• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 26
  • 5
  • 2
  • 2
  • 1
  • Tagged with
  • 57
  • 57
  • 42
  • 42
  • 21
  • 21
  • 14
  • 10
  • 9
  • 9
  • 8
  • 7
  • 7
  • 7
  • 7
  • 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.
31

Long Non-Coding RNA Hotairm1 Promotes S100A9 Support of MDSC Expansion during Sepsis

Alkhateeb, Tuqa, Bah, Isatou, Kumbhare, Ajinkya, Youssef, Dima, Yao, Zhi Q., McCall, Charles E., Gazzar, Mohamed E. 01 January 2020 (has links)
Myeloid-derived suppressor cells (MDSCs) expand during mouse and human sepsis, but the mechanism responsible for this is unclear. We previously reported that nuclear transport of S100A9 protein programs Gr1CD11b myeloid precursors into MDSCs in septic mice. Here, we show that long non-coding RNA Hotairm1 converts MDSCs from an activator to a repressor state. Mechanistically, increased Hotairm1 expression in MDSCs in mice converted S100A9 from a secreted proinflammatory mediator to an immune repressor by binding to and shuttling it from cytosol to nucleus during late sepsis. High Hotairm1 levels were detected in exosomes shed from MDSCs from late septic mice. These exosomes inhibited lipopolysaccharide-stimulated secretion of S100A9 from early sepsis Gr1CD11b cells. Importantly, Hotairm1 knockdown in late sepsis Gr1CD11b MDSCs prevented S100A9 cytosol to nuclear transfer and decreased repression of proimmune T cells. Notably, ectopic expression of Hotairm1 in early sepsis Gr1CD11b cells shuttled S100A9 to the nucleus and promoted the MDSC repressor phenotype. In support of translating the mechanistic concept to human sepsis, we found that Hotairm1 binds S100A9 protein in CD33CD11bHLA-DR MDSCs during established sepsis. Together, these data support that Hotairm1 is a plausible molecular target for treating late sepsis immune suppression in humans and its immune repressor mechanism may be cell autonomous.
32

Mechanism of Myeloid-Derived Suppressor Cell Accumulation in Cancer and Susceptibility to Reversal by Sunitinib

Ko, Jennifer S. 23 December 2009 (has links)
No description available.
33

CHARACTERIZATION OF PORCINE MYELOID DERIVED SUPPRESSOR CELLS

Dhakal, Santosh 14 October 2015 (has links)
No description available.
34

ADAM10 overexpression dysregulates Notch signaling in favor of myeloid derived suppressor cell (MDSC) accumulation that deferentially modulates the host response depending on immune stimuli and interaction with mast cells.

Saleem, Sheinei 08 July 2013 (has links)
Although the physiological consequences of Notch signaling in hematopoiesis have been extensively studied, the differential effects of individual notch cleavage products remain to be elucidated. Given that a disintegrin and metalloproteinase 10 (ADAM10) is a critical regulator of Notch and that its deletion is embryonically lethal, we generated transgenic mice that overexpress ADAM10 at early stages of lymphoid and myeloid development (A10Tg). ADAM10 transgene expression alters hematopoiesis post-hematopoietic Lineage-Sca-1+c-kit+ (LSK) subset differentiation but prior to lineage commitment of progenitor populations. This results in delayed T cell development, abrogated B2 cell development, and dramatic expansion of functionally active myeloid derived suppressor cells (MDSCs) in A10Tg mice. Given ADAM10’s role in Notch signaling, we hypothesized that the observed hematopoietic alterations may be a consequence of perturbed Notch signaling. In fact, blockade of ADAM10 (S2) rescues B cell development and reduces myeloid cells in A10Tg LSKs. Inhibition of γ-secretase (S3) in wild type (WT) LSKs results in enhanced myelopoiesis, mimicking the phenotype of A10Tg mice. Collectively, these findings indicate that the differential cleavage of Notch into S2 and S3 products regulated by ADAM10 is critical for hematopoietic cell-fate determination. Albeit arising in a tumor-free host, A10Tg MDSCs are functionally and phenotypically analogous to tumor-derived MDSCs. A10Tg MDSCs inhibit T cell activation in vitro, and inhibit adoptive immunotherapy (AIT) of metastatic melanoma in vivo, which can be reversed with MDSC depletion. Intriguingly, A10Tg mice are resistant to parasitic infection upon inoculation of Nippostrongylus brasiliensis. However, depletion of MDSCs abrogates this response, while adoptive transfer (AT) of MDSCs into WT mice increases their resistance. This polarized activity of MDSCs is heavily dependent upon interaction with mast cells (MCs). In fact, B16 melanoma cells metastasize more rapidly in WT mice infused with MDSCs when compared to MC-deficient mice (Kit Wsh/Wsh), with or without MDSC AT. Parallel to B16 progression, the ability of MDSCs to promote anti-Nb immunity is significantly diminished in MC-deficient (Kit Wsh/Wsh) mice even with MDSC AT. This augmentation of MDSC activity in the presence of MCs is further corroborated by in vitro co-culture assays that demonstrate a synergistic increase in cytokine production. Furthermore, MDSCs preferentially migrate to the liver in a MC-dependent manner. This interaction is mediated by MC-released histamine. In fact, MDSCs express histamine receptors (HR) and histamine induces MDSC survival, proliferation, and activation. We demonstrate that MDSC activity is abrogated with histamine blockade. Moreover, in humans, allergic patients present with an increase in MDSC population, and MDSCs purified from a stage I breast cancer patient exhibit increased survival in the presence of histamine. Taken together, our studies indicate that MCs and MC-released histamine are critical for the observed functional duality of MDSCs, ranging from immunosuppressive to immunosupportive, depending on the disease state.
35

Immunotherapy of Cancer: Reprogramming Tumor/Immune Cellular Crosstalk to Improve Anti-Tumor Efficacy

Payne, Kyle K. 01 January 2015 (has links)
Immunotherapy of cancer has been shown to be promising in prolonging patient survival. However, complete elimination of cancer and life-long relapse-free survival remain to be major challenge for anti-cancer therapeutics. We have previously reported that ex vivo reprogramming of tumor-sensitized immune cells by bryostatin 1/ionomycin (B/I) and the gamma-chain (γ-c) cytokines IL-2, IL-7, and IL-15 resulted in the generation of memory T cells as well as CD25+ NKT cells and CD25+ NK cells. Adoptive cellular therapy (ACT) utilizing these reprogrammed immune cells protected FVBN202 mice from tumor challenge, and overcame the suppressive functions of myeloid-derived suppressor cells (MDSCs). We then demonstrated that the presence of CD25+ NKT cells was required for anti-tumor efficacy of T cells as well as their resistance to MDSCs. Similar results were obtained by reprogramming of peripheral blood mononuclear cells (PBMC) from patients with early stage breast cancer, demonstrating that an increased frequency of CD25+ NKT cells in reprogrammed immune cells was associated with modulation of MDSCs to CD11b-HLA-DR+ immune stimulatory cells. Here, we tested the efficacy of immunotherapy in a therapeutic setting against established primary breast cancer (Chapter One), experimental metastatic breast cancer (Chapter Three) as well as against minimal residual disease (MRD) in patients with multiple myeloma (Chapter Two). We evaluated the ability of reprogrammed immune cells, including CD25+ NKT cells, to convert MDSCs to myeloid immune stimulatory cells, in vivo; this resulted in the identification and characterization of a novel antigen presenting cell (APC). These novel immune stimulatory cells differed from conventional APCs, including dendritic cells (DCs) and macrophages. We have also demonstrated that enhancing immunogenicity of mammary tumors by treatment with Decitabine (Dec) along with overcoming MDSCs by utilizing reprogrammed T cells and NKT cells in ACT prolongs survival of animals, but fails to eliminate the tumor. However, targeting cancer during a setting of MDR, when tumor cells are dormant, results in objective responses as evidenced in our multiple myeloma studies. This suggests that targeting breast cancer with immunotherapy following conventional therapies, in a setting of residual disease when tumor cells are dormant, may be effective in eliminating such residual cells or maintaining dormancy and extending time-to-relapse for breast cancer patients.
36

AUGMENTATION OF T CELL EXPANSION FOR ADOPTIVE IMMUNOTHERAPY BY ALTERNATE GAMMA CHAIN CYTOKINES AND BY GEMCITABINE MEDIATED INHIBITION OF MYELOID DERIVED SUPPRESSOR CELLS

Le, Hanh 01 January 2008 (has links)
Successful treatment of cancer with adoptive immunotherapy (AIT) is dependent on the ability to produce large numbers of tumor-specific, functional T cells. The purpose of this thesis is to explore ways in which T cell expansion could be augmented. We have focused on exploring alternate gamma chain cytokines as stimulators of T cell proliferation and differentiation in addition to investigating the potential usefulness of gemcitabine (GEM) in abrogating the immunosuppressive effects of myeloid derived suppressor cells (MDSCs). B16 melanoma sensitized draining lymph node cells that have been activated in vitro with bryostatin-1 and ionomycin (B/I) were expanded in either IL-7/15 or in IL-2. We found that IL-7/15 was superior to IL-2 in expanding T cells for AIT of pulmonary metastases. Expansion of antitumor T cells was also improved by suppressing accumulation of MDSCs in mice bearing 4T1 mammary carcinoma using GEM. GEM directly inhibits both 4T1 mammary carcinoma cells and MDSCs. Its inhibition of MDSCs rescued tolerant T cells, augmenting both expansion and response to tumor antigen.
37

Rôle de Tif1gamma dans les différenciations granulo-monocytaire et macrophagique / Role of Tif1gamma in granulomonocytory and macrophagic differentiations

Chretien, Marie-Lorraine 18 December 2015 (has links)
La LMMC est une pathologie clonale de la cellule souche hématopoïétique dont les caractéristiques la classent parmi les syndromes myélodysplasiques/myéloprolifératifs (SMD/SMP). L’invalidation conditionnelle de Tif1γ au niveau hématopoïétique chez la Souris (Tif1γΔ/Δ) est responsable du développement d’un SMD/SMP mimant la LMMC humaine lorsque la souris atteint l’âge de 6 mois, faisant de Tif1γ un gène suppresseur de tumeur. Par ailleurs, malgré une monocytose, la population macrophagique péritonéale de ces souris est diminuée.Les objectifs de mon travail étaient de caractériser chez les souris malades la population myéloïde et d’étudier la différenciation macrophagique. Nous avons identifié chez les souris homozygotes Tif1γΔ/Δ une population morphologiquement immature, associant des caractéristiques granulocytaires et monocytaires. Les propriétés phénotypiques et moléculaires de cette population évoquent celles des cellules myéloïdes suppressives granulocytaires de type PMN-MDSC, faisant de Tif1γ un régulateur négatif de son développement. En sus, la différenciation in vitro des cellules myéloïdes médullaires en macrophages sous l’effet du CSF-1 est altérée. La baisse d’expression du CSF-1R n’explique pas à elle seule ces altérations puisque celle des cellules dendritiques est également perturbée sans modification de l’expression du GM-CSFR. Nous émettons l’hypothèse que l’augmentation d’expression de S100A8 et S100A9 chez les souris malades induit le développement des progéniteurs myéloïdes en cellules proches des PMN-MDSC au détriment des différenciations dendritique et macrophagique. En conclusion, Tif1γ est un régulateur majeur de la myélopoïèse. / Chronic myelomonocytic leukemia (CMML) is a hematologic stem cell disease whose characteristics correspond to myelodysplastic/myeloproliferative syndroms (MDS/MPS). Hematopoietic conditional deletion of Tif1γ in mice leads to the development of a MDS/MPS, mimiking human CMML, when age is comprised between 6 to 10 months, defining Tif1γ as a tumour suppressor gene. Moreover, peritoneal macrophage population in these mice is decreased despite a monocytosis.The aims of my work were first to characterize in sick mice the myeloid population, and second to study macrophage differentiation. The myeloid population in Tif1γΔ/Δ mice is morphologically immature, with granulocytic and monocytic features. We demonstrated that phenotypic and molecular characteristics of this population are close to those observed in PMN-MDSC (polymorphonuclear myeloid-derived suppressor cells), suggesting that Tif1γ is a negative regulator gene of this myeloid subset. Furthermore, we showed that in vitro macrophage differentiation of myeloid progenitors upon CSF-1 treatment is altered. Decreased expression of CSF1-R (CSF-1 receptor) does not totally explain this alteration since dendritic cell differentiation is also abnormal, without alteration in GM-CSFR expression. Therefore, we hypothesize that S100A8 and S100A9 hyperexpression in Tif1γΔ/Δ mice is able to promote PMN-MDSC-like differentiation at the expense of macrophage and dendritic differentiations. In conclusion, Tif1γ is a major myelopoiesis regulator gene.
38

Characteristics and functions of human T lymphocyte subpopulations separated on the basis of theophylline sensitivity of E rosette formation

Divakaran, Sarala. January 1984 (has links) (PDF)
Bibliography: leaves 99-106.
39

Aspects fonctionnels et pronostiques des cellules myéloïdes suppressives et de Foxp3 dans le cancer / Functional and prognostic aspects of myeloid suppressor cells and Foxp3 in cancer

Ladoire, Sylvain 10 May 2011 (has links)
L’échappement des cellules tumorales au processus d’immunosurveillance semble être une condition nécessaire au développement tumoral dans les modèles précliniques, comme chez l’homme. Les mécanismes par lesquels la tumeur parvient à médier une immunosubvertion sont multiples et font intervenir la plupart des cellules du système immunitaire, au sein desquelles, les cellules immunorégulatrices telles les cellules myéloides suppressives (MDSCs) ou les lymphocytes T régulateurs (Tregs, exprimant le facteur de transcription Foxp3), semblent jouer un rôle prépondérant. Les résultats présentés dans ce travail visent à mieux comprendre les rôles fonctionnels et pronostics des cellules myéloïdes suppressives et des Tregs dans le cancer, avec une attention plus particulière sur la façon dont ces cellules peuvent être modulées par la chimiothérapie. Concernant les MDSCs, nos travaux ont permis de mieux comprendre les mécanismes moléculaires présidant à leur accumulation d’une part, et d’autre part à l’acquisition de leur propriétés immunosuppressives, à travers une voie de signalisation impliquant les exosomes d’origine tumorale. Cette découverte, et la propriété d’une molécule d’usage thérapeutique courant, l’amiloride, de diminuer la production d’exosomes, y compris par les cellules tumorales, offrent une nouvelle possibilité de ciblage pharmacologique des MDSCs. Par ailleurs, l’étude des effets cytotoxiques sur les MDSCs de plusieurs molécules de chimiothérapie nous a permis de montrer que le 5-fluorouracile, probablement en raison d’un faible niveau d’expression de sa cible, la thymidilate synthase, dans les MDSCs, possédait une capacité sélective à éliminer ces cellules. Nos travaux d’immunohistochimie conduits sur des prélèvements tumoraux issus de patientes porteuses de cancers du sein localisés traitées par chimiothérapie néoadjuvante ont quand à eux permis de démontrer que la chimiothérapie néoadjuvante s’accompagne de modifications qualitatives de l’infiltration tumorale à la fois en lymphocytes T CD8+ et en lymphocytes T régulateurs Foxp3+. L’existence, après chimiothérapie néoadjuvante, d’une balance favorable de la réponse immunitaire, associant forte infiltration en CD8+ et faible infiltration en Foxp3+ s’accompagne d’une augmentation significative des marqueurs de cytotoxicité à médiation cellulaire, et est significativement corrélée à une éradication complète des cellules tumorales. Cette signature immunologique favorable se traduit également à long terme par une meilleure survie sans récidive et une meilleure survie globale, indépendamment du type de chimiothérapie reçue, de l’obtention ou non d’une réponse complète histologique, et du sous type moléculaire de cancer du sein. La combinaison de cette information immunologique avec la connaissance de la taille du résidu tumoral après traitement permet de considérablement affiner le pronostic des patientes. Enfin, nos travaux préliminaires semblent montrer que l’expression de Foxp3 dans les cellules cancéreuses de tumeurs du sein HER2+++ constitue un facteur de bon pronostic. Ces résultats illustrent donc l’importance non pas seulement des caractéristiques tumorales, mais aussi des caractéristiques de l’hôte, en particulier de la réponse immunitaire qu’il est capable de susciter, et de l’influence de la chimiothérapie sur cette dernière. / Evasion of immune surveillance by certain tumour cells seems to be a basic requirement for tumour development in preclinical models and in humans. The mechanisms by which the tumour mediates its immune evasion are manifold, and involve the majority of immune system cells. Among these, immunoregulatory cells such as myeloid-derived suppressor cells (MDSCs) or regulatory T lymphocytes (T-regs, which express the transcription factor Foxp3) appear to play a predominant role. The results presented in this work aim to improve our understanding of the functional and prognostic roles of myeloid suppressor cells and T-regs in cancer, focussing particularly on how these cells are modulated by chemotherapy. Regarding MDSCs, our work has made it possible to better understand on the one hand the molecular mechanisms underlying their accumulation, and on the other hand, their acquisition of immunosuppressive properties, through a signaling pathway involving exosomes of tumoral origin. This discovery, combined with the ability of amiloride, a molecule in frequent therapeutic use, to reduce the production of exosomes, even by tumour cells, offers new avenues for pharmacological targeting of MDSCs. Indeed, a study of the cytotoxic effects on MDSCs of several chemotherapy compounds made it possible to show that 5-fluorouracil has a selective capacity to eliminate MDSCs, probably due to the low level of expression of its target, thymidylate synthase, in MDSCs. Our immunohistochemical studies on tumour specimens resected from patients with localised breast cancer treated by neoadjuvant chemotherapy have shown that neoadjuvant chemotherapy is associated with qualitative changes in tumour infiltration by both CD8+ T-lymphocytes and Foxp3+ T-regs. The existence of a favourable immune response ratio after neoadjuvant chemotherapy, as defined by high infiltration by CD8+ and a low level of Foxp3+ infiltration, is associated with a significant increase in markers of cell-mediated cytotoxicity, and is also significantly correlated with complete eradication of tumour cells. This favourable immunological profile is reflected in the long-term by improved disease-free survival and better overall survival, regardless of the type of chemotherapy, the achievement or not of complete pathological response, and the molecular sub-type of breast cancer. Combining this immunological information with the data about the extent of tumour residue after treatment makes it possible to considerably refine prognosis in these patients. Finally, our preliminary work suggests that expression of Foxp3 in cancerous cells in HER2+++ breast tumours is a favourable prognostic factor. Overall, these results illustrate the importance not only of the tumour characteristics, but also of the host characteristics, in particular, the type of immune response that it is capable of eliciting, and the effect of chemotherapy on this immune response.
40

