Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Eur J Immunol ; 54(6): e2350878, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38581345

ABSTRACT

Tumor-associated macrophages (TAM) are abundant in several tumor types and usually correlate with poor prognosis. Previously, we demonstrated that anti-inflammatory macrophages (M2) inhibit NK cell effector functions. Here, we explored the impact of TAM on NK cells in the context of clear-cell renal cell carcinoma (ccRCC). Bioinformatics analysis revealed that an exhausted NK cell signature strongly correlated with an M2 signature. Analysis of TAM from human ccRCC samples confirmed that they exhibited an M2-skewed phenotype and inhibited IFN-γ production by NK cells. Moreover, human M0 macrophages cultured with conditioned media from ccRCC cell lines generated macrophages with an M2-skewed phenotype (TAM-like), which alike TAM, displayed suppressive activity on NK cells. Moreover, TAM depletion in the mouse Renca ccRCC model resulted in delayed tumor growth and reduced volume, accompanied by an increased frequency of IFN-γ-producing tumor-infiltrating NK cells that displayed heightened expression of T-bet and NKG2D and reduced expression of the exhaustion-associated co-inhibitory molecules PD-1 and TIM-3. Therefore, in ccRCC, the tumor microenvironment polarizes TAM toward an immunosuppressive profile that promotes tumor-infiltrating NK cell dysfunction, contributing to tumor progression. In addition, immunotherapy strategies targeting TAM may result in NK cell reinvigoration, thereby counteracting tumor progression.


Subject(s)
Carcinoma, Renal Cell , Interferon-gamma , Kidney Neoplasms , Killer Cells, Natural , Tumor-Associated Macrophages , Killer Cells, Natural/immunology , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Interferon-gamma/metabolism , Interferon-gamma/immunology , Humans , Animals , Mice , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Disease Progression , Cell Line, Tumor , Tumor Microenvironment/immunology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Hepatitis A Virus Cellular Receptor 2/metabolism , Hepatitis A Virus Cellular Receptor 2/immunology , Programmed Cell Death 1 Receptor/metabolism
2.
Enferm. nefrol ; 25(4): 337-342, octubre 2022. tab, graf
Article in Spanish | IBECS | ID: ibc-214109

ABSTRACT

Introducción: La infección del orificio de salida del catéter de diálisis peritoneal es una complicación importante en la diálisis peritoneal, que puede evolucionar hacia una tunelitis y/o infección peritoneal, lo que suele conllevar una retirada del mismo.Objetivo: Analizar el uso de mupirocina en la cura del orificio sano y su repercusión en la ratio de infecciones del orificio de salida y en el porcentaje de infecciones peritoneales dependientes de catéter.Material y Método: Estudio observacional retrospectivo, en una unidad de diálisis peritoneal provincial. Se incluyeron a todos los pacientes mayores de 18 años, que realizaron diálisis peritoneal en domicilio del 01/01/2019 hasta el 31/12/2021. Las variables analizadas, fueron: sexo , ratio de infección de orificio anual, gérmenes responsables, infecciones peritoneales dependientes de catéter y aparición de resistencias. Los datos se obtuvieron del registro anual de la unidad.Resultados: Tras la incorporación del uso de mupirocina 2 veces por semana en la cura del orificio de salida sano disminuyó la ratio de infección del orificio de salida, pasando de 0,47 episodio/paciente/año en 2019, a una ratio de 0,32 en 2020 y de 0,16 en 2021, disminuyendo el porcentaje de infecciones peritoneales dependientes de catéter de 38% en 2019, al 30% en 2020 y del 8% en 2021. No se han objetivado resistencias a antibióticos, ni otros efectos indeseados.Conclusiones: El uso de mupirocina en nuestra unidad parece ser eficaz en la reducción de las infecciones del orificio de salida, y de las infecciones peritoneales dependientes de catéter. (AU)


ntroduction: Outflow tract infection is an important compli-cation in peritoneal dialysis, mainly because it can progress to tunelitis and/or catheter-dependent peritoneal infection that often leads to catheter removal. Objective: To analyse the mupirocin application in the mana-gement of the healthy exit site and the impact on the infection rate and the percentage of catheter-dependent peritoneal in-fections.Material and Method: Retrospective observational study in a provincial peritoneal dialysis unit. Patients over 18 years of age who underwent peritoneal dialysis at home from 01/01/2019 to 31/12/2021 were included. Variables analysed were: sex, yearly orifice infection rate, responsible germs, catheter-dependent peritoneal infections and appearance of resistance. Data were obtained from the unit’s annual registry.342338Results:Twice-weekly mupirocin application in healthy peritoneal catheter exit site care has decreased the rate of exit site infections from 0.47 episode/patient/year in 2019 to 0.32 in 2020 and 0.16 in 2021, decreasing the percentage of catheter-dependent peritoneal infections from 38% in 2019 to 30% in 2020 and 8% in 2021. No resistance has been observed.Conclusions:Mupirocin application in our unit appears to be effective in reducing exit site infections and catheter-dependent peritoneal infections. (AU)


Subject(s)
Humans , Peritoneal Dialysis , Catheters , Mupirocin , Dialysis , Nephrology Nursing
3.
Oncoimmunology ; 11(1): 2104991, 2022.
Article in English | MEDLINE | ID: mdl-35936986

ABSTRACT

NKG2D is a major natural killer (NK) cell-activating receptor that recognizes eight ligands (NKG2DLs), including MICA, and whose engagement triggers NK cell effector functions. As NKG2DLs are upregulated on tumor cells but tumors can subvert the NKG2D-NKG2DL axis, NKG2DLs constitute attractive targets for antibody (Ab)-based immuno-oncology therapies. However, such approaches require a deep characterization of NKG2DLs and NKG2D cell surface expression on primary tumor and immune cells. Here, using a bioinformatic analysis, we observed that MICA is overexpressed in renal cell carcinoma (RCC), and we also detected an association between the NKG2D-MICA axis and a diminished overall survival of RCC patients. Also, by flow cytometry (FC), we observed that MICA was the only NKG2DL over-expressed on clear cell renal cell carcinoma (ccRCC) tumor cells, including cancer stem cells (CSC) that also coexpressed NKG2D. Moreover, tumor-infiltrating leukocytes (TIL), but not peripheral blood lymphoid cells (PBL) from ccRCC patients, over-expressed MICA, ULBP3 and ULBP4. In addition, NKG2D was downregulated on peripheral blood NK cells (PBNK) from ccRCC patients but upregulated on tumor-infiltrating NK cells (TINK). These TINK exhibited impaired degranulation that negatively correlated with NKG2D expression, diminished IFN-γ production, upregulation of TIM-3, and an impaired glucose intake upon stimulation with cytokines, indicating that they are dysfunctional, display features of exhaustion and an altered metabolic fitness. We conclude that ccRCC patients exhibit a distorted MICA-NKG2D axis, and MICA emerges as the forefront NKG2DL for the development of targeted therapies in ccRCC.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Carcinoma, Renal Cell/therapy , Histocompatibility Antigens Class I/metabolism , Humans , Kidney Neoplasms/therapy , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Receptors, Natural Killer Cell
4.
Int Tinnitus J ; 25(2): 169-171, 2022 Mar 03.
Article in English | MEDLINE | ID: mdl-35239301

ABSTRACT

This systematic review was carried out in order to highlight the implementation of ozone therapy as a treatment for nocturnal tinnitus. It is important to note that ozone treatment has resolved nocturnal tinnitus with 100% efficacy in previous studies, producing in turn a symptomatic improvement in the patient. From these results, a high degree of efficacy can be inferred in overcoming nocturnal tinnitus treated with ozone therapy in patients with a severe index of perception of tinnitus.


Subject(s)
Tinnitus , Humans , Tinnitus/drug therapy , Tinnitus/etiology , Treatment Outcome
5.
Front Cell Dev Biol ; 9: 777086, 2021.
Article in English | MEDLINE | ID: mdl-34869380

ABSTRACT

To acquire fertilization competence, mammalian sperm must undergo several biochemical and physiological modifications known as capacitation. Despite its relevance, the metabolic pathways that regulate the capacitation-related events, including the development of hyperactivated motility, are still poorly described. Previous studies from our group have shown that temporary energy restriction in mouse sperm enhanced hyperactivation, in vitro fertilization, early embryo development and pregnancy rates after embryo transfer, and it improved intracytoplasmic sperm injection results in the bovine model. However, the effects of starvation and energy recovery protocols on human sperm function have not yet been established. In the present work, human sperm were incubated for different periods of time in medium containing glucose, pyruvate and lactate (NUTR) or devoid of nutrients for the starving condition (STRV). Sperm maintained in STRV displayed reduced percentages of motility and kinematic parameters compared to cells incubated in NUTR medium. Moreover, they did not undergo hyperactivation and showed reduced levels of ATP, cAMP and protein tyrosine phosphorylation. Similar to our results with mouse sperm, starvation induced increased intracellular Ca2+ concentrations. Starved human sperm were capable to continue moving for more than 27 h, but the incubation with a mitochondrial uncoupler or inhibitors of oxidative phosphorylation led to a complete motility loss. When exogenous nutrients were added back (sperm energy recovery (SER) treatment), hyperactivated motility was rescued and there was a rise in sperm ATP and cAMP levels in 1 min, with a decrease in intracellular Ca2+ concentration and no changes in sperm protein tyrosine phosphorylation. The finding that human sperm can remain motile for several hours under starvation due to mitochondrial use of endogenous metabolites implies that other metabolic pathways may play a role in sperm energy production. In addition, full recovery of motility and other capacitation parameters of human sperm after SER suggests that this treatment might be used to modulate human sperm fertilizing ability in vitro.

6.
Front Immunol ; 12: 745939, 2021.
Article in English | MEDLINE | ID: mdl-34616407

ABSTRACT

Natural Killer (NK) cells play a key role in cancer immunosurveillance. However, NK cells from cancer patients display an altered phenotype and impaired effector functions. In addition, evidence of a regulatory role for NK cells is emerging in diverse models of viral infection, transplantation, and autoimmunity. Here, we analyzed clear cell renal cell carcinoma (ccRCC) datasets from The Cancer Genome Atlas (TCGA) and observed that a higher expression of NK cell signature genes is associated with reduced survival. Analysis of fresh tumor samples from ccRCC patients unraveled the presence of a high frequency of tumor-infiltrating PD-L1+ NK cells, suggesting that these NK cells might exhibit immunoregulatory functions. In vitro, PD-L1 expression was induced on NK cells from healthy donors (HD) upon direct tumor cell recognition through NKG2D and was further up-regulated by monocyte-derived IL-18. Moreover, in vitro generated PD-L1hi NK cells displayed an activated phenotype and enhanced effector functions compared to PD-L1- NK cells, but simultaneously, they directly inhibited CD8+ T cell proliferation in a PD-L1-dependent manner. Our results suggest that tumors might drive the development of PD-L1-expressing NK cells that acquire immunoregulatory functions in humans. Hence, rational manipulation of these regulatory cells emerges as a possibility that may lead to improved anti-tumor immunity in cancer patients.


Subject(s)
B7-H1 Antigen/biosynthesis , CD8-Positive T-Lymphocytes/cytology , Carcinoma, Renal Cell/immunology , Kidney Neoplasms/immunology , Killer Cells, Natural/metabolism , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/mortality , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Datasets as Topic , Disease-Free Survival , Gene Expression , Humans , Interferon-gamma/pharmacology , Interleukin-18/pharmacology , K562 Cells , Kaplan-Meier Estimate , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/mortality , Monitoring, Immunologic , Monocytes/metabolism , Recombinant Proteins/pharmacology , Up-Regulation
7.
Front Immunol ; 12: 681615, 2021.
Article in English | MEDLINE | ID: mdl-34149719

ABSTRACT

Although natural killer (NK) cells infiltrate clear cell renal cell carcinomas (ccRCC), the most frequent malignancy of the kidney, tumor progression suggests that they become dysfunctional. As ccRCC-driven subversion of NK cell effector functions is usually accompanied by phenotypic changes, analysis of such alterations might lead to the identification of novel biomarkers and/or targets in immuno-oncology. Consequently, we performed a phenotypic analysis of peripheral blood NK cells (PBNK) and tumor-infiltrating NK cells (TINK) from ccRCC patients. Compared to HD, PBNK from ccRCC patients exhibited features of activated cells as shown by CD25, CD69 and CD62L expression. They also displayed increased expression of DNAM-1, CD48, CD45, MHC-I, reduced expression of NKG2D, and higher frequencies of CD85j+ and PD-1+ cells. In addition, compared to PBNK from ccRCC patients, TINK exhibited higher expression of activation markers, tissue residency features and decreased expression of the activating receptors DNAM-1, NKp30, NKp46, NKp80 and CD16, suggesting a more inhibitory phenotype. Analysis of The Cancer Genome Atlas (TCGA) revealed that CD48, CD45, CD85j and PD-1 are significantly overexpressed in ccRCC and that their expression is associated with an NK cell infiltration signature. Calculation of z-scores revealed that their expression on PBNK, alone or combined, distinguished ccRCC patients from HD. Therefore, these molecules emerge as novel potential biomarkers and our results suggest that they might constitute possible targets for immunotherapy in ccRCC patients.


Subject(s)
Carcinoma, Renal Cell/etiology , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/etiology , Kidney Neoplasms/metabolism , Killer Cells, Natural/immunology , Lymphocyte Count , Lymphocytes, Tumor-Infiltrating/immunology , Aged , Biomarkers , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/therapy , Disease Management , Disease Susceptibility , Female , Gene Expression , Humans , Immunophenotyping , Kidney Neoplasms/pathology , Kidney Neoplasms/therapy , Killer Cells, Natural/metabolism , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Male , Middle Aged , Neoplasm Staging , Nephrectomy
8.
FASEB J ; 35(6): e21478, 2021 06.
Article in English | MEDLINE | ID: mdl-33991146

ABSTRACT

Sperm capacitation is essential to gain fertilizing capacity. During this process, a series of biochemical and physiological modifications occur that allow sperm to undergo acrosomal exocytosis (AE). At the molecular level, hyperpolarization of the sperm membrane potential (Em) takes place during capacitation. This study shows that human sperm incubated under conditions that do not support capacitation (NC) can become ready for an agonist stimulated AE by pharmacologically inducing Em hyperpolarization with Valinomycin or Amiloride. To investigate how Em hyperpolarization promotes human sperm's ability to undergo AE, live single-cell imaging experiments were performed to simultaneously monitor changes in [Ca2+ ]i and the occurrence of AE. Em hyperpolarization turned [Ca2+ ]i dynamics in NC sperm from spontaneously oscillating into a sustained slow [Ca2+ ]i increase. The addition of progesterone (P4) or K+ to Valinomycin-treated sperm promoted that a significant number of cells displayed a transitory rise in [Ca2+ ]i which then underwent AE. Altogether, our results demonstrate that Em hyperpolarization is necessary and sufficient to prepare human sperm for the AE. Furthermore, this Em change decreased Ca2+ oscillations that block the occurrence of AE, providing strong experimental evidence of the molecular mechanism that drives the acquisition of acrosomal responsiveness.


Subject(s)
Acrosome Reaction , Calcium Signaling , Exocytosis , Membrane Potentials , Sperm Capacitation , Spermatozoa/physiology , Humans , Male , Phosphorylation
9.
Obesity (Silver Spring) ; 28(10): 1974-1983, 2020 10.
Article in English | MEDLINE | ID: mdl-32808737

ABSTRACT

OBJECTIVE: Psychopathology in bariatric surgery patients may contribute to adverse postoperative sequelae, including weight regain, substance use, and self-harm. This cross-sectional study aimed to advance the understanding of the risk and protective paths through which weight bias associates with depressive and anxiety symptoms in bariatric surgery candidates (BSC). METHODS: BSC recruited from a surgical clinic (N = 213, 82.2% women, 43 [SD 12] years, mean BMI: 49 [SD 9] kg/m2 ) completed measures of experienced weight bias (EWB), internalized weight bias (IWB), body and internalized shame, and self-compassion; anxiety and depression screeners were accessed from medical charts. Multiple regression and PROCESS bootstrapping estimates tested our hypothesized mediation model as follows: EWB→IWB→body shame→shame→self-compassion→symptoms. RESULTS: After accounting for EWB and IWB, internalized shame accounted for greater variance in both end points than body shame. EWB was associated with greater anxiety through risk paths implicating IWB, body shame, and/or internalized shame. Protective paths associated EWB with fewer depressive and anxiety symptoms among those with higher self-compassion. CONCLUSIONS: The findings suggest a potentially important role for weight bias and shame in psychological health among BSC and implicate self-compassion, a trainable affect-regulation strategy, as a protective factor that may confer some resiliency. Future research using longitudinal and causal designs is warranted.


Subject(s)
Anxiety/psychology , Bariatric Surgery/psychology , Body Weight/genetics , Depression/psychology , Empathy/physiology , Psychopathology/methods , Adult , Bias , Cross-Sectional Studies , Female , Humans , Male , Risk Factors , Shame
10.
J Immunother Cancer ; 8(1)2020 06.
Article in English | MEDLINE | ID: mdl-32518090

ABSTRACT

BACKGROUND: Natural killer and cytotoxic CD8+ T cells are major players during antitumor immunity. They express NKG2D, an activating receptor that promotes tumor elimination through recognition of the MHC class I chain-related proteins A and B (MICA and MICB). Both molecules are overexpressed on a great variety of tumors from different tissues, making them attractive targets for immunotherapy. However, tumors shed MICA and MICB, and the soluble forms of both (sMICA and sMICB) mediate tumor-immune escape. Some reports indicate that anti-MICA antibodies (Ab) can promote the restoration of antitumor immunity through the induction of direct antitumor effects (antibody-dependent cell-mediated cytotoxicity, ADCC) and scavenging of sMICA. Therefore, we reasoned that an active induction of anti-MICA Ab with an immunogenic protein might represent a novel therapeutic and prophylactic alternative to restore antitumor immunity. METHODS: We generated a highly immunogenic chimeric protein (BLS-MICA) consisting of human MICA fused to the lumazine synthase from Brucella spp (BLS) and used it to generate anti-MICA polyclonal Ab (pAb) and to investigate if these anti-MICA Ab can reinstate antitumor immunity in mice using two different mouse tumors engineered to express MICA. We also explored the underlying mechanisms of this expected therapeutic effect. RESULTS: Immunization with BLS-MICA and administration of anti-MICA pAb elicited by BLS-MICA significantly delayed the growth of MICA-expressing mouse tumors but not of control tumors. The therapeutic effect of immunization with BLS-MICA included scavenging of sMICA and the anti-MICA Ab-mediated ADCC, promoting heightened intratumoral M1/proinflammatory macrophage and antigen-experienced CD8+ T cell recruitment. CONCLUSIONS: Immunization with the chimeric protein BLS-MICA constitutes a useful way to actively induce therapeutic anti-MICA pAb that resulted in a reprogramming of the antitumor immune response towards an antitumoral/proinflammatory phenotype. Hence, the BLS-MICA chimeric protein constitutes a novel antitumor vaccine of potential application in patients with MICA-expressing tumors.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibody-Dependent Cell Cytotoxicity/immunology , Histocompatibility Antigens Class I/immunology , Lymphoma/immunology , Recombinant Fusion Proteins/immunology , Urinary Bladder Neoplasms/immunology , Animals , Brucella/enzymology , Female , Histocompatibility Antigens Class I/genetics , Lymphoma/pathology , Lymphoma/therapy , Male , Mice , Mice, Inbred C57BL , Multienzyme Complexes/genetics , Multienzyme Complexes/immunology , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/immunology , Tumor Cells, Cultured , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/therapy
11.
Sensors (Basel) ; 20(5)2020 Feb 28.
Article in English | MEDLINE | ID: mdl-32121271

ABSTRACT

Cancer represents one of the conditions with the most causes of death worldwide. Common methods for its diagnosis are based on tissue biopsies-the extraction of tissue from the primary tumor, which is used for its histological analysis. However, this technique represents a risk for the patient, along with being expensive and time-consuming and so it cannot be frequently used to follow the progress of the disease. Liquid biopsy is a new cancer diagnostic alternative, which allows the analysis of the molecular information of the solid tumors via a body fluid draw. This fluid-based diagnostic method displays relevant advantages, including its minimal invasiveness, lower risk, use as often as required, it can be analyzed with the use of microfluidic-based platforms with low consumption of reagent, and it does not require specialized personnel and expensive equipment for the diagnosis. In recent years, the integration of sensors in microfluidics lab-on-a-chip devices was performed for liquid biopsies applications, granting significant advantages in the separation and detection of circulating tumor nucleic acids (ctNAs), circulating tumor cells (CTCs) and exosomes. The improvements in isolation and detection technologies offer increasingly sensitive and selective equipment's, and the integration in microfluidic devices provides a better characterization and analysis of these biomarkers. These fully integrated systems will facilitate the generation of fully automatized platforms at low-cost for compact cancer diagnosis systems at an early stage and for the prediction and prognosis of cancer treatment through the biomarkers for personalized tumor analysis.


Subject(s)
Early Detection of Cancer/methods , Liquid Biopsy/methods , Microfluidics/methods , Biomarkers, Tumor/blood , Cell-Free Nucleic Acids/blood , Exosomes/pathology , Humans , Lab-On-A-Chip Devices , Neoplastic Cells, Circulating/pathology
12.
Methods Mol Biol ; 2097: 125-136, 2020.
Article in English | MEDLINE | ID: mdl-31776924

ABSTRACT

Natural killer (NK) cells can kill virus-infected cells and tumor cells without prior sensitization and secrete numerous cytokines and chemokines that modulate the activity of different cells of the immune system. The recognition of target cells is mediated by germ line-encoded receptors, and the activity of NK cells can be further regulated by soluble factors such as cytokines and Toll-like receptor ligands. Thus, NK cells display an exciting potential as a powerful immunotherapeutic tool against malignant diseases, and different strategies are being tested aiming to overcome tumor-induced NK cell suppression and restore NK-cell mediated antitumor activity. This section describes different flow cytometry-based protocols to study NK cell effector functions, which can be used to evaluate the immunomodulatory ability of different therapeutic compounds.


Subject(s)
Immunomodulation/immunology , Killer Cells, Natural/immunology , Cell Degranulation , Cell Membrane/metabolism , Cell Separation , Humans , Killer Cells, Natural/physiology , Phenotype , Staining and Labeling
13.
J Immunol ; 200(3): 1008-1015, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29282306

ABSTRACT

NK cells play important roles during immunosurveillance against tumors and viruses as they trigger cytotoxicity against susceptible cells and secrete proinflammatory cytokines such as IFN-γ. In addition, upon activation, macrophages can become proinflammatory (M1) or anti-inflammatory (M2) cells. Although the consequences of the cross-talk between M1 and NK cells are known, the outcome of the cross-talk between M2 and NK cells remains ill-defined. Therefore, in the current work, we investigated the outcome and the underlying mechanisms of the interaction between resting or stimulated human NK cells with M1 or M2. We observed a lower percentage of activated NK cells that produced less IFN-γ upon coculture with M2. Also, CD56dim NK cells cocultured with M2 displayed lower degranulation and cytotoxic activity than NK cells cocultured with M1. Soluble TGF-ß and M2-driven upregulation of CD85j (ILT-2) on NK cells accounted for the diminished IFN-γ production by CD56bright NK cells, whereas M2-driven upregulation of CD85j on NK cells accounted for the generation of hyporesponsive CD56dim NK cells with limited degranulation and cytotoxic capacity. Accordingly, M2 expressed higher amounts of HLA-G, the main ligand for CD85j, than M1. Hyporesponsiveness to degranulation in NK cells was not restored at least for several hours upon removal of M2. Therefore, alternatively activated macrophages restrain NK cell activation and effector functions through different mechanisms, leading to NK cells that display diminished IFN-γ production and at least a transiently impaired degranulation ability. These results unravel an inhibitory circuit of possible relevance in pathological situations.


Subject(s)
Cell Communication/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Macrophage Activation/immunology , Transforming Growth Factor beta/metabolism , Antigens, CD/immunology , CD56 Antigen/metabolism , Cells, Cultured , Coculture Techniques , HLA-G Antigens/metabolism , Humans , Interferon-gamma/immunology , Leukocyte Immunoglobulin-like Receptor B1/immunology , Macrophages/immunology , Transforming Growth Factor beta/immunology
14.
Front Immunol ; 8: 1959, 2017.
Article in English | MEDLINE | ID: mdl-29403472

ABSTRACT

Interleukin (IL)-23 is a member of the IL-12 family of cytokines that, as the other members of this family, is secreted by monocytes, macrophages, and dendritic cells (DC) upon recognition of bacterial, viral, and fungal components. IL-23 is critical during immunity against acute infections, and it is also involved in the development of autoimmune diseases. Although immunoregulatory effects of IL-23 on mouse natural killer (NK) cells have been described, the effect of IL-23 on human NK cells remains ill-defined. In this study, we observed that monocytes stimulated with LPS secreted IL-23 and that blockade of this cytokine during monocyte and NK cell coculture led to a diminished production of IFN-γ by NK cells. Accordingly, rIL-23-induced NK cell activation and stimulated IFN-γ production by CD56bright NK cells. This effect involved MEK1/MEK2, JNK, PI3K, mammalian target of rapamycin, and NF-κB, but not STAT-1, STAT-3, nor p38 MAPK pathways. Moreover, while NK cell-mediated cytotoxicity remained unaltered, antibody-dependent cellular cytotoxicity (ADCC) was enhanced after IL-23 stimulation. In addition, IL-23 displayed a synergistic effect with IL-18 for IFN-γ production by both CD56bright and CD56dim NK cells, and this effect was due to a priming effect of IL-23 for IL-18 responsiveness. Furthermore, NK cells pre-stimulated with IL-18 promoted an increase in CD86 expression and IL-12 secretion by DC treated with LPS, and IL-23 potentiated these effects. Moreover, IL-23-driven enhancement of NK cell "helper" function was dependent on NK cell-derived IFN-γ. Therefore, our results suggest that IL-23 may trigger NK cell-mediated "helper" effects on adaptive immunity, shaping T cell responses during different pathological situations through the regulation of DC maturation.

15.
J Immunol ; 197(3): 953-61, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27342842

ABSTRACT

Despite the classical function of NK cells in the elimination of tumor and of virus-infected cells, evidence for a regulatory role for NK cells has been emerging in different models of autoimmunity, transplantation, and viral infections. However, this role has not been fully explored in the context of a growing tumor. In this article, we show that NK cells can limit spontaneous cross-priming of tumor Ag-specific CD8(+) T cells, leading to reduced memory responses. After challenge with MC57 cells transduced to express the model Ag SIY (MC57.SIY), NK cell-depleted mice exhibited a significantly higher frequency of SIY-specific CD8(+) T cells, with enhanced IFN-γ production and cytotoxic capability. Depletion of NK cells resulted in a CD8(+) T cell population skewed toward an effector memory T phenotype that was associated with enhanced recall responses and delayed tumor growth after a secondary tumor challenge with B16.SIY cells. Dendritic cells (DCs) from NK cell-depleted tumor-bearing mice exhibited a more mature phenotype. Interestingly, tumor-infiltrating and tumor-draining lymph node NK cells displayed an upregulated expression of the inhibitory molecule programmed death ligand 1 that, through interaction with programmed death-1 expressed on DCs, limited DC activation, explaining their reduced ability to induce tumor-specific CD8(+) T cell priming. Our results suggest that NK cells can, in certain contexts, have an inhibitory effect on antitumor immunity, a finding with implications for immunotherapy in the clinic.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cross-Priming/immunology , Dendritic Cells/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Neoplasms, Experimental/immunology , Animals , B7-H1 Antigen/immunology , Cell Line, Tumor , Cell Separation , Flow Cytometry , Humans , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/immunology , Real-Time Polymerase Chain Reaction , Signal Transduction/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...