Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
1.
J Virol ; 95(19): e0070721, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34287053

ABSTRACT

Understanding the earliest events of human immunodeficiency virus (HIV) sexual transmission is critical to developing and optimizing HIV prevention strategies. To gain insights into the earliest steps of HIV rectal transmission, including cellular targets, rhesus macaques were intrarectally challenged with a single-round simian immunodeficiency virus (SIV)-based dual reporter that expresses luciferase and near-infrared fluorescent protein 670 (iRFP670) upon productive transduction. The vector was pseudotyped with the HIV-1 envelope JRFL. Regions of tissue containing foci of luminescent transduced cells were identified macroscopically using an in vivo imaging system, and individual transduced cells expressing fluorescent protein were identified and phenotyped microscopically. This system revealed that anal and rectal tissues are both susceptible to transduction 48 h after the rectal challenge. Detailed phenotypic analysis revealed that, on average, 62% of transduced cells are CCR6-positive (CCR6+) T cells-the vast majority of which express RORγT, a Th17 lineage-specific transcription factor. The second most common target cells were immature dendritic cells at 20%. These two cell types were transduced at rates that are four to five times higher than their relative abundances indicate. Our work demonstrates that Th17 T and immature dendritic cells are preferential initial targets of HIV/SIV rectal transmission. IMPORTANCE Men and women who participate in unprotected receptive anal intercourse are at high risk of acquiring HIV. While in vitro data have developed a framework for understanding HIV cell tropism, the initial target cells in the rectal mucosa have not been identified. In this study, we identify these early host cells by using an innovative rhesus macaque rectal challenge model and methodology, which we previously developed. Thus, by shedding light on these early HIV/SIV transmission events, this study provides a specific cellular target for future prevention strategies.


Subject(s)
Dendritic Cells/virology , HIV Infections/transmission , HIV Infections/virology , HIV-1/physiology , Rectum/virology , Simian Immunodeficiency Virus/physiology , Th17 Cells/virology , Anal Canal/virology , Animals , Female , Intestinal Mucosa/virology , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Virus Replication
2.
Trends Immunol ; 38(7): 498-512, 2017 07.
Article in English | MEDLINE | ID: mdl-28549714

ABSTRACT

Multiple sclerosis (MS) is a T cell driven autoimmune disease of the central nervous system (CNS). Despite its association with Epstein-Barr Virus (EBV), how viral infections promote MS remains unclear. However, there is increasing evidence that the CNS is continuously surveyed by virus-specific T cells, which protect against reactivating neurotropic viruses. Here, we discuss how viral infections could lead to the breakdown of self-tolerance in genetically predisposed individuals, and how the reactivations of viruses in the CNS could induce the recruitment of both autoaggressive and virus-specific T cell subsets, causing relapses and progressive disability. A disturbed immune surveillance in MS would explain several experimental findings, and has important implications for prognosis and therapy.


Subject(s)
Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/immunology , Host-Pathogen Interactions/immunology , Immunologic Surveillance , Molecular Mimicry/immunology , Multiple Sclerosis/virology , Cell Movement , Central Nervous System/immunology , Central Nervous System/virology , Cytokines/genetics , Cytokines/immunology , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/immunology , Gene Expression Regulation , Gene-Environment Interaction , Genetic Predisposition to Disease , Herpesvirus 4, Human/pathogenicity , Humans , Multiple Sclerosis/complications , Multiple Sclerosis/immunology , Receptors, Cytokine/genetics , Receptors, Cytokine/immunology , Th1 Cells/immunology , Th1 Cells/virology , Th17 Cells/immunology , Th17 Cells/virology
3.
J Cell Mol Med ; 23(2): 908-919, 2019 02.
Article in English | MEDLINE | ID: mdl-30378252

ABSTRACT

BACKGROUND: Patients with idiopathic pulmonary fibrosis (IPF) often experience acute exacerbation (AE) after an episode of common cold. AIMS: To establish a mouse model of virus infection-induced AE-IPF and investigate the mechanism underlying the AE-IPF. METHODS: Herpes simplex virus 1 (HSV1) was inoculated intranasally to wild-type (WT) and IL-17A gene knockout (IL-17A-/- ) mice 21 days after intratracheal administration of bleomycin (BLM). RESULTS: HSV1 infection caused acute exacerbation in mice with BLM-induced fibrosis. Compared with the BLM+Saline mice, the mice with BLM+HSV1 showed significantly higher acute lung injury (ALI) score (P < 0.0001), lower survival rate (100% vs 21.4%, P < 0.0001), poorer lung function and higher inflammatory response representing by increased total inflammatory cells in bronchoalveolar lavage fluid (BALF) (P = 0.0323), increased proportion of Th17 cells in peripheral blood (P = 0.0004) and higher inflammatory factors in BALF. In addition, HSV1 infection increased the expression of endoplasmic reticulum stress (ERS)-related proteins in mice with BLM-induced fibrosis. The inhibition of ERS by tauroursodeoxycholic acid (TUDCA, an ERS inhibitor) significantly reduced the IL-17A levels in BALF (P = 0.0140) and TH17 cells in the peripheral blood (P = 0.0084) of mice with BLM+HSV1, suggesting that suppression of ERS may reduce TH17 response in mice with AE-IPF. Compared with WT mice with BLM+HSV1, IL-17A-/- mice with BLM+HSV1 had lower ALI score (P = 0.0119), higher survival rate (78.6% vs 21.4%, P = 0.004), improved lung function, and milder inflammatory response. CONCLUSIONS: HSV1 infection in addition to BLM-induced IPF can successfully establish AE-IPF in mice. IL-17A and ERS promote lung inflammation in AE-IPF development.


Subject(s)
Acute Lung Injury/virology , Endoplasmic Reticulum Stress/immunology , Herpes Simplex/virology , Idiopathic Pulmonary Fibrosis/virology , Interleukin-17/genetics , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Acute Lung Injury/mortality , Animals , Anti-Inflammatory Agents/pharmacology , Antiviral Agents/pharmacology , Bleomycin , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , Disease Models, Animal , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Gene Expression , Herpes Simplex/chemically induced , Herpes Simplex/drug therapy , Herpes Simplex/mortality , Herpesvirus 1, Human , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/mortality , Interleukin-17/deficiency , Interleukin-17/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Respiratory Function Tests , Survival Analysis , Taurochenodeoxycholic Acid/pharmacology , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/virology
4.
Gastroenterology ; 154(4): 1047-1060, 2018 03.
Article in English | MEDLINE | ID: mdl-29229400

ABSTRACT

BACKGROUND AND AIMS: CD4+CD25+Foxp3+ T-regulatory (Treg) cells control immune responses and maintain immune homeostasis. However, under inflammatory conditions, Treg cells produce cytokines that promote inflammation. We investigated production of tumor necrosis factor (TNF) by Treg cells in patients with acute hepatitis A (AHA), and examined the characteristics of these cells and association with clinical factors. METHODS: We analyzed blood samples collected from 63 patients with AHA at the time of hospitalization (and some at later time points) and 19 healthy donors in South Korea. Liver tissues were collected from patients with fulminant AHA during liver transplantation. Peripheral blood mononuclear cells were isolated from whole blood and lymphocytes were isolated from liver tissues and analyzed by flow cytometry. Cytokine production from Treg cells (CD4+CD25+Foxp3+) was measured by immunofluorescence levels following stimulation with anti-CD3 and anti-CD28. Epigenetic stability of Treg cells was determined based on DNA methylation patterns. Phenotypes of Treg cells were analyzed by flow cytometry and an RORγt inhibitor, ML-209, was used to inhibit TNF production. Treg cell suppression assay was performed by co-culture of Treg-depleted peripheral blood mononuclear cells s and isolated Treg cells. RESULTS: A higher proportion of CD4+CD25+Foxp3+ Treg cells from patients with AHA compared with controls produced TNF upon stimulation with anti-CD3 and anti-CD28 (11.2% vs 2.8%). DNA methylation analysis confirmed the identity of the Treg cells. TNF-producing Treg cells had features of T-helper 17 cells, including up-regulation of RORγt, which was required for TNF production. The Treg cells had reduced suppressive functions compared with Treg cells from controls. The frequency of TNF-producing Treg cells in AHA patients' blood correlated with their serum level of alanine aminotransferase. CONCLUSIONS: Treg cells from patients with AHA have altered functions compared with Treg cells from healthy individuals. Treg cells from patients with AHA produce higher levels of TNF, gain features of T-helper 17 cells, and have reduced suppressive activity. The presence of these cells is associated with severe liver injury in patients with AHA.


Subject(s)
Hepatitis A/metabolism , Liver/metabolism , T-Lymphocytes, Regulatory/metabolism , Tumor Necrosis Factor-alpha/metabolism , Acute Disease , Antigens, CD/immunology , Antigens, CD/metabolism , Apyrase/immunology , Apyrase/metabolism , Case-Control Studies , Cells, Cultured , DNA Methylation , Epigenesis, Genetic , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Hepatitis A/diagnosis , Hepatitis A/immunology , Hepatitis A/virology , Hepatitis A virus/immunology , Hepatitis A virus/pathogenicity , Host-Pathogen Interactions , Humans , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Liver/immunology , Liver/pathology , Liver/virology , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Phenotype , Severity of Illness Index , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/virology , Th17 Cells/immunology , Th17 Cells/metabolism , Th17 Cells/virology , Time Factors , Tumor Necrosis Factor-alpha/immunology
5.
J Virol ; 92(8)2018 04 15.
Article in English | MEDLINE | ID: mdl-29437964

ABSTRACT

Measles is characterized by a transient immune suppression, leading to an increased risk of opportunistic infections. Measles virus (MV) infection of immune cells is mediated by the cellular receptor CD150, expressed by subsets of lymphocytes, dendritic cells, macrophages, and thymocytes. Previous studies showed that human and nonhuman primate memory T cells express higher levels of CD150 than naive cells and are more susceptible to MV infection. However, limited information is available about the CD150 expression and relative susceptibility to MV infection of B-cell subsets. In this study, we assessed the susceptibility and permissiveness of naive and memory T- and B-cell subsets from human peripheral blood or tonsils to in vitro MV infection. Our study demonstrates that naive and memory B cells express CD150, but at lower frequencies than memory T cells. Nevertheless, both naive and memory B cells proved to be highly permissive to MV infection. Furthermore, we assessed the susceptibility and permissiveness of various functionally distinct T and B cells, such as helper T (TH) cell subsets and IgG- and IgA-positive memory B cells, in peripheral blood and tonsils. We demonstrated that TH1TH17 cells and plasma and germinal center B cells were the subsets most susceptible and permissive to MV infection. Our study suggests that both naive and memory B cells, along with several other antigen-experienced lymphocytes, are important target cells of MV infection. Depletion of these cells potentially contributes to the pathogenesis of measles immune suppression.IMPORTANCE Measles is associated with immune suppression and is often complicated by bacterial pneumonia, otitis media, or gastroenteritis. Measles virus infects antigen-presenting cells and T and B cells, and depletion of these cells may contribute to lymphopenia and immune suppression. Measles has been associated with follicular exhaustion in lymphoid tissues in humans and nonhuman primates, emphasizing the importance of MV infection of B cells in vivo However, information on the relative susceptibility of B-cell subsets is scarce. Here, we compared the susceptibility and permissiveness to in vitro MV infection of human naive and memory T- and B-cell subsets isolated from peripheral blood or tonsils. Our results demonstrate that both naive and memory B cells are more permissive to MV infection than T cells. The highest infection levels were detected in plasma cells and germinal center B cells, suggesting that infection and depletion of these populations contribute to reduced host resistance.


Subject(s)
B-Lymphocytes/immunology , Immunologic Memory , Measles virus/immunology , Measles/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Adult , B-Lymphocytes/pathology , B-Lymphocytes/virology , Child , Female , Humans , Male , Measles/pathology , Th1 Cells/pathology , Th1 Cells/virology , Th17 Cells/pathology , Th17 Cells/virology
6.
Dig Dis Sci ; 64(3): 773-780, 2019 03.
Article in English | MEDLINE | ID: mdl-30498928

ABSTRACT

BACKGROUND: B cell-activating transcription factor (BATF) contributes to Th17 cell differentiation and pathological inflammatory responses. AIMS: This study explored BATF as a regulator of Th17 differentiation in normal and hepatitis B virus (HBV) transgenic mice. METHODS: Normal mice were divided into control, short hairpin RNA (shRNA) scramble, and shRNA BATF groups. HBV transgenic mice were divided into control, entecavir, shRNA scramble, entecavir + vector control, entecavir + shRNA scramble, shRNA BATF, and entecavir + shRNA BATF groups. Serum concentrations of AST, ALT, HBV-DNA, BATF, IL-17, and IL-22 and Th17 cell frequencies in the liver were compared among the groups. Correlations of serum HBV surface antigen (HBsAg), e-antigen (HBeAg), and core antigen (HBcAg) concentrations with BATF mRNA expression and the proportion of Th17 cells in the livers of HBV transgenic mice were also analyzed. RESULTS: Serum AST, ALT, BATF, IL-17, and IL-22 concentrations and Th17 cell proportions were higher in HBV transgenic mice relative to normal controls. Positive correlations of the HBcAg concentration with BATF mRNA and the proportion of Th17 cells were observed in HBV transgenic mice. BATF interference reduced the proportion of Th17 cells and serum IL-17 and IL-22 concentrations and led to obvious downregulation of AST, ALT, BATF, IL-17, and IL-22 expression and a reduced proportion of Th17 cells when combined with entecavir. CONCLUSION: HBV markedly upregulated BATF expression and promoted Th17 cell activation. By contrast, BATF interference significantly impeded the proliferation of Th17 cells and secretion of IL-17 and IL-22 while alleviating hepatic lesions.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Cell Differentiation , Hepatitis B virus/genetics , Hepatitis B/metabolism , Liver/metabolism , Lymphocyte Activation , Th17 Cells/metabolism , Animals , Antiviral Agents/pharmacology , Basic-Leucine Zipper Transcription Factors/genetics , Cell Differentiation/drug effects , Disease Models, Animal , Guanine/analogs & derivatives , Guanine/pharmacology , Hepatitis B/immunology , Hepatitis B/prevention & control , Hepatitis B/virology , Hepatitis B virus/growth & development , Host-Pathogen Interactions , Interleukin-17/metabolism , Interleukins/metabolism , Liver/drug effects , Liver/immunology , Liver/virology , Lymphocyte Activation/drug effects , Male , Mice, Inbred BALB C , Mice, Transgenic , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/virology , Viral Load , Interleukin-22
7.
Am J Transplant ; 18(11): 2647-2662, 2018 11.
Article in English | MEDLINE | ID: mdl-29659179

ABSTRACT

Human cytomegalovirus (HCMV) donor positive (D+) serostatus with acute rejection is associated with renal allograft loss, but the impact of recipient positive (R+) serostatus is unclear. In an allogeneic renal transplant model, antiviral natural killer (NK) and CD8+ T cell memory responses in murine CMV (MCMV) D+/R+ transplants were compared to D-/R- and D+/R- transplants, with recipient infection varied by MCMV dose and strain. D+/R- transplants had high primary antiviral cytolytic (interferon-γ+) and cytotoxic (granzyme B+) NK responses, whereas NK memory responses were lower in D+/R+ recipients receiving a high primary MCMV dose. Despite MCMV immunity, D+/R+ recipients receiving a low MCMV dose showed primary-like high cytolytic and cytotoxic NK responses. D+/R+ transplants infected with different D/R strains had low cytolytic NK responses but high cytotoxic NK responses. NK memory also induced a novel TNF-α+ NK response among high-dose virus recipients. MCMV+ transplants had greater Th17 responses than MCMV-uninfected transplants and Th17 inhibition ameliorated graft injury. All MCMV+ recipients had similar CD8+ T cell responses. In sum, NK and Th17 responses, but not CD8+ T cells, varied according to conditions of primary recipient infection. This variability could contribute to variable graft outcomes in HCMV D+/R+ renal transplantation.


Subject(s)
Cytomegalovirus Infections/immunology , Graft Rejection/etiology , Kidney Transplantation/adverse effects , Killer Cells, Natural/immunology , Muromegalovirus/classification , Th17 Cells/immunology , Viral Load/immunology , Allografts , Animals , Cytomegalovirus Infections/virology , Graft Rejection/pathology , Killer Cells, Natural/pathology , Killer Cells, Natural/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Muromegalovirus/immunology , Th17 Cells/pathology , Th17 Cells/virology
8.
BMC Infect Dis ; 18(1): 254, 2018 06 04.
Article in English | MEDLINE | ID: mdl-29866105

