GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
  • 2
    Online Resource
    Online Resource
    The American Association of Immunologists ; 2016
    In:  The Journal of Immunology Vol. 196, No. 1_Supplement ( 2016-05-01), p. 140.32-140.32
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 196, No. 1_Supplement ( 2016-05-01), p. 140.32-140.32
    Abstract: Targeting the CD40-CD154 pathway to induce transplantation tolerance has been of interest to transplantation immunologists for over two decades. The efficacy of anti-CD154 antibody (Ab) observed in murine and non-human primate models for preventing allograft rejection and suppressing autoimmunity, generated great interest in the development of this antibody for clinical use. However, clinical trials with anti-CD154 Ab resulted in thromboembolic complications in patients as a result of CD154 expression by human platelets and limited the clinical application. To develop an alternative approach to anti-CD154 Ab, recent efforts have focused on targeting its receptor, CD40. In this study we evaluated the effects of a novel murine antagonistic antibody to CD40 (BI CD40-1) on alloreactive immune responses. Costimulation blockade consisting of donor splenocytes (DST) and BI CD40-1 reduced activation of alloreactive CD8 T cells as shown by inhibition of expansion, decreased cytokine production and decreased killing of allogeneic targets using an in vivo cytotoxicity assay. Moreover DST/BI CD40-1 treatment significantly prolonged skin graft survival in a BALB/c to B6 MHC mismatch model. The efficacy of BI CD40-1 in preventing alloreactive T cell responses was similar to anti-CD154 Ab. These results indicate that anti-CD40 Ab is an attractive candidate for translation to clinical application.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2016
    detail.hit.zdb_id: 1475085-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    The American Association of Immunologists ; 2014
    In:  The Journal of Immunology Vol. 192, No. 1_Supplement ( 2014-05-01), p. 70.4-70.4
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 192, No. 1_Supplement ( 2014-05-01), p. 70.4-70.4
    Abstract: Costimulation blockade targeting the CD40-CD154 pathway is an effective approach to induce tolerance to allografts in murine and non-human primate models. However inflammation generated during infection and exposure to pathogen associated molecular patterns abrogates costimulation blockade induced tolerance. Here we have characterized the alloreactive CD8 T cell response during costimulation blockade in the absence or presence of inflammation. Costimulation blockade consisting of donor splenocytes (DST)/anti-CD154 antibody (MR1) stimulated proliferation of alloreactive CD8 T cells but resulted in deletion of the cells by apoptosis. Exposure to LPS during blockade prevented this deletion, but surprisingly reduced proliferation kinetics of the alloreactive CD8 T cells. These data suggest that LPS increases the frequency of alloreactive CD8 T cells by providing survival signals during cell division. RNA-seq analyses revealed differential expression of genes involved in apoptosis (like Fasl, Perp, Birc5, E2F transcription factors), cell cycle (Ki-67, Bub1, Nusap1), CD8 T cell differentiation and function (granzyme B, T-bet, Eomes) and type I interferon induced genes (Ifi44, Ifit1, Usp18) between the treatment groups. Such phenotypic differences will facilitate identification of pathways that can be targeted to prevent the abrogation of transplantation tolerance in situations of inflammation.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2014
    detail.hit.zdb_id: 1475085-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 138, No. Supplement 1 ( 2021-11-05), p. 1704-1704
    Abstract: Background Approximately 50% of AML patients relapse following allogeneic hematopoietic stem cell transplant therapy, leaving them with very few treatment options (Rautenberg et al. (2019) Int. J. Mol. Sci. 20:228). Rare patients who naturally develop a minor antigen-specific graft-versus-leukemia T cell response show substantially lower relapse rates (Marijt et al. (2003) Proc. Natl. Acad. Sci. U.S.A. 100:2742-2747; Spierings et al. (2013) Biol. Blood Marrow Transplant. 19:1244-1253). HA-2 (YIGEVLVSV, genotype RS_61739531 C/C or T/C) is an HLA-A*02:01- and haematopoietically-restricted minor histocompatibility antigen derived from the class I myosin protein, MYO1G (Pierce et al. (2001) J. Immunol. 