Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 217
Filter
1.
Immunol Rev ; 323(1): 126-137, 2024 May.
Article in English | MEDLINE | ID: mdl-38491842

ABSTRACT

Group 3 innate lymphoid cells (ILC3s) are tissue-resident immune lymphocytes that critically regulate intestinal homeostasis, organogenesis, and immunity. ILC3s possess the capacity to "sense" the inflammatory environment within tissues, especially in the context of pathogen challenges that imprints durable non-antigen-specific changes in ILC3 function. As such, ILC3s become a new actor in the emerging field of trained innate immunity. Here, we summarize recent discoveries regarding ILC3 responses to bacterial challenges and the role these encounters play in triggering trained innate immunity. We further discuss how signaling events throughout ILC3 ontogeny potentially control the development and function of trained ILC3s. Finally, we highlight the open questions surrounding ILC3 "training" the answers to which may reveal new insights into innate immunity. Understanding the fundamental concepts behind trained innate immunity could potentially lead to the development of new strategies for improving immunity-based modulation therapies for inflammation, infectious diseases, and cancer.


Subject(s)
Immunity, Innate , Lymphocytes , Signal Transduction , Humans , Animals , Lymphocytes/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Homeostasis , Inflammation/immunology , Gastrointestinal Microbiome/immunology , Intestines/immunology
2.
Cytometry A ; 105(2): 124-138, 2024 02.
Article in English | MEDLINE | ID: mdl-37751141

ABSTRACT

Flow cytometry is the method of choice for immunophenotyping in the context of clinical, translational, and systems immunology studies. Among the latter, the Milieu Intérieur (MI) project aims at defining the boundaries of a healthy immune response to identify determinants of immune response variation. MI used immunophenotyping of a 1000 healthy donor cohort by flow cytometry as a principal outcome for immune variance at steady state. New generation spectral cytometers now enable high-dimensional immune cell characterization from small sample volumes. Therefore, for the MI 10-year follow up study, we have developed two high-dimensional spectral flow cytometry panels for deep characterization of innate and adaptive whole blood immune cells (35 and 34 fluorescent markers, respectively). We have standardized the protocol for sample handling, staining, acquisition, and data analysis. This approach enables the reproducible quantification of over 182 immune cell phenotypes at a single site. We have applied the protocol to discern minor differences between healthy and patient samples and validated its value for application in immunomonitoring studies. Our protocol is currently used for characterization of the impact of age and environmental factors on peripheral blood immune phenotypes of >400 donors from the initial MI cohort.


Subject(s)
Follow-Up Studies , Humans , Immunophenotyping , Phenotype , Flow Cytometry/methods
3.
Proc Natl Acad Sci U S A ; 120(40): e2306761120, 2023 10 03.
Article in English | MEDLINE | ID: mdl-37756335

ABSTRACT

Natural killer (NK) cells and type 1 innate lymphoid cells (ILC1) require signal transducer and activator of transcription 4 (STAT4) to elicit rapid effector responses and protect against pathogens. By combining genetic and transcriptomic approaches, we uncovered divergent roles for STAT4 in regulating effector differentiation of these functionally related cell types. Stat4 deletion in Ncr1-expressing cells led to impaired NK cell terminal differentiation as well as to an unexpected increased generation of cytotoxic ILC1 during intestinal inflammation. Mechanistically, Stat4-deficient ILC1 exhibited upregulation of gene modules regulated by STAT5 in vivo and an aberrant effector differentiation upon in vitro stimulation with IL-2, used as a prototypical STAT5 activator. Moreover, STAT4 expression in NCR+ innate lymphocytes restrained gut inflammation in the dextran sulfate sodium-induced colitis model limiting pathogenic production of IL-13 from adaptive CD4+ T cells in the large intestine. Collectively, our data shed light on shared and distinctive mechanisms of STAT4-regulated transcriptional control in NK cells and ILC1 required for intestinal inflammatory responses.


Subject(s)
Antineoplastic Agents , STAT5 Transcription Factor , Humans , Immunity, Innate , Cell Differentiation , Killer Cells, Natural , Inflammation , STAT4 Transcription Factor/genetics
4.
Eur J Immunol ; 53(12): e2350454, 2023 12.
Article in English | MEDLINE | ID: mdl-37621208

ABSTRACT

Human immune system (HIS) mice provide a model to study human immune responses in vivo. Currently available HIS mouse models may harbor mouse Fc Receptor (FcR)-expressing cells that exert potent effector functions following administration of human Ig. Previous studies showed that the ablation of the murine FcR gamma chain (FcR-γ) results in loss of antibody-dependent cellular cytotoxicity and antibody-dependent cellular phagocytosis in vivo. We created a new FcR-γ-deficient HIS mouse model to compare host (mouse) versus graft (human) effects underlying antibody-mediated immune responses in vivo. FcR-γ-deficient HIS recipients lack expression and function of mouse activating FcRs and can be stably and robustly reconstituted with human immune cells. By screening blood B-cell depletion by rituximab Ig variants, we found that human FcγRs-mediated IgG1 effects, whereas mouse activating FcγRs were dominant in IgG4 effects. Complement played a role as an IgG1 variant (IgG1 K322A) lacking complement binding activity was largely ineffective. Finally, we provide evidence that FcγRIIIA on human NK cells could mediate complement-independent B-cell depletion by IgG1 K322A. We anticipate that our FcR-γ-deficient HIS model will help clarify mechanisms of action of exogenous administered human antibodies in vivo.


Subject(s)
Receptors, Fc , Receptors, IgG , Humans , Mice , Animals , Receptors, IgG/genetics , Immunoglobulin G , Antibody-Dependent Cell Cytotoxicity , Macrophages , Complement System Proteins , Adaptive Immunity
5.
Nat Immunol ; 23(9): 1317-1323, 2022 09.
Article in English | MEDLINE | ID: mdl-35999393

ABSTRACT

An orchestrated cellular network, including adaptive lymphocytes and group 3 innate lymphoid cells (ILC3s), maintains intestinal barrier integrity and homeostasis. T cells can monitor environmental insults through constitutive circulation, scanning tissues and forming immunological contacts, a process named immunosurveillance. In contrast, the dynamics of intestinal ILC3s are unknown. Using intravital imaging, we observed that villus ILC3s were largely immotile at steady state but acquired migratory 'patrolling' attributes and enhanced cytokine expression in response to inflammation. We showed that T cells, the chemokine CCL25 and bacterial ligands regulated intestinal ILC3 behavior and that loss of patrolling behavior by interleukin-22 (IL-22)-producing ILC3s altered the intestinal barrier through increased epithelial cell death. Collectively, we identified notable differences between the behavior of ILC3s and T cells, with a prominent adaptation of intestinal ILC3s toward mucosal immunosurveillance after inflammation.


Subject(s)
Immunity, Innate , Lymphocytes , Cytokines/metabolism , Humans , Inflammation/metabolism , Intestinal Mucosa , Ligands
6.
Nat Commun ; 13(1): 4344, 2022 07 27.
Article in English | MEDLINE | ID: mdl-35896601

ABSTRACT

Innate lymphoid cells (ILCs) include cytotoxic natural killer cells and distinct groups of cytokine-producing innate helper cells which participate in immune defense and promote tissue homeostasis. Circulating human ILC precursors (ILCP) able to generate all canonical ILC subsets via multi-potent or uni-potent intermediates according to our previous work. Here we show potential cooperative roles for the Notch and IL-23 signaling pathways for human ILC differentiation from blood ILCP using single cell cloning analyses and validate these findings in patient samples with rare genetic deficiencies in IL12RB1 and RORC. Mechanistically, Notch signaling promotes upregulation of the transcription factor RORC, enabling acquisition of Group 1 (IFN-γ) and Group 3 (IL-17A, IL-22) effector functions in multi-potent and uni-potent ILCP. Interfering with RORC or signaling through its target IL-23R compromises ILC3 effector functions but also generally suppresses ILC production from multi-potent ILCP. Our results identify a Notch->RORC- > IL-23R pathway which operates during human ILC differentiation. These observations may help guide protocols to expand functional ILC subsets in vitro with an aim towards novel ILC therapies for human disease.


Subject(s)
Immunity, Innate , Lymphocytes , Cell Differentiation , Humans , Interleukin-23 , Killer Cells, Natural , Lymphoid Progenitor Cells/physiology , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Receptors, Notch/metabolism
7.
Curr Opin Hematol ; 29(4): 209-217, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35787549

ABSTRACT

PURPOSE OF REVIEW: Innate lymphoid cells (ILCs) are specialized immune cells that rapidly sense environmental perturbations and regulate immune responses and tissue homeostasis. ILCs are mainly tissue resident and their crosstalk within tissue microenvironments influences both local and systemic metabolism. Reciprocally, metabolic status conditions ILC phenotype and effector function. In this review, we discuss the role of ILCs as metabolic sentinels and describe how ILC subset-specific activities influence homeostasis and disease. Finally, we highlight emerging challenges in the field of ILC immunometabolism. RECENT FINDINGS: Accumulating evidence suggests that ILCs metabolism, phenotype, and function are shaped by signals from the tissue microenvironment. Dietary, endogenous, and microbial metabolites are sensed by ILC subsets and can impact on ILC-mediated immune responses. Recent studies have found that mitochondria are central regulators of ILC effector function. Furthermore, ILCs have emerged as crucial sensors of metabolic stress, suggesting they might act as metabolic sentinels, coordinating tissue and host metabolism. SUMMARY: Our understanding how ILCs mechanistically regulate host metabolism and defenses is still incomplete. Unraveling critical metabolic features of ILCs may lead to novel therapeutic strategies that target these cells in the context of disease.


Subject(s)
Immunity, Innate , Lymphocytes , Homeostasis/physiology , Humans , Immunotherapy
8.
J Exp Med ; 219(3)2022 03 07.
Article in English | MEDLINE | ID: mdl-35230385

ABSTRACT

Decrypting the B cell ontogeny of HIV-1 broadly neutralizing antibodies (bNAbs) is paramount for vaccine design. Here, we characterized IgA and IgG bNAbs of three distinct B cell lineages in a viremic controller, two of which comprised only IgG+ or IgA+ blood memory B cells; the third combined both IgG and IgA clonal variants. 7-269 bNAb in the IgA-only lineage displayed the highest neutralizing capacity despite limited somatic mutation, and delayed viral rebound in humanized mice. bNAbs in all three lineages targeted the N332 glycan supersite. The 2.8-Å resolution cryo-EM structure of 7-269-BG505 SOSIP.664 complex showed a similar pose as 2G12, on an epitope mainly composed of sugar residues comprising the N332 and N295 glycans. Binding and cryo-EM structural analyses showed that antibodies from the two other lineages interact mostly with glycans N332 and N386. Hence, multiple B cell lineages of IgG and IgA bNAbs focused on a unique HIV-1 site of vulnerability can codevelop in HIV-1 viremic controllers.


Subject(s)
HIV Infections , HIV-1 , Animals , Antibodies, Neutralizing , Broadly Neutralizing Antibodies , Elite Controllers , Epitopes , HIV Antibodies , Humans , Immunoglobulin A , Immunoglobulin G , Mice , Polysaccharides , env Gene Products, Human Immunodeficiency Virus
9.
Science ; 375(6583): 859-863, 2022 02 25.
Article in English | MEDLINE | ID: mdl-35201883

ABSTRACT

Group 3 innate lymphoid cells (ILC3s) are innate immune effectors that contribute to host defense. Whether ILC3 functions are stably modified after pathogen encounter is unknown. Here, we assess the impact of a time-restricted enterobacterial challenge to long-term ILC3 activation in mice. We found that intestinal ILC3s persist for months in an activated state after exposure to Citrobacter rodentium. Upon rechallenge, these "trained" ILC3s proliferate, display enhanced interleukin-22 (IL-22) responses, and have a superior capacity to control infection compared with naïve ILC3s. Metabolic changes occur in C. rodentium-exposed ILC3s, but only trained ILC3s have an enhanced proliferative capacity that contributes to increased IL-22 production. Accordingly, a limited encounter with a pathogen can promote durable phenotypic and functional changes in intestinal ILC3s that contribute to long-term mucosal defense.


Subject(s)
Citrobacter rodentium/immunology , Enterobacteriaceae Infections/immunology , Immunity, Mucosal , Intestinal Mucosa/immunology , Lymphocyte Activation , Lymphocytes/immunology , Adaptive Immunity , Animals , Cell Proliferation , Female , Immunity, Innate , Immunologic Memory , Interleukins/metabolism , Intestines/immunology , Listeria monocytogenes , Listeriosis/immunology , Lymphocytes/metabolism , Male , Metabolic Networks and Pathways , Mice , Mice, Inbred C57BL , Oxygen Consumption , RNA-Seq , Reinfection/immunology , Interleukin-22
10.
Blood ; 139(17): 2585-2600, 2022 04 28.
Article in English | MEDLINE | ID: mdl-35157765

ABSTRACT

Both innate and adaptive lymphocytes have critical roles in mucosal defense that contain commensal microbial communities and protect against pathogen invasion. Here we characterize mucosal immunity in patients with severe combined immunodeficiency (SCID) receiving hematopoietic stem cell transplantation (HSCT) with or without myeloablation. We confirmed that pretransplant conditioning had an impact on innate (natural killer and innate lymphoid cells) and adaptive (B and T cells) lymphocyte reconstitution in these patients with SCID and now show that this further extends to generation of T helper 2 and type 2 cytotoxic T cells. Using an integrated approach to assess nasopharyngeal immunity, we identified a local mucosal defect in type 2 cytokines, mucus production, and a selective local immunoglobulin A (IgA) deficiency in HSCT-treated SCID patients with genetic defects in IL2RG/GC or JAK3. These patients have a reduction in IgA-coated nasopharyngeal bacteria and exhibit microbial dysbiosis with increased pathobiont carriage. Interestingly, intravenous immunoglobulin replacement therapy can partially normalize nasopharyngeal immunoglobulin profiles and restore microbial communities in GC/JAK3 patients. Together, our results suggest a potential nonredundant role for type 2 immunity and/or of local IgA antibody production in the maintenance of nasopharyngeal microbial homeostasis and mucosal barrier function.


Subject(s)
Severe Combined Immunodeficiency , Dysbiosis , Humans , Immunity, Innate , Immunity, Mucosal , Immunoglobulin A , Interleukin Receptor Common gamma Subunit/genetics , Janus Kinase 3/genetics , Lymphocytes/metabolism , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/therapy
11.
J Exp Med ; 219(2)2022 02 07.
Article in English | MEDLINE | ID: mdl-35019940

ABSTRACT

Despite their importance in lung health and disease, it remains unknown how human alveolar macrophages develop early in life. Here we define the ontogeny of human alveolar macrophages from embryonic progenitors in vivo, using a humanized mouse model expressing human cytokines (MISTRG mice). We identified alveolar macrophage progenitors in human fetal liver that expressed the GM-CSF receptor CD116 and the transcription factor MYB. Transplantation experiments in MISTRG mice established a precursor-product relationship between CD34-CD116+ fetal liver cells and human alveolar macrophages in vivo. Moreover, we discovered circulating CD116+CD64-CD115+ macrophage precursors that migrated from the liver to the lung. Similar precursors were present in human fetal lung and expressed the chemokine receptor CX3CR1. Fetal CD116+CD64- macrophage precursors had a proliferative gene signature, outcompeted adult precursors in occupying the perinatal alveolar niche, and developed into functional alveolar macrophages. The discovery of the fetal alveolar macrophage progenitor advances our understanding of human macrophage origin and ontogeny.


Subject(s)
Cell Differentiation , Cell Movement , Macrophages, Alveolar/cytology , Macrophages, Alveolar/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Stem Cells/metabolism , Animals , Biomarkers , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Movement/genetics , Cell Movement/immunology , Fetus , Gene Expression , Genes, myb , Humans , Immunohistochemistry , Immunophenotyping , Liver/cytology , Lung/cytology , Mice , Mice, Transgenic , Stem Cells/cytology
12.
J Biol Chem ; 298(1): 101290, 2022 01.
Article in English | MEDLINE | ID: mdl-34678315

ABSTRACT

The current COVID-19 pandemic illustrates the importance of obtaining reliable methods for the rapid detection of SARS-CoV-2. A highly specific and sensitive diagnostic test able to differentiate the SARS-CoV-2 virus from common human coronaviruses is therefore needed. Coronavirus nucleoprotein (N) localizes to the cytoplasm and the nucleolus and is required for viral RNA synthesis. N is the most abundant coronavirus protein, so it is of utmost importance to develop specific antibodies for its detection. In this study, we developed a sandwich immunoassay to recognize the SARS-CoV-2 N protein. We immunized one alpaca with recombinant SARS-CoV-2 N and constructed a large single variable domain on heavy chain (VHH) antibody library. After phage display selection, seven VHHs recognizing the full N protein were identified by ELISA. These VHHs did not recognize the nucleoproteins of the four common human coronaviruses. Hydrogen Deuterium eXchange-Mass Spectrometry (HDX-MS) analysis also showed that these VHHs mainly targeted conformational epitopes in either the C-terminal or the N-terminal domains. All VHHs were able to recognize SARS-CoV-2 in infected cells or on infected hamster tissues. Moreover, the VHHs could detect the SARS variants B.1.17/alpha, B.1.351/beta, and P1/gamma. We propose that this sandwich immunoassay could be applied to specifically detect the SARS-CoV-2 N in human nasal swabs.


Subject(s)
Enzyme-Linked Immunosorbent Assay/methods , Nucleocapsid Proteins/analysis , SARS-CoV-2/immunology , Single-Domain Antibodies/immunology , Animals , Cricetinae , Electrophoresis, Polyacrylamide Gel , Humans , Limit of Detection , Nucleocapsid Proteins/immunology
14.
Eur J Immunol ; 51(12): 2708-3145, 2021 12.
Article in English | MEDLINE | ID: mdl-34910301

ABSTRACT

The third edition of Flow Cytometry Guidelines provides the key aspects to consider when performing flow cytometry experiments and includes comprehensive sections describing phenotypes and functional assays of all major human and murine immune cell subsets. Notably, the Guidelines contain helpful tables highlighting phenotypes and key differences between human and murine cells. Another useful feature of this edition is the flow cytometry analysis of clinical samples with examples of flow cytometry applications in the context of autoimmune diseases, cancers as well as acute and chronic infectious diseases. Furthermore, there are sections detailing tips, tricks and pitfalls to avoid. All sections are written and peer-reviewed by leading flow cytometry experts and immunologists, making this edition an essential and state-of-the-art handbook for basic and clinical researchers.


Subject(s)
Autoimmune Diseases/immunology , Flow Cytometry , Infections/immunology , Neoplasms/immunology , Animals , Chronic Disease , Humans , Mice , Practice Guidelines as Topic
15.
Nat Commun ; 12(1): 6277, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34725327

ABSTRACT

Several COVID-19 vaccines have now been deployed to tackle the SARS-CoV-2 pandemic, most of them based on messenger RNA or adenovirus vectors.The duration of protection afforded by these vaccines is unknown, as well as their capacity to protect from emerging new variants. To provide sufficient coverage for the world population, additional strategies need to be tested. The live pediatric measles vaccine (MV) is an attractive approach, given its extensive safety and efficacy history, along with its established large-scale manufacturing capacity. We develop an MV-based SARS-CoV-2 vaccine expressing the prefusion-stabilized, membrane-anchored full-length S antigen, which proves to be efficient at eliciting strong Th1-dominant T-cell responses and high neutralizing antibody titers. In both mouse and golden Syrian hamster models, these responses protect the animals from intranasal infectious challenge. Additionally, the elicited antibodies efficiently neutralize in vitro the three currently circulating variants of SARS-CoV-2.


Subject(s)
COVID-19 Vaccines/immunology , COVID-19/immunology , Genetic Vectors , Immunity , Adenoviridae , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , COVID-19/prevention & control , COVID-19 Vaccines/administration & dosage , Cricetinae , Cytokines , Female , Immunization , Immunization, Secondary , Male , Measles Vaccine/immunology , Mesocricetus , Mice , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology
16.
Nat Immunol ; 22(11): 1367-1374, 2021 11.
Article in English | MEDLINE | ID: mdl-34686862

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) represent innate homologs of type 2 helper T cells (TH2) that participate in immune defense and tissue homeostasis through production of type 2 cytokines. While T lymphocytes metabolically adapt to microenvironmental changes, knowledge of human ILC2 metabolism is limited, and its key regulators are unknown. Here, we show that circulating 'naive' ILC2s have an unexpected metabolic profile with a higher level of oxidative phosphorylation (OXPHOS) than natural killer (NK) cells. Accordingly, ILC2s are severely reduced in individuals with mitochondrial disease (MD) and impaired OXPHOS. Metabolomic and nutrient receptor analysis revealed ILC2 uptake of amino acids to sustain OXPHOS at steady state. Following activation with interleukin-33 (IL-33), ILC2s became highly proliferative, relying on glycolysis and mammalian target of rapamycin (mTOR) to produce IL-13 while continuing to fuel OXPHOS with amino acids to maintain cellular fitness and proliferation. Our results suggest that proliferation and function are metabolically uncoupled in human ILC2s, offering new strategies to target ILC2s in disease settings.


Subject(s)
Cell Proliferation , Cytokines/metabolism , Energy Metabolism , Immunity, Innate , Lymphocyte Activation , Mitochondrial Diseases/metabolism , Th2 Cells/metabolism , Amino Acids, Branched-Chain/metabolism , Arginine/metabolism , Case-Control Studies , Cell Proliferation/drug effects , Cells, Cultured , Energy Metabolism/drug effects , Humans , Immunity, Innate/drug effects , Interleukin-33/pharmacology , Lymphocyte Activation/drug effects , Mitochondria/metabolism , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/immunology , Phenotype , Th2 Cells/drug effects , Th2 Cells/immunology
17.
Cell Rep ; 37(4): 109887, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34706233

ABSTRACT

In emergency myelopoiesis (EM), expansion of the myeloid progenitor compartment and increased myeloid cell production are observed and often mediated by the pro-inflammatory cytokine interferon gamma (IFN-γ). Interleukin-10 (IL-10) inhibits IFN-γ secretion, but paradoxically, its therapeutic administration to humans causes hematologic changes similar to those observed in EM. In this work, we use different in vivo systems, including a humanized immune system mouse model, to show that IL-10 triggers EM, with a significant expansion of the myeloid progenitor compartment and production of myeloid cells. Hematopoietic progenitors display a prominent IFN-γ transcriptional signature, and we show that IFN-γ mediates IL-10-driven EM. We also find that IL-10, unexpectedly, reprograms CD4 and CD8 T cells toward an activation state that includes IFN-γ production by these T cell subsets in vivo. Therefore, in addition to its established anti-inflammatory properties, IL-10 can induce IFN-γ production and EM, opening additional perspectives for the design of IL-10-based immunotherapies.


Subject(s)
Interferon-gamma/immunology , Interleukin-10/immunology , Myeloid Progenitor Cells/immunology , Myelopoiesis/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Interferon-gamma/genetics , Interleukin-10/genetics , Mice , Mice, Knockout , Myelopoiesis/genetics
18.
J Clin Invest ; 131(17)2021 09 01.
Article in English | MEDLINE | ID: mdl-34623332

ABSTRACT

We studied a child with severe viral, bacterial, fungal, and parasitic diseases, who was homozygous for a loss-of-function mutation of REL, encoding c-Rel, which is selectively expressed in lymphoid and myeloid cells. The patient had low frequencies of NK, effector memory cells reexpressing CD45RA (Temra) CD8+ T cells, memory CD4+ T cells, including Th1 and Th1*, Tregs, and memory B cells, whereas the counts and proportions of other leukocyte subsets were normal. Functional deficits of myeloid cells included the abolition of IL-12 and IL-23 production by conventional DC1s (cDC1s) and monocytes, but not cDC2s. c-Rel was also required for induction of CD86 expression on, and thus antigen-presenting cell function of, cDCs. Functional deficits of lymphoid cells included reduced IL-2 production by naive T cells, correlating with low proliferation and survival rates and poor production of Th1, Th2, and Th17 cytokines by memory CD4+ T cells. In naive CD4+ T cells, c-Rel is dispensable for early IL2 induction but contributes to later phases of IL2 expression. The patient's naive B cells displayed impaired MYC and BCL2L1 induction, compromising B cell survival and proliferation and preventing their differentiation into Ig-secreting plasmablasts. Inherited c-Rel deficiency disrupts the development and function of multiple myeloid and lymphoid cells, compromising innate and adaptive immunity to multiple infectious agents.


Subject(s)
Genes, rel , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/immunology , Proto-Oncogene Proteins c-rel/deficiency , Proto-Oncogene Proteins c-rel/genetics , Adaptive Immunity/genetics , Adaptive Immunity/immunology , Child , Consanguinity , Female , Hematopoietic Stem Cell Transplantation , Homozygote , Host Microbial Interactions/genetics , Host Microbial Interactions/immunology , Humans , Immunity, Innate/genetics , Immunity, Innate/immunology , Lymphocyte Activation , Lymphocytes/classification , Lymphocytes/immunology , Mutation , Myeloid Cells/immunology , Primary Immunodeficiency Diseases/therapy , Protein Isoforms
19.
Nat Immunol ; 22(11): 1428-1439, 2021 11.
Article in English | MEDLINE | ID: mdl-34471264

ABSTRACT

Coordinated local mucosal and systemic immune responses following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection either protect against coronavirus disease 2019 (COVID-19) pathologies or fail, leading to severe clinical outcomes. To understand this process, we performed an integrated analysis of SARS-CoV-2 spike-specific antibodies, cytokines, viral load and bacterial communities in paired nasopharyngeal swabs and plasma samples from a cohort of clinically distinct patients with COVID-19 during acute infection. Plasma viral load was associated with systemic inflammatory cytokines that were elevated in severe COVID-19, and also with spike-specific neutralizing antibodies. By contrast, nasopharyngeal viral load correlated with SARS-CoV-2 humoral responses but inversely with interferon responses, the latter associating with protective microbial communities. Potential pathogenic microorganisms, often implicated in secondary respiratory infections, were associated with mucosal inflammation and elevated in severe COVID-19. Our results demonstrate distinct tissue compartmentalization of SARS-CoV-2 immune responses and highlight a role for the nasopharyngeal microbiome in regulating local and systemic immunity that determines COVID-19 clinical outcomes.


Subject(s)
COVID-19/immunology , Microbiota/immunology , Nasopharynx/immunology , SARS-CoV-2/physiology , Acute Disease , Adolescent , Adult , Aged , Antibodies, Viral/blood , Cohort Studies , Female , Humans , Immunity, Humoral , Immunity, Mucosal , Interferons/blood , Male , Middle Aged , Nasopharynx/microbiology , Spike Glycoprotein, Coronavirus/immunology , Viral Load , Young Adult
20.
Eur J Immunol ; 51(9): 2110-2111, 2021 09.
Article in English | MEDLINE | ID: mdl-34468027
SELECTION OF CITATIONS
SEARCH DETAIL
...