protective effect on sensitization to casein (specific) or BLG (non-specific), nor on CMP-induced allergic reactions. Surprisingly, eHC+LGG mice demonstrated significantly enhanced humoral and cellular immune responses after sensitization with CMP. Only some subtle changes were evidenced by flow cytometry. Conclusion Neither specific nor nonspecific preventive effects of administration of casein-derived peptides on the development of CMP food allergy were evidenced in our experimental setup. Further studies should be conducted to delineate the mechanisms involved in the immunostimulatory potential of LGG and to clarify its significance in clinical use.
Collapse
Affiliation(s)
- Karine Adel-Patient
- Service de Pharmacologie et d’Immunoanalyse, Département Médicaments et Technologies pour la Santé (DMTS), CEA, INRAE, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Marine Guinot
- Service de Pharmacologie et d’Immunoanalyse, Département Médicaments et Technologies pour la Santé (DMTS), CEA, INRAE, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Blanche Guillon
- Service de Pharmacologie et d’Immunoanalyse, Département Médicaments et Technologies pour la Santé (DMTS), CEA, INRAE, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Hervé Bernard
- Service de Pharmacologie et d’Immunoanalyse, Département Médicaments et Technologies pour la Santé (DMTS), CEA, INRAE, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Amina Chikhi
- Service de Pharmacologie et d’Immunoanalyse, Département Médicaments et Technologies pour la Santé (DMTS), CEA, INRAE, Université Paris-Saclay, Gif-sur-Yvette, France
- Laboratoire de Physiologie de la Nutrition et de Sécurité Alimentaire, Université d’Oran 1 Ahmed Ben Bella, Oran, Algeria
| | - Stéphane Hazebrouck
- Service de Pharmacologie et d’Immunoanalyse, Département Médicaments et Technologies pour la Santé (DMTS), CEA, INRAE, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Christophe Junot
- Service de Pharmacologie et d’Immunoanalyse, Département Médicaments et Technologies pour la Santé (DMTS), CEA, INRAE, Université Paris-Saclay, Gif-sur-Yvette, France
| |
Collapse
|
10
|
Paraimmunobiotic Bifidobacteria Modulate the Expression Patterns of Peptidoglycan Recognition Proteins in Porcine Intestinal Epitheliocytes and Antigen Presenting Cells. Cells 2019; 8:cells8080891. [PMID: 31416116 PMCID: PMC6721749 DOI: 10.3390/cells8080891] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 08/07/2019] [Accepted: 08/09/2019] [Indexed: 02/07/2023] Open
Abstract
Peptidoglycan recognition proteins (PGLYRPs) are a family of pattern recognition receptors (PRRs) that are able to induce innate immune responses through their binding to peptidoglycan (PGN), lipopolysaccharide, or lipoteichoic acid, or by interacting with other PRR-ligands. Recently, progress has been made in understanding the immunobiology of PGLYRPs in human and mice, however, their functions in livestock animals have been less explored. In this study, we characterized the expression patterns of PGLYRPs in porcine intestinal epithelial (PIE) cells and antigen-presenting cells (APCs) and their modulation by the interactions of host cells with PRR-ligands and non-viable immunomodulatory probiotics referred to as paraimmunobiotics. We demonstrated that PGLYRP-1, -2, -3, and -4 are expressed in PIE cells and APCs from Peyer’s patches, being PGLYPR-3 and -4 levels higher than PGLYRP-1 and -2. We also showed that PGLYRPs expression in APCs and PIE cells can be modulated by different PRR agonists. By using knockdown PIE cells for TLR2, TLR4, NOD1, and NOD2, or the four PGLYRPs, we demonstrated that PGLYRPs expressions would be required for activation and functioning of TLR2, TLR4, NOD1, and NOD2 in porcine epitheliocytes, but PGLYRPs activation would be independent of those PRR expressions. Importantly, we reported for the first time that PGLYRPs expression can be differentially modulated by paraimmunobiotic bifidobacteria in a strain-dependent manner. These results provide evidence for the use of paraimmunobiotic bifidobacteria as an alternative for the improvement of resistance to intestinal infections or as therapeutic tools for the reduction of the severity of inflammatory damage in diseases in which a role of PGLYRPs-microbe interaction has been demonstrated.
