Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Front Immunol ; 12: 766272, 2021.
Article in English | MEDLINE | ID: mdl-34912339

ABSTRACT

Bacterial infections are a major cause of morbidity and mortality in chronic lymphocytic leukemia (CLL), and infection risk increases in patients treated with the Bruton's tyrosine kinase (Btk) inhibitor, ibrutinib. Btk and related kinases (like Tec) are expressed in non-leukemic hematopoietic cells and can be targeted by ibrutinib. In platelets, ibrutinib therapy is associated with bleeding complications mostly due to off-target effects. But the ability of platelets to respond to bacteria in CLL, and the potential impact of ibrutinib on platelet innate immune functions remain unknown. FcγRIIA is a tyrosine kinase-dependent receptor critical for platelet activation in response to IgG-coated pathogens. Crosslinking of this receptor with monoclonal antibodies causes downstream activation of Btk and Tec in platelets, however, this has not been investigated in response to bacteria. We asked whether ibrutinib impacts on FcγRIIA-mediated activation of platelets derived from CLL patients and healthy donors after exposure to Staphylococcus aureus Newman and Escherichia coli RS218. Platelet aggregation, α-granule secretion and integrin αIIbß3-dependent scavenging of bacteria were detected in CLL platelets but impaired in platelets from ibrutinib-treated patients and in healthy donor-derived platelets exposed to ibrutinib in vitro. While levels of surface FcγRIIA remained unaffected, CLL platelets had reduced expression of integrin αIIbß3 and GPVI compared to controls regardless of therapy. In respect of intracellular signaling, bacteria induced Btk and Tec phosphorylation in both CLL and control platelets that was inhibited by ibrutinib. To address if Btk is essential for platelet activation in response to bacteria, platelets derived from X-linked agammaglobulinemia patients (lacking functional Btk) were exposed to S. aureus Newman and E. coli RS218, and FcγRIIA-dependent aggregation was observed. Our data suggest that ibrutinib impairment of FcγRIIA-mediated platelet activation by bacteria results from a combination of Btk and Tec inhibition, although off-target effects on additional kinases cannot be discarded. This is potentially relevant to control infection-risk in CLL patients and, thus, future studies should carefully evaluate the effects of CLL therapies, including Btk inhibitors with higher specificity for Btk, on platelet-mediated immune functions.


Subject(s)
Adenine/analogs & derivatives , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Blood Platelets/drug effects , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Piperidines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Receptors, IgG/immunology , Adenine/pharmacology , Adenine/therapeutic use , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Blood Platelets/immunology , Escherichia coli , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/blood , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Male , Middle Aged , Piperidines/pharmacology , Platelet Activation/drug effects , Platelet Aggregation/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrazines/pharmacology , Staphylococcus aureus
2.
Front Immunol ; 12: 707830, 2021.
Article in English | MEDLINE | ID: mdl-34484207

ABSTRACT

The impact of biological sex on T-cell immunity to Cytomegalovirus (CMV) has not been investigated in detail with only one published study comparing CMV-specific T-cell responses in men and women. Many studies, however, have shown an association between CMV infection and immunosenescence, with broad effects on peripheral blood lymphocyte subsets as well as the T and B-cell repertoires. Here, we provide a detailed analysis of CMV-specific T-cell responses in (n=94) CMV+ older people, including 47 women and 47 men aged between 60 and 93 years. We explore sex differences with respect to 16 different CMV proteins arranged in 14 peptide pools (overlapping peptides). Following ex vivo stimulation, CD4 and CD8 T-cells producing IFN-γ, TNF, and IL-2 were enumerated by flow-cytometry (intracellular cytokine staining). T-cell responses were evaluated in terms of each cytokine separately or in terms of cytokines produced simultaneously (polyfunctionality). Surface memory phenotype and CD3 downmodulation were assessed in parallel. The polyfunctionality index and a memory subset differentiation score were used to identify associations between response size, cytokine production, polyfunctionality, and memory subset distribution. While no significant sex differences were found with respect to overall CMV target protein selection, the T-cell response in men appeared more focused and accompanied by a more prominent accumulation of CMV-specific memory CD4 and CD8 T-cells. T-cell polyfunctionality and differentiation were similar in the sexes, however, CMV-specific T-cells in men produced more pro-inflammatory cytokines. Particularly, TNF production by CD4 T-cells was stronger in men than in women. Also, compared with women, men had larger responses to CMV proteins with immediate-early/early kinetics than women, which might have been driven by CMV reactivation. In conclusion, the CMV-specific T-cell response in men was larger and more pro-inflammatory than in women. Our findings may help explain sex differences in CMV-associated pathologies.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Immunosenescence/immunology , Sex Characteristics , Aged , Aged, 80 and over , Antigens, Viral/immunology , Cytomegalovirus/immunology , Female , Humans , Male , Middle Aged
3.
Free Radic Biol Med ; 169: 248-257, 2021 06.
Article in English | MEDLINE | ID: mdl-33862160

ABSTRACT

Many studies reported that redox enzymes, particularly thioredoxin and peroxiredoxin, can be released by cells and act as soluble mediators in immunity. Recently, it became clear that peroxiredoxins can be secreted via the exosome-release route, yet it remains unclear how this exactly happens and why. This review will first introduce briefly the possible redox states of protein cysteines and the role of redox enzymes in their regulation. We will then discuss the studies on the extracellular forms of some of these enzymes, their association with exosomes/extracellular vesicles and with exosome micro-RNAs (miRNAs)/mRNAs involved in oxidative processes, relevant in infection and inflammation.


Subject(s)
Exosomes , Extracellular Vesicles , MicroRNAs , Exosomes/metabolism , Extracellular Vesicles/metabolism , Humans , Inflammation/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Oxidation-Reduction
4.
Theranostics ; 11(12): 5728-5741, 2021.
Article in English | MEDLINE | ID: mdl-33897878

ABSTRACT

Human Cytomegalovirus (CMV) infection is associated with atherosclerosis, higher cardiovascular disease (CVD) risk, and an increase in memory T-cells (Tmem). T-cells have also been implicated in CVD, independently of CMV infection. To better understand the CMV-associated CVD risk, we examined the association between CMV (IgG) serostatus and central aortic (carotid-to-femoral) pulse wave velocity (cfPWV), an early, independent predictor of CVD. We also investigated if such an association might be reflected by the distribution of Tmem and/or other T-cell subsets. Methods: Healthy older volunteers (60-93 years) underwent routine clinical and laboratory evaluation, including assessment of cfPWV in eligible participants. Flow-cytometry was used to assess proportions of memory T-cells, CD28null T-cells, and CMV-specific T-cells. The following associations were examined; CMV serostatus/cfPWV, CMV serostatus/proportion of Tmem, proportion of Tmem/cfPWV, CD28null T-cells/cfPWV, and CMV-specific T-cells/cfPWV. Linear regression models were used to adjust for age, sex, socioeconomic status, smoking, waist-to-hip ratio, cholesterol, and blood pressure as required. Results: Statistically significant positive associations were found (P-values for the fully adjusted models are given); CMV serostatus/cfPWV in men (P ≤ 0.01) but not in women, CMV serostatus/proportions of CD4 Tmem in men (P ≤ 0.05) but not in women; proportions of CD4 Tmem/cfPWV among CMV seropositive (CMV+) people (P ≤ 0.05) but not CMV seronegative (CMV-) people. Conclusion: CMV infection increases the CVD risk of older men by increasing cfPWV. This may be mediated in part by increased proportions of CD4 Tmem, higher numbers of which are found in CMV+ older people and more so among men than women. Given the high prevalence of CMV worldwide, our findings point to a significant global health issue. Novel strategies to mitigate the increased CVD risk associated with CMV may be required.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Carotid Arteries/immunology , Cytomegalovirus Infections/immunology , Immunologic Memory/immunology , Vascular Stiffness/immunology , Aged , Aorta/immunology , Aorta/virology , Atherosclerosis/immunology , Atherosclerosis/virology , Blood Pressure/immunology , CD28 Antigens/immunology , Cardiovascular Diseases/immunology , Cardiovascular Diseases/virology , Carotid Arteries/virology , Female , Humans , Male , Pulse Wave Analysis/methods , Risk Factors
6.
Theranostics ; 8(16): 4509-4519, 2018.
Article in English | MEDLINE | ID: mdl-30214635

ABSTRACT

An increased risk of cardiovascular death in Cytomegalovirus (CMV)-infected individuals remains unexplained, although it might partly result from the fact that CMV infection is closely associated with the accumulation of CD28null T-cells, in particular CD28null CD4 T-cells. These cells can directly damage endothelium and precipitate cardiovascular events. However, the current paradigm holds that the accumulation of CD28null T-cells is a normal consequence of aging, whereas the link between these T-cell populations and CMV infection is explained by the increased prevalence of this infection in older people. Resolving whether CMV infection or aging triggers CD28null T-cell expansions is of critical importance because, unlike aging, CMV infection can be treated. Methods: We used multi-color flow-cytometry, antigen-specific activation assays, and HLA-typing to dissect the contributions of CMV infection and aging to the accumulation of CD28null CD4 and CD8 T-cells in CMV+ and CMV- individuals aged 19 to 94 years. Linear/logistic regression was used to test the effect of sex, age, CMV infection, and HLA-type on CD28null T-cell frequencies. Results: The median frequencies of CD28null CD4 T-cells and CD28null CD8 T-cells were >12-fold (p=0.000) but only approximately 2-fold higher (p=0.000), respectively, in CMV+ (n=136) compared with CMV- individuals (n=106). The effect of CMV infection on these T-cell subsets was confirmed by linear regression. Unexpectedly, aging contributed only marginally to an increase in CD28null T-cell frequencies, and only in CMV+ individuals. Interestingly, the presence of HLA-DRB1*0301 led to an approximately 9-fold reduction of the risk of having CD28null CD4 T-cell expansions (OR=0.108, p=0.003). Over 75% of CMV-reactive CD4 T-cells were CD28null. Conclusion: CMV infection and HLA type are major risk factors for CD28null CD4 T-cell-associated cardiovascular pathology. Increased numbers of CD28null CD8 T-cells are also associated with CMV infection, but to a lesser extent. Aging, however, makes only a negligible contribution to the expansion of these T-cell subsets, and only in the presence of CMV infection. Our results open up new avenues for risk assessment, prevention, and treatment.


Subject(s)
Aging/pathology , CD28 Antigens/analysis , CD4-Positive T-Lymphocytes/chemistry , CD4-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/complications , Heart Failure/epidemiology , Heart Failure/physiopathology , Adult , Aged , Aged, 80 and over , Cytomegalovirus Infections/pathology , Female , Flow Cytometry , Histocompatibility Testing , Humans , Male , Middle Aged , Prevalence , Risk Factors , Young Adult
7.
J Infect Dis ; 217(12): 1997-2007, 2018 05 25.
Article in English | MEDLINE | ID: mdl-29506153

ABSTRACT

Background: Interleukin (IL)-7 promotes the generation, expansion, and survival of memory T cells. Previous mouse and human studies showed that IL-7 can support immune cell reconstitution in lymphopenic conditions, expand tumor-reactive T cells for adoptive immunotherapy, and enhance effector cytokine expression by autoreactive T cells. Whether pathogen-reactive T cells also benefit from IL-7 exposure remains unknown. Methods: In this study, we investigated this issue in cultures of peripheral blood mononuclear cells (PBMCs) derived from patients infected with various endemic pathogens. After short-term exposure to IL-7, we measured PBMC responses to antigens derived from pathogens, such as Mycobacterium tuberculosis, Candida albicans, and cytomegalovirus, and to the superantigen Staphylococcus aureus enterotoxin B. Results: We found that IL-7 favored the expansion and, in some instances, the uncovering of pathogen-reactive CD4 T cells, by promoting pathogen-specific interferon-γ, IL-2, and tumor necrosis factor recall responses. Conclusions: Our findings indicate that IL-7 unveils and supports reactivation of pathogen-specific T cells with possible diagnostic, prognostic, and therapeutic significance of clinical value, especially in conditions of pathogen persistence and chronic infection.


Subject(s)
Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Interleukin-7/immunology , Aged , Candida albicans/immunology , Candidiasis/immunology , Cytomegalovirus/immunology , Cytomegalovirus Infections/immunology , Female , Humans , Immunotherapy, Adoptive/methods , Interferon-gamma/immunology , Interleukin-2/immunology , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/immunology , Male , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Tumor Necrosis Factor-alpha/immunology
8.
Front Immunol ; 8: 489, 2017.
Article in English | MEDLINE | ID: mdl-28503175

ABSTRACT

T follicular helper cells (Tfh) provide crucial signals for germinal center (GC) formation, but Tfh populations are heterogeneous. While PD1hi Tfh are important in the GC response, the function of the PD1lo Tfh-like subset is unknown. We show that these cells, like the PD1hi GC-Tfh, depend upon B cells; however, their entry to follicles is independent of CXCR5 or cognate interactions with B cells. The differentiation into PD1hi Tfh is dependent on MHC class II interactions with B cells and requires CXCR5. Our data suggest a Tfh differentiation pathway that is initially B cell-independent, then dependent on non-cognate B cell interactions, and finally following cognate interaction with B cells and CXCR5-ligands allows the formation of GC-Tfh. The PD1lo Tfh-like cells make early cytokine responses and may represent precursors of CD4 memory cells.

9.
J Infect Dis ; 215(8): 1212-1220, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28199648

ABSTRACT

Cytomegalovirus (CMV) infection sometimes causes large expansions of CMV-specific T cells, particularly in older people. This is believed to undermine immunity to other pathogens and to accelerate immunosenescence. While multiple different CMV proteins are recognized, most publications on age-related T-cell expansions have focused on dominant target proteins UL83 or UL123, and the T-cell activation marker interferon-γ (IFN-γ). We were concerned that this narrow approach might have skewed our understanding of CMV-specific immunity at older ages. We have, therefore, widened the scope of analysis to include in vitro-induced T-cell responses to 19 frequently recognized CMV proteins in "young" and "older" healthy volunteers and a group of "oldest old" long-term survivors (>85 years of age). Polychromatic flow cytometry was used to analyze T-cell activation markers (CD107, CD154, interleukin-2 [IL-2], tumor necrosis factor [TNF], and IFN-γ) and memory phenotypes (CD27, CD45RA). The older group had, on average, larger T-cell responses than the young, but, interestingly, response size differences were relatively smaller when all activation markers were considered rather than IFN-γ or TNF alone. The oldest old group recognized more proteins on average than the other groups, and had even bigger T-cell responses than the older group with a significantly larger central memory CD4 T-cell component.


Subject(s)
Aging , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Immunologic Memory , Adult , Aged , Aged, 80 and over , Biomarkers/metabolism , Cytomegalovirus/genetics , Female , Flow Cytometry , Healthy Volunteers , Humans , Interferon-gamma/metabolism , Male , Middle Aged , Tumor Necrosis Factor-alpha/metabolism , Young Adult
10.
Front Immunol ; 8: 1977, 2017.
Article in English | MEDLINE | ID: mdl-29459855

ABSTRACT

The systemic inflammatory response syndrome (SIRS) is a potentially lethal response triggered by diverse forms of tissue injury and infection. When systemic inflammation is triggered by infection, the term sepsis is used. Understanding how inflammation is mediated and regulated is of enormous medical importance. We previously demonstrated that circulating inflammatory-relevant microRNAs (CIR-miRNAs) are candidate biomarkers for differentiating sepsis from SIRS. Here, we set out to determine how CIR-miRNA levels reflect SIRS severity and whether they derive from activated immune cells. Clinical disease severity scores and markers of red blood cell (RBC) damage or immune cell activation were correlated with CIR-miRNA levels in patients with SIRS and sepsis. The release of CIR-miRNAs modulated during SIRS was assessed in immune cell cultures. We show that severity of non-infective SIRS, but not sepsis is reflected in the levels of miR-378a-3p, miR-30a-5p, miR-30d-5p, and miR-192-5p. These CIR-miRNA levels positively correlate with levels of the redox biomarker, peroxiredoxin-1 (Prdx-1), which has previously been shown to be released by immune cells during inflammation. Furthermore, in vitro activated immune cells produce SIRS-associated miR-378a-3p, miR-30a-5p, miR-30d-5p, and miR-192-5p. Our study furthers the understanding of the origin, role, and trafficking of CIR-miRNAs as potential regulators of inflammation.

11.
J Infect Dis ; 214(9): 1430-1437, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27521364

ABSTRACT

BACKGROUND: Parallel upregulation of several T-cell effector functions (ie, polyfunctionality) is believed to be critical for the protection against viruses but thought to decrease in large T-cell expansions, in particular at older ages. The factors determining T-cell polyfunctionality are incompletely understood. Here we revisit the question of cytomegalovirus (CMV)-specific T-cell polyfunctionality, including a wide range of T-cell target proteins, response sizes, and participant ages. METHODS: Polychromatic flow cytometry was used to analyze the functional diversity (ie, CD107, CD154, interleukin 2, tumor necrosis factor, and interferon γ expression) of CD4+ and CD8+ T-cell responses to 19 CMV proteins in a large group of young and older United Kingdom participants. A group of oldest old people (age >85 years) was included to explore these parameters in exceptional survivors. Polyfunctionality was assessed for each protein-specific response subset, by subset and in aggregate, across all proteins by using the novel polyfunctionality index. RESULTS: Polyfunctionality was not reduced in healthy older people as compared to young people. However, it was significantly related to target protein specificity. For each protein, it increased with response size. In the oldest old group, overall T-cell polyfunctionality was significantly lower. DISCUSSION: Our results give a new perspective on T-cell polyfunctionality and raise the question of whether maintaining polyfunctionality of CMV-specific T cells at older ages is necessarily beneficial.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Adult , Aged , Aged, 80 and over , Female , Flow Cytometry , Humans , Interferon-gamma/immunology , Interleukin-2/immunology , Male , Middle Aged , Tumor Necrosis Factor-alpha/immunology , United Kingdom , Young Adult
12.
Sci Rep ; 6: 28006, 2016 06 20.
Article in English | MEDLINE | ID: mdl-27320175

ABSTRACT

Systemic inflammation in humans may be triggered by infection, termed sepsis, or non-infective processes, termed non-infective systemic inflammatory response syndrome (SIRS). MicroRNAs regulate cellular processes including inflammation and may be detected in blood. We aimed to establish definitive proof-of-principle that circulating microRNAs are differentially affected during sepsis and non-infective SIRS. Critically ill patients with severe (n = 21) or non-severe (n = 8) intra-abdominal sepsis; severe (n = 23) or non-severe (n = 21) non-infective SIRS; or no SIRS (n = 16) were studied. Next-generation sequencing and qRT-PCR were used to measure plasma microRNAs. Detectable blood miRNAs (n = 116) were generally up-regulated in SIRS compared to no-SIRS patients. Levels of these 'circulating inflammation-related microRNAs' (CIR-miRNAs) were 2.64 (IQR: 2.10-3.29) and 1.52 (IQR: 1.15-1.92) fold higher for non-infective SIRS and sepsis respectively (p < 0.0001), hence CIR-miRNAs appeared less abundant in sepsis than in SIRS. Six CIR-miRNAs (miR-30d-5p, miR-30a-5p, miR-192-5p, miR-26a-5p, miR-23a-5p, miR-191-5p) provided good-to-excellent discrimination of severe sepsis from severe SIRS (0.742-0.917 AUC of ROC curves). CIR-miRNA levels inversely correlated with pro-inflammatory cytokines (IL-1, IL-6 and others). Thus, among critically ill patients, sepsis and non-infective SIRS are associated with substantial, differential changes in CIR-miRNAs. CIR-miRNAs may be regulators of inflammation and warrant thorough evaluation as diagnostic and therapeutic targets.


Subject(s)
MicroRNAs/blood , Sepsis/blood , Sepsis/diagnosis , Systemic Inflammatory Response Syndrome/blood , Systemic Inflammatory Response Syndrome/diagnosis , Adult , Aged , Area Under Curve , Biomarkers/blood , C-Reactive Protein/analysis , Female , High-Throughput Nucleotide Sequencing , Humans , Interleukin-1/blood , Interleukin-6/blood , Male , MicroRNAs/chemistry , Middle Aged , Principal Component Analysis , ROC Curve , Sepsis/genetics , Sepsis/pathology , Sequence Analysis, DNA , Severity of Illness Index , Systemic Inflammatory Response Syndrome/genetics , Systemic Inflammatory Response Syndrome/pathology , Up-Regulation
13.
Mech Ageing Dev ; 158: 14-22, 2016 09.
Article in English | MEDLINE | ID: mdl-26957355

ABSTRACT

BACKGROUND: In studies exploring the effects of HCMV infection on immune system aging ('immunosenescence'), after organ transplantation or in other settings, HCMV-specific T-cell responses are often assessed with respect to purportedly 'immunodominant' protein subunits. However, the response structure in terms of recognized antigens and response hierarchies (architecture) is not well understood and actual correlates of immune protection are not known. METHODS: We explored the distribution of T-cell response sizes and dominance hierarchies as well as response breadth in 33 HCMV responders with respect to >200 HCMV proteins. RESULTS: At the individual responder level HCMV-specific T-cell responses were generally arranged in clear dominance hierarchies; interestingly, the number of proteins recognized by an individual correlated closely with the size of their biggest response. Target-specificity varied considerably between donors and across hierarchy levels with the presence, size, and hierarchy position of responses to purportedly 'immunodominant' targets being unpredictable. CONCLUSIONS: Predicting protective immunity based on isolated HCMV subunit-specific T-cell responses is questionable in light of the complex architecture of the overall response. Our findings have important implications for T-cell monitoring, intervention strategies, as well as the application of animal models to the understanding of human infection.


Subject(s)
Aging/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Immunity, Cellular , T-Lymphocytes/immunology , Adult , Aging/pathology , Animals , Cytomegalovirus Infections/pathology , Female , Humans , Male , Middle Aged , T-Lymphocytes/pathology
14.
Methods Mol Biol ; 1396: 181-206, 2016.
Article in English | MEDLINE | ID: mdl-26676048

ABSTRACT

Regulatory T cells (Tregs) that suppress the activation of immune effector cells limit immunopathology and are fast emerging as therapeutic targets for autoimmune and cancer disease. Tools enabling Treg in vitro-induction, expansion, and characterization and manipulation will help future clinical developments. In this chapter, we describe in detail how to use bacterial superantigens to induce human Tregs efficiently from peripheral blood mononuclear cells. How to assess human Treg phenotype and suppressive capacity are also described. Technical details, variations, and alternative experimental conditions are provided.


Subject(s)
Lymphocyte Activation/immunology , Superantigens/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Antigens, Surface/metabolism , Cell Culture Techniques , Cells, Cultured , Coculture Techniques , Flow Cytometry , Humans , Immunomagnetic Separation/methods , Immunomodulation , Immunophenotyping , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Phenotype , Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/immunology , Precursor Cells, T-Lymphoid/metabolism
15.
Crit Rev Immunol ; 32(2): 97-126, 2012.
Article in English | MEDLINE | ID: mdl-23216610

ABSTRACT

Harnessing T-cell responses to constrain tumor growth is a realistic treatment aspiration in tumor medicine, as many tumors express specific tumor associated antigens that are recognized by the adaptive immune system. CD8 T cells have direct cytolytic activity against tumor cells, and CD4 T cells mount a variety of responses that have important influences on tumor growth. We discuss how individual T-cell subsets contribute to antitumor responses and the goals and problems associated with generating and/or maintaining effective multifunctional T-cell responses to provide long-term protection against tumors.


Subject(s)
Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive/methods , Neoplasms/immunology , Adaptive Immunity , Animals , CD4-Positive T-Lymphocytes/transplantation , CD8-Positive T-Lymphocytes/transplantation , Cytotoxicity, Immunologic , Humans , Immunologic Memory , Neoplasms/therapy
16.
PLoS One ; 7(4): e35466, 2012.
Article in English | MEDLINE | ID: mdl-22496920

ABSTRACT

Certain parasites have evolved to evade the immune response and establish chronic infections that may persist for many years. T cell responses in these conditions become muted despite ongoing infection. Upregulation of surface receptors with inhibitory properties provides an immune cell-intrinsic mechanism that, under conditions of chronic infection, regulates immune responses and limits cellular activation and associated pathology. The negative regulator, CD200 receptor, and its ligand, CD200, have been shown to regulate macrophage activation and reduce pathology following infection. We show that CD4 T cells also increase expression of inhibitory CD200 receptors (CD200R) in response to chronic infection. CD200R was upregulated on murine effector T cells in response to infection with bacterial, Salmonella enterica, or helminth, Schistosoma mansoni, pathogens that respectively drive predominant Th1- or Th2-responses. In vitro chronic and prolonged stimuli were required for the sustained upregulation of CD200R, and its expression coincided with loss of multifunctional potential in T effector cells during infection. Importantly, we show an association between IL-4 production and CD200R expression on T effector cells from humans infected with Schistosoma haematobium that correlated effectively with egg burden and, thus infection intensity. Our results indicate a role of CD200R:CD200 in T cell responses to helminths which has diagnostic and prognostic relevance as a marker of infection for chronic schistosomiasis in mouse and man.


Subject(s)
Antigens, CD/immunology , Antigens, Surface/biosynthesis , CD4-Positive T-Lymphocytes/parasitology , Membrane Glycoproteins/biosynthesis , Receptors, Cell Surface/biosynthesis , Schistosomiasis/immunology , Adolescent , Animals , Anthelmintics/therapeutic use , Antigens, Surface/immunology , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Child , Chronic Disease , Cohort Studies , Female , Humans , Interleukin-4/biosynthesis , Interleukin-4/immunology , Male , Membrane Glycoproteins/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Orexin Receptors , Praziquantel/therapeutic use , Receptors, Cell Surface/immunology , Schistosoma haematobium/immunology , Schistosoma haematobium/isolation & purification , Schistosoma mansoni/immunology , Schistosoma mansoni/isolation & purification , Schistosomiasis/drug therapy , Severity of Illness Index
17.
J Immunol ; 185(11): 6545-54, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21048115

