Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 345
Filter
Add more filters

Complementary Medicines
Publication year range
1.
FASEB J ; 36(1): e22082, 2022 01.
Article in English | MEDLINE | ID: mdl-34918389

ABSTRACT

Vitamin D deficiency is associated with risk of several common cancers, including colorectal cancer (CRC). Here we have utilized patient derived epithelial organoids (ex vivo) and CRC cell lines (in vitro) to show that calcitriol (1,25OHD) increased the expression of the CRC tumor suppressor gene, CDH1, at both the transcript and protein level. Whole genome expression analysis demonstrated significant differential expression of a further six genes after 1,25OHD treatment, including genes with established links to carcinogenesis GADD45, EFTUD1 and KIAA1199. Furthermore, gene ontologies relevant to carcinogenesis were enriched by 1,25OHD treatment (e.g., 'regulation of Wnt signaling pathway', 'regulation of cell death'), with common enriched processes across in vitro and ex vivo cultures including 'negative regulation of cell proliferation', 'regulation of cell migration' and 'regulation of cell differentiation'. Our results identify genes and pathways that are modifiable by calcitriol that have links to CRC tumorigenesis. Hence the findings provide potential mechanism to the epidemiological and clinical trial data indicating a causal association between vitamin D and CRC. We suggest there is strong rationale for further well-designed trials of vitamin D supplementation as a novel CRC chemopreventive and chemotherapeutic agent.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinogenesis/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Proteins/biosynthesis , Neoplasms/metabolism , Transcriptome/drug effects , Vitamin D/analogs & derivatives , Caco-2 Cells , HCT116 Cells , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Vitamin D/pharmacology
2.
Int J Mol Sci ; 22(9)2021 Apr 29.
Article in English | MEDLINE | ID: mdl-33947048

ABSTRACT

Hemistepta lyrata (Bunge) Bunge is a biennial medicinal plant possessing beneficial effects including anti-inflammation, and hemistepsin A (HsA) isolated from H. lyrata has been known as a hepatoprotective sesquiterpene lactone. In this report, we explored the cytotoxic effects of H. lyrata on hepatocellular carcinoma (HCC) cells and investigated the associated bioactive compounds and their relevant mechanisms. From the viability results of HCC cells treated with various H. lyrata extracts, HsA was identified as the major compound contributing to the H. lyrata-mediated cytotoxicity. HsA increased expression of cleaved PARP and cells with Sub-G1 phase, Annexin V binding, and TUNEL staining, which imply HsA induces apoptosis. In addition, HsA provoked oxidative stress by decreasing the reduced glutathione/oxidized glutathione ratio and accumulating reactive oxygen species and glutathione-protein adducts. Moreover, HsA inhibited the transactivation of signal transducer and activator of transcription 3 (STAT3) by its dephosphorylation at Y705 and glutathione conjugation. Stable expression of a constitutive active mutant of STAT3 prevented the reduction of cell viability by HsA. Finally, HsA enhanced the sensitivity of sorafenib-mediated cytotoxicity by exaggerating oxidative stress and Y705 dephosphorylation of STAT3. Therefore, HsA will be a promising candidate to induce apoptosis of HCC cells via downregulating STAT3 and sensitizing conventional chemotherapeutic agents.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Gene Expression Regulation, Neoplastic/drug effects , Lactones/pharmacology , Liver Neoplasms/pathology , Neoplasm Proteins/biosynthesis , STAT3 Transcription Factor/biosynthesis , Sesquiterpenes/pharmacology , Transcriptional Activation/drug effects , Cell Line, Tumor , Down-Regulation/drug effects , Drug Screening Assays, Antitumor , Genes, Reporter , Humans , Neoplasm Proteins/genetics , Oxidative Stress , Protein Kinase Inhibitors/pharmacology , Recombinant Proteins/metabolism , STAT3 Transcription Factor/genetics , Sorafenib/pharmacology
3.
Int J Mol Sci ; 22(5)2021 Mar 06.
Article in English | MEDLINE | ID: mdl-33800754

ABSTRACT

The natural isoquinoline alkaloid Berberine (BBR) has been shown to possess several therapeutic effects, including anticancer activity. Different BBR derivatives have been designed and synthesized in order to obtain new compounds with enhanced anticancer efficacy. We previously showed that intraperitoneal (IP) administration of the BBR-derived NAX014 compound was able to counteract HER-2 overexpressing mammary tumors onset and progression in transgenic mice. However, the IP administration was found to induce organ toxicity at doses higher than 2.5 mg/Kg. In this study, we evaluated the effect of intragastric (IG) administration of 20 mg/kg of NAX014 on both safety and anticancer efficacy in HER-2/neu transgenic mice. Furthermore, cancer cell dissemination and migration, tumor cell senescence and immunological changes were examined. Our results demonstrated that IG NAX014 administration delayed the onset of mammary tumors with no negative effects on health and survival. NAX014 reduced HER-2 overexpressing BC cells migration in vitro and the frequency of lung metastasis in HER-2/neu transgenic mice. A statistically significant increase of senescence-associated p16 expression was observed in tumors from NAX014-treated mice, and the induction of cell senescence was observed in HER-2 overexpressing BC cells after in vitro treatment with NAX014. Although NAX014 did not modulate the presence of tumor-infiltrating lymphocytes, the level of circulating TNF-α and VEGF was found to be reduced in NAX014-treated mice. The overall results address the NAX014 compound as potential tool for therapeutic strategies against HER-2 overexpressing breast cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Berberine Alkaloids/therapeutic use , Genes, erbB-2 , Mammary Neoplasms, Experimental/prevention & control , Neoplasm Metastasis/prevention & control , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Berberine Alkaloids/administration & dosage , Berberine Alkaloids/chemistry , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Drug Screening Assays, Antitumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Transgenic , Molecular Structure , Neoplasm Metastasis/drug therapy , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Rats , Tumor Burden/drug effects
4.
Mol Med Rep ; 23(4)2021 04.
Article in English | MEDLINE | ID: mdl-33537819

ABSTRACT

Hyperthermia is one of the most widely employed adjuvant treatments for cancer, especially for hyperthermic intraperitoneal chemotherapy, and has few side effects. Gastric cancer has various hyperthermia sensitivities, but the exact molecular mechanisms remain to be elucidated. In the present study, western blotting was performed to detect differential expression of proteins that have been reported to be upregulated in gastric cancer. Following knockdown of these proteins, apoptosis was measured by Annexin V­FITC/propidium iodide (PI) double staining and hyperthermia treatment was applied. To evaluate the effect of cyclin­dependent kinase 6 (CDK6) on hyperthermia­induced apoptosis, CDK6 was knocked down or inhibited by the addition of a specific inhibitor and subsequent PI staining and cell proliferation, migration and invasion assays were performed. Hyperthermia­induced protein kinase B (AKT) expression and phosphorylation inhibition were detected. As demonstrated in the present study, the hyperthermia­induced proteins kinesin family member 11 (KIF11), cyclin­dependent kinase 6 (CDK6), stromal antigen 2, NIMA­related kinase 2 and karyopherin subunit α 4 were highly expressed in gastric cancer cells, including SH­10­TC and HGC­27 cells. Knockdown of KIF11 significantly increased apoptosis without hyperthermia treatment and CDK6 significantly increased hyperthermia­induced apoptosis, prompting the present study to focus on CDK6. It was further confirmed that CDK6 activity was critical for decreasing hyperthermia­induced apoptosis and for cell proliferation. Hyperthermia­induced AKT expression and phosphorylation inhibition is potentially the main cause of CDK6 transcriptional upregulation. Taken together, these findings demonstrated that CDK6 is upregulated via hyperthermia­induced AKT inhibition and subsequently protected gastric cancer cells from hyperthermia­induced apoptosis, indicating that it is a potential therapeutic target to sensitize gastric cancer cells to hyperthermia­based therapy.


Subject(s)
Cyclin-Dependent Kinase 6/biosynthesis , Gene Expression Regulation, Neoplastic , Hyperthermia, Induced , Neoplasm Proteins/biosynthesis , Stomach Neoplasms/metabolism , Transcription, Genetic , Up-Regulation , Cell Line, Tumor , Cyclin-Dependent Kinase 6/genetics , Humans , Neoplasm Proteins/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Stomach Neoplasms/therapy
5.
J Biochem Mol Toxicol ; 35(5): e22736, 2021 May.
Article in English | MEDLINE | ID: mdl-33512746

ABSTRACT

BACKGROUND: The field of nanotechnology offers great opportunities for cancer therapy. OBJECTIVE: This study aimed to compare the therapeutic impact of Zn oxide nanoparticles (ZnO NPs) and thymoquinone (TQ) alone or as cotherapy in Ehrlich ascites carcinoma (EAC) induced in mice. MATERIALS AND METHODS: This study was performed on 75 female albino mice divided into Group I: EAC-bearing control group, Group II: EAC treated with TQ, Group III: EAC treated with low-dose ZnO NPs, Group IV: EAC treated with high-dose ZnO NPs, Group V: EAC treated with TQ and low-dose ZnO NPs. All groups were subjected to measurement of cell viability, ascites fluid volume, Bcl2 protein expression by Western blot analysis, cyclooxygenase 2 (COX2) gene expression by a real-time polymerase chain reaction, enzyme-linked immunosorbent assay levels of Beclin 1, interferon γ (INFγ), interleukin 13 (IL-13), and estimation of Zn concentrations in EAC cells and liver homogenate to evaluate its toxicity. RESULTS: Cotherapy has an efficient anticancer effect by enhancing apoptosis and autophagy, resulting in reducing tumor cell viability and ascites fluid volume together with downregulation of Bcl2 protein expression. This cotherapy increases Beclin 1 and INFγ and decreases IL-13. ZnO NPs upregulate COX2 expression, whereas TQ downregulates its expression. High-dose ZnO NPs have more toxic effects on liver enzymes. Using TQ together with ZnO NPs can eliminate ZnO NPs liver toxicity. CONCLUSION: The cotherapy has an efficient anticancer effect by enhancing apoptosis and autophagy. High-dose ZnO NPs have more toxic effects on liver enzymes. Using TQ together with ZnO NPs can eliminate ZnO NP liver toxicity.


Subject(s)
Benzoquinones , Carcinoma, Ehrlich Tumor , Gene Expression Regulation, Neoplastic/drug effects , Nanoparticles , Neoplasm Proteins/biosynthesis , Zinc Oxide , Animals , Benzoquinones/chemistry , Benzoquinones/pharmacology , Carcinoma, Ehrlich Tumor/drug therapy , Carcinoma, Ehrlich Tumor/metabolism , Carcinoma, Ehrlich Tumor/pathology , Female , Mice , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Zinc Oxide/chemistry , Zinc Oxide/pharmacology
6.
J Mol Med (Berl) ; 99(3): 373-382, 2021 03.
Article in English | MEDLINE | ID: mdl-33409552

ABSTRACT

Vascular calcification may result from stimulation of osteogenic signalling with upregulation of the transcription factors CBFA1, MSX2 and SOX9, as well as alkaline phosphatase (ALPL), which degrades and thus inactivates the calcification inhibitor pyrophosphate. Osteogenic signalling further involves upregulation of the Ca2+-channel ORAI1. The channel is activated by STIM1 and then accomplishes store-operated Ca2+ entry. ORAI1 and STIM1 are upregulated by the serum & glucocorticoid inducible kinase 1 (SGK1) which is critically important for osteogenic signalling. Stimulators of vascular calcification include vasopressin. The present study explored whether exposure of human aortic smooth muscle cells (HAoSMCs) to vasopressin upregulates ORAI1 and/or STIM1 expression, store-operated Ca2+ entry and osteogenic signalling. To this end, HAoSMCs were exposed to vasopressin (100 nM, 24 h) without or with additional exposure to ORAI1 blocker MRS1845 (10 µM) or SGK1 inhibitor GSK-650394 (1 µM). Transcript levels were measured using q-RT-PCR, cytosolic Ca2+-concentration ([Ca2+]i) by Fura-2-fluorescence, and store-operated Ca2+ entry from increase of [Ca2+]i following re-addition of extracellular Ca2+ after store depletion with thapsigargin (1 µM). As a result, vasopressin enhanced the transcript levels of ORAI1 and STIM1, store-operated Ca2+ entry, as well as the transcript levels of CBFA1, MSX2, SOX9 and ALPL. The effect of vasopressin on store-operated Ca2+ entry as well as on transcript levels of CBFA1, MSX2, SOX9 and ALPL was virtually abrogated by MRS1845 and GSK-650394. In conclusion, vasopressin stimulates expression of ORAI1/STIM1, thus augmenting store-operated Ca2+ entry and osteogenic signalling. In HAoSMCs, vasopressin (VP) upregulates Ca2+ channel ORAI1 and its activator STIM1. VP upregulates store-operated Ca2+ entry (SOCE) and osteogenic signalling (OS). VP-induced SOCE, OS and Ca2+-deposition are disrupted by ORAI1 inhibitor MRS1845. VP-induced SOCE, OS and Ca2+-deposition are disrupted by SGK1 blocker GSK-650394. KEY MESSAGES: • In HAoSMCs, vasopressin (VP) upregulates Ca2+ channel ORAI1 and its activator STIM1. • VP upregulates store-operated Ca2+ entry (SOCE) and osteogenic signalling (OS). • VP-induced SOCE, OS and Ca2+-deposition are disrupted by ORAI1 inhibitor MRS1845. • VP-induced SOCE, OS and Ca2+-deposition are disrupted by SGK1 blocker GSK-650394.


Subject(s)
Calcium Signaling/drug effects , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , ORAI1 Protein/biosynthesis , Vascular Calcification/metabolism , Vasopressins/pharmacology , Aorta/cytology , Benzoates/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Signaling/physiology , Cells, Cultured , Drug Evaluation, Preclinical , Humans , Immediate-Early Proteins/antagonists & inhibitors , Immediate-Early Proteins/physiology , Myocytes, Smooth Muscle/metabolism , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Nitrendipine/analogs & derivatives , Nitrendipine/pharmacology , ORAI1 Protein/antagonists & inhibitors , ORAI1 Protein/genetics , Osteogenesis/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/physiology , Stromal Interaction Molecule 1/biosynthesis , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/physiology , Transcription, Genetic/drug effects , Up-Regulation/drug effects , Vascular Calcification/prevention & control
7.
Cell Death Differ ; 28(4): 1174-1192, 2021 04.
Article in English | MEDLINE | ID: mdl-33110216

ABSTRACT

Hepatic ischemic reperfusion injury (IRI) is a common complication of liver surgery. Although an imbalance between mitochondrial fission and fusion has been identified as the cause of IRI, the detailed mechanism remains unclear. Augmenter of liver regeneration (ALR) was reported to prevent mitochondrial fission by inhibiting dynamin-related protein 1 (Drp1) phosphorylation, contributing partially to its liver protection. Apart from phosphorylation, Drp1 activity is also regulated by small ubiquitin-like modification (SUMOylation), which accelerates mitochondrial fission. This study aimed to investigate whether ALR-mediated protection from hepatic IRI might be associated with an effect on Drp1 SUMOylation. Liver tissues were harvested from both humans and from heterozygous ALR knockout mice, which underwent IRI. The SUMOylation and phosphorylation of Drp1 and their modulation by ALR were investigated. Hepatic Drp1 SUMOylation was significantly increased in human transplanted livers and IRI-livers of mice. ALR-transfection significantly decreased Drp1 SUMOylation, attenuated the IRI-induced mitochondrial fission and preserved mitochondrial stability and function. This study showed that the binding of transcription factor Yin Yang-1 (YY1) to its downstream target gene UBA2, a subunit of SUMO-E1 enzyme heterodimer, was critical to control Drp1 SUMOylation. By interacting with YY1, ALR inhibits its nuclear import and dramatically decreases the transcriptional level of UBA2. Consequently, mitochondrial fission was significantly reduced, and mitochondrial function was maintained. This study showed that the regulation of Drp1 SUMOylation by ALR protects mitochondria from fission, rescuing hepatocytes from IRI-induced apoptosis. These new findings provide a potential target for clinical intervention to reduce the effects of IRI during hepatic surgery.


Subject(s)
DNA-Binding Proteins/biosynthesis , Dynamins/genetics , Mitochondria, Liver/metabolism , Neoplasm Proteins/biosynthesis , Reperfusion Injury/prevention & control , Sumoylation , Animals , Apoptosis , Cell Line , DNA-Binding Proteins/genetics , Dynamins/metabolism , Humans , Liver/metabolism , Liver/pathology , Liver Regeneration , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Liver/pathology , Mitochondrial Dynamics , Neoplasm Proteins/genetics , Phosphorylation , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
8.
Hematol Oncol Stem Cell Ther ; 14(3): 179-191, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33278349

ABSTRACT

Vitamin C (ascorbate) is an essential dietary requirement, with fundamental redox, anti-oxidant functions at physiologic concentrations. Vitamin C is a cofactor for Fe2+ and 2-oxoglutarate-dependent dioxygenases, englobing large families of enzymes, including also epigenetic regulators of DNA and histone methylation. Importantly, vitamin C is involved in the control of the activity of TET (ten-eleven translocation) enzymes, key epigenetic regulators. For this spectrum of activities, often involving pathways deregulated in cancer cells, vitamin C possesses some pharmacologic activities that can be exploited in anticancer therapy. In particular, the capacity of pharmacological doses of vitamin C to target redox imbalance and to rescue deregulated epigenetic program observed in some cancer cells represents a consistent therapeutic potentiality. Several recent studies have identified some cancer subsets that could benefit from the pharmacological activities of vitamin C. The identification of these potentially responsive patients will help to carefully define controlled clinical trials aiming to evaluate the anticancer activity of Vitamin C.


Subject(s)
Antineoplastic Agents/therapeutic use , Ascorbic Acid/therapeutic use , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Proteins/biosynthesis , Neoplasms , Humans , Neoplasms/classification , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology
9.
Molecules ; 25(21)2020 Oct 30.
Article in English | MEDLINE | ID: mdl-33143282

ABSTRACT

Manna is produced from the spontaneous solidification of the sap of some Fraxinus species, and, owing its content in mannitol, is used in medicine as a mild laxative. Manna is also a rich source of characteristic bio-phenols with reducing, antioxidant and anti-inflammatory properties. This study assesses the activity of a hydrophilic extract of manna (HME) on cellular and molecular events in human colon-rectal cancer cells. HME showed a time- and concentration-dependent anti-proliferative activity, measured by MTT assay, in all the cell lines examined, namely Caco-2, HCT-116 and HT-29. The amounts of HME that caused 50% of cell death after a 24 h treatment were 8.51 ± 0.77, 10.73 ± 1.22 and 28.92 ± 1.99 mg manna equivalents/mL, respectively; no toxicity was observed in normally differentiated Caco-2 intestinal cells. Hydroxytyrosol, a component of HME known for its cytotoxic effects on colon cancer cells, was ineffective, at least at the concentration occurring in the extract. Through flow-cytometric techniques and Western blot analysis, we show that HME treatment causes apoptosis, assessed by phosphatidylserine exposure, as well as a loss of mitochondrial membrane potential, an intracellular formation of reactive oxygen species (ROS), increases in the levels of cleaved PARP-1, caspase 3 and Bax, and a decrease in Bcl-2 expression. Moreover, HME interferes with cell cycle progression, with a block at the G1/S transition. In conclusion, the phytocomplex extracted from manna exerts an anti-proliferative activity on human colon cancer cells through the activation of mitochondrial pathway-mediated apoptosis and cell cycle arrest. Our data may suggest that manna could have the potential to exert chemo-preventive effects for the intestine.


Subject(s)
Antineoplastic Agents, Phytogenic , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Colonic Neoplasms/drug therapy , Fraxinus/chemistry , Mitochondria/metabolism , Plant Extracts , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Caco-2 Cells , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , HT29 Cells , Humans , Mitochondria/pathology , Neoplasm Proteins/biosynthesis , Plant Extracts/chemistry , Plant Extracts/pharmacology
10.
Mol Biol Rep ; 47(8): 6053-6065, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32737827

ABSTRACT

Mercuric chloride (MC) is a complex substance which is capable to produce free radicals. Middle Eastern Phoenix dactylifera (MEPD) is a flowering plant of palm family with potent antioxidant feature. Due to the increasing use of herbs in medicine, this study was designed to assess the effects of MEPD and MC on inflammatory apoptogenic, oxidative and histomorphometric alterations in liver. Sixty-four male rats were assigned to 8 groups including: control groups (normal group and MC (50 mg/kg)), MEPD groups (30, 90, 270 mg/kg) and MC + MEPD treated groups. All experimental groups were treated intraperitoneally and orally daily for 5 weeks. The relative expression level of apoptotic genes (p53, Bcl2 and Bax) and hepatocyte apoptotic index were analyzed. Also, Nitrite oxide (NO), lipid peroxidation (LP), Ferric Reducing Ability of Plasma (FRAP) assays were conducted to assess the antioxidant levels. Cytokines involved in inflammation, hepatic enzymes and histomorphometric parameters (hepatocytes diameter (HD) and central hepatic vein (CHV)) were evaluated. All factors showed incremental trends following MC administration (else FRAP level and Bcl2, which were decreased) in MC group than normal group (P < 0.05). In comparison with the MC group, total values in MEPD and MEPD + MC groups were decreased (P < 0.05) (except FRAP level and Bcl2, which were increased). According to the obtained data, the administration of MEPD extract has potent antioxidant property that attenuates the destructive hepatic effects of MC by initiation of cellular antioxidant pathways and restoration of pathological changes into the physiological form.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/therapeutic use , Chemical and Drug Induced Liver Injury/drug therapy , Mercuric Chloride/toxicity , Phoeniceae/chemistry , Phytotherapy , Plant Extracts/therapeutic use , Animals , Anti-Inflammatory Agents/isolation & purification , Antioxidants/isolation & purification , Antioxidants/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/etiology , Cytokines/blood , Fatty Acids/analysis , Flavonoids/analysis , Hepatic Veins/pathology , Hepatocytes/drug effects , Hepatocytes/pathology , Lipid Peroxidation/drug effects , Male , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Nitric Oxide/blood , Oxidation-Reduction , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Random Allocation , Rats , Rats, Wistar
11.
Mol Biol Rep ; 47(8): 6043-6051, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32700127

ABSTRACT

Centaurea bruguierana, of the Asteraceae family, has a long history of use in traditional medicines for the treatment of various ailments. However, the anticancer activity and underlying mechanisms have not yet been assessed. The C. bruguierana was extracted with methanol and fractionated into four different fractions. Different cancer cells and one non-cancerous were used to examine the cytotoxic effects of these fractions using MTT assay. The most potent fraction, C. bruguierana ethyl acetate fraction (CB EtOAc), was explored for its effects on cell cycle progression and apoptosis induction by Hoechst staining and annexin V-PI double staining in MCF-7 cells. The expression of apoptosis-related genes was quantified by RT-PCR. Of all fractions, CB EtOAc was found to have the strongest antiproliferative activity (IC50 = 10 µg/mL) against MCF-7 cells. The antiproliferative activity of the CB EtOAc fraction against MCF-7 was correlated with arrested of cell cycle in the G1 phase, nuclear fragmentation, and the exposure of phosphatidylserine. The induction of apoptosis by CB EtOAc in MCF-7 cells was also associated with an increase in the Bax/Bcl-2 ratio and higher expression of caspases. Overall, our results demonstrated that CB EtOAc showed apoptosis-inducing effects, suggesting that C. bruguierana may be a promising source for a novel chemotherapeutic agents for the treatment of breast cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/isolation & purification , Breast Neoplasms/pathology , Centaurea/chemistry , Plant Extracts/pharmacology , 1-Butanol , A549 Cells , Acetates , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Cell Cycle Checkpoints/drug effects , Cell Division/drug effects , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Chloroform , Drug Screening Assays, Antitumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Human Umbilical Vein Endothelial Cells , Humans , Inhibitory Concentration 50 , MCF-7 Cells , Methanol , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Plant Components, Aerial/chemistry , Solvents
12.
Pak J Biol Sci ; 23(8): 1025-1036, 2020 Jan.
Article in English | MEDLINE | ID: mdl-32700853

ABSTRACT

BACKGROUND AND OBJECTIVE: Rosmarinic acid is considered as one of the most important secondary metabolites in medicinal plants especially of family Lamiaceae. Rosmarinic acid can prevent both the tumor initiation and promotion stages of carcinogenesis. The aim of current study was to evaluate the antiproliferative effects of Hyssopus officinalis and Thymus vulgaris callus crude extracts contained rosmarinic acid on breast cancer cells with correlation to phenylpropanoid biosynthetic pathway genes expression. MATERIALS AND METHODS: Calli of both plants were maintained on Murashige and Skoog medium supplemented with kinetin and 2,4-D. Rosmarinic acid was determined spectrophotometrically in both seed-germinated plants (control) and callus tissues. Transcriptional profiling of rosmarinic acid pathway genes was performed with RT-PCR system. The human breast cancer cell line MCF-7 was treated with different levels of crude extracts at different time intervals in order to show their effects on the cell proliferation using a cell viability colorimetric assay (MTT). RESULTS: The results showed a significant increase of rosmarinic acid content up to 6.5% in callus compared to control. The transcriptional profile of the selected rosmarinic acid genes in callus tissues indicated significant effects on the rosmarinic acid content in both genotypes. T. vulgaris (90 µg mL-1) and H. officinalis (150 µg mL-1) callus extracts had exhibited highest reduction in the cell MCF-7 viability after 48 h of exposure. CONCLUSION: It was concluded that rosmarinic acid production increased in callus tissue, showed the higher gene expression levels and remarkably inhibited growth of human breast cancer cell line.


Subject(s)
Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cinnamates/pharmacology , Depsides/pharmacology , Neoplasm Proteins/biosynthesis , Breast Neoplasms/metabolism , Female , Humans , MCF-7 Cells , Protein Biosynthesis/genetics , Rosmarinic Acid
13.
Int J Mol Sci ; 21(14)2020 Jul 11.
Article in English | MEDLINE | ID: mdl-32664580

ABSTRACT

Bone mass loss occurs with a decrease in osteoblast proliferation and differentiation, or the enhancement of bone resorption, which further leads to the impairment of bone mineral density and increase in bone fracture. Recent studies suggest that some phenolic compounds found in food play positive role in bone metabolism. High content of phenolic compounds with potential beneficial effects on bone metabolism have been identified in the Viburnum opulus fruit. The aim of the study was to determine the influence of V. opulus fresh juice (FJ) and juice purified by solid phase extraction (PJ) on osteogenesis processes with osteosarcoma Saos-2 cell lines. V. opulus purified juice revealed stronger potential as an inducer of Saos-2 osteogenic differentiation. Saos-2 cells matrix mineralization was evaluated with alkaline phosphatase (ALP) activity measurement and alizarin red S staining. Gene expression analysis showed the elevation of the mRNA levels of Runt-related transcription factor 2 (RUNX2), ALP, collagen type 1 and osteonectin, whereas the nuclear factor-κB ligand and osteoprotegerin ratio (RANKL/OPG) decreased. Furthermore, V. opulus was able to diminish the secretion of pro-inflammatory cytokines Il6 and TNFα, however had no effect on vascular endothelial growth factor (VEGF). It decreased intracellular oxidative stress and induced DNA repair, but had no effect on the growth inhibition of lactic acid beneficial microorganisms.


Subject(s)
Bone Density Conservation Agents/pharmacology , Bone Neoplasms/pathology , Fruit and Vegetable Juices/analysis , Osteogenesis/drug effects , Osteosarcoma/pathology , Phenols/pharmacology , Plant Extracts/pharmacology , Viburnum/chemistry , Bacteria/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , DNA Repair/drug effects , Drug Evaluation, Preclinical , Gastrointestinal Microbiome/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-6/metabolism , Mitochondria/drug effects , Mitochondria/enzymology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Osteogenesis/genetics , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/metabolism
14.
Nutrients ; 12(4)2020 Apr 06.
Article in English | MEDLINE | ID: mdl-32268584

ABSTRACT

Prostate cancer (PCa) is a multifactorial disease with an unclear etiology. Due to its high prevalence, long latency, and slow progression, PCa is an ideal target for chemoprevention strategies. Many research studies have highlighted the positive effects of natural flavonoids on chronic diseases, including PCa. Different classes of dietary flavonoids exhibit anti-oxidative, anti-inflammatory, anti-mutagenic, anti-aging, cardioprotective, anti-viral/bacterial and anti-carcinogenic properties. We overviewed the most recent evidence of the antitumoral effects exerted by dietary flavonoids, with a special focus on their epigenetic action in PCa. Epigenetic alterations have been identified as key initiating events in several kinds of cancer. Many dietary flavonoids have been found to reverse DNA aberrations that promote neoplastic transformation, particularly for PCa. The epigenetic targets of the actions of flavonoids include oncogenes and tumor suppressor genes, indirectly controlled through the regulation of epigenetic enzymes such as DNA methyltransferase (DNMT), histone acetyltransferase (HAT), and histone deacetylase (HDAC). In addition, flavonoids were found capable of restoring miRNA and lncRNA expression that is altered during diseases. The optimization of the use of flavonoids as natural epigenetic modulators for chemoprevention and as a possible treatment of PCa and other kinds of cancers could represent a promising and valid strategy to inhibit carcinogenesis and fight cancer.


Subject(s)
DNA Methylation/drug effects , DNA, Neoplasm/metabolism , Epigenesis, Genetic/drug effects , Flavonoids/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Prostatic Neoplasms , Humans , Male , Neoplasm Proteins/biosynthesis , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/prevention & control , RNA, Neoplasm/biosynthesis
15.
J Neurogenet ; 34(2): 216-222, 2020.
Article in English | MEDLINE | ID: mdl-32116108

ABSTRACT

Bone cancer pain is considered to be mechanistically unique compared with inflammatory or neuropathic pain states. Toll-like receptor 4 (TLR4) is a transmembrane receptor protein which has been reported to be involved in neuropathic pain. However, the role of TLR4 in bone cancer pain is still unclear. Therefore, the aim of this study is to investigate the hypothesis that oxytocin may ameliorate bone cancer pain by suppressing TLR4 in spinal cord. Behavioral analysis and molecular biological experiments were carried out. Our data demonstrated that intrathecally delivery of oxytocin significantly ameliorated the mechanical allodynia and thermal hyperalgesia in bone cancer pain rats. Moreover, oxytocin suppressed the up-regulation of TLR4 and proinflammatory cytokines TNFα and IL-1ß in spinal cord of bone cancer pain rats. Therefore, we concluded that intrathecal administration of oxytocin relieves bone cancer pain by suppressing the up-regulation of TLR4, TNFα and IL-1ß in spinal cord. Oxytocin possesses analgesic efficacy against bone cancer pain and deserves further to confirm its effectiveness in clinically relevant of cancer pain.


Subject(s)
Analgesics/therapeutic use , Bone Neoplasms/physiopathology , Carcinosarcoma/physiopathology , Hyperalgesia/drug therapy , Oxytocin/therapeutic use , Spinal Cord/drug effects , Toll-Like Receptor 4/antagonists & inhibitors , Analgesics/administration & dosage , Analgesics/pharmacology , Animals , Cell Line, Tumor , Cytokines/metabolism , Drug Evaluation, Preclinical , Gene Expression Regulation, Neoplastic/drug effects , Hyperalgesia/etiology , Hyperalgesia/physiopathology , Injections, Spinal , Interleukin-1beta/biosynthesis , Interleukin-1beta/genetics , Male , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Transplantation , Oxytocin/administration & dosage , Oxytocin/pharmacology , Rats , Spinal Cord/metabolism , Toll-Like Receptor 4/biosynthesis , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Up-Regulation/drug effects
16.
Mater Sci Eng C Mater Biol Appl ; 110: 110594, 2020 May.
Article in English | MEDLINE | ID: mdl-32204058

ABSTRACT

Short interfering RNA (siRNA) has been investigated as a promising modality of cancer treatment due to its capability to target specific target genes for downregulation. However, the successful application of this strategy depends on producing a safe and effective carrier system for delivering siRNA to the tumor. Thus, investigation of siRNA delivery carriers is a fundamental step in the field of siRNA-based therapeutics. In the current research, the surface of selenium nanoparticles (SeNPs) were modified with the tumor-targeted molecular RGDfC peptide with positive charge to synthetize the biocompatible siRNA carrier RGDfC-SeNPs. Subsequently, KLK12-siRNA was loaded onto the surface of RGDfC-SeNPs to create functionalized nanoparticles (RGDfC-Se@siRNA) that we tested for in vitro and in vivo antitumor efficacy. We measured significantly greater particle uptake in HT-29 colorectal cancer cells relative to HUVECs, providing evidence for the targeted delivery of RGDfC-Se@siRNA. We found that RGDfC-Se@siRNA could enter HT-29 cells primarily via clathrin-mediated endocytosis. Further, these particles experienced faster siRNA release in an acidic microenvironment compared to pH 7.4. The results from quantitative PCR and Western blot assays suggested that the target gene of KLK12 in HT-29 cells were obviously silenced by RGDfC-Se@siRNA. The further biological studies showed that treatment with RGDfC-Se@siRNA had ability to suppress the proliferation and migration/invasion of HT-29 cells, and triggered HT-29 cells apoptosis. RGDfC-Se@siRNA could induce the mitochondrial membrane potential (MMP) disruption and enhance the reactive oxygen species (ROS) generation in HT-29 cells, indicating that RGDfC-Se@siRNA induced the HT-29 cells apoptosis possibly by a ROS-mediated mitochondrial dysfunction pathway. Importantly, the in vivo antitumor study also verified that RGDfC-Se@siRNA could significantly suppress the growth of tumor in vivo. In addition, we did not observe any signs of systemic or tissue-specific toxicity after administration of RGDfC-Se@siRNA in mice. As a whole, these findings suggest that RGDfC-Se@siRNA has promising potential as a therapy for colorectal cancer.


Subject(s)
Colorectal Neoplasms , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing/drug effects , Kallikreins/biosynthesis , Metal Nanoparticles , Neoplasm Proteins/biosynthesis , Oligopeptides , Selenium , Animals , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Female , HT29 Cells , Human Umbilical Vein Endothelial Cells , Humans , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Nude , Oligopeptides/chemistry , Oligopeptides/pharmacology , Selenium/chemistry , Selenium/pharmacology , Xenograft Model Antitumor Assays
17.
Cell Prolif ; 53(1): e12732, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31746080

ABSTRACT

OBJECTIVES: Temozolomide (TMZ) is one of the most commonly used clinical drugs for glioblastoma (GBM) treatment, but its drug sensitivity needs to be improved. Gamabufotalin (CS-6), the primary component of the traditional Chinese medicine "ChanSu," was shown to have strong anti-cancer activity. However, more efforts should be directed towards reducing its toxicity or effective treatment doses. METHODS: Target fishing experiment, Western blotting, PCR, confocal immunofluorescence and molecular cloning techniques were performed to search for possible downstream signalling pathways. In addition, GBM xenografts were used to further determine the potential molecular mechanisms of the synergistic effects of CS-6 and TMZ in vivo. RESULTS: Mechanistic research revealed a negative feedback loop between ATP1A3 and AQP4 through which CS-6 inhibited GBM growth and mediated the synergistic treatment effect of CS-6 and TMZ. In addition, by mutating potential amino acid residues of ATP1A3, which were predicted by modelling and docking to interact with CS-6, we demonstrated that abrogating hydrogen bonding of the amino acid Thr794 interferes with the activation of ATP1A3 by CS-6 and that the Thr794Ala mutation directly affects the synergistic treatment efficacy of CS-6 and TMZ. CONCLUSIONS: As the main potential target of CS-6, ATP1A3 activation critically depends on the hydrogen bonding of Thr794 with CS-6. The combination of CS-6 and TMZ could significantly reduce the therapeutic doses and promote the anti-cancer efficacy of CS-6/TMZ monotherapy.


Subject(s)
Aquaporin 4/metabolism , Bufanolides/pharmacology , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/drug therapy , Neoplasm Proteins/biosynthesis , Sodium-Potassium-Exchanging ATPase/biosynthesis , Temozolomide/pharmacology , Animals , Aquaporin 4/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , Neoplasm Proteins/genetics , Sodium-Potassium-Exchanging ATPase/genetics , Xenograft Model Antitumor Assays
18.
BMC Complement Altern Med ; 19(1): 173, 2019 Jul 12.
Article in English | MEDLINE | ID: mdl-31299960

ABSTRACT

BACKGROUND: This study was aimed to determination the tumor inhibitory effect and explore the potential mechanisms of Lagopsis supine ethanol extract (Ls) on colorectal cancer. METHODS: The cell growth inhibition experiment of Ls in colorectal cancer cell lines was determined by MTT method in the time course of 24, 48 and 72 h in four gradient drug concentrations. The protein expression levels of pSTAT3, pJAK2, STAT3, JAK2, Bcl-2 and caspase 3 were measured by Western blot method. The mRNA levels of the downstream genes of STAT3 were detected through semi-quantitative RT PCR. Sixty Balb/c-nude mice were xenograft with HCT116 colorectal cancer cells through subcutaneously. The xenografts were divided into five groups: model group, positive group (capecitabine 300 mg/kg) and three dosages of Ls treated groups (75, 150 and 300 mg/kg). Tumor size and tumor weight were calculated for evaluation the anti-tumor effects. H & E staining and immunohistochemical analysis were used to determine the histopathological changes and the levels of pSTAT3 and pJAK2 in the tumor tissues. RESULTS: Ls exhibited a significant anti-proliferation effect in HCT116 and SW480 cells in vitro. The protein levels of pSTAT3, pJAK2 and Bcl-2, and the mRNA levels of Bcl-2 and Bak notably reduced with a dose-dependent manner. While the protein levels of caspase 3, and mRNA levels of Bax and caspase-3 remarkably increased in the gradient dosage of Ls in HCT116 cells. HCT116 in vivo xenografts experiment showed that the growth of the tumors significantly inhibited by Ls administration, which with no any significant body weight changes in each experiment group. The histopathology analysis displayed that Ls significantly reduced the inflammatory cells in tumor tissue. Furthermore, Ls also significantly down-regulate the protein levels of pSTAT3 and pJAK2 in the tumor tissues, compared with the model group. CONCLUSIONS: This work shows that Ls inhibited the cell proliferation of colorectal cancer in vitro and significantly reduced the tumor growth in HCT116 xenografts in vivo, which is probably related with the JAK/STAT signal pathway.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Colorectal Neoplasms/drug therapy , Drugs, Chinese Herbal/therapeutic use , Lamiaceae/chemistry , Animals , Antineoplastic Agents, Phytogenic/isolation & purification , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Neoplasm Proteins/biosynthesis , Xenograft Model Antitumor Assays
19.
Cytogenet Genome Res ; 158(1): 17-24, 2019.
Article in English | MEDLINE | ID: mdl-31261155

ABSTRACT

Osteoarthritis (OA) is a degenerative disease characterized by progressive articular cartilage destruction and joint marginal osteophyte formation with different degrees of synovitis. Docosahexaenoic acid (DHA) is an unsaturated fatty acid with anti-inflammatory, antioxidant, and antiapoptotic functions. In this study, the human chondrosarcoma cell line SW1353 was cultured in vitro, and an OA cell model was constructed with inflammatory factor IL-1ß stimulation. After cells were treated with DHA, cell apoptosis was measured. Western blot assay was used to detect protein expression of apoptosis-related factors (Bax, Bcl-2, and cleaved caspase-3) and mitogen-activated protein kinase (MAPK) signaling pathway family members, including extracellular signal-regulated kinase (ERK), c-JUN N-terminal kinase (JNK), and p38 MAPK. Our results show that IL-1ß promotes the apoptosis of SW1353 cells, increases the expression of Bax and cleaved caspase-3, and activates the MAPK signaling pathway. In contrast, DHA inhibits the expression of IL-1ß, inhibits IL-1ß-induced cell apoptosis, and has a certain inhibitory effect on the activation of the MAPK signaling pathway. When the MAPK signaling pathway is inhibited by its inhibitors, the effects of DHA on SW1353 cells are weakened. Thus, DHA enhances the apoptosis of SW1353 cells through the MAPK signaling pathway.


Subject(s)
Apoptosis/drug effects , Bone Neoplasms/pathology , Chondrosarcoma/pathology , Docosahexaenoic Acids/pharmacology , Interleukin-1beta/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Osteoarthritis/drug therapy , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Butadienes/pharmacology , Cell Line, Tumor , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , Enzyme Induction/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-1beta/biosynthesis , Interleukin-1beta/genetics , Interleukin-1beta/pharmacology , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/biosynthesis , Mitogen-Activated Protein Kinases/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Nitriles/pharmacology , Protein Kinase Inhibitors/pharmacology
20.
Biomed Res Int ; 2019: 8084109, 2019.
Article in English | MEDLINE | ID: mdl-31179333

ABSTRACT

Shenqi Fuzheng Injection (SFI) is a traditional Chinese medicine injection with anticancer properties and is mainly composed of ginseng and astragalus. Its efficacy has been confirmed in clinical trials, but the mechanism remains unclear. We investigated the effect of SFI on vascular endothelial growth factor (VEGF) gene expression in hepatocellular carcinoma (HCC) cells and identified its possible mechanism of synergistic effects when combined with the chemotherapeutic drug interferon (IFN-) α. An MTT assay was used to measure the inhibition effects of low-dose IFN-α (6000 IU) with or without SFI (0.5 g/L) on the HCC cell line MHCC97. VEGF-silenced MHCC97L-mir200 cell lines were prepared using lentiviral vectors and evaluated by real-time PCR to determine the inhibition effect. We examined MHCC97L-mir200 and MHCC97L cells by MTT assay, using IFN-α alone or in combination with SFI. The inhibition ratio of IFN-α (6000 IU) was -29.5%, while that for IFN-α (6000 IU) + SFI (0.5 g/L) was 17.0%, which was significantly higher than that for the IFN-α group (P < 0.01). The VEGF gene was silenced successfully in MHCC97-L cells. After interference of VEGF, the inhibition by SFI and IFN-α in MHCC97L-mir200 did not differ from that in MHCC97-L cells (P > 0.05). SFI can reduce the expression of VEGF in HCC, which can increase the efficacy of IFN-α, providing a theoretical basis for clinical application.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Drugs, Chinese Herbal/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , Interferon-alpha/biosynthesis , Liver Neoplasms/metabolism , Neoplasm Proteins/biosynthesis , Vascular Endothelial Growth Factor A/biosynthesis , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Humans , Interferon-alpha/genetics , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Neoplasm Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL