1
|
Sogard AS, Emerson TS, Chandler CA, Cobb EA, Shei RJ, Paris HL, Lindley MR, Mickleborough TD. The role of nutritional factors in exercise-induced bronchoconstriction: a narrative review. Am J Physiol Regul Integr Comp Physiol 2025; 328:R651-R684. [PMID: 40257056 DOI: 10.1152/ajpregu.00249.2024] [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: 10/04/2024] [Revised: 12/20/2024] [Accepted: 04/12/2025] [Indexed: 04/22/2025]
Abstract
Exercise-induced bronchoconstriction (EIB) describes an acute narrowing of the airways that develops following vigorous physical activity. Clinical responses to current asthma therapy, such as leukotriene antagonists and corticosteroids, are heterogeneous, even with optimal treatment. Epidemiological studies indicate an increasing use of complementary and alternative medicine therapy in asthma patients due to the lack of efficacy of conventional treatment, concerns about potentially harmful side effects of pharmacological treatment, cost barriers to asthma care, and the accessibility of complementary and alternative medicine therapy. Plausible physiological mechanisms now exist for many nutrients as potential modifiers of EIB severity, primarily because of their role in inflammatory processes, airway smooth muscle function, and modulation of lung microvascular volume and pressure. Dietary supplementation as a treatment for EIB has generally shown evidence of significant yet incomplete inhibition of EIB with low-salt diets, omega-3 fatty acids, and vitamin C when supplemented for up to 3 weeks. However, larger, randomized, placebo-controlled, double-blinded trials are needed to clarify the effectiveness of nutritional intervention in individuals with EIB. Additionally, many studies have focused on nonathletes with EIB, and therefore, more studies are required to evaluate the efficacy of nutritional intervention on EIB in elite athletes. In conclusion, if dietary supplementation or restriction is prescribed, it should be seen as an option to lessen the reliance on pharmaceutical interventions and not as an alternative to established pharmacotherapies.
Collapse
Affiliation(s)
- Abigail S Sogard
- Department of Kinesiology, School of Public Health-Bloomington, Indiana University, Bloomington, Indiana, United States
| | - Travis S Emerson
- Department of Kinesiology, School of Public Health-Bloomington, Indiana University, Bloomington, Indiana, United States
| | - Christopher A Chandler
- Department of Kinesiology, School of Public Health-Bloomington, Indiana University, Bloomington, Indiana, United States
| | - Emily A Cobb
- Department of Kinesiology, School of Public Health-Bloomington, Indiana University, Bloomington, Indiana, United States
| | - Ren-Jay Shei
- Indiana University Alumni Association, Indiana University, Bloomington, Indiana, United States
| | - Hunter L Paris
- Department of Sports Medicine, Pepperdine University, Malibu, California, United States
| | - Martin R Lindley
- School of Health Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | - Timothy D Mickleborough
- Department of Kinesiology, School of Public Health-Bloomington, Indiana University, Bloomington, Indiana, United States
| |
Collapse
|
2
|
Silva RA, Nahas MJB, Bremer SDC, de Oliveira LW, de Souza AM, Rufino MN, Caetano HRDS, Keller R, Bremer-Neto H. Prebiotics improve parameters indicative of allergy-induced asthma in murines: a systematic review with meta-analysis. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025. [PMID: 40346864 DOI: 10.1002/jsfa.14325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 02/19/2025] [Accepted: 04/13/2025] [Indexed: 05/12/2025]
Abstract
The prevalence of allergic asthma is increasing worldwide and it is important that new treatments are implemented. This study aimed to thoroughly investigate the effect of prebiotics on parameters indicative of allergic asthma induced by ovalbumin (OVA), house dust mites (HDM) and OVA + lipopolysaccharide (LPS). The review was registered in the Open Science Framework (registration ID: osf.io/du4ab). PRISMA and web-app Rayyan were used as tools for the selection of studies collected in seven databases: PubMed; ScienceDirect; Web of Science; Scielo; Scopus; EMBASE; and Google Scholar with the use of predetermined keywords and Medical Subject Heading terms of the National Library of Medicine. Eight studies involving 182 mice and rats were included in the meta-analysis. The results showed a significant reduction in the total number of inflammatory cells and in the isolated number of inflammatory cells, eosinophils, lymphocytes, macrophages and neutrophils (P < 0.05). The results also revealed a significant decrease in the concentration of interleukins 13 and 33 in lung tissue. The present study demonstrated that prebiotic supplementation in the diet of rats and mice, as preclinical models, mitigates indicative parameters, inflammatory cells and interleukins, of allergic asthma induced by OVA, HDM or OVA + LPS. These beneficial results encourage randomized clinical trials to be carried out aiming at the prevention/treatment of allergic asthma. © 2025 Society of Chemical Industry.
Collapse
Affiliation(s)
- Ricardo Augusto Silva
- Food, Nutrition and Basic, Experimental and Clinical Health Research Group, Department of Functional Sciences, Faculty of Medicine, Western São Paulo University (UNOESTE), São Paulo, Brazil
| | - Maria Julia Brolezzi Nahas
- Food, Nutrition and Basic, Experimental and Clinical Health Research Group, Department of Functional Sciences, Faculty of Medicine, Western São Paulo University (UNOESTE), São Paulo, Brazil
| | | | - Luiz Waldemar de Oliveira
- Food, Nutrition and Basic, Experimental and Clinical Health Research Group, Department of Functional Sciences, Faculty of Medicine, Western São Paulo University (UNOESTE), São Paulo, Brazil
| | - Adriano Messias de Souza
- Food, Nutrition and Basic, Experimental and Clinical Health Research Group, Department of Functional Sciences, Faculty of Medicine, Western São Paulo University (UNOESTE), São Paulo, Brazil
| | - Marcos Natal Rufino
- Food, Nutrition and Basic, Experimental and Clinical Health Research Group, Department of Functional Sciences, Faculty of Medicine, Western São Paulo University (UNOESTE), São Paulo, Brazil
| | - Heliard Rodrigues Dos Santos Caetano
- Food, Nutrition and Basic, Experimental and Clinical Health Research Group, Department of Functional Sciences, Faculty of Medicine, Western São Paulo University (UNOESTE), São Paulo, Brazil
| | - Rogéria Keller
- Food, Nutrition and Basic, Experimental and Clinical Health Research Group, Department of Functional Sciences, Faculty of Medicine, Western São Paulo University (UNOESTE), São Paulo, Brazil
| | - Hermann Bremer-Neto
- Food, Nutrition and Basic, Experimental and Clinical Health Research Group, Department of Functional Sciences, Faculty of Medicine, Western São Paulo University (UNOESTE), São Paulo, Brazil
| |
Collapse
|
3
|
Al-Adham ISI, Agha ASAA, Al-Akayleh F, Al-Remawi M, Jaber N, Al Manasur M, Collier PJ. Prebiotics Beyond the Gut: Omics Insights, Artificial Intelligence, and Clinical Trials in Organ-Specific Applications. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10465-x. [PMID: 39878922 DOI: 10.1007/s12602-025-10465-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2025] [Indexed: 01/31/2025]
Abstract
Prebiotics, traditionally linked to gut health, are increasingly recognized for their systemic benefits, influencing multiple organ systems through interactions with the gut microbiota. Compounds like inulin, fructooligosaccharides (FOS), and galactooligosaccharides (GOS) enhance short-chain fatty acid (SCFA) production, benefiting neurocognitive health, cardiovascular function, immune modulation, and skin integrity. Advances in biotechnology, including deep eutectic solvents (DES) for extraction and machine learning (ML) for personalized formulations, have expanded prebiotic applications. Integrating these innovations with "omics" technologies enables precise microbial modulation, fostering personalized nutrition and precision therapies. This review examines organ-specific effects of prebiotics, highlights findings from clinical trials, and explores biotechnological innovations that enhance prebiotic efficacy, laying the groundwork for future personalized therapeutic strategies.
Collapse
Affiliation(s)
- Ibrahim S I Al-Adham
- Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, 11196, Jordan.
| | - Ahmed S A Ali Agha
- School of Pharmacy, Department of Pharmaceutical Sciences, The University of Jordan, Amman, 11942, Jordan
| | - Faisal Al-Akayleh
- Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, 11196, Jordan
| | - Mayyas Al-Remawi
- Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, 11196, Jordan
| | - Nisrein Jaber
- Faculty of Pharmacy, Al Zaytoonah University of Jordan, Amman, 11733, Jordan
| | - Manar Al Manasur
- Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, 11196, Jordan
| | - Phillip J Collier
- Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, 11196, Jordan.
| |
Collapse
|
4
|
Dehghani A, Wang L, Garssen J, Styla E, Leusink-Muis T, Van Ark I, Folkerts G, Van Bergenhenegouwen J, Braber S. Synbiotics, a promising approach for alleviating exacerbated allergic airway immune responses in offspring of a preclinical murine pollution model. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 112:104591. [PMID: 39577477 DOI: 10.1016/j.etap.2024.104591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 11/13/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
Exposure to pollutants like environmental cigarette smoke (CS) poses a major global health risk, affecting individuals from an early age. Therefore, this study explores how postnatal synbiotic supplementation affects allergic asthma symptoms in house-dust-mite (HDM)- challenged offspring maternally exposed to CS. In HDM-allergic offspring of CS-exposed dams, lung resistance was elevated, but synbiotic supplementation effectively reduced this resistance. Elevated eosinophil BALF counts following HDM challenge were intensified in pups maternally exposed to CS. Similarly, Th2 cell activation and serum IgE and IgG1 levels were more pronounced in HDM-allergic offspring of CS-exposed mothers. Synbiotics reduced eosinophil numbers and serum IgE and IgG1, and tended to decrease Th2 cell infiltration and activation. Synbiotics promoted beneficial gut bacteria like Bifidobacterium and Akkermansia. In conclusion, early-life synbiotic intervention mitigated allergic asthma associated with maternal air pollution exposure, highlighting the potential of synbiotics for clinical evaluation as a strategy to prevent allergy development in offspring.
Collapse
Affiliation(s)
- Ali Dehghani
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands; Department of Public and Occupational Health, Amsterdam University of Medical Science, Amsterdam, Netherlands
| | - Lei Wang
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands; Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands; Danone Nutricia Research, Utrecht, Netherlands
| | - Eirini Styla
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Thea Leusink-Muis
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Ingrid Van Ark
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Jeroen Van Bergenhenegouwen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands; Danone Nutricia Research, Utrecht, Netherlands
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands; Danone Nutricia Research, Utrecht, Netherlands.
| |
Collapse
|
5
|
Lee SH, Lee JH, Lee SW. Application of Microbiome-Based Therapies in Chronic Respiratory Diseases. J Microbiol 2024; 62:201-216. [PMID: 38635003 DOI: 10.1007/s12275-024-00124-1] [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: 12/14/2023] [Revised: 02/02/2024] [Accepted: 02/16/2024] [Indexed: 04/19/2024]
Abstract
The application of microbiome-based therapies in various areas of human disease has recently increased. In chronic respiratory disease, microbiome-based clinical applications are considered compelling options due to the limitations of current treatments. The lung microbiome is ecologically dynamic and affected by various conditions, and dysbiosis is associated with disease severity, exacerbation, and phenotype as well as with chronic respiratory disease endotype. However, it is not easy to directly modulate the lung microbiome. Additionally, studies have shown that chronic respiratory diseases can be improved by modulating gut microbiome and administrating metabolites. Although the composition, diversity, and abundance of the microbiome between the gut and lung are considerably different, modulation of the gut microbiome could improve lung dysbiosis. The gut microbiome influences that of the lung via bacterial-derived components and metabolic degradation products, including short-chain fatty acids. This phenomenon might be associated with the cross-talk between the gut microbiome and lung, called gut-lung axis. There are multiple alternatives to modulate the gut microbiome, such as prebiotics, probiotics, and postbiotics ingestion and fecal material transplantation. Several studies have shown that high-fiber diets, for example, present beneficial effects through the production of short-chain fatty acids. Additionally, genetically modified probiotics to secrete some beneficial molecules might also be utilized to treat chronic respiratory diseases. Further studies on microbial modulation to regulate immunity and potentiate conventional pharmacotherapy will improve microbiome modulation techniques, which will develop as a new therapeutic area in chronic respiratory diseases.
Collapse
Affiliation(s)
- Se Hee Lee
- Department of Pulmonology, Allergy and Critical Care Medicine, CHA Bundang Medical Center, CHA University, Seongnam, 13496, Republic of Korea
| | - Jang Ho Lee
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Sei Won Lee
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.
| |
Collapse
|
6
|
Sulaiman I, Okwuofu EO, Mohtarrudin N, Lim JCW, Stanslas J. An Andrographis paniculata Burm. Nees extract standardized for three main Andrographolides prevents house dust mite-induced airway inflammation, remodeling, and hyperreactivity by regulating Th1/Th2 gene expression in mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117082. [PMID: 37652197 DOI: 10.1016/j.jep.2023.117082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 09/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Andrographis paniculata Burm. Nees (AP) is an herb used traditionally in Indian and Chinese traditional medicine for the treatment of various inflammatory and respiratory tract diseases. However, the anti-inflammatory potential of standardized Andrographis paniculata 50% ethanol extract (APEE50) in the murine model of asthma has not been investigated. AIM OF THE STUDY This study aimed to evaluate the protective anti-inflammatory potential and better understand the underlying mechanism of action of APEE50 in a clinically-relevant mouse asthma model. Thereafter, develop the ethanolic extract of AP as a supplement for asthma prophylaxis. MATERIALS AND METHOD APEE50 was prepared and standardized for AGP, NAG, and DDAG using a high-performance liquid chromatography system. Asthma was induced according to a 14-day house dust mite (HDM) induction protocol. The prophylactic potential of APEE50 (50 mg/kg - 200 mg/kg) was determined by assessing cardinal asthma features, which included BALF leukocyte and differential cell count, BALF cytokine assay, histology, gene expression, and airway hyperreactivity study. RESULTS APEE50 significantly inhibited HDM-induced airway eosinophilia and neutrophilia. In addition to decreased levels of IL-4, IL-5, IL-13, and eotaxin in bronchoalveolar fluid, APEE50 abrogated HDM-induced airway mucus over-secretion and airway hyper-responsiveness. Administration of APEE50 downregulated HDM-induced upregulation of the oxidative stress enzyme Duox1 (dual oxidase 1) and marginally induced Nfe2l2 (nuclear factor erythroid 2-related factor 2) gene expressions. Similarly, Th2-related (Serpinb2, Clca3a1, Il4 and Il13) and Muc5ac gene expression were significantly downregulated. CONCLUSION Prophylactic administration of APEE50 prevented the progression of HDM-induced asthmatic responses by down-regulating Th2 cytokine gene expression and oxidative stress level.
Collapse
Affiliation(s)
- Ibrahim Sulaiman
- Pharmacotherapeutic Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Emmanuel Oshiogwe Okwuofu
- Pharmacotherapeutic Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Norhafizah Mohtarrudin
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Jonathan Chee Woei Lim
- Pharmacotherapeutic Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Johnson Stanslas
- Pharmacotherapeutic Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia.
| |
Collapse
|
7
|
Yin Z, Liu X, Guo L, Ren M, Kang W, Ma C, Waterhouse GIN, Sun-Waterhouse D. The potential of dietary fiber in building immunity against gastrointestinal and respiratory disorders. Crit Rev Food Sci Nutr 2023; 64:13318-13336. [PMID: 37837407 DOI: 10.1080/10408398.2023.2266462] [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] [Indexed: 10/16/2023]
Abstract
The numerous health benefits of dietary fibers (DFs) justify their inclusion in human diets and biomedical products. Given the short- and long-term human impacts of the COVID-19 virus on human health, the potential of DFs in building immunity against gastrointestinal and respiratory disorders is currently receiving high attention. This paper reviews the physicochemical properties of DFs, together with their immune functions and effects on the gastrointestinal tract and respiratory system mainly based on research in the last ten years. Possible modes of action of DFs in promoting health, especially building immunity, are explored. We seek to highlight the importance of understanding the exact physical and chemical characteristics and molecular behaviors of DFs in providing specific immune function. This review provides a perspective beyond the existing recognition of DFs' positive effects on human health, and offers a theoretical framework for the development of special DFs components and their application in functional foods and other therapeutic products against gastrointestinal and respiratory disorders. DFs enhance immunity from gastrointestinal and respiratory diseases to promote host health.
Collapse
Affiliation(s)
- Zhenhua Yin
- National R &D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Comprehensive Utilization of Edible and Medicinal Plant Resources Engineering Technology Research Center, Huanghe Science and Technology College, Zhengzhou, China
- Function Food Engineering Technology Research Center, Kaifeng, China
| | - Xiaopeng Liu
- National R &D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Function Food Engineering Technology Research Center, Kaifeng, China
| | - Lin Guo
- National R &D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Function Food Engineering Technology Research Center, Kaifeng, China
| | - Mengjie Ren
- National R &D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Function Food Engineering Technology Research Center, Kaifeng, China
| | - Wenyi Kang
- National R &D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Function Food Engineering Technology Research Center, Kaifeng, China
| | - Changyang Ma
- National R &D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Function Food Engineering Technology Research Center, Kaifeng, China
| | | | | |
Collapse
|
8
|
Illidi CR, Romer LM, Johnson MA, Williams NC, Rossiter HB, Casaburi R, Tiller NB. Distinguishing science from pseudoscience in commercial respiratory interventions: an evidence-based guide for health and exercise professionals. Eur J Appl Physiol 2023; 123:1599-1625. [PMID: 36917254 PMCID: PMC10013266 DOI: 10.1007/s00421-023-05166-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/19/2023] [Indexed: 03/16/2023]
Abstract
Respiratory function has become a global health priority. Not only is chronic respiratory disease a leading cause of worldwide morbidity and mortality, but the COVID-19 pandemic has heightened attention on respiratory health and the means of enhancing it. Subsequently, and inevitably, the respiratory system has become a target of the multi-trillion-dollar health and wellness industry. Numerous commercial, respiratory-related interventions are now coupled to therapeutic and/or ergogenic claims that vary in their plausibility: from the reasonable to the absurd. Moreover, legitimate and illegitimate claims are often conflated in a wellness space that lacks regulation. The abundance of interventions, the range of potential therapeutic targets in the respiratory system, and the wealth of research that varies in quality, all confound the ability for health and exercise professionals to make informed risk-to-benefit assessments with their patients and clients. This review focuses on numerous commercial interventions that purport to improve respiratory health, including nasal dilators, nasal breathing, and systematized breathing interventions (such as pursed-lips breathing), respiratory muscle training, canned oxygen, nutritional supplements, and inhaled L-menthol. For each intervention we describe the premise, examine the plausibility, and systematically contrast commercial claims against the published literature. The overarching aim is to assist health and exercise professionals to distinguish science from pseudoscience and make pragmatic and safe risk-to-benefit decisions.
Collapse
Affiliation(s)
- Camilla R Illidi
- Clinical Exercise and Respiratory Physiology Laboratory, Department of Kinesiology and Physical Education, Faculty of Education, McGill University, Montréal, QC, Canada
| | - Lee M Romer
- Division of Sport, Health and Exercise Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UK
| | - Michael A Johnson
- Exercise and Health Research Group, Sport, Health and Performance Enhancement (SHAPE) Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, Nottinghamshire, UK
| | - Neil C Williams
- Exercise and Health Research Group, Sport, Health and Performance Enhancement (SHAPE) Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, Nottinghamshire, UK
| | - Harry B Rossiter
- Institute of Respiratory Medicine and Exercise Physiology, Division of Respiratory and Critical Care Physiology and Medicine, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, 1124 W. Carson Street, CDCRC Building, Torrance, CA, 90502, USA
| | - Richard Casaburi
- Institute of Respiratory Medicine and Exercise Physiology, Division of Respiratory and Critical Care Physiology and Medicine, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, 1124 W. Carson Street, CDCRC Building, Torrance, CA, 90502, USA
| | - Nicholas B Tiller
- Institute of Respiratory Medicine and Exercise Physiology, Division of Respiratory and Critical Care Physiology and Medicine, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, 1124 W. Carson Street, CDCRC Building, Torrance, CA, 90502, USA.
| |
Collapse
|
9
|
Bozorgmehr T, Boutin RCT, Woodward SE, Donald K, Chow JM, Buck RH, Finlay BB. Early Life Exposure to Human Milk Oligosaccharides Reduces Allergic Response in a Murine Asthma Model. J Immunol Res 2023; 2023:9603576. [PMID: 37545544 PMCID: PMC10404156 DOI: 10.1155/2023/9603576] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/27/2023] [Accepted: 06/29/2023] [Indexed: 08/08/2023] Open
Abstract
Background Studies suggest that early-life gut microbiota composition and intestinal short-chain fatty acids (SCFAs) are linked to future asthma susceptibility. Furthermore, infancy offers a critical time window to modulate the microbiota and associated metabolites through diet-microbe interactions to promote infant health. Human milk oligosaccharides (HMOs), nondigestible carbohydrates abundant in breast milk, are prebiotics selectively metabolized by gut microbiota that consequently modify microbiome composition and SCFA production. Methods Using a house dust mite mouse model of allergy, we investigated the impacts of early oral treatment of pups with biologically relevant doses of 2'-fucosyllactose (2'-FL) and 6'-sialyllactose (6'-SL), two of the most abundant HMOs in human milk, in amelioration of allergic airway disease severity. Results We found that administration of 2'-FL and 6'-SL during early life reduced lung histopathology scores, circulating IgE, cytokine levels, and inflammatory cell infiltration, all hallmark symptoms of allergic asthma. HMO supplementation also increased the relative abundance of intestinal Bacteroidetes and Clostridia, known SCFA producers within the gut. Indeed, we detected increased SCFA concentrations in both the intestine and blood of adult mice who received HMOs prior to weaning. Conclusion We propose a model in which orally administered HMOs delivered during early life shift the microbiota toward increased production of SCFAs, which dampens the allergic immune responses behind allergy and asthma. Overall, these data suggest the potential for HMO supplementation to protect infants against asthma development later in life, with possible benefits against additional atopic diseases such as eczema and food allergies.
Collapse
Affiliation(s)
- Tahereh Bozorgmehr
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Rozlyn C. T. Boutin
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Sarah E. Woodward
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Katherine Donald
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Jo May Chow
- Nutrition Division, Abbott Laboratories, Columbus, OH, USA
| | | | - B. Brett Finlay
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
10
|
Shen Y, Song M, Wu S, Zhao H, Zhang Y. Plant-Based Dietary Fibers and Polysaccharides as Modulators of Gut Microbiota in Intestinal and Lung Inflammation: Current State and Challenges. Nutrients 2023; 15:3321. [PMID: 37571257 PMCID: PMC10420973 DOI: 10.3390/nu15153321] [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: 06/26/2023] [Revised: 07/20/2023] [Accepted: 07/23/2023] [Indexed: 08/13/2023] Open
Abstract
Recent research has underscored the significant role of gut microbiota in managing various diseases, including intestinal and lung inflammation. It is now well established that diet plays a crucial role in shaping the composition of the microbiota, leading to changes in metabolite production. Consequently, dietary interventions have emerged as promising preventive and therapeutic approaches for managing these diseases. Plant-based dietary fibers, particularly polysaccharides and oligosaccharides, have attracted attention as potential therapeutic agents for modulating gut microbiota and alleviating intestinal and lung inflammation. This comprehensive review aims to provide an in-depth overview of the current state of research in this field, emphasizing the challenges and limitations associated with the use of plant-based dietary fibers and polysaccharides in managing intestinal and lung inflammation. By shedding light on existing issues and limitations, this review seeks to stimulate further research and development in this promising area of therapeutic intervention.
Collapse
Affiliation(s)
- Yu Shen
- Heilongjiang Provincial Key Laboratory of New Drug Development and Pharmacotoxicological Evaluation, College of Pharmacy, Jiamusi University, Jiamusi 154007, China; (Y.S.)
| | - Mingming Song
- Heilongjiang Provincial Key Laboratory of New Drug Development and Pharmacotoxicological Evaluation, College of Pharmacy, Jiamusi University, Jiamusi 154007, China; (Y.S.)
| | - Shihao Wu
- Heilongjiang Provincial Key Laboratory of New Drug Development and Pharmacotoxicological Evaluation, College of Pharmacy, Jiamusi University, Jiamusi 154007, China; (Y.S.)
| | - Hongbo Zhao
- College of Rehabilitation Medicine, Jiamusi University, Jiamusi 154007, China
| | - Yu Zhang
- Heilongjiang Provincial Key Laboratory of New Drug Development and Pharmacotoxicological Evaluation, College of Pharmacy, Jiamusi University, Jiamusi 154007, China; (Y.S.)
| |
Collapse
|
11
|
Jin Z, Sheng H, Wang S, Wang Y, Cheng Y. Network pharmacology study to reveal active compounds of Qinggan Yin formula against pulmonary inflammation by inhibiting MAPK activation. JOURNAL OF ETHNOPHARMACOLOGY 2022; 296:115513. [PMID: 35779819 DOI: 10.1016/j.jep.2022.115513] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/21/2022] [Accepted: 06/26/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Pneumonia is common and frequently-occurred disease caused by pathogens which predisposes to lung parenchymal inflammation leading pulmonary dysfunction. To prevent and alleviate the symptoms of pneumonia, Qinggan Yin formula (QGY) was composed based on clinical experience and four classical traditional Chinese medicine prescriptions which frequently applied to treat infectious diseases. AIM OF THE STUDY Traditional Chinese medicine is a complex mixture and it is difficult to distinguish the effective component molecules. The aim of this study is to identify the compounds of QGY with anti-inflammatory effects and investigate the molecular mechanism. MATERIALS AND METHODS The high-resolution mass spectrometry and molecular networking were performed for comprehensive chemical profiling of QGY. Network pharmacology was used to generate "herbal-target-pathway" network for target predictions. The anti-inflammation effects of QGY were evaluated in mice model of lipopolysaccharide (LPS) induced acute inflammation. Tail transected zebrafish was also employed to validate macrophage migration reversed effect of QGY. Based on the molecular enrichment analysis, the active substances of QGY with anti-inflammatory effects were further identified in cellular model of macrophage activation. The mechanisms of active substances were investigated by testing their effects on the expression of correlated proteins by Western blot. RESULTS In total, 71 compounds are identified as major substances of QGY. According to the results of network pharmacology, QGY shows moderate anti-inflammatory effects and inhibit pulmonary injury. MAPK signaling pathway was predicted as the most related pathway regulated by QGY. Moreover, QGY significantly inhibit LPS-induced pulmonary inflammation in mice, and reversed macrophage migration toward the injury site in zebrafish. We also validate that some major compounds in QGY significantly attenuated the release of IL-1β, IL-6 and TNF-α in LPS-stimulated macrophage. Those active substances including acacetin and arctiin can inhibit the phosphorylation of ERK/JNK and down-regulated the protein expression of BCL-2. CONCLUSION Collectively, QGY possessed pronounced anti-inflammation effects. The integration of network pharmacology and experimental results indicated arctiin, iridin, acacetin, liquiritin, and arctigenin are major active substances of QGY with anti-inflammatory effects. The underlying mechanism of QGY involves MAPK signaling pathway and oxidative stress pathway.
Collapse
Affiliation(s)
- Zehua Jin
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hongda Sheng
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Shufang Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yi Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, China.
| | - Yiyu Cheng
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
12
|
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
|
13
|
Janbazacyabar H, van Bergenhenegouwen J, Garssen J, Leusink-Muis T, van Ark I, van Daal MT, Folkerts G, Braber S. Prenatal and Postnatal Cigarette Smoke Exposure Is Associated With Increased Risk of Exacerbated Allergic Airway Immune Responses: A Preclinical Mouse Model. Front Immunol 2022; 12:797376. [PMID: 35003121 PMCID: PMC8732376 DOI: 10.3389/fimmu.2021.797376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/07/2021] [Indexed: 01/29/2023] Open
Abstract
Increased exposure to household air pollution and ambient air pollution has become one of the world’s major environmental health threats. In developing and developed countries, environmental cigarette smoke (CS) exposure is one of the main sources of household air pollution (HAP). Moreover, results from different epidemiological and experimental studies indicate that there is a strong association between HAP, specifically CS exposure, and the development of allergic diseases that often persists into later life. Here, we investigated the impact of prenatal and postnatal CS exposure on offspring susceptibility to the development of allergic airway responses by using a preclinical mouse model. Pregnant BALB/c mice were exposed to either CS or air during pregnancy and lactation and in order to induce allergic asthma the offspring were sensitized and challenged with house dust mite (HDM). Decreased lung function parameters, like dynamic compliance and pleural pressure, were observed in PBS-treated offspring born to CS-exposed mothers compared to offspring from air-exposed mothers. Maternal CS exposure significantly increased the HDM-induced airway eosinophilia and neutrophilia in the offspring. Prenatal and postnatal CS exposure increased the frequency of Th2 cells in the lungs of HDM-treated offspring compared to offspring born to air-exposed mothers. Offspring born to CS-exposed mothers showed increased levels of IL-4, IL-5 and IL-13 in bronchoalveolar lavage fluid compared to offspring from air-exposed mothers. Ex-vivo restimulation of lung cells isolated from HDM-treated offspring born to CS-exposed mothers also resulted in increased IL-4 production. Finally, serum immunoglobulins levels of HDM-specific IgE and HDM-specific IgG1 were significantly increased upon a HDM challenge in offspring born to CS-exposed mothers compared to offspring from air-exposed mothers. In summary, our results reveal a biological plausibility for the epidemiological studies indicating that prenatal and postnatal CS exposure increases the susceptibility of offspring to allergic immune responses.
Collapse
Affiliation(s)
- Hamed Janbazacyabar
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Jeroen van Bergenhenegouwen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands.,Global Center of Excellence Immunology, Danone Nutricia Research, Utrecht, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands.,Global Center of Excellence Immunology, Danone Nutricia Research, Utrecht, Netherlands
| | - Thea Leusink-Muis
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Ingrid van Ark
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Marthe T van Daal
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
14
|
Lad N, Murphy A, Parenti C, Nelson C, Williams N, Sharpe G, McTernan P. Asthma and obesity: endotoxin another insult to add to injury? Clin Sci (Lond) 2021; 135:2729-2748. [PMID: 34918742 PMCID: PMC8689194 DOI: 10.1042/cs20210790] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 11/29/2021] [Accepted: 12/06/2021] [Indexed: 12/20/2022]
Abstract
Low-grade inflammation is often an underlying cause of several chronic diseases such as asthma, obesity, cardiovascular disease, and type 2 diabetes mellitus (T2DM). Defining the mediators of such chronic low-grade inflammation often appears dependent on which disease is being investigated. However, downstream systemic inflammatory cytokine responses in these diseases often overlap, noting there is no doubt more than one factor at play to heighten the inflammatory response. Furthermore, it is increasingly believed that diet and an altered gut microbiota may play an important role in the pathology of such diverse diseases. More specifically, the inflammatory mediator endotoxin, which is a complex lipopolysaccharide (LPS) derived from the outer membrane cell wall of Gram-negative bacteria and is abundant within the gut microbiota, and may play a direct role alongside inhaled allergens in eliciting an inflammatory response in asthma. Endotoxin has immunogenic effects and is sufficiently microscopic to traverse the gut mucosa and enter the systemic circulation to act as a mediator of chronic low-grade inflammation in disease. Whilst the role of endotoxin has been considered in conditions of obesity, cardiovascular disease and T2DM, endotoxin as an inflammatory trigger in asthma is less well understood. This review has sought to examine the current evidence for the role of endotoxin in asthma, and whether the gut microbiota could be a dietary target to improve disease management. This may expand our understanding of endotoxin as a mediator of further low-grade inflammatory diseases, and how endotoxin may represent yet another insult to add to injury.
Collapse
Affiliation(s)
- Nikita Lad
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Alice M. Murphy
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Cristina Parenti
- SHAPE Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Carl P. Nelson
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Neil C. Williams
- SHAPE Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Graham R. Sharpe
- SHAPE Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| | - Philip G. McTernan
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, U.K
| |
Collapse
|
15
|
Dietary Fibers: Effects, Underlying Mechanisms and Possible Role in Allergic Asthma Management. Nutrients 2021; 13:nu13114153. [PMID: 34836408 PMCID: PMC8621630 DOI: 10.3390/nu13114153] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/10/2021] [Accepted: 11/18/2021] [Indexed: 12/13/2022] Open
Abstract
The prevalence of asthma is increasing, but the cause remains under debate. Research currently focuses on environmental and dietary factors that may impact the gut-lung axis. Dietary fibers are considered to play a crucial role in supporting diversity and activity of the microbiome, as well as immune homeostasis in the gut and lung. This review discusses the current state of knowledge on how dietary fibers and their bacterial fermentation products may affect the pathophysiology of allergic asthma. Moreover, the impact of dietary fibers on early type 2 asthma management, as shown in both pre-clinical and clinical studies, is described. Short-chain fatty acids, fiber metabolites, modulate host immunity and might reduce the risk of allergic asthma development. Underlying mechanisms include G protein-coupled receptor activation and histone deacetylase inhibition. These results are supported by studies in mice, children and adults with allergic asthma. Fibers might also exert direct effects on the immune system via yet to be elucidated mechanisms. However, the effects of specific types of fiber, dosages, duration of treatment, and combination with probiotics, need to be explored. There is an urgent need to further valorize the potential of specific dietary fibers in prevention and treatment of allergic asthma by conducting more large-scale dietary intervention trials.
Collapse
|
16
|
Anti-Inflammatory Properties of Fructo-Oligosaccharides in a Calf Lung Infection Model and in Mannheimia haemolytica-Infected Airway Epithelial Cells. Nutrients 2021; 13:nu13103514. [PMID: 34684515 PMCID: PMC8537102 DOI: 10.3390/nu13103514] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/02/2021] [Accepted: 10/04/2021] [Indexed: 01/18/2023] Open
Abstract
Emerging antimicrobial-resistant pathogens highlight the importance of developing novel interventions. Here, we investigated the anti-inflammatory properties of Fructo-oligosaccharides (FOS) in calf lung infections and in airway epithelial cells stimulated with pathogens, and/or bacterial components. During a natural exposure, 100 male calves were fed milk replacer with or without FOS for 8 weeks. Then, immune parameters and cytokine/chemokine levels in the bronchoalveolar lavage fluid (BALF) and blood were measured, and clinical scores were investigated. Calf primary bronchial epithelial cells (PBECs) and human airway epithelial cells (A549) were treated with Mannheimia haemolytica, lipopolysaccharides (LPS), and/or flagellin, with or without FOS pretreatment. Thereafter, the cytokine/chemokine levels and epithelial barrier function were examined. Relative to the control (naturally occurring lung infections), FOS-fed calves had greater macrophage numbers in BALF and lower interleukin (IL)-8, IL-6, and IL-1β concentrations in the BALF and blood. However, FOS did not affect the clinical scores. At slaughter, FOS-fed calves had a lower severity of lung lesions compared to the control. Ex vivo, FOS prevented M. haemolytica-induced epithelial barrier dysfunction. Moreover, FOS reduced M. haemolytica- and flagellin-induced (but not LPS-induced) IL-8, TNF-α, and IL-6 release in PBECs and A549 cells. Overall, FOS had anti-inflammatory properties during the natural incidence of lung infections but had no effects on clinical symptoms.
Collapse
|
17
|
Bottemanne P, Paquot A, Ameraoui H, Guillemot-Legris O, Alhouayek M, Muccioli GG. 25-Hydroxycholesterol metabolism is altered by lung inflammation, and its local administration modulates lung inflammation in mice. FASEB J 2021; 35:e21514. [PMID: 33734509 DOI: 10.1096/fj.202002555r] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/18/2021] [Accepted: 02/24/2021] [Indexed: 12/12/2022]
Abstract
Inflammation is a critical component of many lung diseases including asthma and acute lung injury (ALI). Using high-performance liquid chromatography-mass spectrometry, we quantified the levels of oxysterols in two different murine models of lung diseases. These are lipid mediators derived from cholesterol and known to modulate immunity and inflammation. Interestingly, 25-hydroxycholesterol (25-OHC) was the only oxysterol with altered levels during lung inflammation, and its levels were differently affected according to the model. Therefore, we sought to assess how this oxysterol would affect lung inflammatory responses. In a model of lipopolysaccharide (LPS)-induced acute lung inflammation, 25-OHC levels were increased, and most of the hallmarks of the model (eg, leukocyte recruitment, mRNA expression, and secretion of inflammatory cytokines) were decreased following its intratracheal administration. We also found that, when administered in the lung, 25-OHC is metabolized locally into 25-hydroxycholesterol-3-sulfate and 7α,25-dihydroxycholesterol. Their administration in the lungs did not recapitulate all the effects of 25-OHC. Conversely, in a model of allergic asthma induced by intranasal administration of house dust mites (HDM), 25-OHC levels were decreased, and when intranasally administered, this oxysterol worsened the hallmarks of the model (eg, leukocyte recruitment, tissue remodeling [epithelium thickening and peribranchial fibrosis], and cytokine expression) and induced changes in leukotriene levels. Ex vivo, we found that 25-OHC decreases LPS-induced primary alveolar macrophage activation while having no effect on neutrophil activation. Its sulfated metabolite, 25-hydroxycholesterol-3-sulfate, decreased neutrophil, but not macrophage activation. Taken together, our data support a differential role of 25-OHC in ALI and allergic inflammation models.
Collapse
Affiliation(s)
- Pauline Bottemanne
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Adrien Paquot
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Hafsa Ameraoui
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Owein Guillemot-Legris
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Mireille Alhouayek
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
18
|
Wang L, Pelgrim CE, Swart DH, Krenning G, van der Graaf AC, Kraneveld AD, Leusink-Muis T, van Ark I, Garssen J, Folkerts G, Braber S. SUL-151 Decreases Airway Neutrophilia as a Prophylactic and Therapeutic Treatment in Mice after Cigarette Smoke Exposure. Int J Mol Sci 2021; 22:4991. [PMID: 34066693 PMCID: PMC8125869 DOI: 10.3390/ijms22094991] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/01/2021] [Accepted: 05/06/2021] [Indexed: 12/31/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) caused by cigarette smoke (CS) is featured by oxidative stress and chronic inflammation. Due to the poor efficacy of standard glucocorticoid therapy, new treatments are required. Here, we investigated whether the novel compound SUL-151 with mitoprotective properties can be used as a prophylactic and therapeutic treatment in a murine CS-induced inflammation model. SUL-151 (4 mg/kg), budesonide (500 μg/kg), or vehicle were administered via oropharyngeal instillation in this prophylactic and therapeutic treatment setting. The number of immune cells was determined in the bronchoalveolar lavage fluid (BALF). Oxidative stress response, mitochondrial adenosine triphosphate (ATP) production, and mitophagy-related proteins were measured in lung homogenates. SUL-151 significantly decreased more than 70% and 50% of CS-induced neutrophils in BALF after prophylactic and therapeutic administration, while budesonide showed no significant reduction in neutrophils. Moreover, SUL-151 prevented the CS-induced decrease in ATP and mitochondrial mtDNA and an increase in putative protein kinase 1 expression in the lung homogenates. The concentration of SUL-151 was significantly correlated with malondialdehyde level and radical scavenging activity in the lungs. SUL-151 inhibited the increased pulmonary inflammation and mitochondrial dysfunction in this CS-induced inflammation model, which implied that SUL-151 might be a promising candidate for COPD treatment.
Collapse
Affiliation(s)
- Lei Wang
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (L.W.); (C.E.P.); (A.D.K.); (T.L.-M.); (I.v.A.); (J.G.); (G.F.)
| | - Charlotte E. Pelgrim
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (L.W.); (C.E.P.); (A.D.K.); (T.L.-M.); (I.v.A.); (J.G.); (G.F.)
| | - Daniël H. Swart
- Sulfateq B.V., Admiraal de Ruyterlaan 5, 9726 GN Groningen, The Netherlands; (D.H.S.); (G.K.); (A.C.v.d.G.)
| | - Guido Krenning
- Sulfateq B.V., Admiraal de Ruyterlaan 5, 9726 GN Groningen, The Netherlands; (D.H.S.); (G.K.); (A.C.v.d.G.)
- Cardiovascular Regenerative Medicine, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Adrianus C. van der Graaf
- Sulfateq B.V., Admiraal de Ruyterlaan 5, 9726 GN Groningen, The Netherlands; (D.H.S.); (G.K.); (A.C.v.d.G.)
| | - Aletta D. Kraneveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (L.W.); (C.E.P.); (A.D.K.); (T.L.-M.); (I.v.A.); (J.G.); (G.F.)
- Institute for Risk Assessment Sciences (IRAS), Utrecht University, 3584 CM Utrecht, The Netherlands
| | - Thea Leusink-Muis
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (L.W.); (C.E.P.); (A.D.K.); (T.L.-M.); (I.v.A.); (J.G.); (G.F.)
| | - Ingrid van Ark
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (L.W.); (C.E.P.); (A.D.K.); (T.L.-M.); (I.v.A.); (J.G.); (G.F.)
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (L.W.); (C.E.P.); (A.D.K.); (T.L.-M.); (I.v.A.); (J.G.); (G.F.)
- Nutricia Research, Department of Immunology, 3584 CT Utrecht, The Netherlands
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (L.W.); (C.E.P.); (A.D.K.); (T.L.-M.); (I.v.A.); (J.G.); (G.F.)
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (L.W.); (C.E.P.); (A.D.K.); (T.L.-M.); (I.v.A.); (J.G.); (G.F.)
| |
Collapse
|
19
|
Role of Short Chain Fatty Acids and Apolipoproteins in the Regulation of Eosinophilia-Associated Diseases. Int J Mol Sci 2021; 22:ijms22094377. [PMID: 33922158 PMCID: PMC8122716 DOI: 10.3390/ijms22094377] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/17/2021] [Accepted: 04/19/2021] [Indexed: 02/06/2023] Open
Abstract
Eosinophils are key components of our host defense and potent effectors in allergic and inflammatory diseases. Once recruited to the inflammatory site, eosinophils release their cytotoxic granule proteins as well as cytokines and lipid mediators, contributing to parasite clearance but also to exacerbation of inflammation and tissue damage. However, eosinophils have recently been shown to play an important homeostatic role in different tissues under steady state. Despite the tremendous progress in the treatment of eosinophilic disorders with the implementation of biologics, there is an unmet need for novel therapies that specifically target the cytotoxic effector functions of eosinophils without completely depleting this multifunctional immune cell type. Recent studies have uncovered several endogenous molecules that decrease eosinophil migration and activation. These include short chain fatty acids (SCFAs) such as butyrate, which are produced in large quantities in the gastrointestinal tract by commensal bacteria and enter the systemic circulation. In addition, high-density lipoprotein-associated anti-inflammatory apolipoproteins have recently been shown to attenuate eosinophil migration and activation. Here, we focus on the anti-pathogenic properties of SCFAs and apolipoproteins on eosinophil effector function and provide insights into the potential use of SCFAs and apolipoproteins (and their mimetics) as effective agents to combat eosinophilic inflammation.
Collapse
|
20
|
Ghiamati Yazdi F, Zakeri A, van Ark I, Leusink-Muis T, Braber S, Soleimanian-Zad S, Folkerts G. Crude Turmeric Extract Improves the Suppressive Effects of Lactobacillus rhamnosus GG on Allergic Inflammation in a Murine Model of House Dust Mite-Induced Asthma. Front Immunol 2020; 11:1092. [PMID: 32582180 PMCID: PMC7287160 DOI: 10.3389/fimmu.2020.01092] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 05/06/2020] [Indexed: 12/20/2022] Open
Abstract
There is a strong correlation between dysregulation of the gastrointestinal microbiota and development of allergic diseases. The most prevalent therapies for relieving asthma symptoms are associated with serious side effects, and therefore novel approaches are needed. Our objective was to elucidate whether oral administration of Lactobacillus rhamnosus GG (LGG) as a probiotic or turmeric powder (TP) as a prebiotic or both as a synbiotic mitigate allergic inflammation including lung function, airway inflammatory cell infiltration, Th2 cytokines/chemokine in a murine model of house dust mite (HDM)-induced asthma. BALB/c mice were intranasally sensitized and challenged with HDM received TP (20 mg/Kg mouse), or/and LGG (105 or 107 cfu/ml), or both orally. Interestingly, the synbiotic intervention (HDM-TP-LGG E7) specifically suppress the developement of airway hyperresponsiveness in response to methacholine. Besides, our synbiotic, TP, and LGG strongly down-regulated eosinophilia, IL-5, CCL17, IL-13. In terms of T cell response, CD4+ Th2 cells and CD4+ Th17 population were reduced in the splenocytes of the treatment groups compared to control. The synbiotic group not only elevated CD25+Foxp3+Treg frequency compared to asthmatic group, but also increased T reg cells compared to the probiotic group. The synbiotic also indicated the superior effect in suppressing Th2 cells compared to probiotic. Although, TP and LGG alone displayed suppressive effects, this study showed that the combination therapy consisting of TP and LGG (synbiotic) is more effective in some of the parameters than either of the treatments alone. This novel synbiotic, might be considered as a potential food-based drug for translational medicine and can possibly be used along with corticosteroid treatment.
Collapse
Affiliation(s)
- Fariba Ghiamati Yazdi
- Department of Food Science and Technology, College of Agriculture, Isfahan University of Technology (IUT), Isfahan, Iran.,Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Amin Zakeri
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Ingrid van Ark
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Thea Leusink-Muis
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Sabihe Soleimanian-Zad
- Department of Food Science and Technology, College of Agriculture, Isfahan University of Technology (IUT), Isfahan, Iran
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
21
|
Zuurveld M, van Witzenburg NP, Garssen J, Folkerts G, Stahl B, van't Land B, Willemsen LEM. Immunomodulation by Human Milk Oligosaccharides: The Potential Role in Prevention of Allergic Diseases. Front Immunol 2020; 11:801. [PMID: 32457747 PMCID: PMC7221186 DOI: 10.3389/fimmu.2020.00801] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 04/07/2020] [Indexed: 12/13/2022] Open
Abstract
The prevalence and incidence of allergic diseases is rising and these diseases have become the most common chronic diseases during childhood in Westernized countries. Early life forms a critical window predisposing for health or disease. Therefore, this can also be a window of opportunity for allergy prevention. Postnatally the gut needs to mature, and the microbiome is built which further drives the training of infant's immune system. Immunomodulatory components in breastmilk protect the infant in this crucial period by; providing nutrients that contain substrates for the microbiome, supporting intestinal barrier function, protecting against pathogenic infections, enhancing immune development and facilitating immune tolerance. The presence of a diverse human milk oligosaccharide (HMOS) mixture, containing several types of functional groups, points to engagement in several mechanisms related to immune and microbiome maturation in the infant's gastrointestinal tract. In recent years, several pathways impacted by HMOS have been elucidated, including their capacity to; fortify the microbiome composition, enhance production of short chain fatty acids, bind directly to pathogens and interact directly with the intestinal epithelium and immune cells. The exact mechanisms underlying the immune protective effects have not been fully elucidated yet. We hypothesize that HMOS may be involved in and can be utilized to provide protection from developing allergic diseases at a young age. In this review, we highlight several pathways involved in the immunomodulatory effects of HMOS and the potential role in prevention of allergic diseases. Recent studies have proposed possible mechanisms through which HMOS may contribute, either directly or indirectly, via microbiome modification, to induce oral tolerance. Future research should focus on the identification of specific pathways by which individual HMOS structures exert protective actions and thereby contribute to the capacity of the authentic HMOS mixture in early life allergy prevention.
Collapse
Affiliation(s)
- Marit Zuurveld
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Nikita P. van Witzenburg
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
- Global Centre of Excellence Immunology, Danone Nutricia Research B.V., Utrecht, Netherlands
| | - Gert Folkerts
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Bernd Stahl
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
- Global Centre of Excellence Human Milk Research and Analytical Sciences, Danone Nutricia Research B.V., Utrecht, Netherlands
- Division of Chemical Biology and Drug Discovery, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Belinda van't Land
- Global Centre of Excellence Immunology, Danone Nutricia Research B.V., Utrecht, Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Linette E. M. Willemsen
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
22
|
Janbazacyabar H, van Bergenhenegouwen J, Verheijden KA, Leusink-Muis T, van Helvoort A, Garssen J, Folkerts G, Braber S. Non-digestible oligosaccharides partially prevent the development of LPS-induced lung emphysema in mice. PHARMANUTRITION 2019. [DOI: 10.1016/j.phanu.2019.100163] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
23
|
Prebiotics: Mechanisms and Preventive Effects in Allergy. Nutrients 2019; 11:nu11081841. [PMID: 31398959 PMCID: PMC6722770 DOI: 10.3390/nu11081841] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 12/13/2022] Open
Abstract
Allergic diseases now affect over 30% of individuals in many communities, particularly young children, underscoring the need for effective prevention strategies in early life. These allergic conditions have been linked to environmental and lifestyle changes driving the dysfunction of three interdependent biological systems: microbiota, epithelial barrier and immune system. While this is multifactorial, dietary changes are of particular interest in the altered establishment and maturation of the microbiome, including the associated profile of metabolites that modulate immune development and barrier function. Prebiotics are non-digestible food ingredients that beneficially influence the health of the host by 1) acting as a fermentable substrate for some specific commensal host bacteria leading to the release of short-chain fatty acids in the gut intestinal tract influencing many molecular and cellular processes; 2) acting directly on several compartments and specifically on different patterns of cells (epithelial and immune cells). Nutrients with prebiotic properties are therefore of central interest in allergy prevention for their potential to promote a more tolerogenic environment through these multiple pathways. Both observational studies and experimental models lend further credence to this hypothesis. In this review, we describe both the mechanisms and the therapeutic evidence from preclinical and clinical studies exploring the role of prebiotics in allergy prevention.
Collapse
|
24
|
Abdel-Aziz MI, Vijverberg SJH, Neerincx AH, Kraneveld AD, Maitland-van der Zee AH. The crosstalk between microbiome and asthma: Exploring associations and challenges. Clin Exp Allergy 2019; 49:1067-1086. [PMID: 31148278 PMCID: PMC6852296 DOI: 10.1111/cea.13444] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 12/17/2022]
Abstract
With the advancement of high‐throughput DNA/RNA sequencing and computational analysis techniques, commensal bacteria are now considered almost as important as pathological ones. Understanding the interaction between these bacterial microbiota, host and asthma is crucial to reveal their role in asthma pathophysiology. Several airway and/or gut microbiome studies have shown associations between certain bacterial taxa and asthma. However, challenges remain before gained knowledge from these studies can be implemented into clinical practice, such as inconsistency between studies in choosing sampling compartments and/or sequencing approaches, variability of results in asthma studies, and not taking into account medication intake and diet composition especially when investigating gut microbiome. Overcoming those challenges will help to better understand the complex asthma disease process. The therapeutic potential of using pro‐ and prebiotics to prevent or reduce risk of asthma exacerbations requires further investigation. This review will focus on methodological issues regarding setting up a microbiome study, recent developments in asthma bacterial microbiome studies, challenges and future therapeutic potential.
Collapse
Affiliation(s)
- Mahmoud I Abdel-Aziz
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Clinical Pharmacy, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Susanne J H Vijverberg
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Anne H Neerincx
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Aletta D Kraneveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.,Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Anke H Maitland-van der Zee
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Pediatric Respiratory Medicine, Amsterdam UMC, Emma Children's Hospital, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Untersmayr E, Bax HJ, Bergmann C, Bianchini R, Cozen W, Gould HJ, Hartmann K, Josephs DH, Levi‐Schaffer F, Penichet ML, O'Mahony L, Poli A, Redegeld FA, Roth‐Walter F, Turner MC, Vangelista L, Karagiannis SN, Jensen‐Jarolim E. AllergoOncology: Microbiota in allergy and cancer-A European Academy for Allergy and Clinical Immunology position paper. Allergy 2019; 74:1037-1051. [PMID: 30636005 PMCID: PMC6563061 DOI: 10.1111/all.13718] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 12/27/2018] [Accepted: 12/28/2018] [Indexed: 12/18/2022]
Abstract
The microbiota can play important roles in the development of human immunity and the establishment of immune homeostasis. Lifestyle factors including diet, hygiene, and exposure to viruses or bacteria, and medical interventions with antibiotics or anti-ulcer medications, regulate phylogenetic variability and the quality of cross talk between innate and adaptive immune cells via mucosal and skin epithelia. More recently, microbiota and their composition have been linked to protective effects for health. Imbalance, however, has been linked to immune-related diseases such as allergy and cancer, characterized by impaired, or exaggerated immune tolerance, respectively. In this AllergoOncology position paper, we focus on the increasing evidence defining the microbiota composition as a key determinant of immunity and immune tolerance, linked to the risk for the development of allergic and malignant diseases. We discuss novel insights into the role of microbiota in disease and patient responses to treatments in cancer and in allergy. These may highlight opportunities to improve patient outcomes with medical interventions supported through a restored microbiome.
Collapse
Affiliation(s)
- Eva Untersmayr
- Institute of Pathophysiology and Allergy ResearchCenter of Pathophysiology, Infectiology and ImmunologyMedical University ViennaViennaAustria
| | - Heather J. Bax
- St. John's Institute of DermatologySchool of Basic & Medical BiosciencesKing's College LondonGuy's HospitalLondonUK
- School of Cancer and Pharmaceutical SciencesKing's College LondonGuy's HospitalLondonUK
| | | | - Rodolfo Bianchini
- Comparative MedicineThe Interuniversity Messerli Research InstituteUniversity of Veterinary Medicine ViennaMedical University ViennaUniversity ViennaViennaAustria
| | - Wendy Cozen
- Center for Genetic EpidemiologyDepartment of Preventive MedicineKeck School of Medicine of University of Southern CaliforniaLos AngelesCaliforniaUSA
- Department of PathologyKeck School of Medicine of University of Southern CaliforniaLos AngelesCaliforniaUSA
- Norris Comprehensive Cancer CenterKeck School of Medicine of Los AngelesLos AngelesCaliforniaUSA
| | - Hannah J. Gould
- Randall Centre for Cell and Molecular BiophysicsSchool of Basic & Medical BiosciencesKing's College LondonNew Hunt's HouseLondonUK
- Medical Research Council & Asthma UK Centre in Allergic Mechanisms of AsthmaLondonUK
| | - Karin Hartmann
- Department of DermatologyUniversity of LuebeckLuebeckGermany
| | - Debra H. Josephs
- St. John's Institute of DermatologySchool of Basic & Medical BiosciencesKing's College LondonGuy's HospitalLondonUK
- School of Cancer and Pharmaceutical SciencesKing's College LondonGuy's HospitalLondonUK
| | - Francesca Levi‐Schaffer
- Pharmacology and Experimental Therapeutics UnitSchool of PharmacyFaculty of MedicineThe Institute for Drug ResearchThe Hebrew University of JerusalemJerusalemIsrael
| | - Manuel L. Penichet
- Division of Surgical OncologyDepartment of SurgeryDavid Geffen School of MedicineUniversity of California, Los AngelesCaliforniaUSA
- Department of Microbiology, Immunology and Molecular GeneticsDavid Geffen School of MedicineUniversity of California, Los AngelesCaliforniaUSA
- Jonsson Comprehensive Cancer CenterUniversity of CaliforniaLos AngelesCaliforniaUSA
- The Molecular Biology InstituteUniversity of CaliforniaLos AngelesCaliforniaUSA
- UCLA AIDS InstituteLos AngelesCaliforniaUSA
| | - Liam O'Mahony
- Departments of Medicine and MicrobiologyAPC Microbiome IrelandNational University of IrelandCorkIreland
| | - Aurelie Poli
- Department of Infection and ImmunityLuxembourg Institute of HealthEsch‐sur‐AlzetteLuxembourg
| | - Frank A. Redegeld
- Division of PharmacologyFaculty of ScienceUtrecht Institute for Pharmaceutical SciencesUtrecht UniversityUtrechtThe Netherlands
| | - Franziska Roth‐Walter
- Comparative MedicineThe Interuniversity Messerli Research InstituteUniversity of Veterinary Medicine ViennaMedical University ViennaUniversity ViennaViennaAustria
| | - Michelle C. Turner
- Barcelona Institute for Global Health (ISGlobal)BarcelonaSpain
- Universitat Pompeu Fabra (UPF)BarcelonaSpain
- CIBER Epidemiología y Salud Pública (CIBERESP)MadridSpain
- McLaughlin Centre for Population Health Risk AssessmentUniversity of OttawaOttawaOntarioCanada
| | - Luca Vangelista
- Department of Biomedical SciencesNazarbayev University School of MedicineAstanaKazakhstan
| | - Sophia N. Karagiannis
- St. John's Institute of DermatologySchool of Basic & Medical BiosciencesKing's College LondonGuy's HospitalLondonUK
| | - Erika Jensen‐Jarolim
- Institute of Pathophysiology and Allergy ResearchCenter of Pathophysiology, Infectiology and ImmunologyMedical University ViennaViennaAustria
- Comparative MedicineThe Interuniversity Messerli Research InstituteUniversity of Veterinary Medicine ViennaMedical University ViennaUniversity ViennaViennaAustria
| |
Collapse
|
26
|
Verheijden KAT, Braber S, Leusink-Muis T, Jeurink PV, Thijssen S, Kraneveld AD, Garssen J, Folkerts G, Willemsen LEM. The Combination Therapy of Dietary Galacto-Oligosaccharides With Budesonide Reduces Pulmonary Th2 Driving Mediators and Mast Cell Degranulation in a Murine Model of House Dust Mite Induced Asthma. Front Immunol 2018; 9:2419. [PMID: 30405619 PMCID: PMC6207001 DOI: 10.3389/fimmu.2018.02419] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 10/01/2018] [Indexed: 12/20/2022] Open
Abstract
Background: Dietary non-digestible galacto-oligosaccharides (GOS) suppress allergic responses in mice sensitized and challenged with house dust mite (HDM). Budesonide is the standard therapy for allergic asthma in humans but is not always completely effective. Aim: To compare the efficacy of budesonide or different doses of GOS alone or with a combination therapy of budesonide and GOS on HDM-allergic responses in mice. Methods:BALB/c mice were sensitized and challenged with HDM, while fed a control diet or a diet supplemented with 1 or 2.5 w/w% GOS, and either or not oropharyngeally instilled with budesonide. Systemic and local inflammatory markers, such as mucosal mast cell protease-1 (mMCP-1) in serum, pulmonary CCL17, CCL22, and IL-33 concentrations and inflammatory cell influx in the bronchoalveolar lavage fluid (BALF) were determined. Results: Budesonide or GOS alone suppressed the number of eosinophils in the BALF of HDM allergic mice whereas budesonide either or not combined with GOS lowered both eosinophil and lymphocyte numbers in the BALF of HDM-allergic mice. Both 1 w/w% and 2.5 w/w% GOS and/or budesonide suppressed serum mMCP-1 concentrations. However, budesonide nor GOS alone was capable of reducing Th2 driving chemokines CCL17, CCL22 and IL-33 protein levels in supernatants of lung homogenates of HDM allergic mice, whereas the combination therapy did. Moreover, IL-13 concentrations were only significantly suppressed in mice treated with budesonide while fed GOS. A similar tendency was observed for the frequency of GATA3+CD4+ Th2 and CD4+RORγt+ Th17 cells in the lungs of the allergic mice. Conclusion: Dietary intervention using GOS may be a novel way to further improve the efficacy of anti-inflammatory drug therapy in allergic asthma by lowering Th2 driving mediators and mast cell degranulation.
Collapse
Affiliation(s)
- Kim A T Verheijden
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Department of Nephrology, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Saskia Braber
- Division of Veterinary Pharmacy, Pharmacology and Toxicology, Faculty of Veterinary Sciences, Utrecht University, Utrecht, Netherlands
| | - Thea Leusink-Muis
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | | | - Suzan Thijssen
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Aletta D Kraneveld
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Nutricia Research, Immunology, Utrecht, Netherlands
| | - Gert Folkerts
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Linette E M Willemsen
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
27
|
Strategies to reduce non-communicable diseases in the offspring: negative and positive in utero programming. J Dev Orig Health Dis 2018; 9:642-652. [PMID: 30111388 DOI: 10.1017/s2040174418000569] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Non-communicable diseases (NCDs) are a major problem as they are the leading cause of death and represent a substantial economic cost. The 'Developmental Origins of Health and Disease Hypothesis' proposes that adverse stimuli at different life stages can increase the predisposition to these diseases. In fact, adverse in utero programming is a major origin of these diseases due to the high malleability of embryonic development. This review provides a comprehensive analysis of the scientific literature on in utero programming and NCDs highlighting potential medical strategies to prevent these diseases based upon this programming. We fully address the concept and mechanisms involved in this programming (anatomical disruptions, epigenetic modifications and microbiota alterations). We also examine the negative role of in utero programming on the increased predisposition of NCDs in the offspring, which introduces the passive medical approach that consists of avoiding adverse stimuli including an unhealthy diet and environmental chemicals. Finally, we extensively discuss active medical approaches that target the causes of NCDs and have the potential to significantly and rapidly reduce the incidence of NCDs. These approaches can be classified as direct in utero programming modifications and personalized lifestyle pregnancy programs; they could potentially provide transgenerational NCDs protection. Active strategies against NCDs constitute a promising tool for the reduction in NCDs.
Collapse
|
28
|
Folkerts J, Stadhouders R, Redegeld FA, Tam SY, Hendriks RW, Galli SJ, Maurer M. Effect of Dietary Fiber and Metabolites on Mast Cell Activation and Mast Cell-Associated Diseases. Front Immunol 2018; 9:1067. [PMID: 29910798 PMCID: PMC5992428 DOI: 10.3389/fimmu.2018.01067] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 04/30/2018] [Indexed: 12/14/2022] Open
Abstract
Many mast cell-associated diseases, including allergies and asthma, have seen a strong increase in prevalence during the past decades, especially in Western(ized) countries. It has been suggested that a Western diet may contribute to the prevalence and manifestation of allergies and asthma through reduced intake of dietary fiber and the subsequent production of their metabolites. Indeed, dietary fiber and its metabolites have been shown to positively influence the development of immune disorders via changes in microbiota composition and the regulation of B- and T-cell activation. However, the effects of these dietary components on the activation of mast cells, key effector cells of the inflammatory response in allergies and asthma, remain poorly characterized. Due to their location in the gut and vascularized tissues, mast cells are exposed to high concentrations of dietary fiber and/or its metabolites. Here, we provide a focused overview of current findings regarding the direct effects of dietary fiber and its various metabolites on the regulation of mast cell activity and the pathophysiology of mast cell-associated diseases.
Collapse
Affiliation(s)
- Jelle Folkerts
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands.,Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands.,Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States.,Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ralph Stadhouders
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands.,Department of Cell Biology, Erasmus MC, Rotterdam, Netherlands
| | - Frank A Redegeld
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - See-Ying Tam
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands
| | - Stephen J Galli
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Marcus Maurer
- Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
29
|
Wang Y, Le Y, Zhao W, Lin Y, Wu Y, Yu C, Xiong J, Zou F, Dong H, Cai S, Zhao H. Short Thymic Stromal Lymphopoietin Attenuates Toluene Diisocyanate-induced Airway Inflammation and Inhibits High Mobility Group Box 1-Receptor for Advanced Glycation End Products and Long Thymic Stromal Lymphopoietin Expression. Toxicol Sci 2018; 157:276-290. [PMID: 28329851 DOI: 10.1093/toxsci/kfx043] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Short thymic stromal lymphopoietin (short TSLP), one of TSLP variants, exerts anti-inflammatory activities in endotoxin shock and colitis mouse models. Our latest work reported that short TSLP prevented house dust mite-induced epithelial barrier disruption. Yet the role of short TSLP in toluene diisocyanate (TDI)-induced asthma is unknown. Male BALB/c mice were sensitized and challenged with TDI to generate a chemical-induced asthma model. Synthetic short TSLP peptides were given intranasally or intraperitoneally before each challenge. TDI significantly increased inflammation and hyperresponsiveness of airway, which were suppressed by short TSLP treatment. Levels of mouse TSLP, high mobility group box 1 (HMGB1), and receptor for advanced glycation end products (RAGE) in airway epithelium and whole lung tissues were markedly increased in TDI group compared with control mice, which were decreased after administration of short TSLP. Meanwhile, short TSLP also inhibited STAT5(Y694) phosphorylation, which was highly expressed in airways of TDI-exposure mice. In vitro, both TDI-human serum albumin (HSA) and recombinant human (rh) HMGB1 promoted long TSLP but not short TSLP gene production in human bronchial epithelial cells (16HBE). Cells pre-treated with short TSLP exhibited less expression of RAGE and long TSLP and lower phosphorylation of Akt(S473), p38 MAPK(T180/Y182), and STAT5(Y694) than stimulated with TDI-HSA or rhHMGB1 alone. Results suggest that short TSLP prevents airway inflammation in a chemical-induced asthma model, which might be associated with the inhibitions of HMGB1-RAGE and long TSLP expression and STAT5(Y694) phosphorylation.
Collapse
Affiliation(s)
- Yanhong Wang
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanqing Le
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wenqu Zhao
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yun Lin
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yue Wu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Changhui Yu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jing Xiong
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fei Zou
- School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, China
| | - Hangming Dong
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shaoxi Cai
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haijin Zhao
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
30
|
Zehra S, Khambati I, Vierhout M, Mian MF, Buck R, Forsythe P. Human Milk Oligosaccharides Attenuate Antigen-Antibody Complex Induced Chemokine Release from Human Intestinal Epithelial Cell Lines. J Food Sci 2018; 83:499-508. [PMID: 29377120 DOI: 10.1111/1750-3841.14039] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 10/24/2017] [Accepted: 12/11/2017] [Indexed: 12/17/2022]
Abstract
There has been increased interest in the use of dietary ingredients, including prebiotics such as human-milk oligosaccharides (HMOs), as therapeutic strategies for food allergy. Understanding the mechanisms underlying the beneficial effects of HMOs is important to realizing their therapeutic potential. Here we demonstrate that the HMO, 6'-sialyllactose (6'SL) inhibited chemokine (IL-8 and CCL20) release from T-84 and HT-29 cells stimulated with antigen-antibody complex, TNFα or PGE2 ; an effect that was PPARγ dependent and associated with decreased activity of the transcription factors AP-1 and NFκB. In contrast, 2'-fucosyllactose (2'FL) selectively inhibited CCL20 release in response to antigen antibody complex in a PPARγ independent manner. This study reinforces the concept that structurally different oligosaccharides have distinct biological activities and identifies, for the first time, that the HMOs, 6'SL, and 2'FL, modulate human epithelial cell responses related to allergic disease. These findings encourage further investigation of the therapeutic potential of specific HMOs in food allergy. PRACTICAL APPLICATION This study provides evidence for direct effects of HMOs in addition to their prebiotic role and demonstrates, for the first time, modulation of Ag-IgE complex activation of human epithelial cells that may have important implications for food-allergy. The study also reinforces the concept that structurally different oligosaccharides have distinct biological activities. In determining the composition of infant formula, addition of oligosaccharides with specific structures may provide direct modulation of immune responses and potentially attenuate symptoms or development of food allergy.
Collapse
Affiliation(s)
- Sehrish Zehra
- Dept. of Medicine, Pathology and Molecular Medicine, McMaster Univ., Hamilton, Ont., Canada
| | - Ibrahim Khambati
- Dept. of Medicine, Pathology and Molecular Medicine, McMaster Univ., Hamilton, Ont., Canada
| | - Megan Vierhout
- Dept. of Medicine, Pathology and Molecular Medicine, McMaster Univ., Hamilton, Ont., Canada
| | - M Firoz Mian
- Dept. of Pathology and Molecular Medicine, McMaster Univ., Hamilton, Ont., Canada
| | - Rachael Buck
- Abbott Nutrition: a Division of Abbott Laboratories, Columbus, Ohio, U.S.A
| | - Paul Forsythe
- Dept. of Medicine, Pathology and Molecular Medicine, McMaster Univ., Hamilton, Ont., Canada.,The Firestone Inst. for Respiratory Research, McMaster Univ., Hamilton, Ont., Canada
| |
Collapse
|
31
|
Barrera-Bugueño C, Realini O, Escobar-Luna J, Sotomayor-Zárate R, Gotteland M, Julio-Pieper M, Bravo JA. Anxiogenic effects of a Lactobacillus, inulin and the synbiotic on healthy juvenile rats. Neuroscience 2017; 359:18-29. [DOI: 10.1016/j.neuroscience.2017.06.064] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 06/29/2017] [Accepted: 06/30/2017] [Indexed: 12/26/2022]
|
32
|
McAleer JP, Kolls JK. Contributions of the intestinal microbiome in lung immunity. Eur J Immunol 2017; 48:39-49. [PMID: 28776643 DOI: 10.1002/eji.201646721] [Citation(s) in RCA: 166] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 06/12/2017] [Accepted: 08/01/2017] [Indexed: 12/20/2022]
Abstract
The intestine is a critical site of immune cell development that not only controls intestinal immunity but extra-intestinal immunity as well. Recent findings have highlighted important roles for gut microbiota in shaping lung inflammation. Here, we discuss interactions between the microbiota and immune system including T cells, protective effects of microbiota on lung infections, the role of diet in shaping the composition of gut microbiota and susceptibility to asthma, epidemiologic evidence implicating antibiotic use and microbiota in asthma and clinical trials investigating probiotics as potential treatments for atopy and asthma. The systemic effects of gut microbiota are partially attributed to their generating metabolites including short chain fatty acids, which can suppress lung inflammation through the activation of G protein-coupled receptors. Thus, studying the interactions between microbiota and immune cells can lead to the identification of therapeutic targets for chronic lower respiratory diseases.
Collapse
Affiliation(s)
- Jeremy P McAleer
- Department of Pharmaceutical Science and Research, Marshall University School of Pharmacy, Huntington, WV, USA
| | - Jay K Kolls
- Tulane School of Medicine, Center for Translational Research in Infection and Inflammation, New Orleans, LA, USA
| |
Collapse
|
33
|
Abbring S, Verheijden KAT, Diks MAP, Leusink-Muis A, Hols G, Baars T, Garssen J, van Esch BCAM. Raw Cow's Milk Prevents the Development of Airway Inflammation in a Murine House Dust Mite-Induced Asthma Model. Front Immunol 2017; 8:1045. [PMID: 28894452 PMCID: PMC5581320 DOI: 10.3389/fimmu.2017.01045] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 08/11/2017] [Indexed: 02/03/2023] Open
Abstract
Epidemiological studies show an inverse relation between raw cow’s milk consumption and the development of asthma. This protective effect seems to be abolished by milk processing. However, evidence for a causal relationship is lacking, and direct comparisons between raw and processed milk are hardly studied. Therefore, this study investigated the preventive capacity of raw and heated raw milk on the development of house dust mite (HDM)-induced allergic asthma in mice. Six- to seven-week-old male BALB/c mice were intranasally (i.n.) sensitized with 1 µg HDM or PBS on day 0, followed by an i.n. challenge with 10 µg HDM or PBS on days 7–11. In addition, mice were fed 0.5 mL raw cow’s milk, heated raw cow’s milk, or PBS three times a week throughout the study, starting 1 day before sensitization. On day 14, airway hyperresponsiveness (AHR) in response to increasing doses of methacholine was measured to assess lung function. Bronchoalveolar lavage fluid (BALF) and lungs were furthermore collected to study the extent of airway inflammation. Raw milk prevented both HDM-induced AHR and pulmonary eosinophilic inflammation, whereas heated raw milk did not. Both milk types suppressed the Th2-polarizing chemokine CCL17 in lung homogenates and reduced lung Th2 and Th17 cell frequency. IL-4 and IL-13 production after ex vivo restimulation of lung T cells with HDM was also reduced by both milk types. However, local IL-5 and IL-13 concentrations were only suppressed by raw milk. These findings support the asthma-protective capacity of raw cow’s milk and show the importance of reduced local type 2 cytokine levels. Heated raw milk did not show an asthma-protective effect, which indicates the involvement of heat-sensitive components. Besides causal evidence, this study provides the basis for further mechanistic studies.
Collapse
Affiliation(s)
- Suzanne Abbring
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Kim A T Verheijden
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Mara A P Diks
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Athea Leusink-Muis
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Gert Hols
- Nutricia Research, Utrecht, Netherlands
| | - Ton Baars
- Research Institute of Organic Agriculture (FiBL), Frick, Switzerland
| | - Johan Garssen
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Nutricia Research, Utrecht, Netherlands
| | - Betty C A M van Esch
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Nutricia Research, Utrecht, Netherlands
| |
Collapse
|
34
|
A prebiotic galactooligosaccharide mixture reduces severity of hyperpnoea-induced bronchoconstriction and markers of airway inflammation. Br J Nutr 2017; 116:798-804. [PMID: 27523186 DOI: 10.1017/s0007114516002762] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Gut microbes have a substantial influence on systemic immune function and allergic sensitisation. Manipulation of the gut microbiome through prebiotics may provide a potential strategy to influence the immunopathology of asthma. This study investigated the effects of prebiotic Bimuno-galactooligosaccharide (B-GOS) supplementation on hyperpnoea-induced bronchoconstriction (HIB), a surrogate for exercise-induced bronchoconstriction, and airway inflammation. A total of ten adults with asthma and HIB and eight controls without asthma were randomised to receive 5·5 g/d of either B-GOS or placebo for 3 weeks separated by a 2-week washout period. The peak fall in forced expiratory volume in 1 s (FEV1) following eucapnic voluntary hyperpnoea (EVH) defined HIB severity. Markers of airway inflammation were measured at baseline and after EVH. Pulmonary function remained unchanged in the control group. In the HIB group, the peak post-EVH fall in FEV1 at day 0 (-880 (sd 480) ml) was unchanged after placebo, but was attenuated by 40 % (-940 (sd 460) v. -570 (sd 310) ml, P=0·004) after B-GOS. In the HIB group, B-GOS reduced baseline chemokine CC ligand 17 (399 (sd 140) v. 323 (sd 144) pg/ml, P=0·005) and TNF-α (2·68 (sd 0·98) v. 2·18 (sd 0·59) pg/ml, P=0·040) and abolished the EVH-induced 29 % increase in TNF-α. Baseline C-reactive protein was reduced following B-GOS in HIB (2·46 (sd 1·14) v. 1·44 (sd 0·41) mg/l, P=0·015) and control (2·16 (sd 1·02) v. 1·47 (sd 0·33) mg/l, P=0·050) groups. Chemokine CC ligand 11 and fraction of exhaled nitric oxide remained unchanged. B-GOS supplementation attenuated airway hyper-responsiveness with concomitant reductions in markers of airway inflammation associated with HIB.
Collapse
|
35
|
Lung Homeostasis: Influence of Age, Microbes, and the Immune System. Immunity 2017; 46:549-561. [DOI: 10.1016/j.immuni.2017.04.005] [Citation(s) in RCA: 153] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 04/03/2017] [Accepted: 04/04/2017] [Indexed: 12/24/2022]
|
36
|
Huang YJ, Marsland BJ, Bunyavanich S, O'Mahony L, Leung DYM, Muraro A, Fleisher TA. The microbiome in allergic disease: Current understanding and future opportunities-2017 PRACTALL document of the American Academy of Allergy, Asthma & Immunology and the European Academy of Allergy and Clinical Immunology. J Allergy Clin Immunol 2017; 139:1099-1110. [PMID: 28257972 PMCID: PMC5899886 DOI: 10.1016/j.jaci.2017.02.007] [Citation(s) in RCA: 231] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/02/2017] [Accepted: 02/07/2017] [Indexed: 12/15/2022]
Abstract
PRACTALL is a joint initiative of the American Academy of Allergy, Asthma & Immunology and the European Academy of Allergy and Clinical Immunology to provide shared evidence-based recommendations on cutting-edge topics in the field of allergy and immunology. PRACTALL 2017 is focused on what has been established regarding the role of the microbiome in patients with asthma, atopic dermatitis, and food allergy. This is complemented by outlining important knowledge gaps regarding its role in allergic disease and delineating strategies necessary to fill these gaps. In addition, a review of progress in approaches used to manipulate the microbiome will be addressed, identifying what has and has not worked to serve as a baseline for future directions to intervene in allergic disease development, progression, or both.
Collapse
Affiliation(s)
- Yvonne J Huang
- Division of Pulmonary and Critical Care, Department of Internal Medicine, University of Michigan, Ann Arbor, Mich
| | - Benjamin J Marsland
- Service de Pneumologie, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Supinda Bunyavanich
- Division of Allergy and Immunology, Departments of Pediatrics and Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Liam O'Mahony
- Molecular Immunology, Swiss Institute of Allergy and Asthma Research, University of Zurich, Davos, Switzerland
| | - Donald Y M Leung
- Division of Allergy and Immunology, Department of Pediatrics, National Jewish Health, Denver, Colo
| | - Antonella Muraro
- Food Allergy Referral Centre Veneto Region, Department of Women and Child Health, Padua General University Hospital, Padua, Italy
| | - Thomas A Fleisher
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Md.
| |
Collapse
|
37
|
Rico-Díaz A, Álvarez-Cao ME, Escuder-Rodríguez JJ, González-Siso MI, Cerdán ME, Becerra M. Rational mutagenesis by engineering disulphide bonds improves Kluyveromyces lactis beta-galactosidase for high-temperature industrial applications. Sci Rep 2017; 7:45535. [PMID: 28361909 PMCID: PMC5374532 DOI: 10.1038/srep45535] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 03/01/2017] [Indexed: 12/29/2022] Open
Abstract
Kluyveromyces lactis β-galactosidase (Kl-β-Gal) is one of the most important enzymes in the dairy industry. The poor stability of this enzyme limits its use in the synthesis of galactooligosaccharides (GOS) and other applications requiring high operational temperature. To obtain thermoresistant variants, a rational mutagenesis strategy by introducing disulphide bonds in the interface between the enzyme subunits was used. Two improved mutants, R116C/T270C and R116C/T270C/G818C, had increased half-lives at 45 °C compared to Kl-β-Gal (2.2 and 6.8 fold increases, respectively). Likewise, Tm values of R116C/T270C and R116C/T270C/G818C were 2.4 and 8.5 °C, respectively, higher than Kl-β-Gal Tm. Enrichment in enzymatically active oligomeric forms in these mutant variants also increased their catalytic efficiency, due to the reinforcement of the interface contacts. In this way, using an artificial substrate (p-nitrophenyl-β-D-galactopyranoside), the Vmax values of the mutants were ~1.4 (R116C/T270C) and 2 (R116C/T270C/G818C) fold higher than that of native Kl-β-Gal. Using the natural substrate (lactose) the Vmax for R116C/T270C/G818C almost doubled the Vmax for Kl-β-Gal. Validation of these mutant variants of the enzyme for their use in applications that depend on prolonged incubations at high temperatures was achieved at the laboratory scale by monitoring their catalytic activity in GOS synthesis.
Collapse
Affiliation(s)
- Agustín Rico-Díaz
- Universidade da Coruña. Grupo EXPRELA, Centro de Investigacións Científicas Avanzadas (CICA), Departamento de Bioloxía Celular e Molecular, Facultade de Ciencias, A Coruña, Spain
| | - María-Efigenia Álvarez-Cao
- Universidade da Coruña. Grupo EXPRELA, Centro de Investigacións Científicas Avanzadas (CICA), Departamento de Bioloxía Celular e Molecular, Facultade de Ciencias, A Coruña, Spain
| | - Juan-José Escuder-Rodríguez
- Universidade da Coruña. Grupo EXPRELA, Centro de Investigacións Científicas Avanzadas (CICA), Departamento de Bioloxía Celular e Molecular, Facultade de Ciencias, A Coruña, Spain
| | - María-Isabel González-Siso
- Universidade da Coruña. Grupo EXPRELA, Centro de Investigacións Científicas Avanzadas (CICA), Departamento de Bioloxía Celular e Molecular, Facultade de Ciencias, A Coruña, Spain
| | - M Esperanza Cerdán
- Universidade da Coruña. Grupo EXPRELA, Centro de Investigacións Científicas Avanzadas (CICA), Departamento de Bioloxía Celular e Molecular, Facultade de Ciencias, A Coruña, Spain
| | - Manuel Becerra
- Universidade da Coruña. Grupo EXPRELA, Centro de Investigacións Científicas Avanzadas (CICA), Departamento de Bioloxía Celular e Molecular, Facultade de Ciencias, A Coruña, Spain
| |
Collapse
|
38
|
Bridgman SL, Kozyrskyj AL, Scott JA, Becker AB, Azad MB. Gut microbiota and allergic disease in children. Ann Allergy Asthma Immunol 2016; 116:99-105. [PMID: 26815703 DOI: 10.1016/j.anai.2015.10.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 09/30/2015] [Accepted: 10/02/2015] [Indexed: 12/24/2022]
Affiliation(s)
- Sarah L Bridgman
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Anita L Kozyrskyj
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - James A Scott
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Allan B Becker
- Department of Pediatrics and Child Health, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Meghan B Azad
- Department of Pediatrics and Child Health, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
39
|
Bouchaud G, Castan L, Chesné J, Braza F, Aubert P, Neunlist M, Magnan A, Bodinier M. Maternal exposure to GOS/inulin mixture prevents food allergies and promotes tolerance in offspring in mice. Allergy 2016; 71:68-76. [PMID: 26424001 DOI: 10.1111/all.12777] [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] [Accepted: 09/25/2015] [Indexed: 01/10/2023]
Abstract
BACKGROUND Food allergies affect 4-8% of children and are constantly on the rise, thus making allergies a timely issue. Most importantly, prevention strategies are nonexistent, and current therapeutic strategies have limited efficacy and need to be improved. One alternative to prevent or reduce allergies, particularly during infancy, could consist of modulating maternal immunity and microbiota using nondigestible food ingredients, such as prebiotics. For this purpose, we studied the preventive effects of prebiotics in Balb/c mothers during pregnancy and breastfeeding on food allergy development in offspring mice. METHODS After weaning, the offspring from mothers that were exposed to GOS/inulin mixture or fed a control diet were intraperitoneally sensitized to wheat proteins to induce a systemic allergic response and orally exposed to the same allergen. Immunological, physiological, and microbial parameters were analyzed. RESULTS GOS/inulin mixture diet modified the microbiota of mothers and their offspring. Offspring from mothers that received GOS/inulin prebiotics were protected against food allergies and displayed lower clinical scores, specifically of IgE and histamine levels, compared to offspring from mothers fed a control diet. Moreover, GOS/inulin supplementation for the mother resulted in stronger intestinal permeability in the offspring. Enhancement of the regulatory response to allergic inflammation and changes in the Th2/Th1 balance toward a dampened Th2 response were observed in mice from GOS/inulin mixture-exposed mothers. CONCLUSION The treatment of pregnant and lactating mice with nondigestible GOS/inulin prebiotics promotes a long-term protective effect against food allergies in the offspring.
Collapse
Affiliation(s)
| | - L. Castan
- UR1268 BIA; INRA; Nantes France
- UMR1087; l'institut du thorax; INSERM; Nantes France
- UMR6291; CNRS; Nantes France
- Université de Nantes; Nantes France
| | - J. Chesné
- UR1268 BIA; INRA; Nantes France
- UMR1087; l'institut du thorax; INSERM; Nantes France
- UMR6291; CNRS; Nantes France
- UMR913; Institut des Maladies de l'Appareil Digestif (IMAD); Faculté de Médecine; INSERM; Nantes France
| | - F. Braza
- UR1268 BIA; INRA; Nantes France
- UMR1087; l'institut du thorax; INSERM; Nantes France
- UMR6291; CNRS; Nantes France
- UMR913; Institut des Maladies de l'Appareil Digestif (IMAD); Faculté de Médecine; INSERM; Nantes France
| | - P. Aubert
- UMR6291; CNRS; Nantes France
- UMR913; Institut des Maladies de l'Appareil Digestif (IMAD); Faculté de Médecine; INSERM; Nantes France
- DHU2020 médecine personnalisée des maladies chroniques; Nantes France
| | - M. Neunlist
- UMR6291; CNRS; Nantes France
- UMR913; Institut des Maladies de l'Appareil Digestif (IMAD); Faculté de Médecine; INSERM; Nantes France
- DHU2020 médecine personnalisée des maladies chroniques; Nantes France
| | - A. Magnan
- UMR1087; l'institut du thorax; INSERM; Nantes France
- UMR6291; CNRS; Nantes France
- Université de Nantes; Nantes France
- l'institut du thorax; Service de Pneumologie; CHU de Nantes; Nantes France
- DHU2020 médecine personnalisée des maladies chroniques; Nantes France
| | | |
Collapse
|
40
|
Verheijden KAT, Akbari P, Willemsen LEM, Kraneveld AD, Folkerts G, Garssen J, Fink-Gremmels J, Braber S. Inflammation-induced expression of the alarmin interleukin 33 can be suppressed by galacto-oligosaccharides. Int Arch Allergy Immunol 2015; 167:127-36. [PMID: 26304032 DOI: 10.1159/000437327] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 06/30/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The alarmin interleukin 33 (IL-33) and its receptor ST2 play an important role in mucosal barrier tissues, and seem to be crucial for Th2-cell mediated host defense. Galacto-oligosaccharides (GOS), used in infant formulas, exhibit gut and immune modulatory effects. To enhance our understanding of the immunomodulatory capacity of GOS, this study investigated the impact of dietary GOS intervention on IL-33 and ST2 expression related to intestinal barrier dysfunction and asthma. METHODS B6C3F1 and BALB/c mice were fed a control diet with or without 1% GOS. To simulate intestinal barrier dysfunction, B6C3F1 mice received a gavage with the mycotoxin deoxynivalenol (DON). To mimic asthma-like inflammatory airway responses, BALB/c mice were sensitized on day 0 and challenged on days 7-11 with house-dust mite (HDM) allergen. Samples from the intestines and lungs were collected for IL-33 and ST2 analysis by qRT-PCR, immunoblotting and immunohistochemistry. RESULTS Dietary GOS counteracted the DON-induced IL-33 mRNA expression and changed the IL-33 distribution pattern in the mouse small intestine. The IL-33 mRNA expression was positively correlated to the intestinal permeability. A strong positive correlation was also observed between IL-33 mRNA expression in the lung and the number of bronchoalveolar fluid cells. Reduced levels of IL-33 protein, altered IL-33 distribution and reduced ST2 mRNA expression were observed in the lungs of HDM-allergic mice after GOS intervention. CONCLUSIONS Dietary GOS mitigated IL-33 at the mucosal surfaces in a murine model for intestinal barrier dysfunction and HDM-induced asthma. This promising effect may open up new avenues to use GOS not only as a prebiotic in infant nutrition, but also as a functional ingredient that targets inflammatory processes and allergies associated with IL-33 expression.
Collapse
Affiliation(s)
- Kim A T Verheijden
- Division of Veterinary Pharmacy, Pharmacology and Toxicology, Utrecht University, Utrecht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Verheijden KAT, Willemsen LEM, Braber S, Leusink-Muis T, Jeurink PV, Garssen J, Kraneveld AD, Folkerts G. The development of allergic inflammation in a murine house dust mite asthma model is suppressed by synbiotic mixtures of non-digestible oligosaccharides and Bifidobacterium breve M-16V. Eur J Nutr 2015; 55:1141-51. [PMID: 26003185 PMCID: PMC4819948 DOI: 10.1007/s00394-015-0928-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 05/09/2015] [Indexed: 12/27/2022]
Abstract
Purpose
The incidence and severity of allergic asthma is rising, and novel strategies to prevent or treat this disease are needed. This study investigated the effects of different mixtures of non-digestible oligosaccharides combined with Bifidobacterium breve M-16V (BB) on the development of allergic airway inflammation in an animal model for house dust mite (HDM)-induced allergic asthma. Methods BALB/c mice were sensitized intranasally (i.n.) with HDM and subsequently challenged (i.n.) with PBS or HDM while being fed diets containing different oligosaccharide mixtures in combination with BB or an isocaloric identical control diet. Bronchoalveolar lavage fluid (BALF) inflammatory cell influx, chemokine and cytokine concentrations in lung homogenates and supernatants of ex vivo HDM-restimulated lung cells were analyzed. Results The HDM-induced influx of eosinophils and lymphocytes was reduced by the diet containing the short-chain and long-chain fructo-oligosaccharides and BB (FFBB). In addition to the HDM-induced cell influx, concentrations of IL-33, CCL17, CCL22, IL-6, IL-13 and IL-5 were increased in supernatants of lung homogenates or BALF and IL-4, IFN-γ and IL-10 were increased in restimulated lung cell suspensions of HDM-allergic mice. The diet containing FFBB reduced IL-6, IFN-γ, IL-4 and IL-10 concentrations, whereas the combination of galacto-oligosaccharides and long-chain fructo-oligosaccharides with BB was less potent in this model. Conclusion These findings show that synbiotic dietary supplementation can affect respiratory allergic inflammation induced by HDM. The combination of FFBB was most effective in the prevention of HDM-induced airway inflammation in mice.
Collapse
Affiliation(s)
- K A T Verheijden
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands.
| | - L E M Willemsen
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - S Braber
- Division of Veterinary Pharmacy, Pharmacology and Toxicology, Faculty of Veterinary Sciences, Utrecht University, Utrecht, The Netherlands
| | - T Leusink-Muis
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - P V Jeurink
- Nutricia Research, Immunology, Utrecht, The Netherlands
| | - J Garssen
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands.,Nutricia Research, Immunology, Utrecht, The Netherlands
| | - A D Kraneveld
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - G Folkerts
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| |
Collapse
|
42
|
Verheijden KA, Braber S, Leusink-Muis T, Thijssen S, Boon L, Kraneveld AD, Garssen J, Folkerts G, Willemsen LE. Regulatory T Cell Depletion Abolishes the Protective Effect of Dietary Galacto-Oligosaccharides on Eosinophilic Airway Inflammation in House Dust Mite-Induced Asthma in Mice. J Nutr 2015; 146:831-837. [PMID: 26962188 DOI: 10.3945/jn.115.224402] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 11/07/2015] [Accepted: 02/08/2016] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND In a murine model for house dust mite (HDM)-induced asthma, dietary galacto-oligosaccharides have been shown to suppress allergic symptoms. Previously, CD25+ regulatory T cells (Tregs) induced by nondigestible oligosaccharides were found to protect against allergy development. OBJECTIVE The aim of the current study was to examine the effect of anti-CD25-induced Treg depletion in a murine HDM-induced asthma model and to study the contribution of Tregs in the protective effect of dietary intervention with galacto-oligosaccharides. METHODS Male BALB/c mice (aged 6-8 wk) were intranasally sensitized and challenged with phosphate-buffered saline (PBS) or HDM. Two weeks before sensitization and throughout the whole experiment, mice were fed a control or 1% w/w galacto-oligosaccharide diet. Tregs were depleted by anti-mouse CD25 antibody (intraperitoneally injected). On day 14, T helper cell subtypes in lung and spleen were analyzed and cytokines were measured. Leukocyte subtypes were analyzed in the bronchoalveolar lavage fluid, and interleukin (IL)-33 and chemokines were measured in lung homogenate supernatants. RESULTS Anti-CD25 treatment depleted CD25+ Forkhead box P3+ Tregs in the lung and spleen of control and HDM-allergic mice (P < 0.0001) by >70% while increasing the percentage of activated T helper cells (P < 0.05) and type 2 T helper cells (P < 0.05). This was associated with increased IL-10, IL-4, and IL-13 concentrations in supernatants of ex vivo restimulated lung cells (P < 0.01). Bronchoalveolar lavage fluid leukocyte numbers and percentages of eosinophils and lymphocytes were greater in HDM-allergic mice compared with PBS mice (P < 0.01) but remained unaffected by the anti-CD25 treatment. Galacto-oligosaccharides decreased airway eosinophilia compared with HDM-allergic mice fed the control diet (from 47.8% ± 6.7% to 26.6% ± 8.5%, P < 0.01). This protective effect was lost in anti-CD25-treated mice (P < 0.05). In lung homogenates of HDM-allergic mice, IL-33 was increased compared with PBS mice (from 2.8 ± 0.3 to 5.4 ± 0.6 ng protein/mg, P < 0.01). Galacto-oligosaccharides abrogated the increase in IL-33 compared with HDM-allergic mice fed the control diet (3.0 ± 0.6 ng protein/mg, P < 0.05), which was abolished by the anti-CD25 treatment (P < 0.01). CONCLUSIONS Treg depletion enhances pulmonary type 2 T helper cell frequency and cytokine release in HDM-induced asthma in mice. Galacto-oligosaccharides decreased airway eosinophilia and IL-33 concentrations in the lung, which was abrogated by Treg depletion. This indicates that galacto-oligosaccharides have a beneficial effect in the prevention of HDM-induced allergic asthma by supporting pulmonary Treg function.
Collapse
Affiliation(s)
- Kim At Verheijden
- Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Division of Pharmacology, Utrecht University, Utrecht, Netherlands
| | - Saskia Braber
- Faculty of Veterinary Sciences, Division of Veterinary Pharmacy, Pharmacology, and Toxicology, Utrecht University, Utrecht, Netherlands
| | - Thea Leusink-Muis
- Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Division of Pharmacology, Utrecht University, Utrecht, Netherlands
| | - Suzan Thijssen
- Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Division of Pharmacology, Utrecht University, Utrecht, Netherlands
| | - Louis Boon
- EPIRUS Biopharmaceuticals Netherlands BV, Utrecht, Netherlands
| | - Aletta D Kraneveld
- Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Division of Pharmacology, Utrecht University, Utrecht, Netherlands
| | - Johan Garssen
- Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Division of Pharmacology, Utrecht University, Utrecht, Netherlands.,Nutricia Research, Immunology, Utrecht, Netherlands
| | - Gert Folkerts
- Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Division of Pharmacology, Utrecht University, Utrecht, Netherlands
| | - Linette Em Willemsen
- Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Division of Pharmacology, Utrecht University, Utrecht, Netherlands
| |
Collapse
|