• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 38
  • 16
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • Tagged with
  • 90
  • 90
  • 90
  • 90
  • 32
  • 23
  • 22
  • 22
  • 21
  • 21
  • 19
  • 17
  • 16
  • 16
  • 16
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
41

Perfil fenotípico de linfócitos T CD8+ na fase aguda da dengue / Phenotypic profile of CD8+ T cells in acute dengue infection

Andréia Manso de Matos 31 October 2011 (has links)
A dengue é uma doença infecciosa aguda causada pelo vírus DEN do gênero Flavivirus e é transmitida pela picada de um mosquito vetor, principalmente o Aedes aegypti. Existem quatro sorotipos do vírus da dengue (DEN-1, DEN-2, DEN-3 e DEN-4) e sua incidência tem aumentado dramaticamente nos últimos 50 anos, inclusive no Brasil. O objetivo deste trabalho é caracterizar subpopulações de linfócitos T, principalmente linfócitos T CD8+, provenientes de pacientes infectados quanto a sua capacidade proliferativa, seu estado de ativação e memória celular. Os pacientes foram recrutados no Hospital Ana Costa de Santos, SP, no ano de 2010, após assinarem o Termo de Consentimento Livre e Esclarecido. O diagnóstico de dengue foi realizado utilizando o teste rápido Dengue Duo e os parâmetros imunológicos foram analisados no citômetro de fluxo. Foi coletado sangue periférico para criopreservação de células mononucleares e separação de soro para detecção da carga viral. Pacientes com dengue apresentaram maior proliferação de linfócitos T CD8+ quando comparados com indivíduos saudáveis. Foi ainda observado que tal proliferação celular foi evidente nos dias cinco e seis de sintomas. Quando marcadores de ativação celular foram analisados por citometria de fluxo, observou-se um aumento de linfócitos T CD8+ expressando CD38 e HLA-DR nos pacientes, quando comparados com indivíduos saudáveis. Da mesma forma, a ativação celular também aumentou com o passar dos dias de sintomas com destaque para o quinto e sexto dia. Este aumento na ativação das células juntamente com os dias de sintomas, foi igualmente observado em várias subpopulações de células T de memória Além disso, foi observada uma correlação negativa entre o número absoluto de linfócitos T CD8+ e a carga viral. Juntos, os resultados desse estudo sugerem que a infecção por DEN leva a um aumento da ativação de células T CD8+ / Dengue is an acute disease caused by DEN, a Flavivirus transmitted by a mosquito vector, primarily Aedes aegypti. There are four serotypes of dengue viruses (DEN-1, DEN-2, DEN-3 and DEN-4) and their incidences have increased dramatically over the past 50 years, including in Brazil. The goal of this study is to characterize subpopulations of T lymphocytes, mainly CD8+ T cells, from infected patients. Status of cell activation, and memory cell profiles were assessed. Patients were recruited at Hospital Ana Costa, Santos-SP, Brazil, in 2010, after having signed an informed consent form. The serologic diagnosis of dengue was carryied out using a rapid test and immunological parameters were analyzed in flow cytometer. Peripheral blood was collected for cryopreservation of mononuclear cells and separation of serum for viral load testing. Dengue patients showed higher proliferation of CD8+ T cells compared to healthy subjects. Cell proliferation was more evident in the fifth and sixth days of symptoms. We observed increased frequency of CD8+ T cells expressing activation markers CD38 and HLA-DR in patients, when compared to healthy subjects. Similarly, T cell activation also increased along with the passing days of symptoms hitting on the fifth and the sixth days. Such augment in cellular activation along with the days of symptoms was equally observed in the several memory T cell compartments. Furthermore, we observed a negative correlation between the absolute number of CD8+ T lymphocytes and viral load. Together, the results of this study suggest that dengue virus infection leads to an increased activation of CD8+ T cells
42

The Role of Signal 3 Cytokine Timing in CD8 T Cell Activation: A Dissertation

Urban, Stina L. 16 July 2015 (has links)
During an acute virus infection, antigen-specific CD8 T cells undergo clonal expansion and differentiation into effector cells in order to control the infection. Efficient clonal expansion and differentiation of CD8 T cells are required to develop protective memory CD8 T cells. Antigen specific cells require 3 distinct signals for their activation: TCR engagement of peptide-MHC (signal 1), costimulation between B7 and CD28 (signal 2), and inflammatory cytokines including IL-12 or type 1 IFN (signal 3). CD8 T cells that encounter antigen and costimulation undergo programmed cell division, but these two signals alone are not sufficient for full effector cell differentiation and survival into memory. CD8 T cells need a third signal for efficient clonal expansion, differentiation into various effector populations, acquisition of cytolytic effector functions, and memory formation. The requirements for signal 3 cytokines in CD8 T cell activation have only been recently described; however, the timing of exposure to these signals has yet to be investigated. During the course of an immune response not all T cells will see antigen, costimulation, and inflammatory cytokines at the same time or in the same order. I sought to examine how the timing of signal 3 cytokines affected CD8 T cell activation. I questioned how the order of these signals effected CD8 T cell priming and subsequent activation, expansion and differentiation. In order to study the in vivo effects of out-of-sequence signaling on CD8 T cell activation, I utilized poly(I:C), a dsRNA analogue, which is known to induce a strong type 1 IFN response. Through the use of various congenic transgenic and polyclonal CD8 T cell populations, in conjunction with adoptive transfer models, specific T cells which had been exposed to poly(I:C) induced environments could be identified and tracked over time. I wanted to characterize how out-of-sequence signaling affected T cell activation immediately after cognate antigen stimulation (4-5hours), and after prolonged exposure to cognate antigen (days-weeks). Considering type 1 IFN can have both inhibitory and stimulatory effects on CD8 T cell proliferation, and when type 1 IFN provides signal 3 cytokine activity, it has positive effects on CD8 T cell expansion, I wanted to investigate the role of type 1 IFN as an out-of-sequence signal during CD8 T cell activation. We identified a transient defect in the phosphorylation of downstream STAT molecules after IFNβ signaling within poly(I:C) pretreated CD8 T cells. The inability of poly(I:C) pretreated CD8 T cells to respond to IFNβ signaling makes these cells behave in a manner more similar to T cells that only received 2 signals, rather than ones that received all 3 signals in the appropriate order. Consequently, poly(I:C) pretreated, or out-of-sequence, CD8 T cells were found to have defects in clonal expansion, effector differentiation and function as well as memory generation resulting in reduced efficacy of viral clearance. Out-of-sequence CD8 T cells showed suppression of CD8 T cell responses after prolonged exposure to cognate antigen, but naïve CD8 T cells pre-exposed to poly(I:C) exhibited immediate effector function within hours of cognate antigen stimulation, prior to cell division. Poly(I:C) pretreated naïve CD8 T cells acquired an early activated phenotype associated with alterations of transcription factors and surface markers. Changes in naïve CD8 T cell phenotype are thought to be mediated by poly(I:C)-induced upregulation of self-MHC and costimulatory molecules on APCs through direct type 1 IFN signaling. Inoculating with poly(I:C) enabled naive CD8 T cells to produce effector functions immediately upon stimulation with high density cognate antigen, reduced affinity altered peptide ligands (APLs), and in response to reduced concentrations of cognate antigen. Unlike conventional naïve CD8 T cells, poly(I:C) pretreated naïve CD8 T cells acquired the ability to specifically lyse target cells. These studies identified how the timing of activation signals can dramatically affect the acquisition of CD8 T cell effector function. This thesis describes how CD8 T cell exposure to activation signals in an unconventional order may result in altered response to antigen stimulation. Exposure of naïve CD8 T cells to type 1 IFN and costimulatory molecules in the presence of self-peptides enabled them to respond immediately upon antigen stimulation. Primed naïve CD8 T cells produced multiple cytokines in response to low-affinity, and low-density antigens, and gained ability to specifically lyse target cells. However, immediate effector function may come at the expense of clonal expansion and effector cell differentiation in response to prolonged antigen exposure as out-of-sequence CD8 T cells showed reduced proliferation, effector function and memory formation. The findings presented here may seem contradictory because out-of-sequence signaling can prime T cells to produce immediate effector functions and yet cause defects in T cell expansion and effector differentiation. However, these two models ascertained T cell function at different points after antigen exposure; one where functions were evaluated within hours after seeing cognate antigen, and the other showing T cell responses after days of antigen stimulation. Studies described in this thesis highlight the growing complexity of CD8 T cell activation. Not only do the presence or absence of signals 1-3 contribute to T cell activation, but the timing of these signals also proves to be of great importance. These studies may describe how both latecomer and third party antigen specific T cells behave when and if they encounter cognate antigen in the midst of an ongoing infection. Out-of-sequence exposure to IFN initially stimulates effector function but at the expense of efficient clonal expansion and subsequent memory formation. The immediate effector function that naïve T cells gain during out-of-sequence priming may explain how some individuals are more resistant to superinfections, whereas the impairment in proliferation describes a universal mechanism of virus-induced immune suppression, explaining how other individuals can be more susceptible to secondary infections. Ultimately, results identified here can be applied to developing better and more effective vaccines.
43

