Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Immunology ; 145(4): 485-97, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25772938

RESUMO

Host immune responses must be tightly regulated by an intricate balance between positive and negative signals while fighting pathogens; persistent pathogens may usurp these regulatory mechanisms to dampen host immunity to facilitate survival in vivo. Here we report that Tim-3, a negative signalling molecule expressed on monocytes and T cells, is up-regulated on natural killer (NK) cells in individuals chronically infected with hepatitis C virus (HCV). Additionally, the transcription factor T-bet was also found to be up-regulated and associated with Tim-3 expression in NK cells during chronic HCV infection. MicroRNA-155 (miR-155), an miRNA that inhibits signalling proteins involved in immune responses, was down-regulated in NK cells by HCV infection. This Tim-3/T-bet over-expression and miR-155 inhibition were recapitulated in vitro by incubating primary NK cells or NK92 cell line with Huh-7 hepatocytes expressing HCV. Reconstitution of miR-155 in NK cells from HCV-infected patients led to a decrease in T-bet/Tim-3 expression and an increase in interferon-γ production. Blocking Tim-3 signalling also enhanced interferon-γ production in NK cells by improving signal transducer and activator of transcription-5 phosphorylation. These data indicate that HCV-induced, miR-155-regulated Tim-3 expression regulates NK cell function, suggesting a novel mechanism for balancing immune clearance and immune injury during chronic viral infection.


Assuntos
Hepatite C Crônica/imunologia , Interferon gama/imunologia , Células Matadoras Naturais/imunologia , Proteínas de Membrana/imunologia , MicroRNAs/imunologia , Transdução de Sinais/imunologia , Regulação para Cima/imunologia , Adulto , Idoso , Linhagem Celular , Feminino , Receptor Celular 2 do Vírus da Hepatite A , Hepatite C Crônica/patologia , Hepatócitos/imunologia , Hepatócitos/patologia , Humanos , Células Matadoras Naturais/patologia , Masculino , Pessoa de Meia-Idade , Proteínas com Domínio T/imunologia
2.
J Immunol ; 192(2): 649-57, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24337749

RESUMO

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.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor de Quinase Dependente de Ciclina p27/genética , Vacinas contra Hepatite B/imunologia , Hepatite B/imunologia , Hepatite C/imunologia , Lectinas Tipo C/genética , Transativadores/genética , Envelhecimento/genética , Envelhecimento/imunologia , Linfócitos T CD4-Positivos/virologia , Proliferação de Células , Células Cultivadas , Coinfecção/genética , Coinfecção/imunologia , Ciclina E/genética , Ciclina E/imunologia , Quinase 2 Dependente de Ciclina/genética , Quinase 2 Dependente de Ciclina/imunologia , Inibidor p16 de Quinase Dependente de Ciclina/imunologia , Inibidor de Quinase Dependente de Ciclina p27/imunologia , Hepacivirus/imunologia , Hepatite B/genética , Hepatite B/prevenção & controle , Vírus da Hepatite B/imunologia , Hepatite C/genética , Hepatite C/virologia , Humanos , Interleucina-2/genética , Interleucina-2/imunologia , Lectinas Tipo C/imunologia , Fosforilação/genética , Fosforilação/imunologia , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/imunologia , Receptores Imunológicos , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Transativadores/imunologia
3.
PLoS One ; 8(8): e72488, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23967307

RESUMO

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.


Assuntos
Galectinas/metabolismo , Regulação da Expressão Gênica , Interleucina-12/genética , Interleucina-23/genética , Proteínas de Membrana/metabolismo , Monócitos/citologia , Receptores Toll-Like/metabolismo , Linhagem Celular , Galectinas/deficiência , Galectinas/genética , Inativação Gênica , Receptor Celular 2 do Vírus da Hepatite A , Humanos , Espaço Intracelular/metabolismo , Macrófagos/citologia , Macrófagos/imunologia , Macrófagos/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Fosforilação , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Transcrição Gênica
4.
J Virol ; 87(21): 11626-36, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23966413

RESUMO

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.


Assuntos
Hepatite C Crônica/imunologia , Células Matadoras Naturais/imunologia , Lectinas Tipo C/metabolismo , Transdução de Sinais , Transativadores/metabolismo , Apoptose , Antígeno CD56/análise , Proliferação de Células , Feminino , Humanos , Interferon gama/metabolismo , Células Matadoras Naturais/química , Masculino , Receptores Imunológicos
5.
Vaccine ; 31(18): 2238-45, 2013 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-23499521

RESUMO

Hepatitis B virus (HBV) vaccination is recommended for individuals with hepatitis C virus (HCV) infection given their shared risk factors and increased liver-related morbidity and mortality upon super-infection. Vaccine responses in this setting are often blunted, with poor response rates to HBV vaccinations in chronically HCV-infected individuals compared to healthy subjects. In this study, we investigated the role of T cell immunoglobulin mucin domain-3 (Tim-3)-mediated immune regulation in HBV vaccine responses during HCV infection. We found that Tim-3, a marker for T cell exhaustion, was over-expressed on monocytes, leading to a differential regulation of IL-12/IL-23 production which in turn TH17 cell accumulation, in HCV-infected HBV vaccine non-responders compared to HCV-infected HBV vaccine responders or healthy subjects (HS). Importantly, ex vivo blockade of Tim-3 signaling corrected the imbalance of IL-12/IL-23 as well as the IL-17 bias observed in HBV vaccine non-responders during HCV infection. These results suggest that Tim-3-mediated dysregulation of innate to adaptive immune responses is involved in HBV vaccine failure in individuals with chronic HCV infection, raising the possibility that blocking this negative signaling pathway might improve the success rate of HBV immunization in the setting of chronic viral infection.


Assuntos
Vacinas contra Hepatite B/imunologia , Hepatite B/prevenção & controle , Hepatite C Crônica/imunologia , Interleucina-12/imunologia , Interleucina-23/imunologia , Proteínas de Membrana/imunologia , Células Th17/imunologia , Imunidade Adaptativa , Voluntários Saudáveis , Receptor Celular 2 do Vírus da Hepatite A , Anticorpos Anti-Hepatite B/sangue , Hepatite C Crônica/metabolismo , Humanos , Imunidade Inata , Interleucina-17/imunologia , Fígado/imunologia , Fígado/metabolismo , Fígado/virologia , Masculino , Proteínas de Membrana/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Transdução de Sinais/imunologia
6.
J Virol ; 87(8): 4372-83, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23388728

RESUMO

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.


Assuntos
Diferenciação Celular , Hepatite C Crônica/imunologia , Interleucina-12/biossíntese , Interleucina-23/biossíntese , Proteínas de Membrana/metabolismo , Células Th17/imunologia , Receptor Celular 2 do Vírus da Hepatite A , Humanos , Subpopulações de Linfócitos/imunologia , Subpopulações de Linfócitos/fisiologia , Células Th17/fisiologia
7.
Eur J Immunol ; 43(2): 458-67, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23161469

RESUMO

HCV is remarkable at disrupting human immunity to establish chronic infection. The accumulation of Treg cells at the site of infection and upregulation of inhibitory signaling pathways (such as T-cell Ig and mucin domain protein-3 (Tim-3) and galectin-9 (Gal-9)) play pivotal roles in suppressing antiviral effector T (Teff) cells that are essential for viral clearance. While Tim-3/Gal-9 interactions have been shown to negatively regulate Teff cells, their role in regulating Treg cells is poorly understood. To explore how Tim-3/Gal-9 interactions regulate HCV-mediated Treg-cell development, here we provide pilot data showing that HCV-infected human hepatocytes express higher levels of Gal-9 and TGF-ß, and upregulate Tim-3 expression and regulatory cytokines TGF-ß/IL-10 in co-cultured human CD4(+) T cells, driving conventional CD4(+) T cells into CD25(+) Foxp3(+) Treg cells. Additionally, recombinant Gal-9 protein can transform TCR-activated CD4(+) T cells into Foxp3(+) Treg cells in a dose-dependent manner. Importantly, blocking Tim-3/Gal-9 ligations abrogates HCV-mediated Treg-cell induction by HCV-infected hepatocytes, suggesting that Tim-3/Gal-9 interactions may regulate human Foxp3(+) Treg-cell development and function during HCV infection.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Galectinas/metabolismo , Hepacivirus/imunologia , Hepatite C Crônica/metabolismo , Hepatócitos/virologia , Proteínas de Membrana/metabolismo , Linfócitos T Reguladores/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Células Cultivadas , Técnicas de Cocultura , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Fatores de Transcrição Forkhead/metabolismo , Galectinas/genética , Galectinas/imunologia , Receptor Celular 2 do Vírus da Hepatite A , Hepatite C Crônica/genética , Hepatite C Crônica/imunologia , Hepatite C Crônica/virologia , Hepatócitos/imunologia , Hepatócitos/metabolismo , Humanos , Subunidade alfa de Receptor de Interleucina-2/genética , Subunidade alfa de Receptor de Interleucina-2/imunologia , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/virologia , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/imunologia , Fator de Crescimento Transformador beta/metabolismo
8.
J Immunol ; 189(2): 755-66, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22706088

RESUMO

Hepatitis C virus (HCV) is remarkable at disrupting human immunity to establish chronic infection. Upregulation of inhibitory signaling pathways (such as T cell Ig and mucin domain protein-3 [Tim-3]) and accumulation of regulatory T cells (Tregs) play pivotal roles in suppressing antiviral effector T cell (Teff) responses that are essential for viral clearance. Although the Tim-3 pathway has been shown to negatively regulate Teffs, its role in regulating Foxp3(+) Tregs is poorly explored. In this study, we investigated whether and how the Tim-3 pathway alters Foxp3(+) Treg development and function in patients with chronic HCV infection. We found that Tim-3 was upregulated, not only on IL-2-producing CD4(+)CD25(+)Foxp3(-) Teffs, but also on CD4(+)CD25(+)Foxp3(+) Tregs, which accumulate in the peripheral blood of chronically HCV-infected individuals when compared with healthy subjects. Tim-3 expression on Foxp3(+) Tregs positively correlated with expression of the proliferation marker Ki67 on Tregs, but it was inversely associated with proliferation of IL-2-producing Teffs. Moreover, Foxp3(+) Tregs were found to be more resistant to, and Foxp3(-) Teffs more sensitive to, TCR activation-induced cell apoptosis, which was reversible by blocking Tim-3 signaling. Consistent with its role in T cell proliferation and apoptosis, blockade of Tim-3 on CD4(+)CD25(+) T cells promoted expansion of Teffs more substantially than Tregs through improving STAT-5 signaling, thus correcting the imbalance of Foxp3(+) Tregs/Foxp3(-) Teffs that was induced by HCV infection. Taken together, the Tim-3 pathway appears to control Treg and Teff balance through altering cell proliferation and apoptosis during HCV infection.


Assuntos
Hepatite C Crônica/imunologia , Hepatite C Crônica/patologia , Receptores Imunológicos/fisiologia , Transdução de Sinais/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/virologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/virologia , Proteínas Reguladoras de Apoptose/fisiologia , Proliferação de Células , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/fisiologia , Hepatite C Crônica/metabolismo , Humanos , Subunidade alfa de Receptor de Interleucina-2/biossíntese , Subunidade alfa de Receptor de Interleucina-2/fisiologia , Projetos Piloto , Subpopulações de Linfócitos T/patologia , Linfócitos T Reguladores/patologia , Viremia/imunologia , Viremia/metabolismo , Viremia/patologia
9.
J Leukoc Biol ; 91(2): 189-96, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21844165

RESUMO

Tim-3 and PD-1 are powerful immunoinhibitory molecules involved in immune tolerance, autoimmune responses, and antitumor or antiviral immune evasion. A current model for Tim-3 regulation during immune responses suggests a divergent function, such that Tim-3 acts synergistically with TLR signaling pathways in innate immune cells to promote inflammation, yet the same molecule terminates Th1 immunity in adaptive immune cells. To better understand how Tim-3 might be functioning in innate immune responses, we examined the kinetics of Tim-3 expression in human CD14+ M/M(Ф) in relation to expression of IL-12, a key cytokine in the transition of innate to adaptive immunity. Here, we show that Tim-3 is constitutively expressed on unstimulated peripheral blood CD14+ monocytes but decreases rapidly upon TLR stimulation. Conversely, IL-12 expression is low in these cells but increases rapidly in CD14+ M/M(Ф) in correlation with the decrease in Tim-3. Blocking Tim-3 signaling or silencing Tim-3 expression led to a significant increase in TLR-mediated IL-12 production, as well as a decrease in activation-induced up-regulation of the immunoinhibitor, PD-1; TNF-α production was not altered significantly, but IL-10 production was increased. These results suggest that Tim-3 has a role as a regulator of pro- and anti-inflammatory innate immune responses.


Assuntos
Citocinas/biossíntese , Macrófagos/metabolismo , Proteínas de Membrana/fisiologia , Monócitos/metabolismo , Anticorpos/farmacologia , Citocinas/genética , Regulação para Baixo , Regulação da Expressão Gênica , Receptor Celular 2 do Vírus da Hepatite A , Humanos , Imunidade Inata , Interleucina-10/biossíntese , Interleucina-10/genética , Interleucina-12/biossíntese , Interleucina-12/genética , Receptores de Lipopolissacarídeos/análise , Proteínas de Membrana/antagonistas & inibidores , Receptor de Morte Celular Programada 1/biossíntese , Receptor de Morte Celular Programada 1/genética , RNA Interferente Pequeno/farmacologia , Receptores Toll-Like/efeitos dos fármacos , Receptores Toll-Like/fisiologia , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética , Regulação para Cima
10.
PLoS One ; 6(5): e19664, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21637332

RESUMO

T cell immunoglobulin and mucin domain-containing protein 3 (Tim-3) is a newly identified negative immunomodulator that is up-regulated on dysfunctional T cells during viral infections. The expression and function of Tim-3 on human innate immune responses during HCV infection, however, remains poorly characterized. In this study, we report that Tim-3 is constitutively expressed on human resting CD14(+) monocyte/macrophages (M/M(Ø)) and functions as a cap to block IL-12, a key pro-inflammatory cytokine linking innate and adaptive immune responses. Tim-3 expression is significantly reduced and IL-12 expression increased upon stimulation with Toll-like receptor 4 (TLR4) ligand--lipopolysaccharide (LPS) and TLR7/8 ligand--R848. Notably, Tim-3 is over-expressed on un-stimulated as well as TLR-stimulated M/M(Ø), which is inversely associated with the diminished IL-12 expression in chronically HCV-infected individuals when compared to healthy subjects. Up-regulation of Tim-3 and inhibition of IL-12 are also observed in M/M(Ø) incubated with HCV-expressing hepatocytes, as well as in primary M/M(Ø) or monocytic THP-1 cells incubated with HCV core protein, an effect that mimics the function of complement C1q and is reversible by blocking the HCV core/gC1qR interaction. Importantly, blockade of Tim-3 signaling significantly rescues HCV-mediated inhibition of IL-12, which is primarily expressed by Tim-3 negative M/M(Ø). Tim-3 blockade reduces HCV core-mediated expression of the negative immunoregulators PD-1 and SOCS-1 and increases STAT-1 phosphorylation. Conversely, blocking PD-1 or silencing SOCS-1 gene expression also decreases Tim-3 expression and enhances IL-12 secretion and STAT-1 phosphorylation. These findings suggest that Tim-3 plays a crucial role in negative regulation of innate immune responses, through crosstalk with PD-1 and SOCS-1 and limiting STAT-1 phosphorylation, and may be a novel target for immunotherapy to HCV infection.


Assuntos
Hepacivirus/imunologia , Hepatite C Crônica/imunologia , Subunidade p40 da Interleucina-12/metabolismo , Proteínas de Membrana/metabolismo , Monócitos/metabolismo , Adulto , Antígenos CD/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Transporte/metabolismo , Técnicas de Cocultura , Complemento C1q/metabolismo , Regulação para Baixo , Inativação Gênica , Receptor Celular 2 do Vírus da Hepatite A , Hepatite C Crônica/virologia , Hepatócitos/metabolismo , Hepatócitos/virologia , Humanos , Subunidade p40 da Interleucina-12/biossíntese , Receptores de Lipopolissacarídeos/metabolismo , Ativação de Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Pessoa de Meia-Idade , Proteínas Mitocondriais/metabolismo , Fosforilação , Receptor de Morte Celular Programada 1 , Ligação Proteica , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Proteína 1 Supressora da Sinalização de Citocina , Proteínas Supressoras da Sinalização de Citocina/genética , Regulação para Cima , Proteínas do Core Viral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...