Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Mol Cell Endocrinol ; 590: 112267, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38729597

ABSTRACT

Mammary gland (MG) lactogenic differentiation involves epigenetic mechanisms. We have previously shown that hypothyroidism (HypoT) alters the MG transcriptome in lactation. However, the role of thyroid hormones (T3 and T4 a. k.a. THs) in epigenetic differentiation of MG is still unknown. We used a model of post-lactating HypoT rats to study in MG: a) Methylation and expression level of Gata3, Elf5, Stat6, Stat5a, Stat5b; b) Expression of Lalba, IL-4Rα and Ncoa1 mRNA; c) Histone H3 acetylation and d) Estrogen and progesterone concentration in serum. HypoT increases the estrogen serum level, decreases the progesterone level, promotes methylation of Stat5a, Stat5b and Stat6, decreasing their mRNA level and of its target genes (Lalba and IL-4Rα) and increases the Ncoa1 mRNA expression and histone H3 acetylation level. Our results proved that HypoT alters the post-lactation MG epigenome and could compromise mammary functional differentiation.


Subject(s)
Cell Differentiation , Epigenesis, Genetic , Histones , Hypothyroidism , Mammary Glands, Animal , Animals , Female , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Hypothyroidism/genetics , Hypothyroidism/metabolism , Hypothyroidism/pathology , Histones/metabolism , Cell Differentiation/genetics , Rats , Acetylation , Progesterone/blood , Rats, Wistar , Estrogens/metabolism , DNA Methylation/genetics , Lactation , RNA, Messenger/genetics , RNA, Messenger/metabolism
2.
J Tradit Complement Med ; 13(1): 20-29, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36685075

ABSTRACT

Background: New products with tolerogenic properties on T cell response are necessary to improve current efficacy, cost and side effects of immunosuppressants. Prosopis strombulifera aqueous extract (PsAE) have reported cytotoxic, antitumoral, antiatherogenic and antileishmanial activities, containing phytochemicals with immune-related activities. Despite these, there are no previous studies with respect to PsAE suppressive properties over T cell proliferation and their function. Goal: Because of previous antecedents, this study aims to evaluate the effect of PsAE on T cell activation, proliferation, cytokine production, and to investigate its effect over an in vivo model of type 1 diabetes (T1D). Experimental procedure: Splenocytes and sorted CD4+/CD8+ from wild type C57BL/6 mice were cultured to determine activation, IFN-γ release and T-cell proliferation after polyclonal stimulation. NOD (non-obese diabetic) mice were used to study the effects of orally administered extract on glycemia, insulitis stages and perforin-1 (PRF-1)/granzyme-B (GRZ-B) expression. Results: In primary cultures, the plant extract impairs T cell activation, decreases IFN-γ release, and reduces proliferation after different polyclonal stimuli. In vivo, PsAE improves NOD mice glycemic levels and T1D progression by diminution of pancreas insulitis and reduction of PRF-1 and GRZ-B mRNA expression. To our knowledge, this is the first report characterizing the therapeutic properties of PsAE on T cell activation. Conclusion: The current work provides evidence about in vitro and in vivo immunosuppressive effects of PsAE and promotes this plant extract as a complementary and alternative treatment in autoimmune T-cell mediated diseases as T1D.

3.
Eur Thyroid J ; 10(5): 425-433, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34540713

ABSTRACT

INTRODUCTION: The potential of the thyroid hormone receptor ß (TRß1) selective analog GC-1 has been widely proven in animal models and humans. However, its effect on the reproductive stage of the female rat has not been evaluated. METHODS: The effect of the administration of GC-1 or equimolar doses of triiodothyronine (T3) was evaluated on the reproductive performance of the hypothyroid female rat and the indirect effect on pup thyroid status, weight, and survival. RESULTS: Hypothyroidism reduced the number of embryos implanted in the uterus, whereas T3 and GC-1 treatment in hypothyroid females reestablished the number of implanted embryos to normal. Initiation of labor was delayed by hypothyroidism, and T3 replacement treatment reinstated the normal timing of parturition. The administration of GC-1 alone to the lactating mother did not affect pup survival, weight, or thyroidal status. CONCLUSIONS: Our findings show the differential effect of thyroid hormone selective signaling during gestation and the indirect exposure of the pups; we also emphasize the plausible use of GC-1 for treatment of hypothyroid mothers during the lactation period.

4.
Front Physiol ; 12: 662833, 2021.
Article in English | MEDLINE | ID: mdl-34335290

ABSTRACT

Despite popular usage of medicinal plants, their effects as cardiovascular protective agents have not been totally elucidated. We hypothesized that treatment with aqueous extract from Prosopis strombulifera (AEPs) and Tessaria absinthioides (AETa), Argentinian native plants, produces antioxidant effects on vascular smooth muscle cells (VSMCs) and attenuates atherogenesis on apolipoprotein E-knockout (ApoE-KO) mice. In VSMCs, both extracts (5-40 µg/ml) inhibited 10% fetal calf serum-induced cell proliferation, arrested cell in G2/M phase, reduced angiotensin II-induced reactive oxygen species (ROS) generation, and decreased NADPH oxidase subunit expression. In ApoE-KO mice, extracts significantly reduced triglycerides and lipid peroxidation [plasma thiobarbituric acid reactive substances (TBARS)], increased plasma total antioxidant status (TAS), and improved glutathione peroxidase activity in the liver. Under high-fat diet (HFD), both extracts were able to inhibit O2 - generation in the aortic tissue and caused a significant regression of atheroma plaques (21.4 ± 1.6% HFD group vs. 10.2 ± 1.2%∗ AEPs group and 14.3 ± 1.0%∗ AETa group; ∗ p < 0.01). Consumption of AEPs and AETa produces antioxidant/antimitogenic/anti-atherosclerotic effects, and their use may be beneficial as a complementary strategy regarding cardiovascular disease therapies.

5.
Heliyon ; 6(2): e03353, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32055742

ABSTRACT

The aqueous extract of the Argentinean native plant, Prosopis strombulifera (PsAE), presents cytotoxicity against human cancer cell lines by inducing cytostasis, necrosis and apoptosis; with diminution of clonogenic survival; without genotoxic effects nor oral animal toxicity. Until now, the chemical extract composition and its in vivo antitumoral properties remain unknown; these studies are the aim of the current work. The PsAE was characterized by chemical fingerprinting and the metabolome was identified by tandem UHPLC-PDA-HESI-Q-orbitrap® mass spectrometry. Colorectal tumors were induced by DMH administration and melanomas resulted from B16-F0 S.C. cells injection; then, animals were treated orally with PsEA. To correlate in vivo results with in vitro cytotoxicity, B16-F0 cell were cultured to determine: cell proliferation and viability by dye exclusion assays, MTT and CFSE dilution; cell cycle distribution by flow cytometry; and immunoblotting of p21cip1, PCNA, cleaved caspase 3, cleaved PARP and TUBA1A. Based on UHPLC-OT-MS and PDA analysis, twenty-six compounds were identified, including: 5 simple organic acids, 4 phenolic acids, 4 procyanidins, 11 flavonoids, and 2 oxylipins. On C57BL6 mice, PsAE significantly increases the median survival on colorectal cancer and reduces the final volume and weight of melanomas. Over cultured cells, the treatment induce over-expression of p21, cytostasis by G2/M cell cycle arrest and apoptosis; while, on in vivo melanomas, treatment up-regulates p21 and slightly decreases PCNA. In conclusion, PsAE is composed by phenolic compounds which demonstrate cytotoxic and antitumoral properties when is orally administrated. Presented results support future research of PsAE as a potential phytomedicine for cancer treatment.

6.
IET Nanobiotechnol ; 14(1): 86-93, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31935683

ABSTRACT

Drug encapsulation in nanocarriers such as polymeric nanoparticles (Nps) may help to overcome the limitations associated with cannabinoids. In this study, the authors' work aimed to highlight the use of electrospraying techniques for the development of carrier Nps of anandamide (AEA), an endocannabinoid with attractive pharmacological effects but underestimated due to its unfavourable physicochemical and pharmacokinetic properties added to its undesirable effects at the level of the central nervous system. The authors characterised physicochemically and evaluated in vitro biological activity of anandamide/ɛ-polycaprolactone nanoparticles (Nps-AEA/PCL) obtained by electrospraying in epithelial cells of the human proximal tubule (HK2), to prove the utility of this method and to validate the biological effect of Nps-AEA/PCL. They obtained particles from 100 to 900 nm of diameter with a predominance of 200-400 nm. Their zeta potential was -20 ± 1.86 mV. They demonstrated the stable encapsulation of AEA in Nps-AEA/PCL, as well as its dose-dependent capacity to induce the expression of iNOS and NO levels and to decrease the Na+/K+ ATPase activity in HK2 cells. Obtaining Nps-AEA/PCL by electrospraying would represent a promising methodology for a novel AEA pharmaceutical formulation development with optimal physicochemical properties, physical stability and biological activity on HK2 cells.


Subject(s)
Arachidonic Acids/chemistry , Endocannabinoids/chemistry , Nanoparticles/chemistry , Polyesters/chemistry , Polyunsaturated Alkamides/chemistry , Arachidonic Acids/pharmacology , Cell Line , Cell Survival/drug effects , Chemical Phenomena , Drug Stability , Electrochemical Techniques , Endocannabinoids/pharmacology , Humans , Nanoparticles/toxicity , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Polyunsaturated Alkamides/pharmacology , Sodium-Potassium-Exchanging ATPase/metabolism
7.
Antioxidants (Basel) ; 8(12)2019 Nov 28.
Article in English | MEDLINE | ID: mdl-31795145

ABSTRACT

The decoction of Tessaria absinthioides is used in traditional medicine of South America as hypocholesterolemic, balsamic, and expectorant; but it is also useful for the prevention of hepatitis, renal insufficiency, and diabetes, and is used as digestive. A lyophilized decoction from the aerial parts of this plant (TLD) collected in San Juan (TLDSJ) and Mendoza (TLDM) provinces (Argentina) and one collection from Antofagasta, Chile (TLDCH) were characterized regarding antioxidant and antibacterial activities, phenolics and flavonoids content, and ultrahigh resolution liquid chromatography Orbitrap MS analysis UHPLC-PDA-OT-MS/MS metabolite profiling. The antioxidant properties were carried out "in vitro" using 2,2-diphenyl-1-picrylhydrazyl (DPPH) and trolox equivalent antioxidant activity (TEAC) methods, ferric-reducing antioxidant power (FRAP), and lipoperoxidation in erythrocytes (LP). The antibacterial activity was evaluated following the Clinical and Laboratory Standards Institute (CLSI) rules. TLDSJ, TLDM, and TLDCH displayed a strong DPPH scavenging activity (EC50 = 42, 41.6, and 43 µg/mL, respectively) and inhibition of lipoperoxidation in erythrocytes (86-88% at 250 µg TLD/mL), while a less effect in the FRAP and TEACantioxidant assays was found. Additionally, the decoctions showed a content of phenolics compounds of 94 mg gallic acid equivalents (GAE)/g, 185 GAE/g, and 64 GAE/g, for TLDSJ, TLDM, and TLDCH samples, respectively. Regarding the flavonoid content, the Chilean sample was highlighted with 19 mg quercetin equivalents (QE)/g. In this work, several phenolic compounds, including sesquiterpenes, flavonoids, and phenolic acids, were rapidly identified in TLDSJ, TLDM, and TLDCH extracts by means UHPLC-PDA-OT-MS/MS for the first time, which gave a first scientific support to consider this medicinal decoction from both countries as a valuable source of metabolites with antioxidant effects, some with outstanding potential to improve human health.

8.
Molecules ; 24(6)2019 Mar 19.
Article in English | MEDLINE | ID: mdl-30893865

ABSTRACT

The decoction of the local plant Baccharis grisebachii is used as a digestive, gastroprotective, external cicatrizing agent and antiseptic in Argentine. A lyophilized decoction (BLD) from the aerial parts of this plant was evaluated regarding its anti-ulcer, antioxidant and cytotoxic activities and the bioactivities were supported by UHPLC-MS metabolome fingerprinting which revealed the presence of several small bioactive compounds. The antioxidant properties were evaluated by DPPH, TEAC, FRAP and lipoperoxidation inhibition in erythrocytes methods, and the antibacterial activity was evaluated according to the Clinical and Laboratory Standards Institute (CLSI) guidelines. The BLD showed a moderate free radical scavenging activity in the DPPH (EC50 = 106 µg/mL) and lipid peroxidation in erythrocytes assays (67%, at 250 µg/mL). However, the BLD had the highest gastroprotective effect at a dose of 750 mg/kg with a ninety-three percent inhibition of damage through a mechanism that involve NO and prostaglandins using the ethanol-induced gastric damage in a standard rat model. On the other hand, BLD does not induce cytotoxic changes on human tumor and no-tumor cell lines at the concentrations assayed. Regarding the metabolomic analysis, thirty-one compounds were detected and 30 identified based on UHPLC-OT-MS including twelve flavonoids, eleven cinnamic acid derivatives, one coumarin, one stilbene and two other different phenolic compounds. The results support that the medicinal decoction of Baccharis grisebachii is a valuable natural product with gastroprotective effects and with potential to improve human health that opens a pathway for the development of important phytomedicine products.


Subject(s)
Antioxidants/chemistry , Baccharis/chemistry , Biphenyl Compounds/chemistry , Chromatography, High Pressure Liquid , Cinnamates/chemistry , Flavonoids/chemistry , HCT116 Cells , Humans , Lipid Peroxidation , Mass Spectrometry , Picrates/chemistry
9.
Medicina (B Aires) ; 77(4): 283-290, 2017.
Article in English | MEDLINE | ID: mdl-28825571

ABSTRACT

Higher plants have provided various natural derived drugs used currently in western medicine. Tessaria absinthioides (Hook. & Arn.) DC, Asteraceae, is a native plant from South-America with reported ethnopharmacological and culinary uses. Despite recent scientific reports about plants properties, there is not a well conducted research about its anticancer and potential toxic effects. The current work demonstrates the plant aqueous extract composition; the in vitro induced cytotoxicity, and explores, in vivo, its oral toxicity and antitumoral effects. Composition of aqueous extract was determined by phytochemical reactions. Cytotoxicity was tested in tumoral (Hela, Gli-37, HCT-116 and MCF-7) and non-tumoral (HBL-100) cells, using MTT assay. Oral toxicity and the antitumor activity against colorectal carcinoma were studied in rodents. The chemical analysis revealed the presence of flavonoids, carbohydrates, sterols, terpenes and tannins. Cytotoxicity towards tumoral cells was observed (CV50: 3.0 to 14.8 υg/ml); while in non-tumoral cells, extracts evidenced a selective reduced toxicity (CV50: 29.5 υg/ml). Oral administration of the extract does not induce acute nor dose-repeated toxicity at doses up to 2000 mg/kg and 1000 mg/kg/day, respectively. The antitumoral effect was confirmed by a significant increase in a median survival from 24 weeks (non-treated) to 30 weeks (T. absinthioides treated). The present data indicate that T. absinthioides extract exhibits cytotoxicity against cancer cell lines, with no-toxic effects and significant antitumoral effects in colorectal cancer when is orally administrated. In conclusion, T. absinthioides possesses selective cytotoxicity and antitumoral activities, making its plant derivatives products promising for cancer research and treatment.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Asteraceae/chemistry , Colorectal Neoplasms/drug therapy , Plant Extracts/pharmacology , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Cell Line, Tumor , Colorectal Neoplasms/pathology , Disease Models, Animal , Fluorouracil , Mice , Mice, Inbred BALB C , Plant Extracts/therapeutic use , Rats , Rats, Sprague-Dawley , Tetrazolium Salts , Toxicity Tests
10.
Medicina (B.Aires) ; 77(4): 283-290, ago. 2017. ilus, graf, tab
Article in English | LILACS | ID: biblio-894480

ABSTRACT

Higher plants have provided various natural derived drugs used currently in western medicine. Tessaria absinthioides (Hook. & Arn.) DC, Asteraceae, is a native plant from South-America with reported ethnopharmacological and culinary uses. Despite recent scientific reports about plants properties, there is not a well conducted research about its anticancer and potential toxic effects. The current work demonstrates the plant aqueous extract composition; the in vitro induced cytotoxicity, and explores, in vivo, its oral toxicity and antitumoral effects. Composition of aqueous extract was determined by phytochemical reactions. Cytotoxicity was tested in tumoral (Hela, Gli-37, HCT-116 and MCF-7) and non-tumoral (HBL-100) cells, using MTT assay. Oral toxicity and the antitumor activity against colorectal carcinoma were studied in rodents. The chemical analysis revealed the presence of flavonoids, carbohydrates, sterols, terpenes and tannins. Cytotoxicity towards tumoral cells was observed (CV50: 3.0 to 14.8 μg/ml); while in non-tumoral cells, extracts evidenced a selective reduced toxicity (CV50: 29.5 μg/ml). Oral administration of the extract does not induce acute nor dose-repeated toxicity at doses up to 2000 mg/kg and 1000 mg/kg/day, respectively. The antitumoral effect was confirmed by a significant increase in a median survival from 24 weeks (non-treated) to 30 weeks (T. absinthioides treated). The present data indicate that T. absinthioides extract exhibits cytotoxicity against cancer cell lines, with no-toxic effects and significant antitumoral effects in colorectal cancer when is orally administrated. In conclusion, T. absinthioides possesses selective cytotoxicity and antitumoral activities, making its plant derivatives products promising for cancer research and treatment.


Las plantas superiores han provisto numerosos derivados naturales usados actualmente por la medicina occidental. Tessaria absinthioides (Hook & Arn) DC, Asteraceae, es una planta autóctona de Sudamérica con informes de uso etnofarmacológico y culinario. A pesar de los reportes científicos sobre las propiedades de esta planta, no existen estudios que caractericen sus efectos antitumorales ni sus efectos tóxicos. En el presente trabajo se describe la composición del extracto acuoso de T. absinthioides, sus propiedades citotóxicas in vitro, y explora in vivo la toxicidad oral y su capacidad de afectar la progresión de tumores. La composición se determinó mediante reacciones fitoquímicas. La citotoxicidad se estudió en líneas celulares tumorales (Gli-37, HeLa, HCT-116 y MCF-7) y no tumorales (HBL-100), utilizando el ensayo de MTT. La toxicidad oral de los extractos y su capacidad antitumoral sobre carcinoma colorrectal se analizaron en roedores. El análisis del extracto acuoso evidenció flavonoides, carbohidratos, esteroles, terpenos y taninos. La citotoxicidad sobre células tumorales resultó similar a la observada para el 5-fluoracilo (CV50: 3.0 a 14.8 μg/ml); mientras que, en células no tumorales, el efecto estuvo selectivamente reducido (CV50: 29.5 μg/ml). La administración oral del extracto no indujo toxicidad aguda ni a dosis repetidas (dosis hasta 2000 mg/kg y 1000 mg/kg/día, respectivamente). Los efectos antitumorales se confirmaron mediante un significativo aumento de la supervivencia en el grupo tratado con T. absinthioides. En conclusión, de acuerdo a los resultados obtenidos, T. absinthioides y sus derivados naturales representan un campo prometedor de estudio para la investigación en el tratamiento del cáncer.


Subject(s)
Animals , Rabbits , Rats , Plant Extracts/pharmacology , Colorectal Neoplasms/drug therapy , Asteraceae/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Tetrazolium Salts , Plant Extracts/therapeutic use , Colorectal Neoplasms/pathology , Rats, Sprague-Dawley , Toxicity Tests , Cell Line, Tumor , Disease Models, Animal , Fluorouracil , Mice, Inbred BALB C , Antineoplastic Agents, Phytogenic/therapeutic use
11.
Mol Oncol ; 10(7): 1099-117, 2016 08.
Article in English | MEDLINE | ID: mdl-27233943

ABSTRACT

The synthetic steroid mifepristone blocks the growth of ovarian cancer cells, yet the mechanism driving such effect is not entirely understood. Unbiased genomic and proteomic screenings using ovarian cancer cell lines of different genetic backgrounds and sensitivities to platinum led to the identification of two key genes upregulated by mifepristone and involved in the unfolded protein response (UPR): the master chaperone of the endoplasmic reticulum (ER), glucose regulated protein (GRP) of 78 kDa, and the CCAAT/enhancer binding protein homologous transcription factor (CHOP). GRP78 and CHOP were upregulated by mifepristone in ovarian cancer cells regardless of p53 status and platinum sensitivity. Further studies revealed that the three UPR-associated pathways, PERK, IRE1α, and ATF6, were activated by mifepristone. Also, the synthetic steroid acutely increased mRNA translation rate, which, if prevented, abrogated the splicing of XBP1 mRNA, a non-translatable readout of IRE1α activation. Moreover, mifepristone increased LC3-II levels due to increased autophagic flux. When the autophagic-lysosomal pathway was inhibited with chloroquine, mifepristone was lethal to the cells. Lastly, doses of proteasome inhibitors that are inadequate to block the activity of the proteasomes, caused cell death when combined with mifepristone; this phenotype was accompanied by accumulation of poly-ubiquitinated proteins denoting proteasome inhibition. The stimulation by mifepristone of ER stress and autophagic flux offers a therapeutic opportunity for utilizing this compound to sensitize ovarian cancer cells to proteasome or lysosome inhibitors.


Subject(s)
Autophagy/drug effects , Lysosomes/metabolism , Mifepristone/pharmacology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Proteasome Inhibitors/pharmacology , Protein Biosynthesis/drug effects , Unfolded Protein Response/drug effects , Activating Transcription Factor 4/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chloroquine/pharmacology , Cinnamates/pharmacology , Endoplasmic Reticulum Chaperone BiP , Eukaryotic Initiation Factor-2/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lysosomes/drug effects , Platinum/pharmacology , Protein Biosynthesis/genetics , Puromycin/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Thiourea/analogs & derivatives , Thiourea/pharmacology , Tunicamycin/pharmacology
12.
J Ovarian Res ; 7: 45, 2014.
Article in English | MEDLINE | ID: mdl-24795781

ABSTRACT

BACKGROUND: Antiprogestin compounds have been shown to be effective in blocking the growth of ovarian cancer cells of different genetic backgrounds. Herein we studied the anti-ovarian cancer effect of a series of antiprogestins sharing the chemical backbone of the most characterized antiprogestin, mifepristone, but with unique modifications in position C-17 of the steroid ring. We assessed the effect of mifepristone-like antiprogestins on the growth of ovarian cancer cells sensitive to the standard combination therapy cisplatin-paclitaxel or made double-resistant upon six cycles of pulse-selection with the drugs used at clinically relevant concentrations and exposure times. METHODS: IGROV-1 and SKOV-3 cells were pulsed with 20 µM cisplatin for 1 h followed by 100 nM paclitaxel for 3 h once a week for six weeks. The cells that did not die and repopulate the culture after the chemotherapies were termed Platinum-Taxane-EScape cells (PTES). Parental cells were compared against their PTES derivatives in their responses to further platinum-taxane treatments. Moreover, both ovarian cancer cells and their PTES siblings were exposed to escalating doses of the various antiprogestin derivatives. We assessed cell growth, viability and sub-G1 DNA content using microcapillary cytometry. Cyclin-dependent kinase inhibitors p21(cip1) and p27(kip1) and cleavage of downstream caspase-3 substrate PARP were used to assess whether cell fate, as a consequence of treatment, was limited to cytostasis or progressed to lethality. RESULTS: Cells subjected to six pulse-selection cycles of cisplatin-paclitaxel gave rise to sibling derivatives that displayed ~2-7 fold reduction in their sensitivities to further chemotherapy. However, regardless of the sensitivity the cells developed to the combination cisplatin-paclitaxel, they displayed similar sensitivity to the antiprogestins, which blocked their growth in a dose-related manner, with lower concentrations causing cytostasis, and higher concentrations causing lethality. CONCLUSIONS: Antiprogestins carrying a backbone similar to mifepristone are cytotoxic to ovarian cancer cells in a manner that does not depend on the sensitivity the cells have to the standard ovarian cancer chemotherapeutics, cisplatin and paclitaxel. Thus, antiprogestin therapy could be used to treat ovarian cancer cells showing resistance to both platinum and taxanes.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Mifepristone/pharmacology , Paclitaxel/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Drug Synergism , Estrenes/pharmacology , Female , Furans/pharmacology , Hormone Antagonists/pharmacology , Humans , Inhibitory Concentration 50 , Norpregnadienes/pharmacology
13.
Article in English | MEDLINE | ID: mdl-23420486

ABSTRACT

We have previously shown that the antiprogestin and antiglucocorticoid mifepristone inhibits the growth of ovarian cancer cells. In this work, we hypothesized that cellular stress caused by mifepristone is limited to cytostasis and that cell killing is avoided as a consequence of the persistent activity of the PI3K/Akt survival pathway.To investigate the role of this pathway in mifepristone-induced growth inhibition, human ovarian cancer cells of various histological subtypes and genetic backgrounds were exposed to cytostatic doses of mifepristone in the presence or absence of the PI3K inhibitor, LY294002. The activation of Akt in ovarian cancer cells, as marked by its phosphorylation on Ser473, was not modified by cytostatic concentrations of mifepristone, but it was blocked upon treatment with LY294002. The combination mifepristone/LY294002, but not the individual drugs, killed ovarian cancer cells via apoptosis, as attested by genomic DNA fragmentation and cleavage of caspase-3, and the concomitant down-regulation of anti-apoptotic proteins Bcl-2 and XIAP. From a pharmacological standpoint, when assessing cell growth inhibition using a median-dose analysis algorithm, the interaction between mifepristone and LY294002 was synergistic. The lethality caused by the combination mifepristone/LY294004 in two dimensional cell cultures was recapitulated in organized, tri-dimensional spheroids. This study demonstrates that mifepristone and LY294002, when used individually, cause cell growth arrest, yet when combined, they cause lethality.

14.
BMC Cancer ; 12: 200, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-22642877

ABSTRACT

BACKGROUND: Advanced ovarian cancer is treated with cytoreductive surgery and combination platinum- and taxane-based chemotherapy. Although most patients have acute clinical response to this strategy, the disease ultimately recurs. In this work we questioned whether the synthetic steroid mifepristone, which as monotherapy inhibits the growth of ovarian cancer cells, is capable of preventing repopulation of ovarian cancer cells if given after a round of lethal cisplatin-paclitaxel combination treatment. METHODS: We established an in vitro approach wherein ovarian cancer cells with various sensitivities to cisplatin or paclitaxel were exposed to a round of lethal doses of cisplatin for 1 h plus paclitaxel for 3 h. Thereafter, cells were maintained in media with or without mifepristone, and short- and long-term cytotoxicity was assessed. RESULTS: Four days after treatment the lethality of cisplatin-paclitaxel was evidenced by reduced number of cells, increased hypodiploid DNA content, morphological features of apoptosis, DNA fragmentation, and cleavage of caspase-3, and of its downstream substrate PARP. Short-term presence of mifepristone either enhanced or did not modify such acute lethality. Seven days after receiving cisplatin-paclitaxel, cultures showed signs of relapse with escaping colonies that repopulated the plate in a time-dependent manner. Conversely, cultures exposed to cisplatin-paclitaxel followed by mifepristone not only did not display signs of repopulation following initial chemotherapy, but they also had their clonogenic capacity drastically reduced when compared to cells repopulating after cisplatin-paclitaxel. CONCLUSIONS: Cytostatic concentrations of mifepristone after exposure to lethal doses of cisplatin and paclitaxel in combination blocks repopulation of remnant cells surviving and escaping the cytotoxic drugs.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hormone Antagonists/pharmacology , Mifepristone/pharmacology , Ovarian Neoplasms/drug therapy , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cisplatin/administration & dosage , DNA, Neoplasm/analysis , Female , G1 Phase/drug effects , Humans , Paclitaxel/administration & dosage , Tumor Cells, Cultured
15.
Reprod Biol Endocrinol ; 8: 14, 2010 Feb 11.
Article in English | MEDLINE | ID: mdl-20149258

ABSTRACT

BACKGROUND: Rats made hypothyroid with propilthyouracil start showing abnormal cycling on the second cycle after the start of the treatment, with a high proportion of spontaneous pseudopregnancies and reduced fertility. METHODS: To investigate some of the mechanisms involved in these reproductive abnormalities, hypothyroidism was induced in virgin rats by propilthyouracil (0.1 g/L in the drinking water) and we determined circulating hormones by radioimmunoassay and whole ovary expression of ovarian hormone receptors, growth factors and steroidogenic enzymes using semi-quantitative RT-PCR.The study was performed on days 6 to 9 of treatment, corresponding to diestrus I (at 20.00-22.00 h), diestrus II (at 20.00-22.00 h), proestrus and estrus (both at 8.00-10.00 h and 20.00-22.00 h) of the second estrous cycle after beginning propilthyouracil treatment. Another group of rats was mated on day 8 and the treatment continued through the entire pregnancy to evaluate reproductive performance. RESULTS: Hypothyroidism increased circulating prolactin and estradiol on estrus 5 to 7-fold and 1.2 to 1.4-fold respectively. Growth hormone and insulin-like growth factor 1 diminished 60 and 20% respectively on proestrus morning. Hypothyroidism doubled the ovarian mRNA contents of estrogen receptor-beta on proestrus and estrus evenings, cyp19A1 aromatase mRNA on estrus evening and of growth hormone receptor on proestrus evening. Hypothyroidism did not influence ovulation rate or the number of corpora lutea at term, but a diminished number of implantation sites and pups per litter were observed (Hypothyroid: 11.7 +/- 0.8 vs. CONTROL: 13.9 +/- 0.7). CONCLUSIONS: Short term hypothyroidism alters normal hormone profile in the cycling rat increasing the expression of estrogen receptor-beta and cyp19A1 aromatase on estrus, which in turn may stimulate estradiol and prolactin secretion, favouring corpus luteum survival and the subsequent instauration of pseudopregnancy.


Subject(s)
Estrous Cycle/physiology , Hypothyroidism/physiopathology , Ovary/physiology , Animals , Embryo Implantation/drug effects , Embryo Implantation/physiology , Estrous Cycle/blood , Female , Gonadal Steroid Hormones/blood , Growth Hormone/blood , Hypothyroidism/blood , Hypothyroidism/chemically induced , Insulin-Like Growth Factor I/analysis , Male , Ovary/drug effects , Ovulation/drug effects , Ovulation/physiology , Pregnancy , Prolactin/blood , Propylthiouracil , Rats , Rats, Wistar , Thyroid Hormones/blood , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...