251
|
|
252
|
Chinthrajah RS, Hernandez JD, Boyd SD, Galli SJ, Nadeau KC. Molecular and cellular mechanisms of food allergy and food tolerance. J Allergy Clin Immunol 2016; 137:984-997. [PMID: 27059726 DOI: 10.1016/j.jaci.2016.02.004] [Citation(s) in RCA: 208] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 02/17/2016] [Accepted: 02/18/2016] [Indexed: 02/06/2023]
Abstract
Ingestion of innocuous antigens, including food proteins, normally results in local and systemic immune nonresponsiveness in a process termed oral tolerance. Oral tolerance to food proteins is likely to be intimately linked to mechanisms that are responsible for gastrointestinal tolerance to large numbers of commensal microbes. Here we review our current understanding of the immune mechanisms responsible for oral tolerance and how perturbations in these mechanisms might promote the loss of oral tolerance and development of food allergies. Roles for the commensal microbiome in promoting oral tolerance and the association of intestinal dysbiosis with food allergy are discussed. Growing evidence supports cutaneous sensitization to food antigens as one possible mechanism leading to the failure to develop or loss of oral tolerance. A goal of immunotherapy for food allergies is to induce sustained desensitization or even true long-term oral tolerance to food allergens through mechanisms that might in part overlap with those associated with the development of natural oral tolerance.
Collapse
Affiliation(s)
- R Sharon Chinthrajah
- Department of Medicine, Stanford University School of Medicine, Stanford, Calif; Department of Pediatrics, Stanford University School of Medicine, Stanford, Calif; Sean N. Parker Center for Allergy & Asthma Research, Stanford University School of Medicine, Stanford, Calif
| | - Joseph D Hernandez
- Department of Pediatrics, Stanford University School of Medicine, Stanford, Calif; Department of Pathology, Stanford University School of Medicine, Stanford, Calif; Sean N. Parker Center for Allergy & Asthma Research, Stanford University School of Medicine, Stanford, Calif
| | - Scott D Boyd
- Department of Pathology, Stanford University School of Medicine, Stanford, Calif; Sean N. Parker Center for Allergy & Asthma Research, Stanford University School of Medicine, Stanford, Calif
| | - Stephen J Galli
- Department of Pathology, Stanford University School of Medicine, Stanford, Calif; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, Calif; Sean N. Parker Center for Allergy & Asthma Research, Stanford University School of Medicine, Stanford, Calif
| | - Kari C Nadeau
- Department of Medicine, Stanford University School of Medicine, Stanford, Calif; Department of Pediatrics, Stanford University School of Medicine, Stanford, Calif; Sean N. Parker Center for Allergy & Asthma Research, Stanford University School of Medicine, Stanford, Calif.
| |
Collapse
|
253
|
Marelli G, Belgiovine C, Mantovani A, Erreni M, Allavena P. Non-redundant role of the chemokine receptor CX3CR1 in the anti-inflammatory function of gut macrophages. Immunobiology 2016; 222:463-472. [PMID: 27707514 DOI: 10.1016/j.imbio.2016.07.013] [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: 06/14/2016] [Revised: 07/24/2016] [Accepted: 07/31/2016] [Indexed: 12/20/2022]
Abstract
Mucosal immunity at the intestinal level is constantly challenged by the presence of external food and microbial antigens and must be kept under strict control to avoid the rise of aberrant inflammation. Among cells of the innate immunity, macrophages expressing the chemokine receptor CX3CR1 are strategically located near the gut epithelial barrier. These cells contribute to the maintenance of homeostasis by producing the anti-inflammatory cytokine IL-10; however, their role in the control of full blown inflammation and tissue injury is controversial. In this study we investigated mice proficient or deficient for the expression of the CX3CR1 receptor in a model of dextran sulphate sodium (DSS) induced acute colitis. We found that KO mice (CX3CR1GFP/GFP) had a more severe disease compared to WT mice (CX3CR1GFP/+), both in terms of histological examination of colonic tissues and leukocyte infiltration, with an expansion of macrophages and CD4-Th17 lymphocytes. The expression of several inflammatory mediators (IL-1β, IL-6, IFNγ, iNOS) was also significantly upregulated in KO mice, despite higher IL-10 production. Overall, our study demonstrates that macrophages expressing a functional CX3CR1 receptor have an important and non-redundant role in controlling the abnormal intestinal inflammation that may lead to tissue damage.
Collapse
Affiliation(s)
- G Marelli
- Dpt. Immunology and Inflammation, IRCCS-Humanitas Clinical and Research Center, Via Manzoni, 56, 20089 Rozzano (Milano), Italy.
| | - C Belgiovine
- Dpt. Immunology and Inflammation, IRCCS-Humanitas Clinical and Research Center, Via Manzoni, 56, 20089 Rozzano (Milano), Italy
| | - A Mantovani
- Dpt. Immunology and Inflammation, IRCCS-Humanitas Clinical and Research Center, Via Manzoni, 56, 20089 Rozzano (Milano), Italy; Humanitas University, Via Manzoni, 56, 20089 Rozzano (Milano), Italy
| | - M Erreni
- Dpt. Immunology and Inflammation, IRCCS-Humanitas Clinical and Research Center, Via Manzoni, 56, 20089 Rozzano (Milano), Italy
| | - P Allavena
- Dpt. Immunology and Inflammation, IRCCS-Humanitas Clinical and Research Center, Via Manzoni, 56, 20089 Rozzano (Milano), Italy
| |
Collapse
|
254
|
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
|
255
|
Kim D, Beck BR, Lee SM, Jeon J, Lee DW, Lee JI, Song SK. Pellet feed adsorbed with the recombinant Lactococcus lactis BFE920 expressing SiMA antigen induced strong recall vaccine effects against Streptococcus iniae infection in olive flounder (Paralichthys olivaceus). FISH & SHELLFISH IMMUNOLOGY 2016; 55:374-383. [PMID: 27302864 DOI: 10.1016/j.fsi.2016.06.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 05/31/2016] [Accepted: 06/06/2016] [Indexed: 06/06/2023]
Abstract
The aim of this study was to develop a fish feed vaccine that provides effective disease prevention and convenient application. A lactic acid bacterium (LAB), Lactococcus lactis BFE920, was modified to express the SiMA antigen, a membrane protein of Streptococcus iniae. The antigen was engineered to be expressed under the nisin promoter, which is induced by nisin produced naturally by the host LAB. Various sizes (40 ± 3.5 g, 80 ± 2.1 g, and 221 ± 2.4 g) of olive flounder (Paralichthys olivaceus) were vaccinated by feeding the extruded pellet feed, onto which the SiMA-expressing L. lactis BFE920 (1.0 × 10(7) CFU/g) was adsorbed. Vaccine-treated feed was administered twice a day for 1 week, and priming and boosting were performed with a 1-week interval in between. The vaccinated fish had significantly elevated levels of antigen-specific serum antibodies and T cell marker mRNAs: CD4-1, CD4-2, and CD8a. In addition, the feed vaccine significantly induced T cell effector functions, such as the production of IFN-γ and activation of the transcription factor that induces its expression, T-bet. When the flounder were challenged by intraperitoneal infection and bath immersion with S. iniae, the vaccinated fish showed 84% and 82% relative percent survival (RPS), respectively. Furthermore, similar protective effects were confirmed even 3 months after vaccination in a field study (n = 4800), indicating that this feed vaccine elicited prolonged duration of immunopotency. In addition, the vaccinated flounder gained 21% more weight and required 16% less feed to gain a unit of body weight compared to the control group. The data clearly demonstrate that the L. lactis BFE920-SiMA feed vaccine has strong protective effects, induces prolonged vaccine efficacy, and has probiotic effects. In addition, this LAB-based fish feed vaccine can be easily used to target many different pathogens of diverse fish species.
Collapse
Affiliation(s)
- Daniel Kim
- School of Life Science, Handong University, Pohang 791-708, Republic of Korea
| | - Bo Ram Beck
- School of Life Science, Handong University, Pohang 791-708, Republic of Korea
| | - Sun Min Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Republic of Korea
| | - Jongsu Jeon
- New Business Development Dept., Medytox Inc, Republic of Korea
| | - Dong Wook Lee
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Jae Il Lee
- GyeongSangbuk-Do Fisheries Technology Center, Pohang 791-941, Republic of Korea
| | - Seong Kyu Song
- School of Life Science, Handong University, Pohang 791-708, Republic of Korea.
| |
Collapse
|
256
|
Xu Y, Liu Y, Yang C, Kang L, Wang M, Hu J, He H, Song W, Tang H. Macrophages transfer antigens to dendritic cells by releasing exosomes containing dead-cell-associated antigens partially through a ceramide-dependent pathway to enhance CD4(+) T-cell responses. Immunology 2016; 149:157-71. [PMID: 27278624 DOI: 10.1111/imm.12630] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 05/26/2016] [Accepted: 06/03/2016] [Indexed: 02/06/2023] Open
Abstract
Defects in rapid clearance of apoptotic cells lead to an accumulation of dead cells (late apoptotic or secondary necrotic cells), which results in an aberrant immune response. However, little is known about whether and how macrophages (Mφs) cooperate with dendritic cells (DCs) in the presentation of dead-cell-associated antigens in this process. By transferring high numbers of dead cells to mimic a failure of apoptotic cell clearance in vivo, we found that Mφs and neutrophils were the predominant phagocytes in the uptake of dead cells in the spleen. Moreover, both Mφs and DCs were required for an optimal CD4(+) T-cell response triggered by dead-cell-associated antigens. Importantly, although Mφs alone had a poor capacity for antigen presentation, they could transfer phagocytosed antigens to DCs for potent antigen presentation to enhance T-cell responses. Finally, we found that exosomes released from Mφs acted as a transmitter to convey antigens to DCs partially in a ceramide-dependent manner, since treatment with the neutral sphingomyelinase inhibitor GW4869 and spiroepoxide resulted in a significant reduction of T-cell proliferation in vitro and in vivo. These findings point to a novel pathway of cross-talk between Mφs and DCs, which will be helpful to explain possible mechanisms for autoimmune diseases characterized by increased rates of apoptosis.
Collapse
Affiliation(s)
- Yingping Xu
- Institute of Immunology, Taishan Medical University, Taian, China
| | - Yi Liu
- Institute of Immunology, Taishan Medical University, Taian, China
| | - Chunqing Yang
- Institute of Immunology, Taishan Medical University, Taian, China
| | - Li Kang
- Institute of Immunology, Taishan Medical University, Taian, China
| | - Meixiang Wang
- Institute of Immunology, Taishan Medical University, Taian, China
| | - Jingxia Hu
- Institute of Immunology, Taishan Medical University, Taian, China
| | - Hao He
- Institute of Immunology, Taishan Medical University, Taian, China
| | - Wengang Song
- Institute of Immunology, Taishan Medical University, Taian, China
| | - Hua Tang
- Institute of Immunology, Taishan Medical University, Taian, China
| |
Collapse
|
257
|
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
|
258
|
Isidro RA, Appleyard CB. Colonic macrophage polarization in homeostasis, inflammation, and cancer. Am J Physiol Gastrointest Liver Physiol 2016; 311:G59-73. [PMID: 27229123 PMCID: PMC4967174 DOI: 10.1152/ajpgi.00123.2016] [Citation(s) in RCA: 164] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 05/24/2016] [Indexed: 01/31/2023]
Abstract
Our review focuses on the colonic macrophage, a monocyte-derived, tissue-resident macrophage, and the role it plays in health and disease, specifically in inflammatory conditions such as inflammatory bowel disease and cancer of the colon and rectum. We give special emphasis to macrophage polarization, or phenotype, in these different states. We focus on macrophages because they are one of the most numerous leukocytes in the colon, and because they normally contribute to homeostasis through an anti-inflammatory phenotype. However, in conditions such as inflammatory bowel disease, proinflammatory macrophages are increased in the colon and have been linked to disease severity and progression. In colorectal cancer, tumor cells may employ anti-inflammatory macrophages to promote tumor growth and dissemination, whereas proinflammatory macrophages may antagonize tumor growth. Given the key roles that this cell type plays in homeostasis, inflammation, and cancer, the colonic macrophage is an intriguing therapeutic target. As such, potential macrophage-targeting strategies are discussed.
Collapse
Affiliation(s)
- Raymond A Isidro
- Department of Basic Sciences, Ponce Health Sciences University-Medical School and Ponce Research Institute, 395 Zona Industrial Reparada 2, Ponce, Puerto Rico 00716
| | - Caroline B Appleyard
- Department of Basic Sciences, Ponce Health Sciences University-Medical School and Ponce Research Institute, 395 Zona Industrial Reparada 2, Ponce, Puerto Rico 00716
| |
Collapse
|
259
|
Kabat AM, Pott J, Maloy KJ. The Mucosal Immune System and Its Regulation by Autophagy. Front Immunol 2016; 7:240. [PMID: 27446072 PMCID: PMC4916208 DOI: 10.3389/fimmu.2016.00240] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 06/07/2016] [Indexed: 12/20/2022] Open
Abstract
The gastrointestinal tract presents a unique challenge to the mucosal immune system, which has to constantly monitor the vast surface for the presence of pathogens, while at the same time maintaining tolerance to beneficial or innocuous antigens. In the intestinal mucosa, specialized innate and adaptive immune components participate in directing appropriate immune responses toward these diverse challenges. Recent studies provide compelling evidence that the process of autophagy influences several aspects of mucosal immune responses. Initially described as a “self-eating” survival pathway that enables nutrient recycling during starvation, autophagy has now been connected to multiple cellular responses, including several aspects of immunity. Initial links between autophagy and host immunity came from the observations that autophagy can target intracellular bacteria for degradation. However, subsequent studies indicated that autophagy plays a much broader role in immune responses, as it can impact antigen processing, thymic selection, lymphocyte homeostasis, and the regulation of immunoglobulin and cytokine secretion. In this review, we provide a comprehensive overview of mucosal immune cells and discuss how autophagy influences many aspects of their physiology and function. We focus on cell type-specific roles of autophagy in the gut, with a particular emphasis on the effects of autophagy on the intestinal T cell compartment. We also provide a perspective on how manipulation of autophagy may potentially be used to treat mucosal inflammatory disorders.
Collapse
Affiliation(s)
- Agnieszka M Kabat
- Sir William Dunn School of Pathology, University of Oxford , Oxford , UK
| | - Johanna Pott
- Sir William Dunn School of Pathology, University of Oxford , Oxford , UK
| | - Kevin J Maloy
- Sir William Dunn School of Pathology, University of Oxford , Oxford , UK
| |
Collapse
|
260
|
Bedoui S, Heath WR, Mueller SN. CD
4
+
T‐cell help amplifies innate signals for primary
CD
8
+
T‐cell immunity. Immunol Rev 2016; 272:52-64. [DOI: 10.1111/imr.12426] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Sammy Bedoui
- Department of Microbiology and Immunology The University of Melbourne Peter Doherty Institute for Infection and Immunity Parkville Vic. Australia
| | - William R. Heath
- Department of Microbiology and Immunology The University of Melbourne Peter Doherty Institute for Infection and Immunity Parkville Vic. Australia
- The Australian Research Council Centre of Excellence in Advanced Molecular Imaging The University of Melbourne Parkville Vic. Australia
| | - Scott N. Mueller
- Department of Microbiology and Immunology The University of Melbourne Peter Doherty Institute for Infection and Immunity Parkville Vic. Australia
- The Australian Research Council Centre of Excellence in Advanced Molecular Imaging The University of Melbourne Parkville Vic. Australia
| |
Collapse
|
261
|
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: 29] [Impact Index Per Article: 3.2] [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
|
262
|
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
|
263
|
Hao S, Han X, Wang D, Yang Y, Li Q, Li X, Qiu C. Critical role of CCL22/CCR4 axis in the maintenance of immune homeostasis during apoptotic cell clearance by splenic CD8α(+) CD103(+) dendritic cells. Immunology 2016; 148:174-86. [PMID: 26868141 PMCID: PMC4863574 DOI: 10.1111/imm.12596] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 02/07/2016] [Accepted: 02/08/2016] [Indexed: 12/19/2022] Open
Abstract
Macrophages and dendritic cells (DCs) in murine spleen are essential for the maintenance of immune homeostasis by elimination of blood-borne foreign particles and organisms. It has been reported that splenic DCs, especially CD8α(+) CD103(+) DCs, are responsible for tolerance to apoptosis-associated antigens. However, the molecular mechanism by which these DCs maintain immune homeostasis by blood-borne apoptotic cell clearance remains elusive. Here, we found that the CCL22/CCR4 axis played a critical role in the maintenance of immune homeostasis during apoptotic cell clearance by splenic CD8α(+) CD103(+) DCs. The present results revealed that systemic administration of apoptotic cells rapidly induced a large number of CCL22 and CCR4(+) regulatory T (Treg) cells in the spleen of C57BL/6J mice. Further study demonstrated that CD8α(+) CD103(+) DCs dominantly produce much higher CCL22 than CD8α(+) CD103(-) DCs. Moreover, the transient deletion of CD8α(+) CD103(+) DCs caused a decrease in CCL22 levels together with CCR4(+) Treg cell percentage. Subsequently, the levels of some pro-inflammatory cytokines, such as interleukin-17 and interferon-γ in the spleen with the absence of CD8α(+) CD103(+) DCs increased in response to the administration of apoptotic cells. Hence, intravenous injection of apoptotic cells induced a subsequent increase in CCL22 expression and CCR4(+) Treg cells, which contribute to the maintenance of immune homeostasis at least partially by splenic CD8α(+) CD103(+) DCs.
Collapse
Affiliation(s)
- Shengyu Hao
- Department of Cell BiologyShandong University School of MedicineJinanShandongChina
| | - Xiaolei Han
- Department of Cell BiologyShandong University School of MedicineJinanShandongChina
| | - Dan Wang
- Department of Cell BiologyShandong University School of MedicineJinanShandongChina
| | - Yang Yang
- Department of Cell BiologyShandong University School of MedicineJinanShandongChina
| | - Qiuting Li
- Department of Cell BiologyShandong University School of MedicineJinanShandongChina
| | - Xiangzhi Li
- Department of Cell BiologyShandong University School of MedicineJinanShandongChina
| | - Chun‐Hong Qiu
- Department of Cell BiologyShandong University School of MedicineJinanShandongChina
| |
Collapse
|
264
|
Edwards JP, Hand TW, da Fonseca DM, Glass DD, Belkaid Y, Shevach EM. The GARP/Latent TGF-β1 complex on Treg cells modulates the induction of peripherally derived Treg cells during oral tolerance. Eur J Immunol 2016; 46:1480-9. [PMID: 27062243 PMCID: PMC11022272 DOI: 10.1002/eji.201546204] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 03/01/2016] [Accepted: 03/31/2016] [Indexed: 12/21/2022]
Abstract
Treg cells can secrete latent TGF-β1 (LTGF-β1), but can also utilize an alternative pathway for transport and expression of LTGF-β1 on the cell surface in which LTGF-β1 is coupled to a distinct LTGF-β binding protein termed glycoprotein A repetitions predominant (GARP)/LRRC32. The function of the GARP/LTGF-β1 complex has remained elusive. Here, we examine in vivo the roles of GARP and TGF-β1 in the induction of oral tolerance. When Foxp3(-) OT-II T cells were transferred to wild-type recipient mice followed by OVA feeding, the conversion of Foxp3(-) to Foxp3(+) OT-II cells was dependent on recipient Treg cells. Neutralization of IL-2 in the recipient mice also abrogated this conversion. The GARP/LTGF-β1 complex on recipient Treg cells, but not dendritic cell-derived TGF-β1, was required for efficient induction of Foxp3(+) T cells and for the suppression of delayed hypersensitivity. Expression of the integrin αvβ8 by Treg cells (or T cells) in the recipients was dispensable for induction of Foxp3 expression. Transient depletion of the bacterial flora enhanced the development of oral tolerance by expanding Treg cells with enhanced expression of the GARP/LTGF-β1 complex.
Collapse
Affiliation(s)
- Justin P. Edwards
- Cellular Immunology Section, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Timothy W. Hand
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Denise Morais da Fonseca
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Deborah D. Glass
- Cellular Immunology Section, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yasmine Belkaid
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ethan M. Shevach
- Cellular Immunology Section, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
265
|
Tse G, Lai ETH, Yeo JM, Tse V, Wong SH. Mechanisms of Electrical Activation and Conduction in the Gastrointestinal System: Lessons from Cardiac Electrophysiology. Front Physiol 2016; 7:182. [PMID: 27303305 PMCID: PMC4885840 DOI: 10.3389/fphys.2016.00182] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/06/2016] [Indexed: 12/12/2022] Open
Abstract
The gastrointestinal (GI) tract is an electrically excitable organ system containing multiple cell types, which coordinate electrical activity propagating through this tract. Disruption in its normal electrophysiology is observed in a number of GI motility disorders. However, this is not well characterized and the field of GI electrophysiology is much less developed compared to the cardiac field. The aim of this article is to use the established knowledge of cardiac electrophysiology to shed light on the mechanisms of electrical activation and propagation along the GI tract, and how abnormalities in these processes lead to motility disorders and suggest better treatment options based on this improved understanding. In the first part of the article, the ionic contributions to the generation of GI slow wave and the cardiac action potential (AP) are reviewed. Propagation of these electrical signals can be described by the core conductor theory in both systems. However, specifically for the GI tract, the following unique properties are observed: changes in slow wave frequency along its length, periods of quiescence, synchronization in short distances and desynchronization over long distances. These are best described by a coupled oscillator theory. Other differences include the diminished role of gap junctions in mediating this conduction in the GI tract compared to the heart. The electrophysiology of conditions such as gastroesophageal reflux disease and gastroparesis, and functional problems such as irritable bowel syndrome are discussed in detail, with reference to ion channel abnormalities and potential therapeutic targets. A deeper understanding of the molecular basis and physiological mechanisms underlying GI motility disorders will enable the development of better diagnostic and therapeutic tools and the advancement of this field.
Collapse
Affiliation(s)
- Gary Tse
- Li Ka Shing Faculty of Medicine, School of Biomedical Sciences, University of Hong KongHong Kong, China
| | - Eric Tsz Him Lai
- Li Ka Shing Faculty of Medicine, School of Biomedical Sciences, University of Hong KongHong Kong, China
| | - Jie Ming Yeo
- School of Medicine, Imperial College LondonLondon, UK
| | - Vivian Tse
- Department of Physiology, McGill UniversityMontreal, QC, Canada
| | - Sunny Hei Wong
- Department of Medicine and Therapeutics, Institute of Digestive Disease, LKS Institute of Health Sciences, Chinese University of Hong KongHong Kong, China
| |
Collapse
|
266
|
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: 31] [Impact Index Per Article: 3.4] [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
|
267
|
Swiatczak B, Cohen IR. Gut feelings of safety: tolerance to the microbiota mediated by innate immune receptors. Microbiol Immunol 2016; 59:573-85. [PMID: 26306708 DOI: 10.1111/1348-0421.12318] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 08/09/2015] [Accepted: 08/20/2015] [Indexed: 12/11/2022]
Abstract
To enable microbial colonization of the gut mucosa, the intestinal immune system must not only react to danger signals but also recognize cues that indicate safety. Recognition of safety, paradoxically, is mediated by the same environmental sensors that are involved in signaling danger. Indeed, in addition to their well-established role in inducing inflammation in response to stress signals, pattern recognition receptors and a variety of metabolic sensors also promote gut-microbiota symbiosis by responding to "microbial symbiosis factors", "resolution-associated molecular patterns", markers of energy extraction and other signals indicating the absence of pathogenic infection and tissue damage. Here we focus on how the paradoxical roles of immune receptors and other environmental sensors define the microbiota signature of an individual.
Collapse
Affiliation(s)
- Bartlomiej Swiatczak
- Department of History of Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, 230026, China
| | - Irun R Cohen
- Department of Immunology, Weizmann Institute of Science, Rehovot, 76100, Israel
| |
Collapse
|
268
|
de Jesus ER, Isidro RA, Cruz ML, Marty H, Appleyard CB. Adoptive Transfer of Dendritic Cells Expressing Fas Ligand Modulates Intestinal Inflammation in a Model of Inflammatory Bowel Disease. ACTA ACUST UNITED AC 2016; 7. [PMID: 27274906 PMCID: PMC4892183 DOI: 10.4172/2155-9899.1000411] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Background Inflammatory bowel diseases (IBD) are chronic relapsing inflammatory conditions of unknown cause and likely result from the loss of immunological tolerance, which leads to over-activation of the gut immune system. Gut macrophages and dendritic cells (DCs) are essential for maintaining tolerance, but can also contribute to the inflammatory response in conditions such as IBD. Current therapies for IBD are limited by high costs and unwanted toxicities and side effects. The possibility of reducing intestinal inflammation with DCs genetically engineered to over-express the apoptosis-inducing FasL (FasL-DCs) has not yet been explored. Objective Investigate the immunomodulatory effect of administering FasL-DCs in the rat trinitrobenzene sulfonic acid (TNBS) model of acute colitis. Methods Expression of FasL on DCs isolated from the mesenteric lymph nodes (MLNs) of normal and TNBS-colitis rats was determined by flow cytometry. Primary rat bone marrow DCs were transfected with rat FasL plasmid (FasL-DCs) or empty vector (EV-DCs). The effect of these DCs on T cell IFNγ secretion and apoptosis was determined by ELISPOT and flow cytometry for Annexin V, respectively. Rats received FasL-DCs or EV-DCs intraperitoneally 96 and 48 hours prior to colitis induction with TNBS. Colonic T cell and neutrophil infiltration was determined by immunohistochemistry for CD3 and myeloperoxidase activity assay, respectively. Macrophage number and phenotype was measured by double immunofluorescence for CD68 and inducible Nitric Oxide Synthase. Results MLN dendritic cells from normal rats expressed more FasL than those from colitic rats. Compared to EV-DCs, FasL-DCs reduced T cell IFNγ secretion and increased T cell apoptosis in vitro. Adoptive transfer of FasL-DCs decreased macroscopic and microscopic damage scores and reduced colonic T cells, neutrophils, and proinflammatory macrophages when compared to EV-DC adoptive transfer. Conclusion FasL-DCs are effective at treating colonic inflammation in this model of IBD and represent a possible new treatment for patients with IBD.
Collapse
Affiliation(s)
- Edelmarie Rivera de Jesus
- Ponce Health Sciences University-Medical School and Ponce Research Institute, Ponce, PR 00732, USA; Department of Biology, University of Puerto Rico - Ponce Campus, Ponce, PR 00732, USA
| | - Raymond A Isidro
- Ponce Health Sciences University-Medical School and Ponce Research Institute, Ponce, PR 00732, USA
| | - Myrella L Cruz
- Ponce Health Sciences University-Medical School and Ponce Research Institute, Ponce, PR 00732, USA
| | - Harry Marty
- Ponce Health Sciences University-Medical School and Ponce Research Institute, Ponce, PR 00732, USA
| | - Caroline B Appleyard
- Ponce Health Sciences University-Medical School and Ponce Research Institute, Ponce, PR 00732, USA
| |
Collapse
|
269
|
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
|
270
|
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: 59] [Impact Index Per Article: 6.6] [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
|
271
|
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: 209] [Impact Index Per Article: 23.2] [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
|
272
|
Hoffmann F, Ender F, Schmudde I, Lewkowich IP, Köhl J, König P, Laumonnier Y. Origin, Localization, and Immunoregulatory Properties of Pulmonary Phagocytes in Allergic Asthma. Front Immunol 2016; 7:107. [PMID: 27047494 PMCID: PMC4803735 DOI: 10.3389/fimmu.2016.00107] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 03/08/2016] [Indexed: 01/21/2023] Open
Abstract
Allergic asthma is a chronic inflammatory disease of the airways that is driven by maladaptive T helper 2 (Th2) and Th17 immune responses against harmless, airborne substances. Pulmonary phagocytes represent the first line of defense in the lung where they constantly sense the local environment for potential threats. They comprise two distinct cell types, i.e., macrophages and dendritic cells (DC) that differ in their origins and functions. Alveolar macrophages quickly take up most of the inhaled allergens, yet do not deliver their cargo to naive T cells sampling in draining lymph nodes. In contrast, pulmonary DCs instruct CD4(+) T cells develop into Th2 and Th17 effectors, initiating the maladaptive immune responses toward harmless environmental substances observed in allergic individuals. Unraveling the mechanisms underlying this mistaken identity of harmless, airborne substances by innate immune cells is one of the great challenges in asthma research. The identification of different pulmonary DC subsets, their role in antigen uptake, migration to the draining lymph nodes, and their potential to instruct distinct T cell responses has set the stage to unravel this mystery. However, at this point, a detailed understanding of the spatiotemporal resolution of DC subset localization, allergen uptake, processing, autocrine and paracrine cellular crosstalk, and the humoral factors that define the activation status of DCs is still lacking. In addition to DCs, at least two distinct macrophage populations have been identified in the lung that are either located in the airway/alveolar lumen or in the interstitium. Recent data suggest that such populations can exert either pro- or anti-inflammatory functions. Similar to the DC subsets, detailed insights into the individual roles of alveolar and interstitial macrophages during the different phases of asthma development are still missing. Here, we will provide an update on the current understanding of the origin, localization, and function of the diverse pulmonary antigen-presenting cell subsets, in particular with regard to the development and regulation of allergic asthma. While most data are from mouse models of experimental asthma, we have also included available human data to judge the translational value of the findings obtained in experimental asthma models.
Collapse
Affiliation(s)
| | - Fanny Ender
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Inken Schmudde
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Ian P. Lewkowich
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Giessen, Germany
| | - Peter König
- Institute for Anatomy, University of Lübeck, Lübeck, Germany
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Giessen, Germany
| | - Yves Laumonnier
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| |
Collapse
|
273
|
Ohta T, Sugiyama M, Hemmi H, Yamazaki C, Okura S, Sasaki I, Fukuda Y, Orimo T, Ishii KJ, Hoshino K, Ginhoux F, Kaisho T. Crucial roles of XCR1-expressing dendritic cells and the XCR1-XCL1 chemokine axis in intestinal immune homeostasis. Sci Rep 2016; 6:23505. [PMID: 27005831 PMCID: PMC4804307 DOI: 10.1038/srep23505] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 03/08/2016] [Indexed: 12/26/2022] Open
Abstract
Intestinal immune homeostasis requires dynamic crosstalk between innate and adaptive immune cells. Dendritic cells (DCs) exist as multiple phenotypically and functionally distinct sub-populations within tissues, where they initiate immune responses and promote homeostasis. In the gut, there exists a minor DC subset defined as CD103(+)CD11b(-) that also expresses the chemokine receptor XCR1. In other tissues, XCR1(+) DCs cross-present antigen and contribute to immunity against viruses and cancer, however the roles of XCR1(+) DCs and XCR1 in the intestine are unknown. We showed that mice lacking XCR1(+) DCs are specifically deficient in intraepithelial and lamina propria (LP) T cell populations, with remaining T cells exhibiting an atypical phenotype and being prone to death, and are also more susceptible to chemically-induced colitis. Mice deficient in either XCR1 or its ligand, XCL1, similarly possess diminished intestinal T cell populations, and an accumulation of XCR1(+) DCs in the gut. Combined with transcriptome and surface marker expression analysis, these observations lead us to hypothesise that T cell-derived XCL1 facilitates intestinal XCR1(+) DC activation and migration, and that XCR1(+) DCs in turn provide support for T cell survival and function. Thus XCR1(+) DCs and the XCR1/XCL1 chemokine axis have previously-unappreciated roles in intestinal immune homeostasis.
Collapse
Affiliation(s)
- Tomokazu Ohta
- Laboratory for Immune Regulation, World Premier International Research Center Initiative, Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan.,Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Wakayama 641-8509, Japan.,Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masanaka Sugiyama
- Laboratory for Immune Regulation, World Premier International Research Center Initiative, Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan.,Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan.,Laboratory for Inflammatory Regulation, RIKEN Center for Integrative Medical Science (IMS-RCAI), Yokohama, Kanagawa 230-0045, Japan.,Laboratory for Host Defence, RIKEN Center for Integrative Medical Science (IMS-RCAI), Yokohama, Kanagawa 230-0045, Japan
| | - Hiroaki Hemmi
- Laboratory for Immune Regulation, World Premier International Research Center Initiative, Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan.,Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Wakayama 641-8509, Japan.,Laboratory for Inflammatory Regulation, RIKEN Center for Integrative Medical Science (IMS-RCAI), Yokohama, Kanagawa 230-0045, Japan.,Laboratory for Host Defence, RIKEN Center for Integrative Medical Science (IMS-RCAI), Yokohama, Kanagawa 230-0045, Japan
| | - Chihiro Yamazaki
- Laboratory for Host Defence, RIKEN Center for Integrative Medical Science (IMS-RCAI), Yokohama, Kanagawa 230-0045, Japan.,Department of Immunology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Okayama 700-8558, Japan
| | - Soichiro Okura
- Laboratory for Immune Regulation, World Premier International Research Center Initiative, Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan.,Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Wakayama 641-8509, Japan
| | - Izumi Sasaki
- Laboratory for Immune Regulation, World Premier International Research Center Initiative, Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan.,Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Wakayama 641-8509, Japan.,Laboratory for Host Defence, RIKEN Center for Integrative Medical Science (IMS-RCAI), Yokohama, Kanagawa 230-0045, Japan
| | - Yuri Fukuda
- Laboratory for Immune Regulation, World Premier International Research Center Initiative, Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan.,Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Wakayama 641-8509, Japan.,Laboratory for Host Defence, RIKEN Center for Integrative Medical Science (IMS-RCAI), Yokohama, Kanagawa 230-0045, Japan
| | - Takashi Orimo
- Laboratory for Immune Regulation, World Premier International Research Center Initiative, Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan.,Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Wakayama 641-8509, Japan.,Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Ken J Ishii
- Laboratory of Adjuvant Innovation, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan.,Laboratory of Vaccine Science, World Premier International Research Center Initiative, Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Katsuaki Hoshino
- Laboratory for Immune Regulation, World Premier International Research Center Initiative, Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan.,Laboratory for Inflammatory Regulation, RIKEN Center for Integrative Medical Science (IMS-RCAI), Yokohama, Kanagawa 230-0045, Japan.,Laboratory for Host Defence, RIKEN Center for Integrative Medical Science (IMS-RCAI), Yokohama, Kanagawa 230-0045, Japan.,Department of Immunology, Faculty of Medicine, Kagawa University, Miki, Kagawa 761-0793, Japan
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 138648, Singapore
| | - Tsuneyasu Kaisho
- Laboratory for Immune Regulation, World Premier International Research Center Initiative, Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan.,Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Wakayama 641-8509, Japan.,Laboratory for Inflammatory Regulation, RIKEN Center for Integrative Medical Science (IMS-RCAI), Yokohama, Kanagawa 230-0045, Japan.,Laboratory for Host Defence, RIKEN Center for Integrative Medical Science (IMS-RCAI), Yokohama, Kanagawa 230-0045, Japan
| |
Collapse
|
274
|
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
|
275
|
Abstract
Obesity and insulin resistance are associated with chronic inflammation in metabolic tissues such as adipose tissue and the liver. Recently, growing evidence has implicated the intestinal immune system as an important contributor to metabolic disease. Obesity predisposes to altered intestinal immunity and is associated with changes to the gut microbiota, intestinal barrier function, gut-residing innate and adaptive immune cells, and oral tolerance to luminal antigens. Accordingly, the gut immune system may represent a novel therapeutic target for systemic inflammation in insulin resistance. This review discusses the emerging field of intestinal immunity in obesity-related insulin resistance and how it affects metabolic disease.
Collapse
Affiliation(s)
- Daniel A Winer
- Diabetes Research Group, Division of Cellular and Molecular Biology, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, ON M5G 1L7, Canada; Department of Immunology, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Department of Pathology, University Health Network, 200 Elizabeth Street, Toronto, ON M5G 2C4, Canada.
| | - Helen Luck
- Diabetes Research Group, Division of Cellular and Molecular Biology, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, ON M5G 1L7, Canada; Department of Immunology, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Sue Tsai
- Diabetes Research Group, Division of Cellular and Molecular Biology, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Shawn Winer
- Diabetes Research Group, Division of Cellular and Molecular Biology, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, ON M5G 1L7, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Department of Laboratory Medicine, St. Michael's Hospital, 30 Bond Street, Toronto, ON M5B 1W8, Canada.
| |
Collapse
|
276
|
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
|
277
|
Berin MC, Shreffler WG. Mechanisms Underlying Induction of Tolerance to Foods. Immunol Allergy Clin North Am 2016; 36:87-102. [DOI: 10.1016/j.iac.2015.08.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
278
|
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: 466] [Impact Index Per Article: 51.8] [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
|
279
|
Da Silva N, Barton CR. Macrophages and dendritic cells in the post-testicular environment. Cell Tissue Res 2016; 363:97-104. [PMID: 26337514 PMCID: PMC4703462 DOI: 10.1007/s00441-015-2270-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 08/03/2015] [Indexed: 12/14/2022]
Abstract
Macrophages (MΦ) and dendritic cells (DCs) are heterogeneous families of functionally and developmentally related immune cells that play crucial roles in tissue homeostasis and the regulation of immune responses. During the past 5 years, immunologists have generated a considerable amount of data that challenge dogmas about the ontogeny and functions of these highly versatile cells. The male excurrent duct system plays a critical role in the establishment of fertility by allowing sperm maturation, transport and storage. In addition, it is challenged by pathogens and must establish a protective and tolerogenic environment for a continuous flow of autoantigenic spermatozoa. The post-testicular environment and, in particular, the epididymis contain an intricate network of DCs and MΦ; however, the immunophysiology of this intriguing and highly specialized mucosal system is poorly understood. This review summarizes the current trends in mouse MΦ and DC biology and speculates about their roles in the steady-state epididymis. Unraveling immune cell functions in the male reproductive tract is an essential prerequisite for the design of innovative strategies aimed at controlling male fertility and treating infertility.
Collapse
Affiliation(s)
- Nicolas Da Silva
- Division of Nephrology, Center for Systems Biology and Program in Membrane Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, 185 Cambridge Street, CPZN 8.218, Boston, MA 02114-2790, USA.
| | - Claire R Barton
- Division of Nephrology, Center for Systems Biology and Program in Membrane Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, 185 Cambridge Street, CPZN 8.218, Boston, MA 02114-2790, USA
| |
Collapse
|
280
|
Dolpady J, Sorini C, Di Pietro C, Cosorich I, Ferrarese R, Saita D, Clementi M, Canducci F, Falcone M. Oral Probiotic VSL#3 Prevents Autoimmune Diabetes by Modulating Microbiota and Promoting Indoleamine 2,3-Dioxygenase-Enriched Tolerogenic Intestinal Environment. J Diabetes Res 2016; 2016:7569431. [PMID: 26779542 PMCID: PMC4686713 DOI: 10.1155/2016/7569431] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 06/01/2015] [Accepted: 07/01/2015] [Indexed: 02/07/2023] Open
Abstract
The gut microbiota modulates the autoimmune pathogenesis of type 1 diabetes (T1D) via mechanisms that remain largely unknown. The inflammasome components are innate immune sensors that are highly influenced by the gut environment and play pivotal roles in maintaining intestinal immune homeostasis. In this study we show that modifications of the gut microbiota induced by oral treatment with Lactobacillaceae-enriched probiotic VSL#3, alone or in combination with retinoic acid (RA), protect NOD mice from T1D by affecting inflammasome at the intestinal level. In particular, we show that VSL#3 treatment inhibits IL-1β expression while enhancing release of protolerogenic components of the inflammasome, such as indoleamine 2,3-dioxygenase (IDO) and IL-33. Those modifications of the intestinal microenvironment in VSL#3-treated NOD mice modulate gut immunity by promoting differentiation of tolerogenic CD103(+) DCs and reducing differentiation/expansion of Th1 and Th17 cells in the intestinal mucosa and at the sites of autoimmunity, that is, within the pancreatic lymph nodes (PLN) of VSL#3-treated NOD mice. Our data provide a link between dietary factors, microbiota composition, intestinal inflammation, and immune homeostasis in autoimmune diabetes and could pave the way for new therapeutic approaches aimed at changing the intestinal microenvironment with probiotics to counterregulate autoimmunity and prevent T1D.
Collapse
MESH Headings
- Administration, Oral
- Age Factors
- Animals
- Autoimmunity
- Cellular Microenvironment
- Diabetes Mellitus, Type 1/enzymology
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/microbiology
- Diabetes Mellitus, Type 1/prevention & control
- Disease Models, Animal
- Gastrointestinal Microbiome
- Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Inflammasomes/immunology
- Inflammasomes/metabolism
- Interleukin-1beta/metabolism
- Interleukin-33/metabolism
- Intestines/enzymology
- Intestines/immunology
- Intestines/microbiology
- Lactobacillaceae/growth & development
- Lactobacillaceae/immunology
- Mice, Inbred NOD
- Probiotics/administration & dosage
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Th1 Cells/microbiology
- Th17 Cells/immunology
- Th17 Cells/metabolism
- Th17 Cells/microbiology
- Tretinoin/pharmacology
Collapse
Affiliation(s)
- Jayashree Dolpady
- Experimental Diabetes Unit, Diabetes Research Institute, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Chiara Sorini
- Experimental Diabetes Unit, Diabetes Research Institute, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Caterina Di Pietro
- Experimental Diabetes Unit, Diabetes Research Institute, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Ilaria Cosorich
- Experimental Diabetes Unit, Diabetes Research Institute, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Roberto Ferrarese
- Microbiology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Diego Saita
- Microbiology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Massimo Clementi
- Microbiology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Filippo Canducci
- Microbiology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Marika Falcone
- Experimental Diabetes Unit, Diabetes Research Institute, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
- *Marika Falcone:
| |
Collapse
|
281
|
Koscsó B, Gowda K, Bogunovic M. In vivo depletion and genetic targeting of mouse intestinal CX3CR1(+) mononuclear phagocytes. J Immunol Methods 2015; 432:13-23. [PMID: 26705686 DOI: 10.1016/j.jim.2015.12.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 11/14/2015] [Accepted: 12/11/2015] [Indexed: 12/26/2022]
Abstract
Mononuclear phagocytes (MPs) are an essential component of the intestinal immune system. They are comprised of a few dendritic cell and macrophage subsets, all with the common ability to sample extracellular milieu and to discriminate between dangerous and innocuous signals. Despite the commonality, each MP subset acquires distinct developmental pathways and unique functions, likely to fulfill needs of the tissue in which they reside. Some MP subsets develop from monocytes and are distinguished by their expression of CX3C-chemokine receptor 1 (CX3CR1). This manuscript summarizes our expertise in vivo targeting of intestinal CX3CR1(+) MP subsets. The described tools might be useful for studies of CX3CR1(+) MP function in various murine experimental models, particularly under non-inflammatory conditions.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Biomarkers/metabolism
- CX3C Chemokine Receptor 1
- Cell Lineage
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Down-Regulation
- Gene Targeting/methods
- Genotype
- Hybridomas
- Immunity, Mucosal
- Immunophenotyping
- Integrases/genetics
- Intestinal Mucosa/metabolism
- Intestines/drug effects
- Intestines/immunology
- Macrophages/drug effects
- Macrophages/immunology
- Macrophages/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Muramidase/genetics
- Muramidase/immunology
- Muramidase/metabolism
- Phenotype
- Promoter Regions, Genetic
- Receptors, Chemokine/deficiency
- Receptors, Chemokine/genetics
- Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors
- Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics
- Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology
- Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism
Collapse
Affiliation(s)
- Balázs Koscsó
- Department of Microbiology and Immunology, Penn State University College of Medicine and Milton Hershey Medical Center, Hershey, PA 17033, USA
| | - Kavitha Gowda
- Department of Microbiology and Immunology, Penn State University College of Medicine and Milton Hershey Medical Center, Hershey, PA 17033, USA
| | - Milena Bogunovic
- Department of Microbiology and Immunology, Penn State University College of Medicine and Milton Hershey Medical Center, Hershey, PA 17033, USA.
| |
Collapse
|
282
|
T cell polarizing properties of probiotic bacteria. Immunol Lett 2015; 168:337-42. [DOI: 10.1016/j.imlet.2015.11.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 10/29/2015] [Accepted: 11/04/2015] [Indexed: 12/30/2022]
|
283
|
Kreuger J, Phillipson M. Targeting vascular and leukocyte communication in angiogenesis, inflammation and fibrosis. Nat Rev Drug Discov 2015; 15:125-42. [PMID: 26612664 DOI: 10.1038/nrd.2015.2] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Regulation of vascular permeability, recruitment of leukocytes from blood to tissue and angiogenesis are all processes that occur at the level of the microvasculature during both physiological and pathological conditions. The interplay between microvascular cells and leukocytes during inflammation, together with the emerging roles of leukocytes in the modulation of the angiogenic process, make leukocyte-vascular interactions prime targets for therapeutics to potentially treat a wide range of diseases, including pathological and dysfunctional vessel growth, chronic inflammation and fibrosis. In this Review, we discuss how the different cell types that are present in and around microvessels interact, cooperate and instruct each other, and in this context we highlight drug targets as well as emerging druggable processes that can be exploited to restore tissue homeostasis.
Collapse
Affiliation(s)
- Johan Kreuger
- Department of Medical Cell Biology, Uppsala University, Husargatan 3, Uppsala, 75123, Sweden
| | - Mia Phillipson
- Department of Medical Cell Biology, Uppsala University, Husargatan 3, Uppsala, 75123, Sweden
| |
Collapse
|
284
|
Kayama H, Takeda K. Functions of innate immune cells and commensal bacteria in gut homeostasis. J Biochem 2015; 159:141-9. [PMID: 26615026 DOI: 10.1093/jb/mvv119] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 11/11/2015] [Indexed: 02/06/2023] Open
Abstract
The intestinal immune system remains unresponsive to beneficial microbes and dietary antigens while activating pro-inflammatory responses against pathogens for host defence. In intestinal mucosa, abnormal activation of innate immunity, which directs adaptive immune responses, causes the onset and/or progression of inflammatory bowel diseases. Thus, innate immunity is finely regulated in the gut. Multiple innate immune cell subsets have been identified in both murine and human intestinal lamina propria. Some innate immune cells play a key role in the maintenance of gut homeostasis by preventing inappropriate adaptive immune responses while others are associated with the pathogenesis of intestinal inflammation through development of Th1 and Th17 cells. In addition, intestinal microbiota and their metabolites contribute to the regulation of innate/adaptive immune responses. Accordingly, perturbation of microbiota composition can trigger intestinal inflammation by driving inappropriate immune responses.
Collapse
Affiliation(s)
- Hisako Kayama
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine and Laboratory of Mucosal Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Kiyoshi Takeda
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine and Laboratory of Mucosal Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
285
|
Do J, Visperas A, Freeman ML, Jang E, Kim S, Malissen B, Min B. γδ T cells support gut Ag-reactive colitogenic effector T-cell generation by enhancing Ag presentation by CD11b(+) DCs in the mesenteric LN. Eur J Immunol 2015; 46:340-6. [PMID: 26549797 DOI: 10.1002/eji.201545919] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 10/13/2015] [Accepted: 11/03/2015] [Indexed: 12/23/2022]
Abstract
T cells expressing the γδ TCR are dominant T-cell subsets in the intestinal immune system. We previously demonstrated that γδ T cells play important roles in augmenting Th17-type colitogenic immune responses in a T-cell-induced colitic inflammation model. However, its underlying mechanism remains poorly understood. In this study, an in vitro coculture system using effector T cells enriched in gut Ag-reactive cells was employed as a readout tool to search for gut Ag presenting APCs. We found that the presence of γδ T cells dramatically enhances gut Ag presentation within the mLN in mice. Gut Ag presentation by CD11b(+) DC subsets was particularly controlled by γδ T cells. Interestingly, γδ T-cell entry to the lymph nodes was essential to improve the Ag presentation. Therefore, our results highlight that γδ T cells play a previously unrecognized role to support colitogenic immunity by regulating gut Ag presentation in the draining LN.
Collapse
Affiliation(s)
- Jeongsu Do
- Department of Immunology, Lerner Research Institute, Lerner Research InstituteCleveland Clinic Foundation, Cleveland, OH, USA
| | - Anabelle Visperas
- Department of Immunology, Lerner Research Institute, Lerner Research InstituteCleveland Clinic Foundation, Cleveland, OH, USA
| | - Michael L Freeman
- Department of Immunology, Lerner Research Institute, Lerner Research InstituteCleveland Clinic Foundation, Cleveland, OH, USA
| | - Eunjung Jang
- Department of Immunology, Lerner Research Institute, Lerner Research InstituteCleveland Clinic Foundation, Cleveland, OH, USA
| | - Sohee Kim
- Department of Immunology, Lerner Research Institute, Lerner Research InstituteCleveland Clinic Foundation, Cleveland, OH, USA
| | - Bernard Malissen
- Centre d'Immunologie de Marseille-Luminy, Université de la Méditerranée, Case 906, Institut National de la Santé et de la Recherche Médicale, U631, Centre National de la Recherche Scientifique, UMR6102, Marseille, France
| | - Booki Min
- Department of Immunology, Lerner Research Institute, Lerner Research InstituteCleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
286
|
Berek C. Eosinophils: important players in humoral immunity. Clin Exp Immunol 2015; 183:57-64. [PMID: 26291602 DOI: 10.1111/cei.12695] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2015] [Indexed: 12/13/2022] Open
Abstract
Eosinophils perform numerous tasks. They are involved in inflammatory reactions associated with innate immune defence against parasitic infections and are also involved in pathological processes in response to allergens. Recently, however, it has become clear that eosinophils also play crucial non-inflammatory roles in the generation and maintenance of adaptive immune responses. Eosinophils, being a major source of the plasma cell survival factor APRIL (activation and proliferation-induced ligand), are essential not only for the long-term survival of plasma cells in the bone marrow, but also for the maintenance of these cells in the lamina propria which underlies the gut epithelium. At steady state under non-inflammatory conditions eosinophils are resident cells of the gastrointestinal tract, although only few are present in the major organized lymphoid tissue of the gut - the Peyer's patches (PP). Surprisingly, however, lack of eosinophils abolishes efficient class-switching of B cells to immunoglobulin (Ig)A in the germinal centres of PP. Thus, eosinophils are required to generate and to maintain mucosal IgA plasma cells, and as a consequence their absence leads to a marked reduction of IgA both in serum and in the gut-associated lymphoid tissues (GALT). Eosinophils thus have an essential part in long-term humoral immune protection, as they are crucial for the longevity of antibody-producing plasma cells in the bone marrow and, in addition, for gut immune homeostasis.
Collapse
Affiliation(s)
- C Berek
- B cell Immunology, Deutsches Rheuma Forschungszentrum, Berlin, Germany
| |
Collapse
|
287
|
Affiliation(s)
- M. C. Berin
- Pediatric Allergy and Immunology; Icahn School of Medicine at Mount Sinai; New York NY USA
| |
Collapse
|
288
|
Kedl RM, Tamburini BA. Antigen archiving by lymph node stroma: A novel function for the lymphatic endothelium. Eur J Immunol 2015; 45:2721-9. [PMID: 26278423 DOI: 10.1002/eji.201545739] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 06/01/2015] [Accepted: 08/12/2015] [Indexed: 12/22/2022]
Abstract
Secondary lymphoid stroma performs far more functions than simple structural support for lymphoid tissues, providing a host of soluble and membrane-bound cues to trafficking leukocytes during inflammation and homeostasis. More recently it has become clear that stromal cells can manipulate T-cell responses, either through direct antigen-mediated stimulation of T cells or more indirectly through the retention and management of antigen after viral infection or vaccination. In light of recent data, this review provides an overview of stromal cell subsets and functions during the progression of an adaptive immune response with particular emphasis on antigen capture and retention by follicular dendritic cells as well as the recently described "antigen archiving" function of lymphatic endothelial cells (LECs). Given its impact on the maintenance of protective immune memory, we conclude by discussing the most pressing questions pertaining to LEC antigen capture, archiving and exchange with hematopoetically derived antigen-presenting cells.
Collapse
Affiliation(s)
- Ross M Kedl
- Department of Immunology and Microbiology, University of Colorado Denver, School of Medicine, Aurora, CO, USA
| | - Beth A Tamburini
- Department of Immunology and Microbiology, University of Colorado Denver, School of Medicine, Aurora, CO, USA
| |
Collapse
|
289
|
Fransen F, Zagato E, Mazzini E, Fosso B, Manzari C, El Aidy S, Chiavelli A, D'Erchia AM, Sethi MK, Pabst O, Marzano M, Moretti S, Romani L, Penna G, Pesole G, Rescigno M. BALB/c and C57BL/6 Mice Differ in Polyreactive IgA Abundance, which Impacts the Generation of Antigen-Specific IgA and Microbiota Diversity. Immunity 2015; 43:527-40. [PMID: 26362264 DOI: 10.1016/j.immuni.2015.08.011] [Citation(s) in RCA: 202] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 05/12/2015] [Accepted: 08/10/2015] [Indexed: 12/20/2022]
Abstract
The interrelationship between IgAs and microbiota diversity is still unclear. Here we show that BALB/c mice had higher abundance and diversity of IgAs than C57BL/6 mice and that this correlated with increased microbiota diversity. We show that polyreactive IgAs mediated the entrance of non-invasive bacteria to Peyer's patches, independently of CX3CR1(+) phagocytes. This allowed the induction of bacteria-specific IgA and the establishment of a positive feedback loop of IgA production. Cohousing of mice or fecal transplantation had little or no influence on IgA production and had only partial impact on microbiota composition. Germ-free BALB/c, but not C57BL/6, mice already had polyreactive IgAs that influenced microbiota diversity and selection after colonization. Together, these data suggest that genetic predisposition to produce polyreactive IgAs has a strong impact on the generation of antigen-specific IgAs and the selection and maintenance of microbiota diversity.
Collapse
Affiliation(s)
- Floris Fransen
- Department of Experimental Oncology and Immunotherapy Programme, IEO, Via Adamello 16, 20139 Milan, Italy
| | - Elena Zagato
- Department of Experimental Oncology and Immunotherapy Programme, IEO, Via Adamello 16, 20139 Milan, Italy
| | - Elisa Mazzini
- Department of Experimental Oncology and Immunotherapy Programme, IEO, Via Adamello 16, 20139 Milan, Italy
| | - Bruno Fosso
- Institute of Biomembranes and Bioenergetics, Consiglio Nazionale delle Ricerche, Via Amendola 165/A, 70126 Bari, Italy
| | - Caterina Manzari
- Institute of Biomembranes and Bioenergetics, Consiglio Nazionale delle Ricerche, Via Amendola 165/A, 70126 Bari, Italy
| | - Sahar El Aidy
- Department of Experimental Oncology and Immunotherapy Programme, IEO, Via Adamello 16, 20139 Milan, Italy; Groningen Biomolecular Sciences and Biotechnology, Faculty of Mathematics and Natural Sciences, Nijenborgh 7, 9700 CC Groningen, the Netherlands
| | - Andrea Chiavelli
- Department of Experimental Oncology and Immunotherapy Programme, IEO, Via Adamello 16, 20139 Milan, Italy
| | - Anna Maria D'Erchia
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Via Orabona 4, 70125 Bari, Italy
| | - Maya K Sethi
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Stra. 1, 30625 Hannover, Germany
| | - Oliver Pabst
- Institute of Molecular Medicine, RWTH Aachen University, Carl-Neuberg-Str. 1, 52074 Aachen, Germany
| | - Marinella Marzano
- Institute of Biomembranes and Bioenergetics, Consiglio Nazionale delle Ricerche, Via Amendola 165/A, 70126 Bari, Italy
| | - Silvia Moretti
- Department of Experimental Medicine, University of Perugia, Polo Unico Sant'Andrea delle Fratte, 06132 Perugia, Italy
| | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, Polo Unico Sant'Andrea delle Fratte, 06132 Perugia, Italy
| | - Giuseppe Penna
- Department of Experimental Oncology and Immunotherapy Programme, IEO, Via Adamello 16, 20139 Milan, Italy
| | - Graziano Pesole
- Institute of Biomembranes and Bioenergetics, Consiglio Nazionale delle Ricerche, Via Amendola 165/A, 70126 Bari, Italy; Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Via Orabona 4, 70125 Bari, Italy
| | - Maria Rescigno
- Department of Experimental Oncology and Immunotherapy Programme, IEO, Via Adamello 16, 20139 Milan, Italy; Dipartimento di Scienze della salute, Universita' di Milano, via Rudini 8, 20142 Milan, Italy.
| |
Collapse
|
290
|
Parigi SM, Eldh M, Larssen P, Gabrielsson S, Villablanca EJ. Breast Milk and Solid Food Shaping Intestinal Immunity. Front Immunol 2015; 6:415. [PMID: 26347740 PMCID: PMC4541369 DOI: 10.3389/fimmu.2015.00415] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 07/28/2015] [Indexed: 12/22/2022] Open
Abstract
After birth, the intestinal immune system enters a critical developmental stage, in which tolerogenic and pro-inflammatory cells emerge to contribute to the overall health of the host. The neonatal health is continuously challenged by microbial colonization and food intake, first in the form of breast milk or formula and later in the form of solid food. The microbiota and dietary compounds shape the newborn immune system, which acquires the ability to induce tolerance against innocuous antigens or induce pro-inflammatory immune responses against pathogens. Disruption of these homeostatic mechanisms might lead to undesired immune reactions, such as food allergies and inflammatory bowel disease. Hence, a proper education and maturation of the intestinal immune system is likely important to maintain life-long intestinal homeostasis. In this review, the most recent literature regarding the effects of dietary compounds in the development of the intestinal immune system are discussed.
Collapse
Affiliation(s)
- Sara M Parigi
- Translational Immunology Unit, Department of Medicine Solna, Karolinska Institutet and University Hospital , Stockholm , Sweden
| | - Maria Eldh
- Translational Immunology Unit, Department of Medicine Solna, Karolinska Institutet and University Hospital , Stockholm , Sweden
| | - Pia Larssen
- Translational Immunology Unit, Department of Medicine Solna, Karolinska Institutet and University Hospital , Stockholm , Sweden
| | - Susanne Gabrielsson
- Translational Immunology Unit, Department of Medicine Solna, Karolinska Institutet and University Hospital , Stockholm , Sweden
| | - Eduardo J Villablanca
- Translational Immunology Unit, Department of Medicine Solna, Karolinska Institutet and University Hospital , Stockholm , Sweden
| |
Collapse
|
291
|
Panea C, Farkas AM, Goto Y, Abdollahi-Roodsaz S, Lee C, Koscsó B, Gowda K, Hohl TM, Bogunovic M, Ivanov II. Intestinal Monocyte-Derived Macrophages Control Commensal-Specific Th17 Responses. Cell Rep 2015; 12:1314-24. [PMID: 26279572 DOI: 10.1016/j.celrep.2015.07.040] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 06/11/2015] [Accepted: 07/16/2015] [Indexed: 01/12/2023] Open
Abstract
Generation of different CD4 T cell responses to commensal and pathogenic bacteria is crucial for maintaining a healthy gut environment, but the associated cellular mechanisms are poorly understood. Dendritic cells (DCs) and macrophages (Mfs) integrate microbial signals and direct adaptive immunity. Although the role of DCs in initiating T cell responses is well appreciated, how Mfs contribute to the generation of CD4 T cell responses to intestinal microbes is unclear. Th17 cells are critical for mucosal immune protection and at steady state are induced by commensal bacteria, such as segmented filamentous bacteria (SFB). Here, we examined the roles of mucosal DCs and Mfs in Th17 induction by SFB in vivo. We show that Mfs, and not conventional CD103(+) DCs, are essential for the generation of SFB-specific Th17 responses. Thus, Mfs drive mucosal T cell responses to certain commensal bacteria.
Collapse
Affiliation(s)
- Casandra Panea
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Adam M Farkas
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Yoshiyuki Goto
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Shahla Abdollahi-Roodsaz
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Carolyn Lee
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Balázs Koscsó
- Department of Microbiology and Immunology, College of Medicine and Milton S. Hershey Medical Center, Pennsylvania State University, Hershey, PA 17033, USA
| | - Kavitha Gowda
- Department of Microbiology and Immunology, College of Medicine and Milton S. Hershey Medical Center, Pennsylvania State University, Hershey, PA 17033, USA
| | - Tobias M Hohl
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Milena Bogunovic
- Department of Microbiology and Immunology, College of Medicine and Milton S. Hershey Medical Center, Pennsylvania State University, Hershey, PA 17033, USA
| | - Ivaylo I Ivanov
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
292
|
Dong J, Wong CK, Cai Z, Jiao D, Chu M, Lam CWK. Amelioration of allergic airway inflammation in mice by regulatory IL-35 through dampening inflammatory dendritic cells. Allergy 2015; 70:921-32. [PMID: 25869299 DOI: 10.1111/all.12631] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND IL-35, a new member of the IL-12 family, is an inhibitory cytokine produced by regulatory T and B lymphocytes that play a suppressive role in the inflammatory diseases. This study focuses on the cellular mechanism regulating the anti-inflammatory activity of IL-35 in asthmatic mice. METHODS Ovalbumin-induced asthmatic and humanized asthmatic mice were adopted to evaluate the in vivo anti-inflammatory activities of IL-35. For monitoring the airway, Penh value (% baseline) was measured using a whole-body plethysmograph. RESULTS In this study using ovalbumin-induced asthmatic mice, we observed that intraperitoneal injection of IL-35 during the allergen sensitization stage was more efficient than administration in the challenge stage for the amelioration of airway hyper-responsiveness. This was reflected by the significantly reduced concentration of asthma-related Th2 cytokines IL-5 and IL-13, as well as eosinophil counts in bronchoalveolar lavage fluid (all P < 0.05). IL-35 also significantly attenuated the accumulation of migratory CD11b+CD103(-) dendritic cells (DC) in the mediastinal lymph node (mLN) and lung of mice (all P < 0.05). IL-35 markedly inhibited the ovalbumin-induced conversion of recruited monocytes into inflammatory DC, which were then substantially reduced in mLN to cause less T-cell proliferation (all P < 0.05). Further study using the humanized asthmatic murine model also indicated human IL-35 exhibited a regulatory impact on allergic asthma. CONCLUSION Our findings suggest that IL-35 can act as a crucial regulatory cytokine to inhibit the development of allergic airway inflammation via suppressing the formation of inflammatory DC at the inflammatory site and their accumulation in the draining lymph nodes.
Collapse
Affiliation(s)
- J. Dong
- Department of Chemical Pathology; The Chinese University of Hong Kong; Prince of Wales Hospital; Shatin, NT, Hong Kong China
- Shenzhen Research Institute; The Chinese University of Hong Kong; Shenzhen
| | - C. K. Wong
- Department of Chemical Pathology; The Chinese University of Hong Kong; Prince of Wales Hospital; Shatin, NT, Hong Kong China
- Shenzhen Research Institute; The Chinese University of Hong Kong; Shenzhen
- Institute of Chinese Medicine and State Key Laboratory of Phytochemistry and Plant Resources in West China; The Chinese University of Hong Kong; Hong Kong
| | - Z. Cai
- Department of Chemical Pathology; The Chinese University of Hong Kong; Prince of Wales Hospital; Shatin, NT, Hong Kong China
- Shenzhen Research Institute; The Chinese University of Hong Kong; Shenzhen
| | - D. Jiao
- Department of Chemical Pathology; The Chinese University of Hong Kong; Prince of Wales Hospital; Shatin, NT, Hong Kong China
- Shenzhen Research Institute; The Chinese University of Hong Kong; Shenzhen
| | - M. Chu
- Department of Chemical Pathology; The Chinese University of Hong Kong; Prince of Wales Hospital; Shatin, NT, Hong Kong China
- Shenzhen Research Institute; The Chinese University of Hong Kong; Shenzhen
| | - C. W. K. Lam
- State Key Laboratory of Quality Research in Chinese Medicine; Macau Institute for Applied Research in Medicine and Health; Macau University of Science and Technology; Taipa, Macau China
| |
Collapse
|
293
|
Glass AM, Snyder EG, Taffet SM. Connexins and pannexins in the immune system and lymphatic organs. Cell Mol Life Sci 2015; 72:2899-910. [PMID: 26100515 PMCID: PMC11113820 DOI: 10.1007/s00018-015-1966-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Accepted: 06/11/2015] [Indexed: 12/11/2022]
Abstract
Connexin43 and pannexin1 are found in immune cells. While gap junctional communication has been demonstrated between immune cells, hemichannels have been implicated in many cellular functions. Among the functions involved as being connexin dependent and pannexin dependent are cell migration, phagocytosis, antigen presentation, T-cell reactivity and B-cell responses. Surprisingly, many of these connexin-related and pannexin-related functions are not recapitulated in in vivo models. This is leading to a reevaluation of the role of these proteins in immune function.
Collapse
Affiliation(s)
- Aaron M. Glass
- Department of Microbiology and Immunology, SUNY Upstate Medical University, 750 E Adams Street, Syracuse, NY 13210 USA
| | - Elizabeth G. Snyder
- Department of Microbiology and Immunology, SUNY Upstate Medical University, 750 E Adams Street, Syracuse, NY 13210 USA
| | - Steven M. Taffet
- Department of Microbiology and Immunology, SUNY Upstate Medical University, 750 E Adams Street, Syracuse, NY 13210 USA
| |
Collapse
|
294
|
Tittarelli A, Janji B, Van Moer K, Noman MZ, Chouaib S. The Selective Degradation of Synaptic Connexin 43 Protein by Hypoxia-induced Autophagy Impairs Natural Killer Cell-mediated Tumor Cell Killing. J Biol Chem 2015. [PMID: 26221040 DOI: 10.1074/jbc.m115.651547] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Although natural killer (NK) cells play an important role in the control of melanoma, hypoxic stress in the tumor microenvironment may impair NK-mediated tumor cell killing by mechanisms that are not fully understood. In this study, we investigated the effect of hypoxia on the expression and channel activity of connexin 43 (Cx43) in melanoma cells and its impact on their susceptibility to NK cell-mediated lysis. Our results demonstrated that hypoxic stress increases Cx43 expression in melanoma cells via hypoxia-inducible factor-1α (HIF-1α) transcriptional activity. Hypoxic cells displaying increased Cx43 expression were less susceptible to NK cell-mediated lysis compared with normoxic cells expressing a moderate level of Cx43. Conversely, when overexpressed in normoxic tumor cells, Cx43 improves their susceptibility to N cell-mediated killing. We show that the NK cell immune synapse formed with normoxic melanoma cells is more stable and contains a high level of gap-junctional Cx43 whereas that formed with hypoxic cells is less stable and contains a significant lower level of gap-junctional Cx43. We provide evidence that the activation of autophagy in hypoxic melanoma cells selectively degrades gap-junctional Cx43, leading to the destabilization of the immune synapse and the impairment of NK cell-mediated killing. Inhibition of autophagy by genetic or pharmacological approaches as well as expression of the non-degradable form of Cx43 significantly restore its accumulation at the immune synapse and improves N cell-mediated lysis of hypoxic melanoma cells. This study provides the first evidence that the hypoxic microenvironment negatively affects the immune surveillance of tumors by NK cells through the modulation of Cx43-mediated intercellular communications.
Collapse
Affiliation(s)
- Andrés Tittarelli
- From the INSERM U753, Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, 94805 Villejuif, France and
| | - Bassam Janji
- the Laboratory of Experimental Cancer Research, Department of Oncology, Luxembourg Institute of Health, 1526 Luxembourg City, Luxembourg
| | - Kris Van Moer
- the Laboratory of Experimental Cancer Research, Department of Oncology, Luxembourg Institute of Health, 1526 Luxembourg City, Luxembourg
| | - Muhammad Zaeem Noman
- From the INSERM U753, Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, 94805 Villejuif, France and
| | - Salem Chouaib
- From the INSERM U753, Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, 94805 Villejuif, France and
| |
Collapse
|
295
|
Abstract
The importance of CD4 T lymphocytes in immunity to M. tuberculosis is well established; however, how dendritic cells activate T cells in vivo remains obscure. In this issue of Cell Host & Microbe, Srivastava and Ernst (2014) report a mechanism of antigen transfer for efficient activation of antimycobacterial T cells.
Collapse
|
296
|
Abstract
Leading scientists in microbiome research met at Lake Titisee, Germany, in April 2014 to discuss the current state of the field, the most urgent and unresolved questions, state-of-the-art technological advances, and new avenues of future research. We summarize some of the concepts and themes discussed at this meeting.
Collapse
Affiliation(s)
| | - Eran Elinav
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
297
|
Stary G, Olive A, Radovic-Moreno AF, Gondek D, Alvarez D, Basto PA, Perro M, Vrbanac VD, Tager AM, Shi J, Yethon JA, Farokhzad OC, Langer R, Starnbach MN, von Andrian UH. VACCINES. A mucosal vaccine against Chlamydia trachomatis generates two waves of protective memory T cells. Science 2015; 348:aaa8205. [PMID: 26089520 DOI: 10.1126/science.aaa8205] [Citation(s) in RCA: 290] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Genital Chlamydia trachomatis (Ct) infection induces protective immunity that depends on interferon-γ-producing CD4 T cells. By contrast, we report that mucosal exposure to ultraviolet light (UV)-inactivated Ct (UV-Ct) generated regulatory T cells that exacerbated subsequent Ct infection. We show that mucosal immunization with UV-Ct complexed with charge-switching synthetic adjuvant particles (cSAPs) elicited long-lived protection in conventional and humanized mice. UV-Ct-cSAP targeted immunogenic uterine CD11b(+)CD103(-) dendritic cells (DCs), whereas UV-Ct accumulated in tolerogenic CD11b(-)CD103(+) DCs. Regardless of vaccination route, UV-Ct-cSAP induced systemic memory T cells, but only mucosal vaccination induced effector T cells that rapidly seeded uterine mucosa with resident memory T cells (T(RM) cells). Optimal Ct clearance required both T(RM) seeding and subsequent infection-induced recruitment of circulating memory T cells. Thus, UV-Ct-cSAP vaccination generated two synergistic memory T cell subsets with distinct migratory properties.
Collapse
Affiliation(s)
- Georg Stary
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Andrew Olive
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Aleksandar F Radovic-Moreno
- Harvard-MIT Division of Health Sciences & Technology, Cambridge, MA 02139, USA.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - David Gondek
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - David Alvarez
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Pamela A Basto
- Harvard-MIT Division of Health Sciences & Technology, Cambridge, MA 02139, USA.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Mario Perro
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Vladimir D Vrbanac
- The Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Andrew M Tager
- The Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jinjun Shi
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - Omid C Farokhzad
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,King Abdulaziz University, Jeddah, Saudi Arabia
| | - Robert Langer
- Harvard-MIT Division of Health Sciences & Technology, Cambridge, MA 02139, USA.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Michael N Starnbach
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Ulrich H von Andrian
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA.,The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| |
Collapse
|
298
|
Yin Y, Qin T, Wang X, Lin J, Yu Q, Yang Q. CpG DNA assists the whole inactivated H9N2 influenza virus in crossing the intestinal epithelial barriers via transepithelial uptake of dendritic cell dendrites. Mucosal Immunol 2015; 8:799-814. [PMID: 25492476 DOI: 10.1038/mi.2014.110] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Accepted: 10/09/2014] [Indexed: 02/04/2023]
Abstract
Intestinal mucosa remains a pivotal barrier for the oral vaccine absorption of H9N2 whole inactivated influenza virus (WIV). However, CpG DNA, as an adjuvant, can effectively improve relevant mucosal and systemic immunity. The downstream mechanism is well confirmed, yet the evidence of CpG DNA assisting H9N2 WIV in transepithelial delivery is lacking. Here, we reported both in vitro and in vivo that CpG DNA combined with H9N2 WIV was capable of recruiting additional dendritic cells (DCs) to the intestinal epithelial cells (ECs) to form transepithelial dendrites (TEDs) for luminal viral uptake. Both CD103(+) and CD103(-) DCs participated in this process. The engagement of the chemokine CCL20 from the apical ECs and the DCs drove DC recruitment and TED formation. Virus-loaded CD103(+) but not CD103(-) DCs also quickly migrated into mesenteric lymph nodes within 2 h. Moreover, the mechanism of CpG DNA was independent of epithelial transcytosis and disruption of the epithelial barriers. Finally, the subsequent phenotypic and functional maturation of DCs was also enhanced. Our findings indicated that CpG DNA improved the delivery of H9N2 WIV via TEDs of intestinal DCs, and this may be an important mechanism for downstream effective antigen-specific immune responses.
Collapse
Affiliation(s)
- Y Yin
- Key Lab of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu, People's Republic of China
| | - T Qin
- Key Lab of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu, People's Republic of China
| | - X Wang
- Key Lab of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu, People's Republic of China
| | - J Lin
- Key Lab of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu, People's Republic of China
| | - Q Yu
- Key Lab of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu, People's Republic of China
| | - Q Yang
- Key Lab of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
299
|
Flannigan KL, Geem D, Harusato A, Denning TL. Intestinal Antigen-Presenting Cells: Key Regulators of Immune Homeostasis and Inflammation. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1809-19. [PMID: 25976247 PMCID: PMC4483458 DOI: 10.1016/j.ajpath.2015.02.024] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 02/09/2015] [Accepted: 02/12/2015] [Indexed: 12/31/2022]
Abstract
The microbiota that populate the mammalian intestine are critical for proper host physiology, yet simultaneously pose a potential danger. Intestinal antigen-presenting cells, namely macrophages and dendritic cells (DCs), are integral components of the mucosal innate immune system that maintain co-existence with the microbiota in face of this constant threat. Intestinal macrophages and DCs integrate signals from the microenvironment to orchestrate innate and adaptive immune responses that ultimately lead to durable tolerance of the microbiota. Tolerance is not a default response, however, because macrophages and DCs remain poised to vigorously respond to pathogens that breach the epithelial barrier. In this review, we summarize the salient features of macrophages and DCs in the healthy and inflamed intestine and discuss how signals from the microbiota can influence their function.
Collapse
Affiliation(s)
- Kyle L Flannigan
- Institute for Biomedical Sciences, Center for Inflammation, Immunity, and Infection, Georgia State University, Atlanta
| | - Duke Geem
- Institute for Biomedical Sciences, Center for Inflammation, Immunity, and Infection, Georgia State University, Atlanta; Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - Akihito Harusato
- Institute for Biomedical Sciences, Center for Inflammation, Immunity, and Infection, Georgia State University, Atlanta
| | - Timothy L Denning
- Institute for Biomedical Sciences, Center for Inflammation, Immunity, and Infection, Georgia State University, Atlanta.
| |
Collapse
|
300
|
Abstract
The mechanisms whereby the gut mucosa tolerates trillions of commensal bacteria without developing inflammation remain poorly understood. A recent Science article reveals that gut innate lymphoid cells constrain inflammatory T cell responses to commensal bacteria by adopting a strategy usually deployed by thymic epithelial cells to negatively select self-reactive T cells.
Collapse
Affiliation(s)
- Giuliana Magri
- Institut Hospital del Mar 'Investigacions Mèdiques, Barcelona Biomedical Research Park, Barcelona, Spain
| | - Andrea Cerutti
- Institut Hospital del Mar 'Investigacions Mèdiques, Barcelona Biomedical Research Park, Barcelona, Spain.,Catalan Institute for Research and Advanced Studies (ICREA), Barcelona Biomedical Research Park, Barcelona, Spain.,Immunology Institute, Department of Medicine, Division of Clinical Immunology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|