Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Biol Macromol ; 213: 858-870, 2022 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-35697164

RESUMEN

G-quadruplex regulates a wide spectrum of biological processes, including telomere maintenance, DNA replication and transcription. The development of small molecules to selectively target G-quadruplex and their application remain hotspots in cancer therapy. Here, we explored the biological effect of G-quadruplexes stabilizer Tetra-Pt(bpy) in telomerase-positive cancer cells. Telomere maintenance was evaluated by telomerase repeat amplification protocol, chromosome orientation fluorescence in situ hybridization and telomere restriction fragment assays. We found that Tetra-Pt(bpy) accelerates telomere shortening through dual inhibition of telomerase activity and telomere sister chromatin exchanges mediated by telomeric G-quadruplexes. Consequently, Tetra-Pt(bpy)-treated cancer cells became enriched with extremely short telomeres and produced a strong telomeric DNA damage response following long-term treatment, leading to cell proliferation inhibition and senescence. Experimental evidence from RNA seq and cell migration-related assays showed that Tetra-Pt(bpy) decreased cell-matrix adhesion and inhibited the migration of non-senescent tumor cells. Mechanistically, Tetra-Pt(bpy) induced the formation of G-quadruplexes in focal adhesion kinase (FAK)-encoding gene PTK2, resulting in FAK transcription inhibition. Tetra-Pt(bpy) reduced xenograft tumor formation and inhibited tumor cell growth and migration in mice. This study further elucidates the function of G-quadruplexes in the human genome and reveals the potential of Tetra-Pt(bpy) as a novel chemotherapeutic agent for targeting telomerase-positive cancer cells.


Asunto(s)
Antineoplásicos , G-Cuádruplex , Neoplasias , Telomerasa , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Humanos , Hibridación Fluorescente in Situ , Ratones , Neoplasias/tratamiento farmacológico , Telomerasa/genética , Telomerasa/metabolismo , Telómero/genética , Telómero/metabolismo
2.
Phytomedicine ; 101: 154087, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35429924

RESUMEN

BACKGROUND: Although triple-negative breast cancer (TNBC) accounts for only 15% of breast cancer cases, it is associated with a high relapse rate and poor outcome after standard treatment. Currently, the effective drugs and treatment strategies for TNBC remain limited, and thus, developing effective treatments for TNBC is pressing. Several studies have demonstrated that both chalcone and syringaldehyde have anticancer effect, but their potential anti-TNBC bioactivity are still unknown. PURPOSE: The present study aimed to synthesize a chalcone-syringaldehyde hybrid (CSH1) and explore its potential anti-TNBC effects and the underlying molecular mechanism. METHODS: Cell cytotoxicity was determined by 3-(4,5-dimethythiazol)-2,5-diphenyltetrazolium bromide (MTT). The activity of cell proliferation was measured by colony formation assay and 5-ethynyl-2'-deoxyuridine (EdU) staining assay. Cell cycle distribution and cell apoptosis were determined by fluorescence-activated cell sorter (FACS). The situation of DNA damage was observed using fluorescence microscopy. The ability of cell-matrix adhesion, migration and invasion was detected using cell adhesion assay and transwell assay. Transcriptome sequencing was performed to find out the changed genes. Levels of various signaling proteins were assessed by western blotting. RESULTS: CSH1 treatment triggered DNA damage and inhibited DNA replication, cell cycle arrest, and cell apoptosis via suppressing signal transducer and activator of transcription 3 (STAT3) phosphorylation. Whole genome RNA-seq analysis suggested that 4% of changed genes were correlated to DNA damage and repair, and nearly 18% of changed genes were functionally related to cell adhesion and migration. Experimental evidence indicated that CSH1 treatment significantly affected the distribution of focal adhesion kinase (FAK) and its phosphorylation, resulting in cell-matrix-adhesion reduction and migration inhibition of TNBC cells. Further mechanistic studies indicated that CSH1 inhibited TNBC cell proliferation, adhesion, and migration by inhibiting cytoskeleton-associated protein 2 (CKAP2)-mediated FAK and STAT3 phosphorylation signaling. CONCLUSION: These results suggest that CKAP2-mediated FAK and STAT3 phosphorylation signaling is a valuable target for TNBC treatment, and these findings also reveal the potential of CSH1 as a prospective TNBC drug.


Asunto(s)
Chalcona , Chalconas , Neoplasias de la Mama Triple Negativas , Apoptosis , Benzaldehídos , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Chalcona/farmacología , Chalcona/uso terapéutico , Chalconas/farmacología , Chalconas/uso terapéutico , Proteínas del Citoesqueleto , Citoesqueleto/metabolismo , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Recurrencia Local de Neoplasia/metabolismo , Fosforilación , Factor de Transcripción STAT3/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo
3.
Cell Prolif ; 54(9): e13101, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34296479

RESUMEN

OBJECTIVE: Osteosarcoma (OS) is characterized by high levels of the tumour-associated inflammatory microenvironment. Moreover, in approximately 60% of OS, telomere length is maintained by alternative lengthening of telomeres (ALT) pathway. Whether the ALT pathway can be exploited for OS therapeutic treatment and how the OS inflammatory microenvironment influences the anti-cancer drug effect remains unknown. Here, we examined the biological effects of TMPyP4 and cisplatin in the inflammatory microenvironment of OS cells. MATERIALS AND METHODS: Immunofluorescence in situ hybridization (IF-FISH) and C-circle experiments were used to detect the G-quadruplex and ALT activity. The redox potential of single guanine, G-quadruplex and G-quadruplex/TMPyP4 was evaluated by the lowest unoccupied molecular orbital energy (LUMO), zeta potential and cyclic voltammetry. Cell viability, flow cytometry and apoptosis, Western blot, comet assay, adhesion, transwell and scratch experiments were performed to compare the anti-tumour proliferation and migration effects of TMPyP4 and cisplatin in the inflammatory microenvironment. RESULTS: This study indicated that compared with cisplatin, TMPyP4 could induce the formation of human telomeres and FAK G-quadruplex in vitro and in vivo, and TMPyP4-treated OS cells showed fewer extrachromosomal C-circles and fewer ALT-associated promyelocytic leukaemia bodies. Consequently, the ALT activity and FAK-related cell migration were suppressed by TMPyP4. Mechanistically, the formation of G-quadruplex resulted in both lower redox potential than G within the genome and FAK transcription inhibition, and TMPyP4 could enhance this phenomenon, especially in the inflammatory microenvironment. CONCLUSIONS: Our results reveal that TMPyP4 is more suitable for OS treatment than cisplatin.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , G-Cuádruplex/efectos de los fármacos , Osteosarcoma/tratamiento farmacológico , Porfirinas/farmacología , Microambiente Tumoral/efectos de los fármacos , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Osteosarcoma/metabolismo , Telomerasa/metabolismo , Telómero/efectos de los fármacos
4.
Med Oncol ; 38(6): 59, 2021 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-33880669

RESUMEN

Cancer is one of the diseases that threatens human health and is a leading cause of mortality worldwide. High levels of reactive oxygen species (ROS) have been observed in cancer tissues compared with normal tissues in vivo, and it is not yet known how this influences chemotherapeutic drug action. Cationic porphyrin 5,10,15,20-tetra-(N-methyl-4-pyridyl) porphyrin (TMPyP4) is a photosensitizer used in photodynamic therapy (PDT) and a telomerase inhibitor used in the treatment of telomerase-positive cancer. Here, we investigated the anticancer activity of TMPyP4 in A549 and PANC cells cultured in H2O2. The results showed that compared to TMPyP4 alone, the combination of TMPyP4 and H2O2 exhibited sensitization effects on cell viability and colony formation inhibition and apoptosis in A549 and PANC cells, but had no effect in human normal MIHA cells. Mechanistically, the combination of TMPyP4 and H2O2 activates high ROS and mitochondrial membrane potential in A549 and PANC cells, resulting in intense DNA damage and DNA damage responses. Consequently, compared to TMPyP4 alone, TMPyP4 and H2O2 combined treatment upregulates the expression of BAX, cleaved caspase 3, and p-JNK and downregulates the expression of Bcl-2 in A549 and PANC cells. Taken together, these data suggested that H2O2 enhanced the anticancer activity of TMPyP4-mediated ROS-dependent DNA damage and related apoptotic protein regulation, revealing that the high ROS tumor microenvironment plays an important role in chemotherapeutic drug action.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Mitocondrias/efectos de los fármacos , Neoplasias Pancreáticas/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo , Células A549 , Línea Celular Tumoral , Ensayo Cometa , Daño del ADN , Hepatocitos/efectos de los fármacos , Humanos , Peróxido de Hidrógeno/administración & dosificación , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/genética , Mitocondrias/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Porfirinas/administración & dosificación , Microambiente Tumoral/efectos de los fármacos
5.
J Microencapsul ; 35(3): 272-280, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29671362

RESUMEN

Vitamin C (VC) and folic acid (FA) are the important nutrient and antioxidant in human body. In order to improve their stability, their co-loaded liposomes (VCFA-Lip) and chitosan-coated liposomes (CS-VCFA-Lip) are prepared and characterised. The mean particle size of VCFA-Lip and CS-VCFA-Lip is 138 nm and 249 nm, respectively. The encapsulation efficiencies of both drugs for CS-VCFA-Lip are much higher than those for VCFA-Lip. Furthermore, the experimental results show that the antioxidant activity of CS-VCFA-Lip is higher than that of VCFA-Lip. Moreover, the storage stability study reveals that the chitosan coating can efficiently improve the physical stability of VCFA-Lip. These results indicate that stability of VC and FA can be greatly improved after being wrapped by liposomes. In addition, the performance of CS-VCFA-Lip is better than VCFA-Lip, indicating CS-VCFA-Lip can be applied as a promising delivery system for the antioxidant defence system to the food industry and cosmetic industry.


Asunto(s)
Antioxidantes/administración & dosificación , Ácido Ascórbico/administración & dosificación , Quitosano/química , Ácido Fólico/administración & dosificación , Liposomas/química , Vitaminas/administración & dosificación , Antioxidantes/química , Antioxidantes/farmacología , Ácido Ascórbico/química , Ácido Ascórbico/farmacología , Compuestos de Bifenilo/química , Liberación de Fármacos , Estabilidad de Medicamentos , Ácido Fólico/química , Ácido Fólico/farmacología , Radicales Libres/química , Peróxido de Hidrógeno/química , Picratos/química , Vitaminas/química , Vitaminas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...