Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cell Death Discov ; 10(1): 270, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38830859

ABSTRACT

Cancer stem cells (CSCs) are defined as a rare population of cancer cells related to tumor initiation and maintenance. These cells are primarily responsible for tumor growth, invasion, metastasis, recurrence, and resistance to chemotherapy. In this paper, we demonstrated the ability of Ru(II)-based complexes containing 2-thiouracil derivatives with the chemical formulas trans-[Ru(2TU)(PPh3)2(bipy)]PF6 (1) and trans-[Ru(6m2TU)(PPh3)2(bipy)]PF6 (2) (where 2TU = 2-thiouracil and 6m2TU = 6-methyl-2-thiouracil) to suppress liver CSCs by targeting NF-κB and Akt/mTOR signaling. Complexes 1 and 2 displayed potent cytotoxic effects on cancer cell lines and suppressed liver CSCs from HepG2 cells. Increased phosphatidylserine exposure, loss of mitochondrial transmembrane potential, increased PARP (Asp214) cleavage, DNA fragmentation, chromatin condensation and cytoplasmic shrinkage were detected in HepG2 cells treated with these complexes. Mechanistically, complexes 1 and 2 target NF-κB and Akt/mTOR signaling in HepG2 cells. Cell motility inhibition was also detected in HepG2 cells treated with these complexes. Complexes 1 and 2 also inhibited tumor progression in mice with HepG2 cell xenografts and exhibited tolerable systemic toxicity. Taken together, these results indicate that these complexes are new anti-HCC drug candidates that can suppress liver CSCs.

2.
Cell Death Discov ; 10(1): 201, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38684672

ABSTRACT

Acute myeloid leukemia (AML) is a fatal malignancy of the blood and bone marrow. Leukemic stem cells (LSCs) are a rare subset of leukemic cells that promote the development and progression of AML, and eradication of LSCs is critical for effective control of this disease. Emetine is an FDA-approved antiparasitic drug with antitumor properties; however, little is known about its potential against LSCs. Herein, we explored the antileukemic potential of emetine, focusing on its effects on AML stem/progenitor cells. Emetine exhibited potent cytotoxic activity both in hematologic and solid cancer cells and induced AML cell differentiation. Emetine also inhibited AML stem/progenitor cells, as evidenced by decreased expression of CD34, CD97, CD99, and CD123 in KG-1a cells, indicating anti-AML stem/progenitor cell activities. The administration of emetine at a dosage of 10 mg/kg for two weeks showed no significant toxicity and significantly reduced xenograft leukemic growth in vivo. NF-κB activation was reduced in emetine-treated KG-1a cells, as shown by reduced phospho-NF-κB p65 (S529) and nuclear NF-κB p65. DNA fragmentation, YO-PRO-1 staining, mitochondrial depolarization and increased levels of active caspase-3 and cleaved PARP (Asp214) were detected in emetine-treated KG-1a cells. Moreover, treatment with the pancaspase inhibitor Z-VAD(OMe)-FMK partially prevented the apoptotic cell death induced by emetine. Emetine treatment also increased cellular and mitochondrial reactive oxygen species, and emetine-induced apoptosis in KG-1a cells was partially prevented by the antioxidant N-acetylcysteine, indicating that emetine induces apoptosis, at least in part, by inducing oxidative stress. Overall, these studies indicate that emetine is a novel potential anti-AML agent with promising activity against stem/progenitor cells, encouraging the development of further studies aimed at its clinical application.

3.
Cell Death Discov ; 10(1): 147, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38503729

ABSTRACT

Acute myeloid leukaemia (AML) is a haematological malignancy characterised by the accumulation of transformed myeloid progenitors in the bone marrow. Piplartine (PL), also known as piperlongumine, is a pro-oxidant small molecule extracted from peppers that has demonstrated antineoplastic potential in solid tumours and other haematological malignancies. In this work, we explored the potential of PL to treat AML through the use of a combination of cellular and molecular analyses of primary and cultured leukaemia cells in vitro and in vivo. We showed that PL exhibits in vitro cytotoxicity against AML cells, including CD34+ leukaemia-propagating cells, but not healthy haematopoietic progenitors, suggesting anti-leukaemia selectivity. Mechanistically, PL treatment increased reactive oxygen species (ROS) levels and induced ROS-mediated apoptosis in AML cells, which could be prevented by treatment with the antioxidant scavenger N-acetyl-cysteine and the pancaspase inhibitor Z-VAD(OMe)-FMK. PL treatment reduced NFKB1 gene transcription and the level of NF-κB p65 (pS536), which was depleted from the nucleus of AML cells, indicating suppression of NF-κB p65 signalling. Significantly, PL suppressed AML development in a mouse xenograft model, and its combination with current AML treatments (cytarabine, daunorubicin and azacytidine) had synergistic effects, indicating translational therapeutic potential. Taken together, these data position PL as a novel anti-AML candidate drug that can target leukaemia stem/progenitors and is amenable to combinatorial therapeutic strategies.

4.
Cell Death Dis ; 14(12): 832, 2023 12 15.
Article in English | MEDLINE | ID: mdl-38102125

ABSTRACT

In this work, we describe a novel ruthenium-xanthoxylin complex, [Ru(phen)2(xant)](PF6) (RXC), that can eliminate colorectal cancer (CRC) stem cells by targeting the chaperone Hsp90. RXC exhibits potent cytotoxicity in cancer cell lines and primary cancer cells, causing apoptosis in HCT116 CRC cells, as observed by cell morphology, YO-PRO-1/PI staining, internucleosomal DNA fragmentation, mitochondrial depolarization, and PARP cleavage (Asp214). Additionally, RXC can downregulate the HSP90AA1 and HSP90B1 genes and the expression of HSP90 protein, as well as the expression levels of its downstream/client elements Akt1, Akt (pS473), mTOR (pS2448), 4EBP1 (pT36/pT45), GSK-3ß (pS9), and NF-κB p65 (pS529), implying that these molecular chaperones can be molecular targets for RXC. Moreover, this compound inhibited clonogenic survival, the percentage of the CRC stem cell subpopulation, and colonosphere formation, indicating that RXC can eliminate CRC stem cells. RXC reduced cell migration and invasion, decreased vimentin and increased E-cadherin expression, and induced an autophagic process that appeared to be cytoprotective, as autophagy inhibitors enhanced RXC-induced cell death. In vivo studies showed that RXC inhibits tumor progression and experimental metastasis in mice with CRC HCT116 cell xenografts. Taken together, these results highlight the potential of the ruthenium complex RXC in CRC therapy with the ability to eliminate CRC stem cells by targeting the chaperone Hsp90.


Subject(s)
Colorectal Neoplasms , Ruthenium , Humans , Animals , Mice , Signal Transduction , Glycogen Synthase Kinase 3 beta/metabolism , HCT116 Cells , HSP90 Heat-Shock Proteins/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Cell Proliferation , Cell Line, Tumor
5.
Cell Death Discov ; 9(1): 460, 2023 Dec 16.
Article in English | MEDLINE | ID: mdl-38104089

ABSTRACT

[Ru(5-FU)(PPh3)2(bipy)]PF6 (Ru/5-FU) is a novel ruthenium complex with 5-fluorouracil with promising potential against colorectal cancer (CRC). In the present study, we investigated the molecular mechanism of Ru/5-FU action in HCT116 CRC cells. Ru/5-FU exhibited potent cytotoxicity on a panel of cancer cell lines and on primary cancer cells and induced apoptosis in HCT116 CRC cells. Ru/5-FU reduced AKT1 gene transcripts, as well as the expression of Akt1 and Akt (pS473) and downstream Akt proteins mTOR (pS2448), S6 (pS235/pS236), 4EBP1 (pT36/pT45), GSK-3ß (pS9) and NF-κB p65 (pS529), but not Akt upstream proteins Hsp90 and PI3K p85/p55 (pT458/pT199), indicating an inhibitory action of Akt/mTOR signaling. Ru/5-FU increased LC3B expression and reduced p62/SQSTM1 levels, indicating autophagy induction. Curiously, the autophagy inhibitors 3-methyladenine and chloroquine increased Ru/5-FU-induced cell death, indicating an induction of cytoprotective autophagy by this compound. Ru/5-FU also reduced clonogenic survival, as well as the percentage of CD133+ cells and colonosphere formation, indicating that Ru/5-FU can suppress stem cells in HCT116 cells. Ru/5-FU inhibited cell migration and invasion in wound healing assays and Transwell cell invasion assays, along with a reduction in vimentin expression and an increase in E-cadherin levels, indicating that Ru/5-FU can interfere with epithelial-mesenchymal transition. Ru/5-FU also inhibited in vivo HCT116 cell development and experimental lung metastases in mouse xenograft models. Altogether, these results indicate that Ru/5-FU is an anti-CRC chemotherapy drug candidate with the ability to suppress stemness in CRC cells by inhibiting Akt/mTOR signaling.

6.
Chem Biol Interact ; 343: 109444, 2021 Jul 01.
Article in English | MEDLINE | ID: mdl-33939975

ABSTRACT

Quinones are plant-derived secondary metabolites that present diverse pharmacological properties, including antibacterial, antifungal, antiviral, anti-inflammatory, antipyretic and anticancer activities. In the present study, we evaluated the cytotoxic effect of a new naphthoquinone 6b,7-dihydro-5H-cyclopenta [b]naphtho [2,1-d]furan-5,6 (9aH)-dione) (CNFD) in different tumor cell lines. CNFD displayed cytotoxic activity against different tumor cell lines, especially in MCF-7 human breast adenocarcinoma cells, which showed IC50 values of 3.06 and 0.98 µM for 24 and 48 h incubation, respectively. In wound-healing migration assays, CNFD promoted inhibition of cell migration. We have found typical hallmarks of apoptosis, such as cell shrinkage, chromatin condensation, phosphatidylserine exposure, increase of caspases-9 and-3 activation, increase of internucleosomal DNA fragmentation without affecting the cell membrane permeabilization, increase of ROS production, and loss of mitochondrial membrane potential induced by CNFD. Moreover, gene expression experiments indicated that CNFD increased the expression of the genes CDKN1A, FOS, MAX, and RAC1 and decreased the levels of mRNA transcripts of several genes, including CCND1, CDK2, SOS1, RHOA, GRB2, EGFR and KRAS. The CNFD treatment of MCF-7 cells induced the phosphorylation of c-jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinases (MAPKs) and inactivation of extracellular signal-regulated protein kinase 1/2 (ERK1/2). In a study using melanoma cells in a murine model in vivo, CNFD induced a potent anti-tumor activity. Herein, we describe, for the first time, the cytotoxicity and anti-tumor activity of CNFD and sequential mechanisms of apoptosis in MCF-7 cells. CNFD seems to be a promising candidate for anti-tumor therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , MAP Kinase Signaling System/drug effects , Melanoma/drug therapy , Naphthoquinones/therapeutic use , Reactive Oxygen Species/metabolism , Animals , Antineoplastic Agents/pharmacology , Caspases/metabolism , Cell Line, Tumor , DNA/metabolism , DNA Fragmentation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MAP Kinase Kinase 4/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred C57BL , Mitochondria/metabolism , Naphthoquinones/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Appl Immunohistochem Mol Morphol ; 29(7): e57-e67, 2021 08 01.
Article in English | MEDLINE | ID: mdl-33797435

ABSTRACT

Salivary gland neoplasms represent an important group of cancers in the head and neck and myoepithelial cells play a key role on the development these tumors. This study evaluated the distribution of mast cells and related proteins (PAR-2, TGFß1, IL-6) to the myofibroblastic differentiation in malignant tumors of salivary glands with and without myoepithelial differentiation. Immunohistochemical assessement for tryptase mast cells, SMA, PAR-2, TGFß1, IL-6 was performed in 10 cases of polymorphous low-grade adenocarcinoma, 14 cases of mucoepidermoid carcinoma (MEC) and 10 cases of adenoid cystic carcinoma. When the density of mast cells were compared between tumors, their density was significantly higher in MEC (P=0.08). Tumors with high expression of PAR-2 (79.4%) exhibited a high density of mast cells. Myofibroblasts were more frequent in malignant tumors with low expression (<50%) of cell masts. Individual analysis of the tumors showed no significant difference between the expression of PAR-2, IL-6, TGFß1, and myofibroblasts. When the density of mast cells, myofibroblasts and the expression of PAR-2 protein, IL-6, and TGFß1 were compared, it was no statistically significant difference between tumors with and without myoepithelial differentiation. The results of present study suggest a possible participation of mast cells and especially of PAR-2 in the development and progression of malignant salivary cancers, regardless of myoepithelial content.


Subject(s)
Cell Differentiation , Interleukin-6/metabolism , Mast Cells , Myofibroblasts , Neoplasm Proteins/metabolism , Receptor, PAR-2/metabolism , Salivary Gland Neoplasms , Transforming Growth Factor beta1/metabolism , Humans , Mast Cells/metabolism , Mast Cells/pathology , Myofibroblasts/metabolism , Myofibroblasts/pathology , Salivary Gland Neoplasms/metabolism , Salivary Gland Neoplasms/pathology
8.
Appl Immunohistochem Mol Morphol ; 29(5): 335-344, 2021.
Article in English | MEDLINE | ID: mdl-32769440

ABSTRACT

In oral squamous cell carcinoma (OSCC), involvement and activation of the Hedgehog pathway (HH) may be related to epithelial-mesenchymal transition and cell proliferation. The present study aimed to evaluate epithelial-mesenchymal transition and proliferative potential in OSCC cases demonstrating activation of the HH pathway. Twenty-three GLi-1-positive OSCC cases were submitted to immunohistochemical detection of Snail, Slug, N-cadherin, E-cadherin, ß-catenin, and MCM3 proteins. Clinical-pathologic immunoexpression data were obtained from the invasion front and tumor islets, and then compared. At the invasion front, OSCC cases presented positive Snail, Slug, and MCM3 expression in the nuclei of tumor cells. Loss of membrane and cytoplasmic expression of E-cadherin and ß-catenin was also observed. Positive N-cadherin expression was observed in 31.78% of the cases. GLi-1 immunoexpression was associated with loss of membrane E-cadherin (P<0.001), membrane ß-catenin (P<0.001), and cytoplasmic ß-catenin (P=0.02) expression. In the tumor islets, we observed nuclear expression of GLi-1, Snail, Slug, and MCM3. E-cadherin and ß-catenin showed positivity in tumor cell membranes. Statistically significant positive correlations between GLi-1 and Snail (P=0.05), E-cadherin (P=0.01), and cytoplasmic ß-catenin (P=0.04) were found. GLi-1 was associated with clinical staging, while membrane ß-catenin expression was related to the presence of metastasis in lymph nodes and to clinical staging. The HH pathway may be involved in regulating the expression of the mesenchymal phenotype. The loss of membrane E-cadherin and ß-catenin expression was observed at the tumor front region, whereas cell adhesion protein expression was detected in tumor islets regardless of MCM3.


Subject(s)
Biomarkers, Tumor/biosynthesis , Cell Proliferation , Epithelial-Mesenchymal Transition , Head and Neck Neoplasms , Squamous Cell Carcinoma of Head and Neck , Zinc Finger Protein GLI1/biosynthesis , Female , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Male , Middle Aged , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology
9.
Biomed Pharmacother ; 122: 109713, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31918282

ABSTRACT

Guatteria megalophylla Diels (Annonaceae) is an 8-10 m tall tree that grows near streams and is widely spread throughout Colombian, Ecuadorian, Peruvian, Brazilian and Guianese Amazon rainforest. Herein, we investigated for the first time the chemical composition and in vitro and in vivo anti-leukemia potential of G. megalophylla leaf essential oil (EO) using human promyelocytic leukemia HL-60 cells as model. EO was obtained by a hydrodistillation clevenger-type apparatus and characterized quali- and quantitatively by GC-MS and GC-FID, respectively. In vitro cytotoxic potential of EO was evaluated in human cancer cell lines (HL-60, MCF-7 CAL27, HSC-3, HepG2 and HCT116) and in human non-cancer cell line (MRC-5) by Alamar blue method. Annexin V/propidium iodide staining, cell cycle distribution and reactive oxygen species (ROS) were assessed by flow cytometry for HL-60 cells treated with EO. In vivo efficacy of EO (50 and 100 mg/kg) was evaluated in C.B-17 SCID mice with HL-60 cell xenografts. Chemical composition analyses showed spathulenol, γ-muurolene, bicyclogermacrene, ß-elemene and δ-elemene as main constituents of assayed sample. EO displayed in vitro cytotoxicity, including anti-leukemia effect with IC50 value of 12.51 µg/mL for HL-60 cells. EO treatment caused augment of phosphatidylserine externalization and DNA fragmentation without increasing of ROS in HL-60 cells. In vivo tumor mass inhibition rates of EO was 16.6-48.8 %. These data indicate anti-leukemia potential of G. megalophylla leaf EO.


Subject(s)
Annonaceae/chemistry , Cell Proliferation/drug effects , Guatteria/chemistry , Leukemia, Promyelocytic, Acute/drug therapy , Oils, Volatile/pharmacology , Plant Leaves/chemistry , Plant Preparations/pharmacology , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line , Cell Line, Tumor , HCT116 Cells , HL-60 Cells , Hep G2 Cells , Humans , MCF-7 Cells , Male , Mice, SCID
10.
J Ethnopharmacol ; 231: 516-524, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30445109

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Salacia impressifolia (Miers) A. C. Smith (family Celastraceae) is a traditional medicinal plant found in the Amazon Rainforest known as "miraruíra", "cipó-miraruíra" or "panu" and is traditionally used to treat dengue, flu, inflammation, pain, diabetes, male impotency, renal affections, rheumatism and cancer. AIM OF THE STUDY: The aim of this study was to investigate in vitro and in vivo anti-leukemia activity of the stem bark of S. impressifolia in experimental models. MATERIALS AND METHODS: The in vitro cytotoxic activity of extracts, fractions and quinonemethide triterpenes (22-hydroxytingenone, tingenone and pristimerin) from the stem bark of S. impressifolia in cultured cancer cells was determined. The in vivo antitumor activity of the ethyl acetate extract (EAE) and of its fraction (FEAE.3) from the stem bark of S. impressifolia was assessed in C.B-17 severe combined immunodeficient (SCID) mice engrafted with human promyelocytic leukemia HL-60 cells. RESULTS: The extract EAE, its fraction FEAE.3, and quinonemethide triterpenes exhibited potent cytotoxicity against cancer cell lines, including in vitro anti-leukemia activity against HL-60 and K-562 cells. Moreover, extract EAE and its fraction FEAE.3 inhibited the in vivo development of HL-60 cells engrafted in C.B-17 SCID mice. Tumor mass inhibition rates were measured as 40.4% and 81.5% for the extract EAE (20 mg/kg) and for its fraction FEAE.3 (20 mg/kg), respectively. CONCLUSIONS: Ethyl acetate extract and its fraction from the stem bark of S. impressifolia exhibit in vitro and in vivo anti-leukemia activity that can be attributed to their quinonemethide triterpenes. These data confirm the ethnopharmacological use of this species and may contribute to the development of a novel anticancer herbal medicine.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Leukemia/drug therapy , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Salacia , Animals , Cell Line , Female , Humans , Leukemia/pathology , Mice, SCID , Phytotherapy , Plant Bark , Plant Stems
11.
J Histochem Cytochem ; 66(6): 467-475, 2018 06.
Article in English | MEDLINE | ID: mdl-29553869

ABSTRACT

Mast cells (MCs) can influence the maturation of collagen fibers. This study evaluated the relationship between the distribution and degranulation of MCs and collagen maturation in human gingival tissue in chronic periodontitis. A total of 16 specimens of patients clinically diagnosed as periodontitis and 18 controls clinically diagnosed as healthy or gingivitis were included. Immunohistochemistry and Picrosirius staining were performed to identify MCs and assess collagen fibers, respectively. Chi-square, t test, and Pearson's correlation test ( p<0.05) were used. In control specimens, there was a positive association between MCs in the connective tissue and the presence of immature collagen ( p=0.001); in periodontitis samples, this association was not confirmed ( p≥0.12). There was no significant relationship between periodontal diagnosis and collagen maturation or MC degranulation ( p≥0.35). MC density was significantly higher ( p=0.04) in periodontitis tissue (339.01 ± 188.94 MCs/mm2) than in control tissue (211.14 ± 131.13 MCs/mm2) in the area of connective tissue containing inflammatory infiltrate. There was a correlation between the number of MCs and probing depth ( r = 0.34, p=0.04). MCs are involved in the pathogenesis of periodontal diseases and might be associated with collagen maturation in periodontal tissue during the early stages of periodontal disease pathogenesis.


Subject(s)
Chronic Periodontitis/pathology , Collagen/analysis , Mast Cells/pathology , Adolescent , Adult , Female , Humans , Immunohistochemistry/methods , Male , Middle Aged , Young Adult
12.
Cell Death Dis ; 9(2): 79, 2018 01 23.
Article in English | MEDLINE | ID: mdl-29362398

ABSTRACT

Ruthenium-based compounds have gained great interest due to their potent cytotoxicity in cancer cells; however, much of their potential applications remain unexplored. In this paper, we report the synthesis of a novel ruthenium complex with xanthoxylin (RCX) and the investigation of its cellular and molecular action in human hepatocellular carcinoma HepG2 cells. We found that RCX exhibited a potent cytotoxic effect in a panel of cancer cell lines in monolayer cultures and in a 3D model of multicellular cancer spheroids formed from HepG2 cells. This compound is detected at a high concentration in the cell nuclei, induces DNA intercalation and inhibits DNA synthesis, arresting the cell cycle in the S-phase, which is followed by the activation of the caspase-mediated apoptosis pathway in HepG2 cells. Gene expression analysis revealed changes in the expression of genes related to cell cycle control, apoptosis and the MAPK pathway. In addition, RCX induced the phosphorylation of ERK1/2, and pretreatment with U-0126, an MEK inhibitor known to inhibit the activation of ERK1/2, prevented RCX-induced apoptosis. In contrast, pretreatment with a p53 inhibitor (cyclic pifithrin-α) did not prevent RCX-induced apoptosis, indicating the activation of a p53-independent apoptosis pathway. RCX also presented a potent in vivo antitumor effect in C.B-17 SCID mice engrafted with HepG2 cells. Altogether, these results indicate that RCX is a novel anticancer drug candidate.


Subject(s)
Acetophenones/pharmacology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , MAP Kinase Signaling System/drug effects , Ruthenium/pharmacology , S Phase/drug effects , Tumor Suppressor Protein p53/metabolism , Acetophenones/chemical synthesis , Acetophenones/chemistry , Animals , Antineoplastic Agents/pharmacology , Caspase Inhibitors/pharmacology , Caspases/metabolism , DNA/biosynthesis , Female , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Intercalating Agents/pharmacology , Membrane Potential, Mitochondrial/drug effects , Mice, SCID , Models, Biological , Protein Kinase Inhibitors/pharmacology , Proton Magnetic Resonance Spectroscopy , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Xenograft Model Antitumor Assays
13.
Oncotarget ; 8(61): 104367-104392, 2017 Nov 28.
Article in English | MEDLINE | ID: mdl-29262647

ABSTRACT

Piplartine (piperlongumine) is a plant-derived molecule that has been receiving intense interest due to its anticancer characteristics that target the oxidative stress. In the present paper, two novel piplartine-containing ruthenium complexes [Ru(piplartine)(dppf)(bipy)](PF6)2 (1) and [Ru(piplartine)(dppb)(bipy)](PF6)2 (2) were synthesized and investigated for their cellular and molecular responses on cancer cell lines. We found that both complexes are more potent than metal-free piplartine in a panel of cancer cell lines on monolayer cultures, as well in 3D model of cancer multicellular spheroids formed from human colon carcinoma HCT116 cells. Mechanistic studies uncovered that the complexes reduced the cell growth and caused phosphatidylserine externalization, internucleosomal DNA fragmentation, caspase-3 activation and loss of the mitochondrial transmembrane potential on HCT116 cells. Moreover, the pre-treatment with Z-VAD(OMe)-FMK, a pan-caspase inhibitor, reduced the complexes-induced apoptosis, indicating cell death by apoptosis through caspase-dependent and mitochondrial intrinsic pathways. Treatment with the complexes also caused a marked increase in the production of reactive oxygen species (ROS), including hydrogen peroxide, superoxide anion and nitric oxide, and decreased reduced glutathione levels. Application of N-acetyl-cysteine, an antioxidant, reduced the ROS levels and apoptosis induced by the complexes, indicating activation of ROS-mediated apoptosis pathway. RNA transcripts of several genes, including gene related to the cell cycle, apoptosis and oxidative stress, were regulated under treatment. However, the complexes failed to induce DNA intercalation. In conclusion, the complexes are more potent than piplartine against different cancer cell lines and are able to induce caspase-dependent and mitochondrial intrinsic apoptosis on HCT116 cells by ROS-mediated pathway.

SELECTION OF CITATIONS
SEARCH DETAIL
...