Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Stem Cells Int ; 2023: 8667045, 2023.
Article in English | MEDLINE | ID: mdl-36895785

ABSTRACT

Blood-brain barrier (BBB) is the most important component of central nervous system (CNS) to keep toxins and pathogens from CNS. Although our studies demonstrated that using interleukin-6 antibodies (IL-6-AB) reversed the increased permeability of BBB, IL-6-AB is limited in their application that only could be used a few hours before surgery and seemed delayed the surgical wounds healing process, which urges us to find another more effective method. In this study, we employed the C57BL/6J female mice to investigate the potential effects of umbilical cord-derived mesenchymal stem cells (UC-MSCs) transplantation on BBB dysfunction induced by surgical wound. Compared to IL-6-AB, the transplantation of UC-MSCs more effectively decreased the BBB permeability after surgical wound evaluated by dextran tracer (immunofluorescence imaging and luorescence quantification). In addition, UC-MSCs can largely decrease the ratio of proinflammatory cytokine IL-6 to the anti-inflammatory cytokine IL-10 in both serum and brain tissue after surgical wound. Moreover, UC-MSCs successfully increased the levels of tight junction proteins (TJs) in BBB such as ZO-1, Occludin, and Claudin-5 and extremely decreased the level of matrix metalloproteinase-9 (MMP-9). Interestingly, UC-MSCs treatment also had positive effects on wound healing while protecting the BBB dysfunction induced by surgical wound compared to IL-6-AB treatment. These findings suggest that UC-MSCs transplantation is a highly efficient and promising approach on protecting the integrity of BBB which caused by peripheral traumatic injuries.

2.
Front Immunol ; 14: 1092668, 2023.
Article in English | MEDLINE | ID: mdl-36891306

ABSTRACT

Chronic skin inflammatory diseases including atopic dermatitis (AD) and psoriasis have been considered uncontrolled inflammatory responses, which have usually troubled patients around the world. Moreover, the recent method to treat AD and psoriasis has been based on the inhibition, not regulation, of the abnormal inflammatory response, which can induce a number of side effects and drug resistance in long-term treatment. Mesenchymal stem/stromal cells (MSCs) and their derivatives have been widely used in immune diseases based on their regeneration, differentiation, and immunomodulation with few adverse effects, which makes MSCs a promising treatment for chronic skin inflammatory diseases. As a result, in this review, we aim to systematically discuss the therapeutic effects of various resources of MSCs, the application of preconditioning MSCs and engineering extracellular vesicles (EVs) in AD and psoriasis, and the clinical evaluation of the administration of MSCs and their derivatives, which can provide a comprehensive vision for the application of MSCs and their derivatives in future research and clinical treatment.


Subject(s)
Dermatitis, Atopic , Mesenchymal Stem Cells , Psoriasis , Skin Diseases , Humans , Dermatitis, Atopic/therapy , Skin , Psoriasis/therapy
3.
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
4.
Front Oncol ; 12: 1078272, 2022.
Article in English | MEDLINE | ID: mdl-36727056

ABSTRACT

Background: Trichilemmal carcinoma (TLC) is a rare malignant cutaneous adnexal neoplasm, with no relatively comprehensive research. Objective: The aim of this study is to perform an updated statistical analysis so as to better understand TLC's epidemiology, clinical features, diagnosis, and treatment. Methods: The diagnosis and treatment of three TLC cases in our department were summarized. Then, all TLC cases published in the literature were retrieved for a comprehensive analysis, followed by the analysis of global trends and regional distribution, demographic characteristics, clinical features, pathogenesis, histopathological features, and treatment and prognosis of TLC. Results: Of the 231 cases, the incidence of TLC has shown an upward trend recently, especially in China, in Asia. The susceptible population is men aged 60-80 and women over 80, and the most prone location is head and neck. The phenotype of TLC is not always typical and may be misdiagnosed because of the coexistence of other diseases. There is a linear relationship between the diameter and its duration or thickness. UV, locally present skin lesions, trauma, scarring, organ transplantation, and genetic disorders may trigger the occurrence of TLC. Periodic acid-Schiff staining and CD34, but not Epithelial Membrane Antigen (EMA), were helpful in the diagnosis of TLC. Although effective, surgical excision and Mohs micrographic surgery need further improvement to reduce recurrence of TLC. Carcinoma history is an independent risk factor for TLC recurrence. Limitations: The limitation of this study is the lack of randomized controlled trial on TLC treatment and recurrence. Conclusion: TLC has the possibility of invasive growth and recurrence, especially in patients with longer duration and carcinoma history.

5.
Crit Care ; 24(1): 647, 2020 11 16.
Article in English | MEDLINE | ID: mdl-33198786

ABSTRACT

BACKGROUND: Coronavirus disease 2019 (COVID-19), a highly infectious disease, has been rapidly spreading all over the world and remains a great threat to global public health. Patients diagnosed with severe or critical cases have a poor prognosis. Hence, it is crucial for us to identify potentially severe or critical cases early and give timely treatments for targeted patients. In the clinical practice of treating patients with COVID-19, we have observed that the neutrophil-to-lymphocyte ratio (NLR) of severe patients is higher than that in mild patients. We performed this systematic review and meta-analysis to evaluate the predictive values of NLR on disease severity and mortality in patients with COVID-19. METHODS: We searched PubMed, EMBASE, China National Knowledge Infrastructure (CNKI) and Wanfang databases to identify eligible studies (up to August 11, 2020). Two authors independently screened studies and extracted data. The methodological quality of the included studies was assessed by Quality Assessment of Diagnostic Accuracy Studies 2 (QUADAS-2). RESULTS: Thirteen studies involving 1579 patients reported the predictive value of NLR on disease severity. The pooled sensitivity (SEN), specificity (SPE) and area under curve (AUC) were 0.78 (95% CI 0.70-0.84), 0.78 (95% CI 0.73-0.83) and 0.85 (95% CI 0.81-0.88), respectively. Ten studies involving 2967 patients reported the predictive value of NLR on mortality. The pooled SEN, SPE and AUC were 0.83 (95% CI 0.75-0.89), 0.83 (95% CI 0.74-0.89) and 0.90 (95% CI 0.87-0.92), respectively. CONCLUSIONS: NLR has good predictive values on disease severity and mortality in patients with COVID-19 infection. Evaluating NLR can help clinicians identify potentially severe cases early, conduct early triage and initiate effective management in time, which may reduce the overall mortality of COVID-19. TRIAL REGISTRY: This meta-analysis was prospectively registered on PROSPERO database (Registration number: CRD42020203612).


Subject(s)
Betacoronavirus , Coronavirus Infections/blood , Coronavirus Infections/diagnosis , Lymphocytes/metabolism , Neutrophils/metabolism , Pneumonia, Viral/blood , Pneumonia, Viral/diagnosis , Severity of Illness Index , COVID-19 , Coronavirus Infections/mortality , Humans , Mortality/trends , Pandemics , Pneumonia, Viral/mortality , SARS-CoV-2
6.
Cell Mol Immunol ; 17(3): 247-260, 2020 03.
Article in English | MEDLINE | ID: mdl-30842630

ABSTRACT

Epigenetic modifications to histones dictate the differentiation of naïve CD4+ T cells into different subsets of effector T helper (TH) cells. The histone methyltransferase enhancer of zeste homolog 2 (EZH2) has been implicated in the mechanism regulating the differentiation of TH1, TH2 and regulatory T (Treg) cells. However, whether and how EZH2 regulates follicular helper T (TFH) cell differentiation remain unknown. Using a mouse model of acute lymphocytic choriomeningitis virus (LCMV) infection, we observed abundant EZH2 expression and associated H3K27me3 modifications preferentially in the early committed virus-specific TFH cells compared to those in TH1 cells. Ablation of EZH2 in LCMV-specific CD4+ T cells leads to a selective impairment of early TFH cell fate commitment, but not late TFH differentiation or memory TFH maintenance. Mechanistically, EZH2 specifically stabilizes the chromatin accessibility of a cluster of genes that are important for TFH fate commitment, particularly B cell lymphoma 6 (Bcl6), and thus directs TFH cell commitment. Therefore, we identified the chromatin-modifying enzyme EZH2 as a novel regulator of early TFH differentiation during acute viral infection.


Subject(s)
Cell Differentiation/immunology , Enhancer of Zeste Homolog 2 Protein/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , T-Lymphocytes, Helper-Inducer/immunology , Acute Disease , Animals , Cell Differentiation/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/pathology , Mice , Mice, Transgenic
7.
Biomater Sci ; 7(6): 2533-2544, 2019 May 28.
Article in English | MEDLINE | ID: mdl-30968875

ABSTRACT

Cytotoxic CD8+ T cells (CTLs) are crucial for controlling intracellular pathogens as well as cancer. However, how to promote the cytotoxic activity of CTL cells in vitro and in vivo remains largely unknown. On the other hand, ceria nanoparticles (CNPs) are widely used in biomedical fields, but the role of CNPs in CTL cells is still unclear. In this study, we found that the activated antigen-specific (P14) and nonspecific CD8+ T cells with CNP treatment both produced more cytokines, including interleukin-2 (IL-2) and tumor necrosis factor-α (TNF-α), and released more effector molecules, such as granzyme B and perforin, and then exhibited higher killing activity of P14 cells in vitro and stronger viral clearance capacity of CTL cells in vivo. Mechanistically, the activated P14 cells with CNP treatment inhibited the production of reactive oxygen species, and therefore promoted the activity of NF-κB signaling. Importantly, while the P14 cells were simultaneously treated by IMD-0354, a specific inhibitor of NF-κB signaling, the increases of IL-2 and TNF-α productions and granzyme B and perforin releases were remedied, and the P14 cells eventually exhibited the natural killing activity in vitro. Thus, our results demonstrated that CNP treatment promoted the cytotoxic activity of CTL cells and provide new ideas in the usage of CNPs and fascinating pharmacological potentials for clinical application, especially cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , Cerium/chemistry , Cerium/pharmacology , Metal Nanoparticles/chemistry , NF-kappa B/metabolism , Signal Transduction/drug effects , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytokines/biosynthesis , Female , Hydrophobic and Hydrophilic Interactions , Ligands , Male , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism
8.
Front Immunol ; 10: 606, 2019.
Article in English | MEDLINE | ID: mdl-30984183

ABSTRACT

Follicular helper T cells (TFH cells), known as the primary "helpers" of the germinal center (GC) reaction, promote the humoral immune response to defend against various pathogens. Under conditions of infection by different types of pathogens, many shared transcription factors (TFs), such as Bcl-6, TCF-1, and Maf, are selectively enriched in pathogen-specific TFH cells, orchestrating TFH cell differentiation and function. In addition, TFH cells also coexpress environmentally associated TFs as their conventional T cell counterparts (such as T-bet, GATA-3, or ROR-γt, which are expressed in Th1, Th2, or Th17 cells, respectively). These features likely indicate both the lineage-specificity and environmental adaption of the TFH cell responses. However, the extent to which the TFH cell response relies on these environmentally specific TFs is not completely understood. Here, we found that T-bet was specifically expressed in Type I TFH cells but not Type II TFH cells. While dispensable for the early fate commitment of TFH cells, T-bet was essential for the maintenance of differentiated TFH cells, promoting their proliferation, and inhibiting their apoptosis during acute viral infection. Microarray analysis showed both similarities and differences in transcriptome dependency on T-bet in TFH and TH1 cells, suggesting the distinctive role of T-bet in TFH cells. Collectively, our findings reveal an important and specific supporting role for T-bet in type I TFH cell response, which can help us gain a deeper understanding of TFH cell subsets.


Subject(s)
Gene Expression Regulation/immunology , Germinal Center/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , T-Box Domain Proteins/immunology , T-Lymphocytes, Helper-Inducer/immunology , Acute Disease , Animals , Apoptosis/genetics , Apoptosis/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Germinal Center/pathology , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus/genetics , Mice , Mice, Knockout , T-Box Domain Proteins/genetics , T-Lymphocytes, Helper-Inducer/pathology
9.
Nature ; 567(7749): 525-529, 2019 03.
Article in English | MEDLINE | ID: mdl-30814730

ABSTRACT

T cells become dysfunctional when they encounter self antigens or are exposed to chronic infection or to the tumour microenvironment1. The function of T cells is tightly regulated by a combinational co-stimulatory signal, and dominance of negative co-stimulation results in T cell dysfunction2. However, the molecular mechanisms that underlie this dysfunction remain unclear. Here, using an in vitro T cell tolerance induction system in mice, we characterize genome-wide epigenetic and gene expression features in tolerant T cells, and show that they are distinct from effector and regulatory T cells. Notably, the transcription factor NR4A1 is stably expressed at high levels in tolerant T cells. Overexpression of NR4A1 inhibits effector T cell differentiation, whereas deletion of NR4A1 overcomes T cell tolerance and exaggerates effector function, as well as enhancing immunity against tumour and chronic virus. Mechanistically, NR4A1 is preferentially recruited to binding sites of the transcription factor AP-1, where it represses effector-gene expression by inhibiting AP-1 function. NR4A1 binding also promotes acetylation of histone 3 at lysine 27 (H3K27ac), leading to activation of tolerance-related genes. This study thus identifies NR4A1 as a key general regulator in the induction of T cell dysfunction, and a potential target for tumour immunotherapy.


Subject(s)
Gene Expression Regulation/genetics , Genome , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Acetylation , Animals , Arenaviridae Infections/immunology , Arenaviridae Infections/virology , Cell Line, Tumor , Colitis/immunology , Colitis/pathology , Colitis/therapy , Epigenesis, Genetic , Female , Histones/chemistry , Histones/metabolism , Immune Tolerance/genetics , Immunotherapy , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , T-Lymphocytes/immunology , Transcription Factor AP-1/metabolism , Transcription, Genetic
10.
Front Immunol ; 10: 169, 2019.
Article in English | MEDLINE | ID: mdl-30814995

ABSTRACT

The long-term persistence of viral antigens drives virus-specific CD8 T cell exhaustion during chronic viral infection. Yet exhausted, CD8 T cells are still endowed with certain levels of effector function, by which they can keep viral replication in check in chronic infection. However, the regulatory factors involved in regulating the effector function of exhausted CD8 T cell are largely unknown. Using mouse model of chronic LCMV infection, we found that the deletion of transcription factor TCF-1 in LCMV-specific exhausted CD8 T cells led to the profound reduction in cytokine production and degranulation. Conversely, ectopic expression of TCF-1 or using agonist to activate TCF-1 activities promotes the effector function of exhausted CD8 T cells. Mechanistically, TCF-1 fuels the functionalities of exhausted CD8 T cells by promoting the expression of an array of key effector function-associated transcription regulators, including Foxo1, Zeb2, Id3, and Eomes. These results collectively indicate that targeting TCF-1 mediated transcriptional pathway may represent a promising immunotherapy strategy against chronic viral infections by reinvigorating the effector function of exhausted virus-specific CD8 T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , T Cell Transcription Factor 1/metabolism , Virus Diseases/etiology , Virus Diseases/metabolism , Adoptive Transfer , Animals , Cell Survival , Chronic Disease , Cytokines/metabolism , Disease Models, Animal , Gene Expression , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/metabolism , Lymphocytic Choriomeningitis/therapy , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Transgenic , T Cell Transcription Factor 1/genetics , Transplantation Chimera , Viral Load , Virus Diseases/therapy
11.
Nat Med ; 24(10): 1536-1544, 2018 10.
Article in English | MEDLINE | ID: mdl-30297899

ABSTRACT

Impaired immunity in patients with late-stage cancer is not limited to antitumor responses, as demonstrated by poor vaccination protection and high susceptibility to infection1-3. This has been largely attributed to chemotherapy-induced impairment of innate immunity, such as neutropenia2, whereas systemic effects of tumors on hematopoiesis and adoptive immunity remain incompletely understood. Here we observed anemia associated with severe deficiency of CD8+ T cell responses against pathogens in treatment-naive mice bearing large tumors. Specifically, we identify CD45+ erythroid progenitor cells (CD71+TER119+; EPCs) as robust immunosuppressors. CD45+ EPCs, induced by tumor growth-associated extramedullary hematopoiesis, accumulate in the spleen to become a major population, outnumbering regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs). The CD45+ EPC transcriptome closely resembles that of MDSCs, and, like MDSCs, reactive oxygen species production is a major mechanism underlying CD45+ EPC-mediated immunosuppression. Similarly, an immunosuppressive CD45+ EPC population was detected in patients with cancer who have anemia. These findings identify a major population of immunosuppressive cells that likely contributes to the impaired T cell responses commonly observed in patients with advanced cancer.


Subject(s)
Anemia/immunology , Erythroid Precursor Cells/immunology , Myeloid-Derived Suppressor Cells/immunology , Sarcoma, Myeloid/immunology , Anemia/genetics , Anemia/pathology , Animals , Antigens, CD/immunology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Erythroid Precursor Cells/metabolism , Erythroid Precursor Cells/pathology , Humans , Immune Tolerance , Immunity, Innate/genetics , Leukocyte Common Antigens/immunology , Mice , Neoplasm Staging , Reactive Oxygen Species/metabolism , Receptors, Transferrin/immunology , Sarcoma, Myeloid/metabolism , Sarcoma, Myeloid/pathology , T-Lymphocytes, Regulatory/immunology , Xenograft Model Antitumor Assays
12.
Front Immunol ; 9: 1095, 2018.
Article in English | MEDLINE | ID: mdl-29872434

ABSTRACT

The combination antiretroviral therapeutic (cART) regime effectively suppresses human immunodeficiency virus (HIV) replication and prevents progression to acquired immunodeficiency diseases. However, cART is not a cure, and viral rebound will occur immediately after treatment is interrupted largely due to the long-term presence of an HIV reservoir that is composed of latently infected target cells that maintain a quiescent state or persistently produce infectious viruses. CD4 T cells that reside in B-cell follicles within lymphoid tissues, called follicular helper T cells (TFH), have been identified as a major HIV reservoir. Due to their specialized anatomical structure, HIV-specific CD8 T cells are largely insulated from this TFH reservoir. It is increasingly clear that the elimination of TFH reservoirs is a key step toward a functional cure for HIV infection. Recently, several studies have suggested that a fraction of HIV-specific CD8 T cells can differentiate into a CXCR5-expressing subset, which are able to migrate into B-cell follicles and inhibit viral replication. In this review, we discuss the differentiation and functions of this newly identified CD8 T-cell subset and propose potential strategies for purging TFH HIV reservoirs by utilizing this unique population.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , HIV Infections/immunology , HIV/immunology , Host-Pathogen Interactions , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Communication/immunology , Chronic Disease , Gene Expression , HIV Infections/metabolism , HIV Infections/virology , Host-Pathogen Interactions/immunology , Humans , Receptors, CXCR5/genetics , Receptors, CXCR5/metabolism , T-Cell Antigen Receptor Specificity , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/virology
13.
Cell Mol Immunol ; 15(9): 815-826, 2018 09.
Article in English | MEDLINE | ID: mdl-28287115

ABSTRACT

CD4+ T cells are essential for sustaining CD8+ T cell responses during a chronic infection. The adoptive transfer of virus-specific CD4+ T cells has been shown to efficiently rescue exhausted CD8+ T cells. However, the question of whether endogenous virus-specific CD4+ T cell responses can be enhanced by certain vaccination strategies and subsequently reinvigorate exhausted CD8+ T cells remains unexplored. In this study, we developed a CD4+ T cell epitope-based heterologous prime-boost immunization strategy and examined the efficacy of this strategy using a mouse model of chronic lymphocytic choriomeningitis virus (LCMV) infection. We primed chronically LCMV-infected mice with a Listeria monocytogenes vector that expressed the LCMV glycoprotein-specific I-Ab-restricted CD4+ T cell epitope GP61-80 (LM-GP61) and subsequently boosted the primed mice with an influenza virus A (PR8 strain) vector that expressed the same CD4+ T cell epitope (IAV-GP61). This heterologous prime-boost vaccination strategy elicited strong anti-viral CD4+ T cell responses, which further improved both the quantity and quality of the virus-specific CD8+ T cells and led to better control of the viral loads. The combination of this strategy and the blockade of the programmed cell death-1 (PD-1) inhibitory pathway further enhanced the anti-viral CD8+ T cell responses and viral clearance. Thus, a heterologous prime-boost immunization that selectively induces virus-specific CD4+ T cell responses in conjunction with blockade of the inhibitory pathway may represent a promising therapeutic approach to treating patients with chronic viral infections.


Subject(s)
Epitopes, T-Lymphocyte , Genetic Vectors , Immunization, Secondary , Influenza A virus , Listeria monocytogenes , Lymphocytic Choriomeningitis/prevention & control , Lymphocytic choriomeningitis virus/immunology , Animals , CD4-Positive T-Lymphocytes , Chronic Disease , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus/genetics , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...