Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
J Inorg Biochem ; 247: 112308, 2023 10.
Article in English | MEDLINE | ID: mdl-37441923

ABSTRACT

Structural and biological studies were conducted on the novel complexes [Fe(U)2(H2O)2]Cl3 (FeU) and [Ru(U)2(H2O)2]Cl3 (RuU) (U = 5,6-Diamino-1,3-dimethylpyrimidine-2,4(1H,3H)-dione) to develop an anticancer drug candidate. The two complexes have been synthesized and characterized. Based on our findings, these complexes have octahedral geometry. The DNA-binding study proved that both complexes coordinated with CT-DNA. The docking study confirmed the potency of both complexes in downregulating the topoisomerase I protein through their high binding affinity. Biological studies have established that both complexes can act as potent anticancer agents against three cancer cell lines. RuU or FeU complexes induce apoptosis in breast cancer cells by increasing caspase9 protein and inhibiting proliferating cell nuclear antigen (PCNA) activity. In addition, both complexes down-regulate topoisomerase I expression in breast cancer cells. Therefore, the RuU and FeU complexes' anticancer activities were mediated via both apoptosis induction and topoisomerase I down-regulation. In conclusion, both complexes have dual anticancer activity pathways that may be responsible for the selective cytotoxicity of the complexes. This makes them more suitable for the development of novel cancer treatment strategies.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Coordination Complexes , Ruthenium , Female , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Breast Neoplasms/drug therapy , Cell Line, Tumor , Coordination Complexes/chemistry , DNA/chemistry , DNA Topoisomerases, Type I/metabolism , Iron/chemistry , Ruthenium/chemistry , Uracil
2.
Int J Biol Macromol ; 235: 123804, 2023 Apr 30.
Article in English | MEDLINE | ID: mdl-36842736

ABSTRACT

The liver is the most important organ in the body. Hepatocyte oxidative damage occurs to excess ROS. Liver fibrosis is a mechanism that the immune system uses to treat extreme inflammation by repairing damaged tissue with the creation of a scar. The outcome of fibrosis may be reversed by consuming natural plant extracts with high ROS-scavenging ability. The date palm fruits contain caffeic acid, gallic acid, syringic acid, and ferulic acid, which have anti-inflammatory, antioxidant, and hepatoprotective properties. This study aimed to prepare a date fruit extract, load it onto chitosan nanoparticles, and compare its anti-fibrotic activity with the unloaded crude extract in the CCl4-mouse model. Our findings show that nanocomposite (Cs@FA/DEx) has anti-fibrotic properties and can improve liver function enzymes and endogenous antioxidant enzymes by inhibiting cell apoptosis caused by CCl4-induction in mice. Furthermore, significantly reduced CD95 and ICAM1 levels and down-regulation of TGFß-1 and collagen-α-1 expression demonstrated the anti-fibrotic effects of the Cs@FA/DEx. Therefore, the Cs@FA/DEx might be an innovative supplement for inhibiting liver fibrosis and hepatocyte inflammation induced by chemical toxins. Besides, this nano-supplement could be a promising anti-hepatocellular carcinoma agent as it has potent in vitro anticancer activity against the HePG2 cell line.


Subject(s)
Chitosan , Liver Diseases , Nanoparticles , Phoeniceae , Mice , Animals , Phoeniceae/chemistry , Chitosan/pharmacology , Chitosan/metabolism , Reactive Oxygen Species/metabolism , Plant Extracts/chemistry , Liver Cirrhosis/chemically induced , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Liver , Antioxidants/chemistry , Liver Diseases/metabolism , Disease Models, Animal , Inflammation/pathology , Carbon Tetrachloride/toxicity
3.
Int J Biol Macromol ; 234: 123633, 2023 Apr 15.
Article in English | MEDLINE | ID: mdl-36791938

ABSTRACT

Lung cancer progresses without obvious symptoms and is detected in most patients at late stages, causing a high rate of mortality. Avocado peels (AVP) were thought to be biowaste, but they have antioxidant and anticancer properties in vitro. Chitosan nanoparticles (Cs-NPs) were loaded with various plant extracts, increasing their in vitro and in vivo anticancer activities. Our goal was to load AVP onto Cs-NPs and determine the role of AVP-extract or AVP-loaded Cs-NPs in controlling the progression of lung cancer caused by urethane toxicity. The AVP-loaded chitosan nano-combination (Cs@AVP NC) was synthesized and characterized. Our in vitro results show that Cs@AVP NC has higher anticancer activity than AVP against three human cancer cell lines. The in vivo study proved the activation of apoptosis in lung cancer cells with the Cs@AVP NC oral treatment more than the AVP treatment. Additionally, Cs@AVP NC-treated animals showed significantly higher p53 and Bax-expression levels and lower NF-κB p65 levels in their lung tissues than in positive control animals. In conclusion, our study demonstrated the superior anticancer potency of Cs@AVP NC over AVP extract and its ability to inhibit lung cancer proliferation. Therefore, oral consumption of Cs@AVP NC might be a promising treatment for lung cancer.


Subject(s)
Chitosan , Lung Neoplasms , Nanoparticles , Persea , Mice , Animals , Humans , Urethane , Lung Neoplasms/drug therapy , Plant Extracts/pharmacology
4.
Pharmaceutics ; 14(6)2022 May 29.
Article in English | MEDLINE | ID: mdl-35745733

ABSTRACT

Apigenin (Ap) is one of the most important natural flavonoids that has potent anticancer activity. This study was designed, for the first time, to load Ap into chitosan to improve its hydrophobicity and then it was coated with albumin-folic acid to increase its stability and bioavailability and to target cancer cells. The newly developed encapsulated Ap (Ap-CH-BSA-FANPs) was characterized and tested in vitro. The zeta potential of -17.0 mV was within the recommended range (-30 mV to +30 mV), indicating that encapsulated apigenin would not quickly settle and would be suspended. The in vitro results proved the great anticancer activity of the encapsulated apigenin on HePG-2 cells compared to pure Ap. The treated HePG-2 cells with Ap-CH-BSA-FANPs demonstrated the induction of apoptosis by increasing p53 gene expression, arresting the cell cycle, increasing caspase-9 levels, and decreasing both the MMP9 gene and Bcl-2 protein expression levels. Moreover, the higher antioxidant activity of the encapsulated apigenin treatment was evident through increasing SOD levels and decreasing the CAT concentration. In conclusion, the Ap-CH-BSA-FANPs were easy to produce with low coast, continued drug release, good loading capacity, high solubility in physiological pH, and were more stable than the formerly Ap-loaded liposomes or PLGA. Moreover, Ap-CH-BSA-FANPs may be a promising chemotherapeutic agent in the treatment of HCC.

5.
Colloids Surf B Biointerfaces ; 206: 111935, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34252691

ABSTRACT

Silver nanoparticles (AgNPs) could be employed in the combat against COVID-19, yet are associated with toxicities. In this study, biogenic and biocompatible AgNPs using the agro-waste, non-edible Hibiscus sabdariffa stem were synthesized. Under optimized reaction conditions, synthesized green AgNPs were crystalline, face cubic centered, spherical with a diameter of around 17 nm and a surface charge of -20 mV. Their murine lethal dose 50 (LD50) was 4 folds higher than the chemical AgNPs. Furthermore, they were more murine hepato- and nephro-tolerated than chemical counterparts due to activation of Nrf-2 and HO-1 pathway. They exerted an apoptotic anti-ovarian cancer activity with IC50 value 6 times more than the normal cell line. Being functionalized with polydopamine and conjugated to either moxifloxacin or gatifloxacin, the conjugates exerted an augmented antibiofilm activity against Klebsiella pneumoniae, Pseudomonas aeruginosa, and Acinetobacter baumannii biofilms that was significantly higher than antibiotic alone or functionalized AgNPs suggesting a synergistic activity. In conclusion, this study introduced a facile one-pot synthesis of biogenic and biocompatible AgNPs with preferential anti-cancer activity and could be utilized as antibiotic delivery system for a successful eradication of Gram-negative biofilms.


Subject(s)
Anti-Bacterial Agents , Metal Nanoparticles , Silver , Animals , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Green Chemistry Technology , Hibiscus , Indoles , Mice , Microbial Sensitivity Tests , Polymers , Silver/pharmacology
6.
Int J Biol Macromol ; 185: 134-152, 2021 Aug 31.
Article in English | MEDLINE | ID: mdl-34147524

ABSTRACT

This study was designed to present a new quercetin encapsulated chitosan functionalized copper oxide nanoparticle (CuO-ChNPs-Q) and assessed its anti-breast cancer activity both in vitro and in vivo. The CuO-ChNPs-Q may act as anti-proliferating agent against DMBA-induced mammary carcinoma in female rats. The CuONPs was functionalized with chitosan then quercetin was conjugated with them producing CuO-ChNPs-Q, then characterized. The in vitro anti-proliferating activity of the CuO-ChNPs-Q was evaluated against three human cell line. Then, the anti-breast cancer effect of the CuO-ChNPs-Q was assessed against DMBA-induction compared to both CuONPs and Q in female rat model. The in vitro results proved the potent anticancer activity of the CuO-ChNPs-Q compared to CuONPs and quercetin. The in vivo data showed significant reduction in breast tumors of DMBA-induced rats treated with CuO-ChNPs-Q compared to CuONPs and Q. The CuO-ChNPs-Q treatment had induced apoptosis via increased p53 gene, arrested the cell-cycle, and increased both cytochrome c and caspase-3 levels leading to mammary carcinoma cell death. Also, the CuO-ChNPs-Q treatment had suppressed the PCNA gene which decreased the proliferation of the mammary carcinoma cells. In conclusion, the CuO-ChNPs-Q might be a promising chemotherapeutic agent for treatment of breast cancer with a minimal toxicity on vital organs.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Chitosan/chemistry , Copper/chemistry , Quercetin/administration & dosage , Tumor Suppressor Protein p53/metabolism , 9,10-Dimethyl-1,2-benzanthracene/adverse effects , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Breast Neoplasms/chemically induced , Breast Neoplasms/metabolism , Caco-2 Cells , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Compounding , Female , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , MCF-7 Cells , Metal Nanoparticles , Proliferating Cell Nuclear Antigen/metabolism , Quercetin/chemistry , Quercetin/pharmacology , Rats , Xenograft Model Antitumor Assays
7.
Molecules ; 25(18)2020 Sep 18.
Article in English | MEDLINE | ID: mdl-32962014

ABSTRACT

New anticancer ruthenium(II/III) complexes [RuCl2(DMSO)2(Hapbim)] (1) and [RuCl3(DMSO) (Hapbim)] (2) (Hapbim = 2-aminophenyl benzimidazole) have been synthesized and characterized, and their chemotherapeutic potential evaluated. The interaction of the compounds with DNA was studied by both UV-Visible and fluorescence spectroscopies, revealing intercalation of both the Hapbim ligand and the Ru complexes. The in vitro cytotoxicity of the compounds was tested on human breast cancer (MCF7), human colorectal cancer (Caco2), and normal human liver cell lines (THLE-2), with compound (2) the most potent against cancer cells. The cytotoxic effect of (2) is shown to correlate with the ability of the Ru(III) complex to induce apoptosis and to cause cell-cycle arrest in the G2/M phase. Notably, both compounds were inactive in the noncancerous cell line. The anticancer effect of (2) has also been studied in an EAC (Ehrlich Ascites Carcinoma) mouse model. Significantly, the activity of the complex was more pronounced in vivo, with removal of the cancer burden at doses that resulted in only low levels of hepatotoxicity and nephrotoxicity. An apoptosis mechanism was determined by the observation of increased Bax and caspase 3 and decreased Bcl2 expression. Furthermore, (2) decreased oxidative stress and increased the levels of antioxidant enzymes, especially SOD, suggesting the enhancement of normal cell repair. Overall, compound (2) shows great potential as a chemotherapeutic candidate, with promising activity and low levels of side effects.


Subject(s)
Antineoplastic Agents/chemistry , Coordination Complexes/chemistry , Ruthenium/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antioxidants/metabolism , Apoptosis/drug effects , Benzimidazoles/chemistry , Carcinoma, Ehrlich Tumor/drug therapy , Carcinoma, Ehrlich Tumor/pathology , Cell Line, Tumor , Cell Survival/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/pharmacology , Coordination Complexes/therapeutic use , Dimethyl Sulfoxide/chemistry , Disease Models, Animal , Drug Screening Assays, Antitumor , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Mice , Proto-Oncogene Proteins c-bcl-2/metabolism , Up-Regulation/drug effects , bcl-2-Associated X Protein/metabolism
8.
Int J Biol Macromol ; 160: 1230-1241, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32474075

ABSTRACT

This study aimed to present a new heart of P. dactylifera (HP) extract loaded chitosan nanoparticles and estimate its anticancer, antimicrobial, antioxidant activity and free radical scavenger effect (in vitro). This nano-supplement may prevent doxorubicin cardiotoxicity and nephrotoxicity in rat model. The HP extract was loaded on chitosan nanoparticles producing HP-ChNPs, then characterized. The antioxidant properties of the HP-ChNPs was assessed in vitro. The antibacterial activity against three-gram positive bacteria and two gram-negative bacteria were done. The in vitro studies of cytotoxicity against MCF7, CaCo3, and Hela cell lines were also evaluated. Then, the protective effect of the HP-ChNPs (2 mg/kg, IP) was evaluated against doxorubicin induce organ toxicity in a rat model. The in vitro studies revealed the antibacterial, anticancer and antioxidant activities of the HP-ChNPs. The in vivo study demonstrates reduction of heart and kidney apoptosis with increased programmed cell death protein-1 (PD-1); as the major anticancer drug (doxorubicin) pathway is to release free radicals with decreased PD-1 levels and induction of apoptosis. In conclusion, the HP-ChNPs, in a very small dose, might be a promising supplement to avoid doxorubicin toxicity with improvment the antioxidant enzymes without affecting its anticancer activity.


Subject(s)
Anti-Infective Agents/pharmacology , Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Chitosan/analogs & derivatives , Nanoparticles/chemistry , Plant Extracts/pharmacology , Animals , Anti-Infective Agents/administration & dosage , Antineoplastic Agents/administration & dosage , Antioxidants/administration & dosage , Apoptosis/drug effects , Doxorubicin/toxicity , HeLa Cells , Humans , Kidney/drug effects , Kidney/metabolism , MCF-7 Cells , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oxidants/toxicity , Phoeniceae/chemistry , Plant Extracts/administration & dosage , Rats , Rats, Sprague-Dawley
9.
Food Chem Toxicol ; 135: 111045, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31830546

ABSTRACT

Cardiotoxicity and nephrotoxicity due to the abnormal production of free radicals have been observed in patients treated with the anticancer antibiotic adriamycin (ADR). The aim of the present study was to evaluate the role of the heart of palm extract in preventing oxidative stress, cardiotoxicity and nephrotoxicity induced by ADR. In this work, an aqueous ethanolic extract of the heart of the Phoenix dactylifera tree (HP) was investigated. The polyphenol content was evaluated by gas chromatography-mass spectrometry (GC-MS) and High-performance liquid chromatography (HPLC). The protective effect of the HP-extract (250 and 500 mg/kg, p.o.) was evaluated along with ADR administration (cumulative dose 15 mg/kg, IP) in rats. The HP-extract (500 mg/kg) treated group showed significant reductions in cardiotoxicity and nephrotoxicity serum markers, apoptotic percentage, and caspase-3 and cyclooxygenase-2 level, with an improvement in antioxidant enzymes in both heart and kidney homogenate, compared with the ADR-induction group. The cardiac and kidney programmed cell death protein-1 (PD-1) was increased in high dose HP-extract treated rats after being inhibited by ADR administration. In conclusion, the HP-extract might be a promising food supplement for preventing the cardiotoxicity and nephrotoxicity induced by ADR administration.


Subject(s)
Apoptosis/drug effects , Doxorubicin/toxicity , Heart/drug effects , Kidney/drug effects , Oxidative Stress/drug effects , Phoeniceae/chemistry , Plant Extracts/pharmacology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Dose-Response Relationship, Drug , Male , Plant Extracts/chemistry , Polyphenols/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL