Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 243
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 39: 417-447, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33902312

ABSTRACT

Natural killer (NK) cells are innate lymphocytes that provide critical host defense against pathogens and cancer. Originally heralded for their early and rapid effector activity, NK cells have been recognized over the last decade for their ability to undergo adaptive immune processes, including antigen-driven clonal expansion and generation of long-lived memory. This review presents an overview of how NK cells lithely partake in both innate and adaptive responses and how this versatility is manifest in human NK cell-mediated immunity.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Animals , Humans , Immunity, Cellular , Killer Cells, Natural
2.
Annu Rev Immunol ; 38: 229-247, 2020 04 26.
Article in English | MEDLINE | ID: mdl-31928469

ABSTRACT

Neonatal CD4+ and CD8+ T cells have historically been characterized as immature or defective. However, recent studies prompt a reinterpretation of the functions of neonatal T cells. Rather than a population of cells always falling short of expectations set by their adult counterparts, neonatal T cells are gaining recognition as a distinct population of lymphocytes well suited for the rapidly changing environment in early life. In this review, I will highlight new evidence indicating that neonatal T cells are not inert or less potent versions of adult T cells but instead are a broadly reactive layer of T cells poised to quickly develop into regulatory or effector cells, depending on the needs of the host. In this way, neonatal T cells are well adapted to provide fast-acting immune protection against foreign pathogens, while also sustaining tolerance to self-antigens.


Subject(s)
T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Adaptive Immunity , Animals , Biomarkers , Cell Differentiation/immunology , Host-Pathogen Interactions , Humans , Immunologic Memory , Lymphocyte Activation/immunology , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/immunology , Lymphoid Progenitor Cells/metabolism , Phenotype , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocyte Subsets/cytology
3.
Cell ; 180(1): 92-106.e11, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31866068

ABSTRACT

Repeated exposure to pathogens or their antigens triggers anamnestic antibody responses that are higher in magnitude and affinity than the primary response. These involve reengagement of memory B cell (MBC) clones, the diversity and specificity of which determine the breadth and effectiveness of the ensuing antibody response. Using prime-boost models in mice, we find that secondary responses are characterized by a clonality bottleneck that restricts the engagement of the large diversity of MBC clones generated by priming. Rediversification of mutated MBCs is infrequent within secondary germinal centers (GCs), which instead consist predominantly of B cells without prior GC experience or detectable clonal expansion. Few MBC clones, generally derived from higher-affinity germline precursors, account for the majority of secondary antibody responses, while most primary-derived clonal diversity is not reengaged detectably by boosting. Understanding how to counter this bottleneck may improve our ability to elicit antibodies to non-immunodominant epitopes by vaccination.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , Immunologic Memory/immunology , Adaptive Immunity/immunology , Animals , Antibody Formation/immunology , Antibody Formation/physiology , Antigens/immunology , B-Lymphocytes/metabolism , CHO Cells , Cell Line , Cricetulus , Female , Germinal Center/metabolism , Humans , Immunologic Memory/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal
4.
Immunity ; 57(8): 1848-1863.e7, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-38889716

ABSTRACT

Expression of the transcriptional regulator ZFP318 is induced in germinal center (GC)-exiting memory B cell precursors and memory B cells (MBCs). Using a conditional ZFP318 fluorescence reporter that also enables ablation of ZFP318-expressing cells, we found that ZFP318-expressing MBCs were highly enriched with GC-derived cells. Although ZFP318-expressing MBCs constituted only a minority of the antigen-specific MBC compartment, their ablation severely impaired recall responses. Deletion of Zfp318 did not alter the magnitude of primary responses but markedly reduced MBC participation in recall. CD40 ligation promoted Zfp318 expression, whereas B cell receptor (BCR) signaling was inhibitory. Enforced ZFP318 expression enhanced recall performance of MBCs that otherwise responded poorly. ZFP318-deficient MBCs expressed less mitochondrial genes, had structurally compromised mitochondria, and were susceptible to reactivation-induced cell death. The abundance of ZFP318-expressing MBCs, instead of the number of antigen-specific MBCs, correlated with the potency of prime-boost vaccination. Therefore, ZFP318 controls the MBC recallability and represents a quality checkpoint of humoral immune memory.


Subject(s)
Germinal Center , Immunologic Memory , Memory B Cells , Mitochondria , Animals , Mitochondria/metabolism , Mitochondria/immunology , Mice , Immunologic Memory/genetics , Immunologic Memory/immunology , Memory B Cells/immunology , Memory B Cells/metabolism , Germinal Center/immunology , Receptors, Antigen, B-Cell/metabolism , Receptors, Antigen, B-Cell/genetics , Gene Expression Regulation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Transcription Factors/metabolism , Transcription Factors/genetics , Signal Transduction/immunology , CD40 Antigens/metabolism , CD40 Antigens/genetics , CD40 Antigens/immunology , Immunity, Humoral , Transcription, Genetic , Membrane Proteins , Mitochondrial Proteins
5.
Cell ; 174(1): 117-130.e14, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29909981

ABSTRACT

Heterogeneity is a hallmark feature of the adaptive immune system in vertebrates. Following infection, naive T cells differentiate into various subsets of effector and memory T cells, which help to eliminate pathogens and maintain long-term immunity. The current model suggests there is a single lineage of naive T cells that give rise to different populations of effector and memory T cells depending on the type and amounts of stimulation they encounter during infection. Here, we have discovered that multiple sub-populations of cells exist in the naive CD8+ T cell pool that are distinguished by their developmental origin, unique transcriptional profiles, distinct chromatin landscapes, and different kinetics and phenotypes after microbial challenge. These data demonstrate that the naive CD8+ T cell pool is not as homogeneous as previously thought and offers a new framework for explaining the remarkable heterogeneity in the effector and memory T cell subsets that arise after infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Genes, Developmental , Listeria monocytogenes/pathogenicity , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Cell Line, Tumor , Chromatin/metabolism , Cytokines/pharmacology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/metabolism , Immunologic Memory , Interferon-gamma/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Listeria monocytogenes/metabolism , Mice , Mice, Inbred C57BL , Principal Component Analysis , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Thymus Gland/transplantation , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome
6.
Immunity ; 54(6): 1245-1256.e5, 2021 06 08.
Article in English | MEDLINE | ID: mdl-34004140

ABSTRACT

We examined how baseline CD4+ T cell repertoire and precursor states impact responses to pathogen infection in humans using primary immunization with yellow fever virus (YFV) vaccine. YFV-specific T cells in unexposed individuals were identified by peptide-MHC tetramer staining and tracked pre- and post-vaccination by tetramers and TCR sequencing. A substantial number of YFV-reactive T cells expressed memory phenotype markers and contained expanded clones in the absence of exposure to YFV. After vaccination, pre-existing YFV-specific T cell populations with low clonal diversity underwent limited expansion, but rare populations with a reservoir of unexpanded TCRs generated robust responses. These altered dynamics reorganized the immunodominance hierarchy and resulted in an overall increase in higher avidity T cells. Thus, instead of further increasing the representation of dominant clones, YFV vaccination recruits rare and more responsive T cells. Our findings illustrate the impact of vaccines in prioritizing T cell responses and reveal repertoire reorganization as a key component of effective vaccination.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Yellow Fever Vaccine/immunology , Yellow Fever/immunology , Yellow fever virus/immunology , Adult , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antigens, Viral/immunology , Cells, Cultured , Chlorocebus aethiops , Humans , Receptors, Antigen, T-Cell/immunology , Vaccination/methods , Vero Cells , Yellow Fever/virology
7.
Immunity ; 54(9): 2133-2142.e3, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34453880

ABSTRACT

SARS-CoV-2 mRNA vaccines have shown remarkable clinical efficacy, but questions remain about the nature and kinetics of T cell priming. We performed longitudinal antigen-specific T cell analyses on healthy SARS-CoV-2-naive and recovered individuals prior to and following mRNA prime and boost vaccination. Vaccination induced rapid antigen-specific CD4+ T cell responses in naive subjects after the first dose, whereas CD8+ T cell responses developed gradually and were variable in magnitude. Vaccine-induced Th1 and Tfh cell responses following the first dose correlated with post-boost CD8+ T cells and neutralizing antibodies, respectively. Integrated analysis revealed coordinated immune responses with distinct trajectories in SARS-CoV-2-naive and recovered individuals. Last, whereas booster vaccination improved T cell responses in SARS-CoV-2-naive subjects, the second dose had little effect in SARS-CoV-2-recovered individuals. These findings highlight the role of rapidly primed CD4+ T cells in coordinating responses to the second vaccine dose in SARS-CoV-2-naive individuals.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , COVID-19 Vaccines/immunology , COVID-19/immunology , SARS-CoV-2/physiology , Th1 Cells/immunology , 2019-nCoV Vaccine mRNA-1273 , Adult , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , BNT162 Vaccine , Female , Humans , Immunity, Cellular , Immunity, Humoral , Immunization, Secondary , Immunologic Memory , Lectins, C-Type/metabolism , Lymphocyte Activation , Male , Middle Aged , Peptides/immunology , Spike Glycoprotein, Coronavirus/immunology , Vaccination , Young Adult
8.
Immunity ; 54(9): 2005-2023.e10, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34525339

ABSTRACT

Cell fate decisions during early B cell activation determine the outcome of responses to pathogens and vaccines. We examined the early B cell response to T-dependent antigen in mice by single-cell RNA sequencing. Early after immunization, a homogeneous population of activated precursors (APs) gave rise to a transient wave of plasmablasts (PBs), followed a day later by the emergence of germinal center B cells (GCBCs). Most APs rapidly exited the cell cycle, giving rise to non-GC-derived early memory B cells (eMBCs) that retained an AP-like transcriptional profile. Rapid decline of antigen availability controlled these events; provision of excess antigen precluded cell cycle exit and induced a new wave of PBs. Fate mapping revealed a prominent contribution of eMBCs to the MBC pool. Quiescent cells with an MBC phenotype dominated the early response to immunization in primates. A reservoir of APs/eMBCs may enable rapid readjustment of the immune response when failure to contain a threat is manifested by increased antigen availability.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , Immunity, Humoral/immunology , Immunologic Memory/immunology , Lymphocyte Activation/immunology , Animals , Antigen Presentation/immunology , Cell Differentiation/immunology , Mice , Plasma Cells/immunology , Precursor Cells, B-Lymphoid/immunology
9.
Proc Natl Acad Sci U S A ; 121(9): e2309153121, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38386711

ABSTRACT

The molecular mechanisms leading to the establishment of immunological memory are inadequately understood, limiting the development of effective vaccines and durable antitumor immune therapies. Here, we show that ectopic OCA-B expression is sufficient to improve antiviral memory recall responses, while having minimal effects on primary effector responses. At peak viral response, short-lived effector T cell populations are expanded but show increased Gadd45b and Socs2 expression, while memory precursor effector cells show increased expression of Bcl2, Il7r, and Tcf7 on a per-cell basis. Using an OCA-B mCherry reporter mouse line, we observe high OCA-B expression in CD4+ central memory T cells. We show that early in viral infection, endogenously elevated OCA-B expression prospectively identifies memory precursor cells with increased survival capability and memory recall potential. Cumulatively, the results demonstrate that OCA-B is both necessary and sufficient to promote CD4 T cell memory in vivo and can be used to prospectively identify memory precursor cells.


Subject(s)
CD4-Positive T-Lymphocytes , Memory T Cells , Animals , Mice , Immunologic Memory , Memory , Receptors, Interleukin-7 , Trans-Activators , GADD45 Proteins , Antigens, Differentiation
10.
Proc Natl Acad Sci U S A ; 120(18): e2216918120, 2023 05 02.
Article in English | MEDLINE | ID: mdl-37094168

ABSTRACT

Activation-induced cytidine deaminase (AID) is the essential enzyme for imprinting immunological memory through class switch recombination (CSR) and somatic hypermutation (SHM) of the immunoglobulin (Ig) gene. AID-dependent reduction of Topoisomerase 1 (Top1) promotes DNA cleavage that occurs upon Ig gene diversification, whereas the mechanism behind AID-induced Top1 reduction remains unclear. Here, we clarified the contribution of the microRNA-Ago2 complex in AID-dependent Top1 decrease. Ago2 binds to Top1 3'UTR with two regions of AID-dependent Ago2-binding sites (5'- and 3'dABs). Top1 3'UTR knockout (3'UTRKO) in B lymphoma cells leads to decreases in DNA break efficiency in the IgH gene accompanied by a reduction in CSR and SHM frequencies. Furthermore, AID-dependent Top1 protein reduction and Ago2-binding to Top1 mRNA are down-regulated in 3'UTRKO cells. Top1 mRNA in the highly translated fractions of the sucrose gradient is decreased in an AID-dependent and Top1 3'UTR-mediated manner, resulting in a decrease in Top1 protein synthesis. Both AID and Ago2 localize in the mRNA-binding protein fractions and they interact with each other. Furthermore, we found some candidate miRNAs which possibly bind to 5'- and 3'dAB in Top1 mRNA. Among them, miR-92a-3p knockdown induces the phenotypes of 3'UTRKO cells to wild-type cells whereas it does not impact on 3'UTRKO cells. Taken together, the Ago2-miR-92a-3p complex will be recruited to Top1 3'UTR in an AID-dependent manner and posttranscriptionally reduces Top1 protein synthesis. These consequences cause the increase in a non-B-DNA structure, enhance DNA cleavage by Top1 in the Ig gene and contribute to immunological memory formation.


Subject(s)
MicroRNAs , MicroRNAs/genetics , 3' Untranslated Regions , DNA Cleavage , Cytidine Deaminase/genetics , Immunoglobulin Class Switching , Antibodies/genetics , Somatic Hypermutation, Immunoglobulin
11.
Proc Natl Acad Sci U S A ; 120(34): e2219932120, 2023 08 22.
Article in English | MEDLINE | ID: mdl-37579158

ABSTRACT

Tissue-resident memory CD8+ T cells (TRM) reside at sites of previous infection, providing protection against reinfection with the same pathogen. In the skin, TRM patrol the epidermis, where keratinocytes are the entry site for many viral infections. Epidermal TRM react rapidly to cognate antigen encounter with the secretion of cytokines and differentiation into cytotoxic effector cells, constituting a first line of defense against skin reinfection. Despite the important protective role of skin TRM, it has remained unclear, whether their reactivation requires a professional antigen-presenting cell (APC). We show here, using a model system that allows antigen targeting selectively to keratinocytes in a defined area of the skin, that limited antigen expression by keratinocytes results in rapid, antigen-specific reactivation of skin TRM. Our data identify epidermal Langerhans cells that cross-present keratinocyte-derived antigens, as the professional APC indispensable for the early reactivation of TRM in the epidermal layer of the skin.


Subject(s)
CD8-Positive T-Lymphocytes , Langerhans Cells , Humans , Memory T Cells , Reinfection/metabolism , Epidermis , Antigens , Immunologic Memory
12.
EMBO Rep ; 24(10): e56829, 2023 Oct 09.
Article in English | MEDLINE | ID: mdl-37610043

ABSTRACT

Neonatal health is determined by the transfer of maternal antibodies from the mother to the fetus. Besides antibodies, maternal cells cross the placental barrier and seed into fetal organs. Contrary to maternal antibodies, maternal microchimeric cells (MMc) show a high longevity, as they can persist in the offspring until adulthood. Recent evidence highlights that MMc leukocytes promote neonatal immunity against early-life infections in mice and humans. As shown in mice, this promotion of immunity was attributable to an improved fetal immune development. Besides this indirect effect, MMc may be pathogen-specific and thus, directly clear pathogen threats in the offspring postnatally. By using ovalbumin recombinant Listeria monocytogenes (LmOVA), we here provide evidence that OVA-specific T cells are transferred from the mother to the fetus, which is associated with increased activation of T cells and a milder course of postnatal infection in the offspring. Our data highlight that maternally-derived passive immunity of the neonate is not limited to antibodies, as MMc have the potential to transfer immune memory between generations.

13.
Bioessays ; 45(5): e2200236, 2023 05.
Article in English | MEDLINE | ID: mdl-37009769

ABSTRACT

Pre-existing but untranslated or 'poised' mRNA exists as a means to rapidly induce the production of specific proteins in response to stimuli and as a safeguard to limit the actions of these proteins. The translation of poised mRNA enables immune cells to express quickly genes that enhance immune responses. The molecular mechanisms that repress the translation of poised mRNA and, upon stimulation, enable translation have yet to be elucidated. They likely reflect intrinsic properties of the mRNAs and their interactions with trans-acting factors that direct poised mRNAs away from or into the ribosome. Here, I discuss mechanisms by which this might be regulated.


Subject(s)
Gene Expression Regulation , Lymphocytes , Protein Biosynthesis , RNA, Messenger , RNA, Messenger/chemistry , RNA, Messenger/metabolism , 5' Untranslated Regions , 3' Untranslated Regions , Immunity , Lymphocytes/immunology , Lymphocytes/metabolism , Humans , Animals
14.
Semin Immunol ; 55: 101507, 2021 06.
Article in English | MEDLINE | ID: mdl-34716096

ABSTRACT

Coronaviruses are evolutionarily successful RNA viruses, common to multiple avian, amphibian and mammalian hosts. Despite their ubiquity and potential impact, knowledge of host immunity to coronaviruses remains incomplete, partly owing to the lack of overt pathogenicity of endemic human coronaviruses (HCoVs), which typically cause common colds. However, the need for deeper understanding became pressing with the zoonotic introduction of three novel coronaviruses in the past two decades, causing severe acute respiratory syndromes in humans, and the unfolding pandemic of coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This renewed interest not only triggered the discovery of two of the four HCoVs, but also uncovered substantial cellular and humoral cross-reactivity with shared or related coronaviral antigens. Here, we review the evidence for cross-reactive B cell memory elicited by HCoVs and its potential impact on the puzzlingly variable outcome of SARS-CoV-2 infection. The available data indicate targeting of highly conserved regions primarily in the S2 subunits of the spike glycoproteins of HCoVs and SARS-CoV-2 by cross-reactive B cells and antibodies. Rare monoclonal antibodies reactive with conserved S2 epitopes and with potent virus neutralising activity have been cloned, underscoring the potential functional relevance of cross-reactivity. We discuss B cell and antibody cross-reactivity in the broader context of heterologous humoral immunity to coronaviruses, as well as the limits of protective immune memory against homologous re-infection. Given the bidirectional nature of cross-reactivity, the unprecedented current vaccination campaign against SARS-CoV-2 is expected to impact HCoVs, as well as future zoonotic coronaviruses attempting to cross the species barrier. However, emerging SARS-CoV-2 variants with resistance to neutralisation by vaccine-induced antibodies highlight a need for targeting more constrained, less mutable parts of the spike. The delineation of such cross-reactive areas, which humoral immunity can be trained to attack, may offer the key to permanently shifting the balance of our interaction with current and future coronaviruses in our favour.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Antibodies, Viral , Humans , Immunity, Humoral
15.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Article in English | MEDLINE | ID: mdl-35217611

ABSTRACT

Rapid clonal expansion of antigen-specific T cells is a fundamental feature of adaptive immune responses. It enables the outgrowth of an individual T cell into thousands of clonal descendants that diversify into short-lived effectors and long-lived memory cells. Clonal expansion is thought to be programmed upon priming of a single naive T cell and then executed by homogenously fast divisions of all of its descendants. However, the actual speed of cell divisions in such an emerging "T cell family" has never been measured with single-cell resolution. Here, we utilize continuous live-cell imaging in vitro to track the division speed and genealogical connections of all descendants derived from a single naive CD8+ T cell throughout up to ten divisions of activation-induced proliferation. This comprehensive mapping of T cell family trees identifies a short burst phase, in which division speed is homogenously fast and maintained independent of external cytokine availability or continued T cell receptor stimulation. Thereafter, however, division speed diversifies, and model-based computational analysis using a Bayesian inference framework for tree-structured data reveals a segregation into heritably fast- and slow-dividing branches. This diversification of division speed is preceded already during the burst phase by variable expression of the interleukin-2 receptor alpha chain. Later it is accompanied by selective expression of memory marker CD62L in slower dividing branches. Taken together, these data demonstrate that T cell clonal expansion is structured into subsequent burst and diversification phases, the latter of which coincides with specification of memory versus effector fate.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Cell Lineage , Animals , Antigens, CD/immunology , Biomarkers , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Division , Mice , Mice, Inbred C57BL
16.
J Infect Dis ; 230(1): e30-e33, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39052732

ABSTRACT

Generation of a stable long-lived plasma cell (LLPC) population is the sine qua non of durable antibody responses after vaccination or infection. We studied 20 individuals with a prior coronavirus disease 2019 infection and characterized the antibody response using bone marrow aspiration and plasma samples. We noted deficient generation of spike-specific LLPCs in the bone marrow after severe acute respiratory syndrome coronavirus 2 infection. Furthermore, while the regression model explained 98% of the observed variance in anti-tetanus immunoglobulin G levels based on LLPC enzyme-linked immunospot assay, we were unable to fit the same model with anti-spike antibodies, again pointing to the lack of LLPC contribution to circulating anti-spike antibodies.


Subject(s)
Antibodies, Viral , Bone Marrow , COVID-19 , Plasma Cells , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , COVID-19/immunology , Plasma Cells/immunology , Spike Glycoprotein, Coronavirus/immunology , Antibodies, Viral/blood , SARS-CoV-2/immunology , Male , Middle Aged , Female , Bone Marrow/virology , Adult , Immunoglobulin G/blood , Aged
17.
FASEB J ; 37(4): e22855, 2023 04.
Article in English | MEDLINE | ID: mdl-36906286

ABSTRACT

Chronic uveitis comprises heterogeneous clinical entities characterized by sustained and recurrent intraocular inflammation that is believed to be driven by autoimmune responses. The management of chronic uveitis is challenging with the limited availability of efficacious treatments, and the underlying mechanisms mediating disease chronicity remain poorly understood as the majority of experimental data are derived from the acute phase of the disease (the first 2-3 weeks post-induction). Herein, we investigated the key cellular mechanisms underlying chronic intraocular inflammation using our recently established murine model of chronic autoimmune uveitis. We demonstrate unique long-lived CD44hi IL-7R+ IL-15R+ CD4+ memory T cells in both retina and secondary lymphoid organs after 3 months postinduction of autoimmune uveitis. These memory T cells functionally exhibit antigen-specific proliferation and activation in response to retinal peptide stimulation in vitro. Critically, these effector-memory T cells are capable of effectively trafficking to the retina and accumulating in the local tissues secreting both IL-17 and IFN-γ upon adoptively transferred, leading to retinal structural and functional damage. Thus, our data reveal the critical uveitogenic functions of memory CD4+ T cells in sustaining chronic intraocular inflammation, suggesting that memory T cells can be a novel and promising therapeutic target for treating chronic uveitis in future translational studies.


Subject(s)
Autoimmune Diseases , Retinal Diseases , Uveitis , Mice , Humans , Animals , Disease Models, Animal , CD4-Positive T-Lymphocytes , Inflammation
18.
Immunity ; 43(4): 634-45, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26488815

ABSTRACT

Natural killer (NK) cells have historically been considered short-lived cytolytic cells that can rapidly respond against pathogens and tumors in an antigen-independent manner and then undergo cell death. Recently, however, NK cells have been shown to possess traits of adaptive immunity and can acquire immunological memory in a manner similar to that of T and B cells. In this review, we discuss evidence of NK cell memory and the mechanisms involved in the generation and survival of these innate lymphocytes.


Subject(s)
Immunologic Memory , Killer Cells, Natural/immunology , Adaptive Immunity , Adoptive Transfer , Animals , Antigens, Viral/immunology , Apoptosis/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/immunology , Dermatitis, Contact/immunology , Haptens/immunology , Homeodomain Proteins/immunology , Homeostasis/immunology , Humans , Inflammation/immunology , Killer Cells, Natural/classification , Killer Cells, Natural/transplantation , Lymphocyte Activation , Lymphocyte Subsets/immunology , Lymphocytes/classification , Lymphocytes/immunology , Mice , Models, Immunological , Peyer's Patches/cytology , Peyer's Patches/immunology , Receptors, Natural Killer Cell/immunology , Virus Diseases/immunology
19.
Immunol Invest ; 53(7): 1092-1101, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38994913

ABSTRACT

INTRODUCTION: Memory CD8+ T cells are essential for long-term immune protection in viral infections, including COVID-19. METHODS: This study examined the responses of CD8+ TEM, TEMRA, and TCM subsets from unvaccinated individuals who had recovered from mild and severe COVID-19 by flow cytometry. RESULTS AND DISCUSSION: The peptides triggered a higher frequency of CD8+ TCM cells in the recovered mild group. CD8+ TCM and TEM cells showed heterogeneity in CD137 expression between evaluated groups. In addition, a predominance of CD137 expression in naïve CD8+ T cells, TCM, and TEM was observed in the mild recovered group when stimulated with peptides. Furthermore, CD8+ TCM and TEM cell subsets from mild recovered volunteers had higher TNF-α expression. In contrast, the expression partner of IFN-γ, IL-10, and IL-17 indicated an antiviral signature by CD8+ TEMRA cells. These findings underscore the distinct functional capabilities of each memory T cell subset in individuals who have recovered from COVID-19 upon re-exposure to SARS-CoV-2 antigens.


Subject(s)
CD8-Positive T-Lymphocytes , COVID-19 , Memory T Cells , SARS-CoV-2 , Tumor Necrosis Factor Receptor Superfamily, Member 9 , Tumor Necrosis Factor-alpha , Humans , COVID-19/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , SARS-CoV-2/immunology , CD8-Positive T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/metabolism , Memory T Cells/immunology , Male , Adult , Middle Aged , Female , Immunologic Memory/immunology
20.
Mol Ther ; 31(2): 535-551, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36068918

ABSTRACT

Immune checkpoint blockade can induce potent and durable responses in patients with highly immunogenic mismatch repair-deficient tumors; however, these drugs are ineffective against immune-cold neuroblastoma tumors. To establish a role for a T cell-based therapy against neuroblastoma, we show that T cell and memory T cell-dependent gene expression are associated with improved survival in high-risk neuroblastoma patients. To stimulate anti-tumor immunity and reproduce this immune phenotype in neuroblastoma tumors, we used CRISPR-Cas9 to knockout MLH1-a crucial molecule in the DNA mismatch repair pathway-to induce mismatch repair deficiency in a poorly immunogenic murine neuroblastoma model. Induced mismatch repair deficiency increased the expression of proinflammatory genes and stimulated T cell infiltration into neuroblastoma tumors. In contrast to adult cancers with induced mismatch repair deficiency, neuroblastoma tumors remained unresponsive to anti-PD1 treatment. However, anti-CTLA4 therapy was highly effective against these tumors. Anti-CTLA4 therapy promoted immune memory and T cell epitope spreading in cured animals. Mechanistically, the effect of anti-CTLA4 therapy against neuroblastoma tumors with induced mismatch repair deficiency is CD4+ T cell dependent, as depletion of these cells abolished the effect. Therefore, a therapeutic strategy involving mismatch repair deficiency-based T cell infiltration of neuroblastoma tumors combined with anti-CTLA4 can serve as a novel T cell-based treatment strategy for neuroblastoma.


Subject(s)
Brain Neoplasms , Colorectal Neoplasms , Neuroblastoma , Mice , Animals , Immunologic Memory , Colorectal Neoplasms/pathology , Neuroblastoma/genetics , Neuroblastoma/therapy
SELECTION OF CITATIONS
SEARCH DETAIL