Network


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

Hotspot


Dive into the research topics where Jill Schartner is active.

Publication


Featured researches published by Jill Schartner.


Journal of Neuroscience Research | 2005

Microglia function in brain tumors

Jyoti J. Watters; Jill Schartner; Behnam Badie

Microglia play an important role in inflammatory diseases of the central nervous system (CNS). These cells have also been identified in brain neoplasms; however, as of yet their function largely remains unclear. More recent studies designed to characterize further tumor‐associated microglia suggest that the immune effector function of these cells may be suppressed in CNS tumors. Furthermore, microglia and macrophages can secrete various cytokines and growth factors that may contribute to the successful immune evasion, growth, and invasion of brain neoplasms. A better understanding of microglia and macrophage function is essential for the development of immune‐based treatment strategies against malignant brain tumors.


Neurosurgery | 2000

Flow cytometric characterization of tumor-associated macrophages in experimental gliomas.

Behnam Badie; Jill Schartner

OBJECTIVE Although microglia have been suggested to be a component of the inflammatory reaction to tumors of the central nervous system, their role in glioma biology remains unknown. One obstacle to studying the function of microglia is the inability to effectively separate them from macrophages. Because flow cytometry can effectively discern immune cells with similar surface antigens, we evaluated its role in characterizing the mononuclear cell infiltration in experimental gliomas. METHODS Freshly prepared rat C6, 9L, and RG-2 tumor specimens were labeled ex vivo with monoclonal antibodies against CD11b/c, CD45, and CD8a antigens and analyzed by flow cytometry. The extent of microglia (CD11b/c(high), CD45(low)), macrophage (CD11b/c(high), CD45(high)), and lymphocyte (CD11b/c(negative), CD45(high)) infiltration into tumors, tumor periphery, and contralateral tumor-free hemispheres was measured for each glioma type. RESULTS Microglia, which accounted for 13 to 34% of viable cells, were distributed throughout the central nervous system and were present in the tumors, tumor periphery, and contralateral tumor-free hemispheres. In contrast, macrophages were less prominent within the tumors and tumor periphery (4.2-12%) and were scarce in the contralateral tumor-free hemispheres (0.9-1.1%). Among the tumor types, RG-2 gliomas had the least microglia/macrophage infiltration. The frequency and the distribution pattern of lymphocytes also varied among tumor models. Whereas lymphocytes accounted for more than one-third of the cells in C6 and 9L tumors, they represented only 1% of cells in RG-2 gliomas. CONCLUSION More abundant than macrophages and scattered throughout the central nervous system, microglia account for a significant component of the inflammatory response to experimental gliomas. A better understanding of microglial function in gliomas may be important in the development of immunotherapy strategies.


Glia | 2005

Impaired capacity for upregulation of MHC class II in tumor‐associated microglia

Jill Schartner; Aaron R. Hagar; Michelle Van Handel; Leying Zhang; Nivedita V. Nadkarni; Behnam Badie

Immunotherapy for malignant gliomas is being studied as a possible adjunctive therapy for this highly fatal disease. Thus far, inadequate understanding of brain tumor immunology has hindered the design of such therapies. For instance, the role of microglia and macrophages, which comprise a significant proportion of tumor‐infiltrating inflammatory cells, in the regulation of the local anti‐tumor immune response is poorly understood. To study the response of microglia and macrophages to known activators in brain tumors, we injected CpG oligodeoxynucleotide (ODN), interferon‐γ (IFN‐γ), and IFN‐γ/LPS into normal and intracranial RG2 glioma‐bearing rodents. Microglia/macrophage infiltration and their surface expression of MHC class II B7.1 and B7.2 was examined by flow cytometry. Each agent evaluated yielded a distinct microglia/macrophage response: CpG ODN was the most potent inducer of microglia/macrophage infiltration and B7.1 expression, while IFN‐γ resulted in the highest MHC‐II expression in both normal and tumors. Regardless of the agent injected, however, MHC‐II induction was significantly muted in tumor microglia/macrophage as compared with normal brain. These data suggest that microglia/macrophage responsiveness to activators can vary in brain tumors when compared with normal brain. Understanding the mechanism of these differences may be critical in the development of novel immunotherapies for malignant glioma.


Neurosurgery | 1999

In vitro modulation of microglia motility by glioma cells is mediated by hepatocyte growth factor/scatter factor.

Behnam Badie; Jill Schartner; Jessica Klaver; Jessica Vorpahl

OBJECTIVE Considered as immune effector cells of the central nervous system, microglia represent a major component of the inflammatory cells found in malignant gliomas. Although their role in brain tumor biology is unclear, accumulation of microglia in malignant brain tumors may be mediated through active secretion of cytokines by glioma cells. Because hepatocyte growth factor/scatter factor (HGF/SF) has been shown to modulate glioma motility through an autocrine mechanism, and because microglia have been reported to express the HGF/SF receptor Met, we hypothesized that microglia recruitment by gliomas may also occur through the secretion of HGF/SF. METHODS The effect of glioma cells in augmenting BV-2 murine microglia motility was studied by using an in vitro Boyden chamber migration assay. To determine the chemokines involved in microglia migration, neutralizing monoclonal antibodies against monocyte chemotactic protein-1 and HGF/SF were tested. Immunoblotting was used to check for the expression of HGF/SF by glioma cells, and the expression of Met by BV-2 cells was examined by flow cytometry. RESULTS BV-2 migration was noted within 7 hours of incubation with both human (U251 MG and U373 MG) and murine (GL261) glioma cell lines. This migration corresponded to HGF/SF secretion by glioma cells and was completely inhibited by neutralizing monoclonal antibody against HGF/SF, but not monocyte chemotactic protein-1. Exposure of BV-2 cells to recombinant HGF/SF, but not monocyte chemotactic protein-1, resulted in their migration and down-regulation of Met in a dose-dependent fashion. CONCLUSION HGF/SF, which plays a role in glioma motility and mitogenesis, may also act as a chemokine for microglia and may be responsible for the microglia infiltration in malignant gliomas. This active recruitment of microglia may play an important role in glioma biology.


Journal of Neuroimmunology | 2001

Expression of Fas ligand by microglia: possible role in glioma immune evasion

Behnam Badie; Jill Schartner; Sakthivel Prabakaran; Jasmeet Paul; Jessica Vorpahl

The immune-privileged status of the central nervous system is thought to limit the application of immunotherapy for treatment of malignant brain tumors. Because the Fas pathway has been proposed to play a role in immune evasion, we examined the effect of tumor environment on the expression of Fas ligand (FasL) in a mouse glioma model. Immunoblotting revealed the expression of membrane-bound FasL to nearly double when murine G26 gliomas were propagated intracranially (IC) as compared to subcutaneously (SC). Further analysis by flow cytometry revealed microglia, which were absent in the SC tumors, to account for half of the FasL expression in the IC tumors. Interestingly, when FasL activity was inhibited in IC tumors, the proportion of tumor-infiltrating leukocytes increased three-fold, reaching the same frequency as the SC tumors. These observations suggest that microglia are a major source of FasL expression in brain tumors and possibly contribute to the local immunosuppressive milieu of malignant gliomas.


Experimental Neurology | 2000

Interferon-γ induces apoptosis and augments the expression of Fas and Fas ligand by microglia in vitro

Behnam Badie; Jill Schartner; Jessica Vorpahl; Keith Preston

Activation of microglia by interferon-gamma (IFN-gamma) has been implicated in a number of central nervous system (CNS) inflammatory disease processes. Because IFN-gamma has also been shown to play a role in programmed cell death, we investigated its cytotoxicity and its effect on the Fas apoptotic pathway in microglia. Flow cytometry was used to quantify the IFN-gamma-mediated apoptotic response and Fas and Fas ligand (FasL) expression in two well-characterized murine microglia cell lines (BV-2 and N9). Nuclear fragmentation, suggestive of apoptosis, was noted within 24 h of incubation of microglia with IFN-gamma (10 U/ml). After a 72-h incubation, almost every BV-2 and N9 microglia, but not GL261 glioma cells, underwent cell death and detached from the culture plates. This cytotoxicity occurred even at low IFN-gamma concentrations (1 U/ml) and was inhibited by BAF, a pan-caspase inhibitor. Incubation of BV-2 and N9 microglia, but not GL261 glioma cells, with IFN-gamma also potentiated the expression of Fas and FasL in a similar dose-response and time-course manner, as seen for the apoptotic response. Whereas Fas expression increased by 100% in both microglia cells, FasL upregulation was more pronounced and increased by as much as 200% in the N9 cells. These findings suggest that in addition to its role as a microglia activator, IFN-gamma may also induce apoptosis of microglia, possibly through simultaneous upregulation of Fas and FasL. Interferon-gamma modulation of the Fas pathway and apoptosis in microglia may be important in the pathogenesis of inflammatory CNS disease processes.


Journal of Neuroimmunology | 2002

Differential expression of MHC class II and B7 costimulatory molecules by microglia in rodent gliomas

Behnam Badie; Becky Bartley; Jill Schartner

To assess the immune function of microglia and macrophages in brain tumors, the expression of MHC class II and B7 costimulatory molecules in three rodent glioma models was examined. Microglia and macrophages, which accounted for 5-12% of total cells, expressed B7.1 and MHC class II molecules in the C6 and 9L tumors, but not RG2 gliomas. Interestingly, the expression of B7.1 and MHC class II molecules by microglia and macrophage was associated with an increase in the number of tumor-infiltrating lymphocytes in C6 and 9L tumors. B7.2 expression, which was present at low levels on microglia and macrophages in normal brain, did not significantly change in tumors. Interestingly, the expression of all three surface antigens increased after microglia were isolated from intracranial C6 tumors and cultured for a short period of time. We conclude that microglia immune activity may be suppressed in gliomas and directly correlates to the immunogenecity of experimental brain tumors.


Clinical and Experimental Immunology | 2009

Recurrent superantigen exposure in vivo leads to highly suppressive CD4+CD25+ and CD4+CD25- T cells with anergic and suppressive genetic signatures

Jill Schartner; Anne Marie Singh; Paul Dahlberg; Lauren Nettenstrom; Christine M. Seroogy

Staphylococcal enterotoxin B (SEB) activates T cells via non‐canonical signalling through the T cell receptor and is an established model for T cell unresponsiveness in vivo. In this study, we sought to characterize the suppressive qualities of SEB‐exposed CD4+ T cells and correlate this with genetic signatures of anergy and suppression. SEB‐exposed CD25+ and CD25‐Vβ8+CD4+ T cells expressed forkhead box P3 (FoxP3) at levels comparable to naive CD25+ T regulatory cells and were enriched after exposure in vivo. Gene related to anergy in lymphocytes (GRAIL), an anergy‐related E3 ubiquitin ligase, was up‐regulated in the SEB‐exposed CD25+ and CD25‐FoxP3+Vβ8+CD4+ T cells and FoxP3‐CD25‐Vβ8+CD4+ T cells, suggesting that GRAIL may be important for dominant and recessive tolerance. The SEB‐exposed FoxP3+GRAIL+ T cells were highly suppressive and non‐proliferative independent of CD25 expression level and via a glucocorticoid‐induced tumour necrosis factor R‐related protein‐independent mechanism, whereas naive T regulatory cells were non‐suppressive and partially proliferative with SEB activation in vitro. Lastly, adoptive transfer of conventional T cells revealed that induction of FoxP3+ regulatory cells is not operational in this model system. These data provide a novel paradigm for chronic non‐canonical T cell receptor engagement leading to highly suppressive FoxP3+GRAIL+CD4+ T cells.


Journal of Biological Chemistry | 2009

Gene related to anergy in lymphocytes (GRAIL) expression in CD4+ T cells impairs actin cytoskeletal organization during T cell/antigen-presenting cell interactions.

Jill Schartner; William T. Simonson; Sarah A. Wernimont; Lauren Nettenstrom; Anna Huttenlocher; Christine M. Seroogy

GRAIL (gene related to anergy in lymphocytes), is an E3 ubiquitin ligase with increased expression in anergic CD4+ T cells. The expression of GRAIL has been shown to be both necessary and sufficient for the induction of T cell (T) anergy. To date, several subsets of anergic T cells have demonstrated altered interactions with antigen-presenting cells (APC) and perturbed TCR-mediated signaling. The role of GRAIL in mediating these aspects of T cell anergy remains unclear. We used flow cytometry and confocal microscopy to examine T/APC interactions in GRAIL-expressing T cells. Increased GRAIL expression resulted in reduced T/APC conjugation efficiency as assessed by flow cytometry. Examination of single T/APC conjugates by confocal microscopy revealed altered polarization of polymerized actin and LFA-1 to the T/APC interface. When GRAIL expression was knocked down, actin polarization to the T/APC interface was restored, demonstrating that GRAIL is necessary for alteration of actin cytoskeletal rearrangement under anergizing conditions. Interestingly, proximal TCR signaling including calcium flux and phosphorylation of Vav were not disrupted by expression of GRAIL in CD4+ T cells. In contrast, interrogation of distal signaling events demonstrated significantly decreased JNK phosphorylation in GRAIL-expressing T cells. In sum, GRAIL expression in CD4+ T cells mediates alterations in the actin cytoskeleton during T/APC interactions. Moreover, in this model, our data dissociates proximal T cell signaling events from functional unresponsiveness. These data demonstrate a novel role for GRAIL in modulating T/APC interactions and provide further insight into the cell biology of anergic T cells.


Clinical and Experimental Immunology | 2007

Daily subcutaneous injections of peptide induce CD4+ CD25+ T regulatory cells.

Paul Dahlberg; Jill Schartner; A. Timmel; Christine M. Seroogy

Peptide immunotherapy is being explored to modulate varied disease states; however, the mechanism of action remains poorly understood. In this study, we investigated the ability of a subcutaneous peptide immunization schedule to induce of CD4+ CD25+ T regulatory cells. DO11·10 T cell receptor (TCR) transgenic mice on a Rag 2–/– background were injected subcutaneously with varied doses of purified ovalbumin (OVA323−339) peptide daily for 16 days. While these mice have no CD4+ CD25+ T regulatory cells, following this injection schedule up to 30% of the CD4+ cells were found to express CD25. Real‐time quantitative polymerase chain reaction (QPCR) analysis of the induced CD4+ CD25+ T cells revealed increased expression of forkhead box P3 (FoxP3), suggesting that these cells may have a regulatory function. Proliferation and suppression assays in vitro utilizing the induced CD4+ CD25+ T cells revealed a profound anergic phenotype in addition to potent suppressive capability. Importantly, co‐injection of the induced CD4+ CD25+ T cells with 5,6‐carboxy‐succinimidyl‐fluorescence‐ester (CFSE)‐labelled naive CD4+ T cells (responder cells) into BALB/c recipient mice reduced proliferation and differentiation of the responder cells in response to challenge with OVA323−339 peptide plus adjuvant. We conclude that repeated subcutaneous exposure to low‐dose peptide leads to de novo induction of CD4+ CD25+ FoxP3+ T regulatory cells with potent in vitro and in vivo suppressive capability, thereby suggesting that one mechanism of peptide immunotherapy appears to be induction of CD4+ CD25+ Foxp3+ T regulatory cells.

Collaboration


Dive into the Jill Schartner's collaboration.

Top Co-Authors

Avatar

Behnam Badie

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Christine M. Seroogy

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Jessica Vorpahl

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Becky Bartley

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Lauren Nettenstrom

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Paul Dahlberg

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Sakthivel Prabakaran

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

A. Timmel

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Aaron R. Hagar

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Anna Huttenlocher

University of Wisconsin-Madison

View shared research outputs
Researchain Logo
Decentralizing Knowledge