23 results on '"Corey N. Miller"'
Search Results
2. Combined transient ablation and single-cell RNA-sequencing reveals the development of medullary thymic epithelial cells
- Author
-
Kristen L Wells, Corey N Miller, Andreas R Gschwind, Wu Wei, Jonah D Phipps, Mark S Anderson, and Lars M Steinmetz
- Subjects
single-cell transcriptomics ,medullary thymic epithelial cell ,immune system ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Medullary thymic epithelial cells (mTECs) play a critical role in central immune tolerance by mediating negative selection of autoreactive T cells through the collective expression of the peripheral self-antigen compartment, including tissue-specific antigens (TSAs). Recent work has shown that gene-expression patterns within the mTEC compartment are heterogenous and include multiple differentiated cell states. To further define mTEC development and medullary epithelial lineage relationships, we combined lineage tracing and recovery from transient in vivo mTEC ablation with single-cell RNA-sequencing in Mus musculus. The combination of bioinformatic and experimental approaches revealed a non-stem transit-amplifying population of cycling mTECs that preceded Aire expression. We propose a branching model of mTEC development wherein a heterogeneous pool of transit-amplifying cells gives rise to Aire- and Ccl21a-expressing mTEC subsets. We further use experimental techniques to show that within the Aire-expressing developmental branch, TSA expression peaked as Aire expression decreased, implying Aire expression must be established before TSA expression can occur. Collectively, these data provide a roadmap of mTEC development and demonstrate the power of combinatorial approaches leveraging both in vivo models and high-dimensional datasets.
- Published
- 2020
- Full Text
- View/download PDF
3. SIV clearance from neonatal macaques following transient CCR5 depletion
- Author
-
Jesse D. Deere, David Merriam, Kawthar Machmach Leggat, Wen-Lan William Chang, Gema Méndez-Lagares, Hung Kieu, Joseph Dutra, Justin Fontaine, Wenze Lu, Ning Chin, Connie Chen, Bryant Chi-Thien Tran, Jessica Salinas, Corey N. Miller, Steven G. Deeks, Jeffrey D. Lifson, Kathleen Engelman, Diogo Magnani, Keith Reimann, Mario Stevenson, and Dennis J. Hartigan-O’Connor
- Subjects
Article - Abstract
SUMMARY PARAGRAPHTreatment of people with HIV (PWH) with antiretroviral therapy (ART) results in sustained suppression of viremia, but HIV persists indefinitely as integrated provirus in CD4-expressing cells. Intact persistent provirus, the “rebound competent viral reservoir” (RCVR), is the primary obstacle to achieving a cure. Most variants of HIV enter CD4+T cells by binding to the chemokine receptor, CCR5. The RCVR has been successfully depleted only in a handful of PWH following cytotoxic chemotherapy and bone marrow transplantation from donors with a mutation inCCR5. Here we show that long-term SIV remission and apparent cure can be achieved for infant macaques via targeted depletion of potential reservoir cells that express CCR5. Neonatal rhesus macaques were infected with virulent SIVmac251, then treated with ART beginning one week after infection, followed by treatment with either a CCR5/CD3-bispecific or a CD4-specific antibody, both of which depleted target cells and increased the rate of plasma viremia decrease. Upon subsequent cessation of ART, three of seven animals treated with CCR5/CD3-bispecific antibody rebounded quickly and two rebounded 3 or 6 months later. Remarkably, the other two animals remained aviremic and efforts to detect replication-competent virus were unsuccessful. Our results show that bispecific antibody treatment can achieve meaningful SIV reservoir depletion and suggest that functional HIV cure might be achievable for recently infected individuals having a restricted reservoir.
- Published
- 2023
4. Single-cell transcriptional profiling of human thymic stroma uncovers novel cellular heterogeneity in the thymic medulla
- Author
-
Lauren Byrnes, Julie B. Sneddon, James M. Gardner, Mark S. Anderson, Corey N. Miller, Jessica Chavez, Joe Germino, Trevor D. Burt, Nathan T. Cramer, Audrey Parent, Vasilis Ntranos, Jhoanne L. Bautista, Chun Jimmie Ye, and David I. Berrios
- Subjects
Adult ,0301 basic medicine ,Central tolerance ,T-Lymphocytes ,Mesenchyme ,Science ,Population ,Cell ,Systems analysis ,General Physics and Astronomy ,Autoimmunity ,Thymus Gland ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Immune tolerance ,Mesoderm ,Genetic Heterogeneity ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigen ,Stroma ,medicine ,Animals ,Humans ,Cell Lineage ,education ,education.field_of_study ,Thymocytes ,Multidisciplinary ,Gene Expression Profiling ,Endothelial Cells ,Cell Differentiation ,Epithelial Cells ,General Chemistry ,Thymus ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,bacteria ,Single-Cell Analysis ,Pericytes ,Thymocyte migration - Abstract
The thymus’ key function in the immune system is to provide the necessary environment for the development of diverse and self-tolerant T lymphocytes. While recent evidence suggests that the thymic stroma is comprised of more functionally distinct subpopulations than previously appreciated, the extent of this cellular heterogeneity in the human thymus is not well understood. Here we use single-cell RNA sequencing to comprehensively profile the human thymic stroma across multiple stages of life. Mesenchyme, pericytes and endothelial cells are identified as potential key regulators of thymic epithelial cell differentiation and thymocyte migration. In-depth analyses of epithelial cells reveal the presence of ionocytes as a medullary population, while the expression of tissue-specific antigens is mapped to different subsets of epithelial cells. This work thus provides important insight on how the diversity of thymic cells is established, and how this heterogeneity contributes to the induction of immune tolerance in humans., The thymus supports T cell immunity by providing the environment for thymocyte differentiation. Here the authors profile human thymic stroma at the single cell level, identifying ionocytes as a new medullary population and defining tissue specific antigen expression in multiple stromal cell types.
- Published
- 2021
5. Author response: Combined transient ablation and single-cell RNA-sequencing reveals the development of medullary thymic epithelial cells
- Author
-
Wu Wei, Corey N. Miller, Mark S. Anderson, Jonah D Phipps, Kristen L. Wells, Andreas R Gschwind, and Lars M. Steinmetz
- Subjects
medicine.anatomical_structure ,Medullary cavity ,Chemistry ,medicine.medical_treatment ,Cell ,medicine ,RNA ,Transient (oscillation) ,Ablation ,Cell biology - Published
- 2020
- Full Text
- View/download PDF
6. Combined transient ablation and single cell RNA sequencing reveals the development of medullary thymic epithelial cells
- Author
-
Lars M. Steinmetz, Wu Wei, Mark S. Anderson, Andreas R Gschwind, Kristen L. Wells, Corey N. Miller, and Jonah D Phipps
- Subjects
education.field_of_study ,Cellular differentiation ,Cell ,Population ,RNA ,Biology ,Cell biology ,Immune tolerance ,Negative selection ,medicine.anatomical_structure ,Antigen ,Gene expression ,medicine ,education - Abstract
Medullary thymic epithelial cells (mTECs) play a critical role in central immune tolerance by mediating negative selection of autoreactive T cells through the collective expression of the peripheral self-antigen compartment, including tissue-specific antigens (TSAs). Recent work has shown that gene expression patterns within the mTEC compartment are remarkably heterogenous and include multiple differentiated cell states. To further define mTEC development and medullary epithelial lineage relationships, we combined lineage tracing and recovery from transient in vivo mTEC ablation with single cell RNA-sequencing. The combination of bioinformatic and experimental approaches revealed a non-stem transit-amplifying population of cycling mTECs that preceded Aire expression. Based on our findings, we propose a branching model of mTEC development wherein a heterogeneous pool of transit-amplifying cells gives rise to Aire- and Ccl21a-expressing mTEC subsets. We further use experimental techniques to show that within the Aire-expressing developmental branch, TSA expression peaked as Aire expression decreased, implying Aire expression must be established before TSA expression can occur. Collectively, these data provide a higher order roadmap of mTEC development and demonstrate the power of combinatorial approaches leveraging both in vivo models and high-dimensional datasets.
- Published
- 2020
- Full Text
- View/download PDF
7. Combined transient ablation and single-cell RNA-sequencing reveals the development of medullary thymic epithelial cells
- Author
-
Wu Wei, Kristen L. Wells, Jonah D Phipps, Andreas R Gschwind, Corey N. Miller, Lars M. Steinmetz, and Mark S. Anderson
- Subjects
0301 basic medicine ,Mouse ,Cellular differentiation ,Cell ,Inbred C57BL ,Immune tolerance ,immunology ,Negative selection ,Mice ,0302 clinical medicine ,Immunology and Inflammation ,genetics ,Biology (General) ,education.field_of_study ,General Neuroscience ,medullary thymic epithelial cell ,Cell Differentiation ,General Medicine ,Cell biology ,Mutant Strains ,medicine.anatomical_structure ,Medicine ,Stem Cell Research - Nonembryonic - Non-Human ,Single-Cell Analysis ,Sequence Analysis ,Research Article ,QH301-705.5 ,Science ,1.1 Normal biological development and functioning ,Population ,Thymus Gland ,Biology ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Immune system ,Antigen ,Underpinning research ,medicine ,genomics ,Animals ,Cell Lineage ,education ,mouse ,General Immunology and Microbiology ,Sequence Analysis, RNA ,RNA ,Epithelial Cells ,Genetics and Genomics ,Stem Cell Research ,Mice, Mutant Strains ,Mice, Inbred C57BL ,immune system ,030104 developmental biology ,inflammation ,Biochemistry and Cell Biology ,single-cell transcriptomics ,030217 neurology & neurosurgery - Abstract
Medullary thymic epithelial cells (mTECs) play a critical role in central immune tolerance by mediating negative selection of autoreactive T cells through the collective expression of the peripheral self-antigen compartment, including tissue-specific antigens (TSAs). Recent work has shown that gene-expression patterns within the mTEC compartment are heterogenous and include multiple differentiated cell states. To further define mTEC development and medullary epithelial lineage relationships, we combined lineage tracing and recovery from transient in vivo mTEC ablation with single-cell RNA-sequencing in Mus musculus. The combination of bioinformatic and experimental approaches revealed a non-stem transit-amplifying population of cycling mTECs that preceded Aire expression. We propose a branching model of mTEC development wherein a heterogeneous pool of transit-amplifying cells gives rise to Aire- and Ccl21a-expressing mTEC subsets. We further use experimental techniques to show that within the Aire-expressing developmental branch, TSA expression peaked as Aire expression decreased, implying Aire expression must be established before TSA expression can occur. Collectively, these data provide a roadmap of mTEC development and demonstrate the power of combinatorial approaches leveraging both in vivo models and high-dimensional datasets., eLife digest Specialized cells in the immune system known as T cells protect the body from infection by destroying disease-causing microbes, such as bacteria or viruses. T cells use proteins on their surface called receptors to stick to infectious microbes and remove them from the body. Some newly developed T-cells, however, contain receptors that recognize and bind to cells that belong in the body. If these faulty T cells are released, they can attack healthy tissues and cause an autoimmune disease. After a new T cell is developed, it gets carried to a gland in the chest known as the thymus. Cells in the thymus called mTECs screen T cells for receptors that may bind to the body’s tissues. mTECs do this by presenting T cells with proteins that are commonly found on the surface of healthy cells in the body. If a T cell recognizes any of these ‘tissue specific proteins’, it is destroyed or given a new role in the body. Some faulty T cells, however, still manage to evade detection. One way to uncover why this might happen is to investigate how mTECs develop. Previous work showed that mTECs transition through various stages before reaching their final form. However, the order in which these events occur remained unclear. To gain a better understanding of these developmental steps, Wells, Miller et al. extracted mTECs from the thymus of mice and analyzed the genetic make-up of individual cells. This uncovered a missing link in mTEC development: a new type of cell that is the immediate predecessor of the final mTEC. These ‘predecessor’ cells were actively growing, highlighting that mTECs can be constantly generated in the body. By probing the genes that generate tissue-specific proteins in mTECs, Wells, Miller et al. revealed that these proteins were only produced for short periods and in the late stages of mTEC development. These findings contribute to our understanding of how mTECs develop to screen T cells. Mapping these developmental stages will make it easier to identify when faulty T cells are able to evade mTECs. This will lead to earlier detection of autoimmune diseases which could result in better treatments.
- Published
- 2020
8. Thymic regulatory T cells arise via two distinct developmental programs
- Author
-
Rebecca S. LaRue, Louisa E. Sjaastad, Mark S. Anderson, Calvin B. Williams, Praful Aggarwal, Maria-Luisa Alegre, Ulrich Broeckel, Shawn A. Mahmud, Irina Proekt, Dimitre R. Simeonov, Jason B. Williams, Michael A. Farrar, Weishan Huang, Jonathan C. Jeschke, Corey N. Miller, Roland Ruscher, David L. Owen, Christine Henzler, Justin A. Spanier, Can Hekim, Avery August, Alexander Marson, and Ye Zheng
- Subjects
0301 basic medicine ,Encephalomyelitis, Autoimmune, Experimental ,Transgene ,Immunology ,Freund's Adjuvant ,Mice, Transgenic ,chemical and pharmacologic phenomena ,Thymus Gland ,Biology ,Autoantigens ,T-Lymphocytes, Regulatory ,Article ,Transcriptome ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Antigen ,Immune Tolerance ,Immunology and Allergy ,Animals ,Humans ,IL-2 receptor ,Progenitor cell ,Enhancer ,Interleukin-2 Receptor alpha Subunit ,Forkhead Transcription Factors ,hemic and immune systems ,Cell Differentiation ,Mycobacterium tuberculosis ,Lymphoid Progenitor Cells ,Colitis ,Peptide Fragments ,Cell biology ,Specific Pathogen-Free Organisms ,Disease Models, Animal ,030104 developmental biology ,Apoptosis ,Myelin-Oligodendrocyte Glycoprotein ,Signal transduction ,030215 immunology ,Signal Transduction - Abstract
The developmental programs that generate a broad repertoire of regulatory T cells (Treg cells) able to respond to both self antigens and non-self antigens remain unclear. Here we found that mature Treg cells were generated through two distinct developmental programs involving CD25+ Treg cell progenitors (CD25+ TregP cells) and Foxp3lo Treg cell progenitors (Foxp3lo TregP cells). CD25+ TregP cells showed higher rates of apoptosis and interacted with thymic self antigens with higher affinity than did Foxp3lo TregP cells, and had a T cell antigen receptor repertoire and transcriptome distinct from that of Foxp3lo TregP cells. The development of both CD25+ TregP cells and Foxp3lo TregP cells was controlled by distinct signaling pathways and enhancers. Transcriptomics and histocytometric data suggested that CD25+ TregP cells and Foxp3lo TregP cells arose by coopting negative-selection programs and positive-selection programs, respectively. Treg cells derived from CD25+ TregP cells, but not those derived from Foxp3lo TregP cells, prevented experimental autoimmune encephalitis. Our findings indicate that Treg cells arise through two distinct developmental programs that are both required for a comprehensive Treg cell repertoire capable of establishing immunotolerance.
- Published
- 2019
9. Thymic tuft cells promote an IL4-enriched medulla and shape thymocyte development
- Author
-
Mark S. Anderson, Richard M. Locksley, Lars M. Steinmetz, Corey N. Miller, Joshua L. Pollack, Wint Lwin, Imran S. Khan, Todd C. Metzger, Bruno Kyewski, Aparna R. Rajpurkar, David J. Erle, Kristin Rattay, Irina Proekt, Adam Fries, Haiguang Wang, Kristen L. Wells, Jakob von Moltke, Kristin A. Hogquist, Audrey Parent, and Eric J. Wigton
- Subjects
0301 basic medicine ,Multidisciplinary ,Taste Transduction Pathway ,medicine.medical_treatment ,Lymphocyte differentiation ,Epithelial Cells ,Biology ,Natural killer T cell ,Article ,Cell biology ,Immune tolerance ,03 medical and health sciences ,Thymocyte ,030104 developmental biology ,0302 clinical medicine ,Cytokine ,medicine ,TRPM5 ,Interleukin 4 ,030215 immunology - Abstract
The thymus is responsible for generating a diverse yet self-tolerant pool of T cells1. Although the thymic medulla consists mostly of developing and mature AIRE+ epithelial cells, recent evidence has suggested that there is far greater heterogeneity among medullary thymic epithelial cells than was previously thought2. Here we describe in detail an epithelial subset that is remarkably similar to peripheral tuft cells that are found at mucosal barriers3. Similar to the periphery, thymic tuft cells express the canonical taste transduction pathway and IL-25. However, they are unique in their spatial association with cornified aggregates, ability to present antigens and expression of a broad diversity of taste receptors. Some thymic tuft cells pass through an Aire-expressing stage and depend on a known AIRE-binding partner, HIPK2, for their development. Notably, the taste chemosensory protein TRPM5 is required for their thymic function through which they support the development and polarization of thymic invariant natural killer T cells and act to establish a medullary microenvironment that is enriched in the type 2 cytokine, IL-4. These findings indicate that there is a compartmentalized medullary environment in which differentiation of a minor and highly specialized epithelial subset has a non-redundant role in shaping thymic function.
- Published
- 2018
10. Adaptation to ER stress is mediated by differential stabilities of pro-survival and pro-apoptotic mRNAs and proteins.
- Author
-
D Thomas Rutkowski, Stacey M Arnold, Corey N Miller, Jun Wu, Jack Li, Kathryn M Gunnison, Kazutoshi Mori, Amir A Sadighi Akha, David Raden, and Randal J Kaufman
- Subjects
Biology (General) ,QH301-705.5 - Abstract
The accumulation of unfolded proteins in the endoplasmic reticulum (ER) activates a signaling cascade known as the unfolded protein response (UPR). Although activation of the UPR is well described, there is little sense of how the response, which initiates both apoptotic and adaptive pathways, can selectively allow for adaptation. Here we describe the reconstitution of an adaptive ER stress response in a cell culture system. Monitoring the activation and maintenance of representative UPR gene expression pathways that facilitate either adaptation or apoptosis, we demonstrate that mild ER stress activates all UPR sensors. However, survival is favored during mild stress as a consequence of the intrinsic instabilities of mRNAs and proteins that promote apoptosis compared to those that facilitate protein folding and adaptation. As a consequence, the expression of apoptotic proteins is short-lived as cells adapt to stress. We provide evidence that the selective persistence of ER chaperone expression is also applicable to at least one instance of genetic ER stress. This work provides new insight into how a stress response pathway can be structured to allow cells to avert death as they adapt. It underscores the contribution of posttranscriptional and posttranslational mechanisms in influencing this outcome.
- Published
- 2006
- Full Text
- View/download PDF
11. Exercise Is Medicine On Campus At Michigan Tech: Promoting Physical Activity During The Covid-19 Pandemic
- Author
-
Gregory J. Miodonski, Steven J. Elmer, Emma Huhta, Jamie J. Phillips, Isaac J. Wedig, Corey N. Miller, Grant S. Thivierge, Ari M. Laiho, and Jenna L. Phelps
- Subjects
2019-20 coronavirus outbreak ,Coronavirus disease 2019 (COVID-19) ,business.industry ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Environmental health ,Pandemic ,Physical activity ,Medicine ,Physical Therapy, Sports Therapy and Rehabilitation ,Orthopedics and Sports Medicine ,business - Published
- 2021
- Full Text
- View/download PDF
12. Insights into immune tolerance from AIRE deficiency
- Author
-
Corey N. Miller, Michail S. Lionakis, Irina Proekt, and Mark S. Anderson
- Subjects
0301 basic medicine ,1.1 Normal biological development and functioning ,Immunology ,Thymus Gland ,Biology ,Autoimmune Disease ,Autoantigens ,Article ,Immune tolerance ,Mice ,03 medical and health sciences ,Antigen ,Underpinning research ,Immunity ,Immune Tolerance ,Animals ,Humans ,Immunology and Allergy ,Polyendocrinopathies, Autoimmune ,Transcription factor ,Regulation of gene expression ,Inflammatory and immune system ,Peripheral tolerance ,Phenotype ,030104 developmental biology ,Polyendocrinopathies ,Gene Expression Regulation ,Central tolerance ,Autoimmune ,Transcription Factors - Abstract
AIRE is a well-established master regulator of central tolerance. It plays an essential role in driving expression of tissue-specific antigens in the thymus and shaping the development of positively selected T-cells. Humans and mice with compromised or absent AIRE function have markedly variable phenotypes that include a range of autoimmune manifestations. Recent evidence suggests that this variability stems from cooperation of autoimmune susceptibilities involving both central and peripheral tolerance checkpoints. Here we discuss the broadening understanding of the factors that influence Aire expression, modify AIRE function, and the impact and intersection of AIRE with peripheral immunity. This rapidly expanding body of knowledge will force a reexamination of the definition and clinical management of APS-1 patients as well as provide a foundation for the development of immunomodulatory strategies targeting central tolerance.
- Published
- 2017
- Full Text
- View/download PDF
13. Revisiting the Road Map of Medullary Thymic Epithelial Cell Differentiation
- Author
-
Sheena Pinto, Bruno Kyewski, Benedikt Brors, Corey N. Miller, Chloé Michel, Rita Küchler, and Mark S. Anderson
- Subjects
0301 basic medicine ,Programmed cell death ,Cellular differentiation ,Immunology ,Cell ,Apoptosis ,Thymus Gland ,Rats, Sprague-Dawley ,Mice ,03 medical and health sciences ,Agglutinin ,Pregnancy ,Lectins ,medicine ,Animals ,Humans ,Immunology and Allergy ,Cell Lineage ,Cells, Cultured ,Mice, Knockout ,Regulation of gene expression ,MHC class II ,biology ,Histocompatibility Antigens Class II ,FOXN1 ,Cell Differentiation ,Epithelial Cells ,Rats ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,biology.protein ,Female ,Biomarkers ,Transcription Factors - Abstract
The basic two-step terminal differentiation model of the medullary thymic epithelial cell (mTEC) lineage from immature MHC class II (MHCII)lo to mature MHCIIhi mTECs has recently been extended to include a third stage, namely the post-Aire MHCIIlo subset as identified by lineage-tracing models. However, a suitable surface marker distinguishing the phenotypically overlapping pre- from the post-Aire MHCIIlo stage has been lacking. In this study, we introduce the lectin Tetragonolobus purpureas agglutinin (TPA) as a novel cell surface marker that allows for such delineation. Based on our data, we derived the following sequence of mTEC differentiation: TPAloMHCIIlo → TPAloMHCIIhi → TPAhiMHCIIhi → TPAhiMHCIIlo. Surprisingly, in the steady-state postnatal thymus TPAloMHCIIlo pre-Aire rather than terminally differentiated post-Aire TPAhiMHCIIlo mTECs were marked for apoptosis at an exceptionally high rate of ∼70%. Hence, only the minor cycling fraction of the MHCIIlo subset (
- Published
- 2017
- Full Text
- View/download PDF
14. Tolerance checkpoint bypass permits emergence of pathogenic T cells to neuromyelitis optica autoantigen aquaporin-4
- Author
-
Andrés Cruz-Herranz, Michael J. Levy, Peggy P. Ho, Patricia A. Nelson, Marc H. Levin, Raymond A. Sobel, Mark S. Anderson, Lawrence Steinman, Jeffrey Bennett, Collin M. Spencer, Alan S. Verkman, Corey N. Miller, Ryan C. Winger, Scott S. Zamvil, Ari J. Green, Sarah Hagberg, and Sharon A. Sagan
- Subjects
Central Nervous System ,0301 basic medicine ,T-Lymphocytes ,experimental autoimmune encephalomyelitis ,aquaporin-4 ,Epitopes, T-Lymphocyte ,Neurodegenerative ,Inbred C57BL ,Autoantigens ,Transgenic ,Epitope ,experimental neuromyelitis optica ,Mice ,Epitopes ,0302 clinical medicine ,Leukocytes ,Paralysis ,2.1 Biological and endogenous factors ,T-cell receptor ,Aetiology ,Receptor ,tolerance ,Multidisciplinary ,Neuromyelitis Optica ,Biological Sciences ,Flow Cytometry ,Aquaporin 4 ,Female ,medicine.symptom ,Central tolerance ,Multiple Sclerosis ,T cells ,neuromyelitis optica ,Mice, Transgenic ,Biology ,Autoimmune Disease ,Article ,Autoimmune Diseases ,03 medical and health sciences ,Immune Tolerance ,medicine ,Animals ,Eye Disease and Disorders of Vision ,Autoantibodies ,Cell Proliferation ,Inflammation ,Autoimmune disease ,Neuromyelitis optica ,Inflammatory and immune system ,ENMO ,Neurosciences ,medicine.disease ,Brain Disorders ,Mice, Inbred C57BL ,Good Health and Well Being ,030104 developmental biology ,T-Lymphocyte ,Immunization ,Immunoglobulin G ,Immunology ,Th17 Cells ,sense organs ,Spleen ,Epitope Mapping ,030217 neurology & neurosurgery - Abstract
Aquaporin-4 (AQP4)-specific T cells are expanded in neuromyelitis optica (NMO) patients and exhibit Th17 polarization. However, their pathogenic role in CNS autoimmune inflammatory disease is unclear. Although multiple AQP4 T-cell epitopes have been identified in WT C57BL/6 mice, we observed that neither immunization with those determinants nor transfer of donor T cells targeting them caused CNS autoimmune disease in recipient mice. In contrast, robust proliferation was observed following immunization of AQP4-deficient (AQP4-/-) mice with AQP4 peptide (p) 135-153 or p201-220, peptides predicted to contain I-Ab-restricted T-cell epitopes but not identified in WT mice. In comparison with WT mice, AQP4-/- mice used unique T-cell receptor repertoires for recognition of these two AQP4 epitopes. Donor T cells specific for either determinant from AQP4-/-, but not WT, mice induced paralysis in recipient WT and B-cell-deficient mice. AQP4-specific Th17-polarized cells induced more severe disease than Th1-polarized cells. Clinical signs were associated with opticospinal infiltrates of T cells and monocytes. Fluorescent-labeled donor T cells were detected in CNS lesions. Visual system involvement was evident by changes in optical coherence tomography. Fine mapping of AQP4 p201-220 and p135-153 epitopes identified peptides within p201-220 but not p135-153, which induced clinical disease in 40% of WT mice by direct immunization. Our results provide a foundation to evaluate how AQP4-specific T cells contribute to AQP4-targeted CNS autoimmunity (ATCA) and suggest that pathogenic AQP4-specific T-cell responses are normally restrained by central tolerance, which may be relevant to understanding development of AQP4-reactive T cells in NMO.
- Published
- 2016
- Full Text
- View/download PDF
15. Continuous Antigenic Stimulation of DO11.10 TCR Transgenic Mice in the Presence or Absence of IL-1β: Possible Implications for Mechanisms of T Cell Depletion in HIV Disease
- Author
-
Jeff E. Mold, Claire L. Emson, Bridget K. McEvoy-Hein Asgarian, Kristin Ladell, Corey N. Miller, Mette D. Hazenberg, Robert Busch, Marc K. Hellerstein, David Price, Mark Fitch, Joseph M. McCune, Amsterdam institute for Infection and Immunity, Cancer Center Amsterdam, and Clinical Haematology
- Subjects
CD4-Positive T-Lymphocytes ,Male ,Ovalbumin ,T-Lymphocytes ,1.1 Normal biological development and functioning ,T cell ,Transgene ,Molecular Sequence Data ,Interleukin-1beta ,Immunology ,Mice, Transgenic ,HIV Infections ,Stimulation ,Biology ,Lymphocyte Activation ,T-Lymphocytes, Regulatory ,Article ,Transgenic ,Proinflammatory cytokine ,Mice ,Interleukin 21 ,Underpinning research ,medicine ,Animals ,2.1 Biological and endogenous factors ,Immunology and Allergy ,Cytotoxic T cell ,Amino Acid Sequence ,IL-2 receptor ,Aetiology ,Inflammatory and immune system ,FOXP3 ,Regulatory ,Infectious Diseases ,medicine.anatomical_structure ,HIV/AIDS ,Female ,Lymph Nodes ,Infection ,Spleen - Abstract
Untreated HIV disease is associated with chronic immune activation and CD4+ T cell depletion. A variety of mechanisms have been invoked to account for CD4+ T cell depletion in this context, but the quantitative contributions of these proposed mechanisms over time remains unclear. We turned to the DO11.10 TCR transgenic (tg) mouse model, where OVA is recognized in the context of H-2d, to explore the impact of chronic antigenic stimulation on CD4+ T cell dynamics. To model dichotomous states of persistent antigen exposure in the presence or absence of proinflammatory stimulation, we administered OVA peptide (OVAp) to these mice on a continuous basis with or without the prototypic proinflammatory cytokine, interleukin 1β (IL-1β). In both cases, circulating antigen-specific and non-specific CD4+ T cells were depleted. However, in the absence of IL-1β, there was limited proliferation and effector/memory conversion of antigen-specific T cells, depletion of peripheral CD4+ T cells in hematolymphoid organs, and systemic induction of regulatory FoxP3+CD4+ T cells, as often observed in late-stage HIV disease. By contrast, when OVAp was administered in the presence of IL-1β, effector/memory phenotype T cells expanded and the typical symptoms of heightened immune activation were observed. Acknowledging the imperfect and incomplete relationship between antigen-stimulated DO11.10 TCR tg mice and HIV-infected humans, our data suggest that CD4+ T cell depletion in the setting of HIV disease may reflect, at least in part, chronic antigen exposure in the absence of proinflammatory signals and/or appropriate antigen-presenting cell functions.
- Published
- 2015
- Full Text
- View/download PDF
16. LYN- and AIRE-mediated tolerance checkpoint defects synergize to trigger organ-specific autoimmunity
- Author
-
Mark S. Anderson, Anthony L. DeFranco, Marion Jeanne, Douglas B. Gould, Clifford A. Lowell, Kayla J. Fasano, Irina Proekt, Corey N. Miller, and James J. Moon
- Subjects
0301 basic medicine ,CD4-Positive T-Lymphocytes ,T cell ,Antigen presentation ,Immunology ,Mice, Transgenic ,Autoimmunity ,Posterior ,Biology ,medicine.disease_cause ,Inbred C57BL ,Autoimmune Disease ,Medical and Health Sciences ,Transgenic ,Autoimmune Diseases ,Uveitis ,03 medical and health sciences ,Mice ,LYN ,medicine ,Genetics ,Animals ,2.1 Biological and endogenous factors ,Aetiology ,Eye Proteins ,Autoantibodies ,Autoimmune disease ,Mutation ,Antigen Presentation ,Inflammatory and immune system ,Neurosciences ,Peripheral tolerance ,Uveitis, Posterior ,General Medicine ,Dendritic Cells ,Autoimmune regulator ,medicine.disease ,3. Good health ,Gastrointestinal Microbiome ,Mice, Inbred C57BL ,Retinol-Binding Proteins ,030104 developmental biology ,medicine.anatomical_structure ,src-Family Kinases ,Organ Specificity ,Research Article ,Transcription Factors - Abstract
Studies of the genetic factors associated with human autoimmune disease suggest a multigenic origin of susceptibility; however, how these factors interact and through which tolerance pathways they operate generally remain to be defined. One key checkpoint occurs through the activity of the autoimmune regulator AIRE, which promotes central T cell tolerance. Recent reports have described a variety of dominant-negative AIRE mutations that likely contribute to human autoimmunity to a greater extent than previously thought. In families with these mutations, the penetrance of autoimmunity is incomplete, suggesting that other checkpoints play a role in preventing autoimmunity. Here, we tested whether a defect in LYN, an inhibitory protein tyrosine kinase that is implicated in systemic autoimmunity, could combine with an Aire mutation to provoke organ-specific autoimmunity. Indeed, mice with a dominant-negative allele of Aire and deficiency in LYN spontaneously developed organ-specific autoimmunity in the eye. We further determined that a small pool of retinal protein-specific T cells escaped thymic deletion as a result of the hypomorphic Aire function and that these cells also escaped peripheral tolerance in the presence of LYN-deficient dendritic cells, leading to highly destructive autoimmune attack. These findings demonstrate how 2 distinct tolerance pathways can synergize to unleash autoimmunity and have implications for the genetic susceptibility of autoimmune disease.
- Published
- 2016
17. Detection of Succinate by Intestinal Tuft Cells Triggers a Type 2 Innate Immune Circuit
- Author
-
James B. Jaffe, Jakob von Moltke, Marija S. Nadjsombati, Corey N. Miller, Lucian DiPeso, Joshua L. Pollack, Mary F. Fontana, Miranda R. Lyons-Cohen, G. A. Nagana Gowda, Mark S. Anderson, Christoph Schneider, Daniel Raftery, John W. McGinty, David J. Erle, and Richard M. Locksley
- Subjects
Male ,0301 basic medicine ,chemosensing ,Metabolite ,Succinic Acid ,Small ,Inbred C57BL ,urologic and male genital diseases ,Receptors, G-Protein-Coupled ,ILC2 ,Type 2 immune response ,Mice ,chemistry.chemical_compound ,Intestine, Small ,Receptors ,2.1 Biological and endogenous factors ,Immunology and Allergy ,Nippostrongylus brasiliensis ,Intestinal Mucosa ,Aetiology ,Receptor ,helminth ,Mice, Knockout ,Mucosal ,biology ,Innate lymphoid cell ,female genital diseases and pregnancy complications ,Intestine ,Cell biology ,Tritrichomonas ,Infectious Diseases ,medicine.anatomical_structure ,Organ Specificity ,Female ,Nippostrongylus ,Tuft cell ,protist ,Signal Transduction ,Knockout ,Immunology ,TRPM Cation Channels ,Autoimmune Disease ,Cell Line ,G-Protein-Coupled ,03 medical and health sciences ,Th2 Cells ,Species Specificity ,tuft cell ,nippostrongylus brasiliensis ,tritrichomonas ,medicine ,Animals ,Immunity, Mucosal ,Strongylida Infections ,Protozoan Infections ,Innate immune system ,urogenital system ,Inflammatory and immune system ,Immunity ,type 2 immunity ,succinate ,biology.organism_classification ,Small intestine ,Mice, Inbred C57BL ,030104 developmental biology ,chemistry ,Digestive Diseases ,small intestine - Abstract
In the small intestine, type 2 responses are regulated by a signaling circuit that involves tuft cells and group 2 innate lymphoid cells (ILC2s). Here, we identified the microbial metabolite succinate as an activating ligand for small intestinal (SI) tuft cells. Sequencing analyses of tuft cells isolated from the small intestine, gall bladder, colon, thymus, and trachea revealed that expression of tuft cell chemosensory receptors is tissue specific. SI tuft cells expressed the succinate receptor (SUCNR1), and providing succinate in drinking water was sufficient to induce a multifaceted type 2 immune response via the tuft-ILC2 circuit. The helminth Nippostrongylus brasiliensis and a tritrichomonad protist both secreted succinate as a metabolite. Invivo sensing of the tritrichomonad required SUCNR1, whereas N.brasiliensis was SUCNR1 independent. These findings define a paradigm wherein tuft cells monitor microbial metabolites to initiate type 2 immunity and suggest the existence of othersensing pathways triggering the response to helminths.
- Published
- 2018
- Full Text
- View/download PDF
18. Identification of a novel cis-regulatory element essential for immune tolerance
- Author
-
Kayla J. Fasano, Michael Waterfield, Wint Lwin, Grégory Seumois, Mark S. Anderson, Irina Proekt, Corey N. Miller, Pandurangan Vijayanand, and Taylor N. LaFlam
- Subjects
Sequence Homology ,Regulatory Sequences, Nucleic Acid ,medicine.disease_cause ,Inbred C57BL ,Medical and Health Sciences ,Transgenic ,Immune tolerance ,Autoimmunity ,Mice ,Immunology and Allergy ,Research Articles ,Microscopy ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,NF-kappa B ,Autoimmune regulator ,3. Good health ,Cell biology ,Histone ,Central tolerance ,Protein Binding ,Molecular Sequence Data ,Immunology ,Mice, Transgenic ,Thymus Gland ,Autoimmune Disease ,Fluorescence ,Sequence Homology, Nucleic Acid ,medicine ,Immune Tolerance ,Genetics ,Animals ,Humans ,Gene ,Transcription factor ,Base Sequence ,Nucleic Acid ,Prevention ,Inflammatory and immune system ,HEK 293 cells ,Brief Definitive Report ,Epithelial Cells ,DNA ,Mice, Inbred C57BL ,HEK293 Cells ,Microscopy, Fluorescence ,biology.protein ,Transcriptome ,Regulatory Sequences ,Transcription Factors - Abstract
LaFlam et al. identify a novel and highly conserved noncoding DNA element, ACNS1, essential for Aire expression and immune tolerance regulation in thymic epithelial cells. They show that ACNS1 is an NF-κB–responsive element and that its loss results in development of spontaneous autoimmunity in mice., Thymic central tolerance is essential to preventing autoimmunity. In medullary thymic epithelial cells (mTECs), the Autoimmune regulator (Aire) gene plays an essential role in this process by driving the expression of a diverse set of tissue-specific antigens (TSAs), which are presented and help tolerize self-reactive thymocytes. Interestingly, Aire has a highly tissue-restricted pattern of expression, with only mTECs and peripheral extrathymic Aire-expressing cells (eTACs) known to express detectable levels in adults. Despite this high level of tissue specificity, the cis-regulatory elements that control Aire expression have remained obscure. Here, we identify a highly conserved noncoding DNA element that is essential for Aire expression. This element shows enrichment of enhancer-associated histone marks in mTECs and also has characteristics of being an NF-κB-responsive element. Finally, we find that this element is essential for Aire expression in vivo and necessary to prevent spontaneous autoimmunity, reflecting the importance of this regulatory DNA element in promoting immune tolerance.
- Published
- 2015
19. IL-7 production in murine lymphatic endothelial cells and induction in the setting of peripheral lymphopenia
- Author
-
Myeong Sup Lee, Grace Laidlaw, Joseph M. McCune, Dennis J. Hartigan-O'Connor, Shaun R. Coughlin, Mehrdad Matloubian, Corey N. Miller, Ivo Cornelissen, and Donald M. McDonald
- Subjects
Myeloid ,Stromal cell ,Endothelium ,government.form_of_government ,Immunology ,Green Fluorescent Proteins ,Mice, Transgenic ,Biology ,Lymphatic System ,Mice ,Reticular cell ,Lymphocyte homeostasis ,Lymphopenia ,medicine ,STAT5 Transcription Factor ,Immunology and Allergy ,Animals ,Phosphorylation ,Lymph node ,Interleukin-7 ,Endothelial Cells ,General Medicine ,Cell biology ,Lymphatic Endothelium ,medicine.anatomical_structure ,Lymphatic system ,government ,Featured Article of the Month - Abstract
IL-7 is a required factor for T-cell homeostasis. Because of low expression levels and poor reagent availability, the cellular sources of IL-7 have proven challenging to characterize. In this study, we describe a reporter mouse in which enhanced GFP is expressed from the endogenous Il7 locus. We show that IL-7 is produced by lymphatic endothelial cells (LECs) distributed throughout the systemic lymphatic vasculature as well as by fibroblastic reticular cells, and that phosphorylation of STAT5 in lymphocytes is higher in lymphatics than in blood. Furthermore, in nodes depleted of lymphocytes, Il7 transcription is increased in stromal but not in myeloid subsets. These data support recent findings that lymphocyte homeostasis is influenced by access to secondary lymphoid organs and point to LECs as an important in vivo source of IL-7, bathing trafficking immune cells under both resting and lymphopenic conditions.
- Published
- 2013
20. Antidiabetic and antisteatotic effects of the selective fatty acid synthase (FAS) inhibitor platensimycin in mouse models of diabetes
- Author
-
Margaret Wu, Cai Li, Robert W. Myers, Michael R. Tota, Michael E. Lassman, Christine C. Chung, Maryann Powles, Corey N. Miller, Jun Wang, Bindhu V. Karanam, Gino Salituro, Sheo B. Singh, Kenneth P. Ellsworth, Bei B. Zhang, and Sang Ho Lee
- Subjects
Platensimycin ,Adamantane ,Biology ,Carbohydrate metabolism ,chemistry.chemical_compound ,Mice ,Anti-Infective Agents ,medicine ,Diabetes Mellitus ,Animals ,Humans ,Hypoglycemic Agents ,Aminobenzoates ,Anilides ,Beta oxidation ,Fatty acid synthesis ,Multidisciplinary ,Fatty liver ,Fatty Acids ,Biological Sciences ,medicine.disease ,Mice, Mutant Strains ,Cerulenin ,Fatty Liver ,Fatty acid synthase ,Disease Models, Animal ,Sterols ,Glucose ,chemistry ,Biochemistry ,Liver ,Lipogenesis ,biology.protein ,Fatty Acid Synthases ,Oxidation-Reduction - Abstract
Platensimycin (PTM) is a recently discovered broad-spectrum antibiotic produced by Streptomyces platensis . It acts by selectively inhibiting the elongation-condensing enzyme FabF of the fatty acid biosynthesis pathway in bacteria. We report here that PTM is also a potent and highly selective inhibitor of mammalian fatty acid synthase. In contrast to two agents, C75 and cerulenin, that are widely used as inhibitors of mammalian fatty acid synthase, platensimycin specifically inhibits fatty acid synthesis but not sterol synthesis in rat primary hepatocytes. PTM preferentially concentrates in liver when administered orally to mice and potently inhibits hepatic de novo lipogenesis, reduces fatty acid oxidation, and increases glucose oxidation. Chronic administration of platensimycin led to a net reduction in liver triglyceride levels and improved insulin sensitivity in db/+ mice fed a high-fructose diet. PTM also reduced ambient glucose levels in db/db mice. These results provide pharmacological proof of concept of inhibiting fatty acid synthase for the treatment of diabetes and related metabolic disorders in animal models.
- Published
- 2011
21. Glucagon-like peptide 1/glucagon receptor dual agonism reverses obesity in mice
- Author
-
Gary G. Chicchi, Michael E. Lassman, Lan Zhu, Michael M. Sountis, George J. Eiermann, Xiaobing Du, Antonello Pessi, Alessandro Pocai, Nancy A. Thornberry, Xiaoping Zhang, Paul E. Carrington, Ranabir SinhaRoy, Jennifer R. Adams, Elisabetta Bianchi, Michael Wright, Guoqiang Jiang, Corey N. Miller, Gaochao Zhou, Franklin Liu, Laurie Tota, Donald J. Marsh, Aleksandr Petrov, Alessia Santoprete, and Elena Capito
- Subjects
Agonist ,medicine.medical_specialty ,medicine.drug_class ,Endocrinology, Diabetes and Metabolism ,Injections, Subcutaneous ,Molecular Sequence Data ,CHO Cells ,Biology ,Glucagon ,Glucagon-Like Peptide-1 Receptor ,chemistry.chemical_compound ,Mice ,Cricetulus ,Glucagon-Like Peptide 1 ,Internal medicine ,Cricetinae ,Weight Loss ,Internal Medicine ,medicine ,Receptors, Glucagon ,Animals ,Insulin ,Amino Acid Sequence ,Obesity ,Receptor ,Adiponectin ,Reverse Transcriptase Polymerase Chain Reaction ,Leptin ,Body Weight ,Glucagon-like peptide-1 ,Dietary Fats ,Oxyntomodulin ,Mice, Inbred C57BL ,Endocrinology ,chemistry ,Diabetes Mellitus, Type 2 ,Original Article ,Energy Intake ,Glucagon receptor ,Obesity Studies - Abstract
OBJECTIVE Oxyntomodulin (OXM) is a glucagon-like peptide 1 (GLP-1) receptor (GLP1R)/glucagon receptor (GCGR) dual agonist peptide that reduces body weight in obese subjects through increased energy expenditure and decreased energy intake. The metabolic effects of OXM have been attributed primarily to GLP1R agonism. We examined whether a long acting GLP1R/GCGR dual agonist peptide exerts metabolic effects in diet-induced obese mice that are distinct from those obtained with a GLP1R-selective agonist. RESEARCH DESIGN AND METHODS We developed a protease-resistant dual GLP1R/GCGR agonist, DualAG, and a corresponding GLP1R-selective agonist, GLPAG, matched for GLP1R agonist potency and pharmacokinetics. The metabolic effects of these two peptides with respect to weight loss, caloric reduction, glucose control, and lipid lowering, were compared upon chronic dosing in diet-induced obese (DIO) mice. Acute studies in DIO mice revealed metabolic pathways that were modulated independent of weight loss. Studies in Glp1r−/− and Gcgr−/− mice enabled delineation of the contribution of GLP1R versus GCGR activation to the pharmacology of DualAG. RESULTS Peptide DualAG exhibits superior weight loss, lipid-lowering activity, and antihyperglycemic efficacy comparable to GLPAG. Improvements in plasma metabolic parameters including insulin, leptin, and adiponectin were more pronounced upon chronic treatment with DualAG than with GLPAG. Dual receptor agonism also increased fatty acid oxidation and reduced hepatic steatosis in DIO mice. The antiobesity effects of DualAG require activation of both GLP1R and GCGR. CONCLUSIONS Sustained GLP1R/GCGR dual agonism reverses obesity in DIO mice and is a novel therapeutic approach to the treatment of obesity.
- Published
- 2009
22. Adaptation to ER Stress Is Mediated by Differential Stabilities of Pro-Survival and Pro-Apoptotic mRNAs and Proteins
- Author
-
Kazutoshi Mori, Jack Shiansong Li, Randal J. Kaufman, Amir A. Sadighi Akha, Kathryn M Gunnison, David Raden, D. Thomas Rutkowski, Corey N. Miller, Stacey M. Arnold, and Jun-Dong Wu
- Subjects
Protein Denaturation ,Protein Folding ,QH301-705.5 ,Cell Survival ,Eukaryotes ,Adaptation, Biological ,Apoptosis ,Biology ,Endoplasmic Reticulum ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Stress, Physiological ,Gene expression ,Animals ,RNA, Messenger ,Biology (General) ,Cells, Cultured ,Heat-Shock Proteins ,030304 developmental biology ,Cell Proliferation ,0303 health sciences ,General Immunology and Microbiology ,Cell growth ,General Neuroscience ,Endoplasmic reticulum ,Cell Biology ,Models, Theoretical ,Cell biology ,In Vitro ,Phenotype ,Cell culture ,030220 oncology & carcinogenesis ,Unfolded protein response ,Synopsis ,Phosphorylation ,Protein folding ,Signal transduction ,General Agricultural and Biological Sciences ,Apoptosis Regulatory Proteins ,Signal Transduction - Abstract
The accumulation of unfolded proteins in the endoplasmic reticulum (ER) activates a signaling cascade known as the unfolded protein response (UPR). Although activation of the UPR is well described, there is little sense of how the response, which initiates both apoptotic and adaptive pathways, can selectively allow for adaptation. Here we describe the reconstitution of an adaptive ER stress response in a cell culture system. Monitoring the activation and maintenance of representative UPR gene expression pathways that facilitate either adaptation or apoptosis, we demonstrate that mild ER stress activates all UPR sensors. However, survival is favored during mild stress as a consequence of the intrinsic instabilities of mRNAs and proteins that promote apoptosis compared to those that facilitate protein folding and adaptation. As a consequence, the expression of apoptotic proteins is short-lived as cells adapt to stress. We provide evidence that the selective persistence of ER chaperone expression is also applicable to at least one instance of genetic ER stress. This work provides new insight into how a stress response pathway can be structured to allow cells to avert death as they adapt. It underscores the contribution of posttranscriptional and posttranslational mechanisms in influencing this outcome.
- Published
- 2006
23. The unfolded protein response sensor IRE1alpha is required at 2 distinct steps in B cell lymphopoiesis
- Author
-
Kezhong, Zhang, Hetty N, Wong, Benbo, Song, Corey N, Miller, Donalyn, Scheuner, and Randal J, Kaufman
- Subjects
X-Box Binding Protein 1 ,Protein Denaturation ,Protein Folding ,RNA Splicing ,Plasma Cells ,Immunoglobulins ,Receptors, Antigen, B-Cell ,Bone Marrow Cells ,Regulatory Factor X Transcription Factors ,Protein Serine-Threonine Kinases ,Endoplasmic Reticulum ,Mice ,eIF-2 Kinase ,Endoribonucleases ,Animals ,Gene Rearrangement, B-Lymphocyte ,Bone Marrow Transplantation ,Mice, Knockout ,Lymphopoiesis ,Membrane Proteins ,Nuclear Proteins ,Cell Differentiation ,DNA-Binding Proteins ,Commentary ,Signal Transduction ,Transcription Factors - Abstract
B lymphocyte differentiation is coordinated with the induction of high-level Ig secretion and expansion of the secretory pathway. Upon accumulation of unfolded proteins in the lumen of the ER, cells activate an intracellular signaling pathway termed the unfolded protein response (UPR). Two major proximal sensors of the UPR are inositol-requiring enzyme 1alpha (IRE1alpha), an ER transmembrane protein kinase/endoribonuclease, and ER-resident eukaryotic translation initiation factor 2alpha (eIF2alpha) kinase (PERK). To elucidate whether the UPR plays an important role in lymphopoiesis, we carried out reconstitution of recombinase-activating gene 2-deficient (rag2-/-) mice with hematopoietic cells defective in either IRE1alpha- or PERK-mediated signaling. IRE1alpha-deficient (ire1alpha-/-) HSCs can proliferate and give rise to pro-B cells that home to bone marrow. However, IRE1alpha, but not its catalytic activities, is required for Ig gene rearrangement and production of B cell receptors (BCRs). Analysis of rag2-/- mice transplanted with IRE1alpha trans-dominant-negative bone marrow cells demonstrated an additional requirement for IRE1alpha in B lymphopoiesis: both the IRE1alpha kinase and RNase catalytic activities are required to splice the mRNA encoding X-box-binding protein 1 (XBP1) for terminal differentiation of mature B cells into antibody-secreting plasma cells. Furthermore, UPR-mediated translational control through eIF2alpha phosphorylation is not required for B lymphocyte maturation and/or plasma cell differentiation. These results suggest specific requirements of the IRE1alpha-mediated UPR subpathway in the early and late stages of B lymphopoiesis.
- Published
- 2004
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.