Collapse
|
11
|
Izumi H, Ehara T, Sugahara H, Matsubara T, Mitsuyama E, Nakazato Y, Tsuda M, Shimizu T, Odamaki T, Xiao JZ, Takeda Y. The Combination of Bifidobacterium breve and Three Prebiotic Oligosaccharides Modifies Gut Immune and Endocrine Functions in Neonatal Mice. J Nutr 2019; 149:344-353. [PMID: 30721975 DOI: 10.1093/jn/nxy248] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 04/03/2018] [Accepted: 09/04/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Several types of oligosaccharides are used in infant formula to improve the gut microbiota of formula-fed infants. We previously reported that a combination of 3 oligosaccharides (lactulose, raffinose, and galacto-oligosaccharides; LRG) and Bifidobacterium breve effectively increased B. breve numbers, acetate, and the expression of several immune- and gut hormone-related mRNAs in neonatal mice gut. OBJECTIVE We investigated whether changes in neonatal gut microbiota alter gut immune and endocrine development. METHODS We first compared postnatal day (PD) 14 with PD21 in C57BL/6J male mouse pups to identify the physiologic immune and endocrine changes during development. In a separate study, we administered phosphate-buffered saline (control group; CON), B. breve M-16V (M-16V), or M-16V + LRG to male mouse pups from PD6 to PD13, and analyzed the gut microbiota and immune and endocrine parameters on PD14 to evaluate whether M-16V + LRG accelerates gut immune and endocrine development. RESULTS The proportion of regulatory T (Treg) cells in the CD4+ cells of large intestinal lamina propria lymphocytes (LPLs) was significantly increased (63% higher) at PD21 compared with PD14. The serum glucagon-like peptide (GLP)-1 tended to be lower (P = 0.0515) and that of GLP-2 was significantly lower (58% lower) at PD21 than at PD14. M-16V + LRG significantly increased the Treg proportion in large intestinal LPL CD4+ cells (20% and 29% higher compared with CON and M-16V, respectively) at PD14. M-16V + LRG also caused significant changes in expression of large intestinal mRNAs that are consistent with developmental progression, and increased serum concentrations of GLP-1 (207% and 311% higher compared with CON and M-16V, respectively) and GLP-2 (57% and 97% higher compared with CON and M-16V, respectively) at PD14. CONCLUSIONS Neonatal administration of M-16V + LRG alters the gut microbiota and enhances gut immune and endocrine development in suckling mice.
Collapse
Affiliation(s)
| | - Tatsuya Ehara
- Wellness & Nutrition Science Institute, R&D Division
| | - Hirosuke Sugahara
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., Kanagawa, Japan
| | | | - Eri Mitsuyama
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., Kanagawa, Japan
| | - Yuki Nakazato
- Wellness & Nutrition Science Institute, R&D Division
| | - Muneya Tsuda
- Wellness & Nutrition Science Institute, R&D Division
| | | | - Toshitaka Odamaki
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., Kanagawa, Japan
| | - Jin-Zhong Xiao
- Next Generation Science Institute, R&D Division, Morinaga Milk Industry Co., Ltd., Kanagawa, Japan
| | | |
Collapse
|
12
|
Pescuma M, Hébert E, Font G, Saavedra L, Mozzi F. Hydrolysate of β-lactoglobulin by Lactobacillus delbrueckii subsp. bulgaricus CRL 656 suppresses the immunoreactivity of β-lactoglobulin as revealed by in vivo assays. Int Dairy J 2019. [DOI: 10.1016/j.idairyj.2018.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
13
|
Gouw JW, Jo J, Meulenbroek LAPM, Heijjer TS, Kremer E, Sandalova E, Knulst AC, Jeurink PV, Garssen J, Rijnierse A, Knippels LMJ. Identification of peptides with tolerogenic potential in a hydrolysed whey-based infant formula. Clin Exp Allergy 2018; 48:1345-1353. [PMID: 29974988 DOI: 10.1111/cea.13223] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 05/08/2018] [Accepted: 06/28/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND Failure to induce oral tolerance may result in food allergy. Hydrolysed cow's milk-based infant formulas are recommended in subjects with a high risk of developing allergic disease. Presentation of T cell epitopes is a prerequisite to generate regulatory T cells that could contribute to oral tolerance. OBJECTIVE To investigate whether a specific hydrolysed whey-based infant formula contains peptides that function as T cell epitopes to support the development of oral tolerance to whey. METHODS First, a novel liquid chromatography-mass spectrometry (LC-MS) method was developed to characterize β-lactoglobulin-derived peptides present in a specific infant formula with a focus on region AA#13-48 of β-lactoglobulin, which has previously been described to contain T cell epitopes with tolerogenic potential. Second, the formula was subjected to the ProImmune ProPresent® antigen presentation assay and MHC class II binding algorithm to identify relevant HLA-DRB1-restricted peptides. Third, identified peptides were tested on human cow's milk protein-specific T cell lines to determine T cell recognition. RESULTS Thirteen peptides of minimal 9AAs long that overlap with AA#13-48 of β-lactoglobulin were identified. Six of them were found across all batches analysed. It was further confirmed that these peptides were processed and presented by human dendritic cells. The identified HLA-DRB1-restricted peptides were correlated to AA#11-30 and AA#23-39 of β-lactoglobulin. Importantly, the proliferation assay showed that the synthetic peptides were recognized by cow's milk protein-specific T cell lines and induced T cell proliferation. CONCLUSION AND CLINICAL RELEVANCE This study demonstrates that the tested hydrolysed infant formula contains functional HLA-DRB1-restricted T cell epitopes, which can potentially support the development of oral tolerance to whey.
Collapse
Affiliation(s)
- Joost W Gouw
- Danone Nutricia Research, Utrecht, The Netherlands
| | - Juandy Jo
- Danone Nutricia Research, Singapore.,Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Laura A P M Meulenbroek
- Danone Nutricia Research, Utrecht, The Netherlands.,Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - T Sam Heijjer
- Danone Nutricia Research, Utrecht, The Netherlands.,Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Erica Kremer
- Danone Nutricia Research, Utrecht, The Netherlands
| | - Elena Sandalova
- Danone Nutricia Research, Singapore.,Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - André C Knulst
- Department of Dermatology and Allergology, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Prescilla V Jeurink
- Danone Nutricia Research, Utrecht, The Netherlands.,Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Johan Garssen
- Danone Nutricia Research, Utrecht, The Netherlands.,Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | | | - Léon M J Knippels
- Danone Nutricia Research, Utrecht, The Netherlands.,Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
14
|
Nochi T, Jansen CA, Toyomizu M, van Eden W. The Well-Developed Mucosal Immune Systems of Birds and Mammals Allow for Similar Approaches of Mucosal Vaccination in Both Types of Animals. Front Nutr 2018; 5:60. [PMID: 30050906 PMCID: PMC6052093 DOI: 10.3389/fnut.2018.00060] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 06/19/2018] [Indexed: 01/07/2023] Open
Abstract
The mucosal immune system is a compartmentalized part of the immune system that provides local immunity in the mucosa of the respiratory, gastrointestinal, and digestive tracts. It possesses secondary lymphoid tissues, which contain immune cells, such as T, B, and dendritic cells. Once the cells of the mucosal immune system are stimulated by luminal antigens, including microorganisms, they infiltrate into diffuse areas of mucosal tissues (e.g., respiratory mucosa and lamina propria of intestinal villi) and exhibit immune effector functions. Inducing the antigen-specific immune responses in mucosal tissues by mucosal vaccination would be an ideal strategy for not only humans, but also mammals and birds, to protect against infectious diseases occurring in mucosal tissues (e.g., pneumonia and diarrhea). Infectious diseases cause huge economic losses in agriculture, such as livestock and poultry industries. Since most infectious diseases occur in mucosal tissues, vaccines that are capable of inducing immune responses in mucosal tissues are in high need. In this review, we discuss the current understanding of mucosal immunity in mammals and birds, and recent progress in the development of mucosal vaccines.
Collapse
Affiliation(s)
- Tomonori Nochi
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, Japan.,International Research and Development Center for Mucosal Vaccine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Christine A Jansen
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Masaaki Toyomizu
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, Japan
| | - Willem van Eden
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
15
|
Hulshof L, Overbeek SA, Wyllie AL, Chu MLJN, Bogaert D, de Jager W, Knippels LMJ, Sanders EAM, van Aalderen WMC, Garssen J, Van't Land B, Sprikkelman AB. Exploring Immune Development in Infants With Moderate to Severe Atopic Dermatitis. Front Immunol 2018; 9:630. [PMID: 29966024 PMCID: PMC5884950 DOI: 10.3389/fimmu.2018.00630] [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: 12/04/2017] [Accepted: 03/13/2018] [Indexed: 12/16/2022] Open
Abstract
Background Atopic dermatitis (AD) is the most common chronic inflammatory skin disease in infancy with a complex pathology. In adults, the clinical severity of AD has been associated with increases in T helper cell type (Th) 2, Th22, and Th17 serum markers, including high levels of CC chemokine ligand (CCL) 17 and CCL22 chemokines. Objective To explore the possible association between serum chemokine levels and AD severity in infants with moderate-to-severe AD and elevated immunoglobulin E (IgE). Subjects and methods Serum samples (n = 41) obtained from a randomized, double-blind, and clinical dietary intervention study were used to study biomarkers in infants with AD. Baseline- and post-intervention samples (4 months) were used, six chemokines and nine ratios thereof were analyzed using Luminex and correlated to AD severity. In the initial study, the infants were randomized to receive extensively hydrolyzed whey-based formula without (control) or with short-chain galacto-oligosaccharides/long-chain fructo-oligosaccharides (9:1) and Bifidobacterium breve M-16V (active). Results 31 Infants up to 11 months of age, with an objective-SCORAD score (oSCORAD) ≥ 20 and elevated total-IgE and/or specific-IgE levels were included. In time, the median oSCORAD decreased in both groups by −8 (control, p < 0.05; active, p < 0.01). Irrespective of dietary intervention, several changes in Th2 chemokines (CCL17 and CCL22), inflammatory chemokine (CCL20), and the Th1 chemokine, CXC chemokine ligand (CXCL) 9, were detected over time. Overall CCL17 correlated to oSCORAD (r = 0.446, p < 0.01). After 4 months of dietary intervention, CXCL9 was higher (p < 0.01) in the active group compared with control [active, 2.33 (1.99–2.89); controls, 1.95 (1.77–2.43) log 10 median (range)]. In addition, a reduction in Th2/Th1 chemokine ratios for CCL17/CXCL9, CCL22/CXCL9, CCL20/CXCL10, and CCL20/CXCL11 was detected associated with the active intervention. Conclusion While this study is small and exploratory in nature, these data contribute to immune biomarker profiling and understanding of AD in infants.
Collapse
Affiliation(s)
- Lies Hulshof
- Emma Children's Hospital Academic Medical Centre, Department of Paediatric Respiratory Medicine and Allergy, University of Amsterdam, Amsterdam, Netherlands
| | - Saskia A Overbeek
- Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Nutricia Research, Utrecht, Netherlands
| | - Anne L Wyllie
- Department of Paediatric Immunology and Infectious Diseases, University Medical Centre Utrecht, Wilhelmina Children's Hospital, Utrecht, Netherlands
| | - Mei Ling J N Chu
- Department of Paediatric Immunology and Infectious Diseases, University Medical Centre Utrecht, Wilhelmina Children's Hospital, Utrecht, Netherlands
| | - Debby Bogaert
- Department of Paediatric Immunology and Infectious Diseases, University Medical Centre Utrecht, Wilhelmina Children's Hospital, Utrecht, Netherlands
| | - Wilco de Jager
- Laboratory of Translational Immunology, Department of Paediatric Immunology, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Leon M J Knippels
- Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Nutricia Research, Utrecht, Netherlands
| | - Elisabeth A M Sanders
- Department of Paediatric Immunology and Infectious Diseases, University Medical Centre Utrecht, Wilhelmina Children's Hospital, Utrecht, Netherlands
| | - Wim M C van Aalderen
- Emma Children's Hospital Academic Medical Centre, Department of Paediatric Respiratory Medicine and Allergy, University of Amsterdam, Amsterdam, Netherlands
| | - Johan Garssen
- Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Nutricia Research, Utrecht, Netherlands
| | - Belinda Van't Land
- Nutricia Research, Utrecht, Netherlands.,Department of Paediatric Immunology and Infectious Diseases, University Medical Centre Utrecht, Wilhelmina Children's Hospital, Utrecht, Netherlands
| | - Aline B Sprikkelman
- Emma Children's Hospital Academic Medical Centre, Department of Paediatric Respiratory Medicine and Allergy, University of Amsterdam, Amsterdam, Netherlands.,Department of Paediatric Pulmonology and Paediatric Allergology, University of Groningen, University Medical Centre Groningen, Beatrix Children's Hospital, Groningen, Netherlands
| | | |
Collapse
|