Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 9 de 9
1.
Article En | MEDLINE | ID: mdl-38074985

For effective adaptive immunity, T lymphocytes must rapidly expand and contract in an antigen-specific manner to effectively control invading pathogens and preserve immunological memory, without sustaining excessive collateral damage to host tissues. Starting from initial antigen encounter, carefully calibrated programmed cell death pathways are critical for maintaining homeostasis over distinct phases of the T cell response. Restimulation-induced cell death (RICD), a self-regulatory apoptosis pathway triggered by re-engagement of the T cell receptor (TCR), is particularly important for constraining effector T cell expansion to preclude overt immunopathology; indeed, genetic disorders affecting key molecules involved in RICD execution can manifest in excessive lymphoproliferation, malignancy, and autoimmunity. Herein we review our current knowledge of how RICD sensitivity is ultimately regulated over the course of an immune response, including recent revelations on molecules that tune RICD by enforcing resistance or promoting susceptibility in expanding versus mature effector T cells, respectively. Detailed dissection of the molecular and temporal control of RICD also illuminates novel therapeutic strategies for correcting abnormal T cell responses noted in various immune disorders by ultimately tuning RICD sensitivity.

2.
Sci Rep ; 13(1): 389, 2023 01 08.
Article En | MEDLINE | ID: mdl-36617594

The novel coronavirus SARS-CoV-2 continues to cause death and disease throughout the world, underscoring the necessity of understanding the virus and host immune response. From the start of the pandemic, a prominent pattern of central nervous system (CNS) pathologies, including demyelination, has emerged, suggesting an underlying mechanism of viral mimicry to CNS proteins. We hypothesized that immunodominant epitopes of SARS-CoV-2 share homology with proteins associated with multiple sclerosis (MS). Using PEPMatch, a newly developed bioinformatics package which predicts peptide similarity within specific amino acid mismatching parameters consistent with published MHC binding capacity, we discovered that nucleocapsid protein shares significant overlap with 22 MS-associated proteins, including myelin proteolipid protein (PLP). Further computational evaluation demonstrated that this overlap may have critical implications for T cell responses in MS patients and is likely unique to SARS-CoV-2 among the major human coronaviruses. Our findings substantiate the hypothesis of viral molecular mimicry in the pathogenesis of MS and warrant further experimental exploration.


COVID-19 , Multiple Sclerosis , Humans , SARS-CoV-2 , T-Lymphocytes , Nucleocapsid Proteins , Nucleocapsid
3.
Bio Protoc ; 12(4): e4326, 2022 Feb 20.
Article En | MEDLINE | ID: mdl-35340300

When the body mounts an immune response against a foreign pathogen, the adaptive arm of the immune system relies upon clonal expansion of antigen-specific T cell populations to exercise acquired effector and cytotoxic functions to clear it. However, T cell expansion must be modulated to effectively combat the perceived threat without inducing excessive collateral damage to host tissues. Restimulation-induced cell death (RICD) is an apoptotic program triggered in activated T cells when an abundance of antigen and IL-2 are present, imposing a negative feedback mechanism that constrains the growing T cell population. This autoregulatory process can be detected via increases in caspase activation, Annexin V binding, and loss of mitochondrial membrane potential. However, simple changes in T cell viability through flow cytometric analysis can reliably measure RICD sensitivity in response to T-cell receptor (TCR) restimulation. This protocol describes the in vitro polyclonal activation, expansion, and restimulation of human primary T cells isolated from donor peripheral blood mononuclear cells (PBMC). This simple procedure allows for accurate quantification of RICD via flow cytometry. We also describe strategies for interrogating the role of specific proteins and pathways that may alter RICD sensitivity. This straightforward protocol provides a quick and dependable tool to track RICD sensitivity in culture over time while probing critical factors that control the magnitude and longevity of an adaptive immune response. Graphic abstract: In-vitro simulation of restimulation-induced cell death in activated human T cells.

4.
J Infect Dis ; 224(12): 2010-2019, 2021 12 15.
Article En | MEDLINE | ID: mdl-34673956

BACKGROUND: Characterizing the longevity and quality of cellular immune responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) enhances understanding of coronavirus disease 2019 (COVID-19) immunity that influences clinical outcomes. Prior studies suggest SARS-CoV-2-specific T cells are present in peripheral blood 10 months after infection. Analysis of the function, durability, and diversity of cellular response long after natural infection, over a range of ages and disease phenotypes, is needed to identify preventative and therapeutic interventions. METHODS: We identified participants in our multisite longitudinal, prospective cohort study 12 months after SARS-CoV-2 infection representing a range of disease severity. We investigated function, phenotypes, and frequency of T cells specific for SARS-CoV-2 using intracellular cytokine staining and spectral flow cytometry, and compared magnitude of SARS-CoV-2-specific antibodies. RESULTS: SARS-CoV-2-specific antibodies and T cells were detected 12 months postinfection. Severe acute illness was associated with higher frequencies of SARS-CoV-2-specific CD4 T cells and antibodies at 12 months. In contrast, polyfunctional and cytotoxic T cells responsive to SARS-CoV-2 were identified in participants over a wide spectrum of disease severity. CONCLUSIONS: SARS-CoV-2 infection induces polyfunctional memory T cells detectable at 12 months postinfection, with higher frequency noted in those who experienced severe disease.


COVID-19/immunology , COVID-19/virology , Immunologic Memory , Memory T Cells , SARS-CoV-2/immunology , T-Lymphocyte Subsets/immunology , Adult , Antibodies, Viral , Antigens, Viral , Biomarkers , COVID-19/diagnosis , COVID-19/epidemiology , Female , Humans , Immunity, Cellular , Longitudinal Studies , Male , Middle Aged , Prospective Studies , Severity of Illness Index , T-Lymphocyte Subsets/metabolism , Time Factors
5.
Cell Death Dis ; 12(4): 400, 2021 04 14.
Article En | MEDLINE | ID: mdl-33854046

Immune homeostasis depends upon effective clearance of pathogens while simultaneously preventing autoimmunity and immunopathology in the host. Restimulation-induced cell death (RICD) is one such mechanism where by activated T cells receive subsequent antigenic stimulation, reach a critical signal threshold through the T cell receptor (TCR), and commit to apoptosis. Many details of this process remain unclear, including the role of co-stimulatory and co-inhibitory proteins that influence the TCR signaling cascade. Here we characterize the role of T cell immunoglobulin and mucin domain containing 3 (TIM-3) in RICD regulation. TIM-3 protected newly activated CD8+ effector T cells from premature RICD during clonal expansion. Surprisingly, however, we found that TIM-3 potentiated RICD in late-stage effector T cells. The presence of TIM-3 increased proximal TCR signaling and proapoptotic protein expression in late-stage effector T cells, with no consistent signaling effects noted in newly activated cells with or without TIM-3. To better explain these differences in TIM-3 function as T cells aged, we characterized the temporal pattern of TIM-3 expression in effector T cells. We found that TIM-3 was expressed on the surface of newly activated effector T cells, but remained largely intracellular in late-stage effector cells. Consistent with this, TIM-3 required a ligand to prevent early RICD, whereas ligand manipulation had no effects at later stages. Of the known TIM-3 ligands, carcinoembryonic antigen-related cell adhesion molecule (CEACAM1) showed the greatest difference in surface expression over time and also protected newly activated cells from premature RICD, with no measurable effects in late-stage effectors. Indeed, CEACAM1 enabled TIM-3 surface expression on T cells, implying a co-dependency for these proteins in protecting expanding T cells from premature RICD. Our findings suggest that co-signaling proteins like TIM-3 and CEACAM1 can alter RICD sensitivity at different stages of the effector T cell response, with important implications for checkpoint blockade therapy.


Antigens, CD/metabolism , Apoptosis/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Adhesion Molecules/metabolism , Hepatitis A Virus Cellular Receptor 2/metabolism , Antigens, CD/immunology , Cell Adhesion Molecules/immunology , Humans , Lymphocyte Activation/immunology , Membrane Proteins/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism
6.
Nat Immunol ; 22(2): 128-139, 2021 02.
Article En | MEDLINE | ID: mdl-33398182

Complement hyperactivation, angiopathic thrombosis and protein-losing enteropathy (CHAPLE disease) is a lethal disease caused by genetic loss of the complement regulatory protein CD55, leading to overactivation of complement and innate immunity together with immunodeficiency due to immunoglobulin wasting in the intestine. We report in vivo human data accumulated using the complement C5 inhibitor eculizumab for the medical treatment of patients with CHAPLE disease. We observed cessation of gastrointestinal pathology together with restoration of normal immunity and metabolism. We found that patients rapidly renormalized immunoglobulin concentrations and other serum proteins as revealed by aptamer profiling, re-established a healthy gut microbiome, discontinued immunoglobulin replacement and other treatments and exhibited catch-up growth. Thus, we show that blockade of C5 by eculizumab effectively re-establishes regulation of the innate immune complement system to substantially reduce the pathophysiological manifestations of CD55 deficiency in humans.


Antibodies, Monoclonal, Humanized/therapeutic use , Complement Activation/drug effects , Complement C5/antagonists & inhibitors , Complement Inactivating Agents/therapeutic use , Energy Metabolism/drug effects , Hypoproteinemia/drug therapy , Immunity, Innate/drug effects , Protein-Losing Enteropathies/drug therapy , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , Biomarkers/blood , CD55 Antigens/deficiency , CD55 Antigens/genetics , Complement C5/metabolism , Complement Inactivating Agents/adverse effects , Complement Inactivating Agents/pharmacokinetics , Genetic Predisposition to Disease , Humans , Hypoproteinemia/genetics , Hypoproteinemia/immunology , Hypoproteinemia/metabolism , Mutation , Phenotype , Protein-Losing Enteropathies/genetics , Protein-Losing Enteropathies/immunology , Protein-Losing Enteropathies/metabolism , Treatment Outcome
7.
Cell Mol Immunol ; 18(1): 194-205, 2021 01.
Article En | MEDLINE | ID: mdl-31659245

The adaptive immune response relies on specific apoptotic programs to maintain homeostasis. Conventional effector T cell (Tcon) expansion is constrained by both forkhead box P3 (FOXP3)+-regulatory T cells (Tregs) and restimulation-induced cell death (RICD), a propriocidal apoptosis pathway triggered by repeated stimulation through the T-cell receptor (TCR). Constitutive FOXP3 expression protects Tregs from RICD by suppressing SLAM-associated protein (SAP), a key adaptor protein that amplifies TCR signaling strength. The role of transient FOXP3 induction in activated human CD4 and CD8 Tcons remains unresolved, but its expression is inversely correlated with acquired RICD sensitivity. Here, we describe a novel role for FOXP3 in protecting human Tcons from premature RICD during expansion. Unlike FOXP3-mediated protection from RICD in Tregs, FOXP3 protects Tcons through a distinct mechanism requiring de novo transcription that does not require SAP suppression. Transcriptome profiling and functional analyses of expanding Tcons revealed that FOXP3 enhances expression of the SLAM family receptor CD48, which in turn sustains basal autophagy and suppresses pro-apoptotic p53 signaling. Both CD48 and FOXP3 expression reduced p53 accumulation upon TCR restimulation. Furthermore, silencing FOXP3 expression or blocking CD48 decreased the mitochondrial membrane potential in expanding Tcons with a concomitant reduction in basal autophagy. Our findings suggest that FOXP3 governs a distinct transcriptional program in early-stage effector Tcons that maintains RICD resistance via CD48-dependent protective autophagy and p53 suppression.


CD48 Antigen/metabolism , Cell Death , Forkhead Transcription Factors/metabolism , Lymphocyte Activation , Receptors, Antigen, T-Cell/metabolism , Signaling Lymphocytic Activation Molecule Associated Protein/metabolism , T-Lymphocytes, Regulatory/immunology , Apoptosis , Autophagy , CD48 Antigen/genetics , Forkhead Transcription Factors/genetics , Humans , Receptors, Antigen, T-Cell/genetics , Signal Transduction , Signaling Lymphocytic Activation Molecule Associated Protein/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
8.
Emerg Infect Dis ; 23(8): 1397-1399, 2017 08.
Article En | MEDLINE | ID: mdl-28726612

Baylisascaris procyonis (raccoon roundworm) infection is common in raccoons and can cause devastating pathology in other animals, including humans. Limited information is available on the frequency of asymptomatic human infection. We tested 150 adults from California, USA, for B. procyonis antibodies; 11 were seropositive, suggesting that subclinical infection does occur.


Ascaridida Infections/epidemiology , Ascaridida/immunology , Animals , Ascaridida Infections/history , Ascaridida Infections/immunology , California/epidemiology , Feces/parasitology , Female , History, 21st Century , Humans , Male , Prevalence , Raccoons , Seroepidemiologic Studies , Zoonoses
9.
Environ Toxicol Pharmacol ; 50: 167-174, 2017 Mar.
Article En | MEDLINE | ID: mdl-28214749

Many chemicals have been used to increase the safety of consumer products by reducing their flammability and risk for ignition. Recent focus on brominated flame retardants, such as polybrominated diphenyl ethers (PBDEs) has shown them to contribute to neurobehavioral deficits in children, including learning and memory. As the manufacture and use of PBDEs have been reduced, replacement chemicals, such as hexabromocyclododecane (HBCDD) have been substituted. Our current study evaluated the neurotoxicity of HBCDD, concentrating on dopaminergic innervation to the hippocampus. Using an in vivo model, we exposed male mice to HBCDD and then assessed alterations to the dopamine synapse 6 weeks later. These exposures elicited significant reductions in presynaptic dopaminergic proteins, including TH, COMT, MAO-B, DAT, VMAT2, and alpha-synuclein. In contrast, postsynaptic dopamine receptors were not impaired. These findings suggest that the mesohippocampal dopamine circuit is vulnerable to HBCDD and the dopamine terminal may be a selective target for alteration.


Dopaminergic Neurons/drug effects , Flame Retardants/toxicity , Hippocampus/drug effects , Hydrocarbons, Brominated/toxicity , Animals , Gene Expression Regulation/drug effects , Hippocampus/cytology , Male , Mice , Signal Transduction/drug effects , Synaptic Potentials/drug effects , alpha-Synuclein/metabolism
...