Spelling suggestions: "subject:"T-Cell 1receptor"" "subject:"T-Cell 2receptor""
61 |
Regulation of Immune Cell Activation and Functionby the nBMPp2 Protein andthe CD5 Co-ReceptorFreitas, Claudia Mercedes 01 April 2019 (has links)
According to the centers for disease control and prevention (CDC) and the world healthorganization (WHO), heart disease and immune related diseases such as diabetes and cancer areamong the leading causes of death around the world. Thus, the regulation of the function ofimmune cell plays a key role in health and disease. Calcium (Ca2+) ions play a critical role inimmune cell activation, function and in a robust immune response. Defects in Ca2+ signalinginfluences the development of cardiac disease, Alzheimer disease, immune cell metabolism,muscle dysfunction, and cancer. Each immune cell is unique in its activation and function,making it relevant to understand how activation of each type of immune cell is regulated. Herewe describe the role of the nBMP2 protein in macrophage activation and function and the role ofthe CD5 co-receptor in helper T cell activation and function.The nuclear bone morphogenetic protein 2 (nBMP2) is the nuclear variant of the bonemorphogenetic protein 2 (BMP2), a growth factor important in heart development, neurogenesis,bone, cartilage and muscle development. To better understand the function of nBMP2, transgenicnBMP2 mutant mice were generated. These mice have a slow muscle relaxation and cognitivedeficit caused in part by abnormal Ca2+ mobilization. Mutant nBMP2 mice also have an impairedsecondary immune response to systemic bacterial challenge. Here we have further characterizedmacrophage activation and function from mutant nBMP2 mice before and after bacterialinfection. We describe how nBMP2 influences the Ca2+ mobilization response and phagocytosisin macrophages, revealing a novel role of the nBMP2 protein in immune cell regulation.CD5 is a surface marker on T cells, thymocytes, and the B1 subset of B cells. CD5 isknown to play an important role during thymic development of T cells. CD5 functions as anegative regulator of T cell receptor (TCR) signaling and fine tunes the TCR signaling response.Here we describe our characterization of CD5 regulation of Ca2+ signaling in naïve helper Tcells. We also outline our findings examining how CD5-induced changes in helper T cellactivation influence other biological processes such as immune cell metabolism, the diversity ofthe gut microbiome, and cognitive function and behavior. Thus, this work elucidates theinfluence of the CD5 co-receptor on the functional outcomes in multiple systems when CD5 isaltered.
|
62 |
Mechanisms of IFN-gamma-mediated Resistance against Development of Toxoplasmic EncephalitisWang, Xisheng 07 March 2007 (has links)
Toxoplasma gondii, an obligate intracellular protozoan parasite, establishes a latent, chronic infection by forming cysts preferentially in the brain after replication of tachyzoites in various organs during the acute stage of infection. Chronic infection with T. gondii is one of the most common parasitic diseases in humans. The immune system is required for maintaining the latency of chronic infection. Reactivation of infection can occur in immunocompromised individuals, such as AIDS patients, which results in the development of life-threatening toxoplasmic encephalitis (TE). IFN-gamma-dependent, cell mediated immune responses play an essential role in preventing the reactivation of chronic infection of T. gondii in the brain. In my dissertation study, we examined the mechanisms of IFN-gamma-mediated prevention of TE by using models of reactivation of chronic infection in BALB/c mice. This strain of mouse is genetically resistant to T. gondii infection and establishes a latent chronic infection as do immunocompetent humans, and therefore provides an excellet model for this purpose.
Our laboratory previously demonstrated that both T cells and IFN-gamma-producing non-T cells are required for genetic resistance of BALB/c mice against development of TE. However, the function of T cells required for the resistance is still unclear. Therefore, in the present study, we examined whether IFN-gamma production or perforin-mediated cytotoxicity of T cells play an important role in their protective activity against TE. Immune T cells were obtained from infected IFN-gamma-knockout (IFN-g-/-), perforin-knockout (PO), and wild-type (WT) BALB/c mice, and transferred into infected, sulfadiazine-treated athymic nude mice which lack T cells but have IFN-gamma-producing non-T cells. Control nude mice that had not received any T cells developed severe TE due to reactivation of infection and died after discontinuation of sulfadiazine treatment. Animals that had received immune T cells from either PO or WT mice did not develop TE and survived. In contrast, nude mice that had received immune T cells from IFN-gamma-/- mice developed severe TE and died as early as control nude mice. T cells obtained from spleens of the animals that had received either PO or WT T cells both produced large amounts of IFN-gamma following stimulation with T. gondii antigens in vitro. In addition, the amounts of IFN-gamma mRNA expressed in the brains of PO T-cell recipients did not differ from those of WT T-cell recipients. These results indicate that IFN-gamma production, but not perforin-mediated cytotoxic activity, by T cells is required for prevention of TE in genetically resistant BALB/c mice.
In our attempt to identify a T cell population(s) that produces IFN-gamma in the brain and plays an important role for prevention of TE, we analyzed T cell receptor (TCR) Vb chain usage in T cells expressing IFN-gamma in the brains of infected BALB/c mice. We found T cells bearing TCR V beta8 chain to be the most frequent IFN-g-producing population in the brains of infected animals. To examine the role of IFN-gamma production by this T cell population for prevention of TE, V beta8+ immune T cells purified from spleens of infected BALB/c and IFN-g-/- mice were transferred into infected, sulfadiazine-treated athymic nude mice. After discontinuation of sulfadiazine treatment, control nude mice that had not received any T cells and animals that had received Vb8+ T cells from IFN-g-/- mice all died due to reactivation of infection (TE). In contrast, animals that had received the cells from WT mice survived. These results indicate that IFN-gamma production by Vb8+ T cells in the absence of any other T cell population can prevent reactivation of infection. Thus, V beta8+ T cells play a crucial role in genetic resistance of BALB/c mice to TE through their production of IFN-gamma. When V beta8+ immune T cells were divided into CD4+ and CD8+ subsets, a potent protective activity was observed only in the CD8+ subset whereas a combination of both subsets provided greater protection than did the CD8+Vb8+ population alone. These results indicate that CD8+ subset of V beta8+ T cells is a major afferent limb of IFN-gamma-mediated resistance of BALB/c mice against TE, although the CD4+ subset of the T cell population works additively or synergistically with the CD8+V beta8+ population.
T cells need to enter into the brains of infected mice to demonstrate their protective activity against TE. This migration is mediated, in part, by endothelial adhesion molecules. Since IFN-gamma is essential for preventing reactivation of chronic infection with this parasite in the brain, we examined whether this cytokine plays an important role in expression of lymphocyte and endothelial adhesion molecules and recruitment of T cells into the brain during chronic infection with T. gondii using IFN-g-/- and WT BALB/c mice. Although the number of cerebral vessels expressing intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) increased in both WT and IFN-g-/- mice following infection, there were more VCAM-1+ vessels in brains of infected WT than infected IFN-g-/- mice; in contrast, numbers of ICAM-1+ vessels did not differ between strains. We did not detect endothelial E-selectin, P-selectin, MAdCAM-1 or PNAd in any of the brains. Significantly fewer CD8+ T cells were recruited into brains of infected IFN-g-/- than WT mice. Treatment of infected IFN-g-/- mice with recombinant IFN-gamma restored the expression of VCAM-1 on their cerebral vessels and recruitment of CD8+ T cells into their brains, confirming an importance of this cytokine for up-regulation of VCAM-1 expression and CD8+ T cell trafficking. In infected WT and IFN-g-/- animals, almost all cerebral CD8+ T cells had an effector/memory phenotype (LFA-1high, CD44high and CD62Lneg) and approximately 38% were positive for a4b1 integrin (the ligand for VCAM-1). In adoptive transfer of immune spleen cells, pre-treatment of the cells with a monoclonal antibody against a4 integrin markedly inhibited recruitment of CD8+ T cells into the brain of chronically infected wild-type mice. These results indicate that IFN-g-induced expression of endothelial VCAM-1 and its binding to a4b1 integrin on CD8+ T cells is important for recruitment of the T cells into the brain during the chronic stage of T. gondii infection. Since we found strong expression of ICAM-1 on endothelia and LFA-1 on T cells in the brains of infected mice, LFA-1/ICAM-1 interaction, in addition to a4b1 integrin/VCAM-1 interaction, may also be involved in this process. As mentioned earlier, CD8+ T cells are crucial for prevention of TE in BALB/c mice. Therefore, IFN-gamma-mediated expression of VCAM-1 and its binding to a4b1 integrin for recruitment of CD8+ T cells may play a critical role in genetic resistance of BALB/c mice to development of TE. / Ph. D.
|
63 |
Generation of dual T cell receptor (TCR) T cells by TCR gene transfer for adoptive T cell therapySommermeyer, Daniel 10 February 2010 (has links)
Die Herstellung von T-Zellen mit definierten Spezifitäten durch den Transfer von T-Zellrezeptor (TCR) Genen ist eine effiziente Methode, um Zellen für eine Immuntherapie bereitzustellen. Eine besondere Herausforderung ist dabei, ein ausreichend hohes Expressionsniveau des therapeutischen TCR zu erreichen. Da T-Zellen mit einem zusätzlichen TCR ausgestattet werden, entsteht eine Konkurrenzsituation zwischen dem therapeutischen und dem endogenen TCR. Bevor diese Arbeit begonnen wurde war nicht bekannt, welche TCR nach einem Gen-Transfer exprimiert werden. Daher haben wir Modelle etabliert, in denen TCR Gene in Maus und humane T-Zellen mit definierten endogenen TCR transferiert wurden. Die Expression beider TCR wurde mithilfe von Antikörpern und MHC-Multimeren analysiert. Diese Modelle haben gezeigt, dass bestimmte TCR andere TCR von der Zelloberfläche verdrängen können. Dies führte in einem Fall zu einer vollständigen Umkehr der Antigenspezifität. Aufgrund dieser Ergebnisse haben wir das Konzept von „starken“ (gut exprimierten) und „schwachen“ (schlecht exprimierten) TCR vorgeschlagen. Zusätzlich wurde die Verdrängung „schwacher“ und „starker“ humaner TCR durch Maus TCR beobachtet. Parallel dazu wurde berichtet, dass die konstanten (C) Regionen von Maus TCR für die erhöhte Expression auf humanen Zellen verantwortlich sind. Dies führte zu einer Strategie zur Verbesserung der Expression humaner TCR, die auf dem Austausch der humanen C-Regionen durch die von Maus TCR basiert (Murinisierung). Ein Problem ist dabei die mögliche Immunogenität dieser hybriden Konstrukte. Deshalb haben wir jene Bereiche der Maus C-Regionen identifiziert, die für die erhöhte Expression verantwortlich sind. In der TCRalpha Kette wurden vier und in der TCRbeta Kette fünf Aminosäuren gefunden, die ausreichend für diesen Effekt waren. Primäre humane T-Zellen mit TCR, die diese neun „Maus“ Aminosäuren enthielten, zeigten eine bessere Funktionalität als T-Zellen mit Wildtyp TCR. / The in vitro generation of T cells with a defined antigen specificity by T cell receptor (TCR) gene transfer is an efficient method to create cells for immunotherapy. One major challenge of this strategy is to achieve sufficiently high expression levels of the therapeutic TCR. As T cells expressing an endogenous TCR are equipped with an additional TCR, there is a competition between therapeutic and endogenous TCR. Before this work was started, it was not known which TCR is present on the cell surface after TCR gene transfer. Therefore, we transferred TCR genes into murine and human T cells and analyzed TCR expression of endogenous and transferred TCR by staining with antibodies and MHC-multimers. We found that some TCR have the capability to replace other TCR on the cell surface, which led to a complete conversion of antigen specificity in one model. Based on these findings we proposed the concept of ‘‘strong’’ (well expressed) and “weak” (poorly expressed) TCR. In addition, we found that a mouse TCR is able to replace both “weak” and “strong” human TCR on human cells. In parallel to this result, it was reported that the constant (C)-regions of mouse TCR were responsible for the improved expression of murine TCR on human cells. This led to a strategy to improve human TCR by exchanging the C-regions by their murine counterparts (murinization). However, a problem of these hybrid constructs is the probable immunogenicity. Therefore, we identified the specific parts of the mouse C-regions which are essential to improve human TCR. In the TCRalpha C-region four and in the TCRbeta C-region five amino acids were identified. Primary human T cells modified with TCR containing these nine “murine” amino acids showed an increased function compared to cells modified with wild type TCR. For TCR gene therapy the utilization of these new C-regions will reduce the amount of foreign sequences and thus the risk of immunogenicity of the therapeutic TCR.
|
64 |
A new safeguard eliminates T cell receptor gene-modified auto-reactive T cells after adoptive therapyKieback, Elisa 23 October 2008 (has links)
Der adoptive Transfer von TZR-modifizierten T Zellen ist mit potentiellen Risiken verbunden. Autoimmunreaktionen können auftreten, wenn Tumor-assoziierte Antigene auf normalem Gewebe erkannt werden, Fehlpaarung der TZR-Ketten zur Bildung eines autoreaktiven Rezeptors führen oder ein sonst anerger auto-reaktiver endogener Rezeptor aktiviert wird. Auch besteht das Risiko der malignen Transformation der Zelle durch Insertionsmutagenese. Daher ist es notwendig, die transferierten T Zellen im Fall schwerer Nebenwirkungen eliminieren zu können. Derzeit verfügbare Sicherheitsmechanismen sind für die Therapie mit TZR-modifizierten T Zellen ungeeignet. In dieser Arbeit wurde ein neuer Sicherheitsansatz entwickelt, der auf einem TZR-intrinsischen Depletionsmechanismus beruht und TZR-veränderte T Zellen eliminieren kann. Durch Einfügen eines myc-tags in murine (OT-I, P14) und humane (gp100) TZRs konnten TZR-exprimierende T Zellen in vitro und in vivo mittels eines myc-spezifischen Antikörpers depletiert werden. Die T Zellen behielten vergleichbare Funktionalität hinsichtlich Antigenerkennung und Zytokinsekretion wie Zellen, die den Wild-Typ Rezeptor exprimierten. Die Depletion adoptiv transferierter T Zellen verhinderte lethalen Diabetes in einem Mausversuch. Im verwendeten Modell wurden Splenozyten, die einen myc-getagten OT-I TZR exprimierten, in RIP-mOVA Mäuse injiziert, welche in den Inselzellen des Pankreas das OT-I-spezifische Antigen Ovalbumin exprimieren. Zerstörung der Inselzellen durch die T-Zellen induzierte lethalen Diabetes in unbehandelten Mäusen. Tiere, denen ein myc-spezifischer Antikörper verabreicht wurde, zeigten keine Symptome. Dieser neuartige Sicherheitsmechanismus erlaubt es, adoptive T Zelltherapie abzubrechen, falls schwere Nebenwirkungen auftreten. Im Gegensatz zu früheren Strategien muss kein zusätzliches Sicherheitsgen eingebaut werden und die Sicherheit des Ansatzes wird durch Verlust oder Herunterregulierung des Transgens nicht beeinflusst. / Adoptive transfer of TCR gene-modified T lymphocytes into patients is associated with potential risk factors. First, auto-immunity may occur if a tumor-associated antigen is targeted on normal tissue, if TCR chain mispairing leads to the formation of an auto-reactive receptor or if an otherwise anergic endogenous receptor specific for an auto-antigen becomes activated. Second, retroviral integration could lead to malignant transformation of the T cell. Therefore, it is essential to have the possibility to deplete the transferred T cells in vivo in case of severe side effects. The available safety modalities comprise disadvantages rendering them less feasible for the application in therapy with TCR gene-modified T cells. In this study, a safeguard based on a TCR-intrinsic depletion mechanism has been developed that eliminates auto-reactive TCR-redirected T cells. By introducing a myc-tag into the murine (OT-I, P14) or human (gp100) TCRs it was possible to deplete TCR-expressing T cells in vitro and in vivo with a myc-specific antibody. The T cells maintained equal function compared to cells expressing the wild-type receptor as shown by antigen binding and cytokine secretion. Importantly, the in vivo depletion of adoptively transferred T cells prevented disease in an auto-immune mouse model. Here, splenocytes transduced with a myc-tagged OT-I TCR were injected into RIP-mOVA mice expressing the OT-I-specific antigen ovalbumin in the pancreatic beta-cells. Destruction of these cells by the adoptively transferred T cells led to severe diabetes in untreated mice. Animals receiving a myc-specific antibody after T cell transfer showed no increase in blood glucose levels. The developed safeguard allows termination of adoptive therapy in case of severe side-effects. The strategy is superior to previous ones as it relies on a TCR-intrinsic mechanism which does not require introduction of an additional gene and safety is not hampered by loss or low expression of the transgene.
|
65 |
Liniová plasticita fyziologických a maligních lymfocytárních prekursorů / Lineage plasticity in normal and malignant lymphocyte precursorsRezková Řezníčková, Leona January 2012 (has links)
Klasické schéma vývoje hematopoetických buněk předpokládá časné oddělení lymfoidního a myeloidního prekurzoru. V poslední době jsou navrhovány složitější modely, které předpokládají větší flexibilitu hematopoezy a navrhují existenci progenitorů s lymfoidním i myeloidním potenciálem. Akutní hybridní leukémie jsou malignity, které podle různých kritérií nelze jednoznačně zařadit k lymfoidní nebo k myeloidní linii a jejichž chování spíše dává za pravdu novým modelům hematopoezy. Předkládaná práce se zabývala především výzkumem dětských leukémií s přesmykem z lymfoidní do myeloidní linie během indukční léčby. Jedná se o rozsáhlý projekt, v jehož rámci si diplomová práce si kladla za úkol určit liniové zařazení leukemických blastů pomocí detekce přestaveb genů pro imunoglobuliny a T-buněčné receptory (TCR). Potvrdili jsme, že myeloidní buňky derivované v průběhu léčby pochází u všech pacientů z původního lymfoidního klonu. Dále jsme u těchto případů zkoumali expresi vytipovaných genů ve srovnání s běžnými druhy leukémií. Třetí částí práce byl výzkum prognostického významu přítomnosti přestaveb TCR (a tedy příslušnosti k lymfoidní linii) u leukémií z T-lymfoidní řady.
|
66 |
Avaliação da função tímica em pacientes com diabetes mellitus tipo 1 submetidos ao transplante autólogo de células-tronco hematopoéticas / Evaluation of thymic function in type 1 diabetes mellitus patients following autologous hematopoietic stem cell transplantation.Azevedo, Júlia Teixeira Cottas de 19 August 2013 (has links)
O diabetes mellitus tipo 1 (DM-1) é uma doença autoimune órgão-específica caracterizada pela destruição seletiva das células pancreáticas produtoras de insulina. A imunossupressão em altas doses seguida do transplante autólogo de células-tronco hematopoéticas (TACTH) constitui uma alternativa terapêutica recente e promissora para o DM-1 recém-diagnosticado, impedindo a progressão da destruição das células pancreáticas produtoras de insulina e induzindo independência insulínica por um período prolongado na maioria dos pacientes. O princípio dessa terapia baseia-se na eliminação das células autorreativas pela imunossupressão intensa e na reconstituição de um sistema imunológico novo e tolerante após o transplante. Com o objetivo de avaliar a função do timo e sua contribuição na geração do repertório de células T nos pacientes com DM-1 após o TACTH, nesse trabalho foram avaliados os níveis de T cell receptor excision circles (TRECs) em células T do sangue periférico e a diversidade do repertório de células T dos pacientes com DM-1 (n=23) antes e em diversos períodos após o transplante. A quantificação absoluta dos níveis de TRECs (número de moléculas de TRECs/100g de DNA) foi realizada pela técnica de PCR em tempo real e a avaliação do repertório de células T foi realizada pela técnica de TCRBV CDR3 Spectratyping. Dentre os vinte e três pacientes, vinte alcançaram a independência insulínica por períodos variáveis de tempo e três não responderam ao tratamento. Não foi observada a restrição do repertório de células T nos pacientes com DM-1 no período pré-transplante, ou seja, quando recém-diagnosticados. Foram identificadas cinco famílias V (7, 18, 19, 20 e 22) em expansão clonal nos pacientes com DM-1. As famílias V 7, 18, 19, 20 apresentaram-se em expansão clonal antes do transplante e se mantiveram com frequência elevada após o transplante, enquanto a família V 22 apresentou aumento da frequência somente nos períodos mais tardios após o transplante. Nos primeiros meses após o transplante, houve redução do número de moléculas de TRECs e restrição do repertório de células T. Contudo, um ano após o transplante, o número de moléculas de TRECs atingiram valores normais e o repertório de células T apresentou-se com ampla diversidade. Nossos resultados mostraram que o TACTH foi capaz de induzir mudanças na composição do repertório de células T dos pacientes com DM-1 após a terapia de IAD/TACTH, evidenciadas por alterações qualitativas e quantitativas dos picos de CDR3 do TCR, sugerindo a reconstituição de um repertório de células T diverso até dois anos pós-transplante. Embora tenha ocorrido reativação da função tímica após o transplante, evidenciada pelo aumento dos níveis de TRECs de um ano e meio a cinco anos pós-transplante, a diversidade do repertório das células T diminuiu a partir de dois anos e meio pós-transplante, sugerindo uma reconstituição tímica de novo de células T naive que expressam preferencialmente algumas cadeias V. Estas evidências imunológicas poderiam explicar a melhora clínica (independência insulínica) temporária observada na maioria dos pacientes após a terapia de IAD/TACTH. / Type 1 diabetes mellitus (T1D) is an organ-specific autoimmune disease characterized by insulin-producing pancreatic cell destruction. High-dose immunosuppression followed by autologous hematopoietic stem cell transplantation (AHSCT) is a recent and promising therapeutic approach for treatment of T1D, preventing the progress of destruction of pancreatic cells and inducing insulin independence for a prolonged period in most patients. The rationale of the AHSCT is based on the elimination of autoreactive cells by the intense immunosuppression and on the reconstitution of a new and tolerant immune system after transplantation. Aiming at assessing the thymic role in the production of new T cell repertoire in T1D patients after AHSCT, in this study was evaluated the levels of T cell receptor excision circles (TRECs) in T cells of peripheral blood as well as the clonality and diversity of T cell repertoire in T1D patients (n=23) before and several periods after transplantation. The absolute quantification of TRECs levels (number of molecules of TRECs/100ng of DNA) was performed by real-time PCR and the analysis of T cell repertoire was performed by TCRBV CDR3 Spectratyping. Among the twenty-three patients, twenty achieved insulin independence for variable periods and three did not respond to the treatment. The T cell repertoire in T1D patients was not restricted in pre-transplantation, i.e., when newly diagnosed. It was identified five V families (7, 18, 19, 20 e 22) in the clonal expansion in T1D patients. The V families 7, 18, 19, 20 were in clonal expansion before transplantation and maintained with high frequency after transplantation, whereas the V 22 family increased its frequency only in the later periods after transplantation. It was observed that the numbers of molecules of TRECs decreased and the T cell repertoire was restricted in the early months after transplantation. However, the levels of TRECs were normalized and the T cell repertoire showed diversity one year after transplantation. Our results indicate that AHSCT was able to induce changes in the composition of the T cell repertoire of patients after AHSCT, evidenced by qualitative and quantitative changes in the composition of T-cell receptor -chain CDR3 peaks, suggesting the reconstitution of diverse T cell repertoire up to two years after transplantation. Although there was reactivation of thymic function after transplantation, as evidenced by increased levels of TRECs from one and a half year to five years after transplantation, the diversity of the T cells repertoire decreased from two and a half years after transplantation, suggesting a reconstruction of new naive T cells that preferentially express some V chains. These immunological evidences could explain the temporary clinical improvement (insulin independence) observed in most patients after IAD / AHSCT therapy.
|
67 |
Shb and Its Homologues: Signaling in T Lymphocytes and FibroblastsLindholm, Cecilia January 2002 (has links)
<p>Stimulation of the T cell receptor (TCR) induces tyrosine phosphorylation of numerous intracellular proteins, leading to activation of the interleukin-2 (IL-2) gene in T lymphocytes. Shb is a ubiquitously expressed adapter protein, with the ability to associate with the T cell receptor and several signaling proteins in T cells, including: the TCR ζ-chain, LAT, PLC-γ1, Vav, SLP-76 and Gads. Jurkat T cells expressing Shb with a mutation in the SH2 domain, exhibited reduced phosphorylation of several proteins and abolished activation of the MAP kinases ERK1, ERK2 and JNK, upon CD3 stimulation. The TCR induced Ca<sup>2+</sup> response in these cells was abolished, together with the activation of the IL-2 promoter via the transcription factor NFAT. Consequently, IL-2 production was also perturbed in these cells, compared to normal Jurkat T cells. Shb was also seen to associate with the β and γ chains of the IL-2 receptor, upon IL-2 stimulation, in T and NK cells. This association occurred between the Shb SH2 domain and Tyr-510 of the IL-2R β chain. The proline-rich domains of Shb were found to associate with the tyrosine kinases JAK1 and JAK3, which are important for STAT-mediated proliferation of T and NK cells upon IL-2 stimulation. Shb was also found to be involved in IL-2 mediated regulation of apoptosis. These findings indicate a dual role for Shb in T cells, where Shb is involved in both T cell receptor and IL-2 receptor signaling. </p><p>A Shb homologue, Shf was identified, and seen to associate with the PDGF-α-receptor. Shf shares high sequence homology with Shb and a Shd (also of the Shb family) in the SH2 domain and in four motifs containing putative tyrosine phosphorylation sites. When Shf was overexpressed in fibroblasts, these cells displayed significantly lower rates of apoptosis than control cells in the presence of PDGF-AA. These findings suggest a role for the novel adapter Shf in PDGF-receptor signaling and regulation of apoptosis.</p>
|
68 |
Clinical and ex-vivo studies on the thymotropic properties of the somatotrope growth hormone (GH) / insulin-like growth factor 1 (IGF-1) axisKermani, Hamid 16 February 2011 (has links)
The objective of this thesis was to investigate the effects of the
somatotrope GH/IGF-1 axis upon the thymus. This work included two parts:
1. Translational research study:
Thymus function in adult GH deficiency (AGHD) with and without GH treatment
Background: Despite age-related adipose involution, T cell generation in the thymus (thymopoiesis) is maintained beyond puberty in adults. In rodents, growth hormone (GH), insulin-like growth factor-1 (IGF-1), and GH secretagogues reverse agerelated changes in thymus cytoarchitecture and increase thymopoiesis. GH administration also enhances thymic mass and function in HIV-infected patients. Until now, thymic function has not been investigated in adult GH deficiency (AGHD). The objective of this clinical study was to evaluate thymic function in AGHD, as well as the repercussion upon thymopoiesis of
GH treatment for restoration of GH/IGF-1 physiological levels.
Methodology/Principal Findings: Twenty-two patients with documented AGHD were enrolled in this study. The following parameters were measured: plasma IGF-1 concentrations, signal-joint T-cell receptor excision circle (sjTREC) frequency, and
sj/b TREC ratio. Analyses were performed at three time points: firstly on GH treatment at maintenance dose, secondly one month after GH withdrawal, and thirdly one month after GH resumption. After 1-month interruption of GH treatment, both
plasma IGF-1 concentrations and sjTREC frequency were decreased (p,0.001). Decreases in IGF-1 and sjTREC levels were correlated (r = 0.61, p,0.01). There was also a decrease in intrathymic T cell proliferation as indicated by the reduced sj/b
TREC ratio (p,0.01). One month after reintroduction of GH treatment, IGF-1 concentration and sjTREC frequency regained a level equivalent to the one before GH withdrawal. The sj/b TREC ratio also increased with GH resumption, but did not return to the level measured before GH withdrawal.
Conclusions: In patients with AGHD under GH treatment, GH withdrawal decreases thymic T cell output, as well as intrathymic T cell proliferation. These parameters of thymus function are completely or partially restored one month after GH resumption. These data indicate that the functional integrity of the somatotrope GH/IGF-1 axis is important for the maintenance of a normal thymus function in human adults.
2. Fundamental study:
intrathymic expression of members of the GH/IGF-1 axis and effects of GH on T-cell differentiation in murine fetalthymic organ cultures (FTOC).
We here address the question of expression and role of GH/IGF axis in the thymus.
Methods: Using RT-qPCR, the expression profile of various components of the somatotrope
GH/IGF axis was measured in different thymic cell types and during thymus embryogenesis in Balb/c mice. Effect of GH on T-cell differentiation was explored through thymic organotypic culture.
Results: Transcription of Gh, Igf1, Igf2 and their related receptors predominantly occurred in
thymic epithelial cells (TEC), while a low level of Gh and Igf1r transcription was also evidenced in thymic T cells (thymocytes). Gh, Ghr, Ins2, Igf1, Igf2, and Igfr1, displayed distinct expression profiles depending on the developmental stage. The protein concentration of IGF-1 and IGF-2 were in accordance with the profile of their gene expression. In fetal thymus organ cultures (FTOC) derived from Balb/c mice, treatment with exogenous GH resulted in a significant increase of double negative CD4-CD8- T cells and CD4+ T cells, with a concomitant decrease in double positive CD4+CD8+ T cells. These changes were inhibited by concomitant treatment with GH and GHR antagonist pegvisomant. However, GH treatment also induced a significant decrease in FTOC Gh, Ghr and Igf1 expression.
Conclusion: These data show that the thymotropic properties of the somatotrope GH/IGF-1 axis involve an interaction between exogenous GH and GHR expressed by TEC. Since thymic IGF-1 is not increased by GH treatment, the effects of GH upon T-cell differentiation could implicate a different local growth factor or cytokine.
|
69 |
Shb and Its Homologues: Signaling in T Lymphocytes and FibroblastsLindholm, Cecilia January 2002 (has links)
Stimulation of the T cell receptor (TCR) induces tyrosine phosphorylation of numerous intracellular proteins, leading to activation of the interleukin-2 (IL-2) gene in T lymphocytes. Shb is a ubiquitously expressed adapter protein, with the ability to associate with the T cell receptor and several signaling proteins in T cells, including: the TCR ζ-chain, LAT, PLC-γ1, Vav, SLP-76 and Gads. Jurkat T cells expressing Shb with a mutation in the SH2 domain, exhibited reduced phosphorylation of several proteins and abolished activation of the MAP kinases ERK1, ERK2 and JNK, upon CD3 stimulation. The TCR induced Ca2+ response in these cells was abolished, together with the activation of the IL-2 promoter via the transcription factor NFAT. Consequently, IL-2 production was also perturbed in these cells, compared to normal Jurkat T cells. Shb was also seen to associate with the β and γ chains of the IL-2 receptor, upon IL-2 stimulation, in T and NK cells. This association occurred between the Shb SH2 domain and Tyr-510 of the IL-2R β chain. The proline-rich domains of Shb were found to associate with the tyrosine kinases JAK1 and JAK3, which are important for STAT-mediated proliferation of T and NK cells upon IL-2 stimulation. Shb was also found to be involved in IL-2 mediated regulation of apoptosis. These findings indicate a dual role for Shb in T cells, where Shb is involved in both T cell receptor and IL-2 receptor signaling. A Shb homologue, Shf was identified, and seen to associate with the PDGF-α-receptor. Shf shares high sequence homology with Shb and a Shd (also of the Shb family) in the SH2 domain and in four motifs containing putative tyrosine phosphorylation sites. When Shf was overexpressed in fibroblasts, these cells displayed significantly lower rates of apoptosis than control cells in the presence of PDGF-AA. These findings suggest a role for the novel adapter Shf in PDGF-receptor signaling and regulation of apoptosis.
|
70 |
Modified Glycopeptides Targeting Rheumatoid Arthritis : Exploring molecular interactions in class II MHC/glycopeptide/T-cell receptor complexesAndersson, Ida E. January 2011 (has links)
Rheumatoid arthritis (RA) is an autoimmune inflammatory disease that leads to degradation of cartilage and bone mainly in peripheral joints. In collagen-induced arthritis (CIA), a mouse model for RA, activation of autoimmune CD4+ T cells depends on a molecular recognition system where T-cell receptors (TCRs) recognize a complex between the class II MHC Aq protein and CII259-273, a glycopeptide epitope from type II collagen (CII). Interestingly, vaccination with the Aq/CII259-273 complex can relieve symptoms and cause disease regression in mice. This thesis describes the use of modified glycopeptides to explore interactions important for binding to the Aq protein and recognition by autoimmune T-cell hybridomas obtained from mice with CIA. The CII259-273 glycopeptide was modified by replacement of backbone amides with different amide bond isosteres, as well as substitution of two residues that anchor the glycopeptide in prominent pockets in the Aq binding site. A three-dimensional structure of the Aq/glycopeptide complex was modeled to provide a structural basis for interpretation of the modified glycopeptide’s immunological activities. Overall, it was found that the amide bond isosteres affected Aq binding more than could be explained by the static model of the Aq/glycopeptide complex. Molecular dynamics (MD) simulations, however, revealed that the introduced amide bond isosteres substantially altered the hydrogen-bonding network formed between the N-terminal 259-265 backbone sequence of CII259-273 and Aq. These results indicated that the N-terminal hydrogen-bonding interactions follow a cooperative model, where the strength and presence of individual hydrogen bonds depended on the neighboring interactions. The two important anchor residues Ile260 and Phe263 were investigated using a designed library of CII259-273 based glycopeptides with substitutions by different (non-)natural amino acids at positions 260 and 263. Evaluation of binding to the Aq protein showed that there was scope for improvement in position 263 while Ile was preferred in position 260. The obtained SAR understanding provided a valuable basis for future development of modified glycopeptides with improved Aq binding. Furthermore, the modified glycopeptides elicited varying T-cell responses that generally could be correlated to their ability to bind to Aq. However, in several cases, there was a lack of correlation between Aq binding and T-cell recognition, which indicated that the interactions with the TCRs were determined by other factors, such as presentation of altered epitopes and changes in the kinetics of the TCR’s interaction with the Aq/glycopeptide complex. Several of the modified glycopeptides were also found to bind well to the human RA-associated DR4 protein and elicit strong responses with T-cell hybridomas obtained from transgenic mice expressing DR4 and the human CD4 co-receptor. This encourages future investigations of modified glycopeptides that can be used to further probe the MHC/glycopeptide/TCR recognition system and that also constitute potential therapeutic vaccines for treatment of RA. As a step towards this goal, three modified glycopeptides presented in this thesis have been identified as candidates for vaccination studies using the CIA mouse model.
|
Page generated in 0.0495 seconds