Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 138
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Mol Sci ; 25(9)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38732063

ABSTRACT

Epidermal growth factor receptor (EGFR)-mutant non-small-cell lung cancer (NSCLC) patients treated with EGFR-tyrosine kinase inhibitors (TKIs) inevitably develop resistance through several biological mechanisms. However, little is known on the molecular mechanisms underlying acquired resistance to suboptimal EGFR-TKI doses, due to pharmacodynamics leading to inadequate drug exposure. To evaluate the effects of suboptimal EGFR-TKI exposure on resistance in NSCLC, we obtained HCC827 and PC9 cell lines resistant to suboptimal fixed and intermittent doses of gefitinib and compared them to cells exposed to higher doses of the drug. We analyzed the differences in terms of EGFR signaling activation and the expression of epithelial-mesenchymal transition (EMT) markers, whole transcriptomes byRNA sequencing, and cell motility. We observed that the exposure to low doses of gefitinib more frequently induced a partial EMT associated with an induced migratory ability, and an enhanced transcription of cancer stem cell markers, particularly in the HCC827 gefitinib-resistant cells. Finally, the HCC827 gefitinib-resistant cells showed increased secretion of the EMT inducer transforming growth factor (TGF)-ß1, whose inhibition was able to partially restore gefitinib sensitivity. These data provide evidence that different levels of exposure to EGFR-TKIs in tumor masses might promote different mechanisms of acquired resistance.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Cell Movement , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , ErbB Receptors , Gefitinib , Lung Neoplasms , Protein Kinase Inhibitors , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/genetics , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/metabolism , ErbB Receptors/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Gefitinib/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Antineoplastic Agents/pharmacology , Signal Transduction/drug effects , Transforming Growth Factor beta1/metabolism
2.
Brief Bioinform ; 22(6)2021 11 05.
Article in English | MEDLINE | ID: mdl-34050359

ABSTRACT

MOTIVATION: Assessment of genetic mutations is an essential element in the modern era of personalized cancer treatment. Our strategy is focused on 'multiple network analysis' in which we try to improve cancer diagnostics by using biological networks. Genetic alterations in some important hubs or in driver genes such as BRAF and TP53 play a critical role in regulating many important molecular processes. Most of the studies are focused on the analysis of the effects of single mutations, while tumors often carry mutations of multiple driver genes. The aim of this work is to define an innovative bioinformatics pipeline focused on the design and analysis of networks (such as biomedical and molecular networks), in order to: (1) improve the disease diagnosis; (2) identify the patients that could better respond to a given drug treatment; and (3) predict what are the primary and secondary effects of gene mutations involved in human diseases. RESULTS: By using our pipeline based on a multiple network approach, it has been possible to demonstrate and validate what are the joint effects and changes of the molecular profile that occur in patients with metastatic colorectal carcinoma (mCRC) carrying mutations in multiple genes. In this way, we can identify the most suitable drugs for the therapy for the individual patient. This information is useful to improve precision medicine in cancer patients. As an application of our pipeline, the clinically significant case studies of a cohort of mCRC patients with the BRAF V600E-TP53 I195N missense combined mutation were considered. AVAILABILITY: The procedures used in this paper are part of the Cytoscape Core, available at (www.cytoscape.org). Data used here on mCRC patients have been published in [55]. SUPPLEMENTARY INFORMATION: A supplementary file containing a more detailed discussion of this case study and other cases is available at the journal site as Supplementary Data.


Subject(s)
Biomarkers, Tumor , Computational Biology/methods , Disease Susceptibility , Neoplasms/etiology , Precision Medicine/methods , Gene Regulatory Networks , Humans , Metabolic Networks and Pathways , Neoplasms/metabolism , Protein Interaction Maps , Signal Transduction
3.
BMC Cancer ; 23(1): 1010, 2023 Oct 19.
Article in English | MEDLINE | ID: mdl-37858132

ABSTRACT

BACKGROUND: Metastatic disease in tumors originating from the gastrointestinal tract can exhibit varying degrees of tumor burden at presentation. Some patients follow a less aggressive disease course, characterized by a limited number of metastatic sites, referred to as "oligo-metastatic disease" (OMD). The precise biological characteristics that define the oligometastatic behavior remain uncertain. In this study, we present a protocol designed to prospectively identify OMD, with the aim of proposing novel therapeutic approaches and monitoring strategies. METHODS: The PREDICTION study is a monocentric, prospective, observational investigation. Enrolled patients will receive standard treatment, while translational activities will involve analysis of the tumor microenvironment and genomic profiling using immunohistochemistry and next-generation sequencing, respectively. The first primary objective (descriptive) is to determine the prevalence of biological characteristics in OMD derived from gastrointestinal tract neoplasms, including high genetic concordance between primary tumors and metastases, a significant infiltration of T lymphocytes, and the absence of clonal evolution favoring specific driver genes (KRAS and PIK3CA). The second co-primary objective (analytic) is to identify a prognostic score for true OMD, with a primary focus on metastatic colorectal cancer. The score will comprise genetic concordance (> 80%), high T-lymphocyte infiltration, and the absence of clonal evolution favoring driver genes. It is hypothesized that patients with true OMD (score 3+) will have a lower rate of progression/recurrence within one year (20%) compared to those with false OMD (80%). The endpoint of the co-primary objective is the rate of recurrence/progression at one year. Considering a reasonable probability (60%) of the three factors occurring simultaneously in true OMD (score 3+), using a significance level of α = 0.05 and a test power of 90%, the study requires a minimum enrollment of 32 patients. DISCUSSION: Few studies have explored the precise genetic and biological features of OMD thus far. In clinical settings, the diagnosis of OMD is typically made retrospectively, as some patients who undergo intensive treatment for oligometastases develop polymetastatic diseases within a year, while others do not experience disease progression (true OMD). In the coming years, the identification of true OMD will allow us to employ more personalized and comprehensive strategies in cancer treatment. TRIAL REGISTRATION: ClinicalTrials.gov ID NCT05806151.


Subject(s)
Gastrointestinal Neoplasms , Humans , Prospective Studies , Retrospective Studies , Gastrointestinal Neoplasms/genetics , Tumor Microenvironment
4.
Oncologist ; 27(1): 7-12, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35305107

ABSTRACT

Increasing evidence suggests that liquid biopsy might play a relevant role in the management of metastatic non-small cell lung cancer (NSCLC) patients. Here, we show how the Molecular Tumor Board (MTB) in our cancer center employed liquid biopsy to support therapeutic decisions in a patient with NSCLC carrying a rare EGFR mutation. A 44-year-old woman, never-smoker with an EGFR, ALK, and ROS1-negative lung adenocarcinoma and multiple brain metastases received systemic therapy and surgery before being referred to our Institute. The MTB suggested NGS testing of tumor biopsy that revealed a rare exon-20 EGFR insertion (p.His773dup; c.2315_2316insCCA) and EGFR amplification. The MTB recommended treatment with erlotinib and follow-up with liquid biopsy, by using both cell-free DNA (cfDNA) and circulating tumor cells (CTCs). An increase of EGFR mutation levels in cfDNA revealed resistance to treatment about 6 months before clinical progression. Extremely low levels of EGFR p.T790M were detected at progression. Based on preclinical data suggesting activity of osimertinib against EGFR exon-20 insertions, the MTB recommended treatment with brain and bone radiotherapy and osimertinib. A dramatic reduction of EGFR mutation levels in the cfDNA was observed after 4 weeks of treatment. The PET scan demonstrated a metabolic partial remission that was maintained for 9 months. This case supports the evidence that liquid biopsy can aid in the management of metastatic NSCLC. It also suggests that treatment with osimertinib might be a therapeutic option in patients with EGFR exon-20 insertions when a clinical trial is not available.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Cell-Free Nucleic Acids , Lung Neoplasms , Adult , Aniline Compounds/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , ErbB Receptors/genetics , ErbB Receptors/therapeutic use , Exons/genetics , Female , Humans , Liquid Biopsy , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutation , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics
5.
Immunity ; 39(2): 372-85, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23973224

ABSTRACT

Endogenous tryptophan (Trp) metabolites have an important role in mammalian gut immune homeostasis, yet the potential contribution of Trp metabolites from resident microbiota has never been addressed experimentally. Here, we describe a metabolic pathway whereby Trp metabolites from the microbiota balance mucosal reactivity in mice. Switching from sugar to Trp as an energy source (e.g., under conditions of unrestricted Trp availability), highly adaptive lactobacilli are expanded and produce an aryl hydrocarbon receptor (AhR) ligand-indole-3-aldehyde-that contributes to AhR-dependent Il22 transcription. The resulting IL-22-dependent balanced mucosal response allows for survival of mixed microbial communities yet provides colonization resistance to the fungus Candida albicans and mucosal protection from inflammation. Thus, the microbiota-AhR axis might represent an important strategy pursued by coevolutive commensalism for fine tuning host mucosal reactivity contingent on Trp catabolism.


Subject(s)
Candida albicans/immunology , Interleukins/metabolism , Limosilactobacillus reuteri/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Tryptophan/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Candidiasis/immunology , Energy Metabolism , Female , Gastrointestinal Tract/immunology , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Indoleamine-Pyrrole 2,3,-Dioxygenase/deficiency , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoles/metabolism , Interleukin-17/deficiency , Interleukin-17/genetics , Limosilactobacillus reuteri/growth & development , Limosilactobacillus reuteri/immunology , Metagenome , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Probiotics , Receptors, Aryl Hydrocarbon/deficiency , Receptors, Aryl Hydrocarbon/genetics , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/genetics , Tryptophan/chemistry , Interleukin-22
6.
J Biol Chem ; 295(33): 11866-11876, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32616652

ABSTRACT

Garcinoic acid (GA or δ-T3-13'COOH), is a natural vitamin E metabolite that has preliminarily been identified as a modulator of nuclear receptors involved in ß-amyloid (Aß) metabolism and progression of Alzheimer's disease (AD). In this study, we investigated GA's effects on Aß oligomer formation and deposition. Specifically, we compared them with those of other vitamin E analogs and the soy isoflavone genistein, a natural agonist of peroxisome proliferator-activated receptor γ (PPARγ) that has therapeutic potential for managing AD. GA significantly reduced Aß aggregation and accumulation in mouse cortical astrocytes. Similarly to genistein, GA up-regulated PPARγ expression and apolipoprotein E (ApoE) efflux in these cells with an efficacy that was comparable with that of its metabolic precursor δ-tocotrienol and higher than those of α-tocopherol metabolites. Unlike for genistein and the other vitamin E compounds, the GA-induced restoration of ApoE efflux was not affected by pharmacological inhibition of PPARγ activity, and specific activation of pregnane X receptor (PXR) was observed together with ApoE and multidrug resistance protein 1 (MDR1) membrane transporter up-regulation in both the mouse astrocytes and brain tissue. These effects of GA were associated with reduced Aß deposition in the brain of TgCRND8 mice, a transgenic AD model. In conclusion, GA holds potential for preventing Aß oligomerization and deposition in the brain. The mechanistic aspects of GA's properties appear to be distinct from those of other vitamin E metabolites and of genistein.


Subject(s)
Amyloid beta-Peptides/metabolism , Benzopyrans/pharmacology , Brain/drug effects , Protein Aggregation, Pathological/prevention & control , Vitamin E/analogs & derivatives , Amyloid beta-Peptides/ultrastructure , Animals , Benzopyrans/pharmacokinetics , Brain/metabolism , Brain/pathology , Male , Mice , Protein Aggregates/drug effects , Protein Aggregation, Pathological/pathology , Vitamin E/pharmacokinetics , Vitamin E/pharmacology
7.
Int J Mol Sci ; 22(12)2021 Jun 18.
Article in English | MEDLINE | ID: mdl-34207085

ABSTRACT

Inflammasomes are powerful cytosolic sensors of environmental stressors and are critical for triggering interleukin-1 (IL-1)-mediated inflammatory responses. However, dysregulation of inflammasome activation may lead to pathological conditions, and the identification of negative regulators for therapeutic purposes is increasingly being recognized. Anakinra, the recombinant form of the IL-1 receptor antagonist, proved effective by preventing the binding of IL-1 to its receptor, IL-1R1, thus restoring autophagy and dampening NLR family pyrin domain containing 3 (NLRP3) activity. As the generation of mitochondrial reactive oxidative species (ROS) is a critical upstream event in the activation of NLRP3, we investigated whether anakinra would regulate mitochondrial ROS production. By profiling the activation of transcription factors induced in murine alveolar macrophages, we found a mitochondrial antioxidative pathway induced by anakinra involving the manganese-dependent superoxide dismutase (MnSOD) or SOD2. Molecularly, anakinra promotes the binding of SOD2 with the deubiquitinase Ubiquitin Specific Peptidase 36 (USP36) and Constitutive photomorphogenesis 9 (COP9) signalosome, thus increasing SOD2 protein longevity. Functionally, anakinra and SOD2 protects mice from pulmonary oxidative inflammation and infection. On a preclinical level, anakinra upregulates SOD2 in murine models of chronic granulomatous disease (CGD) and cystic fibrosis (CF). These data suggest that protection from mitochondrial oxidative stress may represent an additional mechanism underlying the clinical benefit of anakinra and identifies SOD2 as a potential therapeutic target.


Subject(s)
Inflammasomes/metabolism , Interleukin 1 Receptor Antagonist Protein/pharmacology , Recombinant Proteins/pharmacology , Superoxide Dismutase/metabolism , Animals , Cells, Cultured , Cystic Fibrosis/etiology , Cystic Fibrosis/metabolism , Cystic Fibrosis/pathology , Disease Models, Animal , Granulomatous Disease, Chronic/etiology , Granulomatous Disease, Chronic/metabolism , Granulomatous Disease, Chronic/pathology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Mice , Mice, Knockout , Mitochondria/metabolism , Oxidation-Reduction , Oxidative Stress , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Transcription Factors/metabolism
8.
Int J Mol Sci ; 21(18)2020 Sep 18.
Article in English | MEDLINE | ID: mdl-32962091

ABSTRACT

Fibroblast growth factor receptors (FGFRs) are tyrosine kinase receptors involved in many biological processes. Deregulated FGFR signaling plays an important role in tumor development and progression in different cancer types. FGFR genomic alterations, including FGFR gene fusions that originate by chromosomal rearrangements, represent a promising therapeutic target. Next-generation-sequencing (NGS) approaches have significantly improved the discovery of FGFR gene fusions and their detection in clinical samples. A variety of FGFR inhibitors have been developed, and several studies are trying to evaluate the efficacy of these agents in molecularly selected patients carrying FGFR genomic alterations. In this review, we describe the most frequent FGFR aberrations in human cancer. We also discuss the different approaches employed for the detection of FGFR fusions and the potential role of these genomic alterations as prognostic/predictive biomarkers.


Subject(s)
Molecular Targeted Therapy/methods , Neoplasms/metabolism , Neoplasms/therapy , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/genetics , Disease Progression , Gene Fusion , Genomics , High-Throughput Nucleotide Sequencing , Humans , Mutation , Neoplasms/diagnosis , Neoplasms/genetics , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction
9.
Mediators Inflamm ; 2018: 7396136, 2018.
Article in English | MEDLINE | ID: mdl-30510489

ABSTRACT

Mast cells are increasingly being recognized as crucial cells in the response of the organism to environmental agents. Interestingly, the ability of mast cells to sense and respond to external cues is modulated by the microenvironment that surrounds mast cells and influences their differentiation. The scenario that is emerging unveils a delicate equilibrium that balances the effector functions of mast cells to guarantee host protection without compromising tissue homeostasis. Among the environmental components able to mold mast cells and fine-tune their effector functions, the microorganisms that colonize the human body, collectively known as microbiome, certainly play a key role. Indeed, microorganisms can regulate not only the survival, recruitment, and maturation of mast cells but also their activity by setting the threshold required for the exploitation of their different effector functions. Herein, we summarize the current knowledge about the mechanisms underlying the ability of the microorganisms to regulate mast cell physiology and discuss potential deviations that result in pathological consequences. We will discuss the pivotal role of the aryl hydrocarbon receptor in sensing the environment and shaping mast cell adaptation at the host-microbe interface.


Subject(s)
Mast Cells/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Animals , Host-Pathogen Interactions/physiology , Humans , Microbiota/physiology
10.
Mediators Inflamm ; 2018: 1601486, 2018.
Article in English | MEDLINE | ID: mdl-29670460

ABSTRACT

Tryptophan (trp) metabolism is an important regulatory component of gut mucosal homeostasis and the microbiome. Metabolic pathways targeting the trp can lead to a myriad of metabolites, of both host and microbial origins, some of which act as endogenous low-affinity ligands for the aryl hydrocarbon receptor (AhR), a cytosolic, ligand-operated transcription factor that is involved in many biological processes, including development, cellular differentiation and proliferation, xenobiotic metabolism, and the immune response. Low-level activation of AhR by endogenous ligands is beneficial in the maintenance of immune health and intestinal homeostasis. We have defined a functional node whereby certain bacteria species contribute to host/microbial symbiosis and mucosal homeostasis. A microbial trp metabolic pathway leading to the production of indole-3-aldehyde (3-IAld) by lactobacilli provided epithelial protection while inducing antifungal resistance via the AhR/IL-22 axis. In this review, we highlight the role of AhR in inflammatory lung diseases and discuss the possible therapeutic use of AhR ligands in cystic fibrosis.


Subject(s)
Cystic Fibrosis/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Animals , Humans , Indoles/metabolism , Lactobacillus/metabolism
11.
Mediators Inflamm ; 2018: 6195958, 2018.
Article in English | MEDLINE | ID: mdl-29692681

ABSTRACT

Phagocytes fight fungi using canonical and noncanonical, also called LC3-associated phagocytosis (LAP), autophagy pathways. However, the outcomes of autophagy/LAP in shaping host immune responses appear to greatly vary depending on fungal species and cell types. By allowing efficient pathogen clearance and/or degradation of inflammatory mediators, autophagy proteins play a broad role in cellular and immune homeostasis during fungal infections. Indeed, defects in autophagic machinery have been linked with aberrant host defense and inflammatory states. Thus, understanding the molecular mechanisms underlying the relationship between the different forms of autophagy may offer a way to identify drugable molecular signatures discriminating between selective recognition of cargo and host protection. In this regard, IFN-γ and anakinra are teaching examples of successful antifungal agents that target the autophagy machinery. This article provides an overview of the role of autophagy/LAP in response to fungi and in their infections, regulation, and therapeutic exploitation.


Subject(s)
Autophagy/physiology , Phagocytosis/physiology , Animals , Humans , Interferon-gamma/metabolism , Interleukin 1 Receptor Antagonist Protein/metabolism , Phagocytes/metabolism , Phagocytes/physiology
12.
Proc Natl Acad Sci U S A ; 111(9): 3526-31, 2014 Mar 04.
Article in English | MEDLINE | ID: mdl-24550444

ABSTRACT

Patients with chronic granulomatous disease (CGD) have a mutated NADPH complex resulting in defective production of reactive oxygen species; these patients can develop severe colitis and are highly susceptible to invasive fungal infection. In NADPH oxidase-deficient mice, autophagy is defective but inflammasome activation is present despite lack of reactive oxygen species production. However, whether these processes are mutually regulated in CGD and whether defective autophagy is clinically relevant in patients with CGD is unknown. Here, we demonstrate that macrophages from CGD mice and blood monocytes from CGD patients display minimal recruitment of microtubule-associated protein 1 light chain 3 (LC3) to phagosomes. This defect in autophagy results in increased IL-1ß release. Blocking IL-1 with the receptor antagonist (anakinra) decreases neutrophil recruitment and T helper 17 responses and protects CGD mice from colitis and also from invasive aspergillosis. In addition to decreased inflammasome activation, anakinra restored autophagy in CGD mice in vivo, with increased Aspergillus-induced LC3 recruitment and increased expression of autophagy genes. Anakinra also increased Aspergillus-induced LC3 recruitment from 23% to 51% (P < 0.01) in vitro in monocytes from CGD patients. The clinical relevance of these findings was assessed by treating CGD patients who had severe colitis with IL-1 receptor blockade using anakinra. Anakinra treatment resulted in a rapid and sustained improvement in colitis. Thus, inflammation in CGD is due to IL-1-dependent mechanisms, such as decreased autophagy and increased inflammasome activation, which are linked pathological conditions in CGD that can be restored by IL-1 receptor blockade.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Colitis/drug therapy , Granulomatous Disease, Chronic/drug therapy , Interleukin 1 Receptor Antagonist Protein/pharmacology , Phagosomes/metabolism , Receptors, Interleukin-1/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/therapeutic use , Aspergillus fumigatus , Autophagy/physiology , Colitis/etiology , Colitis/immunology , Enzyme-Linked Immunosorbent Assay , Granulomatous Disease, Chronic/complications , Granulomatous Disease, Chronic/immunology , Humans , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microtubule-Associated Proteins/metabolism , NADPH Oxidases/deficiency , NADPH Oxidases/genetics , Real-Time Polymerase Chain Reaction , Statistics, Nonparametric
13.
Biochim Biophys Acta ; 1854(5): 410-25, 2015 May.
Article in English | MEDLINE | ID: mdl-25617660

ABSTRACT

Vascular endothelial growth factor A (VEGFA) has different biological activities and plays a central role in tumor proliferation, angiogenesis and metastasis. Different VEGFA isoforms are generated by alternative splice site selection of exons 6, 7 and 8. In this paper, we analyzed the physical and chemical properties of the VEGFA exon 6 sequence, and modeled the three-dimensional structures of the regions corresponding to exons 6, 7 and 8 of six different pro-angiogenic isoforms of VEGFA in comparison to the experimental structure of VEGFA_165 by a combined approach of fold recognition and comparative modeling strategies and molecular dynamics simulations. Our results showed that i) exon 6 is a very flexible polycation with high disordered propensity, features well conserved in all mammals, ii) the structures of all the isoforms are stabilized by H-bond sub-networks organized around HUB residues and, iii) the charge content of exon 6 modulates the intrinsic structural preference of its flexible backbone, which can be described as an ensemble of conformations. Moreover, complexes between NRP-1 and VEGFA isoforms were modeled by molecular docking to study what isoforms are able to bind NRP-1. The analysis of complexes evidenced that VEGFA_121, VEGFA_145, VEGFA_183, VEGFA_189 and VEGFA_206, containing exons 7 and 8a, are able to interact with NRP-1 because they have the key regions of exons 7b and/or 8a. An overview of the isoforms shows how the fluctuations are the main guidance of their biological function. MD simulations also provide insights into factors that stabilize the binding regions of isoforms.


Subject(s)
Carcinogenesis , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/physiology , Amino Acid Sequence , Angiogenesis Inducing Agents/chemistry , Carcinogenesis/genetics , Carcinogenesis/metabolism , Humans , Models, Molecular , Molecular Dynamics Simulation , Molecular Sequence Data , Neuropilin-1/chemistry , Neuropilin-1/metabolism , Protein Interaction Domains and Motifs , Protein Isoforms/chemistry , Protein Isoforms/physiology , Protein Structure, Quaternary , Sequence Alignment , Structure-Activity Relationship
14.
J Cell Physiol ; 231(7): 1514-21, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26542886

ABSTRACT

Vascular endothelial growth factor A (VEGFA) is one of the main mediators of angiogenesis in non-small cell lung cancer (NSCLC). Recently, it has been described an autocrine feed-forward loop in NSCLC cells in which tumor-derived VEGFA promoted the secretion of VEGFA itself, amplifying the proangiogenic signal. In order to investigate the role of VEGFA in lung cancer progression, we assessed the effects of recombinant VEGFA on proliferation, migration, and secretion of other angiogenic factors in A549, H1975, and HCC827 NSCLC cell lines. We found that VEGFA did not affect NSCLC cell proliferation and migration. On the other hand, we demonstrated that VEGFA not only produced a strong and persistent increase of VEGFA itself but also significantly induced the secretion of a variety of angiogenic factors, including follistatin (FST), hepatocyte growth factor (HGF), angiopoietin-2 (ANGPT2), granulocyte-colony stimulating factor (G-CSF), interleukin (IL)-8, leptin (LEP), platelet/endothelial cell adhesion molecule 1 (PECAM-1), and platelet-derived growth factor bb (PDGF-BB). PI3K/AKT, RAS/ERK, and STAT3 signalling pathways were found to mediate the effects of VEGFA in NSCLC cell lines. We also observed that VEGFA regulation mainly occurred at post-transcriptional level and that NSCLC cells expressed different isoforms of VEGFA. Collectively, our data suggested that VEGFA contributes to lung cancer progression by inducing a network of angiogenic factors, which might offer potential for therapeutic intervention.


Subject(s)
Angiogenic Proteins/biosynthesis , Carcinoma, Non-Small-Cell Lung/genetics , Recombinant Proteins/administration & dosage , Vascular Endothelial Growth Factor A/genetics , Angiogenic Proteins/genetics , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Recombinant Proteins/genetics , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/administration & dosage
15.
PLoS Pathog ; 10(3): e1003936, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24603878

ABSTRACT

The galactosaminogalactan (GAG) is a cell wall component of Aspergillus fumigatus that has potent anti-inflammatory effects in mice. However, the mechanisms responsible for the anti-inflammatory property of GAG remain to be elucidated. In the present study we used in vitro PBMC stimulation assays to demonstrate, that GAG inhibits proinflammatory T-helper (Th)1 and Th17 cytokine production in human PBMCs by inducing Interleukin-1 receptor antagonist (IL-1Ra), a potent anti-inflammatory cytokine that blocks IL-1 signalling. GAG cannot suppress human T-helper cytokine production in the presence of neutralizing antibodies against IL-1Ra. In a mouse model of invasive aspergillosis, GAG induces IL-1Ra in vivo, and the increased susceptibility to invasive aspergillosis in the presence of GAG in wild type mice is not observed in mice deficient for IL-1Ra. Additionally, we demonstrate that the capacity of GAG to induce IL-1Ra could also be used for treatment of inflammatory diseases, as GAG was able to reduce severity of an experimental model of allergic aspergillosis, and in a murine DSS-induced colitis model. In the setting of invasive aspergillosis, GAG has a significant immunomodulatory function by inducing IL-1Ra and notably IL-1Ra knockout mice are completely protected to invasive pulmonary aspergillosis. This opens new treatment strategies that target IL-1Ra in the setting of acute invasive fungal infection. However, the observation that GAG can also protect mice from allergy and colitis makes GAG or a derivative structure of GAG a potential treatment compound for IL-1 driven inflammatory diseases.


Subject(s)
Aspergillosis/immunology , Aspergillus fumigatus/immunology , Fungal Polysaccharides/immunology , Interleukin 1 Receptor Antagonist Protein/biosynthesis , Polysaccharides/immunology , Virulence Factors/immunology , Animals , Cytokines/biosynthesis , Cytokines/immunology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Humans , Interleukin 1 Receptor Antagonist Protein/immunology , Leukocytes, Mononuclear/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout
16.
J Cell Biochem ; 116(12): 2778-85, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25959272

ABSTRACT

Although evidence suggests that the RAF/MEK/ERK pathway plays an important role in triple negative breast cancer (TNBC), resistance to MEK inhibitors has been observed in TNBC cells. Different mechanisms have been hypothesized to be involved in this phenomenon, including receptor tyrosine kinase-dependent activation of the PI3K/AKT pathway. In this study, we analyzed the effects of the MEK1/2 inhibitor selumetinib in combination with the epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor gefitinib in a panel of TNBC cell lines that showed different levels of sensitivity to single-agent selumetinib: SUM-149 and MDA-MB-231 cells resulted to be sensitive, whereas SUM-159, MDA-MB-468 and HCC70 cells were relatively resistant to the drug. Treatment of TNBC cells with selumetinib produced an increase of the phosphorylation of the EGFR both in selumetinib-sensitive SUM-149, MDA-MB-231 and in selumetinib-resistant MDA-MB-468 TNBC cells. The combination of selumetinib and gefitinib resulted in a synergistic growth inhibitory effect in all the TNBC cell lines, although the IC50 was not reached in SUM-159 and MDA-MB-468 cells. This effect was associated with an almost complete suppression of ERK1/2 activation and a reduction of selumetinib-induced AKT phosphorylation. In addition, in selumetinib-sensitive TNBC cells the combination of selumetinib and gefitinib induced a significant G0/G1 cell cycle arrest and apoptosis. Taken together, our data demonstrated that blockade of the EGFR might efficiently increase the antitumor activity of selumetinib in a subgroup of TNBC and that this phenomenon might be related to the effects of such combination on both ERK1/2 and AKT activation.


Subject(s)
Benzimidazoles/administration & dosage , ErbB Receptors/antagonists & inhibitors , MAP Kinase Kinase Kinases/antagonists & inhibitors , Quinazolines/administration & dosage , Triple Negative Breast Neoplasms/drug therapy , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , Drug Synergism , ErbB Receptors/biosynthesis , Female , Gefitinib , Humans , MAP Kinase Kinase Kinases/biosynthesis , MAP Kinase Signaling System/drug effects , Oncogene Protein v-akt/genetics , Protein Kinase Inhibitors/administration & dosage , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
17.
Eur J Immunol ; 44(11): 3192-200, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25256754

ABSTRACT

An increased understanding of the importance of microbiota in shaping the host's immune and metabolic activities has rendered fungal interactions with their hosts more complex than previously appreciated. The aryl hydrocarbon receptor (AhR) has a pivotal role in connecting tryptophan catabolism by microbial communities and the host's own pathway of tryptophan metabolite production with the orchestration of T-cell function. AhR activation by a Lactobacillus-derived AhR ligand leads to the production of IL-22 to the benefit of mucosal defense mechanisms, an activity upregulated in the absence of the host tryptophan catabolic enzyme, indoleamine 2,3-dioxygenase 1 (IDO1), which is required for protection from fungal diseases ("disease tolerance"). As AhR activation in turn leads to the activation-in a feedback fashion-of IDO1, the regulatory loop involving AhR and IDO1 may have driven the coevolution of commensal fungi with the mammalian immune system and the microbiota, to the benefit of host survival and fungal commensalism. This review will discuss the essential help the microbiota provides in controlling the balance between the dual nature of the fungal-host relationship, namely, commensalism vs. infection.


Subject(s)
Fungi/immunology , Mycoses/immunology , Receptors, Aryl Hydrocarbon/metabolism , Symbiosis/immunology , Tryptophan/metabolism , Fungi/pathogenicity , Humans , Immune Tolerance/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Interleukins/biosynthesis , Interleukins/immunology , Lactobacillus/metabolism , Microbiota , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Interleukin-22
18.
PLoS Pathog ; 9(7): e1003486, 2013.
Article in English | MEDLINE | ID: mdl-23853597

ABSTRACT

The ability to tolerate Candida albicans, a human commensal of the gastrointestinal tract and vagina, implicates that host defense mechanisms of resistance and tolerance cooperate to limit fungal burden and inflammation at the different body sites. We evaluated resistance and tolerance to the fungus in experimental and human vulvovaginal candidiasis (VVC) as well as in recurrent VVC (RVVC). Resistance and tolerance mechanisms were both activated in murine VVC, involving IL-22 and IL-10-producing regulatory T cells, respectively, with a major contribution by the enzyme indoleamine 2,3-dioxygenase 1 (IDO1). IDO1 was responsible for the production of tolerogenic kynurenines, such that replacement therapy with kynurenines restored immunoprotection to VVC. In humans, two functional genetic variants in IL22 and IDO1 genes were found to be associated with heightened resistance to RVVC, and they correlated with increased local expression of IL-22, IDO1 and kynurenines. Thus, IL-22 and IDO1 are crucial in balancing resistance with tolerance to Candida, their deficiencies are risk factors for RVVC, and targeting tolerance via therapeutic kynurenines may benefit patients with RVVC.


Subject(s)
Candida albicans/immunology , Candidiasis, Vulvovaginal/immunology , Immune Tolerance , Immunity, Mucosal , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interleukins/biosynthesis , T-Lymphocytes, Regulatory/immunology , Animals , Candida albicans/drug effects , Candida albicans/isolation & purification , Candidiasis, Vulvovaginal/genetics , Candidiasis, Vulvovaginal/metabolism , Candidiasis, Vulvovaginal/microbiology , Female , Genetic Association Studies , Genetic Variation , Humans , Immune Tolerance/drug effects , Immunity, Mucosal/drug effects , Immunologic Factors/metabolism , Immunologic Factors/therapeutic use , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Interleukin-10/biosynthesis , Interleukins/genetics , Kynurenine/metabolism , Kynurenine/therapeutic use , Mice , Mice, Inbred C57BL , Mice, SCID , Recurrence , Severe Combined Immunodeficiency/drug therapy , Severe Combined Immunodeficiency/immunology , Severe Combined Immunodeficiency/physiopathology , Specific Pathogen-Free Organisms , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Interleukin-22
19.
Future Oncol ; 11(11): 1611-23, 2015.
Article in English | MEDLINE | ID: mdl-26043215

ABSTRACT

ABSTRACT The presence of EGFR mutations predicts the sensitivity to EGF receptor (EGFR)-tyrosine kinase inhibitors in a molecularly defined subset of non-small-cell lung carcinoma (NSCLC) patients. For this reason, EGFR testing of NSCLC is required to provide personalized treatment options and better outcomes for NSCLC patients. As surgery specimens are not available in the majority of NSCLC, other currently available DNA sources are small biopsies and cytological samples, providing however limited and low-quality material. In order to address this issue, the use of surrogate sources of DNA, such as blood, serum and plasma samples, which often contains circulating free tumor DNA or circulating tumor cells, is emerging as a new strategy for tumor genotyping.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , DNA, Neoplasm/blood , ErbB Receptors/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Biomarkers, Tumor/genetics , Biopsy/methods , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride/therapeutic use , Gefitinib , Genotyping Techniques , Humans , Mutation , Precision Medicine , Protein Kinase Inhibitors/therapeutic use , Quinazolines/therapeutic use , Tumor Cells, Cultured
20.
Immunol Cell Biol ; 92(8): 659-70, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24820384

ABSTRACT

Unlike induced Foxp3(+) regulatory T cells (Foxp3(+) iTreg) that have been shown to play an essential role in the development of protective immunity to the ubiquitous mold Aspergillus fumigatus, type-(1)-regulatory T cells (Tr1) cells have, thus far, not been implicated in this process. Here, we evaluated the role of Tr1 cells specific for an epitope derived from the cell wall glucanase Crf-1 of A. fumigatus (Crf-1/p41) in antifungal immunity. We identified Crf-1/p41-specific latent-associated peptide(+) Tr1 cells in healthy humans and mice after vaccination with Crf-1/p41+zymosan. These cells produced high amounts of interleukin (IL)-10 and suppressed the expansion of antigen-specific T cells in vitro and in vivo. In mice, in vivo differentiation of Tr1 cells was dependent on the presence of the aryl hydrocarbon receptor, c-Maf and IL-27. Moreover, in comparison to Tr1 cells, Foxp3(+) iTreg that recognize the same epitope were induced in an interferon gamma-type inflammatory environment and more potently suppressed innate immune cell activities. Overall, our data show that Tr1 cells are involved in the maintenance of antifungal immune homeostasis, and most likely play a distinct, yet complementary, role compared with Foxp3(+) iTreg.


Subject(s)
Aspergillus fumigatus/immunology , T-Cell Antigen Receptor Specificity/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, Fungal/administration & dosage , Antigens, Fungal/chemistry , Antigens, Fungal/immunology , Aspergillosis/immunology , Aspergillosis/metabolism , Cytokines/metabolism , Cytomegalovirus/immunology , Epitopes, T-Lymphocyte , Female , Forkhead Transcription Factors/metabolism , Healthy Volunteers , Humans , Immunomodulation , Immunophenotyping , Lymphocyte Activation , Mice , Mice, Knockout , Peptides/administration & dosage , Peptides/chemistry , Peptides/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL