Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Drug Res (Stuttg) ; 74(3): 133-144, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38350632

RESUMEN

In this study, the protective efficacy of pentoxifylline (PTX) as a xanthine derivative against arsenic trioxide (ATO)-induced kidney and liver damage in mice was investigated. Thirty-six mice were divided into six groups, receiving intraperitoneal injections of saline, ATO, PTX, or a combination for four weeks. Blood samples were analyzed for serum biochemistry, while hepatic tissue underwent examination for histopathological changes and assessment of oxidative stress markers and antioxidant gene expression through Real-Time PCR. ATO exposure significantly increased serum markers (creatinine, ALT, BUN, ALP, AST) and induced histopathological changes in the liver. Moreover, it elevated renal and hepatic nitric oxide (NO) and lipid peroxidation (LPO) levels, and reduced antioxidant enzyme expression (CAT, GSR, GPx, MPO, SOD), total thiol groups (TTGs), and total antioxidant capacity (TAC). Conversely, PTX treatment effectively lowered serum hepatic and renal markers, improved antioxidant markers, and induced histopathological alterations. Notably, PTX did not significantly affect renal and hepatic NO levels. These findings suggest that PTX offers therapeutic potential in mitigating liver and acute kidney injuries induced by various insults, including exposure to ATO.


Asunto(s)
Alcaloides , Antioxidantes , Ratones , Animales , Antioxidantes/farmacología , Antioxidantes/metabolismo , Trióxido de Arsénico/metabolismo , Trióxido de Arsénico/farmacología , Hígado/metabolismo , Estrés Oxidativo , Alcaloides/farmacología , Xantinas/metabolismo , Xantinas/farmacología
2.
J Med Chem ; 66(24): 16579-16596, 2023 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-38069817

RESUMEN

Arsenic trioxide (ATO) targets PML/RARα and leads to miraculous success in treating acute promyelocytic leukemia. Notably, ATO also targets p53, the most frequently mutated protein in cancers, through a similar binding mechanism. However, p53-targeting ATO trials are challenging due to the poor cellular uptake and cancer selectivity of ATO. Here, we analyzed the structure-activity relationship of arsenicals and rationally developed a novel arsenical (designated AcGlcAs) by conjugating arsenic to sulfur atoms and tetraacetyl-ß-d-thioglucose. AcGlcAs exhibited remarkable cellular uptake through a thiol-mediated pathway (maximally 127-fold higher than ATO), thereby potently targeting PML/RARα and mutant p53. Among the 55 tested cell lines, AcGlcAs preferentially killed cancer lines rather than normal lines. In preclinical studies, AcGlcAs significantly extended the survival of mice bearing a xenograft tumor with p53 mutation while showing high plasma stability and oral bioavailability. Thus, AcGlcAs is a potential clinical candidate for precisely treating numerous p53-mutated cancers.


Asunto(s)
Antineoplásicos , Arsenicales , Leucemia Promielocítica Aguda , Humanos , Ratones , Animales , Proteína p53 Supresora de Tumor/metabolismo , Óxidos/farmacología , Óxidos/metabolismo , Apoptosis , Trióxido de Arsénico/farmacología , Trióxido de Arsénico/metabolismo , Trióxido de Arsénico/uso terapéutico , Arsenicales/farmacología , Arsenicales/uso terapéutico , Leucemia Promielocítica Aguda/tratamiento farmacológico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico
3.
Sci Transl Med ; 15(690): eabn9155, 2023 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-37018419

RESUMEN

Tumor suppressor p53 is inactivated by thousands of heterogeneous mutations in cancer, but their individual druggability remains largely elusive. Here, we evaluated 800 common p53 mutants for their rescue potencies by the representative generic rescue compound arsenic trioxide (ATO) in terms of transactivation activity, cell growth inhibition, and mouse tumor-suppressive activities. The rescue potencies were mainly determined by the solvent accessibility of the mutated residue, a key factor determining whether a mutation is a structural one, and the temperature sensitivity, the ability to reassemble the wild-type DNA binding surface at a low temperature, of the mutant protein. A total of 390 p53 mutants were rescued to varying degrees and thus were termed as type 1, type 2a, and type 2b mutations, depending on the degree to which they were rescued. The 33 type 1 mutations were rescued to amounts comparable to the wild type. In PDX mouse trials, ATO preferentially inhibited growth of tumors harboring type 1 and type 2a mutants. In an ATO clinical trial, we report the first-in-human mutant p53 reactivation in a patient harboring the type 1 V272M mutant. In 47 cell lines derived from 10 cancer types, ATO preferentially and effectively rescued type 1 and type 2a mutants, supporting the broad applicability of ATO in rescuing mutant p53. Our study provides the scientific and clinical communities with a resource of the druggabilities of numerous p53 mutations (www.rescuep53.net) and proposes a conceptual p53-targeting strategy based on individual mutant alleles rather than mutation type.


Asunto(s)
Neoplasias , Proteína p53 Supresora de Tumor , Humanos , Animales , Ratones , Trióxido de Arsénico/metabolismo , Trióxido de Arsénico/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis , Mutación , Neoplasias/genética
4.
Cell Death Differ ; 30(5): 1320-1333, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36894687

RESUMEN

Acute promyelocytic leukemia (APL) is driven by the oncoprotein PML-RARα, which recruits corepressor complexes, including histone deacetylases (HDACs), to suppress cell differentiation and promote APL initiation. All-trans retinoic acid (ATRA) combined with arsenic trioxide (ATO) or chemotherapy highly improves the prognosis of APL patients. However, refractoriness to ATRA and ATO may occur, which leads to relapsed disease in a group of patients. Here, we report that HDAC3 was highly expressed in the APL subtype of AML, and the protein level of HDAC3 was positively associated with PML-RARα. Mechanistically, we found that HDAC3 deacetylated PML-RARα at lysine 394, which reduced PIAS1-mediated PML-RARα SUMOylation and subsequent RNF4-induced ubiquitylation. HDAC3 inhibition promoted PML-RARα ubiquitylation and degradation and reduced the expression of PML-RARα in both wild-type and ATRA- or ATO-resistant APL cells. Furthermore, genetic or pharmacological inhibition of HDAC3 induced differentiation, apoptosis, and decreased cellular self-renewal of APL cells, including primary leukemia cells from patients with resistant APL. Using both cell line- and patient-derived xenograft models, we demonstrated that treatment with an HDAC3 inhibitor or combination of ATRA/ATO reduced APL progression. In conclusion, our study identifies the role of HDAC3 as a positive regulator of the PML-RARα oncoprotein by deacetylating PML-RARα and suggests that targeting HDAC3 could be a promising strategy to treat relapsed/refractory APL.


Asunto(s)
Antineoplásicos , Arsénico , Arsenicales , Leucemia Promielocítica Aguda , Humanos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Arsénico/metabolismo , Arsénico/farmacología , Arsénico/uso terapéutico , Trióxido de Arsénico/farmacología , Trióxido de Arsénico/metabolismo , Trióxido de Arsénico/uso terapéutico , Arsenicales/metabolismo , Arsenicales/farmacología , Arsenicales/uso terapéutico , Diferenciación Celular , Leucemia Promielocítica Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/metabolismo , Proteínas Nucleares/metabolismo , Óxidos/metabolismo , Óxidos/farmacología , Óxidos/uso terapéutico , Factores de Transcripción/metabolismo , Tretinoina/farmacología , Ubiquitinación
5.
Transl Res ; 255: 66-76, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36400307

RESUMEN

Gemcitabine (GEM) is the first-line medication for pancreatic ductal adenocarcinoma (PDAC). However, over some treatment cycles, GEM sensitivity declines and chemotherapeutic resistance develops, resulting in tumor recurrence and metastasis. Therefore, it is critical to elucidate the mechanism of GEM chemoresistance. And a specific drug that is closely related to the mechanism is urgently required to sensitize GEM. Here, tissue inhibitor of matrix metalloproteinases 1 (TIMP1) and phosphorylated mammalian target of rapamycin (p-mTOR) were found to be substantially elevated in PDAC patients and were associated with worse overall survival. The TIMP1/PI3K/AKT/mTOR pathway was found in GEM-resistant PDAC cells and was revealed to be involved in epithelial-mesenchymal transition (EMT) and apoptosis. Furthermore, arsenic trioxide (ATO), a basic therapeutic drug for acute promyelocytic leukemia, mediated TIMP1 reduction by inducing reactive oxygen species generation and hampered the subsequent PI3K/AKT/mTOR axis. Moreover, the combination of ATO and GEM cooperatively suppressed the TIMP1/PI3K/AKT/mTOR pathway, synergistically inhibited EMT and promoted apoptosis. In vitro and in vivo, ATO combined with GEM has a collaborative anticancer effect, inhibiting cancer cell proliferation, migration, invasion, and suppressing tumor growth both in PDAC parental and GEM-resistant cells. Overall, the TIMP1/PI3K/AKT/mTOR pathway is present in PDAC and linked to GEM resistance. ATO suppresses the axis to sensitize GEM and reverse GEM resistance, suggesting a promising treatment for the disease.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Gemcitabina , Trióxido de Arsénico/farmacología , Trióxido de Arsénico/metabolismo , Trióxido de Arsénico/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositol 3-Quinasas/farmacología , Fosfatidilinositol 3-Quinasas/uso terapéutico , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Regulación hacia Abajo , Resistencia a Antineoplásicos , Neoplasias Pancreáticas/patología , Serina-Treonina Quinasas TOR/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Proliferación Celular , Apoptosis , Línea Celular Tumoral , Neoplasias Pancreáticas
6.
Sheng Li Xue Bao ; 74(5): 763-772, 2022 Oct 25.
Artículo en Chino | MEDLINE | ID: mdl-36319099

RESUMEN

The present study was aimed to investigate the effects of circRNA-0028171 on the apoptosis of vascular endothelial cells induced by arsenic trioxide (As2O3). Human umbilical vein endothelial cells (HUVECs) were treated with 0-15 µmol/L As2O3 for 24 h. Then, cellular viability was measured by MTT assay. The expression levels of circRNA-0028171, Bcl-2 and Bax mRNA were detected by real-time quantitative PCR. Bcl-2/Bax protein ratio was detected by Western blot. Whether circRNA-0028171 was involved in the regulation of HUVECs by As2O3 was investigated by transfection with overexpression plasmid of circRNA-0028171 and siRNA. The results showed that compared with the control group, As2O3 group showed decreased cellular viability, reduced Bcl-2/Bax mRNA and protein ratios, and significantly lower expression of circRNA-0028171. Overexpression of circRNA-0028171 inhibited apoptosis of HUVECs induced by As2O3. Knockdown of circRNA-0028171 by siRNA promoted As2O3-induced apoptosis in HUVECs. These results suggest that circRNA-0028171 is involved in the vascular endothelial cell apoptosis induced by As2O3.


Asunto(s)
Apoptosis , ARN Circular , Humanos , Trióxido de Arsénico/metabolismo , Trióxido de Arsénico/farmacología , Proteína X Asociada a bcl-2/metabolismo , ARN Interferente Pequeño/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , ARN Mensajero/metabolismo
7.
Methods Mol Biol ; 2497: 173-184, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35771442

RESUMEN

Arsenic is either notorious toxicant or miracle cure for acute promyelocytic leukemia and several other diseases. It interacts with mitochondria directly or indirectly, by interacting with mitochondrial enzymes, such as respiratory chain complexes and tricarboxylic acid cycle proteins, or affecting mitochondrial homeostasis via ROS or mitochondrial outer membrane permeabilization. Given the ubiquitous presence of mitochondria and indispensable role in cellular metabolism, arsenical-mitochondrial interactions may manifest clinical importance by revealing mechanism of disease curation, preventing severe side effects, and foreseeing potential health issues. Here, we described the interaction between isolated mitochondria and arsenicals.


Asunto(s)
Arsenicales , Apoptosis , Trióxido de Arsénico/metabolismo , Trióxido de Arsénico/toxicidad , Potencial de la Membrana Mitocondrial , Mitocondrias/metabolismo , Óxidos , Especies Reactivas de Oxígeno/metabolismo
8.
Toxicology ; 475: 153237, 2022 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-35714947

RESUMEN

Long term low dose exposure of arsenic has been reported to lead various cells proliferation and malignant transformation. GLUT1, as the key transporter of glucose, has been reported to have association with rapid proliferation of various cells or tumor cells. In our study, we found that low dose exposure to arsenic trioxide (0.1µmol/L As2O3) could induce an increase in glucose uptake and promote cell viability and DNA synthesis. And, 2-DG, a non-metabolized glucose analog, significantly decreased the glucose uptake and cell proliferation of 0.1µmol/L As2O3 treated L-02 cells. However, 4 mmol/L 2-DG was co-utilized with equal dose glucose had no significant effect on the cell proliferation of 0.1µmol/L As2O3 treated L-02 cells. Further studies showed that exposure to 0.1µmol/L As2O3 could promote the expression of GLUT1 on plasma membrane. Inhibition of GLUT1 expression by 5µmol/L BAY-876 significantly decreased the abilities of glucose uptake and cell proliferation in As2O3-treated L-02 cells. Moreover, 0.1µmol/L As2O3 induced the AKT activation indicated by increased the phospho-AKT (Ser473 and Thr308). Knockdown AKT by shRNA or inhibited AKT activation by LY294002 was followed by significantly decreased glucose uptake, GLUT1 plasma membrane expression and cell proliferation in As2O3-treated L-02 cells. All in all, these results demonstrated that arsenic trioxide-induced AKT activation contributed to the cells proliferation through upregulating expression of GLUT1 on plasma membrane that enhanced glucose uptake.


Asunto(s)
Antineoplásicos , Arsenicales , Antineoplásicos/farmacología , Apoptosis , Trióxido de Arsénico/metabolismo , Trióxido de Arsénico/toxicidad , Arsenicales/farmacología , Línea Celular Tumoral , Proliferación Celular , Glucosa , Transportador de Glucosa de Tipo 1/genética , Óxidos/metabolismo , Óxidos/toxicidad , Proteínas Proto-Oncogénicas c-akt/metabolismo
9.
Adv Clin Exp Med ; 31(8): 903-911, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35467087

RESUMEN

BACKGROUND: Neuroblastoma (NB) is one of the most common extracranial tumors with limited therapeutic options. Retinoic acid (RA) has been identified to play anticancer role against NB cells by inducing the differentiation and apoptosis of immature neuroblasts. However, silencing HoxC9 promoter by EZH2-induced H3K27me3 hypermethylation can lead to RA resistance. Previous studies have suggested that arsenic trioxide (ATO), an inhibitor of DNA methylation, could downregulate the expression of EZH2 in breast cancer cells. OBJECTIVES: In our study, we attempted to obtain some insight into the mechanisms of differentiation of RA-resistant NB cells by detecting the expressions of HoxC9 and EZH2 in NB cells treated with ATO, so as to provide a basis for the subsequent treatment of RA-resistant NB by ATO. MATERIAL AND METHODS: Two NB cell lines, SK-N-AS (retinoic acid-resistant neuroblastoma cells) and SK-N-SH (retinoic acid-sensitive neuroblastoma cells), were used in our experiments. Cell proliferation and apoptosis were respectively determined with Cell Counting Kit-8 (CCK-8) assay kit and Annexin V staining. The inverted phase contrast microscope was used to observe cell growth and measure the total length of nerve synapses. We employed label-free quantitative proteomic analysis to profile ATO-dependent changes in the proteome of NB cells. Western blot was used to detect the expressions of HoxC9, HoxD8 and EZH2. RESULTS: Arsenic trioxide inhibited the cell proliferation and increased apoptosis and total length of synapses in two NB cell lines. The expressions of HoxC9 and HoxD8 were upregulated, while the expression of EZH2 was downregulated in the SK-N-AS cell line. No significant changes in the 3 proteins mentioned above were observed in the SK-N-SH cell line after ATO treatment. CONCLUSIONS: Arsenic trioxide may reactivate the expression of HoxC9 by downregulating EZH2, which leads to restoring RA sensitivity and promoting the differentiation and apoptosis of RA-resistant NB cells.


Asunto(s)
Neuroblastoma , Tretinoina , Apoptosis , Trióxido de Arsénico/metabolismo , Trióxido de Arsénico/farmacología , Trióxido de Arsénico/uso terapéutico , Diferenciación Celular , Línea Celular Tumoral , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/uso terapéutico , Humanos , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/genética , Proteómica , Tretinoina/farmacología , Regulación hacia Arriba
10.
Tissue Cell ; 76: 101805, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35487055

RESUMEN

BACKGROUND: Arsenic trioxide (As2O3) as an inorganic compound is used to treat various cancers and other diseases. It has been reported that arsenic trioxide induced cellular apoptosis in certain kinds of cancers, including prostate cancers. The present study aimed to elucidate the crucial cooperative role of arsenic trioxide and Curcumin and their ability to protect against prostate cancers by targeting the epithelial-to-mesenchymal transition and expression of apoptosis-related genes. MATERIAL AND METHODS: The human prostate cell lines (LNCaP and PC3) were treated with different concentrations of Curcumin and As2O3 alone and combined to find effective doses and IC50 values. Percentages of apoptotic cells were evaluated by Annexin/P.I. staining, the proliferative inhibitory effect was assessed by Micro Culture Tetrazolium Test (MTT), and mRNA levels of KLK2, E-cadherin, SNAIL, angiogenesis genes (VEGFA and VEGFC), and apoptosis genes (BAX, Bcl2, and P53) expression were investigated by the real-time PCR method. ANOVA and t-test were used to appraise the results. RESULTS: For the first time, we presented that the combination therapy of Curcumin and As2O3 increases prostate cancer cell apoptosis and inhibits proliferation; Our data displayed that Curcumin (15 µM and 10 µM in PC3 and LNCap), As2O3 (8 µM and 5 µM in PC3 and LNCap), and also their combination (15 µM Curcumin and 8 µM As2O3 in PC3, 10 µM Curcumin and 5 µM As2O3 in LNCap cell lines) significantly increased the percentage of apoptotic cells and inhibited cell growth (P < 0.05) compared with each drug alone. Generally, both cell lines treated with the combination of Curcumin and As2O3 displayed decreased angiogenesis genes (VEGFA and VEGFC), apoptosis genes (BAX and Bcl2), and prostate cancer marker (KLK2), the zinc-finger protein (SNAIL); and an increase in expression (P < 0.05) of cell-cell adhesion molecule (E-cadherin) and tumor suppressor gene (P53) genes. CONCLUSIONS: The antitumor effects of combination therapy with As2O3 and Curcumin have been displayed on prostate cancer cell lines (LNCaP and PC3), which probably originates from their potential to induce apoptosis and inhibit the growth of prostate cancer cells simultaneously.


Asunto(s)
Antineoplásicos , Curcumina , Neoplasias de la Próstata , Antineoplásicos/farmacología , Apoptosis , Trióxido de Arsénico/metabolismo , Trióxido de Arsénico/farmacología , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular Tumoral , Curcumina/farmacología , Curcumina/uso terapéutico , Humanos , Masculino , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Proteína p53 Supresora de Tumor , Proteína X Asociada a bcl-2/metabolismo
11.
Biomed Res Int ; 2022: 9426623, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36619305

RESUMEN

Background: Kirsten rat sarcoma (KRAS) protein is an essential contributor to the development of pancreatic ductal adenocarcinoma (PDAC). KRAS G12D and G12V mutant tumours are significant challenges in cancer therapy due to high resistance to the treatment. Objective: To determine how effective is the ATO/D-VC combination in suppression of PDAC the mouse transgenic model. This study investigated the antitumour effect of a novel combination of arsenic trioxide (ATO) and D-ascorbic acid isomer (D-VC). Such a combination can be used to treat KRAS mutant cancer by inducing catastrophic oxidative stress. Methods: In this study, we examined the effectiveness of ATO and D-VC on xenograft models-AK192 cells transplanted into mice. Previously, it has been shown that a high concentration of Vitamin C (VC) selectively can kill the cells expressing KRAS. Results: The results of this study demonstrated that the combination of VC with a low dose of the oxidizing drug ATO led to the enhancement of the therapeutic effect. These findings suggest that the combined treatment using ATO and D-VC is a promising approach to overcome the limitation of drug selectivity and efficacy.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Ratones , Animales , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Carcinoma Ductal Pancreático/patología , Trióxido de Arsénico/metabolismo , Modelos Animales de Enfermedad , Estrés Oxidativo , Ácido Ascórbico/farmacología , Ácido Ascórbico/uso terapéutico , Combinación de Medicamentos , Oxidación-Reducción , Línea Celular Tumoral , Neoplasias Pancreáticas
12.
Toxicol Ind Health ; 37(12): 727-736, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34730462

RESUMEN

Epidemiological evidence suggests that the metabolic profiles of each individual exposed to arsenic (As) are related to the risk of cancer, coronary heart disease, and diabetes. The arsenite methyltransferase (AS3MT) gene plays a key role in As metabolism. Several single nucleotide polymorphisms in the AS3MT gene may affect both enzyme activity and gene transcription. AS3MT polymorphisms are associated with the proportions of monomethylarsenic acid (MMA) and dimethylarsenic acid (DMA) in urine as well as the incidence of cancer. P21 protein is a cyclin-dependent kinase inhibitor. Mutations of the P21 gene have been found in cancer patients. In our study, we investigate whether polymorphisms of the AS3MT gene alter As methylation capacity and adversely affect the P21 gene in arsenic trioxide plant workers. The DNA damage was examined by the quantitative polymerase chain reaction. Restriction fragment length polymorphism was used to analyze the genotype of the AS3MT gene. The results showed that DNA damage in P21 gene fragments was greater in those individuals exposed to high levels of As. There was a strong positive correlation between the DNA damage to P21 gene fragments and the percentage of MMA in urine. However, DNA damage in P21 gene fragments was negatively associated with the percentage of DMA in urine (%uDMA), primary methylation index (PMI), and secondary methylation index. We found that subjects with the rs7085104 GG or GA allele were associated with higher %uDMA and PMI and less DNA damage. The subjects with the rs11191454 GG+GA or GA allele were also associated with higher %uDMA and PMI and less DNA damage. Our results suggest that rs1191454 and rs7085104 in the AS3MT gene affect the As-induced DNA damage by altering individual metabolic efficiency.


Asunto(s)
Trióxido de Arsénico/efectos adversos , Trióxido de Arsénico/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Metiltransferasas/genética , Exposición Profesional/efectos adversos , Polimorfismo de Nucleótido Simple/efectos de los fármacos , Adulto , Alelos , Arsénico , China , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Femenino , Humanos , Masculino , Metiltransferasas/orina , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa
13.
Chemotherapy ; 66(4): 139-155, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34344008

RESUMEN

BACKGROUND: The aim of this study was to understand physical characteristics of Embosphere microspheres for the clinical use of microsphere chemotherapy embolization of liver cancer. METHODS: The morphology of Embosphere microspheres in different states, including static, oscillating, and in a magnetic field was observed with the naked eye. Ninety-five patients diagnosed with primary hepatocellular carcinoma (HCC) were separated into 3 groups based on the types of embolic material as follows: 32 cases of sole microspheres, 34 cases of iodinated oil (17 cases with additional application of gelatin sponge particle), and 29 cases of iodinated oil + Embosphere microspheres. RESULTS: The diameter of the microspheres ranged from 100 to 300 µm, with a sedimentation rate υ = 0.0375 cm/s in physiological saline. The diameter of microspheres ranged from 300 to 500 µm, with a sedimentation rate υ = 0.1875 cm/s. The swelling rate of microspheres was 90%. Microspheres showed nondirectional movement in a 1.5- or 3.0-T magnetic field during magnetic resonance imaging. A volumetric ratio of 1:1.4-1:1.5 between microspheres and contrast agent resulted in optimal suspension properties. Microspheres appeared circular with a smooth surface upon water adsorption. Microsphere embolism was observable in blood vessels of pathological sections. The surface of microspheres can adsorb 5-fluorouracil and arsenic trioxide. There are statistically significant differences in local-regional tumor control conditions among patients treated with sole microspheres, iodinated oil, and iodinated oil + microspheres during transarterial chemoembolization. CONCLUSIONS: Embosphere microspheres can be used to embolize patients with rupture and hemorrhage of HCC. Embosphere microsphere embolization is superior to iodinated oil and iodinated oil + microsphere for HCC.


Asunto(s)
Portadores de Fármacos/química , Microesferas , Angiografía , Trióxido de Arsénico/química , Trióxido de Arsénico/metabolismo , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/terapia , Quimioembolización Terapéutica , Fluorouracilo/química , Fluorouracilo/metabolismo , Fluorouracilo/uso terapéutico , Gelatina/química , Arteria Hepática/diagnóstico por imagen , Humanos , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/terapia , Aceites/química , Tamaño de la Partícula
14.
FEBS Open Bio ; 11(7): 2019-2032, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34058077

RESUMEN

HnRNP K protein is a heterogeneous nuclear ribonucleoprotein which has been proposed to be involved in the leukemogenesis of acute promyelocytic leukemia (APL), as well as in differentiation induced by all-trans retinoic acid (ATRA). We previously demonstrated a connection between SET and hnRNP K function in head and neck squamous cell carcinoma (HNSCC) cells related to splicing processing. The objective of this study was to characterize the participation of hnRNP K and SET proteins in ATRA-induced differentiation in APL. We observed higher (5- to 40-fold) levels of hnRNP K and SET mRNA in APL patients at the diagnosis phase compared with induction and maintenance phases. hnRNP K knockdown using short-hairpin RNA led to cell death in ATRA-sensitive NB4 and resistant NB4-R2 cells by apoptosis with SET cleavage. In addition, hnRNP K knockdown increased granulocytic differentiation in APL cells, mainly in NB4-R2 with ATRA. hnRNP K knockdown had an effect similar to that of treatment with U0126 (an meiosis-specific serine/threonine protein kinase/ERK inhibitor), mainly in NB4-R2 cells. SET knockdown in APL cells revealed that apoptosis induction in cells with hnRNP K knockdown occurred by SET cleavage rather than by reduction in SET protein. Transplantation of NB4-R2 cells into nude mice confirmed that arsenic trioxide (ATO) combined with U0126 has higher potential against tumor progression when compared to ATO. Therefore, hnRNP K/SET and ERK are potential therapeutic targets for both antineoplastic leukemia therapy and relapsed APL patients with ATRA resistance.


Asunto(s)
Leucemia Promielocítica Aguda , Animales , Trióxido de Arsénico/metabolismo , Trióxido de Arsénico/uso terapéutico , Ribonucleoproteína Heterogénea-Nuclear Grupo K/genética , Humanos , Leucemia Promielocítica Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/metabolismo , Ratones , Ratones Desnudos , Tretinoina/metabolismo , Tretinoina/farmacología
15.
Sci Rep ; 11(1): 3761, 2021 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-33580144

RESUMEN

Arsenic is reportedly a biphasic inorganic compound for its toxicity and anticancer effects in humans. Recent studies have shown that certain arsenic compounds including arsenic hexoxide (AS4O6; hereafter, AS6) induce programmed cell death and cell cycle arrest in human cancer cells and murine cancer models. However, the mechanisms by which AS6 suppresses cancer cells are incompletely understood. In this study, we report the mechanisms of AS6 through transcriptome analyses. In particular, the cytotoxicity and global gene expression regulation by AS6 were compared in human normal and cancer breast epithelial cells. Using RNA-sequencing and bioinformatics analyses, differentially expressed genes in significantly affected biological pathways in these cell types were validated by real-time quantitative polymerase chain reaction and immunoblotting assays. Our data show markedly differential effects of AS6 on cytotoxicity and gene expression in human mammary epithelial normal cells (HUMEC) and Michigan Cancer Foundation 7 (MCF7), a human mammary epithelial cancer cell line. AS6 selectively arrests cell growth and induces cell death in MCF7 cells without affecting the growth of HUMEC in a dose-dependent manner. AS6 alters the transcription of a large number of genes in MCF7 cells, but much fewer genes in HUMEC. Importantly, we found that the cell proliferation, cell cycle, and DNA repair pathways are significantly suppressed whereas cellular stress response and apoptotic pathways increase in AS6-treated MCF7 cells. Together, we provide the first evidence of differential effects of AS6 on normal and cancerous breast epithelial cells, suggesting that AS6 at moderate concentrations induces cell cycle arrest and apoptosis through modulating genome-wide gene expression, leading to compromised DNA repair and increased genome instability selectively in human breast cancer cells.


Asunto(s)
Trióxido de Arsénico/toxicidad , Células MCF-7/efectos de los fármacos , Glándulas Mamarias Humanas/efectos de los fármacos , Apoptosis/efectos de los fármacos , Arsénico/metabolismo , Trióxido de Arsénico/metabolismo , Trióxido de Arsénico/farmacología , Arsenicales/farmacología , Neoplasias de la Mama/genética , Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Glándulas Mamarias Humanas/metabolismo , Cultivo Primario de Células , Células Tumorales Cultivadas
16.
Ecotoxicol Environ Saf ; 208: 111752, 2021 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-33396077

RESUMEN

Arsenic is a toxic heavy metal vastly dispersed all over the earth crust. It manifests several major adverse health issues to millions of arsenic exposed populations. Arsenic is associated with different types of cancer, cardiovascular disorders, diabetes, hypertension and many other diseases. On the contrary, arsenic (arsenic trioxide, As2O3) is used as a chemotherapeutic agent in the treatment of acute promyelocytic leukemia. Balance between arsenic induced cellular proliferations and apoptosis finally decide the outcome of its transformation rate. Arsenic propagates signals via cellular and nuclear pathways depending upon the chemical nature, and metabolic-fates of the arsenical compounds. Arsenic toxicity is propagated via ROS induced stress to DNA-repair mechanism and mitochondrial stability in the cell. ROS induced alteration in p53 regulation and some mitogen/ oncogenic functions determine the transformation outcome influencing cyclin-cdk complexes. Growth factor regulator proteins such as c-Jun, c-fos and c-myc are influenced by chronic arsenic exposure. In this review we have delineated arsenic induced ROS regulations of epidermal growth factor receptor (EGFR), NF-ĸß, MAP kinase, matrix-metalloproteinases (MMPs). The role of these signaling molecules has been discussed in relation to cellular apoptosis, cellular proliferation and neoplastic transformation. The arsenic stimulated pathways which help in proliferation and neoplastic transformation ultimately resulted in cancer manifestation whereas apoptotic pathways inhibited carcinogenesis. Therapeutic strategies against arsenic should be designed taking into account all these factors.


Asunto(s)
Arsénico/fisiología , Proliferación Celular/fisiología , Plásticos/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Arsénico/metabolismo , Trióxido de Arsénico/metabolismo , Trióxido de Arsénico/farmacología , Arsenicales/metabolismo , Proliferación Celular/efectos de los fármacos , Humanos , Mitocondrias/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neoplasias , Óxidos/toxicidad , Transducción de Señal/efectos de los fármacos
17.
Mol Med Rep ; 22(6): 4645-4654, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33174611

RESUMEN

All­trans retinoic acid (ATRA) and arsenic trioxide (As2O3) are currently first­line treatments for acute promyelocytic leukemia (APL). However, a number of patients with APL are resistant to ATRA but still sensitive to As2O3, and the underlying mechanisms of this remain unclear. In the present study, two­dimensional gel electrophoresis, mass spectrometry and other proteomic methods were applied to screen and identify the differentially expressed proteins between the retinoic acid­sensitive cell lines and drug­resistant cell lines. The results demonstrated that in retinoic acid­resistant NB4­R1 cells, the protein expression of cofilin­1 was markedly increased compared with that in the drug­sensitive NB4 cells. Subsequently, the effects of cofilin­1 on As2O3­induced apoptosis in NB4­R1 cells were further investigated. The results revealed that cell viability was markedly suppressed and apoptosis was increased in the As2O3­treated NB4­R1 cells, with increased expression levels of cleaved­poly (ADP­ribose) polymerase and cleaved­caspase 12. Cofilin­1 expression was significantly decreased at both the mRNA and protein levels in the As2O3­treated group compared with the control. Western blotting further revealed that As2O3 treatment decreased the cytoplasmic cofilin­1 level but increased its expression in the mitochondrion. However, the opposite effects of As2O3 on the cytochrome C distribution were found in NB4­R1 cells. This suggested that As2O3 can induce the transfer of cofilin­1 from the cytoplasm to mitochondria and trigger the release of mitochondrial cytochrome C in NB4­R1 cells. Moreover, cofilin­1 knockdown by its specific short hairpin RNA significantly suppressed As2O3­induced NB4­R1 cell apoptosis and inhibited the release of mitochondrial cytochrome C. Whereas, overexpression of cofilin­1 using a plasmid vector carrying cofilin­1 increased the release of cytochrome C into the cytoplasm from the mitochondria in As2O3­treated NB4­R1 cells. In conclusion, cofilin­1 played a role in As2O3­induced NB4­R1 cell apoptosis and it might be a novel target for APL treatment.


Asunto(s)
Cofilina 1/metabolismo , Resistencia a Antineoplásicos/genética , Leucemia Promielocítica Aguda/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Trióxido de Arsénico/metabolismo , Trióxido de Arsénico/uso terapéutico , Muerte Celular/efectos de los fármacos , Cofilina 1/fisiología , Resistencia a Antineoplásicos/fisiología , Humanos , Mitocondrias/metabolismo , Óxidos/farmacología , Proteómica/métodos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptores de Ácido Retinoico/metabolismo , Tretinoina/farmacología , Células Tumorales Cultivadas
18.
Arch Toxicol ; 94(8): 2587-2601, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32435915

RESUMEN

Arsenic is a well-known environmental carcinogen and chronic exposure to arsenic through drinking water has been reported to cause skin, bladder and lung cancers, with arsenic metabolites being implicated in the pathogenesis. In contrast, arsenic trioxide (As2O3) is an effective therapeutic agent for the treatment of acute promyelocytic leukemia, in which the binding of arsenite (iAsIII) to promyelocytic leukemia (PML) protein is the proposed initial step. These findings on the two-edged sword characteristics of arsenic suggest that after entry into cells, arsenic reaches the nucleus and triggers various nuclear events. Arsenic is reduced, conjugated with glutathione, and methylated in the cytosol. These biotransformations, including the production of reactive metabolic intermediates, appear to determine the intracellular dynamics, target organs, and biological functions of arsenic.


Asunto(s)
Antineoplásicos/farmacología , Intoxicación por Arsénico/etiología , Trióxido de Arsénico/farmacología , Arsenicales/farmacología , Leucemia Promielocítica Aguda/tratamiento farmacológico , Animales , Antineoplásicos/metabolismo , Intoxicación por Arsénico/metabolismo , Trióxido de Arsénico/metabolismo , Arsenicales/metabolismo , Biotransformación , Humanos , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patología , Medición de Riesgo , Pruebas de Toxicidad
19.
J Trace Elem Med Biol ; 55: 82-88, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31345371

RESUMEN

BACKGROUND: Exposure to the environmental pollutants poses a serious threat to aquatic organism. The arsenic exposure in fish increases the risk of developing serious alterations from embryo to adult. OBJECTIVES: The present investigation was done to study the toxic effects of heavy metal arsenic [As(III)] on medaka (Oryzias latipes). Morphological alterations, apoptosis, nuclear abnormalities, and genotoxic biomarkers in erythrocytes were used to determine the stress caused by arsenic (As) exposure. METHODS: Medaka was exposed to As for 15 days at two toxic sublethal concentrations (7 ppm and 10 ppm) in combination with Spirulina platensis (SP) treatment as antioxidant algae at 200 mg/L. RESULTS: Results were consistent with a previous study results on tilapia. Exposure of medaka to As resulted in a dose-dependent increase in most the biomarkers used in the current study. Fish exposed to10 ppm As showed highest level of DNA damage. For the first time to our knowledge, using SP to counter the As toxicity in medaka, DNA damage restored to control levels. CONCLUSION: Accordingly, those results suggests that SP can protect medaka in aquaculture against As-induced damage by its ability as reactive oxygen species (ROS) reducer, antioxidant role, and DNA damage scavenger.


Asunto(s)
Trióxido de Arsénico/farmacología , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Oryzias/sangre , Spirulina/metabolismo , Animales , Trióxido de Arsénico/administración & dosificación , Trióxido de Arsénico/metabolismo , Biodegradación Ambiental
20.
Biomed Res Int ; 2019: 3924581, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31355259

RESUMEN

This study investigated the effects of proanthocyanidins (PC) on arsenic methylation metabolism and efflux in human hepatocytes (L-02), as well as the relationships between PC and GSH, MRP1 and other molecules. Cells were randomly divided into blank control group, arsenic trioxide exposure group (ATO, As2O3, 25µmol/L), and PC-treated arsenic exposure group (10, 25, 50mg/L). After 24/48h, the contents of different forms of arsenic were determined, and the methylation indexes were calculated. Intracellular S-adenosyl methionine (SAM), arsenic (+3 oxidation state) methyltransferase (AS3MT), multidrug resistance-associated protein 1 (MRP1), and reduced glutathione (GSH) were ascertained. Changing trends were observed and the correlation between arsenic metabolism and efflux related factors and arsenic metabolites was analyzed. We observed that cells showed increased levels of content/constituent ratio of methyl arsenic, primary/secondary methylation index, methylation growth efficiency/rate, and the difference of methyl arsenic content in cells and culture medium (P<0.05, resp.). Compared with ATO exposure group, the intracellular SAM content in PC-treated group decreased, and the contents of GSH, AS3MT, and MRP1 increased (P<0.05, resp.). There was a positive correlation between the content of intracellular GSH/AS3MT and methyl arsenic. The content of MRP1 was positively correlated with the difference of methyl arsenic content in cell and culture medium; conversely, the SAM content was negatively correlated with intracellular methyl arsenic content (P<0.05, resp.). Taken together, these results prove that PC can promote arsenic methylation metabolism and efflux in L-02 cells, which may be related to the upregulation of GSH, MRP1, and AS3MT levels by PC.


Asunto(s)
Arsénico/metabolismo , Hepatocitos/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , Proantocianidinas/farmacología , Arsénico/química , Trióxido de Arsénico/química , Trióxido de Arsénico/metabolismo , Transporte Biológico , Regulación de la Expresión Génica/efectos de los fármacos , Glutatión/genética , Humanos , Metilación/efectos de los fármacos , Metiltransferasas/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...