Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Lett ; 590: 216843, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38579893

ABSTRACT

Recurrent chemotherapy-induced senescence and resistance are attributed to the polyploidization of cancer cells that involve genomic instability and poor prognosis due to their unique form of cellular plasticity. Autophagy, a pre-dominant cell survival mechanism, is crucial during carcinogenesis and chemotherapeutic stress, favouring polyploidization. The selective autophagic degradation of essential proteins associated with cell cycle progression checkpoints deregulate mitosis fidelity and genomic integrity, imparting polyploidization of cancer cells. In connection with cytokinesis failure and endoreduplication, autophagy promotes the formation, maintenance, and generation of the progeny of polyploid giant cancer cells. The polyploid cancer cells embark on autophagy-guarded elevation in the expression of stem cell markers, along with triggered epithelial and mesenchymal transition and senescence. The senescent polyploid escapers represent a high autophagic index than the polyploid progeny, suggesting regaining autophagy induction and subsequent autophagic degradation, which is essential for escaping from senescence/polyploidy, leading to a higher proliferative phenotypic progeny. This review documents the various causes of polyploidy and its consequences in cancer with relevance to autophagy modulation and its targeting for therapeutic intervention as a novel therapeutic strategy for personalized and precision medicine.


Subject(s)
Autophagy , Cellular Senescence , Neoplasms , Neoplastic Stem Cells , Polyploidy , Humans , Cellular Senescence/drug effects , Neoplasms/pathology , Neoplasms/genetics , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/drug effects , Animals , Epithelial-Mesenchymal Transition
2.
Eur J Pharm Sci ; 147: 105299, 2020 Apr 30.
Article in English | MEDLINE | ID: mdl-32165315

ABSTRACT

The targeting of pro-inflammatory enzymes becomes a therapeutic intervention when acute inflammation is proliferating in pathological conditions. This research is intended to carry out an evaluation of inhibiting and inducing enzymes with inflammatory associations with 28 cyclohexanone analogs based on the ligustrazine. Tests were undertaken with inhibitor screening assay kits using a range of synthetic compounds to investigate how they could inhibit the activity of cyclooxygenase (COX) enzymes, secretory phospholipase A2 (sPLA2), and lipoxygenase (LOX) enzyme. Significant and similar inhibitory activities against sPLA2 with were noted with synthetic compounds which included 1f and 1g (IC50 = 2.2 µM). The optimal inhibitory activity regarding LOX enzyme was shown with compounds 1d (IC50 = 8.1 µM) and 1e (IC50 = 7.5 µM). Additionally, the compounds 1b, 1d, 1e, 2n, and 2o were shown to be significant inhibitors of COX-1 activity with IC50 values 0.09 to 0.7 µM. The outcomes of assays for COX inhibition demonstrated that the same compounds had a further strong inhibitive influence on the COX-2 enzyme, and certain compounds such as 1d, 1e, and 2n demonstrated enhanced potency compared with positive controls.


Subject(s)
Cyclohexanones/pharmacology , Inflammation/drug therapy , Pyrazines/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cyclohexanones/chemistry , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Humans , In Vitro Techniques , Lipoxygenase Inhibitors/pharmacology , Pyrazines/chemistry , Structure-Activity Relationship
3.
Saudi Pharm J ; 27(8): 1203-1209, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31885480

ABSTRACT

There are many mechanisms of resistance, chemoresistance of HeLa cells to anti-cancer agents seems to be autophagy-mediated. While using very effective anti-cancers such as Doxorubicin and cisplatin, cells overcome the cytotoxicity of these drugs through promotion of what so-called cytoprotective autophagy. Here in this study, we sought to introduce a novel platinum-based compound PBC-II that possesses anti-cancer activity. Our data showed that PBC-II is able to induce apoptosis at relatively low concentrations, with no detectable reactive oxygen species (ROS). However, further experiments demonstrated that exposure of HeLa cells to PBC-II did not promote autophagy; rather, it resulted in accumulation of p62 and decrease in LC3-II levels. Autophagy was then promoted in HeLa cells pharmacologically by Doxorubicin and genetically by siRNA IL-10. In order to confirm promotion of autophagy in our model, we performed acridine orange staining to assess for autophagy under microscope as well as via flow cytometry. We then measured protein level of autophagy markers p62 and LC3 by western blot. Our data indicated that PBC-II interferes with therapy-induced autophagy. We also determined PI3K activity while co-incubation of PBC-II with autophagy inducers. It was clear that PI3K activation decreased when PBC-II was co-administered with autophagy inducers. Collectively, PBC-II exerts unique anti-proliferative effects associated with inhibition of autophagy, which indicates that PBC-II is potentially a promising agent to be used in resistant ovarian tumors.

5.
Inflammopharmacology ; 27(4): 817-827, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30600471

ABSTRACT

BACKGROUND: Carfilzomib (CFZ), a proteasome inhibitor approved by the FDA to treat multiple myeloma, may cause nephrotoxicity. HYPOTHESIS: Rutin is a bioflavonoid with antioxidant properties. We aimed to examine whether rutin protects the kidney from CFZ-induced nephrotoxicity. STUDY DESIGN: This study aimed to demonstrate the effect of rutin on CFZ-induced renal injury via the inhibition of oxidative stress and inflammation. METHODS: Wistar albino rats were divided into six groups (n = 6): Group 1 (normal control; NC) was administered normal saline for 3 weeks; Group 2 (CFZ/toxic group) received CFZ [4 mg/kg, intraperitoneal (i.p.) injection] twice weekly for 3 weeks; Group 3 (standard treatment group) was administered CFZ (4 mg/kg, i.p.) and olmesartan (2 mg/kg, p.o.) for 3 weeks; Group 4 was administered CFZ (4 mg/kg, i.p.) and rutin (10 mg/kg, p.o.) for 3 weeks; Group 5 was administered CFZ (4 mg/kg, i.p.) and rutin (20 mg/kg, p.o.) for 3 weeks; and Group 6 was administered CFZ (4 mg/kg, i.p.) and rutin (40 mg/kg, p.o.) for 3 weeks. We carried out haematological and biochemical analyses, determined oxidative stress, caspase-3 activity, and protein levels, and performed a histopathological evaluation to confirm CFZ-induced nephrotoxicity and its prevention by rutin administration. RESULTS: Exposure to only CFZ significantly (p < 0.05) increased white blood cell (WBC) count, Hb%, and HTC% concentration; however, these features were significantly decreased (p < 0.05) when olmesartan and rutin were administered. CFZ administration significantly decreased (p < 0.0001) the level of antioxidant enzymes; whereas, administration of olmesartan and rutin significantly reversed (p < 0.05) their levels toward the normal range. The levels of caspase-3 enzyme significantly increased (p < 0.001) in the CFZ group and were reduced toward the normal values by olmesartan and rutin administration. Furthermore, the results of NOS-2, NF-κB, IkBa, and IL-17 protein estimation and the histopathological evaluation strengthened our findings that rutin exhibits a protective effect against CFZ-induced nephrotoxicity. CONCLUSION: These findings clearly demonstrate that rutin ameliorates CFZ-induced oxidative stress and inflammation in nephrotoxicity via the NOS-mediated NF-κB signaling pathway.


Subject(s)
Inflammation/drug therapy , Nitric Oxide Synthase/metabolism , Oligopeptides/pharmacology , Oxidative Stress/drug effects , Rutin/pharmacology , Signal Transduction/drug effects , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Caspase 3/metabolism , Flavonoids/pharmacology , Imidazoles/pharmacology , Inflammation/metabolism , Kidney/drug effects , Kidney/metabolism , Male , NF-kappa B/metabolism , Rats , Rats, Wistar , Tetrazoles/pharmacology
6.
Pharmacol Rep ; 70(5): 993-1000, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30118964

ABSTRACT

BACKGROUND: Doxorubicin is an effective, potent and commonly used anthracycline-related anticancer drug; however, cardiotoxicity compromises its therapeutic potential. Apremilast, a novel phosphodiesterase type 4-inhibitor, reported to have anti-inflammatory effects and modulating many inflammatory mediators. METHODS: The present study investigated the influence of apremilast against doxorubicin-induced cardiotoxicity in male Wistar rats. A total, 24 animals were divided into four groups of six animal each. Group 1, served as control and received normal saline. Group 2 animals, received doxorubicin (20mgkg-1, ip). Group 3 and 4, treatment group, received doxorubicin (20mgkg-1, ip) with the same schedule as group-2, plus apremilast (10 and 20mgkg-1day-1, po) respectively. Oxidative stress, caspase-3 enzyme activity, gene expression and protein expression were tested. RESULTS: The results of the present study demonstrated that administration of apremilast reversed doxorubicin-induced cardiotoxicity. CONCLUSION: These findings suggested that apremilast can attenuate doxorubicin-induced cardiotoxicity via inhibition of oxidative stress mediated activation of nuclear factor-kappa B signaling pathways.


Subject(s)
Apoptosis/drug effects , Doxorubicin/adverse effects , Doxorubicin/antagonists & inhibitors , Inflammation/chemically induced , NF-kappa B/metabolism , Oxidative Stress/drug effects , Signal Transduction/drug effects , Thalidomide/analogs & derivatives , Animals , Cardiotoxicity/prevention & control , Caspase 3/metabolism , Catalase/metabolism , Dose-Response Relationship, Drug , Glutathione/metabolism , Glutathione Reductase/metabolism , Male , Malondialdehyde/metabolism , Myocardium/metabolism , RNA, Messenger/biosynthesis , Rats , Thalidomide/pharmacology
7.
BMC Complement Altern Med ; 17(1): 472, 2017 Sep 29.
Article in English | MEDLINE | ID: mdl-28962559

ABSTRACT

BACKGROUND: Cisplatin is widely used chemotherapeutic agent for cancer treatment with limited uses due to its neurotoxic side effect. The aim of this study was to determine the potential preventive effects of rutin on the brain of cisplatin- neurotoxic rat model. METHODS: Forty rats were divided into four groups. Group-1 (control group) was intra-peritoneal (IP) injected with 2.5 ml/kg saline. Group-2 (rutin group) was orally administrated 30 mg/kg rutin dissolved in water for 14 days. Group-3 (cisplatin group) was IP received 5 mg/kg cisplatin single dose. Group-4 (rutin and cisplatin group) was orally administrated 30 mg/kg rutin dissolved in water for 14 days with a single dose of 5 mg/kg cisplatin IP on day ten. Brain tissues from frontal cortex was used to extract RNA, the gene expression levels of paraoxonase-1 (PON-1), PON-2, PON-3, peroxisome proliferator-activated receptor delta (PPAR-δ), and glutathione peroxidase (GPx) was investigated by Real-time PCR. RESULTS: Cisplatin significantly decreased the expression levels of PON-1, PON-3, PPAR-δ and GPX whereas significantly increased PON-2 expression levels. Co-administration of Rutin prevented the cisplatin-induced toxicity by restoring the alteration in the studied genes to normal values as in the control group. CONCLUSION: This study showed that Rutin has neuroprotective effect and reduces cisplatin- neurotoxicity with possible mechanism via the antioxidant pathway.


Subject(s)
Brain/drug effects , Cisplatin/adverse effects , Neuroprotective Agents/pharmacology , Rutin/pharmacology , Animals , Body Weight/drug effects , Brain/enzymology , Brain/metabolism , Brain Chemistry/drug effects , Glutathione/metabolism , Glutathione Peroxidase/analysis , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Male , Oxidative Stress/drug effects , PPAR delta/analysis , PPAR delta/genetics , PPAR delta/metabolism , Rats , Rats, Wistar , Thiobarbituric Acid Reactive Substances/metabolism
8.
Asian Pac J Cancer Prev ; 17(11): 4965-4971, 2016 11 01.
Article in English | MEDLINE | ID: mdl-28032724

ABSTRACT

Objective: Breast cancer is global female health problem worldwide. Most of the currently used agents for breast cancer treatment have toxic side-effects. Ginseng root, an oriental medicine, has many health benefits and may exhibit direct anti-cancer properties. This study was performed to assess the effects of ginseng on breast cancer cell lines. Materials and Methods: Cytotoxicity of ginseng extract was measured by MTT assay after exposure of MDA-MB-231, MCF-10A and MCF-7 breast cancer cells to concentrations of 0.25, 0.5, 1, 1.5, 2 and 2.5 mg/well. Expression levels of p21WAF, p16INK4A, Bcl-2, Bax and P53 genes were analyzed by quantitative real time PCR. Results: The treatment resulted in inhibition of cell proliferation in a dose-and time-dependent manner. p53, p21WAF1and p16INK4A expression levels were up-regulated in ginseng treated MDA-MB-231 and MCF-7 cancer cells compared to untreated controls and in MCF-10A cells. The expression levels of Bcl2 in the MDA-MB-231 and MCF-7 cells were down-regulated. In contrast, that of Bax was significantly up-regulated. Conclusion: The results of this study revealed that ginseng may inhibit breast cancer cell growth by activation of the apoptotic pathway.

SELECTION OF CITATIONS
SEARCH DETAIL
...