Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Pharmaceuticals (Basel) ; 14(10)2021 Sep 29.
Article in English | MEDLINE | ID: mdl-34681227

ABSTRACT

Methamphetamine (METH) is a widely abused psychostimulant and a stress-inducing compound, which leads to neurotoxicity for nigrostriatal dopamine (DA) terminals in rodents and primates including humans. In vitro studies indicate that autophagy is a strong modulator of METH toxicity. In detail, suppressing autophagy increases METH toxicity, while stimulating autophagy prevents METH-induced toxicity in cell cultures. In the present study, the role of autophagy was investigated in vivo. In the whole brain, METH alone destroys meso-striatal DA axon terminals, while fairly sparing DA cell bodies within substantia nigra pars compacta (SNpc). No damage to either cell bodies or axons from ventral tegmental area (VTA) is currently documented. According to the hypothesis that ongoing autophagy prevents METH-induced DA toxicity, we tested whether systemic injection of autophagy inhibitors such as asparagine (ASN, 1000 mg/Kg) or glutamine (GLN, 1000 mg/Kg), may extend METH toxicity to DA cell bodies, both within SNpc and VTA, where autophagy was found to be inhibited. When METH (5 mg/Kg × 4, 2 h apart) was administered to C57Bl/6 mice following ASN or GLN, a frank loss of cell bodies takes place within SNpc and a loss of both axons and cell bodies of VTA neurons is documented. These data indicate that, ongoing autophagy protects DA neurons and determines the refractoriness of cell bodies to METH-induced toxicity.

2.
Toxicol Sci ; 123(2): 523-41, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21742779

ABSTRACT

In human neuroblastoma SH-SY5Y cells, hydrogen peroxide (H(2)O(2), 200µM) rapidly (< 5 min) induced autophagy, as shown by processing and vacuolar relocation of light chain 3(LC3). Accumulation of autophagosome peaked at 30 min of H(2)O(2) exposure. The continuous presence of H(2)O(2) eventually (at > 60 min) caused autophagy-dependent annexin V-positive cell death. However, the cells exposed to H(2)O(2) for 30 min and then cultivated in fresh medium could recover and grow, despite ongoing autophagy. H(2)O(2) rapidly (5 min) triggered the formation of dichlorofluorescein-sensitive HO(·)-free radicals within mitochondria, whereas the mitochondria-associated oxidoradicals revealed by MitoSox (O(2)(·-)) became apparent after 30 min of exposure to H(2)O(2). 3-Methyladenine inhibited autophagy and cell death, but not the generation of HO(·). Genetic silencing of beclin-1 prevented bax- and annexin V-positive cell death induced by H(2)O(2), confirming the involvement of canonical autophagy in peroxide toxicity. The lysosomotropic iron chelator deferoxamine (DFO) prevented the mitochondrial generation of both HO(.) and O(2)(·-) and suppressed the induction of autophagy and of cell death by H(2)O(2). Upon exposure to H(2)O(2), Akt was intensely phosphorylated in the first 30 min, concurrently with mammalian target of rapamycin inactivation and autophagy, and it was dephosphorylated at 2 h, when > 50% of the cells were dead. DFO did not impede Akt phosphorylation, which therefore was independent of reactive oxygen species (ROS) generation but inhibited Akt dephosphorylation. In conclusion, exogenous H(2)O(2) triggers two parallel independent pathways, one leading to autophagy and autophagy-dependent apoptosis, the other to transient Akt phosphorylation, and both are inhibited by DFO. The present work establishes HO(·) as the autophagy-inducing ROS and highlights the need for free lysosomal iron for its production within mitochondria in response to hydrogen peroxide.


Subject(s)
Autophagy/drug effects , Hydrogen Peroxide/toxicity , Lysosomes/drug effects , Neuroblastoma/drug therapy , Oncogene Protein v-akt/metabolism , Oxidants/toxicity , Siderophores/pharmacology , Adenine/analogs & derivatives , Adenine/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Beclin-1 , Cell Line, Tumor , Deferoxamine/pharmacology , Free Radicals/metabolism , Gene Silencing , Humans , Hydroxyl Radical/metabolism , Lysosomes/metabolism , Membrane Proteins/genetics , Mitochondria/drug effects , Mitochondria/metabolism , Neuroblastoma/genetics , Neuroblastoma/metabolism , Phosphorylation , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics
3.
PLoS One ; 6(7): e21908, 2011.
Article in English | MEDLINE | ID: mdl-21747967

ABSTRACT

The lysosomal aspartic protease Cathepsin D (CD) is ubiquitously expressed in eukaryotic organisms. CD activity is essential to accomplish the acid-dependent extensive or partial proteolysis of protein substrates within endosomal and lysosomal compartments therein delivered via endocytosis, phagocytosis or autophagocytosis. CD may also act at physiological pH on small-size substrates in the cytosol and in the extracellular milieu. Mouse and fruit fly CD knock-out models have highlighted the multi-pathophysiological roles of CD in tissue homeostasis and organ development. Here we report the first phenotypic description of the lack of CD expression during zebrafish (Danio rerio) development obtained by morpholino-mediated knock-down of CD mRNA. Since the un-fertilized eggs were shown to be supplied with maternal CD mRNA, only a morpholino targeting a sequence containing the starting ATG codon was effective. The main phenotypic alterations produced by CD knock-down in zebrafish were: 1. abnormal development of the eye and of retinal pigment epithelium; 2. absence of the swim-bladder; 3. skin hyper-pigmentation; 4. reduced growth and premature death. Rescue experiments confirmed the involvement of CD in the developmental processes leading to these phenotypic alterations. Our findings add to the list of CD functions in organ development and patho-physiology in vertebrates.


Subject(s)
Air Sacs/enzymology , Cathepsin D/deficiency , Cathepsin D/genetics , Gene Knockdown Techniques , Retinal Pigment Epithelium/enzymology , Zebrafish/genetics , Zebrafish/physiology , Air Sacs/embryology , Air Sacs/growth & development , Animals , Base Sequence , Body Size/genetics , Embryonic Development/genetics , Humans , Longevity/genetics , Mutation , Oligonucleotides/genetics , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retinal Pigment Epithelium/embryology , Retinal Pigment Epithelium/growth & development , Zebrafish/embryology , Zebrafish/growth & development , Zygote/metabolism
4.
J Agric Food Chem ; 59(8): 4264-72, 2011 Apr 27.
Article in English | MEDLINE | ID: mdl-21395220

ABSTRACT

Malignant glioblastoma represents a challenge in the chemotherapy of brain tumors, because of its aggressive behavior characterized by chemoresistance, infiltrative diffusion, and high rate of recurrence and death. In this study, we used cultured human U87MG cells and primary human glioblastoma cultures to test the anticancer properties of resveratrol (RV), a phytoalexin abundantly present in a variety of dietary products. In U87MG cells, 100 µM RV elicited cell growth arrest by 48 h and bax-mediated cell toxicity by 96 h and greatly limited cell migration and invasion through matrigel. Both in U87MG cells and in primary glioblastoma cultures, the chronic administration of RV (100 µM for up to 96 h) decreased the expression of nestin (a brain (cancer) stem cells marker) but increased that of glial acidic fibrillary protein (a mature glial cell marker) and of ßIII-tubulin (a neuronal differentiation marker). Chronic treatment with RV increased the proportion of cells positive for senescence-associated ß-galactosidase activity. This is the first report showing the ability of RV to induce glial-like and neuronal-like differentiation in glioblastoma cells. The beneficial effects of chronic RV supplementation lasted up to 96 h after its withdrawal from the culture medium. The present findings support the introduction of pulsed administration of this food-derived molecule in the chemotherapy regimen of astrocytomas.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Brain Neoplasms/pathology , Cell Differentiation/drug effects , Cell Division/drug effects , Glioblastoma/pathology , Stilbenes/pharmacology , Cell Line, Tumor , Humans , Microscopy, Fluorescence , Resveratrol , Reverse Transcriptase Polymerase Chain Reaction , Wound Healing
5.
Cancer Biomark ; 7(1): 47-64, 2010.
Article in English | MEDLINE | ID: mdl-21045264

ABSTRACT

Three molecular forms of the proteolytic enzyme Cathepsin D (CD) are found in the cell: the precursor (proCD), the intermediate single-chain and the mature double-chain. ProCD, which is found in the Golgi Complex, is enzymatically inactive, while the intermediate and the mature forms, respectively found in endosomes and lysosomes, are enzymatically active. The latter are involved in autophagy and apoptosis pathways thus playing a crucial role in the control of cell and tissue homeostasis. ProCD can be secreted in the extracellular space and, by interacting with membrane receptors, can promote cell proliferation. At slightly acid pH, secreted proCD undergoes partial maturation and becomes active. In the extracellular space, CD can degrade the protein components of the matrix and free growth factors therein embedded, thus favoring tumor growth, invasion and angiogenesis. Based on the multiple tasks performed by CD inside and outside the cell, it is not irrational to hypothesize its involvement in cancer development and progression and a strict link between its tissue expression and the clinico-pathological features of the tumor. Thus, not surprisingly, as many as 519 articles are found in the database of pubmed with the keywords 'cathepsin D, cancer and marker'. Disappointingly, however, in spite of, or because of, this large number of studies, the scientific community has not reached a general agreement on the prognostic value of CD in cancer progression. Here, we will briefly review the relevant literature and offer a possible explanation for the conflicting data.


Subject(s)
Biomarkers, Tumor/metabolism , Cathepsin D/metabolism , Enzyme Precursors/metabolism , Golgi Apparatus/enzymology , Neoplasms/etiology , Breast Neoplasms/enzymology , Colorectal Neoplasms/enzymology , Disease Progression , Endosomes/enzymology , Female , Head and Neck Neoplasms/enzymology , Humans , Lung Neoplasms/enzymology , Lysosomes/enzymology , Male , Melanoma/enzymology , Neoplasms/enzymology , Neoplasms/pathology , Nervous System Neoplasms/enzymology , Ovarian Neoplasms/enzymology , Pancreatic Neoplasms/enzymology , Prostatic Neoplasms/enzymology , Stomach Neoplasms/enzymology , Urinary Bladder Neoplasms/enzymology
6.
Dis Markers ; 28(3): 167-83, 2010.
Article in English | MEDLINE | ID: mdl-20534902

ABSTRACT

The lysosomal protease Cathepsin D (CD) has been implicated in the homeostasis of lymphatic tissues. We investigated whether the level of CD expression influences the progression and the clinical outcome in Non-Hodgkin's Lymphomas (NHLs). The expression of CD was assessed by immunohistochemistry and immunofluorescence in biopsies of Diffuse Large B Cell Lymphomas (DLBCL, 35 cases), Follicular Lymphomas (FL, 9 cases of grade I-II plus 14 cases of grade IIIB), Chronic Lymphocytic Leukaemias (CLL, 17 cases) and Peripheral T-cell Lymphomas (PTCL, 5 cases). CD staining showed a cytoplasmic punctate pattern compatible with its lysosomal localization. Based on the level of CD expression and the proportion of positive cells, lymphomas were classified as 'low expressing' (< 20% of tumor cells) or 'highly expressing' (>or= 20% of tumor cells). Lymphomas highly expressing CD were associated with a worse stage (III-IV) at diagnosis (31/34 cases; p=0.002) and with a poor clinical outcome (i.e., partial remission and death; 28/34 cases; p=0.03). In the subgroup of aggressive/high grade of malignancy lymphomas (i.e., DLBCL, FL IIIB and PTCL), the Kaplan-Meier curve revealed a very low cumulative overall survival probability (approximately 20% at 5 year) for patients bearing a NHL with > 40% CD-positive cells compared to that of patients bearing a NHL with < 20% CD-positive cells ( approximately 70% at 5 year). This correlation was statistically significant (log-rank test, p=0.01). In Cox multivariate analysis CD failed to be a prognosticator independent of pathologic stage, though the hazard ratio confirmed the association of low expression with a better survival probability. These data indicate that the presence of a high percentage of CD-positive tumor cells negatively reflects on the progression of NHLs.


Subject(s)
Cathepsin D/metabolism , Lymphoma, Non-Hodgkin/enzymology , Adult , Aged , Aged, 80 and over , Female , Humans , Immunohistochemistry , Lymphoma, Non-Hodgkin/pathology , Male , Middle Aged , Survival Analysis , Treatment Outcome
7.
Toxicol Sci ; 117(1): 152-62, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20525898

ABSTRACT

Hydrogen peroxide (H(2)O(2)) is an extremely reactive oxidoradical that is normally produced as a by-product of the mitochondrial activity and also under several metabolic stress conditions. Autophagy, a lysosomal degradation pathway, is triggered by oxidative stress as a defensive response. How autophagy and death pathways are coordinated in cells subjected to oxidative stress is still poorly understood. In human neuroblastoma SH-SY5Y cells, 200microM H(2)O(2) rapidly induced the formation of LC3-positive autophagic vacuoles and of beclin1-Vps34 double-positive macroaggregates. Vacuolar LC3 and beclin1 aggregates did not form when oxidative stress was performed in cells pretreated with 3-methyladenine (3MA), an inhibitor of Vps34, or infected with a recombinant adenovirus expressing a dominant-negative mutant of Vps34. H(2)O(2) provoked the permeabilization of lysosomes (at 30 min) and of mitochondria, the concomitant oligomerization of bax, and eventually (at 2 h), cell death in about 50% of the cell culture. Inactivation of Vps34-dependent autophagy in oxidative-stressed cells abrogated lysosome leakage, bax activation, and caspase-dependent apoptosis and conferred protection for as long as 16 h. Inhibition of caspase activity (by ZVAD-fmk) did not trigger an alternative cell death pathway but rather afforded complete protection from oxidative toxicity, despite the ongoing generation of oxidoradicals and the cellular accumulation of autophagic vacuoles and of leaking lysosomes. On long-term (16 h) exposure to H(2)O(2), signs of necrotic cell death became apparent in LC3-positive cells, which could be prevented by ZVAD-fmk. The present data highlight the pivotal role of autophagy in H(2)O(2)-induced cell death in dopaminergic neuroblastoma cells.


Subject(s)
Apoptosis , Autophagy , Dopamine/metabolism , Neuroblastoma/pathology , Oxidative Stress , Phosphoinositide-3 Kinase Inhibitors , Caspases/metabolism , Cell Line, Tumor , Humans , Necrosis , Neuroblastoma/enzymology , Neuroblastoma/metabolism , Phosphatidylinositol 3-Kinases/metabolism
8.
Mod Pathol ; 23(7): 937-50, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20473282

ABSTRACT

The expression of beclin-1, an oncosuppressor monoallelically deleted in >60% epithelial cancers, has been shown to be developmentally regulated in T and B lymphocytes. By interacting with either bcl-2 or class III phosphatidyl-inositol-3-phosphate kinase, beclin-1 regulates apoptosis and autophagy, two processes crucial for lymphatic tissue homeostasis. We analyzed the potential link between beclin-1-mediated autophagy and the malignant behaviour of lymphomas. The tissue expression of beclin-1 was analyzed in a large series of non-Hodgkin lymphomas and correlated with patient's clinical outcome. By immunofluorescence, beclin-1 staining showed faintly detectable and diffusely distributed in the cytoplasm (regarded as negative) or confined to the perinuclear region as large and brilliant puncta suggestive of macro-aggregate reactivity (regarded as positive). The positive expression of beclin-1 well correlated with the presence of LC3-positive autophagic vacuoles and was inversely correlated with the expression of bcl-2. Non-Hodgkin lymphomas in which > or =20% of tumour cells expressed high level of beclin-1 aggregates were associated with a complete (57%) or partial (35%) remission. The 5-year overall survival probability, calculated by the Kaplan-Meier method, was 92% and 42% in beclin-1-expressing non-Hodgkin lymphomas with > or =20% and <20% positive cells, respectively (log-rank test, P<0.000.1). In Cox multivariate analysis, the level of beclin-1 expression, adjusted for patient's age and pathologic stage, revealed to be significantly correlated with patient's survival (P<0.0001). This is the first demonstration of the involvement of beclin-1 and autophagy in the clinical behaviour of non-Hodgkin lymphomas. The present data are compatible with the hypothesis that non-Hodgkin lymphomas with upregulated autophagy are more responsive to chemotherapy and indicate that beclin-1 could be a valuable independent prognostic factor in this heterogeneous group of tumours.


Subject(s)
Apoptosis Regulatory Proteins/biosynthesis , Autophagy/physiology , Biomarkers, Tumor/analysis , Lymphoma, Non-Hodgkin/metabolism , Lymphoma, Non-Hodgkin/mortality , Lymphoma, Non-Hodgkin/pathology , Membrane Proteins/biosynthesis , Adult , Aged , Aged, 80 and over , Beclin-1 , Blotting, Western , Female , Fluorescent Antibody Technique , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Staging , Prognosis , Proportional Hazards Models
9.
Biol Chem ; 391(5): 519-31, 2010 May.
Article in English | MEDLINE | ID: mdl-20302512

ABSTRACT

Arsenic trioxide (arsenite) was the first chemotherapeutic drug to be described and is now being rediscovered in cancer treatment, including glioblastoma multiforme. Arsenite toxicity triggers autophagy in cancer cells, although final stages of the process involve executive caspases, suggesting an interplay between autophagic and apoptotic pathways that awaits to be explained at a molecular level. We evaluated the contribution of the lysosomal cathepsins (Cat) L and B, which are upregulated in glioblastomas, in the mechanism of arsenite toxicity in human glioblastoma cells. Arsenite treatment induced autophagosome formation and permeabilization of mitochondria, followed by caspase 3/7-mediated apoptosis. The autophagy inhibitor 3-methyladenine protected from arsenite toxicity, whereas bafilomycin A1 did not. Furthermore, arsenite significantly decreased CatB levels and selectively inhibited its cellular and recombinant protein activity, while not affecting CatL. However, downregulation of CatL greatly enhanced apoptosis by arsenite. Our results show that arsenite toxicity involves a complex interplay between autophagy and apoptosis in human glioblastoma cells and is associated with inhibition of CatB, and that this toxicity is highly exacerbated by simultaneous CatL inhibition. The latter points to a synergy that could be used in clinical treatment to lower the therapeutic dose, thus avoiding the toxic side effects of arsenite in glioblastoma management.


Subject(s)
Antineoplastic Agents/therapeutic use , Arsenicals/therapeutic use , Autophagy , Cathepsin B/physiology , Cathepsin L/physiology , Glioblastoma/drug therapy , Oxides/therapeutic use , Adenine/analogs & derivatives , Adenine/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Antineoplastic Agents/adverse effects , Apoptosis/drug effects , Arsenic Trioxide , Arsenicals/adverse effects , Autophagy/drug effects , Caspases/metabolism , Cathepsin B/antagonists & inhibitors , Cathepsin L/antagonists & inhibitors , Cell Line, Tumor , Dipeptides/pharmacology , Down-Regulation , Epoxy Compounds/pharmacology , Glioblastoma/metabolism , Humans , Macrolides/pharmacology , Oxides/adverse effects , Pyridines/pharmacology
10.
Cell Oncol ; 31(6): 423-36, 2009.
Article in English | MEDLINE | ID: mdl-19940359

ABSTRACT

BACKGROUND: Plex-B1, the receptor of Sema4D, has been implicated in tumour growth, angiogenesis and metastasis. The binding of Sema4D to Plex-B1 can trigger the activation of Met tyrosine kinase, thereby promoting cell dissociation and invasive growth. We tested the hypothesis that the expression of Plex-B1, either alone or in association with Met, can be of predictive value for tumour progression. METHODS: The expression and distribution of Plex-B1 and Met were investigated by immunohistochemistry and immunofluorescence in 50 human neoplasias originating in the breast and ovary, and correlated with clinical-pathological data at diagnosis. RESULTS: Plex-B1 and Met were individually expressed in 14% and in 24% of the tumours, respectively. Plex-B1 and Met were co-expressed in 24/50 cases (48%), and in the majority of these (83%) Met was tyrosine phosphorylated. The expression of Plex-B1 or Met alone showed no significant correlation with tumour aggressiveness, whereas advanced stage tumours (III-IV) frequently showed Plex-B1-Met double-positive (9/13). Tumours co-expressing Plex-B1 and Met were characterised by worse grading and higher incidence of lymph node metastases. Out of 22 tumours with lymph node metastases, as many as 19 were Plex-B1 and Met double-positive (p=0.0008), and 17 expressed phosphorylated Met (p=0.002). CONCLUSION: Plex-B1 assumes a predictive value for unfavourable outcome when co-expressed with Met.


Subject(s)
Breast Neoplasms/metabolism , Nerve Tissue Proteins/metabolism , Ovarian Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Growth Factor/metabolism , Blotting, Western , Breast Neoplasms/pathology , Disease Progression , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Neoplasm Staging , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins c-met , Risk Factors
11.
J Cell Mol Med ; 13(6): 1096-109, 2009 Jun.
Article in English | MEDLINE | ID: mdl-18657225

ABSTRACT

The ovarian cancer cell lines A2780 (wild-type p53) and NIHOVCAR3 (mutated p53) showed, respectively, sensitivity and resistance towards several chemotherapy drugs. We hypothesized that the two cell lines differ in their ability to activate the intrinsic death pathway and have, therefore, dissected the lysosome-mitochondrion signalling pathway by pharmacological inhibition or genetic manipulation of key regulators and executioners. Biochemical and morphological confocal fluorescence studies showed that: (1) In A2780 cells bcl-2 is expressed at an undetectable level, whereas Bax is expressed at a rather high level; by contrast, bcl-2 is highly expressed and Bax is expressed at extremely low levels in NIHOVCAR3 cells; (2) Chemotherapy treatment reduced the expression of bcl-2 in NIHOVCAR3 cells, yet these cells resisted to drug toxicity; (3) Cathepsin D (CD), not cathepsin B or L, mediates the activation of the mitochondrial intrinsic death pathway in A2780 cells; (4) Lysosome leakage and cytosolic relocation of CD occurs in the chemosensitive A2780 cells, not in the chemoresistant NIHOVCAR3 cells; (5) Bax is essential for the permeabilization of both lysosomes and mitochondria in A2780 cells exposed to chemotherapy drugs; (6) CD activity is mandatory for the oligomerization of Bax on both mitochondrial and lysosomal membranes; (7) Bax activation did not occur in the resistant NIHOVCAR3 cells despite their high content in CD. The present data are consistent with a model in which on treatment with a cytotoxic drug the activation of a CD-Bax loop leads to the generalized permeabilization of lysosomes and eventually of mitochondria, thus reaching the point of no return, and culminates with the activation of the caspase cascade. Our data also imply that dysfunctional permeabilization of lysosomes contributes to the development of chemoresistance.


Subject(s)
Antineoplastic Agents/pharmacology , Cathepsin D/metabolism , Signal Transduction/drug effects , bcl-2-Associated X Protein/metabolism , Apoptosis/drug effects , Cathepsin D/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Dose-Response Relationship, Drug , Etoposide/pharmacology , Female , Humans , Immunoblotting , Lysosomes/drug effects , Lysosomes/metabolism , Membrane Potential, Mitochondrial/drug effects , Microscopy, Fluorescence , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Paclitaxel/pharmacology , Protein Transport/drug effects , RNA Interference , bcl-2-Associated X Protein/genetics
12.
ACS Appl Mater Interfaces ; 1(3): 678-87, 2009 Mar.
Article in English | MEDLINE | ID: mdl-20355990

ABSTRACT

Mesoporous silica nanoparticles are being explored as versatile tools for various biomedical and biotechnological applications including disease diagnosis, drug delivery, and intracellular imaging. In this paper, the synthesis and characterization of a fluorescent hybrid mesoporous silica nanomaterial, which is noncytotoxic and shows great potential for "in-cell" bioimaging applications, will be described. The hybrid mesoporous material has been obtained by confining highly fluorescent organic dyes, belonging to the indocyanine family, within the channels of mesoporous MCM-41. To explore the dispersion of the dye inside the mesoporous channels and the formation of dye aggregates, several hybrid samples with increasing dye/MCM-41 loading (up to 100 mg/g) were prepared. A uniform distribution of monomeric 1,1'-diethyl-3,3,3',3'-tetramethylindocarbocyanine iodide has been achieved at low dye loading (1 mg/g), as evidenced by photoluminescence spectra and lifetime, while a progressive formation of J-aggregates is induced by an increase in the dye loading. To elucidate the properties of the dye immobilized in mesoporous MCM-41, a detailed physical chemical characterization by structural (X-ray diffraction), volumetric and optical (Fourier transform infrared, diffuse-reflectance UV-vis and photoluminescence) techniques has been performed. By ultrasonication of the bulk material, nanoparticles of 2-20 nm diameter were obtained. Biocompatibility, endocytic uptake, and intracellular compartmentalization of such fluorescent nanoparticles were investigated in mammalian cultured cells.


Subject(s)
Carbocyanines/chemistry , Fluorescent Dyes/chemistry , Nanocomposites/chemistry , Nanoparticles/chemistry , Silicon Dioxide/chemistry , Adsorption , Animals , Carbocyanines/pharmacokinetics , Cell Line, Tumor , Cell Survival , Fluorescent Dyes/pharmacokinetics , Humans , Lysosomes/metabolism , Mice , Porosity , Rats , Spectrometry, Fluorescence , X-Ray Diffraction
13.
J Neurochem ; 106(3): 1426-39, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18489716

ABSTRACT

Methamphetamine abuse is toxic to dopaminergic neurons, causing nigrostriatal denervation and striatal dopamine loss. Following methamphetamine exposure, the number of nigral cell bodies is generally preserved, but their cytoplasm features autophagic-like vacuolization and cytoplasmic accumulation of alpha-synuclein-, ubiquitin- and parkin-positive inclusion-like bodies. Whether autophagy is epiphenomenal or it plays a role in the mechanism of methamphetamine toxicity and, in the latter case, whether its role consists of counteracting or promoting the neurotoxic effect remains obscure. We investigated the signaling pathway and the significance (protective vs. toxic) of autophagy activation and the convergence of the autophagic and the ubiquitin-proteasome pathways at the level of the same intracellular bodies in a simple cell model of methamphetamine toxicity. We show that autophagy is rapidly up-regulated in response to methamphetamine. Confocal fluorescence microscopy and immuno-electron microscopy studies demonstrated the presence of alpha-synuclein aggregates in autophagy-lysosomal structures in cells exposed to methamphetamine, a condition compatible with cell survival. Inhibition of autophagy either by pharmacologic or genetic manipulation of the class III Phosphatidylinositol-3 kinase-mediated signaling prevented the removal of alpha-synuclein aggregates and precipitated a bax-mediated mitochondrial apoptosis pathway.


Subject(s)
Autophagy/drug effects , Methamphetamine/administration & dosage , Neurons/cytology , Neurons/drug effects , Animals , Autophagy/physiology , Cell Death/drug effects , Cell Death/physiology , Cell Line, Tumor , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/physiology , Methamphetamine/toxicity , Mitochondria/drug effects , Mitochondria/pathology , Neurons/pathology , PC12 Cells , Permeability/drug effects , Rats , bcl-2-Associated X Protein/physiology
14.
Endocrinology ; 149(8): 4095-105, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18420735

ABSTRACT

Cathepsin D (CD), a lysosomal aspartic protease present in mammary tissue and milk in various molecular forms, is also found in the incubation medium of mammary acini in molecular forms that are proteolytically active on prolactin at a physiological pH. Because prolactin controls the vesicular traffic in mammary cells, we studied, in vivo and in vitro, its effects on the polarized transport and secretion of various forms of CD in the rat mammary gland. CD accumulated in vesicles not involved in endocytosis in the basal region of cells. Prolactin increased this accumulation and the release of endosomal active single-chain CD at the basal side of acini. The CD-mediated proteolysis of prolactin, leading to the antiangiogenic 16-kDa form, at a physiological pH, was observed only in conditioned medium but not milk. These data support the novel concept that an active molecular form of CD, secreted at the basal side of the mammary epithelium, participates in processing blood-borne prolactin outside the cell, this polarized secretion being controlled by prolactin itself.


Subject(s)
Cathepsin D/metabolism , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/metabolism , Prolactin/pharmacology , Animals , Cell Polarity/physiology , Endocytosis/drug effects , Female , Hydrogen-Ion Concentration , Mammary Glands, Animal/cytology , Milk/metabolism , Models, Biological , Protein Processing, Post-Translational/drug effects , Protein Transport , Rats , Rats, Wistar
15.
Carcinogenesis ; 29(2): 381-9, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18048384

ABSTRACT

In human colorectal DLD1 cancer cells, the dietary bioflavonoid resveratrol (RV) rapidly induced autophagy. This effect was reversible (on removal of the drug) and was associated with increased expression and cytosolic redistribution of the proteins Beclin1 and LC3 II. Supplementing the cells with asparagine (Asn) abrogated the Beclin-dependent autophagy. When applied acutely (2 h), RV was not toxic; however, reiterate chronic (48 h) exposure to RV eventually led to annexin V- and terminal deoxinucleotidyl transferase-mediated dUTP-biotin nick end labeling-positive cell death. This toxic effect was autophagy dependent, as it was prevented either by Asn, by expressing a dominant-negative lipid kinase-deficient class III phosphoinositide 3-phosphate kinase, or by RNA interference knockdown of Beclin1. Lamp2b silencing abolished the fusion of autophagosomes with lysosomes and preserved cell viability despite the ongoing formation of autophagosomes in cells chronically exposed to RV. The pan-caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone inhibited RV-induced cell death, but not autophagy. These results uncover a novel pathway of RV cytotoxicity in which autophagy plays a dual role: (i) at first, it acts as a prosurvival stress response and (ii) at a later time, it switches to a caspase-dependent apoptosis pathway. The present data also indicate that genetic or epigenetic inactivation of autophagy proteins in cancer cells may confer resistance to RV-mediated killing.


Subject(s)
Apoptosis , Enzyme Inhibitors/pharmacology , Phagosomes/metabolism , Phosphatidylinositol 3-Kinases/biosynthesis , Phosphotransferases/metabolism , Stilbenes/pharmacology , Autophagy , Cell Line, Tumor , Epigenesis, Genetic , Gene Silencing , Genes, Dominant , Humans , Lipid Metabolism , Lysosomes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Resveratrol
16.
Am J Physiol Endocrinol Metab ; 293(5): E1365-77, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17785503

ABSTRACT

In the corpus luteum (CL), blood vessels develop, stabilize, and regress. This process depends on the ratio of pro- and antiangiogenic factors, which change during the ovarian cycle. The present study focuses on the possible roles of 23,000 (23K) prolactin (PRL) in the bovine CL and its antiangiogenic NH(2)-terminal fragments after extracellular cleavage by cathepsin D (Cath D). PRL RNA and protein were demonstrated in the CL tissue, in luteal endothelial cells, and in steroidogenic cells. Cath D was detected in CL tissue, cell extracts, and corresponding cell supernatants. In the intact CL, 23K PRL levels decreased gradually, whereas Cath D levels concomitantly increased between early and late luteal stages. In vitro, PRL cleavage occurred in the presence of acidified homogenates of CL tissue, cells, and corresponding cell supernatants. Similar fragments were obtained with purified Cath D, and their appearance was inhibited by pepstatin A. The aspartic protease specific substrate MOCAc-GKPILF~FRLK(Dnp)-D-R-NH(2) was cleaved by CL cell supernatants, providing further evidence for Cath D activity. The 16,000 PRL inhibited proliferation of luteal endothelial cells accompanied by an increase in cleaved caspase-3. In conclusion, 1) the bovine CL is able to produce PRL and to process it into antiangiogenic fragments by Cath D activity and 2) PRL cleavage might mediate angioregression during luteolysis.


Subject(s)
Cathepsin D/metabolism , Corpus Luteum/physiology , Estrous Cycle/physiology , Luteolysis/physiology , Prolactin/biosynthesis , Animals , Aspartic Acid Endopeptidases/metabolism , Caspase 3/metabolism , Cattle , Corpus Luteum/cytology , Corpus Luteum/enzymology , Endothelial Cells/enzymology , Endothelial Cells/physiology , Female , Granulosa Cells/enzymology , Granulosa Cells/physiology , Microscopy, Fluorescence/veterinary , Microscopy, Phase-Contrast/veterinary , Oligopeptides/metabolism , Prolactin/genetics , Prolactin/metabolism , Protein Processing, Post-Translational , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
17.
Free Radic Biol Med ; 42(9): 1305-16, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17395004

ABSTRACT

Hydrogen peroxide, the major oxidoradical species in the central nervous system, has been involved in neuronal cell death and associated neurodegenerative diseases. In this study, we have investigated the involvement of the lysosomal pathway in the cytotoxic mechanism of hydrogen peroxide in human neuroblastoma cells. Alteration of lysosomal and mitochondrial membrane integrity was shown to be an early event in the lethal cascade triggered by oxidative stress. Desferrioxamine (DFO), an iron chelator that abolishes the formation of reactive oxygen species within lysosomes, prevented lysosome leakage, mitochondrial permeabilization and caspase-dependent apoptosis in hydrogen peroxide-treated cells. Inhibition of cathepsin D, not of cathepsin B, as well as small-interference RNA-mediated silencing of the cathepsin D gene prevented hydrogen peroxide-induced injury of mitochondria, caspase activation, and TUNEL-positive cell death. Cathepsin D activity was shown indispensable for translocation of Bax onto mitochondrial membrane associated with oxidative stress. DFO abolished both the cytosolic relocation of Cathepsin D and the mitochondrial relocation of Bax in hydrogen peroxide-treated cells. siRNA-mediated down-regulation of Bax expression protected the cells from oxidoradical injury. The present study identifies the lysosome as the primary target and the axis cathepsin D-Bax as the effective pathway of hydrogen peroxide lethal activity in neuroblastoma cells.


Subject(s)
Cathepsin D/metabolism , Deferoxamine/pharmacology , bcl-2-Associated X Protein/metabolism , Cathepsin D/genetics , Cell Line, Tumor , Cell Survival/drug effects , Humans , Hydrogen Peroxide/pharmacology , Neuroblastoma , Oxidative Stress/drug effects , Pepstatins/pharmacology , RNA, Small Interfering/genetics , Siderophores/pharmacology , Transfection , bcl-2-Associated X Protein/genetics
18.
Int J Biochem Cell Biol ; 39(3): 638-49, 2007.
Article in English | MEDLINE | ID: mdl-17188016

ABSTRACT

The precursor of human cathepsin D (CD) is converted into the single-chain and the double-chain active polypeptides by subsequent proteolysis reactions taking place in the endosomal-lysosomal compartment and involving specific aminoacid sequences. We have mutagenized the region of aminoacids (comprising the beta-hairpin loop) involved in the latter proteolytic maturation step and generated a mutant CD that cannot be converted into the mature double-chain form. This mutant CD expressed in rodent cells reaches the lysosome and is stable as single-chain polypeptide, bears high-mannose type sugars, binds to pepstatin A and is enzymatically active, indicating that it is correctly folded. The present work provides new insights on the aminoacid region involved in the terminal processing of human CD and on the function of the processing beta-hairpin loop.


Subject(s)
Cathepsin D/chemistry , Cathepsin D/genetics , Amino Acid Sequence , Animals , CHO Cells , Cathepsin D/antagonists & inhibitors , Cathepsin D/metabolism , Cell Line , Cricetinae , Cricetulus , Humans , Hydrogen-Ion Concentration , Lysosomes/metabolism , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Pepstatins/metabolism , Pepstatins/pharmacology , Protein Binding , Protein Folding , Protein Processing, Post-Translational , Protein Structure, Quaternary , Protein Transport , Rats , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection
19.
Carcinogenesis ; 28(5): 922-31, 2007 May.
Article in English | MEDLINE | ID: mdl-17116725

ABSTRACT

In human colorectal cancer cells, the polyphenol resveratrol (RV) activated the caspase-dependent intrinsic pathway of apoptosis. This effect was not mediated via estrogen receptors. Pepstatin A, an inhibitor of lysosomal cathepsin D (CD), not (2S,3S)-trans-epoxysuccinyl-L-leucylamido-3-methylbutane ethyl ester, an inhibitor of cathepsins B and L, prevented RV cytotoxicity. Similar protection was attained by small interference RNA-mediated knockdown of CD protein expression. RV promoted the accumulation of mature CD, induced lysosome leakage and increased cytosolic immunoreactivity of CD. Inhibition of CD or its post-transcriptional down-regulation precluded Bax oligomerization, permeabilization of mitochondrial membrane, cytosolic translocation of cytochrome c, caspase 3 activation and terminal deoxinucleotidyl transferase-mediated dUTP-biotin nick end labeling positivity occurring in RV-treated cells. The present study identifies the lysosome as a novel target of RV activity and demonstrates a hierarchy of the proteolytic pathways involved in its cytotoxic mechanism in which the lysosomal CD acts upstream of the cytosolic caspase activation. Our data indicate that metabolic, pharmacologic or genetic conditions affecting CD expression and/or activity could reflect on the sensitivity of cancer cells to RV.


Subject(s)
Cathepsin D/metabolism , Colorectal Neoplasms/metabolism , Lysosomes/metabolism , Stilbenes/pharmacology , Caspase Inhibitors , Cathepsin L , Cathepsins/metabolism , Cell Death/drug effects , Cell Line , Colorectal Neoplasms/pathology , Cysteine Endopeptidases/metabolism , Cytochromes c/metabolism , Cytosol/metabolism , Dose-Response Relationship, Drug , HT29 Cells , Humans , Resveratrol , Time Factors
20.
Free Radic Biol Med ; 40(10): 1738-48, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16678013

ABSTRACT

We investigated the signal mediators and the cellular events involved in the nitric oxide (NO)-induced hepatocyte resistance to oxygen deprivation in isolated hepatocytes treated with the NO donor (Z)-1-(N-methyl-N-[6-(N-methylammoniohexyl)amino])diazen-1-ium-1,2-diolate (NOC-9). NOC-9 greatly induced PI3K activation, as tested by phosphorylation of PKB/Akt. This effect was prevented by either 1H-(1,2,4)-oxadiazolo-(4,3)-quinoxalin-1-one, an inhibitor of the soluble guanylate cyclase (sGC), or KT5823, an inhibitor of cGMP-dependent kinase (cGK), as well as by farnesyl protein transferase inhibitor, which blocks the function of Ras GTPase. Bafilomycin A, an inhibitor of the lysosome-type vacuolar H+-ATPase, cytochalasin D, which disrupts the cytoskeleton-dependent organelle traffic, and wortmannin, which inhibits the PI3K-dependent traffic of lysosomes, all abolished the NOC-9-induced hepatocyte protection. The treatment with NOC-9 was associated with the PI3K-dependent peripheral translocation and fusion with the plasma membrane of lysosomes and the appearance at the cell surface of the vacuolar H+-ATPase. Inhibition of sGC, cGK, and Ras, as well as the inhibition of PI3K by wortmannin, prevented the exocytosis of lysosomes and concomitantly abolished the protective effect of NOC-9 on hypoxia-induced pHi and [Na+]i alterations and cell death. These data indicate that NO increases hepatocyte resistance to hypoxic injury by activating a pathway involving Ras, sGC, and cGK that determines PI3K-dependent exocytosis of lysosomes.


Subject(s)
Exocytosis/drug effects , Hepatocytes/drug effects , Ischemic Preconditioning , Lysosomes/drug effects , Nitric Oxide/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Animals , Cell Hypoxia/drug effects , Cyclic GMP-Dependent Protein Kinases/drug effects , Cyclic GMP-Dependent Protein Kinases/metabolism , Enzyme Inhibitors/pharmacology , Exocytosis/physiology , Guanylate Cyclase , Lysosomes/metabolism , Male , Phosphatidylinositol 3-Kinases/drug effects , Rats , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Soluble Guanylyl Cyclase , Triazenes/pharmacology , ras GTPase-Activating Proteins/drug effects , ras GTPase-Activating Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...