Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Sci Rep ; 14(1): 11928, 2024 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-38789508

RESUMEN

Cancer stands as one of the most impactful illnesses in the modern world, primarily owing to its lethal consequences. The fundamental concern in this context likely stems from delayed diagnoses in patients. Hence, detecting various forms of cancer is imperative. A formidable challenge in cancer research has been the diagnosis and treatment of this disease. Early cancer diagnosis is crucial, as it significantly influences subsequent therapeutic steps. Despite substantial scientific efforts, accurately and swiftly diagnosing cancer remains a formidable challenge. It is well known that the field of cancer diagnosis has effectively included electrochemical approaches. Combining the remarkable selectivity of biosensing components-such as aptamers, antibodies, or nucleic acids-with electrochemical sensor systems has shown positive outcomes. In this study, we adapt a novel electrochemical biosensor for cancer detection. This biosensor, based on a glassy carbon electrode, incorporates a nanocomposite of reduced graphene oxide/Fe3O4/Nafion/polyaniline. We elucidated the modification process using SEM, TEM, FTIR, RAMAN, VSM, and electrochemical methods. To optimize the experimental conditions and monitor the immobilization processes, electrochemical techniques such as CV, EIS, and SWV were employed. The calibration graph has a linear range of 102-106 cells mL-1, with a detection limit of 5 cells mL-1.


Asunto(s)
Compuestos de Anilina , Biomarcadores de Tumor , Técnicas Biosensibles , Neoplasias de la Mama , Técnicas Electroquímicas , Polímeros de Fluorocarbono , Grafito , Receptor ErbB-2 , Grafito/química , Humanos , Técnicas Biosensibles/métodos , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/patología , Técnicas Electroquímicas/métodos , Compuestos de Anilina/química , Polímeros de Fluorocarbono/química , Línea Celular Tumoral , Receptor ErbB-2/metabolismo , Receptor ErbB-2/análisis , Femenino , Óxido Ferrosoférrico/química , Límite de Detección , Electrodos
2.
bioRxiv ; 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38617302

RESUMEN

Modern cancer treatment approaches aim at achieving cancer remission by using targeted and personalized therapies, as well as harnessing the power of the immune system to recognize and eliminate the cancer cells. To overcome a relatively short-lived response due to the development of resistance to the administered drugs, combination therapies have been pursued, as well. To expand the outlook of combination therapies, the objective of this study was to use high-throughput data generation technologies such as mass spectrometry and proteomics to investigate the response of HER2+ breast cancer cells to a mixture of two kinase inhibitors that has not been adopted yet as a standard treatment regime. The broader landscape of biological processes that are affected by inhibiting two major pathways that sustain the growth and survival of cancer cells, i.e., EGFR and PI3K/AKT, was investigated by treating SKBR3/HER2+ breast cancer cells with Lapatinib or a mixture of Lapatinib/Ipatasertib small molecule drugs. Changes in protein expression and/or activity in response to the drug treatments were assessed by using two complementary quantitative proteomic approaches based on peak area and peptide spectrum match measurements. Over 900 proteins matched by three unique peptide sequences (FDR<0.05) were affected by the exposure of cells to the drugs. The work corroborated the anti-proliferative activity of Lapatinib and Ipatasertib, and, in addition to cell cycle and growth arrest processes enabled the identification of several multi-functional proteins with roles in cancer-supportive hallmark processes. Among these, immune response, adhesion and migration emerged as particularly relevant to the ability to effectively suppress the proliferation and dissemination of cancer cells. The supplementation of Lapatinib with Ipatasertib further affected the expression or activity of additional transcription factors and proteins involved in gene expression, trafficking, DNA repair, and development of multidrug resistance. Furthermore, over fifty of the affected proteins represented approved or investigational targets in the DrugBank database, which through their protein-protein interaction networks can inform the selection of effective therapeutic partners. Altogether, our findings exposed a broad plethora of yet untapped opportunities that can be further explored for enhancing the anti-cancer effects of each drug as well as of many other multi-drug therapies that target the EGFR/ERBB2 and PI3K/AKT pathways. The data are available via ProteomeXchange with identifier PXD051094.

