Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
J Nutr Biochem ; 107: 109018, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35489658

RESUMEN

Docosahexaenoic acid (DHA) reduces breast cancer tumor growth in preclinical models. To better understand how DHA amplifies the actions of docetaxel (TXT) chemotherapy, we examined the effects of two doses of dietary DHA on tumor size, membrane DHA content and necroptosis using a drug resistant triple negative breast cancer (TNBC) patient derived xenograft (PDX) model. Female NOD.Cb-PrkdcscidIl2rg mice bearing TNBC PDXs were randomized to one of three nutritionally complete diets (20% w/w fat): control (0% DHA), high DHA (3.8% HDHA), or low DHA (1.6% LDHA) with or without intraperitoneal injections of 5 mg/kg TXT, twice weekly for 6 weeks (n=8 per group). Tumors from mice fed either HDHA+TXT or LDHA+TXT were similar in size to each other, but were 36% and 32% smaller than tumors from mice fed control+TXT, respectively (P<.05). A dose effect of DHA incorporation was observed in plasma total phospholipids and in phosphatidylethanolamine and phosphatidylinositol. Both doses of DHA resulted in similarly increased necrotic tissue and decreased NFκB protein expression compared to control tumors, however only the HDHA+TXT had increased expression of necroptosis related proteins: RIPK1, RIPK3 and MLKL (P<.05). Increased MLKL was observed in the lipid raft portion of HDHA+TXT tumor extracts. This work confirms the efficacy of a combination therapy consisting of DHA supplementation and TXT chemotherapy using two doses of DHA as indicated by reduced tumor growth in a TNBC PDX model. Moreover, the results suggest that decreased growth may occur through increased DHA incorporation into tumor phospholipid membranes and necroptosis.


Asunto(s)
Ácidos Docosahexaenoicos , Neoplasias de la Mama Triple Negativas , Animales , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos/metabolismo , Femenino , Xenoinjertos , Ratones , Ratones Endogámicos NOD , Necroptosis , Fosfolípidos/metabolismo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología
2.
Nutr Cancer ; 74(4): 1431-1445, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34286635

RESUMEN

There is a strong rationale for investigating nutritional interventions with docosahexaenoic acid (DHA) in cancer prevention and therapy; however, the effects of DHA on ovarian cancer (OC) have not been well studied. Here, we investigated if DHA alone and in combination with carboplatin reduces OC cell growth in vitro. In vivo, we used a high-grade serous OC patient-derived xenograft (PDX) mouse model to investigate if DHA affects OC growth and enhances the anticancer actions of carboplatin. We showed synergistic cell killing by DHA and carboplatin in DHA-resistant Kuramochi and SKOV3 OC cells, which corresponded with increased DHA incorporation into whole-cell membrane phospholipids (P < 0.05). In vivo, feeding mice a diet supplemented with 3.9% (w/w of fat) DHA resulted in a significant reduction in PDX growth with and without carboplatin (P < 0.05). This reduction in tumor growth was accompanied by an increased tumor necrotic region (P < 0.05) and improved survival. Plasma membranes in tumors and livers excised from mice fed a DHA diet had ∼ twofold increase in DHA incorporation as compared with mice fed a control diet. Our findings indicate that DHA supplementation reduces cancer cell growth and enhances the efficacy of carboplatin in preclinical models of OC through increased apoptosis and necrosis.Supplemental data for this article is available online at https://doi.org/10.1080/01635581.2021.1952453.


Asunto(s)
Ácidos Docosahexaenoicos , Neoplasias Ováricas , Animales , Carboplatino/farmacología , Carcinoma Epitelial de Ovario , Ciclo Celular , Proliferación Celular , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos/farmacología , Femenino , Humanos , Ratones , Neoplasias Ováricas/patología
3.
Front Nutr ; 8: 769293, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34790691

RESUMEN

Background: Dietary long chain polyunsaturated fatty acids (LCPUFA) such as arachidonic acid (ARA) and docosahexaenoic acid (DHA) play an important role in the development of the infant immune system. The role of LCPUFA in the T helper type 2 (Th2) biased immune system is unknown. We aimed to understand the effect of feeding LCPUFA during suckling and post-weaning on immune system development in Th2 bias Brown Norway rat offspring. Methods: Brown Norway dams were randomly assigned to nutritionally adequate maternal diet throughout the suckling period (0-3 weeks), namely, control diet (0% ARA, 0% DHA; n= 8) or ARA + DHA (0.45% ARA, 0.8% DHA; n = 10). At 3 weeks, offspring from each maternal diet group were randomized to either a control (0% ARA, 0% DHA; n = 19) or ARA+DHA post-weaning (0.5% ARA, 0.5% DHA; n = 18) diet. At 8 weeks, offspring were killed, and tissues were collected for immune cell function and fatty acid composition analyses. Results: ARA + DHA maternal diet resulted in higher (p < 0.05) DHA composition in breast milk (4×) without changing ARA levels. This resulted in more mature adaptive immune cells in spleen [T regulatory (Treg) cells and B cells], mesenteric lymph nodes (MLN, lower CD45RA+), and Peyer's patches (PP; higher IgG+, B cells) in the ARA+DHA group offspring at 8 weeks. ARA+DHA post-weaning diet (3-8 weeks) resulted in 2 × higher DHA in splenocyte phospholipids compared to control. This also resulted in higher Th1 cytokines, ~50% higher TNF-α and IFNγ, by PMAi stimulated splenocytes ex vivo, with no differences in Th2 cytokines (IL-4, IL-13, and IL-10) compared to controls. Conclusion: Feeding dams a diet higher in DHA during the suckling period resulted in adaptive immune cell maturation in offspring at 8 weeks. Providing ARA and DHA during the post-weaning period in a Th2 biased Brown Norway offspring model may support Th1 biased immune response development, which could be associated with a lower risk of developing atopic diseases.

4.
Cancers (Basel) ; 13(6)2021 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-33801979

RESUMEN

This scoping review examines the evidence for n-3 long-chain polyunsaturated fatty acid [LCPUFA, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)] supplementation in clinical cancer therapy. A comprehensive literature search was performed to identify relevant clinical intervention studies conducted through August 2020. Fifty-seven unique cancer trials, assessing EPA and/or DHA supplementation pre- or post-treatment, concomitant with neoadjuvant chemotherapy, radiation or surgery, or in palliative therapy were included. Breast, head and neck, gastrointestinal, gastric, colorectal/rectal, esophageal, leukemia/lymphoma, lung, multiple myeloma and pancreatic cancers were investigated. Across the spectrum of cancers, the evidence suggests that supplementation increased or maintained body weight, increased progression-free and overall survival, improved overall quality of life, resulted in beneficial change in immune parameters and decreased serious adverse events. Taken together, the data support that EPA and/or DHA could be used to improve outcomes important to the patient and disease process. However, before incorporation into treatment can occur, there is a need for randomized clinical trials to determine the dose and type of n-3 LCPUFA intervention required, and expansion of outcomes assessed and improved reporting of outcomes.

5.
Curr Dev Nutr ; 5(4): nzab022, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33889794

RESUMEN

BACKGROUND: Studies suggest that fatty acid status influences breast cancer etiology, yet the roles of individual fatty acids in breast cancer risk are unclear, specifically when central adiposity and menopausal status are considered. OBJECTIVES: This study examined the associations of fatty acid status with breast cancer risk including location, menopausal status, and waist-to-hip ratio as key variables. METHODS: Prediagnostic plasma phospholipid fatty acids were measured in women with breast cancer (n = 393) and age-matched controls (n = 786) from a nested case-control prospective study within Alberta's Tomorrow Project (ATP) and British Columbia Generations Project (BCGP) cohorts. Binary logistic regression models were used to evaluate associations of fatty acids and breast cancer risk with subgroup analysis for menopausal status and waist-to-hip ratio. RESULTS: Women from BCGP had a higher n-3 (É·-3) fatty acid status compared with the ATP (6.4% ± 0.08% vs. 5.3% ± 0.06%; P < 0.001), so subsequent analysis was blocked by cohort. Overall, fatty acids had inconsistent associations with risk. In the ATP among premenopausal women, total long-chain n-3 fatty acids (ORQ4vsQ1 = 1.78; 95% CI: 0.58, 5.43; P-trend = 0.007, P-interaction = 0.07) were positively associated with breast cancer risk, whereas in BCGP, DHA (ORQ4vsQ1 = 0.66; 95% CI: 0.28, 1.53; P-trend = 0.03, P-interaction = 0.05) and total long-chain n-3 fatty acids (ORQ4vsQ1 = 0.66; 95% CI: 0.28, 1.54; P-trend = 0.03) were associated with decreased cancer risk when the waist-to-hip ratio was <0.85. CONCLUSIONS: Our findings suggest that regional variations in fatty acid status influence breast cancer risk, resulting in positive associations of total long-chain n-3 fatty acids in premenopausal ATP women and negative associations of these fatty acids in BCGP women with a waist-to-hip ratio below guidelines. This study highlights the complexity and difficulty in using fatty acid status to predict breast cancer risk in diverse populations without the consideration of other risk factors.

6.
Lipids ; 55(5): 549-565, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32588470

RESUMEN

To better understand how docosahexaenoic acid (DHA) improves the effects of doxorubicin (DOX), we examined DHA ± DOX on changes in whole cell and lipid raft phospholipids (PL) of MDA-MB-231 and MCF-7 breast cancer cells. We sought to confirm whether the relative changes in PL DHA content of MDA-MB-231 cells could be extended to PL from MDA-MB-231 tumors grown in mice fed a DHA supplemented diet ±DOX. Treatment with DHA did not change PL composition yet DOX increased the proportion of phosphatidylserine in MCF-7 cell lipid rafts by two-fold (p < 0.001). Regardless of DOX, the relative percent incorporation of DHA was higher in MDA-MB-231 cells compared to MCF-7 cells in phosphatidylserine, phosphatidylethanolamine, and phosphatidylcholine (whole cell and lipid rafts); and higher in phosphatidylethanolamine vs. phosphatidylcholine (4.4-fold in MCF-7 and 6-fold in MDA-MB-231 cells respectively). DHA treatment increased eicosapentaenoic acid and docosapentaenoic acid in MDA-MB-231 cells but not MCF-7 cells. Increased DHA content in MDA-MB-231 cells, MCF-7 cells, and MDA-MB-231 tumors in all PL moieties (except sphingomyelin) corresponded with reduced arachidonic acid (p < 0.05). Feeding mice 2.8% (w/w of fat) DHA ± DOX increased tumor necrotic regions (p < 0.05). This study established differential incorporation of DHA into whole cell and lipid rafts between human breast cancer cell lines. However, within each cell line, this incorporation was not altered by DOX confirming that DOX does not change membrane lipid composition. Furthermore, our findings indicate that membrane changes observed in vitro are translatable to in vivo changes and that DHA + DOX could contribute to the anticancer effects through increased necrosis.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Ácidos Docosahexaenoicos/farmacología , Doxorrubicina/farmacología , Fosfolípidos/farmacología , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ácidos Docosahexaenoicos/química , Doxorrubicina/química , Ácido Eicosapentaenoico/química , Ácido Eicosapentaenoico/farmacología , Femenino , Humanos , Células MCF-7 , Lípidos de la Membrana/química , Lípidos de la Membrana/farmacología , Microdominios de Membrana/química , Ratones , Fosfolípidos/química , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Nutr ; 150(3): 606-615, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31722423

RESUMEN

BACKGROUND: Long-chain n-3 PUFAs (LCPUFAs) improve immune development and reduce atopic disease risk in infants. Stearidonic acid (SDA) can be a substrate for biosynthesis of n-3 LCPUFAs. OBJECTIVE: We aimed to determine the effect of feeding an SDA-enriched diet during suckling and weaning on offspring immunity and ability to develop oral tolerance (OT). METHODS: Pregnant Sprague-Dawley rats were randomly assigned to consume either SDA (3 g SDA/100 g fat) or a control (no SDA) diet, 5 d before parturition and through lactation (21 d). For the OT treatment, 10-d-old pups were fed ovalbumin (Ova; 200 µL of 8 mg/mL) or placebo daily for 5 d. At 21 d, pups (both sexes) were weaned to their respective maternal diet until 6 wk of age or killed. Systemic immunization was induced using Ova (in 3-wk-old pups) or Ova + adjuvant (in 6-wk-old pups). The effect of suckling diet (in 3-wk-old pups) or weaning diet (in 6-wk-old pups) and OT treatment on immune function (main outcome) in spleen and blood was compared using 2-factor ANOVA. RESULTS: An SDA-enriched maternal diet, compared with the control diet, resulted in higher plasma phospholipid (PL) EPA (15 times higher), docosapentaenoic acid (DPA; 3 times higher), and DHA (1.3 times higher) content in 3-wk-old pups, accompanied by higher B-cell function [plasma ovalbumin-specific IgG1 (Ova-IgG1), 2 times higher] ( P < 0.05). Compared with pups fed a control diet, the splenocytes from these pups had more (23%) helper T (Th) cells (CD3+CD4+) and activated (12%) Th cells (CD4+CD28+) (P < 0.02) than controls. At 6 wk, the SDA group had 30% more CD4+CD25+ splenocytes, and when stimulated ex vivo with LPS, produced less inflammatory IL-6 (50%) and TNF-α (30%) and more immunoregulatory IL-10 (45%) cytokines (P < 0.05) than the control group. The Ova-exposed group had less (30%) plasma Ova-IgG1 than the placebo group. Splenocytes and plasma PLs from the 6-wk-old SDA group had more EPA (2x) and DPA (3.5x) (P < 0.05), but not DHA, than the control group. CONCLUSIONS: Feeding SDA during lactation and weaning altered immune responses in directions believed to be beneficial.


Asunto(s)
Animales Recién Nacidos , Grasas Insaturadas en la Dieta/administración & dosificación , Ácidos Grasos Omega-3/administración & dosificación , Sistema Inmunológico/efectos de los fármacos , Animales , Citocinas/biosíntesis , Grasas Insaturadas en la Dieta/análisis , Ácidos Grasos/sangre , Ácidos Grasos/metabolismo , Ácidos Grasos Omega-3/farmacología , Femenino , Sistema Inmunológico/crecimiento & desarrollo , Inmunoglobulina G/sangre , Inmunofenotipificación , Masculino , Fenómenos Fisiológicos de la Nutrición , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Bazo/metabolismo
8.
BMJ Open ; 9(9): e030502, 2019 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-31530611

RESUMEN

INTRODUCTION: Neoadjuvant chemotherapy for breast cancer treatment is prescribed to facilitate surgery and provide confirmation of drug-sensitive disease, and the achievement of pathological complete response (pCR) predicts improved long-term outcomes. Docosahexaenoic acid (DHA) has been shown to reduce tumour growth in preclinical models when combined with chemotherapy and is known to beneficially modulate systemic immune function. The purpose of this trial is to investigate the benefit of DHA supplementation in combination with neoadjuvant chemotherapy in patients with breast cancer. METHODS AND ANALYSIS: This is a double-blind, phase II, randomised controlled trial of 52 women prescribed neoadjuvant chemotherapy to test if DHA supplementation enhances chemotherapy efficacy. The DHA supplementation group will take 4.4 g/day DHA orally, and the placebo group will take an equal fat supplement of vegetable oil. The primary outcome will be change in Ki67 labelling index from prechemotherapy core needle biopsy to definitive surgical specimen. The secondary endpoints include assessment of (1) DHA plasma phospholipid content; (2) systemic immune cell types, plasma cytokines and inflammatory markers; (3) tumour markers for apoptosis and tumour infiltrating lymphocytes; (4) rate of pCR in breast and in axillary nodes; (5) frequency of grade 3 and 4 chemotherapy-associated toxicities; and (6) patient-perceived quality of life. The trial has 81% power to detect a significant between-group difference in Ki67 index with a two-sided t-test of less than 0.0497, and accounts for 10% dropout rate. ETHICS AND DISSEMINATION: This study has full approval from the Health Research Ethics Board of Alberta - Cancer Committee (Protocol #: HREBA.CC-18-0381). We expect to present the findings of this study to the scientific community in peer-reviewed journals and at conferences. The results of this study will provide evidence for supplementing with DHA during neoadjuvant chemotherapy treatment for breast cancer. TRIAL REGISTRATION NUMBER: NCT03831178.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Ácidos Docosahexaenoicos/administración & dosificación , Terapia Neoadyuvante/métodos , Alberta , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Biomarcadores de Tumor/análisis , Neoplasias de la Mama/sangre , Neoplasias de la Mama/patología , Ensayos Clínicos Fase II como Asunto , Citocinas/sangre , Suplementos Dietéticos , Ácidos Docosahexaenoicos/sangre , Método Doble Ciego , Femenino , Humanos , Antígeno Ki-67/metabolismo , Ganglios Linfáticos/patología , Calidad de Vida , Ensayos Clínicos Controlados Aleatorios como Asunto , Resultado del Tratamiento
9.
Breast Cancer Res Treat ; 177(2): 357-367, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31236812

RESUMEN

PURPOSE: The objective of this study was to investigate if DHA dietary supplementation enhances the anticancer actions of docetaxel (TXT) in two different drug resistant triple negative breast cancer (TNBC) patient-derived xenografts (PDX)s. METHODS: In two experiments, female NSG mice bearing TNBC PDXs were randomized to one of two nutritionally adequate diets (20% w/w): control (0% DHA), or DHA (3.9% w/w of total fat) and injected with 0 or 5 mg/kg TXT, twice weekly for 6 weeks (n = 8 per group). Treatment response was determined by significant differences in tumor weight, and apoptotic, proliferation and cell cycle markers at endpoint. RESULTS: Mice bearing MAXF574 xenografts fed DHA diet and treated with TXT had a 57% reduction in tumor weight compared to mice fed control diet (P < 0.004), a 64% reduction compared to control + TXT (P < 0.01) and a 34% reduction compared to DHA with no TXT (P < 0.04). DHA + TXT reduced MAXF401 xenografts growth compared to control and control + TXT (by 43% and 34%, respectively, P < 0.05). In both xenografts, DHA + TXT resulted in a higher expression of proapoptotic proteins Ripk1 and Bid, lower expression of proliferation marker Ki67 and anti-apoptotic proteins Bcl-2 and Parp, and a greater increase in cell cycle arrest as measured by decreased Survivin expression when compared to control + TXT mice (P < 0.05). CONCLUSIONS: This work is the first to confirm that DHA supplementation during chemotherapy treatment improves TXT action in two PDX models of TNBC. The results suggest that decreases in tumor size occurred via changes in apoptosis, cell proliferation, and cell cycle pathways.


Asunto(s)
Antineoplásicos/farmacología , Docetaxel/farmacología , Ácidos Docosahexaenoicos/farmacología , Animales , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Suplementos Dietéticos , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Femenino , Humanos , Ratones , Transducción de Señal/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Nutr ; 149(1): 46-56, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30601995

RESUMEN

Background: Docosahexaenoic acid (DHA) has been shown to reduce growth of breast cancer cells in vitro and in vivo; it may also benefit the action of cytotoxic cancer drugs. The mechanisms for these observations are not completely understood. Objectives: We sought to explore how pretreatment of MDA-MB-231 breast cancer cells with DHA alters gene expression with doxorubicin (DOX) treatment and confirm that feeding DHA to tumor-bearing nu/nu mice improves the efficacy of DOX. Methods: MDA-MB-231 cells were subjected to 4 conditions: a control mixture of 40 µM linoleic and 40 µM oleic acid (OALA), DHA (60 µM plus OALA), OALA DOX (0.41 µM), or DHA DOX (plus OALA) and assessed for effects on viability and function. Female nu/nu mice (6 wk old) bearing MDA-MB-231 tumors were randomly assigned to a nutritionally complete diet (20 g ± 2.8 g DHA/100 g diet) containing a polyunsaturated:saturated fat ratio of 0.5, with or without injections 2 times/wk of 5 mg DOX/kg for 4 wk. Results: Microarray and protein analysis indicated that DHA DOX cells, compared with OALA DOX, had upregulated expression of apoptosis genes, Caspase-10 (1.3-fold), Caspase-9 (1.4-fold), and Receptor (TNFRSF)-interacting serine-threonine kinase 1 (RIPK1) (1.2-fold), while downregulating cell cycle genes, Cyclin B1 (-2.1-fold), WEE1 (-1.6-fold), and cell division cycle 25 homolog C (CDC25C) (-1.8-fold) (P < 0.05). DHA DOX-treated mice had 50% smaller tumors than control mice (P < 0.05). Analysis of proapoptotic proteins from tumors of DHA DOX mice showed increased Caspase-10 (by 68%) and BH3 interacting domain death agonist (Bid) (by 50%), decreased B-cell CLL/lymphoma 2 (BCL2) (by 24%), and decreased cell cycle proteins Cyclin B1 and Cdc25c (both by 42%), compared with control mice (P < 0.05). Conclusions: Supplementation with DHA facilitates the action of DOX in MDA-MB-231 cells and in nu/nu mice, which may occur via amplification of the effect of DOX on apoptosis and cell cycle genes.


Asunto(s)
Ácidos Docosahexaenoicos/farmacología , Ácidos Docosahexaenoicos/farmacocinética , Doxorrubicina/farmacología , Doxorrubicina/farmacocinética , Neoplasias Experimentales/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Neoplasias de la Mama , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Interacciones Farmacológicas , Femenino , Humanos , Ratones , Ratones Desnudos , Distribución Aleatoria
11.
Int J Mol Sci ; 18(8)2017 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-28817068

RESUMEN

Globally, there were 14.1 million new cancer diagnoses and 8.2 million cancer deaths in 2012. For many cancers, conventional therapies are limited in their successes and an improved understanding of disease progression is needed in conjunction with exploration of alternative therapies. The long chain polyunsaturated fatty acid, docosahexaenoic acid (DHA), has been shown to enhance many cellular responses that reduce cancer cell viability and decrease proliferation both in vitro and in vivo. A small number of studies suggest that DHA improves chemotherapy outcomes in cancer patients. It is readily incorporated into cancer cell membranes and, as a result there has been considerable research regarding cell membrane initiated events. For example, DHA has been shown to mediate the induction of apoptosis/reduction of proliferation in vitro and in vivo. However, there is limited research into the effect of DHA on cell cycle regulation in cancer cells and the mechanism(s) by which DHA acts are not fully understood. The purpose of the current review is to provide a critical examination of the literature investigating the ability of DHA to stall progression during different cell cycle phases in cancer cells, as well as the consequences that these changes may have on tumour growth, independently and in conjunction with chemotherapy.


Asunto(s)
Ácidos Docosahexaenoicos/uso terapéutico , Neoplasias/dietoterapia , Neoplasias/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Quimioterapia , Humanos , Neoplasias/patología
12.
Lipids ; 47(11): 1019-30, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23054549

RESUMEN

Docosahexanoic acid (DHA) and eicosapentanoic acid (EPA) have been shown to possess anti-carcinogenic properties in mammary cancers, both in vitro and in vivo. The objective of this study was to investigate the effect of treating three different breast cancer cell lines with DHA or EPA on cellular growth, chemotherapy efficacy, and CD95 expression and localization in the cell. MDA-MB-231, MCF-7 and SKBr-3 cells were incubated with EPA or DHA with or without chemotherapy agents [doxorubicin (dox), Herceptin]. Cell growth was assessed by WST-1 assay and CD95 expression was investigated using flow cytometry, Western blotting and confocal microscopy. DHA and EPA inhibited the growth of all three breast cancer cell lines in a dose-dependent fashion (P < 0.05). DHA, and to a lesser extent EPA, induced the movement and raft clustering of CD95 in the cell membrane (via confocal microscopy) and the surface expression (via flow cytometry) in MDA-MB-231 cells. Neither fatty acid altered the growth/metabolic activity of the non-transformed MCF-12A breast cell line. Pre-treatment with DHA, but not EPA, improved the efficacy of dox in estrogen receptor negative MDA-MB-231 cells (P < 0.05), but not in the other two cell lines. Pre-treating cells with DHA increased CD95 surface expression (threefold) and the plasma membrane raft content of CD95 (2fold) and FADD (>4-fold) after dox treatment, compared to dox treatment alone (P < 0.05). This study demonstrated that pre-treatment of estrogen receptor negative MDA-MB-231 cells with DHA increased the anti-cancer effects of dox and presents evidence to suggest that this may be mediated in part by CD95-induced apoptosis.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Ácidos Docosahexaenoicos/farmacología , Microdominios de Membrana/efectos de los fármacos , Receptores de Estrógenos/metabolismo , Transducción de Señal/efectos de los fármacos , Receptor fas/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Ácido Eicosapentaenoico/farmacología , Femenino , Humanos , Células MCF-7 , Microdominios de Membrana/metabolismo , Receptores de Estrógenos/deficiencia , Relación Estructura-Actividad , Células Tumorales Cultivadas , Receptor fas/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA