Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Nat Immunol ; 24(8): 1345-1357, 2023 08.
Article in English | MEDLINE | ID: mdl-37400675

ABSTRACT

CD4+ T cells play key roles in a range of immune responses, either as direct effectors or through accessory cells, including CD8+ T lymphocytes. In cancer, neoantigen (NeoAg)-specific CD8+ T cells capable of direct tumor recognition have been extensively studied, whereas the role of NeoAg-specific CD4+ T cells is less well understood. We have characterized the murine CD4+ T cell response against a validated NeoAg (CLTCH129>Q) expressed by the MHC-II-deficient squamous cell carcinoma tumor model (SCC VII) at the level of single T cell receptor (TCR) clonotypes and in the setting of adoptive immunotherapy. We find that the natural CLTCH129>Q-specific repertoire is diverse and contains TCRs with distinct avidities as measured by tetramer-binding assays and CD4 dependence. Despite these differences, CD4+ T cells expressing high or moderate avidity TCRs undergo comparable in vivo proliferation to cross-presented antigen from growing tumors and drive similar levels of therapeutic immunity that is dependent on CD8+ T cells and CD40L signaling. Adoptive cellular therapy (ACT) with NeoAg-specific CD4+ T cells is most effective when TCR-engineered cells are differentiated ex vivo with IL-7 and IL-15 rather than IL-2 and this was associated with both increased expansion as well as the acquisition and stable maintenance of a T stem cell memory (TSCM)-like phenotype in tumor-draining lymph nodes (tdLNs). ACT with TSCM-like CD4+ T cells results in lower PD-1 expression by CD8+ T cells in the tumor microenvironment and an increased frequency of PD-1+CD8+ T cells in tdLNs. These findings illuminate the role of NeoAg-specific CD4+ T cells in mediating antitumor immunity via providing help to CD8+ T cells and highlight their therapeutic potential in ACT.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Mice , Animals , Programmed Cell Death 1 Receptor/metabolism , Neoplasms/metabolism , Receptors, Antigen, T-Cell/metabolism , Immunotherapy, Adoptive , Immunotherapy , CD4-Positive T-Lymphocytes , Stem Cells , Tumor Microenvironment
3.
Proc Natl Acad Sci U S A ; 119(10): e2113329119, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35239442

ABSTRACT

SignificanceThe CD4+ Treg response following acute Listeria infection is heterogeneous and deploys two distinct modes of suppression coinciding with initial pathogen exposure and resolution of infection. This bimodal suppression of CD8+ T cells during priming and contraction is mediated by separate Treg lineages. These findings make a significant contribution to our understanding of the functional plasticity inherent within Tregs, which allows these cells to serve as a sensitive and dynamic cellular rheostat for the immune system to prevent autoimmune pathology in the face of inflammation attendant to acute infection, enable expansion of the pathogen-specific response needed to control the infection, and reestablish immune homeostasis after the threat has been contained.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , T-Lymphocytes, Regulatory/immunology , 5'-Nucleotidase/immunology , Acute Disease , Animals , Mice
4.
J Immunol ; 198(6): 2341-2351, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28159899

ABSTRACT

Cross-presentation is a modular series of intracellular events dictating the internalization and subsequent MHC class I (MHC I) display of extracellular Ags. This process has been defined in dendritic cells and plays a fundamental role in the induction of CD8+ T cell immunity during viral, intracellular bacterial, and antitumor responses. Herein, acute viral infection of murine liver with adenovirus, a model for intrahepatic cross-presentation, confirms hepatocytes directly contribute to cross-presentation of Ags and priming the pool of naive CD8+ T cells within the liver microenvironment. Processing of soluble and cell-associated Ags into peptide displayed by MHC I is however defective in hepatocytes lacking collectrin, an intracellular chaperone protein that localizes within the endoplasmic reticulum-Golgi intermediate compartment. Loss of hepatic collectrin expression leads to the diminished cross-priming and expansion of cytolytic antiviral CD8+ T cells. This study demonstrates that collectrin positively regulates processing of engulfed Ags into MHC I:peptide complexes within hepatocytes. Collectrin-mediated cross-presentation supports intrahepatic adaptive antiviral immune responses and may lead to insights into the nature of how the liver acts as a primary site of CD8+ T cell activation.


Subject(s)
Adenoviridae Infections/immunology , Adenoviridae/immunology , CD8-Positive T-Lymphocytes/immunology , Cross-Priming , Hepatocytes/immunology , Liver/immunology , Membrane Glycoproteins/metabolism , Acute Disease , Animals , Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/virology , Extracellular Space/immunology , Hepatocytes/virology , Histocompatibility Antigens Class I/metabolism , Liver/virology , Lymphocyte Activation/genetics , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Solubility , Transplantation Chimera
5.
Hepatology ; 59(4): 1351-65, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24677194

ABSTRACT

UNLABELLED: The liver is a tolerogenic environment exploited by persistent infections, such as hepatitis B (HBV) and C (HCV) viruses. In a murine model of intravenous hepatotropic adenovirus infection, liver-primed antiviral CD8(+) T cells fail to produce proinflammatory cytokines and do not display cytolytic activity characteristic of effector CD8(+) T cells generated by infection at an extrahepatic, that is, subcutaneous, site. Importantly, liver-generated CD8(+) T cells also appear to have a T-regulatory (Treg) cell function exemplified by their ability to limit proliferation of antigen-specific T-effector (Teff ) cells in vitro and in vivo via T-cell immunoglobulin and mucin 3 (Tim-3) expressed by the CD8(+) Treg cells. Regulatory activity did not require recognition of the canonical Tim-3 ligand, galectin-9, but was dependent on CD8(+) Treg cell-surface Tim-3 binding to the alarmin, high-mobility group box 1 (HMGB-1). CONCLUSION: Virus-specific Tim-3(+) CD8(+) T cells operating through HMGB-1 recognition in the setting of acute and chronic viral infections of the liver may act to dampen hepatic T-cell responses in the liver microenvironment and, as a consequence, limit immune-mediated tissue injury or promote the establishment of persistent infections.


Subject(s)
Adaptive Immunity/physiology , Adenoviridae Infections/immunology , Adenoviridae Infections/physiopathology , CD8-Positive T-Lymphocytes/physiology , Galectins/physiology , HMGB1 Protein/physiology , Mucin-3/physiology , Adenoviridae/physiology , Adenoviridae Infections/pathology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Proliferation , Cellular Microenvironment , Disease Models, Animal , Immunoglobulins/physiology , In Vitro Techniques , Liver/pathology , Liver/physiopathology , Liver/virology , Mice , Mice, Inbred Strains , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
7.
J Clin Invest ; 133(17)2023 09 01.
Article in English | MEDLINE | ID: mdl-37655661

ABSTRACT

Therapeutic benefit to immune checkpoint blockade (ICB) is currently limited to the subset of cancers thought to possess a sufficient tumor mutational burden (TMB) to allow for the spontaneous recognition of neoantigens (NeoAg) by autologous T cells. We explored whether the response to ICB of an aggressive low-TMB squamous cell tumor could be improved through combination immunotherapy using functionally defined NeoAg as targets for endogenous CD4+ and CD8+ T cells. We found that, whereas vaccination with CD4+ or CD8+ NeoAg alone did not offer prophylactic or therapeutic immunity, vaccines containing NeoAg recognized by both subsets overcame ICB resistance and led to the eradication of large established tumors that contained a subset of PD-L1+ tumor-initiating cancer stem cells (tCSC), provided the relevant epitopes were physically linked. Therapeutic CD4+/CD8+ T cell NeoAg vaccination produced a modified tumor microenvironment (TME) with increased numbers of NeoAg-specific CD8+ T cells existing in progenitor and intermediate exhausted states enabled by combination ICB-mediated intermolecular epitope spreading. We believe that the concepts explored herein should be exploited for the development of more potent personalized cancer vaccines that can expand the range of tumors treatable with ICB.


Subject(s)
Immune Checkpoint Inhibitors , Vaccination , Epitopes , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes
8.
bioRxiv ; 2023 May 07.
Article in English | MEDLINE | ID: mdl-37205330

ABSTRACT

Therapeutic benefit to immune checkpoint blockade (ICB) is currently limited to the subset of cancers thought to possess a sufficient tumor mutational burden (TMB) to allow for the spontaneous recognition of neoantigens (NeoAg) by autologous T cells. We explored whether the response of an aggressive low TMB squamous cell tumor to ICB could be improved through combination immunotherapy using functionally defined NeoAg as targets for endogenous CD4 + and CD8 + T cells. We found that, whereas vaccination with CD4 + or CD8 + NeoAg alone did not offer prophylactic or therapeutic immunity, vaccines containing NeoAg recognized by both subsets overcame ICB resistance and led to the eradication of large established tumors that contained a subset of PD-L1 + tumor-initiating cancer stem cells (tCSC), provided the relevant epitopes were physically linked. Therapeutic CD4 + /CD8 + T cell NeoAg vaccination produced a modified tumor microenvironment (TME) with increased numbers of NeoAg-specific CD8 + T cells existing in progenitor and intermediate exhausted states enabled by combination ICB-mediated intermolecular epitope spreading. The concepts explored herein should be exploited for the development of more potent personalized cancer vaccines that can expand the range of tumors treatable with ICB.

9.
J Immunol ; 184(5): 2693-701, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20124099

ABSTRACT

The tolerogenic nature of the liver allows daily exposure to gut-derived foreign Ags without causing inflammation, but it may facilitate persistent infection in the liver. NK cells play a central role in innate immunity, as well as in shaping the adaptive immune response. We hypothesized that the naive mouse liver maintains intrahepatic NK cells in a functionally hyporesponsive state. Compared with splenic NK cells, liver NK cells displayed a dampened IFN-gamma response to IL-12/IL-18 stimulation. Importantly, the liver contains a significant population of functionally hyporesponsive NK cells that express high levels of the inhibitory receptor NKG2A and lack expression of MHC class I-binding Ly49 receptors. Adoptively transferred splenic NK cells that migrate to the liver displayed phenotypic and functional changes, suggesting that the liver environment modifies NK cell receptor expression and functional responsiveness. Notably, IL-10 is present at high levels within the liver, and in vivo blockade of IL-10R resulted in a decreased percentage of intrahepatic NKG2A(+)Ly49(-) NK cells. These data suggest that the liver environment regulates NK cell receptor expression and that IL-10 contributes to the regulation of liver NK cells, in part, by maintaining a greater percentage of the hyporesponsive NKG2A(+)Ly49(-) NK cells in the liver.


Subject(s)
Interleukin-10/metabolism , Killer Cells, Natural/metabolism , NK Cell Lectin-Like Receptor Subfamily A/metabolism , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Adoptive Transfer , Animals , Cell Movement , Chemokine CCL3/deficiency , Chemokine CCL3/genetics , Cytotoxicity, Immunologic , Female , Flow Cytometry , Granzymes/metabolism , Interferon-gamma/metabolism , Interleukin-12/pharmacology , Interleukin-18/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Liver/immunology , Liver/metabolism , Mice , Mice, Inbred C57BL , Receptors, Interleukin-10/immunology , Receptors, Interleukin-10/metabolism , Spleen/immunology , Spleen/metabolism
10.
Cancer Immunol Res ; 10(2): 146-153, 2022 02.
Article in English | MEDLINE | ID: mdl-34937730

ABSTRACT

Anti-PD-1/PD-L1 immune checkpoint blockade (ICB) therapy has revolutionized the treatment of many types of cancer over the past decade. The initial therapeutic hypothesis underlying the mechanism of anti-PD-1/PD-L1 ICB was built around the premise that it acts locally in the tumor, reversing the exhaustion of PD-1hiCD8+ T cells by "releasing the brakes." However, recent studies have provided unprecedented insight into the complexity within the CD8+ T-cell pool in the tumor microenvironment (TME). Single-cell RNA sequencing and epigenetic profiling studies have identified novel cell surface markers, revealing heterogeneity within CD8+ T-cell states classified as unique. Moreover, these studies highlighted that following ICB, CD8+ T-cell states within and outside the TME possess a differential capacity to respond, mobilize to the TME, and seed an effective antitumor immune response. In aggregate, these recent developments have led to a reevaluation of our understanding of both the underlying mechanisms and the sites of action of ICB therapy. Here, we discuss the evidence for the reversibility of CD8+ T-cell exhaustion after ICB treatment and its implication for the further development of cancer immunotherapy.


Subject(s)
Immune Checkpoint Inhibitors , Neoplasms , B7-H1 Antigen/pharmacology , CD8-Positive T-Lymphocytes , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Neoplasms/drug therapy , Programmed Cell Death 1 Receptor , Tumor Microenvironment
11.
Front Immunol ; 12: 715234, 2021.
Article in English | MEDLINE | ID: mdl-34354714

ABSTRACT

A paradigm shift in the understanding of the exhausted CD8+ T cell (Tex) lineage is underway. Originally thought to be a uniform population that progressively loses effector function in response to persistent antigen, single-cell analysis has now revealed that CD8+ Tex is composed of multiple interconnected subpopulations. The heterogeneity within the CD8+ Tex lineage is comprised of immune checkpoint blockade (ICB) permissive and refractory subsets termed stem-like and terminally differentiated cells, respectively. These populations occupy distinct peripheral and intratumoral niches and are characterized by transcriptional processes that govern transitions between cell states. This review presents key findings in the field to construct an updated view of the spatial, transcriptional, and functional heterogeneity of anti-tumoral CD8+ Tex. These emerging insights broadly call for (re-)focusing cancer immunotherapies to center on the driver mechanism(s) underlying the CD8+ Tex developmental continuum aimed at stabilizing functional subsets.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunity , Neoplasms/immunology , Animals , Antigens/immunology , Biomarkers , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Disease Management , Disease Susceptibility , Epigenesis, Genetic , Host-Pathogen Interactions/immunology , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Proteins/genetics , Immune Checkpoint Proteins/metabolism , Molecular Targeted Therapy , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Organ Specificity , Single-Cell Analysis , Transcription, Genetic
12.
Cell Rep ; 31(1): 107249, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32268093

ABSTRACT

CD4+ T lymphocytes are crucial for controlling a range of innate and adaptive immune effectors. For CD8+ cytotoxic T lymphocyte (CTL) responses, CD4+ T cells can function as helpers (TH) to amplify magnitude and functionality or as regulatory cells (Treg) capable of profound inhibition. It is unclear what determines differentiation to these phenotypes and whether pathogens provoke alternate programs. We find that, depending on the size of initial dose, Listeria infection drives CD4+ T cells to act as TH or induces rapid polyclonal conversion to immunosuppressive Treg. Conversion to Treg depends on the TLR9 and IL-12 pathways elicited by CD8α+ dendritic cell (DC) sensing of danger-associated neutrophil self-DNA. These findings resolve long-standing questions regarding the conditional requirement for TH amongst pathogens and reveal a remarkable degree of plasticity in the function of CD4+ T cells, which can be quickly converted to Tregin vivo by infection-mediated immune modulation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , DNA/immunology , Listeriosis/immunology , T-Lymphocytes, Regulatory/immunology , Toll-Like Receptor 9/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , DNA/genetics , Dendritic Cells/immunology , Female , Immunity, Cellular/drug effects , Immunity, Cellular/genetics , Immunity, Cellular/immunology , Interleukin-12/biosynthesis , Interleukin-12/genetics , Interleukin-12/immunology , Listeria monocytogenes/immunology , Listeriosis/genetics , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Toll-Like Receptor 9/genetics
13.
Front Immunol ; 9: 2319, 2018.
Article in English | MEDLINE | ID: mdl-30364187

ABSTRACT

There is a need for new vaccine adjuvant strategies that offer both vigorous antibody and T-cell mediated protection to combat difficult intracellular pathogens and cancer. To this aim, we formulated class-B synthetic oligodeoxynucleotide containing unmethylated cytosine-guanine motifs (CpG-ODN) with a nanostructure (Coa-ASC16 or coagel) formed by self-assembly of 6-0-ascorbyl palmitate ester. Our previous results demonstrated that mice immunized with ovalbumin (OVA) and CpG-ODN formulated with Coa-ASC16 (OVA/CpG-ODN/Coa-ASC16) elicited strong antibodies (IgG1 and IgG2a) and Th1/Th17 cellular responses without toxic systemic effects. These responses were superior to those induced by a solution of OVA with CpG-ODN or OVA/CpG-ODN formulated with aluminum salts. In this study, we investigated the capacity of this adjuvant strategy (CpG-ODN/Coa-ASC16) to elicit CD8+ T-cell response and some of the underlying cellular and molecular mechanisms involved in adaptive response. We also analyzed whether this adjuvant strategy allows a switch from an immunization scheme of three-doses to one of single-dose. Our results demonstrated that vaccination with OVA/CpG-ODN/Coa-ASC16 elicited an antigen-specific long-lasting humoral response and importantly-high quality CD8+ T-cell immunity with a single-dose immunization. Moreover, Coa-ASC16 promoted co-uptake of OVA and CpG-ODN by dendritic cells. The CD8+ T-cell response induced by OVA/CpG-ODN/Coa-ASC16 was dependent of type I interferons and independent of CD4+ T-cells, and showed polyfunctionality and efficiency against an intracellular pathogen. Furthermore, the cellular and humoral responses elicited by the nanostructured formulation were IL-6-independent. This system provides a simple and inexpensive adjuvant strategy with great potential for future rationally designed vaccines.


Subject(s)
Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Interferon Type I/metabolism , Oligodeoxyribonucleotides/immunology , Adjuvants, Immunologic , Animals , Antigens/chemistry , Cytokines/metabolism , Humans , Immunity, Humoral , Mice , Mice, Knockout , Nanostructures , Oligodeoxyribonucleotides/chemistry , Ovalbumin/immunology , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
14.
Mol Ther Nucleic Acids ; 2: e72, 2013 Feb 19.
Article in English | MEDLINE | ID: mdl-23423360

ABSTRACT

Effective clinical application of antiviral immunotherapies necessitates enhancing the functional state of natural killer (NK) and CD8(+) T cells. An important mechanism for the establishment of viral persistence in the liver is the activation of the PD-1/PD-L1 inhibitory pathway. To examine the role of hepatic myeloid PD-L1 expression during viral infection, we determined the magnitude and quality of antiviral immune responses by administering PD-L1 short-interfering RNA (siRNA) encapsulated in lipidoid nanoparticles (LNP) in mice. Our studies indicate that Kupffer cells (KC) preferentially engulfed PD-L1 LNP within a short period of time and silenced Pdl1 during adenovirus and MCMV infection leading to enhanced NK and CD8(+) T cell intrahepatic accumulation, effector function (interferon (IFN)-γ and granzyme B (GrB) production), CD8(+) T cell-mediated viral clearance, and memory. Our results demonstrate that PD-L1 knockdown on KCs is central in determining the outcome of liver viral infections, and they represent a new class of gene therapy.Molecular Therapy - Nucleic Acids (2013) 2, e72; doi:10.1038/mtna.2012.63; published online 19 February 2013.

15.
PLoS One ; 4(10): e7619, 2009 Oct 30.
Article in English | MEDLINE | ID: mdl-19876399

ABSTRACT

The liver possesses distinct tolerogenic properties because of continuous exposure to bacterial constituents and nonpathogenic food antigen. The central immune mediators required for the generation of effective immune responses in the liver environment have not been fully elucidated. In this report, we demonstrate that the liver can indeed support effector CD8(+) T cells during adenovirus infection when the T cells are primed in secondary lymphoid tissues. In contrast, when viral antigen is delivered predominantly to the liver via intravenous (IV) adenovirus infection, intrahepatic CD8(+) T cells are significantly impaired in their ability to produce inflammatory cytokines and lyse target cells. Additionally, intrahepatic CD8(+) T cells generated during IV adenovirus infection express elevated levels of PD-1. Notably, lower doses of adenovirus infection do not rescue the impaired effector function of intrahepatic CD8(+) T cell responses. Instead, intrahepatic antigen recognition limits the generation of potent anti-viral responses at both priming and effector stages of the CD8(+) T cell response and accounts for the dysfunctional CD8(+) T cell response observed during IV adenovirus infection. These results also implicate that manipulation of antigen delivery will facilitate the design of improved vaccination strategies to persistent viral infection.


Subject(s)
Antiviral Agents/chemistry , CD8-Positive T-Lymphocytes/immunology , Liver/metabolism , Adenoviridae/metabolism , Adenoviridae Infections , Animals , Bone Marrow Cells/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Coculture Techniques , Cytokines/metabolism , Flow Cytometry/methods , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Spleen/virology
SELECTION OF CITATIONS
SEARCH DETAIL