Network


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

Hotspot


Dive into the research topics where Kathleen A. Hoag is active.

Publication


Featured researches published by Kathleen A. Hoag.


Journal of Leukocyte Biology | 1994

The role of CD4+ and CD8+ T cells in the protective inflammatory response to a pulmonary cryptococcal infection

Gary B. Huffnagle; Mary F. Lipscomb; Julie Lovchik; Kathleen A. Hoag; Nancy E. Street

Moderately virulent strains of Cryptococcusneoformans, inoculated via the trachea, cause a pulmonary infection in BALB/c mice that was gradually resolved by T lymphocyte‐dependent mechanisms. The current studies, using monoclonal antibodies to deplete T cell subsets, demonstrated that CD4+ and CD8+ T cells combined to mediate a prominent pulmonary inflammatory infiltrate that included lymphocytes, macrophages, neutrophils, and eosinophils. The inflammatory response peaked 2 weeks after infection and coincided with the beginning of gradual pulmonary clearance of the infection. CD4/CD8 double, deficiency (4‐8‐) markedly reduced the influx of all cells into the lungs. A CD4 deficiency had a more profound effect on the total number of inflammatory cells recruited to the lungs than a CD8 deficiency. Depletion of either CD8+ or CD4+ T cells significantly decreased pulmonary macrophages and neutrophils, but only a CD4 deficiency prevented the influx of eosinophils. Recruitment of CD8+ T cells occurred independently of CD4+ T cells, but CD4+ T cell recruitment to the lungs was significantly reduced in CD8‐deficient mice. Mitogen‐stimulated infiltrating lung lymphocytes from infected 4+8+ mice secreted both T helper cell type 1 (Th1) [interferon‐γ (IFN‐γ) and interleukin‐2 (IL‐2)] and Th2 (IL‐4, IL‐5, and IL‐10) cytokines. CD4 deficiency resulted in loss of T cells secreting IL‐4, IL‐5, and IL‐10. However, residual CD8+ T cells still secreted IL‐2 and IFN‐γ. Lung T cells from CD8‐deficient mice secreted similar levels of IL‐4, IL‐5, and IL‐10 on a per lung basis compared with 4+8+ mice despite decreased numbers of CD4+ T cells, but secreted reduced levels of IFN‐γ. These experiments indicate that (1) CD4+ T cells play a dominant role in recruiting macrophages and granulocytes to the lung and (2) CD8+ T cells also mediate cellular recruitment, increase the magnitude of CD4+ T cell numbers in the infiltrate, and contribute to the local secretion of IFN‐γ. Thus, these studies demonstrate that CD8+ T cells can independently mediate an inflammatory response to a large, particulate, extracellular antigen, a role heretofore attributed almost solely to CD4+ T cells. J. Leukoc. Biol. 55: 35–42; 1994.


Journal of Neuroimmunology | 2001

Rag-1-dependent cells are necessary for 1,25-dihydroxyvitamin D3 prevention of experimental autoimmune encephalomyelitis

Faye E. Nashold; Kathleen A. Hoag; Joan Goverman; Colleen E. Hayes

Multiple sclerosis (MS) is a demyelinating disease involving genetic and environmental risk factors. Geographic, genetic, and biological evidence suggests that one environmental risk factor may be lack of vitamin D. Here, we investigated how 1,25-dihydroxyvitamin D(3) (1,25-(OH)(2)D(3)) inhibits experimental autoimmune encephalomyelitis (EAE), an MS model. The experiments used adoptive transfer of TCR-transgenic (TCR1) cells specific for myelin basic protein (MBP) peptide into unprimed recipients. When unprimed TCR1 splenocytes were transferred, and the recipients were immunized with peptide, the mock-treated mice developed EAE, but the 1,25-(OH)(2)D(3)-treated recipients remained disease-free. Both groups had TCR1 T cells that proliferated in response to MBP Ac1-11 and produced IFN-gamma but not IL-4 in the lymph node. In the central nervous system (CNS), the mock-treated mice had activated TCR1 T cells that produced IFN-gamma but not IL-4, while the 1,25-(OH)(2)D(3)-treated mice had TCR1 T cells with a non-activated phenotype that did not produce IFN-gamma or IL-4. When activated TCR1 T cells producing IFN-gamma were transferred into unprimed mice, the mock-treated and the 1,25-(OH)(2)D(3)-treated recipients developed EAE. Likewise, the 1,25-(OH)(2)D(3) did not inhibit Th1 cell IFN-gamma production or promote Th2 cell genesis or IL-4 production in vitro. Finally, the 1,25-(OH)(2)D(3) inhibited EAE in MBP-specific TCR-transgenic mice that were Rag-1(+), but not in animals that were Rag-1-null. Together, these data refute the hypothesis that the hormone inhibits Th1 cell genesis or function directly or through an action on antigen-presenting cells, or promotes Th2 cell genesis or function. Instead, the evidence supports a model wherein the 1,25-(OH)(2)D(3) acts through a Rag-1-dependent cell to limit the occurrence of activated, autoreactive T cells in the CNS.


Journal of Nutrition | 2010

Vitamin A as a Regulator of Antigen Presenting Cells

David M. Duriancik; Denise E. Lackey; Kathleen A. Hoag

Vitamin A has been long associated with immune system competence. Vitamin A deficiency is known to compromise many aspects of both innate and adaptive immune responses. Recent advances in retinol uptake and metabolism have identified the antigen presenting cell (APC) as a central immune cell capable of vitamin A metabolism. APC are now known to express retinaldehyde dehydrogenase and secrete retinoic acid. The retinoic acid produced has both autocrine and paracrine effects. Autocrine effects include upregulation of CD1d nonclassical major histocompatibility class I-like molecule and matrix metalloproteinase-9. Paracrine effects influence multiple lymphocyte lineage cell populations. Specifically, retinoic acid increases IgA isotype class switching by B lymphocytes, enhances regulatory T cell differentiation, and directs homing of lymphocytes to mucosa. CD1d lipid antigen presentation expands natural killer T cell populations. Previously, the focus of vitamin A action in adaptive immunity was on lymphocytes, but these recent advances suggest the APC may be the central player in carrying out the immune system functions of vitamin A.


Infection and Immunity | 2009

Campylobacter jejuni-Induced Activation of Dendritic Cells Involves Cooperative Signaling through Toll-Like Receptor 4 (TLR4)-MyD88 and TLR4-TRIF Axes

Vijay A. K. Rathinam; Daniel M. Appledorn; Kathleen A. Hoag; Andrea Amalfitano; Linda S. Mansfield

ABSTRACT Campylobacter jejuni is an important cause of human enteritis and has been linked to the development of autoimmune diseases. Recently we showed that infection of murine dendritic cells (DCs) with C. jejuni resulted in DC activation and induction of Campylobacter-specific Th1-effector responses. Toll-like receptor (TLR) signaling through myeloid differentiation factor 88 (MyD88) and/or Toll-interleukin 1 (IL-1) receptor domain-containing adaptor-inducing beta interferon (IFN-β) (TRIF) is critical in inducing immunity against pathogens. In this study, we investigated the role of TLR2, TLR4, MyD88, and TRIF signaling in C. jejuni-induced inflammatory activation of DCs. DC upregulation of major histocompatibility complex class II and costimulatory molecules after C. jejuni challenge was profoundly impaired by TLR2, TLR4, MyD88, and TRIF deficiencies. Similarly, C. jejuni-induced secretion of IL-12, IL-6, and tumor necrosis factor alpha was significantly inhibited in TLR2−/−, TLR4−/−, MyD88−/−, and TRIF−/− DCs compared to that in wild-type DCs; however, the magnitude of inhibition was greater in MyD88−/−, TRIF−/−, and TLR4−/− DCs than in TLR2−/− DCs. Furthermore, C. jejuni induced interferon regulatory factor 3 phosphorylation and IFN-β secretion by DCs in a TLR4-TRIF-dependent fashion, further demonstrating activation of this pathway by C. jejuni. Importantly, TLR2, TLR4, MyD88, and TRIF deficiencies all markedly impaired the Th1-priming ability of C. jejuni-infected DCs. Thus, our results show that cooperative signaling through the TLR4-MyD88 and TLR4-TRIF axes represents a novel mechanism mediating C. jejuni-induced inflammatory responses of DCs. To our knowledge, such a mechanism has not been demonstrated previously for an intact bacterium.


Microbes and Infection | 2008

Dendritic cells from C57BL/6 mice undergo activation and induce Th1-effector cell responses against Campylobacter jejuni

Vijay A. K. Rathinam; Kathleen A. Hoag; Linda S. Mansfield

Food-borne Campylobacter jejuni (Cj) is an important cause of enteritis. We showed that C57BL/6 and congenic interleukin (IL)-10(-/-) mice serve as models of Cj colonization and enteritis, respectively. Thus, C57BL/6 mice are resistant to Cj induced disease. Because dendritic cells (DCs) are central to regulating adaptive immune responses, we investigated the interaction of Cj with murine bone marrow-derived DCs (BM-DCs) to assess bacterial killing, DC activation, and the ability of Cj-infected BM-DCs to stimulate Campylobacter-specific T cell responses in vitro. BM-DCs challenged with Cj efficiently internalized and killed Cj 11168 and significantly upregulated surface MHC-II, CD40, CD80 and CD86 demonstrating a mature phenotype. Infected BM-DCs secreted significant amounts of tumor necrosis factor-alpha (TNF-alpha), IL-6 and IL-12p70. Formalin-killed Cj also induced maturation of BM-DCs with similar cytokine production but at a significantly lower magnitude than live bacteria. Maximal activation of murine BM-DCs required internalization of Cj; attachment alone was not sufficient to elicit significant responses. Also, various strains of Cj elicited different magnitudes of cytokine production from BM-DCs. Finally, in a coculture system, Cj-infected BM-DCs induced high level interferon-gamma (INF-gamma) production from CD4+T cells indicating Th1 polarization. Thus, DCs from resistant C57BL/6 mice initiate T cell responses against Cj.


Journal of Immunology | 2001

Cutting Edge: A/WySnJ Transitional B Cells Overexpress the Chromosome 15 Proapoptotic Blk Gene and Succumb to Premature Apoptosis

Ian J. Amanna; Karen Clise-Dwyer; Faye E. Nashold; Kathleen A. Hoag; Colleen E. Hayes

Better knowledge of peripheral B lymphocyte homeostasis is needed to address the human hypogammaglobulinemia diseases. A defect in the Bcmd gene shortens the B cell life span and causes B cell deficiency in A/WySnJ mice. Previous genetic mapping placed Bcmd near Srebf2 on chromosome 15. Inspection of the human chromosome 22 syntenic region identified the proapoptotic Bik gene as a candidate. Two mapping methods placed the homologous mouse gene, Blk, near Srebf2. The Blk genomic structure was highly homologous to Bik. Sequence analysis ruled out coding region mutations, but Blk transcripts were overly abundant in sorted A/WySnJ T1 B cells. Moreover, enriched transitional B cells showed a cell-autonomous defect leading to excessive apoptosis. Thus, Bcmd may be a direct mutation in Blk, or in a gene involved in Blk regulation, such that excess expression pushes the A/WySnJ transitional B cells past the apoptosis checkpoint to cell death.


Journal of Nutrition | 2010

Vitamin A Upregulates Matrix Metalloproteinase-9 Activity by Murine Myeloid Dendritic Cells through a Nonclassical Transcriptional Mechanism

Denise E. Lackey; Kathleen A. Hoag

Myeloid dendritic cells (DC) are specialized antigen-presenting immune cells. Upon activation in peripheral tissues, DC migrate to lymph nodes to activate T lymphocytes. Matrix metalloproteinase (MMP)-9 is a gelatinase essential for DC migration. We have previously shown that all-trans retinoic acid (atRA), a bioactive metabolite of vitamin A, significantly augmented DC MMP-9 mRNA and protein production. We investigated the mechanisms by which atRA increased MMP-9 activity in vitro. Mouse myeloid DC cultured with atRA demonstrated increased gelatinase activity compared with cells cultured with retinoic acid receptor (RAR)-alpha antagonist. Adding MMP-9 inhibitor significantly blocked DC gelatinase activity and increased adherence of DC in a dose-dependent manner. AtRA-induced Mmp-9 gene expression in DC was blocked by transcriptional inhibition. Because the Mmp-9 promoter contains no canonical retinoic acid response element (RARE), we performed additional studies to determine how atRA regulated DC Mmp-9 transcription. Electrophoretic mobility shift assays for the consensus Sp1, activating protein-1, and nuclear factor-kappaB binding sites located in the Mmp-9 promoter did not indicate greater nuclear protein binding in response to atRA. Chromatin immunoprecipitation assays indicated RARalpha and histone acetyltransferase p300 recruitment to, and acetylation of, histone H3 at the Mmp-9 promoter was greater after atRA treatment. These data suggest that atRA regulated DC adhesion in vitro partly through MMP-9 gelatinase activity. Mmp-9 expression was enhanced through a transcriptional mechanism involving greater RARalpha promoter binding, recruitment of p300, and subsequent histone H3 acetylation, despite the absence of a consensus RARE.


Cellular Immunology | 2010

Vitamin A deficiency alters splenic dendritic cell subsets and increases CD8+Gr-1+ memory T lymphocytes in C57BL/6J mice ☆

David M. Duriancik; Kathleen A. Hoag

Vitamin A-deficient populations have impaired T cell-dependent antibody responses. Dendritic cells (DCs) are the most proficient antigen-presenting cells to naïve T cells. In the mouse, CD11b(+) myeloid DCs stimulate T helper (Th) 2 antibody immune responses, while CD8α(+) lymphoid DCs stimulate Th1 cell-mediated immune responses. Therefore, we hypothesized that vitamin A-deficient animals would have decreased numbers of myeloid DCs and unaffected numbers of lymphoid DCs. We performed dietary depletion of vitamin A in C57BL/6J male and female mice and used multicolor flow cytometry to quantify immune cell populations of the spleen, with particular focus on DC subpopulations. We show that vitamin A-depleted animals have increased polymorphonuclear neutrophils, lymphoid DCs, and memory CD8(+) T cells and decreased CD4(+) T lymphocytes. Therefore, vitamin A deficiency alters splenic DC subpopulations, which may contribute to skewed immune responses of vitamin A-deficient populations.


Cytometry Part A | 2009

The identification and enumeration of dendritic cell populations from individual mouse spleen and Peyer's patches using flow cytometric analysis

David M. Duriancik; Kathleen A. Hoag

Dendritic cell (DC) research currently involves pooling of tissues from multiple animals followed by enrichment techniques to obtain sufficient numbers of DCs for analysis. Enrichment techniques take advantage of DC adherence, buoyant density properties, and/or positive or negative selection of cell populations using monoclonal antibodies. However, enrichment techniques may significantly change the maturation and/or activation status of DCs or selectively eliminate one or more subpopulations of DCs. To overcome these drawbacks, we designed a multicolor flow cytometric technique for simultaneous analysis of DC populations from tissues of individual mice. The spleens and Peyers patches were mechanically and enzymatically digested, then incubated with a panel of six monoclonal antibody‐fluorochrome direct conjugate reagents. A BD® Biosciences LSR II flow cytometer and FCS Express® software were used to identify three subtypes of mature DCs (myeloid, lymphoid, and plasmacytoid), precursor DCs, polymorphonuclear neutrophils, B lymphocytes, and Gr‐1+/CD8α+ memory T lymphocytes in the spleen. Likewise, we also identified these DC subpopulations and B lymphocytes in the Peyers patches. The three key parameters in analysis of the DC populations were bi‐exponential plotting in data analysis, collection of a minimum of 50,000 total events, and accurate color compensation. This procedure to analyze DCs from individual mice can lead to further understanding of the role of DCs in many other model systems as well as better understanding of how dietary or physiological factors may affect in vivo DC homeostasis.


Clinical and Vaccine Immunology | 2009

Antibodies in the sera of host species with pythiosis recognize a variety of unique immunogens in geographically divergent Pythium insidiosum strains

Ariya Chindamporn; Raquel Vilela; Kathleen A. Hoag; Leonel Mendoza

ABSTRACT Studies by Western blot analyses have shown that antibodies in the sera of host species infected by Pythium insidiosum recognized several prominent proteins expressed by this fungus-like pathogen. Although these studies have utilized sera from infected patients and relevant local strains of P. insidiosum, the results are difficult to compare because of the lack of method standardization. In an effort to resolve this issue, we have utilized standardized methodologies to evaluate six P. insidiosum strains from Asia and the Americas and 15 serum samples from cattle, cats, dogs, horses, and humans with pythiosis from the same geographical regions. Our data show that the antibodies present in these sera recognize a wide variety of unique P. insidiosum immunogenic proteins. Although some of the prominent proteins in this study have been previously reported, several others have yet to be described. For instance, a ∼28-kDa-molecular-mass antigen was detected by the antibodies in all serum samples evaluated. However, this antigen was strongly expressed by only one of the strains evaluated. A diffuse ∼51-kDa protein was not detected by the antibodies in the human sera; but it was recognized by the antibodies in the sera of cattle, cats, dogs, and horses. This antigen was expressed by only two of the strains investigated. Several other similar examples were also observed. The variation of the P. insidiosum protein profile identified by the antibodies in the sera evaluated indicates that some geographically diverged P. insidosum strains expressed some unique immunogens in vitro and that during natural infection (in vivo) P. insidiosum might express a broader number of antigens variably detected by individuals within the same species but especially across species.

Collaboration


Dive into the Kathleen A. Hoag's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Colleen E. Hayes

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Faye E. Nashold

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nancy E. Street

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joan Goverman

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Karen Clise-Dwyer

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge