Transcriptional Pattern Analysis of Virus-Specific CD8+ T Cells in Hepatitis C Infection: Increased Expression of TOX and Eomesodermin During and After Persistent Antigen Recognition

Thymocyte selection-associated high mobility group box (TOX) has been described to be a key regulator in the formation of CD8+ T cell exhaustion. Hepatitis C virus (HCV) infection with different lengths of antigen exposure in acute, chronic, and after resolution of HCV infection is the ideal immunological model to study the expression of TOX in HCV-specific CD8+ T cells with different exposure to antigen. HCV-specific CD8+ T cells from 35 HLA-A*01:01, HLA-A*02:01, and HLA-A*24:02 positive patients were analyzed with a 16-color FACS-panel evaluating the surface expression of lineage markers (CD3, CD8), ectoenzymes (CD39, CD73), markers of differentiation (CD45RO, CCR7, CD127), and markers of exhaustion and activation (TIGIT, PD-1, KLRG1, CD226) and transcription factors (TOX, Eomesodermin, T-bet). Here, we defined on-target T cells as T cells against epitopes without escape mutations and off-target T cells as those against a “historical” antigen mutated in the autologous sequence. TOX+HCV-specific CD8+ T cells from patients with chronic HCV and on-target T cells displayed co-expression of Eomesodermin and were associated with the formation of terminally exhausted CD127-PD1hi, CD39hi, CD73low CD8+ T cells. In contrast, TOX+HCV-specific CD8+ T cells in patients with off-target T cells represented a progenitor memory Tex phenotype characterized by CD127hi expression and a CD39low and CD73hi phenotype. TOX+HCV-specified CD8+ T cells in patients with a sustained virologic response were characterized by a memory phenotype (CD127+, CD73hi) and co-expression of immune checkpoints and Eomesodermin, indicating a key structure in priming of HCV-specific CD8+ T cells in the chronic stage, which persisted as a residual after therapy. Overall, the occurrence of TOX+HCV-specific CD8+ T cells was revealed at each disease stage, which impacted the development of progenitor Tex, intermediate Tex, and terminally exhausted T cell through an individual molecular footprint. In sum, TOX is induced early during acute infection but is modulated by changes in viral sequence and antigen recognition. In the case of antigen persistence, the interaction with Eomesodermin leads to the formation of terminally exhausted virus-specific CD8+ T cells, and there was a direct correlation of the co-expression of TOX and Eomes and terminally exhausted phenotype of virus-specific CD8+ T cells.

[1]  Todd M. Allen,et al.  Epigenetic scars of CD8+ T cell exhaustion persist after cure of chronic infection in humans , 2021, Nature Immunology.

[2]  Semin Lee,et al.  Single-cell RNA sequencing reveals distinct cellular factors for response to immunotherapy targeting CD73 and PD-1 in colorectal cancer , 2021, Journal for ImmunoTherapy of Cancer.

[3]  E. Wherry,et al.  Epigenetic scarring of exhausted T cells hinders memory differentiation upon eliminating chronic antigenic stimulation , 2021, Nature Immunology.

[4]  E. Wherry,et al.  Memory T-Cell Heterogeneity and Terminology. , 2021, Cold Spring Harbor perspectives in biology.

[5]  A. Elorza,et al.  CD73 Ectonucleotidase Restrains CD8+ T Cell Metabolic Fitness and Anti-tumoral Activity , 2021, Frontiers in Cell and Developmental Biology.

[6]  Sagar,et al.  Memory-like HCV-specific CD8+ T cells retain a molecular scar after cure of chronic HCV infection , 2021, Nature Immunology.

[7]  C. Bokemeyer,et al.  Increased frequency of TIGIT+CD73-CD8+ T cells with a TOX+ TCF-1low profile in patients with newly diagnosed and relapsed AML , 2021, Oncoimmunology.

[8]  Sagar,et al.  TOX defines the degree of CD8+ T cell dysfunction in distinct phases of chronic HBV infection , 2020, Gut.

[9]  J. Pawlotsky,et al.  EASL recommendations on treatment of hepatitis C - Final update of the series. , 2020, Journal of hepatology.

[10]  C. Brander,et al.  TOX is expressed by exhausted and polyfunctional human effector memory CD8+ T cells , 2020, Science Immunology.

[11]  Hyun-Dong Chang,et al.  Antigen‐driven PD‐1+TOX+BHLHE40+ and PD‐1+TOX+EOMES+ T lymphocytes regulate juvenile idiopathic arthritis in situ , 2020, European journal of immunology.

[12]  Farshad Mirzavi,et al.  The therapeutic potential of targeting CD73 and CD73-derived adenosine in melanoma. , 2020, Biochimie.

[13]  E. Wherry,et al.  Developmental Relationships of Four Exhausted CD8+ T Cell Subsets Reveals Underlying Transcriptional and Epigenetic Landscape Control Mechanisms. , 2020, Immunity.

[14]  D. Allard,et al.  On the mechanism of anti-CD39 immune checkpoint therapy , 2020, Journal for ImmunoTherapy of Cancer.

[15]  Xu Yixin,et al.  Adenosine Generated by Regulatory T Cells Induces CD8+ T Cell Exhaustion in Gastric Cancer through A2aR Pathway , 2019, BioMed research international.

[16]  G. Freeman,et al.  Proliferating Transitory T Cells with an Effector-like Transcriptional Signature Emerge from PD-1+ Stem-like CD8+ T Cells during Chronic Infection. , 2019, Immunity.

[17]  S. Berger,et al.  TCF-1-Centered Transcriptional Network Drives an Effector versus Exhausted CD8 T Cell-Fate Decision. , 2019, Immunity.

[18]  E. Wherry,et al.  Defining ‘T cell exhaustion’ , 2019, Nature Reviews Immunology.

[19]  A. Lohse,et al.  HCV-specific CD4+ T cells of patients with acute and chronic HCV infection display high expression of TIGIT and other co-inhibitory molecules , 2019, Scientific reports.

[20]  M. Delorenzi,et al.  TOX reinforces the phenotype and longevity of exhausted T cells in chronic viral infection , 2019, Nature.

[21]  S. Berger,et al.  TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion , 2019, Nature.

[22]  Elisabeth F. Heuston,et al.  Single-Cell RNA-Seq Reveals TOX as a Key Regulator of CD8+ T cell Persistence in Chronic Infection , 2019, Nature Immunology.

[23]  Yong Liu,et al.  TOX is a critical regulator of tumour-specific T cell differentiation , 2019, Nature.

[24]  P. Hogan,et al.  TOX and TOX2 transcription factors cooperate with NR4A transcription factors to impose CD8+ T cell exhaustion , 2019, Proceedings of the National Academy of Sciences.

[25]  E. Wherry,et al.  CD8 T Cell Exhaustion During Chronic Viral Infection and Cancer. , 2019, Annual review of immunology.

[26]  M. Smyth,et al.  The role of NK cells and CD39 in the immunological control of tumor metastases , 2019, Oncoimmunology.

[27]  Vijay Kumar,et al.  Lipopolysaccharide-acylating capacity of the gut microbiota and its potential impact on the immunopathogenesis of HIV infection. , 2019, AIDS.

[28]  L. Ni,et al.  High Levels of Eomes Promote Exhaustion of Anti-tumor CD8+ T Cells , 2018, Front. Immunol..

[29]  R. Thimme,et al.  HCV-Specific T Cell Responses During and After Chronic HCV Infection , 2018, Viruses.

[30]  Yue Wang,et al.  [Automatic clustering method of flow cytometry data based on t-distributed stochastic neighbor embedding]. , 2018, Sheng wu yi xue gong cheng xue za zhi = Journal of biomedical engineering = Shengwu yixue gongchengxue zazhi.

[31]  Francesco Ferrari,et al.  High-dimensional single cell analysis identifies stem-like cytotoxic CD8+ T cells infiltrating human tumors , 2018, The Journal of experimental medicine.

[32]  M. Schetinger,et al.  Adenosine signaling and adenosine deaminase regulation of immune responses: impact on the immunopathogenesis of HIV infection , 2018, Purinergic Signalling.

[33]  Evan W. Newell,et al.  Epigenomic‐Guided Mass Cytometry Profiling Reveals Disease‐Specific Features of Exhausted CD8 T Cells , 2018, Immunity.

[34]  C. Sedlik,et al.  CD39 Expression Defines Cell Exhaustion in Tumor-Infiltrating CD8+ T Cells. , 2018, Cancer research.

[35]  R. Kaiser,et al.  A genotype independent, full-genome reverse-transcription protocol for HCV genotyping and resistance testing. , 2017, Journal of clinical virology : the official publication of the Pan American Society for Clinical Virology.

[36]  P. de Vos,et al.  Extracellular ATP and adenosine: The Yin and Yang in immune responses? , 2017, Molecular aspects of medicine.

[37]  P. Knolle,et al.  TCF1+ hepatitis C virus-specific CD8+ T cells are maintained after cessation of chronic antigen stimulation , 2017, Nature Communications.

[38]  M. Manns,et al.  Direct-Acting Antiviral-Induced Hepatitis C Virus Clearance Does Not Completely Restore the Altered Cytokine and Chemokine Milieu in Patients With Chronic Hepatitis C. , 2016, The Journal of infectious diseases.

[39]  S. Berger,et al.  Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade , 2016, Science.

[40]  Matheus C. Bürger,et al.  Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy , 2016, Nature.

[41]  Sandra P. Calderon-Copete,et al.  T Cell Factor 1-Expressing Memory-like CD8(+) T Cells Sustain the Immune Response to Chronic Viral Infections. , 2016, Immunity.

[42]  M. Bono,et al.  CD73 and CD39 ectonucleotidases in T cell differentiation: Beyond immunosuppression , 2015, FEBS letters.

[43]  P. Klenerman,et al.  CD39 Expression Identifies Terminally Exhausted CD8+ T Cells , 2015, PLoS pathogens.

[44]  N. Scherbaum,et al.  Natural prevalence of resistance-associated variants in hepatitis C virus NS5A in genotype 3a-infected people who inject drugs in Germany. , 2015, Journal of clinical virology : the official publication of the Pan American Society for Clinical Virology.

[45]  Piet Demeester,et al.  FlowSOM: Using self‐organizing maps for visualization and interpretation of cytometry data , 2015, Cytometry. Part A : the journal of the International Society for Analytical Cytology.

[46]  E. Wherry,et al.  T cell exhaustion during persistent viral infections. , 2015, Virology.

[47]  X. Liu,et al.  T-cell exhaustion in chronic hepatitis B infection: current knowledge and clinical significance , 2015, Cell Death and Disease.

[48]  J. Haas,et al.  Dysfunctional CD8+ T cells in hepatitis B and C are characterized by a lack of antigen-specific T-bet induction , 2014, The Journal of experimental medicine.

[49]  F. Heinemann,et al.  Impact of Sequence Variation in a Dominant HLA-A*02-Restricted Epitope in Hepatitis C Virus on Priming and Cross-Reactivity of CD8+ T Cells , 2014, Journal of Virology.

[50]  J. Míquel,et al.  Adaptive immune response during hepatitis C virus infection. , 2014, World journal of gastroenterology.

[51]  J. Hauber,et al.  Decreased frequency of CD73+CD8+ T cells of HIV‐infected patients correlates with immune activation and T cell exhaustion , 2013, Journal of leukocyte biology.

[52]  B. Rehermann Pathogenesis of chronic viral hepatitis: differential roles of T cells and NK cells , 2013, Nature Medicine.

[53]  E. Vizi,et al.  CD39 and CD73 in immunity and inflammation. , 2013, Trends in molecular medicine.

[54]  E John Wherry,et al.  Network analysis reveals centrally connected genes and pathways involved in CD8+ T cell exhaustion versus memory. , 2012, Immunity.

[55]  Burton E. Barnett,et al.  Progenitor and Terminal Subsets of CD8+ T Cells Cooperate to Contain Chronic Viral Infection , 2012, Science.

[56]  Jung-Hye Choi,et al.  Overexpression of CD73 in epithelial ovarian carcinoma is associated with better prognosis, lower stage, better differentiation and lower regulatory T cell infiltration , 2012, Journal of gynecologic oncology.

[57]  Todd M. Allen,et al.  Broadly directed virus-specific CD4+ T cell responses are primed during acute hepatitis C infection, but rapidly disappear from human blood with viral persistence , 2012, The Journal of experimental medicine.

[58]  E John Wherry,et al.  T cell exhaustion , 2011 .

[59]  H. Pircher,et al.  Coexpression of PD-1, 2B4, CD160 and KLRG1 on Exhausted HCV-Specific CD8+ T Cells Is Linked to Antigen Recognition and T Cell Differentiation , 2010, PLoS pathogens.

[60]  Todd M. Allen,et al.  Hepatitis C Virus (HCV) Sequence Variation Induces an HCV-Specific T-Cell Phenotype Analogous to Spontaneous Resolution , 2009, Journal of Virology.

[61]  Antonio Polley,et al.  Coregulation of CD8+ T cell exhaustion during chronic viral infection by multiple inhibitory receptors , 2008, Nature immunology.

[62]  Todd M. Allen,et al.  High Level of PD-1 Expression on Hepatitis C Virus (HCV)-Specific CD8+ and CD4+ T Cells during Acute HCV Infection, Irrespective of Clinical Outcome , 2007, Journal of Virology.

[63]  Todd M. Allen,et al.  Viral Sequence Evolution in Acute Hepatitis C Virus Infection , 2007, Journal of Virology.

[64]  H. Pircher,et al.  Analysis of CD127 and KLRG1 Expression on Hepatitis C Virus-Specific CD8+ T Cells Reveals the Existence of Different Memory T-Cell Subsets in the Peripheral Blood and Liver , 2006, Journal of Virology.

[65]  M. Massari,et al.  PD-1 Expression in Acute Hepatitis C Virus (HCV) Infection Is Associated with HCV-Specific CD8 Exhaustion , 2006, Journal of Virology.

[66]  C. Walker,et al.  Adaptive immune responses in acute and chronic hepatitis C virus infection , 2005, Nature.

[67]  F. Chisari,et al.  Determinants of Viral Clearance and Persistence during Acute Hepatitis C Virus Infection , 2001, The Journal of experimental medicine.

[68]  M. Mulligan,et al.  A Significant Number of Human Immunodeficiency Virus Epitope-Specific Cytotoxic T Lymphocytes Detected by Tetramer Binding Do Not Produce Gamma Interferon , 2000, Journal of Virology.

[69]  J. Lieberman,et al.  Impaired function of circulating HIV-specific CD8(+) T cells in chronic human immunodeficiency virus infection. , 2000, Blood.

[70]  B. Walker,et al.  Analysis of Successful Immune Responses in Persons Infected with Hepatitis C Virus , 2000, The Journal of experimental medicine.