837 results on '"Cytostasis"'
Search Results
2. Vitality, viability, long-term clonogenic survival, cytotoxicity, cytostasis and lethality: what do they mean when testing new investigational oncology drugs?
- Author
-
Benjamin N. Forgie, Rewati Prakash, Alicia A. Goyeneche, and Carlos M. Telleria
- Subjects
Viability ,Vitality ,Cytotoxicity ,Cytostasis ,Lethality ,Clonogenicity ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract In the field of experimental therapeutics for oncology purposes researchers are continuously evaluating the toxicity of novel treatment approaches against cancer cells. Within this topic of research, it is highly critical to define parameters of toxicity that denote when cancer cells are perturbed in their functionality by a new investigational drug. As the goal for these approaches is to achieve cellular demise, then what approaches to use and what do they mean in terms of assessing such cell death is of critical importance. In this comment article we highlight the definition of vitality and differentiate it from viability, and further define clonogenic survival in a chronic fashion. Additionally, we highly recommend the use of the term cytotoxicity as a general descriptor indicating toxicity towards a cell, but within that we encourage to sub-classify it as either cytostasis (i.e., when a treatment does not allow a cell to grow but it does not kill it either), or lethality (when a cell dies in response to the treatment). A more precise use of these terms should help advance the field of experimental therapeutics in oncology towards better defining the mechanisms of action of novel investigational drugs.
- Published
- 2024
- Full Text
- View/download PDF
3. Nitric Oxide Prevents Glioblastoma Stem Cells' Expansion and Induces Temozolomide Sensitization.
- Author
-
Salvatori, Luisa, Malatesta, Silvia, Illi, Barbara, Somma, Maria Patrizia, Fionda, Cinzia, Stabile, Helena, Fontanella, Rosaria Anna, and Gaetano, Carlo
- Subjects
- *
STEM cells , *TEMOZOLOMIDE , *NITRIC oxide , *GLIOBLASTOMA multiforme , *DNA damage , *STUNTED growth , *WESTERN immunoblotting , *DNA adducts - Abstract
Glioblastoma multiforme (GBM) has high mortality and recurrence rates. Malignancy resilience is ascribed to Glioblastoma Stem Cells (GSCs), which are resistant to Temozolomide (TMZ), the gold standard for GBM post-surgical treatment. However, Nitric Oxide (NO) has demonstrated anti-cancer efficacy in GBM cells, but its potential impact on GSCs remains unexplored. Accordingly, we investigated the effects of NO, both alone and in combination with TMZ, on patient-derived GSCs. Experimentally selected concentrations of diethylenetriamine/NO adduct and TMZ were used through a time course up to 21 days of treatment, to evaluate GSC proliferation and death, functional recovery, and apoptosis. Immunofluorescence and Western blot analyses revealed treatment-induced effects in cell cycle and DNA damage occurrence and repair. Our results showed that NO impairs self-renewal, disrupts cell-cycle progression, and expands the quiescent cells' population. Consistently, NO triggered a significant but tolerated level of DNA damage, but not apoptosis. Interestingly, NO/TMZ cotreatment further inhibited cell cycle progression, augmented G0 cells, induced cell death, but also enhanced DNA damage repair activity. These findings suggest that, although NO administration does not eliminate GSCs, it stunts their proliferation, and makes cells susceptible to TMZ. The resulting cytostatic effect may potentially allow long-term control over the GSCs' subpopulation. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
4. Half-Sandwich Type Platinum-Group Metal Complexes of C -Glucosaminyl Azines: Synthesis and Antineoplastic and Antimicrobial Activities.
- Author
-
Kacsir, István, Sipos, Adrienn, Major, Evelin, Bajusz, Nikolett, Bényei, Attila, Buglyó, Péter, Somsák, László, Kardos, Gábor, Bai, Péter, and Bokor, Éva
- Subjects
- *
AZINES , *METAL complexes , *PLATINUM compounds , *ANTI-infective agents , *QUINOLINE derivatives , *PYRIDAZINES , *PYRAZINES - Abstract
While platinum-based compounds such as cisplatin form the backbone of chemotherapy, the use of these compounds is limited by resistance and toxicity, driving the development of novel complexes with cytostatic properties. In this study, we synthesized a set of half-sandwich complexes of platinum-group metal ions (Ru(II), Os(II), Ir(III) and Rh(III)) with an N,N-bidentate ligand comprising a C-glucosaminyl group and a heterocycle, such as pyridine, pyridazine, pyrimidine, pyrazine or quinoline. The sugar-containing ligands themselves are unknown compounds and were obtained by nucleophilic additions of lithiated heterocycles to O-perbenzylated 2-nitro-glucal. Reduction of the adducts and, where necessary, subsequent protecting group manipulations furnished the above C-glucosaminyl heterocycles in their O-perbenzylated, O-perbenzoylated and O-unprotected forms. The derived complexes were tested on A2780 ovarian cancer cells. Pyridine, pyrazine and pyridazine-containing complexes proved to be cytostatic and cytotoxic on A2780 cells, while pyrimidine and quinoline derivatives were inactive. The best complexes contained pyridine as the heterocycle. The metal ion with polyhapto arene/arenyl moiety also impacted on the biological activity of the complexes. Ruthenium complexes with p-cymene and iridium complexes with Cp* had the best performance in ovarian cancer cells, followed by osmium complexes with p-cymene and rhodium complexes with Cp*. Finally, the chemical nature of the protective groups on the hydroxyl groups of the carbohydrate moiety were also key determinants of bioactivity; in particular, O-benzyl groups were superior to O-benzoyl groups. The IC50 values of the complexes were in the low micromolar range, and, importantly, the complexes were less active against primary, untransformed human dermal fibroblasts; however, the anticipated therapeutic window is narrow. The bioactive complexes exerted cytostasis on a set of carcinomas such as cell models of glioblastoma, as well as breast and pancreatic cancers. Furthermore, the same complexes exhibited bacteriostatic properties against multiresistant Gram-positive Staphylococcus aureus and Enterococcus clinical isolates in the low micromolar range. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
5. Cripowellins Pause Plasmodium falciparum Intraerythrocytic Development at the Ring Stage.
- Author
-
Butler, Joshua H., Painter, Heather J., Bremers, Emily K., Krai, Priscilla, Llinás, Manuel, and Cassera, Maria B.
- Subjects
- *
PLASMODIUM falciparum , *LIFE cycles (Biology) , *NATURAL products , *MOLECULAR probes , *CELL cycle - Abstract
Cripowellins from Crinum erubescens are known pesticidal and have potent antiplasmodial activity. To gain mechanistic insights to this class of natural products, studies to determine the timing of action of cripowellins within the asexual intraerythrocytic cycle of Plasmodium falciparum were performed and led to the observation that this class of natural products induced reversible cytostasis in the ring stage within the first 24 h of treatment. The transcriptional program necessary for P. falciparum to progress through the asexual intraerythrocytic life cycle is well characterized. Whole transcriptome abundance analysis showed that cripowellin B "pauses" the transcriptional program necessary to progress through the intraerythrocytic life cycle coinciding with the lack of morphological progression of drug treated parasites. In addition, cripowellin B-treated parasites re-enter transcriptional progression after treatment was removed. This study highlights the use of cripowellins as chemical probes to reveal new aspects of cell cycle progression of the asexual ring stage of P. falciparum which could be leveraged for the generation of future antimalarial therapeutics. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
6. In vitro and in silico assessment of cytotoxicity and chromosome instability induced by saxitoxin in human derived neural cell line
- Author
-
JULIANY S. CONSTANTE, JULIANA E. AL KHATEEB, ANA PAULA DE SOUZA, FELIPE U. CONTER, MAURÍCIO LEHMANN, JOÃO S. YUNES, and RAFAEL R. DIHL
- Subjects
STX, micronucleus ,cytostasis ,apoptosis ,systems biology ,Science - Abstract
Abstract In freshwater, saxitoxins (STX) are produced by different cyanobacteria genera, including Raphidiopsis. Data regarding cytogenotoxicity effects of STX on human cells are scarse, this merit further studies of its toxicology. This study assessed the cytotoxicity and the chromosome instability of STX on SHSY-5Y human cell line. The CBMN assay allows the detection of chromosome breaks and abnormal chromosomal segregation. Additionally, in silico systems biology approach, used to search for known and predicted interaction networks, was applied to study the interactions between STX and SHSY-5Y cellular components. The results of the CBMN assay demonstrated that STX concentrations of 2.5 – 10 µg/L induced cytostasis and chromosome instability in a dose-response relationship. Apoptosis was detected after exposure of SHSY-5Y cultured cells to STX concentration of 10 µg/L. The results of the systems biology analysis revealed the interaction of STX with proteins related with acetylcoline pathway, cell cycle regulation and apoptosis. Furthermore, combining the in vitro and in silico approachs, it was possible to suggest a mechanism of action of STX in SHSY-5Y cells. Overall, the data demonstrated the cytotoxicity and mutagenicity of environmentally relevant concentrations of STX. These results should be considered when setting up guidelines for monitoring STX in water supply.
- Published
- 2022
- Full Text
- View/download PDF
7. Cytostatic Effects of Polyethyleneimine Surfaces on the Mesenchymal Stromal Cell Cycle.
- Author
-
Alba, Anna, Villaggio, Giusy, Messina, Grazia Maria Lucia, Caruso, Massimo, Federico, Concetta, Cambria, Maria Teresa, Marletta, Giovanni, and Sinatra, Fulvia
- Subjects
- *
STROMAL cells , *CELL cycle , *BIOLOGICAL interfaces , *POLYETHYLENEIMINE , *POLYELECTROLYTES , *POLYSTYRENE , *CELL adhesion - Abstract
Polyelectrolytes assembled layer-by-layer (PEMs) are commonly used as functional coatings to build-up biological interfaces, particularly suitable as compatible layers for the interaction with a biological medium, providing suitable conditions to promote or prevent cell seeding while maintaining the phenotype. The proper assessment of the biocompatibility of PEMs and the elucidation of the related mechanisms are therefore of paramount importance. In this study, we report in detail the effect of two different PEM endings, polystyrene sulfonate (PSS) and polyethylenimine (PEI), respectively, on the cell adhesion, growth, and viability of human bone mesenchymal stromal cells (MSCs). The results have shown that PSS-ended substrates appear to be the most suitable to drive the cell adhesion and phenotype maintenance of MSCs, showing good biocompatibility. On the contrary, while the cells seem to adhere more quickly and strongly on the PEI-ended surfaces, the interaction with PEI significantly affects the growth and viability, reducing the cell spreading capability, by sequestering the adhesion molecules already in the very early steps of cell–substrate contact. These results point to the promotion of a cytostatic effect of PEI, rather than the often-claimed cytotoxicity. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
8. Flame retardant tris(1,3-dichloro-2-propyl)phosphate (TDCPP) toxicity is attenuated by N-acetylcysteine in human kidney cells
- Author
-
Killilea, David W, Chow, Darryl, Xiao, Sheng Qi, Li, Charles, and Stoller, Marshall L
- Subjects
Environmental Sciences ,Pollution and Contamination ,Clinical Research ,flame retardant ,cytostasis ,cell toxicity ,antioxidant ,cell cycle ,ATSDR ,Agency for Toxic Substances and Disease Registry ,DMEM ,Dulbecco’s Modified Eagle Medium ,DMSO ,dimethyl sulfoxide ,EDTA ,ethylenediamine tetraacetic acid ,FBS ,fetal bovine serum ,N-acetylcysteine ,NAC ,N-acetylcysteine ,SFFCPF ,San Francisco Firefighters Cancer Prevention Foundation ,TDCPP ,tris(1 ,3-dichloro-2-propyl)phosphate ,TR ,thyroid hormone receptor ,Tris(1 ,3-dichloro-2-propyl)phosphate ,Tris ,tris(2 ,3-dibromopropyl)phosphate ,Medicinal and Biomolecular Chemistry ,Other Chemical Sciences ,Clinical Sciences ,Medical biotechnology ,Pharmacology and pharmaceutical sciences ,Ecological applications - Abstract
Prolonged exposure to the flame retardants found in many household products and building materials is associated with adverse developmental, reproductive, and carcinogenic consequences. While these compounds have been studied in numerous epidemiological and animal models, less is known about the effects of flame retardant exposure on cell function. This study evaluated the toxicity of the commonly used fire retardant tris(1,3-dichloro-2-propyl)phosphate (TDCPP) in cell line derived from the kidney, a major tissue target of organohalogen toxicity. TDCPP inhibited cell growth at lower concentrations (IC50 27 μM), while cell viability and toxicity were affected at higher concentrations (IC50 171 μM and 168 μM, respectively). TDCPP inhibited protein synthesis and caused cell cycle arrest, but only at higher concentrations. Additionally, the antioxidant N-acetylcysteine (NAC) reduced cell toxicity in cells treated with TDCPP, suggesting that exposure to TDCPP increased oxidative stress in the cells. In summary, these data show that low concentrations of TDCPP result in cytostasis in a kidney cell line, whereas higher concentrations induce cell toxicity. Furthermore, TDCPP toxicity can be attenuated by NAC, suggesting that antioxidants may be effective countermeasures to some organohalogen exposures.
- Published
- 2017
9. Cripowellins Pause Plasmodium falciparum Intraerythrocytic Development at the Ring Stage
- Author
-
Joshua H. Butler, Heather J. Painter, Emily K. Bremers, Priscilla Krai, Manuel Llinás, and Maria B. Cassera
- Subjects
Plasmodium ,Crinum erubescens ,cripowellin ,ring stage ,cytostasis ,transcription ,Organic chemistry ,QD241-441 - Abstract
Cripowellins from Crinum erubescens are known pesticidal and have potent antiplasmodial activity. To gain mechanistic insights to this class of natural products, studies to determine the timing of action of cripowellins within the asexual intraerythrocytic cycle of Plasmodium falciparum were performed and led to the observation that this class of natural products induced reversible cytostasis in the ring stage within the first 24 h of treatment. The transcriptional program necessary for P. falciparum to progress through the asexual intraerythrocytic life cycle is well characterized. Whole transcriptome abundance analysis showed that cripowellin B “pauses” the transcriptional program necessary to progress through the intraerythrocytic life cycle coinciding with the lack of morphological progression of drug treated parasites. In addition, cripowellin B-treated parasites re-enter transcriptional progression after treatment was removed. This study highlights the use of cripowellins as chemical probes to reveal new aspects of cell cycle progression of the asexual ring stage of P. falciparum which could be leveraged for the generation of future antimalarial therapeutics.
- Published
- 2023
- Full Text
- View/download PDF
10. EMP2 induces cytostasis and apoptosis via the TGFβ/SMAD/SP1 axis and recruitment of P2RX7 in urinary bladder urothelial carcinoma.
- Author
-
Li, Chien-Feng, Chan, Ti-Chun, Pan, Cheng-Tang, Vejvisithsakul, Pichpisith Pierre, Lai, Jia-Chen, Chen, Szu-Yu, Hsu, Ya-Wen, Shiao, Meng-Shin, and Shiue, Yow-Ling
- Subjects
- *
BLADDER , *TRANSITIONAL cell carcinoma , *CELLULAR signal transduction , *DNA replication , *MEMBRANE proteins - Abstract
Purpose: Urinary bladder urothelial carcinoma (UBUC) is a common malignant disease, and its high recurrence rates impose a heavy clinical burden. The objective of this study was to identify signaling pathways downstream of epithelial membrane protein 2 (EMP2), which induces cytostasis and apoptosis in UBUC. Methods: A series of in vitro and in vivo assays using different UBUC-derived cell lines and mouse xenograft models were performed, respectively. In addition, primary UBUC specimens were evaluated by immunohistochemistry. Results: Exogenous expression of EMP2 in J82 UBUC cells significantly decreased DNA replication and altered the expression levels of several TGFβ signaling-related proteins. EMP2 knockdown in BFTC905 UBUC cells resulted in opposite effects. EMP2-dysregulated cell cycle progression was found to be mediated by the TGFβ/TGFBR1/SP1 family member SMAD. EMP2 or purinergic receptor P2X7 (P2RX7) gene expression upregulation induced apoptosis via both intrinsic and extrinsic pathways. In 242 UBUC patient samples, P2RX7 protein levels were found to be significantly and positively correlated with EMP2 protein levels. Low P2RX7 levels conferred poor disease-specific and metastasis-free survival rates, and significantly decreased apoptotic cell rates. EMP2 was found to physically interact with P2RX7. In the presence of a P2RX7 agonist, BzATP, overexpression of both EMP2 and P2RX7 significantly increased apoptotic cell rates compared to overexpression of EMP2 or P2RX7 alone. Conclusions: EMP2 induces cytostasis via the TGFβ/SMAD/SP1 axis and recruits P2RX7 to enhance apoptosis in UBUC. Our data provide new insights that may be employed for the design of UBUC targeting therapies. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
11. Response of neuroblastoma cells to RF currents as a function of the signal frequency
- Author
-
María Luisa Hernández-Bule, Enrique Medel, Clara Colastra, Raquel Roldán, and Alejandro Úbeda
- Subjects
Electric currents ,NB69 ,Capacitive-resistive electric transfer ,Electrothermal therapy ,Subthermal ,Cytostasis ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Capacitive-resistive electric transfer (CRET) is a non-invasive therapeutic strategy that applies radiofrequency electric currents within the 400–600 kHz range to tissue repair and regeneration. Previous studies by our group have shown that 48 h of intermittent exposure to a 570 kHz CRET signal at a subthermal density of 50 μA/mm2 causes significant changes in the expression and activation of cell cycle control proteins, leading to cycle arrest in human cancer cell cultures. The present study investigates the relevance of the signal frequency in the response of the human neuroblastoma cell line NB69 to subthermal electric treatment with four different signal frequency currents within the 350–650 kHz range. Methods Trypan blue assay, flow cytometry, immunofluorescence and immunoblot were used to study the effects of subthermal CRET currents on cell viability, cell cycle progression and the expression of several marker proteins involved in NB69 cell death and proliferation. Results The results reveal that among the frequencies tested, only a 448 kHz signal elicited both proapoptotic and antiproliferative, statistically significant responses. The apoptotic effect would be due, at least in part, to significant changes induced by the 448 kHz signal in the expression of p53, Bax and caspase-3. The cytostatic response was preceded by alterations in the kinetics of the cell cycle and in the expression of proteins p-ERK1/2, cyclin D1 and p27, which is consistent with a potential involvement of the EGF receptor in electrically induced changes in the ERK1/2 pathway. This receives additional support from results indicating that the proapototic and antiproliferative responses to CRET can be transiently blocked when the electric stimulus is applied in the presence of PD98059, a chemical inhibitor of the ERK1/2 pathway. Conclusion The understanding of the mechanisms underlying the ability of slowing down cancer cell growth through electrically-induced changes in the expression of proteins involved in the control of cell proliferation and apoptosis might afford new insights in the field of oncology.
- Published
- 2019
- Full Text
- View/download PDF
12. Mutagenic and cytostatic activities of the Xylopia laevigata essential oil in human lymphocytes.
- Author
-
Pereira, Tais Susane, Machado Esquissato, Giovana Natiele, Costa, Emmanoel Vilaça, Nogueira, Paulo Cesar de Lima, and Castro-Prado, Marialba Avezum Alves de
- Subjects
ESSENTIAL oils ,MUTAGENS ,LYMPHOCYTES ,NUCLEOLUS - Abstract
Hydro-distilled essential oil from leaves of Xylopia laevigata was characterized by GC-MS. Twenty-seven components were identified and the oil's major constituents comprised germacrene D, bicyclogermacrene, (E)-caryophyllene and germacrene B. The cytotoxicity of the essential oil of X. laevigata (EOXL), determined by MTT and mitotic index methods in cultured human lymphocytes was observed in all tested concentrations. Cultures treated with EOXL demonstrated significant increase in the frequencies of micronuclei in the cytokinesis-block micronucleus assay (CBMN) and reduction of the cytokinesis-block proliferation index (CBPI) rates. Results demonstrated the cytostatic and mutagenic effects of EOXL, the latter for the first time. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
13. Nitric Oxide Prevents Glioblastoma Stem Cells’ Expansion and Induces Temozolomide Sensitization
- Author
-
Gaetano, Luisa Salvatori, Silvia Malatesta, Barbara Illi, Maria Patrizia Somma, Cinzia Fionda, Helena Stabile, Rosaria Anna Fontanella, and Carlo
- Subjects
glioblastoma stem cells (GSCs) ,glioblastoma multiforme (GBM) ,nitric oxide (NO) ,Temozolomide (TMZ) ,cytostasis ,adjuvant treatment - Abstract
Glioblastoma multiforme (GBM) has high mortality and recurrence rates. Malignancy resilience is ascribed to Glioblastoma Stem Cells (GSCs), which are resistant to Temozolomide (TMZ), the gold standard for GBM post-surgical treatment. However, Nitric Oxide (NO) has demonstrated anti-cancer efficacy in GBM cells, but its potential impact on GSCs remains unexplored. Accordingly, we investigated the effects of NO, both alone and in combination with TMZ, on patient-derived GSCs. Experimentally selected concentrations of diethylenetriamine/NO adduct and TMZ were used through a time course up to 21 days of treatment, to evaluate GSC proliferation and death, functional recovery, and apoptosis. Immunofluorescence and Western blot analyses revealed treatment-induced effects in cell cycle and DNA damage occurrence and repair. Our results showed that NO impairs self-renewal, disrupts cell-cycle progression, and expands the quiescent cells’ population. Consistently, NO triggered a significant but tolerated level of DNA damage, but not apoptosis. Interestingly, NO/TMZ cotreatment further inhibited cell cycle progression, augmented G0 cells, induced cell death, but also enhanced DNA damage repair activity. These findings suggest that, although NO administration does not eliminate GSCs, it stunts their proliferation, and makes cells susceptible to TMZ. The resulting cytostatic effect may potentially allow long-term control over the GSCs’ subpopulation.
- Published
- 2023
- Full Text
- View/download PDF
14. TGF-β as Tumor Suppressor: In Vitro Mechanistic Aspects of Growth Inhibition
- Author
-
Bartholin, Laurent, Vincent, David F., Valcourt, Ulrich, Moustakas, Aristidis, editor, and Miyazawa, Keiji, editor
- Published
- 2013
- Full Text
- View/download PDF
15. Absence of herb-drug interactions of mistletoe with the tamoxifen metabolite (E/Z)-endoxifen and cytochrome P450 3A4/5 and 2D6 in vitro.
- Author
-
Weissenstein, U., Kunz, M., Oufir, M., Wang, J. T., Hamburger, M., Urech, K., Regueiro, U., and Baumgartner, S.
- Subjects
CELL proliferation ,APOPTOSIS ,BREAST tumors ,CELL cycle ,CELL lines ,ESTRADIOL ,ESTROGEN receptors ,DRUG-herb interactions ,TAMOXIFEN ,VISCUM ,PLANT extracts ,IN vitro studies ,CYTOCHROME P-450 - Abstract
Background: Women diagnosed with breast cancer frequently seek complementary and alternative (CAM) treatment options that can help to cope with their disease and the side effects of conventional cancer therapy. Especially in Europe, breast cancer patients use herbal products containing mistletoe (Viscum album L.). The oldest and one of the most prescribed conventional drugs for the treatment of estrogen receptor positive breast cancer is tamoxifen. Aside from positive clinical experience with the combination of tamoxifen and mistletoe, little is known about possible herb-drug interactions (HDIs) between the two products. In the present in vitro study, we investigated the effect of standardized commercial mistletoe preparations on the activity of endoxifen, the major active metabolite of tamoxifen. Methods: The estrogen receptor positive human breast carcinoma cell line MCF-7 was treated with (E/Z)-endoxifen hydrochloride in the presence and absence of a defined estradiol concentration. Each concentration of the drug was combined with fermented Viscum album L. extracts (VAE) at clinically relevant doses, and proliferation, apoptosis and cell cycle were analyzed. In parallel, possible inhibition of CYP3A4/5 and CYP2D6 was investigated using 50-donor mixed gender pooled human liver microsomes (HLMs). Results: VAE did not inhibit endoxifen induced cytostasis and cytotoxicity. At higher concentrations, VAE showed an additive inhibitory effect. VAE preparations did not cause inhibition of CYP3A4/5 and CYP2D6 catalyzed tamoxifen metabolism. Conclusions: The in vitro results suggest that mistletoe preparations can be used in combination with tamoxifen without the risk of HDIs. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
16. Zoledronic acid induces micronuclei formation, mitochondrial-mediated apoptosis and cytostasis in kidney cells.
- Author
-
Singireesu, Soma Shiva Nageswara Rao, Misra, Sunil, Mondal, Sujan Kumar, and Yerramsetty, Suresh
- Subjects
- *
ZOLEDRONIC acid , *NEPHROTOXICOLOGY , *TOXICITY testing , *EPITHELIAL cells , *MITOCHONDRIA , *THERAPEUTICS - Abstract
Aims Zoledronic acid (ZA), a FDA approved drug has used widely in the treatment of bone metastasis complications, has been linked to renal toxicity with unclear mechanism. The present study is aimed at investigating the genotoxic and cytotoxic effects of ZA in renal epithelial cells. Main methods The genotoxic effect of ZA in Vero and MDCK cells determined by cytokinesis block micronucleus (CBMN) assay. The cytotoxic effect assessed by analysing cell cycle profile, cell death and mitochondrial membrane potential by flow cytometry using propidium iodide, AnnexinV-FITC/PI and JC1 dye staining, respectively, BAX and Bcl-2 expression by Western blotting and caspase activity by spectrofluorimetry. Key findings The cytotoxic effect of ZA based on MTT assay revealed variable sensitivities of Vero and MDCK cells, with IC 50 values of 7.41 and 109.58 μM, respectively. The CBMN assay has shown prominent dose-dependent (IC 10-50 ) induction of micronuclei formation in both cells, indicating ZA's clastogenic and aneugenic potential. Further, the ZA treatment led the cells to apoptosis, evident from dose-dependent increase in the percentage of cells in subG1 phase and display of membranous phosphatidylserine translocation. Studies also confirmed apoptosis through mitochondria, evident from the prominent increase in BAX/Bcl-2 ratio, mitochondrial membrane depolarization and caspase-3/7 activity. In addition, ZA reduces cytokinetic activity of renal cells, evident from dose-wise lowered replicative indices. Significance The study depict ZA's potential genotoxic effect along with cytotoxic effect in renal epithelial cells, could be key factors for the development of renal complications associated with it, which prompts renal safety measures in lieu with ZA usage. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
17. Costunolide induces micronuclei formation, chromosomal aberrations, cytostasis, and mitochondrial-mediated apoptosis in Chinese hamster ovary cells.
- Author
-
Singireesu, Soma Shiva Nageswara Rao, Misra, Sunil, Mondal, Sujan Kumar, Yerramsetty, Suresh, Sahu, Nivedita, and K, Suresh Babu
- Abstract
Costunolide (CE) is a sesquiterpene lactone well-known for its antihepatotoxic, antiulcer, and anticancer activities. The present study focused on the evaluation of the cytogenetic toxicity and cellular death-inducing potential of CE in CHO cells, an epithelial cell line derived from normal ovary cells of Chinese hamster. The cytotoxic effect denoting MTT assay has shown an IC
50 value of 7.56 μM CE, where 50% proliferation inhibition occurs. The oxidative stress caused by CE was confirmed based on GSH depletion induced cell death, conspicuously absent in N-acetylcysteine (GSH precursor) pretreated cells. The evaluation of genotoxic effects of CE using cytokinesis block micronucleus assay and chromosomal aberration test has shown prominent induction of binucleated micronucleated cells and aberrant metaphases bearing chromatid and chromosomal breaks, indicating CE’s clastogenic and aneugenic potential. The apoptotic death in CE treated cells was confirmed by an increase in the number of cells in subG1 phase, exhibiting chromatin condensation and membranous phosphatidylserine translocation. The apoptosis induction follows mitochondrial mediation, evident from an increase in the BAX/Bcl-2 ratio, caspase-3/7 activity, and mitochondrial membrane permeability. CE also induces cytostasis in addition to apoptosis, substantiated by the reduced cytokinetic (replicative indices) and mitotic (mitotic indices and histone H3 Ser-10 phosphorylation) activities. Overall, the cellular GSH depletion and potential genotoxic effects by CE led the CHO cells to commit apoptosis and lowered cell division. The observed sensitivity of CHO cells doubts unintended adverse effects of CE on normal healthy cells, suggesting higher essentiality of further studies in order to establish its safety efficacy in therapeutic explorations. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
18. Diterpenoid natural compound C4 (Crassin) exerts cytostatic effects on triple-negative breast cancer cells via a pathway involving reactive oxygen species.
- Author
-
Richards, Cathy E., Vellanki, Sri H., Smith, Yvonne E., and Hopkins, Ann M.
- Subjects
- *
DITERPENES , *ANTINEOPLASTIC agents , *BREAST cancer , *ESTROGEN receptors , *REACTIVE oxygen species - Abstract
Purpose: Triple-negative breast cancers (TNBC) lack expression of three common cell surface receptors, i.e., estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor-2 (HER2). Accordingly, TNBCs are associated with fewer treatment options and a relatively poor prognosis. Having screened a National Cancer Institute natural compound library, the purpose of this study was to investigate the bioactivity of compound C4 (Crassin) in TNBC cells. Methods: Cell viability assays were performed in two TNBC cell lines, MDA-MB-231 and 4T1, following C4 treatment in the presence or absence of the antioxidant N-acetyl-L-cysteine (NAC). Phosphorylation of Akt and ERK was assessed by Western blotting. Apoptosis, necrosis, autophagy, necroptosis, ferroptosis and cytostasis assays were performed to explain viability deficits resulting from C4 exposure. Results: We found that the viability of the TNBC cells tested decreased in a concentration- and time-dependent fashion following C4 treatment. This decrease coincided with an unexpected increase in the expression of the cell survival effectors pAkt and pERK. In addition, we found that both the decreased cell viability and the increased pAkt/pERK levels could be rescued by the antioxidant NAC, suggesting a central role for reactive oxygen species (ROS) in the mechanism of action of C4. Necrosis, apoptosis, necroptosis and ferroptosis could be ruled out as cell death mechanisms. Instead, we found that C4 induced cytostasis downstream of ROS activation. Finally, we observed a synergistic effect between C4 and the chemotherapeutic drug doxorubicin in TNBC cells. Conclusions: From our in vitro data we conclude that C4 exerts cytostatic effects on triple-negative breast cancer cells via a pathway involving reactive oxygen species. Its potential value in combination with cytotoxic therapies merits deeper investigation in pre-clinical models. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
19. Systematic Analysis of Cytostatic TGF-Beta Response in Mesenchymal-Like Hepatocellular Carcinoma Cell Lines
- Author
-
Serif Senturk, Medine Zeynep Gungor, Merve Uysal, Mehmet Ozturk, Öztürk, Mehmet, and Şentürk, Şerif
- Subjects
Mesenchymal ,Hepatocellular carcinoma ,business.industry ,Mesenchymal stem cell ,Gastroenterology ,Cytostatic response ,SMAD ,Cytostasis ,Transduction (genetics) ,Oncology ,HCC cell lines ,Cell culture ,TGF beta signaling pathway ,TGF-β pathway ,Cancer research ,Medicine ,business ,Intracellular ,Transforming growth factor - Abstract
Background Hepatocellular carcinoma (HCC) is one of the most challenging malignancies, with high morbidity and mortality rates. The transforming growth factor-β (TGF-β) pathway plays a dual role in HCC, acting as both tumor suppressor and promoter. A thorough understanding of the mechanisms underlying its opposing functions is important. The growth suppressive effects of TGF-β remain largely unknown for mesenchymal HCC cells. Using a systematic approach, here we assess the cytostatic TGF-β responses and intracellular transduction of the canonical TGF-β/Smad signaling cascade in mesenchymal-like HCC cell lines. Methods Nine mesenchymal-like HCC cell lines, including SNU182, SNU387, SNU398, SNU423, SNU449, SNU475, Mahlavu, Focus, and Sk-Hep1, were used in this study. The cytostatic effects of TGF-β were evaluated by cell cycle analysis, BrdU labeling, and SA-β-Gal assay. RT-PCR and western blot analysis were utilized to determine the mRNA and protein expression levels of TGF-β signaling components and cytostatic genes. Immunoperoxidase staining and luciferase reporter assays were performed to comprehend the transduction of the canonical TGF-β pathway. Results We report that mesenchymal-like HCC cell lines are resistant to TGF-β-induced growth suppression. The vast majority of cell lines have an active canonical signaling from the cell membrane to the nucleus. Three cell lines had lost the expression of cytostatic effector genes. Conclusion Our findings reveal that cytostatic TGF-β responses have been selectively lost in mesenchymal-like HCC cell lines. Notably, their lack of responsiveness was not associated with a widespread impairment of TGF-β signaling cascade. These cell lines may serve as valuable models for studying the molecular mechanisms underlying the loss of TGF-β-mediated cytostasis during hepatocarcinogenesis.
- Published
- 2021
- Full Text
- View/download PDF
20. EMP2 induces cytostasis and apoptosis via the TGFβ/SMAD/SP1 axis and recruitment of P2RX7 in urinary bladder urothelial carcinoma
- Author
-
Ya-Wen Hsu, Cheng-Tang Pan, Meng-Shin Shiao, Ti-Chun Chan, Yow-Ling Shiue, Jia-Chen Lai, Pichpisith Vejvisithsakul, Szu-Yu Chen, and Chien-Feng Li
- Subjects
Cancer Research ,Gene knockdown ,Chemistry ,Purinergic receptor ,Cell ,General Medicine ,SMAD ,Cytostasis ,medicine.anatomical_structure ,Oncology ,Downregulation and upregulation ,Apoptosis ,Cancer research ,medicine ,Molecular Medicine ,Signal transduction - Abstract
Urinary bladder urothelial carcinoma (UBUC) is a common malignant disease, and its high recurrence rates impose a heavy clinical burden. The objective of this study was to identify signaling pathways downstream of epithelial membrane protein 2 (EMP2), which induces cytostasis and apoptosis in UBUC. A series of in vitro and in vivo assays using different UBUC-derived cell lines and mouse xenograft models were performed, respectively. In addition, primary UBUC specimens were evaluated by immunohistochemistry. Exogenous expression of EMP2 in J82 UBUC cells significantly decreased DNA replication and altered the expression levels of several TGFβ signaling-related proteins. EMP2 knockdown in BFTC905 UBUC cells resulted in opposite effects. EMP2-dysregulated cell cycle progression was found to be mediated by the TGFβ/TGFBR1/SP1 family member SMAD. EMP2 or purinergic receptor P2X7 (P2RX7) gene expression upregulation induced apoptosis via both intrinsic and extrinsic pathways. In 242 UBUC patient samples, P2RX7 protein levels were found to be significantly and positively correlated with EMP2 protein levels. Low P2RX7 levels conferred poor disease-specific and metastasis-free survival rates, and significantly decreased apoptotic cell rates. EMP2 was found to physically interact with P2RX7. In the presence of a P2RX7 agonist, BzATP, overexpression of both EMP2 and P2RX7 significantly increased apoptotic cell rates compared to overexpression of EMP2 or P2RX7 alone. EMP2 induces cytostasis via the TGFβ/SMAD/SP1 axis and recruits P2RX7 to enhance apoptosis in UBUC. Our data provide new insights that may be employed for the design of UBUC targeting therapies.
- Published
- 2021
- Full Text
- View/download PDF
21. Contextual Regulation of TGF-β Signaling in Liver Cancer
- Author
-
Shuo Tu, Wei Huang, Chunhong Huang, Zhijun Luo, and Xiaohua Yan
- Subjects
tgf-β signaling ,smad ,contextual regulation ,liver cancer ,hepatocellular carcinoma ,cytostasis ,Cytology ,QH573-671 - Abstract
Primary liver cancer is one of the leading causes for cancer-related death worldwide. Transforming growth factor beta (TGF-β) is a pleiotropic cytokine that signals through membrane receptors and intracellular Smad proteins, which enter the nucleus upon receptor activation and act as transcription factors. TGF-β inhibits liver tumorigenesis in the early stage by inducing cytostasis and apoptosis, but promotes malignant progression in more advanced stages by enhancing cancer cell survival, EMT, migration, invasion and finally metastasis. Understanding the molecular mechanisms underpinning the multi-faceted roles of TGF-β in liver cancer has become a persistent pursuit during the last two decades. Contextual regulation fine-tunes the robustness, duration and plasticity of TGF-β signaling, yielding versatile albeit specific responses. This involves multiple feedback and feed-forward regulatory loops and also the interplay between Smad signaling and non-Smad pathways. This review summarizes the known regulatory mechanisms of TGF-β signaling in liver cancer, and how they channel, skew and even switch the actions of TGF-β during cancer progression.
- Published
- 2019
- Full Text
- View/download PDF
22. Structure-Based Design of Potent and Orally Active Isoindolinone Inhibitors of MDM2-p53 Protein–Protein Interaction
- Author
-
Benoit Carbain, Keisha Hearn, Ildiko Maria Buck, Burcu Anil, Sarah J. Cully, Gianni Chessari, Jane A. Endicott, John Lunec, Neil T. Thompson, Juan Castro, Roger J. Griffin, Rhian S. Holvey, Karen Haggerty, Charlotte H. Revill, Ruth H. Bawn, Stephen R. Wedge, Christiane Riedinger, Christopher N. Johnson, Bernard T. Golding, Lynsey Fazal, Ian R. Hardcastle, Mladen Vinkovic, Claire E. Jennings, Jong Sook Ahn, Bian Zhang, Pamela A. Williams, Celine Cano, Suzannah J. Harnor, Ben Cons, Stephen J. Hobson, E. Anscombe, Jeffrey D. St. Denis, Steven Howard, David R. Newell, Emiliano Tamanini, Nicola E. Wilsher, Miller Duncan Charles, Huw D. Thomas, Timothy J. Blackburn, Martin E.M. Noble, Judith Reeks, Yan Zhao, and Luke Bevan
- Subjects
Male ,Metabolite ,Mice, Nude ,Antineoplastic Agents ,Bone Neoplasms ,Isoindoles ,Pharmacology ,Crystallography, X-Ray ,01 natural sciences ,Structure-Activity Relationship ,03 medical and health sciences ,chemistry.chemical_compound ,Drug Stability ,In vivo ,Cell Line, Tumor ,Drug Discovery ,Animals ,Humans ,Structure–activity relationship ,neoplasms ,Cell Proliferation ,030304 developmental biology ,Mice, Inbred BALB C ,Osteosarcoma ,0303 health sciences ,Molecular Structure ,biology ,Chemistry ,Proto-Oncogene Proteins c-mdm2 ,Ligand (biochemistry) ,Xenograft Model Antitumor Assays ,Cytostasis ,Small molecule ,In vitro ,0104 chemical sciences ,Macaca fascicularis ,010404 medicinal & biomolecular chemistry ,Microsomes, Liver ,biology.protein ,Molecular Medicine ,Mdm2 ,Female ,Protein Multimerization ,Tumor Suppressor Protein p53 ,Protein Binding - Abstract
Inhibition of murine double minute 2 (MDM2)-p53 protein-protein interaction with small molecules has been shown to reactivate p53 and inhibit tumor growth. Here, we describe rational, structure-guided, design of novel isoindolinone-based MDM2 inhibitors. MDM2 X-ray crystallography, quantum mechanics ligand-based design, and metabolite identification all contributed toward the discovery of potent in vitro and in vivo inhibitors of the MDM2-p53 interaction with representative compounds inducing cytostasis in an SJSA-1 osteosarcoma xenograft model following once-daily oral administration.
- Published
- 2021
- Full Text
- View/download PDF
23. Molecular and Cellular Advantage of Transdifferentiation System for Asexual Reproduction of the Tunicate, Polyandrocarpa misakiensis
- Author
-
Kawamura, Kazuo, Sawada, Hitoshi, editor, Yokosawa, Hideyoshi, editor, and Lambert, Charles C., editor
- Published
- 2001
- Full Text
- View/download PDF
24. Cytostatic Effects of Polyethyleneimine Surfaces on the Mesenchymal Stromal Cell Cycle
- Author
-
Anna Alba, Giusy Villaggio, Grazia Maria Lucia Messina, Massimo Caruso, Concetta Federico, Maria Teresa Cambria, Giovanni Marletta, and Fulvia Sinatra
- Subjects
cell spreading ,polymeric polyelectrolyte multilayers ,bone mesenchymal stromal cells ,QCM-D ,cytostasis ,Polymers and Plastics ,General Chemistry - Abstract
Polyelectrolytes assembled layer-by-layer (PEMs) are commonly used as functional coatings to build-up biological interfaces, particularly suitable as compatible layers for the interaction with a biological medium, providing suitable conditions to promote or prevent cell seeding while maintaining the phenotype. The proper assessment of the biocompatibility of PEMs and the elucidation of the related mechanisms are therefore of paramount importance. In this study, we report in detail the effect of two different PEM endings, polystyrene sulfonate (PSS) and polyethylenimine (PEI), respectively, on the cell adhesion, growth, and viability of human bone mesenchymal stromal cells (MSCs). The results have shown that PSS-ended substrates appear to be the most suitable to drive the cell adhesion and phenotype maintenance of MSCs, showing good biocompatibility. On the contrary, while the cells seem to adhere more quickly and strongly on the PEI-ended surfaces, the interaction with PEI significantly affects the growth and viability, reducing the cell spreading capability, by sequestering the adhesion molecules already in the very early steps of cell–substrate contact. These results point to the promotion of a cytostatic effect of PEI, rather than the often-claimed cytotoxicity.
- Published
- 2022
25. Comparative in vitro investigation of anticancer copper chelating agents.
- Author
-
Gaál, Anikó, Mihucz, Victor G., Bősze, Szilvia, Szabó, Ildikó, Baranyi, Marcell, Horváth, Péter, Streli, Christina, and Szoboszlai, Norbert
- Subjects
- *
CELL-mediated cytotoxicity , *DRUG analysis , *BIOACCUMULATION , *COPPER chelates , *DNA damage , *COMPARATIVE studies ,ANTINEOPLASTIC agent testing - Abstract
Evaluation of in vitro cytotoxicity of several Cu chelating agents - 2,2′-biquinoline, 8-hydroxyquinoline (oxine), ammonium pyrrolidinedithiocarbamate (APDTC), di-2-pyridylketone-4,4,-dimethyl-3-thiosemicarbazone (Dp44mT), dithizone and neocuproine – on HT-29 human colon adenocarcinoma as well as long term in vitro cytostatic effect were assessed in the presence of Cu(II) ions. Intracellular Cu accumulation was observed for each chelating agent. Cytotoxicity was also investigated on HCT-15 and HCT-116 human colon adenocarcinomas, HT-1080 human fibrosarcoma, A-375 malignant melanoma, MCF-7, MDA-MB-231 and ZR-75-1 human breast adenocarcinomas as well as MeT-5A and P31wt mesothelioma. For both short and long term antiproliferative studies, each chelating agent proved to be much more effective in the presence of Cu(II) ions. Generally, the following cytotoxicity order could be established on each cell line: Dp44mT > neocuproine > APDTC > oxine > 2,2′ biquinoline ≈ dithizone. The IC 50 values even showed one order of magnitude difference among the cell lines. Considerable differences were also observed for colony formation and spheroid growth inhibition. Thus, for Dp44mT and neocuproine, practically no resistant cell line could be developed, whereas for the rest of the chelating agents the long term survival was ensured. By raising the Cu(II) concentration from 0.5 μM to 50 μM, dramatically higher apoptotic processes were induced for Dp44mT and oxine after just 20 min. Elevated Cu(II) concentration activated reactive oxygen species generation. The investigated chelating agents restored the DNA damage caused by free Cu(II). Thus, DNA is not the target of the intracellular Cu accumulation. However, DNA intercalation was observed for the Cu(II) and neocuproine combination. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
26. Synthesis and biological evolution of hydrazones derived from 4-(trifluoromethyl)benzohydrazide.
- Author
-
Krátký, Martin, Bősze, Szilvia, Baranyai, Zsuzsa, Stolaříková, Jiřina, and Vinšová, Jarmila
- Subjects
- *
HYDRAZONES , *MYCOBACTERIUM tuberculosis , *TOXICITY testing , *CELL lines , *CAMPHOR - Abstract
Reflecting the known biological activity of isoniazid-based hydrazones, seventeen hydrazones of 4-(trifluoromethyl)benzohydrazide as their bioisosters were synthesized from various benzaldehydes and aliphatic ketones. The compounds were screened for their in vitro activity against Mycobacterium tuberculosis , nontuberculous mycobacteria ( M. avium , M. kansasii ), bacterial and fungal strains. The most antimicrobial potent derivatives were also investigated for their cytostatic and cytotoxic properties against three cell lines. Camphor-based molecule, 4-(trifluoromethyl)- N ′-(1,7,7-trimethylbicyclo[2.2.1]heptan-2-ylidene)benzohydrazide, exhibited the highest and selective inhibition of M. tuberculosis with the minimum inhibitory concentration (MIC) of 4 µM, while N ′-(4-chlorobenzylidene)-4-(trifluoromethyl)benzohydrazide was found to be superior against M. kansasii (MIC = 16 µM). N ′-(5-Chloro-2-hydroxybenzylidene)-4-(trifluoromethyl)benzohydrazide showed the lowest MIC values for gram-positive bacteria including methicillin-resistant Staphylococcus aureus as well as against two fungal strains of Candida glabrata and Trichophyton mentagrophytes within the range of ≤0.49–3.9 µM. The convenient substitution of benzylidene moiety at the position 4 or the presence of 5-chloro-2-hydroxybenzylidene scaffold concomitantly with a sufficient lipophilicity are essential for the noticeable antimicrobial activity. This 5-chlorosalicylidene derivative avoided any cytotoxicity on two mammalian cell cultures (HepG2, BMM Φ ) up to the concentration of 100 µM, but it affected the growth of MonoMac6 cells. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
27. Comparative study on the induction of complex genomic alterations after exposure of mammalian cells to carboplatin and oxaliplatin.
- Author
-
de Souza, Ana Paula, Lehmann, Mauricio, and Dihl, Rafael Rodrigues
- Subjects
- *
METAL complexes , *TUMORS , *CARBOPLATIN , *OXALIPLATIN , *COMPARATIVE method - Abstract
Metal complexes are still broadly used as the first line of the treatment for different types of tumors nowadays. Carboplatin and oxaliplatin were authorized for clinical use, even though there is little information on the mutagenic profile associated to their usage. This study evaluated the cytostatic effects and the induction of complex genomic alterations after 24-h treatment of CHO-K1 cells to concentrations of 12.5–800 μM of carboplatin and oxaliplatin in the cytokinesis-block micronucleus assay (CBMN-Cyt). The results demonstrated that carboplatin and oxaliplatin significantly increased the frequency of micronuclei (MN), nucleoplasmatic bridges (NPBs), and nuclear buds (NBUDs). On one hand, oxaliplatin induces significantly more chromosomal abnormalities than carboplatin at concentrations of 12.5 and 25 μM. On the other hand, carboplatin, in cells exposed to concentrations of 50 and 100 μM, is more efficient than oxaliplatin in the induction of chromosomal instability events. Both drugs cause significant reduction in the cytokinesis-block proliferation index, demonstrating their cytostatic effects at concentrations 50–800 μM. The results of this study shed more light on the characterization of biological effects associated with the exposure to carboplatin and oxaliplatin. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
28. Simultaneous Interference with HER1/EGFR and RAC1 Signaling Drives Cytostasis and Suppression of Survivin in Human Glioma Cells in Vitro.
- Author
-
Karpel-Massler, G., Westhoff, M.-A., Kast, R., Dwucet, A., Karpel-Massler, S., Nonnenmacher, L., Siegelin, M., Wirtz, C., Debatin, K.-M., and Halatsch, M.-E.
- Subjects
- *
GLOMERULAR filtration rate , *SURVIVIN (Protein) , *EPIDERMAL growth factor receptors , *CELLULAR signal transduction , *GLIOMAS , *IN vitro studies - Abstract
We have previously reported that combined inhibition of the epidermal growth factor receptor by erlotinib and of RAC1 by NSC23766 yielded a synergistic antiproliferative effect on established and primary cultured glioblastoma cells. The current study aimed at identifying the molecular mechanism. Staining for annexin V/PI or carboxyfluorescein succinimidyl ester was performed in order to determine the induction of apoptosis, necrosis or cytostasis in established and primary cultured glioblastoma cells. Moreover, expression of Ki-67 was determined by immunofluorescence, and the expression of cell cycle proteins was analysed by Western blot. Our data show that combined treatment with erlotinib and NSC23766 resulted in a reduced number of cell divisions, a significantly decreased Ki-67 expression, increased apoptosis and autophagy when compared to single agent treatments. On the molecular level, concomitant treatment with both agents resulted in a pronounced downregulation of cyclin D1, cyclin-dependent kinases 2, 4 and 6, as well as of survivin when compared to treatments with either agent alone. In conclusion, we demonstrate that combined treatment of human glioma cell lines in vitro with erlotinib and NSC23766 markedly inhibits cell division, induces apoptosis independent of caspase-3 activation and induces autophagy concomitant with suppression of survivin. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
29. Radiotherapy Delivered before CDK4/6 Inhibitors Mediates Superior Therapeutic Effects in ER+ Breast Cancer
- Author
-
Giulia Petroni, Silvia C. Formenti, Aitziber Buqué, Selina Chen-Kiang, Lorenzo Galluzzi, Takahiro Yamazaki, Norma Bloy, and Maurizio Di Liberto
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Therapeutic effect ,Palbociclib ,medicine.disease ,Cytostasis ,Clinical trial ,Radiation therapy ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Breast cancer ,In vivo ,030220 oncology & carcinogenesis ,Internal medicine ,Medicine ,business ,Clonogenic assay - Abstract
Purpose:Recent preclinical data suggest that cyclin-dependent kinase 4/6 (CDK4/6) inhibition may be harnessed to sensitize estrogen receptor–positive (ER+) breast cancer to radiotherapy. However, these findings were obtained in human ER+ breast cancer cell lines exposed to subclinical doses of CDK4/6 inhibitors with limited attention to treatment schedule. We investigated the activity of radiotherapy combined with the prototypic CDK4/6 inhibitor palbociclib placing emphasis on therapeutic schedule.Experimental Design:We combined radiotherapy and palbociclib in various doses and therapeutic schedules in human and mouse models of ER+ and ER-negative (ER−) breast cancer, including an immunocompetent mouse model that recapitulates key features of human luminal B breast cancer in women. We assessed proliferation, cell death, cell-cycle control, and clonogenic survival in vitro, as well as tumor growth, overall survival, and metastatic dissemination in vivo.Results:Radiotherapy and palbociclib employed as standalone agents had partial cytostatic effects in vitro, correlating with suboptimal tumor control in vivo. However, while palbociclib delivered before focal radiotherapy provided minimal benefits as compared with either treatment alone, delivering focal radiotherapy before palbociclib mediated superior therapeutic effects, even in the absence of p53. Such superiority manifested in vitro with enhanced cytostasis and loss of clonogenic potential, as well as in vivo with improved local and systemic tumor control.Conclusions:Our preclinical findings demonstrate that radiotherapy delivered before CDK4/6 inhibitors mediates superior antineoplastic effects compared with alternative treatment schedules, calling into question the design of clinical trials administering CDK4/6 inhibitors before radiotherapy in women with ER+ breast cancer.
- Published
- 2021
- Full Text
- View/download PDF
30. Metabolic Adaptations to MEK and CDK4/6 Cotargeting in Uveal Melanoma
- Author
-
Usman Baqai, Anna Han, Takami Sato, Michael A. Davies, Jessica L.F. Teh, Andrew E. Aplin, Connie Liao, Timothy J. Purwin, Julio A. Aguirre-Ghiso, Nelisa Bechtel, Vivian Chua, and Prem Patel
- Subjects
Uveal Neoplasms ,0301 basic medicine ,Cancer Research ,endocrine system diseases ,MAP Kinase Kinase 1 ,Estrogen receptor ,Apoptosis ,Oxidative Phosphorylation ,Mice ,0302 clinical medicine ,Tumor Cells, Cultured ,Melanoma ,integumentary system ,biology ,Kinase ,Cytostasis ,Gene Expression Regulation, Neoplastic ,Oncology ,030220 oncology & carcinogenesis ,Benzamides ,Female ,biological phenomena, cell phenomena, and immunity ,GNAQ ,Pyridones ,Mice, Nude ,Pyrimidinones ,Palbociclib ,Article ,03 medical and health sciences ,Oxygen Consumption ,Downregulation and upregulation ,Cyclin-dependent kinase ,Biomarkers, Tumor ,medicine ,Animals ,Humans ,Protein Kinase Inhibitors ,neoplasms ,Cell Proliferation ,business.industry ,Gene Expression Profiling ,Diphenylamine ,Cyclin-Dependent Kinase 4 ,Cyclin-Dependent Kinase 6 ,medicine.disease ,Xenograft Model Antitumor Assays ,enzymes and coenzymes (carbohydrates) ,030104 developmental biology ,biology.protein ,Cancer research ,Transcriptome ,business - Abstract
Frequent GNAQ and GNA11 mutations in uveal melanoma hyperactivate the MEK–ERK signaling pathway, leading to aberrant regulation of cyclin-dependent kinases (CDK) and cell-cycle progression. MEK inhibitors (MEKi) alone show poor efficacy in uveal melanoma, raising the question of whether downstream targets can be vertically inhibited to provide long-term benefit. CDK4/6 selective inhibitors are FDA-approved in patients with estrogen receptor (ER)–positive breast cancer in combination with ER antagonists/aromatase inhibitors. We determined the effects of MEKi plus CDK4/6 inhibitors (CDK4/6i) in uveal melanoma. In vitro, palbociclib, a CDK4/6i, enhanced the effects of MEKi via downregulation of cell-cycle proteins. In contrast, in vivo CDK4/6 inhibition alone led to cytostasis and was as effective as MEKi plus CDK4/6i treatment at delaying tumor growth. RNA sequencing revealed upregulation of the oxidative phosphorylation (OxPhos) pathway in both MEKi-resistant tumors and CDK4/6i-tolerant tumors. Furthermore, oxygen consumption rate was increased following MEKi + CDK4/6i treatment. IACS-010759, an OxPhos inhibitor, decreased uveal melanoma cell survival in combination with MEKi + CDK4/6i. These data highlight adaptive upregulation of OxPhos in response to MEKi + CDK4/6i treatment in uveal melanoma and suggest that suppression of this metabolic state may improve the efficacy of MEKi plus CDK4/6i combinations.
- Published
- 2020
- Full Text
- View/download PDF
31. Half-Sandwich Type Platinum-Group Metal Complexes of C-Glucosaminyl Azines: Synthesis and Antineoplastic and Antimicrobial Activities
- Author
-
István Kacsir, Adrienn Sipos, Evelin Major, Nikolett Bajusz, Attila Bényei, Péter Buglyó, László Somsák, Gábor Kardos, Péter Bai, and Éva Bokor
- Subjects
ruthenium ,osmium ,iridium ,rhodium ,half-sandwich complex ,C-glucosaminyl heterocycles ,azines ,Staphylococcus aureus ,Enterococcus ,MRSA ,VRE ,cytostasis ,ovarian cancer ,Chemistry (miscellaneous) ,Organic Chemistry ,Drug Discovery ,Molecular Medicine ,Pharmaceutical Science ,Physical and Theoretical Chemistry ,Analytical Chemistry - Abstract
While platinum-based compounds such as cisplatin form the backbone of chemotherapy, the use of these compounds is limited by resistance and toxicity, driving the development of novel complexes with cytostatic properties. In this study, we synthesized a set of half-sandwich complexes of platinum-group metal ions (Ru(II), Os(II), Ir(III) and Rh(III)) with an N,N-bidentate ligand comprising a C-glucosaminyl group and a heterocycle, such as pyridine, pyridazine, pyrimidine, pyrazine or quinoline. The sugar-containing ligands themselves are unknown compounds and were obtained by nucleophilic additions of lithiated heterocycles to O-perbenzylated 2-nitro-glucal. Reduction of the adducts and, where necessary, subsequent protecting group manipulations furnished the above C-glucosaminyl heterocycles in their O-perbenzylated, O-perbenzoylated and O-unprotected forms. The derived complexes were tested on A2780 ovarian cancer cells. Pyridine, pyrazine and pyridazine-containing complexes proved to be cytostatic and cytotoxic on A2780 cells, while pyrimidine and quinoline derivatives were inactive. The best complexes contained pyridine as the heterocycle. The metal ion with polyhapto arene/arenyl moiety also impacted on the biological activity of the complexes. Ruthenium complexes with p-cymene and iridium complexes with Cp* had the best performance in ovarian cancer cells, followed by osmium complexes with p-cymene and rhodium complexes with Cp*. Finally, the chemical nature of the protective groups on the hydroxyl groups of the carbohydrate moiety were also key determinants of bioactivity; in particular, O-benzyl groups were superior to O-benzoyl groups. The IC50 values of the complexes were in the low micromolar range, and, importantly, the complexes were less active against primary, untransformed human dermal fibroblasts; however, the anticipated therapeutic window is narrow. The bioactive complexes exerted cytostasis on a set of carcinomas such as cell models of glioblastoma, as well as breast and pancreatic cancers. Furthermore, the same complexes exhibited bacteriostatic properties against multiresistant Gram-positive Staphylococcus aureus and Enterococcus clinical isolates in the low micromolar range.
- Published
- 2023
- Full Text
- View/download PDF
32. Molecules
- Author
-
Joshua H. Butler, Heather J. Painter, Emily K. Bremers, Priscilla Krai, Manuel Llinás, and Maria B. Cassera
- Subjects
Chemistry (miscellaneous) ,Organic Chemistry ,Drug Discovery ,Plasmodium ,Crinum erubescens ,cripowellin ,ring stage ,cytostasis ,transcription ,Molecular Medicine ,Pharmaceutical Science ,Physical and Theoretical Chemistry ,Analytical Chemistry - Abstract
Cripowellins from Crinum erubescens are known pesticidal and have potent antiplasmodial activity. To gain mechanistic insights to this class of natural products, studies to determine the timing of action of cripowellins within the asexual intraerythrocytic cycle of Plasmodium falciparum were performed and led to the observation that this class of natural products induced reversible cytostasis in the ring stage within the first 24 h of treatment. The transcriptional program necessary for P. falciparum to progress through the asexual intraerythrocytic life cycle is well characterized. Whole transcriptome abundance analysis showed that cripowellin B “pauses” the transcriptional program necessary to progress through the intraerythrocytic life cycle coinciding with the lack of morphological progression of drug treated parasites. In addition, cripowellin B-treated parasites re-enter transcriptional progression after treatment was removed. This study highlights the use of cripowellins as chemical probes to reveal new aspects of cell cycle progression of the asexual ring stage of P. falciparum which could be leveraged for the generation of future antimalarial therapeutics. Published version
- Published
- 2023
- Full Text
- View/download PDF
33. Interaction of a standardized mistletoe (Viscum album) preparation with antitumor effects of Trastuzumab in vitro.
- Author
-
Weissenstein, U., Kunz, M., Urech, K., Regueiro, U., and Baumgartner, S.
- Subjects
BREAST tumor prevention ,CANCER prevention ,ALTERNATIVE medicine ,ANALYSIS of variance ,ANTINEOPLASTIC agents ,APOPTOSIS ,BIOLOGICAL assay ,BIOLOGICAL models ,CELL cycle ,CELL physiology ,DRUG synergism ,ENZYME-linked immunosorbent assay ,DRUG-herb interactions ,ONCOGENES ,PROBABILITY theory ,RESEARCH funding ,STATISTICS ,VISCUM ,TRASTUZUMAB ,DATA analysis ,STATISTICAL significance ,VASCULAR endothelial growth factors ,DATA analysis software ,IN vitro studies ,PHARMACODYNAMICS - Abstract
Background: Besides conventional anticancer therapy many breast cancer patients use complementary and alternative medicine (CAM) like the medicinal herb mistletoe (Viscum album L.). To gain more knowledge about possible herb-drug interactions between CAM and conventional anticancer medications, in the present in vitro study we investigated the effect of a standardized mistletoe preparation on the action of Trastuzumab, a drug used for the treatment of Her-2 positive breast cancer. Methods: The Her-2 positive human breast carcinoma cell line SK-BR-3 was treated with Trastuzumab. Different doses of the drug were combined with Viscum album extract (VAE) in clinically relevant doses. Proliferation, apoptosis, cell cycle and the secretion of vascular endothelial growth factor (VEGF) were analyzed. Results: No inhibition of antitumor efficacy of Trastuzumab by VAE was detected. VAE and Trastuzumab, either alone or in combination, inhibited proliferation of SK-BR-3 cells in vitro. At higher concentrations VAE induced apoptosis, which was not observed for Trastuzumab. Cells treated with Trastuzumab underwent a G0/G1 cell cycle arrest and cells treated with VAE a G2/M arrest. After application of the two drugs in combination both G0/G1 and G2/M arrest was observed. VEGF secretion of SK-BR-3 cells was significantly inhibited by sole treatment with Trastuzumab or VAE. Combined treatment of Trastuzumab and VAE at clinically relevant doses showed additive inhibitory effects on VEGF secretion. Conclusions: VAE did not interfere with cytostatic effects of Trastuzumab on SK-BR-3 cells in vitro. Our in vitro results suggest that no risk of safety by herb drug interactions has to be expected from the exposition of cancer cells to Trastuzumab and VAE simultaneously. In contrast, VAE and Trastuzumab seem to exhibit complementary anti-cancer effects in vitro. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
34. Metabolic Adaptation of Airway Smooth Muscle Cells to an SPHK2 Substrate Precedes Cytostasis
- Author
-
David Marsolais, Jorge Soliz, Pascale Blais-Lecours, Vincent Joseph, Janette K. Burgess, Christian Arias-Reyes, Andrew J. Halayko, Webster L. Santos, Sofien Laouafa, Restoring Organ Function by Means of Regenerative Medicine (REGENERATE), and Groningen Research Institute for Asthma and COPD (GRIAC)
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,FTY720 ,Stromal cell ,endogenous compound ,sphingosine kinase 2 ,Clinical Biochemistry ,oxidative phosphorylation ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,cytostasis ,controlled study ,fingolimod ,Molecular Biology ,protein expression ,tetrazolium ,Sphingolipids ,Sphingosine ,sphingosine ,Kinase ,Cell growth ,human cell ,article ,Sphingosine Kinase 2 ,AAL-R ,lactate dehydrogenase ,Cell Biology ,respiratory system ,asthma ,protein function ,bronchus hyperreactivity ,Cytostasis ,Sphingolipid ,Cell biology ,enzyme activity ,SPHK2 ,030104 developmental biology ,Metabolism ,cell proliferation ,030228 respiratory system ,chemistry ,stroma cell ,gene expression ,Ki 67 antigen ,sphingolipid ,oxygen ,airway smooth muscle cell - Abstract
Thickening of the airway smooth muscle is central to bronchial hyperreactivity. We have shown that the sphingosine analog (R)-2-amino-4-(4-heptyloxyphenyl)-2-methylbutanol (AAL-R) can reverse preestablished airway hyperreactivity in a chronic asthma model. Because sphingosine analogs can be metabolized by SPHK2 (sphingosine kinase 2), we investigated whether this enzyme was required for AAL-R to perturb mechanisms sustaining airway smooth muscle cell proliferation. We found that AAL-R pretreatment reduced the capacity of live airway smooth muscle cells to use oxygen for oxidative phosphorylation and increased lactate dehydrogenase activity. We also determined that SPHK2 was upregulated in airway smooth muscle cells bearing the proliferation marker Ki67 relative to their Ki67-negative counterpart. Comparing different stromal cell subsets of the lung, we found that high SPHK2 concentrations were associated with the ability of AAL-R to inhibit metabolic activity assessed by conversion of the tetrazolium dye MTT. Knockdown or pharmacological inhibition of SPHK2 reversed the effect of AAL-R on MTT conversion, indicating the essential role for this kinase in the metabolic perturbations induced by sphingosine analogs. Our results support the hypothesis that increased SPHK2 levels in proliferating airway smooth muscle cells could be exploited to counteract airway smooth muscle thickening with synthetic substrates.
- Published
- 2020
- Full Text
- View/download PDF
35. Mutagenic and cytostatic activities of the Xylopia laevigata essential oil in human lymphocytes
- Author
-
Emmanoel V. Costa, Giovana N. M. Esquissato, Tais Susane Pereira, Paulo Cesar de Lima Nogueira, and Marialba Avezum Alves de Castro-Prado
- Subjects
Mitotic index ,Proliferation index ,Traditional medicine ,biology ,010405 organic chemistry ,Chemistry ,Organic Chemistry ,Plant Science ,biology.organism_classification ,01 natural sciences ,Biochemistry ,Cytostasis ,0104 chemical sciences ,Analytical Chemistry ,law.invention ,010404 medicinal & biomolecular chemistry ,chemistry.chemical_compound ,Germacrene ,law ,Micronucleus test ,Cytotoxicity ,Xylopia ,Essential oil - Abstract
Hydro-distilled essential oil from leaves of Xylopia laevigata was characterized by GC-MS. Twenty-seven components were identified and the oil’s major constituents comprised germacrene D, bicyclogermacrene, (E)-caryophyllene and germacrene B. The cytotoxicity of the essential oil of X. laevigata (EOXL), determined by MTT and mitotic index methods in cultured human lymphocytes was observed in all tested concentrations. Cultures treated with EOXL demonstrated significant increase in the frequencies of micronuclei in the cytokinesis-block micronucleus assay (CBMN) and reduction of the cytokinesis-block proliferation index (CBPI) rates. Results demonstrated the cytostatic and mutagenic effects of EOXL, the latter for the first time.
- Published
- 2019
- Full Text
- View/download PDF
36. Combined Aurora Kinase A (AURKA) and WEE1 Inhibition Demonstrates Synergistic Antitumor Effect in Squamous Cell Carcinoma of the Head and Neck
- Author
-
Natalia Issaeva, Dong-Hua Yang, Jeffrey P. Townsend, Ranee Mehra, Stephen G. Gaffney, Elizabeth B. Perry, Kyung Jin Eoh, Janaki Parameswaran, Wendell G. Yarbrough, Fang Zhu, Ja Seok Koo, Teresa Sandoval-Schaefer, Ilya G. Serebriiskii, Roshan Sharma, Jong Woo Lee, Barbara Burtness, and Erica A. Golemis
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Aurora A kinase ,Fluorescent Antibody Technique ,Gene Expression ,Antineoplastic Agents ,Apoptosis ,Cell Cycle Proteins ,Article ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,Animals ,Humans ,Medicine ,Protein Kinase Inhibitors ,Mitotic catastrophe ,Aurora Kinase A ,Neoplasm Staging ,Cyclin-dependent kinase 1 ,biology ,Squamous Cell Carcinoma of Head and Neck ,business.industry ,Drug Synergism ,Protein-Tyrosine Kinases ,Xenograft Model Antitumor Assays ,Cytostasis ,Disease Models, Animal ,Wee1 ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Alisertib ,Cancer research ,biology.protein ,Female ,Neoplasm Grading ,business - Abstract
Purpose: Human papillomavirus (HPV)-negative head and neck squamous cell carcinomas (HNSCC) commonly bear disruptive mutations in TP53, resulting in treatment resistance. In these patients, direct targeting of p53 has not been successful, but synthetic lethal approaches have promise. Although Aurora A kinase (AURKA) is overexpressed and an oncogenic driver, its inhibition has only modest clinical effects in HPV-negative HNSCC. We explored a novel combination of AURKA and WEE1 inhibition to overcome intrinsic resistance to AURKA inhibition. Experimental Design: AURKA protein expression was determined by fluorescence-based automated quantitative analysis of patient specimens and correlated with survival. We evaluated treatment with the AURKA inhibitor alisertib (MLN8237) and the WEE1 inhibitor adavosertib (AZD1775), alone or in combination, using in vitro and in vivo HNSCC models. Results: Elevated nuclear AURKA correlated with worse survival among patients with p16(−) HNSCC. Alisertib caused spindle defects, G2–M arrest and inhibitory CDK1 phosphorylation, and cytostasis in TP53 mutant HNSCC FaDu and UNC7 cells. Addition of adavosertib to alisertib instead triggered mitotic entry and mitotic catastrophe. Moreover, in FaDu and Detroit 562 xenografts, this combination demonstrated synergistic effects on tumor growth and extended overall survival compared with either vehicle or single-agent treatment. Conclusions: Combinatorial treatment with adavosertib and alisertib leads to synergistic antitumor effects in in vitro and in vivo HNSCC models. These findings suggest a novel rational combination, providing a promising therapeutic avenue for TP53-mutated cancers.
- Published
- 2019
- Full Text
- View/download PDF
37. Inhibition of thioredoxin reductase 1 by vulpinic acid suppresses the proliferation and migration of human breast carcinoma.
- Author
-
Kalın, Şeyda Nur, Altay, Ahmet, and Budak, Harun
- Subjects
- *
DOCETAXEL , *HUMAN migrations , *THIOREDOXIN , *CELL migration , *ANTINEOPLASTIC agents , *METABOLITES , *BREAST - Abstract
It was aimed to investigate the thioredoxin reductase 1 (TrxR1)-targeted anticancer effect of vulpinic (VA) and lecanoric (LA) acids, which are lichen secondary metabolites, on breast cancer MCF-7 and MDA-MB-453 cell lines, and to compare the effectiveness of this potential effect against commercial chemotherapeutic drugs carboplatin and docetaxel. The anticancer effects of both lichen metabolites were evaluated by XTT, flow cytometry analysis, cell scratch, and transwell migration assays. Apoptotic results were also confirmed by qPCR and western blot. Changes in TrxR1 were investigated in gene and protein expressions and enzyme activity levels. VA suppressed the proliferation of MCF-7 and MDA-MB-453 cells in a dose- and time-dependent manner, and the IC 50 values were calculated as 22.92 μg/ml and 95.65 μg/ml, respectively. As for LA, it did not have a considerable antiproliferative effect on both cell lines. VA had stronger cytotoxicity than both chemotherapeutic drug in MCF-7 cells and showed antiproliferative activity closer to carboplatin in MDA-MB-453 cells. qPCR, western blot, and flow cytometry analysis results revealed that VA did not induce apoptosis in both cell lines. In contrast, VA caused cell cycle arrest, significantly. Migration assay results showed that VA suppressed migration in both cells. VA induced the gene expression of TrxR1 while inhibiting its protein expression and enzymatic activity in both cell lines. The findings reveal that vulpinic acid may be a novel inhibitor candidate on TrxR1 and could be considered a potential chemotherapeutic agent for breast cancer treatment, especially in MCF-7 cells. [Display omitted] • Vulpinic acid (VA) induced cytotoxicity in human MCF-7 and MDA-MB-453 cell lines. • VA suppressed the cell migration in both cell types. • VA downregulated the protein expression and inhibited enzymatic activity of TrxR1. • VA showed its anticancer effect via targeting TrxR1 in breast cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
38. Targeting prominin2 transcription to overcome ferroptosis resistance in cancer
- Author
-
Peter Chhoy, Emmet R Karner, Caitlin W. Brown, Dimpi Mukhopadhyay, and Arthur M. Mercurio
- Subjects
Drug ,Medicine (General) ,media_common.quotation_subject ,p38 mitogen-activated protein kinases ,QH426-470 ,Article ,Mice ,R5-920 ,Transcription (biology) ,Neoplasms ,Genetics ,Medicine ,Animals ,Ferroptosis ,cancer ,HSF1 ,media_common ,therapy ,Mechanism (biology) ,business.industry ,prominin2 ,Cancer ,Articles ,medicine.disease ,Cytostasis ,heat shock factor 1 ,Cancer cell ,Cancer research ,Molecular Medicine ,Autophagy & Cell Death ,Lipid Peroxidation ,business - Abstract
Understanding how cancer cells resist ferroptosis is a significant problem that impacts ongoing efforts to stimulate ferroptosis as a therapeutic strategy. We reported that prominin2 is induced by ferroptotic stimuli and functions to resist ferroptotic death. Although this finding has significant implications for therapy, specific prominin2 inhibitors are not available. We rationalized that the mechanism by which prominin2 expression is induced by ferroptotic stress could be targeted, expanding the range of options to overcome ferroptosis resistance. Here, we show that that 4‐hydroxynonenal (4HNE), a specific lipid metabolite formed from the products of lipid peroxidation stimulates PROM2 transcription by a mechanism that involves p38 MAP kinase‐mediated activation of HSF1 and HSF1‐dependent transcription of PROM2. HSF1 inhibitors sensitize a wide variety of resistant cancer cells to drugs that induce ferroptosis. Importantly, the combination of a ferroptosis‐inducing drug and an HSF1 inhibitor causes the cytostasis of established tumors in mice, although neither treatment alone is effective. These data reveal a novel approach for the therapeutic induction of ferroptosis in cancer., Stimulating ferroptosis has emerged as a potential therapeutic strategy against cancer. Some tumor cells, however, are resistant to known ferroptosis stimuli. This study identifies mechanisms that contribute to ferroptosis and develops strategies to overcome resistance.
- Published
- 2021
39. Evaluation of Cytotoxic and Mutagenic Effects of the Synthetic Cathinones Mexedrone, α-PVP and α-PHP
- Author
-
Monia Lenzi, Patrizia Hrelia, Veronica Cocchi, Matteo Marti, Sofia Gasperini, Raffaella Arfè, Lenzi M., Cocchi V., Gasperini S., Arfe R., Marti M., and Hrelia P.
- Subjects
0301 basic medicine ,Pyrrolidines ,Mexedrone ,Apoptosis ,01 natural sciences ,Methamphetamine ,chemistry.chemical_compound ,Pentanones ,Cytotoxic T cell ,Biology (General) ,α-PHP ,Cytotoxicity ,Synthetic cathinone ,Spectroscopy ,chemistry.chemical_classification ,LS7_9 ,medicine.diagnostic_test ,mexedrone ,Cell Death ,food and beverages ,ROS ,General Medicine ,Cytostasis ,Computer Science Applications ,Chemistry ,Biochemistry ,novel psychoactive substances ,QH301-705.5 ,Cell Survival ,Socio-culturale ,α-PVP ,Catalysis ,Article ,Flow cytometry ,Cell Line ,Inorganic Chemistry ,03 medical and health sciences ,Alkaloids ,medicine ,Humans ,Mutagenicity ,Novel psychoactive substances ,S9 mix ,Synthetic cathinones ,Physical and Theoretical Chemistry ,Molecular Biology ,QD1-999 ,LS7_5 ,Reactive oxygen species ,synthetic cathinones ,flow cytometry ,010401 analytical chemistry ,Organic Chemistry ,Novel psychoactive substance ,mutagenicity ,0104 chemical sciences ,030104 developmental biology ,chemistry ,Micronucleus ,Reactive Oxygen Species ,Micronucleus, Germline ,DNA ,Mutagens - Abstract
Mexedrone, α-PVP and α-PHP are synthetic cathinones. They can be considered amphetamine-like substances with a stimulating effect. Actually, studies showing their impact on DNA are totally absent. Therefore, in order to fill this gap, aim of the present work was to evaluate their mutagenicity on TK6 cells. On the basis of cytotoxicity and cytostasis results, we selected the concentrations (35–100 µM) to be used in the further analysis. We used the micronucleus (MN) as indicator of genetic damage and analyzed the MNi frequency fold increase by flow cytometry. Mexedrone demonstrated its mutagenic potential contrary to the other two compounds, we then proceeded by repeating the analyzes in the presence of extrinsic metabolic activation in order to check if it was possible to totally exclude the mutagenic capacity for α-PVP and α-PHP. The results demonstrated instead the mutagenicity of their metabolites. We then evaluated reactive oxygen species (ROS) induction as a possible mechanism at the basis of the highlighted effects but the results did not show a statistically significant increase in ROS levels for any of the tested substances. Anyway, our outcomes emphasize the importance of mutagenicity evaluation for a complete assessment of the risk associated with synthetic cathinones exposure.
- Published
- 2021
40. Latent TGF-β Activation Is a Hallmark of the Tenascin Family
- Author
-
Laurent Berthier, Ulrich Valcourt, Sophie Liot, Alexandre Aubert, Raphaël Terreux, Lindsay B Alcaraz, Laura Prigent, Stephanie Aguero, Catherine Moali, Elise Lambert, Bernard Verrier, Perrine Mercier-Gouy, Laboratoire de Biologie Tissulaire et d'ingénierie Thérapeutique UMR 5305 (LBTI), Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS), Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), and Verrier, bernard
- Subjects
0301 basic medicine ,Protein Conformation ,medicine.medical_treatment ,Smad Proteins ,immune cell modulation ,Mice ,0302 clinical medicine ,[SDV.MHEP.MI]Life Sciences [q-bio]/Human health and pathology/Infectious diseases ,Transforming Growth Factor beta ,Immunology and Allergy ,Homeostasis ,Protein Isoforms ,Tissue homeostasis ,Original Research ,[SDV.MP.VIR] Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,chemistry.chemical_classification ,biology ,latent TGF-β activation ,Tenascin ,Cytostasis ,transforming growth factor (TGF)-β ,Recombinant Proteins ,Cell biology ,Molecular Docking Simulation ,Cytokine ,030220 oncology & carcinogenesis ,Tenascin Family ,[SDV.MP.VIR]Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,[SDV.MHEP.MI] Life Sciences [q-bio]/Human health and pathology/Infectious diseases ,[SDV.IMM.IMM] Life Sciences [q-bio]/Immunology/Immunotherapy ,Protein Binding ,Signal Transduction ,[CHIM.POLY] Chemical Sciences/Polymers ,tissue homeostasis ,Immunology ,Molecular Dynamics Simulation ,[SDV.SP.PG] Life Sciences [q-bio]/Pharmaceutical sciences/Galenic pharmacology ,Cell Line ,03 medical and health sciences ,Structure-Activity Relationship ,[SDV.IMM.VAC] Life Sciences [q-bio]/Immunology/Vaccinology ,tenascins ,medicine ,tumor microenvironment ,Animals ,Humans ,Protein Interaction Domains and Motifs ,Amino Acid Sequence ,[SDV.IB.BIO]Life Sciences [q-bio]/Bioengineering/Biomaterials ,Tumor microenvironment ,[SDV.MHEP.HEG]Life Sciences [q-bio]/Human health and pathology/Hépatology and Gastroenterology ,Epithelial Cells ,[SDV.IMM.IMM]Life Sciences [q-bio]/Immunology/Immunotherapy ,RC581-607 ,[SDV.MHEP.HEG] Life Sciences [q-bio]/Human health and pathology/Hépatology and Gastroenterology ,[SDV.IB.BIO] Life Sciences [q-bio]/Bioengineering/Biomaterials ,[CHIM.POLY]Chemical Sciences/Polymers ,[SDV.SP.PG]Life Sciences [q-bio]/Pharmaceutical sciences/Galenic pharmacology ,030104 developmental biology ,chemistry ,biology.protein ,[SDV.IMM.VAC]Life Sciences [q-bio]/Immunology/Vaccinology ,Immunologic diseases. Allergy ,Glycoprotein ,Transforming growth factor - Abstract
International audience; Transforming growth factor-β (TGF-β) isoforms are secreted as inactive complexes formed through non-covalent interactions between bioactive TGF-β entities and their N-terminal pro-domains called latency-associated peptides (LAP). Extracellular activation of latent TGF-β within this complex is a crucial step in the regulation of TGF-β activity for tissue homeostasis and immune cell function. We previously showed that the matrix glycoprotein Tenascin-X (TN-X) interacted with the small latent TGF-β complex and triggered the activation of the latent cytokine into a bioactive TGF-β. This activation most likely occurs through a conformational change within the latent TGF-β complex and requires the C-terminal fibrinogen-like (FBG) domain of the glycoprotein. As the FBG-like domain is highly conserved among the Tenascin family members, we hypothesized that Tenascin-C (TN-C), Tenascin-R (TN-R) and Tenascin-W (TN-W) might share with TN-X the ability to regulate TGF-β bioavailability through their C-terminal domain. Here, we demonstrate that purified recombinant full-length Tenascins associate with the small latent TGF-β complex through their FBG-like domains. This association promotes activation of the latent cytokine and subsequent TGF-β cell responses in mammary epithelial cells, such as cytostasis and epithelial-to-mesenchymal transition (EMT). Considering the pleiotropic role of TGF-β in numerous physiological and pathological contexts, our data indicate a novel common function for the Tenascin family in the regulation of tissue homeostasis under healthy and pathological conditions.
- Published
- 2021
- Full Text
- View/download PDF
41. α-Difluoromethylornithine-Induced Cytostasis is Reversed by Exogenous Polyamines, Not by Thymidine Supplementation
- Author
-
Jouko Vepsäläinen, M. A. Khomutov, Mervi T. Hyvönen, Alex R. Khomutov, and Tuomo A. Keinänen
- Subjects
0301 basic medicine ,Eflornithine ,polyamines ,proliferation ,Spermine ,thymidine ,Biochemistry ,Microbiology ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,DU145 ,Neoplasms ,LNCaP ,Animals ,Humans ,Molecular Biology ,α-difluoromethylornithine ,Cells, Cultured ,S-adenosyl-L-methionine ,Ornithine Decarboxylase Inhibitors ,Cytostatic Agents ,Cytostasis ,QR1-502 ,Cell biology ,Spermidine ,030104 developmental biology ,chemistry ,Cell culture ,030220 oncology & carcinogenesis ,Polyamine ,Thymidine - Abstract
Polyamine spermidine is essential for the proliferation of eukaryotic cells. Administration of polyamine biosynthesis inhibitor α-difluoromethylornithine (DFMO) induces cytostasis that occurs in two phases, the early phase which can be reversed by spermidine, spermine, and some of their analogs, and the late phase which is characterized by practically complete depletion of cellular spermidine pool. The growth of cells at the late phase can be reversed by spermidine and by very few of its analogs, including (S)-1-methylspermidine. It was reported previously (Witherspoon et al. Cancer Discovery 3(9), 1072–81, 2013) that DFMO treatment leads to depletion of cellular thymidine pools, and that exogenous thymidine supplementation partially prevents DFMO-induced cytostasis without affecting intracellular polyamine pools in HT-29, SW480, and LoVo colorectal cancer cells. Here we show that thymidine did not prevent DFMO-induced cytostasis in DU145, LNCaP, MCF7, CaCo2, BT4C, SV40MES13, HepG2, HEK293, NIH3T3, ARPE19 or HT-29 cell lines, whereas administration of functionally active mimetic of spermidine, (S)-1-methylspermidine, did. Thus, the effect of thymidine seems to be specific only for certain cell lines. We conclude that decreased polyamine levels and possibly also distorted pools of folate-dependent metabolites mediate the anti-proliferative actions of DFMO. However, polyamines are necessary and sufficient to overcome DFMO-induced cytostasis, while thymidine is generally not.
- Published
- 2021
42. Synthesis and in vitro biological evaluation of 2-(phenylcarbamoyl)phenyl 4-substituted benzoates.
- Author
-
Krátký, Martin, Bősze, Szilvia, Baranyai, Zsuzsa, Szabó, Ildikó, Stolaříková, Jiřina, Paraskevopoulos, Georgios, and Vinšová, Jarmila
- Subjects
- *
CARBAMOYL compounds , *PHENYL compounds , *SUBSTITUTION reactions , *BENZOATES , *ANTI-infective agents , *DRUG activation , *PHARMACEUTICAL chemistry - Abstract
Based on the previously described antimicrobial activity of salicylanilide derivatives, we designed and synthesized novel 2-(phenylcarbamoyl)phenyl 4-substituted benzoates. The most active salicylanilides were selected for esterification by various 4-substituted benzoic acids. These compounds were evaluated in vitro against Mycobacterium tuberculosis , including multidrug-resistant strains, nontuberculous mycobacteria ( Mycobacterium avium and Mycobacterium kansasii ), and eight bacterial and fungal strains. We also investigated the cytostatic and cytotoxic actions of the esters. The minimum inhibitory concentrations (MICs) against mycobacteria ranged from 0.125 to 8 μM. Interestingly, the drug-resistant strains exhibited the highest susceptibility without any cross-resistance with established drugs. 4-Bromo-2-[4-(trifluoromethyl)phenylcarbamoyl]phenyl 4-nitrobenzoate showed the most potent inhibition with MIC values ranging from 0.25 to 2 μM. Gram-positive bacteria, including methicillin-resistant Staphylococcus aureus , were inhibited by two derivatives with MIC values of at least 0.49 μM, whereas Gram-negative bacteria and most of the tested fungi did not display any marked susceptibility. Benzoates exhibited no cytotoxicity at concentrations up to 50 μM but most caused significant cytostasis with IC 50 values lower than 10 μM. Some cytotoxicity-based selectivity indexes for drug-susceptible and drug-resistant M. tuberculosis as well as Staphylococci were higher than 100. These values indicate that some of these derivatives are promising candidates for future research. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
43. Intra-epithelial non-canonical Activin A signalling safeguards prostate progenitor quiescence
- Author
-
Bertossi A, Francesco Gandolfi, Marco Lorenzoni, Bertossio E, Michael M. Shen, Alessandro Romanel, Alessandra Bisio, Sacha Genovesi, De Felice D, Mattia Barbareschi, Andrea Lunardi, Francesco Berardinelli, Massimo Loda, Celotti M, Alessandro Alaimo, Palumbieri, Gaspari M, Michelangelo Fiorentino, Foletto, Francesco Cambuli, Antoccia A, Mazzero G, and Zaffagni M
- Subjects
medicine.anatomical_structure ,Cell growth ,Prostate ,medicine ,Cancer research ,Biology ,Progenitor cell ,Carcinogenesis ,medicine.disease_cause ,Cytostasis ,Epithelium ,Transforming growth factor ,Progenitor - Abstract
The healthy prostate is a relatively quiescent tissue. Yet, prostate epithelium overgrowth is a common condition during ageing, associated with urinary dysfunction and tumorigenesis. For over thirty years, TGF-β ligands have been known to induce cytostasis in a large variety of epithelia, but the intracellular pathway mediating this signal in the prostate, as well as its relevance for quiescence, have remained elusive.Here, using mouse prostate organoids to model epithelial progenitors, we found that intra-epithelial non-canonical Activin A signalling inhibited cell proliferation in a Smad-independent manner. Mechanistically, Activin A triggered Tak1 and p38 MAPK activity, leading to p16 and p21 nuclear import. Spontaneous evasion from this quiescent state occurred upon prolonged culture, due to reduced Activin A secretion, a condition associated with DNA replication stress and aneuploidy. Organoids capable to escape quiescencein vitrowere also able to implant with increased frequency into immunocompetent mice.Our study demonstrates that non-canonical Activin A signalling safeguards epithelial quiescence in the healthy prostate, with potential implications for the understanding of cancer initiation, and the development of therapies targeting quiescent tumour progenitors.
- Published
- 2021
- Full Text
- View/download PDF
44. Lycorine inhibits cell proliferation, migration and invasion, and primarily exerts in vitro cytostatic effects in human colorectal cancer via activating the ROS/p38 and AKT signaling pathways
- Author
-
Xiaoji Luo, Chunmei Yang, Jinyong Luo, Lulu Zhang, Shengdong Yang, Ping Zhang, Huakun Huang, Tingting Yu, and Xiaohui Yuan
- Subjects
0301 basic medicine ,Cancer Research ,Cell cycle checkpoint ,MAP Kinase Signaling System ,Cell ,Antineoplastic Agents ,Apoptosis ,p38 Mitogen-Activated Protein Kinases ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cyclin D1 ,cytostasis ,Cell Movement ,Cell Line, Tumor ,medicine ,Humans ,Neoplasm Invasiveness ,ROS/p38 ,Protein kinase B ,neoplasms ,Cell Proliferation ,colorectal cancer cells ,Cell growth ,Chemistry ,AKT ,General Medicine ,Articles ,Cell cycle ,Lycorine ,HCT116 Cells ,Cytostasis ,digestive system diseases ,Phenanthridines ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,lycorine ,030220 oncology & carcinogenesis ,Cancer research ,Amaryllidaceae Alkaloids ,Colorectal Neoplasms ,Reactive Oxygen Species ,Proto-Oncogene Proteins c-akt - Abstract
Colorectal cancer (CRC) is a life‑threatening malignant tumor of the digestive tract. Diverse gene mutations and complicated alterations to the signaling pathways in CRC lead to heterogeneity in response to chemotherapy. Moreover, anticancer drugs for CRC chemotherapy are limited due to adverse events. Therefore, developing more effective, tolerable and safe drugs for the treatment of CRC is important. The present study aimed to investigate the effect of lycorine on human CRC cell proliferation, migration, invasion, apoptosis, cell cycle distribution, as well as the underlying molecular mechanism. The crystal violet staining and MTT assay results demonstrated that lycorine suppressed cell proliferation in a dose‑ and time‑dependent manner in the three CRC cell lines, HCT116, LoVo and SW480. Similarly, verified by performing wound healing and Transwell assays, lycorine significantly inhibited HCT116 and LoVo cell migration and invasion in vitro compared with the control group. In LoVo cells, the protein expression levels of matrix metallopeptidases, snail family transcriptional repressor 1, Vimentin and N‑cadherin were significantly downregulated, whereas the protein expression levels of E‑cadherin were significantly upregulated by lycorine treatment compared with the control group. The Hoechst 33258 staining and flow cytometry assay results indicated that lycorine mediated its cytostatic effect on CRC cells potentially via inducing cell cycle arrest, but not apoptosis. Compared with the control group, lycorine significantly induced HCT116 cell cycle arrest at the G2/M phase, but significantly induced LoVo cell cycle arrest at the S and G2/M phases. Furthermore, lycorine significantly downregulated the protein expression levels of cyclin D1 and cyclin E1, but significantly increased p21 and Smad4 protein expression levels in HCT116 and LoVo cells compared with the control group. The intracellular reactive oxygen species (ROS) measurement results also indicated that compared with the control group, lycorine significantly induced ROS accumulation, and increased phosphorylated‑p38 expression levels and AKT phosphorylation. Collectively, the present study suggested that lycorine might induce cell cycle arrest and exert cytostatic effects potentially via activating ROS/p38 and AKT signaling pathways in CRC cells.
- Published
- 2021
45. ABT-751 Induces Multiple Anticancer Effects in Urinary Bladder Urothelial Carcinoma-Derived Cells: Highlighting the Induction of Cytostasis through the Inhibition of SKP2 at Both Transcriptional and Post-Translational Levels
- Author
-
Jasmine Marianne Lee, Cheng-Tang Pan, Yow-Ling Shiue, and Seyedeh Zahra Dehghanian
- Subjects
Catalysis ,Inorganic Chemistry ,lcsh:Chemistry ,cyclin-dependent kinase inhibitors ,cytostasis ,Cyclin-dependent kinase ,S-phase kinase associated protein 2 ,Physical and Theoretical Chemistry ,CHUK ,Molecular Biology ,Mechanistic target of rapamycin ,Protein kinase B ,lcsh:QH301-705.5 ,Spectroscopy ,biology ,Chemistry ,Cell growth ,AKT ,Organic Chemistry ,General Medicine ,Cell cycle ,ABT-751 ,Cytostasis ,Computer Science Applications ,lcsh:Biology (General) ,lcsh:QD1-999 ,biology.protein ,Cancer research ,CDKN1B - Abstract
The objective was to investigate the anti-cancer effects and underlying molecular mechanisms of cytostasis which were activated by an anti-microtubule drug, ABT-751, in two urinary bladder urothelial carcinoma (UBUC)-derived cell lines, BFTC905 and J82, with distinct genetic backgrounds. A series of in vitro assays demonstrated that ABT-751 induced G2/M cell cycle arrest, decreased cell number in the S phase of the cell cycle and suppressed colony formation/independent cell growth, accompanied with alterations of the protein levels of several cell cycle regulators. In addition, ABT-751 treatment significantly hurdled cell migration and invasion along with the regulation of epithelial&ndash, mesenchymal transition-related proteins. ABT-751 triggered autophagy and apoptosis, downregulated the mechanistic target of rapamycin kinase (MTOR) and upregulated several pro-apoptotic proteins that are involved in extrinsic and intrinsic apoptotic pathways. Inhibition of autophagosome and autolysosome enhanced apoptosis was also observed. Through the inhibition of the NF&kappa, B signaling pathway, ABT-751 suppressed S-phase kinase associated protein 2 (SKP2) transcription and subsequent translation by downregulation of active/phospho-AKT serine/threonine kinase 1 (AKT1), component of inhibitor of nuclear factor kappa B kinase complex (CHUK), NFKB inhibitor alpha (NFKBIA), nuclear RELA proto-oncogene, NF&kappa, B subunit (RELA) and maintained a strong interaction between NFKBIA and RELA to prevent RELA nuclear translocation for SKP2 transcription. ABT-751 downregulated stable/phospho-SKP2 including pSKP2(S64) and pSKP2(S72), which targeted cyclin-dependent kinase inhibitors for degradation through the inactivation of AKT. Our results suggested that ABT-751 may act as an anti-cancer drug by inhibiting cell migration, invasion yet inducing cell cycle arrest, autophagy and apoptosis in distinct UBUC-derived cells. Particularly, the upstream molecular mechanism of its anticancer effects was identified as ABT-751-induced cytostasis through the inhibition of SKP2 at both transcriptional and post-translational levels to stabilize cyclin dependent kinase inhibitor 1A (CDKN1A) and CDKN1B proteins.
- Published
- 2021
- Full Text
- View/download PDF
46. 1H-NMR metabolomics reveals a multitarget action of Crithmum maritimum ethyl acetate extract in inhibiting hepatocellular carcinoma cell growth
- Author
-
Francesca Castellaneta, Antonio Mazzocca, Chiara Roberta Girelli, Gianluigi Cesari, Davide Gnocchi, Francesco Paolo Fanizzi, Carlo Sabbà, Laura Del Coco, Gnocchi, Davide, Del Coco, Laura, Girelli, Chiara Roberta, Castellaneta, Francesca, Cesari, Gianluigi, Sabbà, Carlo, Fanizzi, Francesco Paolo, and Mazzocca, Antonio
- Subjects
chemistry.chemical_classification ,Multidisciplinary ,Biochemical networks ,biology ,Hepatocellular carcinoma ,Cell growth ,Cholesterol ,Science ,Pharmacology ,biology.organism_classification ,Cytostasis ,Warburg effect ,Article ,chemistry.chemical_compound ,NMR spectroscopy ,Biosynthesis ,chemistry ,Preclinical research ,Crithmum ,Medicine ,Polyunsaturated fatty acid ,Phosphocholine - Abstract
Hepatocellular carcinoma (HCC) is nowadays the sixth cause of tumour-related deceases worldwide, estimated to become the third in Western countries by 2030. New drugs for HCC treatment still have many adverse effects. Several lines of evidence indicate that plant metabolites offer concrete opportunities for developing new therapeutic strategies for many diseases, including cancer. We previously reported that ethyl acetate extract of a spontaneous edible plant harvested in Apulia, Crithmum maritimum, significantly inhibited cell growth in HCC cells. By 1H-NMR spectroscopy, here we show that Crithmum maritimum ethyl acetate extract counteracts the Warburg effect, by reducing intracellular lactate, inhibits protein anabolism, by decreasing amino acid level, and affects membrane biosynthesis by lowering choline and phosphocholine. Also, we observed an effect on lipid homeostasis, with a reduction in triglycerides, cholesterol, monounsaturated fatty acids (MUFA), and diunsaturated fatty acids (DUFA), and an increase in polyunsaturated fatty acids (PUFA). Taken together, these data demonstrate that Crithmum maritimum-induced cytostasis is exerted through a multi-effect action, targeting key metabolic processes in HCC cells. Overall, our findings highlight the role of Crithmum maritimum as a promising tool for the prevention and the improvement of the therapeutic options for HCC and other types of tumours.
- Published
- 2021
47. Development of an in vitro neuroblastoma 3D model and its application for sterigmatocystin-induced cytotoxicity testing
- Author
-
Maria Rosaria Esposito, María-José Ruiz, Luca Zanella, Veronica Zingales, Noemi Torriero, Mónica Fernández-Franzón, and Elisa Cimetta
- Subjects
endocrine system ,Cytotoxicity ,Sterigmatocystin ,Blotting, Western ,Cell ,Fluorescent Antibody Technique ,Toxicology ,3D cell culture ,chemistry.chemical_compound ,Neuroblastoma ,Cell Movement ,Cell Line, Tumor ,Spheroids, Cellular ,Mechanisms of action ,Toxicity Tests ,medicine ,Humans ,Cell Culture Techniques, Three Dimensional ,Viability assay ,Chemistry ,General Medicine ,Mycotoxins ,3D spheroid ,Cytostasis ,Cell biology ,medicine.anatomical_structure ,Cell culture ,Apoptosis ,Comet Assay ,Reactive Oxygen Species ,Food Science - Abstract
Given the increasing importance of establishing better risk assessments for mycotoxins, novel in vitro tools for the evaluation of their toxicity are mandatory. In this study, an in vitro 3D spheroid model from SH-SY5Y cells, a human neuroblastoma cell line, was developed, optimized and characterized to test the cytotoxic effects caused by the mycotoxin sterigmatocystin (STE). STE induced a concentration- and time-dependent cell viability decrease in spheroids. Spheroids displayed cell disaggregation after STE exposure, increasing in a dose-dependent manner and over time. STE also induced apoptosis as confirmed by immunofluorescence staining and Western blot. Following the decreased proliferation and increased apoptosis, STE cytostasis effects were observed by migration assays both in 2D and 3D cell culture. Increased ROS generation, as well as DNA damage were also observed. Taken together, these data highlight the cytotoxic properties of STE and suggest that cell culture models play a pivotal role in the toxicological risk assessment of mycotoxins. The evaluation of cytotoxicity in spheroids (3D) rather than monolayer cultures (2D) is expected to more accurately reflect in vivo-like cell behaviour.
- Published
- 2021
48. The search for plants with anticancer activity: Pitfalls at the early stages.
- Author
-
Taylor, P., Colman, L., and Bajoon, J.
- Subjects
- *
BIOLOGICAL assay , *IN vitro studies , *ANTINEOPLASTIC agents , *CELL death , *CELL physiology , *EXPERIMENTAL design , *RESEARCH methodology , *MEDICINAL plants , *PLANT extracts , *TREATMENT effectiveness , *PHARMACODYNAMICS - Abstract
Inhibition assays on tumour cells in vitro are commonly used to confirm the activity of extracts, fractions and compounds from plants reported to be antitumoural. The majority of assays report the IC 50 (50% inhibitory concentration), whereas others distinguish between inhibition of cell proliferation (cytostasis) and cell death (cytotoxicity). Here, we offer some suggestions as to the different types of assay, the cell lines that may be used, control cells and drugs, as well as the interpretation of the results. Using both theoretical considerations and experimental data, we specifically question the frequent overinterpretation of reported results regarding the selectivity for cancer cells of the plant extract or compound under study, concluding that this “selectivity” is due to a quantitative difference in cell proliferation rates, rather than a qualitative difference between normal and tumour cells. Inhibition assays will always represent one of the first steps in the discovery of clinically valuable new drugs, but these assays do not allow us to conclude that we have found the “magic bullet”. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
49. Expression of inducible NOS is indispensable for the antiproliferative and proapoptotic effect of imatinib in BCR-ABL positive cells
- Author
-
Sheela Nagarkoti, Tulika Chandra, Anil Kumar Tripathi, Abhishek Singh, Madhu Dikshit, Deepika Awasthi, Megha Dubey, and Manoj Kumar Barthwal
- Subjects
0301 basic medicine ,Adult ,Male ,Adolescent ,Immunology ,Fusion Proteins, bcr-abl ,Nitric Oxide Synthase Type II ,Antineoplastic Agents ,Apoptosis ,Biology ,03 medical and health sciences ,chemistry.chemical_compound ,Young Adult ,0302 clinical medicine ,hemic and lymphatic diseases ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,medicine ,Immunology and Allergy ,Humans ,Kinase activity ,neoplasms ,Protein Kinase Inhibitors ,Aged ,Cell Proliferation ,Myeloid leukemia ,NF-κB ,Imatinib ,Cell Biology ,Middle Aged ,Cytostasis ,030104 developmental biology ,chemistry ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,Imatinib Mesylate ,Female ,Tyrosine kinase ,K562 cells ,medicine.drug - Abstract
Chronic myeloid leukemia (CML) is characterized by constitutive BCR–ABL kinase activity, an aggressive proliferation of immature cells, and reduced differentiation. Targeting tyrosine kinase activity of BCR–ABL with imatinib is an effective therapy for the newly diagnosed CML patients; however, 20%–30% of the patients initially treated with imatinib eventually experience treatment failure. Therefore, early identification of these patients is of high clinical relevance. In the present study, we by undertaking a direct comparison of inducible NOS (iNOS) status in neutrophils from healthy volunteers, newly diagnosed, imatinib responder, and resistant CML patients as well as by conducting in vitro studies in K562 cells demonstrated that inhibition of BCR–ABL by imatinib or siRNA significantly enhanced NO generation and iNOS expression. Indeed, patients exhibiting treatment failure or imatinib resistance were less likely to induce NO generation/iNOS expression. Our findings further demonstrated that imatinib mediated antiproliferative and proapoptotic effect in BCR–ABL+ cells associated with enhanced iNOS expression, and it was significantly prevented in the presence of L-NAME, 1400W, or iNOS siRNA. Overexpression of iNOS in K562 cells expectedly enhanced imatinib sensitivity on cytostasis and apoptosis, even at lower concentration (0.1 μM) of imatinib. Mechanistically, imatinib or BCR–ABL siRNA following deglutathionylation of NF-κB, enhanced its binding to iNOS promoter and induced iNOS transcription. Deglutathionylation of procaspase-3 however associated with increased caspase-3 activity and cell apoptosis. Taken together, results obtained suggest that monitoring NO/iNOS level could be useful to identify patients likely to be responsive or resistant to imatinib and can be used to personalized alternative therapy.
- Published
- 2020
50. Novel Psychoactive Phenethylamines: Impact on Genetic Material
- Author
-
Micaela Tirri, Matteo Marti, Veronica Cocchi, Monia Lenzi, Patrizia Hrelia, Sofia Gasperini, Cocchi V., Gasperini S., Hrelia P., Tirri M., Marti M., and Lenzi M.
- Subjects
25B-NBOMe ,Apoptosis ,Phenethylamines ,Pharmacology ,medicine.disease_cause ,lcsh:Chemistry ,chemistry.chemical_compound ,Flow cytometry ,lcsh:QH301-705.5 ,Spectroscopy ,Micronucleus Tests ,Chemistry ,2C-B ,MDMA ,ROS ,General Medicine ,2C-I ,Cytostasis ,Computer Science Applications ,2C-H ,medicine.drug ,Cell Survival ,N-Methyl-3,4-methylenedioxyamphetamine ,Socio-culturale ,Anisoles ,Article ,Catalysis ,Cell Line ,Inorganic Chemistry ,medicine ,Humans ,Physical and Theoretical Chemistry ,Molecular Biology ,Micronuclei, Chromosome-Defective ,Psychotropic Drugs ,Organic Chemistry ,Acute toxicity ,Genes ,lcsh:Biology (General) ,lcsh:QD1-999 ,Dimethoxyphenylethylamine ,phenethylamine ,genotoxicity ,flow cytometry ,Hallucinogens ,Genotoxicity ,Reactive Oxygen Species ,Micronucleus - Abstract
Psychedelic and stimulating phenethylamines belong to the family of new psychoactive substances (NPS). The acute toxicity framework has begun to be investigated, while studies showing genotoxic potential are very limited or not available. Therefore, in order to fill this gap, the aim of the present work was to evaluate the genotoxicity by treating TK6 cells with 2C-H, 2C-I, 2C-B, 25B-NBOMe, and the popular 3,4-Methylenedioxymethylamphetamine (MDMA). On the basis of cytotoxicity and cytostasis results, we selected the concentrations (6.25&ndash, 35 µ, M) to be used in genotoxicity analysis. We used the micronucleus (MN) as indicator of genetic damage and analyzed the MNi frequency fold increase by an automated flow cytometric protocol. All substances, except MDMA, resulted genotoxic, therefore, we evaluated reactive oxygen species (ROS) induction as a possible mechanism at the basis of the demonstrated genotoxicity. The obtained results showed a statistically significant increase in ROS levels for all genotoxic phenethylamines confirming this hypothesis. Our results highlight the importance of genotoxicity evaluation for a complete assessment of the risk associated also with NPS exposure. Indeed, the subjects who do not have hazardous behaviors or require hospitalization by using active but still &ldquo, safe&rdquo, doses could run into genotoxicity and in the well-known long-term effects associated.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.