Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Hepatology ; 61(4): 1163-73, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25477247

ABSTRACT

UNLABELLED: T cells play a crucial role in viral clearance or persistence; however, the precise mechanisms that control their responses during viral infection remain incompletely understood. MicroRNA (miR) has been implicated as a key regulator controlling diverse biological processes through posttranscriptional repression. Here, we demonstrate that hepatitis C virus (HCV)-mediated decline of miR-181a expression impairs CD4(+) T-cell responses through overexpression of dual specific phosphatase 6 (DUSP6). Specifically, a significant decline of miR-181a expression along with overexpression of DUSP6 was observed in CD4(+) T cells from chronically HCV-infected individuals compared to healthy subjects, and the levels of miR-181a loss were found to be negatively associated with the levels of DUSP6 overexpression in these cells. Importantly, reconstitution of miR-181a or blockade of DUSP6 expression in CD4(+) T cells led to improved T-cell responses including enhanced CD25 and CD69 expression, increased interleukin-2 expression, and improved proliferation of CD4(+) T cells derived from chronically HCV-infected individuals. CONCLUSION: Since a decline of miR-181a concomitant with DUSP6 overexpression is the signature marker for age-associated T-cell senescence, these findings provide novel mechanistic insights into HCV-mediated premature T-cell aging through miR-181a-regulated DUSP6 signaling and reveal new targets for therapeutic rejuvenation of impaired T-cell responses during chronic viral infection.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Dual Specificity Phosphatase 6/biosynthesis , Hepacivirus/physiology , MicroRNAs/physiology , Cells, Cultured , Humans
2.
J Immunol ; 192(2): 649-57, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24337749

ABSTRACT

Coinfection of hepatitis B virus (HBV) with hepatitis C virus (HCV) is quite common, leading to an increase in morbidity and mortality. As such, HBV vaccination is recommended in HCV-infected individuals. However, HBV vaccine responses in HCV-infected individuals are often blunted compared with uninfected populations. The mechanism for this failure of vaccine response in HCV-infected subjects remains unclear. In this study, we investigated the expression and function of an inhibitory receptor, killer cell lectin-like receptor subfamily G member 1 (KLRG1), in the regulation of CD4(+) T cells and HBV vaccine responses during HCV infection. We demonstrated that KLRG1 was overexpressed on CD4(+) T cells from HCV-infected, HBV vaccine nonresponders compared with HBV vaccine responders. The capacity of CD4(+) T cells to proliferate and secrete IL-2 cytokine was inversely associated with the level of KLRG1 expression. Importantly, blocking KLRG1 signaling resulted in a significant improvement in CD4(+) T cell proliferation and IL-2 production in HCV-infected, HBV vaccine nonresponders in response to TCR stimulation. Moreover, blockade of KLRG1 increased the phosphorylation of Akt (Ser(473)) and decreased the expression of cell cycle inhibitors p16(ink4a) and p27(kip1), which subsequently enhanced the expression of cyclin-dependent kinase 2 and cyclin E. These results suggest that the KLRG1 pathway impairs CD4(+) T cell responses to neoantigen and induces a state of immune senescence in individuals with HCV infection, raising the possibility that blocking this negative-signaling pathway might improve HBV vaccine responses in the setting of chronic viral infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Hepatitis B Vaccines/immunology , Hepatitis B/immunology , Hepatitis C/immunology , Lectins, C-Type/genetics , Trans-Activators/genetics , Aging/genetics , Aging/immunology , CD4-Positive T-Lymphocytes/virology , Cell Proliferation , Cells, Cultured , Coinfection/genetics , Coinfection/immunology , Cyclin E/genetics , Cyclin E/immunology , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/immunology , Cyclin-Dependent Kinase Inhibitor p16/immunology , Cyclin-Dependent Kinase Inhibitor p27/immunology , Hepacivirus/immunology , Hepatitis B/genetics , Hepatitis B/prevention & control , Hepatitis B virus/immunology , Hepatitis C/genetics , Hepatitis C/virology , Humans , Interleukin-2/genetics , Interleukin-2/immunology , Lectins, C-Type/immunology , Phosphorylation/genetics , Phosphorylation/immunology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Receptors, Immunologic , Signal Transduction/genetics , Signal Transduction/immunology , Trans-Activators/immunology
3.
J Virol ; 87(21): 11626-36, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23966413

ABSTRACT

In this study, we demonstrate that killer cell lectin-like receptor subfamily G member 1 (KLRG1), a transmembrane protein preferentially expressed on T cells, is highly expressed on CD56(+) NK cells, which are significantly reduced in their numbers and functions in the peripheral blood of patients with chronic hepatitis C virus (HCV) infection compared to subjects without infection. KLRG1 expression is also upregulated on healthy NK cells exposed to Huh-7 hepatocytes infected with HCV in vitro. Importantly, the expression levels of KLRG1 are inversely associated with the capacity of NK cells to proliferate and to produce gamma interferon (IFN-γ) but positively associated with apoptosis of NK cells in response to inflammatory cytokine stimulation. KLRG1(+) NK cells, including CD56(bright) and CD56(dim) subsets, exhibit impaired cell activation and IFN-γ production but increased apoptosis compared to KLRG1(-) NK cells, particularly in HCV-infected individuals. Importantly, blockade of KLRG1 signaling significantly recovered the impaired IFN-γ production by NK cells from HCV-infected subjects. Blockade of KLRG1 also enhanced the impaired phosphorylation of Akt (Ser473) in NK cells from HCV-infected subjects. Taken together, these results indicate that KLRG1 negatively regulates NK cell numbers and functions via the Akt pathway, thus providing a novel marker and therapeutic target for HCV infection.


Subject(s)
Hepatitis C, Chronic/immunology , Killer Cells, Natural/immunology , Lectins, C-Type/metabolism , Signal Transduction , Trans-Activators/metabolism , Apoptosis , CD56 Antigen/analysis , Cell Proliferation , Female , Humans , Interferon-gamma/metabolism , Killer Cells, Natural/chemistry , Male , Receptors, Immunologic
4.
PLoS One ; 8(8): e72488, 2013.
Article in English | MEDLINE | ID: mdl-23967307

ABSTRACT

Human monocytes/macrophages (M/M(Ф)) of the innate immunity sense and respond to microbial products via specific receptor coupling with stimulatory (such as TLR) and inhibitory (such as Tim-3) receptors. Current models imply that Tim-3 expression on M/M(Ø) can deliver negative signaling to TLR-mediated IL-12 expression through trans association with its ligand Galectin-9 (Gal-9) presented by other cells. However, Gal-9 is also expressed within M/M(Ø), and the effect of intracellular Gal-9 on Tim-3 activities and inflammatory responses in the same M/M(Ø) remains unknown. In this study, our data suggest that Tim-3 and IL-12/IL-23 gene transcriptions are regulated by enhanced or silenced Gal-9 expression within monocytes through synergizing with TLR signaling. Additionally, TLR activation facilitates Gal-9/Tim-3 cis association within the same M/M(Ø) to differentially regulate IL-12/IL-23 expressions through STAT-3 phosphorylation. These results reveal a ligand (Gal-9) compartment-dependent regulatory effect on receptor (Tim-3) activities and inflammatory responses via TLR pathways--a novel mechanism underlying cellular responses to external or internal cues.


Subject(s)
Galectins/metabolism , Gene Expression Regulation , Interleukin-12/genetics , Interleukin-23/genetics , Membrane Proteins/metabolism , Monocytes/cytology , Toll-Like Receptors/metabolism , Cell Line , Galectins/deficiency , Galectins/genetics , Gene Silencing , Hepatitis A Virus Cellular Receptor 2 , Humans , Intracellular Space/metabolism , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Monocytes/immunology , Monocytes/metabolism , Phosphorylation , STAT3 Transcription Factor/metabolism , Signal Transduction , Transcription, Genetic
5.
J Virol ; 87(8): 4372-83, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23388728

ABSTRACT

Cytokine production by innate immunity is critical for shaping the adaptive immunity through regulation of T cell differentiation. In this report, we studied T cell immunoglobulin mucin domain protein 3 (Tim-3) expression on monocytes and its regulatory effect on interleukin-12 (IL-12)/IL-23 production by CD14(+) monocytes, as well as IL-17 production by CD4(+) T cells in individuals with chronic hepatitis C virus (HCV) infection. We found that Tim-3 and IL-23p19 are highly expressed and that IL-12p35 is inhibited in human CD14(+) monocytes, while IL-17 expression is upregulated in CD4(+) T cells, in chronically HCV-infected individuals compared to healthy subjects. Interestingly, Tim-3 expression is closely associated with the differential regulation of IL-12/IL-23 expression in CD14(+) monocytes and correlated to IL-17 production by CD4(+) T cells. These Tim-3-associated IL-12/IL-23/IL-17 dysregulations in HCV-infected individuals are also recapitulated in vitro by incubating healthy monocytes or peripheral blood mononuclear cells with Huh-7 hepatoma cells transfected with HCV RNA. Importantly, blocking Tim-3 signaling on monocytes restores the balance of IL-12/IL-23 through the intracellular STAT3 signaling, which in turn reverses the upregulated IL-17 expression both ex vivo and in vitro. Our findings suggest that Tim-3-mediated differential regulation of IL-12/IL-23 drives T(H)17 cell development, a milieu favoring viral persistence and autoimmune phenomenon during HCV infection.


Subject(s)
Cell Differentiation , Hepatitis C, Chronic/immunology , Interleukin-12/biosynthesis , Interleukin-23/biosynthesis , Membrane Proteins/metabolism , Th17 Cells/immunology , Hepatitis A Virus Cellular Receptor 2 , Humans , Lymphocyte Subsets/immunology , Lymphocyte Subsets/physiology , Th17 Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...