Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Front Pharmacol ; 8: 373, 2017.
Article in English | MEDLINE | ID: mdl-28674496

ABSTRACT

Malignant mesothelioma (MM) is a tumor arising from mesothelium. MM patients' survival is poor. The polyphenol 4',5,7,-trihydroxyflavone Apigenin (API) is a "multifunctional drug". Several studies have demonstrated API anti-tumoral effects. However, little is known on the in vitro and in vivo anti-tumoral effects of API in MM. Thus, we analyzed the in vitro effects of API on cell proliferation, cell cycle regulation, pro-survival signaling pathways, apoptosis, and autophagy of human and mouse MM cells. We evaluated the in vivo anti-tumor activities of API in mice transplanted with MM #40a cells forming ascites. API inhibited in vitro MM cells survival, increased reactive oxygen species intracellular production and induced DNA damage. API activated apoptosis but not autophagy. API-induced apoptosis was sustained by the increase of Bax/Bcl-2 ratio, increase of p53 expression, activation of both caspase 9 and caspase 8, cleavage of PARP-1, and increase of the percentage of cells in subG1 phase. API treatment affected the phosphorylation of ERK1/2, JNK and p38 MAPKs in a cell-type specific manner, inhibited AKT phosphorylation, decreased c-Jun expression and phosphorylation, and inhibited NF-κB nuclear translocation. Intraperitoneal administration of API increased the median survival of C57BL/6 mice intraperitoneally transplanted with #40a cells and reduced the risk of tumor growth. Our findings may have important implications for the design of MM treatment using API.

2.
Oncotarget ; 8(21): 34405-34422, 2017 May 23.
Article in English | MEDLINE | ID: mdl-28159921

ABSTRACT

Malignant mesothelioma (MM) is a primary tumor arising from the serous membranes. The resistance of MM patients to conventional therapies, and the poor patients' survival, encouraged the identification of molecular targets for MM treatment. Curcumin (CUR) is a "multifunctional drug". We explored the in vitro effects of CUR on cell proliferation, cell cycle regulation, pro-survival signaling pathways, apoptosis, autophagy of human (MM-B1, H-Meso-1, MM-F1), and mouse (#40a) MM cells. In addition, we evaluated the in vivo anti-tumor activities of CUR in C57BL/6 mice intraperitoneally transplanted with #40a cells forming ascites.CUR in vitro inhibited MM cells survival in a dose- and time-dependent manner and increased reactive oxygen species'intracellular production and induced DNA damage. CUR triggered autophagic flux, but the process was then blocked and was coincident with caspase 8 activation which activates apoptosis. CUR-mediated apoptosis was supported by the increase of Bax/Bcl-2 ratio, increase of p53 expression, activation of caspase 9, cleavage of PARP-1, increase of the percentage of cells in the sub G1 phase which was reduced (MM-F1 and #40a) or abolished (MM-B1 and H-Meso-1) after MM cells incubation with the apoptosis inhibitor Z-VAD-FMK. CUR treatment stimulated the phosphorylation of ERK1/2 and p38 MAPK, inhibited that of p54 JNK and AKT, increased c-Jun expression and phosphorylation and prevented NF-κB nuclear translocation. Intraperitoneal administration of CUR increased the median survival of C57BL/6 mice intraperitoneally transplanted with #40a cells and reduced the risk of developing tumors. Our findings may have important implications for the design of MM treatment using CUR.


Subject(s)
Antinematodal Agents/administration & dosage , Autophagy/drug effects , Curcumin/administration & dosage , Lung Neoplasms/drug therapy , Mesothelioma/drug therapy , Animals , Antinematodal Agents/pharmacology , Caspase 8/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Curcumin/pharmacology , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Humans , MAP Kinase Signaling System/drug effects , Mesothelioma, Malignant , Mice , Mice, Inbred C57BL , Phosphorylation , Xenograft Model Antitumor Assays
3.
Infect Genet Evol ; 46: 28-32, 2016 12.
Article in English | MEDLINE | ID: mdl-27771520

ABSTRACT

Clostridium botulinum is the bacterial agent of botulism, a rare but severe neuro-paralytic disease. Because of its high impact, in Italy botulism is monitored by an ad hoc surveillance system. The National Reference Centre for Botulism, as part of this system, collects and analyzes all demographic, epidemiologic, microbiological, and molecular data recovered during cases and/or outbreaks occurred in Italy. A panel of 312 C. botulinum strains belonging to group I were submitted to MLVA sub-typing. Strains, isolated from clinical specimens, food and environmental samples collected during the surveillance activities, were representative of all forms of botulism from all Italian regions. Through clustering analysis isolates were grouped into 12 main clusters. No regional or temporal clustering was detected, demonstrating the high heterogeneity of strains circulating in Italy. This study confirmed that MLVA is capable of sub-typing C. botulinum strains. Moreover, MLVA is effective at tracing and tracking the source of contamination and is helpful for the surveillance system in terms of planning and upgrading of procedures, activities and data collection forms.


Subject(s)
Botulism/microbiology , Clostridium botulinum/genetics , Molecular Epidemiology/methods , Tandem Repeat Sequences/genetics , Botulism/epidemiology , Cluster Analysis , Humans , Italy/epidemiology , Multilocus Sequence Typing
4.
Oncotarget ; 7(8): 9250-70, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26843616

ABSTRACT

Aberrant Hedgehog (Hh)/glioma-associated oncogene (GLI) signaling has been implicated in cancer progression. Here, we analyzed GLI1, Sonic Hedgehog (Shh) and NF-κB expression in 51 breast cancer (ductal carcinoma) tissues using immunohistochemistry. We found a positive correlation between nuclear GLI1 expression and tumor grade in ductal carcinoma cases. Cytoplasmic Shh staining significantly correlated with a lower tumor grade. Next, the in vitro effects of two Hh signaling pathway inhibitors on breast cancer cell lines were evaluated using the Smoothened (SMO) antagonist GDC-0449 and the direct GLI1 inhibitor GANT-61. GDC-0449 and GANT-61 exhibited the following effects: a) inhibited breast cancer cell survival; b) induced apoptosis; c) inhibited Hh pathway activity by decreasing the mRNA expression levels of GLI1 and Ptch and inhibiting the nuclear translocation of GLI1; d) increased/decreased EGFR and ErbB2 protein expression, reduced p21-Ras and ERK1/ERK2 MAPK activities and inhibited AKT activation; and e) decreased the nuclear translocation of NF-κB. However, GANT-61 exerted these effects more effectively than GDC-0449. The in vivo antitumor activities of GDC-0449 and GANT-61 were analyzed in BALB/c mice that were subcutaneously inoculated with mouse breast cancer (TUBO) cells. GDC-0449 and GANT-61 suppressed tumor growth of TUBO cells in BALB/c mice to different extents. These findings suggest that targeting the Hh pathway using antagonists that act downstream of SMO is a more efficient strategy than using antagonists that act upstream of SMO for interrupting Hh signaling in breast cancer.


Subject(s)
Anilides/therapeutic use , Breast Neoplasms/drug therapy , Carcinoma, Ductal/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Mammary Neoplasms, Experimental/drug therapy , Pyridines/therapeutic use , Pyrimidines/therapeutic use , Smoothened Receptor/antagonists & inhibitors , Zinc Finger Protein GLI1/antagonists & inhibitors , Active Transport, Cell Nucleus/drug effects , Animals , Apoptosis/drug effects , Breast Neoplasms/pathology , Carcinoma, Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Enzyme Activation/drug effects , ErbB Receptors/biosynthesis , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Hedgehog Proteins/antagonists & inhibitors , Humans , MCF-7 Cells , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , NF-kappa B/biosynthesis , Patched-1 Receptor/biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , RNA, Messenger/biosynthesis , Receptor, ErbB-2/biosynthesis , Signal Transduction/drug effects , Zinc Finger Protein GLI1/biosynthesis
5.
Tumour Biol ; 37(3): 3705-17, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26462840

ABSTRACT

Violacein (VIO; 3-[1,2-dihydro-5-(5-hydroxy-1H-indol-3-yl)-2-oxo-3H-pyrrol-3-ylidene]-1,3-dihydro-2H-indol-2-one), an indole-derived purple-colored pigment, produced by a limited number of Gram-negative bacteria species, including Chromobacterium violaceum and Janthinobacterium lividum, has been demonstrated to have anti-cancer activity, as it interferes with survival transduction signaling pathways in different cancer models. Head and neck carcinoma (HNC) represents the sixth most common and one of the most fatal cancers worldwide. We determined whether VIO was able to inhibit head and neck cancer cell growth both in vitro and in vivo. We provide evidence that VIO treatment of human and mouse head and neck cancer cell lines inhibits cell growth and induces autophagy and apoptosis. In fact, VIO treatment increased PARP-1 cleavage, the Bax/Bcl-2 ratio, the inhibition of ERK1 and ERK2 phosphorylation, and the expression of light chain 3-II (LC3-II). Moreover, VIO was able to induce p53 degradation, cytoplasmic nuclear factor kappa B (NF-κB) accumulation, and reactive oxygen species (ROS) production. VIO induced a significant increase in ROS production. VIO administration was safe in BALB/c mice and reduced the growth of transplanted salivary gland cancer cells (SALTO) in vivo and prolonged median survival. Taken together, our results indicate that the treatment of head and neck cancer cells with VIO can be useful in inhibiting in vivo and in vitro cancer cell growth. VIO may represent a suitable tool for the local treatment of HNC in combination with standard therapies.


Subject(s)
Cell Proliferation/drug effects , Head and Neck Neoplasms/drug therapy , Indoles/pharmacology , Oxalobacteraceae/chemistry , Animals , Apoptosis/drug effects , Autophagy/drug effects , Blotting, Western , Caspases/metabolism , Cell Line , Cell Line, Tumor , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Mice, Inbred BALB C , Microscopy, Fluorescence , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Poly (ADP-Ribose) Polymerase-1/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Salivary Gland Neoplasms/drug therapy , Salivary Gland Neoplasms/metabolism , Salivary Gland Neoplasms/pathology , Tumor Burden/drug effects , Tumor Suppressor Protein p53/metabolism
6.
Oncotarget ; 5(21): 10745-62, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25296980

ABSTRACT

The survival rate of head and neck squamous cell carcinomas (HNSCC) patients has not considerably changed over the last two decades. Polyphenols inhibit the growth of cancer cells. We determined whether the combination of Resveratrol (RES) and Curcumin (CUR) enhanced their in vitro and in vivo antitumor activities on HNSCC cell lines compared to the single compounds. We provide evidence that RES potentiated the apoptotic effect and reduced the IC50 of CUR on HNSCC cell lines. The model of compounds interaction indicated the onset of an additive effect of the two compounds compared to the single treatment after decrease of their concentrations. RES+CUR compared to CUR increased the PARP-1 cleavage, the Bax/Bcl-2 ratio, the inhibition of ERK1 and ERK2 phosphorylation, and the expression of LC3 II simultaneously with the formation of autophagic vacuoles. RES and CUR induced cytoplasmic NF-κB accumulation. RES+CUR administrations were safe in BALB/c mice and reduced the growth of transplanted salivary gland cancer cells (SALTO) more efficiently than CUR. Overall, combinations of CUR and RES was more effective in inhibiting in vivo and in vitro cancer growth than the treatment with CUR. Additional studies will be needed to define the therapeutic potential of these compounds in combination.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Carcinoma, Squamous Cell/pathology , Cell Proliferation/drug effects , Drug Synergism , Head and Neck Neoplasms/pathology , Salivary Gland Neoplasms/pathology , Animals , Blotting, Western , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Curcumin/administration & dosage , Fluorescent Antibody Technique , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Humans , In Vitro Techniques , Mice , Mice, Inbred BALB C , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , Resveratrol , Salivary Gland Neoplasms/drug therapy , Salivary Gland Neoplasms/metabolism , Signal Transduction/drug effects , Stilbenes/administration & dosage , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...