Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Front Immunol ; 13: 995432, 2022.
Article in English | MEDLINE | ID: mdl-36225918

ABSTRACT

Parasitic helminth infections remain a significant global health issue and are responsible for devastating morbidity and economic hardships. During infection, helminths migrate through different host organs, which results in substantial tissue damage and the release of diverse effector molecules by both hematopoietic and non-hematopoietic cells. Thus, host protective responses to helminths must initiate mechanisms that help to promote worm clearance while simultaneously mitigating tissue injury. The specialized immunity that promotes these responses is termed type 2 inflammation and is initiated by the recruitment and activation of hematopoietic stem/progenitor cells, mast cells, basophils, eosinophils, dendritic cells, neutrophils, macrophages, myeloid-derived suppressor cells, and group 2 innate lymphoid cells. Recent work has also revealed the importance of neuron-derived signals in regulating type 2 inflammation and antihelminth immunity. These studies suggest that multiple body systems coordinate to promote optimal outcomes post-infection. In this review, we will describe the innate immune events that direct the scope and intensity of antihelminth immunity. Further, we will highlight the recent progress made in our understanding of the neuro-immune interactions that regulate these pathways and discuss the conceptual advances they promote.


Subject(s)
Helminthiasis , Helminths , Animals , Immunity, Innate , Inflammation , Lymphocytes
2.
J Immunol ; 209(10): 1827-1831, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36216513

ABSTRACT

Neutrophils are critical for the direct eradication of Aspergillus fumigatus conidia, but whether they mediate antifungal defense beyond their role as effectors is unclear. In this study, we demonstrate that neutrophil depletion impairs the activation of protective antifungal CCR2+ inflammatory monocytes. In the absence of neutrophils, monocytes displayed limited differentiation into monocyte-derived dendritic cells, reduced formation of reactive oxygen species, and diminished conidiacidal activity. Upstream regulator analysis of the transcriptional response in monocytes predicted a loss of STAT1-dependent signals as the potential basis for the dysfunction seen in neutrophil-depleted mice. We find that conditional removal of STAT1 on CCR2+ cells results in diminished antifungal monocyte responses, whereas exogenous administration of IFN-γ to neutrophil-depleted mice restores monocyte-derived dendritic cell maturation and reactive oxygen species production. Altogether, our findings support a critical role for neutrophils in antifungal immunity not only as effectors but also as important contributors to antifungal monocyte activation, in part by regulating STAT1-dependent functions.


Subject(s)
Monocytes , Neutrophils , Mice , Animals , Antifungal Agents , Reactive Oxygen Species , Aspergillus fumigatus
3.
Proc Natl Acad Sci U S A ; 119(37): e2201645119, 2022 09 13.
Article in English | MEDLINE | ID: mdl-36070344

ABSTRACT

Neuroimmune interactions are crucial for regulating immunity and inflammation. Recent studies have revealed that the central nervous system (CNS) senses peripheral inflammation and responds by releasing molecules that limit immune cell activation, thereby promoting tolerance and tissue integrity. However, the extent to which this is a bidirectional process, and whether peripheral immune cells also promote tolerance mechanisms in the CNS remains poorly defined. Here we report that helminth-induced type 2 inflammation promotes monocyte responses in the brain that are required to inhibit excessive microglial activation and host death. Mechanistically, infection-induced monocytes express YM1 that is sufficient to inhibit tumor necrosis factor production from activated microglia. Importantly, neuroprotective monocytes persist in the brain, and infected mice are protected from subsequent lipopolysaccharide-induced neuroinflammation months after infection-induced inflammation has resolved. These studies demonstrate that infiltrating monocytes promote CNS homeostasis in response to inflammation in the periphery and demonstrate that a peripheral infection can alter the immunologic landscape of the host brain.


Subject(s)
Brain , Encephalitis , Homeostasis , Monocytes , Neuroimmunomodulation , Trichinella spiralis , Trichinellosis , Animals , Brain/immunology , Brain/parasitology , Encephalitis/immunology , Encephalitis/parasitology , Homeostasis/immunology , Lectins/metabolism , Mice , Microglia/immunology , Monocytes/immunology , Trichinella spiralis/immunology , Trichinellosis/immunology , Trichinellosis/pathology , beta-N-Acetylhexosaminidases/metabolism
4.
Nat Immunol ; 23(6): 947-959, 2022 06.
Article in English | MEDLINE | ID: mdl-35552540

ABSTRACT

Inflammation is an important component of fibrosis but immune processes that orchestrate kidney fibrosis are not well understood. Here we apply single-cell sequencing to a mouse model of kidney fibrosis. We identify a subset of kidney tubule cells with a profibrotic-inflammatory phenotype characterized by the expression of cytokines and chemokines associated with immune cell recruitment. Receptor-ligand interaction analysis and experimental validation indicate that CXCL1 secreted by profibrotic tubules recruits CXCR2+ basophils. In mice, these basophils are an important source of interleukin-6 and recruitment of the TH17 subset of helper T cells. Genetic deletion or antibody-based depletion of basophils results in reduced renal fibrosis. Human kidney single-cell, bulk gene expression and immunostaining validate a function for basophils in patients with kidney fibrosis. Collectively, these studies identify basophils as contributors to the development of renal fibrosis and suggest that targeting these cells might be a useful clinical strategy to manage chronic kidney disease.


Subject(s)
Basophils , Renal Insufficiency, Chronic , Animals , Fibrosis , Humans , Kidney/metabolism , Kidney Tubules , Mice , Renal Insufficiency, Chronic/metabolism , Single-Cell Analysis
5.
Cell Rep ; 38(2): 110215, 2022 01 11.
Article in English | MEDLINE | ID: mdl-35021079

ABSTRACT

Macrophages are known to mediate anti-helminth responses, but it remains uncertain which subsets are involved or how macrophages actually kill helminths. Here, we show rapid monocyte recruitment to the lung after infection with the nematode parasite Nippostrongylus brasiliensis. In this inflamed tissue microenvironment, these monocytes differentiate into an alveolar macrophage (AM)-like phenotype, expressing both SiglecF and CD11c, surround invading parasitic larvae, and preferentially kill parasites in vitro. Monocyte-derived AMs (Mo-AMs) express type 2-associated markers and show a distinct remodeling of the chromatin landscape relative to tissue-derived AMs (TD-AMs). In particular, they express high amounts of arginase-1 (Arg1), which we demonstrate mediates helminth killing through L-arginine depletion. These studies indicate that recruited monocytes are selectively programmed in the pulmonary environment to express AM markers and an anti-helminth phenotype.


Subject(s)
Lung/immunology , Macrophages, Alveolar/immunology , Strongylida Infections/immunology , Animals , Arginase/metabolism , Cell Differentiation , Cytokines , Female , Lung/parasitology , Macrophages/immunology , Male , Mice , Mice, Inbred BALB C , Nippostrongylus , Strongylida Infections/parasitology
6.
Semin Immunol ; 53: 101529, 2021 03.
Article in English | MEDLINE | ID: mdl-34815162

ABSTRACT

It has been appreciated that basophilia is a common feature of helminth infections for approximately 50 years. The ability of basophils to secrete IL-4 and other type 2 cytokines has supported the prevailing notion that basophils contribute to antihelminth immunity by promoting optimal type 2 T helper (Th2) cell responses. While this appears to be the case in several helminth infections, emerging studies are also revealing that the effector functions of basophils are extremely diverse and parasite-specific. Further, new reports now suggest that basophils can restrict type 2 inflammation in a manner that preserves the integrity of helminth-affected tissue. Finally, exciting data has also demonstrated that basophils can regulate inflammation by participating in neuro-immune interactions. This article will review the current state of basophil biology and describe how recent studies are transforming our understanding of the role basophils play in the context of helminth infections.


Subject(s)
Basophils , Helminths , Animals , Cytokines/metabolism , Helminths/metabolism , Humans , Inflammation , Th2 Cells
7.
Nat Commun ; 12(1): 3371, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34099671

ABSTRACT

The role of p53 in tumor suppression has been extensively studied and well-established. However, the role of p53 in parasitic infections and the intestinal type 2 immunity is unclear. Here, we report that p53 is crucial for intestinal type 2 immunity in response to the infection of parasites, such as Tritrichomonas muris and Nippostrongylus brasiliensis. Mechanistically, p53 plays a critical role in the activation of the tuft cell-IL-25-type 2 innate lymphoid cell circuit, partly via transcriptional regulation of Lrmp in tuft cells. Lrmp modulates Ca2+ influx and IL-25 release, which are critical triggers of type 2 innate lymphoid cell response. Our results thus reveal a previously unrecognized function of p53 in regulating intestinal type 2 immunity to protect against parasitic infections, highlighting the role of p53 as a guardian of immune integrity.


Subject(s)
Immunity, Innate/immunology , Intestines/immunology , Nippostrongylus/immunology , Parasitic Diseases/immunology , Tritrichomonas/immunology , Tumor Suppressor Protein p53/immunology , Animals , Cell Line, Tumor , Eosinophils/immunology , Eosinophils/parasitology , Gene Expression Regulation , Goblet Cells/immunology , Goblet Cells/parasitology , Host-Parasite Interactions/immunology , Humans , Intestine, Small/immunology , Intestine, Small/metabolism , Intestine, Small/parasitology , Intestines/parasitology , Membrane Proteins/genetics , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Nippostrongylus/physiology , Parasitic Diseases/metabolism , Parasitic Diseases/parasitology , Tritrichomonas/physiology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
9.
Nat Immunol ; 21(10): 1181-1193, 2020 10.
Article in English | MEDLINE | ID: mdl-32807943

ABSTRACT

Type 2 cytokine responses promote parasitic immunity and initiate tissue repair; however, they can also result in immunopathologies when not properly restricted. Although basophilia is recognized as a common feature of type 2 inflammation, the roles basophils play in regulating these responses are unknown. Here, we demonstrate that helminth-induced group 2 innate lymphoid cell (ILC2) responses are exaggerated in the absence of basophils, resulting in increased inflammation and diminished lung function. Additionally, we show that ILC2s from basophil-depleted mice express reduced amounts of the receptor for the neuropeptide neuromedin B (NMB). Critically, NMB stimulation inhibited ILC2 responses from control but not basophil-depleted mice, and basophils were sufficient to directly enhance NMB receptor expression on ILC2s. These studies suggest that basophils prime ILC2s to respond to neuron-derived signals necessary to maintain tissue integrity. Further, these data provide mechanistic insight into the functions of basophils and identify NMB as a potent inhibitor of type 2 inflammation.


Subject(s)
Basophils/immunology , Lung/metabolism , Lymphocytes/immunology , Nippostrongylus/physiology , Strongylida Infections/immunology , Animals , Cell Communication , Cells, Cultured , Cytokines/metabolism , Immunity, Innate , Lung/pathology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurokinin B/analogs & derivatives , Neurokinin B/metabolism , Th2 Cells/immunology , Tryptases/genetics
10.
PLoS Pathog ; 16(5): e1008579, 2020 05.
Article in English | MEDLINE | ID: mdl-32421753

ABSTRACT

Anti-helminth responses require robust type 2 cytokine production that simultaneously promotes worm expulsion and initiates the resolution of helminth-induced wounds and hemorrhaging. However, how infection-induced changes in hematopoiesis contribute to these seemingly distinct processes remains unknown. Recent studies have suggested the existence of a hematopoietic progenitor with dual mast cell-erythrocyte potential. Nonetheless, whether and how these progenitors contribute to host protection during an active infection remains to be defined. Here, we employed single cell RNA-sequencing and identified that the metabolic enzyme, carbonic anhydrase (Car) 1 marks a predefined bone marrow-resident hematopoietic progenitor cell (HPC) population. Next, we generated a Car1-reporter mouse model and found that Car1-GFP positive progenitors represent bipotent mast cell/erythrocyte precursors. Finally, we show that Car1-expressing HPCs simultaneously support mast cell and erythrocyte responses during Trichinella spiralis infection. Collectively, these data suggest that mast cell/erythrocyte precursors are mobilized to promote type 2 cytokine responses and alleviate helminth-induced blood loss, developmentally linking these processes. Collectively, these studies reveal unappreciated hematopoietic events initiated by the host to combat helminth parasites and provide insight into the evolutionary pressure that may have shaped the developmental relationship between mast cells and erythrocytes.


Subject(s)
Erythroid Precursor Cells/immunology , Erythropoiesis/immunology , Mast Cells/immunology , Mastocytosis/immunology , Trichinella spiralis/immunology , Trichinellosis/immunology , Animals , Carbonic Anhydrase I/genetics , Carbonic Anhydrase I/immunology , Erythroid Precursor Cells/parasitology , Erythroid Precursor Cells/pathology , Female , Mast Cells/parasitology , Mast Cells/pathology , Mastocytosis/genetics , Mastocytosis/pathology , Mice , Mice, Transgenic , Trichinellosis/genetics , Trichinellosis/pathology
11.
J Immunol ; 203(5): 1230-1241, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31366712

ABSTRACT

Basophils are innate immune cells associated with type 2 immunity, allergic reactions, and host defense against parasite infections. In this study, we show that the transcription factor PLZF, which is known for its essential role in the function and development of several innate lymphocyte subsets, is also important for the myeloid-derived basophil lineage. PLZF-deficient mice had decreased numbers of basophil progenitors in the bone marrow and mature basophils in multiple peripheral tissues. Functionally, PLZF-deficient basophils were less responsive to IgE activation and produced reduced amounts of IL-4. The altered function of basophils resulted in a blunted Th2 T cell response to a protein allergen. Additionally, PLZF-deficient basophils had reduced expression of the IL-18 receptor, which impacted migration to lungs. PLZF, therefore, is a major player in controlling type 2 immune responses mediated not only by innate lymphocytes but also by myeloid-derived cells.


Subject(s)
Basophils/immunology , Promyelocytic Leukemia Zinc Finger Protein/immunology , Transcription Factors/immunology , Allergens/immunology , Animals , Immunity, Innate/immunology , Immunoglobulin E/immunology , Interleukin-4/immunology , Interleukin-8/immunology , Lymphocyte Subsets/immunology , Mice , Mice, Knockout , Myeloid Cells/immunology , Th2 Cells/immunology
12.
Curr Opin Allergy Clin Immunol ; 19(2): 175-184, 2019 04.
Article in English | MEDLINE | ID: mdl-30672750

ABSTRACT

PURPOSE OF REVIEW: Allergic diseases represent a growing global health concern, especially among pediatric populations. Current strategies for the treatment of allergies and asthma focus on limiting the severity of the symptoms; however, additional research investigating the mechanisms promoting inflammation in the context of allergic reactions may lead to the development of more effective therapeutic strategies. RECENT FINDINGS: Novel studies have highlighted the contributions of innate lymphocytes to the induction of inflammatory responses to allergens. Remarkably, neuron-derived signals, hormones, and even vitamins have been suggested to modulate the activity of innate lymphocytes, opening new windows of opportunity for the treatment of allergic inflammation. SUMMARY: These studies highlight the complex interactions of the nervous, endocrine, and immune system that promote pathology in the context of allergic inflammation. Further studies are required to understand these interactions in order to aid in the development of novel and much-needed therapies to treat allergic conditions.


Subject(s)
Hypersensitivity/immunology , Inflammation/immunology , Lymphocytes/immunology , Allergens/immunology , Animals , Hormones/metabolism , Humans , Immunity, Innate , Immunomodulation , Neuroimmunomodulation
13.
Immunity ; 49(4): 709-724.e8, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30291028

ABSTRACT

B cells thwart antigenic aggressions by releasing immunoglobulin M (IgM), IgG, IgA, and IgE, which deploy well-understood effector functions. In contrast, the role of secreted IgD remains mysterious. We found that some B cells generated IgD-secreting plasma cells following early exposure to external soluble antigens such as food proteins. Secreted IgD targeted basophils by interacting with the CD44-binding protein galectin-9. When engaged by antigen, basophil-bound IgD increased basophil secretion of interleukin-4 (IL-4), IL-5, and IL-13, which facilitated the generation of T follicular helper type 2 cells expressing IL-4. These germinal center T cells enhanced IgG1 and IgE but not IgG2a and IgG2b responses to the antigen initially recognized by basophil-bound IgD. In addition, IgD ligation by antigen attenuated allergic basophil degranulation induced by IgE co-ligation. Thus, IgD may link B cells with basophils to optimize humoral T helper type 2-mediated immunity against common environmental soluble antigens.


Subject(s)
Basophils/immunology , Galectins/immunology , Hyaluronan Receptors/immunology , Immunoglobulin D/immunology , Th2 Cells/immunology , Animals , Basophils/metabolism , Cell Line, Tumor , Cells, Cultured , Galectins/genetics , Galectins/metabolism , Gene Expression Profiling/methods , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Immunoglobulin D/metabolism , Immunoglobulin E/immunology , Immunoglobulin E/metabolism , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Interleukin-4/genetics , Interleukin-4/immunology , Interleukin-4/metabolism , Mice, Inbred BALB C , Protein Binding , Th2 Cells/metabolism
14.
Trends Parasitol ; 34(10): 861-880, 2018 10.
Article in English | MEDLINE | ID: mdl-30177466

ABSTRACT

Helminth infections represent a significant public health concern resulting in devastating morbidity and economic consequences across the globe. Helminths migrate through mucosal sites causing tissue damage and the induction of type 2 immune responses. Antihelminth protection relies on the mobilization and activation of multiple immune cells, including type 2 innate lymphocytes (ILC2s), basophils, mast cells, macrophages, and hematopoietic stem/progenitor cells. Further, epithelial cells and neurons have been recognized as important regulators of type 2 immunity. Collectively, these pathways stimulate host-protective responses necessary for worm expulsion and the healing of affected tissues. In this review we focus on the innate immune pathways that regulate immunity to helminth parasites and describe how better understanding of these pathways may lead to the development of new therapeutic strategies.


Subject(s)
Helminthiasis/immunology , Helminths/immunology , Host-Parasite Interactions/immunology , Immunity, Innate , Animals , Humans
15.
J Autoimmun ; 79: 39-52, 2017 May.
Article in English | MEDLINE | ID: mdl-28126203

ABSTRACT

Regulatory T cells (Tregs) are a subset of CD4+ T cells with suppressive function and are critical for limiting inappropriate activation of T cells. Hence, the expansion of Tregs is an attractive strategy for the treatment of autoimmune diseases. Here, we demonstrate that the skin possesses the remarkable capacity to systemically expand Treg numbers by producing thymic stromal lymphopoietin (TSLP) in response to vitamin D receptor stimulation. An ∼2-fold increase in the proportion and absolute number of Tregs was observed in mice treated topically but not systemically with the Vitamin D3 analog MC903. This expansion of Tregs was dependent on TSLP receptor signaling but not on VDR signaling in hematopoietic cells. However, TSLP receptor expression by Tregs was not required for their proliferation. Rather, skin-derived TSLP promoted Treg expansion through dendritic cells. Importantly, treatment of skin with MC903 significantly lowered the incidence of autoimmune diabetes in non-obese diabetic mice and attenuated disease score in experimental autoimmune encephalomyelitis. Together, these data demonstrate that the skin has the remarkable potential to control systemic immune responses and that Vitamin D-mediated stimulation of skin could serve as a novel strategy to therapeutically modulate the systemic immune system for the treatment of autoimmunity.


Subject(s)
Cytokines/metabolism , Skin/immunology , Skin/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Biomarkers , Cholecalciferol/analogs & derivatives , Cholecalciferol/pharmacology , Cytokines/pharmacology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphocyte Count , Mice , Mice, Knockout , Mice, Transgenic , Models, Biological , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/drug effects , Thymic Stromal Lymphopoietin
16.
Curr Pharmacol Rep ; 3(6): 346-359, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29399438

ABSTRACT

PURPOSE OF REVIEW: It is well established that T helper type 2 (TH2) immune responses are necessary to provide protection against helminth parasites but also to promote the detrimental inflammation associated with allergies and asthma. Given the importance of type 2 immunity and inflammation, many studies have focused on better understanding the factors that regulate TH2 cell development and activation. As a result, significant progress has been made in understanding the signaling pathways and molecular events necessary to promote TH2 cell polarization. In addition to the adaptive compartment, emerging studies are better defining the innate immune pathways needed to promote TH2 cell responses. Given the recent and substantial growth of this field, the purpose of this review is to highlight recent studies defining the innate immune events that promote immunity to helminth parasites and allergic inflammation. RECENT FINDINGS: Emerging studies have begun to elucidate the importance of cytokine alarmins such as thymic stromal lymphopoietin (TSLP), IL-25 (IL-17E) and IL-33 in promoting type 2 immunity and inflammation following helminth challenge or exposure to allergens. Specifically, recent reports have begun to define the complex cellular networks these alarmins activate and their contribution to type 2 immunity and inflammation. SUMMARY: Our increased understanding of the pathways that regulate type 2 cytokine-mediated immunity and inflammation have revealed novel therapeutic targets to treat both helminth infections and allergic disease states.

17.
J Exp Med ; 213(9): 1663-73, 2016 08 22.
Article in English | MEDLINE | ID: mdl-27526715

ABSTRACT

Type 2 cytokine responses are necessary for the development of protective immunity to helminth parasites but also cause the inflammation associated with allergies and asthma. Recent studies have found that peripheral hematopoietic progenitor cells contribute to type 2 cytokine-mediated inflammation through their enhanced ability to develop into mast cells. In this study, we show that carbonic anhydrase (Car) enzymes are up-regulated in type 2-associated progenitor cells and demonstrate that Car enzyme inhibition is sufficient to prevent mouse mast cell responses and inflammation after Trichinella spiralis infection or the induction of food allergy-like disease. Further, we used CRISPR/Cas9 technology and illustrate that genetically editing Car1 is sufficient to selectively reduce mast cell development. Finally, we demonstrate that Car enzymes can be targeted to prevent human mast cell development. Collectively, these experiments identify a previously unrecognized role for Car enzymes in regulating mast cell lineage commitment and suggest that Car enzyme inhibitors may possess therapeutic potential that can be used to treat mast cell-mediated inflammation.


Subject(s)
Carbonic Anhydrases/physiology , Inflammation/etiology , Mast Cells/physiology , Animals , Carbonic Anhydrase Inhibitors/pharmacology , Immunoglobulin E/blood , Mastocytosis/prevention & control , Methazolamide/pharmacology , Mice , Mice, Inbred C57BL
18.
Front Physiol ; 7: 214, 2016.
Article in English | MEDLINE | ID: mdl-27378934

ABSTRACT

Asthma is a heterogeneous disorder that results in recurrent attacks of breathlessness, coughing, and wheezing that affects millions of people worldwide. Although the precise causes of asthma are unclear, studies suggest that a combination of genetic predisposition and environmental exposure to various allergens and pathogens contribute to its development. Currently, the most common treatment to control asthma is a dual combination of ß2-adrenergic receptor agonists and corticosteroids. However, studies have shown that some patients do not respond well to these medications, while others experience significant side effects. It is reported that the majority of asthmas are associated with T helper type 2 (TH2) responses. In these patients, allergen challenge initiates the influx of TH2 cells in the airways leading to an increased production of TH2-associated cytokines and the promotion of allergy-induced asthma. Therefore, biologics that target this pathway may provide an alternative method to treat the allergic airway inflammation associated with asthma. As of now, only two biologics (omalizumab and mepolizumab), which target immunoglobulin E and interleukin-5, respectively, are FDA-approved and being prescribed to asthmatics. However, recent studies have reported that targeting other components of the TH2 response also show great promise. In this review, we will briefly describe the immunologic mechanisms underlying allergic asthma. Furthermore, we will discuss the current therapeutic strategies used to treat asthma including their limitations. Finally, we will highlight the benefits of using biologics to treat asthma-associated allergic airway inflammation with an emphasis on the potential of targeting cytokine alarmins, especially thymic stromal lymphopoietin.

19.
Nat Immunol ; 17(4): 356-63, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27002843

ABSTRACT

Innate cells are responsible for the rapid recognition of infection and mediate essential mechanisms of pathogen elimination, and also facilitate adaptive immune responses. We review here the numerous intricate interactions among innate cells that initiate protective immunity. The efficient eradication of pathogens depends on the coordinated actions of multiple cells, including innate cells and epithelial cells. Rather than acting as isolated effector cells, innate cells are in constant communication with other responding cells of the immune system, locally and distally. These interactions are critically important for the efficient control of primary infections as well for the development of 'trained' innate cells that facilitate the rapid elimination of homologous or heterologous infections.


Subject(s)
Adaptive Immunity/immunology , Cytokines/immunology , Immunity, Innate/immunology , Infections/immunology , Killer Cells, Natural/immunology , Myeloid Cells/immunology , Animals , Basophils/immunology , Eosinophils/immunology , Humans , Macrophages/immunology , Mast Cells/immunology , Monocytes/immunology , Neutrophils/immunology
20.
J Invest Dermatol ; 136(2): 487-496, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26802241

ABSTRACT

Breaches in the skin barrier initiate an inflammatory immune response that is critical for successful wound healing. Innate lymphoid cells (ILCs) are a recently identified population of immune cells that reside at epithelial barrier surfaces such as the skin, lung, and gut, and promote proinflammatory or epithelial repair functions after exposure to allergens, pathogens, or chemical irritants. However, the potential role of ILCs in regulating cutaneous wound healing remains undefined. Here, we demonstrate that cutaneous injury promotes an IL-33-dependent group 2 ILC (ILC2) response and that abrogation of this response impairs re-epithelialization and efficient wound closure. In addition, we provide evidence suggesting that an analogous ILC2 response is operational in acute wounds of human skin. Together, these results indicate that IL-33-responsive ILC2s are an important link between the cutaneous epithelium and the immune system, acting to promote the restoration of skin integrity after injury.


Subject(s)
Immunity, Innate/physiology , Interleukin-33/immunology , Skin/injuries , Wound Healing/immunology , Wounds and Injuries/immunology , Animals , Biopsy, Needle , Disease Models, Animal , Humans , Immunity, Innate/immunology , Immunohistochemistry , Interleukin-33/metabolism , Lymphocytes/cytology , Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Skin/pathology , Wound Healing/physiology , Wounds and Injuries/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...