Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Cells ; 11(16)2022 08 11.
Article in English | MEDLINE | ID: mdl-36010578

ABSTRACT

Photodynamic therapy (PDT) is a promising anticancer strategy based on the light energy stimulation of photosensitizers (PS) molecules within a malignant cell. Among a multitude of recently challenged PS, Rose bengal (RB) has been already reported as an inducer of cytotoxicity in different tumor cells. However, RB displays a low penetration capability across cell membranes. We have therefore developed a short-term amino acids starvation protocol that significantly increases RB uptake in human astrocytoma cells compared to normal rat astrocytes. Following induced starvation uptake, RB is released outside cells by the exocytosis of extracellular vesicles (EVs). Thus, we have introduced a specific pharmacological treatment, based on the GW4869 exosomes inhibitor, to interfere with RB extracellular release. These combined treatments allow significantly reduced nanomolar amounts of administered RB and a decrease in the time interval required for PDT stimulation. The overall conditions affected astrocytoma viability through the activation of apoptotic pathways. In conclusion, we have developed for the first time a combined scheme to simultaneously increase the RB uptake in human astrocytoma cells, reduce the extracellular release of the drug by EVs, and improve the effectiveness of PDT-based treatments. Importantly, this strategy might be a valuable approach to efficiently deliver other PS or chemotherapeutic drugs in tumor cells.


Subject(s)
Astrocytoma , Exosomes , Photochemotherapy , Amino Acids , Animals , Astrocytoma/drug therapy , Humans , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Rats , Rose Bengal/chemistry , Rose Bengal/pharmacology
2.
J Pers Med ; 11(10)2021 Sep 22.
Article in English | MEDLINE | ID: mdl-34683083

ABSTRACT

Photodynamic therapy (PDT) has recently attracted interest as an innovative and adjuvant treatment for different cancers including malignant gliomas. Among these, Glioblastoma (GBM) is the most prevalent neoplasm in the central nervous system. Despite conventional therapeutic approaches that include surgical removal, radiation, and chemotherapy, GBM is characterized by an extremely poor prognosis and a high rate of recurrence. PDT is a physical process that induces tumor cell death through the genesis and accumulation of reactive oxygen species (ROS) produced by light energy interaction with a photosensitizing agent. In this contribution, we explored the potentiality of the plant alkaloid berberine (BBR) as a photosensitizing and cytotoxic agent coupled with a PDT scheme using a blue light source in human established astrocytoma cell lines. Our data mainly indicated for the combined BBR-PDT scheme a potent activation of the apoptosis pathway, through a massive ROS production, a great extent of mitochondria depolarization, and the sub-sequent activation of caspases. Altogether, these results demonstrated that BBR is an efficient photosensitizer agent and that its association with PDT may be a potential anticancer strategy for high malignant gliomas.

3.
Cells ; 9(7)2020 07 06.
Article in English | MEDLINE | ID: mdl-32640653

ABSTRACT

Extracellular vesicles (EVs) are considered as promising nanoparticle theranostic tools in many pathological contexts. The increasing clinical employment of therapeutic nanoparticles is contributing to the development of a new research area related to the design of artificial EVs. To this aim, different approaches have been described to develop mimetic biologically functional nanovescicles. In this paper, we suggest a simplified procedure to generate plasma membrane-derived nanovesicles with the possibility to efficiently encapsulate different drugs during their spontaneously assembly. After physical and molecular characterization by Tunable Resistive Pulse Sensing (TRPS) technology, transmission electron microscopy, and flow cytometry, as a proof of principle, we have loaded into mimetic EVs the isoquinoline alkaloid Berberine chloride and the chemotherapy compounds Temozolomide or Givinostat. We demonstrated the fully functionality of these nanoparticles in drug encapsulation and cell delivery, showing, in particular, a similar cytotoxic effect of direct cell culture administration of the anticancer drugs. In conclusion, we have documented the possibility to easily generate scalable nanovesicles with specific therapeutic cargo modifications useful in different drug delivery contexts.


Subject(s)
Membranes, Artificial , Nanoparticles/chemistry , Drug Delivery Systems/methods , Extracellular Vesicles/chemistry , Nanomedicine/methods
4.
Cells ; 9(5)2020 05 18.
Article in English | MEDLINE | ID: mdl-32443613

ABSTRACT

Adaptation of glioblastoma to caloric restriction induces compensatory changes in tumor metabolism that are incompletely known. Here we show that in human glioblastoma cells maintained in exhausted medium, SHC adaptor protein 3 (SHC3) increases due to down-regulation of SHC3 protein degradation. This effect is reversed by glucose addition and is not present in normal astrocytes. Increased SHC3 levels are associated to increased glucose uptake mediated by changes in membrane trafficking of glucose transporters of the solute carrier 2A superfamily (GLUT/SLC2A). We found that the effects on vesicle trafficking are mediated by SHC3 interactions with adaptor protein complex 1 and 2 (AP), BMP-2-inducible protein kinase and a fraction of poly ADP-ribose polymerase 1 (PARP1) associated to vesicles containing GLUT/SLC2As. In glioblastoma cells, PARP1 inhibitor veliparib mimics glucose starvation in enhancing glucose uptake. Furthermore, cytosol extracted from glioblastoma cells inhibits PARP1 enzymatic activity in vitro while immunodepletion of SHC3 from the cytosol significantly relieves this inhibition. The identification of a new pathway controlling glucose uptake in high grade gliomas represents an opportunity for repositioning existing drugs and designing new ones.


Subject(s)
Adaptation, Physiological , Brain Neoplasms/pathology , Glioblastoma/pathology , Glucose/deficiency , Signal Transduction , Adaptation, Physiological/drug effects , Benzimidazoles/pharmacology , Brain Neoplasms/ultrastructure , Cell Line, Tumor , Endocytosis/drug effects , Glioblastoma/ultrastructure , Glucose Transporter Type 1/metabolism , Glycosylation/drug effects , Humans , Lactic Acid/biosynthesis , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Protein Binding/drug effects , Protein Domains , Protein Stability/drug effects , Protein Transport/drug effects , Signal Transduction/drug effects , Src Homology 2 Domain-Containing, Transforming Protein 3/chemistry , Src Homology 2 Domain-Containing, Transforming Protein 3/metabolism , Transport Vesicles/drug effects , Transport Vesicles/metabolism
5.
Nanomedicine ; 23: 102113, 2020 01.
Article in English | MEDLINE | ID: mdl-31669084

ABSTRACT

C109 is a potent but poorly soluble FtsZ inhibitor displaying promising activity against Burkholderia cenocepacia, a high-risk pathogen for cystic fibrosis (CF) sufferers. To harness C109 for inhalation, we developed nanocrystal-embedded dry powders for inhalation suspension consisting in C109 nanocrystals stabilized with D-α-tocopheryl polyethylene glycol 1000 succinate (TPGS) embedded in hydroxypropyl-ß-cyclodextrin (CD). The powders could be safely re-dispersed in water for in vitro aerosolization. Owing to the presence of a PEG shell, the rod shape and the peculiar aspect ratio, C109 nanocrystals were able to diffuse through artificial CF mucus. The promising technological features were completed by encouraging in vitro/in vivo effects. The formulations displayed no toxicity towards human bronchial epithelial cells and were active against planktonic and sessile B. cenocepacia strains. The efficacy of C109 nanosuspensions in combination with piperacillin was confirmed in a Galleria mellonella infection model, strengthening their potential for combined therapy of B. cenocepacia lung infections.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins/antagonists & inhibitors , Bronchi/microbiology , Burkholderia Infections/drug therapy , Burkholderia cenocepacia/growth & development , Cystic Fibrosis/drug therapy , Cytoskeletal Proteins/antagonists & inhibitors , Drug Delivery Systems , Epithelial Cells/microbiology , Nanoparticles , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Bronchi/metabolism , Bronchi/pathology , Burkholderia Infections/metabolism , Burkholderia Infections/pathology , Cell Line, Tumor , Cystic Fibrosis/metabolism , Cystic Fibrosis/microbiology , Cystic Fibrosis/pathology , Cytoskeletal Proteins/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Nanoparticles/chemistry , Nanoparticles/therapeutic use
6.
Cells ; 8(4)2019 04 12.
Article in English | MEDLINE | ID: mdl-31013754

ABSTRACT

Celiac disease (CD) is a chronic systemic autoimmune disorder that is triggered by the ingestion of gliadin peptides, the alcohol-soluble fraction of wheat gluten. These peptides, which play a key role in the immune response that underlies CD, spontaneously form aggregates and exert a direct toxic action on cells due to the increase in the reactive oxygen species (ROS) levels. Furthermore, peptic-tryptic digested gliadin peptides (PT-gliadin) lead to an impairment in the autophagy pathway in an in vitro model based on Caco-2 cells. Considering these premises, in this study we have analyzed different mTOR-independent inducers, reporting that the disaccharide trehalose, a mTOR-independent autophagy activator, rescued the autophagy flux in Caco-2 cells treated with digested gliadin, as well as improved cell viability. Moreover, trehalose administration to Caco-2 cells in presence of digested gliadin reduced the intracellular levels of these toxic peptides. Altogether, these results showed the beneficial effects of trehalose in a CD in vitro model as well as underlining autophagy as a molecular pathway whose modulation might be promising in counteracting PT-gliadin cytotoxicity.


Subject(s)
Celiac Disease/metabolism , Trehalose/pharmacology , Autophagy/drug effects , Caco-2 Cells , Celiac Disease/immunology , Cell Survival/drug effects , Gliadin/adverse effects , Gliadin/chemistry , Gliadin/toxicity , Glutens , HT29 Cells , Humans , Models, Biological , Peptides , Reactive Oxygen Species , Trehalose/metabolism , Triticum/metabolism
7.
PLoS One ; 13(11): e0205967, 2018.
Article in English | MEDLINE | ID: mdl-30403761

ABSTRACT

The lack of direct neurophysiological recordings from the thalamus and the cortex hampers our understanding of vegetative state/unresponsive wakefulness syndrome and minimally conscious state in humans. We obtained microelectrode recordings from the thalami and the homolateral parietal cortex of two vegetative state/unresponsive wakefulness syndrome and one minimally conscious state patients during surgery for implantation of electrodes in both thalami for chronic deep brain stimulation. We found that activity of the thalamo-cortical networks differed among the two conditions. There were half the number of active neurons in the thalami of patients in vegetative state/unresponsive wakefulness syndrome than in minimally conscious state. Coupling of thalamic neuron discharge with EEG phases also differed in the two conditions and thalamo-cortical cross-frequency coupling was limited to the minimally conscious state patient. When consciousness is physiologically or pharmacologically reversibly suspended there is a significant increase in bursting activity of the thalamic neurons. By contrast, in the thalami of our patients in both conditions fewer than 17% of the recorded neurons showed bursting activity. This indicates that these conditions differ from physiological suspension of consciousness and that increased thalamic inhibition is not prominent. Our findings, albeit obtained in a limited number of patients, unveil the neurophysiology of these conditions at single unit resolution and might be relevant for inspiring novel therapeutic options.


Subject(s)
Consciousness Disorders/diagnostic imaging , Parietal Lobe/diagnostic imaging , Thalamus/diagnostic imaging , Action Potentials/physiology , Consciousness Disorders/physiopathology , Electroencephalography , Humans , Microelectrodes , Neurons/physiology , Parietal Lobe/physiopathology , Persistent Vegetative State/diagnostic imaging , Persistent Vegetative State/physiopathology , Thalamus/physiopathology
8.
Article in English | MEDLINE | ID: mdl-30297366

ABSTRACT

To streamline the elucidation of antibacterial compounds' mechanism of action, comprehensive high-throughput assays interrogating multiple putative targets are necessary. However, current chemogenomic approaches for antibiotic target identification have not fully utilized the multiplexing potential of next-generation sequencing. Here, we used Illumina sequencing of transposon insertions to track the competitive fitness of a Burkholderia cenocepacia library containing essential gene knockdowns. Using this method, we characterized a novel benzothiadiazole derivative, 10126109 (C109), with antibacterial activity against B. cenocepacia, for which whole-genome sequencing of low-frequency spontaneous drug-resistant mutants had failed to identify the drug target. By combining the identification of hypersusceptible mutants and morphology screening, we show that C109 targets cell division. Furthermore, fluorescence microscopy of bacteria harboring green fluorescent protein (GFP) cell division protein fusions revealed that C109 prevents divisome formation by altering the localization of the essential cell division protein FtsZ. In agreement with this, C109 inhibited both the GTPase and polymerization activities of purified B. cenocepacia FtsZ. C109 displayed antibacterial activity against Gram-positive and Gram-negative cystic fibrosis pathogens, including Mycobacterium abscessus C109 effectively cleared B. cenocepacia infection in the Caenorhabditis elegans model and exhibited additive interactions with clinically relevant antibiotics. Hence, C109 is an enticing candidate for further drug development.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Burkholderia cenocepacia/genetics , Cytoskeletal Proteins/antagonists & inhibitors , Drug Evaluation, Preclinical/methods , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Burkholderia Infections/drug therapy , Burkholderia Infections/microbiology , Burkholderia cenocepacia/drug effects , Burkholderia cenocepacia/isolation & purification , Caenorhabditis elegans/microbiology , Cystic Fibrosis/microbiology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Gene Knockdown Techniques , Genes, Essential , Green Fluorescent Proteins/genetics , High-Throughput Nucleotide Sequencing , Humans , Microbial Sensitivity Tests , Mutation
9.
Int J Mol Sci ; 19(2)2018 Feb 23.
Article in English | MEDLINE | ID: mdl-29473905

ABSTRACT

Gliadin, the alcohol-soluble protein fraction of wheat, contains the factor toxic for celiac disease (CD), and its toxicity is not reduced by digestion with gastro-pancreatic enzymes. Importantly, it is proved that an innate immunity to gliadin plays a key role in the development of CD. The immune response induces epithelial stress and reprograms intraepithelial lymphocytes into natural killer (NK)-like cells, leading to enterocyte apoptosis and an increase in epithelium permeability. In this contribution, we have reported that in Caco-2 cells the administration of enzymatically digested gliadin (PT-gliadin) reduced significantly the expression of the autophagy-related marker LC3-II. Furthermore, electron and fluorescent microscope analysis suggested a compromised functionality of the autophagosome apparatus. The rescue of the dysregulated autophagy process, along with a reduction of PT-gliadin toxicity, was obtained with a starvation induction protocol and by 3-methyladenine administration, while rapamycin, a well-known autophagy inducer, did not produce a significant improvement in the clearance of extra- and intra-cellular fluorescent PT-gliadin amount. Altogether, our results highlighted the possible contribution of the autophagy process in the degradation and in the reduction of extra-cellular release of gliadin peptides and suggest novel molecular targets to counteract gliadin-induced toxicity in CD.


Subject(s)
Autophagy/drug effects , Gliadin/pharmacology , Pepsin A/metabolism , Trypsin/metabolism , Apoptosis/drug effects , Caco-2 Cells , Cell Survival/drug effects , Endocytosis/drug effects , Fluorescence , Gliadin/ultrastructure , Humans , Microtubule-Associated Proteins/metabolism , Protein Aggregates/drug effects
10.
Neoplasia ; 19(4): 364-373, 2017 04.
Article in English | MEDLINE | ID: mdl-28319810

ABSTRACT

Glucose transport across glioblastoma membranes plays a crucial role in maintaining the enhanced glycolysis typical of high-grade gliomas and glioblastoma. We tested the ability of two inhibitors of the glucose transporters GLUT/SLC2A superfamily, indinavir (IDV) and ritonavir (RTV), and of one inhibitor of the Na/glucose antiporter type 2 (SGLT2/SLC5A2) superfamily, phlorizin (PHZ), in decreasing glucose consumption and cell proliferation of human and murine glioblastoma cells. We found in vitro that RTV, active on at least three different GLUT/SLC2A transporters, was more effective than IDV, a specific inhibitor of GLUT4/SLC2A4, both in decreasing glucose consumption and lactate production and in inhibiting growth of U87MG and Hu197 human glioblastoma cell lines and primary cultures of human glioblastoma. PHZ was inactive on the same cells. Similar results were obtained when cells were grown in adherence or as 3D multicellular tumor spheroids. RTV treatment but not IDV treatment induced AMP-activated protein kinase (AMPKα) phosphorylation that paralleled the decrease in glycolytic activity and cell growth. IDV, but not RTV, induced an increase in GLUT1/SLC2A1 whose activity could compensate for the inhibition of GLUT4/SLC2A4 by IDV. RTV and IDV pass poorly the blood brain barrier and are unlikely to reach sufficient liquoral concentrations in vivo to inhibit glioblastoma growth as single agents. Isobologram analysis of the association of RTV or IDV and 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) or 4-methyl-5-oxo-2,3,4,6,8-pentazabicyclo[4.3.0]nona-2,7,9-triene-9-carboxamide (TMZ) indicated synergy only with RTV on inhibition of glioblastoma cells. Finally, we tested in vivo the combination of RTV and BCNU on established GL261 tumors. This drug combination increased the overall survival and allowed a five-fold reduction in the dose of BCNU.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Carmustine/pharmacology , Dacarbazine/analogs & derivatives , Glucose Transport Proteins, Facilitative/antagonists & inhibitors , Animals , Biological Transport/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dacarbazine/pharmacology , Disease Models, Animal , Drug Synergism , Female , Glioma/diagnosis , Glioma/drug therapy , Glioma/metabolism , Glioma/mortality , Glucose/metabolism , Glucose Transport Proteins, Facilitative/metabolism , Humans , Mice , Temozolomide , Xenograft Model Antitumor Assays
11.
Sci Rep ; 6: 32487, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27580679

ABSTRACT

Burkholderia cenocepacia, an opportunistic respiratory pathogen particularly relevant for cystic fibrosis patients, is difficult to eradicate due to its high level of resistance to most clinically relevant antimicrobials. Consequently, the discovery of new antimicrobials as well as molecules capable of inhibiting its virulence is mandatory. In this regard quorum sensing (QS) represents a good target for anti-virulence therapies, as it has been linked to biofilm formation and is important for the production of several virulence factors, including proteases and siderophores. Here, we report the discovery of new diketopiperazine inhibitors of the B. cenocepacia acyl homoserine lactone synthase CepI, and report their anti-virulence properties. Out of ten different compounds assayed against recombinant CepI, four were effective inhibitors, with IC50 values in the micromolar range. The best compounds interfered with protease and siderophore production, as well as with biofilm formation, and showed good in vivo activity in a Caenorhabditis elegans infection model. These molecules were also tested in human cells and showed very low toxicity. Therefore, they could be considered for in vivo combined treatments with established or novel antimicrobials, to improve the current therapeutic strategies against B. cenocepacia.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Burkholderia cenocepacia/drug effects , Diketopiperazines/pharmacology , Enzyme Inhibitors/pharmacology , Ligases/antagonists & inhibitors , Quorum Sensing/drug effects , 4-Butyrolactone/analogs & derivatives , 4-Butyrolactone/antagonists & inhibitors , 4-Butyrolactone/biosynthesis , 4-Butyrolactone/genetics , Animals , Anti-Bacterial Agents/chemical synthesis , Biofilms/growth & development , Burkholderia cenocepacia/enzymology , Burkholderia cenocepacia/genetics , Burkholderia cenocepacia/pathogenicity , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/microbiology , Cell Survival/drug effects , Cloning, Molecular , Diketopiperazines/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HeLa Cells , Humans , Ligases/genetics , Ligases/metabolism , Quorum Sensing/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structure-Activity Relationship , Virulence
12.
J Neurooncol ; 120(2): 245-56, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25062668

ABSTRACT

Shc3 protein levels are high in human glioblastoma but they decrease in vitro. We found that SHC3 mRNA and protein increased when glioblastoma cells grew as multicellular tumor spheroid (MTS). Shc3 expression was also induced in adherent cultures by increasing cell density. Among the Shc family members, only Shc2 and Shc3 increased with cell density. Shc3 and focal adhesion kinase (Fak) interact as shown by co-immunoprecipitation. Inhibition of Fak activation reduced Shc3 increase and MTS formation and changed Shc3 phosphorylation pattern. Our results suggest that in gliomas cell density modulates Shc3 protein levels and its activity, at least in part, through Fak activation.


Subject(s)
Apoptosis , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Glioblastoma/pathology , Shc Signaling Adaptor Proteins/metabolism , Blotting, Western , Cell Count , Cell Proliferation , Focal Adhesion Protein-Tyrosine Kinases/genetics , Glioblastoma/genetics , Humans , Immunoprecipitation , Phosphorylation , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Shc Signaling Adaptor Proteins/genetics , Signal Transduction , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Src Homology 2 Domain-Containing, Transforming Protein 2 , Src Homology 2 Domain-Containing, Transforming Protein 3 , Tumor Cells, Cultured
13.
Autophagy ; 7(8): 840-53, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21478678

ABSTRACT

Malignant gliomas are the most common and lethal primary central nervous system neoplasms. Several intriguing lines of evidence have recently emerged indicating that the cellular prion protein (PrPC) may exert neuro- and cyto-protective functions: PrPC overexpression protects cultured neurons and also tumor cell lines exposed to various pro-apoptotic stimuli while, on the contrary, PrPC silencing sensitizes Adriamycin-resistant human breast carcinoma cells to TRAIL-mediated cell death. In order to determine if PrPC is involved in the resistance of glial tumors to cell death, the effects of cellular prion protein downregulation by antisense approach were investigated in different human malignant glioma cell lines. PrPC downregulation induced profound morphological changes and significant cell death. In addition, a significant tumor volume reduction was noted after PrPC silencing in a EGFP-GL261 glioma murine model. Investigations of the molecular effects induced by PrPC silencing were carried out on T98G human glioma cells by analysing autophagic as well as typical apoptotic markers (nuclear morphology, caspase-3/7, p53 and PARP-1). The results indicated that apoptosis was not induced after PrPC downregulation while, on the contrary, electron microscopy analysis, and an accumulation of GFP-LC3-II in autophagosomal membranes of GFP-LC3 transfected cells, indicated a predominant activation of autophagy. PrPC silencing also led to induction of LC3-II, increase in Beclin-1 and a concomitant decrease in p62, Bcl-2 and in the phosphorylation of 4E-BP1, a target of mTOR autophagy signaling. In conclusion, our results show for the first time that interfering with the cellular prion protein expression could modulate autophagy-dependent cell death pathways in glial tumor cells.


Subject(s)
Autophagy/drug effects , DNA/metabolism , Gene Silencing/drug effects , Glioma/pathology , Oligonucleotides, Antisense/pharmacology , PrPC Proteins/genetics , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Protein 7 , Beclin-1 , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Shape/drug effects , Cell Survival/drug effects , Cytoprotection/drug effects , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glioma/genetics , Glioma/ultrastructure , Humans , Membrane Proteins/metabolism , Mice , PrPC Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Ubiquitin-Activating Enzymes/metabolism
14.
J Biomed Biotechnol ; 2010: 301067, 2010.
Article in English | MEDLINE | ID: mdl-20981146

ABSTRACT

Doppel (Dpl) is a membrane-bound glycoprotein mainly expressed in the testis of adult healthy people. It is generally absent in the central nervous system, but its coding gene sequence is ectopically expressed in astrocytoma specimens and in derived cell lines. In this paper, we investigated the expression and the biochemical features of Dpl in a panel of 49 astrocytoma specimens of different WHO malignancy grades. As a result, Dpl was expressed in the majority of the investigated specimens (86%), also including low grade samples. Importantly, Dpl exhibited different cellular localizations and altered glycan moieties composition, depending on the tumor grade. Most low-grade astrocytomas (83%) showed a membrane-bound Dpl, like human healthy testis tissue, whereas the majority of high-grade astrocytomas (75%) displayed a cytosolic Dpl. Deglycosylation studies with N-glycosidase F and/or neuraminidase highlighted defective glycan moieties and an unexpected loss of sialic acid. To find associations between glial tumor progression and Dpl biochemical features, predictive bioinformatics approaches were produced. In particular, Decision tree and Nomogram analysis showed well-defined Dpl-based criteria that separately clustered low-and high-grade astrocytomas. Taken together, these findings show that in astrocytomas, Dpl undergoes different molecular processes that might constitute additional helpful tools to characterize the glial tumor progression.


Subject(s)
Astrocytoma/pathology , Biomarkers, Tumor , Brain Neoplasms/pathology , Prions/chemistry , Prions/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Astrocytoma/metabolism , Blotting, Western , Brain Neoplasms/metabolism , Cell Membrane/metabolism , Child , Cluster Analysis , Cytoplasm/metabolism , Disease Progression , Female , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/isolation & purification , GPI-Linked Proteins/metabolism , Humans , Immunohistochemistry , Male , Middle Aged , N-Acetylneuraminic Acid/metabolism , Predictive Value of Tests , Prions/isolation & purification
15.
J Biomed Biotechnol ; 2010: 670125, 2010.
Article in English | MEDLINE | ID: mdl-20625507

ABSTRACT

Protein interactions are crucial in most biological processes. Several in silico methods have been recently developed to predict them. This paper describes a bioinformatics method that combines sequence similarity and structural information to support experimental studies on protein interactions. Given a target protein, the approach selects the most likely interactors among the candidates revealed by experimental techniques, but not yet in vivo validated. The sequence and the structural information of the in vivo confirmed proteins and complexes are exploited to evaluate the candidate interactors. Finally, a score is calculated to suggest the most likely interactors of the target protein. As an example, we searched for GRB2 interactors. We ranked a set of 46 candidate interactors by the presented method. These candidates were then reduced to 21, through a score threshold chosen by means of a cross-validation strategy. Among them, the isoform 1 of MAPK14 was in silico confirmed as a GRB2 interactor. Finally, given a set of already confirmed interactors of GRB2, the accuracy and the precision of the approach were 75% and 86%, respectively. In conclusion, the proposed method can be conveniently exploited to select the proteins to be experimentally investigated within a set of potential interactors.


Subject(s)
Computational Biology/methods , Protein Interaction Mapping/methods , Amino Acid Motifs , Databases, Protein , GRB2 Adaptor Protein/chemistry , GRB2 Adaptor Protein/metabolism , Humans , Hydrogen Bonding , Mitogen-Activated Protein Kinase 1/chemistry , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 14/chemistry , Mitogen-Activated Protein Kinase 14/metabolism , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Binding , Reproducibility of Results , Sequence Alignment
16.
J Biomed Biotechnol ; 2009: 924565, 2009.
Article in English | MEDLINE | ID: mdl-19657395

ABSTRACT

In this study the mRNA levels of five EGFR indirectly related genes, EGFR, HB-EGF, ADAM17, PTEN, and MMP9, have been assessed by Real-time PCR in a panel of 37 glioblastoma multiforme specimens and in 5 normal brain samples; as a result, in glioblastoma, ADAM17 and PTEN expression was significantly lower than in normal brain samples, and, in particular, a statistically significant inverse correlation was found between PTEN and MMP9 mRNA levels. To verify if this correlation was conserved in gliomas, PTEN and MMP9 expression was further investigated in an additional panel of 16 anaplastic astrocytoma specimens and, in parallel, in different human normal and astrocytic tumor cell lines. In anaplastic astrocytomas PTEN expression was significantly higher than in glioblastoma multiforme, but no significant correlation was found between PTEN and MMP9 expression. PTEN and MMP9 mRNA levels were also employed to identify subgroups of specimens within the different glioma malignancy grades and to define a gene expression-based diagnostic classification scheme. In conclusion, this gene expression survey highlighted that the combined measurement of PTEN and MMP9 transcripts might represent a novel reliable tool for the differential diagnosis of high-grade gliomas, and it also suggested a functional link involving these genes in glial tumors.


Subject(s)
Biomarkers, Tumor/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Glioma/diagnosis , Glioma/enzymology , Matrix Metalloproteinase 9/metabolism , PTEN Phosphohydrolase/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Female , Gene Expression Profiling , Glioma/genetics , Humans , Male , Matrix Metalloproteinase 9/genetics , Middle Aged , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , PTEN Phosphohydrolase/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Young Adult
17.
Cell Oncol ; 30(6): 491-501, 2008.
Article in English | MEDLINE | ID: mdl-18936526

ABSTRACT

Doppel (Dpl) protein is the paralogue of the cellular prion (PrP(C)) protein. In humans, Dpl is expressed almost exclusively in testis where it is involved in spermatogenesis. Recently, the protein has been described to be ectopically expressed in astrocytomas and its potential association to the brain tumor malignancy progression has been advanced. In this study, we aimed to investigate in vitro the potential involvement of Dpl in the tumor cell migration: to this purpose, Dpl expression was reduced in the IPDDC-A2 astrocytoma-derived cell line, by means of antisense and siRNA approaches; migration rates were then evaluated by means of a scratch wound healing assay. As a result, the cellular migration was sensibly reduced after Dpl silencing. Following a complementary approach, in HeLa cells, showing very low endogenous Dpl expression, the protein expression was induced by transfection and stabilization of an eukaryotic expression vector containing the doppel gene coding sequence. These stably Dpl-overexpressing cells revealed a significant increase in the migration rate, compared to untreated and control cells. In addition, Dpl-forced expression induced substantial changes in the cell morphology. Of note, in these cells, viability examination by means of tetrazolium-based assay did not reveal differences in the proliferation; on the contrary, a variation in density-dependent growth, leading to an increase of cell contact inhibition was highlighted. These results, in conclusion, might suggest a potential and functional role for Dpl in tumor cells migratory and morphological behaviours and address to future gene-targeted therapeutic interventions.


Subject(s)
Cell Movement/physiology , Prions/physiology , Astrocytoma/genetics , Astrocytoma/metabolism , Astrocytoma/pathology , Blotting, Western , Cell Line, Tumor , Cell Movement/genetics , GPI-Linked Proteins , HeLa Cells , Humans , Prions/genetics , Prions/metabolism , RNA, Small Interfering/genetics , Transfection
18.
Cell Oncol ; 30(4): 337-47, 2008.
Article in English | MEDLINE | ID: mdl-18607068

ABSTRACT

Doppel, a prion-like protein, is a GPI-membrane anchored protein generally not expressed in the Central Nervous System (CNS) of different mammalian species, including human. Nevertheless, in astrocytomas, a particular kind of glial tumors, the doppel encoding gene (PRND) is over-expressed and the corresponding protein product (Dpl) is ectopically localized in the cytoplasm of the tumor cells. In this study we have analysed the sub-cellular localization of Dpl using double-immunofluorescence staining and confocal microscopy examinations in two astrocytoma-derived human cell lines (IPDDC-A2 and D384-MG). Our results confirmed that Dpl is localized in the cytoplasm of the astrocytoma cells and indicated that it is mostly associated with Lamp-1 and Limp-2 positive lysosomal vesicles and, marginally, to the Golgi apparatus and other cellular organelles. Noticeably, none of the examined tumor cells showed a membrane-Dpl localization. The membrane-associated Dpl expression was restored after the transfection of the astrocytoma cells with mutated Dpl-expression vectors in its glycosylation sites. Additionally, Dpl showed altered expression and traffic using the acidotropic agent ammonium chloride, leading to the accumulation of Dpl in nascent exocytic vesicles. Altogether, these results indicated that in the astrocytic tumor cells Dpl has an altered biosynthetic trafficking, likely derived from abnormal post-translational processes: these modifications do not permit the localization of Dpl in correspondence of the plasma membrane and lead to its intracellular accumulation in the lysosomes. In these proteolytic compartments, the astrocytic tumor cells might provide to the degradation of the excess of a potentially cytotoxic Dpl product.


Subject(s)
Astrocytes/metabolism , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Oligodendroglioma/metabolism , Prions/metabolism , Adult , Ammonium Chloride/pharmacology , Astrocytes/ultrastructure , Brain Neoplasms/pathology , Cell Compartmentation , Cell Culture Techniques , Cell Membrane/metabolism , Cell Membrane/ultrastructure , GPI-Linked Proteins , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Glycosylation , Glycosylphosphatidylinositols/genetics , Glycosylphosphatidylinositols/metabolism , HeLa Cells , Humans , Lysosomes/metabolism , Lysosomes/ultrastructure , Male , Mutant Proteins/analysis , Mutant Proteins/genetics , Mutant Proteins/metabolism , Oligodendroglioma/pathology , Prions/analysis , Prions/genetics , Protein Transport , Recombinant Proteins/analysis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection
19.
Oncol Rep ; 16(6): 1325-32, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17089057

ABSTRACT

Doppel (Dpl) is a paralogue of the mammalian Prion (PrP) protein. It is abundant in testis and, unlike PrP, it is expressed at low levels in the adult central nervous system (CNS). Besides, Dpl overexpression correlates with some prion-disease pathological features, such as ataxia and death of cerebellar neurons. Recently, ectopic expression of doppel was found in two different tumor types, specifically in glial and haematological cancers. In this study the doppel gene (PRND) mRNA and protein expression in PRT-HU2 and IPDDC-A2 astrocytoma-derived cell lines was investigated. Northern blot analysis revealed two equally abundant PRND mRNA isoforms, while real-time PCR, on nuclear and cytoplasmic RNA fractions, and cRNA in situ hybridization, on astrocytoma cells and bioptical specimens, showed a nuclear retention of PRND transcripts. Western blot analysis showed that the amount of protein expressed is low compared to the level of mRNA. Moreover deglycosylation studies indicated that Dpl undergoes unusual glycosylation processes. Immunohistochemistry experiments demonstrated that Dpl was mainly localised in the cytoplasm of the astrocytic tumor cells, and that it failed to be GPI-anchored to the cell membrane. This unusual cellular localization was also confirmed through EGFP-Dpl expression in astrocytomas; on the contrary, HeLa cells exhibited the expected Dpl membrane localization. Our findings suggest an aberrant doppel gene expression pattern, characterized by a substantial nuclear retention of the transcript, an altered post-translational modification of the protein and an unusual cytoplasmic localization.


Subject(s)
Astrocytoma/metabolism , Brain Neoplasms/metabolism , Cell Nucleus/metabolism , Prions/biosynthesis , RNA, Messenger/metabolism , Astrocytoma/genetics , Blotting, Northern , Blotting, Western , Brain Neoplasms/genetics , Cell Line, Tumor , Cytoplasm/metabolism , GPI-Linked Proteins , Gene Expression , Gene Expression Profiling , Humans , Immunohistochemistry , Protein Biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Transfection
20.
Biochem Biophys Res Commun ; 346(1): 108-15, 2006 Jul 21.
Article in English | MEDLINE | ID: mdl-16750514

ABSTRACT

The cellular prion protein (PrP(C)) is a highly conserved protein throughout the evolution of mammals and therefore is thought to play important cellular functions. Despite decades of intensive researches, the physiological function of PrP(C) remains enigmatic. Differently, in particular pathological contexts, generally referred as transmissible spongiform encephalopathies, a conformational isoform of PrP(C), i.e., PrP(Sc), is considered the causative agent of these diseases. In this study, we investigated putative PrP(C) cellular functions through the identification of PrP(C) protein interactants. Using a bacterial two-hybrid approach, we identified a novel interaction between PrP(C) and a two-pore potassium channel protein, TREK-1. This interaction was further verified in transfected eukaryotic cells using co-immunoprecipitation and confocal microscopic analysis of the fluorescent transfected proteins. Importantly, in the cerebellar cortex, the endogenous PrP(C) and TREK-1 proteins exhibited co-localization signals in correspondence of the Purkinje cells. Furthermore, a deletion mapping study defined the carboxyl-terminal regions of the two proteins as the possible determinants of the PrP(C)-TREK-1 interaction. Our results indicated a novel PrP(C) interacting protein and suggested that this complex might be relevant in modulating a variety of electrophysiological-dependent cellular responses.


Subject(s)
Potassium Channels, Tandem Pore Domain/metabolism , PrPC Proteins/metabolism , Animals , Cerebellar Cortex/metabolism , HeLa Cells , Humans , Immunoprecipitation , Mice , Mice, Knockout , Protein Interaction Mapping , Rats , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...