Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21
1.
Immunology ; 171(2): 198-211, 2024 Feb.
Article En | MEDLINE | ID: mdl-37884280

Glioblastoma, isocitrate dehydrogenase-wildtype (GB), is the most common and aggressive primary brain malignancy with poor outcome. Immune checkpoint inhibitors (ICIs) have been tested in GB and, despite disappointing results, the identification of a small subgroup of responders underlies the need to improve our understanding of the tumour microenvironment (TME) immunity. This study aimed to determine whether the expression of selected immune checkpoints on tissue-resident memory T cells (Trm) may predict patient outcome. We conducted a single cohort observational study. Tumour samples were collected from 45 patients with histologically confirmed GB (WHO grade 4) and processed to obtain single-cell suspensions. Patients were assessed for the correlation of Trm phenotype with overall survival (OS) or progression-free survival (PFS) using multiparametric flow cytometry and uni/multivariate analyses. Levels of Trm expressing programmed cell death protein 1 (PD1) and T cell immunoglobulin and mucin domain-containing protein 3 (TIM3) were found to be linked to clinical outcome. Low frequency of Trm expressing PD1 or TIM3 or both markers defined subgroups as independent positive prognostic factors for patient survival. On multivariate analysis, low CD8+CD103+PD1+TIM3+ Trm and Karnofsky performance status (KPS) ≥70 were confirmed to be the most predictive independent factors associated with longer OS (hazard ratios-HR [95%CI]: 0.14 [0.04-0.52] p < 0.001, 0.39 [0.16-0.96] p = 0.04, respectively). The CD8+CD103+ Trm subgroups were also age-related predictors for survival in GB.


Glioblastoma , Hepatitis A Virus Cellular Receptor 2 , Humans , Programmed Cell Death 1 Receptor/metabolism , Prognosis , CD8-Positive T-Lymphocytes , Tumor Microenvironment
2.
Heliyon ; 9(7): e17406, 2023 Jul.
Article En | MEDLINE | ID: mdl-37519681

In recent years, the international community has been strongly committed to promoting energy transition due to the various challenges presented by the global energy market. In this context, renewable sources have increasingly assumed a significant role, representing a viable solution to accelerate decarbonization and promote diversification of energy mix. This paper examines Italian energy profile, its status, and future prospects. Policies and incentives on a European and national scale have been analysed to observe the commitment of institutions in promoting energy transition. Policies and incentives on a European and national scale have been analysed to observe the commitment of institutions in promoting energy transition. Projections on future investments (2030) and economic and employment implications have also been provided. The analysis shows that shared investments are essential to achieve climate goals in the short term and that existing plants need to be converted through the use of new, more effective, cost-efficient technologies. Thus, innovation and investments in research and development are key drivers of energy transition, and with proper management, climate change mitigation can be ensured. The analysis provides some useful implications for policymakers who should ensure tax incentives, feed-in tariffs, and clear regulation at the national and regional levels that do not hinder the deployment of renewable energy infrastructure.

3.
Int J Cancer ; 153(5): 1080-1095, 2023 Sep 01.
Article En | MEDLINE | ID: mdl-37293858

BRAFV600 mutations are the most common oncogenic alterations in melanoma cells, supporting proliferation, invasion, metastasis and immune evasion. In patients, these aberrantly activated cellular pathways are inhibited by BRAFi whose potent antitumor effect and therapeutic potential are dampened by the development of resistance. Here, by using primary melanoma cell lines, generated from lymph node lesions of metastatic patients, we show that the combination of two FDA-approved drugs, the histone deacetylate inhibitor (HDCAi) romidepsin and the immunomodulatory agent IFN-α2b, reduces melanoma proliferation, long-term survival and invasiveness and overcomes acquired resistance to the BRAFi vemurafenib (VEM). Targeted resequencing revealed that each VEM-resistant melanoma cell line and the parental counterpart are characterized by a distinctive and similar genetic fingerprint, shaping the differential and specific antitumor modulation of MAPK/AKT pathways by combined drug treatment. By using RNA-sequencing and functional in vitro assays, we further report that romidepsin-IFN-α2b treatment restores epigenetically silenced immune signals, modulates MITF and AXL expression and induces both apoptosis and necroptosis in sensitive and VEM-resistant primary melanoma cells. Moreover, the immunogenic potential of drug-treated VEM-resistant melanoma cells results significantly enhanced, given the increased phagocytosis rate of these cells by dendritic cells, which in turn exhibit also a selective down-modulation of the immune checkpoint TIM-3. Overall, our results provide evidence that combined epigenetic-immune drugs can overcome VEM resistance of primary melanoma cells by oncogenic and immune pathways reprogramming, and pave the way for rapidly exploiting this combination to improve BRAFi-resistant metastatic melanoma treatment, also via reinforcement of immune checkpoint inhibitor therapy.


Interferon Type I , Melanoma , Humans , Vemurafenib/pharmacology , Vemurafenib/therapeutic use , Drug Resistance, Neoplasm , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Cell Line, Tumor
4.
J Enzyme Inhib Med Chem ; 38(1): 2183810, 2023 Dec.
Article En | MEDLINE | ID: mdl-36916299

Helicobacter pylori (HP) is among the most common pathogens causing infection in humans worldwide. Oxidative stress and gastric inflammation are involved in the progression of HP-related gastric diseases, and they can be targeted by integrating conventional antibiotic treatment with polyphenol-enriched natural products. In this work, we characterised three different propolis extracts and evaluated their stability under in vitro simulated gastric digestion, compared to their main constituents alone. The extract with the highest stability to digestion (namely, the dark propolis extract, DPE) showed a minimum bactericidal concentration (MBC) lower than 1 mg/mL on HP strains with different virulence factors. Finally, since urease is one of the virulence factors contributing to the establishment of a microenvironment that promotes HP infection, we evaluated the possible inhibition of this enzyme by using molecular docking simulations and in vitro colorimetric assay, showing that galangin and pinocembrin may be involved in this activity.


Helicobacter pylori , Propolis , Humans , Propolis/pharmacology , Molecular Docking Simulation , Anti-Bacterial Agents/pharmacology , Virulence Factors/pharmacology , Plant Extracts/pharmacology , Digestion
5.
Cytokine Growth Factor Rev ; 58: 66-74, 2021 04.
Article En | MEDLINE | ID: mdl-33071044

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of the ongoing coronavirus disease 2019 (COVID-19) pandemic, induces severe pneumonia mainly in elderly males. Epidemiological data clearly indicate sex-based differences in disease outcomes, with men accounting for about 70 % of deaths, despite similar susceptibility to infection. It is well known that females are endowed with higher capacity to produce antibodies, which correlates with viral clearance and disease resolution in the context of SARS-Cov-2 infection. Many X-linked immune genes escape X inactivation showing biallelic expression in female immune cells, particularly in plasmacytoid dendritic cells (pDCs). PDCs are more active in females and endowed with high capability to induce IFN-α-mediated B cell activation and differentiation into antibody-producing plasma cells throughout epigenetic mechanisms linked to trained immunity. Thus, we hypothesize that following SARS-CoV-2 infection, epigenetic modifications of X-linked genes involved in pDC-mediated type I IFN (IFN-I) signaling occurs more effectively in females, for inducing neutralizing antibody response as an immune correlate driving sex-biased disease outcome.


Antibody Formation , COVID-19/diagnosis , COVID-19/immunology , Interferon Type I/physiology , SARS-CoV-2/immunology , COVID-19/epidemiology , Female , Humans , Male , Pandemics , Prognosis , Sex Characteristics
6.
J Exp Clin Cancer Res ; 38(1): 432, 2019 Oct 28.
Article En | MEDLINE | ID: mdl-31661001

BACKGROUND: Inefficient T-cell access to the tumor microenvironment (TME) is among the causes of tumor immune-resistance. Previous evidence demonstrated that targeting CXCR4 improves anti-PD-1/PD-L1 efficacy reshaping TME. To evaluate the role of newly developed CXCR4 antagonists (PCT/IB2011/000120/ EP2528936B1/US2013/0079292A1) in potentiating anti-PD-1 efficacy two syngeneic murine models, the MC38 colon cancer and the B16 melanoma-human CXCR4-transduced, were employed. METHODS: Mice were subcutaneously injected with MC38 (1 × 106) or B16-hCXCR4 (5 × 105). After two weeks, tumors bearing mice were intraperitoneally (ip) treated with murine anti-PD-1 [RMP1-14] (5 mg/kg, twice week for 2 weeks), Pep R (2 mg/kg, 5 days per week for 2 weeks), or both agents. The TME was evaluated through immunohistochemistry and flow-cytometry. In addition, the effects of the human-anti-PD-1 nivolumab and/or Peptide-R54 (Pep R54), were evaluated on human melanoma PES43 cells and xenografts treated. RESULTS: The combined treatment, Pep R plus anti-PD-1, reduced the MC38 Relative Tumor Volume (RTV) by 2.67 fold (p = 0.038) while nor anti-PD-1, neither Pep R significantly impacted on tumor growth. Significant higher number of Granzyme B (GZMB) positive cells was detected in MC38 tumors from mice treated with the combined treatment (p = 0.016) while anti-PD-1 determined a modest but significant increase of tumor-infiltrating GZMB positive cells (p = 0.035). Also, a lower number of FoxP3 positive cells was detected (p = 0.022). In the B16-hCXCR4 tumors, two weeks of combined treatment reduced tumor volume by 2.27 fold while nor anti-PD-1 neither Pep R significantly impacted on tumor growth. A significant higher number of GRZB positive cells was observed in B16-hCXCR4 tumors treated with combined treatment (p = 0,0015) as compared to anti-PD-1 (p = 0.028). The combined treatment reduced CXCR4, CXCL12 and PD-L1 expression in MC38 tumors. In addition, flow cytometry on fresh B16-hCXCR4 tumors showed significantly higher Tregs number following anti-PD-1 partially reversed by the combined treatment Pep R and anti-PD-1. Combined treatment determined an increase of CD8/Tregs and CD8/MDSC ratio. To dissect the effect of anti-PD-1 and CXCR4 targeting on PD-1 expressed by human cancer cells, PES43 human melanoma xenograft model was employed. In vitro human anti-PD-1 nivolumab or pembrolizumab (10 µM) reduced PES43 cells growth while nivolumab (10 µM) inhibited pERK1/2, P38 MAPK, pAKT and p4EBP. PES43 xenograft mice were treated with Pep R54, a newly developed Pep R derivative (AcHN-Arg-Ala-[DCys-Arg- Nal(2')-His-Pen]- COOH), plus nivolumab. After 3 weeks of combined treatment a significant reduction in tumor growth was shown (p = 0.038). PES43 lung disseminated tumor cells (DTC) were detected in fresh lung tissues as melanoma positive MCSP-APC+ cells. Although not statistically significant, DTC-PES43 cells were reduced in mice lungs treated with combined treatment while nivolumab or Pep R54 did not affect DTC number. CONCLUSION: Combined treatment with the new developed CXCR4 antagonist, Pep R, plus anti-PD-1, reduced tumor-growth in two syngeneic murine models, anti-PD-1 sensitive and resistant, potentiating Granzyme and reducing Foxp3 cells infiltration. In addition, the human specific CXCR4 antagonist, Pep R54, cooperated with nivolumab in inhibiting the growth of the PD-1 expressing human PES43 melanoma xenograft. This evidence sheds light on PD-1 targeting mechanisms and paves the way for CXCR4/PD-1 targeting combination therapy.


Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptors, CXCR4/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Female , Humans , Mice , Tumor Microenvironment
7.
PLoS One ; 13(1): e0189477, 2018.
Article En | MEDLINE | ID: mdl-29320502

Individuals exposed to Mycobacterium tuberculosis (Mtb) may be infected and remain for the entire life in this condition defined as latent tuberculosis infection (LTBI) or develop active tuberculosis (TB). Among the multiple factors governing the outcome of the infection, dendritic cells (DCs) play a major role in dictating antibacterial immunity. However, current knowledge on the role of the diverse components of human DCs in shaping specific T-cell response during Mtb infection is limited. In this study, we performed a comparative evaluation of peripheral blood circulating DC subsets as well as of monocyte-derived Interferon-α DCs (IFN-DCs) from patients with active TB, subjects with LTBI and healthy donors (HD). The proportion of circulating myeloid BDCA3+ DCs (mDC2) and plasmacytoid CD123+ DCs (pDCs) declined significantly in active TB patients compared to HD, whereas the same subsets displayed a remarkable activation in LTBI subjects. Simultaneously, the differentiation of IFN-DCs from active TB patients resulted profoundly impaired compared to those from LTBI and HD individuals. Importantly, the altered developmental trait of IFN-DCs from active TB patients was associated with down-modulation of IFN-linked genes, marked changes in molecular signaling conveying antigen (Ag) presentation and full inability to induce Ag-specific T cell response. Thus, these data reveal an important role of IFN-α in determining the induction of Mtb-specific immunity.


Dendritic Cells/immunology , Interferon-alpha/immunology , Latent Tuberculosis/pathology , Adult , Antigens, CD/immunology , Down-Regulation , Female , Humans , Latent Tuberculosis/immunology , Male , Middle Aged , Mycobacterium tuberculosis/immunology
8.
Cancer Immunol Res ; 5(7): 604-616, 2017 07.
Article En | MEDLINE | ID: mdl-28615266

Colorectal cancer results from the progressive accumulation of genetic and epigenetic alterations. IFN signaling defects play an important role in the carcinogenesis process, in which the inability of IFN transcription regulatory factors (IRF) to access regulatory sequences in IFN-stimulated genes (ISG) in tumors and in immune cells may be pivotal. We reported that low-dose combination of two FDA-approved epidrugs, azacytidine (A) and romidepsin (R), with IFNα2 (ARI) hampers the aggressiveness of both colorectal cancer metastatic and stem cells in vivo and triggers immunogenic cell death signals that stimulate dendritic cell (DC) function. Here, we investigated the molecular signals induced by ARI treatment and found that this drug combination increased the accessibility to regulatory sequences of ISGs and IRFs that were epigenetically silenced in both colorectal cancer cells and DCs. Likewise, specific ARI-induced histone methylation and acetylation changes marked epigenetically affected ISG promoters in both metastatic cancer cells and DCs. Analysis by ChIP-seq confirmed such ARI-induced epigenetically regulated IFN signature. The activation of this signal endowed DCs with a marked migratory capability. Our results establish a direct correlation between reexpression of silenced ISGs by epigenetic control and ARI anticancer activity and provide new knowledge for the development of innovative combined therapeutic strategies for colorectal cancer. Cancer Immunol Res; 5(7); 604-16. ©2017 AACR.


Carcinogenesis/drug effects , Colorectal Neoplasms/drug therapy , Dendritic Cells/drug effects , Interferon-alpha/immunology , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/immunology , Azacitidine/administration & dosage , Carcinogenesis/immunology , Cell Line, Tumor , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Dendritic Cells/immunology , Depsipeptides/administration & dosage , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factors/genetics , Interferon-alpha/genetics , Interferon-gamma/genetics , Nitric Oxide Synthase Type II/genetics , Receptors, Cytokine/genetics , Receptors, Interferon , Signal Transduction/drug effects
9.
Sci Rep ; 7(1): 1093, 2017 04 24.
Article En | MEDLINE | ID: mdl-28439087

Immunotherapy efficacy relies on the crosstalk within the tumor microenvironment between cancer and dendritic cells (DCs) resulting in the induction of a potent and effective antitumor response. DCs have the specific role of recognizing cancer cells, taking up tumor antigens (Ags) and then migrating to lymph nodes for Ag (cross)-presentation to naïve T cells. Interferon-α-conditioned DCs (IFN-DCs) exhibit marked phagocytic activity and the special ability of inducing Ag-specific T-cell response. Here, we have developed a novel microfluidic platform recreating tightly interconnected cancer and immune systems with specific 3D environmental properties, for tracking human DC behaviour toward tumor cells. By combining our microfluidic platform with advanced microscopy and a revised cell tracking analysis algorithm, it was possible to evaluate the guided efficient motion of IFN-DCs toward drug-treated cancer cells and the succeeding phagocytosis events. Overall, this platform allowed the dissection of IFN-DC-cancer cell interactions within 3D tumor spaces, with the discovery of major underlying factors such as CXCR4 involvement and underscored its potential as an innovative tool to assess the efficacy of immunotherapeutic approaches.


Cell Tracking/methods , Colonic Neoplasms/therapy , Dendritic Cells/immunology , Immunotherapy/methods , Microfluidics/methods , Models, Biological , Cells, Cultured , Humans , Immunologic Factors/metabolism , Interferon-alpha/metabolism , Microscopy/methods , Treatment Outcome
10.
Oncotarget ; 7(18): 26361-73, 2016 May 03.
Article En | MEDLINE | ID: mdl-27028869

Epigenetic alterations, including dysregulated DNA methylation and histone modifications, govern the progression of colorectal cancer (CRC). Cancer cells exploit epigenetic regulation to control cellular pathways, including apoptotic and metastatic signals. Since aberrations in epigenome can be pharmacologically reversed by DNA methyltransferase and histone deacetylase inhibitors, epigenetics in combination with standard agents are currently envisaged as a new therapeutic frontier in cancer, expected to overcome drug resistance associated with current treatments. In this study, we challenged this idea and demonstrated that the combination of azacitidine and romidepsin with IFN-α owns a high therapeutic potential, targeting the most aggressive cellular components of CRC, such as metastatic cells and cancer stem cells (CSCs), via tight control of key survival and death pathways. Moreover, the antitumor efficacy of this novel pharmacological approach is associated with induction of signals of immunogenic cell death. Of note, a previously undisclosed key role of IFN-α in inducing both antiproliferative and pro-apoptotic effects on CSCs of CRC was also found. Overall, these findings open a new frontier on the suitability of IFN-α in association with epigenetics as a novel and promising therapeutic approach for CRC management.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colorectal Neoplasms/pathology , Interferon-alpha/pharmacology , Neoplastic Stem Cells/drug effects , Animals , Apoptosis/drug effects , Azacitidine/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Depsipeptides/pharmacology , Epigenesis, Genetic/drug effects , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/pathology , Xenograft Model Antitumor Assays
11.
PLoS One ; 8(8): e72833, 2013.
Article En | MEDLINE | ID: mdl-23977359

Type I interferon (IFN-I) have emerged as crucial mediators of cellular signals controlling DC differentiation and function. Human DC differentiated from monocytes in the presence of IFN-α (IFN-α DC) show a partially mature phenotype and a special capability of stimulating CD4+ T cell and cross-priming CD8+ T cells. Likewise, plasmacytoid DC (pDC) are blood DC highly specialized in the production of IFN-α in response to viruses and other danger signals, whose functional features may be shaped by IFN-I. Here, we investigated the molecular mechanisms stimulated by IFN-α in driving human monocyte-derived DC differentiation and performed parallel studies on peripheral unstimulated and IFN-α-treated pDC. A specific miRNA signature was induced in IFN-α DC and selected miRNAs, among which miR-23a and miR-125b, proved to be negatively associated with up-modulation of Blimp-1 occurring during IFN-α-driven DC differentiation. Of note, monocyte-derived IFN-α DC and in vitro IFN-α-treated pDC shared a restricted pattern of miRNAs regulating Blimp-1 expression as well as some similar phenotypic, molecular and functional hallmarks, supporting the existence of a potential relationship between these DC populations. On the whole, these data uncover a new role of Blimp-1 in human DC differentiation driven by IFN-α and identify Blimp-1 as an IFN-α-mediated key regulator potentially accounting for shared functional features between IFN-α DC and pDC.


Dendritic Cells/cytology , Dendritic Cells/metabolism , Interferon-alpha/pharmacology , MicroRNAs/genetics , Monocytes/cytology , Repressor Proteins/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Dendritic Cells/drug effects , Gene Expression Profiling , HeLa Cells , Humans , Phenotype , Positive Regulatory Domain I-Binding Factor 1 , Repressor Proteins/metabolism
12.
Neoplasia ; 14(9): 855-67, 2012 Sep.
Article En | MEDLINE | ID: mdl-23019417

Currently approved combination regimens available for the treatment of metastatic tumors, such as breast cancer, have been shown to increase response rates, often at the cost of a substantial increase in toxicity. An ideal combination strategy may consist of agents with different mechanisms of action leading to complementary antitumor activities and safety profiles. In the present study, we investigated the effects of the epigenetic modulator apicidin in combination with the cytotoxic agent docetaxel in tumor breast cell lines characterized by different grades of invasiveness. We report that combined treatment of apicidin and docetaxel, at low toxicity doses, stimulates in metastatic breast cancer cells the expression of CTCF-like protein and other cancer antigens, thus potentially favoring an antitumor immune response. In addition, apicidin and docetaxel co-treatment specifically stimulates apoptosis, characterized by an increased Bax/Bcl-2 ratio and caspase-8 activation. Importantly, following combined exposure to these agents, metastatic cells were also found to induce signals of immunogenic apoptosis such as cell surface expression of calreticulin and release of considerable amounts of high-mobility group box 1 protein, thus potentially promoting the translation of induced cell death into antitumor immune response. Altogether, our results indicate that the combined use of apicidin and docetaxel, at a low toxicity profile, may represent a potential innovative strategy able to activate complementary antitumor pathways in metastatic breast cancer cells, associated with a potential control of metastatic growth and possible induction of antitumor immunity.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/genetics , DNA-Binding Proteins/genetics , HMGB1 Protein/genetics , Peptides, Cyclic/pharmacology , Taxoids/pharmacology , Apoptosis/drug effects , Apoptosis/immunology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , DNA-Binding Proteins/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Docetaxel , Drug Synergism , Female , Gene Expression Regulation, Neoplastic/drug effects , HMGB1 Protein/immunology , Humans , Immunomodulation/drug effects , Neoplasm Metastasis , Peptides, Cyclic/administration & dosage , Taxoids/administration & dosage
13.
J Leukoc Biol ; 88(5): 947-54, 2010 Nov.
Article En | MEDLINE | ID: mdl-20807707

The interaction of PAMPs with cells of the innate immune system shapes the adaptive host response. Here, we report that ß-glucan, a major fungal PAMP purified from Candida albicans, stimulates human DCs to secrete a pro-Th17 cytokine pattern. Notably, ß-glucan induces PGE2 production, which has been shown to play a pivotal role in Th17 cell expansion. Inhibition of PGE2 synthesis or blockade of PGE2 receptors EP2 and EP4 drastically reduces IL-23 production by ß-glucan-activated DCs, suggesting that endogenous PGE2 amplifies IL-23 synthesis in response to the C. albicans PAMP. Moreover ß-glucan promotes the expansion of Th17 cells, which is strongly decreased by EP2 and EP4 receptor blockade on DCs. Our results highlight a novel role for PGE2 in the regulation of innate and adaptive immune response triggered by recognition of a prominent, highly conserved fungal PAMP such as ß-glucan.


Dendritic Cells/immunology , Dinoprostone/physiology , beta-Glucans/pharmacology , Amino Acids/pharmacology , Amphotericin B/pharmacology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Candida albicans/physiology , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dinoprostone/immunology , Dinoprostone/pharmacology , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Immunity, Innate , Interleukin-23/drug effects , Interleukin-23/genetics , Pyruvates/pharmacology
14.
Blood ; 116(22): 4492-500, 2010 Nov 25.
Article En | MEDLINE | ID: mdl-20668227

Endothelial cells (ECs) represent a major source of actively secreted adenosine triphosphate (ATP). Natural killer (NK) cells can mediate vascular injury in several pathologic conditions, including cytomegalovirus infection and vascular leak syndrome. We studied NK-cell expression of P2 receptors and the role of these nucleotide receptors in the regulation of endothelial-NK cell cross-talk. NK cells from healthy subjects expressed P2Y(1,2,4,6,11,12,13,14) and P2X(1,4,5,6,7) receptors. NK cells stimulated with ATP, but not uridine triphosphate, increased intracellular Ca²(+) and chemokinesis. Moreover, ATP, but not uridine triphosphate, inhibited NK chemotaxis in response to CX3CL1, whereas chemotaxis to CXCL12 was increased. CX3CL1 elicited killing of human umbilical vein ECs and human coronary artery ECs by NK cells. However, in the presence of ATP, CX3CL1 failed to stimulate killing of ECs. Such inhibitory effect was lost on exogenous addition of the ATP-hydrolyzing enzyme apyrase or by pharmacologic inhibition of the P2Y11R, and correlated with increased intracellular cyclic adenosine monophosphate concentrations induced by ATP or other P2Y11R agonists, including NAD(+). Extracellular ATP regulates NK-cell cytotoxicity via P2Y11R activation, protecting ECs from CX3CL1-elicited NK cell-mediated killing. These findings point out the P2Y11R as a potential target for pharmacologic intervention aimed at reducing NK-mediated vascular injury.


Adenosine Triphosphate/immunology , Chemokine CX3CL1/immunology , Chemotaxis , Endothelial Cells/immunology , Killer Cells, Natural/immunology , Receptors, Purinergic P2/immunology , Calcium/immunology , Cell Line , Cyclic AMP/immunology , Endothelial Cells/cytology , Gene Expression , Humans , Killer Cells, Natural/cytology , RNA, Messenger/genetics , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X/genetics , Receptors, Purinergic P2X/immunology , Receptors, Purinergic P2Y/genetics , Receptors, Purinergic P2Y/immunology
15.
Blood ; 115(8): 1554-63, 2010 Feb 25.
Article En | MEDLINE | ID: mdl-20009034

The identification of molecules responsible for apoptotic cell (AC) uptake by dendritic cells (DCs) and induction of T-cell immunity against AC-associated antigens is a challenge in immunology. DCs differentiated in the presence of interferon-alpha (IFN-alpha-conditioned DCs) exhibit a marked phagocytic activity and a special attitude in inducing CD8(+) T-cell response. In this study, we found marked overexpression of the scavenger receptor oxidized low-density lipoprotein receptor 1 (LOX-1) in IFN-alpha-conditioned DCs, which was associated with increased levels of genes belonging to immune response families and high competence in inducing T-cell immunity against antigens derived from allogeneic apoptotic lymphocytes. In particular, the capture of ACs by IFN-alpha DCs led to a substantial subcellular rearrangement of major histocompatibility complex class I and class II molecules, along with enhanced cross-priming of autologous CD8(+) T cells and CD4(+) T-cell activation. Remarkably, AC uptake, CD8(+) T-cell cross-priming, and, to a lesser extent, priming of CD4(+) T lymphocytes were inhibited by a neutralizing antibody to the scavenger receptor LOX-1 protein. These results unravel a novel LOX-1-dependent pathway by which IFN-alpha can, under both physiologic and pathologic conditions, render DCs fully competent for presenting AC-associated antigens for cross-priming CD8(+) effector T cells, concomitantly with CD4(+) T helper cell activation.


Antigen Presentation/immunology , Apoptosis/drug effects , Dendritic Cells/immunology , Immunologic Factors/pharmacology , Interferon-alpha/pharmacology , Scavenger Receptors, Class E/immunology , Signal Transduction/drug effects , Apoptosis/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Humans , Immunity, Cellular/drug effects , Immunologic Factors/immunology , Interferon-alpha/immunology , Lymphocyte Activation/drug effects , Signal Transduction/immunology
16.
J Leukoc Biol ; 82(5): 1136-42, 2007 Nov.
Article En | MEDLINE | ID: mdl-17656653

The functional consequences of treating human monocytes with purified and chemically characterized Candida albicans beta-glucan -- a major microbial pathogen associated molecular pattern -- on their differentiation into dendritic cells (DC) were investigated. We show here that beta-glucan-treated monocytes differentiated into mature DC (Glu-MoDC) with altered phenotype and functional behavior, similarly to DC derived from C. albicans germ-tubes-infected monocytes (Gt-MoDC). They failed to express CD1a and to up-regulate CD80 and DR molecules. Moreover, they produced IL-10 but not IL-12 and primed naive T cells without inducing their functional polarization into effector cells. Since C. albicans beta-glucan is a mixture of both beta-(1,3) and beta-(1,6) glucan, we investigated their relative contribution by the use of non-Candida beta-glucan structural analogs. We found that high molecular weight (MW) glucans beta-(1,6) pustulan and beta-(1,3) curdlan totally mimicked the effect of C. albicans beta-glucan, while the low MW beta-(1,3) glucan laminarin did not have any effect. Because beta-glucan is a common constituent of all fungi and is abundantly released in vivo during systemic fungal infection, this novel effect of beta-glucan has potential implications for host-parasite relationship in candidiasis and other mycoses. In particular, our data suggest that beta-glucan could bias noninfected, bystander monocytes, thus aggravating the general immunodeficiency, predisposing to systemic fungal infection.


Candida albicans/immunology , Cell Differentiation , Cell Wall/metabolism , Dendritic Cells/cytology , Monocytes/cytology , beta-Glucans/pharmacology , Antigen Presentation , Candidiasis/immunology , Candidiasis/metabolism , Candidiasis/pathology , Cell Proliferation , Cell Wall/immunology , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/physiology , Humans , Monocytes/physiology , Phenotype , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
17.
Infect Immun ; 72(8): 4385-92, 2004 Aug.
Article En | MEDLINE | ID: mdl-15271894

Dendritic cells (DCs) are critical for initiating a pathogen-specific T-cell response. During chronic infections the pool of tissue DCs must be renewed by recruitment of both circulating DC progenitors and in loco differentiating monocytes. However, the interaction of monocytes with pathogens could affect their differentiation. Mycobacterium tuberculosis has been shown to variably interfere with the generation and function of antigen-presenting cells (APCs). In this study we found that when alpha interferon (IFN-alpha) is used as an inductor of monocyte differentiation, M. tuberculosis inhibits the generation of DCs, forcing the generation of immunoprivileged macrophage-like cells instead. Cells derived from M. tuberculosis-infected monocyte-derived macrophages (M. tuberculosis-infected MoMphi) retained CD14 without acquiring CD1 molecules and partially expressed B7.2 but did not up-regulate B7.1 and major histocompatibility complex (MHC) class I and II molecules. They synthesized tumor necrosis factor alpha and interleukin-10 (IL-10) but not IL-12. They also showed a reduced ability to induce proliferation and functional polarization of allogeneic T lymphocytes. Thus, in the presence of IFN-alpha, M. tuberculosis may hamper the renewal of potent APCs, such as DCs, generating a safe habitat for intracellular growth. M. tuberculosis-infected MoMphi, in fact, showed reduced expression of both signal 1 (CD1, MHC classes I and II) and signal 2 (B7.1 and B7.2), which are essential for mycobacterium-specific T-lymphocyte priming and/or activation. These data further suggest that M. tuberculosis has the ability to specifically interfere with monocyte differentiation. This ability may represent an effective M. tuberculosis strategy for eluding immune surveillance and persisting in the host.


Cell Differentiation/drug effects , Dendritic Cells/immunology , Interferon-alpha/pharmacology , Macrophages/cytology , Monocytes/cytology , Mycobacterium tuberculosis/pathogenicity , Antigen Presentation , Cells, Cultured , Coculture Techniques , Dendritic Cells/cytology , Granulocyte-Macrophage Colony-Stimulating Factor , Humans , Interferon-alpha/metabolism , Lymphocyte Activation , Macrophages/microbiology
18.
Infect Immun ; 72(2): 833-43, 2004 Feb.
Article En | MEDLINE | ID: mdl-14742527

The ability of Candida albicans to convert from the yeast (Y) form to mycelial forms through germ tube (GT) formation is considered a key feature of the transition of the organism from commensalism to virulence. We show here that human monocytes cultured with granulocyte-macrophage colony-stimulating factor and interleukin-4 (IL-4) after phagocytosis of Y forms did not differentiate into dendritic cells (DCs); they retained CD14, did not acquire CD1a, and were unable to express the maturation markers CD83 and CCR7. Moreover, they did not produce IL-12p70 but secreted IL-10. In addition, they spontaneously expressed high levels of tumor necrosis factor alpha (TNF-alpha), IL-6, and IL-8 mRNA transcripts and were able to induce proliferation of alloreactive memory but not naïve T lymphocytes. Conversely, monocytes that had phagocytosed GT forms differentiated into mature CD83+ and CCR7+ DCs; however, there was no up-regulation of CD40, CD80, and major histocompatibility complex class II, irrespective of lipopolysaccharide (LPS) treatment. In addition, these cells were unable to produce IL-12 even after LPS stimulation, but they were not functionally exhausted, as shown by their capacity to express TNF-alpha and IL-8 mRNA transcripts. These cells were able to prime naïve T cells but not to induce their functional polarization into effector cells. These data indicate that phagocytosis of Y and GT forms has profound and distinct effects on the differentiation pathway of monocytes. Thus, the differentiation of human monocytes into DCs appears to be tunable and exploitable by C. albicans to elude immune surveillance.


Candida albicans/immunology , Dendritic Cells/cytology , Monocytes/cytology , Antigen Presentation , Cell Differentiation , Dendritic Cells/physiology , Dendritic Cells/ultrastructure , Humans , Interleukin-12/biosynthesis , Microscopy, Electron , Monocytes/physiology , Monocytes/ultrastructure , Phagocytosis , Protein Subunits/biosynthesis
19.
J Am Acad Child Adolesc Psychiatry ; 42(3): 331-9, 2003 Mar.
Article En | MEDLINE | ID: mdl-12595787

OBJECTIVE: To evaluate short- and long-term effects of time-limited psychodynamic psychotherapy (PP) for children with internalizing disorders. METHOD: Fifty-eight outpatient children (6.3-10.9 years old), seen in a process of routine care and meeting criteria for depressive or anxiety disorder, were assigned to either active treatment or community services. Subjects were measured at baseline, after 6 months, and at a 2-year follow-up, by Children's Global Assessment Scale (C-GAS) and Child Behavior Checklist (CBCL). RESULTS: Major improvements in the experimental group were found in C-GAS and CBCL. These differences are noted at different times, with the C-GAS findings seen at 6 months and the CBCL findings at 2-year follow-up. Significant differences were found also for externalizing syndrome scales. CONCLUSIONS: PP is effective in treating internalizing disorders in routine outpatient care. The benefits of treatment are manifest both immediately and with delayed onset (sleeper effect). The finding that PP patients sought mental health services at a significantly lower rate than comparison conditions represents an important economic impact of PP.


Anxiety Disorders/therapy , Depressive Disorder, Major/therapy , Psychotherapy/methods , Anxiety Disorders/diagnosis , Child , Community Mental Health Services , Depressive Disorder, Major/diagnosis , Diagnostic and Statistical Manual of Mental Disorders , Female , Follow-Up Studies , Humans , Male , Surveys and Questionnaires , Treatment Outcome
20.
Eur J Immunol ; 32(11): 3050-8, 2002 Nov.
Article En | MEDLINE | ID: mdl-12385024

Intracellular pathogens have developed strategies for evading elimination by the defenses of the host immune system. Here we describe an escape mechanism utilized by Mycobacterium tuberculosis that involves the interference with the generation of fully competent DC from monocytes. We show that monocytes infected with live M. tuberculosis differentiated into mature, CD83+ and CCR7+ DC (Mt-MoDC), but were characterized by a selective failure in the expression of the family of CD1 molecules. These cells also showed levels of MHC class II and CD80 (B7.1) that were reduced in comparison with LPS-matured DC. In addition, Mt-MoDC produced TNF-alpha and IL-10, but were unable to secrete IL-12. The generation of Mt-MoDC required the infection of monocytes with live M. tuberculosis, since infection with heat-killed bacteria partially abrogated the effects on monocyte differentiation. Interestingly, Mt-MoDC revealed an impaired antigen-presentation function as assessed by the reduced capability to induce proliferation of cord blood T lymphocytes. Further, naive T lymphocytes expanded by Mt-MoDC were unable to secrete cytokines, in particular IL-4 and IFN-gamma, suggesting that they could be ineffective in helping the macrophage-mediated killing of intracellular mycobacteria. Our results suggest that the interference with monocyte differentiation into fully competent DC is an evasion mechanism of M. tuberculosis that could contribute to its intracellular persistence by avoiding immune recognition.


Dendritic Cells/physiology , Monocytes/physiology , Mycobacterium tuberculosis/physiology , Antigen Presentation , Antigens, CD1/analysis , Cell Differentiation , Dendritic Cells/immunology , Humans , Immunophenotyping , Interferon-gamma/biosynthesis , Interleukin-12/biosynthesis , Monocytes/immunology , Monocytes/microbiology
...