Effettua una ricerca
Antonella Muscella
Ruolo
Ricercatore
Organizzazione
Università del Salento
Dipartimento
Dipartimento di Scienze e Tecnologie Biologiche ed Ambientali
Area Scientifica
Area 06 - Scienze mediche
Settore Scientifico Disciplinare
MED/04 - Patologia Generale
Settore ERC 1° livello
Non Disponibile
Settore ERC 2° livello
Non Disponibile
Settore ERC 3° livello
Non Disponibile
We previously showed that [Pt(O,O'-acac)(γ-acac)(DMS)] ([Pt(acac)2(DMS)]) exerted substantial cytotoxic effects in SH-SY5Y neuroblastoma cells, and decreased metalloproteases (MMPs) production and cells migration in MCF-7 breast cancer cells. The ubiquitously distributed sodium-hydrogen antiporter 1 (NHE1) is involved in motility and invasion of many solid tumours. The present study focuses on the effects of [Pt(acac)2(DMS)] in SH-SY5Y cell migration and also on the possibility that NHE1 may be involved in such effect. After sublethal [Pt(acac)2(DMS)] treatment cell migration was examined by wounding assay and cell invasion by transwell assay. NHE1 activity was measured in BCECF-loaded SH-SY5Y as the rate of Na+-dependent intracellular pH recovery in response to an acute acid pulse. Gelatin zymography for MMP-2/9 activities, Western blottings of MMPs, MAPKs, mTOR, S6 and PKCs and small interfering RNAs to PKC-ε/-δ mRNA were performed. Sublethal concentrations of [Pt(acac)2(DMS)] decreases NHE1 activity, inhibits cell migration and invasion and decreases expression and activity of MMP-2 and -9. [Pt(acac)2(DMS)] administered to SH-SY5Y cells provokes the increment of ROS, generated by NADPH oxidase, responsible for the PKC-ε and PKC-δ activation. Whilst PKC-δ activates p38/MAPK, responsible for the inhibition of MMP-2 and -9 secretion, PKC-ε activates a pathway made of ERK1/2, mTOR and S6K responsible for the inhibition of NHE1 activity and cell migration. In conclusion, we have shown a drastic impairment in tumour cell metastatization in response to inhibition of NHE1 and MMPs activities by [Pt(acac)2(DMS)] occurring through a novel mechanism mediated by PKC-δ/-ε activation.
The selectivity of [Pt(O,O’-acac)(γ-acac)(DMS)] stimulates a more detailed study aimed at pre-clinical investigation of its therapeutic potential in vivo. In this context, we employed a preclinical model based on the subcutaneous injection of MCF-7 breast cancer and ZL55 malignant pleural mesotelioma cell lines in SCID mice. Remarkably, [Pt(O,O’-acac)(γ- acac)(DMS)] stands out for higher anticancer activity than cisplatin toward both the murine tumor models examined, inducing up to 50% inhibition of tumor growth. We also demonstrated enhanced in vivo pharmacokinetics (PK), biodistribution and tolerability of [Pt(O,O’-acac)(γ-acac)(DMS)] when compared to cisplatin administered in Wistar rats. Altogether, these findings suggest that [Pt(O,O’-acac)(γ-acac)(DMS)] is a promising therapeutic agent for preventing growth of cancer, thus providing a solid starting point for its validation as a suitable candidate for further pharmacological testing.
In this study, we used the ovarian cancer cells Skov-3,a cell line resistant to cisplatin, in order to investigate whether [Pt(O,O’-acac)(γ-acac)(DMS)] was able to induce cell death. Skov-3 cells were treated with [Pt(O,O’-acac)(γ-acac)(DMS)] or with cisplatin and cytotoxicity tests were performed, together with western blotting of several proteins involved in apoptosis and autophagy, including LC-3 I/II and Beclin-1, considered important autophagic markers. The results obtained showed that [Pt(O,O’-acac)(γ-acac)(DMS)] induced a significant decrease in Skov-3 cell viability. We assessed the activation of both apoptotic and autophagic processes since western blotting analyses showed a time-dependent increment of the expression levels of autophagic markers Beclin-1 and LC-3, together with the proteolytic activation of caspase 9 and the degradation of PARP. The formation of autophagic vacuoles was detected by visualization of monodansylcadaverine (MDC) in Skov-3 cells incubated with [Pt(O,O’-acac)(gamma-acac)(DMS)]-treated cells, but not in cisplatin-treated cells. In addition, cisplatin caused apoptosis in Skov-3 cells since it activated caspases 7, 3 and 9 with consequent PARP proteolysis whilst it did not have any effect on LC-3II and Beclin-1 activation, thus showing the absence of autophagic processess. In conclusion, both [Pt(O,O’-acac)(gamma-acac)(DMS)] and cisplatin induced apoptotic Skov-3 cell death, but only [Pt(O,O’-acac)(gamma-acac)(DMS)] was able to induce autophagy.
The aim of this study was to compare antitumor activity of [Pt(O,O’-acac)(γ-acac)(DMS)] in epithelioid and sarcomatoid type of MPM. Thus, we employed the human cell line of epithelioid derivation ZL55, and the sarcomatoid cell line ZL34 in vitro and SCID mice. In epithelioid cells, [Pt(O,O’-acac)(γ-acac)(DMS)] was approximately 12-fold more cytotoxic than cisplatin after 24 h of incubation (IC50 were 3.9±0.11 μM for [Pt(O,O’-acac)(γ-acac)(DMS)] and 46.8±0.6 μM for cisplatin, n=6). Similarly, in sarcomatoid cells cisplatin was significantly less cytotoxic than [Pt(O,O’-acac)(γ-acac)(DMS)] (IC50 48.7±1.7 μM and ND n=4, for [Pt(O,O’-acac)(γ-acac)(DMS) and cisplatin, respectively). In addition, we employed a preclinical model based on the subcutaneous injection of ZL55 and ZL34 malignant pleural mesotelioma cell lines in SCID mice. Remarkably, [Pt(O,O’-acac)(γ-acac)(DMS)] stands out for higher anticancer activity than cisplatin toward both the murine tumor models examined, inducing up to 50% inhibition of tumor growth. Mice inoculated with MPM cells showed a statistically significant reduction of tumor volume at every time point in the [Pt(O,O’-acac)(γ-acac)(DMS)] groups compared with both not treated and cisplatin-treated mice (p < 0.05). In summary, our findings show that [Pt(O,O’-acac)(γ-acac)(DMS)] seems to be more potent than cisplatin in MPM, thus providing a solid starting point for its validation as a suitable candidate for further pharmacological testing.
[Pt(O,O′-acac)(γ-acac)(DMS)] can inhibit the metastatic diffusion of breast carcinoma
[Pt(O,O'-acac)(gamma-acac)(DMS)] (PtAcD) is able to induce apoptosis in various human cancer cells, including the cisplatin-resistant human breast carcinoma MCF-7 cells. Here, to confirm that PtAcD has the potentiality for therapeutic intervention, we studied its effects in primary cultured epithelial breast cells obtained from cancers and also from the corresponding histologically proven non-malignant tissue adjacent to the tumor. We demonstrated that PtAcD (1) is more cytotoxic in cancer than in normal breast cells; (2) activated NAD(P)H oxidase, leading to PKC-zeta and PKC-alpha tanslocations; (3) activated antiapoptotic pathways based on the PKC-alpha, ERK1/2 and Akt kinases; (4) activated PKC-zeta and, only in cancer cell PKC-delta, responsible for the sustained phosphorylation of p38 and JNK1/2, kinases both of which are involved in the mitochondrial apoptotic process. Moreover, crosstalk between ERK/Akt and JNK/p38 pathways affected cell death and survival in PtAcD-treated breast cell. In conclusion, this study adds and extends data that highlight the pharmacological potential of PtAcD as an anti breast cancer drug.
We used the human mesothelioma ZL55 cells, which are a reliable model for investigating the therapeutic potential of nucleotides in MPM clinical trials. In addition we attempted to potentiate the cytotoxic effects of cisplatin by ADP. In ZL55 cells ADP is able to block cellular proliferation in a time and dose dependent manner, but evaluating the major apoptotic markers, it is seen that the mechanism underlying this effect is not apoptosis. Cell cycle analysis revealed that inhibition of proliferation of ZL55 cells by ADP was mediated by block of cell cycle progression. Flow cytometry analysis of subconfluent ZL55 cell cultures showed that treatment with ADP (10–100μM) for 24 h caused a concentration dependent increase of cells in the G0/G1 phases. ADP also induced p53 protein levels and increased the levels of p53, p21 and p27 mRNA, whilst cisplatin did not. Cisplatin plus ADP were more efficacious than cisplatin alone in inducing apoptosis characterized by: (a) mitochondria depolarization, (b) increase of bax expression and its cytosol-to-mitochondria translocation and decrease of Bcl-2 expression, (c) activation of caspase-7 and -9 and cleavage of PARP-1. Therefore, the antiproliferative effect of ADP is due to the cell cycle block and increases cisplatin cytotoxicity.
Tumor growth, tumor cell proliferation and microvessel density, in a xenograft model of RCC, developed by injection of Caki-1 cells in BALB/c nude mice, were investigated. Exposure of the Caki-1 cells to cisplatin and to [Pt(O,O’-acac)(γ-acac)(DMS)] resulted in a dose-dependent inhibition of cell survival. [Pt(O,O′-acac)(γ-acac)(DMS)] was much more effective than cisplatin in the inhibition of tumor growth, proliferation and angiogenesis in vivo as well as migration, tube formation and MMP-1, MMP-2 and MMP-9 secretion of endothelial cells, in vitro. Wherease, cisplatin exerted a higher cytotoxic activity on Huvecs, but did not affect tube formation and migration of endothelial cells. In addition, treatment of xenograft mice with [Pt(O,O′-acac)(γ-acac)(DMS)] decreased, VEGF, MMP-1 and MMP-2 expression in tumors.The anti-angiogenic and anti-tumor activities of [Pt(O,O’-acac)(γ-acac)(DMS)] providing a solid starting point for its validation as a suitable candidate for further pharmacological testing.
The higher and selective cytotoxicity of [Pt(O,O′-acac)(γ-acac)(DMS)] towards cancer cell in both immortalized cell lines and in breast cancer cells in primary cultures, stimulated a pre-clinical study in order to evaluate its therapeutic potential in vivo. The efficacy of [Pt(O,O′-acac)(γ-acac)(DMS)] was assessed using a xenograft model of breast cancer developed by injection of MCF-7 cells in the flank of BALB/c nude mice. Treatment of solid tumor-bearing mice with [Pt(O,O′-acac)(γ-acac)(DMS)] induced up to 50% reduction of tumor mass compared to an average 10% inhibition recorded in cisplatin treated animals. Thus, chemotherapy with [Pt(O,O′-acac)(γ-acac)(DMS)] was much more effective than cisplatin. We also demonstrated enhanced in vivo pharmacokinetics, biodistribution, and tolerability of [Pt(O,O′-acac)(γ-acac)(DMS)] when compared to cisplatin administered in Wistar rats. Pharmacokinetics studies with [Pt(O,O′-acac)(γ-acac)(DMS)] revealed prolonged Pt persistence in systemic blood circulation and decreased nefrotoxicity and hepatotoxicity¬, a major target sites of cisplatin toxicity. Overall, [Pt(O,O′-acac)(γ-acac)(DMS)] turned out to be extremely promising in terms of greater in vivo anticancer activity, reduced nephrotoxicity and acute toxicity compared to cisplatin.
BACKGROUND AND PURPOSE It is thought that the mechanism of action of anticancer chemotherapeutic agents is mainly due to a direct inhibition of tumour cell proliferation. In tumour specimens, the endothelial cell proliferation rate increases, suggesting that the therapeutic effects of anticancer agents could also be attributed to inhibition of tumour angiogenesis. Hence, we investigated the potential effects of [Pt (O,O′-acac)(γ-acac)(DMS)] ([Pt(DMS)]), a new platinum drug for non-genomic targets, on human renal carcinoma and compared them with those of the well-established anticancer drug, cisplatin. EXPERIMENTAL APPROACH Tumour growth, tumour cell proliferation and microvessel density were investigated in a xenograft model of renal cell carcinoma, developed by injecting Caki-1 cells into BALB/c nude mice. The antiangiogenic potential of compounds was also investigated using HUVECs. KEY RESULTS Treatment of the Caki-1 cells with cisplatin or [Pt(DMS)] resulted in a dose-dependent inhibition of cell survival, but the cytotoxicity of [Pt(DMS)] was approximately fivefold greater than that of cisplatin. [Pt(DMS)] was much more effective than cisplatin at inhibiting tumour growth, proliferation and angiogenesis in vivo, as well as migration, tube formation and MMP1, MMP2 and MMP9 secretion of endothelial cells in vitro. Whereas, cisplatin exerted a greater cytotoxic effect on HUVECs, but did not affect tube formation or the migration of endothelial cells. In addition, treatment of the xenograft mice with [Pt(DMS)] decreased VEGF, MMP1 and MMP2 expressions in tumours. CONCLUSIONS AND IMPLICATIONS The antiangiogenic and antitumour activities of [Pt(DMS)] provide a solid starting point for its validation as a suitable candidate for further pharmacological testing.
Mesothelioma cancer cells have epithelioid or sarcomatoid morphology. The worst prognosis is associated with sarcomatoid phenotype and resistance to therapy is affected by cells heterogeneity. We recently showed that in ZL55 mesothelioma cell line of epithelioid origin [Pt(O,O'-acac)(γ-acac)(DMS)] (Ptac2S) has an antiproliferative effect in vitro and in vivo. Aim of this work was to extend the study on the effects of Ptac2S on ZL34 cell line, representative of sarcomatoid mesothelioma. ZL34 cells were used to assay the antitumor activity of Ptac2S in a mouse xenograft model in vivo. Then, both ZL34 and ZL55 cells were used in order to assess the involvement of p53 protein in (a) the processes underlying the sensitivity to chemotherapy and (b) the activation of various transduction proteins involved in apoptosis/survival processes. Ptac2S increases ZL34 cell death in vivo compared with cisplatin and, in vitro, Ptac2S was more efficacious than cisplatin in inducing apoptosis. In Ptac2S-treated ZL34 and ZL55 cells, p53 regulated gene products of apoptotic BAX and anti-apoptotic Bcl-2 proteins via transcriptional activation. Ptac2S activated PKC-δ and PKC-ε; their inhibition by PKC-siRNA decreased the apoptotic death of cells. PKC-δ was responsible for JNK1/2 activation that has a role in p53 activation. In addition, PKC-ε activation provoked phosphorylation of p38MAPK, concurring to apoptosis. In ZL34 cells, Ptac2S also activated PKC-α thus provoking ERK1/2 activation; inhibition of PKC-α, or ERK1/2, increased Ptac2S cytotoxicity. Results confirm that Ptac2S is a promising therapeutic agent for malignant mesothelioma, giving a substantial starting point for its further validation.
The relation between the tumor and its microenvironment is one of the most interesting and less understood issues. Recently, we showed a role of CCL20 chemokine in proning the healthy tissue neighboring the tumor to carcinogenesis. Besides, tumor-secreted CCL20 induced proliferation, migration, and EMT of healthy cells. In this context, we have studied here if CCL20 had effects on the migration of cancer cells and the intracellular pathways used in breast epithelial cells in primary culture. Using molecular (siRNA) and pharmacological (inhibitors) techniques, we found multiple signaling kinases to be activated and involved in CCL20-induced tumor breast cell migration. CCL20 provoked a 2.5-fold increase of cell migration and invasion; CCL20 also enhanced MMP- 2 and MMP-9 mRNAs/protein expression and activities. Cell migration and invasiveness due to CCL20 significantly decreased when MMP-2 and MMP-9 were inhibited in CCL20-stimulated cells. CCL20 controlled MMP-2 expression through the JAK2/STAT3 pathway, while the expression of MMP-9 occurred by PKC-α that activated, consequently, c-Src, Akt, and finally NF-kB. These results reveal a role for CCL20 also in tumor breast cell and point to CCL20 as a novel therapeutic target in cancer.
The communication between the tumor cells and the surrounding cells helps drive the process of tumor progression. Since the microenvironment of breast cancer includes CCL20 chemokine, the purpose of this study was to determine whether CCL20 modulates the physiology of healthy breast epithelial cells in areas adjacent to the tumor. Therefore, primary cultures of mammary cells taken from normal peritumoral areaswere used.We assessed that breast cells expressedCCR6CCL20 receptor.Usingmolecular (siRNA) and pharmacological (inhibitors) techniques, we found multiple signaling kinases to be activated by CCR6 and involved in CCL20-induced breast cell proliferation and migration. The binding of 10 ng/ml CCL20 to CCR6 induced cell migration whilst higher concentrations (from 15 to 25 ng/ml) led to cell proliferation. CCL20 controlled cell migration and MMP-9 expression by PKC-alpha that activated Src, which caused the activation of downstream Akt, JNK, and NF-kB pathways. Furthermore, higher CCL20 concentrations increased cycE and decreased p27Kip expression ending in enhanced cell proliferation.Cell proliferation occurred through PKC-epsilon activation that transactivated EGFR and ERK1/2/MAPK pathway.Although activated by differentCCL20 concentrations, these pathways function in parallel and crosstalk to some extent, inasmuch as Akt activation was responsible for ERK1/2 nuclear translocation and enhanced the transcription of of c-fos and c-myc, involved in cell proliferation. In summary, tumor cells exchange signals with the surrounding healthy cellsmodifying the extracellular matrix through enzyme secretion; thus, CCL20 might be a factor involved in the ontogeny of breast carcinoma.
The products obtained by forcing the reaction with nucleosides (guanosine, Guo, and adenosine, Ado) of potential anticancer drugs for nongenomic targets [PtCl(O,O'-acac)(L)] (L = dimethyl sulfoxide, DMSO; dimethyl sulfide, DMS), closely related to their very powerful organometallic analogues [Pt(O,O'-acac)(γ-acac)(L)], have been studied. [PtCl(O,O'-acac)(L)] and [Pt(O,O'-acac)(γ-acac)(L)] complexes were reported unreactive toward nucleobases. Aquo species [Pt(O,O'-acac)H2O(L)]+, obtained from [PtCl(O,O'-acac)(L)] by Ag+ driven coordinated Cl– removal, gave access to [Pt(O,O'- acac)(L)(nucleoside)]+ ([Pt(O,O'-acac)(DMSO)(Guo)]+, [Pt(O,O'-acac)(DMS)(Guo)]+, [Pt(O,O'-acac)(DMSO)(Ado)]+). The effect of the chelate oxygen donor acac (with respect to a chelate diammine), the role of the sulfur ligand (DMSO, DMS), and the influence of the purinic nucleoside itself on the coordinated Guo or Ado dynamic motions in [Pt(O,O'- acac)(L)(nucleoside)]+ complexes have been investigated by NMR spectroscopy. Interestingly, a slow rotation of nucleobase around the Pt–N(7) bond with formation of two rotamers was observed already at room temperature only in the case of [Pt(O,O'- acac)(DMSO)(Guo)]+. On the other hand, no hindered rotation at room temperature was detected in the analogous [Pt(O,O'-acac)(DMS)(Guo)]+ and [Pt(O,O'-acac)(DMSO)(Ado)]+ complexes. Data suggest that rotation of the nucleoside in [Pt(O,O'-acac)(L)(nucleoside)]+ is very different with respect to the analogous [Pt(diammine)(L)(nucleoside)]2+ systems, due to specific interactions between the acac chelate ligand, the DMSO, and the nucleobase
BACKGROUND AND PURPOSE: The aim of this study was to determine whether [platinum (Pt)(O,O'-acetylacetonate (acac))(γ-acac)(dimethylsulphide (DMS))] is differentially cytotoxic in normal and cancer cells, and to measure comparative levels of cytotoxicity compared with cisplatin in the same cells. EXPERIMENTAL APPROACH: We performed experiments on cancerous and normal epithelial breast cells in primary culture obtained from the same patients. The apoptotic effects [Pt(O,O'-acac)(γ-acac)(DMS)] and cisplatin in cancerous and normal breast cells were compared. KEY RESULTS: Cancer cells were more sensitive to [Pt(O,O'-acac)(γ-acac)(DMS)] (IC50 = 5.22 ± 1.2 μmol·L(-1)) than normal cells (IC50 = 116.9 ± 8.8 μmol·L(-1)). However, the difference was less strong when cisplatin was used (IC50 = 96.0 ± 6.9 and 61.9 ± 6.1 μmol·L(-1) for cancer and normal cells respectively). Both compounds caused reactive oxygen species (ROS) production with different mechanisms: [Pt(O,O'-acac)(γ-acac)(DMS)] quickly activated NAD(P)H oxidase while cisplatin caused a slower formation of mitochondrial ROS. Cisplatin and [Pt(O,O'-acac)(γ-acac)(DMS)] caused activation of caspases, proteolysis of PARP and modulation of Bcl-2, Bax and Bid. [Pt(O,O'-acac)(γ-acac)(DMS)] also caused leakage of cytochrome c from the mitochondria. Overall, these processes proceeded more quickly in cells treated with [Pt(O,O'-acac)(γ-acac)(DMS)] compared with cisplatin. [Pt(O,O'-acac)(γ-acac)(DMS)] effects were faster and quantitatively greater in cancer than in normal cells. [Pt(O,O'-acac)(γ-acac)(DMS)] caused a fast decrease of mitochondrial membrane potential, especially in cancer cells. CONCLUSIONS AND IMPLICATIONS: [Pt(O,O'-acac)(γ-acac)(DMS)] was specific to breast cancer cells in primary culture, and this observation makes this compound potentially more interesting than cisplatin.
We investigated the effects of cisplatin (cisPt) on matrix metalloproteinase-2 (MMP-2) gelatinolitic activity in transformed PC E1Araf rat thyroid cells. Cells incubated with increasing cisPt concentrations showed dose- and time-dependent decrease of the MMP-2 protein and activity. CisPt provoked the translocation from the cytosol to the plasma membrane of atypical protein kinase C-zeta (PKC-zeta) and the activation of PKB/AKT. The effect of cisPt on MMP-2 was dependent on PKC-zeta activation since it was potentiated by a myristoylated PKC-zeta pseudo substrate peptide or by PKC-zeta down regulation by siRNA. Moreover, MMP-2 activity modulation by cisPt was also dependent on PKB/AKT activation since it was decreased by PKB/AKT down regulation by siRNA or by pharmacological inhibition of PI3K, thus indicating the importance of the balance of PKB/AKT and PKC-zeta in regulating the cisPt effect on MMP-2 activity. The PC E1Araf cells displayed a migratory capacity that was blocked by MMP-2 down regulation using siRNA or pharmacological inhibition. The inhibition of cell migration was also obtained with cisPt; in cisPt-treated cells the administration of MMP-2 active protein was able to restore cell migration capacity. In conclusion, the decrease of MMP-2 secretion after cisPt was allowed by PKB/AKT and counteracted by PKC-zeta; the cisPt-provoked inhibition of MMP-2 secretion ended in reduction of cell migration.
The effects of a Pt-based drug on human mesothelioma cells NCI-H28 and H2373
Malignant pleural mesothelioma (MPM) is an aggressive malignancy highly resistant to chemotherapy. There is an urgent need for effective therapy inasmuch as resistance, intrinsic and acquired, to conventional therapies is common. Among Pt(II) antitumor drugs, [Pt(O,O'-acac)(γ-acac)(DMS)] (Ptac2S) has recently attracted considerable attention due to its strong in vitro and in vivo antiproliferative activity and reduced toxicity. The purpose of this study was to examine the efficacy of Ptac2S treatment in MPM. We employed the ZL55 human mesothelioma cell line in vitro and in a murine xenograft model in vivo, to test the antitumor activity of Ptac2S. Cytotoxicity assays and Western blottings of different apoptosis and survival proteins were thus performed. Ptac2S increases MPM cell death in vitro and in vivo compared with cisplatin. Ptac2S was more efficacious than cisplatin also in inducing apoptosis characterized by: (a) mitochondria depolarization, (b) increase of bax expression and its cytosol-to-mitochondria translocation and decrease of Bcl-2 expression, (c) activation of caspase-7 and -9. Ptac2S activated full-length PKC-δ and generated a PKC-δ fragment. Full-length PKC-δ translocated to the nucleus and membrane, whilst PKC-δ fragment concentrated to mitochondria. Ptac2S was also responsible for the PKC-ε activation that provoked phosphorylation of p38. Both PKC-δ and PKC-ε inhibition (by PKC-siRNA) reduced the apoptotic death of ZL55 cells. Altogether, our results confirm that Ptac2S is a promising therapeutic agent for malignant mesothelioma, providing a solid starting point for its validation as a suitable candidate for further pharmacological testing.
We assessed the therapeutic potential of [Pt(O,O’-acac)(γ-acac)(DMS)] in vivo using a xenograft model of breast cancer developed by injection of MCF-7 cells in the flank of BALB/c nude mice. Solid tumor-bearing mice showed up to 50% reduction of tumor mass after [Pt(O,O’-acac)(γ-acac)(DMS)] treatment, whereas an average 10% inhibition was recorded in cisplatin treated animals. Moreover, enhanced in vivo pharmacokinetics (PK), biodistribution and tolerability for [Pt(O,O’-acac)(γ-acac)(DMS)] administered Wistar rats have been observed with respect to cisplatin treated.
Extracellular nucleotides can regulate cell proliferation in both normal and tumorigenic tissues. Here, we studied how extracellular nucleotides regulate the proliferation of ZL55 cells, a mesothelioma-derived cell line obtained from bioptic samples of asbestos-exposed patients. ADP and 2-MeS-ADP inhibited ZL55 cell proliferation, whereas ATP, UTP, and UDP were inactive. The nucleotide potency profile and the blockade of the ADP-mediated inhibitory effect by the phospholipase C inhibitor U-73122 suggest that P2Y1 receptor controls ZL55 cell proliferation. The activation of P2Y1 receptor by ADP leads to activation of intracellular transduction pathways involving [Ca(2+) ]i , PKC-δ/PKC-α, and MAPKs, ERK1/2 and JNK1/2. Cell treatment with ADP or 2-MeS-ADP also provokes the activation of p53, causing an accumulation of the G1 cyclin-dependent kinase inhibitors p21(WAF1) and p27(Kip) . Inhibition of ZL55 cell proliferation by ADP was completely reversed by inhibiting MEK1/2, or JNK1/2, or PKC-δ, and PKC-α. Through the inhibition of ADP-activated transductional kinases it was found that PKC-δ was responsible for JNK1/2 activation. JNK1/2 has a role in transcriptional up-regulation of p53, p21(WAF1/CIP1) , and p27(kip1) . Conversely, the ADP-activated PKC-α provoked ERK1/2 phosphorylation. ERK1/2 increased p53 stabilization, required to G1 arrest of ZL55 cells. Concluding, the importance of the study is twofold: first, results shed light on the mechanism of cell cycle inhibition by ADP; second, results suggest that extracellular ADP may inhibit mesothelioma progression.
Risk factors for soccer injuries and prevention have been discussed by several authors, but only few have investigated the effectiveness of preventive interventions, in particular in the last few years. The aim of the present study was to evaluate the effects of a prevention program on the incidence of soccer-related injuries in three different categories of younger players (Allievi, Giovanissimi e Primavera). The soccer teams took part in a prevention program to decrease injury risk, for two agonistic seasons (2005-06, 2006-07). Before identifying training programs, we valued the younger fitness status: each youth soccer players (aged 13 to 19) performed exercise tests of functional capacity: plyometric jump (explosive power) and Lèger test (aerobic resistance). Thus, the temporal connection in loads alternation 1:1 as applied. During the two agonistic seasons all new injuries were registered. The incidence of injury per 1000 hours of training and playing soccer was 3.6 and 1.3 in 2005-06 and 2006-07, respectively. A very high percentage of soccer-related injuries occurred during a game, unlike those occurring during training. Our prevention program had greater effects overall in the last agonistic season taken into account since he injury incidence was significantly lower those reported in other studies. In conclusion, the incidence of youth soccer-related injuries can be reduced by preventive interventions; and coaches and players need better education regarding injury prevention strategies and should include such interventions as part of their regular training.
We previously found that CCL20 induced primarily cultured healthy breast cell proliferation and migration. The objective of this study was to investigate the hypothesis that CCL20 modulated the epithelial-mesenchymal transition (EMT) of primarily cultured healthy breast epithelial cells and the angiogenesis in areas adjacent to the tumor. Key results showed that CCL20 (a) down-regulated E-cadherin and ZO-1; (b) up-regulated N-cadherin, vimentin, and Snail expressions; (c) increased mRNA and secretion of VEGF and (d) increased angiogenic micro vessel sprouting. Thus, the signal transduction pathways evoked by CCL20 were investigated. We showed that NF-kB p65 down-regulation (by small interfering RNA, siRNA) reversed CCL20-induced Snail and blocked the up-regulation of vimentin and N-cadherin mRNAs. Furthermore, PI3K/AKT inhibition (by LY294002) completely blocked CCL20-induced Snail and NF-kB activation. Inhibition of JNK1/2 (by SP60125) or PKC-α (by siRNA) or src (by PP1) blocked NF-kB activation and Snail expression suggesting that these kinases are all upstream of NF-kB/Snail. Inhibition of mTOR (by rapamycin) abolished the effects of CCL20 on N-cadherin and vimentin protein synthesis. Furthermore, siRNA of PKC-δ inhibited the phosphorylation of CCL20-induced mTOR and S6, increased vimentin and N-cadherin expressions and, finally, blocked the CCL20 induced-EMT. CCL20 increased mRNA and secretion of VEGF by healthy breast cells by using PKC-α, src, Akt, NF-kB, and Snail signalling. In summary, tumor cells signal to the surrounding healthy cells through CCL20 inducing the modulation of the expression of molecules involved in EMT and promoting angiogenesis directly and indirectly through the secretion of VEGF, a major contributor to angiogenesis. © 2015 Wiley Periodicals, Inc.
Cisplatin is commonly employed in therapy of mesothelioma but its efficacy is limited and the mechanisms by which induces its effects are not clearly understood. PKCs can regulate cisplatin sensitivity. PKCs effects on cellular sensitivity/resistance depend on the pattern of active PKC isozymes as well as on cellular context. The present study was undertaken to determine if specific PKC isoforms regulate cisplatin-induced apoptosis in the human mesothelioma ZL55 cells. Cells were treated with cisplatin at various concentrations and for different incubation periods. Cytotoxicity assays and Western blottings of various proteins involved in apoptosis and survival were then performed. Exposure of ZL55 cells to cisplatin at concentrations ranging from 1 to 200 μM resulted in a dose-dependent inhibition of cell survival and the activation of the mitochondrial apoptotic pathway. Cisplatin activated full-length PKC-δ and generated a PKC-δ fragment. PKC-δ inhibition (by PKC-δ-siRNA) decreased ZL55 cell apoptosis. Full-length PKC-δ translocated to the nucleus and activated caspase-3 expression, whereas PKC-δ fragment preferentially localized to mitochondria. Cisplatin also provoked the generation of reactive oxygen species (ROS) by NADPH oxidase. ROS increment was responsible for the PKC-α activation that provoked EGFR transactivation and consequential phosphorylation of ERK1/2. The inhibition of this pathway at various level (PKC-α, EGFR or ERK1/2) increased cisplatin-induced cytotoxicity. The results suggest that PKC-δ is an essential part of the apoptotic program in mesothelioma cells, whereas PKC-α mediates a pro-survival response to cisplatin.
We used ZL55 mesothelioma cell line in order to study the cytotoxicity of cisplatin and the cellular pathways eventually ending to the apoptotic process. To this end, ZL55 cells were treated with increasing doses (1-200 μM) and for different incubation times (1-72 hours) with cisplatin and then the number of viable cells was evaluated by the MTT test. The cleavage patterns of caspase-3, -7 and -9, and Poly-ADP ribose polymerase (PARP) by cisplatin were assessed by Western methods. The results showed here suggest that activation of PKC-δ is an essential part of the apoptotic program provoked by cisplatin in mesothelioma cells and that PKC-δ transduces a pro-apoptotic signal in these cells.
We here demonstrated the expression of 4 (P2Y1,4,6,11) purinoceptors mRNAs coupled to Gq protein. The extracellular ATP, UTP, ADP and UDP caused a transient [Ca2+]i peak followed by a sustained lower phase. Removal of extracellular Ca2+ decreased the initial transient and abolished the plateau phase. Ca2+ signal was blocked by the inhibitor of PLCβ, U73122. Through experiments of fluorescence quenching, it was seen that nucleotides increased plasmamembrane Ca2+ permeability. These results indicate that [Ca2+]i increase was due to activation of P2Y receptors exclusively and that Ca2+ is released from intracellular stores and also enters from the extracellular liquid. We also investigated the effects of purinoceptors on cell proliferation. UTP, ATP and UDP had no significant effects, while ADP causes a drastic cell proliferation decrease in a dose- and time-dependent manner. In conclusion, these results indicate that the expressed purinergic receptors act via PLC activation and that ADP may have a therapeutic potential in MM.
Aberrant methylation of CpG islands in the promoter regions of tumour cells results in loss of gene function. In addition to genetic lesions, changes in the methylation profile of the promoters may be considered a factor for tumour-specific aberrant expression of the genes.We investigated the methylation status of E-cadherin gene (CDH1) promoter in low-grade glioma and correlated it with clinical outcome. Eighty-four cases of low-grade glioma (43 diffuse astrocytomas, 27 oligodendrogliomas and 14 oligoastrocytomas) with assessable paraffin-embedded tumour blocks and normal brain tissue, derived from non-cancerous tissue adjacent to tumour and commercially normal brain tissue, were collected, from which we determined CDH1 promoter methylation status and E-cadherin protein expression by methylation-specific polymerase chain reaction (MSP) and immunohistochemistry, respectively. CDH1 promoter was found hypermethylated in 54 out of 84 low grade gliomas (64%) compared with 84 normal brain tissue. CDH1 hypermethylation was found in 65% astrocytomas, 66% oligodendrogliomas and 57% oligoastrocytomas. A significant correlation between hypermethylation status, patient survival and progression-free survival was found (P = 0.04). Survival and progression-free survival were lower in patients with hypermethylated CDH1 promoter. We found that 15 astrocytomas, 9 oligodendrogliomas and 6 oligoastrocytomas were immunoreactive for E-cadherin. The incidence of loss of immunoreactivity for E-cadherin decreased significantly with age, overall survival and progression-free survival (P = 0.001, Kaplan-Meier test). We have demonstrated that CDH1 promoter hypermethylation significantly associated with down-regulated E-cadherin expression and overall survival of patients. This may have a bearing on the prognosis of low-grade glioma.
[Pt(O,O′-acac)(γ-acac)(DMS)] change the expression of 84 proteins as assessed by a proteomic approach in SH-SY5Y cells
BACKGROUND AND PURPOSE: We showed previously that a new Pt(II) complex ([Pt(O,O'-acac)(gamma-acac)(DMS)]) exerted high and fast apoptotic processes in MCF-7 cells. The objective of this study was to investigate the hypothesis that [Pt(O,O'-acac)(gamma-acac)(DMS)] is also able to exert anoikis and alter the migration ability of MCF-7 cells, and to show some of the signalling events leading to these alterations. EXPERIMENTAL APPROACH: Cells were treated with sublethal doses of [Pt(O,O'-acac)(gamma-acac)(DMS)], and the efficiency of colony initiation and anchorage-independent growth was assayed; cell migration was examined by in vitro culture wounding assay. Gelatin zymography for MMP-2 and -9 activities, Western blottings of MMPs, MAPKs, Src, PKC-epsilon and FAK, after [Pt(O,O'-acac)(gamma-acac)(DMS)] treatment, were also performed. KEY RESULTS: Sub-cytotoxic drug concentrations decreased the: (i) anchorage-dependent and -independent growth; (ii) migration ability; and (iii) expression and activity of MMP-2 and MMP-9. [Pt(O,O'-acac)(gamma-acac)(DMS)] provoked the generation of reactive oxygen species (ROS), and the activation of p38MAPK, Src and PKC-epsilon. p38MAPK phosphorylation, cell anoikis and migration due to [Pt(O,O'-acac)(gamma-acac)(DMS)] were blocked by PKC-epsilon inhibition. Furthermore, Src inhibition blocked the [Pt(O,O'-acac)(gamma-acac)(DMS)]-provoked activation of PKC-epsilon, while ROS generation blockage inhibited the activation of Src, and also the decrement of phosphorylated FAK observed in detached [Pt(O,O'-acac)(gamma-acac)(DMS)]-treated cells. CONCLUSIONS AND IMPLICATIONS: Sublethal concentrations of [Pt(O,O'-acac)(gamma-acac)(DMS)] induced anoikis and prevented events leading to metastasis via alterations in cell migration, anchorage independency, stromal interactions and MMP activity. Hence, [Pt(O,O'-acac)(gamma-acac)(DMS)] may be a promising therapeutic agent for preventing growth and metastasis of breast cancer.
The new b-diketonate compounds ([Pt(O,O’-acac)(gamma-acac)(NH3)] and [Pt(O,O’-acac)(g-acac)(py)]) were chemical characterized by NMR spectroscopy and preliminary cytotoxic assays of [Pt(O,O’-acac)(g-acac)(NH3)] on cisplatin resistant MCF-7 breast cancer cell line were performed. Interestingly, [Pt(O,O’-acac)(gamma-acac)(NH3)] resulted in dose dependent inhibition of cell survival comparable to [Pt(O,O’-acac)(γ-acac)(DMS) and approximately 90-fold greater than that observed for cisplatin.
In the present study, we aimed to investigate the anti-angiogenesis effects of [Pt(O,O’-acac)(γ-acac)(DMS)]. [Pt(O,O′-acac)(γ-acac)(DMS)] significantly inhibited human umbilical vein endothelial cell (HUVEC) proliferation. Then, the ex vivo rat aortic ring capillary-network sprouting has been carried out. It was observed that after one week of treatment under regular growth conditions, [Pt(O,O′-acac)(γ-acac)(DMS)] potently inhibited the sprouting as well as capillary-network formation from rat aortic ring in a dose-dependent manner. Compared to control 0,5 and 1 μM doses of [Pt(O,O′-acac)(γ- acac)(DMS)] suppressed capillary-network formation by 68 to 100%, respectively. In addition, in mouse xenograft model of breast cancer, treatment of mice with [Pt(O,O′-acac)(γ-acac)(DMS)], decrease MMP-1 content in tumours. These observations demonstrated that [Pt(O,O′- acac)(γ-acac)(DMS)] could inhibit angiogenesis, which warrants further studies in other in vivo models.
It was previously demonstrated that [Pt(O,O0-acac)(g-acac)(DMS)] exerted toxic effects at high doses, whilst sub-cytotoxic concentrations induced anoikis and decreased cell migration. Aim of this study was to investigate the hypothesis that [Pt(O,O0-acac)(g-acac)(DMS)] alters the [Ca2+]i and that this is linked to its ability to trigger rapid apoptosis in MCF-7 cells. Thus, cells were treated with [Pt(O,O0-acac)(gacac)( DMS)] and its effects on some of the systems regulating Ca2+ homeostasis were studied, also in cells dealing with the complex changes occurring during the Ca2+ signalling evoked by extracellular stimuli. [Pt(O,O0-acac)(g-acac)(DMS)] caused the decrease of PMCA activity (but not SERCA or SPCA) and Ca2+ membrane permeability. These two opposite effects on [Ca2+]i resulted in its overall increase from 102 12 nM to 250 24 nM after 15 min incubation. The effects of [Pt(O,O0-acac)(g-acac)(DMS)] were also evident when cells were stimulated with ATP: the changes in Ca2+ levels caused by purinergic stimulation resulted altered due to decreased PMCA activity and to the closure of Ca2+ channels opened by purinergic receptor. Conversely, [Pt(O,O0-acac)(g-acac)(DMS)] did not affect the store-operated Ca2+ channels opened by thapsigargin or by ATP. [Pt(O,O0-acac)(g-acac)(DMS)] provoked the activation of PKC-a and the production of ROS that were responsible for the Ca2+ permeability and PMCA activity decrease, respectively. The overall effect of [Pt(O,O0-acac)(g-acac)(DMS)] is to increase the [Ca2+]i, an effect that is likely to be linked to its ability to trigger rapid apoptosis in MCF-7 cells. These data reinforce the notion that [Pt(O,O0-acac)(gacac)( DMS)] would be a promising drug in cancer treatment
It was previously shown that [Pt(O,O'-acac)(γ-acac)(DMS)] induces apoptosis in various cancer cells and exerts antimetastatic responses in vitro. In rats, [Pt(O,O'-acac)(γ-acac)(DMS)] reaches the central nervous system in quantities higher than cisplatin causing less excitotoxicity. The aim of the present paper was to investigate whether [Pt(O,O'-acac)(γ-acac)(DMS)] is able to exert cytotoxic effects on SH-SY5Y human neuroblastoma cell line, and to study the intracellular transduction mechanisms underlying these effects. Here we have demonstrated that [Pt(O,O'-acac)(γ-acac)(DMS)] was more effective than cisplatin in provoking apoptosis characterized by: (a) mitochondria depolarization, (b) decrease of Bcl-2 expression and increase of BAX expressions with cytosol-to-mitochondria translocation, (c) activation of caspase-7 and -9 and (d) generation of reactive oxygen species (ROS). [Pt(O,O'-acac)(γ-acac)(DMS)] provoked the activation of the following signalling kinases that were interacting with each other: PKC-δ and -ɛ, ERK1/2, p38MAPK, JNK1/2, NF-κB, c-src and FAK. We found that ROS generated by NADPH oxidase was responsible for the [Pt(O,O'-acac)(γ-acac)(DMS)]-mediated PKC-δ and -ɛ activation and consequential phosphorylation of all MAPKs. [Pt(O,O'-acac)(γ-acac)(DMS)]-induced mitochondrial apoptosis was blocked when p38MAPK and JNK1/2 were inhibited, whilst the effects on Bax/Bcl-2 mRNA and protein levels were blocked inhibiting NF-κB. NF-κB nuclear translocation was blocked inhibiting MEK1/2 activity. In addition to the induction of apoptosis [Pt(O,O'-acac)(γ-acac)(DMS)] downregulated pro-survival pathway. Survival inhibition started from mitochondrial ROS generation which induced c-src, FAK and Akt activation. In conclusion, our results suggest that [Pt(O,O'-acac)(γ-acac)(DMS)] may be considered a promising compound for the treatment of neuroblastoma. Further studies are warranted to explore in detail the therapeutic potential of this compound.
Optic nerve sheath diameter quantification by transbulbar B-mode sonography is a recently validated technique, but its clinical relevance in relapse-free multiple sclerosis patients remains unexplored. In an open-label, comparative, cross-sectional study, we aimed to assess possible differences between patients and healthy controls in terms of optic nerve sheath diameter and its correlation with clinical/paraclinical parameters in this disease. Sixty unselected relapse-free patients and 35 matched healthy controls underwent transbulbar B-mode sonography. Patients underwent routine neurologic examination, brain magnetic resonance imaging and visual evoked potential tests. The mean optic nerve sheath diameter 3 and 5 mm from the eyeball was 22-25% lower in patients than controls and correlated with the Expanded Disability Status Scale (r = -0.34, p = 0.048, and r = -0.32, p = 0.042, respectively). We suggest that optic nerve sheath diameter quantified by transbulbar B-mode sonography should be included in routine assessment of the disease as an extension of the neurologic examination.
This study aimed to evaluate the effect of TGF-β1 on RSC96 Schwann cell migration and to study its intracellular transduction pathways. We found that TGF-β1 induced the activation of key signaling molecules involved in non-Smad (Akt and MAPK) and focal adhesion signaling pathways (FAK and Src) that cooperatively regulated the process of RSC96 cells migration together with MMP-2 and MMP-9 upregulation. These findings indicate a regulatory pathway of TGF-β1 in MMP-9/-2 expression which may be significant in regulating Schwann cell migration and physiology in peripheral nervous sytem injury.
Condividi questo sito sui social