201
|
Chen DC. Gut Microbiota and Intestinal Decolonization of Pathogenic Microorganisms. Chin Med J (Engl) 2017; 129:1639-42. [PMID: 27411449 PMCID: PMC4960951 DOI: 10.4103/0366-6999.185872] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Affiliation(s)
- De-Chang Chen
- Department of Emergency and Critical Care Medicine, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| |
Collapse
|
202
|
Barthels C, Ogrinc A, Steyer V, Meier S, Simon F, Wimmer M, Blutke A, Straub T, Zimber-Strobl U, Lutgens E, Marconi P, Ohnmacht C, Garzetti D, Stecher B, Brocker T. CD40-signalling abrogates induction of RORγt + Treg cells by intestinal CD103 + DCs and causes fatal colitis. Nat Commun 2017; 8:14715. [PMID: 28276457 PMCID: PMC5347138 DOI: 10.1038/ncomms14715] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 01/25/2017] [Indexed: 12/23/2022] Open
Abstract
Immune homeostasis in intestinal tissues depends on the generation of regulatory T (Treg) cells. CD103+ dendritic cells (DCs) acquire microbiota-derived material from the gut lumen for transport to draining lymph nodes and generation of receptor-related orphan γt+ (RORγt+) Helios−-induced Treg (iTreg) cells. Here we show CD40-signalling as a microbe-independent signal that can induce migration of CD103+ DCs from the lamina propria (LP) to the mesenteric lymph nodes. Transgenic mice with constitutive CD11c-specific CD40-signalling have reduced numbers of CD103+ DCs in LP and a low frequency of RORγt+Helios− iTreg cells, exacerbated inflammatory Th1/Th17 responses, high titres of microbiota-specific immunoglobulins, dysbiosis and fatal colitis, but no pathology is detected in other tissues. Our data demonstrate a CD40-dependent mechanism capable of abrogating iTreg cell induction by DCs, and suggest that the CD40L/CD40-signalling axis might be able to intervene in the generation of new iTreg cells in order to counter-regulate immune suppression to enhance immunity. CD103+ dendritic cells induce iTreg cells to maintain immune balance in the gut, but how CD40-signalling regulates this process is unclear. Here the authors show that mice with constitutive CD11c-specific CD40-signalling have altered CD103+ dendritic cell migration, reduced iTreg cell induction, and fatal colitis.
Collapse
Affiliation(s)
- Christian Barthels
- Institute for Immunology, LMU Munich, Großhaderner Strasse 9, Planegg-Martinsried 82152, Germany
| | - Ana Ogrinc
- Institute for Immunology, LMU Munich, Großhaderner Strasse 9, Planegg-Martinsried 82152, Germany
| | - Verena Steyer
- Institute for Immunology, LMU Munich, Großhaderner Strasse 9, Planegg-Martinsried 82152, Germany
| | - Stefanie Meier
- Institute for Immunology, LMU Munich, Großhaderner Strasse 9, Planegg-Martinsried 82152, Germany
| | - Ferdinand Simon
- Institute for Immunology, LMU Munich, Großhaderner Strasse 9, Planegg-Martinsried 82152, Germany
| | - Maria Wimmer
- Center of Allergy Environment (ZAUM), Helmholtz Center and TU Munich, Neuherberg 85764, Germany
| | - Andreas Blutke
- Section of Animal Pathology, Department of Veterinary Clinical Sciences, LMU Munich, Munich 80539, Germany
| | - Tobias Straub
- Bioinformatics core unit, BMC, LMU Munich, Großhaderner Strasse 9, Planegg-Munich 82152, Germany
| | | | - Esther Lutgens
- Institut für Prophylaxe und Epidemiologie der Kreislaufkrankheiten, LMU Munich, Munich 80336, Germany.,Department of Medical Biochemistry, AMC, Amsterdam 1105AZ, The Netherlands
| | - Peggy Marconi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara 44121, Italy
| | - Caspar Ohnmacht
- Center of Allergy Environment (ZAUM), Helmholtz Center and TU Munich, Neuherberg 85764, Germany
| | - Debora Garzetti
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, German Center for Infection Research (DZIF), Partner Site Munich, LMU Munich, Munich 80336, Germany
| | - Bärbel Stecher
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, German Center for Infection Research (DZIF), Partner Site Munich, LMU Munich, Munich 80336, Germany
| | - Thomas Brocker
- Institute for Immunology, LMU Munich, Großhaderner Strasse 9, Planegg-Martinsried 82152, Germany
| |
Collapse
|
203
|
Czarnewski P, Das S, Parigi SM, Villablanca EJ. Retinoic Acid and Its Role in Modulating Intestinal Innate Immunity. Nutrients 2017; 9:nu9010068. [PMID: 28098786 PMCID: PMC5295112 DOI: 10.3390/nu9010068] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 12/26/2016] [Accepted: 01/11/2017] [Indexed: 01/11/2023] Open
Abstract
Vitamin A (VA) is amongst the most well characterized food-derived nutrients with diverse immune modulatory roles. Deficiency in dietary VA has not only been associated with immune dysfunctions in the gut, but also with several systemic immune disorders. In particular, VA metabolite all-trans retinoic acid (atRA) has been shown to be crucial in inducing gut tropism in lymphocytes and modulating T helper differentiation. In addition to the widely recognized role in adaptive immunity, increasing evidence identifies atRA as an important modulator of innate immune cells, such as tolerogenic dendritic cells (DCs) and innate lymphoid cells (ILCs). Here, we focus on the role of retinoic acid in differentiation, trafficking and the functions of innate immune cells in health and inflammation associated disorders. Lastly, we discuss the potential involvement of atRA during the plausible crosstalk between DCs and ILCs.
Collapse
Affiliation(s)
- Paulo Czarnewski
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institutet and University Hospital, Stockholm 171-76, Sweden.
| | - Srustidhar Das
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institutet and University Hospital, Stockholm 171-76, Sweden.
| | - Sara M Parigi
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institutet and University Hospital, Stockholm 171-76, Sweden.
| | - Eduardo J Villablanca
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institutet and University Hospital, Stockholm 171-76, Sweden.
| |
Collapse
|
204
|
Veiga-Fernandes H, Mucida D. Neuro-Immune Interactions at Barrier Surfaces. Cell 2017; 165:801-11. [PMID: 27153494 DOI: 10.1016/j.cell.2016.04.041] [Citation(s) in RCA: 203] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Indexed: 12/23/2022]
Abstract
Multidirectional interactions between the nervous and immune systems have been documented in homeostasis and pathologies ranging from multiple sclerosis to autism, and from leukemia to acute and chronic inflammation. Recent studies have addressed this crosstalk using cell-specific targeting, novel sequencing, imaging, and analytical tools, shedding light on unappreciated mechanisms of neuro-immune regulation. This Review focuses on neuro-immune interactions at barrier surfaces-mostly the gut, but also including the skin and the airways, areas densely populated by neurons and immune cells that constantly sense and adapt to tissue-specific environmental challenges.
Collapse
Affiliation(s)
- Henrique Veiga-Fernandes
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Av. Prof. Egas Moniz, Edifício Egas Moniz, 1649-028 Lisboa, Portugal.
| | - Daniel Mucida
- Laboratory of Mucosal Immunology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
205
|
Sanders TJ, Yrlid U, Maloy KJ. Intestinal Mononuclear Phagocytes in Health and Disease. Microbiol Spectr 2017; 5:10.1128/microbiolspec.mchd-0047-2016. [PMID: 28102120 PMCID: PMC11687448 DOI: 10.1128/microbiolspec.mchd-0047-2016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Indexed: 12/20/2022] Open
Abstract
The intestine is the tissue of the body with the highest constitutive exposure to foreign antigen and is also a common entry portal for many local and systemic pathogens. Therefore, the local immune system has the unenviable task of balancing efficient responses to dangerous pathogens with tolerance toward beneficial microbiota and food antigens. As in most tissues, the decision between tolerance and immunity is critically governed by the activity of local myeloid cells. However, the unique challenges posed by the intestinal environment have necessitated the development of several specialized mononuclear phagocyte populations with distinct phenotypic and functional characteristics that have vital roles in maintaining barrier function and immune homeostasis in the intestine. Intestinal mononuclear phagocyte populations, comprising dendritic cells and macrophages, are crucial for raising appropriate active immune responses against ingested pathogens. Recent technical advances, including microsurgical approaches allowing collection of cells migrating in intestinal lymph, intravital microscopy, and novel gene-targeting approaches, have led to clearer distinctions between mononuclear phagocyte populations in intestinal tissue. In this review, we present an overview of the various subpopulations of intestinal mononuclear phagocytes and discuss their phenotypic and functional characteristics. We also outline their roles in host protection from infection and their regulatory functions in maintaining immune tolerance toward beneficial intestinal antigens.
Collapse
Affiliation(s)
| | - Ulf Yrlid
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, S-405 30 Gothenburg, Sweden
| | - Kevin J Maloy
- Sir William Dunn School of Pathology, Oxford, OX1 3RE, United Kingdom
| |
Collapse
|
206
|
Nowarski R, Jackson R, Flavell RA. The Stromal Intervention: Regulation of Immunity and Inflammation at the Epithelial-Mesenchymal Barrier. Cell 2017; 168:362-375. [DOI: 10.1016/j.cell.2016.11.040] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 10/25/2016] [Accepted: 11/22/2016] [Indexed: 12/24/2022]
|
207
|
Pakalniškytė D, Schraml BU. Tissue-Specific Diversity and Functions of Conventional Dendritic Cells. Adv Immunol 2017; 134:89-135. [DOI: 10.1016/bs.ai.2017.01.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
208
|
Abstract
ABSTRACT
The aim of this review is to provide a coherent framework for understanding dendritic cells (DCs). It has seven sections. The introduction provides an overview of the immune system and essential concepts, particularly for the nonspecialist reader. Next, the “History” section outlines the early evolution of ideas about DCs and highlights some sources of confusion that still exist today. The “Lineages” section then focuses on five different populations of DCs: two subsets of “classical” DCs, plasmacytoid DCs, monocyte-derived DCs, and Langerhans cells. It highlights some cellular and molecular specializations of each, and also notes other DC subsets that have been proposed. The following “Tissues” section discusses the distribution and behavior of different DC subsets within nonlymphoid and secondary lymphoid tissues that are connected by DC migration pathways between them. In the “Tolerance” section, the role of DCs in central and peripheral tolerance is considered, including their ability to drive the differentiation of different populations of regulatory T cells. In contrast, the “Immunity” section considers the roles of DCs in sensing of infection and tissue damage, the initiation of primary responses, the T-cell effector phase, and the induction of immunological memory. The concluding section provides some speculative ideas about the evolution of DCs. It also revisits earlier concepts of generation of diversity and clonal selection in terms of DCs driving the evolution of T-cell responses. Throughout, this review highlights certain areas of uncertainty and suggests some avenues for future investigation.
Collapse
|
209
|
Donaldson DS, Sehgal A, Rios D, Williams IR, Mabbott NA. Increased Abundance of M Cells in the Gut Epithelium Dramatically Enhances Oral Prion Disease Susceptibility. PLoS Pathog 2016; 12:e1006075. [PMID: 27973593 PMCID: PMC5156364 DOI: 10.1371/journal.ppat.1006075] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 11/17/2016] [Indexed: 02/07/2023] Open
Abstract
Many natural prion diseases of humans and animals are considered to be acquired through oral consumption of contaminated food or pasture. Determining the route by which prions establish host infection will identify the important factors that influence oral prion disease susceptibility and to which intervention strategies can be developed. After exposure, the early accumulation and replication of prions within small intestinal Peyer's patches is essential for the efficient spread of disease to the brain. To replicate within Peyer's patches, the prions must first cross the gut epithelium. M cells are specialised epithelial cells within the epithelia covering Peyer's patches that transcytose particulate antigens and microorganisms. M cell-development is dependent upon RANKL-RANK-signalling, and mice in which RANK is deleted only in the gut epithelium completely lack M cells. In the specific absence of M cells in these mice, the accumulation of prions within Peyer's patches and the spread of disease to the brain was blocked, demonstrating a critical role for M cells in the initial transfer of prions across the gut epithelium in order to establish host infection. Since pathogens, inflammatory stimuli and aging can modify M cell-density in the gut, these factors may also influence oral prion disease susceptibility. Mice were therefore treated with RANKL to enhance M cell density in the gut. We show that prion uptake from the gut lumen was enhanced in RANKL-treated mice, resulting in shortened survival times and increased disease susceptibility, equivalent to a 10-fold higher infectious titre of prions. Together these data demonstrate that M cells are the critical gatekeepers of oral prion infection, whose density in the gut epithelium directly limits or enhances disease susceptibility. Our data suggest that factors which alter M cell-density in the gut epithelium may be important risk factors which influence host susceptibility to orally acquired prion diseases.
Collapse
Affiliation(s)
- David S. Donaldson
- The Roslin Institute & Royal (Dick) School of Veterinary Sciences, University of Edinburgh, United Kingdom
| | - Anuj Sehgal
- The Roslin Institute & Royal (Dick) School of Veterinary Sciences, University of Edinburgh, United Kingdom
| | - Daniel Rios
- Dept. Pathology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Ifor R. Williams
- Dept. Pathology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Neil A. Mabbott
- The Roslin Institute & Royal (Dick) School of Veterinary Sciences, University of Edinburgh, United Kingdom
- * E-mail:
| |
Collapse
|
210
|
Williams AR, Klaver EJ, Laan LC, Ramsay A, Fryganas C, Difborg R, Kringel H, Reed JD, Mueller-Harvey I, Skov S, van Die I, Thamsborg SM. Co-operative suppression of inflammatory responses in human dendritic cells by plant proanthocyanidins and products from the parasitic nematode Trichuris suis. Immunology 2016; 150:312-328. [PMID: 27905107 DOI: 10.1111/imm.12687] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 10/29/2016] [Accepted: 11/01/2016] [Indexed: 12/20/2022] Open
Abstract
Interactions between dendritic cells (DCs) and environmental, dietary and pathogen antigens play a key role in immune homeostasis and regulation of inflammation. Dietary polyphenols such as proanthocyanidins (PAC) may reduce inflammation, and we therefore hypothesized that PAC may suppress lipopolysaccharide (LPS) -induced responses in human DCs and subsequent T helper type 1 (Th1) -type responses in naive T cells. Moreover, we proposed that, because DCs are likely to be exposed to multiple stimuli, the activity of PAC may synergise with other bioactive molecules that have anti-inflammatory activity, e.g. soluble products from the helminth parasite Trichuris suis (TsSP). We show that PAC are endocytosed by monocyte-derived DCs and selectively induce CD86 expression. Subsequently, PAC suppress the LPS-induced secretion of interleukin-6 (IL-6) and IL-12p70, while enhancing secretion of IL-10. Incubation of DCs with PAC did not affect lymphocyte proliferation; however, subsequent interferon-γ production was markedly suppressed, while IL-4 production was unaffected. The activity of PAC was confined to oligomers (degree of polymerization ≥ 4). Co-pulsing DCs with TsSP and PAC synergistically reduced secretion of tumour necrosis factor-α, IL-6 and IL-12p70 while increasing IL-10 secretion. Moreover, both TsSP and PAC alone induced Th2-associated OX40L expression in DCs, and together synergized to up-regulate OX40L. These data suggest that PAC induce an anti-inflammatory phenotype in human DCs that selectively down-regulates Th1 response in naive T cells, and that they also act cooperatively with TsSP. Our results indicate a novel interaction between dietary compounds and parasite products to influence immune function, and may suggest that combinations of PAC and TsSP can have therapeutic potential for inflammatory disorders.
Collapse
Affiliation(s)
- Andrew R Williams
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Elsenoor J Klaver
- Department of Molecular Cell Biology and Immunology, VU University Medical Centre, Amsterdam, the Netherlands
| | - Lisa C Laan
- Department of Molecular Cell Biology and Immunology, VU University Medical Centre, Amsterdam, the Netherlands
| | - Aina Ramsay
- Chemistry and Biochemistry Laboratory, University of Reading, Reading, UK
| | - Christos Fryganas
- Chemistry and Biochemistry Laboratory, University of Reading, Reading, UK
| | - Rolf Difborg
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Helene Kringel
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Jess D Reed
- Department of Animal Science, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Søren Skov
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - Irma van Die
- Department of Molecular Cell Biology and Immunology, VU University Medical Centre, Amsterdam, the Netherlands
| | - Stig M Thamsborg
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C, Denmark
| |
Collapse
|
211
|
Worbs T, Hammerschmidt SI, Förster R. Dendritic cell migration in health and disease. Nat Rev Immunol 2016; 17:30-48. [PMID: 27890914 DOI: 10.1038/nri.2016.116] [Citation(s) in RCA: 584] [Impact Index Per Article: 64.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dendritic cells (DCs) are potent and versatile antigen-presenting cells, and their ability to migrate is key for the initiation of protective pro-inflammatory as well as tolerogenic immune responses. Recent comprehensive studies have highlighted the importance of DC migration in the maintenance of immune surveillance and tissue homeostasis, and also in the pathogenesis of a range of diseases. In this Review, we summarize the anatomical, cellular and molecular factors that regulate the migration of different DC subsets in health and disease. In particular, we focus on new insights concerning the role of migratory DCs in the pathogenesis of diseases of the skin, intestine, lung, and brain, as well as in autoimmunity and atherosclerosis.
Collapse
Affiliation(s)
- Tim Worbs
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Swantje I Hammerschmidt
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| |
Collapse
|
212
|
Ren W, Wang K, Yin J, Chen S, Liu G, Tan B, Wu G, Bazer FW, Peng Y, Yin Y. Glutamine-Induced Secretion of Intestinal Secretory Immunoglobulin A: A Mechanistic Perspective. Front Immunol 2016; 7:503. [PMID: 27933057 PMCID: PMC5121228 DOI: 10.3389/fimmu.2016.00503] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 10/27/2016] [Indexed: 12/23/2022] Open
Abstract
Secretory immunoglobulin A (SIgA) is one important line of defense in the intestinal mucosal surface to protect the intestinal epithelium from enteric toxins and pathogenic microorganisms. Multiple factors, such as intestinal microbiota, intestinal cytokines, and nutrients are highly involved in production of SIgA in the intestine. Recently, glutamine has been shown to affect intestinal SIgA production; however, the underlying mechanism by which glutamine stimulates secretion of intestinal SIgA is unknown. Here, we review current knowledge regarding glutamine in intestinal immunity and show that glutamine-enhanced secretion of SIgA in the intestine may involve intestinal microbiota, intestinal antigen sampling and presentation, induction pathways for SIgA production by plasma cells (both T-dependent and T-independent pathway), and even transport of SIgA. Altogether, the glutamine-intestinal SIgA axis has broad therapeutic implications for intestinal SIgA-associated diseases, such as celiac disease, allergies, and inflammatory bowel disease.
Collapse
Affiliation(s)
- Wenkai Ren
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Kai Wang
- Institute of Apicultural Research (IAR), Chinese Academy of Agricultural Sciences (CAAS) , Beijing , China
| | - Jie Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Shuai Chen
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Gang Liu
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences , Changsha , China
| | - Bie Tan
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences , Changsha , China
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University , College Station, TX , USA
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University , College Station, TX , USA
| | - Yuanyi Peng
- Chongqing Key Laboratory of Forage & Herbivore, College of Animal Science and Technology, Southwest University , Chongqing , China
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China; College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
213
|
Okada T, Takahashi S, Ishida A, Ishigame H. In vivo multiphoton imaging of immune cell dynamics. Pflugers Arch 2016; 468:1793-1801. [PMID: 27659161 PMCID: PMC5138265 DOI: 10.1007/s00424-016-1882-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Revised: 09/07/2016] [Accepted: 09/12/2016] [Indexed: 12/20/2022]
Abstract
Multiphoton imaging has been utilized to analyze in vivo immune cell dynamics over the last 15 years. Particularly, it has deepened the understanding of how immune responses are organized by immune cell migration and interactions. In this review, we first describe the following technical advances in recent imaging studies that contributed to the new findings on the regulation of immune responses and inflammation. Improved multicolor imaging of immune cell behavior has revealed that their interactions are spatiotemporally coordinated to achieve efficient and long-term immunity. The use of photoactivatable and photoconvertible fluorescent proteins has increased duration and volume of cell tracking, even enabling the analysis of inter-organ migration of immune cells. In addition, visualization of immune cell activation using biosensors for intracellular calcium concentration and signaling molecule activities has started to give further mechanistic insights. Then, we also introduce recent imaging analyses of interactions between immune cells and non-immune cells including endothelial, fibroblastic, epithelial, and nerve cells. It is argued that future imaging studies that apply updated technical advances to analyze interactions between immune cells and non-immune cells will be important for thorough physiological understanding of the immune system.
Collapse
Affiliation(s)
- Takaharu Okada
- Laboratory for Tissue Dynamics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, 230-0045, Japan.
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Kanagawa, 230-0045, Japan.
- PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama, 332-0012, Japan.
| | - Sonoko Takahashi
- Laboratory for Tissue Dynamics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, 230-0045, Japan
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Kanagawa, 230-0045, Japan
| | - Azusa Ishida
- Laboratory for Tissue Dynamics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, 230-0045, Japan
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Kanagawa, 230-0045, Japan
| | - Harumichi Ishigame
- Laboratory for Tissue Dynamics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, 230-0045, Japan
| |
Collapse
|
214
|
Rossini V, Radulovic K, Riedel CU, Niess JH. Development of an Antigen-driven Colitis Model to Study Presentation of Antigens by Antigen Presenting Cells to T Cells. J Vis Exp 2016. [PMID: 27684040 DOI: 10.3791/54421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammation which affects the gastrointestinal tract (GIT). One of the best ways to study the immunological mechanisms involved during the disease is the T cell transfer model of colitis. In this model, immunodeficient mice (RAG(-/-) recipients) are reconstituted with naive CD4(+) T cells from healthy wild type hosts. This model allows examination of the earliest immunological events leading to disease and chronic inflammation, when the gut inflammation perpetuates but does not depend on a defined antigen. To study the potential role of antigen presenting cells (APCs) in the disease process, it is helpful to have an antigen-driven disease model, in which a defined commensal-derived antigen leads to colitis. An antigen driven-colitis model has hence been developed. In this model OT-II CD4(+) T cells, that can recognize only specific epitopes in the OVA protein, are transferred into RAG(-/-) hosts challenged with CFP-OVA-expressing E. coli. This model allows the examination of interactions between APCs and T cells in the lamina propria.
Collapse
Affiliation(s)
| | | | | | - Jan Hendrik Niess
- Division of Gastroenterology and Hepatology, University Hospital Basel;
| |
Collapse
|
215
|
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
|
216
|
Basson A, Trotter A, Rodriguez-Palacios A, Cominelli F. Mucosal Interactions between Genetics, Diet, and Microbiome in Inflammatory Bowel Disease. Front Immunol 2016; 7:290. [PMID: 27531998 PMCID: PMC4970383 DOI: 10.3389/fimmu.2016.00290] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 07/19/2016] [Indexed: 12/12/2022] Open
Abstract
Numerous reviews have discussed gut microbiota composition changes during inflammatory bowel diseases (IBD), particularly Crohn’s disease (CD). However, most studies address the observed effects by focusing on studying the univariate connection between disease and dietary-induced alterations to gut microbiota composition. The possibility that these effects may reflect a number of other interconnected (i.e., pantropic) mechanisms, activated in parallel, particularly concerning various bacterial metabolites, is in the process of being elucidated. Progress seems, however, hampered by various difficult-to-study factors interacting at the mucosal level. Here, we highlight some of such factors that merit consideration, namely: (1) the contribution of host genetics and diet in altering gut microbiome, and in turn, the crosstalk among secondary metabolic pathways; (2) the interdependence between the amount of dietary fat, the fatty acid composition, the effects of timing and route of administration on gut microbiota community, and the impact of microbiota-derived fatty acids; (3) the effect of diet on bile acid composition, and the modulator role of bile acids on the gut microbiota; (4) the impact of endogenous and exogenous intestinal micronutrients and metabolites; and (5) the need to consider food associated toxins and chemicals, which can introduce confounding immune modulating elements (e.g., antioxidant and phytochemicals in oils and proteins). These concepts, which are not mutually exclusive, are herein illustrated paying special emphasis on physiologically inter-related processes.
Collapse
Affiliation(s)
- Abigail Basson
- Digestive Health Research Institute, Case Western Reserve University , Cleveland, OH , USA
| | - Ashley Trotter
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, USA; University Hospitals Case Medical Center, Cleveland, OH, USA
| | | | - Fabio Cominelli
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, USA; University Hospitals Case Medical Center, Cleveland, OH, USA
| |
Collapse
|
217
|
Koscsó B, Bogunovic M. Analysis and Purification of Mouse Intestinal Dendritic Cell and Macrophage Subsets by Flow Cytometry. ACTA ACUST UNITED AC 2016; 114:14.39.1-14.39.14. [DOI: 10.1002/cpim.11] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Balázs Koscsó
- Penn State University College of Medicine Hershey Pennsylvania
| | | |
Collapse
|
218
|
Donaldson DS, Mabbott NA. The influence of the commensal and pathogenic gut microbiota on prion disease pathogenesis. J Gen Virol 2016; 97:1725-1738. [PMID: 27193137 DOI: 10.1099/jgv.0.000507] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Prion diseases are a unique group of transmissible, chronic, neurodegenerative disorders. Following peripheral exposure (e.g. oral), prions often accumulate first within the secondary lymphoid tissues before they infect the central nervous system (CNS). Prion replication within secondary lymphoid tissues is crucial for the efficient spread of disease to the CNS. Once within the CNS, the responses of innate immune cells within it can have a significant influence on neurodegeneration and disease progression. Recently, there have been substantial advances in our understanding of how cross-talk between the host and the vast community of commensal microorganisms present at barrier surfaces such as the gut influences the development and regulation of the host's immune system. These effects are evident not only in the mucosal immune system in the gut, but also in the CNS. The actions of this microbial community (the microbiota) have many important beneficial effects on host health, from metabolism of nutrients and regulation of host development to protection from pathogen infection. However, the microbiota can also have detrimental effects in some circumstances. In this review we discuss the many and varied interactions between prions, the host and the gut microbiota. Particular emphasis is given to the ways by which changes to the composition of the commensal gut microbiota or congruent pathogen infection may influence prion disease pathogenesis and/or disease susceptibility. Understanding how these factors influence prion pathogenesis and disease susceptibility is important for assessing the risk to infection and the design of novel opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- David S Donaldson
- The Roslin Institute and Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Edinburgh, UK
| | - Neil A Mabbott
- The Roslin Institute and Royal (Dick) School of Veterinary Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
219
|
Hilligan KL, Connor LM, Schmidt AJ, Ronchese F. Activation-Induced TIM-4 Expression Identifies Differential Responsiveness of Intestinal CD103+ CD11b+ Dendritic Cells to a Mucosal Adjuvant. PLoS One 2016; 11:e0158775. [PMID: 27379516 PMCID: PMC4933342 DOI: 10.1371/journal.pone.0158775] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 06/21/2016] [Indexed: 12/23/2022] Open
Abstract
Macrophage and dendritic cell (DC) populations residing in the intestinal lamina propria (LP) are highly heterogeneous and have disparate yet collaborative roles in the promotion of adaptive immune responses towards intestinal antigen. Under steady-state conditions, macrophages are efficient at acquiring antigen but are non-migratory. In comparison, intestinal DC are inefficient at antigen uptake but migrate to the mesenteric lymph nodes (mLN) where they present antigen to T cells. Whether such distinction in the roles of DC and macrophages in the uptake and transport of antigen is maintained under immunostimulatory conditions is less clear. Here we show that the scavenger and phosphatidylserine receptor T cell Immunoglobulin and Mucin (TIM)-4 is expressed by the majority of LP macrophages at steady-state, whereas DC are TIM-4 negative. Oral treatment with the mucosal adjuvant cholera toxin (CT) induces expression of TIM-4 on a proportion of CD103+ CD11b+ DC in the LP. TIM-4+ DC selectively express high levels of co-stimulatory molecules after CT treatment and are detected in the mLN a short time after appearing in the LP. Importantly, intestinal macrophages and DC expressing TIM-4 are more efficient than their TIM-4 negative counterparts at taking up apoptotic cells and soluble antigen ex vivo. Taken together, our results show that CT induces phenotypic changes to migratory intestinal DC that may impact their ability to take up local antigens and in turn promote the priming of mucosal immunity.
Collapse
Affiliation(s)
- Kerry L. Hilligan
- Malaghan Institute of Medical Research, Wellington, New Zealand
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Lisa M. Connor
- Malaghan Institute of Medical Research, Wellington, New Zealand
- * E-mail:
| | | | - Franca Ronchese
- Malaghan Institute of Medical Research, Wellington, New Zealand
| |
Collapse
|
220
|
McKenney ES, Kendall MM. Microbiota and pathogen 'pas de deux': setting up and breaking down barriers to intestinal infection. Pathog Dis 2016; 74:ftw051. [PMID: 27252177 PMCID: PMC5985477 DOI: 10.1093/femspd/ftw051] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 02/04/2016] [Accepted: 05/24/2016] [Indexed: 02/07/2023] Open
Abstract
The gut microbiota plays essential roles in human health and disease. In this review, we focus on the role of the intestinal microbiota in promoting resistance to infection by bacterial pathogens as well as how pathogens overcome this barrier. We discuss how the resident microbiota restricts growth and colonization of invading pathogens by limiting availability of nutrients and through generation of a hostile environment. Additionally, we examine how microbiota-derived signaling molecules interfere with bacterial virulence. In turn, we discuss how pathogens exploit non-competitive metabolites to replicate in vivo as well as to precisely control virulence and cause disease. This bacterial two step of creating and overcoming challenges important in preventing and establishing infection highlights the complexities of elucidating interactions between the commensal bacteria and pathogens. Better understanding of microbiota-pathogen interplay will have significant implications for developing novel therapeutics to treat infectious diseases.
Collapse
Affiliation(s)
- Elizabeth S McKenney
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Melissa M Kendall
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| |
Collapse
|
221
|
Rios D, Wood MB, Li J, Chassaing B, Gewirtz AT, Williams IR. Antigen sampling by intestinal M cells is the principal pathway initiating mucosal IgA production to commensal enteric bacteria. Mucosal Immunol 2016; 9:907-16. [PMID: 26601902 PMCID: PMC4917673 DOI: 10.1038/mi.2015.121] [Citation(s) in RCA: 186] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 10/08/2015] [Indexed: 02/04/2023]
Abstract
Secretory IgA (SIgA) directed against gut resident bacteria enables the mammalian mucosal immune system to establish homeostasis with the commensal gut microbiota after weaning. Germinal centers (GCs) in Peyer's patches (PPs) are the principal inductive sites where naive B cells specific for bacterial antigens encounter their cognate antigens and receive T-cell help driving their differentiation into IgA-producing plasma cells. We investigated the role of antigen sampling by intestinal M cells in initiating the SIgA response to gut bacteria by developing mice in which receptor activator of nuclear factor-κB ligand (RANKL)-dependent M-cell differentiation was abrogated by conditional deletion of Tnfrsf11a in the intestinal epithelium. Mice without intestinal M cells had profound delays in PP GC maturation and emergence of lamina propria IgA plasma cells, resulting in diminished levels of fecal SIgA that persisted into adulthood. We conclude that M-cell-mediated sampling of commensal bacteria is a required initial step for the efficient induction of intestinal SIgA.
Collapse
Affiliation(s)
- D Rios
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - M B Wood
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - J Li
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - B Chassaing
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - A T Gewirtz
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - I R Williams
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
222
|
Bretou M, Kumari A, Malbec O, Moreau HD, Obino D, Pierobon P, Randrian V, Sáez PJ, Lennon-Duménil AM. Dynamics of the membrane-cytoskeleton interface in MHC class II-restricted antigen presentation. Immunol Rev 2016; 272:39-51. [DOI: 10.1111/imr.12429] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Marine Bretou
- Inserm U932, Institut Curie; ANR-10-IDEX-0001-02 PSL* and ANR-11-LABX-0043; Paris France
| | - Anita Kumari
- Inserm U932, Institut Curie; ANR-10-IDEX-0001-02 PSL* and ANR-11-LABX-0043; Paris France
| | - Odile Malbec
- Inserm U932, Institut Curie; ANR-10-IDEX-0001-02 PSL* and ANR-11-LABX-0043; Paris France
| | - Hélène D. Moreau
- Inserm U932, Institut Curie; ANR-10-IDEX-0001-02 PSL* and ANR-11-LABX-0043; Paris France
| | - Dorian Obino
- Inserm U932, Institut Curie; ANR-10-IDEX-0001-02 PSL* and ANR-11-LABX-0043; Paris France
| | - Paolo Pierobon
- Inserm U932, Institut Curie; ANR-10-IDEX-0001-02 PSL* and ANR-11-LABX-0043; Paris France
| | - Violaine Randrian
- Inserm U932, Institut Curie; ANR-10-IDEX-0001-02 PSL* and ANR-11-LABX-0043; Paris France
| | - Pablo J. Sáez
- Inserm U932, Institut Curie; ANR-10-IDEX-0001-02 PSL* and ANR-11-LABX-0043; Paris France
| | | |
Collapse
|
223
|
Barreiro O, Cibrian D, Clemente C, Alvarez D, Moreno V, Valiente Í, Bernad A, Vestweber D, Arroyo AG, Martín P, von Andrian UH, Sánchez Madrid F. Pivotal role for skin transendothelial radio-resistant anti-inflammatory macrophages in tissue repair. eLife 2016; 5. [PMID: 27304075 PMCID: PMC4961461 DOI: 10.7554/elife.15251] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 06/13/2016] [Indexed: 12/20/2022] Open
Abstract
Heterogeneity and functional specialization among skin-resident macrophages are incompletely understood. In this study, we describe a novel subset of murine dermal perivascular macrophages that extend protrusions across the endothelial junctions in steady-state and capture blood-borne macromolecules. Unlike other skin-resident macrophages that are reconstituted by bone marrow-derived progenitors after a genotoxic insult, these cells are replenished by an extramedullary radio-resistant and UV-sensitive Bmi1+ progenitor. Furthermore, they possess a distinctive anti-inflammatory transcriptional profile, which cannot be polarized under inflammatory conditions, and are involved in repair and remodeling functions for which other skin-resident macrophages appear dispensable. Based on all their properties, we define these macrophages as Skin Transendothelial Radio-resistant Anti-inflammatory Macrophages (STREAM) and postulate that their preservation is important for skin homeostasis. DOI:http://dx.doi.org/10.7554/eLife.15251.001 The skin forms an essential barrier that defends our body from external damage. For this reason, it is important to understand the complex mechanisms involved in healing wounds and maintaining healthy skin. This could allow us to find effective treatments for skin diseases such as atopic dermatitis and psoriasis. Immune cells called macrophages can protect the body in different ways depending on the signals they receive. Their protective roles include killing microbes that may cause disease, and helping to repair damaged tissues. Barreiro et al. have now investigated the roles of the macrophages in the skin by means of a number of complementary techniques, including a method called intravital microscopy that can image cells in a living organism. The experiments revealed that a division of labor exists among the macrophages that reside in the skin of mice. Some macrophages help to trigger inflammatory responses in the skin. These immune cells disappear after being exposed to ionizing radiation (such as that used to treat cancer) but can be replaced via a bone marrow transplant. Other macrophages that help to repair tissues can survive being exposed to ionizing radiation but cannot resist high levels of ultraviolet light. Both types of macrophages perform unique and essential roles, and both types are necessary for maintaining healthy skin. Barreiro et al. also discovered that the skin macrophages that help to repair tissues have the ability to move into blood vessels and take up substances from the blood. A question for future investigation is whether the macrophages perform this scavenging process to trigger a protective immune response in the nearby skin. DOI:http://dx.doi.org/10.7554/eLife.15251.002
Collapse
Affiliation(s)
- Olga Barreiro
- Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Danay Cibrian
- Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain.,Servicio de Inmunología, Hospital de la Princesa, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto Investigación Sanitaria Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| | - Cristina Clemente
- Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - David Alvarez
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, United States
| | - Vanessa Moreno
- Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Íñigo Valiente
- Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Antonio Bernad
- Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | | | - Alicia G Arroyo
- Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Pilar Martín
- Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Ulrich H von Andrian
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, United States.,Ragon Institute of MGH, MIT and Harvard, Cambridge, United States
| | - Francisco Sánchez Madrid
- Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain.,Servicio de Inmunología, Hospital de la Princesa, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto Investigación Sanitaria Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
224
|
Allen F, Tong AA, Huang AY. Unique Transcompartmental Bridge: Antigen-Presenting Cells Sampling across Endothelial and Mucosal Barriers. Front Immunol 2016; 7:231. [PMID: 27375624 PMCID: PMC4901051 DOI: 10.3389/fimmu.2016.00231] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 05/27/2016] [Indexed: 12/13/2022] Open
Abstract
Potentially harmful pathogens can gain access to tissues and organ systems through body sites that are in direct contact with the outside environment, such as the skin, the gut, and the airway mucosa. Antigen-presenting cells (APCs) represent a bridge between the innate and adaptive immunity, and their capacity for constant immune surveillance and rapid sampling of incoming pathogens and other potentially harmful antigens is central for mounting an effective and robust protective host response. The classical view is that APCs perform this task efficiently within the tissue to sense invading agents intra-compartmentally. However, recent data based on high resolution imaging support an additional transcompartmental surveillance behavior by APC by reaching across intact physical barriers. In this review, we summarize intravital microscopic evidences of APC to sample antigens transcompartmentally at the gut mucosa and other body sites.
Collapse
Affiliation(s)
- Frederick Allen
- Department of Pathology, Case Western Reserve University School of Medicine , Cleveland, OH , USA
| | - Alexander A Tong
- Department of Pathology, Case Western Reserve University School of Medicine , Cleveland, OH , USA
| | - Alex Y Huang
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Angie Fowler AYA Cancer Institute, University Hospitals Rainbow Babies & Children's Hospital, Cleveland, OH, USA
| |
Collapse
|
225
|
Ihara S, Hirata Y, Serizawa T, Suzuki N, Sakitani K, Kinoshita H, Hayakawa Y, Nakagawa H, Ijichi H, Tateishi K, Koike K. TGF-β Signaling in Dendritic Cells Governs Colonic Homeostasis by Controlling Epithelial Differentiation and the Luminal Microbiota. THE JOURNAL OF IMMUNOLOGY 2016; 196:4603-4613. [DOI: 10.4049/jimmunol.1502548] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Abstract
Dendritic cells (DCs) mediate host immune responses to gut microbes and play critical roles in inflammatory bowel disease. In this study, we examined the role of TGF-β signaling in DCs in colonic homeostasis. CD11c-cre Tgfbr2fl/fl mice developed spontaneous colitis, and CD11c-cre Tgfbr2fl/+ mice exhibited susceptibility to dextran sulfate sodium–induced colitis. Colitis in these mice was characterized by goblet cell depletion and dysbiosis caused by Enterobacteriaceae enrichment. Wild-type mice gavaged with Enterobacteriaceae from CD11c-cre Tgfbr2fl/fl mice feces showed severe colitis after dextran sulfate sodium treatment, whereas those treated with Notch inhibitor exhibited attenuated colonic injury with increased goblet cell numbers, thickened mucus layer, and fewer fecal Enterobacteriaceae. Wild-type mice transplanted with CD11c-cre Tgfbr2fl/fl bone marrow developed colitis showing increased Jagged1 and Jagged2 in DCs, increased Hes1 levels in epithelium, and goblet cell depletion. These findings suggest that TGF-β signaling in DCs regulates intestinal homeostasis by modulating epithelial cell differentiation and fecal microbiota.
Collapse
Affiliation(s)
- Sozaburo Ihara
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Yoshihiro Hirata
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Takako Serizawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Nobumi Suzuki
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Kosuke Sakitani
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Hiroto Kinoshita
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Yoku Hayakawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Hayato Nakagawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Hideaki Ijichi
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Keisuke Tateishi
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| |
Collapse
|
226
|
Ohnmacht C. Tolerance to the Intestinal Microbiota Mediated by ROR(γt)(+) Cells. Trends Immunol 2016; 37:477-486. [PMID: 27255270 DOI: 10.1016/j.it.2016.05.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 05/03/2016] [Accepted: 05/04/2016] [Indexed: 12/21/2022]
Abstract
Harmless microbes colonizing the gut require the establishment of a well-equilibrated symbiosis between this microbiota and its host. However, the immune system is primed to recognize both conserved microbial patterns and foreign antigens, and therefore developed strong tolerance mechanisms to prevent potential fatal immune reactivity to symbiotic microbes. The transcription factor RAR-related orphan-like γt [ROR(γt); encoded by Rorc] plays a key role in the gut for lymphoid tissue organogenesis, development of innate lymphoid cells type 3 (ILC3s) and proinflammatory type 17 T helper (Th17) cells. Surprisingly, recent research has revealed a contribution of ROR(γt)-expressing cells in a variety of tolerance mechanisms in both the innate and adaptive immune system.
Collapse
Affiliation(s)
- Caspar Ohnmacht
- Center of Allergy and Environment (ZAUM), Technische Universität and Helmholtz Center Munich, Biedersteiner Strasse 29, 80802 Munich, Germany.
| |
Collapse
|
227
|
Study of dendritic cell migration using micro-fabrication. J Immunol Methods 2016; 432:30-4. [DOI: 10.1016/j.jim.2015.12.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 11/13/2015] [Accepted: 12/08/2015] [Indexed: 01/22/2023]
|
228
|
Translational aspects of the microbiome-to be exploited. Cell Biol Toxicol 2016; 32:153-6. [PMID: 27098154 DOI: 10.1007/s10565-016-9320-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Accepted: 03/21/2016] [Indexed: 12/19/2022]
|
229
|
Cunningham AL, Guentzel MN, Yu JJ, Hung CY, Forsthuber TG, Navara CS, Yagita H, Williams IR, Klose KE, Eaves-Pyles TD, Arulanandam BP. M-Cells Contribute to the Entry of an Oral Vaccine but Are Not Essential for the Subsequent Induction of Protective Immunity against Francisella tularensis. PLoS One 2016; 11:e0153402. [PMID: 27100824 PMCID: PMC4839702 DOI: 10.1371/journal.pone.0153402] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 03/29/2016] [Indexed: 01/01/2023] Open
Abstract
M-cells (microfold cells) are thought to be a primary conduit of intestinal antigen trafficking. Using an established neutralizing anti-RANKL (Receptor Activator of NF-κB Ligand) antibody treatment to transiently deplete M-cells in vivo, we sought to determine whether intestinal M-cells were required for the effective induction of protective immunity following oral vaccination with ΔiglB (a defined live attenuated Francisella novicida mutant). M-cell depleted, ΔiglB-vaccinated mice exhibited increased (but not significant) morbidity and mortality following a subsequent homotypic or heterotypic pulmonary F. tularensis challenge. No significant differences in splenic IFN-γ, IL-2, or IL-17 or serum antibody (IgG1, IgG2a, IgA) production were observed compared to non-depleted, ΔiglB-vaccinated animals suggesting complementary mechanisms for ΔiglB entry. Thus, we examined other possible routes of gastrointestinal antigen sampling following oral vaccination and found that ΔiglB co-localized to villus goblet cells and enterocytes. These results provide insight into the role of M-cells and complementary pathways in intestinal antigen trafficking that may be involved in the generation of optimal immunity following oral vaccination.
Collapse
Affiliation(s)
- Aimee L. Cunningham
- Department of Biology, South Texas Center for Emerging Infectious Disease, University of Texas at San Antonio, San Antonio, Texas, United States of America
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - M. Neal Guentzel
- Department of Biology, South Texas Center for Emerging Infectious Disease, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Jieh-Juen Yu
- Department of Biology, South Texas Center for Emerging Infectious Disease, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Chiung-Yu Hung
- Department of Biology, South Texas Center for Emerging Infectious Disease, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Thomas G. Forsthuber
- Department of Biology, South Texas Center for Emerging Infectious Disease, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Christopher S. Navara
- Department of Biology, South Texas Center for Emerging Infectious Disease, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Hideo Yagita
- Department of Immunology, Juntendo University, Tokyo, Japan
| | - Ifor R. Williams
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Karl E. Klose
- Department of Biology, South Texas Center for Emerging Infectious Disease, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Tonyia D. Eaves-Pyles
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Bernard P. Arulanandam
- Department of Biology, South Texas Center for Emerging Infectious Disease, University of Texas at San Antonio, San Antonio, Texas, United States of America
- * E-mail:
| |
Collapse
|
230
|
Mononuclear phagocytes contribute to intestinal invasion and dissemination of Yersinia enterocolitica. Int J Med Microbiol 2016; 306:357-66. [PMID: 27107739 DOI: 10.1016/j.ijmm.2016.04.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 04/05/2016] [Accepted: 04/14/2016] [Indexed: 12/30/2022] Open
Abstract
Enteropathogenic Yersinia enterocolitica (Ye) enters the host via contaminated food. After colonisation of the small intestine Ye invades the Peyer's patches (PPs) via M cells and disseminates to the mesenteric lymph nodes (MLNs), spleen and liver. Whether Ye uses other invasion routes and which pathogenicity factors are required remains elusive. Oral infection of lymphotoxin-β-receptor deficient mice lacking PPs and MLNs with Ye revealed similar bacterial load in the spleen 1h post infection as wild-type mice, demonstrating a PP-independent dissemination route for Ye. Immunohistological analysis of the small intestine revealed Ye in close contact with mononuclear phagocytes (MPs), specifically CX3CR1(+) monocyte-derived cells (MCs) as well as CD103(+) dendritic cells (DCs). This finding was confirmed by flow cytometry and imaging flow cytometry analysis of lamina propria (LP) leukocytes showing CD103(+) DCs and MCs with intracellular Ye. Uptake of Ye by LP CD103(+) DCs and MCs was dependent on the pathogenicity factor invasin, whereas the adhesin YadA was dispensable as demonstrated by Ye deletion mutants. Furthermore, Ye were found exclusively associated with CD103(+) DCs in the MLNs from wild-type mice, but not from CCR7(-/-) mice, demonstrating a CCR7 dependent transport of Ye by CD103(+) DCs from LP to the MLNs. In contrast, dissemination of Ye to the spleen was dependent on MCs as significantly less Ye could be recovered from the spleen of CX3CR1(GFP/GFP) mice compared to wild-type mice. Altogether, MCs and CD103(+) DCs contribute to immediate invasion and dissemination of Ye. This together with data from other bacteria suggests MPs as general pathogenic entry site in the intestine.
Collapse
|
231
|
Tanoue T, Atarashi K, Honda K. Development and maintenance of intestinal regulatory T cells. Nat Rev Immunol 2016; 16:295-309. [PMID: 27087661 DOI: 10.1038/nri.2016.36] [Citation(s) in RCA: 415] [Impact Index Per Article: 46.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Gut-resident forkhead box P3 (FOXP3)(+)CD4(+) regulatory T cells (Treg cells) are distinct from those in other organs and have gut-specific phenotypes and functions. Whereas Treg cells in other organs have T cell receptors (TCRs) specific for self antigens, intestinal Treg cells have a distinct set of TCRs that are specific for intestinal antigens, and these cells have pivotal roles in the suppression of immune responses against harmless dietary antigens and commensal microorganisms. The differentiation, migration and maintenance of intestinal Treg cells are controlled by specific signals from the local environment. In particular, certain members of the microbiota continuously provide antigens and immunoregulatory small molecules that modulate intestinal Treg cells. Understanding the development and the maintenance of intestinal Treg cells provides important insights into disease-relevant host-microorganism interactions.
Collapse
Affiliation(s)
- Takeshi Tanoue
- RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Kanagawa 230-0045, Japan
| | - Koji Atarashi
- RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Kanagawa 230-0045, Japan.,Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kenya Honda
- RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Kanagawa 230-0045, Japan.,Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan.,AMED-CREST, Japan Agency for Medical Research and Development, Tokyo 100-0004, Japan
| |
Collapse
|
232
|
Ablating the aryl hydrocarbon receptor (AhR) in CD11c+ cells perturbs intestinal epithelium development and intestinal immunity. Sci Rep 2016; 6:23820. [PMID: 27068235 PMCID: PMC4828637 DOI: 10.1038/srep23820] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 03/09/2016] [Indexed: 12/22/2022] Open
Abstract
Diet and microbiome derived indole derivatives are known to activate the ligand induced transcription factor, the Aryl hydrocarbon Receptor (AhR). While the current understanding of AhR biology has confirmed its role in mucosal lymphocytes, its function in intestinal antigen presenting cells (APCs) is poorly understood. Here, we report that Cre-mediated deletion of AhR in CD11c-expressing cells in C57/BL6 mice is associated with altered intestinal epithelial morphogenesis in vivo. Moreover, when co-cultured with AhR-deficient DCs ex vivo, intestinal organoids showed reduced SRY (sex determining region Y)-box 9 and increased Mucin 2 expression, which correlates with reduced Paneth cells and increased goblet cell differentiation, similar to the data obtained in vivo. Further, characterization of intestinal APC subsets, devoid of AhR, revealed an expression pattern associated with aberrant intrinsic Wnt pathway regulation. At a functional level, the loss of AhR in APCs resulted in a dysfunctional epithelial barrier, associated with a more aggressive chemically induced colitis compared to wild type animals. Our results are consistent with a model whereby the AhR signalling pathway may participate in the regulation of innate immunity through intestinal epithelium development and mucosal immunity.
Collapse
|
233
|
Hudson LE, McDermott CD, Stewart TP, Hudson WH, Rios D, Fasken MB, Corbett AH, Lamb TJ. Characterization of the Probiotic Yeast Saccharomyces boulardii in the Healthy Mucosal Immune System. PLoS One 2016; 11:e0153351. [PMID: 27064405 PMCID: PMC4827847 DOI: 10.1371/journal.pone.0153351] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 03/28/2016] [Indexed: 12/14/2022] Open
Abstract
The probiotic yeast Saccharomyces boulardii has been shown to ameliorate disease severity in the context of many infectious and inflammatory conditions. However, use of S. boulardii as a prophylactic agent or therapeutic delivery vector would require delivery of S. boulardii to a healthy, uninflamed intestine. In contrast to inflamed mucosal tissue, the diverse microbiota, intact epithelial barrier, and fewer inflammatory immune cells within the healthy intestine may all limit the degree to which S. boulardii contacts and influences the host mucosal immune system. Understanding the nature of these interactions is crucial for application of S. boulardii as a prophylactic agent or therapeutic delivery vehicle. In this study, we explore both intrinsic and immunomodulatory properties of S. boulardii in the healthy mucosal immune system. Genomic sequencing and morphological analysis of S. boulardii reveals changes in cell wall components compared to non-probiotic S. cerevisiae that may partially account for probiotic functions of S. boulardii. Flow cytometry and immunohistochemistry demonstrate limited S. boulardii association with murine Peyer’s patches. We also show that although S. boulardii induces a systemic humoral immune response, this response is small in magnitude and not directed against S. boulardii itself. RNA-seq of the draining mesenteric lymph nodes indicates that even repeated administration of S. boulardii induces few transcriptional changes in the healthy intestine. Together these data strongly suggest that interaction between S. boulardii and the mucosal immune system in the healthy intestine is limited, with important implications for future work examining S. boulardii as a prophylactic agent and therapeutic delivery vehicle.
Collapse
Affiliation(s)
- Lauren E. Hudson
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Courtney D. McDermott
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Taryn P. Stewart
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States of America
| | - William H. Hudson
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Daniel Rios
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Milo B. Fasken
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Anita H. Corbett
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Tracey J. Lamb
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States of America
- * E-mail:
| |
Collapse
|
234
|
Mohammed J, Beura LK, Bobr A, Astry B, Chicoine B, Kashem SW, Welty NE, Igyártó BZ, Wijeyesinghe S, Thompson EA, Matte C, Bartholin L, Kaplan A, Sheppard D, Bridges AG, Shlomchik WD, Masopust D, Kaplan DH. Stromal cells control the epithelial residence of DCs and memory T cells by regulated activation of TGF-β. Nat Immunol 2016; 17:414-21. [PMID: 26901152 PMCID: PMC5135085 DOI: 10.1038/ni.3396] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/05/2016] [Indexed: 01/02/2023]
Abstract
Cells of the immune system that reside in barrier epithelia provide a first line of defense against pathogens. Langerhans cells (LCs) and CD8(+) tissue-resident memory T cells (TRM cells) require active transforming growth factor-β1 (TGF-β) for epidermal residence. Here we found that integrins αvβ6 and αvβ8 were expressed in non-overlapping patterns by keratinocytes (KCs) and maintained the epidermal residence of LCs and TRM cells by activating latent TGF-β. Similarly, the residence of dendritic cells and TRM cells in the small intestine epithelium also required αvβ6. Treatment of the skin with ultraviolet irradiation decreased integrin expression on KCs and reduced the availability of active TGF-β, which resulted in LC migration. Our data demonstrated that regulated activation of TGF-β by stromal cells was able to directly control epithelial residence of cells of the immune system through a novel mechanism of intercellular communication.
Collapse
Affiliation(s)
- Javed Mohammed
- Department of Dermatology, Center for Immunology, University of Minnesota, Minneapolis, Minnesota USA
| | - Lalit K Beura
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, Minnesota USA
| | - Aleh Bobr
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Brian Astry
- Department of Dermatology, Center for Immunology, University of Minnesota, Minneapolis, Minnesota USA
| | - Brian Chicoine
- Department of Dermatology, Center for Immunology, University of Minnesota, Minneapolis, Minnesota USA
| | - Sakeen W Kashem
- Department of Dermatology, Center for Immunology, University of Minnesota, Minneapolis, Minnesota USA
| | - Nathan E Welty
- Department of Dermatology, Center for Immunology, University of Minnesota, Minneapolis, Minnesota USA
| | - Botond Z Igyártó
- Department of Dermatology, Center for Immunology, University of Minnesota, Minneapolis, Minnesota USA
| | - Sathi Wijeyesinghe
- Department of Dermatology, Center for Immunology, University of Minnesota, Minneapolis, Minnesota USA
| | - Emily A Thompson
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, Minnesota USA
| | - Catherine Matte
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Laurent Bartholin
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Lyon, France
| | - Alesia Kaplan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Dean Sheppard
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Alina G Bridges
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Warren D Shlomchik
- Department of Medicine, University of Pittsburgh Cancer Center Institute, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh Cancer Center Institute, Pittsburgh, Pennsylvania, USA
| | - David Masopust
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota, Minneapolis, Minnesota USA
| | - Daniel H Kaplan
- Department of Dermatology, Center for Immunology, University of Minnesota, Minneapolis, Minnesota USA
- Department of Dermatology, University of Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pennsylvania, USA
| |
Collapse
|
235
|
Stolp B, Melican K. Microbial pathogenesis revealed by intravital microscopy: pros, cons and cautions. FEBS Lett 2016; 590:2014-26. [PMID: 26938770 DOI: 10.1002/1873-3468.12122] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 02/15/2016] [Accepted: 02/29/2016] [Indexed: 12/22/2022]
Abstract
Intravital multiphoton imaging allows visualization of infections and pathogenic mechanisms within intact organs in their physiological context. Today, most organs of mice and rats are applicable to in vivo or ex vivo imaging, opening completely new avenues for many researchers. Advances in fluorescent labeling of pathogens and infected cells, as well as improved small animal models for human pathogens, led to the increased application of in vivo imaging in infectious diseases research in recent years. Here, we review the latest literature on intravital or ex vivo imaging of viral and bacterial infections and critically discuss requirements, benefits and drawbacks of applied animal models, labeling strategies, and imaged organs.
Collapse
Affiliation(s)
- Bettina Stolp
- Heidelberg University Hospital, Center of Infectious Diseases, Integrative Virology, Heidelberg, Germany
| | - Keira Melican
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
236
|
Esterházy D, Loschko J, London M, Jove V, Oliveira TY, Mucida D. Classical dendritic cells are required for dietary antigen-mediated induction of peripheral T(reg) cells and tolerance. Nat Immunol 2016; 17:545-55. [PMID: 27019226 PMCID: PMC4837106 DOI: 10.1038/ni.3408] [Citation(s) in RCA: 211] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 01/26/2016] [Indexed: 12/12/2022]
Abstract
Oral tolerance prevents pathological inflammatory responses towards innocuous foreign antigens via peripheral regulatory T cells (pTreg cells). However, whether a particular subset of antigen-presenting cells (APCs) is required during dietary antigen exposure to instruct naïve CD4+ T cells to differentiate into pTreg cells has not been defined. Using myeloid lineage-specific APC depletion in mice, we found that monocyte-derived APCs are dispensable, while classical dendritic cells (cDCs) are critical for pTreg cell induction and oral tolerance. CD11b− cDCs from the gut-draining lymph nodes efficiently induced pTreg cells, and conversely, loss of IRF8-dependent CD11b− cDCs impaired their polarization, although oral tolerance remained intact. These data reveal the hierarchy of cDC subsets in pTreg cell induction and their redundancy during oral tolerance development.
Collapse
Affiliation(s)
- Daria Esterházy
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, New York, USA
| | - Jakob Loschko
- Laboratory of Molecular Immunology, The Rockefeller University, New York, New York, USA
| | - Mariya London
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, New York, USA
| | - Veronica Jove
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, New York, USA
| | - Thiago Y Oliveira
- Laboratory of Molecular Immunology, The Rockefeller University, New York, New York, USA
| | - Daniel Mucida
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, New York, USA
| |
Collapse
|
237
|
Abstract
The onslaught of foreign antigens carried by spermatozoa into the epididymis, an organ that has not demonstrated immune privilege, a decade or more after the establishment of central immune tolerance presents a unique biological challenge. Historically, the physical confinement of spermatozoa to the epididymal tubule enforced by a tightly interwoven wall of epithelial cells was considered sufficient enough to prevent cross talk between gametes and the immune system and, ultimately, autoimmune destruction. The discovery of an intricate arrangement of mononuclear phagocytes (MPs) comprising dendritic cells and macrophages in the murine epididymis suggests that we may have underestimated the existence of a sophisticated mucosal immune system in the posttesticular environment. This review consolidates our current knowledge of the physiology of MPs in the steady state epididymis and speculates on possible interactions between auto-antigenic spermatozoa, pathogens and the immune system by drawing on what is known about the immune system in the intestinal mucosa. Ultimately, further investigation will provide valuable information regarding the origins of pathologies arising as a result of autoimmune or inflammatory responses in the epididymis, including epididymitis and infertility.
Collapse
Affiliation(s)
- Nicolas Da Silva
- Massachusetts General Hospital and Harvard Medical School, Division of Nephrology, Center for Systems Biology, Boston, Massachusetts, USA
| | | |
Collapse
|
238
|
Mou C, Zhu L, Xing X, Lin J, Yang Q. Immune responses induced by recombinant Bacillus subtilis expressing the spike protein of transmissible gastroenteritis virus in pigs. Antiviral Res 2016; 131:74-84. [PMID: 26988122 DOI: 10.1016/j.antiviral.2016.02.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 01/25/2016] [Accepted: 02/01/2016] [Indexed: 01/08/2023]
Abstract
Transmissible gastroenteritis (TGE) causes severe diarrhea in suckling piglets, results in enormous economic loss in swine-producing areas of the world. To develop an effective, safe, and convenient vaccine for the prevention of TGE, we have constructed a recombinant Bacillus subtilis strain (B. subtilis CotGSG) displaying the transmissible gastroenteritis virus (TGEV) spike (S) protein and discussed its immune function to intestinal submucosal dendritic cells (DCs). Our results showed that the recombinant B. subtilis had the ability to recruit more DCs to sample B. subtilis CotGSG, migrate to MLNs, and induce immune responses. Immunized piglets with B. subtilis CotGSG could significantly elevate the specific SIgA titers in feces, IgG titers and neutralizing antibodies in serum. Collectively, our results suggested that recombinant B. subtilis CotGSG expressing the TGEV S protein could effectively induce immune responses via DCs, and provided a perspective on potential novel strategy and approach that may be applicable to the development of the next generation of TGEV vaccines.
Collapse
Affiliation(s)
- Chunxiao Mou
- College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, PR China.
| | - Liqi Zhu
- College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, PR China.
| | - Xianping Xing
- College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, PR China.
| | - Jian Lin
- College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, PR China.
| | - Qian Yang
- College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, PR China.
| |
Collapse
|
239
|
Muzaki ARBM, Tetlak P, Sheng J, Loh SC, Setiagani YA, Poidinger M, Zolezzi F, Karjalainen K, Ruedl C. Intestinal CD103(+)CD11b(-) dendritic cells restrain colitis via IFN-γ-induced anti-inflammatory response in epithelial cells. Mucosal Immunol 2016; 9:336-51. [PMID: 26174764 PMCID: PMC4801902 DOI: 10.1038/mi.2015.64] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 06/15/2015] [Indexed: 02/04/2023]
Abstract
A crosstalk between commensals, gut immune cells, and colonic epithelia is required for a proper function of intestinal mucosal barrier. Here we investigated the importance of two distinct intestinal dendritic cell (DC) subsets in controlling intestinal inflammation. We show that Clec9A-diphtheria toxin receptor (DTR) mice after depletion of CD103(+)CD11b(-) DCs developed severe, low-dose dextran sodium sulfate (DSS)-induced colitis, whereas the lack of CD103(+)CD11b(+) DCs in Clec4a4-DTR mice did not exacerbate intestinal inflammation. The CD103(+)CD11b(-) DC subset has gained a functional specialization that able them to repress inflammation via several epithelial interferon-γ (IFN-γ)-induced proteins. Among others, we identified that epithelial IDO1 and interleukin-18-binding protein (IL-18bp) were strongly modulated by CD103(+)CD11b(-) DCs. Through its preferential property to express IL-12 and IL-15, this particular DC subset can induce lymphocytes in colonic lamina propria and in epithelia to secrete IFN-γ that then can trigger a reversible early anti-inflammatory response in intestinal epithelial cells.
Collapse
Affiliation(s)
- A R B M Muzaki
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - P Tetlak
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - J Sheng
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - S C Loh
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Y A Setiagani
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - M Poidinger
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - F Zolezzi
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - K Karjalainen
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - C Ruedl
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore,()
| |
Collapse
|
240
|
Abstract
The intestinal mucosa is a particularly dynamic environment in which the host constantly interacts with trillions of commensal microorganisms, known as the microbiota, and periodically interacts with pathogens of diverse nature. In this Review, we discuss how mucosal immunity is controlled in response to enteric bacterial pathogens, with a focus on the species that cause morbidity and mortality in humans. We explain how the microbiota can shape the immune response to pathogenic bacteria, and we detail innate and adaptive immune mechanisms that drive protective immunity against these pathogens. The vast diversity of the microbiota, pathogens and immune responses encountered in the intestines precludes discussion of all of the relevant players in this Review. Instead, we aim to provide a representative overview of how the intestinal immune system responds to pathogenic bacteria.
Collapse
|
241
|
Ignacio A, Morales CI, Câmara NOS, Almeida RR. Innate Sensing of the Gut Microbiota: Modulation of Inflammatory and Autoimmune Diseases. Front Immunol 2016; 7:54. [PMID: 26925061 PMCID: PMC4759259 DOI: 10.3389/fimmu.2016.00054] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 02/04/2016] [Indexed: 12/13/2022] Open
Abstract
The mammalian gastrointestinal tract harbors a diverse microbial community with which dynamic interactions have been established over millennia of coevolution. Commensal bacteria and their products are sensed by innate receptors expressed in gut epithelia and in gut-associated immune cells, thereby promoting the proper development of mucosal immune system and host homeostasis. Many studies have demonstrated that host–microbiota interactions play a key role during local and systemic immunity. Therefore, this review will focus on how innate sensing of the gut microbiota and their metabolites through inflammasome and toll-like receptors impact the modulation of a distinct set of inflammatory and autoimmune diseases. We believe that a better understanding of the fine-tuning that governs host–microbiota interactions will further improve common prophylactic and therapeutic applications.
Collapse
Affiliation(s)
- Aline Ignacio
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo , São Paulo , Brazil
| | - Camila Ideli Morales
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo , São Paulo , Brazil
| | - Niels Olsen Saraiva Câmara
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil; Department of Medicine, Nephrology Division, Federal University of São Paulo, São Paulo, Brazil; Renal Pathophysiology Laboratory, Department of Clinical Medicine, University of São Paulo, São Paulo, Brazil
| | - Rafael Ribeiro Almeida
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo , São Paulo , Brazil
| |
Collapse
|
242
|
Protective and pro-inflammatory roles of intestinal bacteria. ACTA ACUST UNITED AC 2016; 23:67-80. [PMID: 26947707 DOI: 10.1016/j.pathophys.2016.02.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 02/11/2016] [Accepted: 02/12/2016] [Indexed: 02/06/2023]
Abstract
The intestinal mucosal surface in all vertebrates is exposed to enormous numbers of microorganisms that include bacteria, archaea, fungi and viruses. Coexistence of the host with the gut microbiota represents an active and mutually beneficial relationship that helps to shape the mucosal and systemic immune systems of both mammals and teleosts (ray-finned fish). Due to the potential for enteric microorganisms to invade intestinal tissue and induce local and/or systemic inflammation, the mucosal immune system has developed a number of protective mechanisms that allow the host to mount an appropriate immune response to invading bacteria, while limiting bystander tissue injury associated with these immune responses. Failure to properly regulate mucosal immunity is thought to be responsible for the development of chronic intestinal inflammation. The objective of this review is to present our current understanding of the role that intestinal bacteria play in vertebrate health and disease. While our primary focus will be humans and mice, we also present the new and exciting comparative studies being performed in zebrafish to model host-microbe interactions.
Collapse
|
243
|
Molecular Mechanisms of Induction of Tolerant and Tolerogenic Intestinal Dendritic Cells in Mice. J Immunol Res 2016; 2016:1958650. [PMID: 26981546 PMCID: PMC4766351 DOI: 10.1155/2016/1958650] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 01/06/2016] [Accepted: 01/17/2016] [Indexed: 12/31/2022] Open
Abstract
How does the host manage to tolerate its own intestinal microbiota? A simple question leading to complicated answers. In order to maintain balanced immune responses in the intestine, the host immune system must tolerate commensal bacteria in the gut while it has to simultaneously keep the ability to fight pathogens and to clear infections. If this tender equilibrium is disturbed, severe chronic inflammatory reactions can result. Tolerogenic intestinal dendritic cells fulfil a crucial role in balancing immune responses and therefore creating homeostatic conditions and preventing from uncontrolled inflammation. Although several dendritic cell subsets have already been characterized to play a pivotal role in this process, less is known about definite molecular mechanisms of how intestinal dendritic cells are converted into tolerogenic ones. Here we review how gut commensal bacteria interact with intestinal dendritic cells and why this bacteria-host cell interaction is crucial for induction of dendritic cell tolerance in the intestine. Hereby, different commensal bacteria can have distinct effects on the phenotype of intestinal dendritic cells and these effects are mainly mediated by impacting toll-like receptor signalling in dendritic cells.
Collapse
|
244
|
Hudson LE, Stewart TP, Fasken MB, Corbett AH, Lamb TJ. Transformation of Probiotic Yeast and Their Recovery from Gastrointestinal Immune Tissues Following Oral Gavage in Mice. J Vis Exp 2016:e53453. [PMID: 26890281 PMCID: PMC4781741 DOI: 10.3791/53453] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Development of recombinant oral therapy would allow for more direct targeting of the mucosal immune system and improve the ability to combat gastrointestinal disorders. Adapting probiotic yeast in particular for this approach carries several advantages. These strains have not only the potential to synthesize a wide variety of complex heterologous proteins but are also capable of surviving and protecting those proteins during transit through the intestine. Critically, however, this approach requires expertise in many diverse laboratory techniques not typically used in tandem. Furthermore, although individual protocols for yeast transformation are well characterized for commonly used laboratory strains, emphasis is placed here on alternative approaches and the importance of optimizing transformation for less well characterized probiotic strains. Detailing these methods will help facilitate discussion as to the best approaches for testing probiotic yeast as oral drug delivery vehicles and indeed serve to advance the development of this novel strategy for gastrointestinal therapy.
Collapse
Affiliation(s)
- Lauren E Hudson
- Department of Pediatrics, Emory University School of Medicine
| | - Taryn P Stewart
- Department of Pediatrics, Emory University School of Medicine
| | - Milo B Fasken
- Department of Biochemistry, Emory University School of Medicine
| | - Anita H Corbett
- Department of Biochemistry, Emory University School of Medicine
| | - Tracey J Lamb
- Department of Pediatrics, Emory University School of Medicine;
| |
Collapse
|
245
|
Fonseca DMD, Hand TW, Han SJ, Gerner MY, Glatman Zaretsky A, Byrd AL, Harrison OJ, Ortiz AM, Quinones M, Trinchieri G, Brenchley JM, Brodsky IE, Germain RN, Randolph GJ, Belkaid Y. Microbiota-Dependent Sequelae of Acute Infection Compromise Tissue-Specific Immunity. Cell 2016; 163:354-66. [PMID: 26451485 DOI: 10.1016/j.cell.2015.08.030] [Citation(s) in RCA: 205] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 07/09/2015] [Accepted: 08/11/2015] [Indexed: 02/07/2023]
Abstract
Infections have been proposed as initiating factors for inflammatory disorders; however, identifying associations between defined infectious agents and the initiation of chronic disease has remained elusive. Here, we report that a single acute infection can have dramatic and long-term consequences for tissue-specific immunity. Following clearance of Yersinia pseudotuberculosis, sustained inflammation and associated lymphatic leakage in the mesenteric adipose tissue deviates migratory dendritic cells to the adipose compartment, thereby preventing their accumulation in the mesenteric lymph node. As a consequence, canonical mucosal immune functions, including tolerance and protective immunity, are persistently compromised. Post-resolution of infection, signals derived from the microbiota maintain inflammatory mesentery remodeling and consequently, transient ablation of the microbiota restores mucosal immunity. Our results indicate that persistent disruption of communication between tissues and the immune system following clearance of an acute infection represents an inflection point beyond which tissue homeostasis and immunity is compromised for the long-term. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Denise Morais da Fonseca
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIAID/NIH), Bethesda, MD 20892, USA; Immunity at Barrier Sites Initiative, NIAID/NIH, Bethesda, MD 20892, USA; Department of Biochemistry and Immunology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, 14049-900, Brazil
| | - Timothy W Hand
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIAID/NIH), Bethesda, MD 20892, USA; Immunity at Barrier Sites Initiative, NIAID/NIH, Bethesda, MD 20892, USA
| | - Seong-Ji Han
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIAID/NIH), Bethesda, MD 20892, USA; Immunity at Barrier Sites Initiative, NIAID/NIH, Bethesda, MD 20892, USA
| | - Michael Y Gerner
- Lymphocyte Biology Section, Laboratory of Systems Biology, NIAID/NIH, Bethesda, MD 20892, USA
| | - Arielle Glatman Zaretsky
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIAID/NIH), Bethesda, MD 20892, USA; Immunity at Barrier Sites Initiative, NIAID/NIH, Bethesda, MD 20892, USA
| | - Allyson L Byrd
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIAID/NIH), Bethesda, MD 20892, USA; Immunity at Barrier Sites Initiative, NIAID/NIH, Bethesda, MD 20892, USA; Translational and Functional Genomics Branch, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA; Department of Bioinformatics, Boston University, Boston, MA 02215, USA
| | - Oliver J Harrison
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIAID/NIH), Bethesda, MD 20892, USA; Immunity at Barrier Sites Initiative, NIAID/NIH, Bethesda, MD 20892, USA
| | - Alexandra M Ortiz
- Program in Tissue Immunity and Repair and Immunopathogenesis Section, Laboratory of Molecular Microbiology, NIAID, NIH, Bethesda, MD 20892, USA
| | - Mariam Quinones
- Bioinformatics and Computational Bioscience Branch, NIAID/NIH, Bethesda, MD 20892, USA
| | - Giorgio Trinchieri
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Jason M Brenchley
- Program in Tissue Immunity and Repair and Immunopathogenesis Section, Laboratory of Molecular Microbiology, NIAID, NIH, Bethesda, MD 20892, USA
| | - Igor E Brodsky
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ronald N Germain
- Lymphocyte Biology Section, Laboratory of Systems Biology, NIAID/NIH, Bethesda, MD 20892, USA
| | - Gwendalyn J Randolph
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yasmine Belkaid
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIAID/NIH), Bethesda, MD 20892, USA; Immunity at Barrier Sites Initiative, NIAID/NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
246
|
Gabanyi I, Muller PA, Feighery L, Oliveira TY, Costa-Pinto FA, Mucida D. Neuro-immune Interactions Drive Tissue Programming in Intestinal Macrophages. Cell 2016; 164:378-91. [PMID: 26777404 DOI: 10.1016/j.cell.2015.12.023] [Citation(s) in RCA: 485] [Impact Index Per Article: 53.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 11/24/2015] [Accepted: 12/04/2015] [Indexed: 12/14/2022]
Abstract
Proper adaptation to environmental perturbations is essential for tissue homeostasis. In the intestine, diverse environmental cues can be sensed by immune cells, which must balance resistance to microorganisms with tolerance, avoiding excess tissue damage. By applying imaging and transcriptional profiling tools, we interrogated how distinct microenvironments in the gut regulate resident macrophages. We discovered that macrophages exhibit a high degree of gene-expression specialization dependent on their proximity to the gut lumen. Lamina propria macrophages (LpMs) preferentially expressed a pro-inflammatory phenotype when compared to muscularis macrophages (MMs), which displayed a tissue-protective phenotype. Upon luminal bacterial infection, MMs further enhanced tissue-protective programs, and this was attributed to swift activation of extrinsic sympathetic neurons innervating the gut muscularis and norepinephrine signaling to β2 adrenergic receptors on MMs. Our results reveal unique intra-tissue macrophage specialization and identify neuro-immune communication between enteric neurons and macrophages that induces rapid tissue-protective responses to distal perturbations.
Collapse
Affiliation(s)
- Ilana Gabanyi
- Laboratory of Mucosal Immunology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA; Department of Pathology, School of Veterinary Medicine, Avenue Prof. Orlando Marques de Paiva 87, Cidade Universitária, University of São Paulo, 05508 270 São Paulo, Brazil
| | - Paul A Muller
- Laboratory of Mucosal Immunology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Linda Feighery
- Laboratory of Mucosal Immunology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Thiago Y Oliveira
- Laboratory of Molecular Immunology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Frederico A Costa-Pinto
- Laboratory of Mucosal Immunology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA; Department of Pathology, School of Veterinary Medicine, Avenue Prof. Orlando Marques de Paiva 87, Cidade Universitária, University of São Paulo, 05508 270 São Paulo, Brazil
| | - Daniel Mucida
- Laboratory of Mucosal Immunology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
247
|
Van Beek AA, Hoogerland JA, Belzer C, De Vos P, De Vos WM, Savelkoul HFJ, Leenen PJM. Interaction of mouse splenocytes and macrophages with bacterial strains in vitro: the effect of age in the immune response. Benef Microbes 2015; 7:275-87. [PMID: 26689225 DOI: 10.3920/bm2015.0094] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Probiotics influence the immune system, both at the local and systemic level. Recent findings suggest the relation between microbiota and the immune system alters with age. Our objective was to address direct effects of six bacterial strains on immune cells from young and aged mice: Lactobacillus plantarum WCFS1, Lactobacillus casei BL23, Lactococcus lactis MG1363, Bifidobacterium breve ATCC15700, Bifidobacterium infantis ATCC15697, and Akkermansia muciniphila ATCC BAA-835. We used splenocytes and naïve or interferon-γ-stimulated bone marrow-derived macrophages (BMDM) as responder populations. All tested bacterial strains induced phenotypic and cytokine responses in splenocytes and BMDM. Based on magnitude of the cellular inflammatory response and cytokine profiles, two subgroups of bacteria were identified, i.e. L. plantarum and L. casei versus B. breve, B. infantis, and A. muciniphila. The latter group of bacteria induced high levels of cytokines produced under inflammatory conditions, including tumour necrosis factor (TNF), interleukin (IL)-6 and IL-10. Responses to L. lactis showed features of both subgroups. In addition, we compared responses by splenocytes and BMDM derived from young mice to those of aged mice, and found that splenocytes and BMDM derived from aged mice had an increased IL-10 production and dysregulated IL-6 and TNF production compared to young immune cells. Overall, our study shows differential inflammatory responses to distinct bacterial strains, and profound age-dependent effects. These findings, moreover, support the view that immune environment importantly influences bacterial immune effects.
Collapse
Affiliation(s)
- A A Van Beek
- 1 Top Institute Food and Nutrition, Nieuwe Kanaal 9A, 6709 PA Wageningen, the Netherlands.,2 Cell Biology and Immunology Group, Wageningen University, De Elst 1, 6709 PG Wageningen, the Netherlands.,3 Department of Immunology, Erasmus University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands
| | - J A Hoogerland
- 1 Top Institute Food and Nutrition, Nieuwe Kanaal 9A, 6709 PA Wageningen, the Netherlands.,2 Cell Biology and Immunology Group, Wageningen University, De Elst 1, 6709 PG Wageningen, the Netherlands.,3 Department of Immunology, Erasmus University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands
| | - C Belzer
- 4 Laboratory of Microbiology, Wageningen University, Dreijenplein 10, 6703 HB Wageningen, the Netherlands
| | - P De Vos
- 1 Top Institute Food and Nutrition, Nieuwe Kanaal 9A, 6709 PA Wageningen, the Netherlands.,5 University of Groningen, Pathology and Medical Biology, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - W M De Vos
- 4 Laboratory of Microbiology, Wageningen University, Dreijenplein 10, 6703 HB Wageningen, the Netherlands
| | - H F J Savelkoul
- 2 Cell Biology and Immunology Group, Wageningen University, De Elst 1, 6709 PG Wageningen, the Netherlands
| | - P J M Leenen
- 3 Department of Immunology, Erasmus University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands
| |
Collapse
|
248
|
Martelli S, Pender SLF, Larbi A. Compartmentalization of immunosenescence: a deeper look at the mucosa. Biogerontology 2015; 17:159-76. [PMID: 26689202 DOI: 10.1007/s10522-015-9628-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 12/09/2015] [Indexed: 12/30/2022]
Abstract
Developments in medical care and living conditions led to an astonishing increase in life-span perspective and subsequently a rise in the old population. This can be seen as a success for public health policies but it also challenges society to adapt, in order to cope with the potentially overwhelming cost for the healthcare system. A fast-growing number of older people lose their ability to live independently because of diseases and disabilities, frailty or cognitive impairment. Many require long-term care, including home-based nursing, communities and hospital-based care. Immunosenescence, an age-related deterioration in immune functions, is considered a major contributory factor for the higher prevalence and severity of infectious diseases and the poor efficacy of vaccination in the elderly. When compared with systemic immunosenescence, alterations in the mucosal immune system with age are less well understood. For this reason, this area deserves more extensive and intensive research and support. In this article, we provide an overview of age-associated changes occurring in systemic immunity and discuss the distinct features of mucosal immunosenescence.
Collapse
Affiliation(s)
- Serena Martelli
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,Singapore Immunology Network (SIgN), Aging and Immunity Program, Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Sylvia L F Pender
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Aging and Immunity Program, Agency for Science Technology and Research (A*STAR), Singapore, Singapore.
| |
Collapse
|
249
|
Innate control of actin nucleation determines two distinct migration behaviours in dendritic cells. Nat Cell Biol 2015; 18:43-53. [PMID: 26641718 DOI: 10.1038/ncb3284] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 11/09/2015] [Indexed: 12/13/2022]
Abstract
Dendritic cell (DC) migration in peripheral tissues serves two main functions: antigen sampling by immature DCs, and chemokine-guided migration towards lymphatic vessels (LVs) on maturation. These migratory events determine the efficiency of the adaptive immune response. Their regulation by the core cell locomotion machinery has not been determined. Here, we show that the migration of immature DCs depends on two main actin pools: a RhoA-mDia1-dependent actin pool located at their rear, which facilitates forward locomotion; and a Cdc42-Arp2/3-dependent actin pool present at their front, which limits migration but promotes antigen capture. Following TLR4-MyD88-induced maturation, Arp2/3-dependent actin enrichment at the cell front is markedly reduced. Consequently, mature DCs switch to a faster and more persistent mDia1-dependent locomotion mode that facilitates chemotactic migration to LVs and lymph nodes. Thus, the differential use of actin-nucleating machineries optimizes the migration of immature and mature DCs according to their specific function.
Collapse
|
250
|
Abstract
As the frontiers of immunological research expand, new insights into the pathogenesis of long poorly understood diseases, such as inflammatory bowel disease (IBD), are opening up new possible avenues for treatment. Myeloid-derived cells (i.e., monocytes, macrophages, neutrophils, and dendritic cells), long believed to be effector cells driving the initiation of inflammation, have been increasingly shown to have immunoregulatory effects previously underappreciated. Dysfunction in the immunoregulatory roles of these cells may play a part in the pathogenesis of a subset of patients with IBD. The role of myeloid-derived suppressor cells, initially described in cancer, have been shown to play an important role in the balancing of effector and regulatory T cells in inflammation as well, and their role in IBD is also explored. The potential for future cell-based therapies for IBD is enhanced by the advances being made in the understanding of the innate immune system in the intestine.
Collapse
|