Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Proteomics ; 283-284: 104928, 2023 07 15.
Article in English | MEDLINE | ID: mdl-37207814

ABSTRACT

Tuberous sclerosis complex (TSC) is a rare, multisystem genetic disorder that leads to the development of benign tumors in multiple organs and neurological symptoms. TSC clinical manifestations show a great heterogenicity, with most patients presenting severe neuropsychiatric and neurological disorders. TSC is caused by loss-of-function mutations in either TSC1 or TSC2 genes, leading to overexpression of the mechanistic target of rapamycin (mTOR) and, consequently, abnormal cellular growth, proliferation and differentiation as well as to cell migration defects. Beside the growing interest, TSC remains a disorder poorly understood, with limited perspectives in the field of therapeutic strategies. Here we used murine postnatal subventricular zone (SVZ) neural stem progenitor cells (NSPCs) deficient of Tsc1 gene as a TSC model to unravel novel molecular aspects of the pathophysiology of this disease. 2D-DIGE-based proteomic analysis detected 55 differently represented spots in Tsc1-deficient cells, compared to wild-type counterparts, which were associated with 36 protein entries after corresponding trypsinolysis and nanoLC-ESI-Q-Orbitrap-MS/MS analysis. Proteomic results were validated using various experimental approaches. Bioinformatics associated differently represented proteins with oxidative stress and redox pathways, methylglyoxal biosynthesis, myelin sheath, protein S-nitrosylation and carbohydrate metabolism. Because most of these cellular pathways have already been linked to TSC features, these results were useful to clarify some molecular aspects of TSC etiopathogenesis and suggested novel promising therapeutic protein targets. SIGNIFICANCE: Tuberous Sclerosis Complex (TSC) is a multisystemic disorder caused by inactivating mutations of TSC1 or TSC2 genes, which induce overactivation of the mTOR component. The molecular mechanisms underlying the pathogenesis of TSC remain unclear, probably due to complexity of mTOR signaling network. To have a picture of protein abundance changes occurring in TSC disorder, murine postnatal subventricular zone (SVZ) neural stem progenitor cells (NSPCs) deficient of Tsc1 gene were used as a model of disease. Thus, Tsc1-deficient SVZ NSPCs and wild-type cells were comparatively evaluated by proteomics. This analysis evidenced changes in the abundance of proteins involved in oxidative/nitrosative stress, cytoskeleton remodelling, neurotransmission, neurogenesis and carbohydrate metabolism. These proteins might clarify novel molecular aspects of TSC etiopathogenesis and constitute putative molecular targets for novel therapeutic management of TSC-related disorders.


Subject(s)
Neural Stem Cells , Tuberous Sclerosis , Mice , Humans , Animals , Tuberous Sclerosis/genetics , Tuberous Sclerosis/metabolism , Tuberous Sclerosis/pathology , Tuberous Sclerosis Complex 1 Protein/metabolism , Proteomics , Tandem Mass Spectrometry , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , TOR Serine-Threonine Kinases/metabolism
2.
Int J Mol Sci ; 20(6)2019 Mar 18.
Article in English | MEDLINE | ID: mdl-30889901

ABSTRACT

The monocarbonyl analogue of curcumin (1E,4E)-1,5-Bis(2-methoxyphenyl)penta-1,4-dien-3-one (C1) has been used as a specific activator of the master gene transcription factor EB (TFEB) to correlate the activation of this nuclear factor with the increased activity of lysosomal glycohydrolases and their recruitment to the cell surface. The presence of active lysosomal glycohydrolases associated with the lipid microdomains has been extensively demonstrated, and their role in glycosphingolipid (GSL) remodeling in both physiological and pathological conditions, such as neurodegenerative disorders, has been suggested. Here, we demonstrate that Jurkat cell stimulation elicits TFEB nuclear translocation and an increase of both the expression of hexosaminidase subunit beta (HEXB), hexosaminidase subunit alpha (HEXA), and galactosidase beta 1 (GLB1) genes, and the recruitment of ß-hexosaminidase (Hex, EC 3.2.1.52) and ß-galactosidase (Gal, EC 3.2.1.23) on lipid microdomains. Treatment of Jurkat cells with the curcumin analogue C1 also resulted in an increase of both lysosomal glycohydrolase activity and their targeting to the cell surface. Similar effects of C1 on lysosomal glycohydrolase expression and their recruitment to lipid microdomains was observed by treating the SH-SY5Y neuroblastoma cell line; the effects of C1 treatment were abolished by TFEB silencing. Together, these results clearly demonstrate the existence of a direct link between TFEB nuclear translocation and the transport of Hex and Gal from lysosomes to the plasma membrane.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Membrane/metabolism , Curcumin/analogs & derivatives , Curcumin/pharmacology , Hexosaminidases/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , beta-Galactosidase/metabolism , Cell Membrane/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Exocytosis/drug effects , Humans , Jurkat Cells , Lipid Bilayers/metabolism , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Phytohemagglutinins/pharmacology , Protein Transport/drug effects
3.
J Funct Biomater ; 10(1)2019 Jan 08.
Article in English | MEDLINE | ID: mdl-30626094

ABSTRACT

Polymer nanoparticles (NPs) represent one of the most innovative non-invasive approaches for drug delivery applications. NPs main objective is to convey the therapeutic molecule be they drugs, proteins, or nucleic acids directly into the target organ or tissue. Many polymers are used for the synthesis of NPs and among the currently most employed materials several biocompatible synthetic polymers, namely polylactic acid (PLA), poly lactic-co-glycolic acid (PLGA), and polyethylene glycol (PEG), can be cited. These molecules are made of simple monomers which are naturally present in the body and therefore easily excreted without being toxic. The present review addresses the different approaches that are most commonly adopted to synthetize biocompatible NPs to date, as well as the experimental strategies designed to load them with therapeutic agents. In fact, drugs may be internalized in the NPs or physically dispersed therein. In this paper the various types of biodegradable polymer NPs will be discussed with emphasis on their applications in drug delivery. Close attention will be devoted to the treatment of cancer, where both active and passive targeting is used to enhance efficacy and reduce systemic toxicity, and to diseases affecting the central nervous system, inasmuch as NPs can be modified to target specific cells or cross membrane barriers.

4.
Exp Neurol ; 311: 88-105, 2019 01.
Article in English | MEDLINE | ID: mdl-30243986

ABSTRACT

The discovery that mammalian target of rapamycin (mTOR) inhibition increases lifespan in mice and restores/delays many aging phenotypes has led to the identification of a novel potential therapeutic target for the treatment of Alzheimer's disease (AD). Among mTOR inhibitors, everolimus, which has been developed to improve the pharmacokinetic characteristics of rapamycin, has been extensively profiled in preclinical and clinical studies as anticancer and immunosuppressive agent, but no information is available about its potential effects on neurodegenerative disorders. Using a reliable mouse model of AD (3 × Tg-AD mice), we explored whether short-term treatment with everolimus injected directly into the brain by osmotic pumps was able to modify AD-like pathology with low impact on peripheral organs. We first established in non-transgenic mice the stability of everolimus at 37 °C in comparison with rapamycin and, then, evaluated its pharmacokinetics and pharmacodynamics profiles through either a single peripheral (i.p.) or central (i.c.v.) route of administration. Finally, 6-month-old (symptomatic phase) 3 × Tg-AD mice were treated with continuous infusion of either vehicle or everolimus (0.167 µg/µl/day, i.c.v.) using the osmotic pumps. Four weeks after the beginning of infusion, we tested our hypothesis following an integrated approach, including behavioral (tests for cognitive and depressive-like alterations), biochemical and immunohistochemical analyses. Everolimus (i) showed higher stability than rapamycin at 37 °C, (ii) poorly crossed the blood-brain barrier after i.p. injection, (iii) was slowly metabolized in the brain due to a longer t1/2 in the brain compared to blood, and (iv) was more effective in the CNS when administered centrally compared to a peripheral route. Moreover, the everolimus-induced mTOR inhibition reduced human APP/Aß and human tau levels and improved cognitive function and depressive-like phenotype in the 3 × Tg-AD mice. The intrathecal infusion of everolimus may be effective to treat early stages of AD-pathology through a short and cyclic administration regimen, with short-term outcomes and a low impact on peripheral organs.


Subject(s)
Affect/drug effects , Alzheimer Disease/drug therapy , Cognition Disorders/drug therapy , Cognition/drug effects , Everolimus/administration & dosage , Immunosuppressive Agents/administration & dosage , Affect/physiology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Cell Line, Tumor , Cognition/physiology , Cognition Disorders/genetics , Cognition Disorders/metabolism , Drug Administration Schedule , Humans , Infusion Pumps, Implantable , Injections, Spinal , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic
5.
Int J Mol Sci ; 19(5)2018 May 16.
Article in English | MEDLINE | ID: mdl-29772672

ABSTRACT

The mechanistic target of rapamycin (mTOR), a serine-threonine kinase, plays a pivotal role in regulating cell growth and proliferation. Notably, a great deal of evidence indicates that mTOR signaling is also crucial in controlling proliferation and differentiation of several stem cell compartments. Consequently, dysregulation of the mTOR pathway is often associated with a variety of disease, such as cancer and metabolic and genetic disorders. For instance, hyperactivation of mTORC1 in neural stem cells (NSCs) is associated with the insurgence of neurological manifestation characterizing tuberous sclerosis complex (TSC). In this review, we survey the recent contributions of TSC physiopathology studies to understand the role of mTOR signaling in both neurogenesis and tumorigenesis and discuss how these new insights can contribute to developing new therapeutic strategies for neurological diseases and cancer.


Subject(s)
Neural Stem Cells/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis/metabolism , Animals , Cell Proliferation , Disease Susceptibility , Energy Metabolism , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Signal Transduction/drug effects , Tuberous Sclerosis/drug therapy
6.
Hum Mol Genet ; 26(17): 3303-3312, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28637240

ABSTRACT

Tuberous sclerosis complex (TSC) is an autosomal dominant genetic disorder caused by mutations in either of two genes, TSC1 or TSC2, resulting in the constitutive activation of the mammalian target of rapamycin complex 1 (mTORC1). mTOR inhibitors are now considered the treatment of choice for TSC disease. A major pathological feature of TSC is the development of subependymal giant cell astrocytomas (SEGAs) in the brain. Nowadays, it is thought that SEGAs could be a consequence of aberrant aggregation and migration of neural stem/progenitor cells (NSPCs). Therefore, reactivation of cell migration of NSPCs might be the crucial step for the treatment of patients. In order to identify potential in vitro targets activating migration, we generated Tsc1-deficient NSPCs. These cells summarize most of the biochemical and morphological characteristics of TSC neural cells, such as the mTORC1 activation, the formation of abnormally enlarged astrocytes-like cells, the reduction of autophagy flux and the impairment of cell migration. Moreover, nuclear translocation, namely activation of the transcription factor EB (TFEB) was markedly impaired. Herein, we show that compounds such as everolimus, ionomycin and curcumin, which directly or indirectly stimulate TFEB nuclear translocation, restore Tsc1-deficient NSPC migration. Our data suggest that reduction of TFEB activation, caused by mTORC1 hyperactivation, contributes to the migration deficit characterizing Tsc1-deficient NSPCs. The present work highlights TFEB as a druggable protein target for SEGAs therapy, which can be additionally or alternatively exploited for the mTORC1-directed inhibitory approach.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Neural Stem Cells/metabolism , Animals , Astrocytoma/pathology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Brain/metabolism , Cell Movement/drug effects , Disease Models, Animal , Mice , Mutation/drug effects , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis/metabolism , Tuberous Sclerosis/pathology , Tuberous Sclerosis Complex 1 Protein , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
7.
Nanomaterials (Basel) ; 6(5)2016 May 09.
Article in English | MEDLINE | ID: mdl-28335215

ABSTRACT

Recently, the use of mammalian target of rapamycin (mTOR) inhibitors, in particular rapamycin (Rp), has been suggested to improve the treatment of neurodegenerative diseases. However, as Rp is a strong immunosuppressant, specific delivery to the brain has been postulated to avoid systemic exposure. In this work, we fabricated new Rp loaded solid lipid nanoparticles (Rp-SLN) stabilized with polysorbate 80 (PS80), comparing two different methods and lipids. The formulations were characterized by differential scanning calorimetry (DSC), nuclear magnetic resonance (NMR), wide angle X-ray scattering (WAXS), cryo-transmission electron microscopy (cryo-TEM), dynamic light scattering (DLS) and particle tracking. In vitro release and short-term stability were assessed. Biological behavior of Rp-SLN was tested in SH-SY5Y neuroblastoma cells. The inhibition of mTOR complex 1 (mTORC1) was evaluated over time by a pulse-chase study compared to free Rp and Rp nanocrystals. Compritol Rp-SLN resulted more stable and possessing proper size and surface properties with respect to cetyl palmitate Rp-SLN. Rapamycin was entrapped in an amorphous form in the solid lipid matrix that showed partial crystallinity with stable Lß, sub-Lα and Lß' arrangements. PS80 was stably anchored on particle surface. No drug release was observed over 24 h and Rp-SLN had a higher cell uptake and a more sustained effect over a week. The mTORC1 inhibition was higher with Rp-SLN. Overall, compritol Rp-SLN show suitable characteristics and stability to be considered for further investigation as Rp brain delivery system.

8.
Article in English | MEDLINE | ID: mdl-25682337

ABSTRACT

Isotopic internal standards are increasingly frequent in LC-MS analysis to control biological matrix effects in the quantitation of immunosuppressant drugs, such as everolimus (RAD001). Here we present the evaluation of a LC-MS method, exploiting [(13)C2D4]RAD001 as internal standard, for preclinical determination of RAD001 in mice brain tissue. Samples were purified by solid phase extraction. Brain and blood were collected from vehicle-treated and RAD001-treated mice. The QTOF MS detector was set to select RAD001 ammonium adducts (m/z 975.6152) and [(13)C2D4]RAD001 (m/z 981.6481). Two different UHPLC columns were preliminarily tested. The method showed linear behavior between 4 and 100ng/mL (r(2)=0.99943) and linearity was preserved in the presence of blood (r(2)=0.99107) and brain (r(2)=0.99098) matrix components. Intra-day and inter-day precision (3-19%) and accuracy (82-109%) were comparable between standards and spiked blood and brain samples. As resulting from recovery comparison (82-98%), [(13)C2D4]RAD001 compensated ion suppression phenomena maintaining method performance over a wide range of consecutive analytical runs. The comparison with a HPLC-UV method showed reliability of the method with good correlation between blood (r(2)=0.94319) and brain (r(2)=0.97773) samples and acceptable biases (<15%). This validation suggests that the investigated method could be useful for the preclinical monitoring of RAD001 brain therapeutic concentrations in animal models.


Subject(s)
Brain Chemistry , Carbon Isotopes/analysis , Chromatography, Liquid/methods , Deuterium/analysis , Mass Spectrometry/methods , Sirolimus/analogs & derivatives , Animals , Carbon Isotopes/chemistry , Carbon Isotopes/pharmacokinetics , Deuterium/chemistry , Deuterium/pharmacokinetics , Everolimus , Linear Models , Male , Mice , Mice, Inbred C57BL , Reproducibility of Results , Sensitivity and Specificity , Sirolimus/analysis , Sirolimus/chemistry , Sirolimus/pharmacokinetics
9.
Int J Biochem Cell Biol ; 58: 62-70, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25462158

ABSTRACT

A critical role of endosomal-lysosomal system alteration in neurodegeneration is supported by several studies. Dysfunction of the lysosomal compartment is a common feature also in Alzheimer's disease. Altered expression of lysosomal glycohydrolases has been demonstrated not only in the brain and peripheral tissues of Alzheimer's disease patients, but also in presymptomatic subjects before degenerative phenomenon becomes evident. Moreover, the presence of glycohydrolases associated to the plasma membrane have been widely demonstrated and their alteration in pathological conditions has been documented. In particular, lipid microdomains-associated glycohydrolases can be functional to the maintenance of the proper glycosphingolipids pattern, especially at cell surface level, where they are crucial for the function of cell types such as neurons. In this study we investigated the localization of ß-hexosaminidase and ß-galactosidase glycohydrolases, both involved in step by step degradation of the GM1 to GM3 gangliosides, in lipid microdomains from the cortex of both an early and advanced TgCRND8 mouse model of Alzheimer's disease. Throughout immunoprecipitation experiments of purified cortical lipid microdomains, we demonstrated for the first time that ß-hexosaminidase and ß-galactosidase are associated with post-synaptic vesicles and that their activities are increased at both the early and the advanced stage of Alzheimer's disease. The early increase of lipid microdomain-associated ß-hexosaminidase and ß-galactosidase activities could have relevant implications for the pathophysiology of the disease since their possible pharmacological manipulation could shed light on new reliable targets and biological markers of Alzheimer's disease.


Subject(s)
Alzheimer Disease/enzymology , Cell Membrane/enzymology , Lysosomes/enzymology , beta-Galactosidase/metabolism , Alzheimer Disease/metabolism , Animals , Blotting, Western , Cell Membrane/metabolism , Glycoside Hydrolases/genetics , Glycoside Hydrolases/metabolism , Humans , In Vitro Techniques , Membrane Microdomains/genetics , Membrane Microdomains/metabolism , Mice , Mice, Transgenic , beta-Galactosidase/genetics
10.
PLoS One ; 9(2): e89485, 2014.
Article in English | MEDLINE | ID: mdl-24586816

ABSTRACT

The expression of constitutively active H-RasV12 oncogene has been described to induce proliferative arrest and premature senescence in many cell models. There are a number of studies indicating an association between senescence and lysosomal enzyme alterations, e.g. lysosomal ß-galactosidase is the most widely used biomarker to detect senescence in cultured cells and we previously reported that H-RasV12 up-regulates lysosomal glycohydrolases enzymatic activity in human fibroblasts. Here we investigated the molecular mechanisms underlying lysosomal glycohydrolase ß-hexosaminidase up-regulation in human fibroblasts expressing the constitutively active H-RasV12. We demonstrated that H-Ras activation increases ß-hexosaminidase expression and secretion by a Raf/extracellular signal-regulated protein kinase dependent pathway, through a mechanism that relies on the activity of the transcription factor EB (TFEB). Because of the pivotal role of TFEB in the regulation of lysosomal system biogenesis and function, our results suggest that this could be a general mechanism to enhance lysosomal enzymes activity during oncogene-induced senescence.


Subject(s)
Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Dermis/metabolism , Fibroblasts/metabolism , Gene Expression Regulation, Enzymologic , Proto-Oncogene Proteins p21(ras)/metabolism , beta-N-Acetylhexosaminidases/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Blotting, Western , Cells, Cultured , Cellular Senescence , Chromatin Immunoprecipitation , Dermis/pathology , Electrophoretic Mobility Shift Assay , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/pathology , Humans , Luciferases/metabolism , Lysosomes/enzymology , Mutation/genetics , Proto-Oncogene Proteins p21(ras)/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transcriptional Activation , Up-Regulation , beta-Galactosidase/metabolism , beta-N-Acetylhexosaminidases/metabolism , raf Kinases/genetics , raf Kinases/metabolism
11.
Methods Enzymol ; 534: 25-45, 2014.
Article in English | MEDLINE | ID: mdl-24359946

ABSTRACT

The endosomal-lysosomal system plays important roles in cellular physiology. Beyond the well-known function as terminal degradative compartment, necessary to maintain the health of the cell, lysosomes are critical for many other cellular processes, such as termination of signaling mediated by cell surface receptors and processing of internalized peptides in antigen-presenting cells. Moreover, the intracellular membrane trafficking related to the endosomal-lysosomal system plays a pivotal role in diverse physiological and pathological processes, such as exocytosis, plasma membrane repair, and endocytosis. Increasing evidences suggest that several lysosomal glycohydrolases, together with nonlysosomal glycohydrolases, are associated with cell membranes in their active form, and they are localized into lipid microdomains. The role of these forms in physiological and pathological conditions, such as differentiation and aging, neurodegenerative diseases, and cancer spreading, is under investigation. Here we provide general methods to purify lipid microdomain proteins and to discriminate cell surface lipid microdomains-associated glycohydrolases from those not exposed on cell surface. The methods reported here have been developed to characterize the membrane-associated forms of the acidic glycohydrolases ß-hexosaminidase and ß-galactosidase, but they may be applied to any other protein of interest.


Subject(s)
Endosomes/chemistry , Lysosomes/chemistry , Membrane Microdomains/chemistry , beta-Galactosidase/metabolism , beta-N-Acetylhexosaminidases/metabolism , Biotinylation , Chromatography, Affinity , Chromatography, DEAE-Cellulose , Endosomes/metabolism , G(M1) Ganglioside/chemistry , G(M1) Ganglioside/metabolism , Gene Expression , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Jurkat Cells , Lysosomes/metabolism , Membrane Microdomains/metabolism , Microscopy, Fluorescence , Protein Transport , beta-Galactosidase/chemistry , beta-Galactosidase/isolation & purification , beta-N-Acetylhexosaminidases/chemistry , beta-N-Acetylhexosaminidases/isolation & purification
12.
Mol Neurobiol ; 50(1): 159-67, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24356898

ABSTRACT

Sphingolipidoses are inherited genetic diseases due to mutations in genes encoding proteins involved in the lysosomal catabolism of sphingolipids. Despite a low incidence of each individual disease, altogether, the number of patients involved is relatively high and resolutive approaches for treatment are still lacking. The chaperone therapy is one of the latest pharmacological approaches to these storage diseases. This therapy allows the mutated protein to escape its natural removal and to increase its quantity in lysosomes, thus partially restoring the metabolic functions. Sandhoff disease is an autosomal recessive inherited disorder resulting from ß-hexosaminidase deficiency and characterized by large accumulation of GM2 ganglioside in brain. No enzymatic replacement therapy is currently available, and the use of inhibitors of glycosphingolipid biosynthesis for substrate reduction therapy, although very promising, is associated with serious side effects. The chaperone pyrimethamine has been proposed as a very promising drug in those cases characterized by a residual enzyme activity. In this review, we report the effect of pyrimethamine on the recovery of ß-hexosaminidase activity in cultured fibroblasts from Sandhoff patients.


Subject(s)
Fibroblasts/drug effects , Hexosaminidase B/metabolism , Molecular Chaperones/pharmacology , Pyrimethamine/pharmacology , Sandhoff Disease/drug therapy , Fibroblasts/enzymology , Humans , Molecular Chaperones/therapeutic use , Pyrimethamine/therapeutic use , Sandhoff Disease/enzymology
13.
Biochem Biophys Res Commun ; 440(2): 251-7, 2013 Oct 18.
Article in English | MEDLINE | ID: mdl-24055709

ABSTRACT

Lysosomes are membrane-enclosed organelles containing acid hydrolases. They mediate a variety of physiological processes, such as cellular clearance, lipid homeostasis, energy metabolism and pathogen defence. Lysosomes can secrete their content through a process called lysosome exocytosis in which lysosomes fuse with the plasma membrane realising their content into the extracellular milieu. Lysosomal exocytosis is not only responsible for the secretion of lysosomal enzymes, but it also has a crucial role in the plasma membrane repair. Recently, it has been demonstrated that lysosome response to the physiologic signals is regulated by the transcription factor EB (TFEB). In particular, lysosomal secretion is transcriptionally regulated by TFEB which induces both the docking and fusion of lysosomes with the plasma membrane. In this work we demonstrated that TFEB nuclear translocation is accompanied by an increase of mature glycohydrolases ß-hexosaminidase and ß-galactosidase on cell surface. This evidence contributes to elucidate an unknown TFEB biological function leading the lysosomal glycohydrolases on plasma membrane.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Lysosomes/enzymology , beta-Galactosidase/biosynthesis , beta-N-Acetylhexosaminidases/biosynthesis , Cell Membrane/metabolism , Exocytosis , HEK293 Cells , Humans , Protein Transport
14.
Genes (Basel) ; 4(2): 152-70, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-24705158

ABSTRACT

Exosomes are small extracellular vesicles (30-100 nm) derived from the endosomal system, which have raised considerable interest in the last decade. Several studies have shown that they mediate cell-to-cell communication in a variety of biological processes. Thus, in addition to cell-to-cell direct interaction or secretion of active molecules, they are now considered another class of signal mediators. Exosomes can be secreted by several cell types and retrieved in many body fluids, such as blood, urine, saliva and cerebrospinal fluid. In addition to proteins and lipids, they also contain nucleic acids, namely mRNA and miRNA. These features have prompted extensive research to exploit them as a source of biomarkers for several pathologies, such as cancer and neurodegenerative disorders. In this context, exosomes also appear attractive as gene delivery vehicles. Furthermore, exosome immunomodulatory and regenerative properties are also encouraging their application for further therapeutic purposes. Nevertheless, several issues remain to be addressed: exosome biogenesis and secretion mechanisms have not been clearly understood, and physiological functions, as well as pathological roles, are far from being satisfactorily elucidated.

15.
Biochimie ; 94(3): 684-94, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21978926

ABSTRACT

Growing evidence suggests the presence of active lysosomal enzymes in extra-lysosomal compartments, such as the plasma membrane. Although in the past little attention was paid to glycohydrolases acting on cellular compartments different from lysosomes, there is now increasing interest on plasma membrane-associated glycohydrolases because they should be involved, together with glycosyltransferases, in glycosphingolipids oligosaccharide modification processes regulating cell-to-cell and/or cell-environment interactions in both physiological and pathological conditions. Starting from the previous evidence of the presence of ß-hexosaminidase and ß-galactosidase at the plasma membrane of cultured fibroblasts, we here investigated the association of these glycohydrolases with lipid microdomains of Jurkat T-lymphocytes. Monosialoganglioside GM3 represents the major glycosphingolipid constituent of T-cell plasma membrane and its amount largely increases after T-cell stimulation. ß-hexosaminidase and ß-galactosidase cleave specific ß-linked terminal residues from a wide range of glycoconjugates and in particular are involved in the stepwise degradation of GM1 to GM3 ganglioside. Here we demonstrated that fully processed plasma membrane-associated ß-hexosaminidase and ß-galactosidase co-distribute with the lipid microdomain markers and co-immunoprecipitate with the signalling protein lck in Jurkat T-cell. Furthermore, Jurkat cell stimulation up-regulates the expression and activity of lysosomal ß-hexosaminidase and ß-galactosidase and increases their targeting to lipid microdomains. The non-random distribution of plasma membrane-associated ß-hexosaminidase and ß-galactosidase and their localization within lipid microdomains, suggest a role of these enzymes in the local reorganization of glycosphingolipid-based signalling units.


Subject(s)
Jurkat Cells/metabolism , Membrane Microdomains/metabolism , T-Lymphocytes/metabolism , beta-Galactosidase/metabolism , beta-N-Acetylhexosaminidases/metabolism , Cell Line , Humans , Real-Time Polymerase Chain Reaction , beta-Galactosidase/genetics , beta-N-Acetylhexosaminidases/genetics
16.
Mol Cell Biochem ; 363(1-2): 109-18, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22147196

ABSTRACT

Lysosomes are not only degrading organelles but also involved in other critical cellular processes. In addition, active lysosomal glycohydrolases have been detected in an extra-lysosomal compartment: the presence of glycohydrolases on the plasma membrane (PM) has been widely demonstrated, and a possible role on the modification of the cell surface glycosphingolipids (GSL) participating in the modulation of cell functions such as cell-to-cell interactions and signal transduction pathways has been proposed. On this basis, the coordinated expression of lysosomal glycohydrolases and their translocation to the PM appear to be crucial for many cellular events. In this paper, we report evidence for the existence of a coordinated mechanism regulating the expression/activity of both lysosomal and PM-associated glycohydrolases. We show that the over-expression of the acidic glycohydrolase ß-hexosaminidase α-subunit in mouse NIH/3T3 fibroblasts induces the increased expression of the Hex ß-subunit necessary to form the active isoenzyme dimers as well as of other glycohydrolases participating in the GSL catabolism, such as ß-galactosidase and ß-glucocerebrosidase. More interestingly, this regulatory effect was also extended to the PM-associated hydrolases. In addition, transfected cells displayed a rearrangement of the GSL expression pattern that cannot be simply explained by the increased activity of a single enzyme. These observations clearly indicate that the expression level of metabolically related glycohydrolases is regulated in a coordinated manner and this regulation mechanism also involves the PM-associated isoforms.


Subject(s)
Cell Membrane/enzymology , Fibroblasts/enzymology , Glycoside Hydrolases/metabolism , Glycosphingolipids/metabolism , Lysosomes/enzymology , beta-Hexosaminidase alpha Chain/metabolism , Animals , Exocytosis , Humans , Mice , NIH 3T3 Cells , Transfection , beta-Hexosaminidase alpha Chain/genetics
17.
Biosci Rep ; 31(6): 477-87, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21521175

ABSTRACT

Lysosomal α-D-mannosidase is an exoglycosidase involved in the ordered degradation of N-linked oligosaccharides. It is ubiquitously expressed, although the main transcript is more abundant in peripheral blood leucocytes. Here we report that α-D-mannosidase enzyme activity is very high in the promyelocytic leukaemia cell lines HL60 and NB4, as compared with other leukaemic cell lines or cells from different human sources. The MAN2B1 transcript level correlates with enzyme activity, indicating a transcriptional up-regulation of the α-D-mannosidase gene. The promoter was then characterized in HEK-293 cells (human embryonic kidney 293 cells) and HL60 cells; regulatory sequences crucial for its activity were determined by reporter gene assay in HEK-293 cells and located in the region -101/-71 with respect to the first ATG codon. Supershift assay demonstrated that Sp1 (specificity protein 1) bound to this sequence both in HEK-293 and HL60 cells. However, 5'-RACE (5'-rapid amplification of cDNA ends) indicated the use of multiple upstream TSSs (transcription start sites) in HL60 with respect to HEK-293 cells and gel shift analysis of the sequence -373/-269 demonstrated a specific binding by NF-κB (nuclear factor κB) transcription factor in HL60 but not in HEK-293 cells. We concluded that despite the α-D-mannosidase promoter showing typical features of housekeeping gene promoters, α-D-mannosidase transcription is specifically regulated in HL60 by NF-κB transcription factor.


Subject(s)
Leukemia, Promyelocytic, Acute/enzymology , Lysosomes/enzymology , NF-kappa B/genetics , Promoter Regions, Genetic/genetics , alpha-Mannosidase/genetics , alpha-Mannosidase/metabolism , Base Sequence , Gene Expression Regulation , Glycoside Hydrolases/chemistry , HEK293 Cells , HL-60 Cells , Humans , K562 Cells , Leukemia, Promyelocytic, Acute/pathology , Molecular Sequence Data , NF-kappa B/chemistry , Tumor Cells, Cultured
18.
Recent Pat CNS Drug Discov ; 6(1): 1-19, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21073432

ABSTRACT

Genetic mutations that cause specific lysosomal protein deficiencies account for more than 45 Lysosomal Storage Diseases (LSDs), mostly pre-adult disorders which are associated with neurological symptoms and mental retardation. Interestingly, such diseases are often characterized by intracellular deposition and protein aggregation, events also found in age-related neurodegenerative diseases. During the past twenty years, different approaches have been introduced for the treatment of these disorders, several of which are now in routine clinical use or clinical trials. Among them, enzyme replacement therapy (ERT) represented a major progress. However, the usefulness of ERT is limited due to the fact that enzyme distribution is insufficient and treatment costs are very high. A further novel therapeutic option for LSDs is based on the use of small molecules, that can either inhibit a key enzyme which is responsible for substrate synthesis (substrate reduction) or act as a chaperone to increase the residual activity of the lysosomal enzyme (pharmacological chaperones). In addition, recently various gene therapy approaches have been developed, mostly based on adeno-associated and lentiviral vectors, and strategies based on stem cells administration are beginning their route. This review provides an update of the status of research on LSDs therapeutic approaches, including recent patents in the field.


Subject(s)
Enzyme Replacement Therapy , Genetic Therapy , Lysosomal Storage Diseases/therapy , Lysosomes/drug effects , Molecular Chaperones/therapeutic use , Stem Cell Transplantation , Genetic Vectors , Humans , Hydrolases/genetics , Hydrolases/metabolism , Hydrolases/therapeutic use , Intellectual Disability/etiology , Intellectual Disability/genetics , Lysosomal Storage Diseases/drug therapy , Lysosomal Storage Diseases/genetics , Lysosomes/genetics , Lysosomes/metabolism , Molecular Chaperones/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...