1
|
Pietrasanta C, Carlosama C, Lizier M, Fornasa G, Jost TR, Carloni S, Giugliano S, Silvestri A, Brescia P, De Ponte Conti B, Braga D, Mihula M, Morosi L, Bernardinello A, Ronchi A, Martano G, Mosca F, Penna G, Grassi F, Pugni L, Rescigno M. Prenatal antibiotics reduce breast milk IgA and induce dysbiosis in mouse offspring, increasing neonatal susceptibility to bacterial sepsis. Cell Host Microbe 2024; 32:2178-2194.e6. [PMID: 39603245 DOI: 10.1016/j.chom.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 07/25/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024]
Abstract
Antibiotics (Abx) are administered to 20%-30% of pregnant women, but their effects on neonatal immune development are poorly understood. We show that newborn mice born to Abx-treated dams are more susceptible to late-onset sepsis. This susceptibility is linked to lower maternal breast milk immunoglobulin A (IgA), neonatal fecal IgA, and IgA coating of intestinal bacteria, thus causing the translocation of intestinal pathobionts. Weaned young adults born to Abx-treated mothers had reduced IgA+ plasma cells in the ileum and colon, fecal secretory IgA (SIgA), colonic CD4+ T regulatory lymphocytes and T helper 17-like lymphocytes, and a less diverse fecal microbiome. However, treatment with apyrase, which restores SIgA secretion, prompted IgA production in breast milk and protected pups from sepsis. Additionally, breast milk from untreated mothers rescued the phenotypes of pups born to Abx-treated mothers. Our data highlight the impact of prenatal Abx on breast milk IgA and their long-term influence on intestinal mucosal immune function mediated by breastfeeding.
Collapse
Affiliation(s)
- Carlo Pietrasanta
- Department of Clinical Sciences and Community Health, Dipartimento di Eccellenza 2023-2027, University of Milan, Via della Commenda 19, Milan, Italy; NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via della Commenda 12, Milan, Italy.
| | - Carolina Carlosama
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy
| | - Michela Lizier
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy
| | - Giulia Fornasa
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy
| | - Tanja Rezzonico Jost
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
| | - Sara Carloni
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, Milan 20072, Italy
| | - Silvia Giugliano
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, Milan 20072, Italy
| | | | - Paola Brescia
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, Milan 20072, Italy
| | - Benedetta De Ponte Conti
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
| | - Daniele Braga
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy
| | - Martin Mihula
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy; Department of Medical Biotechnology, Università di Siena, Via Banchi di Sotto 55, 53100 Siena, Italy
| | - Lavinia Morosi
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy
| | - Alessandro Bernardinello
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, Milan 20072, Italy
| | - Andrea Ronchi
- NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via della Commenda 12, Milan, Italy
| | - Giuseppe Martano
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy; Institute of Neuroscience, National Research Council of Italy (CNR) c/o Humanitas Mirasole S.p.A, Via Manzoni 56, Rozzano, Milan, Italy
| | - Fabio Mosca
- Department of Clinical Sciences and Community Health, Dipartimento di Eccellenza 2023-2027, University of Milan, Via della Commenda 19, Milan, Italy; NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via della Commenda 12, Milan, Italy
| | - Giuseppe Penna
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy
| | - Fabio Grassi
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland; Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Lorenza Pugni
- NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via della Commenda 12, Milan, Italy
| | - Maria Rescigno
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, Milan 20072, Italy.
| |
Collapse
|
2
|
Seefeld ML, Templeton EL, Lehtinen JM, Sinclair N, Yadav D, Hartwell BL. Harnessing the potential of the NALT and BALT as targets for immunomodulation using engineering strategies to enhance mucosal uptake. Front Immunol 2024; 15:1419527. [PMID: 39286244 PMCID: PMC11403286 DOI: 10.3389/fimmu.2024.1419527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/08/2024] [Indexed: 09/19/2024] Open
Abstract
Mucosal barrier tissues and their mucosal associated lymphoid tissues (MALT) are attractive targets for vaccines and immunotherapies due to their roles in both priming and regulating adaptive immune responses. The upper and lower respiratory mucosae, in particular, possess unique properties: a vast surface area responsible for frontline protection against inhaled pathogens but also simultaneous tight regulation of homeostasis against a continuous backdrop of non-pathogenic antigen exposure. Within the upper and lower respiratory tract, the nasal and bronchial associated lymphoid tissues (NALT and BALT, respectively) are key sites where antigen-specific immune responses are orchestrated against inhaled antigens, serving as critical training grounds for adaptive immunity. Many infectious diseases are transmitted via respiratory mucosal sites, highlighting the need for vaccines that can activate resident frontline immune protection in these tissues to block infection. While traditional parenteral vaccines that are injected tend to elicit weak immunity in mucosal tissues, mucosal vaccines (i.e., that are administered intranasally) are capable of eliciting both systemic and mucosal immunity in tandem by initiating immune responses in the MALT. In contrast, administering antigen to mucosal tissues in the absence of adjuvant or costimulatory signals can instead induce antigen-specific tolerance by exploiting regulatory mechanisms inherent to MALT, holding potential for mucosal immunotherapies to treat autoimmunity. Yet despite being well motivated by mucosal biology, development of both mucosal subunit vaccines and immunotherapies has historically been plagued by poor drug delivery across mucosal barriers, resulting in weak efficacy, short-lived responses, and to-date a lack of clinical translation. Development of engineering strategies that can overcome barriers to mucosal delivery are thus critical for translation of mucosal subunit vaccines and immunotherapies. This review covers engineering strategies to enhance mucosal uptake via active targeting and passive transport mechanisms, with a parallel focus on mechanisms of immune activation and regulation in the respiratory mucosa. By combining engineering strategies for enhanced mucosal delivery with a better understanding of immune mechanisms in the NALT and BALT, we hope to illustrate the potential of these mucosal sites as targets for immunomodulation.
Collapse
Affiliation(s)
- Madison L Seefeld
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Erin L Templeton
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Justin M Lehtinen
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Noah Sinclair
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Daman Yadav
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Brittany L Hartwell
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
- Center for Immunology, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
3
|
Mostafavi Abdolmaleky H, Zhou JR. Gut Microbiota Dysbiosis, Oxidative Stress, Inflammation, and Epigenetic Alterations in Metabolic Diseases. Antioxidants (Basel) 2024; 13:985. [PMID: 39199231 PMCID: PMC11351922 DOI: 10.3390/antiox13080985] [Citation(s) in RCA: 48] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/05/2024] [Accepted: 08/11/2024] [Indexed: 09/01/2024] Open
Abstract
Gut dysbiosis, resulting from an imbalance in the gut microbiome, can induce excessive production of reactive oxygen species (ROS), leading to inflammation, DNA damage, activation of the immune system, and epigenetic alterations of critical genes involved in the metabolic pathways. Gut dysbiosis-induced inflammation can also disrupt the gut barrier integrity and increase intestinal permeability, which allows gut-derived toxic products to enter the liver and systemic circulation, further triggering oxidative stress, inflammation, and epigenetic alterations associated with metabolic diseases. However, specific gut-derived metabolites, such as short-chain fatty acids (SCFAs), lactate, and vitamins, can modulate oxidative stress and the immune system through epigenetic mechanisms, thereby improving metabolic function. Gut microbiota and diet-induced metabolic diseases, such as obesity, insulin resistance, dyslipidemia, and hypertension, can transfer to the next generation, involving epigenetic mechanisms. In this review, we will introduce the key epigenetic alterations that, along with gut dysbiosis and ROS, are engaged in developing metabolic diseases. Finally, we will discuss potential therapeutic interventions such as dietary modifications, prebiotics, probiotics, postbiotics, and fecal microbiota transplantation, which may reduce oxidative stress and inflammation associated with metabolic syndrome by altering gut microbiota and epigenetic alterations. In summary, this review highlights the crucial role of gut microbiota dysbiosis, oxidative stress, and inflammation in the pathogenesis of metabolic diseases, with a particular focus on epigenetic alterations (including histone modifications, DNA methylomics, and RNA interference) and potential interventions that may prevent or improve metabolic diseases.
Collapse
Affiliation(s)
- Hamid Mostafavi Abdolmaleky
- Nutrition/Metabolism Laboratory, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Jin-Rong Zhou
- Nutrition/Metabolism Laboratory, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
4
|
Gong ZZ, Li T, Yan H, Xu MH, Lian Y, Yang YX, Wei W, Liu T. Exploring the autophagy-related pathogenesis of active ulcerative colitis. World J Clin Cases 2024; 12:1622-1633. [PMID: 38576744 PMCID: PMC10989433 DOI: 10.12998/wjcc.v12.i9.1622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 01/23/2024] [Accepted: 02/27/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND The pathogenesis of ulcerative colitis (UC) is complex, and recent therapeutic advances remain unable to fully alleviate the condition. AIM To inform the development of novel UC treatments, bioinformatics was used to explore the autophagy-related pathogenesis associated with the active phase of UC. METHODS The GEO database was searched for UC-related datasets that included healthy controls who met the screening criteria. Differential analysis was conducted to obtain differentially expressed genes (DEGs). Autophagy-related targets were collected and intersected with the DEGs to identiy differentially expressed autophagy-related genes (DEARGs) associated with active UC. DEARGs were then subjected to KEGG, GO, and DisGeNET disease enrichment analyses using R software. Differential analysis of immune infiltrating cells was performed using the CiberSort algorithm. The least absolute shrinkage and selection operator algorithm and protein-protein interaction network were used to narrow down the DEARGs, and the top five targets in the Dgree ranking were designated as core targets. RESULTS A total of 4822 DEGs were obtained, of which 58 were classified as DEARGs. SERPINA1, BAG3, HSPA5, CASP1, and CX3CL1 were identified as core targets. GO enrichment analysis revealed that DEARGs were primarily enriched in processes related to autophagy regulation and macroautophagy. KEGG enrichment analysis showed that DEARGs were predominantly associated with NOD-like receptor signaling and other signaling pathways. Disease enrichment analysis indicated that DEARGs were significantly linked to diseases such as malignant glioma and middle cerebral artery occlusion. Immune infiltration analysis demonstrated a higher presence of immune cells like activated memory CD4 T cells and follicular helper T cells in active UC patients than in healthy controls. CONCLUSION Autophagy is closely related to the active phase of UC and the potential targets obtained from the analysis in this study may provide new insight into the treatment of active UC patients.
Collapse
Affiliation(s)
- Zhuo-Zhi Gong
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Teng Li
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - He Yan
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Min-Hao Xu
- College of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Beijing 100102, China
| | - Yue Lian
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Yi-Xuan Yang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Wei Wei
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Tao Liu
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| |
Collapse
|
5
|
Martínez-Blanco M, Menchén-Martínez D, Cámara C, López-Fandiño R, Berin MC, Lozano-Ojalvo D. Coculture of Human Dendritic and T Cells for the Study of Specific T Cell-Mediated Responses Against Food Allergens. Methods Mol Biol 2024; 2717:175-190. [PMID: 37737984 DOI: 10.1007/978-1-0716-3453-0_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
Dendritic cells (DCs) connect innate and adaptive immunity by sampling, capturing, processing, and presenting the allergen to distinct subsets of CD4+ T cells. In food allergy, this process leads to the generation of allergen-specific Th2 responses and the production of type 2 cytokines that ultimately induce the synthesis of IgE by allergen-specific B cells. In this chapter, we have described different protocols for the isolation of circulating DCs as well as the generation of DC-like cells derived from autologous peripheral monocytes and the human monocytic THP-1 cell line. Coculture of isolated/generated DCs with CD4+ T cells obtained from PBMCs of allergic subjects allows the study of antigen-specific T cell immune responses against food allergens. Early responses upon allergen recognition can be determined by the upregulation of activation markers such as CD154 (CD40 ligand) and the detection of type 2 cytokines (IL-4, IL-5, IL-9, and IL-13). Delayed allergen-specific CD4+ T cell responses induce the proliferation of these cells and the accumulation of type 2 cytokines in coculture supernatants that can be quantified by different approaches (ELISA, EllaTM, and multiplex assays). Together, the protocols described in this chapter can be used to investigate the features of food proteins to induce food allergy, the influence of environmental factors to generate Th2-polarization, the function of DCs to generate differential immune responses in allergic versus tolerant individuals, and to assess the immunomodulating properties of potential therapeutic substances.
Collapse
Affiliation(s)
- Mónica Martínez-Blanco
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - David Menchén-Martínez
- Instituto de Investigación en Ciencias de la Alimentación (CIAL, CSIC-UAM), Madrid, Spain
| | - Carmen Cámara
- Department of Immunology, Hospital La Paz, Madrid, Spain
| | - Rosina López-Fandiño
- Instituto de Investigación en Ciencias de la Alimentación (CIAL, CSIC-UAM), Madrid, Spain
| | - M Cecilia Berin
- Department of Medicine, Feinberg School of Medicine at Northwestern University, Chicago, IL, USA
| | - Daniel Lozano-Ojalvo
- Instituto de Investigación en Ciencias de la Alimentación (CIAL, CSIC-UAM), Madrid, Spain.
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
6
|
Messerschmidt JL, Azin M, Dempsey KE, Demehri S. TSLP/dendritic cell axis promotes CD4+ T cell tolerance to the gut microbiome. JCI Insight 2023; 8:e160690. [PMID: 37427591 PMCID: PMC10371333 DOI: 10.1172/jci.insight.160690] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/19/2023] [Indexed: 07/11/2023] Open
Abstract
Thymic stromal lymphopoietin (TSLP) overexpression is widely associated with atopy. However, TSLP is expressed in normal barrier organs, suggesting a homeostatic function. To determine the function of TSLP in barrier sites, we investigated the impact of endogenous TSLP signaling on the homeostatic expansion of CD4+ T cells in adult mice. Surprisingly, incoming CD4+ T cells induced lethal colitis in adult Rag1-knockout animals that lacked the TSLP receptor (Rag1KOTslprKO). Endogenous TSLP signaling was required for reduced CD4+ T cell proliferation, Treg differentiation, and homeostatic cytokine production. CD4+ T cell expansion in Rag1KOTslprKO mice was dependent on the gut microbiome. The lethal colitis was rescued by parabiosis between Rag1KOTslprKO and Rag1KO animals and wild-type dendritic cells (DCs) suppressed CD4+ T cell-induced colitis in Rag1KOTslprKO mice. A compromised T cell tolerance was noted in TslprKO adult colon, which was exacerbated by anti-PD-1 and anti-CTLA-4 therapy. These results reveal a critical peripheral tolerance axis between TSLP and DCs in the colon that blocks CD4+ T cell activation against the commensal gut microbiome.
Collapse
|
7
|
Xu K, Cai J, Xing J, Li X, Wu B, Zhu Z, Zhang Z. Broad-spectrum antibiotics associated gut microbiome disturbance impairs T cell immunity and promotes lung cancer metastasis: a retrospective study. BMC Cancer 2022; 22:1182. [DOI: 10.1186/s12885-022-10307-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 11/11/2022] [Indexed: 11/18/2022] Open
Abstract
Abstract
Background
Gut microbiome has been linked to a regulatory role in cancer progression. However, whether broad-spectrum antibiotics (ATB) associated gut microbiome dysbiosis contributes to an impaired T cell immune function, and ultimately promotes lung cancer metastasis is not well known.
Methods
In this study, a retrospective analysis was performed in a cohort of 263 patients initially diagnosed with non-small cell lung cancer (NSCLC) patients, including the ATB group (patients with broad-spectrum antibiotics treatment) (n = 124), and non-ATB group (n = 139) as control. ATB patients were prescribed ATB for over 5 days within 30 days prior to the collection of blood and fecal specimens and followed surgical treatment or first-line therapy. T cell immune function and metastasis-free survival (MFS) were evaluated between the two groups. Gut microbiota was evaluated by 16S rDNA sequencing. The predictive value of T cell immunity for MFS was evaluated by ROC analysis and Cox regression analysis.
Results
Our results suggest that broad-spectrum antibiotics (ATB) impair T cell immune function in patients with either early-stage or advanced NSCLC, which likely contribute to the promotion of lung cancer metastasis. Results of the survival analysis show that metastasis-free survival (MFS) is significantly shorter in the ATB patients than that in the non-ATB patients with stage III NSCLC. The 16S rDNA sequencing shows that ATB administration contributes to a significant dysbiosis of the composition and diversity of gut microbiota. Moreover, ROC analysis results of CD4 (AUC 0.642, p = 0.011), CD8 (AUC was 0.729, p < 0.001), CD16 + 56 + (AUC 0.643, p = 0.003), and the combination of CD4, CD8 and CD16 + 56+ (AUC 0.810, p < 0.001), or Cox regression analysis results of CD4 (HR 0.206, p < 0.001), CD8 (HR 0.555, p = 0.009), which is likely regulated by ATB administration, have significantly predictive values for MFS.
Conclusion
These results provide evidence of gut microbiome disturbance due to ATB administration is involved in the regulation of T cell immunity, and their predictive value for the tumor metastasis in lung cancer patients. Thus, gut microbiota may serve as a therapeutic target for lung cancer. Consequently, caution should be exercised before the long-term administration of broad-spectrum antibiotics in cancer patients.
Collapse
|
8
|
Cerutti A, Filipska M, Fa XM, Tachó-Piñot R. Impact of the mucosal milieu on antibody responses to allergens. J Allergy Clin Immunol 2022; 150:503-512. [PMID: 36075636 DOI: 10.1016/j.jaci.2022.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/30/2022]
Abstract
Respiratory and digestive mucosal surfaces are continually exposed to common environmental antigens, which include potential allergens. Although innocuous in healthy individuals, allergens cause allergy in predisposed subjects and do so by triggering a pathologic TH2 cell response that induces IgE class switching and somatic hypermutation in allergen-specific B cells. The ensuing affinity maturation and plasma cell differentiation lead to the abnormal release of high-affinity IgE that binds to powerful FcεRI receptors on basophils and mast cells. When cross-linked by allergen, FcεRI-bound IgE instigates the release of prestored and de novo-induced proinflammatory mediators. Aside from causing type I hypersensitivity reactions underlying allergy, IgE affords protection against nematodes or venoms from insects and snakes, which raises questions as to the fundamental differences between protective and pathogenic IgE responses. In this review, we discuss the impact of the mucosal environment, including the epithelial and mucus barriers, on the induction of protective IgE responses against environmental antigens. We further discuss how perturbations of these barriers may contribute to the induction of pathogenic IgE production.
Collapse
Affiliation(s)
- Andrea Cerutti
- Catalan Institute for Research and Advanced Studies, Barcelona, Spain; Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona Biomedical Research Park, Barcelona, Spain; Division of Clinical Immunology, Department of Medicine, Mount Sinai School of Medicine, New York.
| | - Martyna Filipska
- Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona Biomedical Research Park, Barcelona, Spain
| | - Xavi Marcos Fa
- Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona Biomedical Research Park, Barcelona, Spain
| | - Roser Tachó-Piñot
- Lydia Becher Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
9
|
Rastall RA, Diez-Municio M, Forssten SD, Hamaker B, Meynier A, Moreno FJ, Respondek F, Stah B, Venema K, Wiese M. Structure and function of non-digestible carbohydrates in the gut microbiome. Benef Microbes 2022; 13:95-168. [PMID: 35729770 DOI: 10.3920/bm2021.0090] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Together with proteins and fats, carbohydrates are one of the macronutrients in the human diet. Digestible carbohydrates, such as starch, starch-based products, sucrose, lactose, glucose and some sugar alcohols and unusual (and fairly rare) α-linked glucans, directly provide us with energy while other carbohydrates including high molecular weight polysaccharides, mainly from plant cell walls, provide us with dietary fibre. Carbohydrates which are efficiently digested in the small intestine are not available in appreciable quantities to act as substrates for gut bacteria. Some oligo- and polysaccharides, many of which are also dietary fibres, are resistant to digestion in the small intestines and enter the colon where they provide substrates for the complex bacterial ecosystem that resides there. This review will focus on these non-digestible carbohydrates (NDC) and examine their impact on the gut microbiota and their physiological impact. Of particular focus will be the potential of non-digestible carbohydrates to act as prebiotics, but the review will also evaluate direct effects of NDC on human cells and systems.
Collapse
Affiliation(s)
- R A Rastall
- Department of Food and Nutritional Sciences, The University of Reading, P.O. Box 226, Whiteknights, Reading, RG6 6AP, United Kingdom
| | - M Diez-Municio
- Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC-UAM), CEI (UAM+CSIC), Nicolás Cabrera 9, 28049 Madrid, Spain
| | - S D Forssten
- IFF Health & Biosciences, Sokeritehtaantie 20, 02460 Kantvik, Finland
| | - B Hamaker
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, 745 Agriculture Mall Drive, West Lafayette, IN 47907-2009, USA
| | - A Meynier
- Nutrition Research, Mondelez France R&D SAS, 6 rue René Razel, 91400 Saclay, France
| | - F Javier Moreno
- Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC-UAM), CEI (UAM+CSIC), Nicolás Cabrera 9, 28049 Madrid, Spain
| | - F Respondek
- Tereos, Zoning Industriel Portuaire, 67390 Marckolsheim, France
| | - B Stah
- Human Milk Research & Analytical Science, Danone Nutricia Research, Uppsalalaan 12, 3584 CT Utrecht, the Netherlands.,Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, the Netherlands
| | - K Venema
- Centre for Healthy Eating & Food Innovation (HEFI), Maastricht University - campus Venlo, St. Jansweg 20, 5928 RC Venlo, the Netherlands
| | - M Wiese
- Department of Microbiology and Systems Biology, TNO, Utrechtseweg 48, 3704 HE, Zeist, the Netherlands
| |
Collapse
|
10
|
Frutos-Rincón L, Gómez-Sánchez JA, Íñigo-Portugués A, Acosta MC, Gallar J. An Experimental Model of Neuro-Immune Interactions in the Eye: Corneal Sensory Nerves and Resident Dendritic Cells. Int J Mol Sci 2022; 23:ijms23062997. [PMID: 35328417 PMCID: PMC8951464 DOI: 10.3390/ijms23062997] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 02/28/2022] [Accepted: 03/04/2022] [Indexed: 12/04/2022] Open
Abstract
The cornea is an avascular connective tissue that is crucial, not only as the primary barrier of the eye but also as a proper transparent refractive structure. Corneal transparency is necessary for vision and is the result of several factors, including its highly organized structure, the physiology of its few cellular components, the lack of myelinated nerves (although it is extremely innervated), the tightly controlled hydration state, and the absence of blood and lymphatic vessels in healthy conditions, among others. The avascular, immune-privileged tissue of the cornea is an ideal model to study the interactions between its well-characterized and dense sensory nerves (easily accessible for both focal electrophysiological recording and morphological studies) and the low number of resident immune cell types, distinguished from those cells migrating from blood vessels. This paper presents an overview of the corneal structure and innervation, the resident dendritic cell (DC) subpopulations present in the cornea, their distribution in relation to corneal nerves, and their role in ocular inflammatory diseases. A mouse model in which sensory axons are constitutively labeled with tdTomato and DCs with green fluorescent protein (GFP) allows further analysis of the neuro-immune crosstalk under inflammatory and steady-state conditions of the eye.
Collapse
Affiliation(s)
- Laura Frutos-Rincón
- Instituto de Neurociencias, Universidad Miguel Hernández—Consejo Superior de Investigaciones Científicas, 03550 San Juan de Alicante, Spain; (L.F.-R.); (A.Í.-P.); (M.C.A.); (J.G.)
- The European University of Brain and Technology-NeurotechEU, 03550 San Juan de Alicante, Spain
| | - José Antonio Gómez-Sánchez
- Instituto de Neurociencias, Universidad Miguel Hernández—Consejo Superior de Investigaciones Científicas, 03550 San Juan de Alicante, Spain; (L.F.-R.); (A.Í.-P.); (M.C.A.); (J.G.)
- Correspondence: ; Tel.: +34-965-91-9594
| | - Almudena Íñigo-Portugués
- Instituto de Neurociencias, Universidad Miguel Hernández—Consejo Superior de Investigaciones Científicas, 03550 San Juan de Alicante, Spain; (L.F.-R.); (A.Í.-P.); (M.C.A.); (J.G.)
| | - M. Carmen Acosta
- Instituto de Neurociencias, Universidad Miguel Hernández—Consejo Superior de Investigaciones Científicas, 03550 San Juan de Alicante, Spain; (L.F.-R.); (A.Í.-P.); (M.C.A.); (J.G.)
- The European University of Brain and Technology-NeurotechEU, 03550 San Juan de Alicante, Spain
| | - Juana Gallar
- Instituto de Neurociencias, Universidad Miguel Hernández—Consejo Superior de Investigaciones Científicas, 03550 San Juan de Alicante, Spain; (L.F.-R.); (A.Í.-P.); (M.C.A.); (J.G.)
- The European University of Brain and Technology-NeurotechEU, 03550 San Juan de Alicante, Spain
- Instituto de Investigación Biomédica y Sanitaria de Alicante, 03010 Alicante, Spain
| |
Collapse
|
11
|
Budiselic J, Sadlek M, Simpson K. Severity of Psychologic Stressors Reflects Course of Crohn's Disease in Two Siblings. Cureus 2021; 13:e16533. [PMID: 34430142 PMCID: PMC8378317 DOI: 10.7759/cureus.16533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2021] [Indexed: 11/05/2022] Open
Abstract
A significant rise in Crohn's disease (CD) cases amongst first-degree relatives strongly suggests that inflammatory bowel disease (IBD) has a genetic component. Adherence to medical management is at the forefront for preventing disease relapse. However, given the role that stress places on the immune system, it is imperative to implore an individual's psychologic stressors to prevent future disease complications. This case of CD in two siblings, male and female, highlights the unique stress experienced by each patient at the time of symptom onset, the severity of their inflammatory symptoms, and their course of disease over several years. The male patient suffered from gender dysphoria and social anxiety for over a decade and had more chronic stress and severe complications of his disease. On the other hand, the stressors faced by his sister were periodic in nature and symptoms resided once stressful periods ended. For select patients, it is indicative that referral for psychotherapy should be considered as an ongoing mainstay of management. This case is intended to highlight the need for including psychotherapy in addition to medical management in order to treat IBD holistically.
Collapse
Affiliation(s)
| | - Mary Sadlek
- Family Medicine, Community Regional Medical Center, Fresno, USA
| | - Keon Simpson
- Clinical Sciences, Saint James School of Medicine, The Quarter, AIA
| |
Collapse
|
12
|
Lerner A. The intestinal luminal sources of α-synuclein: a gastroenterologist perspective. Nutr Rev 2021; 80:282-293. [PMID: 33942062 DOI: 10.1093/nutrit/nuab024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease is characterized by nonmotor/motor dysfunction, midbrain dopaminergic neuronal death, and α-synuclein (aSN) deposits. The current hypothesis is that aSN accumulates in the enteric nervous system to reach the brain. However, invertebrate, vertebrate, and nutritional sources of aSN reach the luminal compartment. Submitted to local amyloidogenic forces, the oligomerized proteins' cargo can be sensed and sampled by a specialized mucosal cell to be transmitted to the adjacent enteric nervous system, starting their upward journey to the brain. The present narrative review extends the current mucosal origin of Parkinson's disease, presenting the possibility that the disease starts in the intestinal lumen. If substantiated, eliminating the nutritional sources of aSN (eg, applying a vegetarian diet) might revolutionize the currently used dopaminergic pharmacologic therapy.
Collapse
Affiliation(s)
- Aaron Lerner
- A. Lerner is with the Zabludowicz Center for Autoimmune Diseases, Chaim Sheba Medical Center, Tel-Hashomer, Israel
| |
Collapse
|
13
|
Aragona P, Baudouin C, Benitez Del Castillo JM, Messmer E, Barabino S, Merayo-Lloves J, Brignole-Baudouin F, Inferrera L, Rolando M, Mencucci R, Rescigno M, Bonini S, Labetoulle M. The ocular microbiome and microbiota and their effects on ocular surface pathophysiology and disorders. Surv Ophthalmol 2021; 66:907-925. [PMID: 33819460 DOI: 10.1016/j.survophthal.2021.03.010] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 12/17/2022]
Abstract
The ocular surface flora perform an important role in the defense mechanisms of the ocular surface system. Its regulation of the immunological activity and the barrier effect against pathogen invasion are remarkable. Composition of the flora differs according to the methods of investigation, because the microbiome, composed of the genetic material of bacteria, fungi, viruses, protozoa, and eukaryotes on the ocular surface, differs from the microbiota, which are the community of microorganisms that colonize the ocular surface. The observed composition of the ocular surface flora depends on harvesting and examining methods, whether with traditional culture or with more refined genetic analysis based on rRNA and DNA sequencing. Environment, diet, sex, and age influence the microbial flora composition, thus complicating the analysis of the baseline status. Moreover, potentially pathogenic organisms can affect its composition, as do various disorders, including chronic inflammation, and therapies applied to the ocular surface. A better understanding of the composition and function of microbial communities at the ocular surface could bring new insights and clarify the epidemiology and pathology of ocular surface dynamics in health and disease. The purpose of this review is to provide an up-to-date overview of knowledge about this topic.
Collapse
Affiliation(s)
- Pasquale Aragona
- Department of Biomedical Sciences, Ophthalmology Clinic, University of Messina, Messina, Italy.
| | - Christophe Baudouin
- Quinze-Vingts National Eye Hospital, IHU ForeSight, Paris Saclay University, Paris, France
| | - Jose M Benitez Del Castillo
- Departamento de Oftalmología, Hospital Clínico San Carlos, Clínica Rementeria, Instituto Investigaciones Oftalmologicas Ramon Castroviejo, Universidad Complutense, Madrid, Spain
| | - Elisabeth Messmer
- Department of Ophthalmology, Ludwig-Maximilians-University, Munich, Germany
| | - Stefano Barabino
- Ocular Surface and Dry Eye Center, Ospedale L. Sacco, University of Milan, Milan, Italy
| | - Jesus Merayo-Lloves
- Instituto Universitario Fernández-Vega, Universidad de Oviedo, Oviedo, Spain
| | - Francoise Brignole-Baudouin
- Sorbonne Université, INSERM UMR_S968, CNRS UMR7210, Institut de la Vision, Paris, France; CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, Laboratoire de Biologie Médicale, Paris, France; Université de Paris, Faculté de Pharmacie de Paris, Département de Chimie-Toxicologie Analytique et Cellulaire, Paris, France
| | - Leandro Inferrera
- Department of Biomedical Sciences, Ophthalmology Clinic, University of Messina, Messina, Italy
| | - Maurizio Rolando
- Ocular Surface and Dry Eye Center, ISPRE Ophthalmics, Genoa, Italy
| | - Rita Mencucci
- Department of Neuroscience, Psychology, Pharmacology and Child Health (NEUROFARBA), Eye Clinic, University of Florence, Florence, Italy
| | - Maria Rescigno
- Humanitas Clinical and Research Center - IRCCS, Humanitas University Department of Biomedical Sciences, Milan, Italy
| | - Stefano Bonini
- Department of Ophthalmology, University of Rome Campus Biomedico, Rome, Italy
| | - Marc Labetoulle
- Ophthalmology Départment, Hôpitaux Universitaires Paris-Sud, APHP, Université Paris-Saclay, IDMIT Infrastructure, Fontenay-aux-Roses Cedex, France
| |
Collapse
|
14
|
de Carvalho MV, Gonçalves-de-Albuquerque CF, Silva AR. PPAR Gamma: From Definition to Molecular Targets and Therapy of Lung Diseases. Int J Mol Sci 2021; 22:E805. [PMID: 33467433 PMCID: PMC7830538 DOI: 10.3390/ijms22020805] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/18/2020] [Accepted: 09/24/2020] [Indexed: 12/15/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear receptor superfamily that regulate the expression of genes related to lipid and glucose metabolism and inflammation. There are three members: PPARα, PPARβ or PPARγ. PPARγ have several ligands. The natural agonists are omega 9, curcumin, eicosanoids and others. Among the synthetic ligands, we highlight the thiazolidinediones, clinically used as an antidiabetic. Many of these studies involve natural or synthetic products in different pathologies. The mechanisms that regulate PPARγ involve post-translational modifications, such as phosphorylation, sumoylation and ubiquitination, among others. It is known that anti-inflammatory mechanisms involve the inhibition of other transcription factors, such as nuclear factor kB(NFκB), signal transducer and activator of transcription (STAT) or activator protein 1 (AP-1), or intracellular signaling proteins such as mitogen-activated protein (MAP) kinases. PPARγ transrepresses other transcription factors and consequently inhibits gene expression of inflammatory mediators, known as biomarkers for morbidity and mortality, leading to control of the exacerbated inflammation that occurs, for instance, in lung injury/acute respiratory distress. Many studies have shown the therapeutic potentials of PPARγ on pulmonary diseases. Herein, we describe activities of the PPARγ as a modulator of inflammation, focusing on lung injury and including definition and mechanisms of regulation, biological effects and molecular targets, and its role in lung diseases caused by inflammatory stimuli, bacteria and virus, and molecular-based therapy.
Collapse
Affiliation(s)
- Márcia V. de Carvalho
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro 21040-900, Brazil;
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro 21040-900, Brazil
| | - Cassiano F. Gonçalves-de-Albuquerque
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro 21040-900, Brazil;
- Laboratório de Imunofarmacologia, Universidade Federal do Estado do Rio de Janeiro (UNIRIO), Rio de Janeiro 20211-010, Brazil
- Programa de Pós-Graduação em Biologia Molecular e Celular, Universidade Federal do Estado do Rio de Janeiro (UNIRIO), Rio de Janeiro 20211-010, Brazil
| | - Adriana R. Silva
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro 21040-900, Brazil;
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro 21040-900, Brazil
| |
Collapse
|
15
|
Oliverio GW, Spinella R, Postorino EI, Inferrera L, Aragona E, Aragona P. Safety and Tolerability of an Eye Drop Based on 0.6% Povidone-Iodine Nanoemulsion in Dry Eye Patients. J Ocul Pharmacol Ther 2020; 37:90-96. [PMID: 33370213 PMCID: PMC7984652 DOI: 10.1089/jop.2020.0085] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Purpose: To evaluate safety and tolerability on the ocular surface of an anti-septic formulation containing 0.6% povidone–iodine (0.6% PVI) for a 4 week period. Methods: An observational, prospective study included 20 mild-moderate dry eye disease (DED) patients who enrolled at the Ocular Surface Disease Unit of the University of Messina, receiving 0.6% PVI eye drops for 28 days, 2 drops twice daily (BID). The assessment included the Ocular Surface Disease Index questionnaire; symptoms score (0 = absent to 3 = severe) for burning, ocular dryness, foreign body sensation, watery eyes, tearing, photophobia, and ocular pain; fluorescein tear break-up time (TBUT); and corneal-conjunctival staining, performed at baseline (T0), after 7 (T7) and 28 (T28). Schirmer I-test, corneal endothelial cell count, intraocular pressure, and fundus examination were performed at T0 and T28. The main outcome measures were TBUT and corneal-conjunctival staining as markers of ocular surface homeostasis. For statistical analysis, Student's T-test and Wilcoxon test were used as appropriate. Results: No significant alterations of the safety parameters were found throughout the study. Further, at T28 a significant improvement of burning, ocular dryness, foreign body sensation, and watery eyes (T0 vs. T28 P < 0.03) were observed; corneal-conjunctival staining improved at T28 (T0 vs. T28 P < 0.0001), and TBUT improved already at T7 (T0 vs. T7 P = 0.0008) lasting so till the end of the study. The only adverse event was mild burning at instillation for the first 3 days of treatment in most of the patients. Conclusions: The treatment with 0.6% PVI was safe and well tolerated in a group of patients with a damaged ocular surface.
Collapse
Affiliation(s)
| | - Rosaria Spinella
- Department of Biomedical Sciences, Ophthalmology Clinic, University of Messina, Messina, Italy
| | - Elisa Imelde Postorino
- Department of Biomedical Sciences, Ophthalmology Clinic, University of Messina, Messina, Italy
| | - Leandro Inferrera
- Department of Biomedical Sciences, Ophthalmology Clinic, University of Messina, Messina, Italy
| | - Emanuela Aragona
- Department of Ophthalmology, Vita-Salute University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Pasquale Aragona
- Department of Biomedical Sciences, Ophthalmology Clinic, University of Messina, Messina, Italy
| |
Collapse
|
16
|
Rinaldi E, Consonni A, Cordiglieri C, Sacco G, Crasà C, Fontana A, Morelli L, Elli M, Mantegazza R, Baggi F. Therapeutic Effect of Bifidobacterium Administration on Experimental Autoimmune Myasthenia Gravis in Lewis Rats. Front Immunol 2019; 10:2949. [PMID: 31956324 PMCID: PMC6951413 DOI: 10.3389/fimmu.2019.02949] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 12/02/2019] [Indexed: 12/13/2022] Open
Abstract
Beneficial effects of probiotics on gut microbiota homeostasis and inflammatory immune responses suggested the investigation of their potential clinical efficacy in experimental models of autoimmune diseases. Indeed, administration of two bifidobacteria and lactobacilli probiotic strains prevented disease manifestations in the Lewis rat model of Myasthenia Gravis (EAMG). Here, we demonstrate the clinical efficacy of therapeutic administration of vital bifidobacteria (i.e., from EAMG onset). The mechanisms involved in immunomodulation were investigated with ex vivo and in vitro experiments. Improvement of EAMG symptoms was associated to decreased anti-rat AChR antibody levels, and differential expression of TGFβ and FoxP3 immunoregulatory transcripts in draining lymph nodes and spleen of treated-EAMG rats. Exposure of rat bone marrow-derived dendritic cells to bifidobacteria or lactobacilli strains upregulated toll-like receptor 2 mRNA expression, a key molecule involved in bacterium recognition via lipotheicoic acid. Live imaging experiments of AChR-specific effector T cells, co-cultured with BMDCs pre-exposed to bifidobacteria, demonstrated increased percentages of motile effector T cells, suggesting a hindered formation of TCR-peptide-MHC complex. Composition of gut microbiota was studied by 16S rRNA gene sequencing, and α and β diversity were determined in probiotic treated EAMG rats, with altered ratios between Tenericutes and Verrucomicrobia (phylum level), and Ruminococcaceae and Lachnospiraceae (family level). Moreover, the relative abundance of Akkermansia genus was found increased compared to healthy and probiotic treated EAMG rats. In conclusion, our findings confirms that the administration of vital bifidobacteria at EAMG onset has beneficial effects on disease progression; this study further supports preclinical research in human MG to evaluate probiotic efficacy as supplementary therapy in MG.
Collapse
Affiliation(s)
- Elena Rinaldi
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Alessandra Consonni
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Chiara Cordiglieri
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Grazia Sacco
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Camilla Crasà
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Alessandra Fontana
- Department for Sustainable Food Process, Università Cattolica del Sacro Cuore, Piacenza, Italy
| | - Lorenzo Morelli
- Department for Sustainable Food Process, Università Cattolica del Sacro Cuore, Piacenza, Italy
| | - Marina Elli
- AAT-Advanced Analytical Technologies, Fiorenzuola d'Arda, Italy
| | - Renato Mantegazza
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Fulvio Baggi
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
17
|
Nešić A, Stam A, Čavić M, Ten Klooster JP, Pieters R, Smit J, Gavrović-Jankulović M. Activation of epithelial cells by the major kiwifruit allergen Act d 1 in human and mouse-derived intestinal model. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.103556] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
18
|
Chen W, Ma L, Li R, Huang S, Xie R, Chen Y, Zhao B, Fei J, Qu H, Chen H, Mao E, Chen EZ. DC-SIGN Expression in Intestinal Epithelial Cells Regulates Sepsis-Associated Acute Intestinal Injury Via Activating ERK1/2-NF-κB/P65 Signaling. Shock 2019; 52:434-442. [PMID: 30335674 DOI: 10.1097/shk.0000000000001277] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVE The aim of the study was to investigate the role of dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin (DC-SIGN) in intestinal epithelial cells (IECs) in regulating sepsis-induced acute intestinal injury and systemic inflammatory response. METHODS To induce sepsis condition, Male C57BL/6 mice were exposed to cecal ligation and puncture (CLP) in vivo, whereas a normal human IECs line (FHs74Int) was stimulated with lipopolysaccharide (LPS) in vitro. DC-SIGN siRNA pretreatment was used to knock down DC-SIGN expression both in vivo and in vitro. The expression of DC-SIGN was detected by western blot and immunohistochemistry. The expression of total and phosphorylation of ERK1/2 and NF-κB/p65 was examined by western blot. The levels of cytokines in serum and culture supernatant were measured by ELISA. The survival rate and organ injures of septic mice were also assessed. RESULTS In vivo, DC-SIGN expression in mouse IECs was time-dependently upregulated by CLP. CLP-induced phosphorylation of ERK1/2 and NF-κB/p65 was effectively inhibited by DC-SIGN siRNA pretreatment, leading to the decrease of systemic inflammatory cytokines (TNF-α, IL-1β, IL-6, IL-10, and IFN-γ), which alleviated multiple organ injuries and increased the survival rate of septic mice. In vitro, DC-SIGN expression in FHs74Int was significantly upregulated by LPS stimulation in a time- and dose-dependent manner. DC-SIGN knockdown abolished LPS-induced ERK1/2 and NF-κB/p65 phosphorylation, resulting in the decrease of cytokines release by FHs74Int. CONCLUSIONS Sepsis-induced DC-SIGN expression in IECs plays a significant role in regulating acute intestinal injury and systemic inflammatory response. The inhibition of DC-SIGN exhibited protective effects on sepsis-associated organ injury and systemic inflammation.
Collapse
Affiliation(s)
- Weiwei Chen
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Li Ma
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ranran Li
- Department of Critical Care Medicine, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shunwei Huang
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Rongli Xie
- Department of Surgery, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ying Chen
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Bing Zhao
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jian Fei
- Department of Surgery, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hongping Qu
- Department of Critical Care Medicine, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hao Chen
- Department of Surgery, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Enqiang Mao
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Er-Zhen Chen
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
19
|
Hawiger J, Zienkiewicz J. Decoding inflammation, its causes, genomic responses, and emerging countermeasures. Scand J Immunol 2019; 90:e12812. [PMID: 31378956 PMCID: PMC6883124 DOI: 10.1111/sji.12812] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/03/2019] [Accepted: 07/29/2019] [Indexed: 12/11/2022]
Abstract
Inflammation is the mechanism of diseases caused by microbial, autoimmune, allergic, metabolic and physical insults that produce distinct types of inflammatory responses. This aetiologic view of inflammation informs its classification based on a cause‐dependent mechanism as well as a cause‐directed therapy and prevention. The genomic era ushered in a new understanding of inflammation by highlighting the cell's nucleus as the centre of the inflammatory response. Exogenous or endogenous inflammatory insults evoke genomic responses in immune and non‐immune cells. These genomic responses depend on transcription factors, which switch on and off a myriad of inflammatory genes through their regulatory networks. We discuss the transcriptional paradigm of inflammation based on denying transcription factors’ access to the nucleus. We present two approaches that control proinflammatory signalling to the nucleus. The first approach constitutes a novel intracellular protein therapy with bioengineered physiologic suppressors of cytokine signalling. The second approach entails control of proinflammatory transcriptional cascades by targeting nuclear transport with a cell‐penetrating peptide that inhibits the expression of 23 out of the 26 mediators of inflammation along with the nine genes required for metabolic responses. We compare these emerging anti‐inflammatory countermeasures to current therapies. The transcriptional paradigm of inflammation offers nucleocentric strategies for microbial, autoimmune, metabolic, physical and other types of inflammation afflicting millions of people worldwide.
Collapse
Affiliation(s)
- Jacek Hawiger
- Immunotherapy Program at Vanderbilt University School of Medicine, Nashville, TN, USA.,Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA.,Department of Veterans Affairs, Tennessee Valley Health Care System, Nashville, TN, USA.,Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jozef Zienkiewicz
- Immunotherapy Program at Vanderbilt University School of Medicine, Nashville, TN, USA.,Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA.,Department of Veterans Affairs, Tennessee Valley Health Care System, Nashville, TN, USA
| |
Collapse
|
20
|
Xiao Y, de Paiva CS, Yu Z, de Souza RG, Li DQ, Pflugfelder SC. Goblet cell-produced retinoic acid suppresses CD86 expression and IL-12 production in bone marrow-derived cells. Int Immunol 2019; 30:457-470. [PMID: 30010888 DOI: 10.1093/intimm/dxy045] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 07/09/2018] [Indexed: 12/22/2022] Open
Abstract
Conjunctival goblet cell loss in ocular surface diseases is accompanied by increased number of interleukin-12 (IL-12)-producing antigen-presenting cells (APCs) and increased interferon-γ (IFN-γ) expression. This study tested the hypothesis that mouse conjunctival goblet cells produce biologically active retinoic acid (RA) that suppresses CD86 expression and IL-12 production by myeloid cells. We found that conditioned media from cultured conjunctival goblet cells (CjCM) suppressed stimulated CD86 expression, NF-κB p65 activation and IL-12 and IFN-γ production in unstimulated and lipopolysaccharide-stimulated cultured bone marrow-derived cells (BMDCs) containing a mixed population of APCs. Goblet cell-conditioned, ovalbumin-loaded APCs suppressed IFN-γ production and increased IL-13 production in co-cultured OTII cells. The goblet cell suppressive activity is due in part to their ability to synthesize RA from retinol. Conjunctival goblet cells had greater expression of aldehyde dehydrogenases Aldh1a1 and a3 and ALDEFLUOR activity than cornea epithelium lacking goblet cells. The conditioning activity was lost in goblet cells treated with an ALDH inhibitor, and a retinoid receptor alpha antagonist blocked the suppressive effects of CjCM on IL-12 production. Similar to RA, CjCM increased expression of suppressor of cytokine signaling 3 (SOCS3) in BMDCs. SOCS3 silencing reversed the IL-12-suppressive effects of CjCM. Our findings indicate that conjunctival goblet cells are capable of synthesizing RA from retinol secreted by the lacrimal gland into tears that can condition APCs. Evidence suggests goblet cell RA may function in maintaining conjunctival immune tolerance and loss of conjunctival goblet cells may contribute to increased Th1 priming in dry eye.
Collapse
Affiliation(s)
- Yangyan Xiao
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA.,Second Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China
| | - Cintia S de Paiva
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| | - Zhiyuan Yu
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| | - Rodrigo G de Souza
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| | - De-Quan Li
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| | - Stephen C Pflugfelder
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
21
|
Molero-Abraham M, Sanchez-Trincado JL, Gomez-Perosanz M, Torres-Gomez A, Subiza JL, Lafuente EM, Reche PA. Human Oral Epithelial Cells Impair Bacteria-Mediated Maturation of Dendritic Cells and Render T Cells Unresponsive to Stimulation. Front Immunol 2019; 10:1434. [PMID: 31316504 PMCID: PMC6611079 DOI: 10.3389/fimmu.2019.01434] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 06/07/2019] [Indexed: 01/03/2023] Open
Abstract
The oral mucosa is a first line of defense against pathogenic organisms and yet tolerates food antigens and resident bacteria. Mucosal epithelial cells are emerging as important regulators of innate and adaptive immune responses. However, the contribution of oral epithelial cells (OECs) determining oral immunity is understudied. Here, we evaluated the ability of H413 and TR146 cells, two OEC lines derived from human oral squamous cell carcinomas, and primary OECs to modulate immune responses to a cocktail of Gram+ and Gram− bacteria known as MV130. OECs expressed CD40 constitutively and class II major histocompatibility complex (MHC II) molecules when stimulated with IFNγ, but not CD80 or CD86. Dendritic cells (DCs) treated with bacteria in co-culture with OECs did not fully mature, as judged by the expression of MHC II, CD80 and CD86, and barely released IL-12 and TNFα, compared to control DCs. Furthermore, in the presence of OECs, DCs were unable to stimulate allogenic naive CD4 T cells to produce IFNγ and TNFα. Similarly, OECs in culture with total CD4 T cells or Th1 cells stimulated with anti-CD3 and anti-CD28 antibodies abrogated CD25 and CD69 expression, T cell proliferation and the release of IFNγ and TNFα. The inhibition on T cell activation by OECs was cell-contact dependent, TGFβ independent and largely irreversible. Overall, this behavior of OECs is likely key to avoid immune system over-reaction against resident bacteria.
Collapse
Affiliation(s)
| | - Jose L Sanchez-Trincado
- Department of Immunology, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Marta Gomez-Perosanz
- Department of Immunology, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Alvaro Torres-Gomez
- Department of Immunology, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | | | - Esther M Lafuente
- Department of Immunology, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Pedro A Reche
- Department of Immunology, School of Medicine, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
22
|
Doonan J, Tarafdar A, Pineda MA, Lumb FE, Crowe J, Khan AM, Hoskisson PA, Harnett MM, Harnett W. The parasitic worm product ES-62 normalises the gut microbiota bone marrow axis in inflammatory arthritis. Nat Commun 2019; 10:1554. [PMID: 30952846 PMCID: PMC6451002 DOI: 10.1038/s41467-019-09361-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 03/07/2019] [Indexed: 12/11/2022] Open
Abstract
The human immune system has evolved in the context of our colonisation by bacteria, viruses, fungi and parasitic helminths. Reflecting this, the rapid eradication of pathogens appears to have resulted in reduced microbiome diversity and generation of chronically activated immune systems, presaging the recent rise of allergic, autoimmune and metabolic disorders. Certainly, gastrointestinal helminths can protect against gut and lung mucosa inflammatory conditions by modulating the microbiome and suppressing the chronic inflammation associated with dysbiosis. Here, we employ ES-62, an immunomodulator secreted by tissue-dwelling Acanthocheilonema viteae to show that helminth-modulation of the gut microbiome does not require live infection with gastrointestinal-based worms nor is protection restricted to mucosal diseases. Specifically, subcutaneous administration of this defined immunomodulator affords protection against joint disease in collagen-induced arthritis, a mouse model of rheumatoid arthritis, which is associated with normalisation of gut microbiota and prevention of loss of intestinal barrier integrity.
Collapse
Affiliation(s)
- James Doonan
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK
| | - Anuradha Tarafdar
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, G12 8TA, UK
| | - Miguel A Pineda
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, G12 8TA, UK
| | - Felicity E Lumb
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK
| | - Jenny Crowe
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, G12 8TA, UK
| | - Aneesah M Khan
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, G12 8TA, UK
| | - Paul A Hoskisson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK
| | - Margaret M Harnett
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, G12 8TA, UK.
| | - William Harnett
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK.
| |
Collapse
|
23
|
Oral administration of a mixture of probiotics protects against food allergy via induction of CD103+ dendritic cells and modulates the intestinal microbiota. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.02.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
24
|
Patente TA, Pinho MP, Oliveira AA, Evangelista GCM, Bergami-Santos PC, Barbuto JAM. Human Dendritic Cells: Their Heterogeneity and Clinical Application Potential in Cancer Immunotherapy. Front Immunol 2019; 9:3176. [PMID: 30719026 PMCID: PMC6348254 DOI: 10.3389/fimmu.2018.03176] [Citation(s) in RCA: 271] [Impact Index Per Article: 45.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 12/24/2018] [Indexed: 12/13/2022] Open
Abstract
Dendritic cells (DC) are professional antigen presenting cells, uniquely able to induce naïve T cell activation and effector differentiation. They are, likewise, involved in the induction and maintenance of immune tolerance in homeostatic conditions. Their phenotypic and functional heterogeneity points to their great plasticity and ability to modulate, according to their microenvironment, the acquired immune response and, at the same time, makes their precise classification complex and frequently subject to reviews and improvement. This review will present general aspects of the DC physiology and classification and will address their potential and actual uses in the management of human disease, more specifically cancer, as therapeutic and monitoring tools. New combination treatments with the participation of DC will be also discussed.
Collapse
Affiliation(s)
- Thiago A Patente
- Laboratory of Tumor Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Mariana P Pinho
- Laboratory of Tumor Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Aline A Oliveira
- Laboratory of Tumor Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Gabriela C M Evangelista
- Laboratory of Tumor Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Patrícia C Bergami-Santos
- Laboratory of Tumor Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - José A M Barbuto
- Laboratory of Tumor Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Discipline of Molecular Medicine, Department of Medicine, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
25
|
Mangalam A, Murray J. Microbial monotherapy with Prevotella histicola for patients with multiple sclerosis. Expert Rev Neurother 2019; 19:45-53. [PMID: 30513004 PMCID: PMC6548683 DOI: 10.1080/14737175.2019.1555473] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 11/30/2018] [Indexed: 02/06/2023]
Abstract
Introduction: The gut microbiome helps to maintain a person's healthy state while perturbations in its function often leading to the development of inflammatory diseases including multiple sclerosis (MS). Consequently, gut-commensals which restore homeostasis have the potential to become novel therapeutic options for treating MS. MS patients have presented gut dysbiosis with a reduction in bacteria belonging to the Prevotella genus. Notably, increased levels of Prevotella are observed when disease-modifying therapies are used. Additionally, Prevotella histicola, an anaerobic bacterium derived from the human, can suppress disease in mice with experimental autoimmune encephalomyelitis, a preclinical MS model. Areas covered: This review compares MS microbiome studies from different geographical regions to identify common gut bacteria. Literature on the potential use of P. histicola as a therapy for MS and the next steps for developing microbial monotherapies in MS is also discussed. Expert commentary: Recent findings presenting an inverse correlation between Prevotella and MS disease severity and ability of P. histicola to suppress disease in preclinical models suggest that P. histicola might provide an additional treatment option for MS patients. However, rigorous testing in well-designed control trials should be performed to determine the safety and efficacy P. histicola in MS patients.
Collapse
Affiliation(s)
- Ashutosh Mangalam
- Interdisciplinary graduate program in Immunology, University of Iowa
- Department of Pathology, Carver College of Medicine University of Iowa, Iowa City, IA-52242
| | - Joseph Murray
- Department of Immunology, Mayo Clinic, Rochester, MN 55901
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55901
| |
Collapse
|
26
|
Lépine AF, de Hilster RHJ, Leemhuis H, Oudhuis L, Buwalda PL, de Vos P. Higher Chain Length Distribution in Debranched Type-3 Resistant Starches (RS3) Increases TLR Signaling and Supports Dendritic Cell Cytokine Production. Mol Nutr Food Res 2019; 63:e1801007. [PMID: 30412339 PMCID: PMC6767581 DOI: 10.1002/mnfr.201801007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 10/20/2018] [Indexed: 12/16/2022]
Abstract
SCOPE Resistant starches (RSs) are classically considered to elicit health benefits through fermentation. However, it is recently shown that RSs can also support health by direct immune interactions. Therefore, it has been hypothesized that the structural traits of RSs might impact the health benefits associated with their consumption. METHODS AND RESULTS Effects of crystallinity, molecular weight, and chain length distribution of RSs are determined on immune Toll-like receptors (TLRs), dendritic cells (DCs), and T-cell cytokines production. To this end, four type-3 RSs (RS3) are compared, namely Paselli WFR, JD150, debranched Etenia, and Amylose fraction V, which are extracted from potatoes and enzymatically modified. Dextrose equivalent seems to be the most important feature influencing immune signaling via activation of TLRs. TLR2 and TLR4 are most strongly stimulated. Especially Paselli WFR is a potent activator of multiple receptors. Moreover, the presence of amylose, even to residual levels, enhances DC and T-cell cytokine responses. Paselli WFR and Amylose fraction V influence T-cell polarization. CONCLUSIONS It has been shown here that chain length and particularly dextrose equivalent are critical features for immune activation. This knowledge might lead to tailoring and design of immune-active RS formulations.
Collapse
Affiliation(s)
- Alexia F.P. Lépine
- ImmunoendocrinologyDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen, University Medical Center GroningenHanzeplein 19700RBGroningenThe Netherlands
- Food and Biobased ResearchWageningen University and Research centerBornse Weilanden 96708WGWageningenThe Netherlands
| | - Roderick H. J. de Hilster
- ImmunoendocrinologyDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen, University Medical Center GroningenHanzeplein 19700RBGroningenThe Netherlands
| | - Hans Leemhuis
- AVEBE Innovation CenterP.O.Box 159640AAVeendamThe Netherlands
| | - Lizette Oudhuis
- AVEBE Innovation CenterP.O.Box 159640AAVeendamThe Netherlands
| | - Piet L. Buwalda
- AVEBE Innovation CenterP.O.Box 159640AAVeendamThe Netherlands
- Biobased Chemistry and TechnologyWageningen University and Research centerBornse Weilanden 96708WGWageningenThe Netherlands
| | - Paul de Vos
- ImmunoendocrinologyDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen, University Medical Center GroningenHanzeplein 19700RBGroningenThe Netherlands
| |
Collapse
|
27
|
McMenamin PG, Saban DR, Dando SJ. Immune cells in the retina and choroid: Two different tissue environments that require different defenses and surveillance. Prog Retin Eye Res 2018; 70:85-98. [PMID: 30552975 DOI: 10.1016/j.preteyeres.2018.12.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 12/03/2018] [Accepted: 12/11/2018] [Indexed: 01/04/2023]
Abstract
In the eye immune defenses must take place in a plethora of differing microenvironments ranging from the corneal and conjunctival epithelia facing the external environment to the pigmented connective tissue of the uveal tract containing smooth muscle, blood vessels and peripheral nerves to the innermost and highly protected neural retina. The extravascular environment of the neural retina, like the brain parenchyma, is stringently controlled to maintain conditions required for neural transmission. The unique physiological nature of the neural retina can be attributed to the blood retinal barriers (BRB) of the retinal vasculature and the retinal pigment epithelium, which both tightly regulate the transport of small molecules and restrict passage of cells and macromolecules from the circulation into the retina in a similar fashion to the blood brain barrier (BBB). The extracellular environment of the neural retina differs markedly from that of the highly vascular, loose connective tissue of the choroid, which lies outside the BRB. The choroid hosts a variety of immune cell types, including macrophages, dendritic cells (DCs) and mast cells. This is in marked contrast to the neural parenchyma of the retina, which is populated almost solely by microglia. This review will describe the current understanding of the distribution, phenotype and physiological role of ocular immune cells behind or inside the blood-retinal barriers and those in closely juxtaposed tissues outside the barrier. The nature and function of these immune cells can profoundly influence retinal homeostasis and lead to disordered immune function that can lead to vision loss.
Collapse
Affiliation(s)
- Paul G McMenamin
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.
| | - Daniel R Saban
- Department of Ophthalmology, Department of Immunology, Duke University School of Medicine, Durham, NC, USA
| | - Samantha J Dando
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
28
|
Lépine A, de Vos P. Synbiotic Effects of the Dietary Fiber Long-Chain Inulin and Probiotic Lactobacillus acidophilus W37 Can be Caused by Direct, Synergistic Stimulation of Immune Toll-Like Receptors and Dendritic Cells. Mol Nutr Food Res 2018; 62:e1800251. [PMID: 29902355 PMCID: PMC6099370 DOI: 10.1002/mnfr.201800251] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/02/2018] [Indexed: 01/24/2023]
Abstract
SCOPE Synbiotic effects of dietary fibers and lactobacilli are usually explained by synergistic modulation of gut microbiota. New insight, however, has demonstrated that both dietary fibers and lactobacilli can directly stimulate immune cells and benefit consumer immunity. Here, the synergistic effects of immune active long-chain inulin (lcITF) and Lactobacillus acidophilus W37 (LaW37) on dendritic cells (DCs) are investigated. METHODS AND RESULTS Effects of lcITF and LaW37 alone or combined were studied on Toll-like receptor (TLRs) signaling and cytokine secretion by DCs in the presence and absence of media of intestinal epithelial cell (IEC) exposed to the ingredients. Also, the effects of DC responses against Salmonella Typhimurium (STM) were investigated. Synergistic effects were observed on TLR2 and 3. Synergistic effects were not always pro-inflammatory. LaW37 was strongly pro-inflammatory, while cytokine responses were regulatory when combined with lcITF. Exposure of DCs to IECs medium changed the DCs' response, which revealed synergistic enhancing effects of lcITF/LaW37 on production of IL-6 and IL-8. DCs' response in the presence of STM and LaW37 were so strong that lcITF had no additional effect. CONCLUSION It is demonstrated that synbiotic effects of dietary fibers and bacteria are not limited to the effects on gut microbiota but can also occur by synergistically directly stimulating IECs and/or immune cells.
Collapse
Affiliation(s)
- Alexia Lépine
- Immunoendocrinology GroupDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity Medical Center Groningen, University of GroningenHanzeplein 19700RBGroningenThe Netherlands,Food and Biobased ResearchWageningen University and ResearchBornse Weilanden 96708WGWageningenThe Netherlands
| | - Paul de Vos
- Immunoendocrinology GroupDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity Medical Center Groningen, University of GroningenHanzeplein 19700RBGroningenThe Netherlands
| |
Collapse
|
29
|
Foulsham W, Coco G, Amouzegar A, Chauhan SK, Dana R. When Clarity Is Crucial: Regulating Ocular Surface Immunity. Trends Immunol 2018; 39:288-301. [PMID: 29248310 PMCID: PMC5880704 DOI: 10.1016/j.it.2017.11.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 11/28/2017] [Accepted: 11/29/2017] [Indexed: 12/23/2022]
Abstract
The ocular surface is a unique mucosal immune compartment in which anatomical, physiological, and immunological features act in concert to foster a particularly tolerant microenvironment. These mechanisms are vital to the functional competence of the eye, a fact underscored by the devastating toll of excessive inflammation at the cornea - blindness. Recent data have elucidated the contributions of specific anatomical components, immune cells, and soluble immunoregulatory factors in promoting homeostasis at the ocular surface. We highlight research trends at this distinctive mucosal barrier and identify crucial gaps in our current knowledge.
Collapse
Affiliation(s)
- William Foulsham
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; University College London (UCL) Institute of Ophthalmology, University College London, London, UK
| | - Giulia Coco
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Afsaneh Amouzegar
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Sunil K Chauhan
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| | - Reza Dana
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
30
|
He L, Liu T, Shi Y, Tian F, Hu H, Deb DK, Chen Y, Bissonnette M, Li YC. Gut Epithelial Vitamin D Receptor Regulates Microbiota-Dependent Mucosal Inflammation by Suppressing Intestinal Epithelial Cell Apoptosis. Endocrinology 2018; 159:967-979. [PMID: 29228157 PMCID: PMC5788002 DOI: 10.1210/en.2017-00748] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 11/28/2017] [Indexed: 02/08/2023]
Abstract
Recent studies show that colonic vitamin D receptor (VDR) signaling protects the mucosal epithelial barrier and suppresses colonic inflammation, but the underlying molecular mechanism remains to be fully understood. To investigate the implication of colonic VDR downregulation seen in patients with inflammatory bowel disease, we assessed the effect of gut epithelial VDR deletion on colonic inflammatory responses in an experimental colitis model. In a 2,4,6-trinitrobenzenesulfonic acid-induced colitis model, mice carrying VDR deletion in gut epithelial cells [VDRflox/flox (VDRf/f);Villin-Cre or VDRΔIEC] or in colonic epithelial cells (VDRf/f;CDX2-Cre or VDRΔCEC) developed more severe clinical colitis than VDRf/f control mice, characterized by more robust T-helper (TH)1 and TH17 responses, with greater increases in mucosal interferon (IFN)-γ+, interleukin (IL)-17+, and IFN-γ+IL-17+ T cells. Accompanying the severe mucosal inflammation was more profound colonic epithelial cell apoptosis in the mutant mice. Treatment with caspase inhibitor Q-VD-OPh dramatically reduced colitis severity and attenuated TH1 and TH17 responses in VDRΔCEC mice. The blockade of cell apoptosis also prevented the increase in mucosal CD11b+CD103+ dendritic cells (DCs), known to be critical for TH17-cell activation. Moreover, depletion of gut commensal bacteria with antibiotics eliminated the robust TH1 and TH17 responses and CD11b+CD103+ DC induction. Taken together, these observations demonstrate that gut epithelial VDR deletion aggravates epithelial cell apoptosis, resulting in increases in mucosal barrier permeability. Consequently, invading luminal bacteria activate CD11b+CD103+ DCs, which promote mucosal TH1 and TH17 responses. Therefore, gut epithelial VDR signaling controls mucosal inflammation by suppressing epithelial cell apoptosis.
Collapse
Affiliation(s)
- Lei He
- Department of Medicine, Division of Biological Sciences, The University of Chicago, Chicago, Illinois 60637
| | - Tianjing Liu
- Department of Medicine, Division of Biological Sciences, The University of Chicago, Chicago, Illinois 60637
- Department of Pediatrics, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110004, China
| | - Yongyan Shi
- Department of Medicine, Division of Biological Sciences, The University of Chicago, Chicago, Illinois 60637
- Department of Pediatrics, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110004, China
| | - Feng Tian
- Department of Medicine, Division of Biological Sciences, The University of Chicago, Chicago, Illinois 60637
- Department of Gastroenterology, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110004, China
| | - Huiyuan Hu
- Department of Medicine, Division of Biological Sciences, The University of Chicago, Chicago, Illinois 60637
- College of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Dilip K. Deb
- Department of Medicine, Division of Biological Sciences, The University of Chicago, Chicago, Illinois 60637
| | - Yinyin Chen
- Department of Medicine, Division of Biological Sciences, The University of Chicago, Chicago, Illinois 60637
- Department of Nephrology, Hunan Provincial People’s Hospital, Changsha, Hunan 410005, China
| | - Marc Bissonnette
- Department of Medicine, Division of Biological Sciences, The University of Chicago, Chicago, Illinois 60637
| | - Yan Chun Li
- Department of Medicine, Division of Biological Sciences, The University of Chicago, Chicago, Illinois 60637
| |
Collapse
|
31
|
Tomlinson ML, Butelli E, Martin C, Carding SR. Flavonoids from Engineered Tomatoes Inhibit Gut Barrier Pro-inflammatory Cytokines and Chemokines, via SAPK/JNK and p38 MAPK Pathways. Front Nutr 2017; 4:61. [PMID: 29326940 PMCID: PMC5741681 DOI: 10.3389/fnut.2017.00061] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 11/24/2017] [Indexed: 12/29/2022] Open
Abstract
Flavonoids are a diverse group of plant secondary metabolites, known to reduce inflammatory bowel disease symptoms. How they achieve this is largely unknown. Our study focuses on the gut epithelium as it receives high topological doses of dietary constituents, maintains gut homeostasis, and orchestrates gut immunity. Dysregulation leads to chronic gut inflammation, via dendritic cell (DC)-driven immune responses. Tomatoes engineered for enriched sets of flavonoids (anthocyanins or flavonols) provided a unique and complex naturally consumed food matrix to study the effect of diet on chronic inflammation. Primary murine colonic epithelial cell-based inflammation assays consist of chemokine induction, apoptosis and proliferation, and effects on kinase pathways. Primary murine leukocytes and DCs were used to assay effects on transmigration. A murine intestinal cell line was used to assay wound healing. Engineered tomato extracts (enriched in anthocyanins or flavonols) showed strong and specific inhibitory effects on a set of key epithelial pro-inflammatory cytokines and chemokines. Chemotaxis assays showed a resulting reduction in the migration of primary leukocytes and DCs. Activation of epithelial cell SAPK/JNK and p38 MAPK signaling pathways were specifically inhibited. The epithelial wound healing-associated STAT3 pathway was unaffected. Cellular migration, proliferation, and apoptosis assays confirmed that wound healing processes were not affected by flavonoids. We show flavonoids target epithelial pro-inflammatory kinase pathways, inhibiting chemotactic signals resulting in reduced leukocyte and DC chemotaxis. Thus, both anthocyanins and flavonols modulate epithelial cells to become hyporesponsive to bacterial stimulation. Our results identify a viable mechanism to explain the in vivo anti-inflammatory effects of flavonoids.
Collapse
Affiliation(s)
- Matthew L. Tomlinson
- Gut Health and Food Safety Research Programme, Quadram Institute, Norwich, United Kingdom
- Martin Laboratory, The John Innes Centre, Norwich, United Kingdom
| | - Eugenio Butelli
- Martin Laboratory, The John Innes Centre, Norwich, United Kingdom
| | - Cathie Martin
- Martin Laboratory, The John Innes Centre, Norwich, United Kingdom
| | - Simon R. Carding
- Gut Health and Food Safety Research Programme, Quadram Institute, Norwich, United Kingdom
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| |
Collapse
|
32
|
Luongo D, Treppiccione L, Sorrentino A, Ferrocino I, Turroni S, Gatti M, Di Cagno R, Sanz Y, Rossi M. Immune-modulating effects in mouse dendritic cells of lactobacilli and bifidobacteria isolated from individuals following omnivorous, vegetarian and vegan diets. Cytokine 2017; 97:141-148. [PMID: 28648868 DOI: 10.1016/j.cyto.2017.06.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 04/28/2017] [Accepted: 06/12/2017] [Indexed: 02/06/2023]
Abstract
Lactobacilli and bifidobacteria play a primary role in modulation of gut immunity. By considering that microbiota composition depends on various factors, including diet, we asked whether functional differences could characterize faecal populations of lactobacilli and bifidobacteria isolated from individuals with different dietary habits. 155 healthy volunteers who followed omnivorous, ovo-lacto-vegetarian or vegan diets were recruited at four Italian centres (Turin, Parma, Bologna and Bari). Faecal samples were collected; lactobacilli and bifidobacteria were isolated on selective media and their immunomodulatory activity was tested in mouse dendritic cells (DCs). Pre-incubation with lactobacilli increased LPS-induced expression of the maturation markers CD80 and CD86, whereas pre-incubation with bifidobacteria decreased such expression. Analysis of the cytokine profile indicated that strains of both genera induced down-regulation of IL-12 and up-regulation of IL-10, whereas expression of TNF-α was not modulated. Notably, analysis of anti-inflammatory potential (IL-10/IL-12 ratio) showed that lactobacilli evoked a greater anti-inflammatory effect than did bifidobacteria in the omnivorous group (P<0.05). We also found significantly reduced anti-inflammatory potential in the bacterial strains isolated from Bari's volunteers in comparison with those from the cognate groups from the other centres. In conclusion, lactobacilli and bifidobacteria showed a genus-specific ability of modulating in vitro innate immunity associated with a specific dietary habit. Furthermore, the geographical area had a significant impact on the anti-inflammatory potential of some components of faecal microbiota.
