Network


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

Hotspot


Dive into the research topics where Kellsey Johannes is active.

Publication


Featured researches published by Kellsey Johannes.


Science | 2008

Deletional Tolerance Mediated by Extrathymic Aire-Expressing Cells

James M. Gardner; Jason DeVoss; Rachel S. Friedman; David J. Wong; Ying X. Tan; Xuyu Zhou; Kellsey Johannes; Maureen A. Su; Howard Y. Chang; Matthew F. Krummel; Mark S. Anderson

The prevention of autoimmunity requires the elimination of self-reactive T cells during their development and maturation. The expression of diverse self-antigens by stromal cells in the thymus is essential to this process and depends, in part, on the activity of the autoimmune regulator (Aire) gene. Here we report the identification of extrathymic Aire-expressing cells (eTACs) resident within the secondary lymphoid organs. These stromally derived eTACs express a diverse array of distinct self-antigens and are capable of interacting with and deleting naïve autoreactive T cells. Using two-photon microscopy, we observed stable antigen-specific interactions between eTACs and autoreactive T cells. We propose that such a secondary network of self-antigen–expressing stromal cells may help reinforce immune tolerance by preventing the maturation of autoreactive T cells that escape thymic negative selection.


Journal of Experimental Medicine | 2006

Spontaneous autoimmunity prevented by thymic expression of a single self-antigen

Jason DeVoss; Yafei Hou; Kellsey Johannes; Wen Lu; Gregory I. Liou; John L. Rinn; Howard Y. Chang; Rachel R. Caspi; Lawrence Fong; Mark S. Anderson

The expression of self-antigen in the thymus is believed to be responsible for the deletion of autoreactive T lymphocytes, a critical process in the maintenance of unresponsiveness to self. The Autoimmune regulator (Aire) gene, which is defective in the disorder autoimmune polyglandular syndrome type 1, has been shown to promote the thymic expression of self-antigens. A clear link, however, between specific thymic self-antigens and a single autoimmune phenotype in this model has been lacking. We show that autoimmune eye disease in aire-deficient mice develops as a result of loss of thymic expression of a single eye antigen, interphotoreceptor retinoid-binding protein (IRBP). In addition, lack of IRBP expression solely in the thymus, even in the presence of aire expression, is sufficient to trigger spontaneous eye-specific autoimmunity. These results suggest that failure of thymic expression of selective single self-antigens can be sufficient to cause organ-specific autoimmune disease, even in otherwise self-tolerant individuals.


Journal of Clinical Investigation | 2008

Mechanisms of an autoimmunity syndrome in mice caused by a dominant mutation in Aire

Maureen A. Su; Karen Giang; Kristina Žumer; Huimin Jiang; Irena Oven; John L. Rinn; Jason DeVoss; Kellsey Johannes; Wen Lu; James M. Gardner; Angela Chang; Paula A. Bubulya; Howard Y. Chang; B. Matija Peterlin; Mark S. Anderson

Homozygous loss-of-function mutations in AIRE cause autoimmune polyglandular syndrome type 1 (APS 1), which manifests in a classic triad of hypoparathyroidism, adrenal insufficiency, and candidiasis. Interestingly, a kindred with a specific G228W AIRE variant presented with an autosomal dominant autoimmune phenotype distinct from APS 1. We utilized a novel G228W-knockin mouse model to show that this variant acted in a dominant-negative manner to cause a unique autoimmunity syndrome. In addition, the expression of a large number of Aire-regulated thymic antigens was partially inhibited in these animals, demonstrating the importance of quantitative changes in thymic antigen expression in determining organ-specific autoimmunity. Furthermore, the dominant-negative effect of the G228W variant was exerted through recruitment of WT Aire away from active sites of transcription in the nucleus of medullary thymic epithelial cells in vivo. Together, these results may demonstrate a mechanism by which autoimmune predisposition to phenotypes distinct from APS 1 can be mediated in a dominant-negative fashion by Aire.


Immunity | 2013

Extrathymic Aire-Expressing Cells Are a Distinct Bone Marrow-Derived Population that Induce Functional Inactivation of CD4+ T Cells

James M. Gardner; Todd Metzger; Eileen J. McMahon; Byron B. Au-Yeung; Anna K. Krawisz; Wen Lu; Jeffrey D. Price; Kellsey Johannes; Ansuman T. Satpathy; Kenneth M. Murphy; Kristin V. Tarbell; Arthur Weiss; Mark S. Anderson

The autoimmune regulator (Aire) is essential for prevention of autoimmunity; its role is best understood in the thymus, where it promotes self-tolerance through tissue-specific antigen (TSA) expression. Recently, extrathymic Aire-expressing cells (eTACs) have been described in murine secondary lymphoid organs, but the identity of such cells and their role in immune tolerance remains unclear. Here we have shown that eTACs are a discrete major histocompatibility complex class II (MHC II)(hi), CD80(lo), CD86(lo), epithelial cell adhesion molecule (EpCAM)(hi), CD45(lo) bone marrow-derived peripheral antigen-presenting cell (APC) population. We also have demonstrated that eTACs can functionally inactivate CD4⁺ T cells through a mechanism that does not require regulatory T cells (Treg) and is resistant to innate inflammatory stimuli. Together, these findings further define eTACs as a distinct tolerogenic cell population in secondary lymphoid organs.


Journal of Immunology | 2008

Effector Mechanisms of the Autoimmune Syndrome in the Murine Model of Autoimmune Polyglandular Syndrome Type 1

Jason DeVoss; Anthony K. Shum; Kellsey Johannes; Wen Lu; Anna K. Krawisz; Peter L. Wang; Ting Yang; Norbert P. LeClair; Cecilia Austin; Erich C. Strauss; Mark S. Anderson

Mutations in the Aire gene result in a clinical phenomenon known as Autoimmune Polyglandular Syndrome (APS) Type I, which classically manifests as a triad of adrenal insufficiency, hypoparathyroidism, and chronic mucocutaneous infections. In addition to this triad, a number of other autoimmune diseases have been observed in APS1 patients including Sjögren’s syndrome, vitiligo, alopecia, uveitis, and others. Aire-deficient mice, the animal model for APS1, have highlighted the role of the thymus in the disease process and demonstrated a failure in central tolerance in aire-deficient mice. However, autoantibodies have been observed against multiple organs in both mice and humans, making it unclear what the specific role of B and T cells are in the pathogenesis of disease. Using the aire-deficient mouse as a preclinical model for APS1, we have investigated the relative contribution of specific lymphocyte populations, with the goal of identifying the cell populations which may be targeted for rational therapeutic design. In this study, we show that T cells are indispensable to the breakdown of self-tolerance, in contrast to B cells which play a more limited role in autoimmunity. Th1 polarized CD4+ T cells, in particular, are major contributors to the autoimmune response. With this knowledge, we go on to use therapies targeted at T cells to investigate their ability to modulate disease in vivo. Depletion of CD4+ T cells using a neutralizing Ab ameliorated the disease process. Thus, therapies targeted specifically at the CD4+ T cell subset may help control autoimmune disease in patients with APS1.


Science Translational Medicine | 2009

Identification of an Autoantigen Demonstrates a Link Between Interstitial Lung Disease and a Defect in Central Tolerance

Anthony K. Shum; Jason DeVoss; Catherine L. Tan; Yafei Hou; Kellsey Johannes; Clodagh S. O'Gorman; Kirk D. Jones; Etienne B. Sochett; Lawrence Fong; Mark S. Anderson

A defect in immune tolerance to self-antigens may underlie the lung damage that accompanies many autoimmune diseases. Autoimmune diseases are intractable and varied, appearing in many guises. Patients with lupus, for example, experience destructive immune reactions that affect the heart, other organs, and the joints. This generalized attack is in stark contrast to the very selective autoimmune destruction of insulin-producing β islet cells in type I diabetes or the antibodies to the acetylcholine receptor that characterize myasthenia gravis. Why are there generalized immune responses in some diseases and very specific reactions in others? Shum et al. have exploited a mutant mouse defective in AIRE, a transcription factor that controls expression of many self-antigens in the thymus. As immune T cells pass through, these self-antigens in the thymus trigger the deletion of cells that react with them, preventing these autoreactive T cells from entering the circulation. Thus, when AIRE is absent or mutated, T cells directed at self-antigens are not deleted and escape into the circulation where they can trigger autoimmunity. This is not only true in mice: Patients with a genetic disease called autoimmune polyglandular syndrome type 1 (APS1) have defective AIRE and suffer from autoimmune disease. One organ that often deteriorates in autoimmune diseases is the lung, which can develop severe fibrosis, preventing effective breathing and oxygen exchange. To investigate the basis of this interstitial lung disease and other similar problems that arise sporadically, Shum et al. explored how lung damage occurs in AIRE-deficient mice and in a patient with APS1. The diseased lungs from both mice and human were infiltrated with T helper 1 CD4+ cells, with smaller numbers of other immune cells. Further work showed that one of the main targets of these cells in mice was, unexpectedly, a protein called vomeromodulin, which the authors determined is expressed in the thymus under AIRE control. Indeed, activated T cells to vomeromodulin cause lung-specific disease when given to immunodeficient mice. In the patient with the same genetic defect as the mice, the lung antigen was not vomeromodulin but was a similar protein called LPLUNC1 (long palate, lung, and nasal epithelium carcinoma–associated protein), located adjacent to the human version of vomeromodulin, which is a pseudogene. This multifaceted study offers several benefits. The lung-specific autoimmune disease seen in the AIRE-deficient mouse closely resembles human interstitial lung disease and can provide a tractable way to understand its progression and develop treatments. This clear example showing how a failure in establishing tolerance to self-antigens can produce an organ-specific disease points to similar mechanisms as possible causes of other autoimmune problems. Finally, the new results elevate vomeromodulin—and its human cousin LPLUNC1—from an interesting secretion product of the nasal epithelium to an autoimmune target in a serious disease, increasing our understanding of how to combat these illnesses. Interstitial lung disease (ILD) is a common manifestation of systemic autoimmunity characterized by progressive inflammation or scarring of the lungs. Patients who develop these complications can exhibit significantly impaired gas exchange that may result in hypoxemia, pulmonary hypertension, and even death. Unfortunately, little is understood about how these diseases arise, including the role of specific defects in immune tolerance. Another key question is whether autoimmune responses targeting the lung parenchyma are critical to ILD pathogenesis, including that of isolated idiopathic forms. We show that a specific defect in central tolerance brought about by mutations in the autoimmune regulator gene (Aire) leads to an autoreactive T cell response to a lung antigen named vomeromodulin and the development of ILD. We found that a human patient and mice with defects in Aire develop similar lung pathology, demonstrating that the AIRE-deficient model of autoimmunity is a suitable translational system in which to unravel fundamental mechanisms of ILD pathogenesis.


Cell Reports | 2013

Lineage tracing and cell ablation identify a post-Aire-expressing thymic epithelial cell population.

Todd Metzger; Imran S. Khan; James M. Gardner; Maria L. Mouchess; Kellsey Johannes; Anna K. Krawisz; Katarzyna M. Skrzypczynska; Mark S. Anderson

Thymic epithelial cells in the medulla (mTECs) play a critical role in enforcing central tolerance through expression and presentation of tissue-specific antigens (TSAs) and deletion of autoreactive thymocytes. TSA expression requires autoimmune regulator (Aire), a transcriptional activator present in a subset of mTECs characterized by high CD80 and major histocompatibility complex II expression and a lack of potential for differentiation or proliferation. Here, using an Aire-DTR transgenic line, we show that short-term ablation specifically targets Aire(+) mTECs, which quickly undergo RANK-dependent recovery. Repeated ablation also affects Aire(-) mTECs, and using an inducible Aire-Cre fate-mapping system, we find that this results from the loss of a subset of mTECs that showed prior expression of Aire, maintains intermediate TSA expression, and preferentially migrates toward the center of the medulla. These results clearly identify a distinct stage of mTEC development and underscore the diversity of mTECs that play a key role in maintaining tolerance.


Journal of Immunology | 2012

Defective Autoimmune Regulator-Dependent Central Tolerance to Myelin Protein Zero Is Linked to Autoimmune Peripheral Neuropathy

Maureen A. Su; Dan Davini; Philip Cheng; Karen Giang; Una Fan; Jason DeVoss; Kellsey Johannes; Lorelei C. Taylor; Anthony K. Shum; Mariella Valenzise; Antonella Meloni; Hélène Bour-Jordan; Mark S. Anderson

Chronic inflammatory demyelinating polyneuropathy is a debilitating autoimmune disease characterized by peripheral nerve demyelination and dysfunction. How the autoimmune response is initiated, identity of provoking Ags, and pathogenic effector mechanisms are not well defined. The autoimmune regulator (Aire) plays a critical role in central tolerance by promoting thymic expression of self-Ags and deletion of self-reactive T cells. In this study, we used mice with hypomorphic Aire function and two patients with Aire mutations to define how Aire deficiency results in spontaneous autoimmune peripheral neuropathy. Autoimmunity against peripheral nerves in both mice and humans targets myelin protein zero, an Ag for which expression is Aire-regulated in the thymus. Consistent with a defect in thymic tolerance, CD4+ T cells are sufficient to transfer disease in mice and produce IFN-γ in infiltrated peripheral nerves. Our findings suggest that defective Aire-mediated central tolerance to myelin protein zero initiates an autoimmune Th1 effector response toward peripheral nerves.


Journal of Immunology | 2010

An Autoimmune Response to Odorant Binding Protein 1a Is Associated with Dry Eye in the Aire-Deficient Mouse

Jason DeVoss; Norbert P. LeClair; Yafei Hou; Navdeep K. Grewal; Kellsey Johannes; Wen Lu; Ting Yang; Craig Meagher; Lawrence Fong; Erich C. Strauss; Mark S. Anderson

Sjögren’s Syndrome (SS) is a human autoimmune disease characterized by immune-mediated destruction of the lacrimal and salivary glands. In this study, we show that the Aire-deficient mouse represents a new tool to investigate autoimmune dacryoadenitis and keratoconjunctivitis sicca, features of SS. Previous work in the Aire-deficient mouse suggested a role for α-fodrin, a ubiquitous Ag, in the disease process. Using an unbiased biochemical approach, however, we have identified a novel lacrimal gland autoantigen, odorant binding protein 1a, targeted by the autoimmune response. This novel autoantigen is expressed in the thymus in an Aire-dependent manner. The results from our study suggest that defects in central tolerance may contribute to SS and provide a new and clinically relevant model to investigate the pathogenic mechanisms in lacrimal gland autoimmunity and associated ocular surface sequelae.


Journal of Experimental Medicine | 2007

Erratum: (Journal of Experimental Medicine (November 27, 2006) 203: 12 (2727-2735))

Jason DeVoss; Yafei Hou; Kellsey Johannes; Wen Lu; Gregory I. Liou; John L. Rinn; Howard Y. Chang; Rachel R. Caspi; Lawrence Fong; Mark S. Anderson

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

Collaboration


Dive into the Kellsey Johannes's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wen Lu

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lawrence Fong

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge