Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.553
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 38: 147-170, 2020 04 26.
Article in English | MEDLINE | ID: mdl-32340573

ABSTRACT

Metabolism is one of the strongest drivers of interkingdom interactions-including those between microorganisms and their multicellular hosts. Traditionally thought to fuel energy requirements and provide building blocks for biosynthetic pathways, metabolism is now appreciated for its role in providing metabolites, small-molecule intermediates generated from metabolic processes, to perform various regulatory functions to mediate symbiotic relationships between microbes and their hosts. Here, we review recent advances in our mechanistic understanding of how microbiota-derived metabolites orchestrate and support physiological responses in the host, including immunity, inflammation, defense against infections, and metabolism. Understanding how microbes metabolically communicate with their hosts will provide us an opportunity to better describe how a host interacts with all microbes-beneficial, pathogenic, and commensal-and an opportunity to discover new ways to treat microbial-driven diseases.


Subject(s)
Disease Susceptibility , Energy Metabolism , Homeostasis , Microbiota , Symbiosis , Animals , Disease Susceptibility/immunology , Host-Pathogen Interactions , Humans , Immune System/immunology , Immune System/metabolism , Microbiota/immunology
2.
Annu Rev Immunol ; 38: 1-21, 2020 04 26.
Article in English | MEDLINE | ID: mdl-31594433

ABSTRACT

It is difficult to believe that in about 1960 practically nothing was known about the thymus and some of its products, T cells bearing αß receptors for antigen. Thus I was lucky to join the field of T cell biology almost at its beginning, when knowledge about the cells was just getting off the ground and there was so much to discover. This article describes findings about these cells made by others and myself that led us all from ignorance, via complete confusion, to our current state of knowledge. I believe I was fortunate to practice science in very supportive institutions and with very collaborative colleagues in two countries that both encourage independent research by independent scientists, while simultaneously ignoring or somehow being able to avoid some of the difficulties of being a woman in what was, at the time, a male-dominated profession.


Subject(s)
Disease Susceptibility , Obsessive-Compulsive Disorder/etiology , Obsessive-Compulsive Disorder/metabolism , Animals , Autoimmunity , Biomarkers , Cell Death , Cytokines/metabolism , Disease Susceptibility/immunology , Histocompatibility Antigens/genetics , Histocompatibility Antigens/immunology , Histocompatibility Antigens/metabolism , Humans , Immunity, Innate , Obsessive-Compulsive Disorder/psychology , Protein Binding , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Superantigens/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism
3.
Annu Rev Immunol ; 36: 755-781, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29677472

ABSTRACT

Inflammatory bowel disease (IBD) defines a spectrum of complex disorders. Understanding how environmental risk factors, alterations of the intestinal microbiota, and polygenetic and epigenetic susceptibility impact on immune pathways is key for developing targeted therapies. Mechanistic understanding of polygenic IBD is complemented by Mendelian disorders that present with IBD, pharmacological interventions that cause colitis, autoimmunity, and multiple animal models. Collectively, this multifactorial pathogenesis supports a concept of immune checkpoints that control microbial-host interactions in the gut by modulating innate and adaptive immunity, as well as epithelial and mesenchymal cell responses. In addition to classical immunosuppressive strategies, we discuss how resetting the microbiota and restoring innate immune responses, in particular autophagy and epithelial barrier function, might be key for maintaining remission or preventing IBD. Targeting checkpoints in genetically stratified subgroups of patients with Mendelian disorder-associated IBD increasingly directs treatment strategies as part of personalized medicine.


Subject(s)
Disease Susceptibility/immunology , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/therapy , Animals , Biomarkers , Chronic Disease , Disease Management , Disease Models, Animal , Drug-Related Side Effects and Adverse Reactions , Dysbiosis , Gastrointestinal Microbiome , Genetic Predisposition to Disease , Humans , Inflammatory Bowel Diseases/prevention & control , Molecular Targeted Therapy , Translational Research, Biomedical
4.
Annu Rev Immunol ; 36: 813-842, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29677477

ABSTRACT

Given the many cell types and molecular components of the human immune system, along with vast variations across individuals, how should we go about developing causal and predictive explanations of immunity? A central strategy in human studies is to leverage natural variation to find relationships among variables, including DNA variants, epigenetic states, immune phenotypes, clinical descriptors, and others. Here, we focus on how natural variation is used to find patterns, infer principles, and develop predictive models for two areas: (a) immune cell activation-how single-cell profiling boosts our ability to discover immune cell types and states-and (b) antigen presentation and recognition-how models can be generated to predict presentation of antigens on MHC molecules and their detection by T cell receptors. These are two examples of a shift in how we find the drivers and targets of immunity, especially in the human system in the context of health and disease.


Subject(s)
Immune System , Immunity , Animals , Antigen Presentation/immunology , Biomarkers , Disease Susceptibility/immunology , Disease Susceptibility/metabolism , Epitopes/immunology , Genomics/methods , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immune System/cytology , Immune System/physiology , Ligands , Major Histocompatibility Complex/genetics , Major Histocompatibility Complex/immunology , Peptides/immunology , Protein Transport , Proteolysis , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
6.
Annu Rev Immunol ; 32: 433-59, 2014.
Article in English | MEDLINE | ID: mdl-24499275

ABSTRACT

Although new activation and regulatory mechanisms are still being identified, the basic architecture of the complement system has been known for decades. Two major roles of complement are to control certain bacterial infections and to promote clearance of apoptotic cells. In addition, although inappropriate complement activation has long been proposed to cause tissue damage in human inflammatory and autoimmune diseases, whether this is indeed true has been uncertain. However, recent studies in humans, especially those using newly available biological therapeutics, have now clearly demonstrated the pathophysiologic importance of the complement system in several rare diseases. Beyond these conditions, recent genetic studies have strongly supported an injurious role for complement in a wide array of human inflammatory, degenerative, and autoimmune diseases. This review includes an overview of complement activation, regulatory, and effector mechanisms. It then focuses on new understandings gained from genetic studies, ex vivo analyses, therapeutic trials, and animal models as well as on new research opportunities.


Subject(s)
Complement System Proteins/immunology , Complement System Proteins/metabolism , Receptors, Complement/metabolism , Animals , Carrier Proteins/metabolism , Complement Activation/immunology , Complement System Proteins/genetics , Disease Models, Animal , Disease Susceptibility/immunology , Genetic Predisposition to Disease , Humans , Protein Binding , Signal Transduction
7.
Cell ; 181(6): 1276-1290.e13, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32402238

ABSTRACT

At the species level, immunity depends on the selection and transmission of protective components of the immune system. A microbe-induced population of RORγ-expressing regulatory T cells (Tregs) is essential in controlling gut inflammation. We uncovered a non-genetic, non-epigenetic, non-microbial mode of transmission of their homeostatic setpoint. RORγ+ Treg proportions varied between inbred mouse strains, a trait transmitted by the mother during a tight age window after birth but stable for life, resistant to many microbial or cellular perturbations, then further transferred by females for multiple generations. RORγ+ Treg proportions negatively correlated with IgA production and coating of gut commensals, traits also subject to maternal transmission, in an immunoglobulin- and RORγ+ Treg-dependent manner. We propose a model based on a double-negative feedback loop, vertically transmitted via the entero-mammary axis. This immunologic mode of multi-generational transmission may provide adaptability and modulate the genetic tuning of gut immune responses and inflammatory disease susceptibility.


Subject(s)
Digestive System/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Disease Susceptibility/immunology , Female , Gastrointestinal Microbiome/immunology , Homeostasis/immunology , Immunoglobulin A/immunology , Inflammation/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Inbred NOD , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology
8.
Nat Immunol ; 22(5): 654-665, 2021 05.
Article in English | MEDLINE | ID: mdl-33888898

ABSTRACT

Controlled human infections provide opportunities to study the interaction between the immune system and malaria parasites, which is essential for vaccine development. Here, we compared immune signatures of malaria-naive Europeans and of Africans with lifelong malaria exposure using mass cytometry, RNA sequencing and data integration, before and 5 and 11 days after venous inoculation with Plasmodium falciparum sporozoites. We observed differences in immune cell populations, antigen-specific responses and gene expression profiles between Europeans and Africans and among Africans with differing degrees of immunity. Before inoculation, an activated/differentiated state of both innate and adaptive cells, including elevated CD161+CD4+ T cells and interferon-γ production, predicted Africans capable of controlling parasitemia. After inoculation, the rapidity of the transcriptional response and clusters of CD4+ T cells, plasmacytoid dendritic cells and innate T cells were among the features distinguishing Africans capable of controlling parasitemia from susceptible individuals. These findings can guide the development of a vaccine effective in malaria-endemic regions.


Subject(s)
Adaptive Immunity/immunology , Disease Susceptibility/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Adaptive Immunity/genetics , Adolescent , Adult , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Black People/genetics , Dendritic Cells/immunology , Disease Susceptibility/blood , Disease Susceptibility/parasitology , Female , Healthy Volunteers , Host-Parasite Interactions/genetics , Host-Parasite Interactions/immunology , Humans , Immunity, Innate/genetics , Immunity, Innate/immunology , Interferon-gamma/metabolism , Malaria, Falciparum/blood , Malaria, Falciparum/parasitology , Male , RNA-Seq , Systems Analysis , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , White People/genetics , Young Adult
9.
Nat Immunol ; 22(6): 687-698, 2021 06.
Article in English | MEDLINE | ID: mdl-33986548

ABSTRACT

The aged adaptive immune system is characterized by progressive dysfunction as well as increased autoimmunity. This decline is responsible for elevated susceptibility to infection and cancer, as well as decreased vaccination efficacy. Recent evidence indicates that CD4+ T cell-intrinsic alteratins contribute to chronic inflammation and are sufficient to accelerate an organism-wide aging phenotype, supporting the idea that T cell aging plays a major role in body-wide deterioration. In this Review, we propose ten molecular hallmarks to represent common denominators of T cell aging. These hallmarks are grouped into four primary hallmarks (thymic involution, mitochondrial dysfunction, genetic and epigenetic alterations, and loss of proteostasis) and four secondary hallmarks (reduction of the TCR repertoire, naive-memory imbalance, T cell senescence, and lack of effector plasticity), and together they explain the manifestation of the two integrative hallmarks (immunodeficiency and inflammaging). A major challenge now is weighing the relative impact of these hallmarks on T cell aging and understanding their interconnections, with the final goal of defining molecular targets for interventions in the aging process.


Subject(s)
Aging/immunology , Immunity, Cellular , T-Lymphocytes/immunology , Aging/genetics , Autoimmunity/genetics , Cell Plasticity/genetics , Cell Plasticity/immunology , Cellular Senescence/genetics , Cellular Senescence/immunology , Disease Susceptibility/immunology , Epigenesis, Genetic/immunology , Gene Expression Regulation/immunology , Humans , Inflammation/genetics , Inflammation/immunology , Proteostasis/genetics , Proteostasis/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , Thymus Gland/immunology , Thymus Gland/physiopathology
10.
Nat Immunol ; 21(6): 605-614, 2020 06.
Article in English | MEDLINE | ID: mdl-32367037

ABSTRACT

Impressive progress has been made over the last several years toward understanding how almost every aspect of the immune system contributes to the expression of systemic autoimmunity. In parallel, studies have shed light on the mechanisms that contribute to organ inflammation and damage. New approaches that address the complicated interaction between genetic variants, epigenetic processes, sex and the environment promise to enlighten the multitude of pathways that lead to what is clinically defined as systemic lupus erythematosus. It is expected that each patient owns a unique 'interactome', which will dictate specific treatment.


Subject(s)
Autoimmunity , Disease Susceptibility/immunology , Lupus Erythematosus, Systemic/diagnosis , Lupus Erythematosus, Systemic/etiology , Animals , Diagnosis, Differential , Environmental Exposure , Genetic Predisposition to Disease , Genetic Variation , Humans , Immunity, Innate , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Organ Specificity , Sex Factors
11.
Nat Immunol ; 21(12): 1585-1596, 2020 12.
Article in English | MEDLINE | ID: mdl-33020659

ABSTRACT

Sepsis is a biphasic disease characterized by an acute inflammatory response, followed by a prolonged immunosuppressive phase. Therapies aimed at controlling inflammation help to reduce the time patients with sepsis spend in intensive care units, but they do not lead to a reduction in overall mortality. Recently, the focus has been on addressing the immunosuppressive phase, often caused by apoptosis of immune cells. However, molecular triggers of these events are not yet known. Using whole-genome CRISPR screening in mice, we identified a triggering receptor expressed on myeloid cells (TREM) family receptor, TREML4, as a key regulator of inflammation and immune cell death in sepsis. Genetic ablation of Treml4 in mice demonstrated that TREML4 regulates calcium homeostasis, the inflammatory cytokine response, myeloperoxidase activation, the endoplasmic reticulum stress response and apoptotic cell death in innate immune cells, leading to an overall increase in survival rate, both during the acute and chronic phases of polymicrobial sepsis.


Subject(s)
Disease Susceptibility , Immunity, Innate , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Sepsis/etiology , Animals , Biomarkers , Cell Death , Clustered Regularly Interspaced Short Palindromic Repeats , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility/immunology , Gene Editing , Gene Knockdown Techniques , Gene Targeting , Genomics/methods , Immunophenotyping , Inflammation/etiology , Inflammation/metabolism , Mice , Mice, Knockout , Neutrophils/immunology , Neutrophils/metabolism , Phenotype , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
12.
Nat Immunol ; 21(7): 746-755, 2020 07.
Article in English | MEDLINE | ID: mdl-32514064

ABSTRACT

Plasma membranes of animal cells are enriched for cholesterol. Cholesterol-dependent cytolysins (CDCs) are pore-forming toxins secreted by bacteria that target membrane cholesterol for their effector function. Phagocytes are essential for clearance of CDC-producing bacteria; however, the mechanisms by which these cells evade the deleterious effects of CDCs are largely unknown. Here, we report that interferon (IFN) signals convey resistance to CDC-induced pores on macrophages and neutrophils. We traced IFN-mediated resistance to CDCs to the rapid modulation of a specific pool of cholesterol in the plasma membrane of macrophages without changes to total cholesterol levels. Resistance to CDC-induced pore formation requires the production of the oxysterol 25-hydroxycholesterol (25HC), inhibition of cholesterol synthesis and redistribution of cholesterol to an esterified cholesterol pool. Accordingly, blocking the ability of IFN to reprogram cholesterol metabolism abrogates cellular protection and renders mice more susceptible to CDC-induced tissue damage. These studies illuminate targeted regulation of membrane cholesterol content as a host defense strategy.


Subject(s)
Bacterial Infections/immunology , Bacterial Toxins/immunology , Hydroxycholesterols/metabolism , Interferons/isolation & purification , Phagocytes/immunology , Streptolysins/immunology , Animals , Bacteria/immunology , Bacteria/metabolism , Bacterial Proteins/administration & dosage , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Cell Membrane/metabolism , Cell Membrane Permeability/immunology , Cells, Cultured , Disease Models, Animal , Disease Susceptibility/immunology , Female , Host Microbial Interactions/immunology , Humans , Intravital Microscopy , Male , Mice , Mice, Transgenic , Phagocytes/cytology , Phagocytes/metabolism , Primary Cell Culture , Steroid Hydroxylases/genetics , Steroid Hydroxylases/metabolism , Streptolysins/administration & dosage , Streptolysins/metabolism
13.
Nat Immunol ; 20(9): 1110-1128, 2019 09.
Article in English | MEDLINE | ID: mdl-31406380

ABSTRACT

In recent years, a population of unconventional T cells called 'mucosal-associated invariant T cells' (MAIT cells) has captured the attention of immunologists and clinicians due to their abundance in humans, their involvement in a broad range of infectious and non-infectious diseases and their unusual specificity for microbial riboflavin-derivative antigens presented by the major histocompatibility complex (MHC) class I-like protein MR1. MAIT cells use a limited T cell antigen receptor (TCR) repertoire with public antigen specificities that are conserved across species. They can be activated by TCR-dependent and TCR-independent mechanisms and exhibit rapid, innate-like effector responses. Here we review evidence showing that MAIT cells are a key component of the immune system and discuss their basic biology, development, role in disease and immunotherapeutic potential.


Subject(s)
Antigen Presentation/immunology , Histocompatibility Antigens Class I/immunology , Minor Histocompatibility Antigens/immunology , Mucosal-Associated Invariant T Cells/immunology , Receptors, Antigen, T-Cell/immunology , Animals , Antigens/immunology , Disease Susceptibility/immunology , Humans , Lymphocyte Activation/immunology , Mice , Neoplasms/immunology
14.
Immunity ; 54(11): 2450-2464, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34758337

ABSTRACT

Sepsis is defined as a life-threatening organ dysfunction caused by a dysregulated host response to an infection. This recently implemented definition does not capture the heterogeneity or the underlying pathophysiology of the syndrome, which is characterized by concurrent unbalanced hyperinflammation and immune suppression. Here, we review current knowledge of aberrant immune responses during sepsis and recent initiatives to stratify patients with sepsis into subgroups that are more alike from a clinical and/or pathobiological perspective, which could be key for identification of patients who are more likely to benefit from specific immune interventions.


Subject(s)
Disease Susceptibility/immunology , Host-Pathogen Interactions , Immunity , Sepsis/etiology , Host-Pathogen Interactions/immunology , Humans
15.
Immunity ; 54(6): 1110-1122, 2021 06 08.
Article in English | MEDLINE | ID: mdl-34107270

ABSTRACT

The interindividual heterogeneity of the immune system likely determines the personal risk for acquiring infections and developing diseases with inflammatory components. In addition to genetic factors, the immune system's heterogeneity is driven by diverging exposures of leukocytes and their progenitors to infections, vaccinations, and health behavior, including lifestyle-related stimuli such as diet, physical inactivity, and psychosocial stress. We review how such experiences alter immune cell responses to concurrent and subsequent challenges, leading to either improved host resilience or disease susceptibility due to a muted or overzealous immune system, with a primary focus on the contribution of innate immune cells. We explore the involvement of diverse mechanisms, including trained immunity, and their relevance for infections and cardiovascular disease, as these prevalent conditions are heavily influenced by immune cell abundance and phenotypic adaptions. Understanding the mechanistic bases of immune modulations by prior or co-exposures may lead to new therapies targeting dysfunctional inflammation.


Subject(s)
Cardiovascular Diseases/immunology , Disease Susceptibility/immunology , Immune System/immunology , Infections/immunology , Animals , Humans , Immunity, Innate/immunology , Inflammation/immunology , Life Style
16.
Immunity ; 54(11): 2578-2594.e5, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34717795

ABSTRACT

Peritoneal immune cells reside unanchored within the peritoneal fluid in homeostasis. Here, we examined the mechanisms that control bacterial infection in the peritoneum using a mouse model of abdominal sepsis following intraperitoneal Escherichia coli infection. Whole-mount immunofluorescence and confocal microscopy of the peritoneal wall and omentum revealed that large peritoneal macrophages (LPMs) rapidly cleared bacteria and adhered to the mesothelium, forming multilayered cellular aggregates composed by sequentially recruited LPMs, B1 cells, neutrophils, and monocyte-derived cells (moCs). The formation of resident macrophage aggregates (resMφ-aggregates) required LPMs and thrombin-dependent fibrin polymerization. E. coli infection triggered LPM pyroptosis and release of inflammatory mediators. Resolution of these potentially inflammatory aggregates required LPM-mediated recruitment of moCs, which were essential for fibrinolysis-mediated resMφ-aggregate disaggregation and the prevention of peritoneal overt inflammation. Thus, resMφ-aggregates provide a physical scaffold that enables the efficient control of peritoneal infection, with implications for antimicrobial immunity in other body cavities, such as the pleural cavity or brain ventricles.


Subject(s)
Bacterial Infections/etiology , Bacterial Infections/metabolism , Host-Pathogen Interactions/immunology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Peritoneal Cavity/microbiology , Animals , Biomarkers , Cellular Microenvironment/immunology , Disease Models, Animal , Disease Susceptibility/immunology , Inflammation Mediators/metabolism , Mice , Peritonitis/etiology , Peritonitis/metabolism , Peritonitis/pathology
17.
Immunity ; 54(10): 2172-2176, 2021 Oct 12.
Article in English | MEDLINE | ID: mdl-34626549

ABSTRACT

The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its associated disease, coronavirus disease 2019 (COVID-19), has caused a devastating pandemic worldwide. Here, we explain basic concepts underlying the transition from an epidemic to an endemic state, where a pathogen is stably maintained in a population. We discuss how the number of infections and the severity of disease change in the transition from the epidemic to the endemic phase and consider the implications of this transition in the context of COVID-19.


Subject(s)
COVID-19/epidemiology , COVID-19/immunology , Endemic Diseases , COVID-19/prevention & control , Disease Susceptibility/epidemiology , Disease Susceptibility/immunology , Epidemics , Humans , Immunity , Prevalence , SARS-CoV-2/immunology , Severity of Illness Index , Vaccination
18.
Immunity ; 54(6): 1200-1218.e9, 2021 06 08.
Article in English | MEDLINE | ID: mdl-33951416

ABSTRACT

Tissue macrophages self-renew during homeostasis and produce inflammatory mediators upon microbial infection. We examined the relationship between proliferative and inflammatory properties of tissue macrophages by defining the impact of the Wnt/ß-catenin pathway, a central regulator of self-renewal, in alveolar macrophages (AMs). Activation of ß-catenin by Wnt ligand inhibited AM proliferation and stemness, but promoted inflammatory activity. In a murine influenza viral pneumonia model, ß-catenin-mediated AM inflammatory activity promoted acute host morbidity; in contrast, AM proliferation enabled repopulation of reparative AMs and tissue recovery following viral clearance. Mechanistically, Wnt treatment promoted ß-catenin-HIF-1α interaction and glycolysis-dependent inflammation while suppressing mitochondrial metabolism and thereby, AM proliferation. Differential HIF-1α activities distinguished proliferative and inflammatory AMs in vivo. This ß-catenin-HIF-1α axis was conserved in human AMs and enhanced HIF-1α expression associated with macrophage inflammation in COVID-19 patients. Thus, inflammatory and reparative activities of lung macrophages are regulated by ß-catenin-HIF-1α signaling, with implications for the treatment of severe respiratory diseases.


Subject(s)
COVID-19/immunology , COVID-19/virology , Cell Self Renewal/immunology , Host-Pathogen Interactions/immunology , Macrophages/immunology , SARS-CoV-2/immunology , Biomarkers , COVID-19/metabolism , Cytokines/metabolism , Disease Susceptibility/immunology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation Mediators/metabolism , Macrophages/cytology , Macrophages/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Signal Transduction
19.
Immunity ; 52(6): 942-956, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32553180

ABSTRACT

Dendritic cells (DCs) are the sentinels of the immune system, sensing a diverse array of pathogens to stimulate a robust and appropriate immune response. To initiate responses to highly disparate challenges, DCs have diversified into multiple phenotypically, anatomically, and functionally distinct cell types. As a result of the application of new single-cell technologies, the full extent of this diversity, as well as the developmental relationships of the DC lineages, is currently undergoing reassessment. Here, we review the cellular and molecular evidence that underpins current models of DC differentiation and functional diversification in the murine and human systems. We discuss these models in the context of the diversity revealed by single-cell studies and propose that understanding DC identity will require defining the regulatory interactions that control gene expression in these cells.


Subject(s)
Cell Differentiation/genetics , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Regulation , Gene Regulatory Networks , Transcription, Genetic , Animals , Biomarkers , Cell Lineage/genetics , Disease Susceptibility/immunology , Epigenesis, Genetic , Genetic Predisposition to Disease , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
20.
Immunity ; 52(2): 275-294.e9, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32075728

ABSTRACT

Type 3 innate lymphoid cells (ILC3s) are critical for lung defense against bacterial pneumonia in the neonatal period, but the signals that guide pulmonary ILC3 development remain unclear. Here, we demonstrated that pulmonary ILC3s descended from ILC precursors that populated a niche defined by fibroblasts in the developing lung. Alveolar fibroblasts produced insulin-like growth factor 1 (IGF1), which instructed expansion and maturation of pulmonary ILC precursors. Conditional ablation of IGF1 in alveolar fibroblasts or deletion of the IGF-1 receptor from ILC precursors interrupted ILC3 biogenesis and rendered newborn mice susceptible to pneumonia. Premature infants with bronchopulmonary dysplasia, characterized by interrupted postnatal alveolar development and increased morbidity to respiratory infections, had reduced IGF1 concentrations and pulmonary ILC3 numbers. These findings indicate that the newborn period is a critical window in pulmonary immunity development, and disrupted lung development in prematurely born infants may have enduring effects on host resistance to respiratory infections.


Subject(s)
Immunity, Innate , Insulin-Like Growth Factor I/metabolism , Lung/immunology , Lymphocytes/cytology , Alveolar Epithelial Cells/metabolism , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/immunology , Cell Differentiation , Cell Proliferation , Disease Susceptibility/immunology , Humans , Infant, Newborn , Infant, Premature , Insulin-Like Growth Factor I/deficiency , Interleukins/metabolism , Lung/cytology , Lung/growth & development , Lymphocytes/metabolism , Mice , Pneumonia/immunology , Promyelocytic Leukemia Zinc Finger Protein/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Signal Transduction , Interleukin-22
SELECTION OF CITATIONS
SEARCH DETAIL