Collapse
Affiliation(s)
| | | | | | - Ilario Ferrocino
- Department of Agricultural, Forest and Food Science, University of Turin, Grugliasco, Italy
| | - Silvia Turroni
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Monica Gatti
- Laboratory of Food Microbiology, Department of Food Science, University of Parma, Parco Area delle Scienze 48/A, Parma, Italy
| | - Raffaella Di Cagno
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Yolanda Sanz
- Microbial Ecology, Nutrition and Health Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| | - Mauro Rossi
- Institute of Food Sciences, CNR, Avellino, Italy.
| |
Collapse
|
33
|
Liu H, Xu W, Yu Q, Yang Q. 4,4'-Diaponeurosporene-Producing Bacillus subtilis Increased Mouse Resistance against Salmonella typhimurium Infection in a CD36-Dependent Manner. Front Immunol 2017; 8:483. [PMID: 28491061 PMCID: PMC5405070 DOI: 10.3389/fimmu.2017.00483] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 04/07/2017] [Indexed: 01/28/2023] Open
Abstract
Deficient mucosal innate immunity is a hallmark of infectious diarrhea, such as Salmonella typhimurium (S. typhimurium)-induced gastroenteritis. Here, we report that oral administration of a 4,4′-diaponeurosporene-producing Bacillus subtilis (B.s-Dia) could improve mice mucosal immunity, as showed by an increased resistance against S. typhimurium infection. Intragastric administration of B.s-Dia for 7 days could increase the secretion of CCL20 by intestinal epithelial cells (IECs) and then recruit more dendritic cells. Meanwhile, the number of CD8αα+ intraepithelial lymphocytes, which play a critical role in downregulating immune responses, was also reduced, probably as a consequence of the decrease of IEC-derived TGFβ. Further study showed that CD36 played a critical role in B.s-Dia-induced immune enhancement, as blocking CD36 signal with a specific antagonist, sulfo-N-succinimidyl oleate, led to the inability of B.s-Dia to enhance mucosal innate immunity.
Collapse
Affiliation(s)
- Haofei Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Wenwen Xu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Qinghua Yu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Qian Yang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
34
|
Bene KP, Kavanaugh DW, Leclaire C, Gunning AP, MacKenzie DA, Wittmann A, Young ID, Kawasaki N, Rajnavolgyi E, Juge N. Lactobacillus reuteri Surface Mucus Adhesins Upregulate Inflammatory Responses Through Interactions With Innate C-Type Lectin Receptors. Front Microbiol 2017; 8:321. [PMID: 28326063 PMCID: PMC5339304 DOI: 10.3389/fmicb.2017.00321] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 02/15/2017] [Indexed: 12/13/2022] Open
Abstract
The vertebrate gut symbiont Lactobacillus reuteri exhibits strain-specific adhesion and health-promoting properties. Here, we investigated the role of the mucus adhesins, CmbA and MUB, upon interaction of L. reuteri ATCC PTA 6475 and ATCC 53608 strains with human monocyte-derived dendritic cells (moDCs). We showed that mucus adhesins increased the capacity of L. reuteri strains to interact with moDCs and promoted phagocytosis. Our data also indicated that mucus adhesins mediate anti- and pro-inflammatory effects by the induction of interleukin-10 (IL-10), tumor necrosis factor alpha (TNF-α), IL-1β, IL-6, and IL-12 cytokines. L. reuteri ATCC PTA 6475 and ATCC 53608 were exclusively able to induce moDC-mediated Th1 and Th17 immune responses. We further showed that purified MUB activates moDCs and induces Th1 polarized immune responses associated with increased IFNγ production. MUB appeared to mediate these effects via binding to C-type lectin receptors (CLRs), as shown using cell reporter assays. Blocking moDCs with antibodies against DC-specific intercellular adhesion molecule 3-grabbing non-integrin (DC-SIGN) or Dectin-2 did not affect the uptake of the MUB-expressing strain, but reduced the production of TNF-α and IL-6 by moDCs significantly, in line with the Th1 polarizing capacity of moDCs. The direct interaction between MUB and CLRs was further confirmed by atomic force spectroscopy. Taken together these data suggest that mucus adhesins expressed at the cell surface of L. reuteri strains may exert immunoregulatory effects in the gut through modulating the Th1-promoting capacity of DCs upon interaction with C-type lectins.
Collapse
Affiliation(s)
- Krisztián P Bene
- Department of Immunology, Faculty of Medicine, University of Debrecen Debrecen, Hungary
| | - Devon W Kavanaugh
- The Gut Health and Food Safety Programme, Institute of Food Research Norwich, UK
| | - Charlotte Leclaire
- The Gut Health and Food Safety Programme, Institute of Food Research Norwich, UK
| | - Allan P Gunning
- The Gut Health and Food Safety Programme, Institute of Food Research Norwich, UK
| | - Donald A MacKenzie
- The Gut Health and Food Safety Programme, Institute of Food Research Norwich, UK
| | | | - Ian D Young
- Food and Health Programme, Institute of Food Research Norwich, UK
| | | | - Eva Rajnavolgyi
- Department of Immunology, Faculty of Medicine, University of Debrecen Debrecen, Hungary
| | - Nathalie Juge
- The Gut Health and Food Safety Programme, Institute of Food Research Norwich, UK
| |
Collapse
|
35
|
Galletti JG, Guzmán M, Giordano MN. Mucosal immune tolerance at the ocular surface in health and disease. Immunology 2017; 150:397-407. [PMID: 28108991 DOI: 10.1111/imm.12716] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/28/2016] [Accepted: 01/12/2017] [Indexed: 12/15/2022] Open
Abstract
The ocular surface is constantly exposed to environmental irritants, allergens and pathogens, against which it can mount a prompt immune response to preserve its integrity. But to avoid unnecessary inflammation, the ocular surface's mucosal immune system must also discriminate between harmless and potentially dangerous antigens, a seemingly complicated task. Despite its unique features, the ocular surface is a mucosal lining, and as such, it shares some homeostatic and pathophysiological mechanisms with other mucosal surfaces. The purpose of this review is to explore the mucosal homeostatic immune function of the ocular surface in both the healthy and diseased states, with a special focus on mucosal immunology concepts. The information discussed in this review has been retrieved by PubMed searches for literature published from January 1981 to October 2016.
Collapse
Affiliation(s)
- Jeremías G Galletti
- Immunology Laboratory, Institute of Experimental Medicine, National Academy of Medicine/CONICET, Buenos Aires, Argentina
| | - Mauricio Guzmán
- Immunology Laboratory, Institute of Experimental Medicine, National Academy of Medicine/CONICET, Buenos Aires, Argentina
| | - Mirta N Giordano
- Immunology Laboratory, Institute of Experimental Medicine, National Academy of Medicine/CONICET, Buenos Aires, Argentina
| |
Collapse
|
36
|
Castronovo G, Clemente AM, Antonelli A, D’Andrea MM, Tanturli M, Perissi E, Paccosi S, Parenti A, Cozzolino F, Rossolini GM, Torcia MG. Differences in Inflammatory Response Induced by Two Representatives of Clades of the Pandemic ST258 Klebsiella pneumoniae Clonal Lineage Producing KPC-Type Carbapenemases. PLoS One 2017; 12:e0170125. [PMID: 28081233 PMCID: PMC5231394 DOI: 10.1371/journal.pone.0170125] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 12/29/2016] [Indexed: 12/20/2022] Open
Abstract
ST258-K. pneumoniae (ST258-KP) strains, the most widespread multidrug-resistant hospital-acquired pathogens, belong to at least two clades differing in a 215 Kb genomic region that includes the cluster of capsule genes. To investigate the effects of the different capsular phenotype on host-pathogen interactions, we studied representatives of ST258-KP clades, KKBO-1 and KK207-1, for their ability to activate monocytes and myeloid dendritic cells from human immune competent hosts. The two ST258-KP strains strongly induced the production of inflammatory cytokines. Significant differences between the strains were found in their ability to induce the production of IL-1β: KK207-1/clade I was much less effective than KKBO-1/clade II in inducing IL-1β production by monocytes and dendritic cells. The activation of NLRP3 inflammasome pathway by live cells and/or purified capsular polysaccharides was studied in monocytes and dendritic cells. We found that glibenclamide, a NLRP3 inhibitor, inhibits more than 90% of the production of mature IL-1β induced by KKBO1 and KK207-1. KK207-1 was always less efficient compared to KKBO-1 in: a) inducing NLRP3 and pro-IL-1β gene and protein expression; b) in inducing caspase-1 activation and pro-IL-1β cleavage. Capsular composition may play a role in the differential inflammatory response induced by the ST258-KP strains since capsular polysaccharides purified from bacterial cells affect NLRP3 and pro-IL-1β gene expression through p38MAPK- and NF-κB-mediated pathways. In each of these functions, capsular polysaccharides from KK207-1 were significantly less efficient compared to those purified from KKBO-1. On the whole, our data suggest that the change in capsular phenotype may help bacterial cells of clade I to partially escape innate immune recognition and IL-1β-mediated inflammation.
Collapse
Affiliation(s)
- Giuseppe Castronovo
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze, Italy
| | - Ann Maria Clemente
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze, Italy
| | - Alberto Antonelli
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze, Italy
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Marco Maria D’Andrea
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze, Italy
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Michele Tanturli
- Department of Experimental and Clinical Biomedical Sciences, University of Firenze, Firenze, Italy
| | - Eloisa Perissi
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze, Italy
| | - Sara Paccosi
- Department of Health Sciences, University of Firenze, Firenze, Italy
| | - Astrid Parenti
- Department of Health Sciences, University of Firenze, Firenze, Italy
| | - Federico Cozzolino
- Department of Experimental and Clinical Biomedical Sciences, University of Firenze, Firenze, Italy
| | - Gian Maria Rossolini
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze, Italy
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Clinical Microbiology and Virology Unit, Careggi University Hospital, Firenze, Italy
- IRCCS Don Carlo Gnocchi Foundation, Firenze, Italy
| | - Maria Gabriella Torcia
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze, Italy
- * E-mail:
| |
Collapse
|
37
|
Adachi T, Kakuta S, Aihara Y, Kamiya T, Watanabe Y, Osakabe N, Hazato N, Miyawaki A, Yoshikawa S, Usami T, Karasuyama H, Kimoto-Nira H, Hirayama K, Tsuji NM. Visualization of Probiotic-Mediated Ca 2+ Signaling in Intestinal Epithelial Cells In Vivo. Front Immunol 2016; 7:601. [PMID: 28018362 PMCID: PMC5159486 DOI: 10.3389/fimmu.2016.00601] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 11/30/2016] [Indexed: 01/28/2023] Open
Abstract
Probiotics, such as lactic acid bacteria (LAB) and Bacillus subtilis var. natto, have been shown to modulate immune responses. It is important to understand how probiotic bacteria impact intestinal epithelial cells (IECs), because IECs are the first line of defense at the mucosal surface barrier and their activities substantially affect the gut microenvironment and immunity. However, to date, their precise mechanism remains unknown due to a lack of analytical systems available for live animal models. Recently, we generated a conditional Ca2+ biosensor Yellow Cameleon (YC3.60) transgenic mouse line and established 5D (x, y, z, time, and Ca2+) intravital imaging systems of lymphoid tissues including those in Peyer’s patches and bone marrow. In the present study, we further advance our intravital imaging system for intestinal tracts to visualize IEC responses against orally administrated food compounds in real time. Using this system, heat-killed B. subtilis natto, a probiotic TTCC012 strain, is shown to directly induce Ca2+ signaling in IECs in mice housed under specific pathogen-free conditions. In contrast, this activation is not observed in the Lactococcus lactis strain C60; however, when we generate germ-free YC3.60 mice and observe the LAB stimulation of IECs in the absence of gut microbiota, C60 is capable of inducing Ca2+ signaling. This is the first study to successfully visualize the direct effect of probiotics on IECs in live animals. These data strongly suggest that probiotic strains stimulate IECs under physiological conditions and that their activity is affected by the microenvironment of the small intestine, such as commensal bacteria.
Collapse
Affiliation(s)
- Takahiro Adachi
- Department of Immunology, Medical Research Institute, Tokyo Medical and Dental University , Tokyo , Japan
| | - Shigeru Kakuta
- Department of Biomedical Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo , Tokyo , Japan
| | - Yoshiko Aihara
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University , Kobe , Japan
| | - Tomonori Kamiya
- Biomedical Research Institute, National Institute for Advanced Industrial Science and Technology (AIST) , Tsukuba , Japan
| | - Yohei Watanabe
- Biomedical Research Institute, National Institute for Advanced Industrial Science and Technology (AIST) , Tsukuba , Japan
| | - Naomi Osakabe
- Department of Bio-science and Engineering, Shibaura Institute of Technology , Saitama , Japan
| | - Naoki Hazato
- Department of Bio-science and Engineering, Shibaura Institute of Technology , Saitama , Japan
| | - Atsushi Miyawaki
- Laboratory for Cell Function and Dynamics, Advanced Technology Development Group, Brain Science Institute, RIKEN , Saitama , Japan
| | - Soichiro Yoshikawa
- Department of Immune Regulation, Tokyo Medical and Dental University , Tokyo , Japan
| | - Takako Usami
- Laboratory of Recombinant Animals, Medical Research Institute, Tokyo Medical and Dental University , Tokyo , Japan
| | - Hajime Karasuyama
- Department of Immune Regulation, Tokyo Medical and Dental University , Tokyo , Japan
| | - Hiromi Kimoto-Nira
- NARO Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba , Ibaraki , Japan
| | - Kazuhiro Hirayama
- Laboratory of Veterinary Public Health, Graduate School of Agricultural and Life Sciences, The University of Tokyo , Tokyo , Japan
| | - Noriko M Tsuji
- Biomedical Research Institute, National Institute for Advanced Industrial Science and Technology (AIST) , Tsukuba , Japan
| |
Collapse
|
38
|
Boucard-Jourdin M, Kugler D, Endale Ahanda ML, This S, De Calisto J, Zhang A, Mora JR, Stuart LM, Savill J, Lacy-Hulbert A, Paidassi H. β8 Integrin Expression and Activation of TGF-β by Intestinal Dendritic Cells Are Determined by Both Tissue Microenvironment and Cell Lineage. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:1968-78. [PMID: 27481847 PMCID: PMC4992650 DOI: 10.4049/jimmunol.1600244] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 06/30/2016] [Indexed: 12/23/2022]
Abstract
Activation of TGF-β by dendritic cells (DCs) expressing αvβ8 integrin is essential for the generation of intestinal regulatory T cells (Tregs) that in turn promote tolerance to intestinal Ags. We have recently shown that αvβ8 integrin is preferentially expressed by CD103(+) DCs and confers their ability to activate TGF-β and generate Tregs. However, how these DCs become specialized for this vital function is unknown. In this study, we show that β8 expression is controlled by a combination of factors that include DC lineage and signals derived from the tissue microenvironment and microbiota. Specifically, our data demonstrate that TGF-β itself, along with retinoic acid and TLR signaling, drives expression of αvβ8 in DCs. However, these signals only result in high levels of β8 expression in cells of the cDC1 lineage, CD8α(+), or CD103(+)CD11b(-) DCs, and this is associated with epigenetic changes in the Itgb8 locus. Together, these data provide a key illustrative example of how microenvironmental factors and cell lineage drive the generation of regulatory αvβ8-expressing DCs specialized for activation of TGF-β to facilitate Treg generation.
Collapse
Affiliation(s)
- Mathilde Boucard-Jourdin
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, 69365 Lyon, France; INSERM, U1111, 69007 Lyon, France; Ecole Normale Supérieure de Lyon, 69007 Lyon, France; Université Claude Bernard Lyon 1, Centre International de Recherche en Infectiologie, Lyon, France; CNRS, UMR5308, 69365 Lyon, France
| | | | - Marie-Laure Endale Ahanda
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, 69365 Lyon, France; INSERM, U1111, 69007 Lyon, France; Ecole Normale Supérieure de Lyon, 69007 Lyon, France; Université Claude Bernard Lyon 1, Centre International de Recherche en Infectiologie, Lyon, France; CNRS, UMR5308, 69365 Lyon, France
| | - Sébastien This
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, 69365 Lyon, France; INSERM, U1111, 69007 Lyon, France; Ecole Normale Supérieure de Lyon, 69007 Lyon, France; Université Claude Bernard Lyon 1, Centre International de Recherche en Infectiologie, Lyon, France; CNRS, UMR5308, 69365 Lyon, France
| | - Jaime De Calisto
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Universidad Mayor, Santiago 8580745, Chile
| | - Ailiang Zhang
- Medical Research Council Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom; and
| | - J Rodrigo Mora
- Gastrointestinal Unit, Massachusetts General Hospital, Boston, MA 02114
| | | | - John Savill
- Medical Research Council Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom; and
| | | | - Helena Paidassi
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, 69365 Lyon, France; INSERM, U1111, 69007 Lyon, France; Ecole Normale Supérieure de Lyon, 69007 Lyon, France; Université Claude Bernard Lyon 1, Centre International de Recherche en Infectiologie, Lyon, France; CNRS, UMR5308, 69365 Lyon, France;
| |
Collapse
|
39
|
Haileselassie Y, Navis M, Vu N, Qazi KR, Rethi B, Sverremark-Ekström E. Lactobacillus reuteri and Staphylococcus aureus differentially influence the generation of monocyte-derived dendritic cells and subsequent autologous T cell responses. IMMUNITY INFLAMMATION AND DISEASE 2016; 4:315-26. [PMID: 27621814 PMCID: PMC5004286 DOI: 10.1002/iid3.115] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 05/20/2016] [Accepted: 06/20/2016] [Indexed: 12/23/2022]
Abstract
Introduction In early‐life, the immature mucosal barrier allows contact between the gut microbiota and the developing immune system. Due to their strategic location and their ability to sample luminal antigen, dendritic cells (DC) play a central role in the interaction of microbes and immune cells in the gut. Here, we investigated how two bacteria associated with opposite immune profiles in children, that is, Lactobacillus (L.) reuteri and Staphylococcus (S.) aureus, influenced the differentiation of monocytes in vitro as well how the generated DC impacted T cell responses. Methods We exposed monocyte cultures to cell‐free supernatants (CFS) from these bacteria during their differentiation to DC. Results The presence of L. reuteri‐CFS during DC differentiation resulted in DC with a more mature phenotype, in terms of up‐regulated surface markers (HLA‐DR, CD86, CD83, CCR7) and enhanced cytokine production (IL6, IL10, and IL23), but had a reduced phagocytic capacity compared with non‐treated monocyte‐derived DC (Mo‐DC). However, upon LPS activation, L. reuteri‐CFS‐generated DC displayed a more regulated phenotype than control Mo‐DC with notable reduction of cytokine responses both at mRNA and protein levels. In contrast, S. aureus‐CFS‐generated DC were more similar to control Mo‐DC both without and after LPS stimulation, but they were still able to induce responses in autologous T cells, in the absence of further T cell stimulation. Conclusions We show that bacterial signals during DC differentiation have a profound impact on DC function and possibly also for shaping the T cell pool.
Collapse
Affiliation(s)
- Yeneneh Haileselassie
- Department of Molecular Biosciences, The Wenner-Gren Institute Stockholm University Stockholm Sweden
| | - Marit Navis
- Department of Molecular Biosciences, The Wenner-Gren Institute Stockholm University Stockholm Sweden
| | - Nam Vu
- Department of Molecular Biosciences, The Wenner-Gren Institute Stockholm University Stockholm Sweden
| | - Khaleda Rahman Qazi
- Department of Molecular Biosciences, The Wenner-Gren Institute Stockholm University Stockholm Sweden
| | - Bence Rethi
- Department of Medicine Karolinska University Hospital Stockholm Sweden
| | - Eva Sverremark-Ekström
- Department of Molecular Biosciences, The Wenner-Gren Institute Stockholm University Stockholm Sweden
| |
Collapse
|
40
|
Haileselassie Y, Navis M, Vu N, Qazi KR, Rethi B, Sverremark-Ekström E. Postbiotic Modulation of Retinoic Acid Imprinted Mucosal-like Dendritic Cells by Probiotic Lactobacillus reuteri 17938 In Vitro. Front Immunol 2016; 7:96. [PMID: 27014275 PMCID: PMC4794487 DOI: 10.3389/fimmu.2016.00096] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 02/29/2016] [Indexed: 01/17/2023] Open
Abstract
Lactobacilli are widely used as probiotics with beneficial effects on infection-associated diarrhea, but also used in clinical trials of e.g., necrotizing enterocolitis and inflammatory bowel diseases. The possibility of using probiotic metabolic products, so-called postbiotics, is desirable as it could prevent possible side effects of live bacteria in individuals with a disturbed gut epithelial barrier. Here, we studied how Lactobacillus reuteri DSM 17938 cell-free supernatant (L. reuteri-CFS) influenced retinoic acid (RA)-driven mucosal-like dendritic cells (DC) and their subsequent effect on T regulatory cells (Treg) in vitro. RA clearly imprinted a mucosal-like DC phenotype with higher IL10 production, increased CD103 and CD1d expression, and a downregulated mRNA expression of several inflammatory-associated genes (NFκB1, RELB, and TNF). Treatment with L. reuteri-CFS further influenced the tolerogenic phenotype of RA-DC by downregulating most genes involved in antigen uptake, antigen presentation, and signal transduction as well as several chemokine receptors, while upregulating IL10 production. L. reuteri-CFS also augmented CCR7 expression on RA-DC. In cocultures, RA-DC increased IL10 and FOXP3 expression in Treg, but pre-treatment with L. reuteri-CFS did not further influence the Treg phenotype. In conclusion, L. reuteri-CFS modulates the phenotype and function of mucosal-like DC, implicating its potential application as postbiotic.
Collapse
Affiliation(s)
- Yeneneh Haileselassie
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Marit Navis
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Nam Vu
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Khaleda Rahman Qazi
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Bence Rethi
- Department of Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Eva Sverremark-Ekström
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| |
Collapse
|
41
|
Oral immune therapy: targeting the systemic immune system via the gut immune system for the treatment of inflammatory bowel disease. Clin Transl Immunology 2016; 5:e60. [PMID: 26900473 PMCID: PMC4735066 DOI: 10.1038/cti.2015.47] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Revised: 11/04/2015] [Accepted: 12/22/2015] [Indexed: 12/13/2022] Open
Abstract
Inflammatory bowel diseases (IBD) are associated with an altered systemic immune response leading to inflammation-mediated damage to the gut and other organs. Oral immune therapy is a method of systemic immune modulation via alteration of the gut immune system. It uses the inherit ability of the innate system of the gut to redirect the systemic innate and adaptive immune responses. Oral immune therapy is an attractive clinical approach to treat autoimmune and inflammatory disorders. It can induce immune modulation without immune suppression, has minimal toxicity and is easily administered. Targeting the systemic immune system via the gut immune system can serve as an attractive novel therapeutic method for IBD. This review summarizes the current data and discusses several examples of oral immune therapeutic methods for using the gut immune system to generate signals to reset systemic immunity as a treatment for IBD.
Collapse
|
42
|
Miniello VL, Colasanto A, Cristofori F, Diaferio L, Ficele L, Lieggi MS, Santoiemma V, Francavilla R. Gut microbiota biomodulators, when the stork comes by the scalpel. Clin Chim Acta 2015; 451:88-96. [PMID: 25668229 DOI: 10.1016/j.cca.2015.01.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 01/22/2015] [Accepted: 01/25/2015] [Indexed: 02/07/2023]
Abstract
The microbial communities that reside in the human gut (microbiota) and their impact on human health and disease are nowadays one of the most exciting new areas of research. A well-balanced microbial intestinal colonization in early postnatal life is necessary for the development of appropriate innate and adaptive immune responses and to establish immune homeostasis later in life. Although the composition and functional characteristics of a 'healthy' gut microbiota remain to be elucidated, perturbations in the microbial colonization of an infant's gastrointestinal tract have been associated with an increased risk of short- and long-term immunologically mediated diseases. Emerging evidence suggests that gut microbiota biomodulators, such as probiotics, prebiotics, synbiotics, and postbiotics may support disease prevention in infants who tend to have a delayed and/or aberrant initial colonization with reduced microbiota diversity (delivery by caesarean section, premature delivery, and excessive use of perinatal antibiotics). Under these dysbiosis conditions probiotics could act as 'surrogate' colonizers to prevent immune-mediated diseases. This review focuses on the influence of delivery mode on the colonization of the infant gastro-intestinal tract. In particular, it examines the manipulation of the gut microbiota composition through the use of gut microbiota biomodulators, in the management of aberrant initial gut colonization and subsequent consequences for the health of the offspring.
Collapse
Affiliation(s)
- Vito Leonardo Miniello
- Department of Paediatrics, Aldo Moro University of Bari-Giovanni XXIII Hospital, Via Amendola 270, 70126 Bari, Italy.
| | - Angela Colasanto
- Department of Paediatrics, Aldo Moro University of Bari-Giovanni XXIII Hospital, Via Amendola 270, 70126 Bari, Italy
| | - Fernanda Cristofori
- Department of Paediatrics, Aldo Moro University of Bari-Giovanni XXIII Hospital, Via Amendola 270, 70126 Bari, Italy
| | - Lucia Diaferio
- Department of Paediatrics, Aldo Moro University of Bari-Giovanni XXIII Hospital, Via Amendola 270, 70126 Bari, Italy
| | - Laura Ficele
- Department of Paediatrics, Aldo Moro University of Bari-Giovanni XXIII Hospital, Via Amendola 270, 70126 Bari, Italy
| | - Maria Serena Lieggi
- Department of Paediatrics, Aldo Moro University of Bari-Giovanni XXIII Hospital, Via Amendola 270, 70126 Bari, Italy
| | - Valentina Santoiemma
- Department of Paediatrics, Aldo Moro University of Bari-Giovanni XXIII Hospital, Via Amendola 270, 70126 Bari, Italy
| | - Ruggiero Francavilla
- Department of Paediatrics, Aldo Moro University of Bari-Giovanni XXIII Hospital, Via Amendola 270, 70126 Bari, Italy
| |
Collapse
|
43
|
Di Giovangiulio M, Verheijden S, Bosmans G, Stakenborg N, Boeckxstaens GE, Matteoli G. The Neuromodulation of the Intestinal Immune System and Its Relevance in Inflammatory Bowel Disease. Front Immunol 2015; 6:590. [PMID: 26635804 PMCID: PMC4653294 DOI: 10.3389/fimmu.2015.00590] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 11/03/2015] [Indexed: 12/18/2022] Open
Abstract
One of the main tasks of the immune system is to discriminate and appropriately react to “danger” or “non-danger” signals. This is crucial in the gastrointestinal tract, where the immune system is confronted with a myriad of food antigens and symbiotic microflora that are in constant contact with the mucosa, in addition to any potential pathogens. This large number of antigens and commensal microflora, which are essential for providing vital nutrients, must be tolerated by the intestinal immune system to prevent aberrant inflammation. Hence, the balance between immune activation versus tolerance should be tightly regulated to maintain intestinal homeostasis and to prevent immune activation indiscriminately against all luminal antigens. Loss of this delicate equilibrium can lead to chronic activation of the intestinal immune response resulting in intestinal disorders, such as inflammatory bowel diseases (IBD). In order to maintain homeostasis, the immune system has evolved diverse regulatory strategies including additional non-immunological actors able to control the immune response. Accumulating evidence strongly indicates a bidirectional link between the two systems in which the brain modulates the immune response via the detection of circulating cytokines and via direct afferent input from sensory fibers and from enteric neurons. In the current review, we will highlight the most recent findings regarding the cross-talk between the nervous system and the mucosal immune system and will discuss the potential use of these neuronal circuits and neuromediators as novel therapeutic tools to reestablish immune tolerance and treat intestinal chronic inflammation.
Collapse
Affiliation(s)
- Martina Di Giovangiulio
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven , Leuven , Belgium
| | - Simon Verheijden
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven , Leuven , Belgium
| | - Goele Bosmans
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven , Leuven , Belgium
| | - Nathalie Stakenborg
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven , Leuven , Belgium
| | - Guy E Boeckxstaens
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven , Leuven , Belgium
| | - Gianluca Matteoli
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven , Leuven , Belgium
| |
Collapse
|
44
|
Buttó LF, Schaubeck M, Haller D. Mechanisms of Microbe-Host Interaction in Crohn's Disease: Dysbiosis vs. Pathobiont Selection. Front Immunol 2015; 6:555. [PMID: 26635787 PMCID: PMC4652232 DOI: 10.3389/fimmu.2015.00555] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 10/16/2015] [Indexed: 12/11/2022] Open
Abstract
Crohn’s disease (CD) is a systemic chronic inflammatory condition mainly characterized by discontinuous transmural pathology of the gastrointestinal tract and frequent extraintestinal manifestations with intermittent episodes of remission and relapse. Genome-wide association studies identified a number of risk loci that, catalyzed by environmental triggers, result in the loss of tolerance toward commensal bacteria based on dysregulated innate effector functions and antimicrobial defense, leading to exacerbated adaptive immune responses responsible for chronic immune-mediated tissue damage. In this review, we discuss the inter-related role of changes in the intestinal microbiota, epithelial barrier integrity, and immune cell functions on the pathogenesis of CD, describing the current approaches available to investigate the molecular mechanisms underlying the disease. Substantial effort has been dedicated to define disease-associated changes in the intestinal microbiota (dysbiosis) and to link pathobionts to the etiology of inflammatory bowel diseases. A cogent definition of dysbiosis is lacking, as well as an agreement of whether pathobionts or complex shifts in the microbiota trigger inflammation in the host. Among the rarely available animal models, SAMP/Yit and TNFdeltaARE mice are the best known displaying a transmural CD-like phenotype. New hypothesis-driven mouse models, e.g., epithelial-specific Caspase8−/−, ATG16L1−/−, and XBP1−/− mice, validate pathway-focused function of specific CD-associated risk genes highlighting the role of Paneth cells in antimicrobial defense. To study the causal role of bacteria in initiating inflammation in the host, the use of germ-free mouse models is indispensable. Unraveling the interactions of genes, immune cells and microbes constitute a criterion for the development of safe, reliable, and effective treatment options for CD.
Collapse
Affiliation(s)
- Ludovica F Buttó
- Chair of Nutrition and Immunology, Technische Universität München , Freising-Weihenstephan , Germany
| | - Monika Schaubeck
- Chair of Nutrition and Immunology, Technische Universität München , Freising-Weihenstephan , Germany
| | - Dirk Haller
- Chair of Nutrition and Immunology, Technische Universität München , Freising-Weihenstephan , Germany
| |
Collapse
|
45
|
Celiac disease: Autoimmunity in response to food antigen. Semin Immunol 2015; 27:343-52. [PMID: 26603490 DOI: 10.1016/j.smim.2015.11.001] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 11/02/2015] [Accepted: 11/02/2015] [Indexed: 02/06/2023]
Abstract
Celiac disease (CD) is an increasingly common disease of the small intestine that occurs in genetically susceptible subjects by ingestion of cereal gluten proteins. Gluten is highly abundant in the modern diet and well tolerated by most individuals. In CD, however, an erroneous but highly specific, adaptive immune response is mounted toward certain parts of the gluten proteome. The resulting intestinal destruction is reversible and resolved upon removal of gluten from the diet. Post-translational modification (deamidation) of gluten peptides by transglutaminase 2 (TG2) is essential for the peptides to act as HLA-DQ-restricted T-cell antigens. Characteristically, deamidated gluten and the self-protein TG2 both become targets of highly disease specific B-cell responses. These antibodies share several peculiar characteristics despite being directed against vastly different antigens, which suggests a common mechanism of development. Importantly, no clear function has been ascribed to the antibodies and their contribution to disease may relate to their function as antigen receptors of the B cells rather than as soluble immunoglobulins. Adaptive immunity against gluten and TG2 appears not to be sufficient for establishment of the disease lesion, and it has been suggested that stress responses in the intestinal epithelium are essential for the development of full-blown disease and tissue damage. In this review we will summarize current concepts of the immune pathology of CD with particular focus on recent advances in our understanding of disease specific B-cell responses.
Collapse
|
46
|
T Lymphocyte Dynamics in Inflammatory Bowel Diseases: Role of the Microbiome. BIOMED RESEARCH INTERNATIONAL 2015; 2015:504638. [PMID: 26583115 PMCID: PMC4637034 DOI: 10.1155/2015/504638] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 05/28/2015] [Indexed: 12/17/2022]
Abstract
Humans have coevolved with a complex community of bacterial species also referred to as the microbiome, which reciprocally provides critical contributions to human metabolism and immune system development. Gut microbiome composition differs significantly between individuals depending on host genetics, diet, and environmental factors. A dysregulation of the symbiotic nature of the intestinal host-microbial relationship and an aberrant and persistent immune response are the fundamental processes involved in inflammatory bowel diseases (IBD). Considering the essential role of T cells in IBD and the contributing role of the microbiome in shaping the immune response during the pathogenesis of IBD, this review focuses on the complex relationship, interplay, and communication between the gut microbiome and T cells, including their differentiation into different subsets of effector or regulatory cells.
Collapse
|
47
|
Soukup K, Halfmann A, Le Bras M, Sahin E, Vittori S, Poyer F, Schuh C, Luger R, Niederreiter B, Haider T, Stoiber D, Blüml S, Schabbauer G, Kotlyarov A, Gaestel M, Felzmann T, Dohnal AM. The MAPK-Activated Kinase MK2 Attenuates Dendritic Cell-Mediated Th1 Differentiation and Autoimmune Encephalomyelitis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 195:541-52. [PMID: 26078274 DOI: 10.4049/jimmunol.1401663] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 05/13/2015] [Indexed: 01/21/2023]
Abstract
Dendritic cell (DC)-mediated inflammation induced via TLRs is promoted by MAPK-activated protein kinase (MK)-2, a substrate of p38 MAPK. In this study we show an opposing role of MK2, by which it consolidates immune regulatory functions in DCs through modulation of p38, ERK1/2-MAPK, and STAT3 signaling. During primary TLR/p38 signaling, MK2 mediates the inhibition of p38 activation and positively cross-regulates ERK1/2 activity, leading to a reduction of IL-12 and IL-1α/β secretion. Consequently, MK2 impairs secondary autocrine IL-1α signaling in DCs, which further decreases the IL-1α/p38 but increases the anti-inflammatory IL-10/STAT3 signaling route. Therefore, the blockade of MK2 activity enables human and murine DCs to strengthen proinflammatory effector mechanisms by promoting IL-1α-mediated Th1 effector functions in vitro. Furthermore, MK2-deficient DCs trigger Th1 differentiation and Ag-specific cytotoxicity in vivo. Finally, wild-type mice immunized with LPS in the presence of an MK2 inhibitor strongly accumulate Th1 cells in their lymph nodes. These observations correlate with a severe clinical course in DC-specific MK2 knockout mice compared with wild-type littermates upon induction of experimental autoimmune encephalitis. Our data suggest that MK2 exerts a profound anti-inflammatory effect that prevents DCs from prolonging excessive Th1 effector T cell functions and autoimmunity.
Collapse
Affiliation(s)
- Klara Soukup
- Department of Immunology, St. Anna Children's Cancer Research Institute, 1090 Vienna, Austria
| | - Angela Halfmann
- Department of Immunology, St. Anna Children's Cancer Research Institute, 1090 Vienna, Austria
| | - Marie Le Bras
- Department of Immunology, St. Anna Children's Cancer Research Institute, 1090 Vienna, Austria
| | - Emine Sahin
- Institute of Physiology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Sarah Vittori
- Department of Immunology, St. Anna Children's Cancer Research Institute, 1090 Vienna, Austria
| | - Fiona Poyer
- Department of Immunology, St. Anna Children's Cancer Research Institute, 1090 Vienna, Austria
| | - Cornelia Schuh
- Department of Immunology, St. Anna Children's Cancer Research Institute, 1090 Vienna, Austria
| | - Romana Luger
- Department of Immunology, St. Anna Children's Cancer Research Institute, 1090 Vienna, Austria
| | - Birgit Niederreiter
- Division of Rheumatology, Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria
| | - Thomas Haider
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Dagmar Stoiber
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; Ludwig Boltzmann Institute for Cancer Research, 1090 Vienna, Austria
| | - Stephan Blüml
- Division of Rheumatology, Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria
| | - Gernot Schabbauer
- Institute of Physiology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Alexey Kotlyarov
- Institute of Physiological Chemistry, Hannover Medical School, 30625 Hannover, Germany; and
| | - Matthias Gaestel
- Institute of Physiological Chemistry, Hannover Medical School, 30625 Hannover, Germany; and
| | | | - Alexander M Dohnal
- Department of Immunology, St. Anna Children's Cancer Research Institute, 1090 Vienna, Austria;
| |
Collapse
|
48
|
Amar Y, Rizzello V, Cavaliere R, Campana S, De Pasquale C, Barberi C, Oliveri D, Pezzino G, Costa G, Meddah AT, Ferlazzo G, Bonaccorsi I. Divergent signaling pathways regulate IL-12 production induced by different species of Lactobacilli in human dendritic cells. Immunol Lett 2015; 166:6-12. [PMID: 25977118 DOI: 10.1016/j.imlet.2015.05.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 04/28/2015] [Accepted: 05/04/2015] [Indexed: 12/21/2022]
Abstract
Recent studies have indicated that different strains of Lactobacilli differ in their ability to regulate IL-12 production by dendritic cells (DCs), as some strains are stronger inducer of IL-12 while other are not and can even inhibit IL-12 production stimulated by IL-12-inducer Lactobacilli. In this report we demonstrate that Lactobacillus reuteri 5289, as previously described for other strains of L. reuteri, can inhibit DC production of IL-12 induced by Lactobacilllus acidophilus NCFM. Remarkably, L. reuteri 5289 was able to inhibit IL-12 production induced not only by Lactobacilli, as so far reported, but also by bacteria of different genera, including pathogens. We investigated in human DCs the signal transduction pathways involved in the inhibition of IL-12 production induced by L. reuteri 5289, showing that this potential anti-inflammatory activity, which is also accompanied by an elevated IL-10 production, is associated to a prolonged phosphorilation of ERK1/2 MAP kinase pathway. Improved understanding of the immune regulatory mechanisms exerted by Lactobacilli is crucial for a more precise employment of these commensal bacteria as probiotics in human immune-mediated pathologies, such as allergies or inflammatory bowel diseases.
Collapse
Affiliation(s)
- Yacine Amar
- Laboratory of Immunology and Biotherapy, Dept. Human Pathology, University of Messina, Messina, Italy; Laboratory of Bioconversion Engineering and Microbiological Food Safety, Department of Biology, University of Mascara, Mascara, Algeria
| | - Valeria Rizzello
- Laboratory of Immunology and Biotherapy, Dept. Human Pathology, University of Messina, Messina, Italy
| | - Riccardo Cavaliere
- Laboratory of Immunology and Biotherapy, Dept. Human Pathology, University of Messina, Messina, Italy; Cell Therapy Program, University Hospital-A.O.U. Policlinico G.Martino, Messina, Italy
| | - Stefania Campana
- Laboratory of Immunology and Biotherapy, Dept. Human Pathology, University of Messina, Messina, Italy
| | - Claudia De Pasquale
- Laboratory of Immunology and Biotherapy, Dept. Human Pathology, University of Messina, Messina, Italy
| | - Chiara Barberi
- Laboratory of Immunology and Biotherapy, Dept. Human Pathology, University of Messina, Messina, Italy
| | - Daniela Oliveri
- Laboratory of Immunology and Biotherapy, Dept. Human Pathology, University of Messina, Messina, Italy
| | - Gaetana Pezzino
- Laboratory of Immunology and Biotherapy, Dept. Human Pathology, University of Messina, Messina, Italy
| | - Gregorio Costa
- Laboratory of Immunology and Biotherapy, Dept. Human Pathology, University of Messina, Messina, Italy; Cell Therapy Program, University Hospital-A.O.U. Policlinico G.Martino, Messina, Italy
| | - Aicha Tirtouil Meddah
- Laboratory of Bioconversion Engineering and Microbiological Food Safety, Department of Biology, University of Mascara, Mascara, Algeria
| | - Guido Ferlazzo
- Laboratory of Immunology and Biotherapy, Dept. Human Pathology, University of Messina, Messina, Italy; Cell Therapy Program, University Hospital-A.O.U. Policlinico G.Martino, Messina, Italy.
| | - Irene Bonaccorsi
- Laboratory of Immunology and Biotherapy, Dept. Human Pathology, University of Messina, Messina, Italy; Cell Therapy Program, University Hospital-A.O.U. Policlinico G.Martino, Messina, Italy
| |
Collapse
|
49
|
Rescigno M. Dendritic cell functions: Learning from microbial evasion strategies. Semin Immunol 2015; 27:119-24. [PMID: 25843245 DOI: 10.1016/j.smim.2015.03.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 03/17/2015] [Indexed: 01/16/2023]
Abstract
Dendritic cells (DCs) are specialized antigen presenting cells (APC) that are fundamental to initiate both immunity and tolerance. DCs play a 'sentinel' role to protect our body from potential pathogens and induce tolerogenic responses toward harmless antigens. The flexibility of DCs or macrophages to adapt to the environment and to respond accordingly can be hijacked by pathogens for their own interest to transform a potentially immunogenic APC into a tolerogenic cell with clear consequences in pathogen clearance. While these immune evasion mechanisms can be detrimental for the host, they can highlight important molecular pathways in DCs necessary for their function. In this review we will mention several mechanisms employed by pathogens to evade DC patrolling function.
Collapse
Affiliation(s)
- Maria Rescigno
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy; Department of Health Sciences, School of Medicine, University of Milan, Milan, Italy.
| |
Collapse
|
50
|
Lei YMK, Nair L, Alegre ML. The interplay between the intestinal microbiota and the immune system. Clin Res Hepatol Gastroenterol 2015; 39:9-19. [PMID: 25481240 PMCID: PMC4423786 DOI: 10.1016/j.clinre.2014.10.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Accepted: 10/08/2014] [Indexed: 02/08/2023]
Abstract
The relationship between commensal microbes and their hosts has been studied for many years. Commensal microorganisms are known to have a significant role in regulating the physiology of their hosts and preventing pathogenic infections while the hosts' immune system is important in determining the composition of the microbiota. More recently, specific effects of the intestinal microbiota on the local and distal immune systems have been uncovered with important consequences for health and disease, and alterations in intestinal microbial composition has been associated with various disease states. Here, we will review the current understanding of the microbiota/immune system crosstalk, highlight the clinical consequences of changes in the microbiota and consider how to harness this symbiotic relationship to improve public health.
Collapse
|