ABSTRACT

The effect of TCR signals on the differentiation of memory T cells is poorly defined. Conventional wisdom suggests that high-avidity interactions are best for the selection of vaccine Ag candidates or T cell specificities for adoptive T cell therapy to stimulate robust responses. However, in conditions of Ag persistence, high-avidity clones might exhaust and fail to form long-lived protective memory. We have manipulated the functional avidity of CD4 T cells by reducing expression of Lck, a key kinase involved in TCR triggering. Using a mouse model, we followed tetramer-positive T cells responding to a tumor Ag expressed by an adenocarcinoma. We show that reducing the functional avidity increased effector-effector memory responses and improved the generation of self-renewing, recirculating, tumor Ag-specific memory phenotype CD4 T cells. Moreover, such cells together with wild type CD8 T cells were better able to control tumor growth. Mechanistically, reducing Lck prolonged IL-2 production and cell turnover in the central memory population while reducing expression of exhaustion markers in the face of chronic Ag. Our data indicate that, in situations of persistent Ag challenge, generating T cells with reduced functional avidity may elicit more effective immune responses.


Subject(s)
Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , Immunologic Memory , Adenocarcinoma/enzymology , Adenocarcinoma/immunology , Amino Acid Sequence , Animals , Antigens, Protozoan/biosynthesis , CD4-Positive T-Lymphocytes/enzymology , CD4-Positive T-Lymphocytes/parasitology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/parasitology , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Line, Tumor , Cells, Cultured , Immunologic Memory/genetics , Leishmania major/immunology , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Models, Immunological , Molecular Sequence Data , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/biosynthesis
19.
Eur J Immunol ; 40(2): 470-9, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19950184

ABSTRACT

It is well established that tumours hinder both natural and vaccine-induced tumour-specific CD4(+) T-cell responses. Adoptive T-cell therapy has the potential to circumvent functional tolerance and enhance anti-tumour protective responses. While protocols suitable for the expansion of cytotoxic CD8(+) T cells are currently available, data on tumour-specific CD4(+) T cells remain scarce. We report here that CD4(+) T cells sensitized to tumour-associated Ag in vivo, proliferate in vitro in response to IL-7 without the need for exogenous Ag stimulation and accumulate several folds while preserving a memory-like phenotype. Both cell proliferation and survival accounts for the outgrowth of tumour-sensitized T cells among other memory and naive lymphocytes following exposure to IL-7. Also IL-2, previously used to expand anti-tumour CTL, promotes tumour-specific CD4(+) T-cell accumulation. However, IL-7 is superior to IL-2 at preserving lymphocyte viability, in vitro and in vivo, maintaining those properties, that are required by helper CD4(+) T cells to confer therapeutic efficacy upon transplantation in tumour-bearing hosts. Together our data support a unique role for IL-7 in retrieving memory-like CD4(+) T cells suitable for adoptive T-cell therapy.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Cell Proliferation/drug effects , Interleukin-2/pharmacology , Interleukin-7/pharmacology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Flow Cytometry , Immunologic Memory/drug effects , Immunologic Memory/immunology , Immunotherapy, Adoptive , Interleukin-15/pharmacology , Interleukin-6/pharmacology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy
20.
Immunol Rev ; 228(1): 9-22, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19290918

ABSTRACT

T-cell development in the thymus and activation of mature T cells in secondary lymphoid organs requires the ability of cells to respond appropriately to environmental signals at multiple stages of their development. The process of thymocyte selection insures a functional T-cell repertoire, while activation of naive peripheral T cells induces proliferation, gain of effector function, and, ultimately, long-lived T-cell memory. The T-cell immune response is initiated upon engagement of the T-cell receptor (TCR) and coreceptor, CD4 or CD8, by cognate antigen/major histocompatibility complexes presented by antigen-presenting cells. TCR/coreceptor engagement induces the activation of biochemical signaling pathways that, in combination with signals from costimulator molecules and cytokine receptors, direct the outcome of the response. Activation of the src-family kinases p56(lck) (Lck) and p59(fyn) (Fyn) is central to the initiation of TCR signaling pathways. This review focuses on our current understanding of the mechanisms by which these two proteins orchestrate T-cell function.


Subject(s)
Immune Tolerance , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes/cytology , Cell Differentiation , Humans , Lymphocyte Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...