167:3223-3230). Patients receiving donor lymphocyte infusion from HA-2-mismatched donors who develop HA-2-specific T cells show a graft vs leukemia response and often experience long-term remission (Marijt et al. (2003) Proc. Natl. Acad. Sci. U.S.A. 100:2742-2747), making HA-2 an ideal candidate for TCR-engineered T cell immunotherapy of liquid tumors. Methods Using TScan's proprietary ReceptorScan platform, we discovered 1,302 HA-2-specific TCRs by screening 237 million naïve CD8 + T cells from 5 healthy HA-2-negative donors. We evaluated these TCRs using our proprietary DexScan platform to select the 15 TCRs with the highest surface expression and greatest affinity for the HA-2 peptide when transferred into primary human T cells. We further tested each TCR individually in our clinical vector backbone for surface expression, selective cytotoxicity, cytokine production, and proliferation using a panel of cell lines that express varying levels of HLA-A*02:01 and MYO1G. Finally, the top 5 TCRs were evaluated for alloreactivity using an array-based screen assessing 108 MHC-I molecules individually, and for off-target cross-reactivity using our proprietary genome-wide TargetScan platform. A lead TCR with limited alloreactivity and a narrow off-target profile was selected as our lead TSC-101 TCR. The avidity of TSC-101 for its putative off-targets was further measured in peptide-pulsed experiments to better appreciate the toxicity risks associated with our lead clinical candidate. Results and Conclusion Of the 1,302 HA-2-specific TCRs identified by our ReceptorScan platform, we identified TSC-101 as the most active TCR. TSC-101 displayed no alloreactivity to 107/108 HLAs tested and limited off-target risks in a genome-wide screens. Potential off-target peptides identified for TSC-101 displayed extremely weak avidities, predicting an absence of toxicity risks for our clinical candidate. Based on these results, TSC-101 has been advanced to IND-enabling activities to prepare for first-in-human testing in 2022. To our knowledge, this is the first clinical grade HA-2-specifc TCR being developed for immunotherapy for liquid tumors. Disclosures Macbeath: TScan Therapeutics: Current Employment, Current equity holder in publicly-traded company.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2021
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    Annual Reviews ; 2017
    In:  Annual Review of Pathology: Mechanisms of Disease Vol. 12, No. 1 ( 2017-01-24), p. 187-215
    In: Annual Review of Pathology: Mechanisms of Disease, Annual Reviews, Vol. 12, No. 1 ( 2017-01-24), p. 187-215
    Abstract: Immunodeficient mice engrafted with functional human cells and tissues, that is, humanized mice, have become increasingly important as small, preclinical animal models for the study of human diseases. Since the description of immunodeficient mice bearing mutations in the IL2 receptor common gamma chain (IL2rg null ) in the early 2000s, investigators have been able to engraft murine recipients with human hematopoietic stem cells that develop into functional human immune systems. These mice can also be engrafted with human tissues such as islets, liver, skin, and most solid and hematologic cancers. Humanized mice are permitting significant progress in studies of human infectious disease, cancer, regenerative medicine, graft-versus-host disease, allergies, and immunity. Ultimately, use of humanized mice may lead to the implementation of truly personalized medicine in the clinic. This review discusses recent progress in the development and use of humanized mice and highlights their utility for the study of human diseases.
    Type of Medium: Online Resource
    ISSN: 1553-4006 , 1553-4014
    URL: Issue
    Language: English
    Publisher: Annual Reviews
    Publication Date: 2017
    detail.hit.zdb_id: 2217576-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2019
    In:  Mammalian Genome Vol. 30, No. 5-6 ( 2019-6), p. 123-142
    In: Mammalian Genome, Springer Science and Business Media LLC, Vol. 30, No. 5-6 ( 2019-6), p. 123-142
    Type of Medium: Online Resource
    ISSN: 0938-8990 , 1432-1777
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2019
    detail.hit.zdb_id: 1459397-X
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    The American Association of Immunologists ; 2015
    In:  The Journal of Immunology Vol. 194, No. 1_Supplement ( 2015-05-01), p. 129.4-129.4
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 194, No. 1_Supplement ( 2015-05-01), p. 129.4-129.4
    Abstract: Tissue transplantation results in the release of endogenous danger molecules that stimulate inflammation by activating the innate immune system. Inflammatory signals regulate the magnitude and functionality of T cell responses during infection, but the direct impact of inflammation on alloreactive T cell responses has not been extensively studied. Understanding the role of inflammation in regulating the activation of alloreactive T cells will aid in the development of novel therapies to prolong allograft survival. We have used a TCR-transgenic alloreactive CD8 T cell model to study the response of alloreactive T cells in the presence of inflammation. Our results show that stimuli inducing different inflammatory profiles have significant effects on the size, phenotype and quality of alloreactive CD8 T cell responses as compared to stimulation with alloantigen alone. Inflammation induced by exposure to poly(I:C) and LPS increased the number of alloreactive CD8 T cells at the peak of the response. The ability of effector alloreactive CD8 T cells to produce IFN-g was enhanced with poly(I:C) but decreased with LPS, while cytotoxic activity was similar. LPS slowed the contraction phase of the alloantigen response, resulting in higher levels of memory CD8 T cells as compared to poly(I:C). These data indicate that inflammatory signals differentially regulate the kinetics and functionality of alloreactive CD8 T cell responses and may determine susceptibility to immune suppression.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2015
    detail.hit.zdb_id: 1475085-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Immunity, Inflammation and Disease, Wiley, Vol. 4, No. 4 ( 2016-12), p. 427-440
    Abstract: Immunodeficient mice engrafted with human immune systems support studies of human hematopoiesis and the immune response to human‐specific pathogens. A significant limitation of these humanized mouse models is, however, a severely restricted ability of human B cells to undergo class switching and produce antigen‐specific IgG after infection or immunization. Methods In this study, we have characterized the development and function of human B cells in NOD‐ scid IL2Rγ null (NSG) mice transgenically expressing human stem cell factor (SCF), granulocyte macrophage colony‐stimulating factor (GM‐CSF), and IL‐3 (NSG‐SGM3) following engraftment with human hematopoietic stem cells, autologous fetal liver, and thymic tissues (bone marrow, liver, thymus or BLT model). The NSG‐SGM3 BLT mice engraft rapidly with human immune cells and develop T cells, B cells, and myeloid cells. Results A higher proportion of human B cells developing in NSG‐SGM3 BLT mice had a mature/naive phenotype with a corresponding decrease in immature/transitional human B cells as compared to NSG BLT mice. In addition, NSG‐SGM3 BLT mice have higher basal levels of human IgM and IgG as compared with NSG BLT mice. Moreover, dengue virus infection of NSG‐SGM3 BLT mice generated higher levels of antigen‐specific IgM and IgG, a result not observed in NSG BLT mice. Conclusions Our studies suggest that NSG‐SGM3 BLT mice show improved human B cell development and permit the generation of antigen‐specific antibody responses to viral infection.
    Type of Medium: Online Resource
    ISSN: 2050-4527 , 2050-4527
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2016
    detail.hit.zdb_id: 2740382-8
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    The American Association of Immunologists ; 2015
    In:  The Journal of Immunology Vol. 194, No. 1_Supplement ( 2015-05-01), p. 198.7-198.7
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 194, No. 1_Supplement ( 2015-05-01), p. 198.7-198.7
    Abstract: CD8+ T cell exhaustion commonly occurs in chronic infections and cancers. During T cell exhaustion there is a progressive and hierarchical loss of effector cytokine production, up-regulation of inhibitory co-stimulatory molecules, and eventual deletion of antigen specific cells by apoptosis. A key factor that regulates T cell exhaustion is persistent TCR stimulation. Loss of this interaction results in restoration of CD8+ T cell effector functions in previously exhausted CD8+ T cells. TCR stimulation is also important for the differentiation of Eomeshi CD8+ effector T cells from T-bethi precursors, both of which are required for optimal viral control. However, the molecular mechanisms regulating the differentiation of these two cell subsets and the relative ratios required for viral clearance have not been described. We show that TCR signal strength regulates the relative expression of T-bet and Eomes in antigen-specific CD8+ T cells by modulating levels of IRF4. Reduced IRF4 expression results in lower proportions and numbers of T-bet+ Eomes- precursors and poor control of LCMV Clone 13 infection. Altering the ratio of T-bet to Eomes by generating compound heterozygotes of Irf4 and Eomes restores the differentiation of T-bet+ Eomes- precursors and leads to efficient viral control. These data highlight a critical role for IRF4 in regulating CD8+ T cell responses by ensuring a protective ratio of T-bet to Eomes, leading to the ultimate control of this chronic viral infection.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2015
    detail.hit.zdb_id: 1475085-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 202, No. 3 ( 2019-02-01), p. 647-651
    Abstract: Apoptosis of CD8 T cells is an essential mechanism that maintains immune system homeostasis, prevents autoimmunity, and reduces immunopathology. CD8 T cell death also occurs early during the response to both inflammation and costimulation blockade (CoB). In this article, we studied the effects of a combined deficiency of Fas (extrinsic pathway) and Bim (intrinsic pathway) on early T cell attrition in response to lymphocytic choriomeningitis virus infection and during CoB during transplantation. Loss of Fas and Bim function in Bcl2l11−/−Faslpr/lpr mice inhibited apoptosis of T cells and prevented the early T cell attrition resulting from lymphocytic choriomeningitis virus infection. Bcl2l11−/−Faslpr/lpr mice were also resistant to prolonged allograft survival induced by CoB targeting the CD40-CD154 pathway. These results demonstrate that both extrinsic and intrinsic apoptosis pathways function concurrently to regulate T cell homeostasis during the early stages of immune responses and allograft survival during CoB.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2019
    detail.hit.zdb_id: 1475085-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...