Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Sara Abromson-Leeman is active.

Publication


Featured researches published by Sara Abromson-Leeman.


Glia | 2002

RANTES stimulates inflammatory cascades and receptor modulation in murine astrocytes.

Yi Luo; Michael A. Berman; Qiwei Zhai; Falko R. Fischer; Sara Abromson-Leeman; Ye Zhang; William A. Kuziel; Craig Gerard; Martin E. Dorf

Cultured mouse astrocytes respond to the CC chemokine RANTES by production of chemokine and cytokine transcripts. Stimulation of astrocytes with 1 nM RANTES or 3–10 nM of the structurally related chemokines (eotaxin, macrophage inflammatory protein‐1α and ‐β [MIP‐1α, MIP‐1β]) induced transcripts for KC, monocyte chemoattractant protein‐1 (MCP‐1), tumor necrosis factor‐α (TNF‐α), MIP‐1α, MIP‐2, and RANTES in a chemokine and cell‐specific fashion. Synthesis of chemokine (KC and MCP‐1) and cytokine (TNF‐α) proteins was also demonstrated. RANTES‐mediated chemokine synthesis was specifically inhibited by pertussis toxin, indicating that G‐protein‐coupled chemokine receptors participated in astrocyte signaling. Astrocytes expressed CCR1 and CCR5 (the redundant RANTES receptors). Astrocytes derived from mice with targeted mutations of either CCR1 or CCR5 respond after RANTES stimulation, suggesting multiple chemokine receptors may separately mediate RANTES responsiveness in astrocytes. Preliminary data suggest activation of the MAP kinase pathway is also critical for RANTES‐mediated signaling in astrocytes. Treatment with RANTES specifically modulated astrocyte receptors upregulating intercellular adhesion molecule 1 (ICAM‐1) and downregulating CX3CR1 expression. Thus, after chemokine treatment, astrocytes release proinflammatory mediators and reprogram their surface molecules. The combined effects of RANTES may serve to amplify inflammatory responses within the central nervous system. GLIA 39:19–30, 2002.


Journal of Neuroimmunology | 2009

Encephalitogenic T cells that stably express both T-bet and RORγt consistently produce IFNγ but have a spectrum of IL-17 profiles

Sara Abromson-Leeman; Roderick T. Bronson; Martin E. Dorf

Th1/Th17 cells, secreting both IFNgamma and IL-17, are often associated with inflammatory pathology. We cloned and studied the cytokine phenotypes of MBP-specific, TCR-identical encephalitogenic CD4+ cells in relationship to Th1- and Th17-associated transcription factors T-bet and RORgammat. IFNgamma-producing cells could be sub-divided into those that are T-bet(+)/RORgammat(-) and those that are T-bet(+)/RORgammat(+). The latter comprises a spectrum of phenotypes, as defined by IL-17 production, and can be induced to up-regulate IL-23R with IL-12 or IL-23. The former, bona fide Th1 cells, lack IL-23R expression under all conditions. In vivo, T-bet(+)/RORgammat(-) and T-bet(+)/RORgammat(+) clones induce EAE equally well.


Immunity | 1995

A signaling pathway coupled to T cell receptor ligation by MMTV superantigen leading to transient activation and programmed cell death

Georg F. Weber; Sara Abromson-Leeman; Harvey Cantor

Stimulation of T cells by retroviral and bacterial super-antigens is followed by specific T cell elimination, in contrast with stimulation of T cells by peptide, which is usually associated with clonal expansion. We show here that this differential response phenotype is apparent at the level of individual T cell clones following TCR ligation with peptide or MTV antigen. We exploited selective coupling of apoptosis to TCR ligation by MTV7 to examine some of the intracellular biochemical events that underlie this response. MTV-dependent activation resulting in apoptosis was associated with activation of phospholipase A2 and the generation of reactive oxygen intermediates. Inhibition of these biochemical events prevented both MTV-dependent activation and apoptosis without affecting the peptide-dependent response of the same T cell clones. These results indicate that clonal expansion or programmed cell death following TCR ligation may be consequences of distinct TCR-coupled signaling pathways.


American Journal of Pathology | 2004

T-cell properties determine disease site, clinical presentation, and cellular pathology of experimental autoimmune encephalomyelitis.

Sara Abromson-Leeman; Rod Bronson; Yi Luo; Michael Berman; Rebecca Leeman; Joshua Leeman; Martin E. Dorf

Two distinct clinical phenotypes of experimental autoimmune encephalomyelitis are observed in BALB interferon-gamma knockout mice immunized with encephalitogenic peptides of myelin basic protein. Conventional disease, characterized by ascending weakness and paralysis, occurs with greater frequency after immunizing with a peptide comprising residues 59 to 76. Axial-rotatory disease, characterized by uncontrolled axial rotation, occurs with greater frequency in mice immunized with a peptide corresponding to exon 2 of the full length 21.5-kd protein. The two clinical phenotypes are histologically distinguishable. Conventional disease is characterized by inflammation and demyelination primarily in spinal cord, whereas axial-rotatory disease involves inflammation and demyelination of lateral medullary areas of brain. Both types have infiltrates in which neutrophils are a predominating component. By isolating T cells and transferring disease to naive recipients, we show here that the type of disease is determined entirely by the inducing T cell. Furthermore, studies using CXCR2 knockout recipients, unable to recruit neutrophils to inflammatory sites, show that although neutrophils are critical for some of these T cells to effect disease, there are also interferon-gamma-deficient T cells that induce disease in the absence of both interferon-gamma and neutrophils. These results highlight the multiplicity of T-cell-initiated effector pathways available for inflammation and demyelination.


Journal of Neuroimmunology | 1998

T-cell responses to myelin basic protein in normal and MBP-deficient mice.

Izumi Yoshizawa; Roderick T. Bronson; Martin E. Dorf; Sara Abromson-Leeman

BALB/c mice are resistant to the development of experimental autoimmune encephalomyelitis (EAE) after immunization with myelin basic protein (MBP). Previous studies of BALB/c mice suggest that MBP-specific T-cells can eventually be cloned from these mice, although they are either initially present in very low frequencies or are functionally anergic. To determine what role endogenous MBP expression plays in shaping the BALB/c T-cell repertoire, MBP-deficient BALB/c mice were constructed by breeding the shiverer (shi/shi) mutation onto the BALB/c background. These mice lack all conventional isoforms of MBP due to a deletion of MBP exons 3-7. Studies of the MBP-directed response of these mice suggest that endogenous MBP expression is directly responsible for EAE resistance in BALB/c mice, by quantitatively affecting expression of the T-cell repertoire. In contrast to wild-type BALB/c T-cells, uncloned T-cells from BALB/c shi/shi mice immunized with MBP proliferate in vitro to MBP and MBP peptides 59-76 and 89-101 and are able to induce severe EAE upon transfer to BALB/c recipients expressing MBP.


Journal of Neuroimmunology | 1993

Granulocyte-macrophage colony stimulating factor inhibits class II major histocompatibility complex expression and antigen presentation by microglia

Masaharu Hayashi; Martin E. Dorf; Sara Abromson-Leeman

Granulocyte-macrophage colony stimulating factor (GM-CSF) modulates various functions of monocytes/macrophages including antigen-presenting capacity. Recently it was found that astrocytes produce GM-CSF in the central nervous system (CNS) and that GM-CSF can induce proliferation and morphological changes of microglia. Here we show that GM-CSF can down regulate the interferon-gamma-mediated induction of major histocompatibility complex (MHC) class II antigens in microglia, but not in astrocytes. GM-CSF pretreatment completely prevents myelin basic protein-specific T cell proliferation induced by microglia not astrocytes. GM-CSF did not affect the cell surface expression on microglia of either MHC class I or cell adhesion molecules. The inhibition of microglial MHC class II expression and antigen-presenting function is specific for GM-CSF, as treatment with a different CSF (interleukin-3) did not modulate microglial phenotype or functional capacity. These data suggest that GM-CSF might be involved in the regulation of immune responses within the central nervous system.


Glia | 2003

Tumor necrosis factor is required for RANTES‐induced astrocyte monocyte chemoattractant protein‐1 production

Yi Luo; Michael A. Berman; Sara Abromson-Leeman; Martin E. Dorf

Astrocytes respond to stimulation with the chemokine RANTES (regulated on activation, normal T cell expressed) by production of a series of cytokines and chemokines, including tumor necrosis factor‐α (TNF‐α) and monocyte chemoattractant protein‐1 (MCP‐1). In the present study we demonstrate that RANTES induces TNF, which in turn stimulates subsequent production of MCP‐1. TNF‐R1 (p55) serves as the principal receptor responsible for MCP‐1 synthesis. The results define an astrocyte proinflammatory cascade that amplifies synthesis of proinflammatory mediators. The implications of these findings to inflammatory diseases of the central nervous system are discussed.


European Journal of Immunology | 2001

CD40-mediated activation of T cells accelerates, but is not required for, encephalitogenic potential of myelin basic protein-recognizing T cells in a model of progressive experimental autoimmune encephalomyelitis.

Sara Abromson-Leeman; Emanual Maverakis; Roderick T. Bronson; Martin E. Dorf

CD40 ligand‐CD40 interactions are important in the development of experimental autoimmune encephalomyelitis (EAE), but it is unclear whether this interaction is critical for de novo recruitment of T cells, entry of T cells into the central nervous system (CNS), or effector function of T cells in vivo. In this report we define the role of CD40 in a model of progressive EAE that does not depend on epitope spread or recruitment of new myelin‐specific T cells into the CNS. Results show that CD40 is not required for trans‐migration of activated T cells through the endothelial blood‐brain barrier, and in its absence T cells will both enter the CNS and induce disease. However, interaction with CD40 is critical for optimal activation and encephalitogenicity of cloned Th1 cells. In its presence, Th1 cells enter the CNS earlier and induce more severe disease. Inclusion of IL‐12 during activation of Th1 cells in the absence of CD40 can override the otherwise suboptimal level of encephalitogenicity observed. The implication of these findings for theapeutic use of agents designed to block this pathway is discussed.


Immunological Reviews | 1988

Comparisons between Helper and Suppressor T-Cell Induction 1

Hiroshi Ishikura; Vljay Kuchroo; Sara Abromson-Leeman; Martin E. Dorf

Although there have been numerous reports of antigen-specific T cells which downregulate or suppress immune responses, the analysis of suppressor T lymphocytes (Ts) has been frustrated by the failure to isolate matiy of the critical elements responsible for their specificity. Nonetheless, tbe demonstration of Ts activity has been readily reproduced in a variety of independent systems. This review is not intetided to resolve the controversies surrounding Ts, but to discuss the pivotal role of tbe antigen-presenting cell (APC) in the induction of Ts activity. This review also examines the mechanism accounting for Ts-restriction specificity, including an analysis of IJ.


Journal of Neuroimmunology | 1998

Differential recognition of MBP epitopes in BALB/c mice determines the site of inflammatory disease induction.

Izumi Yoshizawa; Roderick T. Bronson; Avi Ben-Nun; John R. Richert; Martin E. Dorf; Sara Abromson-Leeman

Although myelin basic protein (MBP)-recognizing T cells are not readily obtained after immunization of BALB/c mice with MBP (reflecting the BALB/c resistance to actively induced experimental autoimmune encephalomyelitis (EAE)), they can be expanded and cloned after several rounds of in vitro culture. The majority of BALB/c-derived clones recognize an epitope defined by MBP peptide 59-76. When transferred to naive BALB/c recipients, these clones cause classical EAE, with characteristic inflammation and demyelination of the central nervous system (CNS). We previously showed that two related clones recognizing a minor epitope, defined by MBP peptide 151-168, cause inflammation and demyelination preferentially of the peripheral nervous system (PNS). Because MBP has alternatively spliced isoforms, residues 151-168 are not present contiguously in all MBP isoforms. In order to determine whether induction of PNS disease is idiosyncratic to these sister clones, or related to their properties of epitope recognition, an independent T-cell line with similar recognition properties was studied. Clone 116F, derived from a BALB/c shiverer mouse, expresses a different T-cell receptor (TCR), with distinct TCR contact residues, but like the previously described T cells, this clone requires residues from both exons 6 and 7 for optimal stimulation. When adoptively transferred to BALB/c recipients, this clone preferentially induces disease of the PNS. A control BALB/c shiverer-derived MBP 59-76-recognizing clone, in contrast, induces CNS disease. These data strongly suggest that the site of disease initiation may correlate with epitope recognition, particularly when alternative isoforms are involved.

Collaboration


Dive into the Sara Abromson-Leeman's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Emanual Maverakis

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge