Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Nature ; 544(7648): 105-109, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28329764

ABSTRACT

Platelets are critical for haemostasis, thrombosis, and inflammatory responses, but the events that lead to mature platelet production remain incompletely understood. The bone marrow has been proposed to be a major site of platelet production, although there is indirect evidence that the lungs might also contribute to platelet biogenesis. Here, by directly imaging the lung microcirculation in mice, we show that a large number of megakaryocytes circulate through the lungs, where they dynamically release platelets. Megakaryocytes that release platelets in the lungs originate from extrapulmonary sites such as the bone marrow; we observed large megakaryocytes migrating out of the bone marrow space. The contribution of the lungs to platelet biogenesis is substantial, accounting for approximately 50% of total platelet production or 10 million platelets per hour. Furthermore, we identified populations of mature and immature megakaryocytes along with haematopoietic progenitors in the extravascular spaces of the lungs. Under conditions of thrombocytopenia and relative stem cell deficiency in the bone marrow, these progenitors can migrate out of the lungs, repopulate the bone marrow, completely reconstitute blood platelet counts, and contribute to multiple haematopoietic lineages. These results identify the lungs as a primary site of terminal platelet production and an organ with considerable haematopoietic potential.


Subject(s)
Blood Platelets/cytology , Hematopoiesis , Hematopoietic Stem Cells/cytology , Lung/blood supply , Lung/cytology , Animals , Bone Marrow , Cell Lineage , Female , Lung/anatomy & histology , Male , Megakaryocytes/cytology , Mice , Microcirculation , Platelet Count , Thrombocytopenia/pathology
2.
Nature ; 531(7595): 513-7, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-26982733

ABSTRACT

Lung metastasis is the lethal determinant in many cancers and a number of lines of evidence point to monocytes and macrophages having key roles in its development. Yet little is known about the immediate fate of incoming tumour cells as they colonize this tissue, and even less known about how they make first contact with the immune system. Primary tumours liberate circulating tumour cells (CTCs) into the blood and we have developed a stable intravital two-photon lung imaging model in mice for direct observation of the arrival of CTCs and subsequent host interaction. Here we show dynamic generation of tumour microparticles in shear flow in the capillaries within minutes of CTC entry. Rather than dispersing under flow, many of these microparticles remain attached to the lung vasculature or independently migrate along the inner walls of vessels. Using fluorescent lineage reporters and flow cytometry, we observed 'waves' of distinct myeloid cell subsets that load differentially and sequentially with this CTC-derived material. Many of these tumour-ingesting myeloid cells collectively accumulated in the lung interstitium along with the successful metastatic cells and, as previously understood, promote the development of successful metastases from surviving tumour cells. Although the numbers of these cells rise globally in the lung with metastatic exposure and ingesting myeloid cells undergo phenotypic changes associated with microparticle ingestion, a consistently sparse population of resident conventional dendritic cells, among the last cells to interact with CTCs, confer anti-metastatic protection. This work reveals that CTC fragmentation generates immune-interacting intermediates, and defines a competitive relationship between phagocyte populations for tumour loading during metastatic cell seeding.


Subject(s)
Cell Movement , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lung/immunology , Lung/pathology , Neoplasm Metastasis/immunology , Neoplasm Metastasis/pathology , Animals , Capillaries/pathology , Cell Line, Tumor , Cell Lineage , Dendritic Cells/cytology , Dendritic Cells/immunology , Female , Genes, Reporter/genetics , Humans , Lung/blood supply , Lung/cytology , Lung Neoplasms/pathology , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Microscopy, Confocal , Myeloid Cells/cytology , Neoplastic Cells, Circulating/pathology
3.
Cell Immunol ; 350: 103862, 2020 04.
Article in English | MEDLINE | ID: mdl-30336937

ABSTRACT

The lung represents a unique immune environment. The primary function of the lung is to enable gas exchange by facilitating the transfer of oxygen into and carbon dioxide out of the blood. However, as a direct byproduct of this process the lung is also constantly exposed to particles, allergens, and pathogens alongside air itself. Due to this, the pulmonary immune system exists in a fine balance between quiescence and inflammation, deviations from which can lead to a failure in respiratory function. A rich history exists attempting to define the critical features of lung immunity, and most recently advances in intravital microscopy have enabled the visualization of intercellular immune dynamics in both steady-state and a variety of disease conditions. In this review, we will summarize a variety of approaches to intravital lung imaging as well as how its application has advanced our understanding of normal lung function as well as disease states such as pulmonary metastasis, asthma, and lung injury.


Subject(s)
Intravital Microscopy/methods , Lung/diagnostic imaging , Lung/immunology , Allergens/immunology , Asthma/immunology , Humans , Immune System/immunology , Inflammation/pathology , Microscopy, Fluorescence, Multiphoton/methods
4.
Trends Immunol ; 35(8): 379-86, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24974157

ABSTRACT

The lung is a complex structure that is interdigitated with immune cells. Understanding the 4D process of normal and defective lung function and immunity has been a centuries-old problem. Challenges intrinsic to the lung have limited adequate microscopic evaluation of its cellular dynamics in real time, until recently. Because of emerging technologies, we now recognize alveolar-to-airway transport of inhaled antigen. We understand the nature of neutrophil entry during lung injury and are learning more about cellular interactions during inflammatory states. Insights are also accumulating in lung development and the metastatic niche of the lung. Here we assess the developing technology of lung imaging, its merits for studies of pathophysiology and areas where further advances are needed.


Subject(s)
Lung Neoplasms/immunology , Lung/immunology , Animals , Cell Communication , Humans , Imaging, Three-Dimensional , Inflammation/immunology , Lung/pathology , Organogenesis
5.
Nature ; 477(7363): 229-33, 2011 Aug 14.
Article in English | MEDLINE | ID: mdl-21841801

ABSTRACT

CD4(+) T-helper type 2 (T(H)2) cells, characterized by their expression of interleukin (IL)-4, IL-5, IL-9 and IL-13, are required for immunity to helminth parasites and promote the pathological inflammation associated with asthma and allergic diseases. Polymorphisms in the gene encoding the cytokine thymic stromal lymphopoietin (TSLP) are associated with the development of multiple allergic disorders in humans, indicating that TSLP is a critical regulator of T(H)2 cytokine-associated inflammatory diseases. In support of genetic analyses, exaggerated TSLP production is associated with asthma, atopic dermatitis and food allergies in patients, and studies in murine systems demonstrated that TSLP promotes T(H)2 cytokine-mediated immunity and inflammation. However, the mechanisms through which TSLP induces T(H)2 cytokine responses remain poorly defined. Here we demonstrate that TSLP promotes systemic basophilia, that disruption of TSLP-TSLPR interactions results in defective basophil responses, and that TSLPR-sufficient basophils can restore T(H)2-cell-dependent immunity in vivo. TSLP acted directly on bone-marrow-resident progenitors to promote basophil responses selectively. Critically, TSLP could elicit basophil responses in both IL-3-IL-3R-sufficient and -deficient environments, and genome-wide transcriptional profiling and functional analyses identified heterogeneity between TSLP-elicited versus IL-3-elicited basophils. Furthermore, activated human basophils expressed TSLPR, and basophils isolated from eosinophilic oesophagitis patients were distinct from classical basophils. Collectively, these studies identify previously unrecognized heterogeneity within the basophil cell lineage and indicate that expression of TSLP may influence susceptibility to multiple allergic diseases by regulating basophil haematopoiesis and eliciting a population of functionally distinct basophils that promote T(H)2 cytokine-mediated inflammation.


Subject(s)
Basophils/cytology , Cytokines/metabolism , Hematopoiesis , Hypersensitivity, Immediate/immunology , Inflammation/immunology , Inflammation/metabolism , Interleukin-3 , Animals , Asthma/immunology , Basophils/metabolism , Cytokines/genetics , Cytokines/immunology , Dermatitis, Atopic/immunology , Food Hypersensitivity/immunology , Humans , Interleukin-3/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Phenotype , Receptors, Cytokine/metabolism , Receptors, Interleukin-3/deficiency , Receptors, Interleukin-3/genetics , Receptors, Interleukin-3/metabolism , Th2 Cells/immunology , Thymic Stromal Lymphopoietin
6.
J Immunol ; 190(3): 904-12, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23275605

ABSTRACT

The epithelial-derived cytokine thymic stromal lymphopoietin (TSLP) has been associated with the promotion of type 2 inflammation and the induction of allergic disease. In humans TSLP is elevated in the lungs of asthma patients and in the lesional skin of individuals with atopic dermatitis, whereas mice lacking TSLP responses are refractory to models of Th2-driven allergic disease. Although several cell types, including dendritic cells, basophils, and CD4 T cells, have been shown to respond to TSLP, its role in macrophage differentiation has not been studied. Type 2 cytokines (i.e., IL-4 and IL-13) can drive the differentiation of macrophages into alternatively activated macrophages (aaMs, also referred to as M2 macrophages). This population of macrophages is associated with allergic inflammation. We therefore reasoned that TSLP/TSLPR signaling may be involved in the differentiation and activation of aaMs during allergic airway inflammation. In this study, we report that TSLP changes the quiescent phenotype of pulmonary macrophages toward an aaM phenotype during TSLP-induced airway inflammation. This differentiation of airway macrophages was IL-13-, but not IL-4-, dependent. Taken together, we demonstrate in this study that TSLP/TSLPR plays a significant role in the amplification of aaMΦ polarization and chemokine production, thereby contributing to allergic inflammation.


Subject(s)
Asthma/immunology , Cytokines/pharmacology , Macrophage Activation , Macrophages/drug effects , Animals , Asthma/chemically induced , Asthma/pathology , Bronchoalveolar Lavage Fluid/cytology , Cell Differentiation , Cells, Cultured/drug effects , Chemokines/analysis , Cytokines/physiology , Disease Models, Animal , Drug Synergism , Female , Immunoglobulins/deficiency , Immunoglobulins/genetics , Immunoglobulins/physiology , Interleukin-13/deficiency , Interleukin-13/physiology , Interleukin-4/deficiency , Interleukin-4/physiology , Lung/pathology , Macrophages/physiology , Mice , Mice, Inbred BALB C , Mice, Knockout , Ovalbumin/toxicity , Receptors, Cytokine/deficiency , Receptors, Cytokine/genetics , Receptors, Cytokine/physiology , STAT6 Transcription Factor/deficiency , STAT6 Transcription Factor/physiology , Signal Transduction , Specific Pathogen-Free Organisms , Thymic Stromal Lymphopoietin
7.
J Allergy Clin Immunol ; 130(5): 1187-1196.e5, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22981788

ABSTRACT

BACKGROUND: Respiratory viral infection, including respiratory syncytial virus (RSV) and rhinovirus, has been linked to respiratory disease in pediatric patients, including severe acute bronchiolitis and asthma exacerbation. OBJECTIVE: The study examined the role of the epithelial-derived cytokine thymic stromal lymphopoietin (TSLP) in the response to RSV infection. METHODS: Infection of human airway epithelial cells was used to examine TSLP induction after RSV infection. Air-liquid interface cultures from healthy children and children with asthma were also tested for TSLP production after infection. Finally, a mouse model was used to directly test the role of TSLP signaling in the response to RSV infection. RESULTS: Infection of airway epithelial cells with RSV led to the production of TSLP via activation of an innate signaling pathway that involved retinoic acid induced gene I, interferon promoter-stimulating factor 1, and nuclear factor-κB. Consistent with this observation, airway epithelial cells from asthmatic children a produced significantly greater levels of TSLP after RSV infection than cells from healthy children. In mouse models, RSV-induced TSLP expression was found to be critical for the development of immunopathology. CONCLUSION: These findings suggest that RSV can use an innate antiviral signaling pathway to drive a potentially nonproductive immune response and has important implications for the role of TSLP in viral immune responses in general.


Subject(s)
Asthma/immunology , Cytokines/metabolism , Respiratory Mucosa/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Asthma/etiology , Cell Line , Child , Cytokines/genetics , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Receptors, Cell Surface , Respiratory Mucosa/virology , Respiratory Syncytial Virus Infections/complications , Signal Transduction/genetics , Signal Transduction/immunology , Th1-Th2 Balance , Th2 Cells/immunology , Thymic Stromal Lymphopoietin
8.
Cell Rep ; 42(6): 112582, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37261951

ABSTRACT

Pre-metastatic niche formation is a critical step during the metastatic spread of cancer. One way by which primary tumors prime host cells at future metastatic sites is through the shedding of tumor-derived microparticles as a consequence of vascular sheer flow. However, it remains unclear how the uptake of such particles by resident immune cells affects their phenotype and function. Here, we show that ingestion of tumor-derived microparticles by macrophages induces a rapid metabolic and phenotypic switch that is characterized by enhanced mitochondrial mass and function, increased oxidative phosphorylation, and upregulation of adhesion molecules, resulting in reduced motility in the early metastatic lung. This reprogramming event is dependent on signaling through the mTORC1, but not the mTORC2, pathway and is induced by uptake of tumor-derived microparticles. Together, these data support a mechanism by which uptake of tumor-derived microparticles induces reprogramming of macrophages to shape their fate and function in the early metastatic lung.


Subject(s)
Lung Neoplasms , Neoplasms , Humans , Macrophages/pathology , Lung/pathology , Neoplasms/pathology , Signal Transduction , Biological Transport , Lung Neoplasms/pathology
9.
J Immunol ; 182(3): 1641-7, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19155513

ABSTRACT

Thymic stromal lymphopoietin (TSLP) is crucial for the development of atopic diseases in humans and mice. Mice that express a lung-specific TSLP transgene (surfactant protein C promoter (SPC)-TSLP) develop a spontaneous and progressive asthma-like disease, suggesting that TSLP expression alone was sufficient for disease development. In this study, we show that, in fact, TSLP alone only causes a weak innate response that is insufficient for development of full airway inflammatory disease. Complete disease development requires both TSLP and antigenic stimulation. These data suggest that the spontaneous lung inflammation observed in SPC-TSLP mice reflects a TSLP-driven predisposition toward the development of aberrant responses against innocuous environmental Ags. This provides evidence that TSLP may act directly to induce susceptibility to the inappropriate allergic responses that characterize atopy and asthma. We additionally show that disease development requires CD4 T cells but not B cells. Further, we reveal a TSLP-driven innate response involving mucus overproduction and goblet cell metaplasia. Taken together, these data suggest a multifaceted model of TSLP-mediated airway inflammation, with an initial activation of resident innate immune cells, followed by activation of the adaptive immune system and full disease development. This study provides new insight into the unique features of the asthma pathology contributed by the innate and adaptive immune responses in response to TSLP stimulation.


Subject(s)
Cytokines/administration & dosage , Epitopes/immunology , Immunity, Innate , Lung/immunology , Lung/pathology , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/pathology , Acute Disease , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Cytokines/biosynthesis , Cytokines/deficiency , Cytokines/genetics , Genetic Predisposition to Disease , Hypersensitivity, Immediate/genetics , Hypersensitivity, Immediate/immunology , Hypersensitivity, Immediate/pathology , Immunity, Innate/genetics , Immunophenotyping , Lung/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Knockout , Mice, Transgenic , Promoter Regions, Genetic , Pulmonary Surfactant-Associated Protein C/genetics , Respiratory Hypersensitivity/genetics , Thymic Stromal Lymphopoietin
10.
Sci Adv ; 7(39): eabg0505, 2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34550730