The Role of Heterologous Immunity in Mediating Natural Resistance to Infection in Human Subjects: A Dissertation

Watkin, Levi B. 13 March 2012 (has links)
Heterologous immunity is a mechanism by which immunological memory within an individual, developed in response to a previous infection, plays a role in the immune response to a subsequent unrelated infection. In murine studies, heterologous immunity facilitated by cross-reactive CD8 T-cell responses can mediate either beneficial (protective immunity) or detrimental effects (e.g. enhanced lung and adipose immunopathology and enhanced viral titers) (Selin et al., 1998; Chen et al., 2001; Welsh and Selin, 2002; Nie et al., 2010; Welsh et al., 2010). Protective heterologous immunity results in enhanced clearance of virus during a subsequent infection with an unrelated pathogen. Such is the case when mice are immunized with lymphocytic choriomeningitis virus (LCMV) and subsequently challenged with Pichinde virus (PV) or vaccinia virus (VACV) (Selin et al., 1998). However, heterologous immunity may also mediate enhanced immunopathology as mice immunized with influenza A virus (IAV) and challenged with LCMV show increased viral titers and enhanced lung immunopathology (Chen et al., 2003). The role heterologous immunity plays during infection is not limited to the murine system. In fact, there have now been several reports of enhanced immunopathology due to heterologous immunity during human infections, involving viruses such as IAV, Epstein-Barr Virus (EBV), hepatitis C virus (HCV), and dengue virus (DENV) (Mathew et al., 1998; Wedemeyer et al., 2001; Acierno et al., 2003; Nilges et al., 2003; Clute et al., 2005; Urbani et al., 2005). Interestingly, in all reported cases in humans, heterologous immunity mediated enhanced immunopathology. Upon infection with EBV the clinical presentation can range from asymptomatic to severe, occasionally fatal, acute infectious mononucleosis (AIM) (Crawford et al., 2006b; Luzuriaga and Sullivan, 2010) which is marked by a massive CD8 lymphocytosis. This lympho-proliferative effect in AIM was shown to be partially mediated by reactivation of cross-reactive IAV-M1 58-66 (IAV-GIL) specific CD8 memory T-cells in HLA-A2 patients reacting to the EBV-BMLF1 280 (EBV-GLC) epitope (Clute et al., 2005). Interestingly, EBV infects ~90% of individuals globally by the third decade of life, establishing a life-long infection (Henle et al., 1969). However, it is unknown why 5-10% of adults remain EBV-sero-negative (EBV-SN), despite the fact that the virus infects the vast majority of the population and is actively shed at high titers even during chronic infection (Hadinoto et al., 2009). Here, we show that EBV-SN HLA-A2+ adults possess cross-reactive IAV-GIL/EBV-GLC memory CD8 T-cells that show highly unique properties. These IAV-GIL cross-reactive memory CD8 T-cells preferentially expand and produce cytokines to EBV antigens at high functional avidity. Additionally, they are capable of lysing EBV-infected targets and show the potential to enter the mucosal epithelial tissue, where infection is thought to initiate, by CD103 expression. This protective capacity of these cross-reactive memory CD8 T-cells may be explained by a unique T-cell receptor (TCR) repertoire that differs by both organization and CDR3 usage from that in EBV-seropositive (EBV-SP) donors. The composition of the CD8 T-cell repertoire is a dynamic process that begins during the stochastic positive selection of the T-cell pool during development in the thymus. Thus, upon egress to the periphery a naïve T-cell pool, or repertoire, is formed that is variable even between genetically identical individuals. This T-cell repertoire is not static, as each new infection leaves its mark on the repertoire once again by stochastically selecting and expanding best-fit effectors and memory populations to battle each new infection while at the same time deleting older memory CD8 T-cells to make room for the new memory cells (Selin et al., 1999). These events induce an altered repertoire that is unique to each individual at each infection. It is this dynamic and variable organization of the T-cell repertoire that leads to private specificity even between genetically identical individuals upon infection with the same pathogens and thus a different fate (Kim et al., 2005; Cornberg et al., 2006a; Nie et al., 2010). It is this private specificity of the TCR repertoire that helps explain why individuals with the same epitope specific cross-reactive response, but composed of different cross-reactive T-cell clones, can either develop AIM or never become infected with EBV. Our results suggest that heterologous immunity may protect EBV-SN adults against the establishment of productive EBV infection, and potentially be the first demonstration of protective T-cell heterologous immunity between unrelated pathogens in humans. Our results also suggest that CD8 T-cell immunity can be sterilizing and that an individual’s TCR repertoire ultimately determines their fate during infection. To conclusively show that heterologous immunity is actively protecting EBV-SN adults from the establishment of a productive EBV infection, one would have to deliberately expose an individual to the virus. Clearly, this is not an acceptable risk, and it could endanger the health of an individual. A humanized mouse model could allow one to address this question. However, before we can even attempt to address the question of heterologous immunity mediating protection from EBV infection in humanized mice, we must first determine whether these mice can be infected with, and build an immune response to the two viruses we are studying, EBV and IAV. We show here that these mice can indeed be infected with and also mount an immune response to EBV. Additionally, these mice can also be infected with IAV. However, at this time the immune responses that are made to these viruses in our established humanized mouse model are not substantial enough to fully mimic a human immune response capable of testing our hypothesis of heterologous immunity mediating protection from EBV infection. Although the immune response in these mice to EBV and IAV infection is not suitable for the testing of our model the data are promising, as the humanized mouse model is constantly improving. Hopefully, with constant improvements being made there will be a model that will duplicate a human immune system in its entirety. This thesis will be divided into 5 major chapters. The first chapter will provide an introduction to both general T-cell biology and also to the role of heterologous immunity in viral infection. The second chapter will provide the details of the experimental procedures that were performed to test our hypothesis. The third chapter will describe the main scientific investigation of the role of heterologous immunity in providing natural resistance to infection in human subjects. This chapter will also consist of the data that will be compiled into a manuscript for publication in a peer-reviewed journal. The fourth chapter will consist of work performed pertaining to the establishment of a humanized mouse model of EBV and IAV infection. The establishment of this model is important for us to be able to show causation for protection from EBV infection mediated by heterologous immunity.
44

The Role of Heterologous Immunity in Viral Co-Infections and Neonatal Immunity: A Dissertation

Kenney, Laurie L. 01 August 2013 (has links)
The dynamics of T cell responses have been extensively studied during single virus infection of naïve mice. During a viral infection, viral antigen is presented in the context of MHC class I molecules on the surface of infected cells. Activated CD8 T cells that recognized viral antigens mediate clearance of virus through lysis of these infected cells. We hypothesize that the balance between the replicating speed of the virus and the efficiency at which the T cell response clears the virus is key in determining the disease outcome of the host. Lower T cell efficiency and delayed viral clearance can lead to extensive T cellmediated immunopathology and death in some circumstances. To examine how the efficiency of the immune response would impact immunopathology we studied several viral infection models where T cell responses were predicted to be less than optimal: 1. a model of co-infection with two viruses that contain a crossreactive epitope, 2. a viral infection model where a high dose infection is known to induce clonal exhaustion of the CD8 T cell response, 3. a neonatal virus infection model where the immune system is immature and 4. A model of beneficial heterologous immunity and T cell crossreactivity where mice are immunized as neonates when the T cell pool is still developing. Model 1. Simultaneous co-infections are common and can occur from mosquito bites, contaminated needle sticks, combination vaccines and the simultaneous administration of multiple vaccines. Using two distantly related arenaviruses, lymphocytic choriomeningitis virus (LCMV) and Pichinde virus (PICV), we questioned if immunological T cell memory and subsequent protection would be altered following a simultaneous co-infection, where two immune responses are generated within the same host at the same time. Coinfection with these two viruses, which require CD8 T cell responses to clear, resulted in decreased immune protection and enhanced immunopathology after challenge with either virus. After primary co-infection, each virus-specific immune response impacted the other as they competed within the same host and resulted in several significant differences in the CD8 T cell responses compared to mice infected with a single virus. Co-infected mice had a dramatic decrease in the overall size of the LCMV-specific CD8 T cell response and variability in which virus-specific response dominated, along with skewing in the immunodominance hierarchies from the normal responses found in single virus infected mice. The reduction in the number of LCMV-specific CD8 memory T cells, specifically cells with an effector memory-like phenotype, was associated with higher viral loads and increased liver pathology in co-infected mice upon LCMV challenge. The variability in the immunodominance hierarchies of co-infected mice resulted in an enhanced cross-reactive response in some mice that mediated enhanced immune-mediated fat pad pathology during PICV challenge. In both viral challenge models, an ineffective memory T cell response in co-infected mice facilitated increased viral replication, possibly leading to enhanced and prolonged accumulation of secondary effector T cells in the tissues, thereby leading to increased immune pathology. Thus, the magnitude and character of memory CD8 T cell responses in simultaneous co-infections differed substantially from those induced by single immunization. This has implications for the design of combination vaccines and scheduling of simultaneous immunizations. Model 2. The balance between protective immunity and immunopathology often determines the fate of the virus-infected host. Several human viruses have been shown to induce a wide range of severity of disease. Patients with hepatitis B virus (HBV), for example, show disease progression ranging from acute resolving infection to a persistent infection and fulminant hepatitis. Certain rapidly replicating viruses have the ability to clonally exhaust the T cell response, such as HBV and hepatitis C virus (HCV) in humans and the clone 13 strain of LCMV in mice. How rapidly virus is cleared is a function of initial viral load, viral replication rate, and efficiency of antigen-specific T cells. By infecting mice with three different inocula of LCMV clone 13, we questioned how the race between virus replication and T cell responses could result in different disease outcomes. A low dose of LCMV generated efficient CD8 T effector cells, which cleared the virus with minimal lung and liver pathology. A high dose of LCMV resulted in clonal exhaustion of T cell responses, viral persistence and little immunopathology. An intermediate dose only partially exhausted the CD8 T cell responses and was associated with significant mortality, and the surviving mice developed viral persistence and massive immunopathology, including necrosis of the lungs and liver. This was a T cell-mediated disease as T cell-deficient mice had no pathology and became persistently infected like mice infected with a high dose of LCMV clone 13. This suggests that for non-cytopathic viruses like LCMV, HCV and HBV, clonal exhaustion may be a protective mechanism preventing severe immunopathology and death. Model 3. Newborns are more susceptible to infections due to their lack of immunological memory and under-developed immune systems. Passive maternal immunity helps protect neonates until their immune systems have matured. We questioned if a noncytolytic virus that produces strong T cell responses in adult mice would also induce an equally effective response in neonatal mice. Neonates were infected with very low doses of LCMV Armstrong and surprisingly the majority succumbed to infection between days 7-11, which is the peak of the T cell response in adult mice infected with LCMV. Death was caused by T cell-dependent pathology and not viral load as 100% of T cell deficient neonates survived with minimal lung and liver pathology. This is similar to the adult model of medium dose LCMV clone 13, but T cell responses in neonates were not partially clonal exhausted. Furthermore, surviving neonates were not persistently infected, clearing virus by day 14 post infection. In adult mice direct intracranial infection leads to LCMV replication and CD8 T cell infiltration in the central nervous system (CNS), causing CD8 T cell-mediated death. However, this does not occur in adults during LCMV intraperitoneal (ip) infections. We questioned if unlike adults LCMV could be gaining access to the CNS in neonates following ip infection. Replicating LCMV was found in the brain of neonates after day 5 post infection along with virus-specific CD8 T cells producing IFNγ at day 9 post infection. Neonates lacking perforin had complete survival when followed until day 14 post infection, suggesting perforin-mediated T cell-dependent immunopathology within the CNS of neonates was causing death after LCMV infection. Passive immunity from LCMV-immune mothers also protected 100% of pups from death by helping control viral load early in infection. We believe that the maternal antibody compensates for the immature innate immune response of neonates and controls viral replication early so the neonatal T cell response induced less immunopathology. Neonates are commonly thought to have less functional immune systems, but these results show that neonates are capable of producing strong T cell responses that contribute to increased mortality. Model 4. Due to their enhanced susceptibility to infection neonatal and infant humans receive multiple vaccines. Several non-specific effects from immunizations have been observed, for example, measles or Bacillus Calmette- Guerin (BCG) vaccines have been linked to decreased death of children from infections other than measles virus or tuberculosis. These studies mirror the concepts of beneficial heterologous immunity, where previous immunization with an unrelated pathogen can result in faster viral clearance. LCMV-immune mice challenged with vaccinia virus (VV) have lower viral loads then naïve mice and survive lethal infections, but some mice do develop fat pad immunopathology in the form of panniculitis or acute fatty necrosis (AFN). We questioned how immunological T cell memory formed during the immature neonatal period would compare to memory generated in fully mature adults during a heterologous viral challenge. Mice immunized as neonates had comparable reduction in VV load and induction of AFN, indicating that heterologous immunity is established during viral infections early in life. Interestingly, the LCMV-specific memory populations that expanded in mice immunized as neonates differed from that of mice immunized as adults. In adult mice 50% of the mice have an expansion of LCMVNP205- specific CD8 T cells while the majority of neonates expanded the LCMVGP34- specific CD8 T cell pool. This alteration in dominant crossreactivities may be due to the limited T cell receptor repertoire of neonatal mice. In naïve neonatal mice we found altered Vβ repertoires within the whole CD8 T cell pool. Furthermore, there was altered Vβ usage within virus-specific responses compared to adult mice and a wide degree of variability between individual neonates, suggesting enhanced private specificity of the TCR repertoire. Beneficial heterologous immunity is maintained in neonates, but there was altered usage of crossreactive responses. As neonatal mice were found to be so sensitive to LCMV infection we questioned if neonates could control another arena virus that did not replicate as efficiently in mice, PICV. Unlike LCMV infection, neonatal mice survived infection with PICV even with adult-like doses. However, viral clearance was protracted in neonates compared to adults, but was cleared from fat pad and kidney by day 11 post infection. The peak of the CD8 T cell response was similarly delayed. PICV infected neonates showed dose-dependent PICV-specific CD8 T cell responses, which were similar to adult responses by frequency, but not total number. As with LCMV infection there were changes in immunodominance hierarchies in neonates. Examination of the immunodominance hierarchies of PICV-infected neonates showed that there were adult-like responses to the dominant NP38- specific response, but a loss of the NP122-specific response. Six weeks post neonatal infection mice were challenged with LCMV Armstrong and there was a strong skewing of the PICV immunodominance hierarchy to the crossreactive NP205-specific response. These data further support the hypothesis that heterologous immunity and crossreactivity develop following neonatal immunization, much as occurs in adults, although TCR repertoire and crossreactive patterns may differ. Changing the balance between T cell efficiency and viral load was found to altered the severity of the developing immunopathology after viral infection.
45

Organ-Dependent and Epitope-Dependent Repertoire Usage and Apoptosis of Antigen-Specific T Cells in Viral Infections: a Dissertation

Wang, Xiaoting Z. 01 April 2004 (has links)
During virus infections, activation of CD8 T cells takes place in secondary lymphoid organs including spleen and lymph nodes. The kinetics of the T cell response in lymphoid tissues has been clearly studied. However, a large number of virus-specific T cells disseminate into various nonlymphoid tissues. As reservoirs for effector and memory cells, nonlymphoid organs play an important role for defending against infections. T cell responses in nonlymphoid organs may differ from lymphoid organs. T cell repertoire usage in lymphoid and nonlymphoid tissues was studied in an acute lymphocytic choriomeningitis virus (LCMV)-infected murine model. The hierarchy of CD8 T cell specificities was examined with cytotoxic T lymphocyte (CTL) sodium 51 chromate (51Cr) release assays and intracellular interferon (IFN)γ assays. T cell receptor (TCR) repertoire usage was determined by complementarity determining region (CDR)3 length spectratyping analysis. Both T cell specificity and TCR repertoire usage revealed some similarities and differences between several organs. Within an epitope-specific CD8 T cell population, the TCR repertoire usage was similar in different organs of the same mouse, but highly heterogeneous between individual mice with genetically identical backgrounds. A very restricted CD4 TCR repertoire was observed in BALB/c mice after secondary respiratory syncytial virus (RSV) infection. Most of the CD4 T cells of BALB/c mice pre-immunized with RSV glycoprotein (GP) predominantly express Vβ14 TCR with discrete oligoclonal CDR3 regions. Depletion of Vβ14 CD4 T cells dramatically reduced immunopathology. The apoptotic phenotype of LCMV-specific CD8 T cells was studied in various lymphoid and nonlymphoid tissues during acute and memory stages of infections. Peripheral tissues (peritoneal cavity (PEC), fat pad, and lung) reacted with a much lower frequency with the early apoptotic marker Annexin V than those in spleen and lymph nodes. This was not due to a TCR-based selection because similar TCR spectratypes were seen in different organs. Activated lymphoid and nonlymphoid T cells from LCMV GP33 transgenic mice, which have identical TCR α and β chains on all T cells, had differential Annexin V binding. When incubated shortly in vitro, most Annexin V+ T cells rapidly fragmented their DNA and became terminal transferase-mediated dUTP nick end-labeling positive (TUNEL+), while much fewer Annexin V- cells became TUNEL+. Therefore, those Annexin-V+ cells were truly in a pre-apoptotic stage. The differential spontaneous apoptosis in different tissues is independent of several death/survival-related molecules, including Fas/Fas ligand (FasL), turner necrosis factor (TNF)α, interleukin (IL-15), perforin, B cell lymphoma (Bcl)-2 and independent of virus tropism. I further investigated the significance of the high Annexin V reactivity of lymphoid T cells. Pre-apoptotic cells were prevented from fragmenting their DNA by anti-CD3 or IL-2 stimulation in vitro. However, this pre-apoptotic phenotype precluded generation of memory. Annexin V reactive cells did not give rise to long-lived memory after being transferred into naïve hosts. The pre-apoptotic phenotype is also an intrinsic property of the epitope. Different proportions of apoptotic cells were found in LCMV effector and, memory T cells specific to two different epitopes, nucleoprotein (NP)396 and GP33. Higher Annexin V reactivity of NP396-specific CD8 T cells was independent of virus tropism and duration of encounter with antigen. Higher expression of IL-7R was found in peripheral, Annexin V- and GP33-specific CD8 T cells, indicating that IL-7-dependent signals may inhibit apoptosis. Nonlymphoid T cells were more resistant than lymphoid T cells to activation-induced cell death (AICD). When stimulated with anti-CD3 in vitro for 40 hours (hr), a significantly reduced number of splenic transgenic T cells were recovered with much higher frequency of Annexin V reactivity and TUNEL staining than transgenic T cells from PEC. Consistent with the finding that Fas and FasL regulates AICD, a much lower expression of Fas and FasL was observed in PEC and lung transgenic T cells than spleen and lymph nodes after short time stimulation. FasL blockage largely increased cell-number recovery and reduced Annexin V and TUNEL staining of spleen transgenic T cells. Interestingly, the leukocyte environment played an important role of deciding the fate of transgenic T cells. When placing activated spleen transgenic T cells with excess infected PEC cells, spleen transgenic cells rapidly reduced their Annexin V staining and TUNEL staining and were recovered with greater number after stimulation. Vice versa, PEC transgenic T cells became Annexin V and TUNEL positive with lower numbers of cells recovered when placed with excess splenocytes. Less detection of Annexin V+ cells in peripheral tissues was not due to rapid phagocytosis by macrophages, because Cytochalasin D, which can inhibit phagocytosis, did not induce equal amount of pre-apoptotic cells in spleen and PEC. This reduced death in the periphery may contribute to the long-term maintenance of nondividing nonlymphoid memory T cells, enabling them to efficiently function without being driven into apoptosis. Overall, this study characterizes in detail the different T cell repertoire usage and apoptosis of virus-specific T cells based on their organ localization and specificities and helps to better understand T cell immunity after infections and vaccine design.
46

The Role of Inducible T Cell Kinase (Itk) in the Development of Innate T Cells and in the Formation of Protective Memory Responses: A Dissertation

Prince, Amanda L. 27 February 2013 (has links)
T cell development in the thymus produces multiple lineages of cells, including conventional naïve CD4+ and CD8+ T cells, regulatory T cells, and innate T cells. Innate T cells encompass γδ T cells, invariant natural killer (iNKT) cells, mucosal-associated invariant T (MAIT) cells, and H2-M3-restricted cells (Berg, 2007). Although they are a minor subset of all thymocytes, innate T cells develop in the thymus and share characteristics of the innate and adaptive immune systems (Berg, 2007). These lymphocytes undergo antigen receptor rearrangement and are able to exert their effector function immediately upon ex vivo stimulation (Berg, 2007). However, in several strains of mice harboring mutations in T cell signaling proteins or transcriptional regulators, conventional CD8+ T cells develop as innate cells that share characteristics with memory T cells (Atherly et al., 2006b; Broussard et al., 2006; Fukuyama et al., 2009; Gordon et al., 2011; Verykokakis et al., 2010b; Weinreich et al., 2010). One of these signaling proteins, inducible T cell kinase (Itk) is a nonreceptor protein tyrosine kinase that signals downstream of the T cell receptor (TCR) (Berg et al., 2005). Upon TCR activation, Itk is activated and recruited to the TCR signaling complex, where Itk interacts with Src homology 2 (SH2) domain-containing leukocyte phosphoprotein of 76 kDa (SLP-76), linker for activation of T cells (LAT), and phospholipase C γ1 (PLCγ1) (Berg et al., 2005). Thus, in Itk-deficient mice, TCR signaling is disrupted, which results in mature CD4- CD8+ (CD8SP) thymocytes that are CD44high, CD62Lhigh, CD122+, and CXCR3+ and that express high levels of the transcription factor, Eomesodermin (Eomes) (Atherly et al., 2006b; Broussard et al., 2006; Weinreich et al., 2010). Recently, it was determined that the development of these innate CD8SP thymocytes in itk-/- mice is dependent on IL-4 produced in the thymic environment by a poorly characterized subset of CD3+ thymocytes expressing the transcriptional regulator, promyelocytic leukemia zinc finger (PLZF) (Gordon et al., 2011; Verykokakis et al., 2010b; Weinreich et al., 2010). Here we show that a sizeable proportion of mature CD4+ CD8- (CD4SP) thymocytes in itk-/- mice also develop as Eomesodermin+ innate T cells. These Eomes+ innate CD4+ T cells are CD44high, CD62Lhigh, CD122+, and CXCR3+ (Atherly et al., 2006b; Broussard et al., 2006; Dubois et al., 2006; Weinreich et al., 2010). Surprisingly, neither CD4SP nor CD8SP innate thymocytes in itk-/- mice are dependent on γδ T cells for their development as was previously hypothesized (Alonzo and Sant'Angelo, 2011). Instead, both subsets of innate itk-/- T cells require the presence of a novel PLZF-expressing, SAP-dependent thymocyte population that is essential for the conversion of conventional CD4+ and CD8+ T cells into Eomesodermin-expressing innate T cells with a memory phenotype. This novel subset of PLZF-expressing SAP-dependent innate T cells preferentially home to the spleen and mesenteric lymph nodes and have a restricted TCR repertoire. Thus, we have christened this subset as CD4+ PLZF + MAIT-like cells. We have characterized multiple subsets of innate T cells that expand in the absence of Itk. Therefore, we were interested in how innate T cells respond to infection. Although Itk KO mice have defects in cytolytic function and cytokine production during an acute infection, these mice are able to clear viral infections (Atherly et al., 2006a; Bachmann et al., 1997). Hence, we hypothesized that Itk-deficient memory CD8+ T cells would be able to provide protection upon a challenge infection. Conversely, we found this not to be true although Itk-deficient memory CD8+ T cells were present in similar frequencies and cell numbers as WT memory CD8+ T cells at 42 days post-infection. Furthermore, Itk-deficient memory CD8+ T cells were able to produce IFNγ and exert cytolytic function upon stimulation. Although the function of Itk-deficient memory CD8+ T cells appeared to be intact, we found that these cells were unable to expand in response to a challenge infection. Remarkably, conventional memory CD8+ T cells lacking Itk were able to expand and form protective memory responses upon challenge. Thus, the inability of Eomes+ innate CD8+ T cells to form protective memory responses does not appear to be intrinsic to cells deficient in Itk. This thesis is divided into six major chapters. The first chapter will provide an introduction to T cell development and the role of Itk in T cell development. Additionally, it will introduce a variety of innate T cell subsets that will be discussed throughout this thesis and will provide an overview of CD4+ and CD8 + T cell differentiation during infection. This section will explain the role of Itk in CD4+ helper T cell differentiation and describe how Itk-deficient CD8+ T cells respond to acute infection. The introduction will also discuss the generation of conventional memory CD8+ T cells. The second chapter will provide the details of the experimental procedures used in this thesis. The third chapter will describe the characterization and development of Eomes+ innate CD4+ T cells that develop in the absence of Itk. Additionally, this chapter will address the subset of PLZF+ innate T cells that induce the expression of Eomes in innate T cells. The fourth chapter will further characterize and explore the development of itk-/- CD4+ PLZF+ MAIT-like T cells. The fifth chapter will examine the role of Eomes + innate CD8+ T cells in protective memory responses. Chapters three through five will display work that is in preparation to be submitted to a peer-reviewed journal. The sixth chapter will discuss the results of this thesis and their implications.
47

Tissue-dependent T Cell Apoptosis and Transcriptional Regulation of Memory CD8+T Cell Differentiation During Viral Infections: A Dissertation

Kapoor, Varun N. 10 December 2013 (has links)
Activation and proliferation of antigen-specific T cells is the hallmark of an anti-viral immune response. Effector T cells generated during an immune response are heterogeneous in regards to their ability to populate the memory pool once the immune response has resolved. Initial T cell activation takes place in the lymphoid organs, after which T cells migrate into the non-lymphoid tissues. The presence of memory T cells at non-lymphoid tissue sites has been shown to be critical for protection against secondary virus challenge. Our lab has previously demonstrated that during and after the resolution of the immune response to Lymphocytic choriomeningitis virus (LCMV) CD8+T cells in the nonlymphoid tissues are more resistant to apoptosis than those in the lymphoid organs. This stability of T cells in the non-lymphoid tissues may be critical in ensuring protection against a secondary virus challenge. Mechanisms regulating tissue-dependent differences in CD8+T cell apoptosis were studied in an acute LCMV infection model. Virus-specific CD8+T cells from lymphoid (spleen, mesenteric lymph nodes (MLN), inguinal lymph nodes (ILN)) and non-lymphoid tissues (peritoneal exudate cells (PEC), fat-pads) were compared for expression of surface antigenic markers known to correlate with a memory phenotype. Non-lymphoid tissues were enriched in IL-7Rhi, KLRG-1lo, CD27hi and CXCR3hi virus-specific CD8+ T cells, and the presence of these antigenic markers correlated with increased memory potential and survival. Transcription factors in addition to cell surface antigens were assessed as correlates of resistance to apoptosis. Virus-specific CD8+T cells in the nonlymphoid tissues were enriched in cells expressing T cell factor-1 (TCF-1), which correlated with increased memory potential and survival. CD8+T cells in the peritoneum of TCF-1-deficient mice had decreased survival during resolution of the immune response to LCMV, suggesting a role for TCF-1 in promoting survival in the non-lymphoid tissues. As an additional mechanism, I investigated whether apoptosis-resistant CD8+T cells migrate to non-lymphoid tissues and contribute to tissue-dependent apoptotic differences. CXCR3+ CD8+T cells resisted apoptosis and accumulated in the lymph nodes of mice treated with FTY720, which blocks the export of lymph node cells into the peripheral tissues. The PECs expressed increased amounts of CXCR3 ligands, CXCL9 and CXCL10, which may have recruited the non-apoptotic cells from the lymph nodes. By adoptively transferring splenic T cells into the spleen or PEC environment I showed that the peritoneal environment through a yet undefined factor promoted survival of CD8+T cells. In this study I have elucidated the mechanisms by which CD8+T cells preferentially survive in the non-lymphoid tissues. I found that non-lymphoid tissues were enriched in memory-phenotype CD8+T cells which were intrinsically resistant to apoptosis irrespective of the tissue environment. Furthermore, apoptosisresistant CD8+T cells may preferentially migrate into the non-lymphoid tissues where the availability of tissue-specific factors may enhance memory cell survival. Few transcription factors have been identified that regulate CD8+T cell effector-memory differentiation during an immune response. In this thesis, I have also studied the mechanism by which the transcription factor Blimp-1 regulates the generation of effector and memory CD8+T cells. Blimp-1 is known to repress a large number of target genes, and ChIP (chromatin immunoprecipitation) sequencing analysis done by Dr. HyunMu Shin in the lab of Dr. Leslie J. Berg identified CD25 (IL-2Rα) and CD27 as potential targets of Blimp-1. I found that Blimp-1-deficient CD8+T cells had sustained expression of CD25 (IL-2Rα) and CD27 during peak and resolution of the immune response to LCMV. By performing adoptive transfers of CD25hi and CD27hi CD8+T cells I showed that CD25 and CD27 expression on CD8+T cells during resolution of the immune response correlates with enhanced survival. Silencing Il2rα and Cd27 expression reduced the Blimp-1-deficient CD8+T cell response, suggesting that sustained expression of CD25 and CD27 was in part responsible for the enhanced CD8+T cell response seen in the Blimp-1-deficient mice. Furthermore, our collaborator Dr. HyunMu Shin showed that CD25 and CD27 are direct targets of Blimp-1, and that Blimp-1 recruits histone modifying enzymes to Il2rα and Cd27 loci to suppress their expression during the peak of the anti-viral immune response. This study identifies one of the mechanisms by which Blimp-1 regulates the balance between generation of effector and memory CD8+T cells. In this thesis work I also studied the function of the transcription factor ROG (Repressor of GATA-3) in regulating in vivo T cell responses during both acute and chronic LCMV infection. ROG-deficient mice had increased CD8+T cell responses during an acute LCMV infection. ROG deficiency also led to the generation of memory T cells with an enhanced recall response compared to WT controls. By using LCMV-specific P14+ TCR transgenic ROG-deficient CD8+T cells these defects were shown to be T cell intrinsic. ROG-deficient mice had enhanced CD8+T cell responses and viral clearance during a persistent high dose LCMV Clone 13 infection. During chronic LCMV infection ROG-deficient mice also had increased lung pathology and mortality. The results indicate that ROG negatively regulates T cell responses and memory generation during both acute and chronic LCMV infection. The studies highlighted in this thesis elucidate the mechanisms promoting CD8+T cell survival in non-lymphoid tissues as well as transcription factormediated regulation of memory CD8+T cell differentiation. Knowledge of this will help us better understand T cell immunity after infections and may eventually help develop better vaccines.
48

Regulation of Type II Responses in Lung Fibrosis and Systemic Autoimmunity: A Dissertation

Brodeur, Tia Bumpus 09 April 2014 (has links)
Preclinical models of lupus indicate that T cell-B cell collaboration drives antinuclear antibody (ANA) production and sustains T cell activation. Autoreactive B lymphocytes are present in the normal repertoire but persist as ignorant or anergic cells. Mechanisms that normally limit T cell activation of autoreactive B cells remain incompletely resolved, but potentially include the absence of autoreactive effector T cell subsets and/or the presence of autoAgspecific regulatory T cells (Tregs). Several studies have addressed this issue by using experimental systems dependent on transgenic autoreactive B cells, but much less is known about the activation of autoreactive B cells present in a polyclonal repertoire. In the second chapter of this thesis, I have explored the role of effector T cells and Tregs using mice that express an inducible pseudoautoAg expressed on B cells and other antigen presenting cells (APCs). In this system, activated Th2 cells, but not naïve T cells, elicit the production of ANAs, but ANA production is severely limited by autoAg-specific Tregs. Bone marrow chimera experiments further demonstrated that this B cell activation is constrained by radioresistant autoantigen-expressing APCs (rAPC) present in the thymus as well as by non-hematopoietic stromal cells located in peripheral lymphoid tissue. Importantly, peripheral rAPC expression of autoAg induced the expansion of a highly effective subset of CD62L+CD69+ Tregs. The third chapter of this thesis focuses on the contribution of CD8+ T cells to fibrosis resulting from sterile lung injury. Type 2 effector production of IL-13 is v a demonstrated requirement in several models of fibrosis, and is routinely ascribed to CD4+ Th2 cells. However, we now demonstrate a major role for pulmonary CD8+ T cells, which mediate an exaggerated wound healing response and fibrosis through robust differentiation into IL-13-producing pro-fibrotic type 2 effectors (Tc2). Remarkably, differentiation of these Tc2 cells in the lung requires IL-21. We further show that the combination of IL-4 and IL-21 skews naïve CD8+ T cells to produce IL-21, which in turn acts in an autocrine manner to support robust IL-13 production. TGF-β negatively regulates production of IL-13 by suppressing CD8+ T cell responsiveness to IL-21. Our data illuminate a novel pathway involved in the onset and regulation of pulmonary fibrosis, and identify Tc2 cells as key mediators of fibrogenesis.
49

Perfil fenotípico e funcional de células Natural Killers induzido por ligantes de receptores Toll-like e células T CD8+ antígeno-específicas em indivíduos expostos e não infectados por HIV-1 / Phenotypic and functional profile of Natural Killer cells induced by Toll-like receptors ligands and antigen-specific CD8+ T cells in HIV-1 exposed uninfected individuals

Lima, Josenilson Feitosa de 14 March 2014 (has links)
Introdução: A resistência a infecção pelo HIV-1 depende de fatores virais, genéticos e imunológicos do hospedeiro, incluindo os componentes da resposta imune inata e adaptativa. As células Natural Killer (NK) e as células T CD8+ são as principais células efetoras que medeiam atividade citotóxica contra células transformadas ou infectadas, que exercem importante papel protetor nos indivíduos expostos e não infectados por HIV-1 (ENI). Objetivo: Avaliar a expressão de receptores de ativação e inibição/exaustão nas células NK e T CD8+, e a capacidade das células NK em secretar citocinas e componentes citotóxicos após estimulação via receptores Toll-like (TLRs), e a resposta de células T CD8+ a peptídeos da Gag do HIV-1 em indivíduos ENI e seus parceiros infectados por HIV-1. Resultados: No grupo ENI foi observado aumento da frequência de células NK CD56bright que expressam moléculas de ativação NKG2D e CD95 na população CD56dim, enquanto no grupo HIV-1 foi mais prevalente a expressão de MIC A/B em ambas populações de células NK, com redução da expressão de NKG2D na população CD56dim. Além disto, foi observado expansão da população de células NK CD56dim que expressam CD94, NKG2C e principalmente de CD57 foi mais prevalente nos indivíduos ENI, com correlação positiva com títulos de anticorpos IgG anti-citomegalovírus humano. Nos indivíduos ENI foi observado que a ativação via TLR-3, TLR-7 ou TLR-7/8 foi capaz de potencializar a expressão de marcadores de desgranulação e de citotoxicidade, CD107a e granzima B, principalmente na população CD56dim, e de IFN-y e TNF nas populações CD56bright e CD56dim. Além disto, somente o grupo ENI, foi detectado aumento da freqüência de células NK secretoras de CD107a, granzima B, IFN-y e TNF, após estimulação com acetato de miristato de forbol e ionomicina. A frequência de expressão de alelos de KIR (killer cell immunoglobulin-like receptors) foi similar entre os grupos analisados. Elevada frequência de células T CD8+ CD38+ e CD8+PD-1+ (programmed cell death protein 1) foi detectado nos grupos ENI e HIV-1, cuja alteração foi observada em todas as fases de maturação celular. Os indivíduos ENI mostraram presença de resposta antígeno-específica de células T CD8+ secretoras de CD107a, granzima B, IFN-y e TNF, semelhante ao grupo HIV-1. Conclusão: Os resultados mostraram que no grupo ENI, as células NK expressam um perfil de ativação, com potente resposta aos estímulos de resposta inata e células NK com perfil de memória. Presença de células TCD8+ antígeno-específica foi evidenciada no grupo ENI, com perfil semelhante, mas de menor magnitude ao detectado no grupo infectado por HIV. Em conjunto, os achados mostraram que no grupo ENI a resposta inata está potencialmente ativa, e que em associação a resposta T CD8+ antígeno-específica podem contribuir para a resistência a infecção pelo HIV-1 / Introduction: Resistance to human immunodeficency virus 1 (HIV-1) is dependent on viral, genetic and immunological host factors, including components of innate and adaptive immune response. Natural Killers cells (NK) and CD8+ T cells are main effectors cells mediating cytotoxic role against transformed or infected cells, playing a crucial role in HIV-1 exposed uninfected individuals (EU). Aim: To evaluate the expression of activation and inhibitory/exhaustion receptors on NK cells and CD8+ T-cells, and to determine the NK cells ability to cytokines and cytotoxic molecules secretion upon Toll-like receptors (TLRs) pathway activation as well as CD8+ T-cells response to HIV Gag peptides in EU individuals and HIV-1 infected partner. Results: Increased frequency of NK CD56bright cells expressing NKG2D and CD95 on CD56dim cells have been observed in EU group, while HIV-1 group was more prevalent MIC A/B expression in both NK cells subsets, with reduced expression of NKG2D in CD56dim cells. Moreover, expansion of NK CD56dim cells expressing CD94, NKG2C, and CD57 was prevalent on ENI group, which positive correlation with anti-human cytomegalovirus IgG serum titers. EU individuals showed that TLR-3, TLR-7 or TLR-7/8 pathway activation was able to enhance CD107a and granzyme B expression in CD56dim cells, and IFN-y and TNF expressions levels in both CD56bright and CD56dim NK cells. Moreover, only in EU group, high frequency of NK cells expressing CD107a, granzyme B, IFN-y and TNF were detected upon phorbol myristate acetate and ionomicyn stimulation. Frequency of KIR alleles (killer cell immunoglobulin-like receptors) was similar between groups. High frequency of CD8+CD38+ and CD8+PD-1+ (programmed cell death protein 1) T-cells were observed in EU and HIV-1 groups, in all stages of cellular differentiation. EU subjects showed presence of antigen-specific response by CD8+ T-cells secreting CD107a, granzyme B, IFN-y and TNF similar to HIV-1 group. Conclusion: The results showed that NK cells in EU subjects express activating profile, with potent ability to innate immune stimuli, as well as NK cells with memory profile. Presence of antigen-specific CD8+ T-cells was detected in EU group, with similar profile, but in less magnitude than HIV-1 group. Taken together, the findings showed an enhanced innate immune response in EU subjects, in association with antigen-specific CD8+ T-cell response can contribute to resistance to HIV-1 infection
50

Caracterização fenotípica e funcional de linfócitos TCD8+ circulantes na síndrome de Sézary / Phenotypic and functional characterization of circulating CD8+ T lymphocytes in Sezary syndrome

Torrealba, Marina Passos 16 September 2016 (has links)
INTRODUÇÃO: A Síndrome de Sézary (SS) é um linfoma cutâneo de células T (LCCT), caracterizado por eritrodermia, linfadenopatia generalizada e presença de células tumorais na pele, linfonodos e sangue periférico. Os linfócitos TCD8+ têm papel fundamental na resposta imune antitumoral, entretanto, há escassos estudos evidenciando seu perfil fenotípico e funcional. Considerando que a resposta imunológica do paciente com SS está suprimida, estratégias para potencializar a imunidade inata e adaptativa com agonistas de receptores Toll-like (TLRs) têm sido exploradas. OBJETIVO: Caracterizar o perfil de marcadores de ativação/inibição das células TCD8+, seus estágios de diferenciação, capacidade de resposta a IL-7/IL-15 e ao agonista de TLR7/TLR8 de pacientes com SS. METODOLOGIA: Foram selecionados 15 pacientes com SS (7 homens e 8 mulheres) com 48-85 anos do Ambulatório de Linfomas Cutâneos, do HC-FMUSP, e um grupo de controle com 24 indivíduos sadios. A análise de marcadores de ativação/inibição e diferenciação celular em células TCD4/TCD8+ do sangue periférico foi realizada por citometria de fluxo. A expressão de marcadores extracelulares e citocinas intracelulares em células mononucleadas do sangue periférico (CMN) após estimulação com o agonista de TLR7/TLR8 foi analisada por citometria de fluxo. Além disto, o efeito de IL-7 e IL-5 em células T foi avaliado pela fosforilação de STAT5, na capacidade de proliferação mitogênica e expressão de BCL-2 em CMNs, como também pelos níveis séricos de IL-7 por citometria de fluxo. RESULTADOS: Os pacientes com SS mostram perfil fenotípico de ativação crônica nos linfócitos TCD8+ periféricos, decorrente do elevadopercentual de células TCD8+ CD38+, redução percentual de TCD8+ CD127+ (IL-7R) e da população naive. Além disso, ocorreu aumento de expressão de PD-1 na população naive de células TCD8+. O marcador de ativação, CD26, até então apenas relacionado com linfócitos TCD4, foi detectado em reduzida percentagem de linfócitos TCD8. A resposta para IL-7/IL-15 parece estar funcionalmente presente tanto nos linfócitos TCD4 quanto nos linfócitos TCD8. Contudo, foi encontrado um perfil diferenciado e heterogêneo de fosforilação de STAT5 assim como de expressão de BCL-2 nos linfócitos TCD8+ de pacientes com SS. O nível sérico de IL-7 reduzido dos pacientes com SS foi inversamente correlacionado com o número absoluto de linfócitos TCD4+. CONCLUSÃO: Os linfócitos TCD8+ dos pacientes com SS encontram-se reduzidos em números absolutos, e possuem um perfil alterado de diferenciação celular e expressão de marcadores extracelulares. A redução percentual da população de TCD8+ naive associada com a presença de moléculas de ativação crônica mostra um perfil de imunosenescência. As células TCD8+ exibem baixa capacidade de resposta aos ligantes de TLR intracelulares, provavelmente devido ao perfil de ativação crônica. Além disso, há resposta parcial dos linfócitos TCD8+ às citocinas ligantes do receptor yc. Nossos resultados evidenciam alterações em linfócitos TCD8+ que debilitam a resposta imune antitumoral e que pode contribuir com a patogênese da síndrome de Sézary / INTRODUCTION: Sézary syndrome (SS) is a cutaneous T cell lymphoma (CTCL), characterized by erythroderma, generalized lymphadenopathy and the presence of tumor cells in the skin, lymph nodes and peripheral blood. The TCD8+ lymphocytes play a key role in anti-tumor immune response, whereas, there are few studies showing its phenotypic and functional profile in SS. Considering that the immune response of SS patient is suppressed, strategies to enhancing the innate and adaptive immunity by Toll-like receptors (TLRs) agonists have been explored. OBJECTIVE: To characterize the profile of activation/inhibition markers of CD8+ T cells, their stages of differentiation, ability of response to IL-7/IL-15 and TLR7/TLR8 agonist of patients with SS. METHODOLOGY: Fifteen SS patients were enrolled (7 men and 8 woman) with 48-85 years from the Clinic of Cutaneous Lymphomas, HC-FMUSP, and a control group of 24 healthy individuals. Analysis of activation/inhibition markers and cellular differentiation in CD4/CD8 T cells from peripheral blood were assessed by flow cytometry. The expression of extracellular markers and intracellular cytokines in mononuclear cells in the peripheral blood (CMN) were evaluated by flow cytometry. Moreover, the effect of IL-7 and IL-15 stimulation in T cells was assessed by the STAT5 phosphorylation, proliferative mitogenic capacity, BCL-2 expression in CMNs as well as serum IL-7 levels by flow cytometry. RESULTS: Patients with SS show a phenotypic CD8 T peripheral lymphocytes profile of chronic activation, due to the high percentage of CD8+CD38+ T cells, reduced percentage of CD8+CD127+ (IL-7R) and naïve population. Furthermore, it was observed an increased PD-1 expression in the naïve CD8+ T cells. The activation marker CD26, previously only associated with CD4 T lymphocyte, was detected at decreased percentage in CD8 T lymphocytes. The TLR7/TLR8 agonist did not affect the IFN-? and TNF secretion of CD8 T lymphocytes of SS patients, in contrast to the control group. The response to IL-7/IL-15 appears to be functional in both CD4 and CD8 T lymphocytes. However, it was founded a differentiated and heterogeneous profile of STAT5 phosphorylation and Bcl-2 expression in the CD8 T lymphocytes in SS patients. The reduced IL-7 serum of patients with SS was inversely correlated with the absolute number of CD4 T lymphocytes. CONCLUSION: CD8 T lymphocytes of patients with SS are reduced in absolute numbers, and show an altered cellular differentiation profile and extracellular markers expression. The reduced percentage of CD8 naïve population associated with chronic activation of molecules reveals an immunosenescence profile. The CD8 T cells exhibit low ability to ligands of intracellular TLR receptors, probably due to chronic activation profile. In addition, there are partial response of CD8 T lymphocytes to the cytokine receptor ?c. Our results show disturbance in CD8 T lymphocytes that may impair the anti-tumor response contributing to the pathogenesis of Sézary syndrome

Page generated in 0.0521 seconds