Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 881
Filter
1.
Anticancer Res ; 43(3): 1139-1148, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36854534

ABSTRACT

BACKGROUND/AIM: Primary effusion lymphoma (PEL) is a rare aggressive B-cell lymphoma associated with HHV-8. With a median survival of fewer than six months, the prognosis of the disease with current standard therapies is usually dismal. Dihydroartemisinin (DHA) is a derivative of artemisinin, originally designed as an antimalarial drug. Several studies have shown that this compound also demonstrates anti-cancer activity in various types of cancer, including hematologic malignancies. MATERIALS AND METHODS: Anti-proliferation activity of DHA on 5 PEL cell lines was assessed by MTT assay. Cell cycle arrest was determined by propidium iodide staining and flow cytometry analysis. DHA-induced PEL apoptosis was shown by annexin V/PI staining and western blotting for cleaved caspases 3, 8, and 9. An inhibitory effect on PEL growth was evaluated in a PEL-xenograft mouse model. A synergistic effect of DHA and doxorubicin combination treatment was shown in vitro. RESULTS: DHA showed anti-proliferative activity on PEL and induced caspase-dependent apoptosis in a time- and dose-dependent manner. DHA-induced cell death appeared to be triggered by increased levels of reactive oxygen species (ROS). N-acetylcysteine treatment inhibited DHA-induced ROS elevation and suppressed expression of cleaved caspases leading to significantly reduced PEL apoptosis. DHA treatment also demonstrated an inhibitory effect on PEL cell growth in an in-vivo xenograft model. Moreover, we found that a combination treatment of DHA and doxorubicin, the standard chemotherapy drug for PEL, demonstrated a synergistic effect on PEL cell lines. CONCLUSION: DHA is a potentially effective candidate drug for PEL treatment.


Subject(s)
Artemisinins , Lymphoma , Pleural Effusion, Malignant , Animals , Humans , Mice , Apoptosis/drug effects , Caspases/drug effects , Lymphoma/drug therapy , Reactive Oxygen Species , Artemisinins/pharmacology , Artemisinins/therapeutic use , Pleural Effusion, Malignant/drug therapy , Pleural Effusion, Malignant/metabolism , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
2.
Int J Mol Sci ; 23(3)2022 Jan 25.
Article in English | MEDLINE | ID: mdl-35163269

ABSTRACT

A growing body of evidence indicates that dietary polyphenols show protective effects against various cancers. However, little is known yet about their activity in brain tumors. Here we investigated the interaction of dietary flavonoid quercetin (QCT) with the human glioblastoma A172 and LBC3 cell lines. We demonstrated that QCT evoked cytotoxic effect in both tested cell lines. Microscopic observations, Annexin V-FITC/PI staining, and elevated expression and activity of caspase 3/7 showed that QCT caused predominantly apoptotic death of A172 cells. Further analyses confirmed enhanced ROS generation, deregulated expression of SOD1 and SOD2, depletion of ATP levels, and an overexpression of CHOP, suggesting the activation of oxidative stress and ER stress upon QCT exposure. Finally, elevated expression and activity of caspase 9, indicative of a mitochondrial pathway of apoptosis, was detected. Conversely, in LBC3 cells the pro-apoptotic effect was observed only after 24 h incubation with QCT, and a shift towards necrotic cell death was observed after 48 h of treatment. Altogether, our data indicate that exposure to QCT evoked cell death via activation of intrinsic pathway of apoptosis in A172 cells. These findings suggest that QCT is worth further investigation as a potential pharmacological agent in therapy of brain tumors.


Subject(s)
Glioblastoma/drug therapy , Oxidative Stress/drug effects , Quercetin/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Brain Neoplasms/metabolism , Caspases/drug effects , Caspases/metabolism , Cell Line, Tumor , Endoplasmic Reticulum Stress/drug effects , Flavonoids/pharmacology , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Glioblastoma/metabolism , Humans , Mitochondria/metabolism , Oxidative Stress/genetics , Preliminary Data , Quercetin/metabolism , Reactive Oxygen Species/metabolism , Stress, Physiological/drug effects , Stress, Physiological/genetics
3.
Mol Biol Rep ; 49(2): 1351-1360, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34806141

ABSTRACT

BACKGROUND: We investigated the apoptotic effects of curcumin in the colon carcinoma cell line SW480. METHODS AND RESULTS: Cells were treated with 40-200 µM curcumin for 24, 48, and 72 h, and the IC50 values were determined for each time interval. BrdU, caspase-3, and TUNEL staining results and the gene expression of FADD, CASP8, and CASP3 were evaluated. Curcumin treatments significantly inhibited cell proliferation and significantly induced apoptosis for 24, 48, and 72 h. The proportion of BrdU-stained cells in the control groups were 58%, 57% and 61% and 28%, 27%, and 30% in the curcumin treatment groups at 24, 48, and 72 h, respectively. The proportion of apoptotic cells was 28%, 29%, and 28% in the control groups and 59%, 61%, and 60% in the curcumin treatment groups at 24, 48, and 72 h, respectively. As expected, caspase-3 staining also revealed a higher number of apoptotic cells in curcumin treatment groups at 24, 48, and 72 h compared to controls. The proportion of Caspase-3-stained cells in the control groups were 23%, 25%, and 24% and 59%, 60%, and 62% in the curcumin treatment groups at 24, 48, and 72 h, respectively. To prove caspase-3 staining results, FADD, CASP8, and CASP3 gene expressions were evaluated by real-time qPCR. Unlike the immunohistochemical results, no statistically significant upregulation was found at 24 and 48 h, while relative gene expressions of FADD, CASP8, and CASP3 was significantly upregulated at 72 h. The expression level increase was 0.88-, 1.19-, and 2.11-fold for FADD, 1.25-, 1.29-, and 1.59-fold for CASP8, and 1.33-, 1.46-, and 3.00-fold for CASP3 at 24, 48, and 72 h, respectively. CONCLUSIONS: These results suggest that curcumin may be a potential protective or treatment agent against colon cancer; however, further studies on curcumin-rich diets and curcumin bioavailability are required.


Subject(s)
Apoptosis/drug effects , Colonic Neoplasms/metabolism , Curcumin/pharmacology , Apoptosis/physiology , Carcinoma , Caspase 3 , Caspase 8 , Caspases/drug effects , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Colon/metabolism , Colonic Neoplasms/drug therapy , Humans
4.
Biomed Pharmacother ; 146: 112567, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34953392

ABSTRACT

Curcumin, a phytochemical derived from the rhizome of turmeric (Curcuma longa L.), has a broad group of substances with antibacterial, anti-inflammatory, anti-oxidant, anticancer activities. The anticancer activity of curcumin and its derivatives are mainly related to its regulation of signal transduction pathways. However, due to the low oral availability of curcumin, fast metabolism and other pharmacokinetic properties limit the application of curcumin in the treatment of cancer. Evidence suggests that curcumin combined with photodynamic therapy can overcome the limitation of curcumin's low bioavailability by acting on apoptosis pathways, such as B-cell lymphoma 2 (Bcl-2) and caspase family, and affecting cell cycle. This paper reviews the structure and pharmacokinetics of curcumin, focusing on the anticancer activity of curcumin combined with photodynamic therapy and the effects on cancer-related signal pathways.


Subject(s)
Antineoplastic Agents/pharmacology , Curcumin/pharmacology , Neoplasms/pathology , Photochemotherapy/methods , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Caspases/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Curcumin/chemistry , Curcumin/pharmacokinetics , Humans , Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/drug effects
5.
Drug Deliv ; 28(1): 2480-2494, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34766543

ABSTRACT

Cisplatin (DDP) is a first-line chemotherapeutic drug applied for the treatment of oral squamous cell carcinoma (OSCC). The anticancer activity of DDP is tightly linked to its intracellular uptake. It is unwise to increase the DDP intake by increasing the dose or shortening the dosing interval because of the severe systemic toxicity (nephrotoxicity, ototoxicity and neurotoxicity) in DDP application. The main uptake pathways of DDP include passive diffusion and active transporter transport. Therefore, finding additional uptake pathways that can improve the effective intracellular concentration of DDP is critical. Macropinocytosis, an endocytic mechanism for extracellular material absorption, contributes to the intracellular uptake of anticancer drugs. No research has been conducted to determine whether macropinocytosis can augment the intracellular uptake of DDP in OSCC cells or not. Based on that, we proved for the first time that silmitasertib (previously CX-4945) could trigger macropinocytosis, which may increase the intracellular uptake of DDP and enhance apoptosis via in vivo and in vitro experiments. We hope that our findings will inspire a new approach for the application of DDP in cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Naphthyridines/pharmacology , Phenazines/pharmacology , Pinocytosis/drug effects , Animals , Apoptosis/drug effects , Carcinoma, Squamous Cell/pathology , Caspases/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cisplatin/pharmacokinetics , Drug Liberation , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mitogen-Activated Protein Kinases/drug effects , Mouth Neoplasms/pathology , Xenograft Model Antitumor Assays
6.
Arch Insect Biochem Physiol ; 108(3): e21844, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34519097

ABSTRACT

Animals maintain homeostasis of cell numbers, constantly creating new cells and eliminating others. Programmed cell death, apoptosis, is a mechanism of cell elimination and it acts in many aspects of animal biology. Drawing on the biomedical background, several signals launch the apoptosis mechanisms, including prostaglandins (PGs). Based on this information, we posed the hypothesis that PGs similarly induce apoptosis in insect cell lines. We used three Spodoptera frugiperda cell lines, including two newly established, BCIRL-SfNS-0518B-YL derived from the central nervous system and BCIRL-Sf4FB-0614-SGS derived from fat body, and the commercially available Sf9 cells. Using a kinetic apoptosis kit, we found treating SfNS cells for 18 h with 15 or 20 µM PGA2 led to decreases in cell numbers, coupled with increased numbers of apoptotic and dead cells. Similar exposures to 10 µM PGA2 (24 h) led to substantial increases in apoptotic cells, confirmed by a terminal deoxynucleotidyl transferase dUTP nick end labeling assay on a flow cytometer. The influence of PGA2 treatments increased with dosage, as we recorded about 20% apoptosis at 24 h post-PGA2 treatments (10 µM) and about 34% apoptosis at 24 h post-30 µM treatments. PGA2 treatments led to 10- to 30-fold increases in messenger RNAs (mRNAs) encoding apoptosis-specific caspases-1, -2, -3, and -5 at 12 h and 40- to 60-fold increases in mRNAs encoding caspases-1 and -2, 10-fold increases for caspases-3 and -5 at 24 h. These findings strongly support our hypothesis that PGs induce apoptosis in an insect cell line and confirm an additional PG action in insect biology.


Subject(s)
Caspases , Prostaglandins A/pharmacology , Sf9 Cells/drug effects , Animals , Apoptosis/drug effects , Caspases/drug effects , Caspases/metabolism , Spodoptera/metabolism
7.
J Chemother ; 33(8): 582-594, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34060437

ABSTRACT

Eight novel ERß selective daidzein analogues (NCE1-8) were synthesized and their anti-cancer activity was evaluated by in vitro and in vivo methods. Cytotoxicity study, Receptor binding studies, Luciferase assay, cMYC & Cyclin D1 expression and Caspase 3, 8 & 9 activities were measured to ascertain the anticancer activity and mechanism. Uterotropic, anti-androgenic and anti-tumour activities were performed in rodents. The results revealed that NCEs produced anti-prostate cancer activity in DU145, LNCaP and PC3 cell lines and 50% more active than genistein. NCEs was significantly down-regulated cMYC & Cyclin D1 genes and elevated caspase 3 & 9 levels and did not show any difference in uterotropic, anti-androgenic activities. The tumour weight was also reduced. The NCE 1 and 2 have shown ERß selectivity in receptor binding studies. Daidzein with methyl substitution at R or R1 position exhibited more ERß selectivity and could be considered as lead molecules for anti-prostate cancer activity.


Subject(s)
Estrogen Receptor beta/drug effects , Isoflavones/pharmacology , Prostatic Neoplasms/pathology , Animals , Caspases/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Cyclin D1/drug effects , Dose-Response Relationship, Drug , Female , Genistein/pharmacology , Humans , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
8.
J Cereb Blood Flow Metab ; 41(4): 857-873, 2021 04.
Article in English | MEDLINE | ID: mdl-33736511

ABSTRACT

Post-stroke neurological deficits and mortality are often associated with vascular disruption and neuronal apoptosis. Galectin-3 (Gal3) is a potent pro-survival and angiogenic factor. However, little is known about its protective role in the cerebral ischemia/reperfusion (I/R) injury. We have previously shown significant up-regulation of Gal3 in the post-stroke rat brain, and that blocking of Gal3 with neutralizing antibody decreases the cerebral blood vessel density. Our current study demonstrates that intracerebral local delivery of the Gal3 into rat brain at the time of reperfusion exerts neuroprotection. Ischemic lesion volume and neuronal cell death were significantly reduced as compared with the vehicle-treated MCAO rat brains. Gal3 increased vessel density and neuronal survival after I/R in rat brains. Importantly, Gal3-treated groups showed significant improvement in motor and sensory functional recovery. Gal3 increased neuronal cell viability under in vitro oxygen-glucose deprivation conditions in association with increased phosphorylated-Akt, decreased phosphorylated-ERK1/2, and reduced caspase-3 activity. Gene expression analysis showed down regulation of pro-apoptotic and inflammatory genes including Fas-ligand, and upregulation of pro-survival and pro-angiogenic genes including Bcl-2, PECAM, and occludin. These results indicate a key role for Gal3 in neuro-vascular protection and functional recovery following ischemic stroke through modulation of angiogenic and apoptotic pathways.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Apoptosis/drug effects , Caspases/drug effects , Galectin 3/therapeutic use , Ischemic Stroke/prevention & control , Neuroprotective Agents/pharmacology , Proto-Oncogene Proteins c-akt/drug effects , Signal Transduction/drug effects , Animals , Brain , Cell Death/drug effects , Galectin 3/administration & dosage , Gene Expression/drug effects , Hypoxia, Brain/drug therapy , Microinjections , Neurons/drug effects , Neurons/pathology , Rats , Rats, Inbred SHR , Reperfusion Injury/prevention & control
9.
Tissue Cell ; 70: 101502, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33582552

ABSTRACT

The probable beneficial effects of mesenchymal stem cells (MSCs) and resveratrol were assessed in an experimental model of Bisphenol-A (BPA)-evident uterine damage in rats. Thirty-five albino rats were involved and equally divided into five groups: Group I: negative control rats received usual diet, Group II: positive control rats received BPA by oral gavage for 15 days, Group III: BPA-treated rats received single oral gavage of resveratrol daily for two weeks, Group IV: BPA-treated rats received a single intravenous dose of MSCs and Group V: BPA-treated rats received combined treatment of resveratrol and MSCs. Oxidative stress markers, apoptosis-related genes, and gonadal hormones were assessed. Histological and immunohistochemical examination of uterine tissue was conducted for TGF-ß 1. Caspases-3, 8, and 9 (Casp3, Casp8, Casp9) genes were assessed in uterine tissues by quantitative real-time PCR. Results revealed that BPA induced significant changes in the endometrial tissue, inflammatory cell infiltration, focal blood extravasation, increase in collagen fibers, decrease in PAS staining, and increase in TGF-ß 1 immunoreactivity. BPA also induced a significant increase in oxidative stress markers; malondialdehyde (MDA), SOD, CAT, and apoptosis-related genes. BPA induced a significant change in blood levels of gonadal hormones; a significant increase in FSH and a significant decrease in estradiol (E2) and progesterone (P). Treatment with either resveratrol, MSCs, or a combination of them resulted in significant enhancement of histological findings, restoration of gonadal hormones to near-normal levels, and a significant decrease in oxidative stress markers and apoptosis genes. Combined treatment with resveratrol and MSCs demonstrated more significant therapeutic effects as regard to the studied parameters in association with rat groups treated with either MSCs or resveratrol separately.


Subject(s)
Endometrium , Mesenchymal Stem Cell Transplantation , Resveratrol/pharmacology , Uterus , Animals , Apoptosis/drug effects , Benzhydryl Compounds/toxicity , Biomarkers/analysis , Caspases/analysis , Caspases/drug effects , Endometrium/drug effects , Endometrium/pathology , Female , Gonadal Hormones/analysis , Mesenchymal Stem Cells/metabolism , Models, Animal , Oxidative Stress/drug effects , Phenols/toxicity , Rats , Resveratrol/therapeutic use , Transforming Growth Factor beta/analysis , Transforming Growth Factor beta/drug effects , Uterus/drug effects , Uterus/pathology
10.
Apoptosis ; 26(3-4): 184-194, 2021 04.
Article in English | MEDLINE | ID: mdl-33515314

ABSTRACT

Previously we have shown inhibition of endometrial cancer cell growth with progesterone and calcitriol. However, the mechanisms by which the two agents attenuate proliferation have not been well characterized yet. Herein, we investigated how progesterone and calcitriol induce apoptosis in cancer cells. DNA fragmentation was upregulated by progesterone and calcitriol in ovarian and endometrial cancer cells. Time-dependent treatment of ovarian cancer cells, ES-2, and TOV-21G with progesterone enhanced caspase -8 activity after 12 h, whereas OV-90, TOV-112D, HEC-1A, and HEC-59 cells showed increased activity after 24 h. Caspase 9 activity was increased in all cell lines after 24 h treatment with calcitriol. Pretreatment of cancer cells with a caspase-8 inhibitor (z-IETD-fmk) or caspase-9 inhibitor (Z-LEHD-fmk) significantly attenuated progesterone and calcitriol induced caspase-8 and caspase-9 expression, respectively. The expression of FasL, Fas, FAD, and pro-caspase-8, which constitute the death-inducing signaling complex (DISC), was upregulated in progesterone treated cancer cells. Knockdown of FAS or FADD with specific siRNAs significantly blocked progesterone-induced caspase-8. Cleavage of the BID was not affected by caspase-8 activation suggesting the absence of cross-talk between caspase-8 and caspase-9 pathways. Calcitriol treatment decreased mitochondrial membrane potential and increased the release of cancer cytochrome C. These findings indicate that progesterone induces apoptosis through activation of caspase-8 and calcitriol through caspase-9 activation in cancer cells. A combination of progesterone-calcitriol activates both extrinsic and intrinsic apoptotic pathways in cancer cells.


Subject(s)
Apoptosis/drug effects , Caspases , Endometrial Neoplasms/metabolism , Ovarian Neoplasms/metabolism , Progesterone/pharmacology , Calcitriol/metabolism , Caspase 8/drug effects , Caspase 8/metabolism , Caspase 9/drug effects , Caspase 9/metabolism , Caspases/drug effects , Caspases/metabolism , Cell Line, Tumor , Cytochromes c/drug effects , Cytochromes c/metabolism , Death Domain Receptor Signaling Adaptor Proteins/drug effects , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Death Domain Superfamily/drug effects , Endometrial Neoplasms/drug therapy , Fas Ligand Protein/drug effects , Fas Ligand Protein/metabolism , Female , Humans , In Vitro Techniques , Membrane Potential, Mitochondrial/drug effects , Ovarian Neoplasms/drug therapy , Signal Transduction/drug effects , fas Receptor/drug effects , fas Receptor/metabolism
11.
Invest New Drugs ; 39(1): 111-121, 2021 02.
Article in English | MEDLINE | ID: mdl-32885355

ABSTRACT

Primary effusion lymphoma (PEL), caused by Kaposi's sarcoma-associated herpesvirus (KSHV), presents as a lymphomatous effusion in body cavities and has a poor prognosis. The anti-malaria drug, artesunate, possesses anti-neoplastic potential. Therefore, we aimed to investigate its effect on KSHV-infected PEL cell lines. Artesunate inhibited cell growth and viability of PEL cells, but its effect on peripheral blood mononuclear cells was less pronounced. Artesunate induced G1 phase arrest by downregulating cyclin D1/D2, CDK2/6 and c-Myc. Artesunate increased reactive oxygen species and DNA damage, but did not affect the expression of latent and lytic genes of KSHV. It exhibited cytotoxicity through caspase-dependent and -independent pathways and reduced Bcl-xL, survivin, XIAP and c-IAP1/2 levels. Furthermore, artesunate suppressed NF-κB and AP-1 by inhibiting IκB kinase and IκBα phosphorylation as well as JunB expression. Finally, artesunate treatment attenuated PEL development in mice. Our data support that artesunate is a potential drug for PEL treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Artesunate/pharmacology , Herpesvirus 8, Human/drug effects , Lymphoma, Primary Effusion/pathology , Animals , Apoptosis/drug effects , Caspases/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Damage/drug effects , Female , Herpesvirus 8, Human/genetics , Humans , I-kappa B Kinase/drug effects , Mice , Mice, SCID , NF-KappaB Inhibitor alpha/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
12.
Invest New Drugs ; 39(1): 98-110, 2021 02.
Article in English | MEDLINE | ID: mdl-32856275

ABSTRACT

Two novel chemotherapeutic chalcones were synthesized and their structures were confirmed by different spectral tools. Theoretical studies such as molecular modeling were done to detect the mechanism of action of these compounds. In vitro cytotoxicity showed a strong effect against all tested cell lines (MCF7, A459, HepG2, and HCT116), and low toxic effect against normal human melanocytes (HFB4). The lung carcinoma cell line was chosen for further molecular studies. Real-time PCR demonstrated that the two compounds upregulated gene expression of (BAX, p53, casp-3, casp-8, casp-9) genes and decreased the expression of anti-apoptotic genes bcl2, CDK4, and MMP1. Flow-cytometry indicated that cell cycle arrest of A459 was induced at the G2/M phase and the apoptotic percentage increased significantly compared to the control sample. Cytochrome c oxidase and VEGF enzyme activity were detected by ELISA assay. SEM tool was used to follow the morphological changes that occurred on the cell surface, cell granulation, and average roughness of the cell surface. The change in the number and morphology of mitochondria, cell shrinkage, increase in the number of cytoplasmic organelles, membrane blebbing, chromatin condensation, and apoptotic bodies were observed using TEM. The obtained data suggested that new chalcones exerted their pathways on lung carcinoma through induction of two pathways of apoptosis. Graphical abstract Novel chalcones were prepared and confirmed by different spectral tools. Docking simulations were done to detect the mechanism of action. In vitro cytotoxicity indicated a strong effect against different cancer cell lines and low toxic effects against normal human melanocytes (HFB4). The lung carcinoma cell line was chosen for further molecular studies that include Real-time PCR, Flow-cytometry, Cytochrome c oxidase, and ELISA assay. SEM and TEM tool were used to follow the morphological changes occurred on the cell surface.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Chalcones/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Caspases/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chalcones/chemistry , Gene Expression/drug effects , Humans , Molecular Docking Simulation , Tumor Suppressor Protein p53/drug effects , bcl-2-Associated X Protein/drug effects
13.
Toxicol Ind Health ; 36(11): 844-851, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32909914

ABSTRACT

Di-2-ethylhexyl phthalate (DEHP) has been widely used as a plasticizer in industry and can affect memory; however, the underlying mechanism remains unclear. In the present study, mouse HT22 cells, an immortalized hippocampal neuronal cell line, was utilized as an in vitro model. We showed that DEHP dramatically inhibited cell viability and increased lactate dehydrogenase (LDH) release from the cells in a dose-dependent manner, suggesting that DEHP could cause cytotoxicity of mouse HT22 cells. The protein levels of cleaved Caspase-8, cleaved Caspase-3, and Bax markedly increased in the DEHP-treated cells, whereas there was a significant decrease in the Bcl-2 protein level, implying that DEHP could induce apoptosis of mouse HT22 cells. DEHP exposure significantly increased the content of malondialdehyde, whereas it markedly decreased the level of glutathione and the activities of glutathione peroxidase and superoxide dismutase, suggesting that DEHP induced oxidative stress of the cells. Compared with the DEHP-treated group, the inhibition of cell viability and the release of LDH were rescued in the N-acetyl-l-cysteine plus DEHP group. Furthermore, inhibition of oxidative stress could rescue the induction of apoptosis by DEHP. Collectively, our results indicated that DEHP could induce apoptosis of mouse HT22 cells via oxidative stress.


Subject(s)
Apoptosis/drug effects , Diethylhexyl Phthalate/toxicity , Neurons/drug effects , Oxidative Stress/drug effects , Animals , Caspases/drug effects , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Glutathione Peroxidase/antagonists & inhibitors , Hippocampus/drug effects , L-Lactate Dehydrogenase/biosynthesis , Mice , Proto-Oncogene Proteins c-bcl-2 , Superoxide Dismutase/antagonists & inhibitors
14.
J Nat Prod ; 83(10): 3050-3057, 2020 10 23.
Article in English | MEDLINE | ID: mdl-32955260

ABSTRACT

Plants in the family Aristolochiaceae contain phenanthrene skeleton-containing chemical constituents that exhibit nephrotoxic, carcinogenic, mutagenic, anti-inflammatory, and cytotoxic effects. Two new phenanthrene-containing 1,2-oxazin-6-ones, designated as asaroidoxazine A (1) and asaroidoxazine B (2), and a known aristolactam, 5-methoxyaristololactam I (3), were isolated from the roots of Asarum asaroides. The structures of compounds 1 and 2 were determined using spectroscopic methods and X-ray crystallography. Treatment of SH-SY5Y human neuroblastoma cells with 1 µM of asaroidoxazine A (1) induced nuclear condensation as well as caspase-3/7 activation, indicating that this compound is a strong apoptosis inducer in neuronal cells. This is the first report of apoptosis induction by phenanthrene-containing oxazines.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Asarum/chemistry , Brain Neoplasms/drug therapy , Neuroblastoma/drug therapy , Plant Roots/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Caspases/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Enzyme Activation/drug effects , Humans , Molecular Structure , Phenanthrenes/chemistry , Phenanthrenes/pharmacology , X-Ray Diffraction
15.
Pharmacol Res ; 160: 105058, 2020 10.
Article in English | MEDLINE | ID: mdl-32619722

ABSTRACT

Despite the discovery of tyrosine kinase inhibitors (TKIs) for the treatment of breakpoint cluster region-Abelson (BCR-ABL)+ cancer types, patients with chronic myeloid leukemia (CML) treated with TKIs develop resistance and severe adverse effects. Combination treatment, especially with a histone deacetylase (HDAC) 6 inhibitor (HDAC6i), appears to be an attractive option to prevent TKI resistance, considering the potential capacity of an HDAC6i to diminish BCR-ABL expression. We first validated the in vivo anti-cancer potential of the compound 7b by significantly reducing the tumor burden of BALB/c mice xenografted with K-562 cells, without notable organ toxicity. Here, we hypothesize that the HDAC6i compound 7b can lead to BCR-ABL downregulation in CML cells and sensitize them to TKI treatment. The results showed that combination treatment with imatinib and 7b resulted in strong synergistic caspase-dependent apoptotic cell death and drastically reduced the proportion of leukemia stem cells, whereas this treatment only moderately affected healthy cells. Ultimately, the combination significantly decreased colony formation in a semisolid methylcellulose medium and tumor mass in xenografted zebrafish compared to each compound alone. Mechanistically, the combination induced BCR-ABL ubiquitination and downregulation followed by disturbance of key proteins in downstream pathways involved in CML proliferation and survival. Taken together, our results suggest that an HDAC6i potentiates the effect of imatinib and could overcome TKI resistance in CML cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Fusion Proteins, bcr-abl/metabolism , Histone Deacetylase 6/antagonists & inhibitors , Imatinib Mesylate/pharmacology , Imatinib Mesylate/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Ubiquitination/drug effects , Animals , Caspases/drug effects , Down-Regulation , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Humans , K562 Cells , Mice , Mice, Inbred BALB C , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
16.
Cells ; 9(4)2020 04 18.
Article in English | MEDLINE | ID: mdl-32325691

ABSTRACT

Aiming to promote cancer cell apoptosis is a mainstream strategy of cancer therapy. The second mitochondria-derived activator of caspase (SMAC)/direct inhibitor of apoptosis protein (IAP)-binding protein with low pI (DIABLO) protein is an essential and endogenous antagonist of inhibitor of apoptosis proteins (IAPs). SMAC mimetics (SMs) are a series of synthetically chemical compounds. Via database analysis and literature searching, we summarize the potential mechanisms of endogenous SMAC inefficiency, degradation, mutation, releasing blockage, and depression. We review the development of SMs, as well as preclinical and clinical outcomes of SMs in solid tumor treatment, and we analyze their strengths, weaknesses, opportunities, and threats from our point of view. We also highlight several questions in need of further investigation.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Inhibitor of Apoptosis Proteins/pharmacology , Mitochondria/drug effects , Mitochondrial Proteins/metabolism , Neoplasms/drug therapy , Animals , Apoptosis Regulatory Proteins/drug effects , Caspases/drug effects , Caspases/metabolism , Humans , Inhibitor of Apoptosis Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/drug effects , Neoplasms/metabolism
17.
J Chemother ; 32(3): 132-143, 2020 May.
Article in English | MEDLINE | ID: mdl-32009586

ABSTRACT

Along with changes in dietary habits and lifestyle, the incidence of esophageal cancer is increasing around the world. Since chemotherapy for esophageal cancer has significant side effects, phytochemicals have attracted attention as an alternative medicine. Licochalcone C (LCC) is a flavonoid compound extracted from Licorice, with a variety of clinical uses including anti-cancer, anti-inflammatory and anti-oxidant effects. Treatment with LCC for 48 h significantly decreased cell viability of esophageal squamous cell carcinoma (ESCC) cells in a dose- and time-dependent manner with IC50 values of 28 µM (KYSE 30), 36 µM (KYSE 70), 19 µM (KYSE 410), 28 µM (KYSE 450) and 26 µM (KYSE 510). LCC induced G1 arrest accompanied by decreased cyclin D1 expression and an increase in the levels of p21 and p27. LCC increased the levels of intracellular ROS, cytochrome C release, and multi-caspase activity, and decreased mitochondrial membrane potential. LCC induced the protein expression of ER stress markers (GRP78 and CHOP) and phosphorylation JNK, c-Jun and p38. We investigated the expression of pro-apoptotic and anti-apoptotic proteins to elucidate the mechanism of apoptosis. Our findings contribute to the understanding of apoptosis mechanism underlying LCC in ESCC cells and provide new insights into the potential clinical opportunities of LCC for ESCC treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Chalcones/pharmacology , Esophageal Squamous Cell Carcinoma/drug therapy , G1 Phase/drug effects , Antineoplastic Agents/administration & dosage , Caspases/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chalcones/administration & dosage , Cytochromes c/biosynthesis , Dose-Response Relationship, Drug , Endoplasmic Reticulum Chaperone BiP , Humans , Membrane Potential, Mitochondrial/drug effects , Reactive Oxygen Species , Time Factors
18.
Nat Commun ; 11(1): 304, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31949141

ABSTRACT

Biological processes in development and disease are controlled by the abundance, localization and modification of cellular proteins. We have developed versatile tools based on recombinant E3 ubiquitin ligases that are controlled by light or drug induced heterodimerization for nanobody or DARPin targeted depletion of endogenous proteins in cells and organisms. We use this rapid, tunable and reversible protein depletion for functional studies of essential proteins like PCNA in DNA repair and to investigate the role of CED-3 in apoptosis during Caenorhabditis elegans development. These independent tools can be combined for spatial and temporal depletion of different sets of proteins, can help to distinguish immediate cellular responses from long-term adaptation effects and can facilitate the exploration of complex networks.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Cytological Techniques , Light , Ubiquitin-Protein Ligases/drug effects , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/radiation effects , Animals , Apoptosis , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/drug effects , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/radiation effects , Caspases/drug effects , Caspases/metabolism , Caspases/radiation effects , Cell Engineering/methods , DNA Damage , DNA Ligase ATP , DNA Repair , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Green Fluorescent Proteins , HeLa Cells , Humans , Lamin Type A/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/genetics
19.
Exp Parasitol ; 209: 107823, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31862270

ABSTRACT

Typically, antimicrobial peptides (AMPs) are short positive charged peptides serving a key role in innate immunity as well as antimicrobial activity. Discovering novel therapeutic agents is considered as an undeniable demand due to increasing microbial species with antibiotic resistance. In this direction, the unique ability of AMPs to modulate immune responses highlighted them as novel drug candidates in the field of microbiology. Patients affected by leishmaniasis; a neglected tropical disease, confront serious problems for their treatment including resistance to common drugs as well as toxicity and high cost of therapy. So, there is a need for development of new drug candidates to control the diseases. Jellein, a peptide derived from royal jelly of honeybee has been shown to have promising effect against several bacterial and fungal species. In current study, anti-leishmanial effect of Jellein and its lauric acid conjugated form was investigated against two forms of Leishmania major (L. major) parasite. Moreover, cytotoxic effect of these peptides was studied in THP1 cell line and human Red Blood Cells (RBCs). Furthermore, the mechanism of action of peptides on L. major promastigotes was assessed through different methods. The results demonstrated that, conjugation of lauric acid to Jellein not only had no effect on the elevation of antimicrobial activity but also halted it completely. Moreover, Jellein caused a limitation in the number of L. major promastigotes by pore formation as well as changing the membrane potential rather than induction of apoptosis or activation of caspases.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Antiprotozoal Agents/pharmacology , Leishmania major/drug effects , Leishmaniasis, Cutaneous/drug therapy , Oligopeptides/chemistry , Antigens, Differentiation, B-Lymphocyte/pharmacology , Antimicrobial Cationic Peptides/therapeutic use , Antimicrobial Cationic Peptides/toxicity , Antiprotozoal Agents/therapeutic use , Antiprotozoal Agents/toxicity , Caspases/drug effects , Caspases/metabolism , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Erythrocytes/drug effects , Escherichia coli/drug effects , Fatty Acids/chemistry , Flow Cytometry , Hemolysis , Histocompatibility Antigens Class II/pharmacology , Humans , Lauric Acids/pharmacology , Lauric Acids/therapeutic use , Lauric Acids/toxicity , Leishmania major/ultrastructure , Membrane Potentials/drug effects , Microscopy, Electron, Scanning , Neglected Diseases/drug therapy , Neglected Diseases/parasitology , Oligopeptides/pharmacology , Oligopeptides/therapeutic use , Oligopeptides/toxicity
20.
Planta Med ; 85(14-15): 1150-1159, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31590196

ABSTRACT

In Europe, especially in German-speaking countries, administration of mistletoe extracts is the most common and popular complementary and alternative therapy approach reported in oncology. Mistletoe therapy is applied to children with cancer for curative and palliative therapeutic regimes with increasing frequency, but at the same time, there are only a few studies on the effectiveness of this therapy. Therefore, we have investigated the response of various pediatric cell lines (acute myeloid leukemia, Ewing's sarcoma, hepatocellular carcinoma, medulloblastoma, neuroblastoma, and osteosarcoma) to mistletoe extract, abnobaVISCUM Fraxini. Effects on cell proliferation, cell cycle distribution as well as on mitochondrial integrity and caspase-mediated apoptosis were investigated in neuroblastoma cell lines, SH-SY5Y and Kelly. Additionally, in vitro tumor cell migration and invasion were studied. In vivo effects of the mistletoe extract were investigated in a syngeneic neuroblastoma mouse model. We could show that tumor cell lines were from 5- to 640-fold more sensitive to abnobaVISCUM Fraxini treatment than non-tumorigenic fibroblasts, whereby neuroblastoma cell lines were the most sensitive. For two neuroblastoma cell lines, SH-SY5Y and Kelly, induction of caspase-9-mediated apoptosis, a decrease of mitochondrial integrity as well as attenuation of migration and invasion were observed. In vivo experiments revealed a reduction of tumor growth and a prolonged survival of tumor-bearing animals. In summary, we can state that these results provide the first preclinical data for cytotoxic activities of abnobaVISCUM Fraxini for a broad panel of pediatric tumor cell lines, in particular, neuroblastoma cells. Thus, it might be a potential remedy for the supportive treatment of neuroblastoma.


Subject(s)
Apoptosis/drug effects , Cytotoxins/pharmacology , Neuroblastoma/drug therapy , Phytotherapy , Plant Extracts/pharmacology , Viscum album/chemistry , Animals , Caspases/drug effects , Caspases/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Humans , Male , Mice , Mitochondria/drug effects , Pediatrics
SELECTION OF CITATIONS
SEARCH DETAIL
...