Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Int J Mol Sci ; 23(6)2022 Mar 16.
Article in English | MEDLINE | ID: mdl-35328630

ABSTRACT

Immune checkpoint inhibitors (ICI) represented a step forward in improving the outcome of patients with various refractory solid tumors and several therapeutic regimens incorporating ICI have already been approved for a variety of tumor entities. However, besides remarkable long-term responses, checkpoint inhibition can trigger severe immune-related adverse events in some patients. In order to improve safety of ICI as well as T cell therapy, we tested the feasibility of combining T cell-based immunotherapy with genetic disruption of checkpoint molecule expression. Therefore, we generated H-Y and ovalbumin antigen-specific CD8+ T cells with abolished PD-1, LAG-3, and TIM-3 expression through CRISPR/Cas9 technology. CD8+ T cells, subjected to PD-1, LAG-3, and TIM-3 genetic editing, showed a strong reduction in immune checkpoint molecule expression after in vitro activation, while no relevant reduction in responsiveness to in vitro stimulation was observed. At the same time, in B16-OVA tumor model, transferred genetically edited OT-1 CD8+ T cells promoted longer survival compared to control T cells and showed enhanced expansion without associated toxicity. Our study supports the notion that antigen-specific adoptive T cell therapy with concomitant genetic disruption of multiple checkpoint inhibitory receptors could represent an effective antitumor immunotherapy approach with improved tolerability profile.


Subject(s)
Neoplasms , Programmed Cell Death 1 Receptor , CD8-Positive T-Lymphocytes , Hepatitis A Virus Cellular Receptor 2/genetics , Hepatitis A Virus Cellular Receptor 2/metabolism , Humans , Immunotherapy , Neoplasms/genetics , Neoplasms/therapy , Programmed Cell Death 1 Receptor/metabolism
2.
Int J Cancer ; 148(12): 3097-3110, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33600609

ABSTRACT

Adoptive T cell therapy (ATT) has revolutionized the treatment of cancer patients. A sufficient number of functional T cells are indispensable for ATT efficacy; however, several ATT dropouts have been reported due to T cell expansion failure or lack of T cell persistence in vivo. With the aim of providing ATT also to those patients experiencing insufficient T cell manufacturing via standard protocol, we evaluated if minimally manipulative prolongation of in vitro expansion (long-term [LT] >3 weeks with IL-7 and IL-15 cytokines) could result in enhanced T cell yield with preserved T cell functionality. The extended expansion resulted in a 39-fold increase of murine CD8+ T central memory cells (Tcm). LT expanded CD8+ and CD4+ Tcm cells retained a gene expression profile related to Tcm and T memory stem cells (Tscm). In vivo transfer of LT expanded Tcm revealed persistence and antitumor capacity. We confirmed our in vitro findings on human T cells, on healthy donors and diffuse large B cell lymphoma patients, undergoing salvage therapy. Our study demonstrates the feasibility of an extended T cell expansion as a practicable alternative for patients with insufficient numbers of T cells after the standard manufacturing process thereby increasing ATT accessibility.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Lymphoma, Large B-Cell, Diffuse/therapy , T-Lymphocytes/cytology , T-Lymphocytes/transplantation , Animals , Case-Control Studies , Cell Culture Techniques , Cell Line, Tumor , Cells, Cultured , Humans , Immunologic Memory , Immunotherapy, Adoptive , Interleukin-15/pharmacology , Interleukin-7/pharmacology , Male , Mice , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
3.
Biomed Pharmacother ; 111: 1399-1407, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30841455

ABSTRACT

Acute Respiratory Distress Syndrome (ARDS) is an inflammatory condition with high mortality rates, and there is still no pharmacological approach with proven effectiveness. In the past few years, several imidazole small molecules have been developed to treat conditions in which inflammation plays a central role. In the present work, we hypothesize that a novel substituted fluorophenyl imidazole synthetized by our research group would present in vivo anti-inflammatory effect in an ARDS murine model induced by LPS. Results shows that the fluorophenyl imidazole has the ability to inhibit leukocyte migration to the bronchoalveolar lavage fluid and lung tissue of animals challenged intranasally with LPS. Furthermore, this inhibition is followed with reduction in myeloperoxidase activity, nitric oxide metabolites generation and cytokines (TNF-α, IL-6, IL-17, IFN-γ and IL-10) secretion. This effect is at least partly related to the capacity of the fluorophenyl imidazole in inhibit p38 MAPK and NF-κB phosphorylation. Finally, fluorophenyl imidazole showed no signs of acute oral toxicity in the toxicological protocol suggested by OECD 423. Taken together, the results shows that fluorophenyl imidazole is a promising prototype for the development of a novel anti-inflammatory drug in which p38 MAPK and NF-κB plays a pivotal role.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Imidazoles/pharmacology , Inflammation/drug therapy , Animals , Bronchoalveolar Lavage Fluid/chemistry , Cytokines/metabolism , Inflammation/metabolism , Inflammation Mediators/pharmacology , Lipopolysaccharides/pharmacology , Lung/drug effects , Lung/metabolism , Male , Mice , Nitric Oxide/metabolism , Phosphorylation/drug effects , Respiratory Distress Syndrome , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Nat Commun ; 9(1): 5232, 2018 12 12.
Article in English | MEDLINE | ID: mdl-30542075

ABSTRACT

PI3K activation plays a central role in the development of pulmonary inflammation and tissue remodeling. PI3K inhibitors may thus offer an improved therapeutic opportunity to treat non-resolving lung inflammation but their action is limited by unwanted on-target systemic toxicity. Here we present CL27c, a prodrug pan-PI3K inhibitor designed for local therapy, and investigate whether inhaled CL27c is effective in asthma and pulmonary fibrosis. Mice inhaling CL27c show reduced insulin-evoked Akt phosphorylation in lungs, but no change in other tissues and no increase in blood glycaemia, in line with a local action. In murine models of acute or glucocorticoid-resistant neutrophilic asthma, inhaled CL27c reduces inflammation and improves lung function. Finally, inhaled CL27c administered in a therapeutic setting protects from bleomycin-induced lung fibrosis, ultimately leading to significantly improved survival. Therefore, local delivery of a pan-PI3K inhibitor prodrug reduces systemic on-target side effects but effectively treats asthma and irreversible pulmonary fibrosis.


Subject(s)
Asthma/drug therapy , Benzene Derivatives/therapeutic use , Enzyme Inhibitors/therapeutic use , Esters/therapeutic use , Phosphoinositide-3 Kinase Inhibitors , Pulmonary Fibrosis/drug therapy , Administration, Inhalation , Animals , Asthma/chemically induced , Asthma/pathology , Benzene Derivatives/administration & dosage , Bleomycin/toxicity , Disease Models, Animal , Enzyme Inhibitors/administration & dosage , Esters/administration & dosage , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Ovalbumin/toxicity , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology
5.
Sci Rep ; 8(1): 7254, 2018 05 08.
Article in English | MEDLINE | ID: mdl-29740022

ABSTRACT

The small-GTPase Rac1 is a key molecular regulator linking extracellular signals to actin cytoskeleton dynamics. Loss-of-function mutations in RAC1 and other genes of the Rac signaling pathway have been implicated in the pathogenesis of Intellectual Disability (ID). The Rac1 activity is negatively controlled by GAP proteins, however the effect of Rac1 hyperactivity on neuronal networking in vivo has been poorly studied. ArhGAP15 is a Rac-specific negative regulator, expressed in the main subtypes of pyramidal cortical neurons. In the absence of ArhGAP15, cortical pyramidal neurons show defective neuritogenesis, delayed axonal elongation, reduced dendritic branching, both in vitro and in vivo. These phenotypes are associated with altered actin dynamics at the growth cone due to increased activity of the PAK-LIMK pathway and hyperphosphorylation of ADF/cofilin. These results can be explained by shootin1 hypo-phosphorylation and uncoupling with the adhesion system. Functionally, ArhGAP15-/- mice exhibit decreased synaptic density, altered electroencephalographic rhythms and cognitive deficits. These data suggest that both hypo- and hyperactivation of the Rac pathway due to mutations in Rac1 regulators can result in conditions of ID, and that a tight regulation of Rac1 activity is required to attain the full complexity of the cortical networks.


Subject(s)
Dendrites/genetics , Neurites/physiology , Neuropeptides/genetics , Pyramidal Cells/physiology , rac1 GTP-Binding Protein/genetics , Actins/genetics , Actins/metabolism , Animals , Axons/metabolism , GTPase-Activating Proteins/genetics , Growth Cones/metabolism , Loss of Function Mutation/genetics , Mice , Neurites/metabolism , Phosphorylation , Pyramidal Cells/metabolism , Signal Transduction/genetics
6.
Food Chem ; 247: 56-65, 2018 May 01.
Article in English | MEDLINE | ID: mdl-29277228

ABSTRACT

Fragaria x ananassa Duch., popularly called strawberry, is known for its worldwide consumption and important biological activities, and these effects are related to its high concentration of anthocyanins. Pelargonidin-3-O-glucoside (P3G) is a major anthocyanin found in strawberry, and was evaluated for its anti-inflammatory action in experimental models. The effect of strawberry extract and P3G, on leukocyte migration, exudation levels and many inflammatory mediators, was therefore evaluated in an in vivo model. An in vitro study was also carried out to characterize the effect of P3G on mitogen-activated protein kinases, and on nuclear transcript factors NF-κB and AP-1. The results revealed that the strawberry and P3G have important anti-inflammatory proprieties, and the anti-inflammatory mechanism of P3G involves the arrest of IkB-α activation and reduction in JNKMAPK phosphorylation. The results reinforce that strawberry fruits are functional foods that can act as an adjuvant in the treatment of inflammatory conditions.


Subject(s)
Anthocyanins/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Fragaria/chemistry , Adenosine Deaminase/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Cell Movement/drug effects , Female , Fruit/chemistry , Leukocytes/drug effects , Mice , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Peroxidase/metabolism , Phosphorylation/drug effects , Plant Extracts/chemistry , Plant Extracts/pharmacology , Pleurisy/drug therapy , Transcription Factor AP-1/metabolism
7.
Cancer Cell ; 32(4): 444-459.e7, 2017 10 09.
Article in English | MEDLINE | ID: mdl-29017056

ABSTRACT

Proper organization of the mitotic spindle is key to genetic stability, but molecular components of inter-microtubule bridges that crosslink kinetochore fibers (K-fibers) are still largely unknown. Here we identify a kinase-independent function of class II phosphoinositide 3-OH kinase α (PI3K-C2α) acting as limiting scaffold protein organizing clathrin and TACC3 complex crosslinking K-fibers. Downregulation of PI3K-C2α causes spindle alterations, delayed anaphase onset, and aneuploidy, indicating that PI3K-C2α expression is required for genomic stability. Reduced abundance of PI3K-C2α in breast cancer models initially impairs tumor growth but later leads to the convergent evolution of fast-growing clones with mitotic checkpoint defects. As a consequence of altered spindle, loss of PI3K-C2α increases sensitivity to taxane-based therapy in pre-clinical models and in neoadjuvant settings.


Subject(s)
Breast Neoplasms/pathology , Genomic Instability , Phosphatidylinositol 3-Kinases/physiology , Spindle Apparatus/physiology , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Cycle Proteins/physiology , Cell Proliferation , Humans , MCF-7 Cells , Mad2 Proteins/physiology , Mice , Microtubule-Associated Proteins/physiology , Nuclear Proteins/physiology , Taxoids/therapeutic use
8.
ChemMedChem ; 12(18): 1542-1554, 2017 09 21.
Article in English | MEDLINE | ID: mdl-28857471

ABSTRACT

Activation of the phosphoinositide 3-kinase (PI3K) pathway is a key signaling event in cancer, inflammation, and other proliferative diseases. PI3K inhibitors are already approved for some specific clinical indications, but their systemic on-target toxicity limits their larger use. In particular, whereas toxicity is tolerable in acute treatment of life-threatening diseases, this is less acceptable in chronic conditions. In the past, the strategy to overcome this drawback was to block selected isoforms mainly expressed in leukocytes, but redundancy within the PI3K family members challenges the effectiveness of this approach. On the other hand, decreasing exposure to selected target cells represents a so-far unexplored alternative to circumvent systemic toxicity. In this manuscript, we describe the generation of a library of triazolylquinolones and the development of the first prodrug pan-PI3K inhibitor.


Subject(s)
Carboxylic Acids/chemistry , Enzyme Inhibitors/chemistry , Phosphoinositide-3 Kinase Inhibitors , Prodrugs/chemistry , Animals , Binding Sites , Carboxylic Acids/metabolism , Carboxylic Acids/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Design , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Hydrogen Bonding , Inhibitory Concentration 50 , Mice , Microsomes/metabolism , Molecular Dynamics Simulation , Phosphatidylinositol 3-Kinases/metabolism , Prodrugs/metabolism , Prodrugs/pharmacology , Protein Binding , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Quinolones/chemistry , Quinolones/metabolism , Quinolones/pharmacology , Structure-Activity Relationship
9.
Sci Signal ; 9(459): ra124, 2016 12 20.
Article in English | MEDLINE | ID: mdl-27999173

ABSTRACT

Mobilization of neutrophils from the bone marrow determines neutrophil blood counts and thus is medically important. Balanced neutrophil mobilization from the bone marrow depends on the retention-promoting chemokine CXCL12 and its receptor CXCR4 and the egression-promoting chemokine CXCL2 and its receptor CXCR2. Both pathways activate the small guanosine triphosphatase Rac, leaving the role of this signaling event in neutrophil retention and egression ambiguous. On the assumption that active Rac determines persistent directional cell migration, we generated a mathematical model to link chemokine-mediated Rac modulation to neutrophil egression time. Our computer simulation indicated that, in the bone marrow, where the retention signal predominated, egression time strictly depended on the time it took Rac to return to its basal activity (namely, adaptation). This prediction was validated in mice lacking the Rac inhibitor ArhGAP15. Neutrophils in these mice showed prolonged Rac adaptation and cell-autonomous retention in the bone marrow. Our model thus demonstrates that mobilization in the presence of two spatially defined opposing chemotactic cues strictly depends on inhibitors shaping the time course of signal adaptation. Furthermore, our findings might help to find new modes of intervention to treat conditions characterized by excessively low or high circulating neutrophils.


Subject(s)
Bone Marrow/enzymology , Neutrophils/enzymology , Signal Transduction/physiology , rac GTP-Binding Proteins/metabolism , Animals , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Mice , Mice, Knockout , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , rac GTP-Binding Proteins/genetics
10.
Sci Rep ; 6: 34877, 2016 10 07.
Article in English | MEDLINE | ID: mdl-27713499

ABSTRACT

During brain development, the small GTPases Rac1/Rac3 play key roles in neuronal migration, neuritogenesis, synaptic formation and plasticity, via control of actin cytoskeleton dynamic. Their activity is positively and negatively regulated by GEFs and GAPs molecules, respectively. However their in vivo roles are poorly known. The ArhGAP15 gene, coding for a Rac-specific GAP protein, is expressed in both excitatory and inhibitory neurons of the adult hippocampus, and its loss results in the hyperactivation of Rac1/Rac3. In the CA3 and dentate gyrus (DG) regions of the ArhGAP15 mutant hippocampus the CR+, PV+ and SST+ inhibitory neurons are reduced in number, due to reduced efficiency and directionality of their migration, while pyramidal neurons are unaffected. Loss of ArhGAP15 alters neuritogenesis and the balance between excitatory and inhibitory synapses, with a net functional result consisting in increased spike frequency and bursts, accompanied by poor synchronization. Thus, the loss of ArhGAP15 mainly impacts on interneuron-dependent inhibition. Adult ArhGAP15-/- mice showed defective hippocampus-dependent functions such as working and associative memories. These findings indicate that a normal architecture and function of hippocampal inhibitory neurons is essential for higher hippocampal functions, and is exquisitely sensitive to ArhGAP15-dependent modulation of Rac1/Rac3.


Subject(s)
Cognition Disorders/genetics , GTPase-Activating Proteins/metabolism , Hippocampus/physiopathology , Neurons/physiology , Neuropeptides/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Behavior, Animal/physiology , Cell Movement/genetics , Cells, Cultured , Cognition Disorders/etiology , Female , GTPase-Activating Proteins/genetics , Gene Expression Regulation, Developmental , Hippocampus/pathology , Interneurons/pathology , Male , Memory, Short-Term/physiology , Mice, Mutant Strains , Neurons/pathology , Neuropeptides/genetics , Rats , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/genetics
11.
Molecules ; 20(9): 17275-87, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26393561

ABSTRACT

A novel series of 4-aryl-3-cyano-2-(3-hydroxyphenyl)-6-morpholino-pyridines have been designed as potential phosphatidylinositol-3-kinase (PI3K) inhibitors. The compounds have been synthesized using the Guareschi reaction to prepare the key 4-aryl-3-cyano-2,6-dihydroxypyridine intermediate. A different selectivity according to the nature of the aryl group has been observed. Compound 9b is a selective inhibitor against the PI3Kα isoform, maintaining a good inhibitory activity. Docking studies were also performed in order to rationalize its profile of selectivity.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Phosphoinositide-3 Kinase Inhibitors , Pyridines/chemical synthesis , Animals , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Mice , Models, Molecular , Molecular Docking Simulation , NIH 3T3 Cells , Pyridines/chemistry , Pyridines/pharmacology , Structure-Activity Relationship
12.
Mediators Inflamm ; 2015: 545417, 2015.
Article in English | MEDLINE | ID: mdl-26185363

ABSTRACT

PURPOSE: Neutrophil-dominated airway inflammation is a key feature of progressive lung damage in cystic fibrosis (CF). Thus, reducing airway inflammation is a major goal to prevent lung damage in CF. However, current anti-inflammatory drugs have shown several limits. PI3Kγ plays a pivotal role in leukocyte recruitment and activation; in the present study we determined the effects of genetic deletion and pharmacologic inhibition of PI3Kγ on airway inflammation and structural lung damage in a mouse model of CF lung disease. METHODS: ßENaC overexpressing mice (ßENaC-Tg) were backcrossed with PI3Kγ-deficient (PI3Kγ (KO)) mice. Tissue damage was assessed by histology and morphometry and inflammatory cell number was evaluated in bronchoalveolar lavage fluid (BALF). Furthermore, we assessed the effect of a specific PI3Kγ inhibitor (AS-605240) on inflammatory cell number in BALF. RESULTS: Genetic deletion of PI3Kγ decreased neutrophil numbers in BALF of PI3Kγ (KO)/ßENaC-Tg mice, and this was associated with reduced emphysematous changes. Treatment with the PI3Kγ inhibitor AS-605240 decreased the number of neutrophils in BALF of ßENaC-Tg mice, reproducing the effect observed with genetic deletion of the enzyme. CONCLUSIONS: These results demonstrate the biological efficacy of both genetic deletion and pharmacological inhibition of PI3Kγ in reducing chronic neutrophilic inflammation in CF-like lung disease in vivo.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/physiology , Cystic Fibrosis/therapy , Inflammation/prevention & control , Lung/pathology , Neutrophil Infiltration , Animals , Class Ib Phosphatidylinositol 3-Kinase/genetics , Cystic Fibrosis/complications , Cystic Fibrosis/pathology , Epithelial Sodium Channels/physiology , Gene Deletion , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphoinositide-3 Kinase Inhibitors
13.
Nat Commun ; 6: 7400, 2015 Jun 23.
Article in English | MEDLINE | ID: mdl-26100075

ABSTRACT

In the liver, insulin-mediated activation of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway is at the core of metabolic control. Multiple PI3K and Akt isoenzymes are found in hepatocytes and whether isoform-selective interplays exist is currently unclear. Here we report that insulin signalling triggers the association of the liver-specific class II PI3K isoform γ (PI3K-C2γ) with Rab5-GTP, and its recruitment to Rab5-positive early endosomes. In these vesicles, PI3K-C2γ produces a phosphatidylinositol-3,4-bisphosphate pool specifically required for delayed and sustained endosomal Akt2 stimulation. Accordingly, loss of PI3K-C2γ does not affect insulin-dependent Akt1 activation as well as S6K and FoxO1-3 phosphorylation, but selectively reduces Akt2 activation, which specifically inhibits glycogen synthase activity. As a consequence, PI3K-C2γ-deficient mice display severely reduced liver accumulation of glycogen and develop hyperlipidemia, adiposity as well as insulin resistance with age or after consumption of a high-fat diet. Our data indicate PI3K-C2γ supports an isoenzyme-specific forking of insulin-mediated signal transduction to an endosomal pool of Akt2, required for glucose homeostasis.


Subject(s)
Aging/genetics , Glycogen/metabolism , Hepatocytes/metabolism , Insulin/metabolism , Liver/metabolism , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , rab5 GTP-Binding Proteins/metabolism , Adiposity/genetics , Animals , Diet, High-Fat , Endosomes/metabolism , Forkhead Transcription Factors/metabolism , Glucose/metabolism , Glycogen Synthase/metabolism , Homeostasis , Hyperlipidemias/genetics , Insulin Resistance/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol Phosphates/metabolism , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction
14.
Small GTPases ; 6(2): 71-80, 2015.
Article in English | MEDLINE | ID: mdl-25942647

ABSTRACT

Rac and PI3Ks are intracellular signal transducers able to regulate multiple signaling pathways fundamental for cell behavior. PI3Ks are lipid kinases that produce phosphorylated lipids which, in turn, transduce extracellular cues within the cell, while Rac is a small G protein that impacts on actin organization. Compelling evidence indicates that in multiple circumstances the 2 signaling pathways appear intermingled. For instance, phosphorylated lipids produced by PI3Ks recruit and activate GEF and GAP proteins, key modulators of Rac function. Conversely, PI3Ks interact with activated Rac, leading to Rac signaling amplification. This review summarizes the molecular mechanisms underlying the cross-talk between Rac and PI3K signaling in 2 different processes, cell migration and ROS production.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Second Messenger Systems , rac GTP-Binding Proteins/metabolism , Animals , Humans , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/genetics , rac GTP-Binding Proteins/chemistry , rac GTP-Binding Proteins/genetics
15.
Methods Enzymol ; 543: 115-40, 2014.
Article in English | MEDLINE | ID: mdl-24924130

ABSTRACT

Phosphoinositide-3-kinase (PI3K) signaling has been implicated in a panoply of cellular responses including survival, proliferation, protein synthesis, migration, and vesicular trafficking. In addition, alterations in the enzymatic activity of PI3Ks have been involved in the pathogenesis of multiple diseases, ranging from cancer to chronic inflammation. The emerging interest in PI3K as a pharmacological target has prompted the development of several molecules with inhibitory activity. In this context, the quantification of the second messenger generated by PI3Ks, phosphoinositide-3-phosphate, offers an opportunity to directly test variations in the lipid kinase activity of PI3K in physiological as well as pathological conditions. Here, we will describe common methods to measure the lipid kinase activity of PI3K in vitro and new techniques to follow the production of phosphoinositide-3-phosphate in vivo. These methods are relevant to study the alterations of the PI3K systems at the interface between signaling and oncometabolism.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Animals , Chromatography, Thin Layer , Enzyme Inhibitors/pharmacology , Humans , Immunoprecipitation , Mice , NIH 3T3 Cells , Phosphoinositide-3 Kinase Inhibitors
16.
Dev Cell ; 28(6): 647-58, 2014 Mar 31.
Article in English | MEDLINE | ID: mdl-24697898

ABSTRACT

Multiple phosphatidylinositol (PtdIns) 3-kinases (PI3Ks) can produce PtdIns3P to control endocytic trafficking, but whether enzyme specialization occurs in defined subcellular locations is unclear. Here, we report that PI3K-C2α is enriched in the pericentriolar recycling endocytic compartment (PRE) at the base of the primary cilium, where it regulates production of a specific pool of PtdIns3P. Loss of PI3K-C2α-derived PtdIns3P leads to mislocalization of PRE markers such as TfR and Rab11, reduces Rab11 activation, and blocks accumulation of Rab8 at the primary cilium. These changes in turn cause defects in primary cilium elongation, Smo ciliary translocation, and Sonic Hedgehog (Shh) signaling and ultimately impair embryonic development. Selective reconstitution of PtdIns3P levels in cells lacking PI3K-C2α rescues Rab11 activation, primary cilium length, and Shh pathway induction. Thus, PI3K-C2α regulates the formation of a PtdIns3P pool at the PRE required for Rab11 and Shh pathway activation.


Subject(s)
Cell Movement/physiology , Cilia/physiology , Endosomes/metabolism , Phosphatidylinositol 3-Kinases/physiology , Phosphatidylinositol Phosphates/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Immunoblotting , Immunoprecipitation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoinositide-3 Kinase Inhibitors , Protein Transport , RNA, Small Interfering/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Transferrin/metabolism , Signal Transduction , Smoothened Receptor
17.
Front Oncol ; 3: 108, 2013.
Article in English | MEDLINE | ID: mdl-23658859

ABSTRACT

The phosphatidylinositol 3-kinase (PI3K) signaling pathway regulates several cellular processes and it's one of the most frequently deregulated pathway in human tumors. Given its prominent role in cancer, there is great interest in the development of inhibitors able to target several members of PI3K signaling pathway in clinical trials. These drug candidates include PI3K inhibitors, both pan- and isoform-specific inhibitors, AKT, mTOR, and dual PI3K/mTOR inhibitors. As novel compounds progress into clinical trials, it's becoming urgent to identify and select patient population that most likely benefit from PI3K inhibition. In this review we will discuss individual PIK3CA mutations as predictors of sensitivity and resistance to targeted therapies, leading to use of novel PI3K/mTOR/AKT inhibitors to a more "personalized" treatment.

18.
Blood ; 119(3): 847-56, 2012 Jan 19.
Article in English | MEDLINE | ID: mdl-22106345

ABSTRACT

Integrin α2ß1-mediated adhesion of human platelets to monomeric type I collagen or to the GFOGER peptide caused a time-dependent activation of PI3K and Akt phosphorylation. This process was abrogated by pharmacologic inhibition of PI3Kß, but not of PI3Kγ or PI3Kα. Moreover, Akt phosphorylation was undetectable in murine platelets expressing a kinase-dead mutant of PI3Kß (PI3Kß(KD)), but occurred normally in PI3Kγ(KD) platelets. Integrin α2ß1 failed to stimulate PI3Kß in platelets from phospholipase Cγ2 (PLCγ2)-knockout mice, and we found that intracellular Ca(2+) linked PLCγ2 to PI3Kß activation. Integrin α2ß1 also caused a time-dependent stimulation of the focal kinase Pyk2 downstream of PLCγ2 and intracellular Ca(2+). Whereas activation of Pyk2 occurred normally in PI3Kß(KD) platelets, stimulation of PI3Kß was strongly reduced in Pyk2-knockout mice. Neither Pyk2 nor PI3Kß was required for α2ß1-mediated adhesion and spreading. However, activation of Rap1b and inside-out stimulation of integrin αIIbß3 were reduced after inhibition of PI3Kß and were significantly impaired in Pyk2-deficient platelets. Finally, both PI3Kß and Pyk2 significantly contributed to thrombus formation under flow. These results demonstrate that Pyk2 regulates PI3Kß downstream of integrin α2ß1, and document a novel role for Pyk2 and PI3Kß in integrin α2ß1 promoted inside-out activation of integrin αIIbß3 and thrombus formation.


Subject(s)
Blood Platelets/metabolism , Focal Adhesion Kinase 2/physiology , Integrin alpha2beta1/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Platelet Adhesiveness , Proto-Oncogene Proteins c-akt/metabolism , Animals , Calcium/metabolism , Collagen/metabolism , Fibrinogen/metabolism , Humans , Immunoblotting , Mice , Mice, Knockout , Phosphorylation , Platelet Aggregation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Signal Transduction
19.
Cardiovasc Res ; 93(1): 200-9, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22038741

ABSTRACT

AIMS: The lipid and protein kinase phosphoinositide 3-kinase γ (PI3Kγ) is abundantly expressed in inflammatory cells and in the cardiovascular tissue. In recent years, its role in inflammation and in cardiac function and remodelling has been unravelled, highlighting the beneficial effects of its pharmacological inhibition. Furthermore, a role for PI3Kγ in the regulation of vascular tone has been emphasized. However, the impact of this signalling in the control of blood pressure is still poorly understood. Our study investigated the effect of a selective inhibition of PI3Kγ, obtained by using two independent small molecules, on blood pressure. Moreover, we dissected the molecular mechanisms involved in control of contraction of resistance arteries by PI3Kγ. METHODS AND RESULTS: We showed that inhibition of PI3Kγ reduced blood pressure in normotensive and hypertensive mice in a concentration-dependent fashion. This effect was dependent on enhanced vasodilatation, documented in vivo by decreased peripheral vascular resistance, and ex vivo by vasorelaxing effects on isolated resistance vessels. The vasorelaxation induced by PI3Kγ inhibition relied on blunted pressure-induced Akt phosphorylation and a myogenic contractile response. Molecular insights revealed that PI3Kγ inhibition affected smooth muscle L-type calcium channel current density and calcium influx by impairing plasma membrane translocation of the α1C L-type calcium channel subunit responsible for channel open-state probability. CONCLUSION: Overall our findings suggest that PI3Kγ inhibition could be a novel tool to modulate calcium influx in vascular smooth muscle cells, thus relaxing resistance arteries and lowering blood pressure.


Subject(s)
Blood Pressure/physiology , Calcium Channels, L-Type/physiology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/physiology , Animals , Blood Pressure/drug effects , Calcium Signaling/drug effects , Class Ib Phosphatidylinositol 3-Kinase/genetics , Enzyme Inhibitors/pharmacology , Hypertension/drug therapy , Hypertension/physiopathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Quinoxalines/pharmacology , Thiazolidinediones/pharmacology , Vasodilation/drug effects , Vasodilation/physiology
20.
Methods Mol Biol ; 795: 55-67, 2012.
Article in English | MEDLINE | ID: mdl-21960215

ABSTRACT

Class IA phosphoinositide-3 kinases (PI3Ks) signaling has recently emerged as a key element in cancer development because of its ability to trigger a complex panoply of cellular responses controlling survival and proliferation. Many cancers show inappropriately activated PI3K pathway, and tumors with high PI3K activity are frequently resistant to traditional chemotherapy. Indeed, preclinical studies demonstrated a prominent role for the PI3K pathway in cancer cell survival and growth, thus validating PI3K as a potential drug target in cancer. The emerging interest in inhibiting PI3Ks in cancer have prompted the aggressive development of new selective PI3K pathway inhibitors as cancer therapy, and many of these molecules are currently in early-phase clinical trials. In this chapter, we describe methods to measure the PI3K lipid kinase activity in vitro, which is the standard procedure to test the efficacy of inhibitors.


Subject(s)
Enzyme Assays/methods , Phosphatidylinositol 3-Kinases/metabolism , Animals , Antibodies, Monoclonal/metabolism , Chromatography, Thin Layer , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Immunoprecipitation , Mice , NIH 3T3 Cells , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...