Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cancer Cell Int ; 22(1): 232, 2022 Jul 21.
Article in English | MEDLINE | ID: mdl-35864494

ABSTRACT

BACKGROUND: Glioblastoma is a highly aggressive brain tumor. A big effort is required to find novel molecules which can cross the blood-brain barrier and efficiently kill these tumor cells. In this perspective, trehalose (α-glucopyranosyl-[1→1]-α-D-glucopyranoside), found in various dietary sources and used as a safe nutrient supplement, attracted our attention for its pleiotropic effects against tumor cells. METHODS: Human glioblastoma cell lines U373-MG and T98G were exposed to trehalose and analyzed at different time points. Cell proliferation was evaluated at medium term, and clonogenic capacity and cell morphology were evaluated at long term. Western blot was used to evaluate biochemical markers of autophagy (also measured in cells co-treated with EIPA or chloroquine), and mTOR, AMPK and ERK 1/2 signalling. Macropinocytosis was evaluated morphologically by bright-field microscopy; in cells loaded with the fluorescein-conjugated fluid-phase tracer dextran, macropinocytic vacuoles were also visualized by fluorescence microscopy, and the extent of macropinocytosis was quantified by flow cytometry. RESULTS: The long-term effect of trehalose on U373-MG and T98G cell lines was impressive, as indicated by a dramatic reduction in clonogenic efficiency. Mechanistically, trehalose proved to be an efficient autophagy inducer in macropinocytosis-deficient T98G cells and an efficient inducer of macropinocytosis and eventual cell death by methuosis in U373-MG glioblastoma cells, proved to be poorly responsive to induction of autophagy. These two processes appeared to act in a mutually exclusive manner; indeed, co-treatment of U373-MG cells with the macropinocytosis inhibitor, EIPA, significantly increased the autophagic response. mTOR activation and AMPK inhibition occurred in a similar way in the two trehalose-treated cell lines. Interestingly, ERK 1/2 was activated only in macropinocytosis-proficient U373-MG cells harbouring loss-of-function mutations in the negative RAS regulator, NF1, suggesting a key role of RAS signalling. CONCLUSIONS: Our results indicate that trehalose is worthy of further study as a candidate molecule for glioblastoma therapy, due to its capacity to induce a sustained autophagic response, ultimately leading to loss of clonogenic potential, and more interestingly, to force macropinocytosis, eventually leading to cell death by methuosis, particularly in tumor cells with RAS hyperactivity. As a further anticancer strategy, stimulation of macropinocytosis may be exploited to increase intracellular delivery of anticancer drugs.

2.
Autoimmunity ; 53(5): 245-252, 2020 08.
Article in English | MEDLINE | ID: mdl-32338081

ABSTRACT

Autoantibodies to nuclear and cytoplasmic antigens are commonly detected by indirect immunofluorescence (IIF) on HEp-2 cells, and three major staining patterns (nuclear, cytoplasmic, and mitotic) are distinguished. Here, we report an atypical cytoplasmic pattern, not described so far, observed in the serum of a patient with a controversial diagnosis of systemic lupus erythematosus (SLE). Moreover, for the first time, we have revealed the presence of autoantibodies against the microtubule-associated light-chain 3 (LC3) protein, which plays a key role in the autophagic process. The target antigen has been identified in IIF by means of a competition test using purified anti-LC3 antibodies on HEp-2 cells, and confirmed by Western blot analysis using cellular or recombinant LC3 as antigen, immunoreacted with the patient's serum. The identification of this atypical pattern and the related autoantibody-antigen system sheds new light on autophagy, which is increasingly considered to be involved in the etiopathogenesis of autoimmune disorders, and could contribute to select more personalized therapies.


Subject(s)
Antibodies, Antinuclear/blood , Autophagy/immunology , Lupus Erythematosus, Systemic/diagnosis , Microtubule-Associated Proteins/immunology , Adult , Cell Line, Tumor , Female , Fluorescent Antibody Technique, Indirect/methods , Humans , Lupus Erythematosus, Systemic/blood
3.
Fertil Steril ; 112(6): 1150-1159, 2019 12.
Article in English | MEDLINE | ID: mdl-31843092

ABSTRACT

OBJECTIVE: To assess the effect of ulipristal acetate (UPA) on the autophagic process of uterine leiomyoma cells. DESIGN: In vitro study in primary cultures of leiomyoma and myometrial cells isolated from biopsy specimen, and gene expression evaluation in biopsy material. SETTING: Cellular pathology laboratory. PATIENT(S): Premenopausal women (without hormonal treatment) undergoing myomectomy or hysterectomy for symptomatic leiomyomas. INTERVENTION(S): Surgical specimens collected from uterine leiomyomas and matched normal myometria. MAIN OUTCOME MEASURE(S): After treatment of myometrial and leiomyoma cells with UPA, autophagy was evaluated by Western blot analysis of the typical biochemical markers, LC3-II, LC3-II:LC3-I ratio, and p62/SQSTM1. The expression level of Atg7 and Atg4D proteins was also assessed by Western blot. RESULT(S): The increase of LC3-II protein, LC3-II:LC3-I ratio, and p62/SQSTM1 protein indicates that UPA treatment up-regulates the autophagic response in leiomyoma cells, whereas these markers were almost unchanged in myometrial cells. Consistently, an increased level of Atg7 and Atg4D proteins was observed only in UPA-treated leiomyoma cells. The autophagic machinery is put into motion selectively in these cells, despite that the basal messenger RNA levels of LC3, SQSTM1, and ATG7 in leiomyoma biopsy specimen were not significantly different from those found in normal myometrial biopsy material. CONCLUSION(S): In vitro UPA treatment stimulates the autophagic response selectively in leiomyoma cells, which adds a novel indication for the clinical use of this selective P receptor (PR) modulator. Autophagy up-regulation may potentially contribute to the leiomyoma shrinkage occurring in UPA-treated patients and warrants further study.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Autophagy-Related Proteins/metabolism , Autophagy/drug effects , Leiomyoma/drug therapy , Norpregnadienes/pharmacology , Uterine Neoplasms/drug therapy , Adult , Female , Humans , Leiomyoma/metabolism , Leiomyoma/pathology , Middle Aged , Signal Transduction , Tumor Cells, Cultured , Uterine Neoplasms/metabolism , Uterine Neoplasms/pathology
4.
J Cell Physiol ; 234(7): 11708-11721, 2019 07.
Article in English | MEDLINE | ID: mdl-30488547

ABSTRACT

Cutaneous melanomas frequently metastasize to the brain, with temozolomide (TMZ) plus radiotherapy (RT) offering little control of these lesions. We tested whether trehalose, a natural glucose disaccharide proved to induce autophagy, could enhance the effect of TMZ and ionizing radiation (IR). In two melanoma cell lines (A375 and SK-Mel-28), which greatly differ in chemosensitivity and radiosensitivity, trehalose significantly inhibited short-term cell proliferation and also enhanced IR-induced cytostasis. Interestingly, in TMZ-resistant SK-Mel-28 cells, trehalose was more effective than TMZ, and combined trehalose + TMZ further reduced cell proliferation. In long-term experiments, colony-forming capacity was dramatically reduced by trehalose, and even more by combined trehalose + TMZ or trehalose + IR. In resistant SK-Mel-28 cells, although growth was inhibited most with trehalose + TMZ + IR-6 Gy combined treatment, it is notable that trehalose + TMZ treatment was also very effective. Along with a direct antiproliferative effect, two further mechanisms may explain how trehalose potentiates TMZ- and IR-induced effects: the remarkable trehalose-stimulated autophagy in A375 cells, which were sensitive to TMZ- and IR-induced apoptosis; and the notable trehalose-stimulated premature senescence in SK-Mel-28 cells, which were resistant to apoptosis and less prone to autophagy. In normal melanocytes, trehalose induced a minor autophagy and cell proliferation inhibition, without affecting cell viability; moreover, when trehalose was used in combination with TMZ, the slight TMZ-induced cytotoxicity was not significantly reinforced. Together, our results suggest that trehalose, a safe nutrient supplement able to cross the blood-brain barrier, is a promising candidate, worthy to be further explored in vivo, to augment the therapeutic efficacy of TMZ and RT in melanoma brain metastases.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Cellular Senescence/drug effects , Melanoma/pathology , Melanoma/radiotherapy , Radiation, Ionizing , Temozolomide/pharmacology , Trehalose/pharmacology , Apoptosis/radiation effects , Autophagy/radiation effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cellular Senescence/radiation effects , Chloroquine/pharmacology , Drug Resistance, Neoplasm/drug effects , Humans , Melanocytes/drug effects , Melanocytes/metabolism , Melanocytes/pathology , Melanocytes/radiation effects , Sequestosome-1 Protein/metabolism
5.
Fertil Steril ; 103(5): 1244-51.e1, 2015 May.
Article in English | MEDLINE | ID: mdl-25772769

ABSTRACT

OBJECTIVE: To evaluate the occurrence of the autophagic process in ovarian endometriomas compared with eutopic endometrium of affected women and with normal endometrium of healthy women. DESIGN: Biochemical and molecular study in tissue extracts. SETTING: University cellular pathology laboratory and university hospital. PATIENT(S): Patients with ovarian endometriosis (n = 13) and healthy women (n = 18). INTERVENTION(S): Specimens of endometrium were obtained by hysteroscopy from patients with endometriosis and from healthy control subjects; specimens of ovarian endometriomas were collected by laparoscopy. All patients underwent surgery after the end of menstrual bleeding, resulting in most of our patients (approximately 80% in each group) being in the proliferative phase. MAIN OUTCOME MEASURE(S): Autophagy was evaluated by Western blot analysis of biochemical markers (LC3-II, LC3-II/LC3-I ratio and p62) and by quantitative real-time polymerase chain reaction of autophagy-related genes (ATG14, BECN1, ATG7, and LC3B); apoptosis-related (p53 and Bcl-2) and oxidative stress-related (heme oxygenase-1) proteins were also evaluated by Western blot analysis. RESULT(S): All tested biochemical markers and messenger RNA levels of autophagy-related genes showed a significant up-regulation of autophagy in ovarian endometriomas compared with eutopic endometria of affected or healthy women. Moreover, a significant decrease of p53 protein and a significant increase of heme oxygenase-1 protein was also evident in endometriomas. CONCLUSION(S): The upregulated autophagic process observed in ovarian endometriomas can be regarded as an integral part of endometriosis pathogenesis, possibly contributing to survival of endometriotic cells in ectopic sites and to lesion maintenance. The decreased susceptibility to apoptosis and the persistent oxidative stress experienced by endometriotic cells could favor autophagy stimulation.


Subject(s)
Autophagy , Endometriosis/enzymology , Heme Oxygenase-1/analysis , Ovary/enzymology , Tumor Suppressor Protein p53/analysis , Adult , Autophagy/genetics , Case-Control Studies , Endometriosis/genetics , Endometriosis/pathology , Endometriosis/surgery , Female , Gene Expression Regulation , Heme Oxygenase-1/genetics , Hospitals, University , Humans , Ovary/pathology , Ovary/surgery , RNA, Messenger/analysis , Signal Transduction , Tumor Suppressor Protein p53/genetics , Young Adult
6.
PLoS One ; 10(2): e0117258, 2015.
Article in English | MEDLINE | ID: mdl-25658320

ABSTRACT

Calpain-3 is an intracellular cysteine protease, belonging to Calpain superfamily and predominantly expressed in skeletal muscle. In human melanoma cell lines and biopsies, we previously identified two novel splicing variants (hMp78 and hMp84) of Calpain-3 gene (CAPN3), which have a significant lower expression in vertical growth phase melanomas and, even lower, in metastases, compared to benign nevi. In the present study, in order to investigate the pathophysiological role played by the longer Calpain-3 variant, hMp84, in melanoma cells, we over-expressed it in A375 and HT-144 cells. In A375 cells, the enforced expression of hMp84 induces p53 stabilization, and modulates the expression of a few p53- and oxidative stress-related genes. Consistently, hMp84 increases the intracellular production of ROS (Reactive Oxygen Species), which lead to oxidative modification of phospholipids (formation of F2-isoprostanes) and DNA damage. Such events culminate in an adverse cell fate, as indicated by the decrease of cell proliferation and by cell death. To a different extent, either the antioxidant N-acetyl-cysteine or the p53 inhibitor, Pifithrin-α, recover cell viability and decrease ROS formation. Similarly to A375 cells, hMp84 over-expression causes inhibition of cell proliferation, cell death, and increase of both ROS levels and F2-isoprostanes also in HT-144 cells. However, in these cells no p53 accumulation occurs. In both cell lines, no significant change of cell proliferation and cell damage is observed in cells over-expressing the mutant hMp84C42S devoid of its enzymatic activity, suggesting that the catalytic activity of hMp84 is required for its detrimental effects. Since a more aggressive phenotype is expected to benefit from down-regulation of mechanisms impairing cell growth and survival, we envisage that Calpain-3 down-regulation can be regarded as a novel mechanism contributing to melanoma progression.


Subject(s)
Calpain/metabolism , Cell Proliferation , Melanoma/pathology , Muscle Proteins/metabolism , Oxidative Stress , Benzothiazoles/pharmacology , Calpain/analysis , Calpain/genetics , Cell Death , Cell Line, Tumor , Humans , Melanoma/genetics , Melanoma/metabolism , Muscle Proteins/analysis , Muscle Proteins/genetics , Mutation , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Toluene/analogs & derivatives , Toluene/pharmacology , Tumor Suppressor Protein p53/metabolism , Up-Regulation
7.
Arch Biochem Biophys ; 564: 26-36, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25305531

ABSTRACT

Calpains are a complex family of ubiquitous or tissue-specific cysteine proteases that proteolyze a variety of substrates (leading to their degradation or functional modulation) and are implicated in several pathophysiological phenomena. In tumor cell biology, calpains are implicated in a triple way: they are involved in different processes crucial for tumor progression, including cell proliferation, apoptotic cell death, survival mechanisms, migration and invasiveness; they have aberrant expression in several human cancers; a variety of anticancer drugs induce cytotoxicity through activation of calpains or the latter can influence response to therapy. This review covers established and recent literature showing these diverse aspects in tumor cells.


Subject(s)
Apoptosis , Calpain/biosynthesis , Cell Proliferation , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , Neoplasms/enzymology , Animals , Antineoplastic Agents/therapeutic use , Calpain/antagonists & inhibitors , Calpain/genetics , Cysteine Proteinase Inhibitors/therapeutic use , Enzyme Activation , Humans , Neoplasm Invasiveness , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology
8.
Eur J Pharm Biopharm ; 88(2): 424-33, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24931342

ABSTRACT

We have developed a series of pH- and temperature-stimuli-sensitive vinyl hydrogels, bearing α-amino acid residues (L-phenylalanine, L-valine) and incorporating magnetic nanoparticles of different chemical compositions (CoFe2O4 and Fe3O4). The goal was to study the potential applications of these nanocomposites in the controlled release of doxorubicin (DOXO), a potent anticancer drug. The strength of the electrostatic interaction between the protonated nitrogen of the DOXO molecule and the ionized carboxylic groups of the hydrogel allowed effective control of the drug release rate in saline solutions. The embedded magnetic nanoparticles were an additional remote control of the drug release under the stimulus of an appropriate external alternating magnetic field (AMF). Data showed that the controlled release of DOXO proceeded for months and followed a diffusion-controlled release mechanism, while maintaining the amount of released drug within acceptable therapeutic windows. The amount of the released DOXO was found in all cases substantially higher than the "control" because the application of the AMF augments in stimulating the nanoparticles within the DOXO-loaded hydrogel. In vitro experiments have shown that the released DOXO is able to induce cell death to cervix adenocarcinoma cells (HeLa cells).


Subject(s)
Amino Acids/chemistry , Antibiotics, Antineoplastic/administration & dosage , Doxorubicin/administration & dosage , Hydrogels , Antibiotics, Antineoplastic/pharmacokinetics , Doxorubicin/pharmacokinetics , HeLa Cells , Humans , In Vitro Techniques , Magnetics , Spectroscopy, Fourier Transform Infrared
9.
PLoS One ; 8(2): e57236, 2013.
Article in English | MEDLINE | ID: mdl-23437349

ABSTRACT

The interplay between a non-lethal autophagic response and apoptotic cell death is still a matter of debate in cancer cell biology. In the present study performed on human melanoma cells, we investigate the role of basal or stimulated autophagy in cisplatin-induced cytotoxicity, as well as the contribution of cisplatin-induced activation of caspases 3/7 and conventional calpains. The results show that, while down-regulating Beclin-1, Atg14 and LC3-II, cisplatin treatment inhibits the basal autophagic response, impairing a physiological pro-survival response. Consistently, exogenously stimulated autophagy, obtained with trehalose or calpains inhibitors (MDL-28170 and calpeptin), protects from cisplatin-induced apoptosis, and such a protection is reverted by inhibiting autophagy with 3-methyladenine or ATG5 silencing. In addition, during trehalose-stimulated autophagy, the cisplatin-induced activation of calpains is abrogated, suggesting the existence of a feedback loop between the autophagic process and calpains. On the whole, our results demonstrate that in human melanoma cells autophagy may function as a beneficial stress response, hindered by cisplatin-induced death mechanisms. In a therapeutic perspective, these findings suggest that the efficacy of cisplatin-based polychemotherapies for melanoma could be potentiated by inhibitors of autophagy.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Cisplatin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Melanoma/genetics , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Proteins , Beclin-1 , Caspase 3/genetics , Caspase 3/metabolism , Caspase 7/genetics , Caspase 7/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Dipeptides/pharmacology , Feedback, Physiological/drug effects , Humans , Melanoma/metabolism , Melanoma/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Signal Transduction/drug effects , Trehalose/pharmacology
10.
Acta Diabetol ; 50(4): 489-95, 2013 Aug.
Article in English | MEDLINE | ID: mdl-21437568

ABSTRACT

An increased oxidative stress and a decreased life span of erythrocytes (RBCs) are reported in patients with diabetes. Aim of this study was to assess in RBCs from patients with type 2 diabetes whether downstream effector mechanisms of apoptosis, such as activation of caspase-3, is operative, and whether an iron-related oxidative imbalance, occurring inside RBCs and in plasma, could be involved in caspase-3 activation. In 26 patients with type 2 diabetes and in 12 healthy subjects, oxidative stress was evaluated by means of different markers; non-protein-bound iron, methemoglobin and glutathione were determined in RBCs, and non-protein-bound iron was also determined in plasma. Erythrocyte caspase-3 activation was evaluated by an immunosorbent enzyme assay. Arterial hypertension, demographic and standard biochemical data were also evaluated. The results show, for the first time, that type 2 diabetic RBCs put into motion caspase-3 activation, which is significantly higher than in control RBCs. Such an effector mechanism of "eryptosis" was positively correlated to blood glucose levels and to the increased plasma NPBI level. Caspase-3 activation was also positively correlated to occurrence of arterial hypertension. The results suggest that an extracellular oxidative milieu can be responsible for erythrocyte caspase-3 activation in patients with type 2 diabetes. In turn, caspase-3 activation can be envisaged as a novel mechanism which, by impairing the maintenance of erythrocyte shape and function, might contribute to the shortened life span of RBCs from patients with type 2 diabetes and to hemorheological disorders observed in these patients.


Subject(s)
Caspase 3/metabolism , Diabetes Mellitus, Type 2/enzymology , Erythrocytes/enzymology , Oxidative Stress , Aged , Aged, 80 and over , Apoptosis , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/physiopathology , Enzyme Activation , Erythrocytes/cytology , Erythrocytes/metabolism , Female , Humans , Male , Middle Aged
11.
Colloids Surf B Biointerfaces ; 88(1): 389-95, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21816582

ABSTRACT

Two acrylic hydrogels, of low cross-linking content and carrying the L-valine residues, were synthesized and studied as a platform to load and release the chemotherapeutic agent cisplatin. The platinum(II)-complex species showed a well-defined stoichiometric ratio in which two carboxylate groups of the collapsing gel coordinate a metal center; this was confirmed by FT-IR spectra. When loaded in water, a zero-order release rate of platinum(II)-species was shown in the physiologic solution (PBS, pH 7.40) for more than one week. Moreover, the amount of platinum(II)-species released from the hydrogel may be improved either by the cross-linking degree and by the temperature. Any increase of the cross-links results in a decreased slope of the straightline Pt(II)/gel (mg/g) versus time, whereas the increasing temperature results in a greater amount of platinum(II)-species in solution. The chemical- and swelling-controlled release are the main mechanisms supervising the whole release process. On the other hand, the loading of cisplatin and temsirolimus in DMF showed a characteristic two phase releasing pattern; the initial burst effect was always followed by the zero-order release rate for a week. In this case only a swelling-controlled mechanism was mainly invoked. The cytotoxic activity towards Me665/2/21 human melanoma cell line, afforded by the cisplatin-loaded hydrogel, was close and in some cases higher compared to the native cisplatin at the same concentration; an interesting synergy in term of cytotoxicity was observed when a combined treatment of temsirolimus and cisplatin was used, although temsirolimus exerted only a moderate inhibition of cell proliferation.


Subject(s)
Antineoplastic Agents/chemistry , Cisplatin/chemistry , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Valine/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cisplatin/pharmacology , Humans , Hydrogen-Ion Concentration , Sirolimus/analogs & derivatives , Sirolimus/chemistry , Sirolimus/pharmacology , Temperature
12.
Biomacromolecules ; 10(4): 944-9, 2009 Apr 13.
Article in English | MEDLINE | ID: mdl-19254026

ABSTRACT

Hydrogels containing alpha-amino acid residues (L-phenylalanine, L-histidine) were used to complex the chemotherapeutic agent cisplatin. The release of the drug in phosphate buffer solution showed an initial burst effect, followed by a near zero-order release phase over the seven days of reported period. Unlike the nonreleasing pattern of the hydrogel poly(N-acryloyl-L-phenylalanine-co-N-isopropylacrylamide) (CP2), the homopolymer poly(N-acryloyl-L-phenylalanine) (P9) hydrogel showed a released amount of cisplatin loaded from a water/DMSO mixture that was three times greater than that loaded from simple water. The hydrogel P9 formed with cisplatinum(II) complex species of well-defined stoichiometry; the drug species was released by a chemically controlled process. The Pt(II)/L (L is the monomeric unit of the polymer) stoichiometric molar ratio of 0.5, corresponding to two close carboxylate groups per Pt(II), was found by the viscometric data on the soluble polymer analogue. The platinum species released from cisplatin-loaded (from water) hydrogel retained its cytotoxic activity toward Me665/2/21 human melanoma cell line, in the same manner shown by the native cisplatin. On the contrary, the platinum species released from cisplatin-loaded (from water/DMSO) hydrogel was devoid of any cytotoxic effect.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Cisplatin/pharmacokinetics , Drug Delivery Systems , Hydrogels/chemistry , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Acrylamides/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Cisplatin/administration & dosage , Cisplatin/pharmacology , Delayed-Action Preparations , Dimethyl Sulfoxide/chemistry , Drug Carriers , Histidine , Humans , Melanoma/metabolism , Melanoma/pathology , Phenylalanine , Polymers/chemistry , Skin Neoplasms/metabolism , Skin Neoplasms/secondary , Tumor Cells, Cultured , Water/chemistry
13.
Int J Oncol ; 28(2): 345-52, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16391788

ABSTRACT

Macrophage migration inhibitory factor (MIF) is a widely expressed cytokine involved in various biological processes. Although MIF's functions in cancer have not been completely elucidated, its expression has usually been correlated with tumour progression and aggressiveness, and it is currently discussed as a new promising target for novel therapies. Recent studies seem to confirm its active role in melanoma pathobiology; however, its expression has not yet been extensively studied in melanocytic tumours. We evaluated MIF protein expression in 126 skin lesions, including benign and atypical nevi, melanoma and melanoma metastases. In 55 cases, we also assessed MIF mRNA expression by real-time RT-PCR. Benign nevi were subdivided into nevocytic and Spitz/blue types; and melanomas into the radial, and vertical growth phase. A strong cytoplasmic MIF positivity was found in most samples, although it was more heterogeneous in malignant tumours; MIF nuclear expression characterized Spitz/blue nevi, atypical nevi, melanomas and metastases. All samples expressed MIF mRNA but it was significantly lower in benign nevi vs atypical nevi, melanomas and metastases (p=0.001; p<0.0001; p=0.002, respectively). Our study shows a widespread distribution of MIF among melanocytic tumours. Whereas we observed a trend towards higher expression levels of mRNA in atypical and malignant tumours, MIF protein was highly expressed in all lesions, although limited to the cytoplasm in most benign nevi. These observations suggest differences in MIF protein storage, subcellular location and properties in most benign nevi vs atypical and malignant tumours that should be confirmed by further investigation and correlation with clinical outcome.


Subject(s)
Macrophage Migration-Inhibitory Factors/metabolism , Melanoma/metabolism , Nevus, Pigmented/metabolism , RNA, Messenger/metabolism , Skin Neoplasms/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , Humans , Immunohistochemistry , Macrophage Migration-Inhibitory Factors/genetics , Melanoma/pathology , Neoplasm Metastasis , Nevus, Pigmented/pathology , Reverse Transcriptase Polymerase Chain Reaction , Skin Neoplasms/pathology
14.
J Biol Chem ; 279(20): 21318-26, 2004 May 14.
Article in English | MEDLINE | ID: mdl-14990581

ABSTRACT

c-Myc plays an essential role in proliferation, differentiation, and apoptosis. Because of its relevance to cancer, most studies have focused on the cellular consequences of c-Myc overexpression. Here, we address the role of physiological levels of c-Myc in drug-induced apoptosis. By using c-MYC null cells we confirm and extend recent reports showing a c-Myc requirement for the induction of apoptosis by a number of anticancer agents. In particular, we show that c-Myc is required for the induction of apoptosis by doxorubicin and etoposide, whereas it is not required for camptothecin-induced cell death. We have investigated the molecular mechanisms involved in executing doxorubicin-induced apoptosis and show caspase-3 activation by both mitochondria-dependent and -independent pathways. Moreover, serine proteases participate in doxorubicin-induced apoptosis partly by contributing to caspase-3 activation. Finally, a complete rescue from doxorubicin-induced apoptosis is obtained only when serine proteases, caspase-3, and mitochondrial activation are inhibited simultaneously. Interestingly, doxorubicin requires c-Myc for the activation of all of these pathways. Our findings therefore support a model in which doxorubicin simultaneously triggers multiple c-Myc-dependent apoptosis pathways.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Doxorubicin/pharmacology , Genes, myc/genetics , Serine Endopeptidases/metabolism , Animals , Antineoplastic Agents/pharmacology , Caspase 3 , Cell Division/drug effects , Cell Line , Enzyme Activation/drug effects , Rats
15.
Exp Cell Res ; 293(2): 302-10, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14729468

ABSTRACT

Apoptosis protease-activating factor-1 (Apaf-1), the central element in the mitochondrial pathway of apoptosis, is frequently absent or poorly expressed in metastatic melanomas, a tumor type showing a low degree of spontaneous apoptosis and a poor response to conventional therapies. In the present study, we used the Apaf-1-positive Me665/2/21 melanoma cell line to investigate the fate of Apaf-1 during cisplatin-induced apoptosis. As novel findings described for the first time in melanoma cells, we observed that Apaf-1 was markedly decreased during apoptosis, already at early stages of cell damage; concurrently, an immunoreactive N-terminal fragment of congruent with 26 kDa was evident. In spite of the remarkable decrease of Apaf-1 in apoptotic cells, caspase-9 was found to be processed and enzymatically active. Both Apaf-1 depletion and its proteolytic cleavage were markedly prevented in presence of the caspase-3/-7 inhibitor ac-DEVD-CHO. In presence of ac-DEVD-CHO, caspase-9 activity was also inhibited, along with a partially different pattern of caspase-9 processing forms. Unexpectedly, the inhibition afforded by ac-DEVD-CHO on several components, that is, caspase-3/-7 and caspase-9 activities, and Apaf-1 proteolytic degradation, did not abrogate the apoptotic morphology and cell detachment, nor the proteolytic degradation of crucial targets, such as poly(ADP-ribose) polymerase (PARP) and lamin B. Together, our results suggest that caspase-3 and -7, proved to be dispensable for the above apoptosis-associated events, play a role on Apaf-1 handling and possibly on apoptosome function.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Cisplatin/pharmacology , Melanoma/enzymology , Proteins/metabolism , Apoptosis/physiology , Apoptotic Protease-Activating Factor 1 , Caspase 3 , Caspase 7 , Caspase Inhibitors , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Line, Tumor , Cytochromes c/metabolism , Down-Regulation/drug effects , Down-Regulation/physiology , Drug Resistance, Neoplasm/physiology , Enzyme Inhibitors/pharmacology , Humans , Lamin Type B/metabolism , Macromolecular Substances , Melanoma/drug therapy , Mitochondria/drug effects , Mitochondria/enzymology , Peptide Fragments/metabolism , Peptide Hydrolases/drug effects , Peptide Hydrolases/metabolism , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases , Proteins/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology
16.
Int J Oncol ; 23(1): 213-20, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12792796

ABSTRACT

Intrinsic and acquired multidrug-resistance (MDR) and the activity of the enzyme telomerase have been demonstrated in human melanoma. A direct regulation of the MDR pathways and of telomerase by interpheron-alpha (IFN-alpha), which is currently used in the therapy of advanced cutaneous melanoma, has also been hypothesized. In this study, we used five melanoma cell lines not selected in vitro for drug resistance (Me665/2/21, Me665/2/60, HT-144, SK-MEL-28, and SK-MEL-5), which in a previous study, had shown different responses to IFN-alpha in terms of proliferation, apoptosis, telomerase activity and expression of mRNA for the human telomerase reverse transcriptase (hTERT). We investigated the expression of the multidrug resistance (MDR1) gene, multidrug resistance protein (MRP), lung resistance protein (LRP), topoisomerase IIalpha (Topo IIalpha), hTERT, and telomerase-associated protein (TEP1), which is shared by telomerase and vault MDR proteins at the mRNA expression level, using the reverse transcription-PCR (RT-PCR). All cell lines showed an intrinsic expression of hTERT, TEP1, and MDR gene transcripts (only MDR1 mRNA was under the detection level in SK-MEL-28 cells). After IFN-alpha exposure, we observed either no effect, a trend towards a decrease of hTERT, MRP, and Topo IIalpha, or an increase of TEP1, MDR1, and LRP mRNA expression in some cell lines. Effects were usually temporary and not always significant. No correlation was found between hTERT and TEP1 mRNA expression, whereas significant positive correlations were found between TEP1 and MDR1 mRNA, and between TEP1 and LRP mRNA. IFN-alpha modulates differently MDR gene transcripts in human melanoma cell lines. Positive correlation between TEP1 and LRP also seems to identify them as common targets of IFN-alpha effects.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Carrier Proteins/biosynthesis , DNA Topoisomerases, Type II/biosynthesis , Gene Expression Regulation, Neoplastic , Interferon-alpha/therapeutic use , Neoplasm Proteins/biosynthesis , Telomerase/biosynthesis , Vault Ribonucleoprotein Particles/biosynthesis , Antigens, Neoplasm , Cell Line, Tumor , DNA Fragmentation , DNA-Binding Proteins , Drug Resistance, Neoplasm , Flow Cytometry , Humans , Melanoma/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
17.
Biochim Biophys Acta ; 1621(1): 76-83, 2003 Apr 07.
Article in English | MEDLINE | ID: mdl-12667613

ABSTRACT

Alterations of protein kinase and protein phosphatase activities have been described in a number of tumors. Redox changes, such as in conditions of oxidant stress, have been reported to affect the cellular protein kinase/phosphatase balance. A basal production of reactive oxygen species (ROS), such as hydrogen peroxide (H(2)O(2)), exists in tumor cells, and the membrane-bound ecto-enzyme gamma-glutamyltransferase (GGT)-overexpressed in a variety of malignant tumors-is one of the mechanisms capable of promoting such a production. The present study was aimed to verify the interactions of GGT activity with protein phosphatase and kinase activities in Me665/2/60 melanoma cells, expressing high levels of this enzyme and exhibiting both basal and GGT-dependent production of hydrogen peroxide. An increase of total phosphatase as well as tyrosine phosphatase activities was observed after treatment of cells with both micromolar H(2)O(2) and GGT stimulation. Accordingly, stimulation of GGT resulted in decreased levels of phosphotyrosine. On the other hand, when serine/threonine phosphatase activities were selectively analyzed, both H(2)O(2) treatment and GGT stimulation caused their down-regulation.The data reported suggest that basal conditions of oxidant stress in melanoma may represent a factor contributing to the redox regulation of protein phosphorylation, and that GGT-mediated prooxidant reactions may participate in the process. As basal oxidant stress and expression of GGT activity are present in a variety of malignant tumors besides melanoma, these phenomena likely represent general mechanisms participating in the alteration of intracellular transduction during carcinogenesis.


Subject(s)
Hydrogen Peroxide/metabolism , Melanoma/enzymology , Phosphoprotein Phosphatases/metabolism , Protein Kinases/metabolism , gamma-Glutamyltransferase/metabolism , Cell Membrane/enzymology , Enzyme Activation , Humans , Hydrogen Peroxide/analysis , Oxidation-Reduction , Oxidative Stress , Phosphoprotein Phosphatases/analysis , Phosphotyrosine/analysis , Protein Serine-Threonine Kinases/analysis , Protein Tyrosine Phosphatases/analysis , Protein-Tyrosine Kinases/analysis , Tumor Cells, Cultured , gamma-Glutamyltransferase/biosynthesis
18.
Ann N Y Acad Sci ; 1010: 200-4, 2003 Dec.
Article in English | MEDLINE | ID: mdl-15033720

ABSTRACT

Apoptosis protease-activating factor-1 (Apaf-1), which plays a central role in the formation of the apoptosome, is absent or poorly expressed (because of a transcriptional silencing by methylation) in a substantial percentage of metastatic melanomas and melanoma cell lines, which are unable to activate caspase-9 and execute the mitochondrial pathway of apoptosis. We studied cisplatin-induced apoptosis of the Apaf-1-positive human metastatic Me665/2/21 melanoma cells. Our results indicate that caspase-7 is already processed in still-adhering cells and such activation, contrary to the common view, precedes caspase-3 processing. As expected by the cytochrome c release into the cytosol, caspase-9 is processed to active forms (p37 and p35), along with a yet-unidentified p28. Interestingly, we also demonstrate a remarkable loss of Apaf-1 protein, along with the appearance of a related immunoreactive fragment of approximate, equals 26 kDa; such proteolytic degradation proves to be a caspase-3/-7-mediated event. Our data also indicate that the inhibition afforded by ac-DEVD-CHO on several components (i.e., caspase-3/-7 and caspase-9 activities), and Apaf-1 proteolytic degradation, does not significantly abrogate either the apoptotic morphology or the cleavage of canonical targets, such as poly(ADP-ribose) polymerase (PARP) and lamin B. These results suggest that caspase-3 and caspase-7 are dispensable for the execution of apoptosis and, in our cellular model, the point of no return could be out of the mitochondrial cascade.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Cisplatin/toxicity , Proteins/metabolism , Apoptotic Protease-Activating Factor 1 , Caspase 3 , Caspase 7 , Cell Line, Tumor , Cysteine Proteinase Inhibitors/pharmacology , Humans , Melanoma/pathology , Neoplasm Metastasis
SELECTION OF CITATIONS
SEARCH DETAIL
...