Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Toxicol Lett ; 401: 13-23, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39197506

RESUMEN

BACKGROUND: Nowadays, the use of food additives, such as Sunset Yellow (SY), is growing, which attracted attention to the potential relationship between some diseases and food additives. AIM: The study aimed to investigate the role of Sunset Yellow during chemically-induced mammary gland carcinogenesis in Sprague-Dawley rats. MATERIAL AND METHODS: Three groups of female rats were intraperitoneally administered with N-methyl-N-nitrosourea (MNU). Group 1 was set on a basal diet. Group 2 was treated with 161.4 mg\kg\day Sunset Yellow (SY). Group 3 was given SY at 80.7 mg\kg\day. Groups 4-6 were not administered MNU; Group 4 received vehicles only. Groups 5 and 6 were administered SY similarly to groups 2 and 3 respectively. RESULTS: Sunset Yellow at both doses exerted a significant dose-dependent increase in tumor incidences, multiplicities, volumes, and decreased tumor latency as compared with control. Immunolabeling indexes of the proliferating cell nuclear antigen, estrogen receptor alpha, and progesterone receptor were significantly increased after SY treatment. Oxidative stress markers, serum estrogen, progesterone, and prolactin levels were significantly modified by SY treatment. The mRNA expression of estrogen receptor alpha and epidermal growth factor was up-regulated in SY groups versus control. CONCLUSION: Collectively, SY has significantly promoted MNU-induced mammary tumors in rats with underlying mechanisms correlating SY consumption with estrogen disruption and subsequent antioxidative stress discrepancy.

2.
BMC Complement Med Ther ; 24(1): 185, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38711049

RESUMEN

BACKGROUND: Cancer is a fatal disease that severely affects humans. Designing new anticancer strategies and understanding the mechanism of action of anticancer agents is imperative. HYPOTHESIS/PURPOSE: In this study, we evaluated the utility of metformin and D-limonene, alone or in combination, as potential anticancer therapeutics using the human liver and breast cancer cell lines HepG2 and MCF-7. STUDY DESIGN: An integrated systems pharmacology approach is presented for illustrating the molecular interactions between metformin and D-limonene. METHODS: We applied a systems-based analysis to introduce a drug-target-pathway network that clarifies different mechanisms of treatment. The combination treatment of metformin and D-limonene induced apoptosis in both cell lines compared with single drug treatments, as indicated by flow cytometric and gene expression analysis. RESULTS: The mRNA expression of Bax and P53 genes were significantly upregulated while Bcl-2, iNOS, and Cox-2 were significantly downregulated in all treatment groups compared with normal cells. The percentages of late apoptotic HepG2 and MCF-7 cells were higher in all treatment groups, particularly in the combination treatment group. Calculations for the combination index (CI) revealed a synergistic effect between both drugs for HepG2 cells (CI = 0.14) and MCF-7 cells (CI = 0.22). CONCLUSION: Our data show that metformin, D-limonene, and their combinations exerted significant antitumor effects on the cancer cell lines by inducing apoptosis and modulating the expression of apoptotic genes.


Asunto(s)
Apoptosis , Neoplasias de la Mama , Proliferación Celular , Limoneno , Neoplasias Hepáticas , Metformina , Humanos , Metformina/farmacología , Limoneno/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Células Hep G2 , Células MCF-7 , Terpenos/farmacología , Femenino , Antineoplásicos/farmacología , Ciclohexenos/farmacología
3.
Sci Rep ; 14(1): 1510, 2024 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-38233443

RESUMEN

Breast cancer ranks as the second leading most significant of mortality for women. Studies have demonstrated the potential benefits of natural compounds in cancer treatment and prevention, either in isolation or in conjunction with chemotherapy. In order to improve Tamoxifen's therapeutic efficacy in in-vivo studies, our research sought to determine the effects of hesperidin, piperine, and bee venom as natural compounds, as well as their combination effect with or without Tamoxifen. First, 132 female albino rats were equally divided into six groups and five subgroups, and breast cancer was induced in the selected groups by xenografting of MCF7 cells. Second, the effect of single and best ratio combinations treatment from previous in vitro studies were selected. Next, tumorous mammary glands were collected for apoptotic and antiapoptotic biomarkers and cell cycle analysis. Single or combined natural products with or without Tamoxifen revealed a significant up-regulation in apoptotic genes Bax and Casp3 and a downregulation of antiapoptotic and angiogenesis genes Bcl-2 and VEGF genes. We found that cell cycle arrest in the G0/G1 phase was exclusively caused by Tamoxifen and/ or hesperidin. However, the cell cycle arrest in the G2/M phase is a result of the combination of piperine and bee venom, with or without Tamoxifen by using the flow cytometric technique. Our research concludes that bee venom, hesperidin, and piperine can synergistically enhance to increase Tamoxifen's efficiency in the management of breast cancer.


Asunto(s)
Alcaloides , Venenos de Abeja , Benzodioxoles , Neoplasias de la Mama , Hesperidina , Piperidinas , Alcamidas Poliinsaturadas , Humanos , Femenino , Ratas , Animales , Tamoxifeno/farmacología , Tamoxifeno/uso terapéutico , Hesperidina/farmacología , Hesperidina/uso terapéutico , Células MCF-7 , Venenos de Abeja/farmacología , Venenos de Abeja/uso terapéutico , Angiogénesis , Apoptosis , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Biomarcadores
4.
Fundam Clin Pharmacol ; 38(3): 550-560, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38258539

RESUMEN

BACKGROUND: Due to the limited success in the treatment of lung adenocarcinomas, new treatment protocols are urgently needed to increase the curability rate and the survival of lung cancer patients. OBJECTIVES: Although statins, like atorvastatin (Ator), and metformin (Met) are widely accepted as hypolipidemic and hypoglycemic drugs, respectively, there are many predictions about their enhancing antitumor effect when they are combined with traditional chemotherapeutics. METHODS: The individual and combined antiproliferative potential of Ator and Met was tested by MTT-assay in non-small cell lung cancer (NSCLC) A549 cell line, compared to the corresponding effect of Gemcitabine (Gem) with implication on the mechanisms of action. RESULTS: Initially, both drugs demonstrated concentration-dependent cytotoxicity in A549 cells. Also, their combination index (CI) indicated their synergistic effect at equi-IC50 concentration (CI = 0.00984). Moreover, Ator and/or Met-treated cells revealed disrupted patterns of SOD, CAT, GSH, MDA, and TAC, developed apoptosis, and larger fractions of the cell population were arrested in G0/G1 phase, particularly in cells dually-treated both Ator and Met. These observations were accompanied by downregulation in the expression of iNOS, HO-1, and the angiogenic marker VEGF, meanwhile, an altered expression of MAPK and AMPK was observed. CONCLUSION: Conclusively, these data suggest that repurposing of Ator and Met demonstrates their individual and combined antiproliferative effect in non-small cell lung cancer and they may adopt a similar mechanism of action.


Asunto(s)
Apoptosis , Atorvastatina , Carcinoma de Pulmón de Células no Pequeñas , Proliferación Celular , Reposicionamiento de Medicamentos , Sinergismo Farmacológico , Neoplasias Pulmonares , Metformina , Humanos , Metformina/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Atorvastatina/farmacología , Proliferación Celular/efectos de los fármacos , Células A549 , Apoptosis/efectos de los fármacos , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Gemcitabina , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Hipoglucemiantes/farmacología
5.
Int J Mol Sci ; 24(4)2023 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-36834960

RESUMEN

Bevacizumab (Bev) a humanized monoclonal antibody that fights vascular endothelial growth factor A (VEGF-A). It was the first specifically considered angiogenesis inhibitor and it has now become the normative first-line therapy for advanced non-small-cell lung cancer (NSCLC). In the current study, polyphenolic compounds were isolated from bee pollen (PCIBP) and encapsulated (EPCIBP) inside moieties of hybrid peptide-protein hydrogel nanoparticles in which bovine serum albumin (BSA) was combined with protamine-free sulfate and targeted with folic acid (FA). The apoptotic effects of PCIBP and its encapsulation (EPCIBP) were further investigated using A549 and MCF-7 cell lines, providing significant upregulation of Bax and caspase 3 genes and downregulation of Bcl2, HRAS, and MAPK as well. This effect was synergistically improved in combination with Bev. Our findings may contribute to the use of EPCIBP simultaneously with chemotherapy to strengthen the effectiveness and minimize the required dose.


Asunto(s)
Antineoplásicos , Bevacizumab , Productos Biológicos , Carcinoma de Pulmón de Células no Pequeñas , Hidrogeles , Animales , Humanos , Células A549/efectos de los fármacos , Células A549/metabolismo , Inhibidores de la Angiogénesis/uso terapéutico , Antineoplásicos/química , Antineoplásicos/farmacología , Abejas/química , Abejas/metabolismo , Bevacizumab/uso terapéutico , Productos Biológicos/química , Productos Biológicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Hidrogeles/química , Hidrogeles/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Células MCF-7/efectos de los fármacos , Células MCF-7/metabolismo , Nanopartículas/química , Nanopartículas/uso terapéutico , Polen/química , Polen/metabolismo , Factor A de Crecimiento Endotelial Vascular/uso terapéutico
6.
Environ Anal Health Toxicol ; 38(4): e2023023-0, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38298042

RESUMEN

A quantitative assessment of silver nanoparticles (AgNPs) in fluids and some organs of pregnant rats as well as their fetal blood were carried out in this study. A single oral dose (1mg/kg) of AgNPs with a size range of 4-20 nm was administered to pregnant rats on the 19th of gestation. Five groups were euthanized after 10 min, 1, 6, 12, and 24 hr as well as the control group. Total Silver (Ag) contents were measured in bloods (maternal and fetal) and several organs using Inductive Coupled Plasma Optical Emission Spectroscopy (ICP-OES) followed by acid digestion. In maternal blood, AgNPs were found to increase time-dependently after 12 and 24 hr into 0.135 and 0.224 µg/ml, but it was slightly higher in fetal blood (0.32 and 0.31 µg/ml) after 10 min and 1 hr. In other samples: kidneys, liver, spleen, placenta, and uterus the data indicated that NPs were rapidly absorbed from the dosing site (gastrointestinal tract) as evidenced by the detection of Ag in the analyzed samples (fluids and tissues). On the other hand, the cumulative percentages of excretion level in urine was 8.25% which was higher than in feces (4.77%) after 24 hr. These findings indicate the ability of AgNPs to accumulate in pregnant rats and transfer to their fetus imposing adverse outcomes and male formation. Thus, further investigations must be followed for direct and/or indirect exposure to such NPs before decision for their practices.

7.
Nanomaterials (Basel) ; 14(1)2023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-38202484

RESUMEN

The widespread biomedical and commercial applications of silver nanoparticles (AgNPs) have increased their potential for human and environmental exposure and toxicity to human health. The bio-distribution and toxicity of AgNPs in rodents following inhalation, intratracheal instillation, and oral ingestion are well documented; however, little is known about the bio-distribution of intravenously (IV)-administered AgNPs and their organ-specific pathophysiological effects. Here, we investigate the pharmacokinetic pattern and tissue distribution of AgNPs in male rats following IV administration. The animals were humanely sacrificed after 10 min, 1 h, 6 h, 12 h, 24 h, and 168 h of AgNP administration, and the silver (Ag) content was measured from blood samples and various tissues following acid digestion. The AgNPs were readily absorbed and subsequently distributed into most organs predominantly in the colon, small intestine, kidney, and heart after 6 h; however, they were the highest in the spinal cord after 168 h. White blood cells (WBCs) were significantly increased (42-60%) in AgNP-administered animals at all time points except 10 min. Regarding platelets, all AgNP-administered animals showed counts 7.8-39.2% lower, with the lowest count at 168 h post-administration. In the case of lymphocytes (LYMs), the AgNP-treated animals exhibited a count 19.5-41% lower at 10 min and 1 h post-administration; however, the animals at 168 h post-administration showed a count 30.5% more. The mean corpuscular hemoglobin (MCH) counts from the AgNP-treated animals were decreased by 50-62%. The concentrations of aspartate transaminase (AST), urea, and creatinine were increased in the AgNP-treated animals. Taken together, the results suggest that the acute IV administration of AgNPs alters metabolic and hematological parameters in animals and may pose a health risk to humans.

8.
Asian Pac J Cancer Prev ; 23(8): 2607-2615, 2022 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-36037113

RESUMEN

OBJECTIVE: Uterine or endometrial cancer affects many women postmenopausal and may reach an advanced stage before signs and symptoms can be noticed. Micro RNAs (miRNAs), non-coding RNAs, play key roles in gene expression regulation and are linked to cancer. This study aimed to elucidate whether some specific types of miRNAs (miRNA133-a, miRNA-21, miRNA-205) can act as prognostic or diagnostic biomarkers for endometrial carcinoma (ER) in Egyptian patients. METHODS: Blood samples from 36 patients suffering from endometrial carcinoma and 15 healthy volunteers were tested for expression levels of miRNA 133a-2, 21 and 205. RESULTS: The expression levels of miRNA133a-2, miRNA-21, and miRNA-205 were significantly elevated in ER patients when compared with the control group, the highest levels were noticed in miRNA133a-2. The CA125 levels were significantly higher in all patients as compared with healthy subjects. CONCLUSION: The findings could support the use of circulating miR133a-2, miR-21 and miR-205 as virtuous prognostic biomarkers for EC in Egyptian patients. The studied miRNA species warrant validation for prospective targeting inhibitory protocols in EC.


Asunto(s)
Neoplasias Endometriales , MicroARNs , Biomarcadores de Tumor/genética , Carcinogénesis/genética , Egipto/epidemiología , Neoplasias Endometriales/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/genética , Pronóstico , Estudios Prospectivos
9.
Nutr Cancer ; 74(9): 3388-3402, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35412401

RESUMEN

Gemcitabine is a chemotherapeutic drug used to treat cancer; however, it has severe side effects. Therefore, we evaluated the anticancer potency of balanitoside, a folk medicine isolated from the edible fruits of Balanites aegyptiaca, using a mouse model of lung cancer induced by Urethane/butylated hydroxytoluene, either alone or in combination with gemcitabine. The results indicated that balanitoside, when administered alone or in combination with gemcitabine, exhibited antitumor activity against lung cancer by reducing tumor incidence, multiplicity, and average tumor size. It also decreased the proliferation of tumor cells, induced apoptosis, triggered cell cycle arrest at the G0/G1 phase, and caused a marked reduction in cancer stem cell markers, aldehyde dehydrogenase (ALDH-1) levels, and the CD133 (+ve) cell population. Balanitoside also modulated the levels of oxidative stress markers in lung tissues. The results indicate that balanitoside enhances the antitumor activity of gemcitabine and may represent a natural adjuvant medication for lung cancer.


Asunto(s)
Desoxicitidina , Neoplasias Pulmonares , Apoptosis , Línea Celular Tumoral , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Sinergismo Farmacológico , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Modelos Teóricos , Saponinas , Esteroides , Gemcitabina
10.
Asian Pac J Cancer Prev ; 22(12): 3735-3740, 2022 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-34973682

RESUMEN

The journal of APJCP (Asian Pacific Journal of Cancer Prevention) focuses to gather relevant and up-to-date novel information's related to cancer sciences. The research methodologies and approaches adopted by the researcher are prone to variation which may be desirable in the context of novel scientific findings however, the reproducibility for these studies needs to be unified and assured. The reproducibility issues are highly concerned when preclinical studies are reported in cancer, for natural products in particular. The natural products and medicinal plants are prone to a wide variation in terms of phytochemistry and phyto-pharmacology, ultimately affecting the end results for cancer studies. Hence the need for specific guidelines to adopt a best-practice in cancer research are utmost essential. The current AIMRDA guidelines aims to develop a consensus-based tool in order to enhance the quality and assure the reproducibility of studies reporting natural products in cancer prevention. A core working committee of the experts developed an initial draft for the guidelines where more focus was kept for the inclusion of specific items not covered in previous published tools. The initial draft was peer-reviewed, experts-views provided, and improved by a scientific committee comprising of field research experts, editorial experts of different journals, and academics working in different organization worldwide. The feedback from continuous online meetings, mail communications, and webinars resulted a final draft in the shape of a checklist tool, covering the best practices related to the field of natural products research in cancer prevention and treatment. It is mandatory for the authors to read and follow the AIMRDA tool, and be aware of the good-practices to be followed in cancer research prior to any submission to APJCP. Though the tool is developed based on experts in the field, it needs to be further updated and validated in practice via implementation in the field.


Asunto(s)
Antineoplásicos , Productos Biológicos , Políticas Editoriales , Revisión por Pares/normas , Proyectos de Investigación/normas , Consenso , Humanos , Reproducibilidad de los Resultados
11.
Arch Physiol Biochem ; 128(4): 970-978, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32212969

RESUMEN

Gemcitabine was loaded in albumin nanoparticles then coated with chitosan. The diameter of GEM-ANPs/CS was 200 ± 4 nm. Gemcitabine was loaded in GEM-ANPs/CS with an efficacy of 75%. The IC50 of GEM-ANPs/CS was found to be 12.98 and 6.08 µg/ml after incubation for 48 and 72 h with MCF-7 cells, respectively. Treatment of MCF-7 cells with IC50 of GEM-ANPS, and GEM-ANPS/CS resulted in membrane damage which led to elevated LDH activity of 4 and 3.4, and increasing GSH level of 4.6 and 9.3, respectively, when compared with untreated cells. DNA fragmentation and up-regulated of caspase-3 and p53 had illustrated the apoptotic effect of MCF-7 treated with GEM-ANPS/CS. The tumour suppressor RRM1 gene expression was down-regulated in MCF-7 cells treated with GEM-ANPS/CS. The modified ANPs coated with chitosan may be used as a promising nanomatrix for gemcitabine delivery and targeting to improve its therapeutic index against MCF-7 cells.


Asunto(s)
Quitosano , Nanopartículas , Humanos , Albúminas/farmacología , Línea Celular Tumoral , Desoxicitidina/análogos & derivados , Gemcitabina
12.
Drug Metab Pers Ther ; 36(4): 299-309, 2021 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-34773731

RESUMEN

OBJECTIVES: This work was designed to compare the sensitizing effects of epigenetic modifiers on cancer cells vs. that of glucocorticoids. Also, to evaluate their effects on genes involved in epigenetic changes and drug metabolism. METHODS: Hepatoma cells (HepG2) were treated with the anticancer drug (Taxol), with a histone deacetylase inhibitor (Trichostatin A [TSA]), DNA methyltransferase inhibitor (5-Aza-dC) or dexamethasone (DEX). Cytotoxicity was assessed by MTT assay and the apoptosis was determined by Annexin V-FITC. The expression levels of HDAC1, HDAC3, Dnmt1, Dnmt3α, CYP1A2, CYP3A4, CYP2B6, CYP2C19 and CYP2D6 were monitored by qRT-PCR. RESULTS: TSA, synergistically enhanced cells sensitivity with the anticancer effect of Taxol more than 5-Aza-dC and DEX. This was evidenced by the relative decrease in IC50 in cells cotreated with Taxol + TSA, Taxol + 5-Aza-dC or Taxol + DEX. Apoptosis was induced in 51.2, 16.9 and 41.3% of cells, respectively. In presence of Taxol, TSA induced four-fold increase in the expression of HDAC1 and downregulated Dnmt1&3α genes. CYP2D6 demonstrated progressive expression (up to 28-fold) with the increasing number of drugs. Moreover, the isoform overexpressed in cells treated with TSA + Taxol > DEX + Taxol > 5-Aza-dC + Taxol (6.4, 4.6 and 2.99, respectively). The investigated genes were clustered in two distinct subsets, where no coregulation was observed between HDAC1 and HDAC3. However, tight pairwise correlation-based cluster was seen between (CYP3A4/Dnmt3α and CYP2D6/CYP2C19). CONCLUSIONS: The data reflects the sensitizing effect of acetylation modification by TSA on the responsiveness of hepatoma cells to anticancer therapy. The effect of histone deacetylase inhibition was more than hypomethylation and glucocorticoid effects. TSA exerts its role through its modulatory role on epigenetics and drugs metabolizing genes. Other modifiers (5-Aza-dC and DEX), however may adopt different mechanisms.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Carcinoma Hepatocelular/tratamiento farmacológico , Decitabina , Dexametasona/farmacología , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ácidos Hidroxámicos , Neoplasias Hepáticas/tratamiento farmacológico , Paclitaxel/farmacología
13.
Drug Metab Pers Ther ; 2021 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-33818027

RESUMEN

OBJECTIVES: This work was designed to compare the sensitizing effects of epigenetic modifiers on cancer cells vs. that of glucocorticoids. Also, to evaluate their effects on genes involved in epigenetic changes and drug metabolism. METHODS: Hepatoma cells (HepG2) were treated with the anticancer drug (Taxol), with a histone deacetylase inhibitor (Trichostatin A [TSA]), DNA methyltransferase inhibitor (5-Aza-dC) or dexamethasone (DEX). Cytotoxicity was assessed by MTT assay and the apoptosis was determined by Annexin V-FITC. The expression levels of HDAC1, HDAC3, Dnmt1, Dnmt3α, CYP1A2, CYP3A4, CYP2B6, CYP2C19 and CYP2D6 were monitored by qRT-PCR. RESULTS: TSA, synergistically enhanced cells sensitivity with the anticancer effect of Taxol more than 5-Aza-dC and DEX. This was evidenced by the relative decrease in IC50 in cells cotreated with Taxol + TSA, Taxol + 5-Aza-dC or Taxol + DEX. Apoptosis was induced in 51.2, 16.9 and 41.3% of cells, respectively. In presence of Taxol, TSA induced four-fold increase in the expression of HDAC1 and downregulated Dnmt1&3α genes. CYP2D6 demonstrated progressive expression (up to 28-fold) with the increasing number of drugs. Moreover, the isoform overexpressed in cells treated with TSA + Taxol > DEX + Taxol > 5-Aza-dC + Taxol (6.4, 4.6 and 2.99, respectively). The investigated genes were clustered in two distinct subsets, where no coregulation was observed between HDAC1 and HDAC3. However, tight pairwise correlation-based cluster was seen between (CYP3A4/Dnmt3α and CYP2D6/CYP2C19). CONCLUSIONS: The data reflects the sensitizing effect of acetylation modification by TSA on the responsiveness of hepatoma cells to anticancer therapy. The effect of histone deacetylase inhibition was more than hypomethylation and glucocorticoid effects. TSA exerts its role through its modulatory role on epigenetics and drugs metabolizing genes. Other modifiers (5-Aza-dC and DEX), however may adopt different mechanisms.

14.
Asian Pac J Cancer Prev ; 21(9): 2739-2750, 2020 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-32986376

RESUMEN

OBJECTIVE: In search for a unique natural combination of highly active biological components for treatment against colon cancer, we used aqueous extract of Ascidia, Styela plicata (ASCex), a marine invertebrate depending on its richness of high levels of biologically active components as indicated in our previous studies, against rat colon cancer, exploring its underlying mechanisms. METHODS: Rats chemically initiated for colon cancer were either non-treated or post-treated with highly saturated ASCex for 32 weeks after initiation, other groups of rats were administered ASCex without cancer initiation or served as normal controls. RESULTS: Rats treated with ASCex alone did not show any signs of non-favored health conditions. Treatment with ASCex after cancer initiation has significantly reduced the average incidences, multiplicities and volumes of colon tumors (adenomas and adenocarcinomas) as compared with the non-treated cancer group. ASCex has also significantly reduced the total numbers of aberrant crypt foci (ACF), surrogate biomarkers for colon cancer as compared with the non-treated cancer group. Moreover, anti-proliferative celluar nucular antigen (PCNA) immunohistochemical staining revealed that ASCex exerted significant antiproliferative characteristics in the carcinogen-treated colonic mucosa as compared with its corresponding control. Also, treatment with ASCex has markedly down-regulated the mRNA expression levels of Nuclear Factor-kappa B (NF-κB), a nuclear transcriptional activator as well as the mRNA expression of the cytoplasmic SOD1 gene which encodes Cu/Zn SOD, the first line defense against superoxide radicals. CONCLUSION: Collectively, ASCex could act as a potent chemotherapeutic drug against colon cancer, likely through the influence of its rich active metabolites which interfere with various biological pathways including inhibition of protein synthesis during cellular growth and marked induction of antioxidative capacity in the colonic mucosa. This role has been extensively discussed herein.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , FN-kappa B/metabolismo , Extractos Vegetales/farmacología , Superóxido Dismutasa-1/metabolismo , Urocordados/química , Animales , Azoximetano/toxicidad , Carcinogénesis/inducido químicamente , Carcinogénesis/metabolismo , Carcinogénesis/patología , Carcinógenos/toxicidad , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Citoplasma/enzimología , Masculino , FN-kappa B/genética , Ratas , Ratas Sprague-Dawley , Superóxido Dismutasa-1/genética
15.
J Environ Pathol Toxicol Oncol ; 38(4): 329-343, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32464004

RESUMEN

This study was designated to explore the role of cancer stem cells (CSCs) during chemically induced mouse colon carcinogenesis (by 1,2- dimethylhydrazine dihydrochloride, DMH) with/or without the treatment with a targeted (anti-COX-2) therapeutic drug, celecoxib. Two experiments were conducted. The first, a short-term, 16-week mouse colon carcinogenesis bioassay, demonstrates the early stages of colon carcinogenesis. The other is a medium-term, 32-week mouse colon cancer experiment that mimics an end point of colon malignancy. Colon tumors were detected in animals after 32 weeks; histopathologically, they varied from benign hyperplastic polyps and adenomas to dysplastic polyps, adenocarcinomas, and invasive carcinomas. The overall colon tumor incidences, multiplicities, and volumes were obviously reduced when treated with celecoxib after DMH initiation. The immunohistochemical (IHC) labeling indexes (L1%) of the proliferating cell nuclear antigen (PCNA) were lower in the colonic epithelium in both experiments after treatment with celecoxib. Also, the IHC expression patterns of CD133 and CD44, known to associate CSCs, showed differential changes depending on the end-point stage of carcinogenesis and celecoxib treatment. Moreover, the biochemical aldehyde dehydrogenase-1 (ALDH-1) activity levels, a known CSC marker in colonic epithelia, were downregulated after 16 weeks but were upregulated after 32 weeks. Flow cytometric analysis showed that numbers of CD133 cells increased in the colonic epithelia of mice after 16 weeks, while the numbers of CD44 but not CD133 cells increased after 32 weeks. Treatment with celecoxib after DMH induced significant increase in apoptotic cell numbers by 47% after 16 weeks, but these numbers had not changed after 32 weeks compared with the corresponding group treated DMH only. Thus, the specific markers and CSC populations targeted by this drug may vary depending on the genetic and phenotypic stages of carcinogenesis. This drug could be useful during targeted therapy for colon cancer patients.


Asunto(s)
Celecoxib/farmacología , Neoplasias del Colon/metabolismo , Inhibidores de la Ciclooxigenasa 2/farmacología , 1,2-Dimetilhidrazina/toxicidad , Animales , Carcinogénesis , Carcinógenos , Masculino , Ratones , Células Madre Neoplásicas , Antígeno Nuclear de Célula en Proliferación
16.
Int J Biol Macromol ; 120(Pt A): 1170-1180, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30172815

RESUMEN

Methotrexate (MTX) is effective therapeutic agent treated many tumors and autoimmune diseases. The aim of our study was to design an effective delivery nanocarrier for methotrexate to improve stability and biodistribution, reduce adverse effects and maximize clinical efficacy. Magnetite nanoparticles (Fe3O4-NPs) were synthesized using Pterocladiella. The size of Fe3O4-NPs, CS-Fe3O4-NPs and MTX/CS-Fe3O4-NPs were 37.6, 61.4 and 150 nm respectively. Methotrexate loading efficiency was 74.15% of total amount of MTX loaded on CS-Fe3O4-NPs and 39.8% of the loaded drug was initially released and the remaining amount was released through 120 h. The IC50 of MTX and MTX/CS-Fe3O4-NPs was 51.4 and 9.7 µg/ml respectively after 72 h. MTX/CS-Fe3O4-NPs caused remarkable damage to the membrane of MCF-7 cells led to increasing the LDH activity 5 fold in MCF-7 cells as compared with MTX treated once. DNA fragmentation and caspase-3 activity were higher in MCF-7 cells treated with MTX/CS-Fe3O4-NPs than that of MTX. Up-regulation of caspase3 and DHFR genes expression was observed in the treatment with MTX/CS-Fe3O4-NPs. The loading of MTX on chitosan coated Fe3O4-NPs improves the release and anticancer efficacy of MTX for effective cancer treatment.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Nanopartículas de Magnetita/administración & dosificación , Metotrexato/administración & dosificación , Neoplasias de la Mama/patología , Quitosano/química , Femenino , Humanos , Hierro/química , Células MCF-7 , Nanopartículas de Magnetita/química , Metotrexato/química
17.
Biomed Pharmacother ; 105: 1335-1343, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30021371

RESUMEN

Despite advances in cancer treatment, breast cancer remains one of the main life threatening diseases in women. Most anti-breast cancer drugs cause severe health complications and multidrug resistance. Although, some natural products, such as hesperidin (Hes), piperine (Pip) and bee venom (BV), showed anti-breast cancer effect when used separately, their combined effect together or with the anti-cancer drug tamoxifen (Tam) has not yet been studied. Herein, we hypothesized that these three natural products could potentiate the therapeutic effect of Tam when used together. First, we studied the cytotoxic effect of Hes, Pip, and BV on MCF7 and T47D cells using MTT assay and found reasonable IC50 comparable to that of Tam. Second, we checked the effect of all combinations (n = 67 for each cell line, prepared as non-constant ratio from fractions of IC50 of the four compounds) and found enhanced anti-proliferative effects on MCF7 and T47D and synergistic effect, revealed by combination index (CI) values below one. Next, the best 5 combinations with lowest Tam doses and CI but with highest cell death were selected for further molecular analysis in comparison to single-drug treatment. All single- and combined-treated groups showed a significant increase in apoptosis (indicated by upregulated mRNA level of the pro-apoptotic marker Bax and downregulated mRNA level of the anti-apoptotic marker Bcl2) and a significant decrease in mRNA level of the two breast cancer related receptors EGFR and ERα, with the best effect in combined groups especially that contained the 4 compounds, as compared to vehicle-treated group. Moreover, Pip, BV and all combinations, except Tam + Hes group, arrested MCF7 and T47D in G2/M phase of cell cycle, while Tam and/or Hes caused G0/G1 phase arrest. These results indicate that Hes, Pip and BV synergistically enhance the anti-cancer effect of Tam and could be used as safe adjuvant/vehicle to Tam in treatment of breast cancer after further confirmatory in vivo investigations.


Asunto(s)
Alcaloides/farmacología , Antineoplásicos/farmacología , Venenos de Abeja/farmacología , Benzodioxoles/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Hesperidina/farmacología , Piperidinas/farmacología , Alcamidas Poliinsaturadas/farmacología , Tamoxifeno/farmacología , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Sinergismo Farmacológico , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Células MCF-7 , ARN Mensajero/metabolismo , Regulación hacia Arriba/efectos de los fármacos
18.
Acta Parasitol ; 63(1): 198-209, 2018 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-29351079

RESUMEN

Accumulating evidence suggest that some infectious agents may interfere in the natural progression of neoplasia. This study examined the association between chronic infection with adult Syphacia muris parasites and 1,2-dimethylhydrazine (DMH)-induced colorectal carcinogenesis in rats. In addition, the conceivable therapeutic effect of Bryostatin-1, a potent extract of the marine Bryozoan, Bugulane ritina, was investigated against this combined effect.DMH administration has induced aberrant crypt foci (ACF), surrogate biomarkers for colorectal carcinogenesis, while the S. muris infection combined with DMH has significantly increased the total numbers of ACF. Nonetheless, treatment with Bryostatin-1 after infection has significantly reduced the ACF numbers particularly larger ones. This inhibition was concomitant with significant inhibition in the immunohistochemical levels of the ki67, Caspase-3 and IgM levels in colorectal epithelium, as well as serum levels of IgM and IgG. Additionally, treatment with Bryostatin-1 after S. muris + DMH has modulated enzymatic antioxidative markers levels of superoxide dismutase and catalase as well as the non-enzymatic antioxidant markers levels of reduced glutathione, lipid peroxidation, nitric oxide and total antioxidant capacity. Further, treatment with Bryostatin-1 has down-regulated the mRNA expression levels of COX-2 and APC genes in colorectal mucosa. In conclusion, infection with S. muris during colorectal carcinogenesis has significantly modulated the oxidative stress markers in the colorectum, while treatment with Bryostatin-1 has exerted significant curative potential. A mechanism could be explained that Bryostatin-1 treatment has reduced oxidative stress markers activities along with affecting host to parasite immunity possibly leading to changes in the COX-2 and APC expression, retarding cellular proliferation and subsequently reducing the colorectal carcinogenesis events.


Asunto(s)
Antineoplásicos/administración & dosificación , Brioestatinas/administración & dosificación , Carcinogénesis , Neoplasias Colorrectales/fisiopatología , Estrés Oxidativo , Oxiuriasis/parasitología , Oxyuroidea/fisiología , Animales , Biomarcadores de Tumor/análisis , Neoplasias Colorrectales/inducido químicamente , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Expresión Génica , Inmunohistoquímica , Oxiuriasis/complicaciones , Ratas , Resultado del Tratamiento
19.
Biomed Pharmacother ; 93: 1310-1319, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28747012

RESUMEN

Grape seed proanthocyanidin extract (GSPE) is known to be effective on broad spectrum of biological pathways in living organisms including oxidative stress. The present study aimed to investigate the effects of proanthocyanidin on preneoplastic lesions and liver cancer induced in rats by Diethylnitrosamine (DEN). 7-8 Week old male Sprague Dawley (S.D.) rats were divided into six groups: The 1st group received no treatment and were -ve controls, the 2nd were treated with a single dose of DEN 200mg/kg intraperitoneally (i.p.) and served as +ve control group. The 3rd and 4th groups were injected with the same dose of DEN as in group 2 and then post treated with 300 or 150mg/kg/b.wt./day GSPE by intrgastroluminal gavage (i.g.) respectively until the end after the 22 weeks. Groups 5 and 6 were treated with the same doses of GSPE as in groups 3 and 4 respectively without DEN administration. The results showed that the immunohistochemical Proliferating Cell Nuclear Antigen (PCNA) labeling indexes (PCNA LI%) were significantly inhibited in liver tissues and tumors by both treatments of GSPE. Furthermore, treatment with GSPE has modified the liver tissue oxidative stress markers levels of SOD, CAT, GSH, GST, GPx, GR and MDA changed by DEN. In conclusion, GSPE has a sufficient therapeutic effect against liver carcinogenesis through their free radical scavenging, inhibition of cellular proliferation.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Extracto de Semillas de Uva/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Extractos Vegetales/farmacología , Proantocianidinas/farmacología , Animales , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Masculino , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
20.
Asian Pac J Cancer Prev ; 17(3): 1023-35, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27039721

RESUMEN

The purpose of this study was to investigate the role of colon cancer stem cells (CSCs) during chemicallyinduced rat multi-step colon carcinogenesis with or without the treatment with a specific cyclooxygenase-2 inhibitor drug (celecoxib). Two experiments were performed, the first, a short term 12 week colon carcinogenesis bioassay in which only surrogate markers for colon cancer, aberrant crypt foci (ACF) lesions, were formed. The other experiment was a medium term colon cancer rat assay in which tumors had developed after 32 weeks. Treatment with celecoxib lowered the numbers of ACF, as well as the tumor volumes and multiplicities after 32 weeks. Immunohistochemical proliferating cell nuclear antigen (PCNA) labeling indexes LI (%) were downregulated after treatment by celecoxib. Also different cell surface antigens known to associate with CSCs such as the epithelial cell adhesion molecule (EpCAM), CD44 and CD133 were compared between the two experiments and showed differential expression patterns depending on the stage of carcinogenesis and treatment with celecoxib. Flow cytometric analysis demonstrated that the numbers of CD133 cells were increased in the colonic epithelium after 12 weeks while those of CD44 but not CD133 cells were increased after 32 weeks. Moreover, aldehyde dehydrogenase-1 activity levels in the colonic epithelium (a known CSC marker) detected by ELISA assay were found down-regulated after 12 weeks, but were up-regulated after 32 weeks. The data have also shown that the protective effect of celecoxib on these specific markers and populations of CSCs and on other molecular processes such as apoptosis targeted by this drug may vary depending on the genetic and phenotypic stages of carcinogenesis. Therefore, uncovering these distinction roles of CSCs during different phases of carcinogenesis and during specific treatment could be useful for targeted therapy.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Carcinogénesis/metabolismo , Celecoxib/farmacología , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Familia de Aldehído Deshidrogenasa 1 , Animales , Bioensayo/métodos , Biomarcadores de Tumor/metabolismo , Carcinogénesis/patología , Colon/efectos de los fármacos , Colon/metabolismo , Colon/patología , Neoplasias del Colon/patología , Inhibidores de la Ciclooxigenasa 2/farmacología , Regulación hacia Abajo/efectos de los fármacos , Receptores de Hialuranos/metabolismo , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Isoenzimas/metabolismo , Masculino , Células Madre Neoplásicas/patología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ratas , Ratas Sprague-Dawley , Retinal-Deshidrogenasa/metabolismo , Lectina 3 Similar a Ig de Unión al Ácido Siálico/metabolismo , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA