21 |
Vliv interferonu gama (IFN-\recke{gamma})a specifických polyklonálních protilátek na průběh experimentální perorální infekce \kur{Encephalitozoon cuniculi in vivo} / The influence of interferon gamma and specific antibodies on the p.o. infection with \kur{Encephalitozoon cuniculi in vivo}JELÍNEK, Jiří January 2007 (has links)
The influence of interferon gamma and specific antibodies on the infection with E. cuniculi in vivo has been studied. Reconstruction of SCID mice with CD4+ T-lymphocytes from BALB/c mice and from mice with defect gene for interferon gamma was used. Effects of the treatment with mouse recombinant interferon gamma and anti-E. cuniculi sera on survival of E. cuniculi infected SCID mice were monitored. The influence of the immunization with E. cuniculi antigen on the survival of E. cuniculi infected mice with defect gene for interferon gamma was examined.
|
22 |
Untersuchungen zur differentiellen Wirkung von verschiedenen Anti-CD4 monoklonalen Antikörpern auf T-ZellenPohlers, Dirk 14 July 2000 (has links)
CD4+-T-Helferzellen sind in großer Zahl in der entzündeten Synovialmembran bei rheumatoider Arthritis (RA) sowie in Arthritismodellen vorhanden und spielen mit großer Wahrscheinlichkeit eine bedeutende Rolle in der Pathogenese von Arthritiden. Bei der präventiven Behandlung mit drei verschiedenen Anti-CD4 monoklonalen Antikörpern (mAk) im Modell der Adjuvansarthritis der Ratte (AA) wurden abhängig von dem jeweils eingesetzten mAk unterschiedliche klinische Verbesserungen beobachtet. Im Mittelpunkt der Untersuchungen stand deshalb die Suche nach Parallelen zwischen der unterschiedlichen klinischen Effizienz der Anti-CD4 mAk W3/25, OX35 (klinisch effizient) und RIB5/2 (klinisch ineffizient) bei der präventiven Therapie der AA und ihren in vitro Effekten auf TZell-Funktionen als Erklärung für die unterschiedlichen Therapieeffekte.
Keine klaren Parallelen zur differentiellen klinischen Effizienz ergaben sich bei den folgenden Untersuchungen: 1.) Bestimmung der Affinitäten der mAk zum CD4-Molekül. 2.) Inhibition der Proliferation in der primären gemischten Lymphozytenkultur (1° MLC) mit CD4+-T-Zellen und CD8+-T-Zellen durch die drei mAk 3.) Beeinflussung der Produktion der Zytokine IL-2, IFNg, IL-10 und IL-4 in verschiedenen experimentellen Ansätzen (sekundäre MLC nach Anwesenheit der mAk in der 1° MLC, Kreuzvernetzung des CD4-Moleküls mittels der mAk nach bzw. vor einer Stimulation von CD4+-T-Zellen über den TZR). 4.) Einfluss der drei Anti-CD4 mAk auf die TZR-vermittelte Apoptose. 5.) Mobilisierung von intrazellulärem Kalzium durch CD4-Kreuzvernetzung mittels der mAk. 6.) Aktivität der Tyrosinkinasen p56lck und p59fyn nach CD4-Kreuzvernetzung mittels der mAk. 7) Phosphorylierung des Shc-Adaptermoleküls durch CD4-Kreuzvernetzung mittels der drei mAk. 8.) Effekte der drei mAk auf die Aktivität der Transkriptionsfaktoren NF-AT und AP-1.
Dagegen ergaben sich bei den Untersuchungen zur Produktion von TNFa und zur Aktivität des Transkriptionsfaktors NF-kB eindeutige Parallelen zur differentiellen klinischen Effizienz: 1.) Die Kreuzvernetzung des CD4-Moleküls mittels des mAk RIB5/2 nach bzw. vor einer Stimulation von CD4+-T-Zellen über den TZR induzierte eine signifikant höhere Sekretion von TNFa als mit den mAk W3/25 und OX35. 2.) Die Kreuzvernetzung des CD4-Moleküls mittels des mAk RIB5/2 vor einer Stimulation von CD4+-T-Zellen über den TZR führte zu einer signifikant stärkeren Erhöhung der Aktivität von NF-kB als mit den mAk W3/25 und OX35. Beide differentiellen Effekte könnten daher die Erklärung für die unterschiedliche klinische Effizienz der drei Anti-CD4 mAk darstellen.
|
23 |
The Role of IkZF Factors in Mediating TH1/TFH Development and FlexibilityBharath Krishnan Nair, Sreekumar 24 January 2020 (has links)
The ability of cells within the adaptive immune system to develop into specialized subsets allow for a robust and tailored immune response in the advent of an infection or injury. Here, CD4+ T-cells are a crucial component within this system, with subsets such as TH1, TH2, TH17, TFH and TREG cells playing vital roles in propagating cell-mediated immunity. For example, TH1 cells are essential in combating intracellular pathogens such as viruses, while TFH cells communicate with B-cells to optimize antibody responses against an invading pathogen. The development (and functionality) of these subsets is ultimately dictated by the appropriate integration of extracellular cues such as cytokines with cell intrinsic transcription factors, thereby promoting the necessary gene profile. Moreover, the observation that T-helper cells could exhibit a flexible nature (i.e having shared gene profiles and effector functions) not only demonstrate the efficiency of our immune system but also how such flexibility could have unintended consequences during adverse events such as autoimmunity. An important mediator of such flexibility is cytokines. However, the complete network of factors that come together to co-ordinate cytokine mediated plasticity remain unknown. Thus, the work in this dissertation hope to delineate the factors that collaborate to regulate cytokine induced T-helper cell flexibility. As such, we see that in the presence of IL-2, the Ikaros Zinc Finger (IkZF) transcription factor Eos is upregulated in TH1 cells, with this factor playing a significant role in promoting regulatory and effector functions of TH1 cells. Moreover, we show that Eos forms a novel protein complex with STAT5 and promotes STAT5 activity in TH1 cells. However, depleting IL-2 from the micro-environment leads to the upregulation of two other members within the IkZF family, Ikaros and Aiolos. Aiolos in turn collaborate with STAT3, induces Bcl-6 expression within these cells, thus promoting these cells to exhibit characteristic features of TFH cells. The work in this dissertation hopes to advance our understanding of the regulatory mechanisms involved in cytokine mediated T-cell flexibility thereby hoping to open new avenues for the development of novel therapeutic strategies in the event of autoimmunity. / Ph. D. / T-helper (TH) cells are an important component of the immune system, as these cells aid in the fight against pathogens by secreting factors that either accentuate the inflammatory response during infection or attenuate immune responses post infection. Such effects are made possible because T-helper cells can differentiate into a variety of subsets, with each subset being an important mediator in maintaining immune homeostasis. For example, the T-helper cell subset called TH1 plays a vital role in the fight against intracellular pathogens such as viruses and certain parasites, while T-follicular helper (TFH) cells aid in the production of antibodies specific to the invading pathogen. The development of such subsets occur when cell extrinsic signals, called cytokines, lead to the activation or induction of cell intrinsic proteins called transcription factors. Interestingly, research over the years have shown that T-helper cells are highly adaptable in nature, with one subset having the ability to attain certain characteristic features of other subsets. This malleable nature of T-helper cells relies on several factors, with cytokines within the micro-environment being an important one. Although this form of flexibility is efficient and beneficial at times, it can also be detrimental, as such flexibility is known to promote certain autoimmune diseases such as multiple sclerosis, rheumatoid arthritis and type 1 diabetes. Such detrimental effects are thought to be due to cytokines within the environment. Therefore understanding how cytokines influence the flexible nature of T-helper cells is important; as controlling such flexibility (either by regulating cytokines or the transcription factors activated as a consequence) could prevent the propagation of undesired T-helper cell functions. As such, the work in this dissertation hopes to uncover how one such cytokine, termed Interleukin-2 (IL-2) mediates the flexibility between TH1 and TFH cells. The work highlighted in this dissertation broadens our understanding of how cytokines influence T-helper cell development and flexibility, and consequently allows the design of novel therapeutic strategies to combat autoimmune diseases.
|
24 |
Antigenpräsentation und Aktivierung von T-Zellen in der LeberDerkow, Katja 24 January 2011 (has links)
Die Ätiologie und Pathogenese autoimmuner Lebererkrankungen sind nur unvollständig verstanden. Bei der primär sklerosierenden Cholangitis bzw. bei der autoimmunen Hepatitis sind Cholangiozyten der größeren Gallengänge und Hepatozyten die Zielzellen der Autoimmunreaktion in der Leber. Mausmodelle sind zur Analyse initialer pathophysiologischer Prozesse notwendig und tragen zum besseren Verständnis der immunologischen Vorgänge in der Leber bei. Mit Hilfe transgener Mauslinien, die das Modellantigen Ovalbumin gewebespezifisch in den Cholangiozyten (ASBT-OVA) oder in den Hepatozyten (TF-OVA) exprimieren, sowie adoptiven Transfers antigenspezifischer CD4+ und CD8+ T-Zellen wurden Untersuchungen zur Antigenpräsentation, T-Zell-Aktivierung und Toleranzinduktion in der Leber durchgeführt. Die Expression von Ovalbumin in Cholangiozyten resultierte in einer Aktivierung der CD8+ T-Zellen in der Leber und den Lymphknoten. Im Gegensatz dazu ignorierten naive CD4+ T-Zellen das Antigen und wurden nicht aktiviert. Die Expression von Ovalbumin in Hepatozyten resultierte in einer vollständigen Aktivierung der CD8+ T-Zellen zu Effektorzellen über Kreuzpräsentation durch professionelle antigenpräsentierende Zellen (APZ) in der Leber. Diese Aktivierung war transient und selbst-limitiert. Die Induktion von CD4+Foxp3+ regulatorischen T-Zellen trug entscheidend zur Limitierung der induzierten Autoimmunität und Kontrolle der Expansion von antigenspezifischen CD8+ T-Zellen bei. Naive CD4+ T-Zellen benötigten die Aktivierung durch APZ in einem anderen Organ, bevor sie in die Leber relokalisierten und wiesen keinen Effektorphänotyp auf. Beide Modelle repräsentieren nicht die chronische Eigenschaft humaner autoimmuner Lebererkrankungen, ermöglichen jedoch Untersuchungen zum besseren Verständnis der Rolle verschiedener T-Zell-Populationen in der Pathogenese autoimmuner Lebererkrankungen sowie der Antigenpräsentation und Aktivierung von T-Zellen durch hepatisches Antigen. / Aetiology and pathogenesis of autoimmune liver diseases are still incompletely understood. Cholangiocytes of the larger bile ducts and hepatocytes are the target structures of autoimmune reactions in the liver in primary sclerosing cholangitis and autoimmune hepatitis, respectively. Mouse models are necessary to analyse initial pathophysiological processes and contribute to a better understanding of immunological processes in the liver. With the help of transgenic mouse strains, in which the model antigen ovalbumin is expressed specifically in the cholangiocytes (ASBT-OVA) or in hepatocytes (TF-OVA), as well as the adoptive transfer of antigen specific CD4+ and CD8+ T cells, antigen presentation, T cell activation and tolerance induction in the liver, were analyzed. Expression of ovalbumin in cholangiocytes resulted in activation of CD8+ T cells in the liver and lymph nodes. In contrary, naïve antigen specific CD4+ T cells ignored the antigen expressed by cholangiocytes and were not activated. Expression of ovalbumin in hepatocytes resulted in complete activation of CD8+ T cells to become effector cells by crosspresentation depending on professional antigen presenting cells (APCs) in the liver. This activation was transient and self-limiting. Induction of CD4+Foxp3+ regulatory T cells played a crucial role in limiting autoimmunity and controlling the expansion of antigen specific CD8+ T cells in the liver. By contrast, naïve CD4+ T cells required activation by professional APCs in a different organ before relocating to the liver and did not display an effector phenotype. Both models do not represent the chronic characteristics of human autoimmune liver diseases, but help to gain a better understanding regarding the role of specific T cell populations in the pathogenesis of autoimmune liver diseases, as well as regarding antigen presentation and activation of T cells by hepatic antigen.
|
25 |
Deciphering the generation of bone marrow resident memory CD4 T cells in the spleenSarkander, Jana 18 October 2019 (has links)
Langlebige Gedächtnis-CD4 T Lymphozyten spielen eine entscheidende Rolle für die Bildung, Erhaltung und Reaktivierung anderer Gedächtnislymphozyten. Im Verlauf einer Immunreaktion wandern einige antigen-erfahrene CD4 T Zellen aus den sekundär lymphoiden Organen (SLO) ins Knochenmark (KM), wo sie als professionelle Gedächtnis-CD4 T Zellen ruhen und überdauern. Es ist jedoch weitgehend unverstanden wie die Vorläuferzellen in SLO gebildet werden. Der erste Teil dieser Arbeit identifiziert aktivierte CD49b+T-bet+/CXCR3+ CD4 T Zellen der Milz als Vorläuferzellen von KM-Gedächtnis-CD4 T Zellen. Der zweite Teil der Arbeit zeigt, dass die Vorläuferzellen nach einer verstärkten Zellproliferation und längerer kognitiver Interaktion mit dendritischen Zellen während der späten Aktivierungsphase der primären Immunantwort entstehen. Die Behandlung mit einem Zytostatikum oder die späte Blockade des kostimulatorischen CD28/B7-Signalweges verhindert wiederum deren Generierung. Fluoreszenzfarbstoffmarkierungsexperimente zeigen, dass mit zunehmender Zellteilung die Expression des Chemokinrezeptors CCR7 in den Vorläuferzellen verringert ist und die Expression des Zytokinrezeptors IL-2Rb erhöht ist. CCR7 ist für die Persistenz in der T-Zellzone von SLO entscheidend, sowie IL-2Rb für das langfristige Überleben der Zellen. Der dritte Teil dieser Arbeit untersucht die Rolle von B Zellen für die Etablierung des CD4 T-Zellgedächtnisses im KM. B Zellen wirken sich in der frühen Phase einer Immunantwort negativ auf die Akkumulation von Gedächtnis CD4 T Vorläuferzellen im KM aus, beeinflussen jedoch nicht die Proliferation von aktivierten CD4 T Zellen in der Milz während der Aktivierungsphase. Die Ergebnisse dieser Arbeit liefern neue Einblicke in die Generierung von Gedächtnis CD4 T Zellen des KM, die für neue Ansätze zur therapeutischen Stärkung des Immungedächtnisses im Rahmen von Impfungen oder dessen Ablation bei Autoimmunerkrankungen beitragen können. / Long-lived memory CD4 T lymphocytes play a crucial role in the generation, maintenance and reactivation of other memory lymphocytes. During an immune reaction, some antigen-experienced CD4 T cells relocate from secondary lymphoid organs (SLOs) to the bone marrow (BM) and reside and rest there as professional memory CD4 T cells. However, it remains elusive how the precursors of BM memory CD4 T cells are generated in SLOs. The first part of this thesis identifies splenic CD49b+T-bet+/CXCR3+ activated CD4 T cells as the precursors of BM memory CD4 T cells. The second part of this thesis describes that precursors of BM memory CD4 T cells are generated following enhanced cell proliferation and prolonged cognate interactions with dendritic cells (DCs) during the late activation phase of a primary immune response. Treatment with a cytostatic drug or blockage of the CD28/B7 costimulatory pathway in the late activation phase in turn abrogates the generation of precursors of BM memory CD4 T cells. Fluorescent-dye labeling experiments demonstrate that the more CD49b+CXCR3+ activated CD4 T cells divide, the more they lose the expression of CCR7, a chemokine receptor crucial for the persistence in the T cell zone of SLOs, and gain the expression of IL-2Rb, a cytokine receptor crucial for long-term survival. The third part of this thesis investigates the role of B cells for the establishment of resting CD4 T cell memory in the BM. B cells negatively impact the accumulation of memory CD4 T cell precursors in the BM during the early phase of an immune response but do not affect the cell division of activated CD4 T cells in the spleen during the activation phase. In sum, the results obtained in this thesis provide new insight into the generation of BM memory CD4 T cells that may help for the therapeutic strengthening of immune memory in the context of vaccination or its abolishment within the scope of autoimmune diseases.
|
26 |
HIV-1 transmission between T cells and macrophages : consequences for viral pathogenesisBaxter, Amy Elizabeth January 2013 (has links)
Within the paradigm of HIV-1 infection, macrophages play a crucial role as long-lived viral reservoirs. However, cell-free virus infection is inefficient and is unlikely to explain the levels of infection observed in vivo. To investigate the hypothesis that macrophages might be infected via direct contact with HIV-1-infected T cells, macrophage and HIV-1-infected T cell cocultures were imaged in real time. I observed that macrophages preferentially phagocytosed HIV-1-infected T cells and, using long-term culture assays, I established that following coculture the macrophage became productively infected. Phagocytosis of HIV-1-infected cells occurred independently of viral tropism; however, productive infection following T cell phagocytosis was restricted by viral tropism. Imaging flow cytometry showed that macrophages primarily phagocytose dying HIV-1-infected T cells. However, a significant population of HIV-1-infected 'healthy' cells were also taken up. Furthermore, ICAM-1 was identified as mediating the uptake of HIV-1-infected T cells. These results indicate that apoptosis plays a significant, but not sufficient, role in the mechanism for recognition and uptake of HIV-1-infected T cells. The response of macrophages to HIV-1 infection remains controversial. Using both primary macrophages and a monocyte/macrophage NFκB reporter line assay, I demonstrated that macrophages are activated in response to HIV-1-infected T cells. In addition, during coculture with HIV-1-infected T cells, macrophages upregulated secretion of Th1 cytokines, with associated dysregulation of regulatory cytokines. Finally, data presented suggest that polarisation of macrophages towards M1 and M2 phenotypes alters the susceptibility to HIV-1 infection in the cell-to-cell route.
|
27 |
The Role of Id Proteins in the Development and Function of T and B LymphocytesLin, Yen-Yu January 2014 (has links)
<p>E and Id proteins are members of the basic helix-loop-helix (bHLH) transcription regulator family. These proteins control a broad range of lymphocyte biology, from the development of multiple lineages to execution of their effector functions. With the development of new experiment models, novel functions of E and Id proteins continued to be discovered. In this thesis, I focused my study on the role of Id2 in gamma delta T cells and CD4<super>+</super> alpha beta T cells, as well as the role of Id3 in B cells.</p><p> Id proteins have been shown to control gamma delta T cell development. Id3 knockout mice demonstrate a dramatic expansion of innate-like Vgamma1.1<super>+</super> Vdelta6.3<super>+</super> T cells in the neonatal stage, suggesting that Id3 is an inhibitor of their development. Interestingly, Id3 knockout mice with a B6/129 mix background have much less expansion of the Vgamma1.1<super>+</super> Vdelta6.3<super>+</super> T cells compared to mice with pure B6 background. Genetic studies showed that this difference is strongly influences by a chromosome region very close to the Id2 locus. Using the Id2<super>f/f</super> CD4Cre<super>+</super> mice, I found that Id2 is also an inhibitor of gamma delta T cell development. Deletion of Id2 alone is sufficient to enhance the maturation of these cells in the thymus and induce a moderate expansion of gamma delta T cells in the periphery. This study demonstrated the delicate balance of transcription control in cells of the immune system.</p><p> The Id2<super>f/f</super> CD4Cre<super>+</super> mice also enabled me to study the role of Id2 in peripheral CD4<super>+</super> alpha beta T cell functions, which was difficult in the past because Id2 knockout mice lack lymph node development. I found that CD4 T cells in these mice have a profound defect in mounting immune responses, demonstrated by a complete resistance to induction of experimental autoimmune encephalomyelitis (EAE). I found that Id2-deficient CD4 T cells fail to infiltrate the central nervous system, and the effector CD4 T cell population is smaller compared to that in control mice. Id2 is important for the survival and proliferation of effector CD4 T cells, and this phenotype was correlated with an increased expression of <italic>Bim</italic> and <italic>SOCS3</italic>. This study revealed a novel role of Id2 in the functioning of CD4<super>+ </super>alpha beta T cells.</p><p> Switching my focus to B cells, recent next generation sequencing of human Burkitt lymphoma samples revealed that a significant proportion of them have mutations of Id3. This finding suggests that Id3 may be a tumor suppressor gene in the lymphoid system. Utilizing various Id3 knockout and conditional knockout mouse models, I showed that Id3 deficiency can accelerate lymphoid tumor genesis driven by the over-expression of oncogene c-Myc. This work may lead to development of a more realistic mouse model of human Burkitt lymphoma, allowing more mechanistic studies and perhaps preclinical tests of new therapies.</p> / Dissertation
|
28 |
BZIP Transcription Factors BATF and c-Maf are Essential for Type-2 InflammationBao, Katherine January 2016 (has links)
<p>Helminth exposure, allergy and asthma each induce cellular responses in lymphoid and peripheral tissues that give rise to type-2 inflammation. Essential molecular mediators of this response are type-2 cytokines interleukin(IL)-4 and IL-13 derived from various subsets of immune cells. In lymphoid tissues, CD4+ Tfh cells make IL-4 to elicit IgE and high-affinity IgG1 production. In peripheral sites of infection, group 2 innate lymphoid (ILC2) cells make IL-13 and Th2 cells make both IL-13 and IL-4. Together, these cells mediate smooth muscle contraction, mucus production and recruitment of other innate effector cells, all of which are hallmarks of type-2 inflammation. As central mediators of type-2 inflammation, understanding the cell-specific expression and molecular regulation of type-2 cytokines in CD4+ T cells and ILC2 cells may lead to new therapies that ameliorate allergic disease and helminth infections. </p><p>The AP-1 factor basic leucine zipper transcription factor ATF-like (BATF) has been identified as a pioneer factor in in vitro-generated Th17 cells. BATF facilitates chromatin remodeling at the IL-17 locus as well as loci of key Th17-associated lineage specifying factors. It has also been deemed essential to the generation of functional humoral immunity through the development of follicular helper T (Tfh) cells and germinal center B cells. However, the role of BATF in the development and function of other CD4+ T helper subsets and innate immune cells in vivo has remained unclear. I show here that mice deficient in BATF do not develop type-2 inflammation after exposure to the parasitic helminth Nippostongylus brasiliensis. Since type-2 cytokine expression by Th2 and ILC2 cells is essential for expedient helminth expulsion, I hypothesized that BATF likely has a role in the development and/or induction of cytokine expression in CD4+ Th2 cells and ILC2 cells. Consistent with this hypothesis, I found that BATF utilizes a novel mechanism to control Th2 cytokine expression in Th2 cells. Specifically, BATF promotes permissive epigenetic modifications to alter the chromatin landscape early during Th2 cell differentiation. In addition, my data show that BATF deficiency inhibits the activation of ILC2 cells, preventing ILC2-mediated helminth clearance. </p><p>In addition to uncovering BATF-mediated regulations of type-2 inflammation, my work has revealed new insight into the role of a second bZIP transcription factor, cMaf, during type-2 immunity. As mentioned above, helminth exposure elicits IL-4 production by both CD4+ Tfh and Th2 cells. Although type-2 cytokine transcription has been well characterized in Th2 cells, Tfh cell-mediated IL-4 production has yet to be fully defined. Importantly, I show that IL-4 production by Tfh cells is sustained upon deletion of classical IL-4 regulatory factors signal transducer and activator of transcription 6 (STAT6) and STAT5 and is not dependent on high GATA-3 expression. In sum, Tfh-driven IL-4 production is induced independent of classical pathways in Th2 cells. </p><p>Presently, the non-canonical transcription factors involved in IL-4 production by Tfh cells remain unclear. C-Maf works with BCL6, the master regulator of Tfh cells, to elicit Tfh formation. However, the precise role of c-Maf in Tfh cell fate and function remains unclear. So far, it has been shown that in Th2 cells, c-Maf binds to the IL-4 promoter and in Tfh cells, c-Maf binds to the CNS2 enhancer of the IL-4 locus to regulate IL-4 expression. Therefore, I hypothesized that c-Maf is important in non-canonical, GATA-3-independent IL-4 production by Tfh cells. </p><p>Here, I show that Tfh cells lacking canonical Th2 pathways for IL-4 expression express high levels of c-Maf and IL-4 transcript. Deletion of c-Maf in CD4+ T cells resulted in normal induction of BCL6 expression. Thus the initial stages of Tfh cell generation were induced. However, cMaf-deficient CD4+ T cells did not express important molecules associated with Tfh cell migration. Immunohistochemistry also confirmed that c-Maf deficiency inhibited CD4+ T cell migration from the paracortex into the B cell follicle. </p><p>These defects did not inhibit cMaf-deficient CD4+ T cells from making IL-4 transcript; however, IL-4 protein production was significantly impaired. Together, these results demonstrate that c-Maf is essential for Tfh cell-mediated immunity by promoting CD4+ T cell migration to the B cell follicles and the production of IL-4 protein in the germinal centers. </p><p>Collectively, the objective of my thesis research is to define the roles of the bZIP transcription factors BATF and c-Maf in type-2 inflammation. My data demonstrate that BATF is essential for the differentiation and function of Tfh, Th2, and ILC2 cells during helminth infection. Additionally, I have shown that c-Maf is required for Tfh function and CD4+ T cell migration to the B cell follicle. Thus, BATF and c-Maf are central to the development of humoral and peripheral type-2 inflammatory responses against helminth infection. Given the wide spectrum of disorders associated with type-2 inflammation, the identification of factors relevant to the development and function of Th2-, ILC2- and Tfh-driven allergic pathologies is broadly relevant. A comprehensive characterization of core factors like BATF and c-Maf provide new avenues in which to explore novel therapies to modulate type-2 inflammatory responses.</p> / Dissertation
|
29 |
Molecular Insights of CD4+ T Cell Differentiation, Effector Formation and Helper FunctionLiu, Siqi January 2016 (has links)
<p>CD4+ T cells play a crucial in the adaptive immune system. They function as the central hub to orchestrate the rest of immunity: CD4+ T cells are essential governing machinery in antibacterial and antiviral responses by facilitating B cell affinity maturation and coordinating the innate and adaptive immune systems to boost the overall immune outcome; on the contrary, hyperactivation of the inflammatory lineages of CD4+ T cells, as well as the impairments of suppressive CD4+ regulatory T cells, are the etiology of various autoimmunity and inflammatory diseases. The broad role of CD4+ T cells in both physiological and pathological contexts prompted me to explore the modulation of CD4+ T cells on the molecular level.</p><p>microRNAs (miRNAs) are small RNA molecules capable of regulating gene expression post-transcriptionally. miRNAs have been shown to exert substantial regulatory effects on CD4+ T cell activation, differentiation and helper function. Specifically, my lab has previously established the function of the miR-17-92 cluster in Th1 differentiation and anti-tumor responses. Here, I further analyzed the role of this miRNA cluster in Th17 differentiation, specifically, in the context of autoimmune diseases. Using both gain- and loss-of-function approaches, I demonstrated that miRNAs in miR-17-92, specifically, miR-17 and miR-19b in this cluster, is a crucial promoter of Th17 differentiation. Consequently, loss of miR-17-92 expression in T cells mitigated the progression of experimental autoimmune encephalomyelitis and T cell-induced colitis. In combination with my previous data, the molecular dissection of this cluster establishes that miR-19b and miR-17 play a comprehensive role in promoting multiple aspects of inflammatory T cell responses, which underscore them as potential targets for oligonucleotide-based therapy in treating autoimmune diseases. </p><p>To systematically study miRNA regulation in effector CD4+ T cells, I devised a large-scale miRNAome profiling to track in vivo miRNA changes in antigen-specific CD4+ T cells activated by Listeria challenge. From this screening, I identified that miR-23a expression tightly correlates with CD4+ effector expansion. Ectopic expression and genetic deletion strategies validated that miR-23a was required for antigen-stimulated effector CD4+ T cell survival in vitro and in vivo. I further determined that miR-23a targets Ppif, a gatekeeper of mitochondrial reactive oxygen species (ROS) release that protects CD4+ T cells from necrosis. Necrosis is a type of cell death that provokes inflammation, and it is prominently triggered by ROS release and its consequent oxidative stress. My finding that miR-23a curbs ROS-mediated necrosis highlights the essential role of this miRNA in maintaining immune homeostasis. </p><p>A key feature of miRNAs is their ability to modulate different biological aspects in different cell populations. Previously, my lab found that miR-23a potently suppresses CD8+ T cell cytotoxicity by restricting BLIMP1 expression. Since BLIMP1 has been found to inhibit T follicular helper (Tfh) differentiation by antagonizing the master transcription factor BCL6, I investigated whether miR-23a is also involved in Tfh differentiation. However, I found that miR-23a does not target BLIMP1 in CD4+ T cells and loss of miR-23a even fostered Tfh differentiation. This data indicate that miR-23a may target other pathways in CD4+ T cells regarding the Tfh differentiation pathway.</p><p>Although the lineage identity and regulatory networks for Tfh cells have been defined, the differentiation path of Tfh cells remains elusive. Two models have been proposed to explain the differentiation process of Tfh cells: in the parallel differentiation model, the Tfh lineage is segregated from other effector lineages at the early stage of antigen activation; alternatively, the sequential differentiation model suggests that naïve CD4+ T cells first differentiate into various effector lineages, then further program into Tfh cells. To address this question, I developed a novel in vitro co-culture system that employed antigen-specific CD4+ T cells, naïve B cells presenting cognate T cell antigen and BAFF-producing feeder cells to mimic germinal center. Using this system, I were able to robustly generate GC-like B cells. Notably, well-differentiated Th1 or Th2 effector cells also quickly acquired Tfh phenotype and function during in vitro co-culture, which suggested a sequential differentiation path for Tfh cells. To examine this path in vivo, under conditions of classical Th1- or Th2-type immunizations, I employed a TCRβ repertoire sequencing technique to track the clonotype origin of Tfh cells. Under both Th1- and Th2- immunization conditions, I observed profound repertoire overlaps between the Teff and Tfh populations, which strongly supports the proposed sequential differentiation model. Therefore, my studies establish a new platform to conveniently study Tfh-GC B cell interactions and provide insights into Tfh differentiation processes.</p> / Dissertation
|
30 |
Estudo funcional de microRNAs na infecção pelo HTLV-1 / miRNAs functional study in HTLV-1 infectionOtaguiri, Katia Kaori 14 March 2013 (has links)
O vírus linfotrópico de células T humanas (HTLV-1) foi o primeiro retrovírus descrito e está etiologicamente ligado a duas principais doenças: a leucemia/linfoma de célula T do adulto (ATLL) e a mielopatia associada ao HTLV-1/paraparesia espástica tropical (HAM/TSP). Apenas 0,3 a 5% dos indivíduos infectados desenvolvem essas doenças associadas, enquanto a maioria permanece assintomática. A HAM/TSP é uma manifestação inflamatória do sistema nervoso central e o mecanismo pelo qual o HTLV-1 induz o surgimento de HAM/TSP ainda não está totalmente esclarecido. Atualmente, uma abordagem promissora no entendimento de mecanismos, bem como na fisiopatogênese das infecções virais tem sido a avaliação da função de microRNAs (miRNAs). Há poucos dados na literatura envolvendo estas moléculas na infecção pelo HTLV-1 em linfócitos T CD4+ bem como no estabelecimento da doença HAM/TSP. No presente estudo, foi avaliada a expressão de miRNAs dos linfócitos T CD4+ isolados de portadores sem HAM/TSP (HAC), pacientes HAM/TSP e indivíduos sadios (CT) por meio de PCR em tempo real. A análise do perfil de expressão dos miRNAs nessas células revelou que 56 e 10 miRNAs apresentavamse mais 1,5 vezes aumentados no grupo HAM/TSP e HAC, respectivamente. O miR- 125b-1-1 apresentou expressão significamente maior no grupo HAC e o miR-146a, no grupo HAM/TSP. A análise in silico de predição de alvo demonstrou que o gene IFNG era potencialmente alvo do miR-125b-1-1 e os genes IRAK1 e TRAF6 do miR- 146a. Foi demonstrado que a expressão do IFNG no grupo HAC era 1,3 vezes mais elevado que o grupo CT e 1,8 vezes mais elevado no grupo HAM que no grupo CT. Houve um aumento na expressão de TRAF6 de 15,7 e 1,5 vezes nos grupos HAM/TSP e HAC, respectivamente. Não foi observada diferença na expressão de IRAK1 entre os três grupos. O ensaios de superexpressão do miR-125b-1-1 alterou a expressão do IFNG e do miR-146a alterou a expressão do gene IRAK1 e sua proteína. Os resultados evidenciados neste trabalho ressaltam a importância dos miRNAs na modulação de genes e proteínas importantes durante a infeção pelo HTLV-1. A correlação entre o miR-125b-1-1 e gene IFNG sugere que este miRNA esteja envolvido nos mecanismos de desenvolvimento de HAM/TSP. Além disso, a interação entre o miR-146a e os genes IRAK1 e TRAF6 sugerem que este miRNA esteja relacionado a mecanismos de persistência viral da infecção pelo HTLV-1 em linfócitos T CD4+. / Human T-cell lymphotropic vírus type 1 (HTLV-1) was the first human retrovirus discovered and it is related with two major diseases: adult T cell lymphoma/leukaemia (ATLL) and HTLV-1 -associated myelopathy/tropical spastic paraparesis (HAM/TS). About 0.3 to 5% of infected individuals will develop HTLV-1 related diseases, while the majority will remain life-long asymptomatic carriers of the virus. HAM/TSP is an inflammatory manifestation of central nervous system and the mechanism involved in HAM/TSP development is noy well elucidated. Currently, a promising approach on understanding the mechanisms as well as physiopathogenesis of viral infections has been the evaluation of the role of microRNAs (miRNAs) roles. There are few data involving CD4+ T cells miRNA expression in HTLV-1 infection as well as HAM/TSP establishment. To identify miRNAs differentially expressed in CD4+ T cells among non-infected individuals (CT), asymptomatic (HAC) and HAM/TSP patients we applied quantitative real time PCR. The analysis of miRNA expression profile in these cells showed 56 and 10 miRNAs upregulated 1.5 times in HAM/TSP and HAC groups, respectively. miR- 125b-1-1 was upregulated in HAC group and miR-146a in HAM/TSP. In silico analysis of target prediction showed that IFNG was a potentially miR-125b-1-1 target and IRAK1 and TRAF6 were miR-146a targets. IFNG expression was 1.3 higher in HAC than CT group and 1.8 higher in HAM/TSP than CT group. It was observed that TRAF6 expression was 15.7 and 1.5 times higher in HAM/TSP and HAC groups, respectively. There was no difference of IRAK1 expression among the three groups. Overexpression assays of miR-125b-1-1 altered IFNG expression and overexpression of miR-146a altered IRAK1 gene and protein expression. The results revealed that miRNAs modulate genes and proteins during HTLV-1 infection. miR- 125b-1-1 and IFNG gene correlation suggests that miR-125b-1-1 seems to contribute to HAM/TSP development. Besides, miR-146a and IRAK1 and TRAF6 interaction suggests that miT-146a seems to contribute to HTLV-1 establishment in CD4+ T cells.
|
Page generated in 0.0319 seconds