ABSTRACT

Modern immunologic research increasingly requires high-dimensional analyses to understand the complex milieu of cell types that comprise the tissue microenvironments of disease. To achieve this, we developed Infinity Flow combining hundreds of overlapping flow cytometry panels using machine learning to enable the simultaneous analysis of the coexpression patterns of hundreds of surface-expressed proteins across millions of individual cells. In this study, we demonstrate that this approach allows the comprehensive analysis of the cellular constituency of the steady-state murine lung and the identification of previously unknown cellular heterogeneity in the lungs of melanoma metastasis­bearing mice. We show that by using supervised machine learning, Infinity Flow enhances the accuracy and depth of clustering or dimensionality reduction algorithms. Infinity Flow is a highly scalable, low-cost, and accessible solution to single-cell proteomics in complex tissues.

11.
Cell Rep ; 36(1): 109309, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34233193

ABSTRACT

αvß8 integrin, a key activator of transforming growth factor ß (TGF-ß), inhibits anti-tumor immunity. We show that a potent blocking monoclonal antibody against αvß8 (ADWA-11) causes growth suppression or complete regression in syngeneic models of squamous cell carcinoma, mammary cancer, colon cancer, and prostate cancer, especially when combined with other immunomodulators or radiotherapy. αvß8 is expressed at the highest levels in CD4+CD25+ T cells in tumors, and specific deletion of ß8 from T cells is as effective as ADWA-11 in suppressing tumor growth. ADWA-11 increases expression of a suite of genes in tumor-infiltrating CD8+ T cells normally inhibited by TGF-ß and involved in tumor cell killing, including granzyme B and interferon-γ. The in vitro cytotoxic effect of tumor CD8 T cells is inhibited by CD4+CD25+ cells, and this suppressive effect is blocked by ADWA-11. These findings solidify αvß8 integrin as a promising target for cancer immunotherapy.


Subject(s)
Immunity , Immunotherapy , Integrins/metabolism , Models, Biological , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes/immunology , Animals , Antibodies, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/immunology , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Granzymes/metabolism , Interferon-gamma/metabolism , Lymphocyte Depletion , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Neoplasms/genetics , Neoplasms/pathology , Signal Transduction , Smad3 Protein/metabolism , Survival Analysis , T-Lymphocytes, Cytotoxic/immunology , Transforming Growth Factor beta/metabolism , Tumor Microenvironment/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
12.
J Immunol ; 181(8): 5189-93, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18832669

ABSTRACT

The epithelial-derived cytokine thymic stromal lymphopoietin (TSLP) has important roles in the initiation of allergic airway inflammation and the activation of dendritic cells. We have shown that the human TSLP gene is regulated in a NF-kappaB-dependent manner; however the factors that negatively regulate TSLP expression are not known. In this study we demonstrate that 9-cis-retinoic acid (9-cis-RA) is a negative regulator of TSLP expression in airway epithelial cells. This inhibition is manifested as a block in the IL-1beta-mediated recruitment of NF-kappaB to the human TSLP promoter. 9-cis-RA-mediated inhibition is not restricted to TSLP gene expression but rather reflects a general inhibition of NF-kappaB activation, as other NF-kappaB-regulated-genes were also inhibited in a similar manner by 9-cis-RA treatment. Taken as a whole, these data demonstrate that inhibition of IL-1beta-dependent genes by active retinoid X receptors involves antagonism of NF-kappaB signaling.


Subject(s)
Cytokines/immunology , Gene Expression Regulation/immunology , NF-kappa B/immunology , Respiratory Hypersensitivity/immunology , Respiratory Mucosa/immunology , Retinoid X Receptors/immunology , Signal Transduction/immunology , Alitretinoin , Antineoplastic Agents/pharmacology , Cell Line , Cytokines/biosynthesis , Epithelial Cells/immunology , Epithelial Cells/metabolism , Gene Expression Regulation/drug effects , Humans , Inflammation/immunology , Inflammation/metabolism , Interleukin-1beta/pharmacology , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Promoter Regions, Genetic/immunology , Respiratory Hypersensitivity/metabolism , Respiratory Mucosa/metabolism , Retinoid X Receptors/agonists , Retinoid X Receptors/antagonists & inhibitors , Retinoid X Receptors/metabolism , Signal Transduction/drug effects , Tretinoin/pharmacology , Thymic Stromal Lymphopoietin
13.
J Immunol ; 181(9): 6557-62, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18941246

ABSTRACT

Lung-specific thymic stromal lymphopoietin (TSLP) expression is sufficient for the development of an asthma-like chronic airway inflammatory disease. However, the nature of the downstream pathways that regulate disease development are not known. In this study, we used IL-4- and Stat6-deficient mice to establish the role of Th2-type responses downstream of TSLP. IL-4 deficiency greatly reduced, but did not eliminate, TSLP-induced airway hyperresponsiveness, airway inflammation, eosinophilia, and goblet cell metaplasia, while Stat6 deficiency eliminated these asthma-like symptoms. We further demonstrate, using the chronic model of TSLP-mediated airway inflammation, that blockade of both IL-4 and IL-13 responses, through administration of an anti-IL-4R alpha mAb, reversed asthma-like symptoms, when given to mice with established disease. Collectively these data provide insight into the pathways engaged in TSLP-driven airway inflammation and demonstrate that simultaneous blockade of IL-4 and IL-13 can reverse established airway disease, suggesting that this may be an effective approach for the therapy of Th2-mediated inflammatory respiratory disease.


Subject(s)
Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/prevention & control , Cytokines/toxicity , Th2 Cells/immunology , Thymus Gland/immunology , Thymus Gland/pathology , Animals , Bronchial Hyperreactivity/pathology , Chronic Disease , Cytokines/antagonists & inhibitors , Cytokines/physiology , Eosinophilia/genetics , Eosinophilia/immunology , Eosinophilia/prevention & control , Humans , Interleukin-13/antagonists & inhibitors , Interleukin-13/metabolism , Interleukin-4/deficiency , Interleukin-4/genetics , Interleukin-4/physiology , Interleukin-6/deficiency , Interleukin-6/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Signal Transduction/genetics , Signal Transduction/physiology , Stromal Cells/immunology , Stromal Cells/metabolism , Stromal Cells/pathology , Th2 Cells/metabolism , Thymus Gland/metabolism , Thymic Stromal Lymphopoietin
14.
J Exp Med ; 217(9)2020 09 07.
Article in English | MEDLINE | ID: mdl-32735669

ABSTRACT

The response to the COVID-19 crisis across most research institutions mandated ceasing nonessential research activities in order to minimize the spread of the virus in our communities. With minimal notice, experiments were terminated, cell lines were frozen, mouse colonies were culled, and trainees were prevented from performing bench research. Still, despite the interruption of experimental productivity, the shutdown has proven for many PIs and trainees that doing and thinking science are not activities that are bound to the laboratory. Furthermore, the shutdowns have solidified important emerging trends and forced us to further innovate to get the most out of working remotely. We hope that some of these innovations, hard-gained in this difficult time, will persist and develop into new paradigms-lessons that will improve our science and our relationship to the climate and community beyond the current pandemic.


Subject(s)
Biomedical Research/trends , Coronavirus Infections/epidemiology , Pneumonia, Viral/epidemiology , Animals , Betacoronavirus , COVID-19 , Communicable Disease Control/methods , Congresses as Topic , Humans , Interinstitutional Relations , Pandemics , Research Personnel , SARS-CoV-2
15.
Sci Immunol ; 4(36)2019 06 07.
Article in English | MEDLINE | ID: mdl-31175176

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) are enriched in mucosal tissues (e.g., lung) and respond to epithelial cell-derived cytokines initiating type 2 inflammation. During inflammation, ILC2 numbers are increased in the lung. However, the mechanisms controlling ILC2 trafficking and motility within inflamed lungs remain unclear and are crucial for understanding ILC2 function in pulmonary immunity. Using several approaches, including lung intravital microscopy, we demonstrate that pulmonary ILC2s are highly dynamic, exhibit amoeboid-like movement, and aggregate in the lung peribronchial and perivascular spaces. They express distinct chemokine receptors, including CCR8, and actively home to CCL8 deposits located around the airway epithelium. Within lung tissue, ILC2s were particularly motile in extracellular matrix-enriched regions. We show that collagen-I drives ILC2 to markedly change their morphology by remodeling their actin cytoskeleton to promote environmental exploration critical for regulating eosinophilic inflammation. Our study provides previously unappreciated insights into ILC2 migratory patterns during inflammation and highlights the importance of environmental guidance cues in the lung in controlling ILC2 dynamics.


Subject(s)
Lung/immunology , Lymphocytes/immunology , Animals , Cell Movement/drug effects , Collagen/immunology , Eosinophils/immunology , Extracellular Matrix/immunology , Female , Fibronectins/immunology , Humans , Immunity, Innate , Inflammation/immunology , Interleukin-33/pharmacology , Lymphocytes/drug effects , Mice, Inbred BALB C , Mice, Transgenic , Recombinant Proteins/pharmacology
16.
Elife ; 72018 06 04.
Article in English | MEDLINE | ID: mdl-29862966

ABSTRACT

Chimeric antigen receptors (CARs) are synthetic receptors that reprogram T cells to kill cancer. The success of CAR-T cell therapies highlights the promise of programmed immunity and suggests that applying CAR strategies to other immune cell lineages may be beneficial. Here, we engineered a family of Chimeric Antigen Receptors for Phagocytosis (CAR-Ps) that direct macrophages to engulf specific targets, including cancer cells. CAR-Ps consist of an extracellular antibody fragment, which can be modified to direct CAR-P activity towards specific antigens. By screening a panel of engulfment receptor intracellular domains, we found that the cytosolic domains from Megf10 and FcRÉ£ robustly triggered engulfment independently of their native extracellular domain. We show that CAR-Ps drive specific engulfment of antigen-coated synthetic particles and whole human cancer cells. Addition of a tandem PI3K recruitment domain increased cancer cell engulfment. Finally, we show that CAR-P expressing murine macrophages reduce cancer cell number in co-culture by over 40%.


Subject(s)
Phagocytosis , Receptors, Chimeric Antigen/metabolism , Animals , Antigens, CD19/metabolism , Antigens, Neoplasm/metabolism , Cell Line, Tumor , Humans , Immunological Synapses , Macrophages/metabolism , Mice , Microspheres , NIH 3T3 Cells , Phosphorylation , Signal Transduction , Silicon Dioxide
17.
Int Immunopharmacol ; 7(6): 734-43, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17466907

ABSTRACT

We report that the novel anthracycline analog, 13-deoxy, 5-iminodoxorubicin (DIDOX), represents a potentially new class of immunosuppressive agents. DIDOX has been structurally modified from the parent compound, doxorubicin, to remove the carbonyl group at carbon-13 and the quinone moiety at carbon-5 since these structures likely mediate the cardiotoxic side effects of this family of chemotherapeutic drugs. Our studies demonstrate that DIDOX inhibits T cell proliferation and the expression of the T cell activation molecules, CD25 and CD40L. DIDOX also inhibits the production of the pro-inflammatory cytokine, TNF-alpha and IL-2. Studies using animal models demonstrate that DIDOX inhibits the inflammation accompanying contact hypersensitivity reactions and possesses reduced cardiotoxicity compared to doxorubicin. These findings indicate that DIDOX has important immunosuppressive activities that may warrant the development of this new and improved anthracycline for the treatment of T cell-mediated inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , CD4-Positive T-Lymphocytes/drug effects , Doxorubicin/analogs & derivatives , Immunosuppressive Agents/pharmacology , Animals , Anti-Inflammatory Agents/blood , Anti-Inflammatory Agents/pharmacokinetics , CD4-Positive T-Lymphocytes/immunology , CD40 Ligand/immunology , Cell Proliferation/drug effects , Cells, Cultured , Dermatitis, Contact/drug therapy , Dermatitis, Contact/pathology , Dinitrofluorobenzene , Doxorubicin/blood , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Echocardiography , Heart Atria/drug effects , Humans , Immunosuppressive Agents/blood , Immunosuppressive Agents/pharmacokinetics , Interleukin-2/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Leukocyte Count , Mice , Mice, Inbred BALB C , Rabbits , Tumor Necrosis Factor-alpha/immunology
18.
J Immunother Cancer ; 5(1): 77, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28923102

ABSTRACT

Understanding how murine models can elucidate the mechanisms underlying antitumor immune responses and advance immune-based drug development is essential to advancing the field of cancer immunotherapy. The Society for Immunotherapy of Cancer (SITC) convened a workshop titled, "Challenges, Insights, and Future Directions for Mouse and Humanized Models in Cancer Immunology and Immunotherapy" as part of the SITC 31st Annual Meeting and Associated Programs on November 10, 2016 in National Harbor, MD. The workshop focused on key issues in optimizing models for cancer immunotherapy research, with discussions on the strengths and weaknesses of current models, approaches to improve the predictive value of mouse models, and advances in cancer modeling that are anticipated in the near future. This full-day program provided an introduction to the most common immunocompetent and humanized models used in cancer immunology and immunotherapy research, and addressed the use of models to evaluate immune-targeting therapies. Here, we summarize the workshop presentations and subsequent panel discussion.


Subject(s)
Disease Models, Animal , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , Animals , Humans , Mice , Molecular Targeted Therapy , Societies, Scientific , Tumor Microenvironment
19.
Cancer Cell ; 30(2): 324-336, 2016 08 08.
Article in English | MEDLINE | ID: mdl-27424807

ABSTRACT

Intratumoral dendritic cells (DC) bearing CD103 in mice or CD141 in humans drive intratumoral CD8(+) T cell activation. Using multiple strategies, we identified a critical role for these DC in trafficking tumor antigen to lymph nodes (LN), resulting in both direct CD8(+) T cell stimulation and antigen hand-off to resident myeloid cells. These effects all required CCR7. Live imaging demonstrated direct presentation to T cells in LN, and CCR7 loss specifically in these cells resulted in defective LN T cell priming and increased tumor outgrowth. CCR7 expression levels in human tumors correlate with signatures of CD141(+) DC, intratumoral T cells, and better clinical outcomes. This work identifies an ongoing pathway to T cell priming, which should be harnessed for tumor therapies.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Melanoma/immunology , Receptors, CCR7/immunology , Animals , Antigen Presentation , Antigens, CD/immunology , Antigens, Neoplasm/immunology , Antigens, Surface/immunology , Cell Movement/immunology , Dendritic Cells/pathology , Humans , Integrin alpha Chains/immunology , Lymph Nodes/immunology , Lymph Nodes/pathology , Melanoma/pathology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Thrombomodulin
20.
Expert Rev Clin Immunol ; 5(5): 547-556, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-20436950

ABSTRACT

Thymic stromal lymphopoietin (TSLP) is an IL-7-related cytokine expressed predominantly by barrier epithelial cells. TSLP is a potent activator of several cell types, including myeloid-derived dendritic cells, monocytes/macrophages and mast cells. Recent studies have revealed an important role for TSLP in the initiation and progression of allergic inflammatory diseases. In this review, we will discuss the role of TSLP in atopic diseases, as well as its function in immune homeostasis.

SELECTION OF CITATIONS
SEARCH DETAIL