Expansion of Myeloid-Derived Suppressor Cells Promotes Differentiation of Regulatory T Cells in HIV-1+ Individuals

Wang, Ling, Zhao, Juan, Ren, Junping P., Wu, Xiao Y., Morrison, Zheng D., El Gazzar, Mohamed A., Ning, Shunbin, Moorman, Jonathan P., Yao, Zhi Q. 19 June 2016 (has links) (PDF)
Objective: Regulatory T cells (Tregs) contribute to HIV-1 disease progression by impairing antiviral immunity; however, the precise mechanisms responsible for the development of Tregs in the setting of HIV-1 infection are incompletely understood. Design: In this study, we provide evidence that HIV-induced expansion of monocytic myeloid-derived suppressor cells (M-MDSCs) promote the differentiation of Foxp3+ Tregs. Methods: We measured MDSC induction and cytokine expression by flow cytometry and analyzed their functions by coculturing experiments. Results: We observed a dramatic increase in M-MDSC frequencies in the peripheral blood of HIV-1 seropositive (HIV-1+) individuals, even in those on antiretroviral therapy with undetectable viremia, when compared with healthy participants. We also observed increases in M-MDSCs after incubating healthy peripheral mononuclear cells (PBMCs) with HIV-1 proteins (gp120 or Tat) or Toll-like receptor 4 ligand lipopolysaccharides in vitro, an effect that could be abrogated in the presence of the phosphorylated signal transducer and activator of transcription 3 inhibitor, STA-21. Functional analyses indicated that M-MDSCs from HIV-1+ individuals express higher levels of IL-10, tumor growth factor-β, IL-4 receptor α, p47phex, programmed death-ligand 1, and phosphorylated signal transducer and activator of transcription 3 – all of which are known mediators of myelopoiesis and immunosuppression. Importantly, incubation of healthy CD4+ T cells with MDSCs derived from HIV-1+ individuals significantly increased differentiation of Foxp3+ Tregs. In addition, depletion of MDSCs from PBMCs of HIV-1+ individuals led to a significant reduction of Foxp3+ Tregs and increase of IFNγ production by CD4+ T effector cells. Conclusions: These results suggest that HIV-induced MDSCs promote Treg cell development and inhibit T cell function – a hallmark of many chronic infectious diseases.

Page generated in 0.0671 seconds