Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
1.
J Dermatolog Treat ; 33(1): 401-407, 2022 Feb.
Article in English | MEDLINE | ID: mdl-32349568

ABSTRACT

BACKGROUND: Cyclosporine A (CsA) is one of the systemic therapeutic options for moderate-to-severe psoriasis, based on its efficacy and rapidity of action. The current study investigated the response to CsA in patients with moderate-to-severe plaque psoriasis. MATERIALS AND METHODS: TRANSITION was an observational, cross-sectional, multicentre study which evaluated the proportion of partial- and suboptimal-responders among patients with moderate-to-severe plaque psoriasis treated with continuous CsA for ≥12 weeks. Patients demonstrating a Psoriasis Area and Severity Index (PASI) response of ≥90, ≥75 and <90, ≥50 and <75 and <50 were defined as responders, suboptimal-responders, partial-responders, and non-responders, respectively. RESULTS: A total of 196 patients (mean age, 46.6 years; 62.8% males) from 14 sites in Italy were evaluated. At the study visit, the mean (SD) PASI score was 4.2(5.5) compared with 15.3(7.1) prior to the last CsA cycle. For response categories, 39.8%, 22.4%, 16.8%, and 20.9% of patients were responders, suboptimal-responders, partial-responders, and non-responders to CsA treatment. Overall, 28.6% of patients permanently discontinued treatment with CsA (lack of efficacy [10.2%], poor tolerability and voluntary discontinuation [3.6% each], and other [11.7%]). CONCLUSION: Patients were only partially satisfied with CsA treatment, reporting measurable impact on quality of life. Only 40% patients showed a satisfactory response to CsA.


Subject(s)
Cyclosporine , Psoriasis , Cross-Sectional Studies , Cyclosporine/therapeutic use , Female , Humans , Male , Middle Aged , Psoriasis/drug therapy , Quality of Life , Severity of Illness Index , Treatment Outcome
2.
Science ; 361(6403)2018 08 17.
Article in English | MEDLINE | ID: mdl-30115782

ABSTRACT

The coordinated expression of highly related homoeologous genes in polyploid species underlies the phenotypes of many of the world's major crops. Here we combine extensive gene expression datasets to produce a comprehensive, genome-wide analysis of homoeolog expression patterns in hexaploid bread wheat. Bias in homoeolog expression varies between tissues, with ~30% of wheat homoeologs showing nonbalanced expression. We found expression asymmetries along wheat chromosomes, with homoeologs showing the largest inter-tissue, inter-cultivar, and coding sequence variation, most often located in high-recombination distal ends of chromosomes. These transcriptionally dynamic genes potentially represent the first steps toward neo- or subfunctionalization of wheat homoeologs. Coexpression networks reveal extensive coordination of homoeologs throughout development and, alongside a detailed expression atlas, provide a framework to target candidate genes underpinning agronomic traits in wheat.


Subject(s)
Gene Expression Regulation, Plant , Polyploidy , Transcription, Genetic , Triticum/genetics , Bread , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genome, Plant , RNA, Plant/genetics , Sequence Analysis, RNA , Triticum/growth & development
4.
J Biol Regul Homeost Agents ; 27(1): 143-54, 2013.
Article in English | MEDLINE | ID: mdl-23489694

ABSTRACT

Glioblastoma multiforme (GBM) is among the most devastating human tumors being rapidly fatal despite aggressive surgery, radiation and chemotherapies. It is characterized by extensive dissemination of tumor cells within the brain that hinders complete surgical resection. GBM tumor initiating-cells (TICs) are a rare subpopulation of cells responsible for tumor development, growth, invasiveness and recurrence after chemotherapy. TICs from human GBM can be selected in vitro using the same conditions permissive for the growth of normal neural cells, of which share some features including marker expression, self-renewal capacity, long-term proliferation, and ability to differentiate into neuronal and glial cells. EGFR overexpression and its constitutive activation is one of the most important signaling alteration identified in GBM, and its pharmacological targeting represents an attractive therapeutic goal. We previously demonstrated that human GBM TICs have different sensitivity to the EGFR kinase inhibitors erlotinib and gefitinib, depending on the differential modulation of downstream signaling cascades. In this work we investigated the mechanisms of resistance to erlotinib in two human GBM TIC cultures, analyzing EGF and bFGF individual contribution to proliferation, clonogenicity, and migration. We demonstrated the presence of a small cell subpopulation whose proliferation is supported by EGF and a larger one mainly dependent on bFGF. Thus, insensitivity to EGFR kinase inhibitors as far as TIC proliferation results from a predominant FGFR activation that hides the inhibitory effects induced on EGFR signaling. Conversely, EGF and bFGF induced cell migration with similar efficacy. In addition, unlike neural stem/progenitors cells, the removal of chondroitin sulphate proteoglycans from cell surface was unable to discern EGF- and bFGF-dependent subpopulations in GBM TICs.


Subject(s)
Epidermal Growth Factor/pharmacology , ErbB Receptors/antagonists & inhibitors , Fibroblast Growth Factor 2/pharmacology , Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Protein Kinase Inhibitors/pharmacology , Aged , Cell Count , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Chondroitin Sulfates/metabolism , Clone Cells , Drug Screening Assays, Antitumor , ErbB Receptors/metabolism , Glioblastoma/metabolism , Humans , Male , Middle Aged , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Time Factors , Tumor Stem Cell Assay
5.
Oncogene ; 32(21): 2670-81, 2013 May 23.
Article in English | MEDLINE | ID: mdl-22797070

ABSTRACT

Amplification and rearrangements of the epidermal growth factor receptor (EGFR) gene are frequently found in glioblastoma multiforme (GBM). The most common variant is EGFR variant III (EGFRvIII). Research suggests that EGFRvIII could be a marker for a cancer stem cell or tumor-initiating population. If amplification and rearrangement are early events in tumorigenesis, this implies that they should be preserved throughout the tumor. However, in primary GBM, EGFRvIII expression is focal and sporadic. Unexpectedly, we found EGFR amplification and rearrangement throughout the tumor, including regions with no EGFRvIII expression, suggesting that mechanisms exist to modulate EGFRvIII expression even in the presence of high gene amplification. To study this phenomenon, we characterized three GBM cell lines with endogenous EGFRvIII. EGFRvIII expression was heterogeneous, with both positive and negative populations maintaining the genetic alterations, akin to primary tumors. Furthermore, EGFRvIII defined a hierarchy where EGFRvIII-positive cells gave rise to additional positive and negative cells. Only cells that had recently lost EGFRvIII expression could re-express EGFRvIII, providing an important buffer for maintaining EGFRvIII-positive cell numbers. Epigenetic mechanisms had a role in maintaining heterogeneous EGFRvIII expression. Demethylation induced a 20-60% increase in the percentage of EGFRvIII-positive cells, indicating that some cells could re-express EGFRvIII. Surprisingly, inhibition of histone deacetylation resulted in a 50-80% reduction in EGFRvIII expression. Collectively, this data demonstrates that EGFR amplification and rearrangement are early events in tumorigenesis and EGFRvIII follows a model of hierarchical expression. Furthermore, EGFRvIII expression is restricted by epigenetic mechanisms, suggesting that drugs that modulate the epigenome might be used successfully in glioblastoma tumors.


Subject(s)
Cell Transformation, Neoplastic , Epigenesis, Genetic , ErbB Receptors , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Gene Rearrangement , Glioblastoma , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Gene Amplification , Glioblastoma/enzymology , Glioblastoma/genetics , Glioblastoma/pathology , Humans
6.
Cell Death Dis ; 2: e138, 2011 Mar 31.
Article in English | MEDLINE | ID: mdl-21451573

ABSTRACT

To define the mechanisms by which hPrP90-231 induces cell death, we analyzed its interaction with living cells and monitored its intracellular fate. Treatment of SH-SY5Y cells with fluorescein-5-isothiocyanate (FITC)-conjugated hPrP90-231 caused the accumulation of cytosolic aggregates of the prion protein fragment that increased in number and size in a time-dependent manner. The formation of large intracellular hPrP90-231 aggregates correlated with the activation of apoptosis. hPrP90-231 aggregates occurred within lysotracker-positive vesicles and induced the formation of activated cathepsin D (CD), indicating that hPrP90-231 is partitioned into the endosomal-lysosomal system structures, activating the proteolytic machinery. Remarkably, the inhibition of CD activity significantly reduced hPrP-90-231-dependent apoptosis. Internalized hPrP90-231 forms detergent-insoluble and SDS-stable aggregates, displaying partial resistance to proteolysis. By confocal microscopy analysis of lucifer yellow (LY) intracellular partition, we show that hPrP90-231 accumulation induces lysosome destabilization and loss of lysosomal membrane impermeability. In fact, although control cells evidenced a vesicular pattern of LY fluorescence (index of healthy lysosomes), hPrP90-231-treated cells showed diffuse cytosolic fluorescence, indicating LY diffusion through damaged lysosomes. In conclusion, these data indicate that exogenously added hPrP90-231 forms intralysosomal deposits having features of insoluble, protease-resistant aggregates and could trigger a lysosome-mediated apoptosis by inducing lysosome membrane permeabilization, followed by the release of hydrolytic enzymes.


Subject(s)
Apoptosis , Endopeptidase K/metabolism , Lysosomes/metabolism , PrPC Proteins/chemistry , PrPC Proteins/metabolism , Prion Diseases/metabolism , Cell Death , Cell Line, Tumor , Cytosol/chemistry , Cytosol/metabolism , Humans , Lysosomes/chemistry , PrPC Proteins/genetics , Prion Diseases/genetics , Prion Diseases/physiopathology , Solubility
7.
G Ital Dermatol Venereol ; 145(3): 319-22, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20461041

ABSTRACT

AIM AND METHODS: The treatment of mild-moderate acne with topical drugs in association with appropriate cosmetics is currently the golden standard. The tolerability and efficacy of a cream formulated with a new mix of alpha-hydroxy acids (Hyseac AHA cream) in 248 patients with mild-moderate acne (comedonic, inflammatory, or mixed) have been investigated in a multicenter, non-randomized, open study by 10 dermatologists from different Italian areas during their routine practice. The medication with Hyseac AHA cream was prescribed at first consultation for 60 days, twice a day, either as a monotherapy (49.2% patients) or in association with a pharmacological treatment (50.2%). RESULTS: The tolerability was good to excellent in 92.3% patients, without significant differences between patients using AHA cream in monotherapy (90.0%) or associated with concomitant pharmacological treatment (97.6%). The efficacy was overall high in 64.2% patients, again without significant differences related to concomitant pharmacological treatment or not (64.8% vs. 63.3%) and/or the acne clinical type (comedonic vs. inflammatory vs. mixed: 69.2% vs. 66.7% vs. 58%). CONCLUSION: The results of this study, while confirming the high tolerability and efficacy of this AHA cream in the treatment of mild/moderate acne, reasonably suggest its possible use also in monotherapy. Furthermore, its use can be reasonably hypothesized as a maintenance treatment after specific pharmacological treatment even in more severe acne types.


Subject(s)
Acne Vulgaris/drug therapy , Hydroxy Acids/therapeutic use , Adolescent , Adult , Child , Female , Humans , Male , Middle Aged , Severity of Illness Index , Young Adult
8.
J Biol Regul Homeost Agents ; 24(1): 27-39, 2010.
Article in English | MEDLINE | ID: mdl-20385069

ABSTRACT

In this paper we analyzed the determinants and the structural effects of the interaction of human prion protein fragment 90-231 (HuPrP) with humic substances, (HS) including humic (HA) and fulvic (FA) acids, natural refractory organic polyanions widely diffused in soils and waters. We show that this interaction is mainly driven by non-specific electrostatic attraction involving regions situated within alpha-helix A and beta-sheet S1 of human PrP. FA binding to HuPrP altered its ability to acquire some PrPSc-like characteristics induced by the mild thermal denaturation of the peptide (1 h at 53 degrees C). In particular, in the presence of FA, HuPrP shows a reduced amount of beta-sheet content (as demonstrated by the reduced binding of thioflavin T), an increased sensitivity to protease K and an inhibition of the entering in the fibrillogenic pathway. FA/HuPrP interaction caused the aggregation of the peptide in unstructured macrocomplexes, as demonstrated by the altered electrophoretic migration in semi-denaturing detergent-agarose gel assay. Importantly, in the presence of FA the rate of internalization of HuPrP in human neuroblastoma cells was significantly reduced as compared to that of the beta-structured peptide. Therefore, HS inhibited the acquisition of PrP(Sc)-like structural properties that, in turn, are responsible for HuPrP intracellular accumulation and lead to neuronal death. Important implications of these data are that HuPrP-HS complexes, being unable to be internalized in living cells may represent a molecular mechanism for the reduced transmission of prion transmission from HS-rich soil also in the presence of contamination from infected animals.


Subject(s)
Peptide Fragments/chemistry , Peptide Fragments/metabolism , Prions/chemistry , Prions/metabolism , Amino Acid Sequence , Benzopyrans/metabolism , Benzopyrans/pharmacology , Benzothiazoles , Cell Line , Endopeptidase K/metabolism , Humans , Humic Substances , In Vitro Techniques , Models, Molecular , Molecular Sequence Data , Peptide Fragments/genetics , Peptide Fragments/toxicity , PrPC Proteins/chemistry , PrPC Proteins/metabolism , PrPSc Proteins/chemistry , PrPSc Proteins/metabolism , Prions/genetics , Prions/toxicity , Protein Binding , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Soil/analysis , Spectrometry, Fluorescence , Static Electricity , Thiazoles/metabolism
9.
Br J Pharmacol ; 155(2): 198-209, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18587421

ABSTRACT

BACKGROUND AND PURPOSE: Cyclooxygenase-2 (COX-2) is expressed in colonic neoplasms, where it supports cell proliferation via prostaglandin E(2) (PGE(2)) production. This study investigated the effects of somatostatin-14 on COX-2 expression, PGE(2) production and proliferation in colon cancer cells. EXPERIMENTAL APPROACH: Human colon adenocarcinoma cell lines Caco-2, HT-29 and HCT116 were used. The following techniques were employed: colourimetric assay for cell growth; 5-bromo-2'-deoxyuridine assay for DNA synthesis; enzyme immunoassay for PGE(2); COX-2 mRNA silencing; RT-PCR or Western blot for somatostatin receptor subtypes, cyclooxygenase isoforms, phosphorylated-ERK-1/ERK-2 and phosphorylated-Akt. KEY RESULTS: HT-29 and Caco-2 cells expressed COX-2 and somatostatin receptors (sst(3/4/5) and sst(3/5), respectively). HCT116 cells did express somatostatin receptors (sst(2/3/5)), but not COX-2. Somatostatin-14 inhibited basal COX-2 expression, PGE(2) production, DNA synthesis and growth in Caco-2 cells and these effects were prevented by BN81658 (sst(3) receptor antagonist). Basal proliferation of HT-29, HCT116 and COX-2-silenced Caco-2 cells was not affected by somatostatin-14. Stimulation of HT-29 cells with gastrin-17 elicited increments of ERK-1/ERK-2 and Akt phosphorylation, COX-2 expression, PGE(2) production, DNA synthesis and cell growth, which were all counteracted by somatostatin-14. Somatostatin-14-induced inhibition of COX-2 expression, PGE(2) production and DNA synthesis were blocked by BIM23056 (sst(5) receptor antagonist). CONCLUSIONS AND IMPLICATIONS: Somatostatin decreases COX-2 expression and function in colon cancer cells via activation of sst(3) or sst(5) receptors, and these effects contribute to the inhibitory action of somatostatin on cell proliferation. These findings can be relevant to the development of therapeutic strategies based on the modulation of the COX-2 pathway.


Subject(s)
Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/metabolism , Somatostatin/pharmacology , Caco-2 Cells , Colon/pathology , Cyclooxygenase 2/genetics , Down-Regulation/drug effects , Down-Regulation/physiology , Gastrins/metabolism , HT29 Cells , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Oligopeptides/metabolism , Protein Kinase C/metabolism , Protein Tyrosine Phosphatases/metabolism
10.
Amino Acids ; 32(3): 395-400, 2007.
Article in English | MEDLINE | ID: mdl-17279309

ABSTRACT

Imatinib, a tyrosine kinase inhibitor directed against the enzymatic domain of KIT protein, was found to produce dramatic clinical responses in metastatic gastrointestinal stromal tumors (GISTs). However, resistance usually develops thus determining treatment failure. The present study was performed to analyse the expression of somatostatin receptor (SSTR) subtypes, modulators of tissue transglutaminase, in a series of GISTs and leiomyosarcomas by immunohistochemistry to identify a new potential therapeutic target. Sixteen cases (8 males and 8 females, age range: 38-73; 11 GISTs, 4 leiomyosarcomas, 1 leiomyoma) were studied. Immunohistochemical detection of the relevant SSTRs was performed on paraffin-embedded tissue sections, stained with polyclonal antibodies directed against the five somatostatin receptor subtypes. We found 7 out of 16 (44%) tumors expressing all SSTRs and 14 out of 16 (87%) tumors positive for at least 3 subtypes. SSTR2A was the most represented subtype in the tumors studied, being expressed in approximately 70% of cases exhibiting an intense labeling in most of these cases. The significant expression of SSTRs shown in this series of GISTs and gastrointestinal leiomyosarcomas suggests a potential therapeutic target to be explored alone and/or in combination with other therapeutic agents in the setting of refractory GI stromal tumors.


Subject(s)
Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/enzymology , Leiomyoma/drug therapy , Leiomyoma/enzymology , Leiomyosarcoma/drug therapy , Leiomyosarcoma/enzymology , Somatostatin/therapeutic use , Transglutaminases/biosynthesis , Adult , Aged , Benzamides , Drug Resistance, Neoplasm/drug effects , Enzyme Activation/drug effects , Enzyme Induction/drug effects , Female , Gastrointestinal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Imatinib Mesylate , Leiomyoma/pathology , Leiomyosarcoma/pathology , Male , Mesoderm/enzymology , Mesoderm/pathology , Middle Aged , Piperazines/therapeutic use , Protein Kinase Inhibitors/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Pyrimidines/therapeutic use , Receptors, Somatostatin/biosynthesis , Somatostatin/analogs & derivatives
11.
Neuroscience ; 142(4): 991-7, 2006 Nov 03.
Article in English | MEDLINE | ID: mdl-16949212

ABSTRACT

Hereditary creatine transporter deficiency causes brain damage, despite the brain having the enzymes to synthesize creatine. Such damage occurring despite an endogenous synthesis is not easily explained. This condition is incurable, because creatine may not be delivered to the brain without its transporter. Creatine-derived compounds that crossed the blood-brain barrier in a transporter-independent fashion would be useful in the therapy of hereditary creatine transporter deficiency, and possibly also in neuroprotection against brain anoxia or ischemia. We tested the double hypothesis that: (1) the creatine carrier is needed to make creatine cross the plasma membrane of brain cells and (2) creatine-derived molecules may cross this plasma membrane independently of the creatine carrier. In in vitro mouse hippocampal slices, incubation with creatine increased creatine and phosphocreatine content of the tissue. Inhibition of the creatine transporter with 3-guanidinopropionic acid (GPA) dose-dependently prevented this increase. Incubation with creatine benzyl ester (CrOBzl) or phosphocreatine-Mg-complex acetate (PCr-Mg-CPLX) increased tissue creatine content, not phosphocreatine. This increase was not prevented by GPA. Thus, the creatine transporter is required for creatine uptake through the plasma membrane. Since there is a strong indication that creatine in the brain is mainly synthesized by glial cells and transferred to neurons, this might explain why hereditary transporter deficiency is attended by severe brain damage despite the possibility of an endogenous synthesis. CrOBzl and PCr-Mg-CPLX cross the plasma membrane in a transporter-independent way, and might be useful in the therapy of hereditary creatine transporter deficiency. They may also prove useful in the therapy of brain anoxia or ischemia.


Subject(s)
Brain/metabolism , Cell Membrane/metabolism , Creatine/deficiency , Membrane Transport Proteins/metabolism , Neurons/metabolism , Animals , Biological Transport, Active/drug effects , Biological Transport, Active/physiology , Brain/drug effects , Brain Diseases, Metabolic/drug therapy , Brain Diseases, Metabolic/metabolism , Brain Diseases, Metabolic/physiopathology , Cell Membrane/drug effects , Creatine/analogs & derivatives , Creatine/pharmacology , Enzyme Inhibitors/pharmacology , Guanidines/pharmacology , Male , Membrane Transport Proteins/drug effects , Mice , Mice, Inbred ICR , Neurons/drug effects , Organ Culture Techniques , Propionates/pharmacology
12.
Int J Immunopathol Pharmacol ; 19(2): 339-56, 2006.
Article in English | MEDLINE | ID: mdl-16831301

ABSTRACT

The transition of prion protein from a mainly alpha-structured isoform (PrPC) to a beta sheet-containing protein (PrPSc) represents a major pathogenetic mechanism in prion diseases. To study the role of PrP structural conformation in prion-dependent neurodegeneration, we analysed the neurotoxicity of PrP in alpha and beta conformations, using a recombinant protein encompassing amino acids 90-231 of the human PrP (hPrP90-231). Using controlled thermal denaturation (53 degrees C, 1h) we converted hPrP90-231 in a structural isoform displaying PrPSc-related characteristics: high beta sheet content, increased aggregability and a slight increase in the resistance to protease K. In virtue of these structural changes, hPrP90-231 powerfully affected the survival of SH-SY5Y cells, inducing a caspase-3 and p38- dependent apoptosis. Conversely, in the native alpha-helix-rich conformation, hPrP90-231 did not show significant cell toxicity. The relationship between the structural state of hPrP90-231 and its neurotoxicity was demonstrated, inducing the thermal denaturation of the peptide in the presence of Congo red that prevented both the transition of hPrP90-231 into a beta-rich isoform and the acquisition of toxic properties. In conclusion, we report that the toxicity of hPrP90-231 is dependent on its three-dimensional structure, as is supposed to occur for the pathogen PrP during TSE.


Subject(s)
Apoptosis/drug effects , PrPC Proteins/chemistry , PrPC Proteins/pharmacology , Amyloid/biosynthesis , Benzothiazoles , Caspase 3 , Caspase 7 , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Circular Dichroism , Endopeptidase K/chemistry , Fluorescent Dyes , Humans , Hydrolysis , Immunoblotting , L-Lactate Dehydrogenase/metabolism , Microscopy, Electron , Necrosis , Protein Conformation , Protein Denaturation , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Structure-Activity Relationship , Tetrazolium Salts , Thiazoles/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Int J Immunopathol Pharmacol ; 18(1): 95-112, 2005.
Article in English | MEDLINE | ID: mdl-15698515

ABSTRACT

Peptides corresponding to three alpha helices present in the C-terminal region of the human prion protein have been synthesized and their structural autonomy analyzed by circular dichroism (CD) and NMR spectroscopy. The results obtained indicate that the protein fragment corresponding to the alpha 3-helix, in contrast to alpha 1 and alpha 2 peptides, shows a complete structural autonomy. The chemical shifts values found for NH and CHalpha resonance of the isolated alpha 3 peptide, formed by 30 aminoacid residues, were markedly and surprisingly similar to the corresponding values of the alpha 3-helix in the protein. The structural autonomy of the alpha 3-helix is profoundly determined by the presence of the conserved capping box and, in part, by the ionic bond formed between Glu200 and Lys204. On the basis of these observations a novel PrP consensus pattern, centered on the alpha 3-helix region, has been defined. The data indicate that this autonomous and highly conserved region of the PrPc likely plays a critical role in folding and stability. This gives an explanation of why many of pathogenic mutations occur in this part of the molecule, sharing relevant effects on the overall protein conformation. In particular the D202N capping mutation almost completely destabilizes the isolated alpha 3 peptide. While it is well known that the D202N substitution is associated with a GSS disease, the possible structural basis of this fatal pathology has never been investigated. We propose that a lower alpha 3-helical propensity leading to a major destabilization of the PrPc molecule initiates the pathogenic process associated with D202N capping mutation.


Subject(s)
Mutation/physiology , Peptide Fragments/genetics , Peptide Fragments/metabolism , Prions/genetics , Prions/metabolism , Amino Acid Sequence , Animals , Circular Dichroism , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Conformation , Molecular Sequence Data , Peptide Fragments/chemical synthesis , Prions/chemical synthesis , Protein Conformation
14.
Endocrinology ; 144(10): 4298-305, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12959980

ABSTRACT

The Src homology 2-containing tyrosine phosphatase, Shp-2, is a crucial enzyme that mediates intracellular signaling and is implicated in cell proliferation and differentiation. Here we investigated the involvement of the Shp-2 tyrosine phosphatase in determining the downstream signaling pathways initiated by the Ret oncogene, carrying either the cysteine 634 to tyrosine or the methionine 918 to threonine substitutions. These mutations convert the receptor tyrosine kinase, Ret, into a dominant transforming protein and induce constitutive activation of its intrinsic tyrosine kinase activity leading to congenital and sporadic cancers in neuroendocrine organs. Using the PC12, rat pheochromocytoma cell line, as model system, we show that Shp-2 mediates immediate-early gene expression if induced by either of the mutant alleles. Furthermore, we show that Shp-2 activity is required for RetM918T-induced Akt activation. The results indicate that Shp-2 is a downstream mediator of the mutated receptors RetC634Y and RetM918T, thus suggesting that it may act as a limiting factor in Ret-associated endocrine tumors, in the neoplastic syndromes multiple endocrine neoplasia types 2A and 2B.


Subject(s)
Intracellular Membranes/physiology , Mutation/physiology , Oncogene Proteins/genetics , Protein Tyrosine Phosphatases/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction/physiology , Animals , Cell Differentiation/physiology , Cell Line , Cell Survival/physiology , Glial Cell Line-Derived Neurotrophic Factor , Intracellular Signaling Peptides and Proteins , Nerve Growth Factors/metabolism , Oncogene Proteins/metabolism , PC12 Cells/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Proto-Oncogene Proteins c-ret , Rats , Receptor Protein-Tyrosine Kinases/metabolism
15.
J Neural Transm (Vienna) ; 110(7): 733-47, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12811634

ABSTRACT

Extracellular electrophysiological recordings of neurons of the parafascicular nucleus of the thalamus were done in normal rats and in rats bearing lesions of either the cerebellar nuclei or the entopeduncular nucleus to investigate the functional control of the pedunculopontine nucleus on the parafascicular nucleus. A total of 97 neurons were recorded in the parafascicular nucleus in intact rats, 83 in rats bearing a chronic electrolytic lesion of the ipsilateral deep cerebellar nuclei, and 69 in rats bearing an ibotenate lesion of the ipsilateral entopeduncular nucleus. Lesions of the cerebellar nuclei or the entopeduncular nucleus were made to evaluate the participation of cerebellothalamic fibers or of polysynaptic basal ganglia circuits in the responses recorded in parafascicular neurons following electrical microstimulation of the ipsilateral pedunculopontine nucleus. Two types of excitation and one type of inhibition were the main responses observed in neurons of the parafascicular nucleus following stimulation of the pedunculopontine nucleus. The first type of excitation, observed in 49.5% of neurons recorded in normal rats, had an onset of 1.8 +/- 0.6 ms, lasted 9.2 +/- 0.8 ms and was able to follow high frequency stimulation over 300 Hz. The second type of excitation, observed in a smaller percentage of neurons recorded (3.1%), was a long-latency (8.3 +/- 0.7 ms) activation lasting 19.0 +/- 4.5 ms. It did not follow stimulation frequencies higher than 50-100 Hz. The inhibitory response was observed in 17.5% of the neurons recorded. The latency of this inhibition was 4.5 +/- 1.8 ms and the duration 41.9 +/- 6.8 ms. In rats bearing a lesion of the deep cerebellar nuclei or of the entopeduncular nucleus, the short-latency activation was still present in 24.1% and 31.9% of neurons recorded, respectively. However, the occurrence of the long-latency excitation rats bearing lesions of either the cerebellum or the entopeduncular nucleus increased to 12.1% and to 17.4%, respectively, while the occurrence of the inhibition rose to 22.9% and to 28.9%. These results show that an excitatory influence on the parafascicular nucleus is exerted by the pedunculopontine nucleus irrespectively of the presence of cerebellofugal fibers. This influence appears to be also independent from the integrity of basal ganglia circuits having a relay at the level of the entopeduncular nucleus. However, the variety of responses recorded suggests that the influences of the pedunculopontine nucleus on the parafascicular nucleus are by far more complex than those exerted on its basal ganglia targets such as the substantia nigra. The results are discussed according to a model of functioning of pedunculopontine fibers directed to thalamic and basal ganglia nuclei.


Subject(s)
Intralaminar Thalamic Nuclei/physiology , Mesencephalon/physiology , Neural Pathways/physiology , Pons/physiology , Animals , Cerebellum/pathology , Electric Stimulation , Electrophysiology , Entopeduncular Nucleus/pathology , Male , Neurons/physiology , Rats , Rats, Sprague-Dawley
16.
Endocrinology ; 144(4): 1574-84, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12639942

ABSTRACT

Somatostatin was reported to inhibit Kaposi's sarcoma (KS) cell (KS-Imm) xenografts through an antiangiogenic activity. Here, we show that somatostatin blocks growth of established KS-Imm tumors with the same efficacy as adriamycin, a clinically effective cytotoxic drug. Whereas KS-Imm cells do not express somatostatin receptors (SSTRs), endothelial cells express several SSTRs, in particular SSTR3. We investigated the molecular mechanisms and receptor specificity of somatostatin inhibition of angiogenesis. Somatostatin significantly inhibited angiogenesis in vivo in the matrigel sponge assay; this inhibition was mimicked by the SSTR3 agonist L-796778 and reversed by the SSTR3 antagonist BN81658, demonstrating involvement of SSTR3. In vitro experiments showed that somatostatin directly affected different endothelial cell line proliferation through a block of growth-factor-stimulated MAPK and endothelial nitric oxide (NO) synthase (eNOS) activities. BN81658 reversed somatostatin inhibition of cell proliferation, NO production, and MAPK activity, indicating that SSTR3 activation is required for the effects of somatostatin in vitro. Finally in vivo angiogenesis assays demonstrated that eNOS inhibition was a prerequisite for the antiangiogenic effects of somatostatin, because high concentrations of sodium nitroprusside, an NO donor, abolished the somatostatin effects. In conclusion, we demonstrate that somatostatin is a powerful antitumor agent in vivo that inhibits tumor angiogenesis through SSTR3-mediated inhibition of both eNOS and MAPK activities.


Subject(s)
Hormones/pharmacology , Neovascularization, Pathologic/drug therapy , Nitric Oxide Synthase/metabolism , Receptors, Somatostatin/metabolism , Somatostatin/pharmacology , Amides/pharmacology , Animals , Cell Division/drug effects , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Enzyme Activation/drug effects , Humans , In Vitro Techniques , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Nitrobenzenes/pharmacology , Receptors, Somatostatin/agonists , Receptors, Somatostatin/antagonists & inhibitors , Umbilical Veins/cytology
17.
Ann N Y Acad Sci ; 1010: 610-22, 2003 Dec.
Article in English | MEDLINE | ID: mdl-15033801

ABSTRACT

Prion diseases are neurodegenerative disorders of the central nervous system of humans and animals, characterized by spongiform degeneration of the central nervous system, astrogliosis, and deposition of amyloid into the brain. The conversion of a cellular glycoprotein (prion protein, PrP(C)) into an altered isoform (PrP(Sc)) has been proposed to represent the causative event responsible for these diseases. The peptide corresponding to the residues 106-126 of PrP sequence (PrP106-126) is largely used to explore the neurotoxic mechanisms underlying the prion diseases. We investigated the intracellular signaling responsible for PrP106-126-dependent cell death in the SH-SY5Y human neuroblastoma cell line. In these cells, PrP106-126 treatment induced apoptotic cell death and the activation of caspase-3. The p38 MAP-kinase blockers (SB203580 and PD169316) prevented the apoptotic cell death evoked by PrP106-126 and Western blot analysis revealed that the exposure of the cells to the peptide induced p38 activation. However, whether the neuronal toxicity of PrP106-126 is caused by a soluble or fibrillar form of this peptide is still unknown. In this study, we correlated the structural state of this peptide with its neurotoxicity. We show that the two conserved glycines in position 114 and 119 prevent the peptide to assume a structured conformation, favoring its aggregation in amyloid fibrils. The substitution of both glycines with alanine residues (PrP106-126AA) generates a soluble nonamyloidogenic peptide, that retained its toxic properties when incubated with neuroblastoma cells. These data show that the amyloid aggregation is not necessary for the induction of the toxic effects of PrP106-126.


Subject(s)
Amyloid beta-Peptides/biosynthesis , Apoptosis/physiology , Mitogen-Activated Protein Kinases/metabolism , Peptide Fragments/pharmacology , Prions/pharmacology , Amino Acid Sequence , Apoptosis/drug effects , Cell Line, Tumor , Humans , Molecular Sequence Data , Neuroblastoma/pathology , Peptide Fragments/chemistry , Prions/chemistry , p38 Mitogen-Activated Protein Kinases
19.
Neurochem Int ; 41(1): 55-63, 2002 Jul.
Article in English | MEDLINE | ID: mdl-11918972

ABSTRACT

Prion diseases are fatal neurodegenerative disorders of the CNS of men and animals, characterized by spongiform degeneration of the CNS, astrogliosis and deposition of amyloid into the brain. The conversion of a cellular glycoprotein (the prion protein, PrP(C)) into an altered isoform (the prion scrapie, PrP(Sc)), which accumulates within the brain tissue by virtue of its resistance to the intracellular catabolism, is currently believed to represent the etiologic agent responsible for these diseases. Synthetic or recombinant polypeptides are commonly used to elucidate the mechanism of proteins involved in neurodegenerative diseases. Here we describe a procedure, which allows the synthesis and purification in its native folding, of the human prion protein fragment 90-231, corresponding to the protease resistant core of PrP(Sc). We synthesized the polypeptides 90-231 of both the wild type and the E200K mutant isoforms of PrP. Using a gluthatione S-transferase (GST) fusion protein approach, milligram amounts of polypeptides were obtained after expression in E. coli. The recovery of the purified fusion protein was monitored following the evaluation of the GST activity. The PrP fragment was released from the fusion protein immobilized on a glutathione-coupled agarose resin by direct cleavage with thrombin. The recombinant protein was identified by comassie stained acrylamide gel and by immunoblotting employing a monoclonal anti-PrP antibody. The peptide purified by gel filtration chromatography showed mainly an alpha-helix structure, as analysed by circular dichroism (CD) and an intact disulfide bridge. The same procedure was also successfully employed to synthesize and purify the E200K mutant PrP fragment.


Subject(s)
Escherichia coli/genetics , Prions/genetics , Base Sequence , Blotting, Western , Chromatography, Liquid , Circular Dichroism , DNA Primers , Humans , Mass Spectrometry , Prions/isolation & purification , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Spectrometry, Fluorescence
20.
Minerva Endocrinol ; 26(3): 91-102, 2001 Sep.
Article in Italian | MEDLINE | ID: mdl-11753230

ABSTRACT

A number of studies have been carried out over the past year to characterise the physiological functions and possible therapeutic applications of somatostatin (sst). Somatostatin is one of the most widespread peptides in the body and it fulfils a very wide spectrum of actions in the various organs and tissues. The aim of this review is to make a critical analysis of knowledge of the structure and physiology of sst and its receptors, together with the possible role played by this peptide in cell proliferation in animal models and the treatment of human tumours. In particular, the authors discuss the most recent findings on cell and intracell mechanisms regulated by somatostatin receptors and how this knowledge may encourage the development of more effective and selective new molecules in terms of therapy.


Subject(s)
Somatostatin/physiology , Animals , Apoptosis , Cell Division , Cells, Cultured , Humans , Rats , Receptors, Somatostatin/physiology , Somatostatin/analogs & derivatives , Somatostatin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...