51
|
Nam W, Kim H, Kim J, Nam B, Bae C, Kim J, Park S, Lee J, Sim J. Lactic Acid Bacteria and Natural Product Complex Ameliorates Ovalbumin-Induced Airway Hyperresponsiveness in Mice. J Med Food 2021; 24:517-526. [PMID: 34009021 DOI: 10.1089/jmf.2020.4853] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The incidence of respiratory diseases, such as asthma, has substantially increased in recent times owing to environmental changes, such as air pollution. Induction of a chronic inflammatory response begins with production of biologically active mediators from the airway epithelium, which attracts and recruits inflammatory cells into the lung airway. In our previous study, we confirmed that Lactobacillus casei HY2782 and Bifidobacterium animalis spp. lactis HY8002 could improve lung inflammation in the COPD animal model. In this study, we investigated the effect of the HY2782 complex against airway hyperresponsiveness by using an ovalbumin (OVA)-induced animal model. An orally administered HY2782 complex on OVA-induced allergic asthma in a BALB/c mouse model was used. The present results showed that the HY2782 complex suppressed total immunoglobulin E in serum and bronchoalveolar lavage fluid (BALF). The cytokine production profile in BALF and serum revealed that the HY2782 complex showed reduced levels of Th2 cytokines among immune factors released due to the elevated allergic response. Levels of inflammatory mediators in BALF, MCP-1, MIP-2, and CXCL-9 were decreased by oral administration of the HY2782 complex. Lower numbers of eosinophils and neutrophils in BALF suggested that inflammation was ameliorated by the HY2782 complex. Histological observation of lung sections also showed infiltration of fewer cells. From results, we suggested that the HY2782 complex effectively responds to improvement of the immune response and airway hypersensitivity reaction because of the anti-inflammatory effect of the Pueraria lobata root extract and antioxidant effect of HY2782.
Collapse
Affiliation(s)
- Woo Nam
- R&D Center, Korea Yakult Co. Ltd., Yongin, Korea
| | - Hyeonji Kim
- R&D Center, Korea Yakult Co. Ltd., Yongin, Korea
| | - Jisoo Kim
- R&D Center, Korea Yakult Co. Ltd., Yongin, Korea
| | - Bora Nam
- R&D Center, Korea Yakult Co. Ltd., Yongin, Korea
| | - Chuhyun Bae
- R&D Center, Korea Yakult Co. Ltd., Yongin, Korea
| | - Jooyun Kim
- R&D Center, Korea Yakult Co. Ltd., Yongin, Korea
| | - Soodong Park
- R&D Center, Korea Yakult Co. Ltd., Yongin, Korea
| | | | - Jaehun Sim
- R&D Center, Korea Yakult Co. Ltd., Yongin, Korea
| |
Collapse
|
52
|
Yun L, Li W, Liu Y, Wu T, Zhang M. Regulation of wheat germ polysaccharides in the immune response of mice from newborn to adulthood associated with intestinal microbiota. Food Funct 2021; 11:9662-9674. [PMID: 33211056 DOI: 10.1039/d0fo02031a] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The aim of this study was to determine the effects of wheat germ polysaccharides (WGPs), which are indigestible carbohydrate fibers, on mice in early life, and the changes leading to long-lasting consequences. We determined the influences of early life ceftriaxone and WGP treatment on intestinal microbiota and immunity both in newborn and adulthood mice. The results showed that ceftriaxone significantly altered the intestinal microbiota, short-chain fatty acids' (SCFAs) metabolism, organ index, and serum OVA-specific IgE levels in newborn mice. Comparing adulthood mice to ceftriaxone-treated mice, the diversity and composition of intestinal microbiota were significantly improved after WGP treatment. In addition, the levels of OVA-specific IgE in the WGP-treated mice were significantly decreased, and the expression of cytokines (IL-2, IL-4, IL-6, IFN-γ, and TNF-α) were obviously increased. Therefore, we speculate that the mechanism of action of the indigestible carbohydrate fibers of WGPs is through maintaining immune homeostasis in newborns, which may partly last into adulthood. More importantly, this may be closely related to alterations in the intestinal microbiota.
Collapse
Affiliation(s)
- Liyuan Yun
- State Key Laboratory of Food Nutrition and Safety, Food Biotechnology Engineering Research Center of Ministry of Education, College of food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China.
| | | | | | | | | |
Collapse
|
53
|
Allam VSRR, Chellappan DK, Jha NK, Shastri MD, Gupta G, Shukla SD, Singh SK, Sunkara K, Chitranshi N, Gupta V, Wich PR, MacLoughlin R, Oliver BGG, Wernersson S, Pejler G, Dua K. Treatment of chronic airway diseases using nutraceuticals: Mechanistic insight. Crit Rev Food Sci Nutr 2021; 62:7576-7590. [PMID: 33977840 DOI: 10.1080/10408398.2021.1915744] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Respiratory diseases, both acute and chronic, are reported to be the leading cause of morbidity and mortality, affecting millions of people globally, leading to high socio-economic burden for the society in the recent decades. Chronic inflammation and decline in lung function are the common symptoms of respiratory diseases. The current treatment strategies revolve around using appropriate anti-inflammatory agents and bronchodilators. A range of anti-inflammatory agents and bronchodilators are currently available in the market; however, the usage of such medications is limited due to the potential for various adverse effects. To cope with this issue, researchers have been exploring various novel, alternative therapeutic strategies that are safe and effective to treat respiratory diseases. Several studies have been reported on the possible links between food and food-derived products in combating various chronic inflammatory diseases. Nutraceuticals are examples of such food-derived products which are gaining much interest in terms of its usage for the well-being and better human health. As a consequence, intensive research is currently aimed at identifying novel nutraceuticals, and there is an emerging notion that nutraceuticals can have a positive impact in various respiratory diseases. In this review, we discuss the efficacy of nutraceuticals in altering the various cellular and molecular mechanisms involved in mitigating the symptoms of respiratory diseases.
Collapse
Affiliation(s)
- Venkata Sita Rama Raju Allam
- Department of Medical Biochemistry and Microbiology, Biomedical Centre (BMC), Uppsala University, Uppsala, Sweden
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University (IMU), Kuala Lumpur, Malaysia
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida, Uttar Pradesh, India
| | - Madhur D Shastri
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Tasmania, Australia
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Jaipur, India
| | - Shakti D Shukla
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, Newcastle, New South Wales, Australia
| | - Sachin K Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Krishna Sunkara
- Emergency Clinical Management, Intensive Care Unit, John Hunter Hospital, Newcastle, New South Wales, Australia
| | - Nitin Chitranshi
- Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, New South Wales, Australia
| | - Vivek Gupta
- Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, New South Wales, Australia
| | - Peter R Wich
- School of Chemical Engineering, University of New South Wales, Sydney, New South Wales, Australia.,Centre for Nanomedicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Ronan MacLoughlin
- Aerogen, IDA Business Park, Dangan, Galway, Ireland.,School of Pharmacy & Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland.,School of Pharmacy and Pharmaceutical Sciences, Trinity College, Dublin, Ireland
| | - Brian Gregory George Oliver
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, Australia.,Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia
| | - Sara Wernersson
- Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, Biomedical Centre (BMC), Uppsala University, Uppsala, Sweden.,Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, Australia
| |
Collapse
|
54
|
Al-Hadidi A, Navarro J, Goodman SD, Bailey MT, Besner GE. Lactobacillus reuteri in Its Biofilm State Improves Protection from Experimental Necrotizing Enterocolitis. Nutrients 2021; 13:nu13030918. [PMID: 33809097 PMCID: PMC8000340 DOI: 10.3390/nu13030918] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 12/11/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a devastating disease predominately found in premature infants that is associated with significant morbidity and mortality. Despite decades of research, medical management with broad spectrum antibiotics and bowel rest has remained relatively unchanged, with no significant improvement in patient outcomes. The etiology of NEC is multi-factorial; however, gastrointestinal dysbiosis plays a prominent role in a neonate's vulnerability to and development of NEC. Probiotics have recently emerged as a new avenue for NEC therapy. However, current delivery methods are associated with potential limitations, including the need for at least daily administration in order to obtain any improvement in outcomes. We present a novel formulation of enterally delivered probiotics that addresses the current limitations. A single enteral dose of Lactobacillus reuteri delivered in a biofilm formulation increases probiotic survival in acidic gastric conditions, increases probiotic adherence to gastrointestinal epithelial cells, and reduces the incidence, severity, and neurocognitive sequelae of NEC in experimental models.
Collapse
Affiliation(s)
- Ameer Al-Hadidi
- Department of Pediatric Surgery, Nationwide Children’s Hospital, The Ohio State University College of Medicine, Center for Perinatal Research, The Research Institute at Nationwide Children’s Hospital, Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA;
| | - Jason Navarro
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA; (J.N.); (S.D.G.); (M.T.B.)
| | - Steven D. Goodman
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA; (J.N.); (S.D.G.); (M.T.B.)
| | - Michael T. Bailey
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA; (J.N.); (S.D.G.); (M.T.B.)
| | - Gail E. Besner
- Department of Pediatric Surgery, Nationwide Children’s Hospital, The Ohio State University College of Medicine, Center for Perinatal Research, The Research Institute at Nationwide Children’s Hospital, Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA;
- Correspondence: ; Tel.: +1-614-722-3914
| |
Collapse
|
55
|
Dysbiosis in Pediatrics Is Associated with Respiratory Infections: Is There a Place for Bacterial-Derived Products? Microorganisms 2021; 9:microorganisms9020448. [PMID: 33671586 PMCID: PMC7926342 DOI: 10.3390/microorganisms9020448] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/12/2021] [Accepted: 02/17/2021] [Indexed: 12/22/2022] Open
Abstract
Respiratory tract infections (RTIs) are common in childhood because of the physiologic immaturity of the immune system, a microbial community under development in addition to other genetic, physiological, environmental and social factors. RTIs tend to recur and severe lower viral RTIs in early childhood are not uncommon and are associated with increased risk of respiratory disorders later in life, including recurrent wheezing and asthma. Therefore, a better understanding of the main players and mechanisms involved in respiratory morbidity is necessary for a prompt and improved care as well as for primary prevention. The inter-talks between human immune components and microbiota as well as their main functions have been recently unraveled; nevertheless, more is still to be discovered or understood in the above medical conditions. The aim of this review paper is to provide the most up-to-date overview on dysbiosis in pre-school children and its association with RTIs and their complications. The potential role of non-harmful bacterial-derived products, according to the old hygiene hypothesis and the most recent trained-innate immunity concept, will be discussed together with the need of proof-of-concept studies and larger clinical trials with immunological and microbiological endpoints.
Collapse
|
56
|
Liu Y, Li Z, Wu Y, Jing X, Li L, Fang X. Intestinal Bacteria Encapsulated by Biomaterials Enhance Immunotherapy. Front Immunol 2021; 11:620170. [PMID: 33643302 PMCID: PMC7902919 DOI: 10.3389/fimmu.2020.620170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/31/2020] [Indexed: 12/12/2022] Open
Abstract
The human intestine contains thousands of bacterial species essential for optimal health. Aside from their pathogenic effects, these bacteria have been associated with the efficacy of various treatments of diseases. Due to their impact on many human diseases, intestinal bacteria are receiving increasing research attention, and recent studies on intestinal bacteria and their effects on treatments has yielded valuable results. Particularly, intestinal bacteria can affect responses to numerous forms of immunotherapy, especially cancer therapy. With the development of precision medicine, understanding the factors that influence intestinal bacteria and how they can be regulated to enhance immunotherapy effects will improve the application prospects of intestinal bacteria therapy. Further, biomaterials employed for the convenient and efficient delivery of intestinal bacteria to the body have also become a research hotspot. In this review, we discuss the recent findings on the regulatory role of intestinal bacteria in immunotherapy, focusing on immune cells they regulate. We also summarize biomaterials used for their delivery.
Collapse
Affiliation(s)
- Yilun Liu
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhongmin Li
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yuanyu Wu
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xiabin Jing
- Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Lin Li
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xuedong Fang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
57
|
Engevik MA, Ruan W, Esparza M, Fultz R, Shi Z, Engevik KA, Engevik AC, Ihekweazu FD, Visuthranukul C, Venable S, Schady DA, Versalovic J. Immunomodulation of dendritic cells by Lactobacillus reuteri surface components and metabolites. Physiol Rep 2021; 9:e14719. [PMID: 33463911 PMCID: PMC7814497 DOI: 10.14814/phy2.14719] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Lactic acid bacteria are commensal members of the gut microbiota and are postulated to promote host health. Secreted factors and cell surface components from Lactobacillus species have been shown to modulate the host immune system. However, the precise role of L. reuteri secreted factors and surface proteins in influencing dendritic cells (DCs) remains uncharacterized. HYPOTHESIS We hypothesize that L. reuteri secreted factors will promote DC maturation, skewing cells toward an anti-inflammatory phenotype. In acute colitis, we speculate that L. reuteri promotes IL-10 and dampens pro-inflammatory cytokine production, thereby improving colitis. METHODS & RESULTS Mouse bone marrow-derived DCs were differentiated into immature dendritic cells (iDCs) via IL-4 and GM-CSF stimulation. iDCs exposed to L. reuteri secreted factors or UV-irradiated bacteria exhibited greater expression of DC maturation markers CD83 and CD86 by flow cytometry. Additionally, L. reuteri stimulated DCs exhibited phenotypic maturation as denoted by cytokine production, including anti-inflammatory IL-10. Using mouse colonic organoids, we found that the microinjection of L. reuteri secreted metabolites and UV-irradiated bacteria was able to promote IL-10 production by DCs, indicating potential epithelial-immune cross-talk. In a TNBS-model of acute colitis, L. reuteri administration significantly improved histological scoring, colonic cytokine mRNA, serum cytokines, and bolstered IL-10 production. CONCLUSIONS Overall these data demonstrate that both L. reuteri secreted factors and its bacterial components are able to promote DC maturation. This work points to the specific role of L. reuteri in modulating intestinal DCs. NEW & NOTEWORTHY Lactobacillus reuteri colonizes the mammalian gastrointestinal tract and exerts beneficial effects on host health. However, the mechanisms behind these effects have not been fully explored. In this article, we identified that L. reuteri ATTC PTA 6475 metabolites and surface components promote dendritic cell maturation and IL-10 production. In acute colitis, we also demonstrate that L. reuteri can promote IL-10 and suppress inflammation. These findings may represent a crucial mechanism for maintaining intestinal immune homeostasis.
Collapse
Affiliation(s)
- Melinda A Engevik
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.,Department of Pathology, Texas Children's Hospital, Houston, TX, USA
| | - Wenly Ruan
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,Section of Gastroenterology, Hepatology, and Nutrition, Texas Children's Hospital, Houston, TX, USA
| | - Magdalena Esparza
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.,Department of Pathology, Texas Children's Hospital, Houston, TX, USA
| | - Robert Fultz
- Department of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, Galveston, TX, USA
| | - Zhongcheng Shi
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.,Department of Pathology, Texas Children's Hospital, Houston, TX, USA
| | - Kristen A Engevik
- Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Amy C Engevik
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Faith D Ihekweazu
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,Section of Gastroenterology, Hepatology, and Nutrition, Texas Children's Hospital, Houston, TX, USA
| | - Chonnikant Visuthranukul
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.,Pediatric Nutrition Research Unit, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Susan Venable
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.,Department of Pathology, Texas Children's Hospital, Houston, TX, USA
| | - Deborah A Schady
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.,Department of Pathology, Texas Children's Hospital, Houston, TX, USA
| | - James Versalovic
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.,Department of Pathology, Texas Children's Hospital, Houston, TX, USA
| |
Collapse
|
58
|
Champagne-Jorgensen K, Jose TA, Stanisz AM, Mian MF, Hynes AP, Bienenstock J. Bacterial membrane vesicles and phages in blood after consumption of lacticaseibacillus rhamnosus JB-1. Gut Microbes 2021; 13:1993583. [PMID: 34747333 PMCID: PMC8583084 DOI: 10.1080/19490976.2021.1993583] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/17/2021] [Accepted: 10/06/2021] [Indexed: 02/04/2023] Open
Abstract
Gut microbiota have myriad roles in host physiology, development, and immunity. Though confined to the intestinal lumen by the epithelia, microbes influence distal systems via poorly characterized mechanisms. Recent work has considered the role of extracellular vesicles in interspecies communication, but whether they are involved in systemic microbe-host interaction is unclear. Here, we show that distinctive nanoparticles can be isolated from mouse blood within 2.5 h of consuming Lacticaseibacillus rhamnosus JB-1. In contrast to blood nanoparticles from saline-fed mice, they reproduced lipoteichoic acid-mediated immune functions of the original bacteria, including activation of TLR2 and increased IL-10 expression by dendritic cells. Like the fed bacteria, they also reduced IL-8 induced by TNF in an intestinal epithelial cell line. Though enriched for host neuronal proteins, these isolated nanoparticles also contained proteins and viral (phage) DNA of fed bacterial origin. Our data strongly suggest that oral consumption of live bacteria rapidly leads to circulation of their membrane vesicles and phages and demonstrate a nanoparticulate pathway whereby beneficial bacteria and probiotics may systemically affect their hosts.
Collapse
Affiliation(s)
- Kevin Champagne-Jorgensen
- Neuroscience Graduate Program, McMaster University, Hamilton, Canada
- Brain-Body Institute, St. Joseph’s Healthcare Hamilton, Hamilton, Canada
| | - Tamina A. Jose
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Canada
| | - Andrew M. Stanisz
- Brain-Body Institute, St. Joseph’s Healthcare Hamilton, Hamilton, Canada
| | - M. Firoz Mian
- Brain-Body Institute, St. Joseph’s Healthcare Hamilton, Hamilton, Canada
| | - Alexander P. Hynes
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Canada
- Department of Medicine, McMaster University, Hamilton, Canada
| | - John Bienenstock
- Brain-Body Institute, St. Joseph’s Healthcare Hamilton, Hamilton, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| |
Collapse
|
59
|
Sohn KH, Baek MG, Choi SM, Bae B, Kim RY, Kim YC, Kim HY, Yi H, Kang HR. Alteration of Lung and Gut Microbiota in IL-13-Transgenic Mice Simulating Chronic Asthma. J Microbiol Biotechnol 2020; 30:1819-1826. [PMID: 33046682 PMCID: PMC9728179 DOI: 10.4014/jmb.2009.09019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/15/2022]
Abstract
Increasing evidence suggests a potential role of microbial colonization in the inception of chronic airway diseases. However, it is not clear whether the lung and gut microbiome dysbiosis is coincidental or a result of mutual interaction. In this study, we investigated the airway microbiome in interleukin 13 (IL-13)-rich lung environment and related alterations of the gut microbiome. IL-13- overexpressing transgenic (TG) mice presented enhanced eosinophilic inflammatory responses and mucus production, together with airway hyperresponsiveness and subepithelial fibrosis. While bronchoalveolar lavage fluid and cecum samples obtained from 10-week-old IL-13 TG mice and their C57BL/6 wild-type (WT) littermates showed no significant differences in alpha diversity of lung and gut microbiome, they presented altered beta diversity in both lung and gut microbiota in the IL-13 TG mice compared to the WT mice. Lung-specific IL-13 overexpression also altered the composition of the gut as well as the lung microbiome. In particular, IL-13 TG mice showed an increased proportion of Proteobacteria and Cyanobacteria and a decreased amount of Bacteroidetes in the lungs, and depletion of Firmicutes and Proteobacteria in the gut. The patterns of polymicrobial interaction within the lung microbiota were different between WT and IL-13 TG mice. For instance, in IL-13 TG mice, lung Mesorhizobium significantly affected the alpha diversity of both lung and gut microbiomes. In summary, chronic asthma-like pathologic changes can alter the lung microbiota and affect the gut microbiome. These findings suggest that the lung-gut microbial axis might actually work in asthma.
Collapse
Affiliation(s)
- Kyoung-Hee Sohn
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul 08826, Republic of Korea,Division of Pulmonology, Allergy and Critical Care, Department of Internal Medicine, Kyung Hee University Medical Center, Seoul 0447, Republic of Korea
| | - Min-gyung Baek
- Department of Public Health Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Sung-Mi Choi
- Department of Public Health Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Boram Bae
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul 08826, Republic of Korea
| | - Ruth Yuldam Kim
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul 08826, Republic of Korea
| | - Young-Chan Kim
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul 08826, Republic of Korea
| | - Hye-Young Kim
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul 08826, Republic of Korea,Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 08826, Republic of Korea
| | - Hana Yi
- Department of Public Health Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea,School of Biosystems and Biomedical Sciences, Korea University, Seoul 02841, Republic of Korea,Corresponding authors H.Yi Phone: +82-2-3290-5644 Fax: +82-2-940-2849 E-mail:
| | - Hye-Ryun Kang
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul 08826, Republic of Korea,Division of Allergy and Clinical Immunology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul 0882, Republic of Korea,H-R.Kang Phone: 82-2-2072-0820 Fax: 82-2-742-3291 E-mail:
| |
Collapse
|
60
|
Geuking MB, Burkhard R. Microbial modulation of intestinal T helper cell responses and implications for disease and therapy. Mucosal Immunol 2020; 13:855-866. [PMID: 32792666 DOI: 10.1038/s41385-020-00335-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/20/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023]
Abstract
Induction of intestinal T helper cell subsets by commensal members of the intestinal microbiota is an important component of the many immune adaptations required to establish host-microbial homeostasis. Importantly, altered intestinal T helper cell profiles can have pathological consequences that are not limited to intestinal sites. Therefore, microbial-mediated modulation of the intestinal T helper cell profile could have strong therapeutic potentials. However, in order to develop microbial therapies that specifically induce the desired alterations in the intestinal T helper cell compartment one has to first gain a detailed understanding of how microbial composition and the metabolites derived or induced by the microbiota impact on intestinal T helper cell responses. Here we summarize the milestone findings in the field of microbiota-intestinal T helper cell crosstalk with a focus on the role of specific commensal bacteria and their metabolites. We discuss mechanistic mouse studies and are linking these to human studies where possible. Moreover, we highlight recent advances in the field of microbial CD4 T cell epitope mimicry in autoimmune diseases and the role of microbially-induced CD4 T cells in cancer immune checkpoint blockade therapy.
Collapse
Affiliation(s)
- Markus B Geuking
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| | - Regula Burkhard
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
61
|
Midha A, Ebner F, Schlosser-Brandenburg J, Rausch S, Hartmann S. Trilateral Relationship: Ascaris, Microbiota, and Host Cells. Trends Parasitol 2020; 37:251-262. [PMID: 33008723 DOI: 10.1016/j.pt.2020.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/02/2020] [Accepted: 09/04/2020] [Indexed: 12/11/2022]
Abstract
Ascariasis is a globally spread intestinal nematode infection of humans and a considerable concern in pig husbandry. Ascaris accomplishes a complex body migration from the intestine via the liver and lung before returning to the intestine. Tissue migration and the habitat shared with a complex microbial community pose the question of how the nematode interacts with microbes and host cells from various tissues. This review addresses the current knowledge of the trilateral relationship between Ascaris, its microbial environment, and host cells, and discusses novel approaches targeting these interactions to combat this widespread infection of livestock and man.
Collapse
Affiliation(s)
- Ankur Midha
- Institute of Immunology, Freie Universität Berlin, Robert von Ostertag-Str. 7-13, D-14163 Berlin, Germany
| | - Friederike Ebner
- Institute of Immunology, Freie Universität Berlin, Robert von Ostertag-Str. 7-13, D-14163 Berlin, Germany
| | | | - Sebastian Rausch
- Institute of Immunology, Freie Universität Berlin, Robert von Ostertag-Str. 7-13, D-14163 Berlin, Germany
| | - Susanne Hartmann
- Institute of Immunology, Freie Universität Berlin, Robert von Ostertag-Str. 7-13, D-14163 Berlin, Germany.
| |
Collapse
|
62
|
Early life microbial exposures and allergy risks: opportunities for prevention. Nat Rev Immunol 2020; 21:177-191. [PMID: 32918062 DOI: 10.1038/s41577-020-00420-y] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2020] [Indexed: 02/07/2023]
Abstract
Allergies, including asthma, food allergy and atopic dermatitis, are increasing in prevalence, particularly in westernized countries. Although a detailed mechanistic explanation for this increase is lacking, recent evidence indicates that, in addition to genetic predisposition, lifestyle changes owing to modernization have an important role. Such changes include increased rates of birth by caesarean delivery, increased early use of antibiotics, a westernized diet and the associated development of obesity, and changes in indoor and outdoor lifestyle and activity patterns. Most of these factors directly and indirectly impact the formation of a diverse microbiota, which includes bacterial, viral and fungal components; the microbiota has a leading role in shaping (early) immune responses. This default programme is markedly disturbed under the influence of environmental and lifestyle risk factors. Here, we review the most important allergy risk factors associated with changes in our exposure to the microbial world and the application of this knowledge to allergy prevention strategies.
Collapse
|
63
|
Alaqil AA, Abbas AO, El-Beltagi HS, El-Atty HKA, Mehaisen GMK, Moustafa ES. Dietary Supplementation of Probiotic Lactobacillus acidophilus Modulates Cholesterol Levels, Immune Response, and Productive Performance of Laying Hens. Animals (Basel) 2020; 10:ani10091588. [PMID: 32899938 PMCID: PMC7552304 DOI: 10.3390/ani10091588] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/02/2020] [Accepted: 09/04/2020] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Chicken eggs provide a considerable source of high-quality nutrients for human food and health. However, egg consumption may harm some people for its high contents of cholesterol and association with cardiovascular disease risk. Therefore, we investigated the possible effect of using Lactobacillus acidophilus (LA) as a probiotic additive in the laying hens’ diets on lowering egg-yolk cholesterol and also evaluated the immune responses and the productive performance of laying hens. The obtained results display the ability of LA, when supplemented in hen diets, to decrease the plasma triglycerides and cholesterol levels and the liver and egg yolk cholesterols in the hen. The beneficial effects of LA were also explored on some important humoral and cell-mediated immune responses in the laying hens. These positive effects led to an improvement in the productive performance of laying hens. Therefore, dietary LA supplementation could be recommended as a nutritional strategy for commercial lower-cholesterol egg production in addition to positive impacts on the performance and health of laying hens. Abstract This study examines the effect of dietary supplementation with Lactobacillus acidophilus (LA) on the cholesterol levels, immune response, and productive performance of laying hens. A total of 216, 40-week-old, commercial Hy-Line brown chicken layers were randomly assigned into four treatment groups (18 birds × three replicates per group) and fed diet supplemented with 0 (control), 1 × 109, 21 × 109, and 31 × 109 colony forming units (CFUs) of Lactobacillus acidophilus (LA) per kg of feed for six consecutive weeks. Results show that plasma triglycerides, low-density lipoprotein (LDL) and total cholesterols became lesser, while high-density lipoprotein (HDL) cholesterol became higher in LA-supplemented groups compared to the control. In addition, a significant reduction occurred in the liver and egg yolk cholesterol by LA supplementation. Moreover, the immunological parameters including antibody titer against sheep red blood cells (SRBCs), phytohemagglutinin (PHA)-wattle swelling test, and T- & B-lymphocyte proliferation were enhanced in laying hens supplemented with LA compared to the control hens. While the heterophil to lymphocyte (H/L) ratio decreased with LA supplementation, indicating low stress conditions in the treated hens. These positive effects for LA were further reflected on the productive performance of laying hens and improved egg production, egg weight, egg mass, and feed efficiency. Our findings indicate that LA probiotic could be recommended in laying hens’ diets for lowering egg yolk cholesterol with positive impacts on health and performance.
Collapse
Affiliation(s)
- Abdulaziz A. Alaqil
- Department of Animal and Fish Production, College of Agricultural and Food Sciences, King Faisal University, P.O. Box 420, Al-Ahsa 31982, Saudi Arabia; (A.A.A.); (A.O.A.)
| | - Ahmed O. Abbas
- Department of Animal and Fish Production, College of Agricultural and Food Sciences, King Faisal University, P.O. Box 420, Al-Ahsa 31982, Saudi Arabia; (A.A.A.); (A.O.A.)
- Department of Animal Production, Faculty of Agriculture, Cairo University, Gamma St., Giza 12613, Egypt
| | - Hossam S. El-Beltagi
- Department of Agricultural Biotechnology, College of Agricultural and Food Sciences, King Faisal University, P.O. Box 420, Al-Ahsa 31982, Saudi Arabia;
- Department of Biochemistry, Faculty of Agriculture, Cairo University, Gamma St., Giza 12613, Egypt
| | - Hanaa. K. Abd El-Atty
- Department of Poultry Breeding, Animal Production Research Institute, Agricultural Research Center, Dokki, Giza 12611, Egypt;
| | - Gamal M. K. Mehaisen
- Department of Animal Production, Faculty of Agriculture, Cairo University, Gamma St., Giza 12613, Egypt
- Correspondence: (G.M.K.M.); (E.S.M.); Tel.: +20-122-6797270 (G.M.K.M.)
| | - Eman S. Moustafa
- Department of Animal Production, Faculty of Agriculture, Cairo University, Gamma St., Giza 12613, Egypt
- Correspondence: (G.M.K.M.); (E.S.M.); Tel.: +20-122-6797270 (G.M.K.M.)
| |
Collapse
|
64
|
Liu Y, Mian MF, McVey Neufeld KA, Forsythe P. CD4 +CD25 + T Cells are Essential for Behavioral Effects of Lactobacillus rhamnosus JB-1 in Male BALB/c mice. Brain Behav Immun 2020; 88:451-460. [PMID: 32276029 DOI: 10.1016/j.bbi.2020.04.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 04/06/2020] [Accepted: 04/06/2020] [Indexed: 01/01/2023] Open
Abstract
Over the past decade there has been increasing interest in the involvement of the microbiota-gut-brain axis in mental health. However, there are major gaps in our knowledge regarding the complex signaling systems through which gut microbes modulate the CNS. The immune system is a recognized mediator in the bidirectional communication continuously occurring between gut and brain. We previously demonstrated that Lactobacillus rhamnosus JB-1 (JB-1), a bacterial strain that has anxiolytic- and antidepressant-like effects in mice, modulates the immune system through induction of immunosuppressive T regulatory cells. Here we examined a potential causal relationship between JB-1 induced regulatory T cells and the observed effects on behaviour. We found that depletion of regulatory T cells, via treatment with monoclonal antibody against CD25, inhibited the antidepressant- and anxiolytic-like effects induced by 4-week oral administration of JB-1 in mice. Ly6Chi monocytes were found to be decreased in JB-1 fed mice with intact regulatory T cells, but not in JB-1 fed mice following depletion. Furthermore, adoptive transfer of CD4+CD25+ cells, from JB-1 treated donor mice, but not from controls, induced antidepressant- and anxiolytic-like effects in recipient mice. Ly6Chi monocytes were also significantly decreased in mice receiving CD4+CD25+ cells from JB1 fed donors. This study identifies cells within the CD4+CD25+ population, most likely regulatory T cells, as both necessary and sufficient in JB-1-induced antidepressant- and anxiolytic-like effects in mice, providing novel mechanistic insight into microbiota-gut-brain communication in addition to highlighting the potential for immunotherapy in psychiatric disorders.
Collapse
Affiliation(s)
- Yunpeng Liu
- McMaster Brain-Body Institute, The Research Institute of St. Joseph's Hamilton, Hamilton, Ontario, Canada
| | - M Firoz Mian
- McMaster Brain-Body Institute, The Research Institute of St. Joseph's Hamilton, Hamilton, Ontario, Canada
| | - Karen-Anne McVey Neufeld
- McMaster Brain-Body Institute, The Research Institute of St. Joseph's Hamilton, Hamilton, Ontario, Canada; Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Paul Forsythe
- McMaster Brain-Body Institute, The Research Institute of St. Joseph's Hamilton, Hamilton, Ontario, Canada; Department of Medicine, McMaster University, Hamilton, Ontario, Canada; Firestone Institute for Respiratory Health, St. Joseph's Healthcare and Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
65
|
Kayyal M, Javkar T, Firoz Mian M, Binyamin D, Koren O, McVey Neufeld KA, Forsythe P. Sex dependent effects of post-natal penicillin on brain, behavior and immune regulation are prevented by concurrent probiotic treatment. Sci Rep 2020; 10:10318. [PMID: 32587382 PMCID: PMC7316860 DOI: 10.1038/s41598-020-67271-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/29/2020] [Indexed: 12/23/2022] Open
Abstract
There is increasing awareness of the need to consider potential long-term effects of antibiotics on the health of children. In addition to being associated with immune and metabolic diseases, there is evidence that early-life antibiotic exposure can affect neurodevelopment. Here we investigated the effect of low dose of penicillin V on mice when administered for 1 week immediately prior to weaning. We demonstrated that exposure to the antibiotic during the pre-weaning period led to long-term changes in social behaviour, but not anxiety-like traits, in male mice only. The change in behaviour of males was associated with decreased hippocampal expression of AVPR1A and AVPR1B while expression of both receptors was increased in females. Spleens of male mice also showed an increase in the proportion of activated dendritic cells and a corresponding decrease in regulatory T cells with penicillin exposure. All changes in brain, behaviour and immune cell populations, associated with penicillin exposure, were absent in mice that received L. rhamnosus JB-1 supplementation concurrent with the antibiotic. Our study indicates that post-natal exposure to a clinically relevant dose of antibiotic has long-term, sex dependent effects on the CNS and may have implications for the development of neuropsychiatric disorders. Importantly, we also provide further evidence that probiotic based strategies may be of use in counteracting detrimental effects of early-life antibiotics on neurodevelopment.
Collapse
Affiliation(s)
- Marya Kayyal
- McMaster Brain-Body Institute at St Joseph's Healthcare Hamilton, Hamilton, Canada
| | - Tanvi Javkar
- McMaster Brain-Body Institute at St Joseph's Healthcare Hamilton, Hamilton, Canada
| | - M Firoz Mian
- McMaster Brain-Body Institute at St Joseph's Healthcare Hamilton, Hamilton, Canada
| | - Dana Binyamin
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Omry Koren
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Karen-Anne McVey Neufeld
- McMaster Brain-Body Institute at St Joseph's Healthcare Hamilton, Hamilton, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| | - Paul Forsythe
- McMaster Brain-Body Institute at St Joseph's Healthcare Hamilton, Hamilton, Canada.
- Department of Medicine, McMaster University, Hamilton, Canada.
- Firestone Institute for Respiratory Health, St Joseph's Healthcare Hamilton, Hamilton, Canada.
| |
Collapse
|
66
|
Manirarora JN, Kosiewicz MM, Alard P. Feeding lactobacilli impacts lupus progression in (NZBxNZW)F1 lupus-prone mice by enhancing immunoregulation. Autoimmunity 2020; 53:323-332. [PMID: 32552071 DOI: 10.1080/08916934.2020.1777282] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Although the relationship between autoimmunity and microorganisms is complex, there is evidence that microorganisms can prevent the development of various autoimmune diseases. Lactobacilli are beneficial gut bacteria that play an important role in immune system development. The goals of this study were to assess the ability of three different strains of lactobacilli (L. casei B255, L. reuteri DSM 17509 and L. plantarum LP299v) to control lupus development/progression in (NZBxNZW)F1 (BWF1) lupus-prone mice before and after disease onset, and identify the mechanisms mediating protection. BWF1 mice fed with individual L. casei or L. reuteri before disease onset exhibited delayed lupus onset and increased survival, while feeding L. plantarum had little impact. In vitro treatment of BWF1 dendritic cells with individual lactobacilli strains upregulated IL-10 production to various extents, with L. casei being the most effective. The protection mediated by L. casei was associated with upregulation of B7-1 and B7-2 by antigen presenting cells, two costimulatory molecules important for regulatory T cell (Treg) induction. Moreover, feeding L. casei lead to increased percentages of CD4+Foxp3+ Tregs and IL10-producing T cells in the lymphoid organs of treated mice. More importantly, mice fed L. casei after disease onset remained stable for several months, i.e. exhibited delayed anti-nucleic acid production and kidney disease progression, and increased survival. Therefore, feeding lactobacilli appears to delay lupus progression possibly via mechanisms involving Treg induction and IL-10 production. Altogether, these data support the notion that ingestion of lactobacilli, with immunoregulatory properties, may be a viable strategy for controlling disease development and progression in patients with lupus, i.e. extending remission length and reducing flare frequency.
Collapse
Affiliation(s)
- Jean N Manirarora
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA
| | - Michele M Kosiewicz
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA
| | - Pascale Alard
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA
| |
Collapse
|
67
|
The link “Cancer and autoimmune diseases” in the light of microbiota: Evidence of a potential culprit. Immunol Lett 2020; 222:12-28. [DOI: 10.1016/j.imlet.2020.03.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/20/2020] [Accepted: 03/03/2020] [Indexed: 12/15/2022]
|
68
|
Talepoor AG, Fouladseresht H, Khosropanah S, Doroudchi M. Immune-Inflammation in Atherosclerosis: A New Twist in an Old Tale. Endocr Metab Immune Disord Drug Targets 2020; 20:525-545. [DOI: 10.2174/1871530319666191016095725] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/26/2019] [Accepted: 09/23/2019] [Indexed: 12/27/2022]
Abstract
Background and Objective:Atherosclerosis, a chronic and progressive inflammatory disease, is triggered by the activation of endothelial cells followed by infiltration of innate and adaptive immune cells including monocytes and T cells in arterial walls. Major populations of T cells found in human atherosclerotic lesions are antigen-specific activated CD4+ effectors and/or memory T cells from Th1, Th17, Th2 and Treg subsets. In this review, we will discuss the significance of T cell orchestrated immune inflammation in the development and progression of atherosclerosis.Discussion:Pathogen/oxidative stress/lipid induced primary endothelial wound cannot develop to a full-blown atherosclerotic lesion in the absence of chronically induced inflammation. While the primary inflammatory response might be viewed as a lone innate response, the persistence of such a profound response over time must be (and is) associated with diverse local and systemic T cell responses. The interplay between T cells and innate cells contributes to a phenomenon called immuneinflammation and has an impact on the progression and outcome of the lesion. In recent years immuneinflammation, an old term, has had a comeback in connecting the puzzle pieces of chronic inflammatory diseases.Conclusion:Taking one-step back and looking from afar at the players of immune-inflammation may help us provide a broader perspective of these complicated interactions. This may lead to the identification of new drug targets and the development of new therapies as well as preventative measures.
Collapse
Affiliation(s)
- Atefe Ghamar Talepoor
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamed Fouladseresht
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shahdad Khosropanah
- Department of Cardiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehrnoosh Doroudchi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
69
|
Relationship between T cells and microbiota in health and disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 171:95-129. [PMID: 32475529 DOI: 10.1016/bs.pmbts.2020.03.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In the past decades, the fields of microbiology and immunology have largely advanced by using germ-free animals and next-generation sequencing. Many studies revealed the relationship among gut microbiota, activation of immune system, and various diseases. Especially, some gut commensals can generate their antigen-specific T cells. It is becoming clear that commensal bacteria have important roles in various autoimmune and inflammatory diseases, such as autism, rheumatoid arthritis (RA), and inflammatory bowel diseases (IBD). Recently, it was reported that commensals contribute to the cancer immune therapy. However, how commensal-specific T cells contribute to the disease development and cancer treatment are not fully understood yet. In this chapter, we will summarize the decade history of the studies associated with commensal-induced T cells and commensal-causing diseases.
Collapse
|
70
|
Zazara DE, Wegmann M, Giannou AD, Hierweger AM, Alawi M, Thiele K, Huber S, Pincus M, Muntau AC, Solano ME, Arck PC. A prenatally disrupted airway epithelium orchestrates the fetal origin of asthma in mice. J Allergy Clin Immunol 2020; 145:1641-1654. [PMID: 32305348 DOI: 10.1016/j.jaci.2020.01.050] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 12/27/2019] [Accepted: 01/29/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Prenatal challenges such as maternal stress perception increase the risk and severity of asthma during childhood. However, insights into the trajectories and targets underlying the pathogenesis of prenatally triggered asthma are largely unknown. The developing lung and immune system may constitute such targets. OBJECTIVE Here we have aimed to identify the differential sex-specific effects of prenatal challenges on lung function, immune response, and asthma severity in mice. METHODS We generated bone marrow chimeric (BMC) mice harboring either prenatally stress-exposed lungs or a prenatally stress-exposed immune (hematopoietic) system and induced allergic asthma via ovalbumin. Next-generation sequencing (RNA sequencing) of lungs and assessment of airway epithelial barrier function in ovalbumin-sensitized control and prenatally stressed offspring was also performed. RESULTS Profoundly enhanced airway hyperresponsiveness, inflammation, and fibrosis were exclusively present in female BMC mice with prenatally stress-exposed lungs. These effects were significantly perpetuated if both the lungs and the immune system had been exposed to prenatal stress. A prenatally stress-exposed immune system alone did not suffice to increase the severity of these asthma features. RNA sequencing analysis of lungs from prenatally stressed, non-BMC, ovalbumin-sensitized females unveiled a deregulated expression of genes involved in asthma pathogenesis, tissue remodeling, and tight junction formation. It was also possible to independently confirm a tight junction disruption. In line with this, we identified an altered perinatal and/or postnatal expression of genes involved in lung development along with an impaired alveolarization in female prenatally stressed mice. CONCLUSION Here we have shown that the fetal origin of asthma is orchestrated by a disrupted airway epithelium and further perpetuated by a predisposed immune system.
Collapse
Affiliation(s)
- Dimitra E Zazara
- Department of Obstetrics and Prenatal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Wegmann
- Division of Asthma Exacerbation & Regulation, Priority Area Asthma and Allergy, Leibniz Lung Center Borstel, Airway Research Center North, Member of the German Center for Lung Research, Borstel, Germany
| | - Anastasios D Giannou
- I. Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Alexandra Maximiliane Hierweger
- Department of Obstetrics and Prenatal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Institute for Immunology, Center for Diagnostics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Malik Alawi
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kristin Thiele
- Department of Obstetrics and Prenatal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Samuel Huber
- I. Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Maike Pincus
- Pediatrics and Pediatric Pneumology Practice, Berlin, Germany
| | - Ania C Muntau
- University Children's Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maria Emilia Solano
- Department of Obstetrics and Prenatal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Petra C Arck
- Department of Obstetrics and Prenatal Medicine, Laboratory for Experimental Feto-Maternal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
71
|
Tashiro H, Kasahara DI, Osgood RS, Brown T, Cardoso A, Cho Y, Shore SA. Sex Differences in the Impact of Dietary Fiber on Pulmonary Responses to Ozone. Am J Respir Cell Mol Biol 2020; 62:503-512. [PMID: 31913653 PMCID: PMC7110971 DOI: 10.1165/rcmb.2019-0124oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 01/08/2020] [Indexed: 12/16/2022] Open
Abstract
Ozone causes airway hyperresponsiveness, a defining feature of asthma. We have reported that the gut microbiome contributes to sex differences in ozone-induced airway hyperresponsiveness. Altering dietary fiber affects the gut microbiome. The purpose of this study was to determine the effects of dietary fiber on pulmonary responses to ozone and whether these effects differ by sex. We fed male and female mice fiber-free diets or diets enriched in one of two types of dietary fiber, cellulose and pectin, for 3 days before ozone exposure. Compared with control diets or pectin-enriched diets, cellulose-enriched diets attenuated ozone-induced airway hyperresponsiveness in male but not female mice. In contrast, fiber-free diets augmented responses to ozone in female but not male mice. Analysis of 16S rRNA sequencing of fecal DNA also indicated sex differences in the impact of dietary fiber on the gut microbiome and identified bacterial taxa that were associated with ozone-induced airway hyperresponsiveness. Our data suggest that microbiome-based therapies such as prebiotics may provide an alternative therapeutic strategy for air pollution-triggered asthma, but they indicate that such therapeutics may need to be tailored differently for males and females.
Collapse
Affiliation(s)
- Hiroki Tashiro
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - David I Kasahara
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Ross S Osgood
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Traci Brown
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Aline Cardoso
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Youngji Cho
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Stephanie A Shore
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
72
|
Lactobacillus reuteri 5454 and Bifidobacterium animalis ssp. lactis 5764 improve colitis while differentially impacting dendritic cells maturation and antimicrobial responses. Sci Rep 2020; 10:5345. [PMID: 32210304 PMCID: PMC7093418 DOI: 10.1038/s41598-020-62161-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 02/26/2020] [Indexed: 12/11/2022] Open
Abstract
Crohn’s disease is linked to a decreased diversity in gut microbiota composition as a potential consequence of an impaired anti-microbial response and an altered polarization of T helper cells. Here, we evaluated the immunomodulatory properties of two potential probiotic strains, namely a Bifidobacterium animalis spp. lactis Bl 5764 and a Lactobacillus reuteri Lr 5454 strains. Both strains improved colitis triggered by either 2,4,6-trinitrobenzenesulfonic acid (TNBS) or Citrobacter rodentium infection in mice. Training of dendritic cells (DC) with Lr 5454 efficiently triggered IL-22 secretion and regulatory T cells induction in vitro, while IL-17A production by CD4+ T lymphocytes was stronger when cultured with DCs that were primed with Bl 5764. This strain was sufficient for significantly inducing expression of antimicrobial peptides in vivo through the Crohn’s disease predisposing gene encoding for the nucleotide-binding oligomerization domain, containing protein 2 (NOD2). In contrast, NOD2 was dispensable for the impact on antimicrobial peptide expression in mice that were monocolonized with Lr 5454. In conclusion, our work highlights a differential mode of action of two potential probiotic strains that protect mice against colitis, providing the rational for a personalized supportive preventive therapy by probiotics for individuals that are genetically predisposed to Crohn’s disease.
Collapse
|
73
|
Shavandi A, Saeedi P, Gérard P, Jalalvandi E, Cannella D, Bekhit AED. The role of microbiota in tissue repair and regeneration. J Tissue Eng Regen Med 2020; 14:539-555. [PMID: 31845514 DOI: 10.1002/term.3009] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 09/15/2019] [Accepted: 10/28/2019] [Indexed: 12/22/2022]
Abstract
A comprehensive understanding of the human body endogenous microbiota is essential for acquiring an insight into the involvement of microbiota in tissue healing and regeneration process in order to enable development of biomaterials with a better integration with human body environment. Biomaterials used for biomedical applications are normally germ-free, and the human body as the host of the biomaterials is not germ-free. The complexity and role of the body microbiota in tissue healing/regeneration have been underestimated historically. Traditionally, studies aiming at the development of novel biomaterials had focused on the effects of environment within the target tissue, neglecting the signals generated from the microbiota and their impact on tissue regeneration. The significance of the human body microbiota in relation to metabolism, immune system, and consequently tissue regeneration has been recently realised and is a growing research field. This review summarises recent findings on the role of microbiota and mechanisms involved in tissue healing and regeneration, in particular skin, liver, bone, and nervous system regrowth and regeneration highlighting the potential new roles of microbiota for development of a new generation of biomaterials.
Collapse
Affiliation(s)
- Amin Shavandi
- BioMatter-BTL, École interfacultaire de Bioingénieurs (EIB), Université Libre de Brussels, Brussels, Belgium
| | - Pouya Saeedi
- Department of Human Nutrition, University of Otago, Dunedin, New Zealand
| | - Philippe Gérard
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| | - Esmat Jalalvandi
- School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, UK
| | - David Cannella
- PhotoBioCatalysis Unit - BTL - École interfacultaire de Bioingénieurs (EIB), Université Libre de Brussels, Brussels, Belgium
| | | |
Collapse
|
74
|
Stockert K. Synopsis. ALLERGIEPRÄVENTION 2020. [PMCID: PMC7121829 DOI: 10.1007/978-3-662-58140-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Akute entzündliche Reaktionen bzw. der akute Infekt mit Restitutio ad integrum laufen in einer perfekt modulierten Kaskade ab, bei dem eine akute inflammatorische Einleitungsphase von einer antiinflammatorischen Phase und einer Entzündungsauflösungsphase abgelöst werden.
Collapse
|
75
|
RORγt + Treg to Th17 ratios correlate with susceptibility to Giardia infection. Sci Rep 2019; 9:20328. [PMID: 31889073 PMCID: PMC6937251 DOI: 10.1038/s41598-019-56416-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 11/15/2019] [Indexed: 12/20/2022] Open
Abstract
Infections with Giardia are among the most common causes of food and water-borne diarrheal disease worldwide. Here, we investigated Th17, Treg and IgA responses, and alterations in gut microbiota in two mouse lines with varying susceptibility to Giardia muris infection. Infected BALB/c mice shed significantly more cysts compared with C57BL/6 mice. Impaired control of infection in BALB/c mice was associated with lower Th17 activity and lower IgA levels compared with C57BL/6 mice. The limited metabolic activity, proliferation and cytokine production of Th17 cells in BALB/c mice was associated with higher proportions of intestinal Foxp3+RORγt+ regulatory T cells and BALB/c mice developed increased RORγt+ Treg:Th17 ratios in response to G. muris infection. Furthermore, G. muris colonization led to a significantly reduced evenness in the gut microbial communities of BALB/c mice. Our data indicate that differential susceptibility to Giardia infections may be related to RORγt+ Treg controlling Th17 activity and that changes in the microbiota composition upon Giardia infection partially depend on the host background.
Collapse
|
76
|
Lung Microbiome in Asthma: Current Perspectives. J Clin Med 2019; 8:jcm8111967. [PMID: 31739446 PMCID: PMC6912699 DOI: 10.3390/jcm8111967] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 11/12/2019] [Indexed: 12/20/2022] Open
Abstract
A growing body of evidence implicates the human microbiome as a potentially influential player actively engaged in shaping the pathogenetic processes underlying the endotypes and phenotypes of chronic respiratory diseases, particularly of the airways. In this article, we specifically review current evidence on the characteristics of lung microbiome, and specifically the bacteriome, the modes of interaction between lung microbiota and host immune system, the role of the “lung–gut axis”, and the functional effects thereof on asthma pathogenesis. We also attempt to explore the possibilities of therapeutic manipulation of the microbiome, aiming at the establishment of asthma prevention strategies and the optimization of asthma treatment.
Collapse
|
77
|
Yu S, Su C, Luo X. Impact of infection on transplantation tolerance. Immunol Rev 2019; 292:243-263. [PMID: 31538351 PMCID: PMC6961566 DOI: 10.1111/imr.12803] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/29/2019] [Accepted: 09/03/2019] [Indexed: 12/12/2022]
Abstract
Allograft tolerance is the ultimate goal of organ transplantation. Current strategies for tolerance induction mainly focus on inhibiting alloreactive T cells while promoting regulatory immune cells. Pathogenic infections may have direct impact on both effector and regulatory cell populations, therefore can alter host susceptibility to transplantation tolerance induction as well as impair the quality and stability of tolerance once induced. In this review, we will discuss existing data demonstrating the effect of infections on transplantation tolerance, with particular emphasis on the role of the stage of infection (acute, chronic, or latent) and the stage of tolerance (induction or maintenance) in this infection-tolerance interaction. While the deleterious effect of acute infection on tolerance is mainly driven by proinflammatory cytokines induced shortly after the infection, chronic infection may generate exhausted T cells that could in fact facilitate transplantation tolerance. In addition to pathogenic infections, commensal intestinal microbiota also has numerous significant immunomodulatory effects that can shape the host alloimmunity following transplantation. A comprehensive understanding of these mechanisms is crucial for the development of therapeutic strategies for robustly inducing and stably maintaining transplantation tolerance while preserving host anti-pathogen immunity in clinically relevant scenarios.
Collapse
Affiliation(s)
- Shuangjin Yu
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, United States
- Division of Organ transplantation, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Chang Su
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, United States
| | - Xunrong Luo
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, United States
- Duke Transplant Center, Duke University School of Medicine, Durham, NC 27710, United States
| |
Collapse
|
78
|
Wypych TP, Wickramasinghe LC, Marsland BJ. The influence of the microbiome on respiratory health. Nat Immunol 2019; 20:1279-1290. [PMID: 31501577 DOI: 10.1038/s41590-019-0451-9] [Citation(s) in RCA: 313] [Impact Index Per Article: 62.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 06/18/2019] [Indexed: 02/06/2023]
Abstract
The revolution in microbiota research over the past decade has provided invaluable knowledge about the function of the microbial species that inhabit the human body. It has become widely accepted that these microorganisms, collectively called 'the microbiota', engage in networks of interactions with each other and with the host that aim to benefit both the microbial members and the mammalian members of this unique ecosystem. The lungs, previously thought to be sterile, are now known to harbor a unique microbiota and, additionally, to be influenced by microbial signals from distal body sites, such as the intestine. Here we review the role of the lung and gut microbiotas in respiratory health and disease and highlight the main pathways of communication that underlie the gut-lung axis.
Collapse
Affiliation(s)
- Tomasz P Wypych
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia.
| | - Lakshanie C Wickramasinghe
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Benjamin J Marsland
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
79
|
Haahtela T. A biodiversity hypothesis. Allergy 2019; 74:1445-1456. [PMID: 30835837 DOI: 10.1111/all.13763] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 02/11/2019] [Accepted: 02/18/2019] [Indexed: 02/06/2023]
Abstract
Biodiversity hypothesis states that contact with natural environments enriches the human microbiome, promotes immune balance and protects from allergy and inflammatory disorders. We are protected by two nested layers of biodiversity, microbiota of the outer layer (soil, natural waters, plants, animals) and inner layer (gut, skin, airways). The latter inhabits our body and is colonized from the outer layer. Explosion of human populations along with cultural evolution is profoundly changing our environment and lifestyle. Adaptive immunoregulatory circuits and dynamic homeostasis are at stake in the newly emerged urban surroundings. In allergy, and chronic inflammatory disorders in general, exploring the determinants of immunotolerance is the key for prevention and more effective treatment. Loss of immunoprotective factors, derived from nature, is a new kind of health risk poorly acknowledged until recently. The paradigm change has been implemented in the Finnish allergy programme (2008-2018), which emphasized tolerance instead of avoidance. The first results are promising, as allergy burden has started to reduce. The rapidly urbanizing world is facing serious biodiversity loss with global warming, which are interconnected. Biodiversity hypothesis of health and disease has societal impact, for example, on city planning, food and energy production and nature conservation. It has also a message for individuals for health and well-being: take nature close, to touch, eat, breathe, experience and enjoy. Biodiverse natural environments are dependent on planetary health, which should be a priority also among health professionals.
Collapse
Affiliation(s)
- Tari Haahtela
- Skin and Allergy Hospital, Helsinki University Hospital University of Helsinki Helsinki Finland
| |
Collapse
|
80
|
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: 9.2] [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
|
81
|
Nunes CF, Nogueira JS, Vianna PHO, Ciambarella BT, Rodrigues PM, Miranda KR, Lobo LA, Domingues RMCP, Busch M, Atella GC, Vale AM, Bellio M, Nóbrega A, Canto FB, Fucs R. Probiotic treatment during neonatal age provides optimal protection against experimental asthma through the modulation of microbiota and T cells. Int Immunol 2019; 30:155-169. [PMID: 29420746 DOI: 10.1093/intimm/dxy011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 02/02/2018] [Indexed: 02/06/2023] Open
Abstract
The incidence of allergic diseases, which increased to epidemic proportions in developed countries over the last few decades, has been correlated with altered gut microbiota colonization. Although probiotics may play a critical role in the restoration of gut homeostasis, their efficiency in the control of allergy is controversial. Here, we aimed to investigate the effects of probiotic treatment initiated at neonatal or adult ages on the suppression of experimental ovalbumin (OVA)-induced asthma. Neonatal or adult mice were orally treated with probiotic bacteria and subjected to OVA-induced allergy. Asthma-like symptoms, microbiota composition and frequencies of the total CD4+ T lymphocytes and CD4+Foxp3+ regulatory T (Treg) cells were evaluated in both groups. Probiotic administration to neonates, but not to adults, was necessary and sufficient for the absolute prevention of experimental allergen-induced sensitization. The neonatally acquired tolerance, transferrable to probiotic-untreated adult recipients by splenic cells from tolerant donors, was associated with modulation of gut bacterial composition, augmented levels of cecum butyrate and selective accumulation of Treg cells in the airways. Our findings reveal that a cross-talk between a healthy microbiota and qualitative features inherent to neonatal T cells, especially in the Treg cell subset, might support the beneficial effect of perinatal exposure to probiotic bacteria on the development of long-term tolerance to allergens.
Collapse
Affiliation(s)
- Caroline Fraga Nunes
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Goés (IMPG) - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Departamento de Imunobiologia, Instituto de Biologia - Universidade Federal Fluminense, Niterói - RJ, Brazil
| | - Jeane S Nogueira
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Goés (IMPG) - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Departamento de Imunobiologia, Instituto de Biologia - Universidade Federal Fluminense, Niterói - RJ, Brazil
| | - Pedro Henrique Oliveira Vianna
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Goés (IMPG) - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Karla Rodrigues Miranda
- Faculdade de Farmácia - Universidade Federal do Rio de Janeiro, Rio de Janeiro/Campus Macaé, Macaé - RJ, Brazil
| | - Leandro Araújo Lobo
- Departamento de Microbiologia Médica, Instituto de Microbiologia Paulo de Goés (IMPG) - Universidade Federal do Rio de Janeiro, Rio de Janeiro - RJ, Brazil
| | | | - Mileane Busch
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro - RJ, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, INCT-EM, Rio de Janeiro - RJ, Brazil
| | - Georgia Correa Atella
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro - RJ, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, INCT-EM, Rio de Janeiro - RJ, Brazil
| | - André Macedo Vale
- Laboratório de Imunorreceptores e Sinalização, Instituto de Biofísica Carlos Chagas Filho - Universidade Federal do Rio de Janeiro, Rio de Janeiro - RJ, Brazil
| | - Maria Bellio
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Goés (IMPG) - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alberto Nóbrega
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Goés (IMPG) - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fábio B Canto
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Goés (IMPG) - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Departamento de Imunobiologia, Instituto de Biologia - Universidade Federal Fluminense, Niterói - RJ, Brazil
| | - Rita Fucs
- Departamento de Imunobiologia, Instituto de Biologia - Universidade Federal Fluminense, Niterói - RJ, Brazil
| |
Collapse
|
82
|
Barcik W, Pugin B, Brescó MS, Westermann P, Rinaldi A, Groeger D, Van Elst D, Sokolowska M, Krawczyk K, Frei R, Ferstl R, Wawrzyniak M, Altunbulakli C, Akdis CA, O'Mahony L. Bacterial secretion of histamine within the gut influences immune responses within the lung. Allergy 2019; 74:899-909. [PMID: 30589936 DOI: 10.1111/all.13709] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 11/20/2018] [Accepted: 11/30/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Histamine is an important immunomodulator influencing both the innate and adaptive immune system. Certain host cells express the histidine decarboxylase enzyme (HDC), which is responsible for catalysing the decarboxylation of histidine to histamine. We and others have shown that bacterial strains can also express HDC and secrete histamine; however, the influence of bacterial-derived histamine on the host immune responses distant to the gut is unclear. METHODS The Escherichia coli BL21 (E coli BL21) strain was genetically modified to express the Morganella morganii (M morganii)-derived HDC gene (E coli BL21_HTW). E coli BL21 and E coli BL21_HTW were gavaged to ovalbumin (OVA) sensitized and challenged mice to investigate the effect of bacterial-derived histamine on lung inflammatory responses. RESULTS Oral administration of E coli BL21_HTW, which is able to secrete histamine, to wild-type mice reduced lung eosinophilia and suppressed ex vivo OVA-stimulated cytokine secretion from lung cells in the OVA respiratory inflammation mouse model. In histamine receptor 2 (H2R)-deficient mice, administration of histamine-secreting bacteria also reduced inflammatory cell numbers in bronchoalveolar lavage (BAL). However, the suppressive effect of bacterial-derived histamine on BAL inflammation was lost in HDC-deficient mice. This loss of activity was associated with increased expression of histamine degrading enzymes and reduced histamine receptor expression. CONCLUSION Histamine secretion from bacteria within the gut can have immunological consequences at distant mucosal sites, such as within the lung. These effects are influenced by host histamine receptor expression and the expression of histamine degrading enzymes.
Collapse
Affiliation(s)
- Weronika Barcik
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Benoit Pugin
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Marina Sabaté Brescó
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Patrick Westermann
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Arturo Rinaldi
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Christine Kühne‐Center for Allergy Research and Education Davos Switzerland
| | - David Groeger
- Alimentary Health Pharma Davos (AHPD) Davos Switzerland
| | - Dries Van Elst
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Christine Kühne‐Center for Allergy Research and Education Davos Switzerland
| | - Krzysztof Krawczyk
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Remo Frei
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Christine Kühne‐Center for Allergy Research and Education Davos Switzerland
| | - Ruth Ferstl
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Christine Kühne‐Center for Allergy Research and Education Davos Switzerland
| | - Marcin Wawrzyniak
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Can Altunbulakli
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Christine Kühne‐Center for Allergy Research and Education Davos Switzerland
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Christine Kühne‐Center for Allergy Research and Education Davos Switzerland
| | - Liam O'Mahony
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Departments of Medicine and Microbiology APC Microbiome Ireland National University of Ireland Cork Ireland
| |
Collapse
|
83
|
Litus O, Derkach N, Litus V, Bisyuk Y, Lytvynenko B. Efficacy of Probiotic Therapy on Atopic Dermatitis in Adults Depends on the C-159T Polymorphism of the CD14 Receptor Gene - A Pilot Study. Open Access Maced J Med Sci 2019; 7:1053-1058. [PMID: 31049080 PMCID: PMC6490491 DOI: 10.3889/oamjms.2019.242] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/22/2019] [Accepted: 03/23/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND: The C-159T polymorphism of the CD14 receptor gene can be associated with the development of atopic dermatitis. Probiotics can modulate chronic inflammation through activation of the CD14 receptor. So, the efficacy of probiotic therapy can be dependent on this genetic polymorphism. AIM: The purpose of the study was to investigate the efficacy of adding probiotic (Lactobacillus acidophilus, LA-5 and Bifidobacterium animalis subsp. lactis, BB-12) to standard treatment (ointment of fluticasone propionate 0.005% and emollient) of atopic dermatitis in adults during 28 days, depending on the stratification of patients on CC or TT genotypes of the CD14 receptor gene. MATERIAL AND METHODS: The study included 37 adult patients with AD. There were identified 19 patients with exogenous (IgE-dependent) and 18 with endogenous (IgE-dependent) AD. To evaluate the efficacy of the probiotics all patients were divided into three groups for both exogenous and endogenous AD. The first group was selected from patients with CC genotype (C-159T) who received standard therapy (ointment of fluticasone propionate 0.005% – 2 times a day, emollients – 2 times a day) and probiotic (Lactobacillus acidophilus, LA-5 and Bifidobacterium animalis subsp. lactis, BB-12 - 1 capsule 2 times per day) The second group included patients with CC genotype, who received only standard therapy. The third group was presented by patients with TT genotype (C-159T) who received standard therapy and probiotic. The SCORAD and DLQI parameters were evaluated on Day 0, 14 and 28. The level of IL-4, IL-5, IL-10, TGF-β cytokines was determined on Day 0 and Day 28. RESULTS: The results of our study found that the addition of probiotics (Lactobacillus acidophilus, LA-5 and Bifidobacterium animalis subsp. lactis, BB-12) to standard treatment (ointment of fluticasone propionate 0.005%, emollient) significantly increased the effectiveness of treatment of atopic dermatitis in adults with exogenous form and CC genotype (C-159T), confirmed by clinical (a significant decrease of SCORAD and DLQI indices) and immunological criteria (a significant decrease of IL-4 and an increase of TGF-β). CONCLUSION: Simultaneous determination of the exogenous or endogenous form, identification of the C-159T genotypes, evaluation of the serum level of IL-4 and TGF-β can serve as an algorithm for the personalised treatment of patients with atopic dermatitis.
Collapse
Affiliation(s)
- Oleksandr Litus
- Shupyk National Medical Academy of Postgraduate Education, Kiev, Ukraine
| | - Nadiya Derkach
- Shupyk National Medical Academy of Postgraduate Education, Kiev, Ukraine
| | - Viktor Litus
- Shupyk National Medical Academy of Postgraduate Education, Kiev, Ukraine
| | - Yuriy Bisyuk
- Shupyk National Medical Academy of Postgraduate Education, Kiev, Ukraine
| | - Bohdan Lytvynenko
- Shupyk National Medical Academy of Postgraduate Education, Kiev, Ukraine
| |
Collapse
|
84
|
Kim JY, Kim EM, Yi MH, Lee J, Lee S, Hwang Y, Yong D, Sohn WM, Yong TS. Chinese liver fluke Clonorchis sinensis infection changes the gut microbiome and increases probiotic Lactobacillus in mice. Parasitol Res 2019; 118:693-699. [PMID: 30623233 DOI: 10.1007/s00436-018-6179-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 12/11/2018] [Indexed: 12/26/2022]
Abstract
Chinese liver fluke Clonorchis sinensis changes the host's immune system. Recently, it has been reported that helminths including C. sinensis can ameliorate immune-related diseases such as allergy. In addition, recent studies showed that helminth infection can alleviate immune-mediated disorders by altering the gut microbiome. However, changes in the gut microbiome due to C. sinensis have not been reported yet. In this study, changes in the gut microbiome of C57BL/6 mice infected with C. sinensis metacercariae were evaluated over time. Stool was analyzed by 16S rRNA amplicon analysis using high-throughput sequencing technology. There was no apparent difference in species richness and diversity between the infected and control groups. However, the composition of the microbiome was different between the infected and control groups at 20 days and 30 days post-infection, and the difference disappeared at 50 days post-infection. In particular, this microbiome alteration was associated with a change in the relative abundance of genus Lactobacillus and the probiotic Lactobacillus species that are known to have an immune-modulation role in immune-mediated diseases.
Collapse
Affiliation(s)
- Ju Yeong Kim
- Department of Environmental Medical Biology, Institute of Tropical Medicine, and Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Seoul, 03722, Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Eun-Min Kim
- Department of Environmental Medical Biology, Institute of Tropical Medicine, and Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Myung-Hee Yi
- Department of Environmental Medical Biology, Institute of Tropical Medicine, and Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Jinyoung Lee
- Department of Environmental Medical Biology, Institute of Tropical Medicine, and Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Seogwon Lee
- Department of Environmental Medical Biology, Institute of Tropical Medicine, and Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Younjee Hwang
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Korea.,Department of Laboratory Medicine and Research Institute of Bacterial Resistance, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Dongeun Yong
- Department of Laboratory Medicine and Research Institute of Bacterial Resistance, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Woon-Mok Sohn
- Department of Parasitology and Tropical Medicine, and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, 52727, Korea
| | - Tai-Soon Yong
- Department of Environmental Medical Biology, Institute of Tropical Medicine, and Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Seoul, 03722, Korea.
| |
Collapse
|
85
|
Moura JCV, Moura ICG, Gaspar GR, Mendes GMS, Faria BAV, Jentzsch NS, do Carmo Friche Passos M, Kurdi A, Godman B, Almeida AM. The use of probiotics as a supplementary therapy in the treatment of patients with asthma: a pilot study and implications. Clinics (Sao Paulo) 2019; 74:e950. [PMID: 31411278 PMCID: PMC6683305 DOI: 10.6061/clinics/2019/e950] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 04/17/2019] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES Evaluate the use of probiotics as an additional therapy in the treatment of children and adolescents with asthma in Belo Horizonte, MG-Brazil. METHODS A pilot longitudinal, experimental and nonrandomized study with 30 patients from six to 17 years old from Belo Horizonte. In the baseline appointment, all patients received beclomethasone, and one group also received a probiotic containing Lactobacillus reuteri (n=14). The patients were reassessed after at least 60 days with the Asthma Control Test, spirometry and self-report of the symptoms they experienced associated with asthma. RESULTS A predominance of male patients (56.7%) and a mean age of 10.6 years were observed. The groups using probiotics did not differ in terms of sex, age or atopy. In the longitudinal evaluation, an increase in the Asthma Control Test scores and a reduction in the number of symptoms were observed in the probiotic group. There was an increase in the peak expiratory flow among those who used probiotics. CONCLUSIONS This pilot study supports the hypothesis that the administration of probiotics as a supplementary therapy for the treatment of children and adolescents with asthma improves the clinical condition of the patients. Further studies are needed to confirm the efficacy of probiotics in asthma treatment.
Collapse
Affiliation(s)
| | - Isabel Cristina Gomes Moura
- Programa de Pos Graduacao em Ciencias da Saude, Faculdade de Ciencias Medicas de Minas Gerais, Belo Horizonte, MG, BR
- *Corresponding author. E-mail:
| | | | | | | | - Nulma Souto Jentzsch
- Faculdade de Medicina, Faculdade de Ciencias Medicas de Minas Gerais, Belo Horizonte, MG, BR
| | - Maria do Carmo Friche Passos
- Programa de Pos Graduacao em Ciencias da Saude, Faculdade de Ciencias Medicas de Minas Gerais, Belo Horizonte, MG, BR
| | - Amanj Kurdi
- Pharmacoepidemiology and Pharmacy Practice, Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Scotland, UK
- Department of pharmacology, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | - Brian Godman
- Pharmacoepidemiology and Pharmacy Practice, Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Scotland, UK
- Division of Clinical Pharmacology, Karolinska Institute, Stockholm, Sweden
- Department of Public Health Pharmacy and Management, School of Pharmacy, Sefako Makgatho Health Sciences University, Garankuwa, South Africa
| | - Alessandra Maciel Almeida
- Programa de Pos Graduacao em Ciencias da Saude, Faculdade de Ciencias Medicas de Minas Gerais, Belo Horizonte, MG, BR
| |
Collapse
|
86
|
Abstract
The lung and gut microbiome are factors in asthma risk or protection. Relevant elements of the microbiome within both niches include the importance of the early life window for microbiome establishment, the diversity of bacteria, richness of bacteria, and effect of those bacteria on the local epithelium and immune system. Mechanisms of protection include direct anti-inflammatory action or induction of non-type 2 inflammation by certain bacterial colonies. The gut microbiome further impacts asthma risk through the contribution of metabolic products. This article reviews the mechanisms that connect the lung and gut microbiota to asthma development and severity.
Collapse
Affiliation(s)
- Tara F Carr
- Department of Medicine, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ 85724-5030, USA.
| | - Rhonda Alkatib
- Department of Medicine, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ 85724-5030, USA
| | - Monica Kraft
- Department of Medicine, University of Arizona, 1501 North Campbell Avenue, Tucson, AZ 85724-5030, USA
| |
Collapse
|
87
|
Qiu D, Xia Z, Jiao X, Deng J, Zhang L, Li J. Altered Gut Microbiota in Myasthenia Gravis. Front Microbiol 2018; 9:2627. [PMID: 30483222 PMCID: PMC6241162 DOI: 10.3389/fmicb.2018.02627] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 10/15/2018] [Indexed: 01/01/2023] Open
Abstract
Myasthenia gravis (MG) is an autoimmune-mediated disorder, the etiology of which involves both environmental factors and genetics. While the exact factors responsible for predisposition to MG remain elusive, it is hypothesized that gut microbiota play a critical role in the pathogenesis of MG. This study investigated whether gut microbiota are altered in MG patients by comparing the fecal microbiota profiles of MG patients to those of age- and sex-matched healthy controls. Phylotype profiles of gut microbial populations were generated using hypervariable tag sequencing of the V4 region of the 16S ribosomal RNA gene. Fecal short-chain fatty acids (SCFAs) were assessed by gas chromatographic analyses. The results demonstrated that, compared to the healthy cohort, the gut microbiota of the MG group was changed in terms of the relative abundances of bacterial taxa, with sharply reduced microbial richness, particularly in the genus Clostridium. The fecal SCFA content was significantly lower in the MG group. Furthermore, microbial dysbiosis was closely related to the levels of inflammatory biomarkers in the sera of MG patients.
Collapse
Affiliation(s)
- Dongxu Qiu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Neurology of Hunan Province, Changsha, China
| | - Zhiwei Xia
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Neurology of Hunan Province, Changsha, China
| | - Xiao Jiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Neurology of Hunan Province, Changsha, China
| | - Jun Deng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Neurology of Hunan Province, Changsha, China
| | - Lei Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Jing Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Neurology of Hunan Province, Changsha, China
| |
Collapse
|
88
|
The clinical role of the TME in solid cancer. Br J Cancer 2018; 120:45-53. [PMID: 30413828 PMCID: PMC6325164 DOI: 10.1038/s41416-018-0327-z] [Citation(s) in RCA: 397] [Impact Index Per Article: 66.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 10/02/2018] [Accepted: 10/09/2018] [Indexed: 12/14/2022] Open
Abstract
The highly complex and heterogenous ecosystem of a tumour not only contains malignant cells, but also interacting cells from the host such as endothelial cells, stromal fibroblasts, and a variety of immune cells that control tumour growth and invasion. It is well established that anti-tumour immunity is a critical hurdle that must be overcome for tumours to initiate, grow and spread and that anti-tumour immunity can be modulated using current immunotherapies to achieve meaningful anti-tumour clinical responses. Pioneering studies in melanoma, ovarian and colorectal cancer have demonstrated that certain features of the tumour immune microenvironment (TME)-in particular, the degree of tumour infiltration by cytotoxic T cells-can predict a patient's clinical outcome. More recently, studies in renal cell cancer have highlighted the importance of assessing the phenotype of the infiltrating T cells to predict early relapse. Furthermore, intricate interactions with non-immune cellular players such as endothelial cells and fibroblasts modulate the clinical impact of immune cells in the TME. Here, we review the critical components of the TME in solid tumours and how they shape the immune cell contexture, and we summarise numerous studies evaluating its clinical significance from a prognostic and theranostic perspective.
Collapse
|
89
|
Vitetta L, Vitetta G, Hall S. Immunological Tolerance and Function: Associations Between Intestinal Bacteria, Probiotics, Prebiotics, and Phages. Front Immunol 2018; 9:2240. [PMID: 30356736 PMCID: PMC6189397 DOI: 10.3389/fimmu.2018.02240] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/10/2018] [Indexed: 12/12/2022] Open
Abstract
Post-birth there is a bacterial assault on all mucosal surfaces. The intestinal microbiome is an important participant in health and disease. The pattern of composition and concentration of the intestinal microbiome varies greatly. Therefore, achieving immunological tolerance in the first 3-4 years of life is critical for maintaining health throughout a lifetime. Probiotic bacteria are organisms that afford beneficial health effects to the host and in certain instances may protect against the development of disease. The potential benefits of modifying the composition of the intestinal microbial cohort for therapeutic benefit is evident in the use in high risks groups such as premature infants, children receiving antibiotics, rotavirus infections in non-vaccinated children and traveler's diarrhea in adults. Probiotics and prebiotics are postulated to have immunomodulating capabilities by influencing the intestinal microbial cohort and dampening the activity of pathobiont intestinal microbes, such as Klebsiella pneumonia and Clostridia perfringens. Lactobacilli and Bifidobacteria are examples of probiotics found in the large intestine and so far, the benefits afforded to probiotics have varied in efficacy. Most likely the efficacy of probiotic bacteria has a multifactorial dependency, namely on a number of factors that include agents used, the dose, the pattern of dosing, and the characteristics of the host and the underlying luminal microbial environment and the activity of bacteriophages. Bacteriophages, are small viruses that infect and lyse intestinal bacteria. As such it can be posited that these viruses display an effective local protective control mechanism for the intestinal barrier against commensal pathobionts that indirectly may assist the host in controlling bacterial concentrations in the gut. A co-operative activity may be envisaged between the intestinal epithelia, mucosal immunity and the activity of bacteriophages to eliminate pathobiots, highlighting the potential role of bacteriophages in assisting with maintaining intestinal homeostasis. Hence bacteriophage local control of inflammation and immune responses may be an additional immunological defense mechanism that exploits bacteriophage-mucin glycoprotein interactions that controls bacterial diversity and abundance in the mucin layers of the gut. Moreover, and importantly the efficacy of probiotics may be dependent on the symbiotic incorporation of prebiotics, and the abundance and diversity of the intestinal microbiome encountered. The virome may be an important factor that determines the efficacy of some probiotic formulations.
Collapse
Affiliation(s)
- Luis Vitetta
- Discipline of Pharmacology, Faculty of Medicine and Health, School of Medicine, The University of Sydney, Camperdown, NSW, Australia
- Medlab Clinical Ltd., Sydney, NSW, Australia
| | | | - Sean Hall
- Medlab Clinical Ltd., Sydney, NSW, Australia
| |
Collapse
|
90
|
Fuglsang E, Krych L, Lundsager MT, Nielsen DS, Frøkiaer H. Postnatal Administration of Lactobacillus rhamnosus HN001 Ameliorates Perinatal Broad-Spectrum Antibiotic-Induced Reduction in Myelopoiesis and T Cell Activation in Mouse Pups. Mol Nutr Food Res 2018; 62:e1800510. [PMID: 30211987 DOI: 10.1002/mnfr.201800510] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/03/2018] [Indexed: 12/31/2022]
Abstract
SCOPE This study addresses whether administration of Lactobacillus rhamnosus HN001 could mitigate the effects of a compromised gut microbiota on the composition of mature leukocytes and granulocyte-macrophage progenitor cells (GMPs) in newborn mice. METHODS AND RESULTS Pregnant dams receive oral broad-spectrum antibiotics, which dramatically decrease the gut microbial composition analyzed by 16S rRNA sequencing. Perinatal antibiotic treatment decreases the proportions of bone marrow (BM) GMPs (postnatal day (PND2): 0.5% vs 0.8%, PND4: 0.2% to 0.6%) and mature granulocytes (33% vs 24% at PND2), and spleen granulocytes (7% vs 17% at PND2) and B cells (PND2:18% vs 28%, PND4:11% vs 22%). At PND35, T helper (Th) cells (20% vs 14%) and cytotoxic T (Tc) cells (10% vs 8%) decrease in the spleen. Oral administration of L. rhamnosus HN001 to neonatal pups (PND1-7) restores the antibiotic-induced changes of GMPs and granulocytes in BM and spleen, and further increases splenic granulocytes in control pups. At PND35, splenic proportions of B and Th but not Tc cells are restored. CONCLUSION Postnatal administration of a single bacterial strain efficiently restores granulopoiesis and most T cell activation in neonatal mice that suffer from a perinatal antibiotic-induced compromised gut microbiota at birth.
Collapse
Affiliation(s)
- Eva Fuglsang
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Lukasz Krych
- Department of Food Science, University of Copenhagen, Frederiksberg, Denmark
| | - Mia Thorup Lundsager
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | - Hanne Frøkiaer
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
91
|
Hajavi J, Esmaeili SA, Varasteh AR, Vazini H, Atabati H, Mardani F, Momtazi-Borojeni AA, Hashemi M, Sankian M, Sahebkar A. The immunomodulatory role of probiotics in allergy therapy. J Cell Physiol 2018; 234:2386-2398. [PMID: 30192002 DOI: 10.1002/jcp.27263] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 07/24/2018] [Indexed: 12/14/2022]
Abstract
The increased incidence of allergic disorders may be the result of a relative fall in microbial induction in the intestinal immune system during infancy and early childhood. Probiotics have recently been proposed as viable microorganisms for the prevention and treatment of specific allergic diseases. Different mechanisms have been considered for this probiotic property, such as generation of cytokines from activated pro-T-helper type 1 after bacterial contact. However, the effects of its immunomodulatory potential require validation for clinical applications. This review will focus on the currently available data on the benefits of probiotics in allergy disease.
Collapse
Affiliation(s)
- Jafar Hajavi
- Department of Basic Sciences, Faculty of Allied Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Research Center, BuAli Research Institute, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abdol-Reza Varasteh
- Allergy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Vazini
- Nursing Department, Basic Sciences Faculty, Hamedan Branch, Islamic Azad University, Hamedan, Iran
| | - Hadi Atabati
- Leishmaniasis Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Fatemeh Mardani
- Immunology Research Center, BuAli Research Institute, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir A Momtazi-Borojeni
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology, Nanotechnology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Hashemi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojtaba Sankian
- Immunology Research Center, BuAli Research Institute, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
92
|
Kim JY, Kim EM, Yi MH, Lee J, Lee S, Hwang Y, Yong D, Sohn WM, Yong TS. Intestinal fluke Metagonimus yokogawai infection increases probiotic Lactobacillus in mouse cecum. Exp Parasitol 2018; 193:45-50. [PMID: 30149004 DOI: 10.1016/j.exppara.2018.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 07/23/2018] [Accepted: 08/23/2018] [Indexed: 02/03/2023]
Abstract
Helminth infection can alleviate immune-mediated disorders such as allergies and autoimmune diseases, by altering the gut microbiome. However, changes in gut microbiome due to intestinal trematodes remain unelucidated. Here, we evaluated the changes in the gut microbiome of ICR mice infected with Metagonimus yokogawai, a hypo-virulent intestinal trematode. Four weeks after infection, mouse cecal content was analyzed by 16S rRNA amplicon analysis. Although there was no apparent difference in species richness and diversity, the microbiome composition was different in the infected and control groups. Furthermore, several Lactobacillus species with known immunomodulatory role in immune-mediated diseases were increased in the infected group.
Collapse
Affiliation(s)
- Ju Yeong Kim
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Seoul, 03722, South Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| | - Eun-Min Kim
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| | - Myung-Hee Yi
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| | - Jinyoung Lee
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| | - Seogwon Lee
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| | - Younjee Hwang
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea; Department of Laboratory Medicine and Research Institute of Bacterial Resistance, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| | - Dongeun Yong
- Department of Laboratory Medicine and Research Institute of Bacterial Resistance, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| | - Woon-Mok Sohn
- Department of Parasitology and Tropical Medicine, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, 52727, South Korea.
| | - Tai-Soon Yong
- Department of Environmental Medical Biology, Institute of Tropical Medicine, Arthropods of Medical Importance Resource Bank, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| |
Collapse
|
93
|
Yasushi Y, Haraguchi Y, Shikano A, Kuda T, Takahashi H, Kimura B. Induction of gut Lactobacillus reuteri
in normal ICR mice by oral administration of L. plantarum
AN1. J Food Biochem 2018. [DOI: 10.1111/jfbc.12589] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Yokota Yasushi
- Department of Food Science and Technology; Tokyo University of Marine Science and Technology; Tokyo Japan
| | - Yutaka Haraguchi
- Department of Food Science and Technology; Tokyo University of Marine Science and Technology; Tokyo Japan
| | - Ayane Shikano
- Department of Food Science and Technology; Tokyo University of Marine Science and Technology; Tokyo Japan
| | - Takashi Kuda
- Department of Food Science and Technology; Tokyo University of Marine Science and Technology; Tokyo Japan
| | - Hajime Takahashi
- Department of Food Science and Technology; Tokyo University of Marine Science and Technology; Tokyo Japan
| | - Bon Kimura
- Department of Food Science and Technology; Tokyo University of Marine Science and Technology; Tokyo Japan
| |
Collapse
|
94
|
Microbial Insights into Asthmatic Immunopathology. A Forward-Looking Synthesis and Commentary. Ann Am Thorac Soc 2018; 14:S316-S325. [PMID: 29161080 DOI: 10.1513/annalsats.201707-534aw] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Asthma is an aberrant inflammatory condition of the airways affecting approximately 1 in 10 children in affluent countries. An increasing body of evidence suggests that microbial exposures during a "critical window" of development in early life play a central role in determining future asthma susceptibility. However, like the disease itself, considerable heterogeneity exists among studies in which researchers have investigated the associations between particular microbial taxa and asthma immunology. As our understanding of asthmatic pathology evolves to enable clearer definition of asthma endotypes, it will be important to consider the impact of various environmental factors on each endotype. Given the strong evidence in support of the hypothesis that early-life microbial exposures predict later disease states such as asthma, consideration of these endotypes when establishing experimental outcomes in epidemiological studies could allow for increased precision when determining exposure-outcome associations and engaging in more focused follow-up mechanistic investigations.
Collapse
|
95
|
Spacova I, Ceuppens JL, Seys SF, Petrova MI, Lebeer S. Probiotics against airway allergy: host factors to consider. Dis Model Mech 2018; 11:11/7/dmm034314. [PMID: 30037806 PMCID: PMC6078401 DOI: 10.1242/dmm.034314] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The worldwide prevalence of allergic diseases has drastically increased in the past decades. Recent studies underline the importance of microbial exposure for the development of a balanced immune system. Consequently, probiotic bacteria are emerging as a safe and natural strategy for allergy prevention and treatment. However, clinical probiotic intervention studies have so far yielded conflicting results. There is increasing awareness about the importance of host-associated factors that determine whether an individual will respond to a specific probiotic treatment, and it is therefore crucial to promote a knowledge-based instead of an empirical selection of promising probiotic strains and their administration regimen.In this Review, we summarize the insights from animal model studies of allergic disease, which reveal how host-related factors - such as genetic makeup, sex, age and microbiological status - can impact the outcomes of preventive or curative probiotic treatment. We explore why and how these factors can influence the results of probiotic studies and negatively impact the reproducibility in animal experiments. These same factors might profoundly influence the outcomes of human clinical trials, and can potentially explain the conflicting results from probiotic intervention studies. Therefore, we also link these host-related factors to human probiotic study outcomes in the context of airway allergies.
Collapse
Affiliation(s)
- Irina Spacova
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, 2020 Antwerp, Belgium.,Centre of Microbial and Plant Genetics, Department of Microbial and Molecular Systems (M²S), KU Leuven, Belgium
| | - Jan L Ceuppens
- Laboratory of Clinical Immunology, Department of Microbiology and Immunology, KU Leuven, 3000 Leuven, Belgium
| | - Sven F Seys
- Laboratory of Clinical Immunology, Department of Microbiology and Immunology, KU Leuven, 3000 Leuven, Belgium
| | - Mariya I Petrova
- Centre of Microbial and Plant Genetics, Department of Microbial and Molecular Systems (M²S), KU Leuven, Belgium
| | - Sarah Lebeer
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, 2020 Antwerp, Belgium .,Centre of Microbial and Plant Genetics, Department of Microbial and Molecular Systems (M²S), KU Leuven, Belgium
| |
Collapse
|
96
|
Zahirović A, Lunder M. Microbial Delivery Vehicles for Allergens and Allergen-Derived Peptides in Immunotherapy of Allergic Diseases. Front Microbiol 2018; 9:1449. [PMID: 30013543 PMCID: PMC6036130 DOI: 10.3389/fmicb.2018.01449] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/11/2018] [Indexed: 12/13/2022] Open
Abstract
Allergen-specific immunotherapy represents the only available curative approach to allergic diseases. The treatment has proven effective, but it requires repetitive administrations of allergen extracts over 3-5 years and is often associated with adverse events. This implies the need for novel therapeutic strategies with reduced side effects and decreased treatment time, which would improve patients' compliance. Development of vaccines that are molecularly well defined and have improved safety profile in comparison to whole allergen extracts represents a promising approach. Molecular allergy vaccines are based on major allergen proteins or allergen-derived peptides. Often, such vaccines are associated with lower immunogenicity and stability and therefore require an appropriate delivery vehicle. In this respect, viruses, bacteria, and their protein components have been intensively studied for their adjuvant capacity. This article provides an overview of the microbial delivery vehicles that have been tested for use in allergy immunotherapy. We review in vitro and in vivo data on the immunomodulatory capacity of different microbial vehicles for allergens and allergen-derived peptides and evaluate their potential in development of allergy vaccines. We also discuss relevant aspects and challenges concerning the use of microbes and their components in immunotherapy of allergic diseases.
Collapse
Affiliation(s)
- Abida Zahirović
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Mojca Lunder
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
97
|
Luo X, Pan Z, Luo S, Liu Q, Huang S, Yang G, Nong F, Fu Y, Deng X, Zhou L. Effects of ceftriaxone-induced intestinal dysbacteriosis on regulatory T cells validated by anaphylactic mice. Int Immunopharmacol 2018; 60:221-227. [PMID: 29772494 DOI: 10.1016/j.intimp.2018.05.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 05/07/2018] [Accepted: 05/08/2018] [Indexed: 12/27/2022]
Abstract
Both probiotics and pathogens in the human gut express pathogen-associated molecular patterns (PAMPs) and die with the release of endotoxin and bacterial DNA, which can stimulate our immune system and cause immune reaction. However, it's interesting and fascinating to address why the normal intestinal flora will not generate immunological rejection like the pathogen does. By investigating the changes in cells and molecules relevant to immune tolerance in mice with ceftriaxone-induced dysbacteriosis, our study discovered that both the Evenness indexes and Shannon Wiener index of intestinal flora showed a decrease in dysbacteriosis mice. Moreover, the proportion of αβ+TCR+CD3+CD4-CD8- cells, CD3+γδTCR+ cells and CD4+CD25+FoxP3+ cells in the Peyer's patches (PPs), mesenteric lymph nodes (MLNs) and spleen (SP) and the level of TGF-β1, IL-2, IL-4 and IL-10 in the serum also changed. Intestinal dysbacteriosis in an asthma murine model resulted in enhancement of immunologic response to the allergen ovalbumin (OVA), which was an agent that aggravates asthma symptoms. In summary, it is integral to maintain a certain amount or variety of intestinal microflora for regulatory T cells to act in averting hypersensitivity.
Collapse
Affiliation(s)
- Xia Luo
- Institute: School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, China
| | - Zengfeng Pan
- Institute: School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, China
| | - Shuang Luo
- Institute: School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, China
| | - Qi Liu
- Institute: School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, China
| | - Shaowei Huang
- Institute: School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, China
| | - Guanghua Yang
- Institute: School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, China
| | - Feifei Nong
- Institute: School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, China
| | - Yajun Fu
- Institute: School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, China
| | - Xiangliang Deng
- Infinitus Chinese Herbal Immunity Research Centre, Guangzhou, China
| | - Lian Zhou
- Institute: School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, China.
| |
Collapse
|
98
|
McVey Neufeld KA, Kay S, Bienenstock J. Mouse Strain Affects Behavioral and Neuroendocrine Stress Responses Following Administration of Probiotic Lactobacillus rhamnosus JB-1 or Traditional Antidepressant Fluoxetine. Front Neurosci 2018; 12:294. [PMID: 29867313 PMCID: PMC5952003 DOI: 10.3389/fnins.2018.00294] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/16/2018] [Indexed: 01/01/2023] Open
Abstract
Currently, there is keen interest in the development of alternative therapies in the treatment of depression. Given the explosion of research focused on the microbiota-gut-brain axis, consideration has turned to the potential of certain probiotics to improve patient outcomes for those suffering from mood disorders. Here we examine the abilities of a known antidepressant, fluoxetine, and the probiotic Lactobacillus rhamnosus JB-1™, to attenuate responses to two established criteria for depressive-like behavior in animal models, the tail suspension test (TST) and the corticosterone response to an acute restraint stressor. We examine two different strains of mice known to differ in the extent to which they express both anxiety-like behavior and measures of despair—BALB/c and Swiss Webster—with respectively high and normal behavioral phenotypes for each. While adult male BALB/c mice responded with increased antidepressive-like behavior to both fluoxetine and L. rhamnosus JB-1 in both the TST and the corticosterone stress response, SW mice did not respond to either treatment as compared to controls. These findings highlight the importance of investigating putative antidepressants in mouse strains known to express face validity for some markers of depression. Clinical studies examining the activity of L. rhamnosus JB-1 in patients suffering from mood disorders are warranted, as well as further pre-clinical work examining how interactions between host genotype and intestinal microbial alterations may impact behavioral responses. This study adds to the literature supporting the possibility that modifying the intestinal microbiota via probiotics represents a promising potential therapeutic breakthrough in the treatment of psychiatric disease.
Collapse
Affiliation(s)
- Karen-Anne McVey Neufeld
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada.,Brain-Body Institute, St. Joseph's Healthcare at McMaster University, Hamilton, ON, Canada
| | - Sebastian Kay
- Brain-Body Institute, St. Joseph's Healthcare at McMaster University, Hamilton, ON, Canada
| | - John Bienenstock
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada.,Brain-Body Institute, St. Joseph's Healthcare at McMaster University, Hamilton, ON, Canada
| |
Collapse
|
99
|
Vestergaard DV, Holst GJ, Basinas I, Elholm G, Schlünssen V, Linneberg A, Šantl-Temkiv T, Finster K, Sigsgaard T, Marshall IPG. Pig Farmers' Homes Harbor More Diverse Airborne Bacterial Communities Than Pig Stables or Suburban Homes. Front Microbiol 2018; 9:870. [PMID: 29765370 PMCID: PMC5938556 DOI: 10.3389/fmicb.2018.00870] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 04/16/2018] [Indexed: 12/30/2022] Open
Abstract
Airborne bacterial communities are subject to conditions ill-suited to microbial activity and growth. In spite of this, air is an important transfer medium for bacteria, with the bacteria in indoor air having potentially major consequences for the health of a building’s occupants. A major example is the decreased diversity and altered composition of indoor airborne microbial communities as a proposed explanation for the increasing prevalence of asthma and allergies worldwide. Previous research has shown that living on a farm confers protection against development of asthma and allergies, with airborne bacteria suggested as playing a role in this protective effect. However, the composition of this beneficial microbial community has still not been identified. We sampled settled airborne dust using a passive dust sampler from Danish pig stables, associated farmers’ homes, and from suburban homes (267 samples in total) and carried out quantitative PCR measurements of bacterial abundance and MiSeq sequencing of the V3–V4 region of bacterial 16S rRNA genes found in these samples. Airborne bacteria had a greater diversity and were significantly more abundant in pig stables and farmers’ homes than suburban homes (Wilcoxon rank sum test P < 0.05). Moreover, bacterial taxa previously suggested to contribute to a protective effect had significantly higher relative and absolute abundance in pig stables and farmers’ homes than in suburban homes (ALDEx2 with P < 0.05), including Firmicutes, Peptostreptococcaceae, Prevotellaceae, Lachnospiraceae, Ruminococcaceae, Ruminiclostridium, and Lactobacillus. Pig stables had significantly lower airborne bacterial diversity than farmers’ homes, and there was no discernable direct transfer of airborne bacteria from stable to home. This study identifies differences in indoor airborne bacterial communities that may be an important component of this putative protective effect, while showing that pig stables themselves do not appear to directly contribute to the airborne bacterial communities in the homes of farmers. These findings improve our understanding of the role of airborne bacteria in the increasing prevalence of asthma and allergy.
Collapse
Affiliation(s)
- Ditte V Vestergaard
- Section for Environment, Occupation and Health, Department of Public Health, Aarhus University, Aarhus, Denmark.,Section for Microbiology, Department of Bioscience, Aarhus University, Aarhus, Denmark
| | - Gitte J Holst
- Section for Environment, Occupation and Health, Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Ioannis Basinas
- Centre for Human Exposure Science, Institute of Occupational Medicine, Edinburgh, United Kingdom
| | - Grethe Elholm
- Section for Environment, Occupation and Health, Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Vivi Schlünssen
- Section for Environment, Occupation and Health, Department of Public Health, Aarhus University, Aarhus, Denmark.,National Research Centre for the Working Environment, Copenhagen, Denmark
| | - Allan Linneberg
- Department of Clinical Experimental Research, Rigshospitalet, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Research Centre for Prevention and Health, Rigshospitalet Glostrup, Glostrup, Denmark
| | - Tina Šantl-Temkiv
- Section for Microbiology, Department of Bioscience, Aarhus University, Aarhus, Denmark
| | - Kai Finster
- Section for Microbiology, Department of Bioscience, Aarhus University, Aarhus, Denmark
| | - Torben Sigsgaard
- Section for Environment, Occupation and Health, Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Ian P G Marshall
- Section for Microbiology, Department of Bioscience, Aarhus University, Aarhus, Denmark
| |
Collapse
|
100
|
Mu Q, Tavella VJ, Luo XM. Role of Lactobacillus reuteri in Human Health and Diseases. Front Microbiol 2018; 9:757. [PMID: 29725324 PMCID: PMC5917019 DOI: 10.3389/fmicb.2018.00757] [Citation(s) in RCA: 393] [Impact Index Per Article: 65.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 04/04/2018] [Indexed: 12/13/2022] Open
Abstract
Lactobacillus reuteri (L. reuteri) is a well-studied probiotic bacterium that can colonize a large number of mammals. In humans, L. reuteri is found in different body sites, including the gastrointestinal tract, urinary tract, skin, and breast milk. The abundance of L. reuteri varies among different individuals. Several beneficial effects of L. reuteri have been noted. First, L. reuteri can produce antimicrobial molecules, such as organic acids, ethanol, and reuterin. Due to its antimicrobial activity, L. reuteri is able to inhibit the colonization of pathogenic microbes and remodel the commensal microbiota composition in the host. Second, L. reuteri can benefit the host immune system. For instance, some L. reuteri strains can reduce the production of pro-inflammatory cytokines while promoting regulatory T cell development and function. Third, bearing the ability to strengthen the intestinal barrier, the colonization of L. reuteri may decrease the microbial translocation from the gut lumen to the tissues. Microbial translocation across the intestinal epithelium has been hypothesized as an initiator of inflammation. Therefore, inflammatory diseases, including those located in the gut as well as in remote tissues, may be ameliorated by increasing the colonization of L. reuteri. Notably, the decrease in the abundance of L. reuteri in humans in the past decades is correlated with an increase in the incidences of inflammatory diseases over the same period of time. Direct supplementation or prebiotic modulation of L. reuteri may be an attractive preventive and/or therapeutic avenue against inflammatory diseases.
Collapse
Affiliation(s)
| | | | - Xin M. Luo
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|