Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Molecules ; 26(23)2021 Nov 30.
Article in English | MEDLINE | ID: mdl-34885849

ABSTRACT

Colorectal cancer (CRC) is a leading cause of cancer-related death. The demand for new therapeutic approaches has increased attention paid toward therapies with high targeting efficiency, improved selectivity and few side effects. Porphyrins are powerful molecules with exceptional properties and multifunctional uses, and their special affinity to cancer cells makes them the ligands par excellence for anticancer drugs. Porphyrin derivatives are used as the most important photosensitizers (PSs) for photodynamic therapy (PDT), which is a promising approach for anticancer treatment. Nevertheless, the lack of solubility and selectivity of the large majority of these macrocycles led to the development of different photosensitizer complexes. In addition, targeting agents or nanoparticles were used to increase the efficiency of these macrocycles for PDT applications. On the other hand, gold tetrapyrrolic macrocycles alone showed very interesting chemotherapeutic activity without PDT. In this review, we discuss the most important porphyrin derivatives, alone or associated with other drugs, which have been found effective against CRC, as we describe their modifications and developments through substitutions and delivery systems.


Subject(s)
Colorectal Neoplasms/drug therapy , Porphyrins/therapeutic use , Humans , Nanoparticles/chemistry , Photochemotherapy , Photosensitizing Agents/therapeutic use , Porphyrins/chemistry
2.
Bioorg Med Chem Lett ; 41: 128024, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33845130

ABSTRACT

Two protoporphyrin IX (PpIX) adamantane derivatives were synthesized and then metallated with zinc. The Zn-PpIX derivatives, exhibiting a high singlet oxygen quantum yield, were tested for their photodynamic activity against the HT-29 cell line. In order to enhance their water-solubility and their cellular bioavailability, these photosensitizers were encapsulated into the hydrophobic cavity of cyclodextrins (CD) previously attached to cellulose nanocrystals (CNCs) via electrostatic interactions. Under illumination, the encapsulated adamantanyl-porphyrins exerted an enhanced in vitro cytotoxicity, as compared with the corresponding free photosensitizers.


Subject(s)
Adamantane/pharmacology , Antineoplastic Agents/pharmacology , Cellulose/pharmacology , Cyclodextrins/pharmacology , Photochemotherapy , Photosensitizing Agents/pharmacology , Protoporphyrins/pharmacology , Adamantane/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Cellulose/chemistry , Cyclodextrins/chemistry , Dose-Response Relationship, Drug , Drug Design , Drug Screening Assays, Antitumor , HT29 Cells , Humans , Molecular Structure , Nanoparticles/chemistry , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/chemistry , Protoporphyrins/chemistry , Structure-Activity Relationship
3.
J Am Chem Soc ; 141(46): 18444-18454, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31625740

ABSTRACT

Marine alkaloid rigidins are cytotoxic compounds known to kill cancer cells at nanomolar concentrations by targeting the microtubule network. Here, a rigidin analogue containing a thioether group was "caged" by coordination of its thioether group to a photosensitive ruthenium complex. In the dark, the coordinated ruthenium fragment prevented the rigidin analogue from inhibiting tubulin polymerization and reduced its toxicity in 2D cancer cell line monolayers, 3D lung cancer tumor spheroids (A549), and a lung cancer tumor xenograft (A549) in nude mice. Photochemical activation of the prodrug upon green light irradiation led to the photosubstitution of the thioether ligand by water, thereby releasing the free rigidin analogue capable of inhibiting the polymerization of tubulin. In cancer cells, such photorelease was accompanied by a drastic reduction of cell growth, not only when the cells were grown in normoxia (21% O2) but also remarkably in hypoxic conditions (1% O2). In vivo, low toxicity was observed at a dose of 1 mg·kg-1 when the compound was injected intraperitoneally, and light activation of the compound in the tumor led to 30% tumor volume reduction, which represents the first demonstration of the safety and efficacy of ruthenium-based photoactivated chemotherapy compounds in a tumor xenograft.


Subject(s)
Alkaloids/chemistry , Alkaloids/therapeutic use , Lung Neoplasms/drug therapy , Pyrimidines/chemistry , Pyrimidines/therapeutic use , Pyrroles/chemistry , Pyrroles/therapeutic use , Tubulin Modulators/chemistry , Tubulin Modulators/therapeutic use , A549 Cells , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Proliferation/drug effects , Humans , Light , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice, Nude , Microtubules/drug effects , Microtubules/metabolism , Microtubules/pathology , Oxygen/metabolism , Prodrugs/chemistry , Prodrugs/therapeutic use , Tumor Hypoxia/drug effects
4.
J Geriatr Oncol ; 9(1): 60-67, 2018 01.
Article in English | MEDLINE | ID: mdl-28851511

ABSTRACT

OBJECTIVES: Geriatric patients with hematologic malignancies (HMs) are prescribed targeted and supportive care treatments that add to the preexisting polypharmacy (PP). PP is associated with an increased risk of potentially inappropriate medications (PIM) and drug-drug interactions (DDI) resulting in increased hospitalization and mortality in the elderly. As very few data exist on these medication issues in the context of HMs, the objective of this study was to evaluate prevalence of PP, DDI and PIM use at baseline and 3months among elderly patients with HMs who received baseline geriatric assessment. METHODS: PP, DDI and PIM use were assessed by a clinical pharmacist at two time points in patients over 75years with HMs undergoing chemotherapy. PP was defined as the concurrent use of five or more medications. DDIs were evaluated according to the literature and prescription analysis software. PIMs were assessed according to the Laroche list. RESULTS: 122 patients (mean age 81.5; 6.6 medications) were included and after 3months, 86 patients (5.8 medications) were available for a second assessment. Prevalence of PP, PIM and DDI at inclusion was 75.4%, 34.4% and 71.3%, respectively. PP was the only medication risk that was significantly reduced (p<0.05) at 3months (65.1%) compared to admission. CONCLUSION: This observational study highlighted that PP decreased over time but neither DDI nor PIM use were reduced. A pharmacist-led evaluation might help to manage these medication issues.


Subject(s)
Drug Interactions , Hematologic Neoplasms/drug therapy , Polypharmacy , Aged , Aged, 80 and over , Female , Geriatric Assessment , Hematologic Neoplasms/complications , Humans , Longitudinal Studies , Male , Medication Reconciliation/organization & administration , Pharmacists , Potentially Inappropriate Medication List/statistics & numerical data
6.
Bioorg Med Chem Lett ; 27(18): 4354-4357, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28838697

ABSTRACT

The aim of this study is to synthesize chalcone-polyamine conjugates in order to enhance bioavailability and selectivity of chalcone core towards cancer cells, using polyamine-based vectors. 3-hydroxy-3',4,4',5'-tetramethoxychalcone (1) and 3',4,4',5'-tetramethoxychalcone (2) were selected as parent chalcones since they were found to be efficient anti-proliferative agents on various cancer cells. A series of ten chalcone-polyamine conjugates was obtained by reacting carboxychalcones with different polyamine tails. Chalcones 1 and 2 showed a strong cytotoxic activity against two prostatic cancer (PC-3 and DU-145) and two colorectal cancer (HT-29 and HCT-116) cell lines. Then, chalcone-spermine conjugates 7d and 8d were shown to be the most active of the series and could be considered as promising compounds for colon and prostatic cancer adjuvant therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Chalcone/pharmacology , Drug Design , Polyamines/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Chalcone/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Polyamines/chemistry , Structure-Activity Relationship
7.
Med Sci Monit Basic Res ; 23: 270-284, 2017 Aug 03.
Article in English | MEDLINE | ID: mdl-28769026

ABSTRACT

BACKGROUND In the present study, phytochemical screening, antioxidant, anti-inflammatory, and antiproliferative capacities of 3 extracts from leaves of Lebanese Crataegus azarolus L. were evaluated. MATERIAL AND METHODS Fresh leaves were dissolved in 3 different solvents: distilled water, ethanol, and methanol. The chemical composition was determined using high-performance liquid chromatography (HPLC) and the content of essential oil of this plant was examined by gas chromatography (GC) coupled with mass spectrometry (MS). The antioxidant potential was evaluated using DPPH radical scavenging and Fe2+ chelating activity assays. Anti-inflammatory effect was investigated by measuring the secreted amounts of the proinflammatory mediator PGE2 using ELISA technique, as well as by assaying the mRNA levels of the proinflammatory cytokines (IL-α, IL-ß, and Il-6), chemokines (CCL3 and CCL4) and inflammation-sensitive COX2 and iNOS enzymes using quantitative real-time PCR (qRT-PCR). The antiproliferative effect was evaluated using the XTT viability assay. RESULTS The obtained results show that alcohol (methanol and ethanol) extracts were rich in bioactive molecules with medical relevance and exerted substantial antioxidant, anti-inflammatory, and antiproliferative capacities. On the other hand, aqueous extract contained fewer chemical components and exhibited less therapeutic efficiency. CONCLUSIONS Our observations indicate that Crataegus azarolus L. could be used for treating diseases related to oxidative stress, inflammatory reactions, and uncontrolled cell growth.


Subject(s)
Crataegus/chemistry , Plant Extracts/chemistry , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Cell Proliferation/drug effects , Chromatography, High Pressure Liquid , Gas Chromatography-Mass Spectrometry/methods , Phytochemicals/analysis , Phytotherapy , Plant Extracts/pharmacology , Plant Leaves/chemistry
8.
Bioorg Med Chem Lett ; 26(10): 2503-2506, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27040657

ABSTRACT

A new anti-cancer drug delivery system, based on gold nanoparticles, has been designed for hydrophobic active compounds. The system is a conjugate of gold/polyethyleneimine (AuNPs/PEI) nanoparticles and sulphated ß-cyclodextrin (CD). Anionic cyclodextrin was attached to the positively charged AuNPs/PEI nanoparticles by ionic bonds. Tanshinone IIA and α-mangostin were extracted, purified and encapsulated into the AuNPs/PEI/CD nanoparticles. In vitro preliminary cell viability assays against prostate cancer cell lines PC-3 and DU145 showed that encapsulation resulted in increased cytotoxicity.


Subject(s)
Abietanes/administration & dosage , Drug Delivery Systems/methods , Polyethyleneimine/chemistry , Prostatic Neoplasms/drug therapy , Xanthones/administration & dosage , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/chemistry , Cell Line, Tumor , Cyclodextrins/chemistry , DNA Fragmentation/drug effects , Gold/chemistry , Humans , Male , Microscopy, Electron, Transmission , Nanoparticles/chemistry , Polyethyleneimine/administration & dosage , Prostatic Neoplasms/pathology , Xanthones/chemistry
9.
Bioorg Med Chem Lett ; 26(3): 941-945, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26739777

ABSTRACT

The synthesis of curcumin-cyclodextrin/cellulose nanocrystals (CNCx) nano complexes was performed. CNCx were functionalized by ionic association with cationic ß-cyclodextrin (CD) and CD/CNCx complexes were used to encapsulate curcumin. Preliminary in vitro results showed that the resulting curcumin-CD/CNCx complexes exerted antiproliferative effect on colorectal and prostatic cancer cell lines, with IC50s lower than that of curcumin alone.


Subject(s)
Cellulose/chemistry , Curcumin/chemistry , Nanoparticles/chemistry , beta-Cyclodextrins/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Curcumin/toxicity , Drug Carriers/chemistry , HT29 Cells , Humans , Microscopy, Confocal
10.
Int J Oncol ; 47(1): 220-30, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25997834

ABSTRACT

The aim of this study was to describe and understand the relationship between cyclooxygenase-2 (COX-2) expression and apoptosis rate in erythroleukemia cells after apoptosis induction by Berberis libanotica (Bl) extract. To achieve this goal we used erythroleukemia cell lines expressing COX­2 (HEL cell line) or not (K562 cell line). Moreover, we made use of COX­2 cDNA to overexpress COX­2 in K562 cells. In light of the reported chemopreventive and chemosensitive effects of natural products on various tumor cells and animal models, we postulated that our Bl extract may mediate their effects through apoptosis induction with suppression of cell survival pathways. Our study is the first report on the specific examination of intrinsic apoptosis and Akt/NF-κB/COX­2 pathways in human erythroleukemia cells upon Bl extract exposure. Even if Bl extract induced apoptosis of three human erythroleukemia cell lines, a dominant effect of Bl extract treatment on K562 cells was observed resulting in activation of the late markers of apoptosis with caspase-3 activation, PARP cleavage and DNA fragmentation. Whereas, we showed that Bl extract reduced significantly expression of COX­2 by a dose-dependent manner in HEL and K562 (COX­2+) cells. Furthermore, in regard to our results, it is clear that the simultaneous inhibition of Akt and NF-κB signalling can significantly contribute to the anticancer effects of Bl extract in human erythroleukemia cells. We observed that the Bl extract is clearly more active than the berberine alone on the induction of DNA fragmentation in human erythro-leukemia cells.


Subject(s)
Berberis/chemistry , Leukemia, Erythroblastic, Acute/metabolism , Plant Extracts/pharmacology , Signal Transduction/drug effects , Apoptosis , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cyclooxygenase 2/metabolism , DNA Fragmentation , Humans , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism
11.
Anticancer Drugs ; 26(1): 74-84, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25192452

ABSTRACT

Prostate cancer is the most common malignant cancer in men and the second leading cause of cancer deaths. Previously, we have shown that 2'-hydroxy-4-methylsulfonylchalcone (RG003) induced apoptosis in prostate cancer cell lines PC-3 and DU145. Although tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising anticancer agent, some cancer cells are resistant to TRAIL treatment. PC-3 and LNCaP prostatic cancer cell lines have been reported to be resistant to TRAIL-induced apoptosis. Here, we show for the first time that RG003 overcomes TRAIL resistance in prostate cancer cells. RG003 can enhance TRAIL-induced apoptosis through DR5 upregulation and downregulation of Bcl-2, PI3K/Akt, NF-κB, and cyclooxygenase-2 (COX-2) survival pathways. When used in combined treatment, RG003 and TRAIL amplified TRAIL-induced activation of apoptosis effectors and particularly activation of caspase-8 and the executioner caspase-3, leading to increased poly-ADP-ribose polymerase cleavage and DNA fragmentation in prostate cancer cells. Furthermore, we showed that RG003 reduced COX-2 expression in cells. Previously, we showed that COX-2 was involved in resistance to an apoptosis mechanism; then, its inhibition by RG003 could render cells more sensitive to TRAIL treatment. We showed that nuclear factor-κB activation was inhibited after RG003 treatment. This inhibition was correlated with reduction in COX-2 expression and induction of apoptosis. Overall, we conclude, for the first time, that RG003 can enhance TRAIL-induced apoptosis in human prostate cancer cells. The significance of our in-vitro study with RG003 and TRAIL combined is very encouraging, suggesting the relevance of testing this combined treatment in xenograft animal models.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Chalcones/pharmacology , Prostatic Neoplasms/pathology , Sulfones/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm , Drug Synergism , Humans , Male , NF-kappa B/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Signal Transduction , Transcription Factor AP-1/metabolism
12.
Int J Oncol ; 43(4): 1160-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23877542

ABSTRACT

Limited success has been achieved in extending the survival of patients with metastatic and hormone-refractory prostate cancer (HRPC). There is a strong need for novel agents in the treatment and prevention of HRPC. In the present study, the apoptotic mechanism of action of RG003 (2'-hydroxy-4-methylsulfonylchalcone) and RG005 (4'-chloro-2'-hydroxy-4-methylsulfonylchalcone) in association with intracellular signalling pathways was investigated in the hormone-independent prostate carcinoma cells PC-3 and DU145. We showed that these compounds induced apoptosis through the intrinsic pathway but not through the extrinsic one. We showed that synthetic chalcones induced an activation of caspase-9 but not caspase-8 in PC-3 cells. Even if both chalcones induced apoptosis in PC-3 cells, a dominant effect of RG003 treatment was observed resulting in a disruption of ∆ψm, caspase-9 and caspase-3 activation, PARP cleavage and DNA fragmentation. Furthermore, in regard to our results, it is clear that the simultaneous inhibition of Akt and NF-κB signalling can significantly contribute to the anticancer effects of RG003 and RG005 in PC-3 prostate cancer cells. NF-κB inhibition was correlated with the reduction of COX-2 expression and induction of apoptosis. Our results clearly indicate for the first time that RG003 and RG005 exert their potent anti­proliferative and pro-apoptotic effects through the modulation of Akt/NF-κB/COX-2 signal transduction pathways in PC-3 prostate cancer cells with a dominant effect for RG003.


Subject(s)
Apoptosis/drug effects , Caspase 8/genetics , Caspase 9/genetics , Chalcones/administration & dosage , Poly(ADP-ribose) Polymerases/metabolism , Sulfones/administration & dosage , Antineoplastic Agents , Cell Line, Tumor , Cell Proliferation , Chalcones/chemistry , Cyclooxygenase 2/metabolism , DNA Fragmentation , Humans , Male , NF-kappa B/metabolism , Oncogene Protein v-akt/metabolism , Prostatic Neoplasms , Signal Transduction , Sulfones/chemistry
13.
Cancer Res ; 72(14): 3463-70, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22593187

ABSTRACT

Integrins play a role in the resistance of advanced cancers to radiotherapy and chemotherapy. In this study, we show that high expression of the α5 integrin subunit compromises temozolomide-induced tumor suppressor p53 activity in human glioblastoma cells. We found that depletion of the α5 integrin subunit increased p53 activity and temozolomide sensitivity. However, when cells were treated with the p53 activator nutlin-3a, the protective effect of α5 integrin on p53 activation and cell survival was lost. In a functional p53 background, nutlin-3a downregulated the α5 integrin subunit, thereby increasing the cytotoxic effect of temozolomide. Clinically, α5ß1 integrin expression was associated with a more aggressive phenotype in brain tumors, and high α5 integrin gene expression was associated with decreased survival of patients with high-grade glioma. Taken together, our findings indicate that negative cross-talk between α5ß1 integrin and p53 supports glioma resistance to temozolomide, providing preclinical proof-of-concept that α5ß1 integrin represents a therapeutic target for high-grade brain tumors. Direct activation of p53 may remain a therapeutic option in the subset of patients with high-grade gliomas that express both functional p53 and a high level of α5ß1 integrin.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm , Glioblastoma/drug therapy , Glioblastoma/metabolism , Integrin alpha5beta1/physiology , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Tumor , Dacarbazine/therapeutic use , Humans , Imidazoles/pharmacology , Integrin alpha5beta1/metabolism , Mice , Piperazines/pharmacology , Temozolomide , Treatment Outcome
14.
Pharm Biol ; 48(3): 269-74, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20645812

ABSTRACT

Aristolochia baetica L. (Aristolochiaceae) and Origanum compactum Benth. (Lamiaceae) are native plants of Morocco used in traditional medicine. In order to systematically evaluate their potential activity on human breast cancer, four different polarity extracts from each plant were assessed in vitro for their antiproliferative effect on MCF-7 cells. As a result, several extracts of those plants showed potent cell proliferation inhibition on MCF-7 cells. Chloroform extract of A. baetica (IC50: 216.06 +/- 15 microg/mL) and ethyl acetate of O. compactum (IC50: 279.51 +/- 16 microg/mL) were the most active. Thin layer chromatography examination of the bioactive extracts of A. baetica and O. compactum showed the presence of aristolochic acid and betulinic acid, respectively. These results call for further studies of these extracts.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Aristolochia/chemistry , Breast Neoplasms/drug therapy , Origanum/chemistry , Phytotherapy , Plant Extracts/therapeutic use , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Aristolochic Acids/analysis , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Inhibitory Concentration 50 , Medicine, African Traditional , Morocco , Pentacyclic Triterpenes , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Solvents/chemistry , Triterpenes/analysis , Betulinic Acid
15.
Int J Cancer ; 127(5): 1240-8, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20099278

ABSTRACT

The alpha5beta1 integrin represent a new therapeutic target for glioblastoma, which are malignant brain tumors difficult to cure with conventional therapies. Glioblastoma are known to be highly resistant to chemotherapy. We, therefore, investigated whether blocking alpha5beta1 integrin with specific nonpeptidic antagonists concomitantly with chemotherapy (ellipticine and temozolomide) may impact the response to chemotherapy of human glioblastoma. Here we show that inhibiting alpha5beta1 integrin with 2 selective ligands (SJ749 and K34c) decreases chemotherapy-induced premature senescence and facilitates cell apoptosis in a functional p53 background (U87MG cells). When p53 is mutated and inactive (U373 cells), chemotherapy induces p53-independent cell apoptosis instead of senescence that is not improved by integrin antagonists. Silencing p53 in U87MG cells with siRNA as well as evaluating HCT116 p53+/+ and p53-/- colon carcinoma cell behavior support the hypothesis of an as yet unknown effect of alpha5beta1 integrin antagonists on the control of chemotherapy-induced premature senescence and apoptosis. alpha5beta1 integrin antagonists modulate the p53 signaling induced by chemotherapy. Our results highlight a new role of the alpha5beta1 integrin in the control of glioblastoma aggressiveness and responsiveness to chemotherapy, which may have a crucial impact in the clinical management of patients suffering from brain tumors.


Subject(s)
Aging/drug effects , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Glioblastoma/pathology , Integrin alpha5beta1/antagonists & inhibitors , Propionates/pharmacology , Pyridines/pharmacology , Spiro Compounds/pharmacology , Tumor Suppressor Protein p53/metabolism , Blotting, Western , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , Integrin alpha5beta1/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics
16.
Fundam Clin Pharmacol ; 23(5): 549-56, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19656204

ABSTRACT

Many natural components of plants extract are studied for their beneficial effects on health and particularly on carcinogenesis chemoprevention. In this study, we investigated the effect of citral (3,7-dimethyl-2,6-octadienal), a key component of essential oils extracted from several herbal plants, on the proliferation rate, cell cycle distribution, and apoptosis of the human breast cancer cell line MCF-7. The effects of this compound were also tested on cyclo-oxygenase activity. Citral treatment caused inhibition of MCF-7 cell growth (IC(50)-48 h: 18 x 10(-5)m), with a cycle arrest in G(2)/M phase and apoptosis induction. Moreover, we observed a decrease in prostaglandin E(2) synthesis 48 h after citral treatment. These findings suggest that citral has a potential chemopreventive effect.


Subject(s)
Anticarcinogenic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Monoterpenes/pharmacology , Acyclic Monoterpenes , Cell Culture Techniques , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/pathology , Cyclooxygenase 2/biosynthesis , DNA Fragmentation/drug effects , Dinoprostone/biosynthesis , Dose-Response Relationship, Drug , Humans , Regression Analysis , Time Factors
17.
Int J Mol Med ; 20(4): 451-60, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17786275

ABSTRACT

We conducted our study to assess the antiproliferative and proapoptotic potential of hecogenin and tigogenin, two saponins which are structurally similar to diosgenin. We particularly focused our attention on mitogen-activated protein kinase (MAPK) activation in relation to apoptosis but also with the COX-2 expression and activity. Rheumatoid arthritis (RA) synoviocytes were isolated from fresh synovial biopsies obtained from five RA patients undergoing hip arthroplasty. Measurement of cell proliferation was determined using the MTT assay. Apoptosis was evaluated by studying caspase-8, caspase-9 and caspase-3 activities but also by quantification of DNA fragmentation. Quantification of human phospho-MAPKs was realized by ELISA. COX-2 expression was demonstrated by Western blot analysis and COX-2 activity by assay of endogenous prostaglandin E2 (PGE2) production. Tigogenin was more effective than hecogenin in inducing apoptosis in human RA fibroblast-like synoviocytes (FLS) which was caspase dependent but poly(ADP-ribose) polymerase independent and characterized by DNA fragmentation. Our results demonstrated hecogenin- and tigogenin-induced apoptosis through activation of p38 without affecting the JNK and ERK pathways. Indeed, pretreatment with a p38 inhibitor decreased saponin-induced apoptosis with a significant decrease in DNA fragmentation. Furthermore, the rate of apoptosis induced by hecogenin or tigogenin was associated with overexpression of COX-2 correlated with overproduction of endogenous PGE2. These new results provide strong evidence that a family of structurally similar plant steroids is capable of inducing apoptosis in human RA FLS with different rates and different signalling pathways. This study also confirms the discussed appearance of the downregulation or upregulation of COX-2 in cell apoptosis as a function of cell type.


Subject(s)
Apoptosis/drug effects , Arthritis, Rheumatoid/pathology , Cyclooxygenase 2/genetics , Sapogenins/pharmacology , Spirostans/pharmacology , Synovial Fluid/cytology , p38 Mitogen-Activated Protein Kinases/metabolism , Arthritis, Rheumatoid/enzymology , Caspase 8/metabolism , Caspase 9/metabolism , Cell Death/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Cell Survival/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/drug effects , Fibroblasts/pathology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Middle Aged , Poly(ADP-ribose) Polymerases/metabolism , Sapogenins/chemistry , Saponins/pharmacology , Spirostans/chemistry , Synovial Fluid/drug effects , Synovial Fluid/enzymology , Up-Regulation/drug effects
18.
Int J Mol Med ; 19(1): 113-22, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17143555

ABSTRACT

In the present study, we investigated the signalling pathways involved in diosgenin-induced apoptosis in human rheumatoid arthritis (RA) fibroblast-like synoviocytes (FLS) in vitro with particular interest on Akt and MAPKs activation in relation to arachidonic acid metabolism via COX-2 pathway. MAPK activation was measured by ELISA quantification in diosgenin-treated human RA FLS. Expression of Akt and phospho-Akt was analyzed by Western blot analysis. Nuclear factor-kappaB (NF-kappaB) translocation was evaluated by electromobility shift assay. The prostanoid production (COX-2 activity) was measured by quantitative ELISA. Diosgenin-induced apoptosis in the presence of MAPK or Akt inhibitors was detected by a quantitative determination of DNA fragmentation. Treatment of human RA FLS with 40 microM diosgenin caused an activation of p38 and JNK and an inhibition of ERK phosphorylation. Akt and NF-kappaB are potentially required for diosgenin-induced apoptosis in human RA FLS because 40 microM diosgenin abrogated Akt phosphorylation which correlated with an inhibition of NF-kappaB nuclear translocation. SB203580 and SP600125 (p38 and JNK inhibitors) reduced diosgenin-induced DNA fragmentation whereas U0126 and LY294002 (MEK and PI3 kinase/Akt inhibitors) caused an amplification of proapoptotic effect of diosgenin. Diosgenin increased COX-2 activity resulting in PGE2 and 6-keto-PGF1alpha overproduction in human RA FLS. All MAPK inhibitors markedly reduced diosgenin-induced PGE2 and 6-keto-PGF1alpha synthesis except for SP600125 on 6-keto-PGF1alpha production. These results provide, for the first time, strong evidence that a combined association implicating a MEK inhibitor (U0126) and diosgenin is the most effective in inducing very strong apoptosis with down-regulation of COX-2 expression and activity in human RA FLS.


Subject(s)
Arthritis, Rheumatoid/metabolism , Cyclooxygenase 2/metabolism , Diosgenin/pharmacology , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Prostaglandins/biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , Aged , Apoptosis , Cells, Cultured , Fibroblasts/metabolism , Humans , MAP Kinase Signaling System , Middle Aged , Models, Biological , NF-kappa B/metabolism , Synovial Membrane/cytology
19.
Int J Oncol ; 28(1): 201-7, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16327997

ABSTRACT

Megakaryocytopoiesis is characterized by progressive polyploidization and acquisition of megakaryo-cytic markers. MAPK pathways play a key role during megakaryocytic differentiation of megakaryocyte precursors or leukemic cells. Apoptosis is the physiological fate of normal megakaryocyte after differentiation and maturation. The aim of this study was to investigate the signaling pathways involved in diosgenin-induced differentiation and the fate of diosgenin-differentiated HEL cells. The present report shows that diosgenin induced megakaryocytic differentiation of HEL cells through a combined activation of ERK and inhibition of the p38 MAPK pathways. Inhibition of ERK activation by a MEK inhibitor abrogated diosgenin-induced differentiation. Afterwards, differentiated cells showed a marked inhibition of expression of survival factors NF-kappaB, Akt and Bcl-xL and activation of caspase-3 together with PARP cleavage leading to apoptotic death of diosgenin-differentiated cells.


Subject(s)
Apoptosis/genetics , Cell Differentiation , Megakaryocytes/physiology , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-kappa B/physiology , Diosgenin/pharmacology , Humans , Leukemia, Myeloid, Acute/pathology , Signal Transduction , Tumor Cells, Cultured
20.
Int J Mol Med ; 16(6): 1095-101, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16273292

ABSTRACT

Diosgenin is a steroidal sapogenin with antitumor properties. We previously showed that diosgenin induced apoptosis in human erythroleukemia (HEL) cells. In order to elucidate the mechanism of its apoptotic activity, we investigated the effect of diosgenin on nuclear factor-kappaB (NF-kappaB) binding and on three groups of human mitogen-activated protein kinases (MAPKs) in relation to diosgenin-induced apoptosis in different erythroleukemia cell lines (K562 and HEL). Our results showed that diosgenin decreased DNA binding of NF-kappaB in K562 and HEL cells after 48-h diosgenin treatment. This inhibition of NF-kappaB binding was correlated with strong apoptosis in both erythroleukemia cell lines. Diosgenin-induced apoptosis was associated with cyclo-oxygenase-2 (COX-2) up-regulation in HEL cells but not in K562 cells which are COX-2 deficient. Furthermore, diosgenin inhibited extracellular signal-regulated kinase (ERK) activation only in HEL cells. However, diosgenin activated p38 MAPK in both cell lines and activated c-jun NH2-terminal kinases (JNKs) only in HEL cells. Pre-treatment with a selective p38 inhibitor inhibited diosgenin-induced DNA fragmentation in K562 cells. For the first time, our results suggest that inhibition of NF-kappaB nuclear binding and p38 MAPK activation are involved in the diosgenin-mediated signal cascades in K562 cells for inducing/regulating DNA fragmentation.


Subject(s)
Apoptosis/drug effects , Cyclooxygenase 2/deficiency , Diosgenin/pharmacology , MAP Kinase Signaling System/drug effects , NF-kappa B/antagonists & inhibitors , Phytosterols/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Caspase 3 , Caspases/metabolism , Cell Line, Tumor , Cyclooxygenase 2/metabolism , DNA Fragmentation/drug effects , Enzyme Activation/drug effects , Humans , Imidazoles/pharmacology , K562 Cells , NF-kappa B/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Pyridines/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...