3.
Stud Health Technol Inform ; 308: 329-340, 2023 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-38007757

RESUMEN

Various damage-associated molecular patterns (DAMPs) associated with immunogenic cell death (ICD) have been discovered, potentially leading to cancer cell elimination. Certain platinum-based compounds can trigger both cancer cell apoptosis and ICD. This study aims to investigate the effect of the therapy of anti- PDL1 with Oxaliplatin by increasing amount and increasing treatment duration of anti-PDL1 with Oxaliplatin in SK-Br-3, both in vitro and in vivo conditions. The study will use HER-2 (3+) breast cancer cell line, SKBr3. The cells will be treated with increasing concentrations of Oxaliplatin with anti-PDL1 for different durations. In vitro death of cancer cells will be measured by MTT assay, HMGB1 will be measured by western blot. Additionally, ATP release will be measured, mice will be injected with SK-Br-3 and treated with the combo therapy of anti-PDL1 with Oxaliplatin, and in vivo tumor growth will be recorded weekly for xenograft. The positive control for the experiments is cisplatin, and the negative control is IgG solution instead of aPDL1 and Oxaliplatin in PBS.There are three main possible results: (1) The combo therapy of Oxaliplatin with anti-PDL1 induces robust ICD in HER-2 triple positive breast cancer cells. (2) The combo therapy of Oxaliplatin with anti-PDL1 act as a stimulant for robust ICD in HER-2(3+) positive breast cancer cells. (3) The combo-therapy of Oxaliplatin with anti-PDL1 has no significant effect on inducing robust ICD in HER-2(3+) positive breast cancer cells. The result of the study will provide important insight into the preclinical effectiveness of Oxaliplatin with anti-PDL1 in treating HER-2 (3+) breast cancer, and it also sets the basis for future clinical studies of the drug. Future studies should focus on investigating the mechanism underlying Oxaliplatin with anti-PDL1 effectiveness in SK-Br-3.


Asunto(s)
Neoplasias de la Mama , Humanos , Ratones , Animales , Femenino , Oxaliplatino/farmacología , Oxaliplatino/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Muerte Celular Inmunogénica , Línea Celular Tumoral
4.
Arch Microbiol ; 205(6): 220, 2023 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-37148384

RESUMEN

Targeted delivery of a toxin substance to cancer cells is one of the most recent cancer treatment options. Mistletoe Lectin-1 (ML1) in Viscum album L. is a Ribosome-inactivating proteins with anticancer properties. Therefore, it appears that a recombinant protein with selective permeability can be generated by fusing ML1 protein with Shiga toxin B, which can bind to Gb3 receptor that is abundantly expressed on cancer cells. In this study, we sought to produce and purify a fusion protein containing ML1 fused to STxB and evaluate its cytotoxic activities. The ML1-STxB fusion protein coding sequence was cloned into the pET28a plasmid, then was transformed into E. coli BL21-DE3 cells. Following induction of protein expression, Ni-NTA affinity chromatography was used to purify the protein. Using SDS-PAGE and western blotting, the expression and purification processes were validated. On the SkBr3 cell line, the cytotoxic effects of the recombinant proteins were evaluated. On SDS-PAGE and western blotting membrane, analysis of purified proteins revealed a band of approximately 41 kDa for rML1-STxB. Ultimately, statistical analysis demonstrated that rML1-STxB exerted significant cytotoxic effects on SkBr3 cells at 18.09 and 22.52 ng/L. The production, purification, and encapsulation of rML1-STxB fusion protein with potential cancer cell-specific toxicity were successful. However, additional research must be conducted on the cytotoxic effects of this fusion protein on other malignant cell lines and in vivo cancer models.


Asunto(s)
Antineoplásicos , Productos Biológicos , Muérdago , Viscum album , Lectinas , Escherichia coli/genética , Escherichia coli/metabolismo , Muérdago/metabolismo , Viscum album/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/metabolismo , Antineoplásicos/farmacología , Productos Biológicos/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología
5.
Pharmaceutics ; 15(4)2023 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-37111671

RESUMEN

The anticancer drug ibrutinib (IB), also known as PCI-32765, is a compound that irreversibly inhibits Bruton's tyrosine kinase (BTK) and was initially developed as a treatment option for B-cell lineage neoplasms. Its action is not limited to B-cells, as it is expressed in all hematopoietic lineages and plays a crucial role in the tumor microenvironment. However, clinical trials with the drug have resulted in conflicting outcomes against solid tumors. In this study, folic acid-conjugated silk nanoparticles were used for the targeted delivery of IB to the cancer cell lines HeLa, BT-474, and SKBR3 by exploiting the overexpression of folate receptors on their surfaces. The results were compared with those of control healthy cells (EA.hy926). Cellular uptake studies confirmed total internalization of the nanoparticles functionalized by this procedure in the cancer cells after 24 h, compared to nanoparticles not functionalized with folic acid, suggesting that cellular uptake was mediated by folate receptors overexpressed in the cancer cells. The results indicate that the developed nanocarrier can be used for drug targeting applications by enhancing IB uptake in cancer cells with folate receptor overexpression.

6.
Data Brief ; 46: 108880, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36687151

RESUMEN

To obtain this dataset, two human HER2-positive breast cancer cell lines (SKBR3 and MDA-MB-453 cell lines) were cultured in basal growth media to 80% confluence. Cells were passaged and total RNA extracted, RNA converted to cDNA and diluted to a working concentration of 40 ng/µL. Gene expression panels of cancer markers including Fibroblast growth factors (FGF), FGF receptors (FGFRs), cyclin-dependent kinases, cytokeratins, and WNT pathway components were then examined using Q-PCR. Gene expression was normalised against the expression of the endogenous gene 18S. This article describes the data used in the research article "Syndecan-4 regulates the HER2-positive breast cancer cell proliferation cells via CK19/AKT signaling" [1]. The data presented demonstrates the range of gene expression profiles of these cells and aims to provide more detail of all gene expression changes observed in these cell lines.

7.
Biochimie ; 207: 49-61, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36460206

RESUMEN

Despite the use of the highly specific anti-HER2 receptor (trastuzumab) therapy, HER2-positive breast cancers account for 20-30% of all breast cancer carcinomas, with HER2 status a challenge to treatment interventions. The heparan sulfate proteoglycans (HSPGs) are prominently expressed in the extracellular matrix (ECM), mediate breast cancer proliferation, development, and metastasis with most studies to date conducted in animal models. This study examined HSPGs in HER2-positive human breast cancer cell lines and their contribution to cancer cell proliferation. The study examined the cells following enhancement (via the addition of heparin) and knockdown (KD; using short interfering RNA, siRNA) of HSPG core proteins. The interaction of HSPG core proteins and AKT signalling molecules was examined to identify any influence of this signalling pathway on cancer cell proliferation. Our findings illustrated the HSPG syndecan-4 (SDC4) core protein significantly regulates cell proliferation with increased BC cell proliferation following heparin addition to cultures and decreased cell number following SDC4 KD. In addition, along with SDC4, significant changes in CK19/AKT signalling were identified as mediators of BC HER2-positive BC cell proliferation. This study provides evidence for a cell growth regulatory axis involving HSPGs/CK19 and AKT that represents a potential molecular target to prevent proliferation of HER2-positive breast cancer cells.


Asunto(s)
Neoplasias de la Mama , Animales , Humanos , Femenino , Neoplasias de la Mama/metabolismo , Proteoglicanos de Heparán Sulfato/genética , Proteoglicanos de Heparán Sulfato/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sindecano-4 , Proliferación Celular , Línea Celular Tumoral , Heparina
8.
Molecules ; 27(14)2022 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-35889487

RESUMEN

Breast cancer is one of the most diffuse cancers in the world and despite the availability of the different drugs employed against it, the need for new and particularly more specific molecules is ever growing. In this framework, natural products are increasingly assuming an important role as new anticancer drugs. Aloe-emodin (AE) is one of the best characterized molecules in this field. The functionalization of bioactive natural products with selected peptide sequences to enhance their bioavailability and specificity of action is a powerful and promising strategy. In this study, we analyzed the cell specificity, cell viability effects, intracellular distribution, and immune cell response of a new peptide conjugate of Aloe-emodin in SKBR3 and A549 cell lines by means of viability tests, flow cytometry, and confocal microscopy. The conjugate proved to be more effective at reducing cell viability than AE in both cell lines. Furthermore, the results showed that it was mainly internalized within the SKBR3 cells, showing a nuclear localization, while A459 cells displayed mainly a cytoplasmic distribution. A preserving effect of the conjugate on NKs' cell function was also observed. The designed conjugate showed a promising specific activity towards HER2-expressing cells coupled with an enhanced water solubility and a higher cytotoxicity; thus, the resulting proof-of-concept molecule can be further improved as an anticancer compound.


Asunto(s)
Aloe , Antineoplásicos , Productos Biológicos , Neoplasias de la Mama , Emodina , Aloe/química , Antraquinonas/farmacología , Antineoplásicos/farmacología , Apoptosis , Productos Biológicos/farmacología , Emodina/farmacología , Femenino , Humanos , Péptidos/farmacología
9.
Saudi Pharm J ; 30(5): 485-493, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35693435

RESUMEN

The repurposing strategy of converting nimesulide from an anti-fever drug to an anti-cancer agent by modifying its main structure targeting HSP27 is gaining great attention these days. The goal of this study focuses on synthesizing a new nimesulide derivative with new ligands that have biological anti-cancer activities in different cancer models using the in-vitro assay. Nimesulide derivative L1 was synthesized, characterized by 1H NMR, 13C NMR, FTIR, melting point, mass spectra, and TGA analysis. A single crystal was diffracted and showed colorless block group P-1. The results revealed that L1 demonstrates potent anti-cancer activity with lung (H292), ovarian (SKOV3), and breast (SKBR3) cancer cell lines in-vitro models with IC50 values below 8.8 µM.

10.
Nan Fang Yi Ke Da Xue Xue Bao ; 42(5): 740-746, 2022 May 20.
Artículo en Chino | MEDLINE | ID: mdl-35673919

RESUMEN

OBJECTIVE: To investigate the effect of metformin on the proliferation and apoptosis of HER-2-positive breast cancer cell line SKBR3 and explore the possible mechanism of its action. METHODS: SKBR3 cells were treated with different concentrations (20-120 µmol/L) of metformin, and the changes in cell proliferation and colony formation ability were assessed using CCK-8 assay and crystal violet staining, respectively. Flow cytometry was performed to analyze cell apoptosis and cell cycle changes. Real-time fluorescent quantitative PCR (qRT-PCR) was used to detect mRNA expressions of YAP, TAZ, EGFR, CTGF, CYR61, E-cadherin, N-cadherin, vimentin and fibronectin in the treated cells, and the protein expressions of YAP and TAZ were detected using Western blotting; immunofluorescence assay was used to observe YAP/TAZ nuclear translocation in the cells. RESULTS: Metformin treatment significantly inhibited the proliferation of SKBR3 cells (P < 0.05) in a concentration- and time-dependent manner. The results of flow cytometry showed that metformin significantly promoted apoptosis and caused cell cycle arrest at G1 phase in SKBR3 cells. Metformin treatment significantly down-regulated the mRNA expressions of YAP, TAZ, EGFR, CTGF and CYR61, N-cadherin, vimentin and fibronectin (P < 0.05) and up-regulated the expression of E-cadherin (P < 0.05); Western blotting results showed that YAP and TAZ protein expressions were significantly down-regulated in the cells after metformin treatment (P < 0.05). Immunofluorescence assay revealed that metformin treatment caused the concentration of YAP and TAZ in the cytoplasm, and significantly reduced their amount in the cell nucleus. CONCLUSION: Metformin can inhibit proliferation and promote apoptosis and epithelal-mesenchymal transition of HER-2 positive breast cancer cells possibly by that inhibing YAP and TAZ expression and their nuclear localization.


Asunto(s)
Metformina , Neoplasias , Apoptosis , Cadherinas , Proliferación Celular , Receptores ErbB , Fibronectinas , Metformina/farmacología , Proteínas Serina-Treonina Quinasas , ARN Mensajero , Factores de Transcripción/metabolismo , Vimentina
11.
Chem Asian J ; 17(14): e202200301, 2022 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-35510693

RESUMEN

Cell-targeted peptides are recommended for precision cancer treatment due to their comparable targeting properties, small molecular size, and good biocompatibility. However, unpredictable bioactivity, low penetration rate and poor stability greatly limit its efficacy. Supramolecular self-assembly based on synthetic peptide has great potential to solve related problems and achieve better therapeutic effects. Herein, we report and compare the effects of two different assembly pathway, heating-cooling, and enzyme instruction, on the penetrability of SKBR-3 cell targeted peptides. It was found that enzyme-instructed self-assembly (EISA) resulted in hydrogels composed of uniform supramolecular nanofibers, whereas heating-cooling resulted in solutions and precipitations composed of slightly different nanoparticles. The nanofibers formed by EISA showed enhanced cellular uptake (2.54 µM), which was significantly higher than the 1.06 µM of the nanoparticles formed by temperature regulation. Thus, EISA is a promising strategy to improve the cell penetration rate of targeted peptides and could provide a better solution for precision cancer treatment.


Asunto(s)
Nanofibras , Hidrogeles/química , Hidrogeles/farmacología , Nanofibras/química , Péptidos/química , Péptidos/farmacología
12.
Cancer Genomics Proteomics ; 19(1): 79-93, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34949661

RESUMEN

BACKGROUND/AIM: Trastuzumab and tamoxifen are two of the most widely prescribed anti-cancer drugs for breast cancer (BC). To date, few studies have explored the impact of anticancer drugs on metabolic pathways in BC. Metabolomics is an emerging technology that can identify new biomarkers for tracking therapy response and novel therapeutic targets. MATERIALS AND METHODS: We employed ultra-high-performance liquid chromatography-quadrupole time of flight mass spectrometry (UHPLC-QTOF-MS) to investigate changes in MCF-7 and SkBr3 cell lines treated with either tamoxifen, trastuzumab or a combination of both. The Bruker Human Metabolome Database (HMDB) metabolite library was used to match spectra and the MetaboScape software to assign each feature with a putative metabolite name or molecular formula for metabolite annotation. RESULTS: A total of 98 metabolites were found to significantly differ in abundance in MCF-7 and SkBr3 treated cells. Moreover, the metabolic profile of the combination medication is similar to that of tamoxifen alone, according to functional enrichment analysis. CONCLUSION: Tamoxifen/trastuzumab treatment had a significant effect on pathways essential for the control of energy-production, which have previously been linked to cancer progression, and aggressiveness.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Biomarcadores de Tumor/análisis , Neoplasias de la Mama/tratamiento farmacológico , Tamoxifeno/farmacología , Trastuzumab/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/patología , Cromatografía Líquida de Alta Presión/métodos , Monitoreo de Drogas/métodos , Metabolismo Energético/efectos de los fármacos , Humanos , Células MCF-7 , Metaboloma/efectos de los fármacos , Metabolómica/métodos , Tamoxifeno/uso terapéutico , Espectrometría de Masas en Tándem/métodos , Trastuzumab/uso terapéutico
13.
Pharmaceuticals (Basel) ; 14(4)2021 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-33918832

RESUMEN

Three-dimensional spheroids of non-malignant MCF10A and malignant SKBR3 breast cells were used for subsequent 3D Cell-SELEX to generate aptamers for specific binding and treatment of breast cancer cells. Using 3D Cell-SELEX combined with Next-Generation Sequencing and bioinformatics, ten abundant aptamer families with specific structures were identified that selectively bind to SKBR3, and not to MCF10A cells. Multivalent aptamer polymers were synthesized by co-polymerization and analyzed for binding performance as well as therapeutic efficacy. Binding performance was determined by confocal fluorescence imaging and revealed specific binding and efficient internalization of aptamer polymers into SKBR3 spheroids. For therapeutic purposes, DNA sequences that intercalate the cytotoxic drug doxorubicin were co-polymerized into the aptamer polymers. Viability tests show that the drug-loaded polymers are specific and effective in killing SKBR3 breast cancer cells. Thus, the 3D-selected aptamers enhanced the specificity of doxorubicin against malignant over non-malignant breast cells. The innovative modular DNA aptamer platform based on 3D Cell SELEX and polymer multivalency holds great promise for diagnostics and treatment of breast cancer.

14.
J Pharm Drug Deliv Res ; 10(1)2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33681397

RESUMEN

OBJECTIVE: The aim of this study is to design, fabricate and determine the cytotoxic effects of dual loaded paclitaxel and 17-AAG in stealth polymeric nanoparticles. The nanoparticles were fabricated by dispersion polymerization. METHODS: Two breast cancer cell lines (MCF-7 and SKBR-3) were cultured and treated with media only, blank nanoparticles, paclitaxel (as a free drug), 17-AAG (free drug), paclitaxel + 17-AAG combination (as free drugs), and paclitaxel + 17-AAG combination loaded in poly-ε-caprolactone stealth nanoparticles. Each drug in the combination was half the concentration of the single free drug. RESULTS: The cytotoxic effects of the paclitaxel treatment and that of the combination (free drug) were found to be similar in both SKBR3 and MCF7 cell lines. Similar cytotoxic effects were observed for the drug combination both in the drug loaded nanoparticles formulation and in free drug form for both cell lines. CONCLUSION: Both paclitaxel and 17-AAG were effectively loaded and released from the polymeric nanoparticles. Paclitaxel (free drug), paclitaxel-17AAG combination (free drug), and dual drug-loaded nanoparticles had similar cytotoxic effects on both cell lines. Paclitaxel and 17-AAG combination resulted in synergistic effect: paclitaxel in the combination with 17-AAG was half its original concentration and yielded similar cytotoxic effect. The dose of paclitaxel was reduced without lowering its therapeutic efficacy.

15.
Biosens Bioelectron ; 178: 113023, 2021 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-33529862

RESUMEN

Ultrasensitive monitoring of cancer cells, especially metastatic ones, has a great interest in human medicine. Despite the early diagnosis of diseases, there is an essential need for any prediction in the severity of side effects for therapeutic outcomes like metastasis. Therefore, the inhibition of cancer cells metastasis to other organs is of utmost importance for cancer suffering patients. In this regard, we developed an electrochemiluminescence (ECL)-based cytosensor for the quantification of metastatic breast cancer cells, namely SKBR-3. Silica-based mesoporous materials have a great potential for application in ECL biosensors due to their high loading capacity and mechanical strength. Herein, a silica-based electrode was prepared via in situ electrosyntheses of mesoporous silica as an environmentally friendly approach. In this protocol, luminol (as luminophore) was combined with chitosan (as attachment biomolecule) to produce a stable lumino-composite film on the electrode surface. At the optimum experimental conditions, the lower limit of quantitation (LLOQ) and linear dynamic range (LDR) were obtained as 20 cells/mL and 20 to 2000 cells/mL, individually. The specificity was desirably examined in the presence of other breast cancer cell lines such as MCF-7 and MDA-MB-231, as a model of early-stage and invasive phases of breast cancer cells. The repeatability was successfully examined for five repetitive measurements and the acceptable relative standard deviation (RSD) was calculated as about 1.6% for 500 cells/mL. As a proof of concept, the presented cytosensor has a high ability to use in clinical laboratories for the detection and separation of metastatic cells via the combination with microfluidic systems.


Asunto(s)
Técnicas Biosensibles , Neoplasias de la Mama , Neoplasias de la Mama/diagnóstico , Técnicas Electroquímicas , Humanos , Límite de Detección , Mediciones Luminiscentes , Luminol , Células MCF-7
16.
Sci Total Environ ; 775: 145814, 2021 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-33621883

RESUMEN

Bisphenol AF (BPAF) is a known estrogen disruptor of the ERα pathway. The aim of the present study was to characterize the proliferation effects of BPAF on ERα-negative SKBR-3 breast cancer cells with mechanistic insights. BPAF at low concentrations (0.001-0.1 µM) significantly induced the proliferation of SKBR-3 cells. In a SKBR-3 tumor model in BALB/c nude mice, BPAF at 100 mg/kg body weight/day also significantly promoted the growth of SKBR-3 tumors. Low concentrations of BPAF markedly increased the expression of G protein-coupled estrogen receptor (GPER1), c-Myc, CyclinD1 and c-Fos proteins, and enhanced phosphorylation of extracellular signal-regulated kinase (Erk) and protein kinase B (Akt) in SKBR-3 cells. Further, BPAF significantly upregulated mRNA levels of related target genes in SKBR-3 cells and SKBR-3 tumor tissues in nude mice. The GPER1 inhibitor G15 and phosphatidylinositide 3-kinase (PI3K) inhibitor wortmannin (WM) inhibited phosphorylation of Erk and Akt. The specific signal inhibitors also markedly decreased the expression of target genes and weakened the cell proliferation induced by low-concentration BPAF. The findings showed that GPER1 could independently regulate BPAF-induced proliferation of SKBR-3 cells without requiring ERα. These results provide mechanistic insights into the effects of BPAF regarding ERα-negative human breast cancer development.


Asunto(s)
Neoplasias de la Mama , Receptor alfa de Estrógeno , Animales , Compuestos de Bencidrilo , Línea Celular Tumoral , Proliferación Celular , Receptor alfa de Estrógeno/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Fenoles
17.
Breast Cancer Res Treat ; 185(1): 95-106, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32974790

RESUMEN

PURPOSE: Dysregulation of HER2 signaling pathway in breast cancer is well documented. Our bioinformatics analysis predicted hsa-miR-512-3p (miR-512-3p) as a bona fide regulator of HER2 as well as HER3, PIK3R2, and AKT1 genes. Then, we intended to examine the effect of miR-512-3p on the predicted target genes that are involved in HER2 signaling pathway. METHODS AND RESULTS: RT-qPCR results indicated lower expression of miR-512-3p in breast cancer specimens, compared to their normal pairs. Overexpression of miR-512-3p resulted in HER2, HER3, PIK3R2, and AKT1 gene downregulation, detected by RT-qPCR and the result was confirmed by western analysis and ELIZA test against p-AKT, BAX, FADD, and HER2 proteins in SKBR3 cells, respectively. Then, dual-luciferase assay supported the direct interaction of miR-512-3p with 3'UTR sequences of HER2, HER3, PIK3R2, and AKT1 target genes. When miR-512-3p was overexpressed, BAX/BCL2 expression ratio and proportion of sub-G1 cell population were increased in transfected SKBR3 cells, detected by RT-qPCR and flow cytometry, respectively. These results were consistent with the decreased viability of transfected cells, documented by MTT assay. In addition, results were consistent with the upregulation of BAX, BAK, BOK, PTEN, P53, and P21 genes and downregulation of CCND1 gene in SKBR3 cells. Although the overexpression of miR-512 resulted in cell cycle arrest at Sub-G1 stage in MDA-MB-231 cells, this effect seemed independent of targeting HER2, HER3, PIK3R2, and AKT1 target genes. CONCLUSION: Overall, results indicated that miR-512-3p acts as a cell-type-specific tumor suppressor, through targeting HER2, HER3, PIK3R2, and AKT1 transcripts. These results suggest miR-512-3p as a potential candidate marker for breast cancer diagnosis.


Asunto(s)
Neoplasias de la Mama , MicroARNs , Neoplasias de la Mama/genética , Femenino , Humanos , MicroARNs/genética , Transducción de Señal , Regulación hacia Arriba
18.
Front Pharmacol ; 11: 537265, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32973536

RESUMEN

BACKGROUND: We explored the therapeutic and prognostic effect of YAP/TAZ intensityinHER2-positive breast cancer patients. We also investigated the relationship between YAP/TAZ expression and Trastuzumab-resistance. METHODS: We collected clinicopathological information from 397 cases. We evaluated therapeutic and prognostic effect of YAP/TAZ and other variables. We also cultivated Trastuzumab-resistance cell lines and explored relationship between YAP/TAZ and Trastuzumab-resistance. RESULTS: Over-expression of YAP/TAZ was remarkable in Trastuzumab-resistant cells, and so did HER3 and HER2/HER3 heterodimer. Inhibition of YAP/TAZ expression reversed Trastuzumab-resistance.YAP/TAZ deficiency contributed to favorable therapeutic response, and so did hormone receptor insufficiency and chemotherapy dosage inferiority. Deficient YAP/TAZ intensity and abundant hormone receptor intensity contributed to better survival. Over-expression of YAP/TAZ was obvious in recurrent cases in comparison with their matching primary lesions. Prognostic superiority of insufficient YAP/TAZ intensity was more outstanding in hormone receptor negative cases. Over-expression of YAP/TAZ and HER3 was generally synchronous. Absence of HER3 expression in residual lesions might correlate with better breast cancer-free survival. CONCLUSIONS: Over-expression of YAP/TAZ as well as HER-3 and HER2/HER3 heterodimer was synchronously remarkable in Trastuzumab-resistant cell lines. Inhibition of YAP/TAZ expression reversed Trastuzumab resistance. Deficient YAP/TAZ intensity as well as insufficient hormone receptor intensity and high chemotherapy dosage contributed to favorable therapeutic response. Deficient YAP/TAZ intensity and abundant hormone receptor intensity contributed to better survival, and so did absence of HER3expression in residual lesions. Prognostic superiority of YAP/TAZ expression depended on hormone receptor status. Cases with synchronous over-expression of YAP/TAZ and HER3 suffered poor survival, which revealed the potential effect of YAP/TAZ-HER2/HER3 crosstalk in prognosis of HER2-positive patients.

19.
Rev. biol. trop ; 68(3)sept. 2020.
Artículo en Inglés | LILACS, SaludCR | ID: biblio-1507707

RESUMEN

Introduction: The Vismia genus belongs to the Hypericaceae family and comprises around 57 species of which 17 have been located in Venezuela. Previous investigations have been carried out in extracts as well as pure isolated compounds, revealing antimicrobial, antioxidant and anti-HIV, among other, biological activities. Objective: This investigation aims to determine the cytotoxic activity of essential oils from leaves of Vismia baccifera Triana & Planch (VBJ and VBV) and Vismia macrophylla Kunth (VM) collected in three different locations of the Venezuelan Andean region. Methods: Essential oils obtained by hydrodistillation were analyzed using gas chromatography-mass spectrometry (GC-MS) and their cytotoxic activity was analyzed following the MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide) assay. Human tumor cell lines from SKBr3, MCF-7 and PANC-1, two breast carcinomas and one pancreatic adenocarcinoma of ductal type, were tested with the oil samples and human dermis fibroblasts were used as non-tumor cells. Results: β-caryophyllene and trans-caryophyllene were present as major components in VBJ and VBV, respectively, while γ-bisabolene was the main component in the VM sample. Anticancer activity was observed on V. baccifera essential oil against SKBr3, MCF-7 and PANC-1. The selectivity index showed that VBV is highly selective against the SKBr3 cell line and has no activity against non-tumor cells. Conclusions: These results are considered a contribution to natural products research and may provide supportive data for future studies on cancer.


Introducción: El género Vismia pertenece a la familia Hypericaceae y comprende alrededor de 57 especies de las cuales 17 han sido ubicadas en Venezuela. Se han realizado investigaciones previas tanto en extractos como en compuestos puros aislados revelando actividad antimicrobiana, antioxidante y anti-VIH, entre otras actividades biológicas. Objetivo: El propósito de esta investigación es determinar la actividad citotóxica de los aceites esenciales de las hojas de Vismia baccifera Triana & Planch (VBJ y VBV) y Vismia macrophylla Kunth (VM) recolectadas en tres localidades de la región andina venezolana. Métodos: Aceites esenciales obtenidos por hidrodestilación fueron analizados por cromatografía de gases-espectrometría de masas y su actividad citotóxica fue analizada siguiendo el método MTT (bromuro de 3-[4,5-dimetiltiazol-2-il]-2,5-difeniltetrazolio). Los aceites esenciales fueron ensayados frente a células tumorales humanas SKBr3, MCF-7 y PANC-1, dos carcinomas de mama y un adenocarcinoma pancreático del tipo ductal, usando cultivos primarios de fibroblastos de dermis humana como células no tumorales. Resultados: β-cariofileno y trans-cariofileno estuvieron presentes como compuestos mayoritarios en VBJ y VBV, respectivamente, mientras que γ-bisaboleno fue el componente principal en la muestra VM. El aceite esencial de V. baccifera mostró actividad anticancerígena frente a SKBr3, MCF-7 y PANC-1. El índice de selectividad reveló que VBV es altamente selectivo frente a la línea celular SKBr3 y no presenta actividad contra las células no tumorales. Conclusiones: Estos resultados se consideran una contribución a la investigación de los productos naturales y los datos pueden ser de utilidad en futuras investigaciones sobre el cáncer.


Asunto(s)
Cromatografía de Gases , Clusiaceae/toxicidad , Plantas Medicinales , Venezuela
20.
Mar Drugs ; 18(5)2020 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-32438569

RESUMEN

Astaxanthin (AST) is related to apoptosis but the details of the mechanism of how AST makes apoptosis is not clear. The present study investigated apoptotic effects of AST to SKBR3, a breast cancer cell line in detail. Cell viability assay showed cellular proliferation and morphological changes of the cells were observed under AST treatment. FACS analysis indicated that AST blocked cell cycle progression at G0/G1, suppressed proliferation dose-dependently, and induced apoptosis of the cells. The apoptosis of the cells by AST was further demonstrated through the decreased expression level of mutp53 and cleaved a PARP-1 fragment, respectively. In addition, AST induced the intrinsic apoptosis of the cells by activation of Bax/Bcl2, cleaved caspase-3, and cleaved caspase-9 as well as the phosphorylation of ERK1/2, JNK, and p38. Furthermore, AST decreased production of intracellular reactive oxygen species as well as modulated expressions of superoxide dismutases and Pontin, an anti-apoptotic factor. Co-immunoprecipitation assay revealed AST reduced interaction between Pontin and mutant p53. Taken together, these studies proved that AST regulates the expression of apoptotic molecules to induce intrinsic apoptosis of the cells, suggesting AST therapy might provide an alternative for improving the efficacies of other anti-cancer therapies for breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Línea Celular Tumoral/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos , Xantófilas/farmacología , Xantófilas/uso terapéutico , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA