Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Cell ; 185(22): 4049-4066.e25, 2022 10 27.
Article in English | MEDLINE | ID: mdl-36208623

ABSTRACT

Blocking PD-1/PD-L1 signaling transforms cancer therapy and is assumed to unleash exhausted tumor-reactive CD8+ T cells in the tumor microenvironment (TME). However, recent studies have also indicated that the systemic tumor-reactive CD8+ T cells may respond to PD-1/PD-L1 immunotherapy. These discrepancies highlight the importance of further defining tumor-specific CD8+ T cell responders to PD-1/PD-L1 blockade. Here, using multiple preclinical tumor models, we revealed that a subset of tumor-specific CD8+ cells in the tumor draining lymph nodes (TdLNs) was not functionally exhausted but exhibited canonical memory characteristics. TdLN-derived tumor-specific memory (TTSM) cells established memory-associated epigenetic program early during tumorigenesis. More importantly, TdLN-TTSM cells exhibited superior anti-tumor therapeutic efficacy after adoptive transfer and were characterized as bona fide responders to PD-1/PD-L1 blockade. These findings highlight that TdLN-TTSM cells could be harnessed to potentiate anti-tumor immunotherapy.


Subject(s)
B7-H1 Antigen , Neoplasms , Humans , Programmed Cell Death 1 Receptor , CD8-Positive T-Lymphocytes , Immune Checkpoint Inhibitors , Tumor Microenvironment , Neoplasms/therapy , Neoplasms/pathology , Lymph Nodes/pathology
2.
Nat Immunol ; 23(2): 303-317, 2022 02.
Article in English | MEDLINE | ID: mdl-34949833

ABSTRACT

Antigen-specific memory CD4+ T cells can persist and confer rapid and efficient protection from microbial reinfection. However, the mechanisms underlying the long-term maintenance of the memory CD4+ T cell pool remain largely unknown. Here, using a mouse model of acute infection with lymphocytic choriomeningitis virus (LCMV), we found that the serine/threonine kinase complex mammalian target of rapamycin complex 2 (mTORC2) is critical for the long-term persistence of virus-specific memory CD4+ T cells. The perturbation of mTORC2 signaling at memory phase led to an enormous loss of virus-specific memory CD4+ T cells by a unique form of regulated cell death (RCD), ferroptosis. Mechanistically, mTORC2 inactivation resulted in the impaired phosphorylation of downstream AKT and GSK3ß kinases, which induced aberrant mitochondrial reactive oxygen species (ROS) accumulation and ensuing ferroptosis-causative lipid peroxidation in virus-specific memory CD4+ T cells; furthermore, the disruption of this signaling cascade also inhibited glutathione peroxidase 4 (GPX4), a major scavenger of lipid peroxidation. Thus, the mTORC2-AKT-GSK3ß axis functions as a key signaling hub to promote the longevity of virus-specific memory CD4+ T cells by preventing ferroptosis.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Ferroptosis/immunology , Immunologic Memory/immunology , Longevity/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Mechanistic Target of Rapamycin Complex 2/immunology , Animals , Glycogen Synthase Kinase 3 beta/immunology , Lipid Peroxidation/immunology , Lymphocyte Activation/immunology , Lymphocyte Count/methods , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/immunology
3.
Nat Immunol ; 16(9): 991-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26214740

ABSTRACT

Induction of the transcriptional repressor Bcl-6 in CD4(+) T cells is critical for the differentiation of follicular helper T cells (T(FH) cells), which are essential for B cell-mediated immunity. In contrast, the transcription factor Blimp1 (encoded by Prdm1) inhibits T(FH) differentiation by antagonizing Bcl-6. Here we found that the transcription factor TCF-1 was essential for both the initiation of T(FH) differentiation and the effector function of differentiated T(FH) cells during acute viral infection. Mechanistically, TCF-1 bound directly to the Bcl6 promoter and Prdm1 5' regulatory regions, which promoted Bcl-6 expression but repressed Blimp1 expression. TCF-1-null T(FH) cells upregulated genes associated with non-T(FH) cell lineages. Thus, TCF-1 functions as an important hub upstream of the Bcl-6-Blimp1 axis to initiate and secure the differentiation of T(FH) cells during acute viral infection.


Subject(s)
Cell Differentiation/immunology , DNA-Binding Proteins/immunology , Hepatocyte Nuclear Factor 1-alpha/immunology , Orthomyxoviridae Infections/immunology , T-Lymphocytes, Helper-Inducer/immunology , Transcription Factors/immunology , Animals , Cell Differentiation/genetics , DNA-Binding Proteins/genetics , Germinal Center/immunology , Germinal Center/metabolism , Hepatocyte Nuclear Factor 1-alpha/genetics , Influenza A virus , Mice , Mice, Knockout , Positive Regulatory Domain I-Binding Factor 1 , Proto-Oncogene Proteins c-bcl-6 , T-Lymphocytes, Helper-Inducer/metabolism , Transcription Factors/genetics
4.
Immunity ; 47(3): 538-551.e5, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28930662

ABSTRACT

Follicular regulatory T (Tfr) cells differentiate from conventional regulatory T (Treg) cells and suppress excessive germinal center (GC) responses by acting on both GC B cells and T follicular helper (Tfh) cells. Here, we examined the impact of mTOR, a serine/threonine protein kinase that senses and integrates diverse environmental cues, on the differentiation and functional competency of Tfr cells in response to protein immunization or viral infection. By genetically deleting Rptor or Rictor, essential components for mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2), respectively, we found that mTORC1 but not mTORC2 is essential for Tfr differentiation. Mechanistically, mTORC1-mediated phosphorylation of the transcription factor STAT3 induced the expression of the transcription factor TCF-1 by promoting STAT3 binding to the Tcf7 5'-regulatory region. Subsequently, TCF-1 bound to the Bcl6 promoter to induce Bcl6 expression, which launched the Tfr cell differentiation program. Thus, mTORC1 initiates Tfr cell differentiation by activating the TCF-1-Bcl-6 axis during immunization or infection.


Subject(s)
Immunomodulation , Multiprotein Complexes/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Biomarkers , Cell Differentiation/immunology , Cluster Analysis , Gene Expression Profiling , Hepatocyte Nuclear Factor 1-alpha/metabolism , Immunization , Immunophenotyping , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Transgenic , Multiprotein Complexes/genetics , Proto-Oncogene Proteins c-bcl-6/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/cytology , TOR Serine-Threonine Kinases/genetics
5.
Clin Infect Dis ; 76(3): e336-e341, 2023 02 08.
Article in English | MEDLINE | ID: mdl-35666466

ABSTRACT

BACKGROUND: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern (VOCs), especially the Delta and Omicron variants, have been reported to show significant resistance to approved neutralizing monoclonal antibodies (mAbs) and vaccines. We previously identified a mAb named 35B5 that harbors broad neutralization to SARS-CoV-2 VOCs. Herein, we explored the protection efficacy of a 35B5-based nasal spray against SARS-CoV-2 VOCs in a small-scale clinical trial. METHODS: We enrolled 30 healthy volunteers who were nasally administered the modified 35B5 formulation. At 12, 24, 48, and 72 hours after nasal spray, the neutralization efficacy of nasal mucosal samples was assayed with pseudoviruses coated with SARS-CoV-2 spike protein of the wild-type strain or the Alpha, Beta, Delta, or Omicron variants. RESULTS: The nasal mucosal samples collected within 24 hours after nasal spray effectively neutralized SARS-CoV-2 VOCs (including Delta and Omicron). Meanwhile, the protection efficacy was 60% effective and 20% effective at 48 and 72 hours after nasal spray, respectively. CONCLUSIONS: A single nasal spray of 35B5 formation conveys 24-hour effective protection against SARS-CoV-2 VOCs, including the Alpha, Beta, Delta, or Omicron variants. Thus, 35B5 nasal spray might be potential in strengthening SARS-CoV-2 prevention, especially in high-risk populations. CLINICAL TRIALS REGISTRATION: 2022-005-02-KY.


Subject(s)
COVID-19 , Humans , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing , Antibodies, Viral , COVID-19/prevention & control , Nasal Sprays , SARS-CoV-2/genetics
6.
Nature ; 537(7620): 412-428, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27501245

ABSTRACT

During chronic viral infection, virus-specific CD8(+) T cells become exhausted, exhibit poor effector function and lose memory potential. However, exhausted CD8(+) T cells can still contain viral replication in chronic infections, although the mechanism of this containment is largely unknown. Here we show that a subset of exhausted CD8(+) T cells expressing the chemokine receptor CXCR5 has a critical role in the control of viral replication in mice that were chronically infected with lymphocytic choriomeningitis virus (LCMV). These CXCR5(+) CD8(+) T cells were able to migrate into B-cell follicles, expressed lower levels of inhibitory receptors and exhibited more potent cytotoxicity than the CXCR5(-) [corrected] subset. Furthermore, we identified the Id2-E2A signalling axis as an important regulator of the generation of this subset. In patients with HIV, we also identified a virus-specific CXCR5(+) CD8(+) T-cell subset, and its number was inversely correlated with viral load. The CXCR5(+) subset showed greater therapeutic potential than the CXCR5(-) [corrected] subset when adoptively transferred to chronically infected mice, and exhibited synergistic reduction of viral load when combined with anti-PD-L1 treatment. This study defines a unique subset of exhausted CD8(+) T cells that has a pivotal role in the control of viral replication during chronic viral infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Germinal Center/cytology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Receptors, CXCR5/metabolism , Adoptive Transfer , Animals , B-Lymphocytes/immunology , Basic Helix-Loop-Helix Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/transplantation , Cell Differentiation , Chronic Disease , Female , Germinal Center/immunology , HIV Infections/immunology , HIV Infections/virology , Humans , Inhibitor of Differentiation Protein 2/metabolism , Lymphocytic choriomeningitis virus/growth & development , Male , Mice , Receptors, CXCR5/deficiency , Signal Transduction , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/transplantation , Viral Load/immunology , Virus Replication/immunology
7.
J Virol ; 91(4)2017 02 15.
Article in English | MEDLINE | ID: mdl-27974559

ABSTRACT

mTOR has important roles in regulation of both innate and adaptive immunity, but whether and how mTOR modulates humoral immune responses have yet to be fully understood. To address this issue, we examined the effects of rapamycin, a specific inhibitor of mTOR, on B cell and CD4 T cell responses during acute infection with lymphocytic choriomeningitis virus. Rapamycin treatment resulted in suppression of virus-specific B cell responses by inhibiting proliferation of germinal center (GC) B cells. In contrast, the number of memory CD4 T cells was increased in rapamycin-treated mice. However, the drug treatment caused a striking bias of CD4 T cell differentiation into Th1 cells and substantially impaired formation of follicular helper T (Tfh) cells, which are essential for humoral immunity. Further experiments in which mTOR signaling was modulated by RNA interference (RNAi) revealed that B cells were the primary target cells of rapamycin for the impaired humoral immunity and that reduced Tfh formation in rapamycin-treated mice was due to lower GC B cell responses that are essential for Tfh generation. Additionally, we found that rapamycin had minimal effects on B cell responses activated by lipopolysaccharide (LPS), which stimulates B cells in an antigen-independent manner, suggesting that rapamycin specifically inhibits B cell responses induced by B cell receptor stimulation with antigen. Together, these findings demonstrate that mTOR signals play an essential role in antigen-specific humoral immune responses by differentially regulating B cell and CD4 T cell responses during acute viral infection and that rapamycin treatment alters the interplay of immune cell subsets involved in antiviral humoral immunity. IMPORTANCE: mTOR is a serine/threonine kinase involved in a variety of cellular activities. Although its specific inhibitor, rapamycin, is currently used as an immunosuppressive drug in transplant patients, it has been reported that rapamycin can also stimulate pathogen-specific cellular immunity in certain circumstances. However, whether and how mTOR regulates humoral immunity are not well understood. Here we found that rapamycin treatment predominantly inhibited GC B cell responses during viral infection and that this led to biased helper CD4 T cell differentiation as well as impaired antibody responses. These findings suggest that inhibition of B cell responses by rapamycin may play an important role in regulation of allograft-specific antibody responses to prevent organ rejection in transplant recipients. Our results also show that consideration of antibody responses is required in cases where rapamycin is used to stimulate vaccine-induced immunity.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Host-Pathogen Interactions/immunology , Immunity, Humoral , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Apoptosis/drug effects , B-Lymphocyte Subsets/drug effects , Cell Line , Cell Survival/drug effects , Germinal Center/immunology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Immunization , Immunologic Memory , Immunomodulation/drug effects , Mice , Mice, Transgenic , Signal Transduction , Sirolimus/pharmacology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transduction, Genetic , Virus Diseases/immunology , Virus Diseases/metabolism
8.
Immunology ; 152(2): 276-286, 2017 10.
Article in English | MEDLINE | ID: mdl-28557002

ABSTRACT

The mammalian target of rapamycin (mTOR) is a serine-threonine kinase that has been shown to be essential for the differentiation and function of various immune cells. Earlier in vitro studies showed that mTOR signalling regulates B-cell biology by supporting their activation and proliferation. However, how mTOR signalling temporally regulates in vivo germinal centre B (GCB) cell development and differentiation into short-lived plasma cells, long-lived plasma cells and memory cells is still not well understood. In this study, we used a combined conditional/inducible knock-out system to investigate the temporal regulation of mTOR complex 1 (mTORC1) in the GCB cell response to acute lymphocytic choriomeningitis virus infection by deleting Raptor, a main component of mTORC1, specifically in B cells in pre- and late GC phase. Early Raptor deficiency strongly inhibited GCB cell proliferation and differentiation and plasma cell differentiation. Nevertheless, late GC Raptor deficiency caused only decreases in the size of memory B cells and long-lived plasma cells through poor maintenance of GCB cells, but it did not change their differentiation. Collectively, our data revealed that mTORC1 signalling supports GCB cell responses at both early and late GC phases during viral infection but does not regulate GCB cell differentiation into memory B cells and plasma cells at the late GC stage.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , B-Lymphocytes/enzymology , Germinal Center/enzymology , Lymphocytic Choriomeningitis/enzymology , Lymphocytic choriomeningitis virus/immunology , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/transplantation , B-Lymphocytes/virology , Bone Marrow Transplantation , Cell Differentiation , Cell Proliferation , Disease Models, Animal , Genetic Predisposition to Disease , Germinal Center/immunology , Germinal Center/virology , Host-Pathogen Interactions , Immunity, Humoral , Immunologic Memory , Lymphocyte Activation , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/pathogenicity , Mechanistic Target of Rapamycin Complex 1 , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes/deficiency , Multiprotein Complexes/genetics , Multiprotein Complexes/immunology , Phenotype , Plasma Cells/enzymology , Plasma Cells/immunology , Plasma Cells/virology , Regulatory-Associated Protein of mTOR , Signal Transduction , TOR Serine-Threonine Kinases/deficiency , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/immunology , Time Factors , Transplantation Chimera
10.
Curr Med Imaging ; 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38874027

ABSTRACT

BACKGROUND: Accurate identification of vascular lumen region founded the base of bubble detection and bubble grading, which played a significant role in the detection of vascular gas emboli for the diagnosis of decompression sickness. OBJECTIVES: To assist in the detection of vascular bubbles, it is crucial to develop an automatic algorithm that could identify vascular lumen areas in ultrasound videos with the interference of bubble presence. METHODS: This article proposed an automated vascular lumen region recognition (VLRR) algorithm that could sketch the accurate boundary between vessel lumen and tissues from dynamic 2D ultrasound videos. It adopts 2D ultrasound videos of the lumen area as input and outputs the frames with circled vascular lumen boundary of the videos. Normalized cross-correlation method, distance transform technique, and region growing technique were adopted in this algorithm. Results A double-blind test was carried out to test the recognition accuracy of the algorithm on 180 samples in the images of 6 different grades of bubble videos, during which, intersection over union and pixel accuracy were adopted as evaluation metrics. The average IOU on the images of different bubble grades reached 0.76. The mean PA on 6 of the images of bubble grades reached 0.82. CONCLUSION: It is concluded that the proposed method could identify the vascular lumen with high accuracy, potentially applicable to assist clinicians in the measurement of the severity of vascular gas emboli in clinics.

11.
J Vis Exp ; (203)2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38345257

ABSTRACT

Tumor antigen-specific CD8+ T cells from draining lymph nodes gain an accumulating importance in mounting anti-tumor immune response during tumorigenesis. However, in many cases, cancer cells form metastatic loci in lymph nodes before further metastasizing to distant organs. To what extent the local and systematic CD8+ T cell responses were influenced by LN metastasis remains obscure. To this end, we set up a murine LN metastasis model combined with a B16F10-GP melanoma cell line expressing the surrogate neoantigen derived from lymphocytic choriomeningitis virus (LCMV), glycoprotein (GP), and P14 transgenic mice harboring T cell receptors (TCRs) specific to GP-derived peptide GP33-41 presented by the class I major histocompatibility complex (MHC) molecule H-2Db. This protocol enables the study of antigen-specific CD8+ T cell responses during LN metastasis. In this protocol, C57BL/6J mice were subcutaneously implanted with B16F10-GP cells, followed by adoptive transfer with naive P14 cells. When the subcutaneous tumor grew to approximately 5 mm in diameter, the primary tumor was excised, and B16F10-GP cells were directly injected into the tumor draining lymph node (TdLN). Then, the dynamics of CD8+ T cells were monitored during the process of LN metastasis. Collectively, this model has provided an approach to precisely investigate the antigen-specific CD8+ T cell immune responses during LN metastasis.


Subject(s)
Antigens , CD8-Positive T-Lymphocytes , Mice , Animals , Lymphatic Metastasis , Mice, Inbred C57BL , Mice, Transgenic , Antigens/metabolism , Lymphocytic choriomeningitis virus , Glycoproteins/metabolism , Carcinogenesis/metabolism , Lymph Nodes
12.
Nat Cancer ; 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38609488

ABSTRACT

Tumor-specific T cells are crucial in anti-tumor immunity and act as targets for cancer immunotherapies. However, these cells are numerically scarce and functionally exhausted in the tumor microenvironment (TME), leading to inefficacious immunotherapies in most patients with cancer. By contrast, emerging evidence suggested that tumor-irrelevant bystander T (TBYS) cells are abundant and preserve functional memory properties in the TME. To leverage TBYS cells in the TME to eliminate tumor cells, we engineered oncolytic virus (OV) encoding TBYS epitopes (OV-BYTE) to redirect the antigen specificity of tumor cells to pre-existing TBYS cells, leading to effective tumor inhibition in multiple preclinical models. Mechanistically, OV-BYTE induced epitope spreading of tumor antigens to elicit more diverse tumor-specific T cell responses. Remarkably, the OV-BYTE strategy targeting human severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific T cell memory efficiently inhibited tumor progression in a human tumor cell-derived xenograft model, providing important insights into the improvement of cancer immunotherapies in a large population with a history of SARS-CoV-2 infection or coronavirus disease 2019 (COVID-19) vaccination.

13.
Yi Chuan ; 35(9): 1049-57, 2013 Sep.
Article in Zh | MEDLINE | ID: mdl-24400477

ABSTRACT

According to the types of stem cells and considering tumor evolution, one of the most significant theories about stem cells is derived from cancer stem cells (CSCs), which, similar to normal adult stem cells, possess the capacity of self-renewal and potential of differentiation. Over the past few years, compelling evidence has emerged in support of the CSC model for many tumors. The CSCs are posited to be responsible not only for tumor initiation but also for tumor metastasis, relapse and therapyresistance. Thus, understanding the mechanisms that govern the generation and maintenance of this special population of cells is of great importance. Despite the current progress in basic genetic research, the latest work implies that epigenetic mechanisms, from DNA methylation, histone modifications and chromatin-remodeling to the wide discovered miRNAs, play critical roles in the regulation of CSC features. This review focuses on the key epigenetic mechanisms that regulate and define the unique CSC properties.


Subject(s)
Epigenesis, Genetic , Neoplasms/genetics , Neoplastic Stem Cells/metabolism , Animals , DNA Methylation , Humans , Neoplasms/metabolism , Neoplasms/physiopathology
14.
Vaccine ; 41(34): 4986-4995, 2023 07 31.
Article in English | MEDLINE | ID: mdl-37400286

ABSTRACT

The COVID-19 vaccinations are crucial in protecting against the global pandemic. However, accumulating studies revealed the severely blunted COVID-19 vaccine effectiveness in cancer patients. The PD-1/PD-L1 immune checkpoint blockade (ICB) therapy leads to durable therapeutic responses in a subset of cancer patients and has been approved to treat a wide spectrum of cancers in the clinic. In this regard, it is pivotal to explore the potential impact of PD-1/PD-L1 ICB therapy on COVID-19 vaccine effectiveness during ongoing malignancy. In this study, using preclinical models, we found that the tumor-suppressed COVID-19 vaccine responses are largely reverted in the setting of PD-1/PD-L1 ICB therapy. We also identified that the PD-1/PD-L1 blockade-directed restoration of COVID-19 vaccine effectiveness is irrelevant to anti-tumor therapeutic outcomes. Mechanistically, the restored COVID-19 vaccine effectiveness is entwined with the PD-1/PD-L1 blockade-driven preponderance of follicular helper T cell and germinal center responses during ongoing malignancy. Thus, our findings indicate that PD-1/PD-L1 blockade will greatly normalize the responses of cancer patients to COVID-19 vaccination, while regardless of its anti-tumor efficacies on these patients.


Subject(s)
COVID-19 , Neoplasms , Humans , COVID-19 Vaccines , Immune Checkpoint Inhibitors/therapeutic use , B7-H1 Antigen , Programmed Cell Death 1 Receptor , COVID-19/prevention & control , Neoplasms/therapy , Immunotherapy
15.
Front Immunol ; 13: 875718, 2022.
Article in English | MEDLINE | ID: mdl-35784297

ABSTRACT

Cytotoxic CD8+ T cells are the main focus of efforts to understand anti-tumor immunity and immunotherapy. The adoptive transfer of tumor-reactive cytotoxic CD8+ T lymphocytes expanded and differentiated in vitro has long been considered the primary strategy in adaptive anti-tumor immunity, however, the majority of the transferred tumor antigen-specific CD8+ T cells differentiated into CD39+CD69+ exhausted progenies, limiting its effects in repressing tumor growth. Contrarily, less attention has been addressed to the role of CD4+ T cells during tumorigenesis. Using a mouse model of metastatic melanoma, we found that transferring tumor-specific CD4+ T cells into recipients induces substantial regression of the established metastatic tumors. Notably, in vitro activated CD4+ T cells developed into cytotoxic CD4- T cells in vivo and get exhausted gradually. The blockade of PD-L1 signaling resulted in an expansion of tumor specific CD4+ T cells, which could better control the established metastatic melanoma. Moreover, the tumor-specific memory CD4+ T cell can prevent mice from tumor metastasis, and the tumor-specific effector CD4+ T cells can also mitigate the established metastatic tumor. Overall, our findings suggest a novel mechanism of CD4+ T cells in curtailing tumor metastasis and confirm their therapeutic role in combination with PD-L1 blockade in cancer immunotherapy. Hence, a better understanding of cytotoxic CD4- T cell-mediated tumor regression could provide an alternative choice for patients exhibiting suboptimal or no response to CD8+ T cell-based immunotherapies.


Subject(s)
Antineoplastic Agents , Melanoma , Neoplasms, Second Primary , B7-H1 Antigen , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Humans
16.
J Immunol Methods ; 505: 113266, 2022 06.
Article in English | MEDLINE | ID: mdl-35398062

ABSTRACT

Follicular regulatory T cells (Tfrs), a specialized subset of regulatory T cells (Tregs), have a particular role in the control of follicular helper T cell-driven germinal center (GC) responses. Following differentiation signals similar to those received by follicular helper T cells (Tfhs), Tfrs gain expression of characteristic chemokine receptors and transcription factors, such as CXCR5 and Bcl-6, allowing them to migrate into the B-cell follicle and perform in situ suppression. Thus, together with Tfhs, Tfrs help maintaining an optimized GC-reaction. However, the mechanism underlying the Treg-to-Tfr transition remains obscure. Here, we established a highly reproducible protocol for investigating the differentiation of Tregs into Tfrs by constructing spleen-chimeric mice combined with retrovirus transduction. We demonstrated that using this strategy, over 4 folds of Tregs could differentiate into Tfrs in Bcl-6 overexpression group compared to control counterparts (Migr1), and Bcl-6 could efficiently promote Tfr differentiation during acute viral infection. Hence, this method provides us an easy access to investigate the factors that regulate the differentiation program that converts Tregs into Tfrs, which will enhance our understanding of the networks regulating GC-reaction and shed new light on the molecular basis of immune homeostasis.


Subject(s)
T-Lymphocytes, Regulatory , Virus Diseases , Animals , B-Lymphocytes , Germinal Center , Mice , Receptors, CXCR5/metabolism , T-Lymphocytes, Helper-Inducer , Virus Diseases/metabolism
17.
MedComm (2020) ; 3(1): e111, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35281793

ABSTRACT

Exhausted CD8+ T (Tex) cells are caused by persistent antigenic stimulation during chronic viral infection or tumorigenesis. Tex cells upregulate and sustain the expressions of multiple immune inhibitory receptors (IRs). Blocking IRs of Tex cells, exemplified by PD-1, can partially restore their effector functions and thus lead to viral suppression or tumor remission. Tex cells derived from chronic viral infections share the expression spectrum of IRs with Tex cells derived from tumors; however, whether any IRs are selectively expressed by tumor-derived Tex cells or virus-derived Tex cells remains to be learnt. In the study, we found that Tex cells upregulate IR natural killer cell lectin-like receptor isoform A (NKG2A) specifically in the context of tumor but not chronic viral infection. Moreover, the NKG2A expression is attributed to tumor antigen recognition and thus bias expressed by tumor-specific Tex cells in the tumor microenvironment instead of their counterparts in the periphery. Such dichotomous NKG2A expression further dictates the differential responsiveness of Tex cells to NKG2A immune checkpoint blockade. Therefore, our study highlighted NKG2A as a disease-dependent IR and provided novel insights into the distinct regulatory mechanisms underlying T cell exhaustion between tumor and chronic viral infection.

18.
J Immunother Cancer ; 10(5)2022 05.
Article in English | MEDLINE | ID: mdl-35580929

ABSTRACT

BACKGROUND: Antitumor therapeutic vaccines are generally based on antigenic epitopes presented by major histocompatibility complex (MHC-I) molecules to induce tumor-specific CD8+ T cells. Paradoxically, continuous T cell receptor (TCR) stimulation from tumor-derived CD8+ T-cell epitopes can drive the functional exhaustion of tumor-specific CD8+ T cells. Tumor-specific type-I helper CD4+ T (TH1) cells play an important role in the population maintenance and cytotoxic function of exhausted tumor-specific CD8+ T cells in the tumor microenvironment. Nonetheless, whether the vaccination strategy targeting MHC-II-restricted CD4+ T-cell epitopes to induce tumor-specific TH1 responses can confer effective antitumor immunity to restrain tumor growth is not well studied. Here, we developed a heterologous prime-boost vaccination strategy to effectively induce tumor-specific TH1 cells and evaluated its antitumor efficacy and its capacity to potentiate PD-1/PD-L1 immunotherapy. METHODS: Listeria monocytogenes vector and influenza A virus (PR8 strain) vector stably expressing lymphocytic choriomeningitis virus (LCMV) glycoprotein-specific I-Ab-restricted CD4+ T cell epitope (GP61-80) or ovalbumin-specific CD4+ T cell epitope (OVA323-339) were constructed and evaluated their efficacy against mouse models of melanoma and colorectal adenocarcinoma expressing lymphocytic choriomeningitis virus glycoprotein and ovalbumin. The impact of CD4+ T cell epitope-based heterologous prime-boost vaccination was detected by flow-cytometer, single-cell RNA sequencing and single-cell TCR sequencing. RESULTS: CD4+ T cell epitope-based heterologous prime-boost vaccination efficiently suppressed both mouse melanoma and colorectal adenocarcinoma. This vaccination primarily induced tumor-specific TH1 response, which in turn enhanced the expansion, effector function and clonal breadth of tumor-specific CD8+ T cells. Furthermore, this vaccination strategy synergized PD-L1 blockade mediated tumor suppression. Notably, prime-boost vaccination extended the duration of PD-L1 blockade induced antitumor effects by preventing the re-exhaustion of tumor-specific CD8+ T cells. CONCLUSION: CD4+ T cell epitope-based heterologous prime-boost vaccination elicited potent both tumor-specific TH1 and CTL response, leading to the efficient tumor control. This strategy can also potentiate PD-1/PD-L1 immune checkpoint blockade (ICB) against cancer.


Subject(s)
Adenocarcinoma , Colorectal Neoplasms , Melanoma , Animals , B7-H1 Antigen/pharmacology , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Epitopes, T-Lymphocyte , Glycoproteins , Humans , Immune Checkpoint Inhibitors , Immunotherapy , Mice , Ovalbumin , Programmed Cell Death 1 Receptor , Receptors, Antigen, T-Cell , Tumor Microenvironment , Vaccination
19.
Signal Transduct Target Ther ; 7(1): 114, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35383141

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a global pandemic of novel coronavirus disease (COVID-19). The neutralizing monoclonal antibodies (mAbs) targeting the receptor-binding domain (RBD) of SARS-CoV-2 are among the most promising strategies to prevent and treat COVID-19. However, SARS-CoV-2 variants of concern (VOCs) profoundly reduced the efficacies of most of mAbs and vaccines approved for clinical use. Herein, we demonstrated mAb 35B5 efficiently neutralizes both wild-type (WT) SARS-CoV-2 and VOCs, including B.1.617.2 (delta) variant, in vitro and in vivo. Cryo-electron microscopy (cryo-EM) revealed that 35B5 neutralizes SARS-CoV-2 by targeting a unique epitope that avoids the prevailing mutation sites on RBD identified in circulating VOCs, providing the molecular basis for its pan-neutralizing efficacy. The 35B5-binding epitope could also be exploited for the rational design of a universal SARS-CoV-2 vaccine.


Subject(s)
Antibodies, Monoclonal , Antibodies, Viral , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Antibodies, Monoclonal/chemistry , Antibodies, Viral/chemistry , COVID-19 , Cryoelectron Microscopy , Humans , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/immunology
20.
Front Immunol ; 12: 751584, 2021.
Article in English | MEDLINE | ID: mdl-34630430

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a global pandemic of novel coronavirus disease (COVID-19). Though vaccines and neutralizing monoclonal antibodies (mAbs) have been developed to fight COVID-19 in the past year, one major concern is the emergence of SARS-CoV-2 variants of concern (VOCs). Indeed, SARS-CoV-2 VOCs such as B.1.1.7 (UK), B.1.351 (South Africa), P.1 (Brazil), and B.1.617.1 (India) now dominate the pandemic. Herein, we found that binding activity and neutralizing capacity of sera collected from convalescent patients in early 2020 for SARS-CoV-2 VOCs, but not non-VOC variants, were severely blunted. Furthermore, we observed evasion of SARS-CoV-2 VOCs from a VH3-30 mAb 32D4, which was proved to exhibit highly potential neutralization against wild-type (WT) SARS-CoV-2. Thus, these results indicated that SARS-CoV-2 VOCs might be able to spread in convalescent patients and even harbor resistance to medical countermeasures. New interventions against these SARS-CoV-2 VOCs are urgently needed.


Subject(s)
COVID-19/immunology , Mutation/genetics , SARS-CoV-2/physiology , Adult , Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/metabolism , Antibodies, Viral/metabolism , COVID-19/genetics , COVID-19/therapy , Female , Humans , Immune Evasion , Immunization, Passive , Male , Middle Aged , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/metabolism , COVID-19 Serotherapy
SELECTION OF CITATIONS
SEARCH DETAIL