228 results on '"Jeremy M Boss"'
Search Results
2. Murine gammaherpesvirus infection is skewed toward Igλ+ B cells expressing a specific heavy chain V-segment.
- Author
-
Christopher M Collins, Christopher D Scharer, Thomas J Murphy, Jeremy M Boss, and Samuel H Speck
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
One of the defining characteristics of the B cell receptor (BCR) is the extensive diversity in the repertoire of immunoglobulin genes that make up the BCR, resulting in broad range of specificity. Gammaherpesviruses are B lymphotropic viruses that establish life-long infection in B cells, and although the B cell receptor plays a central role in B cell biology, very little is known about the immunoglobulin repertoire of gammaherpesvirus infected cells. To begin to characterize the Ig genes expressed by murine gammaherpesvirus 68 (MHV68) infected cells, we utilized single cell sorting to sequence and clone the Ig variable regions of infected germinal center (GC) B cells and plasma cells. We show that MHV68 infection is biased towards cells that express the Igλ light chain along with a single heavy chain variable gene, IGHV10-1*01. This population arises through clonal expansion but is not viral antigen specific. Furthermore, we show that class-switching in MHV68 infected cells differs from that of uninfected cells. Fewer infected GC B cells are class-switched compared to uninfected GC B cells, while more infected plasma cells are class-switched compared to uninfected plasma cells. Additionally, although they are germinal center derived, the majority of class switched plasma cells display no somatic hypermutation regardless of infection status. Taken together, these data indicate that selection of infected B cells with a specific BCR, as well as virus mediated manipulation of class switching and somatic hypermutation, are critical aspects in establishing life-long gammaherpesvirus infection.
- Published
- 2020
- Full Text
- View/download PDF
3. IFNγ induces epigenetic programming of human T-bethi B cells and promotes TLR7/8 and IL-21 induced differentiation
- Author
-
Esther Zumaquero, Sara L Stone, Christopher D Scharer, Scott A Jenks, Anoma Nellore, Betty Mousseau, Antonio Rosal-Vela, Davide Botta, John E Bradley, Wojciech Wojciechowski, Travis Ptacek, Maria I Danila, Jeffrey C Edberg, S Louis Bridges Jr, Robert P Kimberly, W Winn Chatham, Trenton R Schoeb, Alexander F Rosenberg, Jeremy M Boss, Ignacio Sanz, and Frances E Lund
- Subjects
interferons ,B lymphocytes ,antibody secreting cells ,T-bet ,systemic lupus erythematosus ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Although B cells expressing the IFNγR or the IFNγ-inducible transcription factor T-bet promote autoimmunity in Systemic Lupus Erythematosus (SLE)-prone mouse models, the role for IFNγ signaling in human antibody responses is unknown. We show that elevated levels of IFNγ in SLE patients correlate with expansion of the T-bet expressing IgDnegCD27negCD11c+CXCR5neg (DN2) pre-antibody secreting cell (pre-ASC) subset. We demonstrate that naïve B cells form T-bethi pre-ASCs following stimulation with either Th1 cells or with IFNγ, IL-2, anti-Ig and TLR7/8 ligand and that IL-21 dependent ASC formation is significantly enhanced by IFNγ or IFNγ-producing T cells. IFNγ promotes ASC development by synergizing with IL-2 and TLR7/8 ligands to induce genome-wide epigenetic reprogramming of B cells, which results in increased chromatin accessibility surrounding IRF4 and BLIMP1 binding motifs and epigenetic remodeling of IL21R and PRDM1 loci. Finally, we show that IFNγ signals poise B cells to differentiate by increasing their responsiveness to IL-21.
- Published
- 2019
- Full Text
- View/download PDF
4. Balancing selection on a regulatory region exhibiting ancient variation that predates human-neandertal divergence.
- Author
-
Omer Gokcumen, Qihui Zhu, Lubbertus C F Mulder, Rebecca C Iskow, Christian Austermann, Christopher D Scharer, Towfique Raj, Jeremy M Boss, Shamil Sunyaev, Alkes Price, Barbara Stranger, Viviana Simon, and Charles Lee
- Subjects
Genetics ,QH426-470 - Abstract
Ancient population structure shaping contemporary genetic variation has been recently appreciated and has important implications regarding our understanding of the structure of modern human genomes. We identified a ∼36-kb DNA segment in the human genome that displays an ancient substructure. The variation at this locus exists primarily as two highly divergent haplogroups. One of these haplogroups (the NE1 haplogroup) aligns with the Neandertal haplotype and contains a 4.6-kb deletion polymorphism in perfect linkage disequilibrium with 12 single nucleotide polymorphisms (SNPs) across diverse populations. The other haplogroup, which does not contain the 4.6-kb deletion, aligns with the chimpanzee haplotype and is likely ancestral. Africans have higher overall pairwise differences with the Neandertal haplotype than Eurasians do for this NE1 locus (p
- Published
- 2013
- Full Text
- View/download PDF
5. Multiple histone methyl and acetyltransferase complex components bind the HLA-DRA gene.
- Author
-
Nancy M Choi and Jeremy M Boss
- Subjects
Medicine ,Science - Abstract
Major histocompatibility complex class II (MHC-II) genes are fundamental components that contribute to adaptive immune responses. While characterization of the chromatin features at the core promoter region of these genes has been studied, the scope of histone modifications and the modifying factors responsible for activation of these genes are less well defined. Using the MHC-II gene HLA-DRA as a model, the extent and distribution of major histone modifications associated with active expression were defined in interferon-γ induced epithelial cells, B cells, and B-cell mutants for MHC-II expression. With active transcription, nucleosome density around the proximal regulatory region was diminished and histone acetylation and methylation modifications were distributed throughout the gene in distinct patterns that were dependent on the modification examined. Irrespective of the location, the majority of these modifications were dependent on the binding of either the X-box binding factor RFX or the class II transactivator (CIITA) to the proximal regulatory region. Importantly, once established, the modifications were stable through multiple cell divisions after the activating stimulus was removed, suggesting that activation of this system resulted in an epigenetic state. A dual crosslinking chromatin immunoprecipitation method was used to detect histone modifying protein components that interacted across the gene. Components of the MLL methyltransferase and GCN5 acetyltransferase complexes were identified. Some MLL complex components were found to be CIITA independent, including MLL1, ASH2L and RbBP5. Likewise, GCN5 containing acetyltransferase complex components belonging to the ATAC and STAGA complexes were also identified. These results suggest that multiple complexes are either used or are assembled as the gene is activated for expression. Together the results define and illustrate a complex network of histone modifying proteins and multisubunit complexes participating in MHC-II transcription.
- Published
- 2012
- Full Text
- View/download PDF
6. The ‘omics of obesity in B-cell acute lymphoblastic leukemia
- Author
-
Delaney K Geitgey, Miyoung Lee, Kirsten A Cottrill, Maya Jaffe, William Pilcher, Swati Bhasin, Jessica Randall, Anthony J Ross, Michelle Salemi, Marisol Castillo-Castrejon, Matthew B Kilgore, Ayjha C Brown, Jeremy M Boss, Rich Johnston, Anne M Fitzpatrick, Melissa L Kemp, Robert English, Eric Weaver, Pritha Bagchi, Ryan Walsh, Christopher D Scharer, Manoj Bhasin, Joshua D Chandler, Karmella A Haynes, Elizabeth A Wellberg, and Curtis J Henry
- Subjects
Cancer Research ,Oncology - Abstract
The obesity pandemic currently affects more than 70 million Americans and more than 650 million individuals worldwide. In addition to increasing susceptibility to pathogenic infections (eg, SARS-CoV-2), obesity promotes the development of many cancer subtypes and increases mortality rates in most cases. We and others have demonstrated that, in the context of B-cell acute lymphoblastic leukemia (B-ALL), adipocytes promote multidrug chemoresistance. Furthermore, others have demonstrated that B-ALL cells exposed to the adipocyte secretome alter their metabolic states to circumvent chemotherapy-mediated cytotoxicity. To better understand how adipocytes impact the function of human B-ALL cells, we used a multi-omic RNA-sequencing (single-cell and bulk transcriptomic) and mass spectroscopy (metabolomic and proteomic) approaches to define adipocyte-induced changes in normal and malignant B cells. These analyses revealed that the adipocyte secretome directly modulates programs in human B-ALL cells associated with metabolism, protection from oxidative stress, increased survival, B-cell development, and drivers of chemoresistance. Single-cell RNA sequencing analysis of mice on low- and high-fat diets revealed that obesity suppresses an immunologically active B-cell subpopulation and that the loss of this transcriptomic signature in patients with B-ALL is associated with poor survival outcomes. Analyses of sera and plasma samples from healthy donors and those with B-ALL revealed that obesity is associated with higher circulating levels of immunoglobulin-associated proteins, which support observations in obese mice of altered immunological homeostasis. In all, our multi-omics approach increases our understanding of pathways that may promote chemoresistance in human B-ALL and highlight a novel B-cell–specific signature in patients associated with survival outcomes.
- Published
- 2023
- Full Text
- View/download PDF
7. IL-6/STAT3 Signaling Axis Enhances and Prolongs Pdcd1 Expression in Murine CD8 T Cells
- Author
-
Michael D. Powell, Peiyuan Lu, Dennis K. Neeld, Anna K. Kania, Lou-Ella M. M. George-Alexander, Alexander P. R. Bally, Christopher D. Scharer, and Jeremy M. Boss
- Subjects
Mice ,Interleukin-6 ,Programmed Cell Death 1 Receptor ,Immunology ,Receptors, Antigen, T-Cell ,Animals ,Immunology and Allergy ,General Medicine ,CD8-Positive T-Lymphocytes ,Lymphocytic Choriomeningitis ,Apoptosis Regulatory Proteins ,Signal Transduction - Abstract
CD8 cytotoxic T cells are a potent line of defense against invading pathogens. To aid in curtailing aberrant immune responses, the activation status of CD8 T cells is highly regulated. One mechanism in which CD8 T cell responses are dampened is via signaling through the immune-inhibitory receptor Programmed Cell Death Protein-1, encoded by Pdcd1. Pdcd1 expression is regulated through engagement of the TCR, as well as by signaling from extracellular cytokines. Understanding such pathways has influenced the development of numerous clinical treatments. In this study, we showed that signals from the cytokine IL-6 enhanced Pdcd1 expression when paired with TCR stimulation in murine CD8 T cells. Mechanistically, signals from IL-6 were propagated through activation of the transcription factor STAT3, resulting in IL-6–dependent binding of STAT3 to Pdcd1 cis-regulatory elements. Intriguingly, IL-6 stimulation overcame B Lymphocyte Maturation Protein 1–mediated epigenetic repression of Pdcd1, which resulted in a transcriptionally permissive landscape marked by heightened histone acetylation. Furthermore, in vivo–activated CD8 T cells derived from lymphocytic choriomeningitis virus infection required STAT3 for optimal Programmed Cell Death Protein-1 surface expression. Importantly, STAT3 was the only member of the STAT family present at Pdcd1 regulatory elements in lymphocytic choriomeningitis virus Ag-specific CD8 T cells. Collectively, these data define mechanisms by which the IL-6/STAT3 signaling axis can enhance and prolong Pdcd1 expression in murine CD8 T cells.
- Published
- 2022
- Full Text
- View/download PDF
8. Persistent Antigen Harbored by Alveolar Macrophages Enhances the Maintenance of Lung-Resident Memory CD8+ T Cells
- Author
-
Jenna L. Lobby, Ida Uddbäck, Christopher D. Scharer, Tian Mi, Jeremy M. Boss, Allan R. Thomsen, Jan P. Christensen, and Jacob E. Kohlmeier
- Subjects
Homeodomain Proteins ,Immunology ,Nerve Tissue Proteins ,CD8-Positive T-Lymphocytes ,Mice ,Nucleoproteins ,Influenza Vaccines ,Influenza, Human ,Macrophages, Alveolar ,Animals ,Humans ,Immunology and Allergy ,Immunologic Memory ,Lung - Abstract
Lung tissue-resident memory T cells are crucial mediators of cellular immunity against respiratory viruses; however, their gradual decline hinders the development of T cell–based vaccines against respiratory pathogens. Recently, studies using adenovirus (Ad)-based vaccine vectors have shown that the number of protective lung-resident CD8+ TRMs can be maintained long term. In this article, we show that immunization of mice with a replication-deficient Ad serotype 5 expressing influenza (A/Puerto Rico/8/34) nucleoprotein (AdNP) generates a long-lived lung TRM pool that is transcriptionally indistinct from those generated during a primary influenza infection. In addition, we demonstrate that CD4+ T cells contribute to the long-term maintenance of AdNP-induced CD8+ TRMs. Using a lineage tracing approach, we identify alveolar macrophages as a cell source of persistent NP Ag after immunization with AdNP. Importantly, depletion of alveolar macrophages after AdNP immunization resulted in significantly reduced numbers of NP-specific CD8+ TRMs in the lungs and airways. Combined, our results provide further insight to the mechanisms governing the enhanced longevity of Ag-specific CD8+ lung TRMs observed after immunization with recombinant Ad.
- Published
- 2022
- Full Text
- View/download PDF
9. Distinct transcriptomic and epigenomic modalities underpin human memory T cell subsets and their activation potential
- Author
-
James R. Rose, Bagdeser Akdogan-Ozdilek, Andrew R. Rahmberg, Michael D. Powell, Sakeenah L. Hicks, Christopher D. Scharer, and Jeremy M. Boss
- Subjects
Medicine (miscellaneous) ,General Agricultural and Biological Sciences ,General Biochemistry, Genetics and Molecular Biology - Abstract
Human memory T cells (MTC) are poised to rapidly respond to antigen re-exposure. Here, we derived the transcriptional and epigenetic programs of resting and ex vivo activated, circulating CD4+ and CD8+ MTC subsets. A progressive gradient of gene expression from naïve to TCM to TEM is observed, which is accompanied by corresponding changes in chromatin accessibility. Transcriptional changes suggest adaptations of metabolism that are reflected in altered metabolic capacity. Other differences involve regulatory modalities comprised of discrete accessible chromatin patterns, transcription factor binding motif enrichment, and evidence of epigenetic priming. Basic-helix-loop-helix factor motifs for AHR and HIF1A distinguish subsets and predict transcription networks to sense environmental changes. Following stimulation, primed accessible chromatin correlate with an augmentation of MTC gene expression as well as effector transcription factor gene expression. These results identify coordinated epigenetic remodeling, metabolic, and transcriptional changes that enable MTC subsets to ultimately respond to antigen re-encounters more efficiently.
- Published
- 2023
- Full Text
- View/download PDF
10. Data from Yin Yang 1 Regulates the Expression of Snail through a Distal Enhancer
- Author
-
Jeremy M. Boss, Paul A. Wade, Hyesuk Yoon, John C. Cooper, Parimal Majumder, and Matthew B. Palmer
- Abstract
Expression of the Snail gene is required for the epithelial-mesenchymal transitions that accompany mammalian gastrulation, neural crest migration, and organ formation. Pathologic expression of Snail contributes to the migratory capacity of invasive tumors, including melanomas. To investigate the mechanism of Snail up-regulation in human melanoma cells, a conserved enhancer located 3′ of the Snail gene was analyzed. An overlapping Ets and yin yang 1 (YY1) consensus sequence, in addition to a SOX consensus sequence, was required for full enhancer activity. Proteins specifically binding these sequences were detected by electrophoretic mobility shift assay. The Ets/YY1 binding activity was purified by DNA-affinity chromatography and identified as YY1. Although ubiquitously expressed, YY1 was bound at the Snail 3′ enhancer in vivo in Snail-expressing cells but not in cells that did not express Snail. Knockdown of YY1 in A375 cells led to decreased Snail expression. These results identify a role for YY1 in regulating transcription of Snail in melanoma cells through binding to the Snail 3′ enhancer. (Mol Cancer Res 2009;7(2):221–9)
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Figure S1 from Yin Yang 1 Regulates the Expression of Snail through a Distal Enhancer
- Author
-
Jeremy M. Boss, Paul A. Wade, Hyesuk Yoon, John C. Cooper, Parimal Majumder, and Matthew B. Palmer
- Abstract
Supplementary Figure S1 from Yin Yang 1 Regulates the Expression of Snail through a Distal Enhancer
- Published
- 2023
- Full Text
- View/download PDF
12. Aiolos represses CD4+ T cell cytotoxic programming via reciprocal regulation of TFH transcription factors and IL-2 sensitivity
- Author
-
Kaitlin A. Read, Devin M. Jones, Srijana Pokhrel, Emily D. S. Hales, Aditi Varkey, Jasmine A. Tuazon, Caprice D. Eisele, Omar Abdouni, Abbey Saadey, Melissa R. Leonard, Robert T. Warren, Michael D. Powell, Jeremy M. Boss, Emily A. Hemann, Jacob S. Yount, Gang Xin, Hazem E. Ghoneim, Chan-Wang J. Lio, Aharon G. Freud, Patrick L. Collins, and Kenneth J. Oestreich
- Subjects
Multidisciplinary ,General Physics and Astronomy ,General Chemistry ,General Biochemistry, Genetics and Molecular Biology - Abstract
During intracellular infection, T follicular helper (TFH) and T helper 1 (TH1) cells promote humoral and cell-mediated responses, respectively. Another subset, CD4-cytotoxic T lymphocytes (CD4-CTLs), eliminate infected cells via functions typically associated with CD8+ T cells. The mechanisms underlying differentiation of these populations are incompletely understood. Here, we identify the transcription factor Aiolos as a reciprocal regulator of TFH and CD4-CTL programming. We find that Aiolos deficiency results in downregulation of key TFH transcription factors, and consequently reduced TFH differentiation and antibody production, during influenza virus infection. Conversely, CD4-CTL programming is elevated, including enhanced Eomes and cytolytic molecule expression. We further demonstrate that Aiolos deficiency allows for enhanced IL-2 sensitivity and increased STAT5 association with CD4-CTL gene targets, including Eomes, effector molecules, and IL2Ra. Thus, our collective findings identify Aiolos as a pivotal regulator of CD4-CTL and TFH programming and highlight its potential as a target for manipulating CD4+ T cell responses.
- Published
- 2023
- Full Text
- View/download PDF
13. Deletion of histone demethylase Lsd1 (Kdm1a) during retinal development leads to defects in retinal function and structure
- Author
-
Salma Ferdous, Debresha A. Shelton, Tatiana E. Getz, Micah A. Chrenek, Nancy L’Hernault, Jana T. Sellers, Vivian R. Summers, P. Michael Iuvone, Jeremy M. Boss, Jeffrey H. Boatright, and John M. Nickerson
- Subjects
Cellular and Molecular Neuroscience - Abstract
PurposeThe purpose of this study was to investigate the role of Lysine specific demethylase 1 (Lsd1) in murine retinal development. LSD1 is a histone demethylase that can demethylate mono- and di-methyl groups on H3K4 and H3K9. Using Chx10-Cre and Rho-iCre75 driver lines, we generated novel transgenic mouse lines to delete Lsd1 in most retinal progenitor cells or specifically in rod photoreceptors. We hypothesize that Lsd1 deletion will cause global morphological and functional defects due to its importance in neuronal development.MethodsWe tested the retinal function of young adult mice by electroretinogram (ERG) and assessed retinal morphology by in vivo imaging by fundus photography and SD-OCT. Afterward, eyes were enucleated, fixed, and sectioned for subsequent hematoxylin and eosin (H&E) or immunofluorescence staining. Other eyes were plastic fixed and sectioned for electron microscopy.ResultsIn adult Chx10-Cre Lsd1fl/fl mice, we observed a marked reduction in a-, b-, and c-wave amplitudes in scotopic conditions compared to age-matched control mice. Photopic and flicker ERG waveforms were even more sharply reduced. Modest reductions in total retinal thickness and outer nuclear layer (ONL) thickness were observed in SD-OCT and H&E images. Lastly, electron microscopy revealed significantly shorter inner and outer segments and immunofluorescence showed modest reductions in specific cell type populations. We did not observe any obvious functional or morphological defects in the adult Rho-iCre75 Lsd1fl/fl animals.ConclusionLsd1 is necessary for neuronal development in the retina. Adult Chx10-Cre Lsd1fl/fl mice show impaired retinal function and morphology. These effects were fully manifested in young adults (P30), suggesting that Lsd1 affects early retinal development in mice.
- Published
- 2023
- Full Text
- View/download PDF
14. An IRF4–MYC–mTORC1 Integrated Pathway Controls Cell Growth and the Proliferative Capacity of Activated B Cells during B Cell Differentiation In Vivo
- Author
-
Christopher D. Scharer, James R. Rose, Madeline J. Price, Anna K. Kania, Jeremy M. Boss, and Dillon G. Patterson
- Subjects
Adoptive cell transfer ,Cell division ,Chemistry ,Cell growth ,Immunology ,mTORC1 ,Article ,Chromatin ,Cell biology ,medicine.anatomical_structure ,medicine ,Immunology and Allergy ,Reprogramming ,Transcription factor ,B cell - Abstract
Cell division is an essential component of B cell differentiation to Ab-secreting plasma cells, with critical reprogramming occurring during the initial stages of B cell activation. However, a complete understanding of the factors that coordinate early reprogramming events in vivo remain to be determined. In this study, we examined the initial reprogramming by IRF4 in activated B cells using an adoptive transfer system and mice with a B cell–specific deletion of IRF4. IRF4-deficient B cells responding to influenza, 4-hydroxy-3-nitrophenylacetyl–Ficoll, and LPS divided but stalled during the proliferative response. Gene expression profiling of IRF4-deficient B cells at discrete divisions revealed IRF4 was critical for inducing MYC target genes, oxidative phosphorylation, and glycolysis. Moreover, IRF4-deficient B cells maintained an inflammatory gene expression signature. Complementary chromatin accessibility analyses established a hierarchy of IRF4 activity and identified networks of dysregulated transcription factor families in IRF4-deficient B cells, including E-box binding bHLH family members. Indeed, B cells lacking IRF4 failed to fully induce Myc after stimulation and displayed aberrant cell cycle distribution. Furthermore, IRF4-deficient B cells showed reduced mTORC1 activity and failed to initiate the B cell activation unfolded protein response and grow in cell size. Myc overexpression in IRF4-deficient cells was sufficient to overcome the cell growth defect. Together, these data reveal an IRF4–MYC–mTORC1 relationship critical for controlling cell growth and the proliferative response during B cell differentiation.
- Published
- 2021
- Full Text
- View/download PDF
15. Adipocyte-mediated epigenomic instability in human T-ALL cells is cytotoxic and phenocopied by epigenetic-modifying drugs
- Author
-
Miyoung Lee, Delaney K. Geitgey, Jamie A. G. Hamilton, Jeremy M. Boss, Christopher D. Scharer, Jennifer M. Spangle, Karmella A. Haynes, and Curtis J. Henry
- Subjects
Cell Biology ,Developmental Biology - Abstract
The world’s population with obesity is reaching pandemic levels. If current trends continue, it is predicted that there will be 1.5 billion people with obesity by 2030. This projection is alarming due to the association of obesity with numerous diseases including cancer, with recent studies demonstrating a positive association with acute myeloid leukemia (AML) and B cell acute lymphoblastic leukemia (B-ALL). Interestingly, several epidemiological studies suggest the converse relationship may exist in patients with T cell acute lymphoblastic leukemia (T-ALL). To determine the relationship between obesity and T-ALL development, we employed the diet-induced obesity (DIO) murine model and cultured human T-ALL cells in adipocyte-conditioned media (ACM), bone marrow stromal cell-conditioned media, stromal conditioned media (SCM), and unconditioned media to determine the functional impact of increased adiposity on leukemia progression. Whereas only 20% of lean mice transplanted with T-ALL cells survived longer than 3 months post-inoculation, 50%–80% of obese mice with leukemia survived over this same period. Furthermore, culturing human T-ALL cells in ACM resulted in increased histone H3 acetylation (K9/K14/K18/K23/K27) and methylation (K4me3 and K27me3) posttranslational modifications (PTMs), which preceded accelerated cell cycle progression, DNA damage, and cell death. Adipocyte-mediated epigenetic changes in human T-ALL cells were recapitulated with the H3K27 demethylase inhibitor GSK-J4 and the pan-HDAC inhibitor vorinostat. These drugs were also highly cytotoxic to human T-ALL cells at low micromolar concentrations. In summary, our data support epidemiological studies demonstrating that adiposity suppresses T-ALL pathogenesis. We present data demonstrating that T-ALL cell death in adipose-rich microenvironments is induced by epigenetic modifications, which are not tolerated by leukemia cells. Similarly, GSK-J4 and vorinostat treatment induced epigenomic instability and cytotoxicity profiles that phenocopied the responses of human T-ALL cells to ACM, which provides additional support for the use of epigenetic modifying drugs as a treatment option for T-ALL.
- Published
- 2022
- Full Text
- View/download PDF
16. Epigenetic gene regulation in plasma cells
- Author
-
Dillon G. Patterson, Jeremy M. Boss, Christopher D. Scharer, Zhihong Zuo, and Anna K. Kania
- Subjects
0301 basic medicine ,Cell division ,Plasma Cells ,Immunology ,Naive B cell ,Cell ,Biology ,Article ,Epigenesis, Genetic ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Immunology and Allergy ,Epigenetics ,Antibody-Producing Cells ,B cell ,Regulation of gene expression ,B-Lymphocytes ,Cell Differentiation ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Reprogramming ,030215 immunology - Abstract
Humoral immunity provides protection from pathogenic infection and is mediated by antibodies following the differentiation of naive B cells (nBs) to antibody-secreting cells (ASCs). This process requires substantial epigenetic and transcriptional rewiring to ultimately repress the nB program and replace it with one conducive to ASC physiology and function. Notably, these reprogramming events occur within the framework of cell division. Efforts to understand the relationship of cell division with reprogramming and ASC differentiation in vivo have uncovered the timing and scope of reprogramming, as well as key factors that influence these events. Herein, we discuss the unique physiology of ASC and how nBs undergo epigenetic and genome architectural reorganization to acquire the necessary functions to support antibody production. We also discuss the stage-wise manner in which reprogramming occurs across cell divisions and how key molecular determinants can influence B cell fate outcomes.
- Published
- 2021
- Full Text
- View/download PDF
17. Extrafollicular IgD+ B cells generate IgE antibody secreting cells in the nasal mucosa
- Author
-
Sanjeev Gumber, Chris Fucile, Jeremy M. Boss, Christopher D. Scharer, Merin Kuruvilla, John M. DelGaudio, Richard P. Ramonell, Greg Gibson, Kelly R. Magliocca, Alexander F. Rosenberg, Matthew C. Woodruff, Hao Wu, Sarah K. Wise, Christopher M. Tipton, Deepak Tomar, Joshua M. Levy, Alessia Corrado, F. Eun-Hyung Lee, Iñaki Sanz, and Swetha Garimalla
- Subjects
0301 basic medicine ,Adult ,Immunology ,Cell ,Population ,Naive B cell ,B-Lymphocyte Subsets ,chemical and pharmacologic phenomena ,Immunoglobulin E ,Immunoglobulin D ,Article ,Immunophenotyping ,03 medical and health sciences ,0302 clinical medicine ,Nasal Polyps ,medicine ,Hypersensitivity ,Immunology and Allergy ,Humans ,education ,Antibody-Producing Cells ,B cell ,education.field_of_study ,biology ,Chemistry ,Gene Expression Profiling ,breakpoint cluster region ,Germinal center ,Computational Biology ,High-Throughput Nucleotide Sequencing ,hemic and immune systems ,Germinal Center ,Immunoglobulin Class Switching ,Immunoglobulin Isotypes ,Nasal Mucosa ,030104 developmental biology ,medicine.anatomical_structure ,Antibody Formation ,biology.protein ,Pollen ,Seasons ,Somatic Hypermutation, Immunoglobulin ,030215 immunology - Abstract
Increased IgE is a typical feature of allergic rhinitis. Local class-switch recombination has been intimated but B cell precursors and mechanisms remain elusive. Here we describe the dynamics underlying the generation of IgE-antibody secreting cells (ASC) in human nasal polyps (NP), mucosal tissues rich in ASC without germinal centers (GC). Using VH next generation sequencing, we identified an extrafollicular (EF) mucosal IgD+ naive-like intermediate B cell population with high connectivity to the mucosal IgE ASC. Mucosal IgD+ B cells, express germline epsilon transcripts and predominantly co-express IgM. However, a small but significant fraction co-express IgG or IgA instead which also show connectivity to ASC IgE. Phenotypically, NP IgD+ B cells display an activated profile and molecular evidence of BCR engagement. Transcriptionally, mucosal IgD+ B cells reveal an intermediate profile between naive B cells and ASC. Single cell IgE ASC analysis demonstrates lower mutational frequencies relative to IgG, IgA, and IgD ASC consistent with IgE ASC derivation from mucosal IgD+ B cell with low mutational load. In conclusion, we describe a novel mechanism of GC-independent, extrafollicular IgE ASC formation at the nasal mucosa whereby activated IgD+ naive B cells locally undergo direct and indirect (through IgG and IgA), IgE class switch.
- Published
- 2021
18. The Murine MHC Class II Super Enhancer IA/IE-SE Contains a Functionally Redundant CTCF-Binding Component and a Novel Element Critical for Maximal Expression
- Author
-
Christopher D. Scharer, Alexander P. R. Bally, Dillon G. Patterson, Parimal Majumder, Jeremy M. Boss, Joshua T. Lee, and Benjamin G. Barwick
- Subjects
Mice, Knockout ,CCCTC-Binding Factor ,biology ,Genes, MHC Class II ,Immunology ,Promoter ,Insulator (genetics) ,Article ,HLA-DQ alpha-Chains ,Cell biology ,Chromatin ,Mice, Inbred C57BL ,Mice ,Histone ,Super-enhancer ,CTCF ,Gene expression ,biology.protein ,Animals ,Immunology and Allergy ,Gene ,HLA-DRB1 Chains - Abstract
In both humans and mice, CTCF-binding elements form a series of interacting loops across the MHC class II (MHC-II) locus, and CTCF is required for maximal MHC-II gene expression. In humans, a CTCF-bound chromatin insulator termed XL9 and a super enhancer (SE) DR/DQ-SE situated in the intergenic region between HLA-DRB1 and HLA-DQA1 play critical roles in regulating MHC-II expression. In this study, we identify a similar SE, termed IA/IE-SE, located between H2-Eb1 and H2-Aa of the mouse that contains a CTCF site (C15) and a novel region of high histone H3K27 acetylation. A genetic knockout of C15 was created and its role on MHC-II expression tested on immune cells. We found that C15 deletion did not alter MHC-II expression in B cells, macrophages, and macrophages treated with IFN-γ because of functional redundancy of the remaining MHC-II CTCF sites. Surprisingly, embryonic fibroblasts derived from C15-deleted mice failed to induce MHC-II gene expression in response to IFN-γ, suggesting that at least in this developmental lineage, C15 was required. Examination of the three-dimensional interactions with C15 and the H2-Eb1 and H2-Aa promoters identified interactions within the novel region of high histone acetylation within the IA/IE-SE (termed N1) that contains a PU.1 binding site. CRISPR/Cas9 deletion of N1 altered chromatin interactions across the locus and resulted in reduced MHC-II expression. Together, these data demonstrate the functional redundancy of the MHC-II CTCF elements and identify a functionally conserved SE that is critical for maximal expression of MHC-II genes.
- Published
- 2021
- Full Text
- View/download PDF
19. A transcriptionally distinct subset of influenza-specific effector memory B cells predicts long-lived antibody responses to vaccination in humans
- Author
-
Anoma Nellore, Esther Zumaquero, Christopher D. Scharer, Christopher F. Fucile, Christopher M. Tipton, R. Glenn King, Tian Mi, Betty Mousseau, John E. Bradley, Fen Zhou, Stuti Mutneja, Paul A. Goepfert, Jeremy M. Boss, Troy D. Randall, Ignacio Sanz, Alexander F. Rosenberg, and Frances E. Lund
- Subjects
Infectious Diseases ,Immunology ,Immunology and Allergy - Published
- 2023
- Full Text
- View/download PDF
20. The Regulation of Immunity
- Author
-
Jeremy M. Boss
- Subjects
Immunology ,Immunology and Allergy ,Immunity, Innate - Abstract
In their AAI President's Addresses reproduced in this issue, Jeremy M. Boss, Ph.D. (AAI '94; AAI president 2019–2020), and Jenny P.-Y. Ting, Ph.D. (AAI '97; AAI president 2020–2021), welcomed attendees to the AAI annual meeting, Virtual IMMUNOLOGY2021™. Due to the SARS-CoV-2 pandemic and the cancellation of IMMUNOLOGY2020™, Dr. Boss and Dr. Ting each presented their respective president's address to open the meeting.
- Published
- 2022
21. Selective DNA Demethylation Accompanies T Cell Homeostatic Proliferation and Gene Regulation in Lupus-Prone lpr Mice
- Author
-
Karen A. Fortner, Christopher D. Scharer, Julie A. Dragon, Ralph C. Budd, Scott Tighe, and Jeremy M. Boss
- Subjects
Receptors, Antigen, T-Cell, alpha-beta ,T-Lymphocytes ,T cell ,Immunology ,Biology ,Lymphocyte Activation ,medicine.disease_cause ,Article ,Mice ,immune system diseases ,medicine ,Animals ,Lupus Erythematosus, Systemic ,Immunology and Allergy ,fas Receptor ,Epigenetics ,skin and connective tissue diseases ,Cell Proliferation ,Mice, Knockout ,Regulation of gene expression ,Mutation ,Systemic lupus erythematosus ,General Medicine ,medicine.disease ,DNA Demethylation ,Mice, Inbred C57BL ,medicine.anatomical_structure ,DNA demethylation ,Gene Expression Regulation ,DNA methylation ,Cancer research ,CD8 - Abstract
Systemic lupus erythematosus (SLE) is characterized by increased DNA demethylation in T cells, although it is unclear whether this occurs primarily in a subset of SLE T cells. The process driving the DNA demethylation and the consequences on overall gene expression are also poorly understood and whether this represents a secondary consequence of SLE or a primary contributing factor. Lupus-prone lpr mice accumulate large numbers of T cells with age because of a mutation in Fas (CD95). The accumulating T cells include an unusual population of CD4−CD8−TCR-αβ+ (DN) T cells that arise from CD8+ precursors and are also found in human SLE. We have previously observed that T cell accumulation in lpr mice is due to dysregulation of T cell homeostatic proliferation, which parallels an increased expression of numerous genes in the DN subset, including several proinflammatory molecules and checkpoint blockers. We thus determined the DNA methylome in lpr DN T cells compared with their CD8+ precursors. Our findings show that DN T cells manifest discrete sites of extensive demethylation throughout the genome, and these sites correspond to the location of a large proportion of the upregulated genes. Thus, dysregulated homeostatic proliferation in lpr mice and consequent epigenetic alterations may be a contributing factor to lupus pathogenesis.
- Published
- 2020
- Full Text
- View/download PDF
22. Transcriptomic and epigenomic dynamics associated with development of human iPSC-derived GABAergic interneurons
- Author
-
Zhexing Wen, Ying Zhou, George Andrew S Inglis, Christopher D. Scharer, Jeremy M. Boss, Dillon G. Patterson, Andrew Escayg, and Yanfei Han
- Subjects
AcademicSubjects/SCI01140 ,0301 basic medicine ,Induced Pluripotent Stem Cells ,Biology ,Transcriptome ,Epigenome ,03 medical and health sciences ,0302 clinical medicine ,Interneurons ,parasitic diseases ,Gene expression ,Genetics ,Humans ,GABAergic Neurons ,Induced pluripotent stem cell ,Molecular Biology ,Transcription factor ,Gene ,Genetics (clinical) ,Epigenomics ,Computational Biology ,Cell Differentiation ,General Medicine ,Chromatin ,GINS ,030104 developmental biology ,General Article ,Neuroscience ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
GABAergic interneurons (GINs) are a heterogeneous population of inhibitory neurons that collectively contribute to the maintenance of normal neuronal excitability and network activity. Identification of the genetic regulatory elements and transcription factors that contribute toward GIN function may provide new insight into the pathways underlying proper GIN activity while also indicating potential therapeutic targets for GIN-associated disorders, such as schizophrenia and epilepsy. In this study, we examined the temporal changes in gene expression and chromatin accessibility during GIN development by performing transcriptomic and epigenomic analyses on human induced pluripotent stem cell-derived neurons at 22, 50 and 78 days (D) post-differentiation. We observed 13 221 differentially accessible regions (DARs) of chromatin that associate with temporal changes in gene expression at D78 and D50, relative to D22. We also classified families of transcription factors that are increasingly enriched at DARs during differentiation, indicating regulatory networks that likely drive GIN development. Collectively, these data provide a resource for examining the molecular networks regulating GIN functionality.
- Published
- 2020
- Full Text
- View/download PDF
23. Environmental cues regulate epigenetic reprogramming of airway-resident memory CD8+ T cells
- Author
-
Christopher D. Scharer, Sarah L. Hayward, Jeremy M. Boss, Jacob E. Kohlmeier, Emily K. Cartwright, Shiki Takamura, and Zheng-Rong Tiger Li
- Subjects
0301 basic medicine ,Male ,Programmed cell death ,Cellular immunity ,Cell Survival ,Immunology ,Cell ,Apoptosis ,Biology ,CD8-Positive T-Lymphocytes ,Immunological memory ,Article ,Epigenesis, Genetic ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Orthomyxoviridae Infections ,medicine ,Immunology and Allergy ,Integrated stress response ,Cytotoxic T cell ,Animals ,Epigenetics ,Lung ,respiratory system ,Cellular Reprogramming ,Cell biology ,respiratory tract diseases ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Cellular Microenvironment ,Mucosal immunology ,Female ,Reprogramming ,Immunologic Memory ,030215 immunology - Abstract
Tissue-resident memory T cells (TRM cells) are critical for cellular immunity to respiratory pathogens and reside in both the airways and the interstitium. In the present study, we found that the airway environment drove transcriptional and epigenetic changes that specifically regulated the cytolytic functions of airway TRM cells and promoted apoptosis due to amino acid starvation and activation of the integrated stress response. Comparison of airway TRM cells and splenic effector-memory T cells transferred into the airways indicated that the environment was necessary to activate these pathways, but did not induce TRM cell lineage reprogramming. Importantly, activation of the integrated stress response was reversed in airway TRM cells placed in a nutrient-rich environment. Our data defined the genetic programs of distinct lung TRM cell populations and show that local environmental cues altered airway TRM cells to limit cytolytic function and promote cell death, which ultimately leads to fewer TRM cells in the lung., Kohlmeier and colleagues showed that the airway environment drove transcriptional and epigenetic changes that regulated the cytolytic functions of airway TRM cells and promoted their apoptosis due to amino acid starvation and activation of the integrated stress response.
- Published
- 2020
24. Germline ETV6 mutation promotes inflammation and disrupts lymphoid development of early hematopoietic progenitors
- Author
-
Chengjing Zhou, Rizvan Uluisik, Jesse W. Rowley, Camille David, Courtney L. Jones, Christopher D. Scharer, Leila Noetzli, Marlie H. Fisher, Gregory D. Kirkpatrick, Katrina Bark, Jeremy M. Boss, Curtis J. Henry, Eric M. Pietras, Jorge Di Paola, and Christopher C. Porter
- Subjects
Inflammation ,Cancer Research ,Proto-Oncogene Proteins c-ets ,Cell Biology ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Thrombopoiesis ,Repressor Proteins ,Mice ,Germ Cells ,Mutation ,Genetics ,Animals ,Humans ,Molecular Biology ,Germ-Line Mutation - Abstract
Germline mutations in ETV6 are associated with a syndrome of thrombocytopenia and leukemia predisposition, and ETV6 is among the most commonly mutated genes in leukemias, especially childhood B-cell acute lymphoblastic leukemia. However, the mechanisms underlying disease caused by ETV6 dysfunction are poorly understood. To address these gaps in knowledge, using CRISPR/Cas9, we developed a mouse model of the most common recurrent, disease-causing germline mutation in ETV6. We found defects in hematopoiesis related primarily to abnormalities of the multipotent progenitor population 4 (MPP4) subset of hematopoietic progenitor cells and evidence of sterile inflammation. Expression of ETV6 in Ba/F3 cells altered the expression of several cytokines, some of which were also detected at higher levels in the bone marrow of the mice with Etv6 mutation. Among these, interleukin-18 and interleukin-13 abrogated B-cell development of sorted MPP4 cells, but not common lymphoid progenitors, suggesting that inflammation contributes to abnormal hematopoiesis by impairing lymphoid development. These data, along with those from humans, support a model in which ETV6 dysfunction promotes inflammation, which adversely affects thrombopoiesis and promotes leukemogenesis.
- Published
- 2022
25. Generation of human long-lived plasma cells by developmentally regulated epigenetic imprinting
- Author
-
Chester J Joyner, Ariel M Ley, Doan C Nguyen, Mohammad Ali, Alessia Corrado, Christopher Tipton, Christopher D Scharer, Tian Mi, Matthew C Woodruff, Jennifer Hom, Jeremy M Boss, Meixue Duan, Greg Gibson, Danielle Roberts, Joel Andrews, Sagar Lonial, Inaki Sanz, and F Eun-Hyung Lee
- Subjects
Adult ,Male ,Resource ,Time Factors ,Adolescent ,Cell Survival ,Health, Toxicology and Mutagenesis ,Plasma Cells ,Apoptosis ,Plant Science ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Epigenesis, Genetic ,Immunophenotyping ,Genetic Heterogeneity ,Genomic Imprinting ,Young Adult ,Humans ,Ecology ,Histocytochemistry ,Middle Aged ,Resources ,Gene Expression Regulation ,Antibody Formation ,Female ,Biomarkers - Abstract
This study shows that the generation of human long-lived plasma cells requires blood antibody secreting cells to undergo epigenetic and transcriptional reprogramming in response to the bone marrow microniche to become apoptosis resistant and survive to secrete antibodies for a lifetime., Antibody secreting cells (ASCs) circulate after vaccination and infection and migrate to the BM where a subset known as long-lived plasma cells (LLPCs) persists and secrete antibodies for a lifetime. The mechanisms by which circulating ASCs become LLPCs are not well elucidated. Here, we show that human blood ASCs have distinct morphology, transcriptomes, and epigenetics compared with BM LLPCs. Compared with blood ASCs, BM LLPCs have decreased nucleus/cytoplasm ratio but increased endoplasmic reticulum and numbers of mitochondria. LLPCs up-regulate pro-survival genes MCL1, BCL2, and BCL-XL while simultaneously down-regulating pro-apoptotic genes HRK1, CASP3, and CASP8. Consistent with reduced gene expression, the pro-apoptotic gene loci are less accessible in LLPCs. Of the pro-survival genes, only BCL2 is concordant in gene up-regulation and loci accessibility. Using a novel in vitro human BM mimetic, we show that blood ASCs undergo similar morphological and molecular changes that resemble ex vivo BM LLPCs. Overall, our study demonstrates that early-minted blood ASCs in the BM microniche must undergo morphological, transcriptional, and epigenetic changes to mature into apoptotic-resistant LLPCs.
- Published
- 2021
26. H3K27me3 Demethylase UTX Restrains Plasma Cell Formation
- Author
-
Anna K. Kania, Madeline J. Price, Lou-Ella George-Alexander, Dillon G. Patterson, Sakeenah L. Hicks, Christopher D. Scharer, and Jeremy M. Boss
- Subjects
Histone Demethylases ,Histones ,Jumonji Domain-Containing Histone Demethylases ,Mice ,Immunology ,Plasma Cells ,Immunology and Allergy ,Animals ,Methylation ,Chromatin ,Article - Abstract
B cell differentiation is associated with substantial transcriptional, metabolic, and epigenetic remodeling, including redistribution of histone 3 lysine 27 trimethylation (H3K27me3), which is associated with a repressive chromatin state and gene silencing. Although the role of the methyltransferase EZH2 (Enhancer of zeste homolog 2) in B cell fate decisions has been well established, it is not known whether H3K27me3 demethylation is equally important. In this study, we showed that simultaneous genetic deletion of the two H3K27 demethylases UTX and JMJD3 (double-knockout [Utxfl/flJmjd3fl/flCd19cre/+] [dKO]) led to a significant increase in plasma cell (PC) formation after stimulation with the T cell–independent Ags LPS and NP-Ficoll. This phenotype occurred in a UTX-dependent manner as UTX single-knockout mice, but not JMJD3 single-knockout mice, mimicked the dKO. Although UTX- and JMJD3-deficient marginal zone B cells showed increased proliferation, dKO follicular B cells also showed increased PC formation. PCs from dKO mice upregulated genes associated with oxidative phosphorylation and exhibited increased spare respiratory capacity. Mechanistically, deletion of Utx and Jmjd3 resulted in higher levels of H3K27me3 at proapoptotic genes and resulted in reduced apoptosis of dKO PCs in vivo. Furthermore, UTX regulated chromatin accessibility at regions containing ETS and IFN regulatory factor (IRF) transcription factor family motifs, including motifs of known repressors of PC fate. Taken together, these data demonstrate that the H3K27me3 demethylases restrain B cell differentiation.
- Published
- 2021
27. Phosphoinositide 3-Kinase Signaling Can Modulate MHC Class I and II Expression
- Author
-
Jeremy M. Boss, Edmund K. Waller, Sanjay Chandrasekaran, David A. Gutman, Christopher D. Scharer, Dillon G. Patterson, Kelly R. Magliocca, Sergei V. Kotenko, Lee Ad Cooper, Gregory B. Lesinski, Brian Pollack, Carlos S. Moreno, John T. Seykora, Susan N. Thomas, Robert A. Swerlick, Maiko Sasaki, Haydn T. Kissick, and Bishu Sapkota
- Subjects
0301 basic medicine ,Cancer Research ,Genes, MHC Class II ,Antigen presentation ,Genes, MHC Class I ,chemical and pharmacologic phenomena ,Major histocompatibility complex ,Article ,Interferon-gamma ,03 medical and health sciences ,0302 clinical medicine ,MHC class I ,Humans ,STAT1 ,Protein Kinase Inhibitors ,Molecular Biology ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Regulation of gene expression ,Antigen Presentation ,Binding Sites ,Phosphoinositide 3-kinase ,biology ,Squamous Cell Carcinoma of Head and Neck ,Antigen processing ,PTEN Phosphohydrolase ,Genomics ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,STAT1 Transcription Factor ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Phosphatidylinositol 3-Kinase ,Proto-Oncogene Proteins c-akt ,Protein Binding ,Signal Transduction - Abstract
Molecular events activating the PI3K pathway are frequently detected in human tumors and the activation of PI3K signaling alters numerous cellular processes including tumor cell proliferation, survival, and motility. More recent studies have highlighted the impact of PI3K signaling on the cellular response to interferons and other immunologic processes relevant to antitumor immunity. Given the ability of IFNγ to regulate antigen processing and presentation and the pivotal role of MHC class I (MHCI) and II (MHCII) expression in T-cell–mediated antitumor immunity, we sought to determine the impact of PI3K signaling on MHCI and MHCII induction by IFNγ. We found that the induction of cell surface MHCI and MHCII molecules by IFNγ is enhanced by the clinical grade PI3K inhibitors dactolisib and pictilisib. We also found that PI3K inhibition increases STAT1 protein levels following IFNγ treatment and increases accessibility at genomic STAT1-binding motifs. Conversely, we found that pharmacologic activation of PI3K signaling can repress the induction of MHCI and MHCII molecules by IFNγ, and likewise, the loss of PTEN attenuates the induction of MHCI, MHCII, and STAT1 by IFNγ. Consistent with these in vitro studies, we found that within human head and neck squamous cell carcinomas, intratumoral regions with high phospho-AKT IHC staining had reduced MHCI IHC staining. Implications: Collectively, these findings demonstrate that MHC expression can be modulated by PI3K signaling and suggest that activation of PI3K signaling may promote immune escape via effects on antigen presentation.
- Published
- 2019
- Full Text
- View/download PDF
28. Epigenetic programming underpins B cell dysfunction in human SLE
- Author
-
Chungwen Wei, Benjamin G. Barwick, Jeremy M. Boss, Christopher D. Scharer, Bridget E. Neary, Tsuneo Deguchi, Iñaki Sanz, Tian Mi, Arezou Khosroshahi, F. Eun-Hyung Lee, Scott A. Jenks, Dillon G. Patterson, Emily L. Blalock, Sakeenah L. Hicks, and Kevin S. Cashman
- Subjects
0301 basic medicine ,Immunology ,B-Lymphocyte Subsets ,Biology ,Article ,Epigenesis, Genetic ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,immune system diseases ,medicine ,Immunology and Allergy ,Humans ,Lupus Erythematosus, Systemic ,Epigenetics ,skin and connective tissue diseases ,B cell ,Epigenesis ,Lupus erythematosus ,Effector ,Epigenome ,DNA Methylation ,medicine.disease ,Chromatin Assembly and Disassembly ,Chromatin ,Cell biology ,Transcription Factor AP-1 ,030104 developmental biology ,medicine.anatomical_structure ,Early Growth Response Transcription Factors ,030215 immunology - Abstract
Systemic lupus erythematosus (SLE) is characterized by the expansion of extrafollicular pathogenic B cells derived from newly activated naive cells. Although these cells express distinct markers, their epigenetic architecture and how it contributes to SLE remain poorly understood. To address this, we determined the DNA methylomes, chromatin accessibility profiles and transcriptomes from five human B cell subsets, including a newly defined effector B cell subset, from subjects with SLE and healthy controls. Our data define a differentiation hierarchy for the subsets and elucidate the epigenetic and transcriptional differences between effector and memory B cells. Importantly, an SLE molecular signature was already established in resting naive cells and was dominated by enrichment of accessible chromatin in motifs for AP-1 and EGR transcription factors. Together, these factors acted in synergy with T-BET to shape the epigenome of expanded SLE effector B cell subsets. Thus, our data define the molecular foundation of pathogenic B cell dysfunction in SLE.
- Published
- 2019
29. An IRF4-MYC-mTORC1 integrated pathway controls cell growth and the proliferative capacity of activated B cells during B cell differentiationin vivo
- Author
-
Jeremy M. Boss, Anna K. Kania, Madeline J. Price, Christopher D. Scharer, James R. Rose, and Dillon G. Patterson
- Subjects
Adoptive cell transfer ,medicine.anatomical_structure ,Cell division ,Cell growth ,medicine ,mTORC1 ,Biology ,Reprogramming ,Transcription factor ,B cell ,Chromatin ,Cell biology - Abstract
Cell division is an essential component of B cell differentiation to antibody-secreting plasma cells, with critical reprogramming occurring during the initial stages of B cell activation. However, a complete understanding of the factors that coordinate early reprogramming events in vivo remain to be determined. In this study, we examined the initial reprogramming by IRF4 in activated B cells using an adoptive transfer system and mice with a B cell-specific deletion of IRF4. IRF4-deficient B cells responding to influenza, NP-Ficoll and LPS divided, but stalled during the proliferative response. Gene expression profiling of IRF4-deficient B cells at discrete divisions revealed IRF4 was critical for inducing MYC target genes, oxidative phosphorylation, and glycolysis. Moreover, IRF4-deficient B cells maintained an inflammatory gene expression signature. Complementary chromatin accessibility analyses established a hierarchy of IRF4 activity and identified networks of dysregulated transcription factor families in IRF4-deficient B cells, including E-box binding bHLH family members. Indeed, B cells lacking IRF4 failed to fully induceMycafter stimulation and displayed aberrant cell cycle distribution. Furthermore, IRF4-deficient B cells showed reduced mTORC1 activity and failed to initiate the B cell-activation unfolded protein response and grow in cell size.Mycoverexpression in IRF4-deficient was sufficient to overcome the cell growth defect. Together, these data reveal an IRF4-MYC-mTORC1 relationship critical for controlling cell growth and the proliferative response during B cell differentiation.
- Published
- 2021
- Full Text
- View/download PDF
30. Maturation of Human Long-lived Plasma Cells Results in Resistance to Apoptosis by Transcriptional and Epigenetic Regulation
- Author
-
Jeremy M. Boss, Christopher D. Scharer, Mohammed K. Ali, Tian Mi, Sagar Lonial, Danielle Roberts, Chester J Joyner, Meixue Duan, Matthew C. Woodruff, Greg Gibson, Ignacio Sanz, Christopher M. Tipton, Frances Eun-Hyung Lee, Joel Andrews, Ariel Ley, Alessia Corrado, Jennifer Hom, and Doan Nguyen
- Subjects
Transcriptome ,medicine.anatomical_structure ,Downregulation and upregulation ,biology ,Apoptosis ,Gene expression ,biology.protein ,medicine ,Epigenetics ,Bone marrow ,Antibody ,Gene ,Cell biology - Abstract
Antibody secreting cells (ASC) circulate after vaccination and migrate to the bone marrow (BM) where a subset known as long-lived plasma cells (LLPC) persist and secrete antibodies for a lifetime. The mechanisms of how circulating ASC become LLPC are not well elucidated. Here, we show that human blood ASCs have distinct morphology, transcriptomes, and epigenetics compared to BM LLPC. LLPC acquire transcriptional and epigenetic changes in the apoptosis pathway to support their survival. Upregulation of pro-survival gene expression accompanies downregulation of pro-apoptotic gene expression in LLPC. While pro-apoptotic gene loci are less accessible, pro-survival gene loci are not always accompanied by accessibility changes. Importantly, we show similar LLPC morphological and transcriptional maturation of blood ASC in response to the novel in vitro BM mimetic. In all, our study demonstrates that blood ASC in the BM microniche must undergo morphological and molecular changes to mature into apoptotic-resistant LLPC.
- Published
- 2021
- Full Text
- View/download PDF
31. Somatic Diversification of Rearranged Antibody Gene Segments by Intra- and Interchromosomal Templated Mutagenesis
- Author
-
Gordon A. Dale, Daniel J. Wilkins, Jordan Rowley, Christopher D. Scharer, Christopher M. Tipton, Jennifer Hom, Jeremy M. Boss, Victor Corces, Ignacio Sanz, and Joshy Jacob
- Subjects
Genes, Immunoglobulin ,Mutagenesis ,Immunology ,Mutation ,Gene Conversion ,Immunogenetics ,Immunology and Allergy ,Humans ,Germinal Center - Abstract
The ability of the humoral immune system to generate Abs capable of specifically binding a myriad of Ags is critically dependent on the somatic hypermutation program. This program induces both templated mutations (i.e., gene conversion) and untemplated mutations. In humans, somatic hypermutation is widely believed to result in untemplated point mutations. In this study, we demonstrate detection of large-scale templated events that occur in human memory B cells and circulating plasmablasts. We find that such mutations are templated intrachromosomally from IGHV genes and interchromosomally from IGHV pseudogenes as well as other homologous regions unrelated to IGHV genes. These same donor regions are used in multiple individuals, and they predominantly originate from chromosomes 14, 15, and 16. In addition, we find that exogenous sequences placed at the IgH locus, such as LAIR1, undergo templated mutagenesis and that homology appears to be the major determinant for donor choice. Furthermore, we find that donor tracts originate from areas in proximity with open chromatin, which are transcriptionally active, and are found in spatial proximity with the IgH locus during the germinal center reaction. These donor sequences are inserted into the Ig gene segment in association with overlapping activation-induced cytidine deaminase hotspots. Taken together, these studies suggest that diversity generated during the germinal center response is driven by untemplated point mutations as well as templated mutagenesis using local and distant regions of the genome.
- Published
- 2021
32. Conserved epigenetic programming and enhanced heme metabolism drive memory B cell reactivation
- Author
-
Christopher D. Scharer, Jeremy M. Boss, Anna K. Kania, Troy D. Randall, and Madeline J. Price
- Subjects
Immunology ,Chemotaxis ,Cell cycle ,Plasma cell ,bacterial infections and mycoses ,Chromatin ,Cell biology ,Transcriptome ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine.anatomical_structure ,chemistry ,polycyclic compounds ,medicine ,bacteria ,Immunology and Allergy ,Epigenetics ,Signal transduction ,Memory B cell ,Heme ,Transcription factor ,030215 immunology - Abstract
Memory B cells (MBCs) have enhanced capabilities to differentiate to plasma cells and generate a rapid burst of antibodies upon secondary stimulation. To determine if MBCs harbor an epigenetic landscape that contributes to increased differentiation potential, we derived the chromatin accessibility and transcriptomes of influenza-specific IgM and IgG MBCs compared to naïve cells. MBCs possessed an accessible chromatin architecture surrounding plasma cell specific genes, as well as altered expression of transcription factors and genes encoding cell cycle, chemotaxis, and signal transduction processes. Intriguingly, this MBC signature was conserved between humans and mice. MBCs of both species possessed a heightened heme signature compared to naïve cells. Differentiation in the presence of hemin enhanced oxidative phosphorylation metabolism and MBC differentiation into antibody secreting plasma cells. Thus, these data define conserved MBC transcriptional and epigenetic signatures that include a central role for heme and multiple other pathways in augmenting MBC reactivation potential.Key PointsInfluenza-specific memory B cells have accessible chromatin structure.Human and mouse memory B cells upregulate heme metabolic pathways.Heme enhances PC differentiation and augments mitochondrial metabolism in ex vivo.
- Published
- 2021
- Full Text
- View/download PDF
33. Epigenome accessibility changes before and after activation reveal distinct and progressive differentiation for human memory T cell subsets
- Author
-
James R Rose, Andrew R Rahmberg, Michael D Powell, Christopher D Scharer, and Jeremy M Boss
- Subjects
Immunology ,Immunology and Allergy - Abstract
Memory T cells (MTC), an indispensable part of adaptive immune memory, have historically been categorized by the cell surface proteins CCR7 and CD45RA into specialized subtypes (TCM, TEM, and TEMRA), each with unique functions. However, the epigenetic characteristics that distinguish the MTC subsets as well as the gene regulatory networks governing each MTC subsets’ response to activation remain poorly understood. Here we define and categorize the transcriptional and epigenetic differences of MTC and their respective primary subsets found in human blood, both in a resting state and after ex-vivo stimulation. Resting TCM were found to be relatively more similar to naïve cells in both CD4 and CD8 lineages, while TEM exhibited greater numbers of differentially expressed genes (DEG) and alterations to chromatin accessibility. Differentially accessible regions (DAR) discerning memory subsets contained binding motifs for factors thought to regulate memory formation from the bZIP, T-box and HMG families, as well as sites for novel bHLH factors MSC and AHR that may function in MTC development. Examining the effect of stimulation on MTC gene expression and chromatin identified unique DAR and DEG modules, some of which may have initially been primed by previous activation events in naive progenitors. Primed DAR were correlated with augmented expression of important genes in MTC after stimulation, suggesting an epigenetic mechanism of regulation. Ultimately these results describe the accumulation of epigenetic alterations during primary activation and during memory development that distinguish MTC from naïve T cells, enable differentiation into distinct subsets, and influence how MTC respond to secondary activation. Supported by grants from NIH (RO1 AI113021,T32 GM0008490)
- Published
- 2022
- Full Text
- View/download PDF
34. Memory B cells are a heterogenous population regulated by epigenetic programming
- Author
-
Keenan J Wiggins, Christopher Scharer, and Jeremy M Boss
- Subjects
Immunology ,Immunology and Allergy - Abstract
Memory B cells (MBC) are a heterogenous population that consist of immunoglobin class switched and non-class switched MBC, and these populations can arise via germinal center dependent or independent mechanisms. The timing of MBC development influences the fate outcome of MBC populations. These different populations of MBC are regulated by cell signaling, but it is not clearly defined what epigenetic factors influence MBC differentiation. EZH2 is an important histone methyltransferase that catalyzes H3K27me3 resulting in gene repression. EZH2 has been shown to regulate B cell differentiation into germinal centers and plasma cells; however, it is unknown if EZH2 regulates MBC development. To address this, a knockout model has been established where EZH2 is conditionally deleted using the CD19 and AICDA driven CRE expression. Here we used the influenza PR8 model to ascertain the kinetics of MBC differentiation and formation in the spleen, dLN, and lungs following a live infection in wild-type and EZH2-KO cohorts. Using B cell tetramers, antigen-specific MBC will be analyzed to define the kinetics of MBC development and determine the similarities and differences between the different populations of MBC. Overall, these data define the early kinetics of MBC establishment and the developmental timing that EZH2-dependent H3K27me3 remodeling is required for MBC formation. Supported by NIH/NIAID to CDS (R01 AI148471)
- Published
- 2022
- Full Text
- View/download PDF
35. Defining the cis- and trans-regulatory architecture for human PD-1 in T Follicular Helper Cells
- Author
-
Michael Duane Powell, James R Rose, Dennis Neeld, Andrew R Rahmberg, Christopher D Scharer, and Jeremy M Boss
- Subjects
Immunology ,Immunology and Allergy - Abstract
The surface immune checkpoint inhibitor Programed Death-1 (PD-1), encoded by PDCD1, is expressed throughout the immune system. Notably, PD-1 expression is amongst its highest in a specialized subset of CD4+ T follicular helper (TFH) cells. TFH cells primarily function by engaging with cognate B cells, facilitating the generation of high affinity vaccine- and pathogen-specific antibodies. Despite the proven clinical benefit of therapies targeting the PD-1 pathway, very little is known regarding the mechanisms by which human PDCD1 is transcriptionally regulated in humans. Here, we seek to elucidate both the cis- and trans-regulatory mechanisms that govern PDCD1 expression in TFH cells. Human TFH cells were sourced from discarded tonsil tissue, circulating blood, or through an ex vivo differentiation model to generate ‘TFH-like’ cells. RNA-seq analysis revealed that ex vivo-derived ‘TFH-like’ cells resembled bona-fide in vivo TFH cells, including induction of PDCD1. Furthermore, ATAC-sequencing experiments identified 8 conserved regions in the PDCD1 locus that exhibited altered accessibility in TFH cells. These cis-regulatory elements were determined to have distinct functions in controlling PDCD1 expression. Additionally, bioinformatic analysis revealed a core TFH gene program, implicating the transcription factors BCL-6, ASCL2, TOX, TOX2, and AIOLOS as regulators of PDCD1 and TFH fate. Taken together, these findings uncover the molecular mechanisms that control PDCD1 expression, informing the design of future therapies aimed at manipulating PD-1. Supported by National Institute of Health grants: RO1 AI113021 to J.M.B, F32 AI161857 to M.D.P
- Published
- 2022
- Full Text
- View/download PDF
36. Antibody-secreting cell destiny emerges during the initial stages of B-cell activation
- Author
-
Madeline J. Price, Christopher D. Scharer, Tian Mi, Jeremy M. Boss, Sakeenah L. Hicks, and Dillon G. Patterson
- Subjects
0301 basic medicine ,Cell division ,animal diseases ,Cell ,General Physics and Astronomy ,02 engineering and technology ,Lymphocyte Activation ,Transcriptome ,Mice ,L-Selectin ,lcsh:Science ,Mice, Knockout ,Plasma cells ,B-Lymphocytes ,Multidisciplinary ,biology ,hemic and immune systems ,Cell Differentiation ,Epigenetics in immune cells ,021001 nanoscience & nanotechnology ,Cell biology ,medicine.anatomical_structure ,Interferon Regulatory Factors ,L-selectin ,0210 nano-technology ,Science ,Antigens, CD19 ,chemical and pharmacologic phenomena ,Cell fate determination ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Antigen ,medicine ,Animals ,Transcriptomics ,Antibody-Producing Cells ,B cell ,Immunity ,General Chemistry ,eye diseases ,Mice, Inbred C57BL ,Humoral immunity ,030104 developmental biology ,biology.protein ,lcsh:Q ,Positive Regulatory Domain I-Binding Factor 1 ,IRF4 - Abstract
Upon stimulation, B cells assume heterogeneous cell fates, with only a fraction differentiating into antibody-secreting cells (ASC). Here we investigate B cell fate programming and heterogeneity during ASC differentiation using T cell-independent models. We find that maximal ASC induction requires at least eight cell divisions in vivo, with BLIMP-1 being required for differentiation at division eight. Single cell RNA-sequencing of activated B cells and construction of differentiation trajectories reveal an early cell fate bifurcation. The ASC-destined branch requires induction of IRF4, MYC-target genes, and oxidative phosphorylation, with the loss of CD62L expression serving as a potential early marker of ASC fate commitment. Meanwhile, the non-ASC branch expresses an inflammatory signature, and maintains B cell fate programming. Finally, ASC can be further subseted based on their differential responses to ER-stress, indicating multiple development branch points. Our data thus define the cell division kinetics of B cell differentiation in vivo, and identify the molecular trajectories of B cell fate and ASC formation., The development of activated B cells into antibody-secreting cells (ASC) is a critical step for humoral immunity. Here the authors show, using adoptive transfers and single cell RNA sequencing, that commitment to ASC occurs soon following B cell activation, and is coordinated by specific transcriptome programs and proliferation kinetics.
- Published
- 2020
37. Signaling through the inhibitory Fc receptor Fc gamma RIIB Induces CD8+ T Cell apoptosis to limit T Cell immunity
- Author
-
Paolo Cravedi, Clara R. Farley, Mark S. Cragg, Anna B. Morris, David F. Pinelli, Christopher D. Scharer, Layne E. Adams, Miguel Fribourg, Jeremy M. Boss, Mandy L. Ford, and Peter S. Heeger
- Subjects
0301 basic medicine ,biology ,medicine.medical_treatment ,T cell ,Immunology ,Fc receptor ,FCGR2B ,Acquired immune system ,FGL2 ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Infectious Diseases ,Cytokine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,medicine ,biology.protein ,Immunology and Allergy ,Cytotoxic T cell ,CD8 - Abstract
Effector CD8+ T cells are important mediators of adaptive immunity, and receptor-ligand interactions that regulate their survival may have therapeutic potential. Here, we identified a subset of effector CD8+ T cells that expressed the inhibitory fragment crystallizable (Fc) receptor FcγRIIB following activation and multiple rounds of division. CD8+ T cell-intrinsic genetic deletion of Fcgr2b increased CD8+ effector T cell accumulation, resulting in accelerated graft rejection and decreased tumor volume in mouse models. Immunoglobulin G (IgG) antibody was not required for FcγRIIB-mediated control of CD8+ T cell immunity, and instead, the immunosuppressive cytokine fibrinogen-like 2 (Fgl2) was a functional ligand for FcγRIIB on CD8+ T cells. Fgl2 induced caspase-3/7-mediated apoptosis in Fcgr2b+, but not Fcgr2b-/-, CD8+ T cells. Increased expression of FcγRIIB correlated with freedom from rejection following withdrawal from immunosuppression in a clinical trial of kidney transplant recipients. Together, these findings demonstrate a cell-intrinsic coinhibitory function of FcγRIIB in regulating CD8+ T cell immunity.
- Published
- 2020
38. Distinct Effector B Cells Induced by Unregulated Toll-like Receptor 7 Contribute to Pathogenic Responses in Systemic Lupus Erythematosus
- Author
-
Esther Zumaquero, Matthew C. Woodruff, Christopher M. Tipton, Regina Bugrovsky, Xiaoqian Wang, Deepak Tomar, S. Sam Lim, F. Eun-Hyung Lee, Emily L. Blalock, Scott A. Jenks, Michelle Petri, Chungwen Wei, Frances E. Lund, Aakash V. Patel, Timothy B. Niewold, Greg Gibson, Christopher D. Scharer, Urko M. Marigorta, Jeremy M. Boss, Kevin S. Cashman, Jennifer H. Anolik, Ignacio Sanz, and Zoe Simon
- Subjects
0301 basic medicine ,Adult ,Male ,Immunology ,Naive B cell ,Plasma Cells ,B-Lymphocyte Subsets ,CD11c ,medicine.disease_cause ,Immunoglobulin D ,CXCR5 ,Article ,Autoimmunity ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Immunity ,medicine ,Immunology and Allergy ,Humans ,Lupus Erythematosus, Systemic ,Gene Regulatory Networks ,Aged ,Aged, 80 and over ,Toll-like receptor ,B-Lymphocytes ,biology ,Effector ,Autoantibody ,TLR7 ,Middle Aged ,Cell biology ,030104 developmental biology ,Infectious Diseases ,Toll-Like Receptor 7 ,biology.protein ,Female ,Transcriptome ,030215 immunology - Abstract
Summary Systemic Lupus Erythematosus (SLE) is characterized by B cells lacking IgD and CD27 (double negative; DN). We show that DN cell expansions reflected a subset of CXCR5- CD11c+ cells (DN2) representing pre-plasma cells (PC). DN2 cells predominated in African-American patients with active disease and nephritis, anti-Smith and anti-RNA autoantibodies. They expressed a T-bet transcriptional network; increased Toll-like receptor-7 (TLR7); lacked the negative TLR regulator TRAF5; and were hyper-responsive to TLR7. DN2 cells shared with activated naive cells (aNAV), phenotypic and functional features, and similar transcriptomes. Their PC differentiation and autoantibody production was driven by TLR7 in an interleukin-21 (IL-21)-mediated fashion. An in vivo developmental link between aNAV, DN2 cells, and PC was demonstrated by clonal sharing. This study defines a distinct differentiation fate of autoreactive naive B cells into PC precursors with hyper-responsiveness to innate stimuli, as well as establishes prominence of extra-follicular B cell activation in SLE, and identifies therapeutic targets.
- Published
- 2020
39. Plasma cell differentiation is controlled by multiple cell division-coupled epigenetic programs
- Author
-
Muyao Guo, Alexander P. R. Bally, Christopher D. Scharer, Jeremy M. Boss, and Benjamin G. Barwick
- Subjects
0301 basic medicine ,Cell division ,Science ,Naive B cell ,Plasma Cells ,Primary Cell Culture ,General Physics and Astronomy ,Mice, Transgenic ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Epigenesis, Genetic ,Histones ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Plasma cell differentiation ,medicine ,Animals ,Enhancer of Zeste Homolog 2 Protein ,Epigenetics ,Promoter Regions, Genetic ,lcsh:Science ,Transcription factor ,B cell ,Multidisciplinary ,Promoter ,Cell Differentiation ,General Chemistry ,DNA Methylation ,Cell biology ,Chromatin ,Immunity, Humoral ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,lcsh:Q ,Positive Regulatory Domain I-Binding Factor 1 ,Cell Division ,030215 immunology - Abstract
The genomic loci associated with B cell differentiation that are subject to transcriptional and epigenetic regulation in vivo are not well defined, leaving a gap in our understanding of the development of humoral immune responses. Here, using an in vivo T cell independent B cell differentiation model, we define a cellular division-dependent cis-regulatory element road map using ATAC-seq. Chromatin accessibility changes correlate with gene expression and reveal the reprogramming of transcriptional networks and the genes they regulate at specific cell divisions. A subset of genes in naive B cells display accessible promoters in the absence of transcription and are marked by H3K27me3, an EZH2 catalyzed repressive modification. Such genes encode regulators of cell division and metabolism and include the essential plasma cell transcription factor Blimp-1. Chemical inhibition of EZH2 results in enhanced plasma cell formation, increased expression of the above gene set, and premature expression of Blimp-1 ex vivo. These data provide insights into cell-division coupled epigenetic and transcriptional processes that program plasma cells., During B cell differentiation, the role of different genomic loci in transcriptional and epigenetic regulation in vivo is not well defined. Here the authors use an in vivo B cell differentiation model to map cellular division-dependent cis-regulatory element road map with ATAC-seq.
- Published
- 2018
40. The molecular signature of murine T cell homeostatic proliferation reveals both inflammatory and immune inhibition patterns
- Author
-
James W. Austin, Karen A. Fortner, Jeremy M. Boss, Ralph C. Budd, and Jeffrey P. Bond
- Subjects
Cytotoxicity, Immunologic ,0301 basic medicine ,Cell Survival ,T cell ,Programmed Cell Death 1 Receptor ,Immunology ,Receptors, Antigen, T-Cell ,Mice, Transgenic ,Biology ,Lymphocyte Activation ,Article ,Immunomodulation ,Mice ,03 medical and health sciences ,Interleukin 21 ,0302 clinical medicine ,Downregulation and upregulation ,T-Lymphocyte Subsets ,medicine ,Animals ,Homeostasis ,Immunology and Allergy ,Cytotoxic T cell ,Antigen-presenting cell ,Cell Proliferation ,Inflammation ,Gene Expression Profiling ,CD44 ,Computational Biology ,Fas receptor ,Cell biology ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,biology.protein ,Female ,Transcriptome ,Biomarkers ,CD8 ,030215 immunology - Abstract
T lymphocyte homeostatic proliferation, driven by the engagement of T cell antigen receptor with self-peptide/major histocompatibility complexes, and signaling through the common γ-chain-containing cytokine receptors, is critical for the maintenance of the T cell compartment and is regulated by the Fas death receptor (Fas, CD95). In the absence of Fas, Fas-deficient lymphoproliferation spontaneous mutation (Ipr) mice accumulate homeostatically expanded T cells. The functional consequences of sequential rounds of homeostatic expansion are not well defined. We thus examined the gene expression profiles of murine wild-type and Fas-deficient Ipr CD8+ T cell subsets that have undergone different amounts of homeostatic proliferation as defined by their level of CD44 expression, and the CD4−CD8−TCRαβ+ T cell subset that results from extensive homeostatic expansion of CD8+ T cells. Our studies show that recurrent T cell homeostatic proliferation results in global gene expression changes, including the progressive upregulation of both cytolytic proteins such as Fas-Ligand and granzyme B as well as inhibitory proteins such as programmed cell death protein 1 (PD-1) and lymphocyte activating 3 (Lag3). These findings provide an explanation for how augmented T cell homeostatic expansion could lead to the frequently observed clinical paradox of simultaneous autoinflammatory and immunodeficiency syndromes and provide further insight into the regulatory programs that control chronically stimulated T cells.
- Published
- 2017
- Full Text
- View/download PDF
41. Cutting Edge: Chromatin Accessibility Programs CD8 T Cell Memory
- Author
-
Christopher D. Scharer, Bhanu Gandham, Alexander P. R. Bally, and Jeremy M. Boss
- Subjects
0301 basic medicine ,Genetics ,Effector ,Immunology ,Biology ,Lymphocytic choriomeningitis ,medicine.disease ,Chromatin ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Transcription (biology) ,Gene expression ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Gene ,ChIA-PET ,030215 immunology - Abstract
CD8 T cell memory is characterized by rapid recall of effector function, increased proliferation, and reduced activation requirements. Despite the extensive functional characterization, the molecular mechanisms that facilitate these enhanced properties are not well characterized. In this study, the assay for transposase-accessible chromatin sequencing was employed to map the cis-regulatory elements in CD8 T cells responding to acute and chronic lymphocytic choriomeningitis virus infections. Integration of chromatin accessibility profiles with gene expression data identified unique regulatory modules that were enriched for distinct combinations of transcription factor–binding motifs. Memory CD8 T cells displayed a chromatin accessibility structure that was absent from other acute and exhausted cells types and included key effector and proliferative genes. Stimulation of memory cells revealed enhanced transcription of “memory-primed” genes compared with naive cells. Thus, memory CD8 T cells display a preprogrammed chromatin accessibility profile and maintain a molecular history of cis-element usage, thereby reducing the steps necessary to revive effector functions.
- Published
- 2017
- Full Text
- View/download PDF
42. Conserved Region C Functions To Regulate PD-1 Expression and Subsequent CD8 T Cell Memory
- Author
-
Joshua T. Lee, Brian D. Evavold, Ryan J. Martinez, Alexander P. R. Bally, Benjamin G. Barwick, Yan Tang, and Jeremy M. Boss
- Subjects
0301 basic medicine ,T cell ,ZAP70 ,Immunology ,Biology ,Jurkat cells ,Cell biology ,TCIRG1 ,03 medical and health sciences ,Interleukin 21 ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,IL-2 receptor ,Antigen-presenting cell ,030215 immunology - Abstract
Expression of programmed death 1 (PD-1) on CD8 T cells promotes T cell exhaustion during chronic Ag exposure. During acute infections, PD-1 is transiently expressed and has the potential to modulate CD8 T cell memory formation. Conserved region C (CR-C), a promoter proximal cis-regulatory element that is critical to PD-1 expression in vitro, responds to NFATc1, FoxO1, and/or NF-κB signaling pathways. Here, a CR-C knockout mouse was established to determine its role on PD-1 expression and the corresponding effects on T cell function in vivo. Deletion of CR-C decreased PD-1 expression on CD4 T cells and Ag-specific CD8 T cells during acute and chronic lymphocytic choriomeningitis virus challenges, but did not affect the ability to clear an infection. Following acute lymphocytic choriomeningitis virus infection, memory CD8 T cells in the CR-C knockout mouse were formed in greater numbers, were more functional, and were more effective at responding to a melanoma tumor than wild-type memory cells. These data implicate a critical role for CR-C in governing PD-1 expression, and a subsequent role in guiding CD8 T cell differentiation. The data suggest the possibility that titrating PD-1 expression during CD8 T cell activation could have important ramifications in vaccine development and clinical care.
- Published
- 2017
- Full Text
- View/download PDF
43. A super enhancer controls expression and chromatin architecture within the MHC class II locus
- Author
-
Andrew R. Rahmberg, Jeremy M. Boss, Christopher D. Scharer, Parimal Majumder, Joshua T. Lee, Tian Mi, and Gaurav Kumar
- Subjects
0301 basic medicine ,musculoskeletal diseases ,CD4-Positive T-Lymphocytes ,CCCTC-Binding Factor ,Immunology ,Genes, MHC Class II ,Locus (genetics) ,chemical and pharmacologic phenomena ,Blood Donors ,Biology ,Polymorphism, Single Nucleotide ,Article ,HLA-DQ alpha-Chains ,03 medical and health sciences ,0302 clinical medicine ,Super-enhancer ,Cell Line, Tumor ,Immunology and Allergy ,CRISPR ,Humans ,Enhancer ,skin and connective tissue diseases ,Promoter Regions, Genetic ,Gene ,Research Articles ,Genetics ,Regulation of gene expression ,B-Lymphocytes ,Binding Sites ,Promoter ,Chromatin Assembly and Disassembly ,Burkitt Lymphoma ,Chromatin ,030104 developmental biology ,Haplotypes ,Genetic Loci ,CRISPR-Cas Systems ,030217 neurology & neurosurgery ,Gene Deletion ,HLA-DRB1 Chains - Abstract
A super enhancer termed DR/DQ-SE within the MHC-II locus was examined by CRISPR mutagenesis. DR/DQ-SE was found to regulate HLA-DRB1, -DQA1, and -DQB1 gene expression by controlling chromatin interactions/looping between MHC-II gene promoters and CTCF binding elements., Super enhancers (SEs) play critical roles in cell type–specific gene regulation. The mechanisms by which such elements work are largely unknown. Two SEs termed DR/DQ-SE and XL9-SE are situated within the human MHC class II locus between the HLA-DRB1 and HLA-DQA1 genes and are highly enriched for disease-causing SNPs. To test the function of these elements, we used CRISPR/Cas9 to generate a series of mutants that deleted the SE. Deletion of DR/DQ-SE resulted in reduced expression of HLA-DRB1 and HLA-DQA1 genes. The SEs were found to interact with each other and the promoters of HLA-DRB1 and HLA-DQA1. DR/DQ-SE also interacted with neighboring CTCF binding sites. Importantly, deletion of DR/DQ-SE reduced the local chromatin interactions, implying that it functions as the organizer for the local three-dimensional architecture. These data provide direct mechanisms by which an MHC-II SE contributes to expression of the locus and suggest how variation in these SEs may contribute to human disease and altered immunity., Graphical Abstract
- Published
- 2019
44. Understanding and measuring human B-cell tolerance and its breakdown in autoimmune disease
- Author
-
F. Eun-Hyung Lee, Christopher D. Scharer, Iñaki Sanz, Scott A. Jenks, Matthew C. Woodruff, Deepak Tomar, Christopher M. Tipton, Jeremy M. Boss, and Kevin S. Cashman
- Subjects
0301 basic medicine ,Future studies ,Immunology ,Autoimmunity ,Biology ,Human b cell ,medicine.disease_cause ,Lymphocyte Activation ,Autoantigens ,Article ,Immune tolerance ,Autoimmune Diseases ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Immune Tolerance ,Immunology and Allergy ,Animals ,Humans ,Epigenetics ,B cell ,Autoimmune disease ,B-Lymphocytes ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Immune System ,Neuroscience ,030215 immunology - Abstract
The maintenance of immunological tolerance of B lymphocytes is a complex and critical process that must be implemented as to avoid the detrimental development of autoreactivity and possible autoimmunity. Murine models have been invaluable to elucidate many of the key components in B-cell tolerance; however, translation to human homeostatic and pathogenic immune states can be difficult to assess. Functional autoreactive, flow cytometric, and single-cell cloning assays have proven to be critical in deciphering breaks in B-cell tolerance within autoimmunity; however, newer approaches to assess human B-cell tolerance may prove to be vital in the further exploration of underlying tolerance defects. In this review, we supply a comprehensive overview of human immune tolerance checkpoints with associated mechanisms of enforcement, and highlight current and future methodologies which are likely to benefit future studies into the mechanisms that become defective in human autoimmune conditions.
- Published
- 2019
45. LSD1 Cooperates with Noncanonical NF-κB Signaling to Regulate Marginal Zone B Cell Development
- Author
-
Christopher D. Scharer, Robert R. Haines, Jenna L. Lobby, and Jeremy M. Boss
- Subjects
animal structures ,Immunology ,B-Lymphocyte Subsets ,Article ,Epigenesis, Genetic ,03 medical and health sciences ,Mice ,0302 clinical medicine ,NF-kappa B p52 Subunit ,Marginal zone B-cell ,medicine ,Immunology and Allergy ,Animals ,Follicular B cell ,Epigenetics ,Transcription factor ,B cell ,Histone Demethylases ,Mice, Knockout ,biology ,Chromatin ,Cell biology ,medicine.anatomical_structure ,biology.protein ,Demethylase ,Signal transduction ,030215 immunology ,Signal Transduction - Abstract
Marginal zone B cells (MZB) are a mature B cell subset that rapidly respond to blood-borne pathogens. Although the transcriptional changes that occur throughout MZB development are known, the corresponding epigenetic changes and epigenetic modifying proteins that facilitate these changes are poorly understood. The histone demethylase LSD1 is an epigenetic modifier that promotes plasmablast formation, but its role in B cell development has not been explored. In this study, a role for LSD1 in the development of B cell subsets was examined. B cell–conditional deletion of LSD1 in mice resulted in a decrease in MZB whereas follicular B cells and bone marrow B cell populations were minimally affected. LSD1 repressed genes in MZB that were normally upregulated in the myeloid and follicular B cell lineages. Correspondingly, LSD1 regulated chromatin accessibility at the motifs of transcription factors known to regulate splenic B cell development, including NF-κB motifs. The importance of NF-κB signaling was examined through an ex vivo MZB development assay, which showed that both LSD1-deficient and NF-κB–inhibited transitional B cells failed to undergo full MZB development. Gene expression and chromatin accessibility analyses of in vivo– and ex vivo–generated LSD1-deficient MZB indicated that LSD1 regulated the downstream target genes of noncanonical NF-κB signaling. Additionally LSD1 was found to interact with the noncanonical NF-κB transcription factor p52. Together, these data reveal that the epigenetic modulation of the noncanonical NF-κB signaling pathway by LSD1 is an essential process during the development of MZB.
- Published
- 2019
46. PD-1 Expression during Acute Infection Is Repressed through an LSD1-Blimp-1 Axis
- Author
-
Alexander P. R. Bally, Jeremy M. Boss, Peiyuan Lu, Parimal Majumder, Yan Tang, Benjamin G. Barwick, Qing Wang, and Dennis K. Neeld
- Subjects
T cell ,Immunology ,Programmed Cell Death 1 Receptor ,Melanoma, Experimental ,CD8-Positive T-Lymphocytes ,Lymphocytic Choriomeningitis ,Lymphocyte Activation ,Article ,Histones ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,Lymphocytic choriomeningitis virus ,Melanoma ,Regulation of gene expression ,Histone Demethylases ,Mice, Knockout ,biology ,Wild type ,Acetylation ,Neoplasms, Experimental ,Cell biology ,Mice, Inbred C57BL ,Histone ,medicine.anatomical_structure ,Gene Expression Regulation ,Regulatory sequence ,DNA methylation ,Acute Disease ,Chronic Disease ,biology.protein ,Positive Regulatory Domain I-Binding Factor 1 ,Ex vivo ,030215 immunology ,Signal Transduction - Abstract
During prolonged exposure to Ags, such as chronic viral infections, sustained TCR signaling can result in T cell exhaustion mediated in part by expression of programmed cell death–1 (PD-1) encoded by the Pdcd1 gene. In this study, dynamic changes in histone H3K4 modifications at the Pdcd1 locus during ex vivo and in vivo activation of CD8 T cells suggested a potential role for the histone H3 lysine 4 demethylase LSD1 in regulating PD-1 expression. CD8 T cells lacking LSD1 expressed higher levels of Pdcd1 mRNA following ex vivo stimulation as well as increased surface levels of PD-1 during acute, but not chronic, infection with lymphocytic choriomeningitis virus (LCMV). Blimp-1, a known repressor of PD-1, recruited LSD1 to the Pdcd1 gene during acute, but not chronic, LCMV infection. Loss of DNA methylation at Pdcd1’s promoter-proximal regulatory regions is highly correlated with its expression. However, following acute LCMV infection, in which PD-1 expression levels return to near baseline, LSD1-deficient CD8 T cells failed to remethylate the Pdcd1 locus to the levels of wild-type cells. Finally, in a murine melanoma model, the frequency of PD-1–expressing tumor-infiltrating LSD1-deficient CD8 T cells was greater than in wild type. Thus, LSD1 is recruited to the Pdcd1 locus by Blimp-1, downregulates PD-1 expression by facilitating the removal of activating histone marks, and is important for remethylation of the locus. Together, these data provide insight into the complex regulatory mechanisms governing T cell immunity and regulation of a critical T cell checkpoint gene.
- Published
- 2019
47. PD-1 expression during acute infection is repressed through a LSD1- Blimp-1 axis
- Author
-
Peiyuan Lu, Benjamin G. Barwick, Parimal Majumder, Jeremy M. Boss, Dennis K. Neeld, Alexander P. R. Bally, Yan Tang, and Qing Wang
- Subjects
0303 health sciences ,animal structures ,biology ,T cell ,T-cell receptor ,Repressor ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,Histone ,medicine.anatomical_structure ,Antigen ,Regulatory sequence ,DNA methylation ,biology.protein ,medicine ,Cytotoxic T cell ,030304 developmental biology ,030215 immunology - Abstract
During prolonged exposure to antigens, such as chronic viral infections, sustained T cell receptor (TCR) signaling can result in T cell exhaustion mediated in part by expression of Programmed cell death-1 (PD-1) encoded by the Pdcd1 gene. Here, dynamic changes in histone H3K4 modifications at the Pdcd1 locus during ex vivo and in vivo activation of CD8 T cells, suggested a potential role for the histone H3 lysine 4 demethylase LSD1 in regulating PD-1 expression. CD8 T cells lacking LSD1 expressed higher levels of Pdcd1 mRNA following ex vivo stimulation, as well as increased surface levels of PD-1 during acute but not chronic infection with lymphocytic choriomeningitis virus (LCMV). Blimp-1, a known repressor of PD-1, recruited LSD1 to the Pdcd1 gene during acute but not chronic LCMV infection. Loss of DNA methylation at Pdcd1’s promoter proximal regulatory regions is highly correlated with its expression. However, following acute LCMV infection where PD-1 expression levels return to near base line, LSD1-deficient CD8 T cells failed to remethylate the Pdcd1 locus to the levels of wild-type cells. Finally, in a murine melanoma model, the frequency of PD-1 expressing tumor infiltrating LSD1-deficient CD8 T cells was greater than wild-type. Thus, LSD1 is recruited to the Pdcd1 locus by Blimp-1, downregulates PD-1 expression by facilitating the removal of activating histone marks, and is important for remethylation of the locus. Together, these data provide insight into the complex regulatory mechanisms governing T cell immunity and the regulation of a critical T cell checkpoint gene.Key PointsLSD1 suppress PD-1 expression following acute infection or transient induction.Blimp-1 binding to the Pdcd1 locus is required to recruit LSD1.LSD1 is required to fully remethylate the PD-1 proximal promoter region.
- Published
- 2019
- Full Text
- View/download PDF
48. Influenza-specific effector memory B cells predict long-lived antibody responses to vaccination in humans
- Author
-
Christopher D. Scharer, John E. Bradley, Iñaki Sanz, Christopher F. Fucile, Betty Mousseau, Paul A. Goepfert, Frances E. Lund, Rodney G. King, Tian Mi, Alexander F. Rosenberg, Fen Zhou, Anoma Nellore, Troy D. Randall, Christopher M. Tipton, Esther Zumaquero, and Jeremy M. Boss
- Subjects
0303 health sciences ,education.field_of_study ,Effector ,Population ,Hemagglutinin (influenza) ,Biology ,In vitro ,3. Good health ,Vaccination ,03 medical and health sciences ,0302 clinical medicine ,Humoral immunity ,Immunology ,biology.protein ,Epigenetics ,Antibody ,education ,030304 developmental biology ,030215 immunology - Abstract
Seasonal influenza vaccination elicits hemagglutinin (HA)-specific CD27+ memory B cells (Bmem) that differ in expression of T-bet, BACH2 and TCF7. T-bethiBACH2loTCF7lo Bmem are transcriptionally similar to effector-like memory cells while T-betloBACH2+TCF7+ Bmem exhibit stem-like central memory properties. T-bethi Bmem do not express plasma cell-specific transcription factors but do exhibit transcriptional, epigenetic, metabolic and functional changes that poise the cells for antibody production. Consistent with these changes, D7 HA+ T-bethi Bmem express intracellular immunoglobulin and T-bethi Bmem differentiate more rapidly into ASCs in vitro. The T-bethi Bmem response positively correlates with long-lived humoral immunity and clonotypes from T-bethi Bmem are represented in the early secondary ASC response to repeat vaccination, suggesting that this effector-like population can be used to predict vaccine durability and recall potential.
- Published
- 2019
- Full Text
- View/download PDF
49. IFNγ induces epigenetic programming of human T-bethi B cells and promotes TLR7/8 and IL-21 induced differentiation
- Author
-
W. Winn Chatham, Christopher D. Scharer, Ignacio Sanz, Frances E. Lund, Scott A. Jenks, Trenton R. Schoeb, Alexander F. Rosenberg, John E. Bradley, Antonio Rosal-Vela, Robert P. Kimberly, Wojciech Wojciechowski, S. Louis Bridges, Travis Ptacek, Jeremy M. Boss, Esther Zumaquero, Maria I. Danila, Davide Botta, Jeffrey C. Edberg, Betty Mousseau, Sara L. Stone, and Anoma Nellore
- Subjects
0301 basic medicine ,QH301-705.5 ,Science ,Naive B cell ,chemical and pharmacologic phenomena ,Biology ,T-bet ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Autoimmunity ,03 medical and health sciences ,0302 clinical medicine ,systemic lupus erythematosus ,PRDM1 ,medicine ,Biology (General) ,Transcription factor ,030304 developmental biology ,0303 health sciences ,General Immunology and Microbiology ,General Neuroscience ,hemic and immune systems ,General Medicine ,TLR7 ,3. Good health ,Cell biology ,Chromatin ,interferons ,030104 developmental biology ,antibody secreting cells ,Medicine ,Reprogramming ,B lymphocytes ,030215 immunology ,IRF4 - Abstract
Although B cells expressing the IFNγR or the IFNγ-inducible transcription factor T-bet drive autoimmunity in Systemic Lupus Erythematosus (SLE)-prone mouse models, the role for IFNγ signaling in human antibody responses is unknown. We show that elevated levels of IFNγ in SLE patients correlate with expansion of the T-bet expressing IgDnegCD27negCD11c+CXCR5neg (DN2) pre-antibody secreting cell (pre-ASC) subset. We demonstrate that naïve B cells form T-bethi pre-ASCs following stimulation with either Th1 cells or with IFNγ, IL-2, anti-Ig and TLR7/8 ligand and that IL-21 dependent ASC formation is significantly enhanced by IFNγ or IFNγ-producing T cells. IFNγ promotes ASC development by synergizing with IL-2 and TLR7/8 ligands to induce genome-wide epigenetic reprogramming of B cells, which results in increased chromatin accessibility surrounding IRF4 and BLIMP1 binding motifs and epigenetic remodeling of IL21R and PRDM1 loci. Finally, we show that IFNγ signals poise B cells to differentiate by increasing their responsiveness to IL-21.
- Published
- 2019
- Full Text
- View/download PDF
50. T-bet transcription factor promotes antibody secreting cell differentiation by limiting the inflammatory effects of IFNγ on B cells
- Author
-
Alexander F. Rosenberg, Zhenhao Qi, Amy S. Weinmann, Huiping Jiang, Ravi S. Misra, Christopher D. Scharer, Ananda W. Goldrath, André Ballesteros-Tato, Scott R. Brodeur, Bingfei Yu, Jeremy M. Boss, Wojciech Wojciechowski, Christopher A. Risley, Betty Mousseau, Troy D. Randall, Danielle A. Chisolm, Jessica N. Peel, Sara L. Stone, Adedayo Hanidu, Frances E. Lund, and Michael D. Schultz
- Subjects
0301 basic medicine ,Immunology ,Cell ,chemical and pharmacologic phenomena ,Biology ,Article ,03 medical and health sciences ,Interferon-gamma ,Mice ,0302 clinical medicine ,Influenza A Virus, H1N1 Subtype ,Downregulation and upregulation ,Orthomyxoviridae Infections ,Interferon ,medicine ,Immunology and Allergy ,Animals ,Antibody-Producing Cells ,Transcription factor ,B cell ,Cells, Cultured ,Receptors, Interferon ,Strongylida Infections ,Mice, Knockout ,B-Lymphocytes ,Nematospiroides dubius ,Effector ,Influenza A Virus, H3N2 Subtype ,hemic and immune systems ,Cell Differentiation ,T-Lymphocytes, Helper-Inducer ,Chromatin ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Antibody-Secreting Cells ,Positive Regulatory Domain I-Binding Factor 1 ,T-Box Domain Proteins ,medicine.drug - Abstract
Although viral infections elicit robust interferon-γ (IFN-γ) and long-lived antibody-secreting cell (ASC) responses, the roles for IFN-γ and IFN-γ-induced transcription factors (TFs) in ASC development are unclear. We showed that B cell intrinsic expression of IFN-γR and the IFN-γ-induced TF T-bet were required for T-helper 1 cell-induced differentiation of B cells into ASCs. IFN-γR signaling induced Blimp1 expression in B cells but also initiated an inflammatory gene program that, if not restrained, prevented ASC formation. T-bet did not affect Blimp1 upregulation in IFN-γ-activated B cells but instead regulated chromatin accessibility within the Ifng and Ifngr2 loci and repressed the IFN-γ-induced inflammatory gene program. Consistent with this, B cell intrinsic T-bet was required for formation of long-lived ASCs and secondary ASCs following viral, but not nematode, infection. Therefore, T-bet facilitates differentiation of IFN-γ-activated inflammatory effector B cells into ASCs in the setting of IFN-γ-, but not IL-4-, induced inflammatory responses.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.