151
|
Microbiome and Asthma: What Have Experimental Models Already Taught Us? J Immunol Res 2015; 2015:614758. [PMID: 26266269 PMCID: PMC4525458 DOI: 10.1155/2015/614758] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 07/02/2015] [Indexed: 02/07/2023] Open
Abstract
Asthma is a chronic inflammatory disease that imposes a substantial burden on patients, their families, and the community. Although many aspects of the pathogenesis of classical allergic asthma are well known by the scientific community, other points are not yet understood. Experimental asthma models, particularly murine models, have been used for over 100 years in order to better understand the immunopathology of asthma. It has been shown that human microbiome is an important component in the development of the immune system. Furthermore, the occurrence of many inflammatory diseases is influenced by the presence of microbes. Again, experimental models of asthma have helped researchers to understand the relationship between the microbiome and respiratory inflammation. In this review, we discuss the evolution of murine models of asthma and approach the major studies involving the microbiome and asthma.
Collapse
|
152
|
Probiotic fermented milk consumption modulates the allergic process induced by ovoalbumin in mice. Br J Nutr 2015; 114:566-76. [PMID: 26179751 DOI: 10.1017/s0007114515001981] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Orally administered probiotic micro-organisms are able to regulate the exacerbated immune response during the antigenic sensitisation process. The aim of the present study was to evaluate the potential efficacy of probiotic fermented milk (PFM) in preventing or treating allergy in an experimental model, and to investigate its underlying mechanisms. Ovoalbumin (OVA)-sensitised BALB/c mice were fed with PFM before the sensitisation procedure or fed continuously with PFM. At 7 and 15 d post-sensitisation, anti-OVA-specific IgE, IgG, IgG1 and IgG2a concentrations were measured in the serum and broncho-alveolar lavage fluid (BALF). Concentrations of interferon-γ (IFN-γ), IL-4, IL-10 and total secretory IgA (S-IgA) were measured in the supernatants of macerated lungs or in the BALF. The levels of IgA+, CD4+ and CD8+ T lymphocytes and F4/80+ cells were measured in the lungs by immunofluorescence. Inducible CD4+/CD25/Foxp3+ regulatory T (Treg) cells were evaluated in the lungs. PFM shifted the T helper (Th)2 profile response towards a Th1 response that led to the production of IgG instead of IgE, with increasing levels of IL-10 and IFN-γ that play an important role in immunomodulation exerted by PFM administration in sensitised mice. Anti-OVA-specific IgE levels were significantly decreased; however, there was no modification in the levels of anti-OVA-specific IgG and total S-IgA. PFM did not influence Treg cells in treated mice. Consumption of PFM could be a promising strategy in the amelioration of airway allergies, considering that the effect is mediated by the production of IgG through the activation of Th1 instead of the direct activation of Th2 cells to produce IgE.
Collapse
|
153
|
Kashiwagi I, Morita R, Schichita T, Komai K, Saeki K, Matsumoto M, Takeda K, Nomura M, Hayashi A, Kanai T, Yoshimura A. Smad2 and Smad3 Inversely Regulate TGF-β Autoinduction in Clostridium butyricum-Activated Dendritic Cells. Immunity 2015; 43:65-79. [PMID: 26141582 DOI: 10.1016/j.immuni.2015.06.010] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 03/03/2015] [Accepted: 06/07/2015] [Indexed: 12/13/2022]
Abstract
Colonization with a mixture of Clostridium species has been shown to induce accumulation of induced regulatory T (iTreg) cells in the colon. Transforming growth factor-β (TGF-β) is an essential factor for iTreg cell induction; however, the relationship between Clostridium species and TGF-β remains to be clarified. Here we demonstrated that a gram-positive probiotic bacterial strain, Clostridium butyricum (C. butyricum), promoted iTreg cell generation in the intestine through induction of TGF-β1 from lamina propria dendritic cells (LPDCs). C. butyricum-mediated TGF-β1 induction was mainly Toll-like receptor 2 (TLR2) dependent, and the ERK-AP-1 kinase pathway played an important role. In addition, the autocrine TGF-β-Smad3 transcription factor signal was necessary for robust TGF-β expression in DCs, whereas Smad2 negatively regulated TGF-β expression. Smad2-deficient DCs expressed higher concentrations of TGF-β and were tolerogenic for colitis models. This study reveals a novel mechanism of TGF-β induction by Clostridia through a cooperation between TLR2-AP-1 and TGF-β-Smad signaling pathways.
Collapse
Affiliation(s)
- Ikkou Kashiwagi
- Department of Microbiology and Immunology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan, and Japan Science and Technology Agency, CREST, Tokyo 102-0076, Japan
| | - Rimpei Morita
- Department of Microbiology and Immunology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan, and Japan Science and Technology Agency, CREST, Tokyo 102-0076, Japan
| | - Takashi Schichita
- Department of Microbiology and Immunology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan, and Japan Science and Technology Agency, CREST, Tokyo 102-0076, Japan; PRESTO (Precursory Research for Embryonic Science and Technology), Chiyoda-ku, Tokyo 102-0075, Japan
| | - Kyoko Komai
- Department of Microbiology and Immunology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan, and Japan Science and Technology Agency, CREST, Tokyo 102-0076, Japan
| | - Keita Saeki
- Department of Microbiology and Immunology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan, and Japan Science and Technology Agency, CREST, Tokyo 102-0076, Japan
| | - Makoto Matsumoto
- Department of Immunology and Medical Zoology, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, 663-8501, Japan
| | - Kiyoshi Takeda
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masatoshi Nomura
- Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Atsushi Hayashi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan, and Japan Science and Technology Agency, CREST, Tokyo 102-0076, Japan; Miyarisan Pharmaceutical Co., Ltd., Research Laboratory, Tokyo 114-0016, Japan
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan, and Japan Science and Technology Agency, CREST, Tokyo 102-0076, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan, and Japan Science and Technology Agency, CREST, Tokyo 102-0076, Japan.
| |
Collapse
|
154
|
Forsythe P. Microbes taming mast cells: Implications for allergic inflammation and beyond. Eur J Pharmacol 2015; 778:169-75. [PMID: 26130124 DOI: 10.1016/j.ejphar.2015.06.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 05/21/2015] [Accepted: 06/17/2015] [Indexed: 12/30/2022]
Abstract
There is increasing awareness of a relationship between our microbiota and the pathogenesis of allergy and other inflammatory diseases. In investigating the mechanisms underlying microbiota modulation of allergy the focus has been on the induction phase; alterations in the phenotype and function of antigen presenting cells, induction of regulatory T cells and shifts in Th1/Th2 balance. However there is evidence that microbes can influence the effector phase of disease, specifically that certain potentially beneficial bacteria can attenuate mast cell activation and degranulation. Furthermore, it appears that different non-pathogenic bacteria can utilize distinct mechanisms to stabilize mast cells, acting locally though direct interaction with the mast cell at mucosal sites or attenuating systemic mast cell dependent responses, likely through indirect signaling mechanisms. The position of mast cells on the frontline of defense against pathogens also suggests they may play an important role in fostering the host-microbiota relationship. Mast cells are also conduits of neuro-immuo-endocrine communication, suggesting the ability of microbes to modulate cell responses may have implications for host physiology beyond immunology. Further investigation of mast cell regulation by non-pathogenic or symbiotic bacteria will likely lead to a greater understanding of host microbiota interaction and the role of the microbiome in health and disease.
Collapse
Affiliation(s)
- Paul Forsythe
- McMaster Brain-Body Institute at St. Joseph's Healthcare, Hamilton, The Firestone Institute for Respiratory Health and Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
155
|
Ten years of subproteome investigations in lactic acid bacteria: A key for food starter and probiotic typing. J Proteomics 2015; 127:332-9. [PMID: 25957532 DOI: 10.1016/j.jprot.2015.04.028] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 04/22/2015] [Accepted: 04/26/2015] [Indexed: 12/29/2022]
Abstract
The definition of safety and efficacy of food-employed bacteria as well as probiotic strains is a continuous, often unattended, challenge. Proteomic techniques such as 2DE, DIGE and LC/LC-MS/MS are suitable and powerful tools to reveal new aspects (positive and negative) of "known" and "unknown" strains that can be employed in food making and as nutraceutical supplements for human health. Unfortunately, these techniques are not used as extensively as it should be wise. The present report describes the most significant results obtained by our research group in 10years of study on subproteomes in bacteria, chiefly lactic acid bacteria. Production of desired and undesired metabolites, differences between strains belonging to same species but isolated from different ecological niches, the effect of cryoprotectants on survival to lyophilization as well as the adhesive capability of strains, were elucidated by analysis of cytosolic, membrane-enriched, surface and extracellular proteomes. The present review opens a window on a yet largely underexplored field and highlights the huge potential of subproteome investigations for more rational choice of microbial strains as food starters, probiotics and for production of nutraceuticals. These analyses will hopefully contribute to manufacturing safer and healthier food and food supplements in the near future. This article is part of a Special Issue entitled: HUPO 2014.
Collapse
|
156
|
Holm JB, Sorobetea D, Kiilerich P, Ramayo-Caldas Y, Estellé J, Ma T, Madsen L, Kristiansen K, Svensson-Frej M. Chronic Trichuris muris Infection Decreases Diversity of the Intestinal Microbiota and Concomitantly Increases the Abundance of Lactobacilli. PLoS One 2015; 10:e0125495. [PMID: 25942314 PMCID: PMC4420551 DOI: 10.1371/journal.pone.0125495] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 03/13/2015] [Indexed: 12/26/2022] Open
Abstract
The intestinal microbiota is vital for shaping the local intestinal environment as well as host immunity and metabolism. At the same time, epidemiological and experimental evidence suggest an important role for parasitic worm infections in maintaining the inflammatory and regulatory balance of the immune system. In line with this, the prevalence of persistent worm infections is inversely correlated with the incidence of immune-associated diseases, prompting the use of controlled parasite infections for therapeutic purposes. Despite this, the impact of parasite infection on the intestinal microbiota, as well as potential downstream effects on the immune system, remain largely unknown. We have assessed the influence of chronic infection with the large-intestinal nematode Trichuris muris, a close relative of the human pathogen Trichuris trichiura, on the composition of the murine intestinal microbiota by 16S ribosomal-RNA gene-based sequencing. Our results demonstrate that persistent T. muris infection dramatically affects the large-intestinal microbiota, most notably with a drop in the diversity of bacterial communities, as well as a marked increase in the relative abundance of the Lactobacillus genus. In parallel, chronic T. muris infection resulted in a significant shift in the balance between regulatory and inflammatory T cells in the intestinal adaptive immune system, in favour of inflammatory cells. Together, these data demonstrate that chronic parasite infection strongly influences the intestinal microbiota and the adaptive immune system. Our results illustrate the complex interactions between these factors in the intestinal tract, and contribute to furthering the understanding of this interplay, which is of crucial importance considering that 500 million people globally are suffering from these infections and their potential use for therapeutic purposes.
Collapse
Affiliation(s)
- Jacob Bak Holm
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Daniel Sorobetea
- Immunology Section, Department of Experimental Medical Sciences, Medical Faculty, Lund University, Lund, Sweden
| | - Pia Kiilerich
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | | | - Jordi Estellé
- INRA, UMR1313 Génétique Animale et Biologie Intégrative (GABI), Jouy-en-Josas, France
| | - Tao Ma
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Lise Madsen
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark; National Institute of Nutrition and Seafood Research, Bergen, Norway
| | - Karsten Kristiansen
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Marcus Svensson-Frej
- Immunology Section, Department of Experimental Medical Sciences, Medical Faculty, Lund University, Lund, Sweden
| |
Collapse
|
157
|
Kearney SC, Dziekiewicz M, Feleszko W. Immunoregulatory and immunostimulatory responses of bacterial lysates in respiratory infections and asthma. Ann Allergy Asthma Immunol 2015; 114:364-9. [DOI: 10.1016/j.anai.2015.02.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 01/30/2015] [Accepted: 02/09/2015] [Indexed: 01/22/2023]
|
158
|
Abstract
Genetic and environmental studies implicate immune pathologies in schizophrenia. The body's largest immune organ is the gastrointestinal (GI) tract. Historical associations of GI conditions with mental illnesses predate the introduction of antipsychotics. Current studies of antipsychotic-naïve patients support that gut dysfunction may be inherent to the schizophrenia disease process. Risk factors for schizophrenia (inflammation, food intolerances, Toxoplasma gondii exposure, cellular barrier defects) are part of biological pathways that intersect those operant in the gut. Central to GI function is a homeostatic microbial community, and early reports show that it is disrupted in schizophrenia. Bioactive and toxic products derived from digestion and microbial dysbiosis activate adaptive and innate immunity. Complement C1q, a brain-active systemic immune component, interacts with gut-related schizophrenia risk factors in clinical and experimental animal models. With accumulating evidence supporting newly discovered gut-brain physiological pathways, treatments to ameliorate brain symptoms of schizophrenia should be supplemented with therapies to correct GI dysfunction.
Collapse
Affiliation(s)
- Emily G Severance
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Blalock 1105, Baltimore, MD, 21287-4933, USA,
| | | | | | | |
Collapse
|
159
|
Ai C, Zhang Q, Ren C, Wang G, Liu X, Tian F, Zhao J, Zhang H, Chen YQ, Chen W. Protective effect of Streptococcus thermophilus CCFM218 against house dust mite allergy in a mouse model. Food Control 2015. [DOI: 10.1016/j.foodcont.2014.08.040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
160
|
Konieczna P, Schiavi E, Ziegler M, Groeger D, Healy S, Grant R, O’Mahony L. Human dendritic cell DC-SIGN and TLR-2 mediate complementary immune regulatory activities in response to Lactobacillus rhamnosus JB-1. PLoS One 2015; 10:e0120261. [PMID: 25816321 PMCID: PMC4376398 DOI: 10.1371/journal.pone.0120261] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 01/21/2015] [Indexed: 02/07/2023] Open
Abstract
The microbiota is required for optimal host development and ongoing immune homeostasis. Lactobacilli are common inhabitants of the mammalian large intestine and immunoregulatory effects have been described for certain, but not all, strains. The mechanisms underpinning these protective effects are beginning to be elucidated. One such protective organism is Lactobacillus rhamnosus JB-1 (Lb. rhamnosus JB-1). Lb. murinus has no such anti-inflammatory protective effects and was used as a comparator organism. Human monocyte-derived dendritic cells (MDDCs) were co-incubated with bacteria and analysed over time for bacterial adhesion and intracellular processing, costimulatory molecule expression, cytokine secretion and induction of lymphocyte polarization. Neutralising antibodies were utilized to identify the responsible MDDC receptors. Lb. rhamnosus JB-1 adhered to MDDCs, but internalization and intracellular processing was significantly delayed, compared to Lb. murinus which was rapidly internalized and processed. Lb. murinus induced CD80 and CD86 expression, accompanied by high levels of cytokine secretion, while Lb. rhamnosus JB-1 was a poor inducer of costimulatory molecule expression and cytokine secretion. Lb. rhamnosus JB-1 primed MDDCs induced Foxp3 expression in autologous lymphocytes, while Lb. murinus primed MDDCs induced Foxp3, T-bet and Ror-γt expression. DC-SIGN was required for Lb. rhamnosus JB-1 adhesion and influenced IL-12 secretion, while TLR-2 influenced IL-10 and IL-12 secretion. Here we demonstrate that the delayed kinetics of bacterial processing by MDDCs correlates with MDDC activation and stimulation of lymphocytes. Thus, inhibition or delay of intracellular processing may be a novel strategy by which certain commensals may avoid the induction of proinflammatory responses.
Collapse
Affiliation(s)
- Patrycja Konieczna
- Swiss Institute of Allergy and Asthma Research, University of Zurich, Davos, Switzerland
| | - Elisa Schiavi
- Swiss Institute of Allergy and Asthma Research, University of Zurich, Davos, Switzerland
| | - Mario Ziegler
- Swiss Institute of Allergy and Asthma Research, University of Zurich, Davos, Switzerland
| | | | | | - Ray Grant
- Alimentary Health Pharma Davos, Davos, Switzerland
| | - Liam O’Mahony
- Swiss Institute of Allergy and Asthma Research, University of Zurich, Davos, Switzerland
- * E-mail:
| |
Collapse
|
161
|
The microbiome in asthma. J Allergy Clin Immunol 2015; 135:25-30. [PMID: 25567040 DOI: 10.1016/j.jaci.2014.11.011] [Citation(s) in RCA: 187] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 10/31/2014] [Accepted: 11/04/2014] [Indexed: 12/27/2022]
Abstract
The application of recently developed sensitive, specific, culture-independent tools for identification of microbes is transforming concepts of microbial ecology, including concepts of the relationships between the vast complex populations of microbes associated with ourselves and with states of health and disease. Although most work initially focused on the community of microbes (microbiome) in the gastrointestinal tract and its relationship to gastrointestinal disease, interest has expanded to include study of the relationships of the airway microbiome to asthma and its phenotypes and to the relationships between the gastrointestinal microbiome, development of immune function, and predisposition to allergic sensitization and asthma. Here we provide our perspective on the findings of studies of differences in the airway microbiome between asthmatic patients and healthy subjects and of studies of relationships between environmental microbiota, gut microbiota, immune function, and asthma development. In addition, we provide our perspective on how these findings suggest the broad outline of a rationale for approaches involving directed manipulation of the gut and airway microbiome for the treatment and prevention of allergic asthma.
Collapse
|
162
|
Ai C, Zhang Q, Ding J, Ren C, Wang G, Liu X, Tian F, Zhao J, Zhang H, Chen YQ, Chen W. Suppression of dust mite allergy by mucosal delivery of a hypoallergenic derivative in a mouse model. Appl Microbiol Biotechnol 2015; 99:4309-19. [PMID: 25661808 DOI: 10.1007/s00253-015-6407-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 01/09/2015] [Accepted: 01/13/2015] [Indexed: 10/24/2022]
Abstract
Allergic asthma caused by house dust mite (HDM) is becoming a public health problem. Specific immunotherapy is considered to be the only curative treatment, but it is always associated with IgE-mediated side effects in the therapy process. A few studies showed that the disruption of allergen IgE epitopes could reduce IgE reactivity and thus reduce allergenic activity. In this study, a hypoallergenic derivative of the major HDM allergen Der p2 was constructed by genetic engineering. This derivative was confirmed to have a considerably reduced IgE reactivity compared with Der p2. For its application in vivo, recombinant Lactococcus lactis (LL-DM) was engineered to deliver the Der p2 derivative to the intestinal mucosal surface. Oral administration of LL-DM significantly alleviated Der p2-induced airway inflammation, as shown by reduced inflammatory infiltration and a reduction in Th2 cytokines in bronchoalveolar lavage. This protective effect was associated with an up-regulation of specific IgG2a and a decrease in IL-4 level in the spleen which may affect specific IgE response. Moreover, the levels of regulatory T cells in the mesenteric lymph nodes and spleen were markedly increased in mice fed with LL-DM, suggesting that LL-DM can inhibit allergic responses via the induction of regulatory T cell. Our results indicate that the Der p2 derivative is a promising therapeutic molecule for specific immunotherapy and recombinant lactic acid bacteria could be developed as a promising treatment or prevention strategy for allergic diseases.
Collapse
Affiliation(s)
- Chunqing Ai
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
163
|
Sze MA, Utokaparch S, Elliott WM, Hogg JC, Hegele RG. Loss of GD1-positive Lactobacillus correlates with inflammation in human lungs with COPD. BMJ Open 2015; 5:e006677. [PMID: 25652802 PMCID: PMC4322209 DOI: 10.1136/bmjopen-2014-006677] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVES The present study assesses the relationship between contents of GD1 (glycerol dehydratase)-positive Lactobacillus, presence of Lactobacillus and the inflammatory response measured in host lung tissue in mild to moderate chronic obstructive pulmonary disease (COPD). We hypothesise that there will be a loss of GD1 producing Lactobacillus with increasing severity of COPD and that GD1 has anti-inflammatory properties. SETTING Secondary care, 1 participating centre in Vancouver, British Columbia, Canada. PARTICIPANTS 74 individuals who donated non-cancerous portions of their lungs or lobes removed as treatment for lung cancer (normal lung function controls (n=28), persons with mild (GOLD 1) (n=21) and moderate (GOLD 2) COPD (n=25)). OUTCOME MEASURES Primary outcome measure was GD1 positivity within each group and whether or not this impacted quantitative histological measures of lung inflammation. Secondary outcome measures included Lactobacillus presence and quantification, and quantitative histological measurements of inflammation and remodelling in early COPD. RESULTS Total bacterial count (p>0.05) and prevalence of Lactobacillus (p>0.05) did not differ between groups. However, the GD1 gene was detected more frequently in the controls (14%) than in either mild (5%) or moderate (0%) COPD (p<0.05) samples. Macrophage and neutrophil volume fractions (0.012±0.005 (mean±SD) vs 0.026±0.017 and 0.005±0.002 vs 0.015±0.014, respectively) in peripheral lung tissue were reduced in samples positive for the GD1 gene (p<0.0035). CONCLUSIONS A reduction in GD1 positivity is associated with an increased tissue immune inflammatory response in early stage COPD. There is potential for Lactobacillus to be used as a possible therapeutic, however, validation of these results need to be completed before an anti-inflammatory role of Lactobacillus in COPD can be confirmed.
Collapse
Affiliation(s)
- Marc A Sze
- Departments of Medicine and Pathology and Laboratory Medicine, Centre for Heart Lung Innovation, Providence Heart-Lung Institute at St Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Soraya Utokaparch
- Departments of Medicine and Pathology and Laboratory Medicine, Centre for Heart Lung Innovation, Providence Heart-Lung Institute at St Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - W Mark Elliott
- Departments of Medicine and Pathology and Laboratory Medicine, Centre for Heart Lung Innovation, Providence Heart-Lung Institute at St Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - James C Hogg
- Departments of Medicine and Pathology and Laboratory Medicine, Centre for Heart Lung Innovation, Providence Heart-Lung Institute at St Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Richard G Hegele
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada
| |
Collapse
|
164
|
Krych Ł, Nielsen DS, Hansen AK, Hansen CHF. Gut microbial markers are associated with diabetes onset, regulatory imbalance, and IFN-γ level in NOD mice. Gut Microbes 2015; 6:101-9. [PMID: 25648687 PMCID: PMC4615729 DOI: 10.1080/19490976.2015.1011876] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Gut microbiota regulated imbalances in the host's immune profile seem to be an important factor in the etiology of type 1 diabetes (T1D), and identifying bacterial markers for T1D may therefore be useful in diagnosis and prevention of T1D. The aim of the present study was to investigate the link between the early gut microbiota and immune parameters of non-obese diabetic (NOD) mice in order to select alleged bacterial markers of T1D. Gut microbial composition in feces was analyzed with 454/FLX Titanium (Roche) pyro-sequencing and correlated with diabetes onset age and immune cell populations measured in diabetic and non-diabetic mice at 30 weeks of age. The early gut microbiota composition was found to be different between NOD mice that later in life were classified as diabetic or non-diabetic. Those differences were further associated with changes in FoxP3(+) regulatory T cells, CD11b(+) dendritic cells, and IFN-γ production. The model proposed in this work suggests that operational taxonomic units classified to S24-7, Prevotella, and an unknown Bacteriodales (all Bacteroidetes) act in favor of diabetes protection whereas members of Lachnospiraceae, Ruminococcus, and Oscillospira (all Firmicutes) promote pathogenesis.
Collapse
Key Words
- CD, cluster of differentiation
- DC, dendritic cell
- FoxP3, forkhead box
- IFN, interferon
- IFN-γ
- MLN, mesenteric lymph node
- NKT, natural killer T cell
- NOD mice
- PCA, principal component analysis
- PCoA, principal coordinate analysis
- PLN, pancreatic lymph node
- Treg, regulatory T cell
- Type 1 diabetes
- gut microbiota
- regulatory immunity
- siLP, small intestinal lamina propria
Collapse
Affiliation(s)
- Ł Krych
- Department of Food Science; Faculty of Science; University of Copenhagen; Copenhagen, Denmark,Correspondence to: Ł Krych;
| | - DS Nielsen
- Department of Food Science; Faculty of Science; University of Copenhagen; Copenhagen, Denmark
| | - AK Hansen
- Department of Veterinary Disease Biology; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen, Denmark
| | - CHF Hansen
- Department of Veterinary Disease Biology; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen, Denmark
| |
Collapse
|
165
|
Affiliation(s)
- Takeshi Tanoue
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Kenya Honda
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan
- RIKEN Center for Integrative Medical Sciences (IMS-RCAI), Yokohama, Kanagawa, Japan
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
166
|
|
167
|
Gnotobiology and the Study of Complex Interactions between the Intestinal Microbiota, Probiotics, and the Host. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00008-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
168
|
Rook GAW, Raison CL, Lowry CA. Microbiota, immunoregulatory old friends and psychiatric disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 817:319-56. [PMID: 24997041 DOI: 10.1007/978-1-4939-0897-4_15] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Regulation of the immune system is an important function of the gut microbiota. Increasing evidence suggests that modern living conditions cause the gut microbiota to deviate from the form it took during human evolution. Contributing factors include loss of helminth infections, encountering less microbial biodiversity, and modulation of the microbiota composition by diet and antibiotic use. Thus the gut microbiota is a major mediator of the hygiene hypothesis (or as we prefer, "Old Friends" mechanism), which describes the role of organisms with which we co-evolved, and that needed to be tolerated, as crucial inducers of immunoregulation. At least partly as a consequence of reduced exposure to immunoregulatory Old Friends, many but not all of which resided in the gut, high-income countries are undergoing large increases in a wide range of chronic inflammatory disorders including allergies, autoimmunity and inflammatory bowel diseases. Depression, anxiety and reduced stress resilience are comorbid with these conditions, or can occur in individuals with persistently raised circulating levels of biomarkers of inflammation in the absence of clinically apparent peripheral inflammatory disease. Moreover poorly regulated inflammation during pregnancy might contribute to brain developmental abnormalities that underlie some cases of autism spectrum disorders and schizophrenia. In this chapter we explain how the gut microbiota drives immunoregulation, how faulty immunoregulation and inflammation predispose to psychiatric disease, and how psychological stress drives further inflammation via pathways that involve the gut and microbiota. We also outline how this two-way relationship between the brain and inflammation implicates the microbiota, Old Friends and immunoregulation in the control of stress resilience.
Collapse
Affiliation(s)
- Graham A W Rook
- Centre for Clinical Microbiology, UCL (University College London), Royal Free Campus, Rowland Hill Street, London, NW3 2PF, UK,
| | | | | |
Collapse
|
169
|
Al-Nedawi K, Mian MF, Hossain N, Karimi K, Mao YK, Forsythe P, Min KK, Stanisz AM, Kunze WA, Bienenstock J. Gut commensal microvesicles reproduce parent bacterial signals to host immune and enteric nervous systems. FASEB J 2014; 29:684-95. [PMID: 25392266 DOI: 10.1096/fj.14-259721] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Ingestion of a commensal bacteria, Lactobacillus rhamnosus JB-1, has potent immunoregulatory effects, and changes nerve-dependent colon migrating motor complexes (MMCs), enteric nerve function, and behavior. How these alterations occur is unknown. JB-1 microvesicles (MVs) are enriched for heat shock protein components such as chaperonin 60 heat-shock protein isolated from Escherichia coli (GroEL) and reproduce regulatory and neuronal effects in vitro and in vivo. Ingested labeled MVs were detected in murine Peyer's patch (PP) dendritic cells (DCs) within 18 h. After 3 d, PP and mesenteric lymph node DCs assumed a regulatory phenotype and increased functional regulatory CD4(+)25(+)Foxp3+ T cells. JB-1, MVs, and GroEL similarly induced phenotypic change in cocultured DCs via multiple pathways including C-type lectin receptors specific intercellular adhesion molecule-3 grabbing non-integrin-related 1 and Dectin-1, as well as TLR-2 and -9. JB-1 and MVs also decreased the amplitude of neuronally dependent MMCs in an ex vivo model of peristalsis. Gut epithelial, but not direct neuronal application of, MVs, replicated functional effects of JB-1 on in situ patch-clamped enteric neurons. GroEL and anti-TLR-2 were without effect in this system, suggesting the importance of epithelium neuron signaling and discrimination between pathways for bacteria-neuron and -immune communication. Together these results offer a mechanistic explanation of how Gram-positive commensals and probiotics may influence the host's immune and nervous systems.
Collapse
Affiliation(s)
- Khalid Al-Nedawi
- *Division of Nephrology, Departments of Medicine, Psychiatry and Behavioral Neurosciences, and Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada; and McMaster Brain-Body Institute at St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - M Firoz Mian
- *Division of Nephrology, Departments of Medicine, Psychiatry and Behavioral Neurosciences, and Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada; and McMaster Brain-Body Institute at St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Nazia Hossain
- *Division of Nephrology, Departments of Medicine, Psychiatry and Behavioral Neurosciences, and Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada; and McMaster Brain-Body Institute at St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Khalil Karimi
- *Division of Nephrology, Departments of Medicine, Psychiatry and Behavioral Neurosciences, and Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada; and McMaster Brain-Body Institute at St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Yu-Kang Mao
- *Division of Nephrology, Departments of Medicine, Psychiatry and Behavioral Neurosciences, and Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada; and McMaster Brain-Body Institute at St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Paul Forsythe
- *Division of Nephrology, Departments of Medicine, Psychiatry and Behavioral Neurosciences, and Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada; and McMaster Brain-Body Institute at St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Kevin K Min
- *Division of Nephrology, Departments of Medicine, Psychiatry and Behavioral Neurosciences, and Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada; and McMaster Brain-Body Institute at St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Andrew M Stanisz
- *Division of Nephrology, Departments of Medicine, Psychiatry and Behavioral Neurosciences, and Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada; and McMaster Brain-Body Institute at St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Wolfgang A Kunze
- *Division of Nephrology, Departments of Medicine, Psychiatry and Behavioral Neurosciences, and Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada; and McMaster Brain-Body Institute at St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - John Bienenstock
- *Division of Nephrology, Departments of Medicine, Psychiatry and Behavioral Neurosciences, and Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada; and McMaster Brain-Body Institute at St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| |
Collapse
|
170
|
Kolwijck E, van de Veerdonk FL. The potential impact of the pulmonary microbiome on immunopathogenesis of Aspergillus-related lung disease. Eur J Immunol 2014; 44:3156-65. [PMID: 25256637 DOI: 10.1002/eji.201344404] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 07/17/2014] [Accepted: 09/23/2014] [Indexed: 02/04/2023]
Abstract
Aspergillosis is an infection or allergic response caused by fungi of the genus Aspergillus. The most common forms of aspergillosis are allergic bronchopulmonary aspergillosis, chronic pulmonary aspergillosis, and invasive pulmonary aspergillosis. Aspergillus also plays an important role in fungal sensitized asthma. Humans inhale Aspergillus spores every day and when the host is immunocompromised, Aspergillus spp. may cause severe pulmonary disease. There is increasing evidence that the microbiome plays a significant role in immune regulation, chronic inflammatory diseases, metabolism, and other physiological processes, including recovery from the effects of antibiotic treatment. Bacterial microbiome mediated resistance mechanisms probably play a major role in limiting fungal colonization of the lungs, and may therefore prevent humans from contracting Aspergillus-related diseases. In this perspective, we review this emerging area of research and discuss the role of the microbiome in aspergillosis, role of Aspergillus in the microbiome, and the influence of the microbiome on anti-Aspergillus host defense and its role in preventing aspergillosis.
Collapse
Affiliation(s)
- Eva Kolwijck
- Department of Medical Microbiology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | |
Collapse
|
171
|
Rosser EC, Oleinika K, Tonon S, Doyle R, Bosma A, Carter NA, Harris KA, Jones SA, Klein N, Mauri C. Regulatory B cells are induced by gut microbiota-driven interleukin-1β and interleukin-6 production. Nat Med 2014; 20:1334-9. [PMID: 25326801 DOI: 10.1038/nm.3680] [Citation(s) in RCA: 333] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Accepted: 08/06/2014] [Indexed: 11/10/2022]
Abstract
Regulatory B cells (Breg cells) differentiate in response to inflammation and subsequently restrain excessive immune responses via the release of interleukin-10 (IL-10). However, the precise inflammatory signals governing their differentiation remain to be elucidated. Here we show that the gut microbiota promotes the differentiation of Breg cells in the spleen as well as in the mesenteric lymph nodes. Perturbation of the gut microbiome imposed either by antibiotic treatment or by changes in the sterility of housing conditions reduces the number and function of Breg cells. Following the induction of arthritis, IL-1β and IL-6 are produced only in conventionally housed mice and both cytokines directly promote Breg cell differentiation and IL-10 production. Mice lacking IL-6 receptor (IL-6R) or IL-1 receptor 1 (IL-1R1) specifically on B cells have a reduced number of IL-10-producing B cells and develop exacerbated arthritis compared to control animals. Thus, in response to inflammatory signals induced by both the gut flora and arthritis, Breg cells increase in number and restrain excessive inflammation.
Collapse
Affiliation(s)
- Elizabeth C Rosser
- Centre for Rheumatology, Division of Medicine, University College London, London, UK
| | - Kristine Oleinika
- Centre for Rheumatology, Division of Medicine, University College London, London, UK
| | - Silvia Tonon
- Universita' degli Studi di Udine, Dipartimento di Medicina e Scienze Biologiche, Udine, Italia
| | - Ronan Doyle
- Infectious Diseases and Microbiology Unit, Institute of Child Health, University College London, London, UK
| | - Anneleen Bosma
- Centre for Rheumatology, Division of Medicine, University College London, London, UK
| | - Natalie A Carter
- Centre for Rheumatology, Division of Medicine, University College London, London, UK
| | - Kathryn A Harris
- Department of Microbiology, Virology and Infection Control, Great Ormond Street Hospital National Health Service Foundation Trust, London, UK
| | - Simon A Jones
- Cardiff Institute for Infection &Immunity, The School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Nigel Klein
- Infectious Diseases and Microbiology Unit, Institute of Child Health, University College London, London, UK
| | - Claudia Mauri
- Centre for Rheumatology, Division of Medicine, University College London, London, UK
| |
Collapse
|
172
|
Ai C, Zhang Q, Ren C, Wang G, Liu X, Tian F, Zhao J, Zhang H, Chen YQ, Chen W. Genetically engineered Lactococcus lactis protect against house dust mite allergy in a BALB/c mouse model. PLoS One 2014; 9:e109461. [PMID: 25290938 PMCID: PMC4188596 DOI: 10.1371/journal.pone.0109461] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 09/10/2014] [Indexed: 11/30/2022] Open
Abstract
Background Mucosal vaccine based on lactic acid bacteria is an attractive concept for the prevention and treatment of allergic diseases, but their mechanisms of action in vivo are poorly understood. Therefore, we sought to investigate how recombinant major dust mite allergen Der p2-expressing Lactococcus lactis as a mucosal vaccine induced the immune tolerance against house dust mite allergy in a mouse model. Methods Three strains of recombinant L. lactis producing Der p2 in different cell components (extracellular, intracellular and cell wall) were firstly constructed. Their prophylactic potential was evaluated in a Der p2-sensitised mouse model, and immunomodulation properties at the cellular level were determined by measuring cytokine production in vitro. Results Der p2 expressed in the different recombinant L. lactis strains was recognized by a polyclonal anti-Der p2 antibody. Oral treatment with the recombinant L. lactis prior sensitization significantly prevented the development of airway inflammation in the Der p2-sensitized mice, as determined by the attenuation of inflammatory cells infiltration in the lung tissues and decrease of Th2 cytokines IL-4 and IL-5 levels in bronchoalveolar lavage. In addition, the serum allergen-specific IgE levels were significantly reduced, and the levels of IL-4 in the spleen and mesenteric lymph nodes cell cultures were also markedly decreased upon allergen stimulation in the mice fed with the recombinant L. lactis strains. These protective effects correlated with a significant up-regulation of regulatory T cells in the mesenteric lymph nodes. Conclusion Oral pretreatment with live recombinant L. lactis prevented the development of allergen-induced airway inflammation primarily by the induction of specific mucosal immune tolerance.
Collapse
Affiliation(s)
- Chunqing Ai
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Qiuxiang Zhang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
- * E-mail: (QZ); (WC)
| | - Chengcheng Ren
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Gang Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Xiaoming Liu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Yong Q. Chen
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
- Synergistic Innovation Center for Food Safety and Nutrition, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
- Synergistic Innovation Center for Food Safety and Nutrition, Jiangnan University, Wuxi, Jiangsu, P. R. China
- * E-mail: (QZ); (WC)
| |
Collapse
|
173
|
Intestine-derived Clostridium leptum induces murine tolerogenic dendritic cells and regulatory T cells in vitro. Hum Immunol 2014; 75:1232-8. [PMID: 25300998 DOI: 10.1016/j.humimm.2014.09.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Revised: 09/27/2014] [Accepted: 09/27/2014] [Indexed: 01/17/2023]
Abstract
Patients with autoimmune and allergic diseases frequently present with reduced numbers and functionally impaired regulatory T cells (Tregs) and/or tolerogenic dendritic cells (tDCs). tDC-mediated regulation of Treg proliferation (numbers) and activation is crucial to establishing and maintaining an appropriate level of immune tolerance. Colonic colonization of Clostridium spp. is associated with accumulation of Tregs, which inhibits development of inflammatory lesions. To investigate whether infection with the Clostridium leptum sp. can specifically induce Tregs and/or tDCs bone marrow-derived dendritic cells were cultured in the presence or absence of C. leptum then co-cultured with CD4(+)CD25(-) T cells or not. Changes in tDC numbers, Treg numbers, percentages of T cell subsets, and expression of cytokines related to Tregs (IL-10 and transforming growth factor-beta (TGF-β1)), DCs (IL-12p40 and IL-6) and effector T cells (IFN-γ, IL-4, IL-5, IL-13, and IL-17A) were measured. In the co-culture system, C. leptum-stimulated tDCs were able to increase the percentage and total number of Tregs attenuate activation of T helper cells (Th1, Th2, and Th17), and decrease the amount of secreted IL-4, IL-5, IL-13, IFN-γ and IL-17A. Thus, C. leptum exposure can induce the tDC-mediated stimulation of Tregs while disrupting the immune inflammatory response mediated by Th1, Th2 and Th17 cells.
Collapse
|
174
|
Zeng H, Chi H. Metabolic control of regulatory T cell development and function. Trends Immunol 2014; 36:3-12. [PMID: 25248463 DOI: 10.1016/j.it.2014.08.003] [Citation(s) in RCA: 195] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 08/10/2014] [Accepted: 08/15/2014] [Indexed: 02/07/2023]
Abstract
Foxp3(+) regulatory T cells (Tregs) maintain immune tolerance and play an important role in immunological diseases and cancers. Recent studies have revealed an intricate relationship between Treg biology and host and microbial metabolism. Various metabolites or nutrients produced by host and commensal microbes, such as vitamins and short-chain fatty acids (SCFAs), regulate Treg generation, trafficking, and function. Furthermore, cell intrinsic metabolic programs, orchestrated by mTOR and other metabolic sensors, modulate Foxp3 induction and Treg suppressive activity. Conversely, Tregs are crucial in regulating obesity-associated inflammation and host metabolic balance, and in shaping homeostasis of gut microbiota. We review here the interplay between Tregs and metabolism, with a particular focus on how host, commensal, and cellular metabolism impinge upon Treg homeostasis and function.
Collapse
Affiliation(s)
- Hu Zeng
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
175
|
Abstract
There is increasing interest in the potential for microbe-based therapeutic approaches to asthma and respiratory infection. However, to date, clinical trials of probiotics in the treatment of respiratory disease have met with limited success. It is becoming clear that to identify the true therapeutic potential of microbes we must move away from a purely empirical approach to clinical trials and adopt knowledge-based selection of candidate probiotics strains, dose, and means of administration. Animal models have played a key role in the identification of mechanisms underlying the immunomodulatory capacity of specific bacteria. Microbe-induced changes in dendritic cell phenotype and function appear key to orchestrating the multiple pathways, involving inter alia, T cells, natural killer cells, and alveolar macrophages, associated with the protective effect of probiotics. Moving forward, the development of knowledge-based strategies for microbe-based therapeutics in respiratory disease will be aided by greater understanding of how specific bacterial structural motifs activate unique combinations of pattern recognition receptors on dendritic cells and thus direct desired immune responses.
Collapse
|
176
|
Abstract
Many important human diseases, such as asthma, have their developmental origins in early life. Respiratory infections in particular may alter the course of asthma and may either protect against or promote the development of this disease. It is likely that the nature of the effects depends on the type and age of infection and is determined by the impact of infection on the immune and respiratory systems. Immunity in early life is plastic and can be moulded by antigen encounter, which may enhance or reinforce the asthmatic phenotype of early life, or induce protective responses. Chlamydial respiratory infections have specific effects and may increase asthma severity in early life by promoting systemic interleukin 13 responses and causing permanent changes in lung structure. Respiratory viral infections, such as those of respiratory syncytial virus and rhinovirus, promote pro-asthmatic responses in early life that contribute to the induction of asthma. By contrast, probiotics or infection or exposure to certain bacteria, such as Streptococcus pneumoniae, may have protective effects in asthma by increasing the numbers and activity of regulatory T cells. Here, we review the impact of infections on the developmental origins of asthma. Understanding these effects may lead to new therapeutic approaches for asthma that either target deleterious infections or utilize beneficial ones.
Collapse
|
177
|
Rook GAW, Raison CL, Lowry CA. Microbial 'old friends', immunoregulation and socioeconomic status. Clin Exp Immunol 2014; 177:1-12. [PMID: 24401109 PMCID: PMC4089149 DOI: 10.1111/cei.12269] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2013] [Indexed: 12/13/2022] Open
Abstract
The immune system evolved to require input from at least three sources that we collectively term the ‘old friends’: (i) the commensal microbiotas transmitted by mothers and other family members; (ii) organisms from the natural environment that modulate and diversify the commensal microbiotas; and (iii) the ‘old’ infections that could persist in small isolated hunter-gatherer groups as relatively harmless subclinical infections or carrier states. These categories of organism had to be tolerated and co-evolved roles in the development and regulation of the immune system. By contrast, the ‘crowd infections’ (such as childhood virus infections) evolved later, when urbanization led to large communities. They did not evolve immunoregulatory roles because they either killed the host or induced solid immunity, and could not persist in hunter-gatherer groups. Because the western lifestyle and medical practice deplete the ‘old’ infections (for example helminths), immunoregulatory disorders have increased, and the immune system has become more dependent upon microbiotas and the natural environment. However, urbanization maintains exposure to the crowd infections that lack immunoregulatory roles, while accelerating loss of exposure to the natural environment. This effect is most pronounced in individuals of low socioeconomic status (SES) who lack rural second homes and rural holidays. Interestingly, large epidemiological studies indicate that the health benefits of living close to green spaces are most pronounced for individuals of low SES. Here we discuss the immunoregulatory role of the natural environment, and how this may interact with, and modulate, the proinflammatory effects of psychosocial stressors in low SES individuals.
Collapse
Affiliation(s)
- G A W Rook
- Centre for Clinical Microbiology, Department of Infection, University College London (UCL), London, UK
| | | | | |
Collapse
|
178
|
Inflammatory cytokine-associated depression. Brain Res 2014; 1617:113-25. [PMID: 25003554 DOI: 10.1016/j.brainres.2014.06.032] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 06/23/2014] [Accepted: 06/26/2014] [Indexed: 02/07/2023]
Abstract
Inflammatory cytokines can sometimes trigger depression in humans, are often associated with depression, and can elicit some behaviors in animals that are homologous to major depression. Moreover, these cytokines can affect monoaminergic and glutamatergic systems, supporting an overlapping pathoetiology with major depression. This suggests that there could be a specific major depression subtype, inflammatory cytokine-associated depression (ICAD), which may require different therapeutic approaches. However, most people do not develop depression, even when exposed to sustained elevations in inflammatory cytokines. Thus several vulnerabilities and sources of resilience to inflammation-associated depression have been identified. These range from genetic differences in neurotrophic and serotonergic systems to sleep quality and omega-3 fatty acid levels. Replicating these sources of resilience as treatments could be one approach for preventing "ICAD". This article is part of a Special Issue entitled SI: Neuroimmunology in Health And Disease.
Collapse
|
179
|
Reynolds LA, Smith KA, Filbey KJ, Harcus Y, Hewitson JP, Redpath SA, Valdez Y, Yebra MJ, Finlay BB, Maizels RM. Commensal-pathogen interactions in the intestinal tract: lactobacilli promote infection with, and are promoted by, helminth parasites. Gut Microbes 2014; 5:522-32. [PMID: 25144609 PMCID: PMC4822684 DOI: 10.4161/gmic.32155] [Citation(s) in RCA: 185] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The intestinal microbiota are pivotal in determining the developmental, metabolic and immunological status of the mammalian host. However, the intestinal tract may also accommodate pathogenic organisms, including helminth parasites which are highly prevalent in most tropical countries. Both microbes and helminths must evade or manipulate the host immune system to reside in the intestinal environment, yet whether they influence each other's persistence in the host remains unknown. We now show that abundance of Lactobacillus bacteria correlates positively with infection with the mouse intestinal nematode parasite, Heligmosomoides polygyrus, as well as with heightened regulatory T cell (Treg) and Th17 responses. Moreover, H. polygyrus raises Lactobacillus species abundance in the duodenum of C57BL/6 mice, which are highly susceptible to H. polygyrus infection, but not in BALB/c mice, which are relatively resistant. Sequencing of samples at the bacterial gyrB locus identified the principal Lactobacillus species as L. taiwanensis, a previously characterized rodent commensal. Experimental administration of L. taiwanensis to BALB/c mice elevates regulatory T cell frequencies and results in greater helminth establishment, demonstrating a causal relationship in which commensal bacteria promote infection with an intestinal parasite and implicating a bacterially-induced expansion of Tregs as a mechanism of greater helminth susceptibility. The discovery of this tripartite interaction between host, bacteria and parasite has important implications for both antibiotic and anthelmintic use in endemic human populations.
Collapse
Affiliation(s)
- Lisa A Reynolds
- Centre for Immunity, Infection and Evolution, and Institute of Immunology and Infection Research; Ashworth Laboratories; University of Edinburgh; Edinburgh, UK
- Michael Smith Laboratories; University of British Columbia; Vancouver, BC Canada
| | - Katherine A Smith
- Centre for Immunity, Infection and Evolution, and Institute of Immunology and Infection Research; Ashworth Laboratories; University of Edinburgh; Edinburgh, UK
| | - Kara J Filbey
- Centre for Immunity, Infection and Evolution, and Institute of Immunology and Infection Research; Ashworth Laboratories; University of Edinburgh; Edinburgh, UK
| | - Yvonne Harcus
- Centre for Immunity, Infection and Evolution, and Institute of Immunology and Infection Research; Ashworth Laboratories; University of Edinburgh; Edinburgh, UK
| | - James P Hewitson
- Centre for Immunity, Infection and Evolution, and Institute of Immunology and Infection Research; Ashworth Laboratories; University of Edinburgh; Edinburgh, UK
| | - Stephen A Redpath
- Department of Microbiology and Immunology; University of British Columbia; Vancouver, BC Canada
| | - Yanet Valdez
- Michael Smith Laboratories; University of British Columbia; Vancouver, BC Canada
| | - María J Yebra
- Laboratorio de Bacterias Lácticas y Probióticos; Instituto de Agroquímica y Tecnología de los Alimentos; IATA-CSIC; Valencia, Spain
| | - B Brett Finlay
- Michael Smith Laboratories; University of British Columbia; Vancouver, BC Canada
- Department of Microbiology and Immunology; University of British Columbia; Vancouver, BC Canada
- Department of Biochemistry and Molecular Biology; University of British Columbia; Vancouver, BC Canada
| | - Rick M Maizels
- Centre for Immunity, Infection and Evolution, and Institute of Immunology and Infection Research; Ashworth Laboratories; University of Edinburgh; Edinburgh, UK
| |
Collapse
|
180
|
Forsberg A, Abrahamsson TR, Björkstén B, Jenmalm MC. Pre- and postnatal administration of Lactobacillus reuteri decreases TLR2 responses in infants. Clin Transl Allergy 2014; 4:21. [PMID: 25002964 PMCID: PMC4083862 DOI: 10.1186/2045-7022-4-21] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 06/22/2014] [Indexed: 11/30/2022] Open
Abstract
Background Mice models indicate that intact Toll like receptor (TLR) signaling may be essential for the allergy protective effects of diverse bacterial exposure observed in clinical trials and epidemiological studies. Probiotic supplementation with Lactobacillus reuteri from pregnancy week 36 and to the infant through the first year of life decreased the prevalence of IgE-associated eczema at two years (ClinicalTrials.gov NCT01285830). The effect of this supplementation on innate immune responses to bacterial products and the expression of associated TLRs were explored. Methods Blood mononuclear cells were collected at birth, 6, 12 and 24 months from 61 infants and cultured with TLR2, 4 and 9 ligands. Cytokine and chemokine secretion was determined as well as TLR2, 4 and 9 mRNA expression. Results Probiotic supplementation was associated with decreased LTA (lipoteichoic acid) induced CCL4, CXCL8, IL-1β and IL-6 responses at 12 months and decreased CCL4 and IL-1β secretion at 24 months. TLR2 mRNA expression was not affected by probiotic treatment. Conclusions Decreased responses to TLR2, the main receptor for LTA from Gram positive bacteria, in probiotic treated children seem to be dependent on factors downstream of TLR mRNA expression.
Collapse
Affiliation(s)
- Anna Forsberg
- Department of Clinical and Experimental Medicine, Unit of Autoimmunity and Immune Regulation, Division of Clinical Immunology, Linköping University, SE-581 85 Linköping, Sweden
| | - Thomas R Abrahamsson
- Department of Clinical and Experimental Medicine, Division of Pediatrics, Linköping University, Linköping, Sweden
| | - Bengt Björkstén
- Institute of Environmental Medicine, Karolinska Institutet, Solna, Stockholm, Sweden ; School of Health and Medical Sciences, Örebro University, Örebro, Sweden
| | - Maria C Jenmalm
- Department of Clinical and Experimental Medicine, Unit of Autoimmunity and Immune Regulation, Division of Clinical Immunology, Linköping University, SE-581 85 Linköping, Sweden
| |
Collapse
|
181
|
Spinler JK, Sontakke A, Hollister EB, Venable SF, Oh PL, Balderas MA, Saulnier DMA, Mistretta TA, Devaraj S, Walter J, Versalovic J, Highlander SK. From prediction to function using evolutionary genomics: human-specific ecotypes of Lactobacillus reuteri have diverse probiotic functions. Genome Biol Evol 2014; 6:1772-89. [PMID: 24951561 PMCID: PMC4122935 DOI: 10.1093/gbe/evu137] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The vertebrate gut symbiont Lactobacillus reuteri has diversified into separate clades reflecting host origin. Strains show evidence of host adaptation, but how host–microbe coevolution influences microbial-derived effects on hosts is poorly understood. Emphasizing human-derived strains of L. reuteri, we combined comparative genomic analyses with functional assays to examine variations in host interaction among genetically distinct ecotypes. Within clade II or VI, the genomes of human-derived L. reuteri strains are highly conserved in gene content and at the nucleotide level. Nevertheless, they share only 70–90% of total gene content, indicating differences in functional capacity. Human-associated lineages are distinguished by genes related to bacteriophages, vitamin biosynthesis, antimicrobial production, and immunomodulation. Differential production of reuterin, histamine, and folate by 23 clade II and VI strains was demonstrated. These strains also differed with respect to their ability to modulate human cytokine production (tumor necrosis factor, monocyte chemoattractant protein-1, interleukin [IL]-1β, IL-5, IL-7, IL-12, and IL-13) by myeloid cells. Microarray analysis of representative clade II and clade VI strains revealed global regulation of genes within the reuterin, vitamin B12, folate, and arginine catabolism gene clusters by the AraC family transcriptional regulator, PocR. Thus, human-derived L. reuteri clade II and VI strains are genetically distinct and their differences affect their functional repertoires and probiotic features. These findings highlight the biological impact of microbe:host coevolution and illustrate the functional significance of subspecies differences in the human microbiome. Consideration of host origin and functional differences at the subspecies level may have major impacts on probiotic strain selection and considerations of microbial ecology in mammalian species.
Collapse
Affiliation(s)
- Jennifer K Spinler
- Texas Children's Microbiome Center, Department of Pathology, Texas Children's Hospital, Houston, TXDepartment of Pathology & Immunology, Baylor College of Medicine, Houston, TX
| | - Amrita Sontakke
- Texas Children's Microbiome Center, Department of Pathology, Texas Children's Hospital, Houston, TXDepartment of Pathology & Immunology, Baylor College of Medicine, Houston, TX
| | - Emily B Hollister
- Texas Children's Microbiome Center, Department of Pathology, Texas Children's Hospital, Houston, TXDepartment of Pathology & Immunology, Baylor College of Medicine, Houston, TX
| | - Susan F Venable
- Texas Children's Microbiome Center, Department of Pathology, Texas Children's Hospital, Houston, TXDepartment of Pathology & Immunology, Baylor College of Medicine, Houston, TX
| | - Phaik Lyn Oh
- Department of Food Science and Technology, University of Nebraska, Lincoln
| | - Miriam A Balderas
- Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX
| | - Delphine M A Saulnier
- Texas Children's Microbiome Center, Department of Pathology, Texas Children's Hospital, Houston, TXDepartment of Pathology & Immunology, Baylor College of Medicine, Houston, TXPresent address: Department of Gastrointestinal Microbiology, German Institute of Human Nutrition, Nuthetal, Germany
| | - Toni-Ann Mistretta
- Texas Children's Microbiome Center, Department of Pathology, Texas Children's Hospital, Houston, TXDepartment of Pathology & Immunology, Baylor College of Medicine, Houston, TX
| | - Sridevi Devaraj
- Texas Children's Microbiome Center, Department of Pathology, Texas Children's Hospital, Houston, TXDepartment of Pathology & Immunology, Baylor College of Medicine, Houston, TX
| | - Jens Walter
- Department of Food Science and Technology, University of Nebraska, LincolnPresent address: Departments of Agricultural, Food, & Nutritional Science and Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - James Versalovic
- Texas Children's Microbiome Center, Department of Pathology, Texas Children's Hospital, Houston, TXDepartment of Pathology & Immunology, Baylor College of Medicine, Houston, TXDepartment of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX
| | - Sarah K Highlander
- Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TXHuman Genome Sequencing Center, Baylor College of Medicine, Houston, TXPresent address: Genomic Medicine, J. Craig Venter Institute, La Jolla, CA
| |
Collapse
|
182
|
Belkaid Y, Hand TW. Role of the microbiota in immunity and inflammation. Cell 2014; 157:121-41. [PMID: 24679531 DOI: 10.1016/j.cell.2014.03.011] [Citation(s) in RCA: 3089] [Impact Index Per Article: 308.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 03/10/2014] [Accepted: 03/11/2014] [Indexed: 02/06/2023]
Abstract
The microbiota plays a fundamental role on the induction, training, and function of the host immune system. In return, the immune system has largely evolved as a means to maintain the symbiotic relationship of the host with these highly diverse and evolving microbes. When operating optimally, this immune system-microbiota alliance allows the induction of protective responses to pathogens and the maintenance of regulatory pathways involved in the maintenance of tolerance to innocuous antigens. However, in high-income countries, overuse of antibiotics, changes in diet, and elimination of constitutive partners, such as nematodes, may have selected for a microbiota that lack the resilience and diversity required to establish balanced immune responses. This phenomenon is proposed to account for some of the dramatic rise in autoimmune and inflammatory disorders in parts of the world where our symbiotic relationship with the microbiota has been the most affected.
Collapse
Affiliation(s)
- Yasmine Belkaid
- Immunity at Barrier Sites Initiative, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Timothy W Hand
- Immunity at Barrier Sites Initiative, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
183
|
Abstract
The impact of the gut microbiota on immune homeostasis within the gut and, importantly, also at systemic sites has gained tremendous research interest over the last few years. The intestinal microbiota is an integral component of a fascinating ecosystem that interacts with and benefits its host on several complex levels to achieve a mutualistic relationship. Host-microbial homeostasis involves appropriate immune regulation within the gut mucosa to maintain a healthy gut while preventing uncontrolled immune responses against the beneficial commensal microbiota potentially leading to chronic inflammatory bowel diseases (IBD). Furthermore, recent studies suggest that the microbiota composition might impact on the susceptibility to immune-mediated disorders such as autoimmunity and allergy. Understanding how the microbiota modulates susceptibility to these diseases is an important step toward better prevention or treatment options for such diseases.
Collapse
|
184
|
Relationship between gut microbiota and development of T cell associated disease. FEBS Lett 2014; 588:4195-206. [PMID: 24681103 DOI: 10.1016/j.febslet.2014.03.019] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 03/11/2014] [Accepted: 03/11/2014] [Indexed: 12/11/2022]
Abstract
The interplay between the immune response and the gut microbiota is complex. Although it is well-established that the gut microbiota is essential for the proper development of the immune system, recent evidence indicates that the cells of the immune system also influence the composition of the gut microbiota. This interaction can have important consequences for the development of inflammatory diseases, including autoimmune diseases and allergy, and the specific mechanisms by which the gut commensals drive the development of different types of immune responses are beginning to be understood. Furthermore, sex hormones are now thought to play a novel role in this complex relationship, and collaborate with both the gut microbiota and immune system to influence the development of autoimmune disease. In this review, we will focus on recent studies that have transformed our understanding of the importance of the gut microbiota in inflammatory responses.
Collapse
|
185
|
Abstract
Chronic obstructive pulmonary disease (COPD) is currently the third leading cause of death in the world. Although smoking is the main risk factor for this disease, only a minority of smokers develop COPD. Why this happens is largely unknown. Recent discoveries by the human microbiome project have shed new light on the importance and richness of the bacterial microbiota at different body sites in human beings. The microbiota plays a particularly important role in the development and functional integrity of the immune system. Shifts or perturbations in the microbiota can lead to disease. COPD is in part mediated by dysregulated immune responses to cigarette smoke and other environmental insults. Although traditionally the lung has been viewed as a sterile organ, by using highly sensitive genomic techniques, recent reports have identified diverse bacterial communities in the human lung that may change in COPD. This review summarizes the current knowledge concerning the lung microbiota in COPD and its potential implications for pathogenesis of the disease.
Collapse
Affiliation(s)
- Marc A Sze
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - James C Hogg
- Department of Pathology and Laboratory Medicine, The James Hogg Research Centre, Providence Heart-Lung Institute, St Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Don D Sin
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
186
|
Abstract
The pathogenetic mechanisms leading to asthma are likely to be diverse, influenced by multiple genetic polymorphisms as well as elements of the environment. Recent data on the microbiome of the airway have revealed intriguing differences between the number and diversity of microbial populations in healthy persons and asthmatics. There is convincing evidence that early viral infections, particularly with human rhinovirus and respiratory syncytial virus, are often associated with the development of chronic asthma and with exacerbations. Recent studies suggest that two unrelated types of atypical bacteria, Mycoplasma pneumoniae (Mpn) and Chlamydia pneumoniae, are present in the airways of a substantial proportion of the population, bringing up the possibility that the persistent presence of the organism may contribute to the asthmatic phenotype in a subset of patients. This review will examine the current data regarding a possible role for infection in chronic asthma with a particular focus on atypical bacterial infections.
Collapse
Affiliation(s)
- T Prescott Atkinson
- Children's of Alabama CPP M220, 1601 4th Ave South, Birmingham, AL, 35233, USA,
| |
Collapse
|
187
|
Huang YJ. Asthma microbiome studies and the potential for new therapeutic strategies. Curr Allergy Asthma Rep 2014; 13:453-61. [PMID: 23709178 DOI: 10.1007/s11882-013-0355-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Recent applications of culture-independent tools for microbiome profiling have revealed significant relationships between asthma and microbiota associated with the environment, gut, or airways. Studies of the airway microbiome in particular represent a new frontier in pulmonary research. Although these studies are relatively new, current evidence suggests the possibility of new therapeutic strategies for the treatment or prevention of asthma. In this article, recent literature on microbiota and asthma are critically reviewed, with a particular focus on studies of the airway microbiome. Perspectives are presented on how growing knowledge of relationships between the microbiome and asthma is likely to translate into improved understanding of asthma pathogenesis, its heterogeneity, and opportunities for novel treatment approaches.
Collapse
Affiliation(s)
- Yvonne J Huang
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, University of California San Francisco, 505 Parnassus Avenue, Box 0130, San Francisco, CA, 94143-0130, USA,
| |
Collapse
|
188
|
Lakritz JR, Poutahidis T, Levkovich T, Varian BJ, Ibrahim YM, Chatzigiagkos A, Mirabal S, Alm EJ, Erdman SE. Beneficial bacteria stimulate host immune cells to counteract dietary and genetic predisposition to mammary cancer in mice. Int J Cancer 2014; 135:529-40. [PMID: 24382758 PMCID: PMC4131439 DOI: 10.1002/ijc.28702] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 12/23/2013] [Indexed: 12/16/2022]
Abstract
Recent studies suggest health benefits including protection from cancer after eating fermented foods such as probiotic yogurt, though the mechanisms are not well understood. Here we tested mechanistic hypotheses using two different animal models: the first model studied development of mammary cancer when eating a Westernized diet, and the second studied animals with a genetic predilection to breast cancer. For the first model, outbred Swiss mice were fed a Westernized chow putting them at increased risk for development of mammary tumors. In this Westernized diet model, mammary carcinogenesis was inhibited by routine exposure to Lactobacillus reuteri ATCC-PTA-6475 in drinking water. The second model was FVB strain erbB2 (HER2) mutant mice, genetically susceptible to mammary tumors mimicking breast cancers in humans, being fed a regular (non-Westernized) chow diet. We found that oral supplement with these purified lactic acid bacteria alone was sufficient to inhibit features of mammary neoplasia in both models. The protective mechanism was determined to be microbially-triggered CD4+CD25+ lymphocytes. When isolated and transplanted into other subjects, these L. reuteri-stimulated lymphocytes were sufficient to convey transplantable anti-cancer protection in the cell recipient animals. These data demonstrate that host immune responses to environmental microbes significantly impact and inhibit cancer progression in distal tissues such as mammary glands, even in genetically susceptible mice. This leads us to conclude that consuming fermentative microbes such as L. reuteri may offer a tractable public health approach to help counteract the accumulated dietary and genetic carcinogenic events integral in the Westernized diet and lifestyle.
Collapse
Affiliation(s)
- Jessica R Lakritz
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA
| | | | | | | | | | | | | | | | | |
Collapse
|
189
|
Abstract
Although traditionally thought to be sterile, accumulating evidence now supports the concept that our airways harbor a microbiome. Thus far, studies have focused upon characterizing the bacterial constituents of the airway microbiome in both healthy and diseased lungs, but what perhaps provides the greatest impetus for the exploration of the airway microbiome is that different bacterial phyla appear to dominate diseased as compared with healthy lungs. As yet, there is very limited evidence supporting a functional role for the airway microbiome, but continued research in this direction is likely to provide such evidence, particularly considering the progress that has been made in understanding host-microbe mutualism in the intestinal tract. In this review, we highlight the major advances that have been made discovering and describing the airway microbiome, discuss the experimental evidence that supports a functional role for the microbiome in health and disease, and propose how this emerging field is going to impact clinical practice.
Collapse
Affiliation(s)
- Benjamin J Marsland
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, CHUV, Lausanne, Switzerland.
| | - Koshika Yadava
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, CHUV, Lausanne, Switzerland
| | - Laurent P Nicod
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, CHUV, Lausanne, Switzerland
| |
Collapse
|
190
|
The murine lung microbiome in relation to the intestinal and vaginal bacterial communities. BMC Microbiol 2013; 13:303. [PMID: 24373613 PMCID: PMC3878784 DOI: 10.1186/1471-2180-13-303] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 12/23/2013] [Indexed: 12/27/2022] Open
Abstract
Background This work provides the first description of the bacterial population of the lung microbiota in mice. The aim of this study was to examine the lung microbiome in mice, the most used animal model for inflammatory lung diseases such as COPD, cystic fibrosis and asthma. Bacterial communities from broncho-alveolar lavage fluids and lung tissue were compared to samples taken from fecal matter (caecum) and vaginal lavage fluid from female BALB/cJ mice. Results Using a customized 16S rRNA sequencing protocol amplifying the V3-V4 region our study shows that the mice have a lung microbiome that cluster separately from mouse intestinal microbiome (caecum). The mouse lung microbiome is dominated by Proteobacteria, Firmicutes, Actinobacteria, Bacteroidetes and Cyanobacteria overlapping the vaginal microbiome. We also show that removal of host tissue or cells from lung fluid during the DNA extraction step has an impact on the resulting bacterial community profile. Sample preparation needs to be considered when choosing an extraction method and interpreting data. Conclusions We have consistently amplified bacterial DNA from mouse lungs that is distinct from the intestinal microbiome in these mice. The gut microbiome has been extensively studied for its links to development of disease. Here we suggest that also the lung microbiome could be important in relation to inflammatory lung diseases. Further research is needed to understand the contribution of the lung microbiome and the gut-lung axis to the development of lung diseases such as COPD and asthma.
Collapse
|
191
|
Microbial symbionts accelerate wound healing via the neuropeptide hormone oxytocin. PLoS One 2013; 8:e78898. [PMID: 24205344 PMCID: PMC3813596 DOI: 10.1371/journal.pone.0078898] [Citation(s) in RCA: 198] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 09/17/2013] [Indexed: 02/07/2023] Open
Abstract
Wound healing capability is inextricably linked with diverse aspects of physical fitness ranging from recovery after minor injuries and surgery to diabetes and some types of cancer. Impact of the microbiome upon the mammalian wound healing process is poorly understood. We discover that supplementing the gut microbiome with lactic acid microbes in drinking water accelerates the wound-healing process to occur in half the time required for matched control animals. Further, we find that Lactobacillus reuteri enhances wound-healing properties through up-regulation of the neuropeptide hormone oxytocin, a factor integral in social bonding and reproduction, by a vagus nerve-mediated pathway. Bacteria-triggered oxytocin serves to activate host CD4+Foxp3+CD25+ immune T regulatory cells conveying transplantable wound healing capacity to naive Rag2-deficient animals. This study determined oxytocin to be a novel component of a multi-directional gut microbe-brain-immune axis, with wound-healing capability as a previously unrecognized output of this axis. We also provide experimental evidence to support long-standing medical traditions associating diet, social practices, and the immune system with efficient recovery after injury, sustained good health, and longevity.
Collapse
|
192
|
Regulation of the immune system by biodiversity from the natural environment: an ecosystem service essential to health. Proc Natl Acad Sci U S A 2013; 110:18360-7. [PMID: 24154724 DOI: 10.1073/pnas.1313731110] [Citation(s) in RCA: 373] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Epidemiological studies suggest that living close to the natural environment is associated with long-term health benefits including reduced death rates, reduced cardiovascular disease, and reduced psychiatric problems. This is often attributed to psychological mechanisms, boosted by exercise, social interactions, and sunlight. Compared with urban environments, exposure to green spaces does indeed trigger rapid psychological, physiological, and endocrinological effects. However, there is little evidence that these rapid transient effects cause long-term health benefits or even that they are a specific property of natural environments. Meanwhile, the illnesses that are increasing in high-income countries are associated with failing immunoregulation and poorly regulated inflammatory responses, manifested as chronically raised C-reactive protein and proinflammatory cytokines. This failure of immunoregulation is partly attributable to a lack of exposure to organisms ("Old Friends") from mankind's evolutionary past that needed to be tolerated and therefore evolved roles in driving immunoregulatory mechanisms. Some Old Friends (such as helminths and infections picked up at birth that established carrier states) are almost eliminated from the urban environment. This increases our dependence on Old Friends derived from our mothers, other people, animals, and the environment. It is suggested that the requirement for microbial input from the environment to drive immunoregulation is a major component of the beneficial effect of green space, and a neglected ecosystem service that is essential for our well-being. This insight will allow green spaces to be designed to optimize health benefits and will provide impetus from health systems for the preservation of ecosystem biodiversity.
Collapse
|
193
|
|
194
|
Asano K, Yoshimura S, Nakane A. Alteration of intestinal microbiota in mice orally administered with salmon cartilage proteoglycan, a prophylactic agent. PLoS One 2013; 8:e75008. [PMID: 24040376 PMCID: PMC3767651 DOI: 10.1371/journal.pone.0075008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 08/08/2013] [Indexed: 12/20/2022] Open
Abstract
Proteoglycan (PG) extracted from salmon nasal cartilage has potential to be a prophylactic agent. Daily oral administration of the PG attenuates systemic inflammatory response in the experimental mouse models. In this study, we applied the culture-independent approach to investigate an alteration of intestinal microbiota composition in PG-administered mice. The results indicated that the population level of bacilli increased in the small and large intestine upon PG administration. On the other hand, the population level of clostridia decreased in the large intestine. The proportion of bacteria that are able to ferment saccharides and produce short-chain fatty acids increased in the small intestine and decreased in the large intestine. Importantly, population level of probiotic lactobacilli and bacteria exhibiting the immunomodulatory effect increased in the PG-administered mice. In addition, several disease-associated bacteria decreased upon PG administration. These results provided an understanding of the specific role of PG involved in host immune modulation and supported our hypothesis that daily oral administration of PG improves the overall balance in composition of the intestinal microbial community.
Collapse
Affiliation(s)
- Krisana Asano
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Sayuri Yoshimura
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Akio Nakane
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
- * E-mail:
| |
Collapse
|
195
|
Gollwitzer ES, Marsland BJ. Microbiota abnormalities in inflammatory airway diseases - Potential for therapy. Pharmacol Ther 2013; 141:32-9. [PMID: 23969226 DOI: 10.1016/j.pharmthera.2013.08.002] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 07/29/2013] [Indexed: 02/07/2023]
Abstract
Increasingly the development of novel therapeutic strategies is taking into consideration the contribution of the intestinal microbiota to health and disease. Dysbiosis of the microbial communities colonizing the human intestinal tract has been described for a variety of chronic diseases, such as inflammatory bowel disease, obesity and asthma. In particular, reduction of several so-called probiotic species including Lactobacilli and Bifidobacteria that are generally considered to be beneficial, as well as an outgrowth of potentially pathogenic bacteria is often reported. Thus a tempting therapeutic approach is to shape the constituents of the microbiota in an attempt to restore the microbial balance towards the growth of 'health-promoting' bacterial species. A twist to this scenario is the recent discovery that the respiratory tract also harbors a microbiota under steady-state conditions. Investigators have shown that the microbial composition of the airway flora is different between healthy lungs and those with chronic lung diseases, such as asthma, chronic obstructive pulmonary disease as well as cystic fibrosis. This is an emerging field, and thus far there is very limited data showing a direct contribution of the airway microbiota to the onset and progression of disease. However, should future studies provide such evidence, the airway microbiota might soon join the intestinal microbiota as a target for therapeutic intervention. In this review, we highlight the major advances that have been made describing the microbiota in chronic lung disease and discuss current and future approaches concerning manipulation of the microbiota for the treatment and prevention of disease.
Collapse
Affiliation(s)
- Eva S Gollwitzer
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, CHUV, Lausanne, Switzerland
| | - Benjamin J Marsland
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, CHUV, Lausanne, Switzerland.
| |
Collapse
|
196
|
Huang YJ, Charlson ES, Collman RG, Colombini-Hatch S, Martinez FD, Senior RM. The role of the lung microbiome in health and disease. A National Heart, Lung, and Blood Institute workshop report. Am J Respir Crit Care Med 2013; 187:1382-7. [PMID: 23614695 DOI: 10.1164/rccm.201303-0488ws] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Study of the human lung microbiome in the context of pulmonary health and disease is an area of emerging research interest that is being driven by several contributing factors. These factors include increased recognition of the diversity of human-associated microbiota, their roles in health and in diseases associated with chronic inflammation, and advancements in technologies and tools that have facilitated such discoveries about the microbiota in organ systems outside of the lung. Therefore, the overarching goals of lung microbiome research are: to identify and characterize microbial populations associated with the respiratory tract and lungs; to understand their roles in lung health and disease; and, we hope, to allow the development of improved approaches for diagnosing and treating chronic respiratory diseases in which the microbiome has a role. Recent studies of the lung microbiome have yielded a number of interesting findings but also highlighted questions and challenges for researchers and clinicians. In December 2011, the National Heart, Lung, and Blood Institute convened a workshop to identify key issues and areas for further attention or development to advance research on the lung microbiome. Current knowledge and the state of research on the lung and related areas of human microbiome investigation were reviewed and discussed.
Collapse
Affiliation(s)
- Yvonne J Huang
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | | | | | | | | | | |
Collapse
|
197
|
|
198
|
Probiotics, prebiotics and immunomodulation of gut mucosal defences: homeostasis and immunopathology. Nutrients 2013; 5:1869-912. [PMID: 23760057 PMCID: PMC3725482 DOI: 10.3390/nu5061869] [Citation(s) in RCA: 290] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 05/08/2013] [Accepted: 05/09/2013] [Indexed: 12/13/2022] Open
Abstract
Probiotics are beneficial microbes that confer a realistic health benefit on the host, which in combination with prebiotics, (indigestible dietary fibre/carbohydrate), also confer a health benefit on the host via products resulting from anaerobic fermentation. There is a growing body of evidence documenting the immune-modulatory ability of probiotic bacteria, it is therefore reasonable to suggest that this is potentiated via a combination of prebiotics and probiotics as a symbiotic mix. The need for probiotic formulations has been appreciated for the health benefits in "topping up your good bacteria" or indeed in an attempt to normalise the dysbiotic microbiota associated with immunopathology. This review will focus on the immunomodulatory role of probiotics and prebiotics on the cells, molecules and immune responses in the gut mucosae, from epithelial barrier to priming of adaptive responses by antigen presenting cells: immune fate decision-tolerance or activation? Modulation of normal homeostatic mechanisms, coupled with findings from probiotic and prebiotic delivery in pathological studies, will highlight the role for these xenobiotics in dysbiosis associated with immunopathology in the context of inflammatory bowel disease, colorectal cancer and hypersensitivity.
Collapse
|
199
|
Mortaz E, Adcock IM, Folkerts G, Barnes PJ, Paul Vos A, Garssen J. Probiotics in the management of lung diseases. Mediators Inflamm 2013; 2013:751068. [PMID: 23737654 PMCID: PMC3662166 DOI: 10.1155/2013/751068] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Revised: 03/14/2013] [Accepted: 03/28/2013] [Indexed: 12/15/2022] Open
Abstract
The physiology and pathology of the respiratory and gastrointestinal tracts are closely related. This similarity between the two organs may underlie why dysfunction in one organ may induce illness in the other. For example, smoking is a major risk factor for COPD and IBD and increases the risk of developing Crohn's disease. Probiotics have been defined as "live microorganisms which, when administered in adequate amounts, confer health benefits on the host." In model systems probiotics regulate innate and inflammatory immune responses. Commonly used probiotics include lactic acid bacteria, particularly Lactobacillus, Bifidobacterium, and Saccharomyces, and these are often used as dietary supplements to provide a health benefit in gastrointestinal diseases including infections, inflammatory bowel disease, and colon cancer. In this respect, probiotics probably act as immunomodulatory agents and activators of host defence pathways which suggest that they could influence disease severity and incidence at sites distal to the gut. There is increasing evidence that orally delivered probiotics are able to regulate immune responses in the respiratory system. This review provides an overview of the possible role of probiotics and their mechanisms of action in the prevention and treatment of respiratory diseases.
Collapse
Affiliation(s)
- Esmaeil Mortaz
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
- Chronic Respiratory Diseases Research Center and National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Department of Immunology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ian M. Adcock
- Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Peter J. Barnes
- Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Arjan Paul Vos
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
- Danone Research Centre for Specialised Nutrition, Wageningen, The Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
- Danone Research Centre for Specialised Nutrition, Wageningen, The Netherlands
| |
Collapse
|
200
|
Felger JC, Lotrich FE. Inflammatory cytokines in depression: neurobiological mechanisms and therapeutic implications. Neuroscience 2013; 246:199-229. [PMID: 23644052 DOI: 10.1016/j.neuroscience.2013.04.060] [Citation(s) in RCA: 723] [Impact Index Per Article: 65.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 04/08/2013] [Accepted: 04/27/2013] [Indexed: 12/28/2022]
Abstract
Mounting evidence indicates that inflammatory cytokines contribute to the development of depression in both medically ill and medically healthy individuals. Cytokines are important for development and normal brain function, and have the ability to influence neurocircuitry and neurotransmitter systems to produce behavioral alterations. Acutely, inflammatory cytokine administration or activation of the innate immune system produces adaptive behavioral responses that promote conservation of energy to combat infection or recovery from injury. However, chronic exposure to elevated inflammatory cytokines and persistent alterations in neurotransmitter systems can lead to neuropsychiatric disorders and depression. Mechanisms of cytokine behavioral effects involve activation of inflammatory signaling pathways in the brain that results in changes in monoamine, glutamate, and neuropeptide systems, and decreases in growth factors, such as brain-derived neurotrophic factor. Furthermore, inflammatory cytokines may serve as mediators of both environmental (e.g. childhood trauma, obesity, stress, and poor sleep) and genetic (functional gene polymorphisms) factors that contribute to depression's development. This review explores the idea that specific gene polymorphisms and neurotransmitter systems can confer protection from or vulnerability to specific symptom dimensions of cytokine-related depression. Additionally, potential therapeutic strategies that target inflammatory cytokine signaling or the consequences of cytokines on neurotransmitter systems in the brain to prevent or reverse cytokine effects on behavior are discussed.
Collapse
Affiliation(s)
- J C Felger
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30306, United States.
| | | |
Collapse
|