Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 38
1.
Neuropathol Appl Neurobiol ; 50(3): e12977, 2024 Jun.
Article En | MEDLINE | ID: mdl-38680020

AIM: Leigh syndrome (LS), the most common paediatric presentation of genetic mitochondrial dysfunction, is a multi-system disorder characterised by severe neurologic and metabolic abnormalities. Symmetric, bilateral, progressive necrotizing lesions in the brainstem are defining features of the disease. Patients are often symptom free in early life but typically develop symptoms by about 2 years of age. The mechanisms underlying disease onset and progression in LS remain obscure. Recent studies have shown that the immune system causally drives disease in the Ndufs4(-/-) mouse model of LS: treatment of Ndufs4(-/-) mice with the macrophage-depleting Csf1r inhibitor pexidartinib prevents disease. While the precise mechanisms leading to immune activation and immune factors involved in disease progression have not yet been determined, interferon-gamma (IFNγ) and interferon gamma-induced protein 10 (IP10) were found to be significantly elevated in Ndufs4(-/-) brainstem, implicating these factors in disease. Here, we aimed to explore the role of IFNγ and IP10 in LS. METHODS: To establish the role of IFNγ and IP10 in LS, we generated IFNγ and IP10 deficient Ndufs4(-/-)/Ifng(-/-) and Ndufs4(-/-)/IP10(-/-) double knockout animals, as well as IFNγ and IP10 heterozygous, Ndufs4(-/-)/Ifng(+/-) and Ndufs4(-/-)/IP10(+/-), animals. We monitored disease onset and progression to define the impact of heterozygous or homozygous loss of IFNγ and IP10 in LS. RESULTS: Loss of IP10 does not significantly impact the onset or progression of disease in the Ndufs4(-/-) model. IFNγ loss significantly extends survival and delays disease progression in a gene dosage-dependent manner, though the benefits are modest compared to Csf1r inhibition. CONCLUSIONS: IFNγ contributes to disease onset and progression in LS. Our findings suggest that IFNγ targeting therapies may provide some benefits in genetic mitochondrial disease, but targeting IFNγ alone would likely yield only modest benefits in LS.


Disease Progression , Electron Transport Complex I , Interferon-gamma , Leigh Disease , Animals , Mice , Brain Stem/pathology , Brain Stem/metabolism , Disease Models, Animal , Electron Transport Complex I/genetics , Electron Transport Complex I/deficiency , Interferon-gamma/metabolism , Leigh Disease/pathology , Leigh Disease/genetics , Mice, Inbred C57BL , Mice, Knockout
2.
bioRxiv ; 2023 Sep 01.
Article En | MEDLINE | ID: mdl-37693598

Hydrogels generally have broad utilization in healthcare due to their tunable structures, high water content, and inherent biocompatibility. FDA-approved applications of hydrogels include spinal cord regeneration, skin fillers, and local therapeutic delivery. Drawbacks exist in the clinical hydrogel space, largely pertaining to inconsistent therapeutic exposure, short-lived release windows, and difficulties inserting the polymer into tissue. In this study, we engineered injectable, biocompatible hydrogels that function as a local protein therapeutic depot with a high degree of user-customizability. We showcase a PEG-based hydrogel functionalized with bioorthogonal strain-promoted azide-alkyne cycloaddition (SPAAC) handles for its polymerization and functionalization with a variety of payloads. Small-molecule and protein cargos, including chemokines and antibodies, were site-specifically modified with hydrolysable "azidoesters" of varying hydrophobicity via direct chemical conjugation or sortase-mediated transpeptidation. These hydrolysable esters afforded extended release of payloads linked to our hydrogels beyond diffusion; with timescales spanning days to months dependent on ester hydrophobicity. Injected hydrogels polymerize in situ and remain in tissue over extended periods of time. Hydrogel-delivered protein payloads elicit biological activity after being modified with SPAAC-compatible linkers, as demonstrated by the successful recruitment of murine T-cells to a mouse melanoma model by hydrolytically released murine CXCL10. These results highlight a highly versatile, customizable hydrogel-based delivery system for local delivery of protein therapeutics with payload release profiles appropriate for a variety of clinical needs.

3.
Brain Pathol ; 33(6): e13192, 2023 11.
Article En | MEDLINE | ID: mdl-37552802

Subacute necrotizing encephalopathy, or Leigh syndrome (LS), is the most common pediatric presentation of genetic mitochondrial disease. LS is a multi-system disorder with severe neurologic, metabolic, and musculoskeletal symptoms. The presence of progressive, symmetric, and necrotizing lesions in the brainstem are a defining feature of the disease, and the major cause of morbidity and mortality, but the mechanisms underlying their pathogenesis have been elusive. Recently, we demonstrated that high-dose pexidartinib, a CSF1R inhibitor, prevents LS CNS lesions and systemic disease in the Ndufs4(-/-) mouse model of LS. While the dose-response in this study implicated peripheral immune cells, the immune populations involved have not yet been elucidated. Here, we used a targeted genetic tool, deletion of the colony-stimulating Factor 1 receptor (CSF1R) macrophage super-enhancer FIRE (Csf1rΔFIRE), to specifically deplete microglia and define the role of microglia in the pathogenesis of LS. Homozygosity for the Csf1rΔFIRE allele ablates microglia in both control and Ndufs4(-/-) animals, but onset of CNS lesions and sequalae in the Ndufs4(-/-), including mortality, are only marginally impacted by microglia depletion. The overall development of necrotizing CNS lesions is not altered, though microglia remain absent. Finally, histologic analysis of brainstem lesions provides direct evidence of a causal role for peripheral macrophages in the characteristic CNS lesions. These data demonstrate that peripheral macrophages play a key role in the pathogenesis of disease in the Ndufs4(-/-) model.


Leigh Disease , Mitochondrial Diseases , Humans , Mice , Animals , Child , Leigh Disease/genetics , Leigh Disease/pathology , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Macrophages/pathology , Brain Stem/pathology , Disease Models, Animal
4.
Sports Med Health Sci ; 5(2): 137-145, 2023 Jun.
Article En | MEDLINE | ID: mdl-37424529

High intensity interval training (HIIT) causes oxidative stress and haematological alteration. Present study was aimed to evaluate the effect of 8 weeks' supplementation of vitamin C and E on HIIT induced changes in lipid profile parameters and haematological variables. Hundred six male adolescent players were randomly assigned into five age-matched groups, i.e., Control (no exercise+placebo), HIIT (placebo), HIIT â€‹+ â€‹vitamin-C (1 000 â€‹mg/day), HIIT â€‹+ â€‹vitamin-E 400 IU/day) and combined HIIT â€‹+ â€‹vitamin C and E. Morning and evening sessions (90 â€‹min) of HIIT included 4 phases (15 â€‹min each) with 3 sets (4 â€‹min each). Each 4 â€‹min HIIT set consisted of 2 â€‹min intense sprint workout (90%-95% of heart rate maximum [HRmax]) followed by 1 â€‹min active recovery (60%-70% HRmax) followed by 1 â€‹min of complete rest (1:1 work-rest ratio). Lipid profile parameters, haematological variables, endurance capacity and vertical jump were evaluated by standard protocols. Significant decrease in body weight, fat%, total cholesterol, triglyceride, Total Cholesterol/High Density Lipoprotein-Cholesterol and significant increase in High Density Lipoprotein-Cholesterol, maximal oxygen consumption, vertical jump were observed for all four intervention groups. White blood cell count, red blood cell count, haemoglobin percentage and haematocrit values were significantly decreased while platelet count and platelet-to-leukocyte ratio (PLR) ratio were increased significantly only for HIIT group. Blood level of tocopherol and ascorbic acid was significantly increased (values were within the normal range) in all the respective vitamin supplemented groups. Supplementation of vitamin C and E secures health protection with suppressed haemolysis and improved inflammatory blood variables with enhanced explosive leg strength and lipid profile parameters without any concomitant change in endurance capacity.

5.
Cell Rep ; 39(2): 110632, 2022 04 12.
Article En | MEDLINE | ID: mdl-35417685

Differential interleukin-2 (IL-2) signaling and production are associated with disparate effector and memory fates. Whether the IL-2 signals perceived by CD8 T cells come from autocrine or paracrine sources, the timing of IL-2 signaling and their differential impact on CD8 T cell responses remain unclear. Using distinct models of germline and conditional IL-2 ablation in post-thymic CD8 T cells, this study shows that paracrine IL-2 is sufficient to drive optimal primary expansion, effector and memory differentiation, and metabolic function. In contrast, autocrine IL-2 is uniquely required during primary expansion to program robust secondary expansion potential in memory-fated cells. This study further shows that IL-2 production by antigen-specific CD8 T cells is largely independent of CD4 licensing of dendritic cells (DCs) in inflammatory infections with robust DC activation. These findings bear implications for immunizations and adoptive T cell immunotherapies, where effector and memory functions may be commandeered through IL-2 programming.


Immunologic Memory , Interleukin-2 , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Interleukin-2/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
6.
Virology ; 567: 77-86, 2022 02.
Article En | MEDLINE | ID: mdl-35032866

Type-I interferon (IFN-I) signals exert a critical role in disease progression during viral infections. However, the immunomodulatory mechanisms by which IFN-I dictates disease outcomes remain to be fully defined. Here we report that IFN-I signals mediate thymic atrophy in viral infections, with more severe and prolonged loss of thymic output and unique kinetics and subtypes of IFN-α/ß expression in chronic infection compared to acute infection. Loss of thymic output was linked to inhibition of early stages of thymopoiesis (DN1-DN2 transition, and DN3 proliferation) and pronounced apoptosis during the late DP stage. Notably, infection-associated thymic defects were largely abrogated upon ablation of IFNαßR and partially mitigated in the absence of CD8 T cells, thus implicating direct as well as indirect effects of IFN-I on thymocytes. These findings provide mechanistic underpinnings for immunotherapeutic strategies targeting IFN-1 signals to manipulate disease outcomes during chronic infections and cancers.


Atrophy/virology , Interferon-alpha/immunology , Interferon-beta/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Thymocytes/virology , Thymus Gland/virology , Animals , Atrophy/genetics , Atrophy/immunology , Atrophy/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Chronic Disease , Female , Gene Expression Regulation , Humans , Immunologic Memory , Interferon-alpha/genetics , Interferon-beta/genetics , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymph Nodes/virology , Lymphocyte Depletion , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus/pathogenicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , Signal Transduction/immunology , Single-Cell Analysis , Thymocytes/immunology , Thymocytes/pathology , Thymus Gland/immunology , Thymus Gland/pathology
7.
Front Immunol ; 12: 706150, 2021.
Article En | MEDLINE | ID: mdl-34867942

Robust T cell responses are crucial for effective anti-tumor responses and often dictate patient survival. However, in the context of solid tumors, both endogenous T cell responses and current adoptive T cell therapies are impeded by the immunosuppressive tumor microenvironment (TME). A multitude of inhibitory signals, suppressive immune cells, metabolites, hypoxic conditions and limiting nutrients are believed to render the TME non-conducive to sustaining productive T cell responses. In this study we conducted an in-depth phenotypic and functional comparison of tumor-specific T cells and tumor-nonspecific bystander memory T cells within the same TME. Using two distinct TCR transgenic and solid-tumor models, our data demonstrate that despite exposure to the same cell-extrinsic factors of the TME, the tumor-nonspecific bystander CD8 T cells retain the complete panoply of memory markers, and do not share the same exhaustive phenotype as tumor-reactive T cells. Compared to tumor-specific T cells, bystander memory CD8 T cells in the TME also retain functional effector cytokine production capabilities in response to ex vivo cognate antigenic stimulation. Consistent with these results, bystander memory T cells isolated from tumors showed enhanced recall responses to secondary bacterial challenge in a T cell transplant model. Importantly, the tumor-resident bystander memory cells could also efficiently utilize the available resources within the TME to elaborate in situ recall effector functions following intra-tumoral peptide antigen injection. Additionally, CRISPR-Cas9 gene deletion studies showed that CXCR3 was critical for the trafficking of both tumor antigen-specific and bystander memory T cells to solid tumors. Collectively, these findings that T cells can persist and retain their functionality in distinct solid tumor environments in the absence of cognate antigenic stimulation, support the notion that persistent antigenic signaling is the central driver of T cell exhaustion within the TME. These studies bear implications for programming more efficacious TCR- and CAR-T cells with augmented therapeutic efficacy and longevity through regulation of antigen and chemokine receptors.


Memory T Cells/immunology , Tumor Microenvironment/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Immunophenotyping , Immunotherapy, Adoptive , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Immunological , Neoplasms, Experimental/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, CXCR3/immunology , Receptors, Chimeric Antigen/immunology
8.
Sci Transl Med ; 13(615): eaba6006, 2021 Oct 13.
Article En | MEDLINE | ID: mdl-34644150

Inhibitory signaling in dysfunctional CD8 T cells through the programmed cell death 1 (PD-1) axis is well established in chronic viral infections and cancers. PD-1 is also transiently induced to high concentrations during priming of acute infections and immunizations, yet its impact on the development of long-lived antigen-independent T cell memory remains unclear. In addition to its expected role in restraining clonal effector expansion, here, we show that PD-1 expression on antigen-specific CD8 T cells is required for the development of a durable CD8 T cell memory pool after antigen clearance. Loss of T cell­specific PD-1 signaling led to increased contraction and a defect in antigen-independent renewal of memory CD8 T cells in response to homeostatic cytokine signals, thus resulting in attrition of the memory pool over time. Whereas exhausted CD8 T cells regain function after PD-1 checkpoint blockade during chronic viral infection, the preexisting pool of resting functional bystander memory CD8 T cells established in response to a previously administered immunogen decreased. Metabolically, PD-1 signals were necessary for regulating the critical balance of mTOR-dependent anabolic glycolysis and fatty acid oxidation programs to meet the bioenergetic needs of quiescent CD8 T cell memory. These results define PD-1 as a key metabolic regulator of protective T cell immunity. Furthermore, these results have potential clinical implications for preexisting CD8 T cell memory during PD-1 checkpoint blockade therapy.


Immunologic Memory , Programmed Cell Death 1 Receptor , Animals , CD8-Positive T-Lymphocytes , Mice , Mice, Inbred C57BL , Signal Transduction
9.
Apunts, Med. esport (Internet) ; 56(210)April - June 2021. tab
Article En | IBECS | ID: ibc-214800

Introduction: High-intensity interval training (HIIT) has previously been reported having the effect of training period on altering oxidant status, muscle damage and performance. The present study was aimed to understand and evaluate the adaptive response of 8 weeks HIIT on muscle damage indices, inflammatory markers, oxidative stress variables and physical fitness parameters.MethodsForty young endurance male players [i.e., football (n=20) and field hockey (n=20)] were recruited under two groups i.e., control and HIIT. 8 weeks long 3h/day of sprint-HIIT was intervened for thrice/week. HIIT workouts includes total 4 sets/session (divided into 2 phase×2 sets×2min) of all-out sprint workout (at 90–95% of HRmax with work: rest=1:1). Muscle damage indices (CK and LDH), inflammatory markers (IL-6 and TNF-α), oxidative stress variables (MDA, SOD, GSH and GPx) and physical fitness variables (VO2max, Wpeak and VJ) were assessed via following standard protocols.ResultThe HIIT resulted to significantly(p<0.001) increase BMI (1.1%), LDH (15.0%), CK (14.4%), cortisol (9.4%), IL-6 (15.7%), TNF-α (18.2%), MDA (29.5%), VO2max (13.6%), Wpeak (11.6%), VJ (11.2%) and GPx (0.4%) along with significant (p<0.001) reduction in BF% (7.6%), SOD (11.1%), GSH (10.8%) content of athletes.ConclusionAntioxidant redox imbalance confers inflammatory oxidative stress condition which further leads to muscle damage and that may cause due to HIIT induced temporary hypoxic condition which contrarily induced overtraining effect but with improving performance variables. (AU)


Humans , Male , High-Intensity Interval Training , Antioxidants , Oxidative Stress , Athletes , Muscle Fatigue , Muscle Weakness
10.
J Leukoc Biol ; 110(4): 809-819, 2021 10.
Article En | MEDLINE | ID: mdl-33464639

Vitamin D, a key nutrient/prohormone classically associated with skeletal health, is also an important immunomodulator, with pleotropic effects on innate and adaptive immune cells. Outcomes of several chronic, autoimmune, and infectious diseases are linked to vitamin D. Emergent correlations of vitamin D insufficiency with coronavirus-induced disease 2019 (COVID-19) severity, alongside empirical and clinical evidence of immunoregulation by vitamin D in other pulmonary diseases, have prompted proposals of vitamin D supplementation to curb the COVID-19 public health toll. In this review paper, we engage an immunological lens to discuss potential mechanisms by which vitamin D signals might regulate respiratory disease severity in severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) infections, vis a vis other pulmonary infections. It is proposed that vitamin D signals temper lung inflammatory cascades during SARS-CoV2 infection, and insufficiency of vitamin D causes increased inflammatory cytokine storm, thus leading to exacerbated respiratory disease. Additionally, analogous to studies of reduced cancer incidence, the dosage of vitamin D compounds administered to patients near the upper limit of safety may serve to maximize immune health benefits and mitigate inflammation and disease severity in SARS-CoV2 infections. We further deliberate on the importance of statistically powered clinical correlative and interventional studies, and the need for in-depth basic research into vitamin D-dependent host determinants of respiratory disease severity.


COVID-19/complications , Cytokine Release Syndrome/pathology , Inflammation/pathology , SARS-CoV-2/isolation & purification , Vitamin D Deficiency/physiopathology , COVID-19/immunology , COVID-19/virology , Cytokine Release Syndrome/etiology , Humans , Inflammation/etiology , Severity of Illness Index , Vitamin D Deficiency/immunology
11.
J Immunol ; 201(12): 3641-3650, 2018 12 15.
Article En | MEDLINE | ID: mdl-30455400

IL-1, generally considered an amplifier of adaptive immune responses, has been proposed for use as adjuvant during immunization with weak immunogens. However, its effects on memory T cell function remain largely undefined. Using the murine model of acute viral infection, in this paper, we show that in addition to augmenting the size of the Ag-specific pool, IL-1 signals act directly on CD8 T cells to promote the quality of effector and memory responses. Ablation of IL-1R1 or MyD88 signaling in T cells led to functional impairment; both the ability to produce multiple cytokines on a per cell basis (polyfunctionality) and the potential for recall proliferation in response to antigenic restimulation were compromised. IL-1 supplementation during priming augmented the expansion of Ag-specific CD8 T cells through the MyD88-IRAK1/4 axis, resulting in a larger memory pool capable of robust secondary expansion in response to rechallange. Together, these findings demonstrate a critical role of the IL-1-MyD88 axis in programming the quantity and quality of memory CD8 T cell responses and support the notion that IL-1 supplementation may be exploited to enhance adoptive T cell therapies against cancers and chronic infections.


CD8-Positive T-Lymphocytes/physiology , Interleukin-1/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/physiology , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cellular Reprogramming , Cellular Reprogramming Techniques , Humans , Immunologic Memory , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Differentiation Factor 88/metabolism , Receptors, Interleukin-1 Type I/genetics , Signal Transduction
12.
Front Immunol ; 9: 2987, 2018.
Article En | MEDLINE | ID: mdl-30619342

Interleukin-2 (IL-2) regulates key aspects of CD8 T cell biology-signaling through distinct pathways IL-2 triggers critical metabolic and transcriptional changes that lead to a spectrum of physiological outcomes such as cell survival, proliferation, and effector differentiation. In addition to driving effector differentiation, IL-2 signals are also critical for formation of long-lived CD8 T cell memory. This review discusses a model of rheostatic control of CD8 T cell effector and memory differentiation by IL-2, wherein the timing, duration, dose, and source of IL-2 signals are considered in fine-tuning the balance of key transcriptional regulators of cell fate.


Cell Differentiation/immunology , Immunologic Memory , Interleukin-2/metabolism , Lymphocyte Activation , T-Lymphocytes, Cytotoxic/physiology , Animals , Humans , Interleukin-2/immunology , Mice , Signal Transduction/immunology
13.
PLoS One ; 12(12): e0190384, 2017.
Article En | MEDLINE | ID: mdl-29284034

Progress towards a safe and effective vaccine for the prevention of tularemia has been hindered by a lack of knowledge regarding the correlates of protective adaptive immunity and a lack of tools to generate this knowledge. CD8+ T cells are essential for protective immunity against virulent strains of Francisella tularensis, but to-date, it has not been possible to study these cells in an antigen-specific manner. Here, we report the development of a tool for expression of the model antigen ovalbumin (OVA) in F. tularensis, which allows for the study of CD8+ T cell responses to the bacterium. We demonstrate that in response to intranasal infection with the F. tularensis Live Vaccine Strain, adoptively transferred OVA-specific CD8+ T cells expand after the first week and produce IFN-γ but not IL-17. Effector and central memory subsets develop with disparate kinetics in the lungs, draining lymph node and spleen. Notably, OVA-specific cells are poorly retained in the lungs after clearance of infection. We also show that intranasal vaccination leads to more antigen-specific CD8+ T cells in the lung-draining lymph node compared to scarification vaccination, but that an intranasal booster overcomes this difference. Together, our data show that this novel tool can be used to study multiple aspects of the CD8+ T cell response to F. tularensis. Use of this tool will enhance our understanding of immunity to this deadly pathogen.


Bacterial Vaccines/immunology , CD8-Positive T-Lymphocytes/immunology , Francisella tularensis/immunology , Vaccines, Attenuated/immunology , Animals , Mice , Mice, Inbred C57BL , Ovalbumin/immunology
14.
PLoS One ; 11(9): e0162674, 2016.
Article En | MEDLINE | ID: mdl-27627450

MicroRNAs constitute a major post-transcriptional mechanism for controlling protein expression, and are emerging as key regulators during T cell development and function. Recent reports of augmented CD8 T cell activation and effector differentiation, and aberrant migratory properties upon ablation of Dicer/miRNAs in naïve cells have established a regulatory role of miRNAs during priming. Whether miRNAs continue to exert similar functions or are dispensable during later stages of CD8 T cell expansion and memory differentiation remains unclear. Here, we report a critical role of Dicer/miRNAs in regulating the balance of long-lived memory and short-lived terminal effector fates during the post-priming stages when CD8 T cells undergo clonal expansion to generate a large cytotoxic T lymphocyte (CTL) pool and subsequently differentiate into a quiescent memory state. Conditional ablation of Dicer/miRNAs in early effector CD8 T cells following optimal activation and expression of granzyme B, using unique dicerfl/fl gzmb-cre mice, led to a strikingly diminished peak effector size relative to wild-type antigen-specific cells in the same infectious milieu. Diminished expansion of Dicer-ablated CD8 T cells was associated with lack of sustained antigen-driven proliferation and reduced accumulation of short-lived effector cells. Additionally, Dicer-ablated CD8 T cells exhibited more pronounced contraction after pathogen clearance and comprised a significantly smaller proportion of the memory pool, despite significantly higher proportions of CD127Hi memory precursors at the effector peak. Combined with previous reports of dynamic changes in miRNA expression as CD8 T cells differentiate from naïve to effector and memory states, these findings support distinct stage-specific roles of miRNA-dependent gene regulation during CD8 T cell differentiation.


CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/physiology , Ribonuclease III/physiology , Animals , CD8-Positive T-Lymphocytes/physiology , Cell Lineage/immunology , Cell Lineage/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/physiology
15.
J Virol ; 90(19): 8934-46, 2016 10 01.
Article En | MEDLINE | ID: mdl-27466420

UNLABELLED: PD-1 is an inhibitory receptor that has a major role in T cell dysfunction during chronic infections and cancer. While demethylation of the PD-1 promoter DNA is observed in exhausted T cells isolated from chronically infected individuals, little is known about when this stable demethylation of PD-1 promoter DNA is programmed during the course of a chronic infection. To assess if PD-1 promoter DNA demethylation is impacted by prolonged stimulation during effector phase of chronic infection, we adoptively transferred virus-specific day 8 effector CD8 T cells from mice infected with lymphocytic choriomeningitis virus (LCMV) clone 13 into recipient mice that had cleared an acute infection. We observed that LCMV-specific CD8 T cells from chronically infected mice maintained their surface expression of PD-1 even after transfer into acute immune mice until day 45 posttransfer. Interestingly, the PD-1 transcriptional regulatory region continued to remain unmethylated in these donor CD8 T cells generated from a chronic infection. The observed maintenance of PD-1 surface expression and the demethylated PD-1 promoter were not a result of residual antigen in the recipient mice, because similar results were seen when chronic infection-induced effector cells were transferred into mice infected with a variant strain of LCMV (LCMV V35A) bearing a mutation in the cognate major histocompatibility complex class I (MHC-I) epitope that is recognized by the donor CD8 T cells. Importantly, the maintenance of PD-1 promoter demethylation in memory CD8 T cells was coupled with impaired clonal expansion and higher PD-1 re-expression upon secondary challenge. These data show that the imprinting of the epigenetic program of the inhibitory receptor PD-1 occurs during the effector phase of chronic viral infection. IMPORTANCE: Since PD-1 is a major inhibitory receptor regulating T cell dysfunction during chronic viral infection and cancers, a better understanding of the mechanisms that regulate PD-1 expression is important. In this work, we demonstrate that the PD-1 epigenetic program in antigen-specific CD8 T cells is fixed during the priming phase of chronic infection.


CD8-Positive T-Lymphocytes/physiology , DNA Methylation , Gene Expression Regulation , Lymphocytic choriomeningitis virus/immunology , Programmed Cell Death 1 Receptor/biosynthesis , Programmed Cell Death 1 Receptor/genetics , Promoter Regions, Genetic , Adoptive Transfer , CD8-Positive T-Lymphocytes/metabolism , Chronic Disease , Epigenesis, Genetic , Lymphocytic Choriomeningitis/immunology
16.
Immunity ; 45(1): 10-2, 2016 07 19.
Article En | MEDLINE | ID: mdl-27438762

Highly active killer T cells mediate a stable standoff during controlled persistent infections. In this issue of Immunity, Robey and colleagues describe a unique antigen-addicted T cell population bearing characteristics of both effector and memory CD8(+) T cells that provides a continuous supply of potent killer T cells to curb Toxoplasma gondii growth during latency.


CD8-Positive T-Lymphocytes/immunology , Toxoplasma/immunology , Humans , T-Lymphocytes, Cytotoxic/immunology
17.
Crit Rev Clin Lab Sci ; 53(2): 132-45, 2016.
Article En | MEDLINE | ID: mdl-26479950

The discovery of vitamin D receptor (VDR) expression in immune cells has opened up a new area of research into immunoregulation by vitamin D, a niche that is distinct from its classical role in skeletal health. Today, about three decades since this discovery, numerous cellular and molecular targets of vitamin D in the immune system have been delineated. Moreover, strong clinical associations between vitamin D status and the incidence/severity of many immune-regulated disorders (e.g. infectious diseases, cancers and autoimmunity) have prompted the idea of using vitamin D supplementation to manipulate disease outcome. While much is known about the effects of vitamin D on innate immune responses and helper T (T(H)) cell immunity, there has been relatively limited progress on the frontier of cytotoxic T lymphocyte (CTL) immunity--an arm of host cellular adaptive immunity that is crucial for the control of such intracellular pathogens as human immunodeficiency virus (HIV), tuberculosis (TB), malaria, and hepatitis C virus (HCV). In this review, we discuss the strong historical and clinical link between vitamin D and infectious diseases that involves cytotoxic T lymphocyte (CTL) immunity, present our current understanding as well as critical knowledge gaps in the realm of vitamin D regulation of host CTL responses, and highlight potential regulatory connections between vitamin D and effector and memory CD8 T cell differentiation events during infections.


Communicable Diseases/immunology , Immunity , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Vitamin D/metabolism , Animals , Dietary Supplements , Humans , Neoplasms/pathology
18.
Immunity ; 42(6): 1116-29, 2015 Jun 16.
Article En | MEDLINE | ID: mdl-26084026

Immune memory cells are poised to rapidly expand and elaborate effector functions upon reinfection yet exist in a functionally quiescent state. The paradigm is that memory T cells remain inactive due to lack of T cell receptor (TCR) stimuli. Here, we report that regulatory T (Treg) cells orchestrate memory T cell quiescence by suppressing effector and proliferation programs through inhibitory receptor, cytotoxic-T-lymphocyte-associated protein-4 (CTLA-4). Loss of Treg cells resulted in activation of genome-wide transcriptional programs characteristic of effector T cells and drove transitioning as well as established memory CD8(+) T cells toward terminally differentiated KLRG-1(hi)IL-7Rα(lo)GzmB(hi) phenotype, with compromised metabolic fitness, longevity, polyfunctionality, and protective efficacy. CTLA-4 functionally replaced Treg cells in trans to rescue memory T cell defects and restore homeostasis. These studies present the CTLA-4-CD28-CD80/CD86 axis as a potential target to accelerate vaccine-induced immunity and improve T cell memory quality in current cancer immunotherapies proposing transient Treg cell ablation.


CD28 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/metabolism , Immunologic Memory , T-Lymphocytes, Regulatory/immunology , Abatacept , Animals , B7 Antigens/metabolism , Cells, Cultured , Cytotoxicity, Immunologic/drug effects , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Granzymes/metabolism , Homeostasis/drug effects , Immunoconjugates/administration & dosage , Immunologic Memory/drug effects , Immunologic Memory/genetics , Lectins, C-Type , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Targeted Therapy , Receptors, Immunologic/metabolism , Receptors, Interleukin-7/metabolism , Signal Transduction/genetics
19.
J Cell Biochem ; 116(8): 1500-12, 2015 Aug.
Article En | MEDLINE | ID: mdl-25694395

It is now well known that in the mammalian body vitamin D is converted by successive hydroxylations to 1,25-dihydroxyvitamin D (1,25D), a steroid-like hormone with pleiotropic properties. These include important contributions to the control of cell proliferation, survival and differentiation, as well as the regulation of immune responses in disease. Here, we present recent advances in current understanding of the role of 1,25D in myelopoiesis and lymphopoiesis, and the potential of 1,25D and analogs (vitamin D derivatives; VDDs) for the control of hematopoietic malignancies. The reasons for the unimpressive results of most clinical studies of the therapeutic effects of VDDs in leukemia and related diseases may include the lack of a precise rationale for the conduct of these studies. Further, clinical trials to date have generally used extremely heterogeneous patient populations and, in many cases, small numbers of patients, generally without controls. Although low calcemic VDDs have been used and combined with agents that can increase the leukemia cell killing or differentiation effects in acute leukemias, the sequencing of agents used for combination therapy should to be more clearly delineated. Most importantly, it is recommended that in future clinical trials the rationale for the basis of the enhancing action of drug combinations should be clearly articulated and the effects on anticancer immunity should also be evaluated.


Hematopoiesis/drug effects , Leukemia/drug therapy , Vitamin D/administration & dosage , Vitamins/administration & dosage , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Differentiation/drug effects , Clinical Trials as Topic , Humans , Leukemia/pathology , Vitamin D/analogs & derivatives , Vitamin D/pharmacology , Vitamins/pharmacology
20.
Cell Mol Immunol ; 12(4): 400-8, 2015 Jul.
Article En | MEDLINE | ID: mdl-25066419

Early after priming, effector CD8 T cells are distinguished into memory precursor and short-lived effector cell subsets (MPECs and SLECs). Here, we delineated a distinct in vivo heterogeneity in killer cell lectin-like receptor G1 (KLRG-1) expression, which was strongly associated with diverse MPEC and SLEC fates. These in vivo MPECs and SLECs expressed equivalent levels of cytotoxic molecules and effector cytokines. Using a unique in vivo degranulation assay, we found that the MPECs and SLECs similarly encountered infected target cells and elaborated equivalent levels of cytotoxicity in vivo. These data provide direct in vivo evidence that memory-fated cells pass through a robust effector phase. Additionally, the preferential localization of the MPECs in the lymph nodes, where a lesser degree of cytotoxicity was elaborated, suggests that the MPECs may be protected from excessive stimulation and terminal differentiation by virtue of their differential tissue localization. These data provide novel mechanistic insights into the linear decreasing potential model of memory differentiation.


CD8-Positive T-Lymphocytes/immunology , Cell Degranulation/immunology , Cell Differentiation/immunology , Immunologic Memory/physiology , Models, Immunological , Precursor Cells, T-Lymphoid/immunology , T-Lymphocyte Subsets/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , Lectins, C-Type , Mice , Precursor Cells, T-Lymphoid/cytology , Receptors, Immunologic/immunology , T-Lymphocyte Subsets/cytology
...