Network


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

Hotspot


Dive into the research topics where Julie K. Olson is active.

Publication


Featured researches published by Julie K. Olson.


Journal of Immunology | 2004

Microglia initiate central nervous system innate and adaptive immune responses through multiple TLRs

Julie K. Olson; Stephen D. Miller

Microglia are the resident macrophage-like population in the CNS. Microglia remain quiescent until injury or infection activates the cells to perform effector inflammatory and APC functions. Our previous studies have shown that microglia infected with a neurotropic strain of Theiler’s murine encephalomyelitis virus secreted innate immune cytokines and up-regulated costimulatory molecules and MHC class II, enabling the cells to present viral and myelin Ags to CD4+ T cells. Recently, TLRs have been shown to recognize pathogen-associated molecular patterns and initiate innate immune responses upon interaction with infectious agents. We examined TLR expression on brain microglia and their functional responses upon stimulation with various TLR agonists. We report that mouse microglia express mRNA for all of the recently identified TLRs, TLR1–9, used for recognition of bacterial and viral molecular patterns. Furthermore, stimulation of quiescent microglia with various TLR agonists, including LPS (TLR4), peptidoglycan (TLR2), polyinosinic-polycytidylic acid (TLR3), CpG DNA (TLR9), and infection with viable Theiler’s murine encephalomyelitis virus, activated the cells to up-regulate unique patterns of innate and effector immune cytokines and chemokines at the mRNA and protein levels. In addition, TLR stimulation activated up-regulation of MHC class II and costimulatory molecules, enabling the microglia to efficiently present myelin Ags to CD4+ T cells. Thus, microglia appear to be a unique and important component of both the innate and adaptive immune response, providing the CNS with a means to rapidly and efficiently respond to a wide variety of pathogens.


Glia | 2005

Differential activation of astrocytes by innate and adaptive immune stimuli

Pamela A. Carpentier; Wendy Smith Begolka; Julie K. Olson; Adam Elhofy; William J. Karpus; Stephen D. Miller

The immunologic privilege of the central nervous system (CNS) makes it crucial that CNS resident cells be capable of responding rapidly to infection. Astrocytes have been reported to express Toll‐like receptors (TLRs), hallmark pattern recognition receptors of the innate immune system, and respond to their ligation with cytokine production. Astrocytes have also been reported to respond to cytokines of the adaptive immune system with the induction of antigen presentation functions. Here we have compared the ability of TLR stimuli and the adaptive immune cytokines interferon‐γ (IFN‐γ) and tumor necrosis factor‐α (TNF‐α) to induce a variety of immunologic functions of astrocytes. We show that innate signals LPS– and poly I:C lead to stronger upregulation of TLRs and production of the cytokines IL‐6 and TNF‐α as well as innate immune effector molecules IFN‐α4, IFN‐β, and iNOS compared with cytokine‐stimulated astrocytes. Both innate stimulation and adaptive stimulation induce similar expression of the chemokines CCL2, CCL3, and CCL5, as well as similar enhancement of adhesion molecule ICAM‐1 and VCAM‐1 expression by astrocytes. Stimulation with adaptive immune cytokines, however, was unique in its ability to induce upregulation of MHC II and the functional ability of astrocytes to activate CD4+ T cells. These results indicate potentially important and changing roles for astrocytes during the progression of CNS infection.


Journal of Clinical Investigation | 2001

A virus-induced molecular mimicry model of multiple sclerosis

Julie K. Olson; J. Ludovic Croxford; Miriam Calenoff; Mauro C. Dal Canto; Stephen D. Miller

Molecular mimicry is the process by which virus infection activates T cells that are cross-reactive with self antigens. Infection of SJL/J mice with the neurotropic picornavirus Theilers murine encephalomyelitis virus (TMEV) leads to a progressive CD4(+) T cell-mediated demyelinating disease similar to multiple sclerosis. To study the potential of virus-induced molecular mimicry to initiate autoimmune demyelination, a nonpathogenic TMEV variant was engineered to encode a 30-mer peptide encompassing the immunodominant encephalitogenic myelin proteolipid protein (PLP139-151) epitope. Infection with the PLP139-151-encoding TMEV led within 10-14 days to a rapid-onset paralytic demyelinating disease characterized by PLP139-151-specific CD4(+) Th1 responses; insertion of a non-self ovalbumin sequence led to restoration of the normal late-onset disease. Early-onset disease was also observed in mice infected with a TMEV encoding PLP139-151 with an amino acid substitution at the secondary T cell receptor (TCR) contact residue (H147A), but not in mice infected with TMEV encoding a PLP139-151 substitution at the primary TCR contact (W144A). Most significantly, mice infected with TMEV encoding a Haemophilus influenzae mimic peptide, sharing only 6 of 13 amino acids with PLP139-151, displayed rapid-onset disease and developed cross-reactive PLP139-151-specific CD4(+) Th1 responses. To our knowledge, this is the first study showing that a naturally infectious virus encoding a myelin epitope mimic can directly initiate organ-specific T cell-mediated autoimmunity.


Journal of Virology | 2001

Direct Activation of Innate and Antigen-Presenting Functions of Microglia following Infection with Theiler's Virus

Julie K. Olson; Ann M. Girvin; Stephen D. Miller

ABSTRACT Microglia are resident central nervous system (CNS) macrophages. Theilers murine encephalomyelitis virus (TMEV) infection of SJL/J mice causes persistent infection of CNS microglia, leading to the development of a chronic-progressive CD4+ T-cell-mediated autoimmune demyelinating disease. We asked if TMEV infection of microglia activates their innate immune functions and/or activates their ability to serve as antigen-presenting cells for activation of T-cell responses to virus and endogenous myelin epitopes. The results indicate that microglia lines can be persistently infected with TMEV and that infection significantly upregulates the expression of cytokines involved in innate immunity (tumor necrosis factor alpha, interleukin-6 [IL-6], IL-18, and, most importantly, type I interferons) along with upregulation of major histocompatibility complex class II, IL-12, and various costimulatory molecules (B7-1, B7-2, CD40, and ICAM-1). Most significantly, TMEV-infected microglia were able to efficiently process and present both endogenous virus epitopes and exogenous myelin epitopes to inflammatory CD4+Th1 cells. Thus, TMEV infection of microglia activates these cells to initiate an innate immune response which may lead to the activation of naive and memory virus- and myelin-specific adaptive immune responses within the CNS.


Viral Immunology | 2001

Virus-induced autoimmunity: potential role of viruses in initiation, perpetuation, and progression of T-cell-mediated autoimmune disease.

Julie K. Olson; J. L. Croxford; Stephen D. Miller

Virus infections have been implicated in the initiation of multiple human autoimmune diseases. This article focuses on reviewing the role of viruses in initiation, progression, and perpetuation of autoimmune diseases. Various mechanisms by which virus infections can induce autoimmune responses including molecular mimicry, epitope spreading, direct bystander activation, and release of cryptic epitopes are discussed. Evidence implicating virus infections in the pathogenesis of various human autoimmune diseases is reviewed. Last, the characteristics of animal models that have been developed for the study of the potential role of viruses in the initiation and progression of autoimmune disease are reviewed.


Journal of Immunology | 2000

Temporal Development of Autoreactive Th1 Responses and Endogenous Presentation of Self Myelin Epitopes by Central Nervous System-Resident APCs in Theiler’s Virus-Infected Mice

Yael Katz-Levy; Katherine L. Neville; Josette Padilla; Sandra Rahbe; Wendy Smith Begolka; Ann M. Girvin; Julie K. Olson; Carol L. Vanderlugt; Stephen D. Miller

Theiler’s murine encephalomyelitis virus (TMEV)-induced demyelinating disease is a chronic-progressive, immune-mediated CNS demyelinating disease and a relevant model of multiple sclerosis. Myelin destruction is initiated by TMEV-specific CD4+ T cells targeting persistently infected CNS-resident APCs leading to activation of myelin epitope-specific CD4+ T cells via epitope spreading. We examined the temporal development of virus- and myelin-specific T cell responses and acquisition of virus and myelin epitopes by CNS-resident APCs during the chronic disease course. CD4+ T cell responses to virus epitopes arise within 1 wk after infection and persist over a >300-day period. In contrast, myelin-specific T cell responses are first apparent ∼50–60 days postinfection, appear in an ordered progression associated with their relative encephalitogenic dominance, and also persist. Consistent with disease initiation by virus-specific CD4+ T cells, CNS mononuclear cells from TMEV-infected SJL mice endogenously process and present virus epitopes throughout the disease course, while myelin epitopes are presented only after initiation of myelin damage (>50–60 days postinfection). Activated F4/80+ APCs expressing high levels of MHC class II and B7 costimulatory molecules and ingested myelin debris chronically accumulate in the CNS. These results suggest a process of autoimmune induction in which virus-specific T cell-mediated bystander myelin destruction leads to the recruitment and activation of infiltrating and CNS-resident APCs that process and present endogenous myelin epitopes to autoreactive T cells in a hierarchical order.


Glia | 2002

Expression of functional NK‐1 receptors in murine microglia

Amy Rasley; Kenneth L. Bost; Julie K. Olson; Stephen D. Miller; Ian Marriott

Cells of myeloid origin such as microglia have the potential to contribute significantly to the development of inflammatory responses in the CNS. The ability of the neuropeptide substance P to augment proinflammatory responses by other myeloid cell types such as macrophages and dendritic cells is well recognized. In the present study, we demonstrate the presence of mRNA encoding NK‐1 (substance P) receptors in murine microglia cell lines. Importantly, we have utilized specific antibodies developed by our laboratory to detect the expression of the NK‐1 receptor protein in murine microglia cell lines by Western blot analysis and flow cytometry. Furthermore, we have investigated the presence of this receptor on primary murine microglia and report the presence of authentic NK‐1 receptors as determined by Western blot analysis and flow cytometry. In addition, we demonstrate that NK‐1 receptors expressed on microglia are functional as demonstrated by the ability of nanomolar concentrations of substance P to initiate activation of the transcriptional activator, NF‐κB. Given the weight of evidence supporting the role of substance P–substance P receptor interactions in the initiation of optimal proinflammatory responses by myeloid cells, the demonstration of authentic and functional NK‐1 receptors in microglia identifies this neuropeptide as a potentially important contributor to CNS inflammatory responses during disease states. GLIA 37:258–267, 2002.


Experimental Neurology | 2012

Immune responses of microglia in the spinal cord: contribution to pain states.

Dominic Schomberg; Julie K. Olson

The role of microglia and their contribution to the development and maintenance of pain states has emerged as an attractive field of study. Sensitization of central nociceptors and interneurons is thought to be responsible for the symptoms of chronic neuropathic pain states. Microglia interact with these neurons at the site of injury or disease as well as remotely. Microglia can be activated by phagocytosis or through the activation of a number of constitutively expressed cell surface molecules. Once activated, microglia participate in both innate and adaptive immune responses and remain active indefinitely. Activated microglia contribute to pain states through the production of pro-inflammatory cytokines, chemokines and extracellular proteases. Activated microglia also exhibit a modulated cell surface receptor and ion channel profile. The activation of several intracellular pathways in microglia has also been implicated in pain states. Attenuation of microglia activity is being presented as a viable therapeutic approach with regard to not only the reduction of pain symptoms but also in preventing the development of chronic pain states.


Autoimmunity Reviews | 2002

Epitope spreading and molecular mimicry as triggers of autoimmunity in the Theiler's virus-induced demyelinating disease model of multiple sclerosis.

J. Ludovic Croxford; Julie K. Olson; Stephen D. Miller

The pathogenesis of multiple sclerosis (MS), a human demyelinating disease of the central nervous system (CNS), is currently unknown. It is widely thought that MS is an autoimmune disease which is supported by animal studies showing that myelin-specific CD4+ T cells can induce similar clinical disease in mice as observed in MS. However, the mechanism(s) of activation of these autoreactive CD4+ T cells are unknown. Although genetic susceptibility is important, other factors may be involved. Viral infections have long thought to be involved in the pathogenesis of MS although there exists little or no direct evidence implicating a role for a specific virus in MS pathogenesis. This review will discuss two models of virus-induced CNS autoimmunity, molecular mimicry and epitope spreading. These two mechanisms of activation of autoreactive T cells are presented in the context of MS.


Journal of Immunology | 2012

Endoplasmic Reticulum Stress Regulates the Innate Immunity Critical Transcription Factor IRF3

Yi-Ping Liu; Ling Zeng; Austin Tian; Ashley Bomkamp; Daniel Rivera; Delia Gutman; Glen N. Barber; Julie K. Olson; Judith A. Smith

IFN regulatory factor 3 (IRF3) regulates early type I IFNs and other genes involved in innate immunity. We have previously shown that cells undergoing an endoplasmic reticulum (ER) stress response called the unfolded protein response produce synergistically augmented IFN-β when stimulated with pattern recognition receptor agonists such as LPS. Concomitant ER stress and LPS stimulation resulted in greater recruitment of the IRF3 transcription factor to ifnb1 gene regulatory elements. In this study, we used murine cells to demonstrate that both oxygen–glucose deprivation and pharmacologic unfolded protein response inducers trigger phosphorylation and nuclear translocation of IRF3, even in the absence of exogenous LPS. Different ER stressors used distinct mechanisms to activate IRF3: IRF3 phosphorylation due to calcium-mobilizing ER stress (thapsigargin treatment, oxygen–glucose deprivation) critically depended upon stimulator of IFN gene, an ER-resident nucleic acid-responsive molecule. However, calcium mobilization alone by ionomycin was insufficient for IRF3 phosphorylation. In contrast, other forms of ER stress (e.g., tunicamycin treatment) promote IRF3 phosphorylation independently of stimulator of IFN gene and TANK-binding kinase 1. Rather, IRF3 activation by tunicamycin and 2-deoxyglucose was inhibited by 4-(2-aminoethyl)-benzenesulfonyl fluoride hydrochloride, a serine protease inhibitor that blocks activating transcription factor 6 processing. Interfering with ER stress-induced IRF3 activation abrogated IFN-β synergy. Together, these data suggest ER stress primes cells to respond to innate immune stimuli by activating the IRF3 transcription factor. Our results also suggest certain types of ER stress accomplish IRF3 phosphorylation by co-opting existing innate immune pathogen response pathways. These data have implications for diseases involving ER stress and type I IFN.

Collaboration


Dive into the Julie K. Olson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Charles Grose

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jenna L. Bowen

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Wendy Smith Begolka

American Academy of Dermatology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Colleen E. Hayes

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Dominic Schomberg

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge