Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Nature ; 614(7947): 334-342, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36697826

RESUMEN

The liver is bathed in bacterial products, including lipopolysaccharide transported from the intestinal portal vasculature, but maintains a state of tolerance that is exploited by persistent pathogens and tumours1-4. The cellular basis mediating this tolerance, yet allowing a switch to immunity or immunopathology, needs to be better understood for successful immunotherapy of liver diseases. Here we show that a variable proportion of CD8+ T cells compartmentalized in the human liver co-stain for CD14 and other prototypic myeloid membrane proteins and are enriched in close proximity to CD14high myeloid cells in hepatic zone 2. CD14+CD8+ T cells preferentially accumulate within the donor pool in liver allografts, among hepatic virus-specific and tumour-infiltrating responses, and in cirrhotic ascites. CD14+CD8+ T cells exhibit increased turnover, activation and constitutive immunomodulatory features with high homeostatic IL-10 and IL-2 production ex vivo, and enhanced antiviral/anti-tumour effector function after TCR engagement. This CD14+CD8+ T cell profile can be recapitulated by the acquisition of membrane proteins-including the lipopolysaccharide receptor complex-from mononuclear phagocytes, resulting in augmented tumour killing by TCR-redirected T cells in vitro. CD14+CD8+ T cells express integrins and chemokine receptors that favour interactions with the local stroma, which can promote their induction through CXCL12. Lipopolysaccharide can also increase the frequency of CD14+CD8+ T cells in vitro and in vivo, and skew their function towards the production of chemotactic and regenerative cytokines. Thus, bacterial products in the gut-liver axis and tissue stromal factors can tune liver immunity by driving myeloid instruction of CD8+ T cells with immunomodulatory ability.


Asunto(s)
Linfocitos T CD8-positivos , Tolerancia Inmunológica , Receptores de Lipopolisacáridos , Lipopolisacáridos , Hígado , Células Mieloides , Humanos , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Células Mieloides/inmunología , Células Mieloides/metabolismo , Neoplasias/inmunología , Neoplasias/patología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Tolerancia Inmunológica/efectos de los fármacos , Tolerancia Inmunológica/inmunología , Hígado/efectos de los fármacos , Hígado/inmunología , Hígado/patología , Hígado/virología , Interleucina-2/biosíntesis , Interleucina-2/inmunología , Quimiotaxis de Leucocito , Bacterias/inmunología , Intestinos/inmunología , Intestinos/microbiología
2.
Hepatology ; 72(5): 1528-1540, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32770836

RESUMEN

BACKGROUND AND AIMS: Therapies for chronic hepatitis B virus (HBV) infection are urgently needed because of viral integration, persistence of viral antigen expression, inadequate HBV-specific immune responses, and treatment regimens that require lifelong adherence to suppress the virus. Immune mobilizing monoclonal T Cell receptors against virus (ImmTAV) molecules represent a therapeutic strategy combining an affinity-enhanced T Cell receptor with an anti-CD3 T Cell-activating moiety. This bispecific fusion protein redirects T cells to specifically lyse infected cells expressing the target virus-derived peptides presented by human leukocyte antigen (HLA). APPROACH AND RESULTS: ImmTAV molecules specific for HLA-A*02:01-restricted epitopes from HBV envelope, polymerase, and core antigens were engineered. The ability of ImmTAV-Env to activate and redirect polyclonal T cells toward cells containing integrated HBV and cells infected with HBV was assessed using cytokine secretion assays and imaging-based killing assays. Elimination of infected cells was further quantified using a modified fluorescent hybridization of viral RNA assay. Here, we demonstrate that picomolar concentrations of ImmTAV-Env can redirect T cells from healthy and HBV-infected donors toward hepatocellular carcinoma (HCC) cells containing integrated HBV DNA resulting in cytokine release, which could be suppressed by the addition of a corticosteroid in vitro. Importantly, ImmTAV-Env redirection of T cells induced cytolysis of antigen-positive HCC cells and cells infected with HBV in vitro, causing a reduction of hepatitis B e antigen and specific loss of cells expressing viral RNA. CONCLUSIONS: The ImmTAV platform has the potential to enable the elimination of infected cells by redirecting endogenous non-HBV-specific T cells, bypassing exhausted HBV-specific T cells. This represents a promising therapeutic option in the treatment of chronic hepatitis B, with our lead candidate now entering trials.


Asunto(s)
Virus de la Hepatitis B/inmunología , Hepatitis B Crónica/tratamiento farmacológico , Receptores de Antígenos de Linfocitos T/uso terapéutico , Proteínas Recombinantes de Fusión/farmacología , Linfocitos T/efectos de los fármacos , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Complejo CD3/antagonistas & inhibidores , Línea Celular Tumoral , Epítopos/inmunología , Antígeno HLA-A2/inmunología , Antígenos de Superficie de la Hepatitis B/inmunología , Virus de la Hepatitis B/aislamiento & purificación , Hepatitis B Crónica/inmunología , Hepatitis B Crónica/virología , Hepatocitos , Humanos , Inmunoconjugados/genética , Inmunoconjugados/inmunología , Inmunoconjugados/farmacología , Inmunoconjugados/uso terapéutico , Activación de Linfocitos/efectos de los fármacos , Cultivo Primario de Células , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/uso terapéutico , Linfocitos T/inmunología
3.
J Immunol ; 197(11): 4283-4291, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27798170

RESUMEN

Human liver contains an Eomeshi population of NK cells that is not present in the blood. In this study, we show that these cells are characterized by a molecular signature that mediates their retention in the liver. By examining liver transplants where donors and recipients are HLA mismatched, we distinguish between donor liver-derived and recipient-derived leukocytes to show that Eomeslo NK cells circulate freely whereas Eomeshi NK cells are unable to leave the liver. Furthermore, Eomeshi NK cells are retained in the liver for up to 13 y. Therefore, Eomeshi NK cells are long-lived liver-resident cells. We go on to show that Eomeshi NK cells can be recruited from the circulation during adult life and that circulating Eomeslo NK cells are able to upregulate Eomes and molecules mediating liver retention under cytokine conditions similar to those in the liver. This suggests that circulating NK cells are a precursor of their liver-resident counterparts.


Asunto(s)
Células Asesinas Naturales/inmunología , Hígado/inmunología , Proteínas de Dominio T Box/inmunología , Regulación hacia Arriba/inmunología , Adulto , Femenino , Humanos , Células Asesinas Naturales/citología , Hígado/patología , Trasplante de Hígado , Masculino , Factores de Tiempo , Donantes de Tejidos
4.
Eur J Immunol ; 45(12): 3431-40, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26420375

RESUMEN

Early production of pro-inflammatory cytokines, including IFN-γ, is essential for control of blood-stage malaria infections. We have shown that IFN-γ production can be induced among human natural killer (NK) cells by coculture with Plasmodium falciparum infected erythrocytes, but the importance of this response is unclear. To further explore the role of NK cells during malaria infection, we have characterized the NK-cell response of C57BL/6 mice during lethal (PyYM) or nonlethal (Py17XNL) P. yoelii infection. Ex vivo flow cytometry revealed that NK cells are activated within 24 h of Py17XNL blood-stage infection, expressing CD25 and producing IFN-γ; this response was blunted and delayed during PyYM infection. CD25 expression and IFN-γ production were highly correlated, suggesting a causal relationship between the two responses. Subsequent in vitro experiments revealed that IL-18 signaling is essential for induction of CD25 and synergizes with IL-12 to enhance CD25 expression on splenic NK cells. In accordance with this, Py17XNL-infected erythrocytes induced NK-cell CD25 expression and IFN-γ production in a manner that is completely IL-18- and partially IL-12-dependent, and IFN-γ production is enhanced by IL-2. These data suggest that IL-2 signaling via CD25 amplifies IL-18- and IL-12-mediated NK-cell activation during malaria infection.


Asunto(s)
Interferón gamma/biosíntesis , Interleucina-18/farmacología , Células Asesinas Naturales/efectos de los fármacos , Malaria/inmunología , Plasmodium yoelii , Receptores de Interleucina-2/metabolismo , Animales , Femenino , Interleucina-12/farmacología , Subunidad alfa del Receptor de Interleucina-2/análisis , Células Asesinas Naturales/inmunología , Ratones , Ratones Endogámicos C57BL
5.
Sci Rep ; 14(1): 14077, 2024 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-38890397

RESUMEN

Extracorporeal haemofiltration devices that selectively remove cytokines could represent an adjunctive treatment in inflammatory diseases. One such device is the "IL-6-Sieve", wherein magnetic Anti-IL-6 Beads are introduced into an extracorporeal circuit via a Bead Adapter and then removed along with any surface-bound interleukin (IL)-6 by a Filter deployed in a Magnet, before the blood is returned to the patient. We report here on a series of animal studies, and a first-in-human study, on the safety of the IL-6-Sieve. Evaluations focused on the: (a) safety of Filter and Magnet placed in an extracorporeal circuit in sheep; (b) safety of Anti-IL-6 Beads-directly infused intravenously as worst case scenario of misuse; or injected into an extracorporeal circuit using the Bead Adapter, Filter, and Magnet as intended-in sheep; (c) biodistribution of Anti-IL-6 Beads intravenously infused in mice; and (d) safety of Filter and Magnet placed in an extracorporeal circuit in healthy volunteers. No serious adverse events or significant changes in vital signs or routine laboratory parameters occurred in any of the animals or humans. Although safety of the IL-6-Sieve requires further study, these initial evaluations represent a promising start for the translation of this new blood purification modality into clinical use.


Asunto(s)
Hemofiltración , Interleucina-6 , Hemofiltración/instrumentación , Hemofiltración/métodos , Animales , Humanos , Ovinos , Ratones , Interleucina-6/sangre , Femenino , Masculino , Adulto
6.
J Infect Dis ; 205(9): 1351-62, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22457290

RESUMEN

BACKGROUND: Natural killer (NK) cells are an important component of the innate immune defense against viruses, including hepatitis C virus (HCV). The cell culture system using HCV-permissive Huh-7.5 cells make studies on interaction of NK cells and HCV-infected target cells possible. We used this system to characterize interactions of HCV-infected Huh-7.5 cells and NK cells from healthy controls and patients with acute HCV infection. METHODS: IFNα- and IL-2 stimulated NK cells were cultured with HCV-infected hepatoma cells and subsequently analyzed (for degranulation and cytokine production) via multicolour flow cytometry. Luciferase assyas have been used to study inhibition of HCV replication. Further, PBMC from patients with acute hepatitis C as well as HCV-infected Huh7.5 cells have been analyzed via flow cytometry for expression of NK cell receptors and ligands, respectively. RESULTS: After interferon (IFN) α stimulation, NK cells from healthy controls and patients with acute hepatitis C efficiently recognized both HCV-infected and uninfected hepatoma cells. Subsequent dissection of receptor-ligand interaction revealed a dominant role for DNAM-1 and a complementary contribution of NKG2D for NK cell activation in this setting. Furthermore, IFN-α-stimulated NK cells effectively inhibited HCV replication in a DNAM-1-dependent manner. CONCLUSIONS: Human NK cells recognize HCV-infected hepatoma cells after IFN-α stimulation in a DNAM-1-dependent manner. Furthermore, interaction of IFN-α-stimulated NK cells with HCV-infected hepatoma cells efficiently reduced HCV replication. This study opens up future studies of NK cell interaction with HCV-infected hepatocytes to gain further insight into the pathogenesis of human HCV infection and the therapeutic effects of IFN-α.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/metabolismo , Antivirales/uso terapéutico , Hepacivirus/efectos de los fármacos , Hepatitis C/patología , Interferón-alfa/uso terapéutico , Células Asesinas Naturales/inmunología , Enfermedad Aguda , Línea Celular , Hepacivirus/crecimiento & desarrollo , Hepacivirus/patogenicidad , Hepatitis C/inmunología , Hepatocitos/metabolismo , Hepatocitos/virología , Humanos , Inmunidad Innata , Interleucina-2/uso terapéutico , Células Asesinas Naturales/metabolismo , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Activación de Linfocitos , Receptores de Células Asesinas Naturales/metabolismo
8.
STAR Protoc ; 3(2): 101356, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35516846

RESUMEN

With the growing appreciation of tissue-resident immunity, studying tissue-specific immune cells contributing to both homeostasis and disease is imperative. Here, we provide a protocol for the isolation of human intrahepatic leukocytes (IHL) maximizing viability, purity, and yield. Our protocol is scalable by tissue weight, allowing for reproducible and efficient IHL liberation suitable for functional characterization, cell isolation, and profiling by flow (or mass) cytometry. Furthermore, we provide a "guide" to determine an expected IHL yield per gram of tissue processed. For complete details on the use and execution of this protocol, please refer to Stegmann et al. (2016), Pallett et al. (2017), Easom et al. (2018), Swadling et al. (2020), Pallett et al. (2020), and Zakeri et al. (2022).


Asunto(s)
Leucocitos , Linfocitos , Separación Celular/métodos , Citometría de Flujo/métodos , Humanos
9.
Sci Transl Med ; 14(640): eabi4670, 2022 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-35417187

RESUMEN

A better understanding of mechanisms that regulate CD8+T cell responses to therapeutic vaccines is needed to develop approaches to enhance vaccine efficacy for chronic viral infections and cancers. We show here that NK cell depletion enhanced antigen-specific T cell responses to chimp adenoviral vector (ChAdOx) vaccination in a mouse model of chronic HBV infection (CHB). Probing the mechanism underlying this negative regulation, we observed that CHB drove parallel up-regulation of programmed cell death ligand 1 (PD-L1) on liver-resident NK cells and programmed cell death 1 (PD-1) on intrahepatic T cells. PD-L1-expressing liver-resident NK cells suppressed PD-1hiCD8+T cells enriched within the HBV-specific response to therapeutic vaccination. Cytokine activation of NK cells also induced PD-L1, and combining cytokine activation with PD-L1 blockade resulted in conversion of NK cells into efficient helpers that boosted HBV-specific CD8+T cell responses to therapeutic vaccination in mice and to chronic infection in humans. Our findings delineate an immunotherapeutic combination that can boost the response to therapeutic vaccination in CHB and highlight the broader importance of PD-L1-dependent regulation of T cells by cytokine-activated NK cells.


Asunto(s)
Antígeno B7-H1 , Vacunas , Animales , Antígeno B7-H1/metabolismo , Linfocitos T CD8-positivos , Citocinas/metabolismo , Células Asesinas Naturales , Ratones , Receptor de Muerte Celular Programada 1/metabolismo
10.
Gastroenterology ; 138(5): 1885-97, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20334827

RESUMEN

BACKGROUND & AIMS: Pegylated interferon-alpha (PEG-IFNalpha), in combination with ribavirin, controls hepatitis C virus (HCV) infection in approximately 50% of patients by mechanisms that are not completely understood. Beside a direct antiviral effect, different immunomodulatory effects have been discussed. Natural killer (NK) cells might be associated with control of HCV infection. We examined the effects of IFNalpha on human NK cells and its relevance to HCV infection. METHODS: We performed gene expression profiling studies of NK cells following stimulation of peripheral blood mononuclear cells with IFNalpha. We evaluated IFNalpha-induced tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) expression using flow cytometry analyses of NK cells isolated from patients with acute or chronic hepatitis C that had received PEG-IFNalpha therapy. RESULTS: TRAIL was among the most up-regulated genes after IFNalpha stimulation of NK cells from healthy controls. After in vitro stimulation with IFNalpha, CD56(dim) NK cells from patients who had responded to PEG-IFNalpha therapy expressed higher levels of TRAIL than cells from patients with chronic hepatitis C. TRAIL expression, ex vivo, was inversely correlated with HCV-RNA levels during the early phase of PEG-IFNalpha therapy. In patients with acute hepatitis C, TRAIL expression on CD56(bright) NK cells increased significantly compared with cells from controls. In in vitro studies, IFNalpha-stimulated NK cells eliminated HCV-replicating hepatoma cells by a TRAIL-mediated mechanism. CONCLUSIONS: IFNalpha-induced expression of TRAIL on NK cells is associated with control of HCV infection; these observations might account for the second-phase decline in HCV-RNA levels during PEG-IFNalpha therapy.


Asunto(s)
Antivirales/uso terapéutico , Citotoxicidad Inmunológica/efectos de los fármacos , Hepatitis C Crónica/tratamiento farmacológico , Interferón-alfa/uso terapéutico , Células Asesinas Naturales/efectos de los fármacos , Polietilenglicoles/uso terapéutico , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Adulto , Antígeno CD56/análisis , Línea Celular Tumoral , Técnicas de Cocultivo , Relación Dosis-Respuesta a Droga , Femenino , Perfilación de la Expresión Génica , Hepacivirus/efectos de los fármacos , Hepacivirus/genética , Hepacivirus/patogenicidad , Hepatitis C Crónica/genética , Hepatitis C Crónica/inmunología , Humanos , Interferón alfa-2 , Células Asesinas Naturales/inmunología , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , ARN Viral/sangre , Proteínas Recombinantes , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Factores de Tiempo , Resultado del Tratamiento , Regulación hacia Arriba , Carga Viral , Replicación Viral/efectos de los fármacos
11.
Mol Ther Methods Clin Dev ; 19: 149-161, 2020 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-33102612

RESUMEN

Emerging base editing technology exploits CRISPR RNA-guided DNA modification effects for highly specific C > T conversion, which has been used to efficiently disrupt gene expression. These tools can enhance synthetic T cell immunity by restricting specificity, addressing histocompatibility leukocyte antigen (HLA) barriers, and promoting persistence. We report lentiviral delivery of a hepatitis B-virus (HBV)-specific recombinant T cell receptor (rTCR) and a linked CRISPR single-guide RNA for simultaneous disruption of endogenous TCRs (eTCRs) when combined with transient cytosine deamination. Discriminatory depletion of eTCR and coupled expression of rTCR resulted in enrichment of HBV-specific populations from 55% (SEM, ±2.4%) to 95% (SEM, ±0.5%). Intensity of rTCR expression increased 1.8- to 2.9-fold compared to that in cells retaining their competing eTCR, and increased cytokine production and killing of HBV antigen-expressing hepatoma cells in a 3D microfluidic model were exhibited. Molecular signatures confirmed that seamless conversion of C > T (G > A) had created a premature stop codon in TCR beta constant 1/2 loci, with no notable activity at predicted off-target sites. Thus, targeted disruption of eTCR by cytosine deamination and discriminatory enrichment of antigen-specific T cells offers the prospect of enhanced, more specific T cell therapies against HBV-associated hepatocellular carcinoma (HCC) as well as other viral and tumor antigens.

12.
Cell Rep ; 30(3): 687-698.e6, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31968246

RESUMEN

Tissue-resident memory T cells have critical roles in long-term pathogen and tumor immune surveillance in the liver. We investigate the role of autophagy in equipping human memory T cells to acquire tissue residence and maintain functionality in the immunosuppressive liver environment. By performing ex vivo staining of freshly isolated cells from human liver tissue, we find that an increased rate of basal autophagy is a hallmark of intrahepatic lymphocytes, particularly liver-resident CD8+ T cells. CD8+ T cells with increased autophagy are those best able to proliferate and mediate cytotoxicity and cytokine production. Conversely, blocking autophagy induction results in the accumulation of depolarized mitochondria, a feature of exhausted T cells. Primary hepatic stellate cells or the prototypic hepatic cytokine interleukin (IL)-15 induce autophagy in parallel with tissue-homing/retention markers. Inhibition of T cell autophagy abrogates tissue-residence programming. Thus, upregulation of autophagy adapts CD8+ T cells to combat mitochondrial depolarization, optimize functionality, and acquire tissue residence.


Asunto(s)
Autofagia , Linfocitos T CD8-positivos/citología , Memoria Inmunológica , Hígado/citología , Hígado/inmunología , Diferenciación Celular , Proliferación Celular , Humanos , Mitocondrias/metabolismo
13.
J Med Virol ; 81(12): 2079-88, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19856479

RESUMEN

Parvovirus B19 (B19V) has been detected in the liver of Asian patients infected with HBV and may contribute to acute and chronic liver disease. This study aimed to investigate the impact of B19V infection in European patients with viral hepatitis. B19V DNA was detected in 1/91 and 0/50 serum samples from patients with chronic hepatitis C and B, respectively. In contrast, B19V DNA was amplified frequently from explanted end-stage liver tissues (37/50, 74%) and from routine biopsy samples (14/32, 44%) (P < 0.05). However, there was no significant difference in B19V copy number per cell between these two groups. B19V-specific CD4(+) T-cell responses to two dominant MHC-class-restricted epitopes were detected in a similar frequency in healthy anti-B19V-positive individuals (3/19; 16%) and patients with chronic hepatitis C (3/13; 23%). These results indicate that B19V can persist in the liver. However, there is no evidence that B19V is a "hepatitis virus" worsening liver disease in European patients with chronic hepatitis C.


Asunto(s)
Hepatitis B Crónica/virología , Hepatitis C Crónica/virología , Hígado/virología , Infecciones por Parvoviridae/virología , Parvovirus B19 Humano/aislamiento & purificación , Adulto , Anciano , Anticuerpos Antivirales/inmunología , Linfocitos T CD4-Positivos/inmunología , ADN Viral/genética , ADN Viral/aislamiento & purificación , Progresión de la Enfermedad , Femenino , Alemania , Hepatitis B Crónica/complicaciones , Hepatitis B Crónica/inmunología , Hepatitis B Crónica/patología , Hepatitis C Crónica/complicaciones , Hepatitis C Crónica/inmunología , Hepatitis C Crónica/patología , Humanos , Hígado/patología , Masculino , Persona de Mediana Edad , Infecciones por Parvoviridae/complicaciones , Infecciones por Parvoviridae/inmunología , Parvovirus B19 Humano/genética , Parvovirus B19 Humano/inmunología , Factores de Tiempo
14.
Front Immunol ; 9: 1009, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29867983

RESUMEN

NK cells have potent antitumor capacity. They are enriched in the human liver, with a large subset specialized for tissue-residence. The potential for liver-resident versus liver-infiltrating NK cells to populate, and exert antitumor functions in, human liver tumors has not been studied. We examined liver-resident and liver-infiltrating NK cells directly ex vivo from human hepatocellular carcinomas (HCCs) and liver colorectal (CRC) metastases, compared with matched uninvolved liver tissue. We found that NK cells were highly prevalent in both HCC and liver CRC metastases, although at lower frequencies than unaffected liver. Up to 79% of intratumoral NK cells had the CXCR6+CD69+ liver-resident phenotype. Direct ex vivo staining showed that liver-resident NK cells had increased NKG2D expression compared to their non-resident counterparts, but both subsets had NKG2D downregulation within liver tumors compared to uninvolved liver. Proliferation of intratumoral NK cells (identified by Ki67) was selectively impaired in those with the most marked NKG2D downregulation. Human liver tumor NK cells were functionally impaired, with reduced capacity for cytotoxicity and production of cytokines, even when compared to the hypo-functional tissue-resident NK cells in unaffected liver. Coculture of human liver NK cells with the human hepatoma cell line PLC/PRF/5, or with autologous HCC, recapitulated the defects observed in NK cells extracted from tumors, with downmodulation of NKG2D, cytokine production, and target cell cytotoxicity. Transwells and conditioned media confirmed a requirement for cell contact with PLC/PRF/5 to impose NK cell inhibition. IL-15 was able to recover antitumor functionality in NK cells inhibited by in vitro exposure to HCC cell lines or extracted directly from HCC. In summary, our data suggest that the impaired antitumor function of local NK cells reflects a combination of the tolerogenic features inherent to liver-resident NK cells together with additional contact-dependent inhibition imposed by HCC itself. The demonstration that IL-15 can recover hepatic NK cell function following tumor exposure supports its inclusion in immunotherapy strategies.


Asunto(s)
Carcinoma Hepatocelular/inmunología , Interleucina-15/farmacología , Células Asesinas Naturales/efectos de los fármacos , Neoplasias Hepáticas/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/genética , Carcinoma Hepatocelular/complicaciones , Línea Celular Tumoral , Movimiento Celular , Técnicas de Cocultivo , Citocinas/inmunología , Citotoxicidad Inmunológica , Regulación hacia Abajo , Humanos , Inmunoterapia , Células Asesinas Naturales/patología , Neoplasias Hepáticas/terapia , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Transducción de Señal
15.
J Exp Med ; 214(6): 1567-1580, 2017 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-28526759

RESUMEN

The liver provides a tolerogenic immune niche exploited by several highly prevalent pathogens as well as by primary and metastatic tumors. We have sampled healthy and hepatitis B virus (HBV)-infected human livers to probe for a subset of T cells specialized to overcome local constraints and mediate immunity. We characterize a population of T-betloEomesloBlimp-1hiHobitlo T cells found within the intrahepatic but not the circulating memory CD8 T cell pool expressing liver-homing/retention markers (CD69+CD103+ CXCR6+CXCR3+). These tissue-resident memory T cells (TRM) are preferentially expanded in patients with partial immune control of HBV infection and can remain in the liver after the resolution of infection, including compartmentalized responses against epitopes within all major HBV proteins. Sequential IL-15 or antigen exposure followed by TGFß induces liver-adapted TRM, including their signature high expression of exhaustion markers PD-1 and CD39. We suggest that these inhibitory molecules, together with paradoxically robust, rapid, cell-autonomous IL-2 and IFNγ production, equip liver CD8 TRM to survive while exerting local noncytolytic hepatic immunosurveillance.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Interleucina-2/metabolismo , Hígado/inmunología , Hígado/virología , Antígenos/inmunología , Antígenos CD/metabolismo , Comunicación Autocrina/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Granzimas/metabolismo , Virus de la Hepatitis B/efectos de los fármacos , Hepatitis B Crónica/inmunología , Hepatitis B Crónica/patología , Hepatitis B Crónica/virología , Humanos , Memoria Inmunológica/efectos de los fármacos , Interleucina-15/farmacología , Hígado/efectos de los fármacos , Hígado/patología , Fenotipo , Receptor de Muerte Celular Programada 1/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores CXCR6 , Receptores de Quimiocina/metabolismo , Receptores Virales/metabolismo , Factor de Crecimiento Transformador beta/farmacología
16.
Sci Rep ; 6: 26157, 2016 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-27210614

RESUMEN

Natural killer cells (NK) are highly enriched in the human liver, where they can regulate immunity and immunopathology. We probed them for a liver-resident subset, distinct from conventional bone-marrow-derived NK. CXCR6+ NK were strikingly enriched in healthy and diseased liver compared to blood (p < 0.0001). Human hepatic CXCR6+ NK had an immature phenotype (predominantly CD56(bright)CD16-CD57-), and expressed the tissue-residency marker CD69. CXCR6+ NK produced fewer cytotoxic mediators and pro-inflammatory cytokines than the non-liver-specific CXCR6- fraction. Instead CXCR6+ NK could upregulate TRAIL, a key death ligand in hepatitis pathogenesis. CXCR6 demarcated liver NK into two transcriptionally distinct populations: T-bet(hi)Eomes(lo)(CXCR6-) and T-bet(lo)Eomes(hi)(CXCR6+); the latter was virtually absent in the periphery. The small circulating CXCR6+ subset was predominantly T-bet(hi)Eomes(lo), suggesting its lineage was closer to CXCR6- peripheral than CXCR6+ liver NK. These data reveal a large subset of human liver-resident T-bet(lo)Eomes(hi) NK, distinguished by their surface expression of CXCR6, adapted for hepatic tolerance and inducible anti-viral immunity.


Asunto(s)
Células Asesinas Naturales/inmunología , Hígado/inmunología , Subgrupos Linfocitarios/inmunología , Receptores CXCR6/análisis , Proteínas de Dominio T Box/análisis , Adolescente , Adulto , Anciano , Citocinas/metabolismo , Femenino , Humanos , Inmunofenotipificación , Células Asesinas Naturales/química , Células Asesinas Naturales/clasificación , Subgrupos Linfocitarios/química , Subgrupos Linfocitarios/clasificación , Masculino , Persona de Mediana Edad , Adulto Joven
17.
Front Immunol ; 2: 88, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22566877

RESUMEN

Natural killer (NK) cells are large granular lymphocytes that express a diverse array of germline encoded inhibitory and activating receptors for MHC Class I and Class I-like molecules, classical co-stimulatory ligands, and cytokines. The ability of NK cells to be very rapidly activated by inflammatory cytokines, to secrete effector cytokines, and to kill infected or stressed host cells, suggests that they may be among the very early responders during infection. Recent studies have also identified a small number of pathogen-derived ligands that can bind to NK cell surface receptors and directly induce their activation. Here we review recent studies that have begun to elucidate the various pathways by which viral, bacterial, and parasite pathogens activate NK cells. We also consider two emerging themes of NK cell-pathogen interactions, namely their contribution to adaptive immune responses and their potential to take on regulatory and immunomodulatory functions.

18.
J Leukoc Biol ; 85(3): 452-61, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19095736

RESUMEN

Prevention of overwhelming immune reactions is essential for an organism to survive. Adenosine, a ribonucleoside produced by various cell types during inflammatory processes, has been shown to inhibit effector functions of different immune cells. Here, we show that the adenosine A(3) receptor agonist iodobenzyl methylcarboxamidoadenosine potently inhibited proliferation, IFN-gamma production, and cytotoxicity of activated human lymphoid cells. Stimulation of the A(3) receptor also caused apoptosis of activated PBMC. However, when PBMC were stimulated with IFN-alpha, adenosine did not decrease, but synergistically increased, the IFN-gamma production of NK cells. This effect was also mediated mainly via the A(3) receptor. Thus, our data suggest that adenosine differentially contributes to the regulation of immune responses during inflammatory processes: It may increase effector functions of NK cells in combination with IFN-alpha but also prevents overwhelming immune responses by inhibiting proliferation and induction of apoptosis of activated lymphoid cells. Future studies need to define the role of the different adenosine receptors in more detail.


Asunto(s)
Interferón-alfa/farmacología , Interferón gamma/biosíntesis , Células Asesinas Naturales/inmunología , Receptor de Adenosina A3/fisiología , Adenosina/análisis , Apoptosis , Proliferación Celular , Humanos , Inmunidad , Inflamación , Células Asesinas Naturales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA