80 results on '"Lichen Jing"'
Search Results
2. CD4 and CD8 co-receptors modulate functional avidity of CD1b-restricted T cells
- Author
-
Charlotte A. James, Yuexin Xu, Melissa S. Aguilar, Lichen Jing, Erik D. Layton, Martine Gilleron, Adriaan J. Minnaard, Thomas J. Scriba, Cheryl L. Day, Edus H. Warren, David M. Koelle, and Chetan Seshadri
- Subjects
Science - Abstract
CD4 and CD8 co-receptors are routinely used to define distinct functional and phenotypic lineages of T cells. Here the authors show CD4 and CD8 also modulate the functional avidity of the CD1b-restricted response to mycobacterial lipid antigens
- Published
- 2022
- Full Text
- View/download PDF
3. Transcriptional and functional analyses of neoantigen-specific CD4 T cells during a profound response to anti-PD-L1 in metastatic Merkel cell carcinoma
- Author
-
Vladimir Makarov, Paul Nghiem, Jaehyuk Choi, Timothy A Chan, Kimberly S Smythe, Jean S Campbell, Robert H Pierce, Raphael Gottardo, Candice Church, David M Koelle, Thomas Pulliam, Natalie Longino, Song Y Park, Nadeem Riaz, Lichen Jing, and Robert Amezquita
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background Merkel cell carcinoma (MCC) often responds to PD-1 pathway blockade, regardless of tumor-viral status (~80% of cases driven by the Merkel cell polyomavirus (MCPyV)). Prior studies have characterized tumor-specific T cell responses to MCPyV, which have typically been CD8, but little is known about the T cell response to UV-induced neoantigens.Methods A patient in her mid-50s with virus-negative (VN) MCC developed large liver metastases after a brief initial response to chemotherapy. She received anti-PD-L1 (avelumab) and had a partial response within 4 weeks. Whole exome sequencing (WES) was performed to determine potential neoantigen peptides. Characterization of peripheral blood neoantigen T cell responses was evaluated via interferon-gamma (IFNγ) ELISpot, flow cytometry and single-cell RNA sequencing. Tumor-resident T cells were characterized by multiplexed immunohistochemistry.Results WES identified 1027 tumor-specific somatic mutations, similar to the published average of 1121 for VN-MCCs. Peptide prediction with a binding cut-off of ≤100 nM resulted in 77 peptides that were synthesized for T cell assays. Although peptides were predicted based on class I HLAs, we identified circulating CD4 T cells targeting 5 of 77 neoantigens. In contrast, no neoantigen-specific CD8 T cell responses were detected. Neoantigen-specific CD4 T cells were undetectable in blood before anti-PD-L1 therapy but became readily detectible shortly after starting therapy. T cells produced robust IFNγ when stimulated by neoantigen (mutant) peptides but not by the normal (wild-type) peptides. Single cell RNAseq showed neoantigen-reactive T cells expressed the Th1-associated transcription factor (T-bet) and associated cytokines. These CD4 T cells did not significantly exhibit cytotoxicity or non-Th1 markers. Within the pretreatment tumor, resident CD4 T cells were also Th1-skewed and expressed T-bet.Conclusions We identified and characterized tumor-specific Th1-skewed CD4 T cells targeting multiple neoantigens in a patient who experienced a profound and durable partial response to anti-PD-L1 therapy. To our knowledge, this is the first report of neoantigen-specific T cell responses in MCC. Although CD4 and CD8 T cells recognizing viral tumor antigens are often detectible in virus-positive MCC, only CD4 T cells recognizing neoantigens were detected in this patient. These findings suggest that CD4 T cells can play an important role in the response to anti-PD-(L)1 therapy.
- Published
- 2022
- Full Text
- View/download PDF
4. T cell receptor sequencing identifies prior SARS-CoV-2 infection and correlates with neutralizing antibodies and disease severity
- Author
-
Rebecca Elyanow, Thomas M. Snyder, Sudeb C. Dalai, Rachel M. Gittelman, Jim Boonyaratanakornkit, Anna Wald, Stacy Selke, Mark H. Wener, Chihiro Morishima, Alexander L. Greninger, Michael Gale Jr., Tien-Ying Hsiang, Lichen Jing, Michael R. Holbrook, Ian M. Kaplan, H. Jabran Zahid, Damon H. May, Jonathan M. Carlson, Lance Baldo, Thomas Manley, Harlan S. Robins, and David M. Koelle
- Subjects
COVID-19 ,Infectious disease ,Medicine - Abstract
BACKGROUND Measuring the immune response to SARS-CoV-2 enables assessment of past infection and protective immunity. SARS-CoV-2 infection induces humoral and T cell responses, but these responses vary with disease severity and individual characteristics.METHODS A T cell receptor (TCR) immunosequencing assay was conducted using small-volume blood samples from 302 individuals recovered from COVID-19. Correlations between the magnitude of the T cell response and neutralizing antibody (nAb) titers or indicators of disease severity were evaluated. Sensitivity of T cell testing was assessed and compared with serologic testing.RESULTS SARS-CoV-2–specific T cell responses were significantly correlated with nAb titers and clinical indicators of disease severity, including hospitalization, fever, and difficulty breathing. Despite modest declines in depth and breadth of T cell responses during convalescence, high sensitivity was observed until at least 6 months after infection, with overall sensitivity ~5% greater than serology tests for identifying prior SARS-CoV-2 infection. Improved performance of T cell testing was most apparent in recovered, nonhospitalized individuals sampled > 150 days after initial illness, suggesting greater sensitivity than serology at later time points and in individuals with less severe disease. T cell testing identified SARS-CoV-2 infection in 68% (55 of 81) of samples with undetectable nAb titers (
- Published
- 2022
- Full Text
- View/download PDF
5. T cell response to intact SARS-CoV-2 includes coronavirus cross-reactive and variant-specific components
- Author
-
Lichen Jing, Xia Wu, Maxwell P. Krist, Tien-Ying Hsiang, Victoria L. Campbell, Christopher L. McClurkan, Sydney M. Favors, Lawrence A. Hemingway, Charmie Godornes, Denise Q. Tong, Stacy Selke, Angela C. LeClair, Chu-Woo Pyo, Daniel E. Geraghty, Kerry J. Laing, Anna Wald, Michael Gale Jr., and David M. Koelle
- Subjects
Infectious disease ,Medicine - Abstract
SARS-CoV-2 provokes a robust T cell response. Peptide-based studies exclude antigen processing and presentation biology, which may influence T cell detection studies. To focus on responses to whole virus and complex antigens, we used intact SARS-CoV-2 and full-length proteins with DCs to activate CD8 and CD4 T cells from convalescent people. T cell receptor (TCR) sequencing showed partial repertoire preservation after expansion. Resultant CD8 T cells recognize SARS-CoV-2–infected respiratory tract cells, and CD4 T cells detect inactivated whole viral antigen. Specificity scans with proteome-covering protein/peptide arrays show that CD8 T cells are oligospecific per subject and that CD4 T cell breadth is higher. Some CD4 T cell lines enriched using SARS-CoV-2 cross-recognize whole seasonal coronavirus (sCoV) antigens, with protein, peptide, and HLA restriction validation. Conversely, recognition of some epitopes is eliminated for SARS-CoV-2 variants, including spike (S) epitopes in the Alpha, Beta, Gamma, and Delta variant lineages.
- Published
- 2022
- Full Text
- View/download PDF
6. HSV-2-Specific Human Female Reproductive Tract Tissue Resident Memory T Cells Recognize Diverse HSV Antigens
- Author
-
David M. Koelle, Lichun Dong, Lichen Jing, Kerry J. Laing, Jia Zhu, Lei Jin, Stacy Selke, Anna Wald, Dana Varon, Meei-Li Huang, Christine Johnston, Lawrence Corey, and Christine M. Posavad
- Subjects
HSV-2 ,CD8 ,CD4 ,epitope ,dendritic cell ,female reproductive tract immunology ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Antigen-specific TRM persist and protect against skin or female reproductive tract (FRT) HSV infection. As the pathogenesis of HSV differs between humans and model organisms, we focus on humans with well-characterized recurrent genital HSV-2 infection. Human CD8+ TRM persisting at sites of healed human HSV-2 lesions have an activated phenotype but it is unclear if TRM can be cultivated in vitro. We recovered HSV-specific TRM from genital skin and ectocervix biopsies, obtained after recovery from recurrent genital HSV-2, using ex vivo activation by viral antigen. Up to several percent of local T cells were HSV-reactive ex vivo. CD4 and CD8 T cell lines were up to 50% HSV-2-specific after sorting-based enrichment. CD8 TRM displayed HLA-restricted reactivity to specific HSV-2 peptides with high functional avidities. Reactivity to defined peptides persisted locally over several month and was quite subject-specific. CD4 TRM derived from biopsies, and from an extended set of cervical cytobrush specimens, also recognized diverse HSV-2 antigens and peptides. Overall we found that HSV-2-specific TRM are abundant in the FRT between episodes of recurrent genital herpes and maintain competency for expansion. Mucosal sites are accessible for clinical monitoring during immune interventions such as therapeutic vaccination.
- Published
- 2022
- Full Text
- View/download PDF
7. Tissue-Resident-Memory CD8+ T Cells Bridge Innate Immune Responses in Neighboring Epithelial Cells to Control Human Genital Herpes
- Author
-
Tao Peng, Khamsone Phasouk, Catherine N. Sodroski, Sijie Sun, Yon Hwangbo, Erik D. Layton, Lei Jin, Alexis Klock, Kurt Diem, Amalia S. Magaret, Lichen Jing, Kerry Laing, Alvason Li, Meei-Li Huang, Max Mertens, Christine Johnston, Keith R. Jerome, David M. Koelle, Anna Wald, David M. Knipe, Lawrence Corey, and Jia Zhu
- Subjects
tissue-resident-memory T cells (TRM) ,innate antiviral response ,cell-intrinsic immunity ,IFI16 restriction factor ,tissue microenvironment ,human genital herpes ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Tissue-resident-memory T cells (TRM) populate the body’s barrier surfaces, functioning as frontline responders against reencountered pathogens. Understanding of the mechanisms by which CD8TRM achieve effective immune protection remains incomplete in a naturally recurring human disease. Using laser capture microdissection and transcriptional profiling, we investigate the impact of CD8TRM on the tissue microenvironment in skin biopsies sequentially obtained from a clinical cohort of diverse disease expression during herpes simplex virus 2 (HSV-2) reactivation. Epithelial cells neighboring CD8TRM display elevated and widespread innate and cell-intrinsic antiviral signature expression, largely related to IFNG expression. Detailed evaluation via T-cell receptor reconstruction confirms that CD8TRM recognize viral-infected cells at the specific HSV-2 peptide/HLA level. The hierarchical pattern of core IFN-γ signature expression is well-conserved in normal human skin across various anatomic sites, while elevation of IFI16, TRIM 22, IFITM2, IFITM3, MX1, MX2, STAT1, IRF7, ISG15, IFI44, CXCL10 and CCL5 expression is associated with HSV-2-affected asymptomatic tissue. In primary human cells, IFN-γ pretreatment reduces gene transcription at the immediate-early stage of virus lifecycle, enhances IFI16 restriction of wild-type HSV-2 replication and renders favorable kinetics for host protection. Thus, the adaptive immune response through antigen-specific recognition instructs innate and cell-intrinsic antiviral machinery to control herpes reactivation, a reversal of the canonical thinking of innate activating adaptive immunity in primary infection. Communication from CD8TRM to surrounding epithelial cells to activate broad innate resistance might be critical in restraining various viral diseases.
- Published
- 2021
- Full Text
- View/download PDF
8. Varicella zoster virus productively infects human peripheral blood mononuclear cells to modulate expression of immunoinhibitory proteins and blocking PD-L1 enhances virus-specific CD8+ T cell effector function.
- Author
-
Dallas Jones, Christina N Como, Lichen Jing, Anna Blackmon, Charles Preston Neff, Owen Krueger, Andrew N Bubak, Brent E Palmer, David M Koelle, and Maria A Nagel
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Varicella zoster virus (VZV) is a lymphotropic alpha-herpesvirinae subfamily member that produces varicella on primary infection and causes zoster, vascular disease and vision loss upon reactivation from latency. VZV-infected peripheral blood mononuclear cells (PBMCs) disseminate virus to distal organs to produce clinical disease. To assess immune evasion strategies elicited by VZV that may contribute to dissemination of infection, human PBMCs and VZV-specific CD8+ T cells (V-CD8+) were mock- or VZV-infected and analyzed for immunoinhibitory protein PD-1, PD-L1, PD-L2, CTLA-4, LAG-3 and TIM-3 expression using flow cytometry. All VZV-infected PBMCs (monocytes, NK, NKT, B cells, CD4+ and CD8+ T cells) and V-CD8+ showed significant elevations in PD-L1 expression compared to uninfected cells. VZV induced PD-L2 expression in B cells and V-CD8+. Only VZV-infected CD8+ T cells, NKT cells and V-CD8+ upregulated PD-1 expression, the immunoinhibitory receptor for PD-L1/PD-L2. VZV induced CTLA-4 expression only in V-CD8+ and no significant changes in LAG-3 or TIM-3 expression were observed in V-CD8+ or PBMC T cells. To test whether PD-L1, PD-L2 or CTLA-4 regulates V-CD8+ effector function, autologous PBMCs were VZV-infected and co-cultured with V-CD8+ cells in the presence of blocking antibodies against PD-L1, PD-L2 or CTLA-4; ELISAs revealed significant elevations in IFNγ only upon blocking of PD-L1. Together, these results identified additional immune cells that are permissive to VZV infection (monocytes, B cells and NKT cells); along with a novel mechanism for inhibiting CD8+ T cell effector function through induction of PD-L1 expression.
- Published
- 2019
- Full Text
- View/download PDF
9. A Novel Approach of Identifying Immunodominant Self and Viral Antigen Cross-Reactive T Cells and Defining the Epitopes They Recognize
- Author
-
Junbao Yang, Lichen Jing, Eddie A. James, John A. Gebe, David M. Koelle, and William W. Kwok
- Subjects
molecular mimicry ,T cell cross-reactivity ,influenza ,glutamate decarboxylase 65 ,autoreactive T cells ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Infection and vaccination can lead to activation of autoreactive T cells, including the activation of cross-reactive T cells. However, detecting these cross-reactive T cells and identifying the non-self and self-antigen epitopes is difficult. The current study demonstrates the utility of a novel approach that effectively accomplishes both. We utilized surface expression of CD38 on newly activated CD4 memory T cells as a strategy to identify type 1 diabetes associated autoreactive T cells activated by influenza vaccination in healthy subjects. We identified an influenza A matrix protein (MP) specific CD4+ T cell clone that cross-recognizes an immunodominant epitope from Glutamic Acid Decarboxylase 65 (GAD65) protein. The sequences of the MP and GAD65 peptides are rather distinct, with only 2 identical amino acids within the HLA-DR binding region. This result suggests that activation of autoreactive T cells by microbial infection under certain physiological conditions can occur amongst peptides with minimum amino acid sequence homology. This novel strategy also provides a new research pathway in which to examine activation of autoreactive CD4+ T cells after vaccination or natural infection.
- Published
- 2018
- Full Text
- View/download PDF
10. Experimental Oral Herpes Simplex Virus-1 (HSV-1) Co-infection in Simian Immunodeficiency Virus (SIV)-Infected Rhesus Macaques
- Author
-
Meropi Aravantinou, Olga Mizenina, Giulia Calenda, Jessica Kenney, Ines Frank, Jeffrey D. Lifson, Moriah Szpara, Lichen Jing, David M. Koelle, Natalia Teleshova, Brooke Grasperge, James Blanchard, Agegnehu Gettie, Elena Martinelli, and Nina Derby
- Subjects
HSV-1 ,SIV ,non-human primate model ,oral infection ,immune response ,Microbiology ,QR1-502 - Abstract
Herpes simplex virus 1 and 2 (HSV-1/2) similarly initiate infection in mucosal epithelia and establish lifelong neuronal latency. Anogenital HSV-2 infection augments the risk for sexual human immunodeficiency virus (HIV) transmission and is associated with higher HIV viral loads. However, whether oral HSV-1 infection contributes to oral HIV susceptibility, viremia, or oral complications of HIV infection is unknown. Appropriate non-human primate (NHP) models would facilitate this investigation, yet there are no published studies of HSV-1/SIV co-infection in NHPs. Thus, we performed a pilot study for an oral HSV-1 infection model in SIV-infected rhesus macaques to describe the feasibility of the modeling and resultant immunological changes. Three SIV-infected, clinically healthy macaques became HSV-1-infected by inoculation with 4 × 108 pfu HSV-1 McKrae on buccal, tongue, gingiva, and tonsils after gentle abrasion. HSV-1 DNA was shed in oral swabs for up to 21 days, and shedding recurred in association with intra-oral lesions after periods of no shedding during 56 days of follow up. HSV-1 DNA was detected in explant cultures of trigeminal ganglia collected at euthanasia on day 56. In the macaque with lowest baseline SIV viremia, SIV plasma RNA increased following HSV-1 infection. One macaque exhibited an acute pro-inflammatory response, and all three animals experienced T cell activation and mobilization in blood. However, T cell and antibody responses to HSV-1 were low and atypical. Through rigorous assessesments, this study finds that the virulent HSV-1 strain McKrae resulted in a low level HSV-1 infection that elicited modest immune responses and transiently modulated SIV infection.
- Published
- 2017
- Full Text
- View/download PDF
11. Local CD4 and CD8 T-cell reactivity to HSV-1 antigens documents broad viral protein expression and immune competence in latently infected human trigeminal ganglia.
- Author
-
Monique van Velzen, Lichen Jing, Albert D M E Osterhaus, Alessandro Sette, David M Koelle, and Georges M G M Verjans
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Herpes simplex virus type 1 (HSV-1) infection results in lifelong chronic infection of trigeminal ganglion (TG) neurons, also referred to as neuronal HSV-1 latency, with periodic reactivation leading to recrudescent herpetic disease in some persons. HSV-1 proteins are expressed in a temporally coordinated fashion during lytic infection, but their expression pattern during latent infection is largely unknown. Selective retention of HSV-1 reactive T-cells in human TG suggests their role in controlling reactivation by recognizing locally expressed HSV-1 proteins. We characterized the HSV-1 proteins recognized by virus-specific CD4 and CD8 T-cells recovered from human HSV-1-infected TG. T-cell clusters, consisting of both CD4 and CD8 T-cells, surrounded neurons and expressed mRNAs and proteins consistent with in situ antigen recognition and antiviral function. HSV-1 proteome-wide scans revealed that intra-TG T-cell responses included both CD4 and CD8 T-cells directed to one to three HSV-1 proteins per person. HSV-1 protein ICP6 was targeted by CD8 T-cells in 4 of 8 HLA-discordant donors. In situ tetramer staining demonstrated HSV-1-specific CD8 T-cells juxtaposed to TG neurons. Intra-TG retention of virus-specific CD4 T-cells, validated to the HSV-1 peptide level, implies trafficking of viral proteins from neurons to HLA class II-expressing non-neuronal cells for antigen presentation. The diversity of viral proteins targeted by TG T-cells across all kinetic and functional classes of viral proteins suggests broad HSV-1 protein expression, and viral antigen processing and presentation, in latently infected human TG. Collectively, the human TG represents an immunocompetent environment for both CD4 and CD8 T-cell recognition of HSV-1 proteins expressed during latent infection. HSV-1 proteins recognized by TG-resident T-cells, particularly ICP6 and VP16, are potential HSV-1 vaccine candidates.
- Published
- 2013
- Full Text
- View/download PDF
12. Mapping of Mcs30, a new mammary carcinoma susceptibility quantitative trait locus (QTL30) on rat chromosome 12: identification of fry as a candidate Mcs gene.
- Author
-
Xuefeng Ren, Jessica C Graham, Lichen Jing, Andrei M Mikheev, Yuan Gao, Jenny Pan Lew, Hong Xie, Andrea S Kim, Xiuling Shang, Cynthia Friedman, Graham Vail, Ming Zhu Fang, Yana Bromberg, and Helmut Zarbl
- Subjects
Medicine ,Science - Abstract
Rat strains differ dramatically in their susceptibility to mammary carcinogenesis. On the assumption that susceptibility genes are conserved across mammalian species and hence inform human carcinogenesis, numerous investigators have used genetic linkage studies in rats to identify genes responsible for differential susceptibility to carcinogenesis. Using a genetic backcross between the resistant Copenhagen (Cop) and susceptible Fischer 344 (F344) strains, we mapped a novel mammary carcinoma susceptibility (Mcs30) locus to the centromeric region on chromosome 12 (LOD score of ∼8.6 at the D12Rat59 marker). The Mcs30 locus comprises approximately 12 Mbp on the long arm of rat RNO12 whose synteny is conserved on human chromosome 13q12 to 13q13. After analyzing numerous genes comprising this locus, we identified Fry, the rat ortholog of the furry gene of Drosophila melanogaster, as a candidate Mcs gene. We cloned and determined the complete nucleotide sequence of the 13 kbp Fry mRNA. Sequence analysis indicated that the Fry gene was highly conserved across evolution, with 90% similarity of the predicted amino acid sequence among eutherian mammals. Comparison of the Fry sequence in the Cop and F344 strains identified two non-synonymous single nucleotide polymorphisms (SNPs), one of which creates a putative, de novo phosphorylation site. Further analysis showed that the expression of the Fry gene is reduced in a majority of rat mammary tumors. Our results also suggested that FRY activity was reduced in human breast carcinoma cell lines as a result of reduced levels or mutation. This study is the first to identify the Fry gene as a candidate Mcs gene. Our data suggest that the SNPs within the Fry gene contribute to the genetic susceptibility of the F344 rat strain to mammary carcinogenesis. These results provide the foundation for analyzing the role of the human FRY gene in cancer susceptibility and progression.
- Published
- 2013
- Full Text
- View/download PDF
13. Spike-specific T cells are enriched in breastmilk following SARS-CoV-2 mRNA vaccination
- Author
-
Blair Armistead, Yonghou Jiang, Marc Carlson, Emily S. Ford, Saumya Jani, John Houck, Xia Wu, Lichen Jing, Tiffany Pecor, Alisa Kachikis, Winnie Yeung, Tina Nguyen, Rene Coig, Nana Minkah, Sasha E. Larsen, Rhea N. Coler, David M. Koelle, and Whitney E. Harrington
- Subjects
Immunology ,Immunology and Allergy - Published
- 2023
14. Data from Prevalent and Diverse Intratumoral Oncoprotein-Specific CD8+ T Cells within Polyomavirus-Driven Merkel Cell Carcinomas
- Author
-
David M. Koelle, Paul Nghiem, Shailender Bhatia, Aude G. Chapuis, Kelly G. Paulson, Alexander L. Greninger, Hong Xie, Maclean M. Cook, Aric Colunga, Olga K. Afanasiev, Jayasri G. Iyer, Dafina Ibrani, Rima M. Kulikauskas, Candice D. Church, Mariliis Ott, and Lichen Jing
- Abstract
Merkel cell carcinoma (MCC) is often caused by persistent expression of Merkel cell polyomavirus (MCPyV) T-antigen (T-Ag). These non-self proteins comprise about 400 amino acids (AA). Clinical responses to immune checkpoint inhibitors, seen in about half of patients, may relate to T-Ag–specific T cells. Strategies to increase CD8+ T-cell number, breadth, or function could augment checkpoint inhibition, but vaccines to augment immunity must avoid delivery of oncogenic T-antigen domains. We probed MCC tumor-infiltrating lymphocytes (TIL) with an artificial antigen-presenting cell (aAPC) system and confirmed T-Ag recognition with synthetic peptides, HLA-peptide tetramers, and dendritic cells (DC). TILs from 9 of 12 (75%) subjects contained CD8+ T cells recognizing 1–8 MCPyV epitopes per person. Analysis of 16 MCPyV CD8+ TIL epitopes and prior TIL data indicated that 97% of patients with MCPyV+ MCC had HLA alleles with the genetic potential that restrict CD8+ T-cell responses to MCPyV T-Ag. The LT AA 70–110 region was epitope rich, whereas the oncogenic domains of T-Ag were not commonly recognized. Specific recognition of T-Ag–expressing DCs was documented. Recovery of MCPyV oncoprotein–specific CD8+ TILs from most tumors indicated that antigen indifference was unlikely to be a major cause of checkpoint inhibition failure. The myriad of epitopes restricted by diverse HLA alleles indicates that vaccination can be a rational component of immunotherapy if tumor immune suppression can be overcome, and the oncogenic regions of T-Ag can be modified without impacting immunogenicity.
- Published
- 2023
15. Supplementary Tables from Prevalent and Diverse Intratumoral Oncoprotein-Specific CD8+ T Cells within Polyomavirus-Driven Merkel Cell Carcinomas
- Author
-
David M. Koelle, Paul Nghiem, Shailender Bhatia, Aude G. Chapuis, Kelly G. Paulson, Alexander L. Greninger, Hong Xie, Maclean M. Cook, Aric Colunga, Olga K. Afanasiev, Jayasri G. Iyer, Dafina Ibrani, Rima M. Kulikauskas, Candice D. Church, Mariliis Ott, and Lichen Jing
- Abstract
Sup Tables 1-6
- Published
- 2023
16. Supplementary Figures from Prevalent and Diverse Intratumoral Oncoprotein-Specific CD8+ T Cells within Polyomavirus-Driven Merkel Cell Carcinomas
- Author
-
David M. Koelle, Paul Nghiem, Shailender Bhatia, Aude G. Chapuis, Kelly G. Paulson, Alexander L. Greninger, Hong Xie, Maclean M. Cook, Aric Colunga, Olga K. Afanasiev, Jayasri G. Iyer, Dafina Ibrani, Rima M. Kulikauskas, Candice D. Church, Mariliis Ott, and Lichen Jing
- Abstract
Sup Figures 1-13
- Published
- 2023
17. Data from Tumor-Infiltrating Merkel Cell Polyomavirus-Specific T Cells Are Diverse and Associated with Improved Patient Survival
- Author
-
Paul Nghiem, David M. Koelle, Thomas Blankenstein, Martin McIntosh, Gerald Willimsky, Ioannis Gavvovidis, Michi Shinohara, Lichen Jing, Kristina Lachance, Matthew P. Fitzgibbon, David Crispin, Lichun Dong, Candice D. Church, and Natalie J. Miller
- Abstract
Tumor-infiltrating CD8+ T cells are associated with improved survival of patients with Merkel cell carcinoma (MCC), an aggressive skin cancer causally linked to Merkel cell polyomavirus (MCPyV). However, CD8+ T-cell infiltration is robust in only 4% to 18% of MCC tumors. We characterized the T-cell receptor (TCR) repertoire restricted to one prominent epitope of MCPyV (KLLEIAPNC, “KLL”) and assessed whether TCR diversity, tumor infiltration, or T-cell avidity correlated with clinical outcome. HLA-A*02:01/KLL tetramer+ CD8+ T cells from MCC patient peripheral blood mononuclear cells (PBMC) and tumor-infiltrating lymphocytes (TIL) were isolated via flow cytometry. TCRβ (TRB) sequencing was performed on tetramer+ cells from PBMCs or TILs (n = 14) and matched tumors (n = 12). Functional avidity of T-cell clones was determined by IFNγ production. We identified KLL tetramer+ T cells in 14% of PBMC and 21% of TIL from MCC patients. TRB repertoires were strikingly diverse (397 unique TRBs were identified from 12 patients) and mostly private (only one TCRb clonotype shared between two patients). An increased fraction of KLL-specific TIL (>1.9%) was associated with significantly increased MCC-specific survival P = 0.0009). T-cell cloning from four patients identified 42 distinct KLL-specific TCRa/b pairs. T-cell clones from patients with improved MCC-specific outcomes were more avid (P < 0.05) and recognized an HLA-appropriate MCC cell line. T cells specific for a single MCPyV epitope display marked TCR diversity within and between patients. Intratumoral infiltration by MCPyV-specific T cells was associated with significantly improved MCC-specific survival, suggesting that augmenting the number or avidity of virus-specific T cells may have therapeutic benefit. Cancer Immunol Res; 5(2); 137–47. ©2017 AACR.
- Published
- 2023
18. Data from Merkel Cell Polyomavirus-Specific CD8+ and CD4+ T-cell Responses Identified in Merkel Cell Carcinomas and Blood
- Author
-
Paul Nghiem, David M. Koelle, Cassian Yee, Denise Galloway, David Byrd, Erik Farrar, Ivy Lai, Joseph Carter, Lichun Dong, Joshua O. Marshak, Lichen Jing, Kotaro Nagase, Kelly Paulson, Christopher McClurkan, Olga K. Afanasiev, and Jayasri G. Iyer
- Abstract
Purpose: Merkel cell polyomavirus (MCPyV) is prevalent in the general population, integrates into most Merkel cell carcinomas (MCC), and encodes oncoproteins required for MCC tumor growth. We sought to characterize T-cell responses directed against viral proteins that drive this cancer as a step toward immunotherapy.Experimental Design: Intracellular cytokine cytometry, IFN-γ enzyme-linked immunospot (ELISPOT) assay, and a novel HLA-A*2402–restricted MCPyV tetramer were used to identify and characterize T-cell responses against MCPyV oncoproteins in tumors and blood of MCC patients and control subjects.Results: We isolated virus-reactive CD8 or CD4 T cells from MCPyV-positive MCC tumors (2 of 6) but not from virus-negative tumors (0 of 4). MCPyV-specific T-cell responses were also detected in the blood of MCC patients (14 of 27) and control subjects (5 of 13). These T cells recognized a broad range of peptides derived from capsid proteins (2 epitopes) and oncoproteins (24 epitopes). HLA-A*2402–restricted MCPyV oncoprotein processing and presentation by mammalian cells led to CD8-mediated cytotoxicity. Virus-specific CD8 T cells were markedly enriched among tumor infiltrating lymphocytes as compared with blood, implying intact T-cell trafficking into the tumor. Although tetramer-positive CD8 T cells were detected in the blood of 2 of 5 HLA-matched MCC patients, these cells failed to produce IFN-γ when challenged ex vivo with peptide.Conclusions: Our findings suggest that MCC tumors often develop despite the presence of T cells specific for MCPyV T-Ag oncoproteins. The identified epitopes may be candidates for peptide-specific vaccines and tumor- or virus-specific adoptive immunotherapies to overcome immune evasion mechanisms in MCC patients. Clin Cancer Res; 17(21); 6671–80. ©2011 AACR.
- Published
- 2023
19. 50 Merkel cell polyomavirus-specific CD8 T cells in blood, but not in tumors, correlate with immunotherapy response in merkel cell carcinoma
- Author
-
Thomas Pulliam, Saumya Jani, Lichen Jing, Jiajia Zhang, Rima Kulikauskas, Candice Church, Charlie Garnett-Benson, Kelly Paulson, Kellie Smith, Andrew Pardoll, David Koelle, Suzanne Topalian, and Paul Nghiem
- Published
- 2022
20. CD8+ T cell clonotypes from prior SARS-CoV-2 infection predominate during the cellular immune response to mRNA vaccination
- Author
-
Emily S. Ford, Koshlan Mayer-Blackwell, Lichen Jing, Anton M. Sholukh, Russell St. Germain, Emily L. Bossard, Hong Xie, Thomas H. Pulliam, Saumya Jani, Stacy Selke, Carlissa J. Burrow, Christopher L. McClurkan, Anna Wald, Michael R. Holbrook, Brett Eaton, Elizabeth Eudy, Michael Murphy, Elena Postnikova, Harlan S. Robins, Rebecca Elyanow, Rachel M. Gittelman, Matyas Ecsedi, Elise Wilcox, Aude G. Chapuis, Andrew Fiore-Gartland, and David M. Koelle
- Abstract
Almost three years into the SARS-CoV-2 pandemic, hybrid immunity is highly prevalent worldwide and more protective than vaccination or prior infection alone. Given emerging resistance of variant strains to neutralizing antibodies (nAb), it is likely that T cells contribute to this protection. To understand how sequential SARS-CoV-2 infection and mRNA-vectored SARS-CoV-2 spike (S) vaccines affect T cell clonotype-level expansion kinetics, we identified and cross-referenced TCR sequences from thousands of S-reactive single cells against deeply sequenced peripheral blood TCR repertoires longitudinally collected from persons during COVID-19 convalescence through booster vaccination. Successive vaccinations recalled memory T cells and elicited antigen-specific T cell clonotypes not detected after infection. Vaccine-related recruitment of novel clonotypes and the expansion of S-specific clones were most strongly observed for CD8+ T cells. Severe COVID-19 illness was associated with a more diverse CD4+ T cell response to SARS-CoV-2 both prior to and after mRNA vaccination, suggesting imprinting of CD4+ T cells by severe infection. TCR sequence similarity search algorithms revealed myriad public TCR clusters correlating with human leukocyte antigen (HLA) alleles. Selected TCRs from distinct clusters functionally recognized S in the predicted HLA context, with fine viral peptide requirements differing between TCRs. Most subjects tested had S-specific T cells in the nasal mucosa after a 3rd mRNA vaccine dose. The blood and nasal T cell responses to vaccination revealed by clonal tracking were more heterogeneous than nAb boosts. Analysis of bulk and single cell TCR sequences reveals T cell kinetics and diversity at the clonotype level, without requiring prior knowledge of T cell epitopes or HLA restriction, providing a roadmap for rapid assessment of T cell responses to emerging pathogens.
- Published
- 2022
21. Spike-specific T cells are enriched in breastmilk following SARS-CoV-2 mRNA vaccination
- Author
-
Blair Armistead, Yonghou Jiang, Marc Carlson, Emily S Ford, Saumya Jani, John Houck, Xia Wu, Lichen Jing, Tiffany Pecor, Alisa Kachikis, Winnie Yeung, Tina Nguyen, Nana Minkah, Sasha E Larsen, Rhea N Coler, David M Koelle, and Whitney E Harrington
- Subjects
Article - Abstract
Human breastmilk is rich in T cells; however, their specificity and function are largely unknown. We compared the phenotype, diversity, and antigen specificity of T cells in the breastmilk and peripheral blood of lactating individuals who received SARS-CoV-2 mRNA vaccination. Relative to blood, breastmilk contained higher frequencies of T effector and central memory populations that expressed mucosal-homing markers. T cell receptor (TCR) sequence overlap was limited between blood and breastmilk. Overabundan t breastmilk clones were observed in all individuals, were diverse, and contained CDR3 sequences with known epitope specificity including to SARS-CoV-2 Spike. Spike-specific TCRs were more frequent in breastmilk compared to blood and expanded in breastmilk following a third mRNA vaccine dose. Our observations indicate that the lactating breast contains a distinct T cell population that can be modulated by maternal vaccination with potential implications for infant passive protection.One-Sentence SummaryThe breastmilk T cell repertoire is distinct and enriched for SARS-CoV-2 Spike-specificity after maternal mRNA vaccination.
- Published
- 2022
22. Transcriptional and functional analyses of neoantigen-specific CD4 T cells during a profound response to anti-PD-L1 in metastatic Merkel cell carcinoma
- Author
-
Candice Church, Thomas Pulliam, Natalie Longino, Song Y Park, Kimberly S Smythe, Vladimir Makarov, Nadeem Riaz, Lichen Jing, Robert Amezquita, Jean S Campbell, Raphael Gottardo, Robert H Pierce, Jaehyuk Choi, Timothy A Chan, David M Koelle, and Paul Nghiem
- Subjects
Pharmacology ,CD4-Positive T-Lymphocytes ,Cancer Research ,Skin Neoplasms ,Immunology ,Programmed Cell Death 1 Receptor ,Carcinoma, Merkel Cell ,Interferon-gamma ,Oncology ,Merkel cell polyomavirus ,Molecular Medicine ,Immunology and Allergy ,Humans ,Female ,Antigens, Viral, Tumor ,Transcription Factors - Abstract
BackgroundMerkel cell carcinoma (MCC) often responds to PD-1 pathway blockade, regardless of tumor-viral status (~80% of cases driven by the Merkel cell polyomavirus (MCPyV)). Prior studies have characterized tumor-specific T cell responses to MCPyV, which have typically been CD8, but little is known about the T cell response to UV-induced neoantigens.MethodsA patient in her mid-50s with virus-negative (VN) MCC developed large liver metastases after a brief initial response to chemotherapy. She received anti-PD-L1 (avelumab) and had a partial response within 4 weeks. Whole exome sequencing (WES) was performed to determine potential neoantigen peptides. Characterization of peripheral blood neoantigen T cell responses was evaluated via interferon-gamma (IFNγ) ELISpot, flow cytometry and single-cell RNA sequencing. Tumor-resident T cells were characterized by multiplexed immunohistochemistry.ResultsWES identified 1027 tumor-specific somatic mutations, similar to the published average of 1121 for VN-MCCs. Peptide prediction with a binding cut-off of ≤100 nM resulted in 77 peptides that were synthesized for T cell assays. Although peptides were predicted based on class I HLAs, we identified circulating CD4 T cells targeting 5 of 77 neoantigens. In contrast, no neoantigen-specific CD8 T cell responses were detected. Neoantigen-specific CD4 T cells were undetectable in blood before anti-PD-L1 therapy but became readily detectible shortly after starting therapy. T cells produced robust IFNγ when stimulated by neoantigen (mutant) peptides but not by the normal (wild-type) peptides. Single cell RNAseq showed neoantigen-reactive T cells expressed the Th1-associated transcription factor (T-bet) and associated cytokines. These CD4 T cells did not significantly exhibit cytotoxicity or non-Th1 markers. Within the pretreatment tumor, resident CD4 T cells were also Th1-skewed and expressed T-bet.ConclusionsWe identified and characterized tumor-specific Th1-skewed CD4 T cells targeting multiple neoantigens in a patient who experienced a profound and durable partial response to anti-PD-L1 therapy. To our knowledge, this is the first report of neoantigen-specific T cell responses in MCC. Although CD4 and CD8 T cells recognizing viral tumor antigens are often detectible in virus-positive MCC, only CD4 T cells recognizing neoantigens were detected in this patient. These findings suggest that CD4 T cells can play an important role in the response to anti-PD-(L)1 therapy.
- Published
- 2022
23. Prevalent and Diverse Intratumoral Oncoprotein-Specific CD8+ T Cells within Polyomavirus-Driven Merkel Cell Carcinomas
- Author
-
Aude G. Chapuis, Rima M. Kulikauskas, Aric Colunga, Hong Xie, Kelly G. Paulson, Paul Nghiem, Maclean M. Cook, Alexander L. Greninger, Olga K. Afanasiev, Jayasri G. Iyer, David M. Koelle, Lichen Jing, Mariliis Ott, Candice D. Church, Dafina Ibrani, and Shailender Bhatia
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,Skin Neoplasms ,Carcinogenesis ,Immunology ,Merkel cell polyomavirus ,chemical and pharmacologic phenomena ,Human leukocyte antigen ,CD8-Positive T-Lymphocytes ,Article ,Epitope ,Epitopes ,Young Adult ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Immune system ,Antigen ,medicine ,Humans ,Cytotoxic T cell ,Antigens, Viral, Tumor ,Aged ,biology ,Merkel cell carcinoma ,Middle Aged ,biology.organism_classification ,medicine.disease ,Carcinoma, Merkel Cell ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Immunotherapy ,Merkel cell - Abstract
Merkel cell carcinoma (MCC) is often caused by persistent expression of Merkel cell polyomavirus (MCPyV) T-antigen (T-Ag). These non-self proteins comprise about 400 amino acids (AA). Clinical responses to immune checkpoint inhibitors, seen in about half of patients, may relate to T-Ag–specific T cells. Strategies to increase CD8+ T-cell number, breadth, or function could augment checkpoint inhibition, but vaccines to augment immunity must avoid delivery of oncogenic T-antigen domains. We probed MCC tumor-infiltrating lymphocytes (TIL) with an artificial antigen-presenting cell (aAPC) system and confirmed T-Ag recognition with synthetic peptides, HLA-peptide tetramers, and dendritic cells (DC). TILs from 9 of 12 (75%) subjects contained CD8+ T cells recognizing 1–8 MCPyV epitopes per person. Analysis of 16 MCPyV CD8+ TIL epitopes and prior TIL data indicated that 97% of patients with MCPyV+ MCC had HLA alleles with the genetic potential that restrict CD8+ T-cell responses to MCPyV T-Ag. The LT AA 70–110 region was epitope rich, whereas the oncogenic domains of T-Ag were not commonly recognized. Specific recognition of T-Ag–expressing DCs was documented. Recovery of MCPyV oncoprotein–specific CD8+ TILs from most tumors indicated that antigen indifference was unlikely to be a major cause of checkpoint inhibition failure. The myriad of epitopes restricted by diverse HLA alleles indicates that vaccination can be a rational component of immunotherapy if tumor immune suppression can be overcome, and the oncogenic regions of T-Ag can be modified without impacting immunogenicity.
- Published
- 2020
24. Expansion of the HSV-2-specific T cell repertoire in skin after immunotherapeutic HSV-2 vaccine
- Author
-
Emily S. Ford, Alvason Li, Christine Johnston, Lichun Dong, Kurt Diem, Lichen Jing, Kerry J. Laing, Alexis Klock, Krithi Basu, Mariliis Ott, Jim Tartaglia, Sanjay Gurunathan, Jack L. Reid, Matyas Ecsedi, Aude G. Chapuis, Meei-Li Huang, Amalia S. Magaret, Jia Zhu, David M. Koelle, and Lawrence Corey
- Abstract
The skin at the site of HSV-2 reactivation is enriched for HSV-2 specific T cells. To evaluate whether an immunotherapeutic vaccine could elicit skin-based memory T cells we studied skin biopsies and HSV-2-reactive CD4+ T cells from peripheral blood mononuclear cells (PBMCs) by T-cell receptor (TCR) sequencing before and after vaccination with a replication-incompetent whole virus HSV-2 vaccine candidate (HSV529). The representation of HSV-2-reactive CD4+ T cell sequences from PBMCs increased from a median of 0.03% (range 0-0.09%) to 0.6% (range 0-1.3%) of the total skin TCR repertoire after the first vaccine dose. We found sustained expansion after vaccination in unique, skin-based T-cell clonotypes that were not detected in HSV-2-reactive CD4+ T cells isolated from PBMCs. While detection of skin clonotypes in the blood was related to abundance in the skin it was not related to expansion after vaccination. In one participant a switch in immunodominance was observed after vaccination with the emergence of a newly dominant TCRa/b pair in skin that was not detected in blood. We confirmed that the newly dominant clonotype was derived from an HSV-specific CD4+ T cell by creation of a synthetic TCR in a Jurkat-based cell line with a NR4A1-mNeonGreen reporter system. Our data indicate that the skin in areas of HSV-2 reactivation possesses an oligoclonal TCR repertoire that is distinct from the circulation with prominent clonotypes infrequently detected in the circulation by standard methods. Defining the influence of therapeutic vaccination on the HSV-2-specific TCR repertoire requires tissue-based evaluation.
- Published
- 2022
25. Viral Shedding 1 Year Following First-Episode Genital HSV-1 Infection
- Author
-
Christine, Johnston, Amalia, Magaret, Hyunju, Son, Michael, Stern, Molly, Rathbun, Daniel, Renner, Moriah, Szpara, Sarah, Gunby, Mariliis, Ott, Lichen, Jing, Victoria L, Campbell, Meei-Li, Huang, Stacy, Selke, Keith R, Jerome, David M, Koelle, and Anna, Wald
- Subjects
Adult ,Male ,Herpes Genitalis ,Herpesvirus 2, Human ,HIV Infections ,Herpes Simplex ,Herpesvirus 1, Human ,General Medicine ,Middle Aged ,Virus Shedding ,Pregnancy ,Humans ,Female ,Prospective Studies ,Genitalia - Abstract
ImportanceHerpes simplex virus type 1 (HSV-1) is the leading cause of first-episode genital herpes in many countries.ObjectiveTo inform counseling messages regarding genital HSV-1 transmission, oral and genital viral shedding patterns among persons with first-episode genital HSV-1 infection were assessed. The trajectory of the development of HSV-specific antibody and T-cell responses was also characterized.Design, Setting, and ParticipantsProspective cohort followed up for up to 2 years, with 82 participants followed up between 2013 and 2018. Participants were recruited from sexual health and primary care clinics in Seattle, Washington. Persons with laboratory-documented first-episode genital HSV-1 infection, without HIV infection or current pregnancy, were referred for enrollment.ExposuresFirst-episode genital HSV-1 infection.Main Outcomes and MeasuresGenital and oral HSV-1 shedding and lesion rates at 2 months, 11 months, and up to 2 years after initial genital HSV-1 infection. Participants self-collected oral and genital swabs for HSV polymerase chain reaction testing for 30 days at 2 and 11 months and up to 2 years after diagnosis of genital HSV-1. Blood samples were collected at serial time points to assess immune responses to HSV-1. Primary HSV-1 infection was defined as absent HSV antibody at baseline or evolving antibody profile using the University of Washington HSV Western Blot. HSV-specific T-cell responses were detected using interferon γ enzyme-linked immunospot.ResultsAmong the 82 participants, the median (range) age was 26 (16-64) years, 54 (65.9%) were women, and 42 (51.2%) had primary HSV-1 infection. At 2 months, HSV-1 was detected from the genital tract in 53 participants (64.6%) and in the mouth in 24 participants (29.3%). Genital HSV-1 shedding was detected on 275 of 2264 days (12.1%) at 2 months and declined significantly to 122 of 1719 days (7.1%) at 11 months (model-predicted rate, 6.2% [95% CI, 4.3%-8.9%] at 2 months vs 3.2% [95% CI, 1.8%-5.7%] at 11 months; relative risk, 0.52 [95% CI, 0.29-0.93]). Genital lesions were rare, reported on 65 of 2497 days (2.6%) at 2 months and 72 of 1872 days (3.8%) at 11 months. Oral HSV-1 shedding was detected on 88 of 2247 days (3.9%) at 2 months. Persons with primary HSV-1 infection had a higher risk of genital shedding compared with those with nonprimary infection (model-predicted rate, 7.9% [95% CI, 5.4%-11.7%] vs 2.9% [95% CI, 1.7%-5.0%]; relative risk, 2.75 [95% CI, 1.40-5.44]). Polyfunctional HSV-specific CD4+ and CD8+ T-cell responses were maintained during the follow-up period.Conclusions and RelevanceGenital HSV-1 shedding was frequent after first-episode genital HSV-1, particularly among those with primary infection, and declined rapidly during the first year after infection.
- Published
- 2022
26. Conservation of molecular and cellular phenotypes of invariant NKT cells between humans and non-human primates
- Author
-
Willie J. Swanson, Krystle K. Q. Yu, Charlotte A. James, Patricia A. Darrah, Malisa T. Smith, Chetan Seshadri, Mario Roederer, Joshua A Hackney, Kathryn E. Foulds, Lichen Jing, Damien B. Wilburn, Robert A. Seder, and David M. Koelle
- Subjects
Male ,Primates ,0301 basic medicine ,Immunology ,Receptors, Antigen, T-Cell ,CD1 ,chemical and pharmacologic phenomena ,Biology ,Major histocompatibility complex ,Jurkat cells ,Antigens, CD1 ,Evolution, Molecular ,Jurkat Cells ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Genetics ,Animals ,Humans ,Amino Acid Sequence ,Conserved Sequence ,iNKT cells ,T-cell receptor ,hemic and immune systems ,CD1D ,Natural killer T cell ,Macaca mulatta ,Non-human primate ,Cell biology ,Killer Cells, Natural ,Phenotype ,030104 developmental biology ,Cell culture ,biology.protein ,Female ,Original Article ,lipids (amino acids, peptides, and proteins) ,T cell receptor ,030215 immunology - Abstract
Invariant NKT (iNKT) cells in both humans and non-human primates are activated by the glycolipid antigen, α-galactosylceramide (α-GalCer). However, the extent to which the molecular mechanisms of antigen recognition and in vivo phenotypes of iNKT cells are conserved among primate species has not been determined. Using an evolutionary genetic approach, we found a lack of diversifying selection in CD1 genes over 45 million years of evolution, which stands in stark contrast to the history of the MHC system for presenting peptide antigens to T cells. The invariant T cell receptor (TCR)-α chain was strictly conserved across all seven primate clades. Invariant NKT cells from rhesus macaques (Macaca mulatta) bind human CD1D-α-GalCer tetramer and are activated by α-GalCer-loaded human CD1D transfectants. The dominant TCR-β chain cloned from a rhesus-derived iNKT cell line is nearly identical to that found in the human iNKT TCR, and transduction of the rhesus iNKT TCR into human Jurkat cells show that it is sufficient for binding human CD1D-α-GalCer tetramer. Finally, we used a 20-color flow cytometry panel to probe tissue phenotypes of iNKT cells in a cohort of rhesus macaques. We discovered several tissue-resident iNKT populations that have not been previously described in non-human primates but are known in humans, such as TCR-γδ iNKTs. These data reveal a diversity of iNKT cell phenotypes despite convergent evolution of the genes required for lipid antigen presentation and recognition in humans and non-human primates. Electronic supplementary material The online version of this article (10.1007/s00251-019-01118-9) contains supplementary material, which is available to authorized users.
- Published
- 2019
27. T cell receptor sequencing identifies prior SARS-CoV-2 infection and correlates with neutralizing antibodies and disease severity
- Author
-
Rebecca Elyanow, Thomas M. Snyder, Sudeb C. Dalai, Rachel M. Gittelman, Jim Boonyaratanakornkit, Anna Wald, Stacy Selke, Mark H. Wener, Chihiro Morishima, Alexander L. Greninger, Michael Gale, Tien-Ying Hsiang, Lichen Jing, Michael R. Holbrook, Ian M. Kaplan, H. Jabran Zahid, Damon H. May, Jonathan M. Carlson, Lance Baldo, Thomas Manley, Harlan S. Robins, and David M. Koelle
- Subjects
SARS-CoV-2 ,Receptors, Antigen, T-Cell ,COVID-19 ,Humans ,General Medicine ,Antibodies, Viral ,Antibodies, Neutralizing ,Severity of Illness Index ,United States ,Article - Abstract
Measuring the adaptive immune response to SARS-CoV-2 can enable the assessment of past infection as well as protective immunity and the risk of reinfection. While neutralizing antibody (nAb) titers are one measure of protection, such assays are challenging to perform at a large scale and the longevity of the SARS-CoV-2 nAb response is not fully understood. Here, we apply a T-cell receptor (TCR) sequencing assay that can be performed on a small volume standard blood sample to assess the adaptive T-cell response to SARS-CoV-2 infection. Samples were collected from a cohort of 302 individuals recovered from COVID-19 up to 6 months after infection. Previously published findings in this cohort showed that two commercially available SARS-CoV-2 serologic assays correlate well with nAb testing. We demonstrate that the magnitude of the SARS-CoV-2-specific T-cell response strongly correlates with nAb titer, as well as clinical indicators of disease severity including hospitalization, fever, or difficulty breathing. While the depth and breadth of the T-cell response declines during convalescence, the T-cell signal remains well above background with high sensitivity up to at least 6 months following initial infection. Compared to serology tests detecting binding antibodies to SARS-CoV-2 spike and nucleoprotein, the overall sensitivity of the TCR-based assay across the entire cohort and all timepoints was approximately 5% greater for identifying prior SARS-CoV-2 infection. Notably, the improved performance of T-cell testing compared to serology was most apparent in recovered individuals who were not hospitalized and were sampled beyond 150 days of their initial illness, suggesting that antibody testing may have reduced sensitivity in individuals who experienced less severe COVID-19 illness and at later timepoints. Finally, T-cell testing was able to identify SARS-CoV-2 infection in 68% (55/81) of convalescent samples having nAb titers below the lower limit of detection, as well as 37% (13/35) of samples testing negative by all three antibody assays. These results demonstrate the utility of a TCR-based assay as a scalable, reliable measure of past SARS-CoV-2 infection across a spectrum of disease severity. Additionally, the TCR repertoire may be useful as a surrogate for protective immunity with additive clinical value beyond serologic or nAb testing methods.
- Published
- 2021
28. Clinical, laboratory, and temporal predictors of neutralizing antibodies to SARS-CoV-2 after COVID-19
- Author
-
Susan L. Fink, Andrew Bryan, Michael R. Holbrook, Lichen Jing, David M. Koelle, Chihiro Morishima, Stacy Selke, Adrienne E Shapiro, Robin Gross, Steven Mazur, Terry Gernsheimer, Victoria L. Campbell, Christopher L. McClurkan, Jim Boonyaratanakornkit, Anna Wald, Mark H. Wener, Elena Postnikova, Alexander L. Greninger, Janie Liang, Sarah A. McGuffin, Danniel Zamora, Marie K. Das, Anu Chaudhary, and Keith R. Jerome
- Subjects
medicine.medical_specialty ,Convalescent plasma ,biology ,Coronavirus disease 2019 (COVID-19) ,Receiver operating characteristic ,business.industry ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Disease ,Gastroenterology ,Article ,Titer ,Immune system ,Internal medicine ,biology.protein ,Medicine ,Clinical Medicine ,Antibody ,business - Abstract
BACKGROUND: SARS-CoV-2–specific antibodies may protect from reinfection and disease, providing rationale for administration of plasma containing SARS-CoV-2–neutralizing antibodies (nAbs) as a treatment for COVID-19. Clinical factors and laboratory assays to streamline plasma donor selection, and the durability of nAb responses, are incompletely understood. METHODS: Potential convalescent plasma donors with virologically documented SARS-CoV-2 infection were tested for serum IgG against SARS-CoV-2 spike protein S1 domain and against nucleoprotein (NP), and for nAb. RESULTS: Among 250 consecutive persons, including 27 (11%) requiring hospitalization, who were studied a median of 67 days since symptom onset, 97% were seropositive on 1 or more assays. Sixty percent of donors had nAb titers ≥1:80. Correlates of higher nAb titers included older age (adjusted OR [AOR] 1.03 per year of age, 95% CI 1.00–1.06), male sex (AOR 2.08, 95% CI 1.13–3.82), fever during illness (AOR 2.73, 95% CI 1.25–5.97), and disease severity represented by hospitalization (AOR 6.59, 95% CI 1.32–32.96). Receiver operating characteristic analyses of anti-S1 and anti-NP antibody results yielded cutoffs that corresponded well with nAb titers, with the anti-S1 assay being slightly more predictive. nAb titers declined in 37 of 41 paired specimens collected a median of 98 days (range 77–120) apart (P < 0.001). Seven individuals (2.8%) were persistently seronegative and lacked T cell responses. CONCLUSION: nAb titers correlated with COVID-19 severity, age, and sex. SARS-CoV-2 IgG results can serve as useful surrogates for nAb testing. Functional nAb levels declined, and a small proportion of convalescent individuals lacked adaptive immune responses. FUNDING: The project was supported by the Frederick National Laboratory for Cancer Research with support from the NIAID under contract number 75N91019D00024, and was supported by the Fred Hutchinson Joel Meyers Endowment, Fast-Grants, a New Investigator award from the American Society for Transplantation and Cellular Therapy, and NIH contracts 75N93019C0063, 75N91019D00024, and HHSN272201800013C, and NIH grants T32-AI118690, T32-AI007044, K08-AI119142, and K23-AI140918.
- Published
- 2020
29. Clinical, laboratory, and temporal predictors of neutralizing antibodies against SARS-CoV-2 among COVID-19 convalescent plasma donor candidates
- Author
-
Michael R. Holbrook, Andrew Bryan, Christopher L. McClurkan, David M. Koelle, Anna Wald, Mark H. Wener, Lichen Jing, Janie Liang, Susan L. Fink, Stacy Selke, Chihiro Morishima, Elena Postnikova, Sarah A. McGuffin, Danniel Zamora, Adrienne E Shapiro, Marie K. Das, Anu Chaudhary, Alexander L. Greninger, Steven Mazur, Terry Gernsheimer, Vladimir V. Lukin, Jim Boonyaratanakornkit, Robin Gross, Victoria L. Campbell, and Keith R. Jerome
- Subjects
0301 basic medicine ,Adult ,Male ,medicine.medical_specialty ,T cell ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Blood Donors ,Disease ,Antibodies, Viral ,Immunoglobulin G ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Internal medicine ,Medicine ,Humans ,COVID-19 Serotherapy ,Aged ,Aged, 80 and over ,biology ,business.industry ,SARS-CoV-2 ,Immunization, Passive ,COVID-19 ,General Medicine ,Middle Aged ,Antibodies, Neutralizing ,Transplantation ,Titer ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,biology.protein ,Female ,Antibody ,business - Abstract
BACKGROUNDSARS-CoV-2-specific antibodies may protect from reinfection and disease, providing rationale for administration of plasma containing SARS-CoV-2-neutralizing antibodies (nAbs) as a treatment for COVID-19. Clinical factors and laboratory assays to streamline plasma donor selection, and the durability of nAb responses, are incompletely understood.METHODSPotential convalescent plasma donors with virologically documented SARS-CoV-2 infection were tested for serum IgG against SARS-CoV-2 spike protein S1 domain and against nucleoprotein (NP), and for nAb.RESULTSAmong 250 consecutive persons, including 27 (11%) requiring hospitalization, who were studied a median of 67 days since symptom onset, 97% were seropositive on 1 or more assays. Sixty percent of donors had nAb titers ≥1:80. Correlates of higher nAb titers included older age (adjusted OR [AOR] 1.03 per year of age, 95% CI 1.00-1.06), male sex (AOR 2.08, 95% CI 1.13-3.82), fever during illness (AOR 2.73, 95% CI 1.25-5.97), and disease severity represented by hospitalization (AOR 6.59, 95% CI 1.32-32.96). Receiver operating characteristic analyses of anti-S1 and anti-NP antibody results yielded cutoffs that corresponded well with nAb titers, with the anti-S1 assay being slightly more predictive. nAb titers declined in 37 of 41 paired specimens collected a median of 98 days (range 77-120) apart (P < 0.001). Seven individuals (2.8%) were persistently seronegative and lacked T cell responses.CONCLUSIONnAb titers correlated with COVID-19 severity, age, and sex. SARS-CoV-2 IgG results can serve as useful surrogates for nAb testing. Functional nAb levels declined, and a small proportion of convalescent individuals lacked adaptive immune responses.FUNDINGThe project was supported by the Frederick National Laboratory for Cancer Research with support from the NIAID under contract number 75N91019D00024, and was supported by the Fred Hutchinson Joel Meyers Endowment, Fast-Grants, a New Investigator award from the American Society for Transplantation and Cellular Therapy, and NIH contracts 75N93019C0063, 75N91019D00024, and HHSN272201800013C, and NIH grants T32-AI118690, T32-AI007044, K08-AI119142, and K23-AI140918.
- Published
- 2020
30. Mechanisms of Endogenous HIV-1 Reactivation by Endocervical Epithelial Cells
- Author
-
Gretchen M. Lentz, Jessica Wagoner, Michael F. Fialkow, Germán G. Gornalusse, Stephen J. Polyak, M. Juliana McElrath, Florian Hladik, Anna C Kirby, Gabriella Fenkart, David M. Koelle, David N. Fredricks, Fernanda Calienes, Sean M. Hughes, Claire N. Levy, Anna Wald, Erin J. dela Cruz, Lamar M. Fleming, Urvashi Pandey, Lucia Vojtech, Rogelio Valdez, and Lichen Jing
- Subjects
CD4-Positive T-Lymphocytes ,Gene Expression Regulation, Viral ,medicine.medical_treatment ,Primary Cell Culture ,Immunology ,Acyclovir ,HIV Infections ,Viremia ,Cervix Uteri ,Biology ,Virus Replication ,medicine.disease_cause ,Antiviral Agents ,Microbiology ,Virus ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Virology ,HIV Seropositivity ,medicine ,Humans ,030304 developmental biology ,0303 health sciences ,Innate immune system ,Epithelial Cells ,Provirus ,medicine.disease ,Virus Latency ,Immunosurveillance ,CCL20 ,Cytokine ,Herpes simplex virus ,Anti-Retroviral Agents ,Insect Science ,HIV-1 ,Pathogenesis and Immunity ,Female ,Virus Activation ,030217 neurology & neurosurgery - Abstract
Pharmacological HIV-1 reactivation to reverse latent infection has been extensively studied. However, HIV-1 reactivation also occurs naturally, as evidenced by occasional low-level viremia (“viral blips”) during antiretroviral treatment (ART). Clarifying where blips originate from and how they happen could provide clues to stimulate latency reversal more effectively and safely or to prevent viral rebound following ART cessation. We studied HIV-1 reactivation in the female genital tract, a dynamic anatomical target for HIV-1 infection throughout all disease stages. We found that primary endocervical epithelial cells from several women reactivated HIV-1 from latently infected T cells. The endocervical cells’ HIV-1 reactivation capacity further increased upon Toll-like receptor 3 stimulation with poly(I·C) double-stranded RNA or infection with herpes simplex virus 2 (HSV-2). Notably, acyclovir did not eliminate HSV-2-induced HIV-1 reactivation. While endocervical epithelial cells secreted large amounts of several cytokines and chemokines, especially tumor necrosis factor alpha (TNF-α), CCL3, CCL4, and CCL20, their HIV-1 reactivation capacity was almost completely blocked by TNF-α neutralization alone. Thus, immunosurveillance activities by columnar epithelial cells in the endocervix can cause endogenous HIV-1 reactivation, which may contribute to viral blips during ART or rebound following ART interruption. IMPORTANCE A reason that there is no universal cure for HIV-1 is that the virus can hide in the genome of infected cells in the form of latent proviral DNA. This hidden provirus is protected from antiviral drugs until it eventually reactivates to produce new virions. It is not well understood where in the body or how this reactivation occurs. We studied HIV-1 reactivation in the female genital tract, which is often the portal of HIV-1 entry and which remains a site of infection throughout the disease. We found that the columnar epithelial cells lining the endocervix, the lower part of the uterus, are particularly effective in reactivating HIV-1 from infected T cells. This activity was enhanced by certain microbial stimuli, including herpes simplex virus 2, and blocked by antibodies against the inflammatory cytokine TNF-α. Avoiding HIV-1 reactivation could be important for maintaining a functional HIV-1 cure when antiviral therapy is stopped.
- Published
- 2020
31. Viral Genetics Modulate Orolabial Herpes Simplex Virus Type 1 Shedding in Humans
- Author
-
Anqi Cheng, Anna Wald, David M. Koelle, Kerry J. Laing, Meei-Li Huang, Lichen Jing, Hong Xie, Ronnie M. Russell, Stacy Selke, Amalia Magaret, Eric Strachan, Tran Tran, Pavitra Roychoudhury, Alexander L. Greninger, Meena Ramchandani, and Keith R. Jerome
- Subjects
Adult ,CD4-Positive T-Lymphocytes ,Male ,0301 basic medicine ,Host immunity ,Genotype ,viruses ,Herpesvirus 1, Human ,Biology ,medicine.disease_cause ,Positive correlation ,Major Articles and Brief Reports ,Open Reading Frames ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Viral genetics ,Twins, Dizygotic ,medicine ,Humans ,Immunology and Allergy ,030212 general & internal medicine ,Viral shedding ,Phylogeny ,Aged ,Mouth ,Twins, Monozygotic ,Middle Aged ,Virology ,Virus Shedding ,Open reading frame ,030104 developmental biology ,Infectious Diseases ,Herpes simplex virus ,Female ,Herpes Labialis ,Viral genotype - Abstract
BACKGROUND: Orolabial herpes simplex virus type 1 (HSV-1) infection has a wide spectrum of severity in immunocompetent persons. To study the role of viral genotype and host immunity, we characterized oral HSV-1 shedding rates and host cellular response, and genotyped viral strains, in monozygotic (MZ) and dizygotic (DZ) twins. METHODS: A total of 29 MZ and 22 DZ HSV-1–seropositive twin pairs were evaluated for oral HSV-1 shedding for 60 days. HSV-1 strains from twins were genotyped as identical or different. CD4(+) T-cell responses to HSV-1 proteins were studied. RESULTS: The median per person oral HSV shedding rate was 9% of days that a swab was obtained (mean, 10.2% of days). A positive correlation between shedding rates was observed within all twin pairs, and in the MZ and DZ twins. In twin subsets with sufficient HSV-1 DNA to genotype, 15 had the same strain and 14 had different strains. Viral shedding rates were correlated for those with the same but not different strains. The median number of HSV-1 open reading frames recognized per person was 16. The agreement in the CD4(+) T-cell response to specific HSV-1 open reading frames was greater between MZ twins than between unrelated persons (P = .002). CONCLUSION: Viral strain characteristics likely contribute to oral HSV-1 shedding rates.
- Published
- 2018
32. T-cell Responses to HSV-1 in Persons Who Have Survived Childhood Herpes Simplex Encephalitis
- Author
-
Lazaro Lorenzo, Lichen Jing, Mariliis Ott, Jean-Laurent Casanova, Shen-Ying Zhang, and David M. Koelle
- Subjects
CD4-Positive T-Lymphocytes ,0301 basic medicine ,Microbiology (medical) ,Enzyme-Linked Immunospot Assay ,Adolescent ,viruses ,T cell ,Mucocutaneous zone ,Herpesvirus 1, Human ,Human leukocyte antigen ,CD8-Positive T-Lymphocytes ,Article ,Virus ,Cohort Studies ,Epitopes ,Viral Proteins ,03 medical and health sciences ,Blood serum ,Immunity ,medicine ,Humans ,Child ,business.industry ,Infant ,medicine.disease ,Virology ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,Child, Preschool ,Pediatrics, Perinatology and Child Health ,Encephalitis, Herpes Simplex ,business ,Encephalitis ,CD8 - Abstract
Background Herpes simplex encephalitis (HSE) after primary herpes simplex virus (HSV)-1 infection can occur in children due to inborn errors of cell-intrinsic immunity in the central nervous system. Paradoxically, symptomatic mucocutaneous HSV-1 recurrences are rare survivors of childhood HSE. T-cell-acquired immunity is thought to be involved in control of recurrent mucocutaneous HSV infection. We thus tested HSV-1-specific immunity in HSE survivors. Methods We obtained serum and peripheral blood mononuclear cells (PBMCs) from participants a median of 13.5 years after HSE. HSV-1 and HSV-2 IgG was detected by type-specific immunoblot. PBMCs from subjects passing quality control criteria were tested using enzyme-linked immunospot assay for CD4 interferon-γ responses with an HSV-1 lysate and for CD8 responses using pooled synthetic HSV-1 peptide CD8 T-cell epitopes. Healthy adult PBMCs were used to standardize assays and as comparators. Results All participants were HSV-1 seropositive. Most (23/24) HSE survivors had human leukocyte antigen class I types matching the human leukocyte antigen restriction of the pooled peptides. We detected HSV-specific CD8 T-cell responses in 14 of 24 (58%) HSE survivors and in 9 of 9 healthy HSV-1 seropositive adults. HSV-specific CD4 T-cell responses were present in all 5 HSE subjects tested and in 8 of 9 healthy adults. Response magnitudes were overlapping between subject groups. Conclusions The defects in cell-intrinsic immunity leading to failure to control primary central nervous system HSV-1 infection do not preclude the acquisition of specific immunity or the control of recurrent mucocutaneous HSV infections. The rarity and lack of severe or recurrent mucocutaneous HSV infection in survivors of childhood HSE corresponds with intact adaptive T-cell immunity.
- Published
- 2017
33. Genome-Wide Approach to the CD4 T-Cell Response to Human Herpesvirus 6B
- Author
-
Olga Tsvetkova, Derek J. Hanson, Alexander L. Greninger, Guilhem F. Rerolle, Lichen Jing, Allesandro Sette, Victoria L. Campbell, and David M. Koelle
- Subjects
CD4-Positive T-Lymphocytes ,Herpesvirus 6, Human ,viruses ,medicine.medical_treatment ,Immunology ,Population ,Epitopes, T-Lymphocyte ,Roseolovirus Infections ,Biology ,medicine.disease_cause ,Microbiology ,Epitope ,Cell Line ,Open Reading Frames ,03 medical and health sciences ,Immune system ,Antigen ,Virology ,medicine ,Humans ,education ,Antigens, Viral ,030304 developmental biology ,0303 health sciences ,education.field_of_study ,030306 microbiology ,virus diseases ,Cytomegalovirus ,Immunotherapy ,Acquired immune system ,biology.organism_classification ,Insect Science ,Pathogenesis and Immunity ,Human herpesvirus 6 ,Epitope Mapping ,Genome-Wide Association Study - Abstract
Human herpesvirus 6 (HHV-6) and cytomegalovirus (CMV) are population-prevalent betaherpesviruses with intermittent lytic replication that can be pathogenic in immunocompromised hosts. Elucidation of the adaptive immune response is valuable for understanding pathogenesis and designing novel treatments. Knowledge of T-cell antigens has reached the genome-wide level for CMV and other human herpesviruses, but study of HHV-6 is at an earlier stage. Using rare-cell enrichment combined with an HLA-agnostic, proteome-wide approach, we queried HHV-6B-specific CD4 T cells from 18 healthy donors with each known HHV-6B protein. We detected a low abundance of HHV-6-specific CD4 T cells in blood; however, the within-person CD4 T-cell response is quite broad: the median number of open reading frame (ORF) products recognized was nine per person. Overall, the data expand the number of documented HHV-6B CD4 T-cell antigens from approximately 11 to 60. Epitopes in the proteins encoded by U14, U90, and U95 were mapped with synthetic peptides, and HLA restriction was defined for some responses. Intriguingly, CD4 T-cell antigens newly described in this report are among the most population prevalent, including U73, U72, U95, and U30. Our results indicate that selection of HHV-6B ORFs for immunotherapy should consider this expanded panel of HHV-6B antigens. IMPORTANCE Human herpesvirus 6 is highly prevalent and maintains chronic infection in immunocompetent individuals, with the potential to replicate widely in settings of immunosuppression, leading to clinical disease. Antiviral compounds may be ineffective and/or pose dose-limiting toxicity, and therefore, immune-based therapies have garnered increased interest in recent years. Attempts at addressing this unmet medical need begin with understanding the cellular response to HHV-6 at the individual and population levels. The present study provides a comprehensive assessment of HHV-6-specific T-cell responses that may inform the development of cell-based therapies directed at this virus.
- Published
- 2019
34. Varicella zoster virus productively infects human peripheral blood mononuclear cells to modulate expression of immunoinhibitory proteins and blocking PD-L1 enhances virus-specific CD8+ T cell effector function
- Author
-
Maria A. Nagel, Anna M. Blackmon, David M. Koelle, Brent E. Palmer, Christina N. Como, Owen Krueger, Charles Preston Neff, Dallas Jones, Andrew N. Bubak, and Lichen Jing
- Subjects
CD4-Positive T-Lymphocytes ,Herpesvirus 3, Human ,B Cells ,viruses ,Programmed Cell Death 1 Receptor ,NK cells ,CD8-Positive T-Lymphocytes ,medicine.disease_cause ,Antibodies, Viral ,Biochemistry ,B7-H1 Antigen ,Monocytes ,White Blood Cells ,Spectrum Analysis Techniques ,Animal Cells ,Medicine and Health Sciences ,Cytotoxic T cell ,CTLA-4 Antigen ,Biology (General) ,Hepatitis A Virus Cellular Receptor 2 ,0303 health sciences ,Immune System Proteins ,biology ,T Cells ,030302 biochemistry & molecular biology ,virus diseases ,Natural killer T cell ,Flow Cytometry ,Lymphocyte Activation Gene 3 Protein ,3. Good health ,medicine.anatomical_structure ,Virus Diseases ,Spectrophotometry ,Cytophotometry ,Antibody ,Cellular Types ,Research Article ,QH301-705.5 ,T cell ,Immune Cells ,Immunology ,Cytotoxic T cells ,Research and Analysis Methods ,Microbiology ,Herpes Zoster ,03 medical and health sciences ,Immune system ,Antigen ,Antigens, CD ,Virology ,Genetics ,medicine ,Humans ,Antibody-Producing Cells ,Molecular Biology ,030304 developmental biology ,Blood Cells ,Varicella zoster virus ,Biology and Life Sciences ,Proteins ,Cell Biology ,RC581-607 ,Programmed Cell Death 1 Ligand 2 Protein ,Coculture Techniques ,Gene Expression Regulation ,biology.protein ,Leukocytes, Mononuclear ,Parasitology ,Immunologic diseases. Allergy ,CD8 - Abstract
Varicella zoster virus (VZV) is a lymphotropic alpha-herpesvirinae subfamily member that produces varicella on primary infection and causes zoster, vascular disease and vision loss upon reactivation from latency. VZV-infected peripheral blood mononuclear cells (PBMCs) disseminate virus to distal organs to produce clinical disease. To assess immune evasion strategies elicited by VZV that may contribute to dissemination of infection, human PBMCs and VZV-specific CD8+ T cells (V-CD8+) were mock- or VZV-infected and analyzed for immunoinhibitory protein PD-1, PD-L1, PD-L2, CTLA-4, LAG-3 and TIM-3 expression using flow cytometry. All VZV-infected PBMCs (monocytes, NK, NKT, B cells, CD4+ and CD8+ T cells) and V-CD8+ showed significant elevations in PD-L1 expression compared to uninfected cells. VZV induced PD-L2 expression in B cells and V-CD8+. Only VZV-infected CD8+ T cells, NKT cells and V-CD8+ upregulated PD-1 expression, the immunoinhibitory receptor for PD-L1/PD-L2. VZV induced CTLA-4 expression only in V-CD8+ and no significant changes in LAG-3 or TIM-3 expression were observed in V-CD8+ or PBMC T cells. To test whether PD-L1, PD-L2 or CTLA-4 regulates V-CD8+ effector function, autologous PBMCs were VZV-infected and co-cultured with V-CD8+ cells in the presence of blocking antibodies against PD-L1, PD-L2 or CTLA-4; ELISAs revealed significant elevations in IFNγ only upon blocking of PD-L1. Together, these results identified additional immune cells that are permissive to VZV infection (monocytes, B cells and NKT cells); along with a novel mechanism for inhibiting CD8+ T cell effector function through induction of PD-L1 expression., Author summary The burden of disease produced by VZV is significant, since 90% of the world population harbors latent virus. At least 50% of infected individuals will reactivate by 85 years of age to develop zoster, which is an established risk factor for stroke and myocardial infarction, as well as multisystem diseases with or without rash. VZV-infected PBMCs disseminate virus to distal organs to produce clinical disease. PD-L1 is an immunoinhibitory protein that interacts with PD-1, its receptor, expressed mainly on T cells to prevent their activation and subsequent clearance of virus- or malignantly transformed cells. We show here that all peripheral blood mononuclear cells have a dramatic induction of PD-L1 expression upon infection with VZV, which is combined with the induction of PD-1 expression on CD8+ T cells, NKT cells and VZV-specific CD8+ T cells. Blocking PD-L1 during co-culturing of VZV-specific CD8+ T cells with autologous VZV-infected PBMCs enhanced IFNγ levels almost 2-fold compared to isotype controls. These results indicate that blocking PD-L1 expression during varicella or zoster may restore CD8+ T cell effector function, enabling effective clearance of virus-infected cells to reduce viral spread and multisystem disease.
- Published
- 2019
35. A Novel Approach of Identifying Immunodominant Self and Viral Antigen Cross-Reactive T Cells and Defining the Epitopes They Recognize
- Author
-
John A. Gebe, William W. Kwok, David M. Koelle, Junbao Yang, Eddie A. James, and Lichen Jing
- Subjects
0301 basic medicine ,CD4-Positive T-Lymphocytes ,lcsh:Immunologic diseases. Allergy ,Glutamate decarboxylase ,Immunology ,Epitopes, T-Lymphocyte ,autoreactive T cells ,T cell cross-reactivity ,Viral antigen ,Biology ,CD38 ,Cross Reactions ,medicine.disease_cause ,Epitope ,03 medical and health sciences ,0302 clinical medicine ,Influenza, Human ,medicine ,Methods ,Humans ,Immunology and Allergy ,Amino Acid Sequence ,molecular mimicry ,glutamate decarboxylase 65 ,Antigens, Viral ,chemistry.chemical_classification ,Viral matrix protein ,Glutamate Decarboxylase ,Immunodominant Epitopes ,HLA-DR Antigens ,Virology ,ADP-ribosyl Cyclase 1 ,3. Good health ,Amino acid ,Vaccination ,Molecular mimicry ,030104 developmental biology ,Diabetes Mellitus, Type 1 ,chemistry ,Influenza A virus ,Influenza Vaccines ,Peptides ,influenza ,lcsh:RC581-607 ,030215 immunology - Abstract
Infection and vaccination can lead to activation of autoreactive T cells, including the activation of cross-reactive T cells. However, detecting these cross-reactive T cells and identifying the non-self and self-antigen epitopes is difficult. The current study demonstrates the utility of a novel approach that effectively accomplishes both. We utilized surface expression of CD38 on newly activated CD4 memory T cells as a strategy to identify type 1 diabetes associated autoreactive T cells activated by influenza vaccination in healthy subjects. We identified an influenza A matrix protein (MP) specific CD4+ T cell clone that cross-recognizes an immunodominant epitope from Glutamic Acid Decarboxylase 65 (GAD65) protein. The sequences of the MP and GAD65 peptides are rather distinct, with only 2 identical amino acids within the HLA-DR binding region. This result suggests that activation of autoreactive T cells by microbial infection under certain physiological conditions can occur amongst peptides with minimum amino acid sequence homology. This novel strategy also provides a new research pathway in which to examine activation of autoreactive CD4+ T cells after vaccination or natural infection.
- Published
- 2018
36. Extensive CD4 and CD8 T Cell Cross-Reactivity between Alphaherpesviruses
- Author
-
Simon Mallal, Christine M. Posavad, Anna Wald, David M. Koelle, Lichen Jing, Ronnie M. Russell, Russell S. Barlow, Katie D. White, Meena Ramchandani, Kerry J. Laing, Christine Johnston, Elizabeth J. Phillips, Søren Buus, Lichun Dong, Alec J. Redwood, and Juergen Haas
- Subjects
0301 basic medicine ,integumentary system ,viruses ,Immunology ,Antigen presentation ,T-cell receptor ,Varicella zoster virus ,virus diseases ,Biology ,medicine.disease_cause ,Virology ,Epitope ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Antigen ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,CD8 ,030215 immunology - Abstract
The Alphaherpesvirinae subfamily includes HSV types 1 and 2 and the sequence-divergent pathogen varicella zoster virus (VZV). T cells, controlled by TCR and HLA molecules that tolerate limited epitope amino acid variation, might cross-react between these microbes. We show that memory PBMC expansion with either HSV or VZV enriches for CD4 T cell lines that recognize the other agent at the whole-virus, protein, and peptide levels, consistent with bidirectional cross-reactivity. HSV-specific CD4 T cells recovered from HSV-seronegative persons can be explained, in part, by such VZV cross-reactivity. HSV-1–reactive CD8 T cells also cross-react with VZV-infected cells, full-length VZV proteins, and VZV peptides, as well as kill VZV-infected dermal fibroblasts. Mono- and cross-reactive CD8 T cells use distinct TCRB CDR3 sequences. Cross-reactivity to VZV is reconstituted by cloning and expressing TCRA/TCRB receptors from T cells that are initially isolated using HSV reagents. Overall, we define 13 novel CD4 and CD8 HSV–VZV cross-reactive epitopes and strongly imply additional cross-reactive peptide sets. Viral proteins can harbor both CD4 and CD8 HSV/VZV cross-reactive epitopes. Quantitative estimates of HSV/VZV cross-reactivity for both CD4 and CD8 T cells vary from 10 to 50%. Based on these findings, we hypothesize that host herpesvirus immune history may influence the pathogenesis and clinical outcome of subsequent infections or vaccinations for related pathogens and that cross-reactive epitopes and TCRs may be useful for multi-alphaherpesvirus vaccine design and adoptive cellular therapy.
- Published
- 2016
37. Genome Sequencing and Analysis of Geographically Diverse Clinical Isolates of Herpes Simplex Virus 2
- Author
-
Robert C. Colgrove, Fernando Diaz, Susanna L. Lamers, Stuart C. Ray, Brian Weiner, Thomas C. Quinn, Ruchi M. Newman, David M. Koelle, Sakina Saif, Jeffrey I. Cohen, Kening Wang, Sarah Young, Aaron A.R. Tobian, Oliver Laeyendecker, Matthew Henn, Lichen Jing, and David M. Knipe
- Subjects
Recombination, Genetic ,Genetics ,Geography ,Transmission (medicine) ,Herpesvirus 2, Human ,viruses ,Immunology ,Genetic Variation ,Genome, Viral ,Biology ,medicine.disease_cause ,Microbiology ,Genome ,DNA sequencing ,genomic DNA ,Herpes simplex virus ,Genetic Diversity and Evolution ,Virology ,Insect Science ,Genetic variation ,medicine ,Humans ,Genetic variability ,Reference genome - Abstract
Herpes simplex virus 2 (HSV-2), the principal causative agent of recurrent genital herpes, is a highly prevalent viral infection worldwide. Limited information is available on the amount of genomic DNA variation between HSV-2 strains because only two genomes have been determined, the HG52 laboratory strain and the newly sequenced SD90e low-passage-number clinical isolate strain, each from a different geographical area. In this study, we report the nearly complete genome sequences of 34 HSV-2 low-passage-number and laboratory strains, 14 of which were collected in Uganda, 1 in South Africa, 11 in the United States, and 8 in Japan. Our analyses of these genomes demonstrated remarkable sequence conservation, regardless of geographic origin, with the maximum nucleotide divergence between strains being 0.4% across the genome. In contrast, prior studies indicated that HSV-1 genomes exhibit more sequence diversity, as well as geographical clustering. Additionally, unlike HSV-1, little viral recombination between HSV-2 strains could be substantiated. These results are interpreted in light of HSV-2 evolution, epidemiology, and pathogenesis. Finally, the newly generated sequences more closely resemble the low-passage-number SD90e than HG52, supporting the use of the former as the new reference genome of HSV-2. IMPORTANCE Herpes simplex virus 2 (HSV-2) is a causative agent of genital and neonatal herpes. Therefore, knowledge of its DNA genome and genetic variability is central to preventing and treating genital herpes. However, only two full-length HSV-2 genomes have been reported. In this study, we sequenced 34 additional HSV-2 low-passage-number and laboratory viral genomes and initiated analysis of the genetic diversity of HSV-2 strains from around the world. The analysis of these genomes will facilitate research aimed at vaccine development, diagnosis, and the evaluation of clinical manifestations and transmission of HSV-2. This information will also contribute to our understanding of HSV evolution.
- Published
- 2015
38. Selective Expression of CCR10 and CXCR3 by Circulating Human Herpes Simplex Virus-Specific CD8 T Cells
- Author
-
Amalia Magaret, Kerry J. Laing, Anqi Cheng, M. T. Hensel, Anna Wald, Tao Peng, Lichun Dong, Stephen C. De Rosa, Lichen Jing, and David M. Koelle
- Subjects
0301 basic medicine ,Antigens, Differentiation, T-Lymphocyte ,Male ,Chemokine ,Herpesvirus 4, Human ,Receptors, CXCR3 ,Lymphocyte ,Herpesvirus 2, Human ,Immunology ,Cytomegalovirus ,Receptors, CCR10 ,CD8-Positive T-Lymphocytes ,CXCR3 ,Lymphocyte Activation ,Microbiology ,03 medical and health sciences ,Chemokine receptor ,Antigen ,Virology ,medicine ,Cytotoxic T cell ,Humans ,RNA, Messenger ,Skin ,Membrane Glycoproteins ,biology ,Chemokine CCL27 ,Herpes Simplex ,Flow Cytometry ,Molecular biology ,030104 developmental biology ,medicine.anatomical_structure ,Insect Science ,biology.protein ,Pathogenesis and Immunity ,CCL27 ,Female ,Immunologic Memory ,Homing (hematopoietic) - Abstract
Herpes simplex virus (HSV) infection is restricted to epithelial cells and neurons and is controlled by CD8 T cells. These cells both traffic to epithelial sites of recurrent lytic infection and to ganglia and persist at the dermal-epidermal junction for up to 12 weeks after lesion resolution. We previously showed that cutaneous lymphocyte-associated antigen (CLA), a functional E-selectin ligand (ESL), is selectively expressed on circulating HSV-2-specific CD8 T cells. CLA/ESL mediates adhesion of T cells to inflamed vascular endothelium. Later stages in T-cell homing involve chemokines (Ch) and lymphocyte chemokine receptors (ChR) for vascular wall arrest and diapedesis. Several candidate ChR have been implicated in skin homing. We measured cell surface ChR on HSV-specific human peripheral blood CD8 T cells and extended our studies to HSV-1. We observed preferential cell surface expression of CCR10 and CXCR3 by HSV-specific CD8 T cells compared to CD8 T cells specific for control viruses, Epstein-Barr virus (EBV) and cytomegalovirus (CMV), and compared to bulk memory CD8 T cells. CXCR3 ligand mRNA levels were selectively increased in skin biopsy specimens from persons with recurrent HSV-2, while the mRNA levels of the CCR10 ligand CCL27 were equivalent in lesion and control skin. Our data are consistent with a model in which CCL27 drives baseline recruitment of HSV-specific CD8 T cells expressing CCR10, while interferon-responsive CXCR3 ligands recruit additional cells in response to virus-driven inflammation. IMPORTANCE HSV-2 causes very localized recurrent infections in the skin and genital mucosa. Virus-specific CD8 T cells home to the site of recurrent infection and participate in viral clearance. The exit of T cells from the blood involves the use of chemokine receptors on the T-cell surface and chemokines that are present in infected tissue. In this study, circulating HSV-2-specific CD8 T cells were identified using specific fluorescent tetramer reagents, and their expression of several candidate skin-homing-associated chemokine receptors was measured using flow cytometry. We found that two chemokine receptors, CXCR3 and CCR10, are upregulated on HSV-specific CD8 T cells in blood. The chemokines corresponding to these receptors are also expressed in infected tissues. Vaccine strategies to prime CD8 T cells to home to HSV lesions should elicit these chemokine receptors if possible to increase the homing of vaccine-primed cells to sites of infection.
- Published
- 2017
39. Worldwide circulation of HSV-2 × HSV-1 recombinant strains
- Author
-
Lichen Jing, Christine Johnston, Stacy Selke, Hong Xie, Meei-Li Huang, David M. Koelle, Amalia Magaret, Matthew Fitzgibbon, Anna Wald, Connie Celum, Peter Norberg, Ronnie M. Russell, Jairam R. Lingappa, Kurt Diem, Larry Stensland, Keith R. Jerome, and Alexander L. Greninger
- Subjects
0301 basic medicine ,Male ,Simplexvirus ,food.ingredient ,Genotype ,viruses ,Herpesvirus 2, Human ,Herpesvirus 1, Human ,Biology ,medicine.disease_cause ,Article ,law.invention ,03 medical and health sciences ,chemistry.chemical_compound ,food ,law ,Phylogenetics ,medicine ,Humans ,Gene ,Phylogeny ,Recombination, Genetic ,Multidisciplinary ,Herpes Simplex ,South America ,Virology ,3. Good health ,030104 developmental biology ,Herpes simplex virus ,chemistry ,Homo sapiens ,Africa ,North America ,Recombinant DNA ,Female ,DNA - Abstract
Homo sapiens harbor two distinct, medically significant species of simplexviruses, herpes simplex virus (HSV)-1 and HSV-2, with estimated divergence 6–8 million years ago (MYA). Unexpectedly, we found that circulating HSV-2 strains can contain HSV-1 DNA segments in three distinct genes. Using over 150 genital swabs from North and South America and Africa, we detected recombinants worldwide. Common, widely distributed gene UL39 genotypes are parsimoniously explained by an initial >457 basepair (bp) HSV-1 × HSV-2 crossover followed by back-recombination to HSV-2. Blocks of >244 and >539 bp of HSV-1 DNA within genes UL29 and UL30, respectively, have reached near fixation, with a minority of strains retaining sequences we posit as ancestral HSV-2. Our data add to previous in vitro and animal work, implying that in vivo cellular co-infection with HSV-1 and HSV-2 yields viable interspecies recombinants in the natural human host.
- Published
- 2017
40. Tumor-infiltrating Merkel cell polyomavirus-specific T cells are diverse and associated with improved patient survival
- Author
-
K. Lachance, Paul Nghiem, David M. Koelle, Matthew Fitzgibbon, Gerald Willimsky, Martin W. McIntosh, Candice D. Church, Natalie J. Miller, Ioannis Gavvovidis, Thomas Blankenstein, Michi M. Shinohara, Lichen Jing, David A. Crispin, and Lichun Dong
- Subjects
0301 basic medicine ,Cancer Research ,Skin Neoplasms ,Receptors, Antigen, T-Cell, alpha-beta ,Immunology ,Merkel cell polyomavirus ,Epitopes, T-Lymphocyte ,chemical and pharmacologic phenomena ,T-Cell Antigen Receptor Specificity ,CD8-Positive T-Lymphocytes ,Peripheral blood mononuclear cell ,Epitope ,Article ,Flow cytometry ,Clonal Evolution ,03 medical and health sciences ,0302 clinical medicine ,Lymphocytes, Tumor-Infiltrating ,T-Lymphocyte Subsets ,medicine ,Humans ,Avidity ,biology ,medicine.diagnostic_test ,Merkel cell carcinoma ,T-cell receptor ,food and beverages ,Genetic Variation ,hemic and immune systems ,Sequence Analysis, DNA ,biology.organism_classification ,medicine.disease ,Prognosis ,Carcinoma, Merkel Cell ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,CD8 - Abstract
Tumor-infiltrating CD8+ T cells are associated with improved survival of patients with Merkel cell carcinoma (MCC), an aggressive skin cancer causally linked to Merkel cell polyomavirus (MCPyV). However, CD8+ T-cell infiltration is robust in only 4% to 18% of MCC tumors. We characterized the T-cell receptor (TCR) repertoire restricted to one prominent epitope of MCPyV (KLLEIAPNC, “KLL”) and assessed whether TCR diversity, tumor infiltration, or T-cell avidity correlated with clinical outcome. HLA-A*02:01/KLL tetramer+ CD8+ T cells from MCC patient peripheral blood mononuclear cells (PBMC) and tumor-infiltrating lymphocytes (TIL) were isolated via flow cytometry. TCRβ (TRB) sequencing was performed on tetramer+ cells from PBMCs or TILs (n = 14) and matched tumors (n = 12). Functional avidity of T-cell clones was determined by IFNγ production. We identified KLL tetramer+ T cells in 14% of PBMC and 21% of TIL from MCC patients. TRB repertoires were strikingly diverse (397 unique TRBs were identified from 12 patients) and mostly private (only one TCRb clonotype shared between two patients). An increased fraction of KLL-specific TIL (>1.9%) was associated with significantly increased MCC-specific survival P = 0.0009). T-cell cloning from four patients identified 42 distinct KLL-specific TCRa/b pairs. T-cell clones from patients with improved MCC-specific outcomes were more avid (P < 0.05) and recognized an HLA-appropriate MCC cell line. T cells specific for a single MCPyV epitope display marked TCR diversity within and between patients. Intratumoral infiltration by MCPyV-specific T cells was associated with significantly improved MCC-specific survival, suggesting that augmenting the number or avidity of virus-specific T cells may have therapeutic benefit. Cancer Immunol Res; 5(2); 137–47. ©2017 AACR.
- Published
- 2017
41. Cross-presentation and genome-wide screening reveal candidate T cells antigens for a herpes simplex virus type 1 vaccine
- Author
-
David M. Koelle, Jürgen Haas, Georges M. G. M. Verjans, Christopher L. McClurkan, Joseph J. Bruckner, Anna Wald, Lichun Dong, Tori N. Yamamoto, Susanne M. Bailer, Joshua O. Marshak, Lichen Jing, Tiana M. Chong, Greg C. Dann, Kerry J. Laing, Publica, and Virology
- Subjects
Adult ,CD4-Positive T-Lymphocytes ,Cytotoxicity, Immunologic ,Male ,viruses ,T cell ,Herpesvirus 1, Human ,Human leukocyte antigen ,Streptamer ,CD8-Positive T-Lymphocytes ,Biology ,medicine.disease_cause ,Genome ,Interferon-gamma ,Tumor Necrosis Factor Receptor Superfamily, Member 9 ,Young Adult ,Cross-Priming ,SDG 3 - Good Health and Well-being ,Antigen ,HLA Antigens ,Clinical investigation ,medicine ,Humans ,Antigens, Viral ,Pan-T antigens ,Cells, Cultured ,Medicine(all) ,Viral Vaccine ,Cross-presentation ,Herpes Simplex ,Viral Vaccines ,General Medicine ,Middle Aged ,Virology ,Herpes simplex virus ,medicine.anatomical_structure ,Technical Advance ,Immunology ,Female ,Corrigendum ,CD8 - Abstract
Herpes simplex virus type 1 (HSV-1) not only causes painful recurrent oral-labial infections, it can also cause permanent brain damage and blindness. There is currently no HSV-1 vaccine. An effective vaccine must stimulate coordinated T cell responses, but the large size of the genome and the low frequency of HSV-1-specific T cells have hampered the search for the most effective T cell antigens for inclusion in a candidate vaccine. We have now developed what we believe to be novel methods to efficiently generate a genome-wide map of the responsiveness of HSV-1-specific T cells, and demonstrate the applicability of these methods to a second complex microbe, vaccinia virus. We used cross-presentation and CD 137 activation-based FACS to enrich for polyclonal CD8(+) T effector T cells. The HSV-1 proteome was prepared in a flexible format for analyzing both CD8(+) and CD4(+) T cells from study participants. Scans with participant-specific panels of artificial APCs identified an oligospecific response in each individual. Parallel CD137-based CD4(+) T cell research showed discrete oligospecific recognition of HSV-1 antigens. Unexpectedly, the two HSV-1 proteins not previously considered as vaccine candidates elicited both CD8(+) and CD4(+) T cell responses in most HSV-1-infected individuals. In this era of microbial genomics, our methods also - demonstrated in principle for vaccinia virus for both CD8(+) and CD4(+) T cells - should be broadly applicable to the selection of T cell antigens for inclusion in candidate vaccines for many pathogens.
- Published
- 2012
42. Mucosal Shedding and Cellular Immune Response After First Episode Genital Herpes Simplex Virus Type 1 Infection
- Author
-
Lichen Jing, Stacy Selke, Hyunju Son, Christine Johnston, David M. Koelle, Sarah A. Gunby, Michael Stern, Anna Wald, Amalia Magaret, Meei-Li Huang, Mariliis Ott, and Keith R. Jerome
- Subjects
0301 basic medicine ,First episode ,business.industry ,Mucous membrane ,Virology ,Phenotype ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Infectious Diseases ,Immune system ,medicine.anatomical_structure ,Ribonucleotide reductase ,Oncology ,Virus type ,Medicine ,Viral shedding ,business ,Etoposide ,030215 immunology ,medicine.drug - Published
- 2017
43. Patients with atopic dermatitis and history of eczema herpeticum elicit herpes simplex virus–specific type 2 immune responses
- Author
-
Lichen Jing, Thomas Werfel, David M. Koelle, Gabriele Begemann, Petra Kienlin, Stephan Traidl, and Lennart M. Roesner
- Subjects
Male ,0301 basic medicine ,Allergy ,medicine.medical_specialty ,Herpesvirus 2, Human ,T-Lymphocytes ,Immunology ,Kaposi Varicelliform Eruption ,medicine.disease_cause ,Article ,Dermatitis, Atopic ,Atopy ,030207 dermatology & venereal diseases ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Immunity ,medicine ,Eczema herpeticum ,Humans ,Immunology and Allergy ,Immunity, Cellular ,Extramural ,business.industry ,Atopic dermatitis ,medicine.disease ,Dermatology ,030104 developmental biology ,Herpes simplex virus ,Cytokines ,Female ,business - Published
- 2018
44. Identification of novel Mycobacterium tuberculosis CD4 T-cell antigens via high throughput proteome screening
- Author
-
David R. Sherman, Xiaowu Liang, Kaustuv Nayak, D. Huw Davies, Kaja Murali-Krishna, Anmol Chandele, William W. Kwok, Lichen Jing, Gary Hermanson, Syed Fazil Ahamed, Junbao Yang, Ronnie M. Russell, John Kenneth, David M. Koelle, and Douglas M. Molina
- Subjects
CD4-Positive T-Lymphocytes ,Proteomics ,Epitopes, T-Lymphocyte ,Cell Separation ,Lymphocyte Activation ,Medical and Health Sciences ,Epitopes ,T-cell ,CD137 ,Tuberculosis Vaccines ,Cells, Cultured ,Cultured ,biology ,Latent tuberculosis ,Bacterial ,respiratory system ,Middle Aged ,Infectious Diseases ,medicine.anatomical_structure ,Antigen ,Proteome ,HIV/AIDS ,Cytokines ,Infection ,Tuberculosis vaccines ,Biotechnology ,Microbiology (medical) ,Adult ,Cells ,T cell ,Immunology ,chemical and pharmacologic phenomena ,Cross Reactions ,Microbiology ,Article ,Vaccine Related ,Mycobacterium tuberculosis ,Open Reading Frames ,Rare Diseases ,Bacterial Proteins ,Clinical Research ,Latent Tuberculosis ,Biodefense ,medicine ,Tuberculosis ,Humans ,Antigens ,Cell Proliferation ,Antigens, Bacterial ,Prevention ,T-cell receptor ,biology.organism_classification ,medicine.disease ,bacterial infections and mycoses ,Malate synthase ,Virology ,CD4 ,High-Throughput Screening Assays ,Orphan Drug ,Good Health and Well Being ,T-Lymphocyte ,Immunization ,Cytokine secretion ,Tetramer - Abstract
Elicitation of CD4 IFN-gamma T cell responses to Mycobacterium tuberculosis (MTB) is a rational vaccine strategy to prevent clinical tuberculosis. Diagnosis of MTB infection is based on T-cell immune memory to MTB antigens. The MTB proteome contains over four thousand open reading frames (ORFs). We conducted a pilot antigen identification study using 164 MTB proteins and MTB-specific T-cells expanded in vitro from 12 persons with latent MTB infection. Enrichment of MTB-reactive T-cells from PBMC used cell sorting or an alternate system compatible with limited resources. MTB proteins were used as single antigens or combinatorial matrices in proliferation and cytokine secretion readouts. Overall, our study found that 44 MTB proteins were antigenic, including 27 not previously characterized as CD4 T-cell antigens. Antigen truncation, peptide, NTM homology, and HLA class II tetramer studies confirmed malate synthase G (encoded by gene Rv1837) as a CD4 T-cell antigen. This simple, scalable system has potential utility for the identification of candidate MTB vaccine and biomarker antigens.
- Published
- 2015
45. Phenotypic Anchoring of Global Gene Expression Profiles Induced by N-Hydroxy-4-acetylaminobiphenyl and Benzo[a]pyrene Diol Epoxide Reveals Correlations between Expression Profiles and Mechanism of Toxicity
- Author
-
Wenhong Fan, Hong Xie, Lue Ping Zhao, Paul Vouros, Wen Luo, Helmut Zarbl, Elaine Ricicki, and Lichen Jing
- Subjects
DNA repair ,7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide ,Biology ,Toxicology ,Cell Line ,chemistry.chemical_compound ,Gene expression ,Aminobiphenyl Compounds ,Humans ,Carcinogen ,Oligonucleotide Array Sequence Analysis ,Microarray analysis techniques ,Gene Expression Profiling ,Mutagenesis ,General Medicine ,Molecular biology ,Gene expression profiling ,Phenotype ,Benzo(a)pyrene ,chemistry ,Biochemistry ,Mutation ,Toxicity ,Carcinogens ,Regression Analysis ,Mutagens - Abstract
The goal of this study was to compare changes in gene expression induced by exposure to different carcinogens and to anchor these changes to the induced levels of toxicity and mutagenesis. The human TK6 lymphoblastoid cell line was used as an in vitro model system, and reactive metabolites of two human carcinogens, benzo[a]pyrene and 4-aminobiphenyl, were used as model compounds. We first determined the toxicity of the model compounds N-hydroxy-4-acetylaminobiphenyl (N-OH-AABP) and benzo[a]pyrene diol epoxide (BPDE) in TK6 cells. BPDE was about 1000-fold more toxic and mutagenic than N-OH-AABP in TK6 cells on a molar basis. We next treated cells with three doses of each compound that resulted in low, medium, and high toxicities (5, 15, and 40%) and harvested cells at different times after exposure. Using comparable levels of toxicity as the phenotypic anchor, we compared the patterns of gene expression induced by each reactive metabolite using printed cDNA microarrays comprising approximately 18,000 human gene/EST sequences. The microarray data from the N-OH-AABP and BPDE treatment groups were compared using self-organizing map clustering algorithms, as well as a statistical regression modeling approach. While subsets of genes indicative of a generalized stress response [Hsp 40 homologue (DNAJ), Hsp70, Hsp105, and Hsp 125] were detected after exposure to both compounds at all concentrations, there were also many differentially regulated genes, including phase I xenobiotic metabolism [e.g., glutathione transferase omega (GSTTLp28) and antioxidant enzymes (Apxl)]. Other differentially regulated genes included those encoding proteins involved in all major DNA repair pathways, including excision repair (e.g., ERCC5), mismatch repair (e.g., MLH3), damage specific DNA binding protein (e.g., DDB2), and cisplatin resistance-associated overexpressed protein (LUC7A, CRA). Differences in the transcriptional response of TK6 cells to N-OH-AABP or BPDE exposure may explain the dramatic differences in the toxicity and mutagenicity of these human carcinogens.
- Published
- 2005
46. Virologic and Immunologic Evidence of Multifocal Genital Herpes Simplex Virus 2 Infection
- Author
-
Anna Wald, Lichen Jing, Stacy Selke, Christine Johnston, David M. Koelle, Amalia Magaret, Youyi Fong, Christopher M. McClurkan, Lawrence Corey, Elizabeth Tronstein, William W. Kwok, Kerry J. Laing, Meei Li Huang, Lei Jin, Alexis Klock, Jia Zhu, Kurt Diem, and Jeffrey D. Stanaway
- Subjects
Adult ,CD4-Positive T-Lymphocytes ,Male ,Adolescent ,viruses ,Biopsy ,Herpesvirus 2, Human ,Immunology ,Biology ,CD8-Positive T-Lymphocytes ,Microbiology ,Asymptomatic ,Polymerase Chain Reaction ,Virus ,Young Adult ,Immune system ,Virology ,medicine ,Humans ,Viral shedding ,Subclinical infection ,Aged ,Herpes Genitalis ,medicine.diagnostic_test ,Middle Aged ,Virus Shedding ,medicine.anatomical_structure ,Microscopy, Fluorescence ,Insect Science ,Asymptomatic Diseases ,DNA, Viral ,Pathogenesis and Immunity ,Cytokines ,Female ,Virus Activation ,medicine.symptom ,Sacral ganglia ,CD8 - Abstract
Genital herpes simplex virus (HSV) reactivation is thought to be anatomically and temporally localized, coincident with limited ganglionic infection. Short, subclinical shedding episodes are the most common form of HSV-2 reactivation, with host clearance mechanisms leading to rapid containment. The anatomic distribution of shedding episodes has not been characterized. To precisely define patterns of anatomic reactivation, we divided the genital tract into a 22-region grid and obtained daily swabs for 20 days from each region in 28 immunocompetent, HSV-2-seropositive persons. HSV was detected via PCR, and sites of asymptomatic HSV shedding were subjected to a biopsy procedure within 24 h. CD4 + and CD8 + T cells were quantified by immunofluorescence, and HSV-specific CD4 + T cells were identified by intracellular cytokine cytometry. HSV was detected in 868 (7%) of 11,603 genital swabs at a median of 12 sites per person (range, 0 to 22). Bilateral HSV detection occurred on 83 (67%) days with shedding, and the median quantity of virus detected/day was associated with the number of sites positive ( P < 0.001). In biopsy specimens of asymptomatic shedding sites, we found increased numbers of CD8 + T cells compared to control tissue (27 versus 13 cells/mm 2 , P = 0.03) and identified HSV-specific CD4 + T cells. HSV reactivations emanate from widely separated anatomic regions of the genital tract and are associated with a localized cellular infiltrate that was demonstrated to be HSV specific in 3 cases. These data provide evidence that asymptomatic HSV-2 shedding contributes to chronic inflammation throughout the genital tract. IMPORTANCE This detailed report of the anatomic patterns of genital HSV-2 shedding demonstrates that HSV-2 reactivation can be detected at multiple bilateral sites in the genital tract, suggesting that HSV establishes latency throughout the sacral ganglia. In addition, genital biopsy specimens from sites of asymptomatic HSV shedding have increased numbers of CD8 + T cells compared to control tissue, and HSV-specific CD4 + T cells are found at sites of asymptomatic shedding. These findings suggest that widespread asymptomatic genital HSV-2 shedding is associated with a targeted host immune response and contributes to chronic inflammation throughout the genital tract.
- Published
- 2014
47. Local CD4 and CD8 T-Cell Reactivity to HSV-1 Antigens Documents Broad Viral Protein Expression and Immune Competence in Latently Infected Human Trigeminal Ganglia
- Author
-
David M. Koelle, Monique van Velzen, Albert D. M. E. Osterhaus, Alessandro Sette, Georges M. G. M. Verjans, Lichen Jing, and Virology
- Subjects
CD4-Positive T-Lymphocytes ,Viral Diseases ,Anatomy and Physiology ,viruses ,Neuroimmunology ,Immunofluorescence ,Fluorescent Antibody Technique ,Antigen Processing and Recognition ,Herpesvirus 1, Human ,CD8-Positive T-Lymphocytes ,Adaptive Immunity ,medicine.disease_cause ,Major Histocompatibility Complex ,Molecular cell biology ,DNA amplification ,0302 clinical medicine ,Infectious Diseases of the Nervous System ,Virus latency ,Antigens, Viral ,Immune Response ,lcsh:QH301-705.5 ,Aged, 80 and over ,Neurons ,0303 health sciences ,T Cells ,Middle Aged ,Flow Cytometry ,Virus Latency ,Viral Persistence and Latency ,3. Good health ,Host-Pathogen Interaction ,Nucleic acids ,Infectious Diseases ,Trigeminal Ganglion ,Lytic cycle ,Cytokines ,Medicine ,Autopsy ,Cellular Types ,Immunohistochemical Analysis ,Research Article ,lcsh:Immunologic diseases. Allergy ,Viral protein ,Immune Cells ,Immunology ,Antigen presentation ,Human leukocyte antigen ,Biology ,Microbiology ,Neurological System ,Viral Proteins ,03 medical and health sciences ,Antigen ,SDG 3 - Good Health and Well-being ,Virology ,Genetics ,medicine ,Humans ,Immunoassays ,Antigen-presenting cell ,Immunity to Infections ,Molecular Biology ,Aged ,030304 developmental biology ,Inflammation ,Immunity ,Herpes Simplex ,Immune Defense ,DNA ,Immunologic Subspecialties ,medicine.disease ,Herpes simplex virus ,lcsh:Biology (General) ,Immune System ,DNA, Viral ,Immunologic Techniques ,Ganglia ,Clinical Immunology ,Parasitology ,lcsh:RC581-607 ,Cytometry ,030215 immunology - Abstract
Herpes simplex virus type 1 (HSV-1) infection results in lifelong chronic infection of trigeminal ganglion (TG) neurons, also referred to as neuronal HSV-1 latency, with periodic reactivation leading to recrudescent herpetic disease in some persons. HSV-1 proteins are expressed in a temporally coordinated fashion during lytic infection, but their expression pattern during latent infection is largely unknown. Selective retention of HSV-1 reactive T-cells in human TG suggests their role in controlling reactivation by recognizing locally expressed HSV-1 proteins. We characterized the HSV-1 proteins recognized by virus-specific CD4 and CD8 T-cells recovered from human HSV-1–infected TG. T-cell clusters, consisting of both CD4 and CD8 T-cells, surrounded neurons and expressed mRNAs and proteins consistent with in situ antigen recognition and antiviral function. HSV-1 proteome-wide scans revealed that intra-TG T-cell responses included both CD4 and CD8 T-cells directed to one to three HSV-1 proteins per person. HSV-1 protein ICP6 was targeted by CD8 T-cells in 4 of 8 HLA-discordant donors. In situ tetramer staining demonstrated HSV-1-specific CD8 T-cells juxtaposed to TG neurons. Intra-TG retention of virus-specific CD4 T-cells, validated to the HSV-1 peptide level, implies trafficking of viral proteins from neurons to HLA class II-expressing non-neuronal cells for antigen presentation. The diversity of viral proteins targeted by TG T-cells across all kinetic and functional classes of viral proteins suggests broad HSV-1 protein expression, and viral antigen processing and presentation, in latently infected human TG. Collectively, the human TG represents an immunocompetent environment for both CD4 and CD8 T-cell recognition of HSV-1 proteins expressed during latent infection. HSV-1 proteins recognized by TG-resident T-cells, particularly ICP6 and VP16, are potential HSV-1 vaccine candidates., Author Summary HSV-1 is an endemic human herpesvirus worldwide that establishes a lifelong latent infection of neurons in the trigeminal ganglion (TG), allowing intermittent reactivation resulting in recurrent disease in some persons. Studies in HSV-1 models suggest a central role of TG-infiltrating virus-specific CD8 T-cells to control reactivation. In humans, however, the functional properties and fine specificity of intra-TG T-cell responses remain enigmatic. The current study used molecular, immunological and in situ analysis platforms on human cadaveric TG obtained within hours after death to characterize the local HSV-1 specific T-cell response in latently infected human TG in detail. We identified that CD4 and CD8 T-cells were juxtaposed to TG neurons and expressed host transcripts and proteins consistent with in situ antigen recognition and antiviral function. The intra-TG T-cell response, involving both CD4 and CD8 T-cells, was directed to a limited set of HSV-1 proteins per person, which was not limited to a specific kinetic or structural class of viral proteins. Collectively, the data indicate that the human TG is an immunocompetent environment for CD4 and CD8 T-cell recognition of diverse HSV-1 proteins expressed during latent infection and that the viral antigens identified herein are rational candidates for HSV-1 subunit vaccines.
- Published
- 2013
48. CD4 T-Cell Memory Responses to Viral Infections of Humans Show Pronounced Immunodominance Independent of Duration or Viral Persistence
- Author
-
Anna Wald, Joseph J. Bruckner, Phillip L. Felgner, David M. Koelle, Joshua T. Schiffer, Juergen Haas, Lichen Jing, Tiana M. Chong, Georges M. G. M. Verjans, D. Huw Davies, and Virology
- Subjects
CD4-Positive T-Lymphocytes ,Viral protein ,viruses ,Immunology ,Vaccinia virus ,Antigens, CD137 ,Immunodominance ,Herpesvirus 1, Human ,Biology ,CD8-Positive T-Lymphocytes ,medicine.disease_cause ,Vaccines, Attenuated ,Microbiology ,Epitope ,Virus ,chemistry.chemical_compound ,Interferon-gamma ,Tumor Necrosis Factor Receptor Superfamily, Member 9 ,Antigen ,SDG 3 - Good Health and Well-being ,Virology ,medicine ,Humans ,Cells, Cultured ,Immunity, Cellular ,Tumor Necrosis Factor-alpha ,Herpes Simplex ,Chronic infection ,Herpes simplex virus ,chemistry ,Insect Science ,Pathogenesis and Immunity ,Interleukin-2 ,Vaccinia ,Immunologic Memory ,Smallpox Vaccine - Abstract
Little is known concerning immunodominance within the CD4 T-cell response to viral infections and its persistence into long-term memory. We tested CD4 T-cell reactivity against each viral protein in persons immunized with vaccinia virus (VV), either recently or more than 40 years ago, as a model self-limited viral infection. Similar tests were done with persons with herpes simplex virus 1 (HSV-1) infection as a model chronic infection. We used an indirect method capable of counting the CD4 T cells in blood reactive with each individual viral protein. Each person had a clear CD4 T-cell dominance hierarchy. The top four open reading frames accounted for about 40% of CD4 virus-specific T cells. Early and long-term memory CD4 T-cell responses to vaccinia virus were mathematically indistinguishable for antigen breadth and immunodominance. Despite the chronic intermittent presence of HSV-1 antigen, the CD4 T-cell dominance and diversity patterns for HSV-1 were identical to those observed for vaccinia virus. The immunodominant CD4 T-cell antigens included both long proteins abundantly present in virions and shorter, nonstructural proteins. Limited epitope level and direct ex vivo data were also consistent with pronounced CD4 T-cell immunodominance. We conclude that human memory CD4 T-cell responses show a pattern of pronounced immunodominance for both chronic and self-limited viral infections and that this pattern can persist over several decades in the absence of antigen.
- Published
- 2013
49. TCR immune profiling reveals intrinsic clonal selection of tissue resident memory T cells in genital mucosa during human HSV-2 infection
- Author
-
Jia Zhu, Trevor Bedford, Anthony Williams, Kerry Laing, Lichen Jing, Alvason Li, Kurt Diem, Christine Johnston, Anna wald, David M Koelle, and Lawrence Corey
- Subjects
Immunology ,Immunology and Allergy - Abstract
Tissue resident memory (TRM) T cells persist in human genital mucosa long after healing of herpes simplex virus 2 (HSV-2) infection. These TRM T cells provide effective protection to rapidly contain reactivating viruses before clinical manifestation occurs, making them excellent candidates for developing novel approaches to controlling HSV2 reactivation. To understand the dynamics of TCR repertoire and the kinetics of T cell clones during disease resolution, sequential biopsies were obtained in three individuals at the time of active lesions and 2, 4, 8 weeks post-healing, and were subjected to high-throughput sequencing of the TCRβ chain hypervariable CDR3 region. Oligoclonal immune profiles were evidenced in all biopsy tissues with a few TCR sequences constituting 30–40% of the TCR repertoire. The usage pattern of various TCRVB genes was similar among tissues of active lesion and post healed biopsies of the same subject. However, the TCRVB usage differed dramatically between the TCR repertoire in tissues and those of PBMC derived HSV-2 specific T cells taken at the same time point within each individual. This difference was more extreme when evaluating the TCR sequences that selectively persisted in post-healing biopsies. The top ten predominant TCR sequences persisting in tissues during clinical quiescence 4–8 weeks post healing did not overlap with HSV-2 specific T cells isolated from PBMC. These data suggest a TCR intrinsic property might dictate the TRM repertoire, and that the antigen specificity of TRM might differ considerably from circulating HSV-2 specific T cells.
- Published
- 2016
50. Merkel cell polyomavirus-specific CD8⁺ and CD4⁺ T-cell responses identified in Merkel cell carcinomas and blood
- Author
-
Paul Nghiem, Joshua O. Marshak, Joseph J. Carter, Erik A. Farrar, Olga K. Afanasiev, Ivy Lai, Lichun Dong, Kotaro Nagase, Christopher M. McClurkan, Kelly G. Paulson, Denise A. Galloway, David R. Byrd, Cassian Yee, Jayasri G. Iyer, Lichen Jing, and David M. Koelle
- Subjects
CD4-Positive T-Lymphocytes ,Cancer Research ,Skin Neoplasms ,medicine.medical_treatment ,Merkel cell polyomavirus ,Epitopes, T-Lymphocyte ,HLA-A24 Antigen ,Biology ,CD8-Positive T-Lymphocytes ,Epitope ,Article ,Interferon-gamma ,Lymphocytes, Tumor-Infiltrating ,Antigen ,Chlorocebus aethiops ,medicine ,Cytotoxic T cell ,Animals ,Humans ,Antigens, Viral, Tumor ,Polyomavirus Infections ,Tumor-infiltrating lymphocytes ,food and beverages ,Immunotherapy ,biology.organism_classification ,Carcinoma, Merkel Cell ,Tumor Virus Infections ,medicine.anatomical_structure ,Oncology ,Immunology ,COS Cells ,Merkel cell ,Peptides ,CD8 ,Epitope Mapping - Abstract
Purpose: Merkel cell polyomavirus (MCPyV) is prevalent in the general population, integrates into most Merkel cell carcinomas (MCC), and encodes oncoproteins required for MCC tumor growth. We sought to characterize T-cell responses directed against viral proteins that drive this cancer as a step toward immunotherapy. Experimental Design: Intracellular cytokine cytometry, IFN-γ enzyme-linked immunospot (ELISPOT) assay, and a novel HLA-A*2402–restricted MCPyV tetramer were used to identify and characterize T-cell responses against MCPyV oncoproteins in tumors and blood of MCC patients and control subjects. Results: We isolated virus-reactive CD8 or CD4 T cells from MCPyV-positive MCC tumors (2 of 6) but not from virus-negative tumors (0 of 4). MCPyV-specific T-cell responses were also detected in the blood of MCC patients (14 of 27) and control subjects (5 of 13). These T cells recognized a broad range of peptides derived from capsid proteins (2 epitopes) and oncoproteins (24 epitopes). HLA-A*2402–restricted MCPyV oncoprotein processing and presentation by mammalian cells led to CD8-mediated cytotoxicity. Virus-specific CD8 T cells were markedly enriched among tumor infiltrating lymphocytes as compared with blood, implying intact T-cell trafficking into the tumor. Although tetramer-positive CD8 T cells were detected in the blood of 2 of 5 HLA-matched MCC patients, these cells failed to produce IFN-γ when challenged ex vivo with peptide. Conclusions: Our findings suggest that MCC tumors often develop despite the presence of T cells specific for MCPyV T-Ag oncoproteins. The identified epitopes may be candidates for peptide-specific vaccines and tumor- or virus-specific adoptive immunotherapies to overcome immune evasion mechanisms in MCC patients. Clin Cancer Res; 17(21); 6671–80. ©2011 AACR.
- Published
- 2011
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.