Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 14(1): 7410, 2024 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-38548749

RESUMEN

Mitogen-activated protein kinase kinase kinase kinase 4 (MAP4K4) has recently emerged as a promising therapeutic target in cancer. In this study, we explored the biological function of MAP4K4 in radioresistant breast cancer cells using two MAP4K4 inhibitors, namely PF06260933 and GNE-495. Radioresistant SR and MR cells were established by exposing SK-BR-3 and MCF-7 breast cancer cells to 48-70 Gy of radiation delivered at 4-5 Gy twice a week over 10 months. Surprisingly, although radioresistant cells were derived from two different subtypes of breast cancer cell lines, MAP4K4 was significantly elevated regardless of subtype. Inhibition of MAP4K4 with PF06260933 or GNE-495 selectively targeted radioresistant cells and improved the response to irradiation. Furthermore, MAP4K4 inhibitors induced apoptosis through the accumulation of DNA damage by inhibiting DNA repair systems in radioresistant cells. Notably, Inhibition of MAP4K4 suppressed the expressions of ACSL4, suggesting that MAP4K4 functioned as an upstream effector of ACSL4. This study is the first to report that MAP4K4 plays a crucial role in mediating the radioresistance of breast cancer by acting upstream of ACSL4 to enhance DNA damage response and inhibit apoptosis. We hope that our findings provide a basis for the development of new drugs targeting MAP4K4 to overcome radioresistance.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Neoplasias de la Mama/genética , Línea Celular Tumoral , Tolerancia a Radiación/genética , Reparación del ADN , Células MCF-7 , Apoptosis/efectos de la radiación , Proteínas Serina-Treonina Quinasas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo
2.
Int J Mol Sci ; 24(15)2023 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-37569361

RESUMEN

Platelets play crucial roles in cardiovascular diseases (CVDs) by regulating hemostasis and blood coagulation at sites of blood vessel damage. Accumulating evidence indicates daidzein inhibits platelet activation, but the mechanism involved has not been elucidated. Thus, in this study, we investigated the mechanism responsible for the inhibition of collagen-induced platelet aggregation by daidzein. We found that in collagen-induced platelets, daidzein suppressed the production of thromboxane A2 (TXA2), a molecule involved in platelet activation and aggregation, by inhibiting the cytosolic phospholipase A2 (cPLA2) signaling pathway. However, daidzein did not affect cyclooxygenase-1 (COX-1). Furthermore, daidzein attenuated the PI3K/PDK1/Akt/GSK3αß and MAPK (p38, ERK) signaling pathways, increased the phosphorylation of inositol trisphosphate receptor1 (IP3R1) and vasodilator-stimulated phosphoprotein (VASP), and increased the level of cyclic adenosine monophosphate (cAMP). These results suggest that daidzein inhibits granule release (ATP, serotonin, P-selectin), integrin αIIbß3 activation, and clot retraction. Taken together, our study demonstrates that daidzein inhibits collagen-induced platelet aggregation and suggests that daidzein has therapeutic potential for the treatment of platelet aggregation-related diseases such as atherosclerosis and thrombosis.


Asunto(s)
Activación Plaquetaria , Inhibidores de Agregación Plaquetaria , Humanos , Inhibidores de Agregación Plaquetaria/farmacología , Agregación Plaquetaria , Plaquetas/metabolismo , Fosforilación , Tromboxanos/metabolismo , Colágeno/metabolismo
3.
IUBMB Life ; 75(9): 765-777, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37492896

RESUMEN

Chemotherapy failure is often caused by drug resistance, for which no effective treatment strategy has been established. Many studies have been undertaken with the aim of overcoming drug resistance using natural products. Arctigenin (ATG), a natural product, has been investigated for its anti-cancer effects in HER2-overexpressing, ER-positive, and triple-negative breast cancer cells. We investigated the efficacy of ATG against self-established doxorubicin (DOX)-resistant breast cancer cells (MCF-DR and MDA-DR cells) derived from MCF-7 and MDA-MB-231 cells, respectively. ATG was found to increase DOX intracellular levels by downregulating multidrug Resistance 1 (MDR1) mRNA expression in DOX-resistant cells. In addition, combined treatment with DOX and ATG (DOX/ATG) reduced the viability of and colony formation by DOX-resistant cells. DOX/ATG also significantly induced G2/M cell cycle arrest by suppressing the Cyclin D1/CDK4/RB pathways and suppressed the expressions of MDR1 and Cyclin D1 by inhibiting the Mitogen-activated protein kinase (MAPK)/Activating protein-1 (AP-1) signaling pathways. Furthermore, DOX/ATG induced DNA damage and attenuated the expressions of RAD51 and Ku80. However, PARP1 (Poly [ADP-ribose] polymerase1) cleavage and AIF (Apoptosis-inducing factor) induced apoptosis did not occur despite DNA damage-induced cell death. Rather, flow cytometry showed that DOX/ATG caused necrosis. In summary, DOX/ATG increased intracellular DOX levels by inhibiting MDR1 and inducing G2/M arrest by inhibiting the Cyclin D1/CDK4/RB pathways and causing necrosis by damaging DNA. Our results suggest that ATG might be used as an adjuvant to enhance the efficacy of DOX in DOX-resistant breast cancer.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Ciclina D1 , Apoptosis , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Puntos de Control de la Fase G2 del Ciclo Celular , Doxorrubicina/farmacología , Puntos de Control del Ciclo Celular , Necrosis
4.
Pharmaceuticals (Basel) ; 16(5)2023 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-37242455

RESUMEN

Considerable emphasis is being placed on combinatorial chemotherapeutic/natural treatments for breast cancer. This study reveals the synergistic anti-tumor activity of morin and Doxorubicin (Dox) co-treatment on MDA-MB-231 triple-negative breast cancer (TNBC) cell proliferation. Morin/Dox treatment promoted Dox uptake and induced DNA damage and formation of nuclear foci of p-H2A.X. Furthermore, DNA repair proteins, RAD51 and survivin, and cell cycle proteins, cyclin B1 and forkhead Box M1 (FOXM1), were induced by Dox alone but attenuated by morin/Dox co-treatment. In addition, Annexin V/7-AAD analysis revealed that necrotic cell death after co-treatment and apoptotic cell death by Dox alone were associated with the induction of cleaved PARP and caspase-7 without Bcl-2 family involvement. FOXM1 inhibition by thiostrepton showed that co-treatment caused FOXM1-mediated cell death. Furthermore, co-treatment downregulated the phosphorylation of EGFR and STAT3. Flow cytometry showed that the accumulation of cells in the G2/M and S phases might be linked to cellular Dox uptake, p21 upregulation, and cyclin D1 downregulation. Taken together, our study shows that the anti-tumor effect of morin/Dox co-treatment is due to the suppression of FOXM1 and attenuation of EGFR/STAT3 signaling pathways in MDA-MB-231 TNBC cells, which suggests that morin offers a means of improving therapeutic efficacy in TNBC patients.

5.
Pharmaceuticals (Basel) ; 17(1)2023 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-38256858

RESUMEN

Anthocyanin oligomers (AOs) are phytochemicals synthesized by fermenting anthocyanins extracted from grape skins and are more biologically active than monomeric anthocyanins. In this study, we evaluate the effects of an AO on triple-negative MDA-MB-231 and HER2-overexpressing SK-BR-3 breast cancer cells. The cell viability of MDA-MB-231 and SK-BR-3 cells was significantly inhibited in a concentration-dependent manner by AO treatment for 24 h, while delphinidin (a monomeric anthocyanin) had no effect on cell viability. In addition, the AO increased H2A.X phosphorylation (a marker of DNA damage), reduced RAD51 (a DNA repair protein) and survivin (a cell survival factor) protein levels, and induced apoptosis by caspase-3-dependent PARP1 cleavage in both cell lines. Surprisingly, the AO induced autophagy by increasing intracellular LC3-II puncta and LC3-II and p62 protein levels. In addition, the AO inhibited the mTOR pathway in MDA-MB-231 and SK-BR-3 cells by suppressing the HER2, EGFR1, and AKT pathways. These results demonstrate that the anti-cancer effect of the AO was due to the induction of apoptosis and autophagy via cleaved caspase-3-mediated PARP1 cleavage and mTOR pathway inhibition, respectively. Furthermore, our results suggest that anthocyanin oligomers could be considered potential candidates for breast cancer treatment.

6.
Cancer Biother Radiopharm ; 37(7): 544-552, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33259733

RESUMEN

Background: Recently, several clinical studies have reported that combination treatments of radiation therapy (RT) and immunotherapy in patients with multiple lesions can improve tumor regression at a distance from the irradiated site, known as the abscopal effect. However, when RT and immunotherapy are concurrently applied, it is hard to distinguish the pure systemic effects of RT from those of the immunotherapy drug. In this preclinical study, the authors investigated the systemic antitumor effects of RT alone according to fraction dose size and splitting schedules. Materials and Methods: 4T1 mouse breast cancer cells were implanted into the right and left sides of mammary gland fat pads of BALB/c mice, followed by irradiation with 6 Gy × 3, 8 Gy × 2, and 13 Gy × 1 fractions when the right-side tumors were palpable. Results: The different irradiation schedules produced similar antitumor effects in irradiated right-side tumors and unirradiated left-side tumors. However, 8 Gy × 2 and 13 Gy × 1 fractions exhibited better antimetastatic potential than that from irradiation using 6 Gy × 3 fractions. Furthermore, 8 Gy × 2 and 13 Gy × 1 fractions produced higher expressions of HMGB1 and lower expressions of the proinflammatory cytokines, IFN-γ, TNF-α, IL-6, and IL-1ß, from the irradiated tumor tissues. Conclusions: These findings suggest that 8 Gy × 2 and 13 Gy × 1 fractions can provide better systemic antitumor effects than 6 Gy × 3 fractions. The authors hope these results provide clues to optimize RT dose regimens to make the abscopal effect clinically more relevant in future combination treatments.


Asunto(s)
Inmunoterapia , Neoplasias , Animales , Ratones , Ratones Endogámicos BALB C , Factor de Necrosis Tumoral alfa
7.
Pharmacol Rep ; 73(5): 1315-1327, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33993438

RESUMEN

BACKGROUND: Morin, a flavonoid extracted from Moraceace family and exhibits several pharmacological activities including anti-cancer activity. Although the anticancer activity of morin in breast cancer was estimated in some investigations, the pharmaceutical mechanism has not been fully elucidated. Therefore, we investigated to unveil the detail signaling pathway in morin-treated in MDA-MB-231 triple-negative breast cancer cells. METHODS: The cytotoxicity of morin in MDA-MB-231 cells was confirmed by sulforhodamine B (SRB) assay and colony formation assay. Flow cytometry was performed to examine the cell cycle and cell death patterns and the protein expression and phosphorylation were detected by western blotting. RESULTS: Our results showed that morin inhibited MDA-MB-231 cells proliferation in time and concentration-dependent manner. Morphological changes were observed when treated with various concentration of morin in MDA-MB-231 cells. In regard to protein expression, morin induced the phosphorylation of ERK and p-H2A.X and decreased the level of DNA repair markers, RAD51 and survivin. In addition, flow cytometry showed S and G2/M arrest by morin that was associated with the decrease in the protein expression of cyclin A2 and cyclin B1 and upregulation of p21. Interestingly, annexin V/PI staining result clearly showed that morin induced cell death without apoptosis. Furthermore, attenuated FoxM1 by morin was co-related with cell cycle regulators including p21, cyclin A2 and cyclin B1. CONCLUSION: Taken together, our study indicates that morin-induced cell death of MDA-MB-231 is caused by sustained cell cycle arrest via the induction of p21 expression by activation of ERK and repression of FOXM1 signaling pathways.


Asunto(s)
Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Flavonoides/farmacología , Neoplasias de la Mama Triple Negativas , Antioxidantes/farmacología , Autofagia/efectos de los fármacos , Butadienos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Estructura Molecular , Nitrilos/farmacología
8.
Oncol Rep ; 46(1)2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33982761

RESUMEN

Morin (2',3,4',5,7­pentahydroxyflavone), a flavonoid isolated from members of the Moraceae family and the leaves of Cudranaia tricuspidata Buread, is a well­known natural substance with anti­inflammatory, antioxidative, antimetastasis, and anticancer effects. However, its anticancer activity has not been comprehensively investigated in human epidermal growth factor receptor 2 (HER2)­overexpressing breast cancer cells. Here, we evaluated the effects of morin on metastasis and cell viability in HER­2­overexpressing human breast cancer SK­BR­3 cells. Our results revealed that morin (150­200 µM) prevented endothelial growth factor (EGF)­induced metastatic potential and suppressed cell migration and MMP­9 activity by inhibiting the EGFR signaling pathway in SK­BR­3 cells by gelatin zymography, wound healing assay and western blotting. Interestingly, morin­induced reductions in cell viability were found to be associated with inhibition of the HER2/EGFR signaling pathway by sulforhodamine B assay and western blotting. Morin also induced the phosphorylation of H2A.X and downregulated the expression levels of RAD51 and survivin, which implied morin­induced DNA damage and that this damage accumulated in HER­2­overexpressing SK­BR­3 cells. Western blot analysis and fluorescent immunocytochemisty showed that morin also activated autophagy after 24 h of treatment and this was maintained at 48 h when activation of apoptosis via PARP cleavage resulted in the activation of caspase­3 and ­7, which was associated with the release of cytochrome c to the cytosol from mitochondria. In addition, the phosphorylation of p38 and JNK was enhanced in the HER­2­overexpressing SK­BR­3 cells by morin after 24 and 48 h of treatment, which suggested p38 and JNK participate in morin­induced cell death. Taken together, the present investigation indicates that morin is a powerful therapeutic candidate for the treatment of HER2­overexpressing breast cancer because it suppresses the EGFR signaling pathway, induces cell death by inhibiting the HER2/EGFR signaling pathway, and suppresses metastatic potential.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Neoplasias de la Mama/metabolismo , Flavonoides/farmacología , Receptor ErbB-2/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Metaloproteinasa 9 de la Matriz/metabolismo , Metástasis de la Neoplasia/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
9.
Biochem Pharmacol ; 190: 114635, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34058187

RESUMEN

Since trastuzumab-resistance remains a major obstacle to the successful treatment of HER2-positive breast cancer, a detailed understanding of the mechanisms responsible is required to direct future pharmacotherapeutic strategies. Recently, several studies have indicated that the quiescent natures of cancer stem cells contribute to treatment resistance and tumor recurrence. Thus, in this study, we investigated the mechanism underlying trastuzumab resistance in a quiescent cell population using tumorsphere cultures and explored better therapeutic strategies to overcome trastuzumab resistance in HER2-positive breast cancer patients. We observed that most cells in SK-BR-3 tumorspheres were quiescent, showing the accumulation of cells at the G0/G1 phase as compared to cells in monolayer culture. Furthermore, SK-BR-3 tumorspheres exhibited enhanced EGFR/HER2 signaling, which was incompletely inhibited by trastuzumab, and subsequently led to trastuzumab-resistance. Interestingly, cytoplasmic estrogen receptor α (ERα) expression was markedly elevated in tumorspheres and was associated with enhanced EGFR/HER2 signaling. Accordingly, inhibition of ERα with tamoxifen selectively targeted tumorspheres rather than cells in monolayer culture and overcame trastuzumab resistance in tumorspheres. Taken together, our findings indicate that crosstalk between cytoplasmic ERα and the HER2/EGFR signaling pathway can be considered a novel therapeutic target for quiescent cell populations within HER2-positive breast cancer and that simultaneous inhibition of ER and the EGFR/HER2 pathway may prevent trastuzumab resistance. We hope that these results provide a basis for the use of combinations of tamoxifen and trastuzumab in HER2-positive breast cancer patients.


Asunto(s)
Resistencia a Antineoplásicos , Receptores ErbB/metabolismo , Receptor alfa de Estrógeno/metabolismo , Receptor ErbB-2/metabolismo , Tamoxifeno/farmacología , Trastuzumab/farmacología , Antineoplásicos Hormonales/farmacología , Antineoplásicos Inmunológicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Citoplasma , Receptores ErbB/genética , Estradiol/farmacología , Receptor alfa de Estrógeno/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Lapatinib/farmacología , Receptor ErbB-2/genética , Transducción de Señal
10.
J Ethnopharmacol ; 274: 114081, 2021 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-33798660

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Chaga mushrooms (Inonotus obliquus) are commonly used in traditional treatments in Eastern Europe and Asia due to their diverse pharmacological effects, including anti-tumor and immunologic effects. Thus, many cancer patients take Chaga mushrooms as a complementary medicine, even during chemotherapy or radiotherapy. However, few studies have investigated the effects or molecular targets of Chaga mushrooms in breast cancer. AIM OF THE STUDY: Herein, we examined the anticancer effects of Chaga mushrooms in different types of breast cancer cell lines, and explored the underlying molecular mechanism to better understand their effects and benefits. MATERIALS AND METHODS: Chaga mushroom extract (CME) was prepared by extracting Chaga mushrooms with 70% ethanol. The cytotoxic effects of CME were assessed by MTT assay and protein expressions were evaluated by western blotting. To evaluate in vivo anti-tumor effects of CME, CME (2 g/kg) was orally administered to 4T1 tumor-bearing BALB/c mice every other day over 30 days (15 administrations), and tumor sizes were measured. Silica gel column chromatography was used to fractionate CME, and major constituents responsible for cytotoxic effects of CME were identified by 1H/13C-NMR and LC-MS. RESULTS: CME inhibited the proliferation of 4T1 mouse breast cancer cells in a dose and time-dependent manner. The expression of LC3 and phosphorylation of AMPK were increased by CME, while the phosphorylation of mTOR, S6, and S6K1 were suppressed, suggesting that CME induced autophagy by activating AMPK and inhibiting mTOR signaling pathways. Consistent with its observed cytotoxic effect in vitro, CME effectively suppressed tumor growth in 4T1 tumor-bearing BALB/c mice. In addition, inotodiol and trametenolic acid were identified as the major constituents responsible for the cytotoxic effects of CME on breast cancer cells. Moreover, inotodiol and trametenolic acid-enriched fractions both exhibited cytotoxic effects regardless of breast cancer cell subtypes and did not interfere with the cytotoxic effects of conventional drugs. CONCLUSIONS: Taken together, Chaga mushroom extract induced autophagy by activating AMPK and inhibiting the mTOR signaling pathway. Our data suggest Chaga mushrooms may be a beneficial complementary medicine for breast cancer patients.


Asunto(s)
Agaricales , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Mezclas Complejas/uso terapéutico , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Mezclas Complejas/química , Mezclas Complejas/farmacología , Femenino , Humanos , Lanosterol/análogos & derivados , Lanosterol/análisis , Lanosterol/farmacología , Ratones Endogámicos BALB C , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Triterpenos/análisis , Triterpenos/farmacología
11.
Pharmacol Rep ; 73(2): 629-641, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33677703

RESUMEN

BACKGROUND: Human epidermal growth factor receptor 2 (HER2) is well-known as the therapeutic marker in breast cancer. Therefore, we evaluated anti-cancer activity of arctigenin (ATG) on in SK-BR-3 HER2-overexpressing human breast cancer cells. METHODS: Cell viability and cytotoxicity were analyzed with MTT and colony-forming assay and cell cycle analysis was performed by flow cytometry. The expression and/or phosphorylation of proteins in whole cell lysate and mitochondrial fraction were analyzed by Western blotting. Cellular levels of LC3 and sequestosome 1 (SQSTM1/P62) were observed by immunofluorescence analysis. RESULTS: The result showed that ATG decreased cell viability of SK-BR-3 cells in a concentration-dependent manner. Moreover, ATG increased the sub G1 population linked to the suppression of HER2/EGFR1 signaling pathway. Furthermore, ATG increased the phosphorylation of H2AX and down-regulated RAD51 and survivin expressions, indicating that ATG induced DNA damage and inhibited the DNA repair system. We also found that cleavages of caspase-7 and PARP by releasing mitochondrial cytochrome c into the cytoplasm were induced by ATG treatment for 72 h through the reduction of Bcl-2 and Bcl-xL levels in mitochondria. In contrast, the levels of LC-3 and SQSTM1/P62 were increased by ATG for 24 h through the Akt/mTOR and AMPK signaling pathway. CONCLUSIONS: Taken together, this study indicates that autophagy-linked apoptosis is responsible for the anti-cancer activity of ATG in SK-BR-3 cells, and suggests that ATG is considered a potential therapeutic for the treatment of HER2-overexpressing breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Furanos/farmacología , Lignanos/farmacología , Receptor ErbB-2/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Receptor ErbB-2/genética , Transducción de Señal/efectos de los fármacos
12.
Biochem Pharmacol ; 188: 114519, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33737052

RESUMEN

Mitogen-activated protein kinase kinase kinase kinase 4 (MAP4K4) is implicated in type 2 diabetes mellitus, insulin tolerance, inflammation, cancer, and atherosclerosis. We found that GNE 495 and PF 06260933 (both potent and selective MAP4K4 inhibitors) regulated human platelet activation. Immunoblotting revealed human platelets express MAP4K4, and that GNE 495 and PF 06260933 inhibited collagen-, ADP-, and thrombin-induced platelet aggregation and eventually suppressed granule release, TXA2 generation, integrin αIIbß3 activation, and clot retraction. In addition, both inhibitors elevated intracellular levels of cAMP, and coincubation with GNE 495 and aspirin or dipyridamole (a phosphodiesterase inhibitor) synergistically inhibited collagen-induced platelet aggregation and TXA2 generation. Moreover, both inhibitors phosphorylated VASP (ser157), IP3 receptor, and PKA and attenuated MAPK and PI3K/Akt/GSK3ß signaling pathways. This study is the first to demonstrate that MAP4K4 inhibitors reduce thrombus formation by inhibiting platelet activation. These findings also suggest MAP4K4 be considered an emerging target protein for the treatment of thrombosis.


Asunto(s)
Aminopiridinas/farmacología , Retracción del Coagulo/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Agregación Plaquetaria/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Adolescente , Adulto , Retracción del Coagulo/fisiología , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Persona de Mediana Edad , Agregación Plaquetaria/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Adulto Joven
13.
Biomed Pharmacother ; 130: 110535, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32707441

RESUMEN

Pathophysiological reaction of platelets in the blood vessel is an indispensable part of thrombosis and cardiovascular disease, which is the most common cause of death in the world. In this study, we performed in vitro assays to evaluate antiplatelet activity of arctigenin in human platelets and attempted to identify the mechanism responsible for thromboxane A2 (TXA2) generation, integrin αIIbß3 activation and cAMP pathway. Arctigenin exhibited obvious inhibitory effects on collagen-, thrombin-, and ADP-induced human platelet aggregation, granule secretion, TXA2 generation, integrin αIIbß3 activation, and clot retraction. Additionally, we found that arctigenin attenuated PI3K/Akt/mTOR/GSK-3ß and MAPK signaling pathways, and increased cAMP level. Accordingly, the findings support that arctigenin attenuates thrombotic events through the inhibition of platelet activation and platelet plug formation. Therefore, we suggest that arctigenin may have therapeutic potential as an antiplatelet and antithrombotic agent.


Asunto(s)
Retracción del Coagulo/efectos de los fármacos , AMP Cíclico , Fibrinolíticos/farmacología , Furanos/farmacología , Lignanos/farmacología , Activación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Transducción de Señal/efectos de los fármacos , Tromboxano A2/biosíntesis , Plaquetas/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta/efectos de los fármacos , Humanos , Técnicas In Vitro , Proteínas Quinasas Activadas por Mitógenos , Proteína Oncogénica v-akt/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Serina-Treonina Quinasas TOR/efectos de los fármacos
14.
Thromb Res ; 195: 87-94, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32682003

RESUMEN

Caffeic acid (CA) is a polyphenol widely distributed in the plant kingdom. Studies have shown CA possesses antithrombotic activity in mouse cerebral arterioles in vivo and inhibits platelet aggregation in vitro. However, little is known regarding the effects of CA on platelet-mediated clot retraction. We investigated the effects of CA on platelet activation and clot retraction in response to thrombin. CA inhibited thrombin-induced platelet aggregation, calcium mobilization, adenosine 1,4,5-tri-phosphate (ATP) release, P-selectin expression and fibrinogen binding to integrin αIIbß3 activation without inducing any cytotoxic effect, and inhibited the phosphorylations of protein kinase B (Akt) and extracellular signal-regulated kinase (ERK) in thrombin-stimulated platelets. In addition, CA enhanced cyclic adenosine monophosphate (cAMP) generation, which led to the phosphorylations of vasodilator-stimulated phosphoprotein (VASP) and inositol trisphosphate (IP3) receptor, and reduced clot retraction without any anticoagulation effect. Dipyridamole, a phosphodiesterase 3 (PDE3) inhibitor, reduced clot retraction, which suggested CA-mediated cAMP generation is the main signaling pathway responsible for its inhibition of clot retraction. Taken together, the findings of the present study suggest that CA may have potential as a therapeutic for the prevention of thrombotic disorders.


Asunto(s)
Plaquetas , Fibrinolíticos , Animales , Ácidos Cafeicos , Retracción del Coagulo , Fibrinolíticos/farmacología , Humanos , Ratones , Agregación Plaquetaria
15.
J Med Food ; 23(8): 886-894, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32522066

RESUMEN

Deep seawater (DSW) has been investigated for its lipid-lowering effects, but clinical evidence is still far from conclusive. Therefore, this study was conducted to examine the effects of refined DSW (RDSW) on hypercholesterolemia. In this randomized, double-blind, placebo-controlled trial, 78 Korean participants were randomized to either an RDSW group that drank RDSW for 8 weeks or a placebo group. Clinical laboratory information was collected from all subjects at 0, 4, and 8 weeks. Both groups showed a significant reduction in total cholesterol (TC), whereas only the RDSW group demonstrated a significant decrease in low-density lipoprotein cholesterol (LDL-c) during the study. Stratified analysis of both groups revealed a significant reduction of TC in the moderately high TC subgroup. However, only the RDSW exhibited a significant decline of LDL-c in the high LDL-c subgroup. In addition, lipoprotein(a) decreased significantly in the RDSW group, but not in the placebo. RDSW did not affect other lipid profiles, including high-density lipoprotein cholesterol (HDL-c), triglyceride, free fatty acid, apolipoproteins, and other markers including inflammation marker, hematological parameters, blood and urine chemistry, and vital signs. RDSW improved lipid profiles by decreasing TC and LDL-c while maintaining HDL-c levels in people with hypercholesterolemia.


Asunto(s)
Anticolesterolemiantes/uso terapéutico , Hipercolesterolemia , Agua de Mar/química , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Método Doble Ciego , Humanos , Hipercolesterolemia/tratamiento farmacológico , Lípidos/sangre , República de Corea
16.
Int J Mol Sci ; 21(8)2020 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-32340377

RESUMEN

Several reports have described the anti-cancer activity of arctigenin, a lignan extracted from Arctium lappa L. Here, we investigated the effect of arctigenin (ATG) on doxorubicin (DOX)-induced cell death using MDA-MB-231 human breast cancer cells. The results showed that DOX-induced cell death was enhanced by ATG/DOX co-treatment in a concentration-dependent manner and that this was associated with increased DOX uptake and the suppression of multidrug resistance-associated protein 1 (MRP1) gene expression in MDA-MB-231 cells. ATG enhanced DOX-induced DNA damage and decreased the phosphorylation of signal transducer and activator of transcription 3 (STAT3) and the expressions of RAD51 and survivin. Cell death caused by ATG/DOX co-treatment was mediated by the nuclear translocation of apoptosis inducing factor (AIF), reductions in cellular and mitochondrial Bcl-2 and Bcl-xL, and increases in mitochondrial BAX levels. However, caspase-3 and -7 did not participate in DOX/ATG-induced cell death. We also found that DOX/ATG-induced cell death was linked with activation of the p38 signaling pathway and suppressions of the phosphorylations and expressions of Akt and c-Jun N-terminal kinase. Taken together, these results show that ATG enhances the cytotoxic activity of DOX in MDA-MB-231 human breast cancer cells by inducing prolonged p21 expression and p38-mediated AIF-dependent cell death. In conclusion, our findings suggest that ATG might alleviate the side effects and improve the therapeutic efficacy of DOX.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Doxorrubicina/farmacología , Furanos/farmacología , Lignanos/farmacología , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Mitocondrias/metabolismo , Fosforilación
17.
Mol Med Rep ; 21(3): 1374-1382, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32016480

RESUMEN

Arctigenin is a natural lignan that is found in burdock with anti­viral, ­oxidative, ­inflammatory and anti­tumor activities. In the current study, the effect of arctigenin on metastatic potential was examined in 4T­1 mouse triple­negative breast cancer cells. The results indicated that arctigenin inhibited cell motility and invasiveness, which was determined using wound healing and transwell invasion assays. Arctigenin suppressed matrix metalloprotease­9 (MMP­9) activity via gelatin zymography, and protein expression of cyclooxygenase­2 (COX­2) and MMP­3. Furthermore, arctigenin attenuated the mRNA expression of metastatic factors, including MMP­9, MMP­3 and COX­2. Based on these results, the effect of arctigenin on the mitogen­activated protein kinase (MAPK)/activating protein­1 (AP­1) signaling pathway was assessed in an attempt to identify the regulatory mechanism responsible for its anti­metastatic effects. Arctigenin was demonstrated to inhibit the phosphorylation of extracellular signal­regulated protein kinase (ERK) and c­Jun N­terminal kinase (JNK), and the nuclear translocations of the AP­1 subunits, c­Jun and c­Fos. In summary, the present study demonstrated that in 4T­1 mouse triple­negative breast cancer cells the anti­metastatic effect of arctigenin is mediated by the inhibition of MMP­9 activity and by the inhibition of the metastasis­enhancing factors MMP­9, MMP­3 and COX­2, due to the suppression of the MAPK/AP­1 signaling pathway. The results of the current study demonstrated that arctigenin exhibits a potential for preventing cell migration and invasion in triple negative breast cancer.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Arctium/química , Furanos/farmacología , Lignanos/farmacología , Sistema de Señalización de MAP Quinasas , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Factor de Transcripción AP-1/metabolismo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Femenino , Humanos , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Invasividad Neoplásica , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
18.
Int J Mol Med ; 45(2): 658-668, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31894254

RESUMEN

Mineral­balanced deep sea water (MBDSW), an unlimited natural sea source, has been demonstrated to minimize the risk of developing cardiovascular diseases, such as obesity, hypertension, inflammation and hyperlipidemia. This study investigated the effects of MBDSW [magnesium (Mg):calcium (Ca) ratio, 3:1] on platelet activation. MBDSW significantly inhibited the collagen­ and thrombin­induced platelet aggregation of human platelets. In collagen­induced platelets, MBDSW inhibited intracellular calcium mobilization, granule secretion [serotonin, adenosine triphosphate (ATP) and P­selectin expression] and thromboxane A2 (TXA2) production. Moreover, MBDSW markedly inhibited Akt and extracellular signal­regulated kinase (ERK) phosphorylation, but not that of c­Jun N­terminal kinase (JNK) and p38. Moreover, MBDSW phosphorylated inositol 1,4,5­triphosphate receptor (IP3R) and vasodilator­stimulated phosphoprotein (VASP), and it increased the cyclic adenosine monophosphate (cAMP) level in collagen­induced human platelets. Dipyridamole, a phosphodiesterase (PDE) inhibitor, significantly increased the cAMP level and regulated the Akt, ERK and VASP (Ser157) levels in a manner similar to that of MBDSW. In addition, LY294002, an Akt inhibitor, inhibited the phosphorylation of ERK, and U0126, an ERK inhibitor, inhibited the phosphorylation of Akt. Taken together, the results of the present investigation suggest that the inhibitory effects of MBDSW on platelet aggregation may be associated with the cross­inhibition of Akt and ERK phosphorylation. These results strongly indicate that MBDSW may have preventive or therapeutic potential for platelet aggregation­mediated diseases, such as thrombosis, atherosclerosis and myocardial infarction.


Asunto(s)
Aguas Minerales , Inhibidores de Agregación Plaquetaria/farmacología , Agregación Plaquetaria/efectos de los fármacos , Agua de Mar , Plaquetas/citología , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Aguas Minerales/análisis , Inhibidores de Agregación Plaquetaria/análisis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Agua de Mar/análisis , Transducción de Señal/efectos de los fármacos
19.
Int J Oncol ; 56(2): 630-640, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31939617

RESUMEN

Plant flavonoid 2',3,4',5,7­pentahydroxyflavone (morin hydrate), isolated from the family Moraceae (Morus alba L.), is known to have anti­inflammatory and anticancer effects. However, its pharmaceutical effects on metastasis have not been fully elucidated to date. Therefore, the current study investigated the effects of morin hydrate on cancer metastasis in MCF­7 human breast cancer cells. The results showed that morin hydrate suppressed 12­O­tetradecanoylphorbol­13­acetate (TPA)­induced cell migration and invasion via the inhibition of matrix metalloproteinase (MMP)­9 activity. Furthermore, gene expression level of MMP­9, MMP­7, urokinase plasminogen activator (uPA), uPA receptor (uPAR) and fibronectin were significantly decreased by morin hydrate treatment. Morin hydrate inhibited the phosphorylation of Akt and glycogen synthase kinase (GSK)­3ß, and downregulated the expression of an activator protein­1 subunit c­Fos. In addition, the GSK­3ß phosphorylation and c­Fos expression were suppressed by PI3K/Akt pathway inhibitors, LY294002 and wortmannin. Taken together, these results demonstrated that morin hydrate reduced the metastatic potential in TPA­treated MCF­7 human breast cancer cells via the inhibition of MMPs, uPA and uPAR, and the underlying Akt/GSK­3ß/c­Fos pathway. Therefore, the present investigation suggested that morin hydrate may be a natural substance with a preventive potential for metastasis in breast cancer cells.


Asunto(s)
Neoplasias de la Mama/patología , Flavonoides/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Acetato de Tetradecanoilforbol/toxicidad , Neoplasias de la Mama/metabolismo , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Células MCF-7 , Invasividad Neoplásica , Metástasis de la Neoplasia , Fosforilación , Proteínas Proto-Oncogénicas c-fos/metabolismo
20.
Eur J Pharmacol ; 865: 172734, 2019 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-31614139

RESUMEN

Morin hydrate is an active constituent of Morus alba L, Prunus dulcis, and Cudrania tricuspidata and has been reported to inhibit platelet activation in vivo and in vitro, but no reports have been issued on its regulation of αIIbß3, a platelet-specific integrin and thromboxane A2 (TXA2), positive feedback molecule. In this study, we investigated the anti-platelet activity of morin hydrate in collagen- and thrombin-induced human platelets and attempted to identify the mechanism responsible for integrin αIIbß3 activation and TXA2 generation. Our results demonstrated that morin hydrate (25-100 µM) inhibited collagen- and thrombin-induced platelet aggregation, granule secretion (P-selectin expression, ATP, and serotonin release), calcium mobilization, TXA2 production, integrin αIIbß3 activation, and clot retraction. Additionally, morin hydrate attenuated the phosphorylations of phospholipase Cγ2 (PLCγ2), cytosolic phospholipase A2 (cPLA2), phosphoinositide 3-kinase (PI3K), Akt, c-Jun N-terminal kinase (JNK), and extracellular signal-regulated kinase (ERK), and enhanced the phosphorylations of inositol trisphosphate receptor (IP3 receptor) and cyclic adenosine monophosphate (cAMP) generation. However, it had no effect on the coagulation pathway. Taken together, these observations indicate morin hydrate inhibits platelet-mediated thrombosis by down-regulating TXA2 production and integrin αIIbß3 activation, and by upregulating cAMP generation, and thus, inhibits clot retraction. These results suggest morin hydrate may have therapeutic potential as a treatment for platelet-activation-related diseases.


Asunto(s)
Retracción del Coagulo/efectos de los fármacos , AMP Cíclico/metabolismo , Flavonoides/farmacología , Activación Plaquetaria/efectos de los fármacos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Tromboxano A2/metabolismo , Coagulación Sanguínea/efectos de los fármacos , Colágeno/farmacología , Humanos , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Fosforilación/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Trombina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...