ABSTRACT

BACKGROUND: Female gender and favorable IFNL3 genotypes are the primary independent predictors of spontaneous clearance of HCV infection. However, chronic hepatitis C infection occurs in numerous women carrying favorable IFNL3 genotypes, indicating that other host and/or virological factors contribute to the prognosis of infection. METHODS: A cohort of 137 anti-HCV-positive female Han Chinese cases, including 64 chronic HCV carriers and 73 HCV spontaneous resolvers, was recruited in the study. 111 SNPs in 23 genes encoding HCV co-receptors, transcription factors, Toll-like receptors, co-stimulating molecules, and cytokines were selected for SNP analysis. RESULTS: After comparison of genotypes and allelotype frequencies of 111 SNPs in 23 genes in the primary cohort, the SNPs rs9826 (P = 0.024 for CC/TT/CT; P = 0.015 for C allele/T allele) and rs1521177 (P = 0.017 for GG/TT/GT; P = 0.006 for G allele/T allele) in the RORC gene were significantly associated with spontaneous HCV clearance. In the sub-cohort carrying favorable IFNL3 genotypes (rs12979860CC, rs8099917 TT, rs12980275 AA), rs1521177 (genotype: P = 0.040; allelotype: P = 0.021) remained significantly associated with spontaneous HCV clearance. Importantly, the most common RORC haplotype rs9826-T/rs1521177-T was presented at significantly different frequencies in resolvers and carriers in both the primary cohort (P = 0.0027) and the IFNL3 favorable sub-cohort (P = 0.0117). CONCLUSIONS: This study indicates that genetic polymorphisms in human Th17-related RORC gene are associated with different natural prognosis of HCV infection. The RORC haplotype, rs9826-T/rs1521177-T, was favorable for spontaneous clearance of HCV infection.


Subject(s)
Hepatitis C, Chronic/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Polymorphism, Single Nucleotide , Th17 Cells/physiology , Alleles , Asian People/genetics , Female , Genotype , Haplotypes , Humans , Interferons , Interleukins/genetics , Middle Aged , Remission, Spontaneous , Th17 Cells/virology
9.
Gene Ther ; 24(1): 21-30, 2017 01.
Article in English | MEDLINE | ID: mdl-27779616

ABSTRACT

The anti-tumour effects associated with oncolytic virus therapy are mediated significantly through immune-mediated mechanisms, which depend both on the type of virus and the route of delivery. Here, we show that intra-tumoral oncolysis by Reovirus induced the priming of a CD8+, Th1-type anti-tumour response. By contrast, systemically delivered Vesicular Stomatitis Virus expressing a cDNA library of melanoma antigens (VSV-ASMEL) promoted a potent anti-tumour CD4+ Th17 response. Therefore, we hypothesised that combining the Reovirus-induced CD8+ T cell response, with the VSV-ASMEL CD4+ Th17 helper response, would produce enhanced anti-tumour activity. Consistent with this, priming with intra-tumoral Reovirus, followed by an intra-venous VSV-ASMEL Th17 boost, significantly improved survival of mice bearing established subcutaneous B16 melanoma tumours. We also show that combination of either therapy alone with anti-PD-1 immune checkpoint blockade augmented both the Th1 response induced by systemically delivered Reovirus in combination with GM-CSF, and also the Th17 response induced by VSV-ASMEL. Significantly, anti-PD-1 also uncovered an anti-tumour Th1 response following VSV-ASMEL treatment that was not seen in the absence of checkpoint blockade. Finally, the combination of all three treatments (priming with systemically delivered Reovirus, followed by double boosting with systemic VSV-ASMEL and anti-PD-1) significantly enhanced survival, with long-term cures, compared to any individual, or double, combination therapies, associated with strong Th1 and Th17 responses to tumour antigens. Our data show that it is possible to generate fully systemic, highly effective anti-tumour immunovirotherapy by combining oncolytic viruses, along with immune checkpoint blockade, to induce complementary mechanisms of anti-tumour immune responses.


Subject(s)
Cell Cycle Checkpoints , Immunotherapy/methods , Melanoma/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cell Line, Tumor , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Melanoma-Specific Antigens/genetics , Melanoma-Specific Antigens/immunology , Mice , Oncolytic Viruses/genetics , Reoviridae/genetics , Reoviridae/immunology , Th1 Cells/cytology , Th1 Cells/immunology , Th1 Cells/virology , Th17 Cells/cytology , Th17 Cells/immunology , Th17 Cells/virology , Vesiculovirus/genetics , Vesiculovirus/immunology
10.
J Virol ; 90(17): 7833-47, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27334595

ABSTRACT

UNLABELLED: Human immunodeficiency virus (HIV) infects and depletes CD4(+) T cells, but subsets of CD4(+) T cells vary in their susceptibility and permissiveness to infection. For example, HIV preferentially depletes interleukin-17 (IL-17)-producing T helper 17 (Th17) cells and T follicular helper (Tfh) cells. The preferential loss of Th17 cells during the acute phase of infection impairs the integrity of the gut mucosal barrier, which drives chronic immune activation-a key determinant of disease progression. The preferential loss of Th17 cells has been attributed to high CD4, CCR5, and CXCR4 expression. Here, we show that Th17 cells also exhibit heightened permissiveness to productive HIV infection. Primary human CD4(+) T cells were sorted, activated under Th17- or Th0-polarizing conditions and infected, and then analyzed by flow cytometry. Th17-polarizing cytokines increased HIV infection, and HIV infection was disproportionately higher among Th17 cells than among IL-17(-) or gamma interferon-positive (IFN-γ(+)) cells, even upon infection with a replication-defective HIV vector with a pseudotype envelope. Further, Th17-polarized cells produced more viral capsid protein. Our data also reveal that Th17-polarized cells have diminished expression of RNase A superfamily proteins, and we report for the first time that RNase 6 inhibits HIV. Thus, our findings link Th17 polarization to increased HIV replication. IMPORTANCE: Our study compares the intracellular replicative capacities of several different HIV isolates among different T cell subsets, providing a link between the differentiation of Th17 cells and HIV replication. Th17 cells are of key importance in mucosal integrity and in the immune response to certain pathogens. Based on our findings and the work of others, we propose a model in which HIV replication is favored by the intracellular environment of two CD4(+) T cell subsets that share several requirements for their differentiation: Th17 and Tfh cells. Characterizing cells that support high levels of viral replication (rather than becoming latently infected or undergoing cell death) informs the search for new therapeutics aimed at manipulating intracellular signaling pathways and/or transcriptional factors that affect HIV replication.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , HIV/growth & development , Ribonuclease, Pancreatic/biosynthesis , Th17 Cells/immunology , Th17 Cells/virology , CD4-Positive T-Lymphocytes/enzymology , Cells, Cultured , Gene Expression Profiling , HIV/physiology , Humans , T-Lymphocyte Subsets/enzymology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/virology , Th17 Cells/enzymology , Virus Replication
11.
J Cardiovasc Pharmacol ; 69(5): 305-313, 2017 May.
Article in English | MEDLINE | ID: mdl-28207428

ABSTRACT

Myocarditis is a heterogeneous group of disorders defined by inflammation of the heart muscle with an excessively activated immune response. Numerous interventions have been investigated for the treatment of myocarditis while success is limited. Interleukin-37 (IL-37), a novel member of the IL-1 cytokine family, is a natural inhibitor of innate immunity associated with autoimmune diseases. However, the modulatory effect of IL-37 in myocarditis is unknown. In this study, we investigated the immunological regulation of IL-37 in the coxsackievirus B3-induced model of murine viral myocarditis. The results show that IL-37 significantly ameliorates the signs of myocarditis with increased survival rate and bodyweight, improved histological changes, reduced activities of MB isoenzyme of creatine kinase and cardiac troponin I, and a suppressed response of Th17 cells and enhanced response of regulatory T cells (Tregs) in the spleen. Moreover, IL-37 down-regulates the expression of Th17-related cytokines IL-6 and IL-17A, while promoting Treg-related cytokine IL-10 levels in the heart. Therefore, IL-37 may exhibit anti-inflammatory activity in the murine model of myocarditis by regulating the balance between Th17 and Treg cells, thereby providing a possible novel therapeutic target in myocarditis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Coxsackievirus Infections/prevention & control , Enterovirus B, Human/pathogenicity , Interleukin-1/pharmacology , Myocarditis/prevention & control , Myocardium/immunology , T-Lymphocytes, Regulatory/drug effects , Th17 Cells/drug effects , Animals , Coxsackievirus Infections/immunology , Coxsackievirus Infections/virology , Disease Models, Animal , Enterovirus B, Human/immunology , Interleukin-10/metabolism , Interleukin-17/metabolism , Interleukin-6/metabolism , Male , Mice, Inbred BALB C , Myocardial Contraction/drug effects , Myocarditis/immunology , Myocarditis/virology , Myocardium/metabolism , Myocardium/pathology , Spleen/drug effects , Spleen/immunology , Spleen/metabolism , Spleen/virology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/virology , Th17 Cells/immunology , Th17 Cells/metabolism , Th17 Cells/virology , Time Factors , Ventricular Function, Left/drug effects
12.
AIDS Res Ther ; 14(1): 49, 2017 Sep 12.
Article in English | MEDLINE | ID: mdl-28893286

ABSTRACT

Circumcision reduces heterosexual HIV-1 acquisition in men by at least 60%. However, the biological mechanisms by which circumcision is protective remain incompletely understood. We test the hypothesis that the sub-preputial microenvironment created by the foreskin drives immune activation in adjacent foreskin tissues, facilitating HIV-1 infection through a combination of epithelial barrier disruption, enhanced dendritic cell maturation, and the recruitment/activation of neutrophils and susceptible CD4 T cell subsets such as Th17 cells. Furthermore, we provide evidence that the genital microbiome may be an important driver of this immune activation. This suggests that new modalities to reduce genital immune activation and/or alter the genital microbiome, used alone or in combination with topical microbicides, may be of significant benefit to HIV prevention.


Subject(s)
Circumcision, Male , Disease Susceptibility , HIV Infections/prevention & control , HIV-1/physiology , Adult , Animals , CD4-Positive T-Lymphocytes/immunology , Chemokines/immunology , Foreskin/immunology , Foreskin/virology , HIV Infections/immunology , HIV Infections/virology , HIV-1/isolation & purification , Heterosexuality , Humans , Lymphocyte Activation , Male , Microbiota/immunology , Penis/cytology , Penis/immunology , Penis/virology , Primates , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/physiology , Th17 Cells/immunology , Th17 Cells/virology
13.
Retrovirology ; 13(1): 59, 2016 08 24.
Article in English | MEDLINE | ID: mdl-27553844

ABSTRACT

BACKGROUND: Th17 cells are permissive to HIV-1 infection and their depletion from the gut of infected individuals leads to microbial translocation, a major cause for non-AIDS co-morbidities. Most recent evidence supports the contribution of long-lived Th17 cells to HIV persistence during antiretroviral therapy (ART). However, the identity of long-lived Th17 cells remains unknown. RESULTS: Here, we performed an in-depth transcriptional and functional characterization of four distinct Th17 subsets and investigated their contribution to HIV reservoir persistence during ART. In addition to the previously characterized CCR6(+)CCR4(+) (Th17) and CCR6(+)CXCR3(+) (Th1Th17) subsets, we reveal the existence of two novel CCR6(+) subsets, lacking (double negative, CCR6(+)DN) or co-expressing CXCR3 and CCR4 (double positive, CCR6(+)DP). The four subsets shared multiple Th17-polarization markers, a fraction of cells proliferated in response to C. albicans, and exhibited lineage commitment and plasticity when cultured under Th17 and Th1 conditions, respectively. Of note, fractions of CCR6(+)DN and Th17 demonstrated stable Th17-lineage commitment under Th1-polarization conditions. Among the four subsets, CCR6(+)DN expressed a unique transcriptional signature indicative of early Th17 development (IL-17F, STAT3), lymph-node homing (CCR7, CD62L), follicular help (CXCR5, BCL6, ASCL2), and self-renewal (LEFI, MYC, TERC). Cross sectional and longitudinal studies demonstrated that CCR6(+)DN cells were the most predominant CCR6(+) subset in the blood before and after ART initiation; high frequencies of these cells were similarly observed in inguinal lymph nodes of individuals receiving long-term ART. Importantly, replication competent HIV was isolated from CCR6(+)DN of ART-treated individuals. CONCLUSIONS: Together, these results provide new insights into the functional heterogeneity of Th17-polarized CCR6(+)CD4(+) T-cells and support the major contribution of CCR6(+)DN cells to HIV persistence during ART.


Subject(s)
Antiretroviral Therapy, Highly Active , HIV Infections/drug therapy , HIV-1/drug effects , Th17 Cells/drug effects , Th17 Cells/physiology , Cross-Sectional Studies , HIV Infections/immunology , HIV Infections/virology , HIV-1/isolation & purification , HIV-1/physiology , Humans , Immunologic Memory , Longitudinal Studies , Receptors, CCR4/analysis , Receptors, CCR6/analysis , Receptors, CXCR3/analysis , Th17 Cells/virology , Virus Replication/drug effects
14.
Biochem Biophys Res Commun ; 473(2): 558-64, 2016 Apr 29.
Article in English | MEDLINE | ID: mdl-27021682

ABSTRACT

Viral myocarditis (VMC) is an inflammation of heart muscle in infants and young adolescents. This study explored the function of halofuginone (HF) in Coxsackievirus B3 (CVB3) -treated suckling mice. HF-treated animal exhibited higher survival rate, lower heart/body weight, and more decreased blood sugar concentration than CVB3 group. HF also reduced the expressions of interleukin(IL)-17 and IL-23 and the numbers of Th17 cells. Moreover, HF downregulated pro-inflammatory cytokine levels and increased anti-inflammatory cytokine levels. The expressions of transforming growth factor(TGF-ß1) and nuclear factor kappa-light-chain-enhancer of activated B (NF-κB) p65/ tumor necrosis factor-α (TNF-α) proteins were decreased by HF as well. Finally, the overexpression of TGF-ß1 counteracted the protection effect of HF in CVB3-treated suckling mice. In summary, our study suggests HF increases the survival of CVB3 suckling mice, reduces the Th17 cells and pro-inflammatory cytokine levels, and may through downregulation of the TGF-ß1-mediated expression of NF-κB p65/TNF-α pathway proteins. These results offer a potential therapeutic strategy for the treatment of VMC.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Coxsackievirus Infections/complications , Enterovirus B, Human/immunology , Myocarditis/drug therapy , Myocarditis/virology , Piperidines/therapeutic use , Quinazolinones/therapeutic use , Transforming Growth Factor beta1/antagonists & inhibitors , Animals , Coxsackievirus Infections/drug therapy , Coxsackievirus Infections/immunology , Mice , Mice, Inbred BALB C , Myocarditis/immunology , NF-kappa B/immunology , Signal Transduction/drug effects , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/virology , Transforming Growth Factor beta1/immunology
15.
J Virol ; 89(22): 11284-93, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26339043

ABSTRACT

UNLABELLED: The ability to persist long term in latently infected CD4 T cells represents a characteristic feature of HIV-1 infection and the predominant barrier to efforts aiming at viral eradication and cure. Yet, increasing evidence suggests that only small subsets of CD4 T cells with specific developmental and maturational profiles are able to effectively support HIV-1 long-term persistence. Here, we analyzed how the functional polarization of CD4 T cells shapes and structures the reservoirs of HIV-1-infected cells. We found that CD4 T cells enriched for a Th1/17 polarization had elevated susceptibilities to HIV-1 infection in ex vivo assays, harbored high levels of HIV-1 DNA in persons treated with antiretroviral therapy, and made a disproportionately increased contribution to the viral reservoir relative to their contribution to the CD4 T memory cell pool. Moreover, HIV-1 DNA levels in Th1/17 cells remained stable over many years of antiretroviral therapy, resulting in a progressively increasing contribution of these cells to the viral reservoir, and phylogenetic studies suggested preferential long-term persistence of identical viral sequences during prolonged antiretroviral treatment in this cell compartment. Together, these data suggest that Th1/17 CD4 T cells represent a preferred site for HIV-1 DNA long-term persistence in patients receiving antiretroviral therapy. IMPORTANCE: Current antiretroviral therapy is very effective in suppressing active HIV-1 replication but does not fully eliminate virally infected cells. The ability of HIV-1 to persist long term despite suppressive antiretroviral combination therapy represents a perplexing aspect of HIV-1 disease pathogenesis, since most HIV-1 target cells are activated, short-lived CD4 T cells. This study suggests that CD4 T helper cells with Th1/17 polarization have a preferential role as a long-term reservoir for HIV-1 infection during antiretroviral therapy, possibly because these cells may imitate some of the functional properties traditionally attributed to stem cells, such as the ability to persist for extremely long periods of time and to repopulate their own pool size through homeostatic self-renewal. These observations support the hypothesis that HIV-1 persistence is driven by small subsets of long-lasting stem cell-like CD4 T cells that may represent particularly promising targets for clinical strategies aiming at HIV-1 eradication and cure.


Subject(s)
Anti-HIV Agents/therapeutic use , Cell Polarity/immunology , HIV Infections/drug therapy , Th1 Cells/immunology , Th17 Cells/immunology , Virus Latency/genetics , Adult , Base Sequence , CD4 Lymphocyte Count , Cells, Cultured , DNA, Viral/genetics , Female , HIV Infections/virology , HIV-1/genetics , HIV-1/immunology , Humans , Male , Middle Aged , Sequence Analysis, DNA , Th1 Cells/virology , Th17 Cells/virology , Th2 Cells/immunology , Th2 Cells/virology , Virus Replication/genetics
16.
Retrovirology ; 12: 102, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26654242

ABSTRACT

BACKGROUND: The HIV-1 infection is characterized by profound CD4(+) T cell destruction and a marked Th17 dysfunction at the mucosal level. Viral suppressive antiretroviral therapy restores Th1 but not Th17 cells. Although several key HIV dependency factors (HDF) were identified in the past years via genome-wide siRNA screens in cell lines, molecular determinants of HIV permissiveness in primary Th17 cells remain to be elucidated. RESULTS: In an effort to orient Th17-targeted reconstitution strategies, we investigated molecular mechanisms of HIV permissiveness in Th17 cells. Genome-wide transcriptional profiling in memory CD4(+) T-cell subsets enriched in cells exhibiting Th17 (CCR4(+)CCR6(+)), Th1 (CXCR3(+)CCR6(-)), Th2 (CCR4(+)CCR6(-)), and Th1Th17 (CXCR3(+)CCR6(+)) features revealed remarkable transcriptional differences between Th17 and Th1 subsets. The HIV-DNA integration was superior in Th17 versus Th1 upon exposure to both wild-type and VSV-G-pseudotyped HIV; this indicates that post-entry mechanisms contribute to viral replication in Th17. Transcripts significantly enriched in Th17 versus Th1 were previously associated with the regulation of TCR signaling (ZAP-70, Lck, and CD96) and Th17 polarization (RORγt, ARNTL, PTPN13, and RUNX1). A meta-analysis using the NCBI HIV Interaction Database revealed a set of Th17-specific HIV dependency factors (HDFs): PARG, PAK2, KLF2, ITGB7, PTEN, ATG16L1, Alix/AIP1/PDCD6IP, LGALS3, JAK1, TRIM8, MALT1, FOXO3, ARNTL/BMAL1, ABCB1/MDR1, TNFSF13B/BAFF, and CDKN1B. Functional studies demonstrated an increased ability of Th17 versus Th1 cells to respond to TCR triggering in terms of NF-κB nuclear translocation/DNA-binding activity and proliferation. Finally, RNA interference studies identified MAP3K4 and PTPN13 as two novel Th17-specific HDFs. CONCLUSIONS: The transcriptional program of Th17 cells includes molecules regulating HIV replication at multiple post-entry steps that may represent potential targets for novel therapies aimed at protecting Th17 cells from infection and subsequent depletion in HIV-infected subjects.


Subject(s)
HIV Infections/virology , HIV-1/physiology , Receptors, Antigen, T-Cell/immunology , Th17 Cells/immunology , Th17 Cells/virology , Virus Replication , Adult , Cells, Cultured , Female , Gene Expression Profiling , Humans , Immunity, Mucosal , Immunologic Memory , MAP Kinase Kinase Kinase 4/genetics , MAP Kinase Kinase Kinase 4/metabolism , Male , NF-kappa B/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 13/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 13/metabolism , RNA Interference , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Receptors, CCR4/immunology , Receptors, CCR6/immunology , T-Lymphocyte Subsets/virology , Th1 Cells/immunology , Th1 Cells/virology , Th17 Cells/classification , Transcriptome
17.
PLoS Pathog ; 9(7): e1003471, 2013.
Article in English | MEDLINE | ID: mdl-23853592

ABSTRACT

In pathogenic HIV and SIV infections of humans and rhesus macaques (RMs), preferential depletion of CD4⁺ Th17 cells correlates with mucosal immune dysfunction and disease progression. Interleukin (IL)-21 promotes differentiation of Th17 cells, long-term maintenance of functional CD8⁺ T cells, and differentiation of memory B cells and antibody-secreting plasma cells. We hypothesized that administration of IL-21 will improve mucosal function in the context of pathogenic HIV/SIV infections. To test this hypothesis, we infected 12 RMs with SIV(mac239) and at day 14 post-infection treated six of them with rhesus rIL-21-IgFc. IL-21-treatment was safe and did not increase plasma viral load or systemic immune activation. Compared to untreated animals, IL-21-treated RMs showed (i) higher expression of perforin and granzyme B in total and SIV-specific CD8⁺ T cells and (ii) higher levels of intestinal Th17 cells. Remarkably, increased levels of Th17 cells were associated with reduced levels of intestinal T cell proliferation, microbial translocation and systemic activation/inflammation in the chronic infection. In conclusion, IL-21-treatment in SIV-infected RMs improved mucosal immune function through enhanced preservation of Th17 cells. Further preclinical studies of IL-21 may be warranted to test its potential use during chronic infection in conjunction with antiretroviral therapy.


Subject(s)
Bacterial Translocation/drug effects , Immunity, Mucosal/drug effects , Immunoglobulin Fc Fragments/therapeutic use , Interleukins/therapeutic use , Intestinal Mucosa/drug effects , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Immunodeficiency Virus/drug effects , Th17 Cells/drug effects , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/drug effects , Down-Regulation/drug effects , Female , Granzymes/biosynthesis , Granzymes/genetics , Granzymes/metabolism , Immunoglobulin Fc Fragments/adverse effects , Immunoglobulin Fc Fragments/genetics , Interleukins/adverse effects , Interleukins/genetics , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestinal Mucosa/virology , Macaca mulatta , Perforin/biosynthesis , Perforin/genetics , Perforin/metabolism , Random Allocation , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/metabolism , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/isolation & purification , Simian Immunodeficiency Virus/physiology , Th17 Cells/immunology , Th17 Cells/pathology , Th17 Cells/virology , Up-Regulation/drug effects , Viral Load/drug effects
18.
Blood ; 121(7): 1136-44, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23258923

ABSTRACT

In HIV infection, CD4 responses to opportunistic pathogens such as Candida albicans are lost early, but CMV-specific CD4 response persists. Little is currently known about HIV infection of CD4 T cells of different pathogen/antigen specificities. CFSE-labeled PBMCs were stimulated with CMV, tetanus toxoid (TT), and C albicans antigens and subsequently exposed to HIV. HIV infection was monitored by intracellular p24 in CFSE(low) population. We found that although TT- and C albicans-specific CD4 T cells were permissive, CMV-specific CD4 T cells were highly resistant to both R5 and X4 HIV. Quantification of HIV DNA in CFSE(low) cells showed a reduction of strong-stop and full-length DNA in CMV-specific cells compared with TT- and C albicans-specific cells. ß-Chemokine neutralization enhanced HIV infection in TT- and C albicans-specific cells, whereas HIV infection in CMV-specific cells remained low despite increased entry by ß-chemokine neutralization, suggesting postentry HIV restriction by CMV-specific cells. Microarray analysis (Gene Expression Omnibus accession number: GSE42853) revealed distinct transcriptional profiles that involved selective up-regulation of comprehensive innate antiviral genes in CMV-specific cells, whereas TT- and C albicans-specific cells mainly up-regulated Th17 inflammatory response. Our data suggest a mechanism for the persistence of CMV-specific CD4 response and earlier loss of mucosal Th17-associated TT- and C albicans-specific CD4 response in AIDS.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , HIV Infections/genetics , HIV Infections/immunology , HIV-1 , Candida albicans/immunology , Candida albicans/pathogenicity , Cytomegalovirus/immunology , Cytomegalovirus/pathogenicity , HIV Infections/virology , HIV-1/immunology , HIV-1/pathogenicity , HIV-1/physiology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate/genetics , Immunity, Mucosal/genetics , Tetanus Toxoid/immunology , Th17 Cells/immunology , Th17 Cells/virology , Transcriptome , Virus Internalization , Virus Replication
19.
Respir Res ; 16: 10, 2015 Feb 05.
Article in English | MEDLINE | ID: mdl-25651926

ABSTRACT

BACKGROUND: Influenza is a common respiratory virus and Staphylococcus aureus frequently causes secondary pneumonia during influenza infection, leading to increased morbidity and mortality. Influenza has been found to attenuate subsequent Type 17 immunity, enhancing susceptibility to secondary bacterial infections. IL-27 is known to inhibit Type 17 immunity, suggesting a potential critical role for IL-27 in viral and bacterial co-infection. METHODS: A murine model of influenza and Staphylococcus aureus infection was used to mimic human viral, bacterial co-infection. C57BL/6 wild-type, IL-27 receptor α knock-out, and IL-10 knock-out mice were infected with Influenza H1N1 (A/PR/8/34) or vehicle for 6 days followed by challenge with Staphylococcus aureus or vehicle for 24 hours. Lung inflammation, bacterial burden, gene expression, and cytokine production were determined. RESULTS: IL-27 receptor α knock-out mice challenged with influenza A had increased morbidity compared to controls, but no change in viral burden. IL-27 receptor α knock-out mice infected with influenza displayed significantly decreased IL-10 production compared to wild-type. IL-27 receptor α knock-out mice co-infected with influenza and S. aureus had improved bacterial clearance compared to wild-type controls. Importantly, there were significantly increased Type 17 responses and decreased IL-10 production in IL-27 receptor α knock-out mice. Dual infected IL-10-/- mice had significantly less bacterial burden compared to dual infected WT mice. CONCLUSIONS: These data reveal that IL-27 regulates enhanced susceptibility to S. aureus pneumonia following influenza infection, potentially through the induction of IL-10 and suppression of IL-17.


Subject(s)
Coinfection , Influenza A Virus, H1N1 Subtype/immunology , Interleukins/immunology , Lung/immunology , Orthomyxoviridae Infections/immunology , Pneumonia, Staphylococcal/immunology , Staphylococcus aureus/immunology , Animals , Bacterial Load , Disease Models, Animal , Host-Pathogen Interactions , Immunity, Cellular , Influenza A Virus, H1N1 Subtype/pathogenicity , Interleukin-10/deficiency , Interleukin-10/genetics , Interleukin-17/immunology , Lung/microbiology , Lung/virology , Male , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/virology , Pneumonia, Staphylococcal/genetics , Pneumonia, Staphylococcal/microbiology , Receptors, Cytokine/deficiency , Receptors, Cytokine/genetics , Receptors, Interleukin , Staphylococcus aureus/pathogenicity , Th17 Cells/immunology , Th17 Cells/microbiology , Th17 Cells/virology , Time Factors , Viral Load
20.
Pediatr Res ; 78(5): 483-91, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26267154

ABSTRACT

The respiratory syncytial virus (RSV) represents the leading cause of viral bronchiolitis and pneumonia in children worldwide and is associated with high morbidity, hospitalization rate, and significant mortality rates. The immune response elicited by RSV is one of the main factors contributing to the pathogenesis of the disease. Two subsets of the cellular immune response, the T helper 17 cell (Th17) and the regulatory T-cell (Treg), and more particularly the balance between these two subsets, might play a significant role in the pathogenesis of the RSV infection. The developmental pathways of Th17 and Treg cells are closely and reciprocally interconnected and plasticity has been demonstrated from Treg toward Th17. During an RSV infection, the functions of both subsets are opposed to one another regarding viral clearance and clinical severity. Th17 and Treg cells offer a promising new view on the pathogenesis of an RSV infection and deserve further exploration.


Subject(s)
Lung/immunology , Lymphocyte Activation , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Cell Communication , Cell Differentiation , Cytokines/immunology , Cytokines/metabolism , Humans , Lung/metabolism , Lung/virology , Phenotype , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Viruses/metabolism , Respiratory Syncytial Viruses/pathogenicity , Signal Transduction , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/virology , Th17 Cells/metabolism , Th17 Cells/virology
SELECTION OF CITATIONS
SEARCH DETAIL