Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Sci Rep ; 14(1): 18974, 2024 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-39152142

RESUMEN

The deubiquitinase tripartite motif containing 44 (TRIM44) plays a critical role in linking the proteotoxic stress response with autophagic degradation, which is significant in the context of cancer and neurological diseases. Although TRIM44 is recognized as a prognostic marker in various cancers, the complex molecular mechanisms through which it facilitates autophagic degradation, particularly under oxidative stress conditions, have not been fully explored. In this study, we demonstrate that TRIM44 significantly enhances autophagy in response to oxidative stress, reducing cytotoxicity in cancer cells treated with arsenic trioxide. Our research emphasizes the critical role of the posttranslational modification of sequestosome-1 (SQSTM1) and its importance in improving sequestration during autophagic degradation under oxidative stress. We found that TRIM44 notably promotes SQSTM1 oligomerization in both PB1 domain-dependent and oxidation-dependent manners. Furthermore, TRIM44 amplifies the interaction between protein kinase A and oligomerized SQSTM1, leading to enhanced phosphorylation of SQSTM1 at S349. This phosphorylation event activates NFE2L2, a key transcription factor in the oxidative stress response, highlighting the importance of TRIM44 in modulating SQSTM1-mediated autophagy. Our findings support that TRIM44 plays pivotal roles in regulating autophagic sensitivity to oxidative stress, with implications for cancer, aging, aging-associated diseases, and neurodegenerative disorders.


Asunto(s)
Autofagia , Factor 2 Relacionado con NF-E2 , Estrés Oxidativo , Proteína Sequestosoma-1 , Proteínas de Motivos Tripartitos , Proteína Sequestosoma-1/metabolismo , Humanos , Proteínas de Motivos Tripartitos/metabolismo , Proteínas de Motivos Tripartitos/genética , Factor 2 Relacionado con NF-E2/metabolismo , Fosforilación , Trióxido de Arsénico/farmacología , Multimerización de Proteína , Línea Celular Tumoral , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células HEK293
2.
J Coll Physicians Surg Pak ; 34(8): 974-980, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39113519

RESUMEN

OBJECTIVE: To describe the prognostic variables and course of paediatric acute promyelocytic leukaemia (APL) in Pakistan. STUDY DESIGN: Cohort study. Place and Duration of the Study: Department of Paediatric Oncology, Combined Military Hospital, Rawalpindi, Pakistan, from January 2012 to December 2022. METHODOLOGY: Patients aged 1-15 years, clinically confirmed APL with promyelocytic leukaemia- retinoic acid receptor alpha (PML-RARA) were enrolled. Initial admission included a thorough examination, recording demographic and clinical data, reporting time, prior treatment, and socioeconomic status. Statistical analysis used SPSS 25.0, with significance at p <0.05. RESULTS: This study included 50 cases of APL. Out of which, 32 (64%) were males and 18 (34%) were females. The mean age at diagnosis was 7.02 ± 3.86 years. Pallor (96%) and fever (88%) were common presentations. The average white blood cell count was 28.70 ± 35.39 x109/L. Treatment protocols include 48% International Consortium for Childhood (ICC)-APL, and 52% arsenic trioxide (ATO). High-risk cases were 54%. Neutropenic fever and differentiation syndrome were common induction complications. Delays over one month increased induction deaths (6.7 to 35%, p = 0.011), reducing disease-free survival (DFS), (76.7 to 35%, p = 0.001), and overall survival (OS), (80 to 45%, p = 0.007). After 40.90 ± 45.19 months' follow-up, 10-year OS and DFS were 66.0% and 60.0%, respectively. The best OS and DFS, at 80%, were observed in standard-risk cases treated with ATO. CONCLUSION: Neutropenic fever and bleeding were the primary causes of mortality in paediatric APL induction. Treatment delay was a key prognostic factor. ATO-based therapy offered safer, improved DFS, and OS suitable for primary healthcare settings. KEY WORDS: Acute promyelocytic leukaemia, Chemotherapy, Neutropenic fever.


Asunto(s)
Leucemia Promielocítica Aguda , Humanos , Leucemia Promielocítica Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/mortalidad , Masculino , Femenino , Pakistán/epidemiología , Niño , Preescolar , Adolescente , Lactante , Pronóstico , Estudios de Cohortes , Trióxido de Arsénico/uso terapéutico , Resultado del Tratamiento , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico
3.
Proc Natl Acad Sci U S A ; 121(29): e2313851121, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-38976734

RESUMEN

Mass spectrometry-based omics technologies are increasingly used in perturbation studies to map drug effects to biological pathways by identifying significant molecular events. Significance is influenced by fold change and variation of each molecular parameter, but also by multiple testing corrections. While the fold change is largely determined by the biological system, the variation is determined by experimental workflows. Here, it is shown that memory effects of prior subculture can influence the variation of perturbation profiles using the two colon carcinoma cell lines SW480 and HCT116. These memory effects are largely driven by differences in growth states that persist into the perturbation experiment. In SW480 cells, memory effects combined with moderate treatment effects amplify the variation in multiple omics levels, including eicosadomics, proteomics, and phosphoproteomics. With stronger treatment effects, the memory effect was less pronounced, as demonstrated in HCT116 cells. Subculture homogeneity was controlled by real-time monitoring of cell growth. Controlled homogeneous subculture resulted in a perturbation network of 321 causal conjectures based on combined proteomic and phosphoproteomic data, compared to only 58 causal conjectures without controlling subculture homogeneity in SW480 cells. Some cellular responses and regulatory events were identified that extend the mode of action of arsenic trioxide (ATO) only when accounting for these memory effects. Controlled prior subculture led to the finding of a synergistic combination treatment of ATO with the thioredoxin reductase 1 inhibitor auranofin, which may prove useful in the management of NRF2-mediated resistance mechanisms.


Asunto(s)
Proteómica , Humanos , Proteómica/métodos , Línea Celular Tumoral , Células HCT116 , Técnicas de Cultivo de Célula/métodos , Neoplasias del Colon/metabolismo , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Trióxido de Arsénico/farmacología , Auranofina/farmacología , Proliferación Celular/efectos de los fármacos , Espectrometría de Masas/métodos
4.
Clin Toxicol (Phila) ; 62(7): 468-471, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38966917

RESUMEN

INTRODUCTION: From at least the fifteenth to late nineteenth centuries, peasants in the Austrian province of Styria ate up to several hundred milligrams of arsenic trioxide or sulfide daily or weekly for periods up to a number of years. Taking these doses of arsenic was believed to increase muscular power and enhance the beauty and sexual attractiveness of peasant girls. There do not appear to be contemporaneous records of the known consequences of chronic arsenic exposure. The historical records of arsenic eating there are reviewed and appear to be valid. The benefits are subjective judgements by arsenic eaters. The lack of objective reports of the anticipated external and internal clinical and pathological effects of arsenic poisoning depends on a smaller number of clinical accounts and autopsy reports and the general medical literature of those times, so it is weaker, but it is consistent. CAN THE CLAIMED BENEFITS OF ARSENIC EATING AND THE APPARENT ABSENCE OF HARMFUL TOXIC EFFECTS BE TRUE?: Why the arsenic eaters did not show the well-known consequences of prolonged exposure to high doses of arsenic is not known. Possible explanations include increases in detoxifying metabolism in the consumers due to induced genomic changes and selection in people and in the gut microbiome, as shown in other populations. Whether these effects would suffice to protect people against their high doses of arsenic has not been explored. CONCLUSION: Although the nature and mechanisms of arsenic toxicity have been extensively described, much still remains to be discovered.


Asunto(s)
Intoxicación por Arsénico , Humanos , Historia del Siglo XIX , Historia del Siglo XVII , Historia del Siglo XVIII , Historia del Siglo XV , Historia del Siglo XVI , Arsénico/toxicidad , Femenino , Contaminación de Alimentos , Historia del Siglo XX , Trióxido de Arsénico , Arsenicales/efectos adversos
5.
Phytomedicine ; 132: 155891, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39059093

RESUMEN

BACKGROUD: Arsenic trioxide (ATO), the first-line drug in treating acute premyelogenous leukemia, has the profound side effect of inducing endothelial mesenchymal transition (EndMT) and causing cardiac fibrosis. Diosgenin (DIO), a pharmaceutical compound found in Paris polyphylla, exhibits promising potential in safeguarding cardiovascular health by mitigating EndMT. PURPOSE: This study aims to explore the role and mechanism of DIO in ATO-induced myocardial fibrosis to provide a novel therapeutic agent for ATO-induced cardiac fibrosis. METHODS: Wistar rats were given DIO by gavage and ATO by tail vein. Cardiac function and fibrosis were evaluated by echocardiography and Masson's trichrome staining in rats. Human aortic endothelial cells (HAECs) were utilized to analyze ATO-induced EndMT in vitro. The cytoskeleton of HAECs was visualized using F-actin staining to observe cell morphology, while Dil-Ac-LDL staining was employed to assess cell functionality. EndMT-related factors (CD31 and α-SMA), glucocorticoid receptor (GR) and interleukin-6 (IL-6) were detected by immunofluorescence and Western blot in vivo and in vitro. Furthermore, GR was knocked down by si-GR, and IL-6 was blocked by IL-6 neutralizing antibody to verify their role in the effect of DIO on ATO-induced EndMT in HAECs. RESULTS: DIO exhibited significant efficacy in ATO-induced damage to both cardiac diastolic and systolic function, along with mitigating cardiac fibrosis. Additionally, DIO alleviated the loss of cytoskeletal anisotropy and enhanced the uptake of Dil-Ac-LDL in HAECs. Furthermore, it reversed the ATO-induced downregulation of endothelial-specific markers CD31 and GR, while suppressing the upregulation of mesenchymal markers α-SMA and IL-6, both in vivo and in vitro. Notably, the protective effect of DIO was compromised upon knockdown of GR, which also led to a reversal of DIO-induced IL-6 downregulation. Furthermore, the neutralization of IL-6 with specific antibodies abolished the ATO-induced changes related to EndMT. CONCLUSION: In this study, we clarified the protective effect of DIO on ATO-induced myocardial fibrosis against EndMT via the GR/IL-6 axis for the first time and provided a potential therapeutic agent for preventing heart damage caused by ATO.


Asunto(s)
Trióxido de Arsénico , Diosgenina , Células Endoteliales , Fibrosis , Ratas Wistar , Animales , Fibrosis/tratamiento farmacológico , Humanos , Masculino , Diosgenina/farmacología , Diosgenina/análogos & derivados , Células Endoteliales/efectos de los fármacos , Ratas , Interleucina-6/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Actinas/metabolismo , Miocardio/patología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Transición Endotelial-Mesenquimatosa
6.
Adv Exp Med Biol ; 1459: 321-339, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39017850

RESUMEN

The transformation of acute promyelocytic leukemia (APL) from the most fatal to the most curable subtype of acute myeloid leukemia (AML), with long-term survival exceeding 90%, has represented one of the most exciting successes in hematology and in oncology. APL is a paradigm for oncoprotein-targeted cure.APL is caused by a 15/17 chromosomal translocation which generates the PML-RARA fusion protein and can be cured by the chemotherapy-free approach based on the combination of two therapies targeting PML-RARA: retinoic acid (RA) and arsenic. PML-RARA is the key driver of APL and acts by deregulating transcriptional control, particularly RAR targets involved in self-renewal or myeloid differentiation, also disrupting PML nuclear bodies. PML-RARA mainly acts as a modulator of the expression of specific target genes: genes whose regulatory elements recruit PML-RARA are not uniformly repressed but also may be upregulated or remain unchanged. RA and arsenic trioxide directly target PML-RARA-mediated transcriptional deregulation and protein stability, removing the differentiation block at promyelocytic stage and inducing clinical remission of APL patients.


Asunto(s)
Leucemia Promielocítica Aguda , Proteínas de Fusión Oncogénica , Tretinoina , Humanos , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patología , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Tretinoina/uso terapéutico , Tretinoina/farmacología , Trióxido de Arsénico/uso terapéutico , Trióxido de Arsénico/farmacología , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Antineoplásicos/uso terapéutico , Antineoplásicos/farmacología , Arsenicales/uso terapéutico , Arsenicales/farmacología , Óxidos/uso terapéutico , Óxidos/farmacología , Animales
7.
Oncol Rep ; 52(2)2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38963046

RESUMEN

Arsenic trioxide (ATO) is expected to be a chemical drug with antitumor activity against acute promyelocytic leukemia (APL), a type of acute myeloid leukemia. In Japan, its antitumor effects were confirmed in clinical trials for APL, and it has been approved in various countries around the world. However, there have been no reports on ATO's antitumor effects on radioresistant leukemia cells, which can be developed during radiotherapy and in combination with therapeutic radiation beams. The present study sought to clarify the antitumor effect of ATO on APL cells with radiation resistance and determine its efficacy when combined with ionizing radiation (IR). The radiation­resistant HL60 (Res­HL60) cell line was generated by subjecting the native cells to 4­Gy irradiation every week for 4 weeks. The half­maximal inhibitory concentration (IC50) for cell proliferation by ATO on native cell was 0.87 µM (R2=0.67), while the IC50 for cell proliferation by ATO on Res­HL60 was 2.24 µM (R2=0.91). IR exposure increased the sub­G1 and G2/M phase ratios in both cell lines. The addition of ATO resulted in a higher population of G2/M after 24 h rather than 48 h. When the rate of change in the sub­G1 phase was examined in greater detail, the sub­G1 phase in both control cells without ATO significantly increased by exposure to IR at 24 h, but only under the condition of 2 Gy irradiation, it had continued to increase at 48 h. Res­HL60 supplemented with ATO showed a higher rate of sub­G1 change at 24 h; however, 2 Gy irradiation resulted in a decrease compared with the control. There was a significant increase in the ratio of the G2/M phase in cells after incubation with ATO for 24 h, and exposure to 2 Gy irradiation caused an even greater increase. To determine whether the inhibition of cell proliferation and cell cycle disruptions is related to reactive oxygen species (ROS) activity, intracellular ROS levels were measured with a flow cytometric assay. Although the ROS levels of Res­HL60 were higher than those of native cells in the absence of irradiation, they did not change after 0.5 or 2 Gy irradiation. Furthermore, adding ATO to Res­HL60 reduced intracellular ROS levels. These findings provide important information that radioresistant leukemia cells respond differently to the antitumor effect of ATO and the combined effect of IR.


Asunto(s)
Trióxido de Arsénico , Arsenicales , Proliferación Celular , Leucemia Promielocítica Aguda , Óxidos , Radiación Ionizante , Humanos , Trióxido de Arsénico/farmacología , Leucemia Promielocítica Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/patología , Leucemia Promielocítica Aguda/radioterapia , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Células HL-60 , Arsenicales/farmacología , Óxidos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Tolerancia a Radiación/efectos de los fármacos , Antineoplásicos/farmacología , Especies Reactivas de Oxígeno/metabolismo
8.
Int J Pharm ; 661: 124426, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38972519

RESUMEN

In recent years, the use of arsenic trioxide (ATO) in the context of ovarian cancer chemotherapy has attracted significant attention. However, ATO's limited biocompatibility and the occurrence of severe toxic side effects hinder its clinical application. A nanoparticle (NP) drug delivery system using ATO as a therapeutic agent is reported in this study. Achieving a synergistic effect by combining starvation therapy, chemodynamic therapy, and chemotherapy for the treatment of ovarian cancer was the ultimate goal of this system. This nanotechnology-based drug delivery system (NDDS) introduced arsenic-manganese complexes into cancer cells, leading to the subsequent release of lethal arsenic ions (As3+) and manganese ions (Mn2+). The acidic microenvironment of the tumor facilitated this process, and MR imaging offered real-time monitoring of the ATO dose distribution. Simultaneously, to produce reactive oxygen species that induced cell death through a Fenton-like reaction, Mn2+ exploited the surplus of hydrogen peroxide (H2O2) within tumor cells. Glucose oxidase-based starvation therapy further supported this mechanism, which restored H2O2 and lowered the cellular acidity. Consequently, this approach achieved self-enhanced chemodynamic therapy. Homologous targeting of the NPs was facilitated through the use of SKOV3 cell membranes that encapsulated the NPs. Hence, the use of a multimodal NDDS that integrated ATO delivery, therapy, and monitoring exhibited superior efficacy and biocompatibility compared with the nonspecific administration of ATO. This approach presents a novel concept for the diagnosis and treatment of ovarian cancer.


Asunto(s)
Antineoplásicos , Trióxido de Arsénico , Nanopartículas , Neoplasias Ováricas , Femenino , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Humanos , Concentración de Iones de Hidrógeno , Línea Celular Tumoral , Trióxido de Arsénico/administración & dosificación , Trióxido de Arsénico/química , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/farmacología , Nanopartículas/química , Manganeso/química , Manganeso/administración & dosificación , Peróxido de Hidrógeno , Especies Reactivas de Oxígeno/metabolismo , Ratones , Materiales Biomiméticos/química , Materiales Biomiméticos/administración & dosificación , Ratones Desnudos , Sistemas de Liberación de Medicamentos/métodos , Sistema de Administración de Fármacos con Nanopartículas/química , Glucosa Oxidasa/administración & dosificación , Ratones Endogámicos BALB C
9.
PeerJ ; 12: e17559, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38854798

RESUMEN

Background: To investigate the effects of arsenic trioxide (ATO) on human colorectal cancer cells (HCT116) growth and the role of transient receptor potential melastatin 4 (TRPM4) channel in this process. Methods: The viability of HCT116 cells was assessed using the CCK-8 assay. Western blot analysis was employed to examine the protein expression of TRPM4. The apoptosis of HCT116 cells was determined using TUNEL and Flow cytometry. Cell migration was assessed through the cell scratch recovery assay and Transwell cell migration assay. Additionally, Transwell cell invasion assay was performed to determine the invasion ability of HCT116 cells. Results: ATO suppressed the viability of HCT116 cells in a dose-dependent manner, accompanied by a decline in cell migration and invasion, and an increase in apoptosis. 9-phenanthroline (9-Ph), a specific inhibitor of TRPM4, abrogated the ATO-induced upregulation of TRPM4 expression. Additionally, blocking TRPM4 reversed the effects of ATO on HCT116 cells proliferation, including restoration of cell viability, migration and invasion, as well as the inhibition of apoptosis. Conclusion: ATO inhibits CRC cell growth by inducing TRPM4 expression, our findings indicate that ATO is a promising therapeutic strategy and TRPM4 may be a novel target for the treatment of CRC.


Asunto(s)
Apoptosis , Trióxido de Arsénico , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Neoplasias Colorrectales , Canales Catiónicos TRPM , Humanos , Canales Catiónicos TRPM/metabolismo , Canales Catiónicos TRPM/antagonistas & inhibidores , Canales Catiónicos TRPM/genética , Trióxido de Arsénico/farmacología , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/metabolismo , Células HCT116 , Movimiento Celular/efectos de los fármacos , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Óxidos/farmacología , Antineoplásicos/farmacología , Invasividad Neoplásica , Arsenicales/farmacología
10.
Cell Death Dis ; 15(6): 408, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38862470

RESUMEN

The cavity-creating p53 cancer mutation Y220C is an ideal paradigm for developing small-molecule drugs based on protein stabilization. Here, we have systematically analyzed the structural and stability effects of all oncogenic Tyr-to-Cys mutations (Y126C, Y163C, Y205C, Y220C, Y234C, and Y236C) in the p53 DNA-binding domain (DBD). They were all highly destabilizing, drastically lowering the melting temperature of the protein by 8-17 °C. In contrast, two non-cancerous mutations, Y103C and Y107C, had only a moderate effect on protein stability. Differential stabilization of the mutants upon treatment with the anticancer agent arsenic trioxide and stibogluconate revealed an interesting proximity effect. Crystallographic studies complemented by MD simulations showed that two of the mutations, Y234C and Y236C, create internal cavities of different size and shape, whereas the others induce unique surface lesions. The mutation-induced pockets in the Y126C and Y205C mutant were, however, relatively small compared with that of the already druggable Y220C mutant. Intriguingly, our structural studies suggest a pronounced plasticity of the mutation-induced pocket in the frequently occurring Y163C mutant, which may be exploited for the development of small-molecule stabilizers. We point out general principles for reactivating thermolabile cancer mutants and highlight special cases where mutant-specific drugs are needed for the pharmacological rescue of p53 function in tumors.


Asunto(s)
Mutación , Neoplasias , Proteína p53 Supresora de Tumor , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Humanos , Mutación/genética , Neoplasias/genética , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Neoplasias/metabolismo , Trióxido de Arsénico/farmacología , Simulación de Dinámica Molecular , Estabilidad Proteica/efectos de los fármacos , Antineoplásicos/farmacología , Antineoplásicos/química
11.
Expert Opin Drug Deliv ; 21(6): 867-880, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38913024

RESUMEN

INTRODUCTION: Arsenicals have a special place in the history of human health, acting both as poison and medicine. Having been used to treat a variety of diseases in the past, the success of arsenic trioxide (ATO) in treating acute promyelocytic leukemia (APL) in the last century marked its use as a drug in modern medicine. To expand their role against cancer, there have been clinical uses of arsenicals worldwide and progress in the development of drug delivery for various malignancies, especially solid tumors. AREAS COVERED: In this review, conducted on Google Scholar [1977-2024], we start with various forms of arsenicals, highlighting the well-known ATO. The mechanism of action of arsenicals in cancer therapy is then overviewed. A summary of the research progress in developing new delivery approaches (e.g. polymers, inorganic frameworks, and biomacromolecules) in recent years is provided, addressing the challenges and opportunities in treating various malignant tumors. EXPERT OPINION: Reducing toxicity and enhancing therapeutic efficacy are guidelines for designing and developing new arsenicals and drug delivery systems. They have shown potential in the fight against cancer and emerging pathogens. New technologies and strategies can help us harness the potency of arsenicals and make better products.


Asunto(s)
Antineoplásicos , Trióxido de Arsénico , Arsenicales , Sistemas de Liberación de Medicamentos , Neoplasias , Humanos , Trióxido de Arsénico/administración & dosificación , Trióxido de Arsénico/uso terapéutico , Trióxido de Arsénico/farmacocinética , Neoplasias/tratamiento farmacológico , Arsenicales/administración & dosificación , Arsenicales/uso terapéutico , Arsenicales/farmacocinética , Antineoplásicos/administración & dosificación , Animales , Desarrollo de Medicamentos , Diseño de Fármacos , Leucemia Promielocítica Aguda/tratamiento farmacológico
12.
Colloids Surf B Biointerfaces ; 241: 114014, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38850742

RESUMEN

Arsenic trioxide (ATO) has gained significant attention due to its promising therapeutic effects in treating different diseases, particularly acute promyelocytic leukemia (APL). Its potent anticancer mechanisms have been extensively studied. Despite the great efficacy ATO shows in fighting cancers, drawbacks in the clinical use are obvious, especially for solid tumors, which include rapid renal clearance and short half-life, severe adverse effects, and high toxicity to normal cells. Recently, the emergence of nanomedicine offers a potential solution to these limitations. The enhanced biocompatibility, excellent targeting capability, and desirable effectiveness have attracted much interest. Therefore, we summarized various nanocarriers for targeted delivery of ATO to solid tumors. We also provided detailed anticancer mechanisms of ATO in treating cancers, its clinical trials and shortcomings as well as the combination therapy of ATO and other chemotherapeutic agents for reduced drug resistance and synergistic effects. Finally, the future study direction and prospects were also presented.


Asunto(s)
Antineoplásicos , Trióxido de Arsénico , Portadores de Fármacos , Neoplasias , Trióxido de Arsénico/química , Trióxido de Arsénico/administración & dosificación , Trióxido de Arsénico/farmacología , Humanos , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/administración & dosificación , Portadores de Fármacos/química , Neoplasias/tratamiento farmacológico , Nanopartículas/química , Animales , Sistemas de Liberación de Medicamentos
13.
EMBO Mol Med ; 16(6): 1324-1351, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38730056

RESUMEN

Clear-cell renal cell carcinoma (ccRCC), the major subtype of RCC, is frequently diagnosed at late/metastatic stage with 13% 5-year disease-free survival. Functional inactivation of the wild-type p53 protein is implicated in ccRCC therapy resistance, but the detailed mechanisms of p53 malfunction are still poorly characterized. Thus, a better understanding of the mechanisms of disease progression and therapy resistance is required. Here, we report a novel ccRCC dependence on the promyelocytic leukemia (PML) protein. We show that PML is overexpressed in ccRCC and that PML depletion inhibits cell proliferation and relieves pathologic features of anaplastic disease in vivo. Mechanistically, PML loss unleashed p53-dependent cellular senescence thus depicting a novel regulatory axis to limit p53 activity and senescence in ccRCC. Treatment with the FDA-approved PML inhibitor arsenic trioxide induced PML degradation and p53 accumulation and inhibited ccRCC expansion in vitro and in vivo. Therefore, by defining non-oncogene addiction to the PML gene, our work uncovers a novel ccRCC vulnerability and lays the foundation for repurposing an available pharmacological intervention to restore p53 function and chemosensitivity.


Asunto(s)
Carcinoma de Células Renales , Senescencia Celular , Neoplasias Renales , Proteína de la Leucemia Promielocítica , Proteína p53 Supresora de Tumor , Proteína de la Leucemia Promielocítica/metabolismo , Proteína de la Leucemia Promielocítica/genética , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/tratamiento farmacológico , Humanos , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Senescencia Celular/efectos de los fármacos , Animales , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Neoplasias Renales/genética , Neoplasias Renales/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Trióxido de Arsénico/farmacología , Ratones
14.
Nucleic Acids Res ; 52(11): 6472-6489, 2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-38752489

RESUMEN

Orphan nuclear receptors (NRs), such as COUP-TF1, COUP-TF2, EAR2, TR2 and TR4, are implicated in telomerase-negative cancers that maintain their telomeres through the alternative lengthening of telomeres (ALT) mechanism. However, how telomere association of orphan NRs is involved in ALT activation remains unclear. Here, we demonstrate that telomeric tethering of orphan NRs in human fibroblasts initiates formation of ALT-associated PML bodies (APBs) and features of ALT activity, including ALT telomere DNA synthesis, telomere sister chromatid exchange, and telomeric C-circle generation, suggesting de novo ALT induction. Overexpression of orphan NRs exacerbates ALT phenotypes in ALT cells, while their depletion limits ALT. Orphan NRs initiate ALT via the zinc finger protein 827, suggesting the involvement of chromatin structure alterations for ALT activation. Furthermore, we found that orphan NRs and deficiency of the ALT suppressor ATRX-DAXX complex operate in concert to promote ALT activation. Moreover, PML depletion by gene knockout or arsenic trioxide treatment inhibited ALT induction in fibroblasts and ALT cancer cells, suggesting that APB formation underlies the orphan NR-induced ALT activation. Importantly, arsenic trioxide administration abolished APB formation and features of ALT activity in ALT cancer cell line-derived mouse xenografts, suggesting its potential for further therapeutic development to treat ALT cancers.


Asunto(s)
Fibroblastos , Proteína de la Leucemia Promielocítica , Homeostasis del Telómero , Humanos , Animales , Proteína de la Leucemia Promielocítica/metabolismo , Proteína de la Leucemia Promielocítica/genética , Ratones , Fibroblastos/metabolismo , Telómero/metabolismo , Telómero/genética , Proteína Nuclear Ligada al Cromosoma X/genética , Proteína Nuclear Ligada al Cromosoma X/metabolismo , Proteínas Co-Represoras/genética , Proteínas Co-Represoras/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Intercambio de Cromátides Hermanas , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Línea Celular Tumoral , Trióxido de Arsénico/farmacología , Chaperonas Moleculares
15.
PLoS One ; 19(5): e0302701, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38728286

RESUMEN

Although the toxicity of arsenic depends on its chemical forms, few studies have taken into account the ambiguous phenomenon that sodium arsenite (NaAsO2) acts as a potent carcinogen while arsenic trioxide (ATO, As2O3) serves as an effective therapeutic agent in lymphoma, suggesting that NaAsO2 and As2O3 may act via paradoxical ways to either promote or inhibit cancer pathogenesis. Here, we compared the cellular response of the two arsenical compounds, NaAsO2 and As2O3, on the Burkitt lymphoma cell model, the Epstein Barr Virus (EBV)-positive P3HR1 cells. Using flow cytometry and biochemistry analyses, we showed that a NaAsO2 treatment induces P3HR1 cell death, combined with drastic drops in ΔΨm, NAD(P)H and ATP levels. In contrast, As2O3-treated cells resist to cell death, with a moderate reduction of ΔΨm, NAD(P)H and ATP. While both compounds block cells in G2/M and affect their protein carbonylation and lipid peroxidation, As2O3 induces a milder increase in superoxide anions and H2O2 than NaAsO2, associated to a milder inhibition of antioxidant defenses. By electron microscopy, RT-qPCR and image cytometry analyses, we showed that As2O3-treated cells display an overall autophagic response, combined with mitophagy and an unfolded protein response, characteristics that were not observed following a NaAsO2 treatment. As previous works showed that As2O3 reactivates EBV in P3HR1 cells, we treated the EBV- Ramos-1 cells and showed that autophagy was not induced in these EBV- cells upon As2O3 treatment suggesting that the boost of autophagy observed in As2O3-treated P3HR1 cells could be due to the presence of EBV in these cells. Overall, our results suggest that As2O3 is an autophagic inducer which action is enhanced when EBV is present in the cells, in contrast to NaAsO2, which induces cell death. That's why As2O3 is combined with other chemicals, as all-trans retinoic acid, to better target cancer cells in therapeutic treatments.


Asunto(s)
Trióxido de Arsénico , Arsenicales , Arsenitos , Autofagia , Mitocondrias , Estrés Oxidativo , Óxidos , Compuestos de Sodio , Trióxido de Arsénico/farmacología , Arsenitos/farmacología , Arsenitos/toxicidad , Humanos , Estrés Oxidativo/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Compuestos de Sodio/farmacología , Arsenicales/farmacología , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Óxidos/farmacología , Muerte Celular/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Herpesvirus Humano 4/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Peróxido de Hidrógeno/farmacología , Peroxidación de Lípido/efectos de los fármacos , Linfoma de Burkitt/virología , Linfoma de Burkitt/metabolismo , Linfoma de Burkitt/patología , Linfoma de Burkitt/tratamiento farmacológico
16.
Am J Emerg Med ; 80: 226.e1-226.e3, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38702220

RESUMEN

A 53-year-old male patient presented to a regional hospital Emergency Department approximately 2 h post an intentional ingestion of Coopers Instant Wetting Powder Sheep Dip (66% arsenic trioxide, 23% sulphur and 0.42% rotenone), mixed in 600 mL water, as a suicide attempt. On arrival to the Emergency Department, the patient had nausea, vomiting and diarrhoea. Seven hours post ingestion, hypotension developed (BP 90/60 mmHg) and intravenous fluids were commenced. He later developed QTc prolongation. He was treated with 2,3-Dimercapto-1-propanesulfonic acid (DMPS) and N-acetylcysteine and improved without development of neurology. Further investigation of NAC efficacy in humans in the setting of acute arsenic poisoning is required and the optimal duration of treatment and dosing needs to be established. This case highlights an uncommon poisoning which presented to the Emergency Department, the acute symptoms of arsenic toxicity and considerations for management.


Asunto(s)
Acetilcisteína , Intoxicación por Arsénico , Arsenicales , Intento de Suicidio , Masculino , Humanos , Persona de Mediana Edad , Acetilcisteína/uso terapéutico , Trióxido de Arsénico/envenenamiento , Óxidos/envenenamiento , Antídotos/uso terapéutico , Unitiol/uso terapéutico
17.
Immunopharmacol Immunotoxicol ; 46(3): 408-416, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38816179

RESUMEN

BACKGROUND: Myelodysplastic syndrome (MDS) is a prevalent hematological neoplastic disorder in clinics and its immunopathogenesis has garnered growing interest. Oral and intravenous arsenic agents have long been used to treat hematological malignancies. The main component of oral arsenic is realgar (arsenic disulfide), while arsenic trioxide is the main component of intravenous arsenic. METHODS: This study aimed to assess the effects of ATO and Realgar on the enhancement of peripheral blood, drug safety, and T cell immune status in the NUP98-HOXD13 (NHD13) mice model of MDS, specifically in the peripheral blood, spleen, and liver. RESULTS: The study findings indicate that realgar and arsenic trioxide (ATO) can improve peripheral hemogram in mice, whereas realgar promotes higher peripheral blood cell production than ATO. Furthermore, the clinical administration method and dose did not cause significant toxicity or side effects and thus can be considered safe. Coexistence and interconversion of hyperimmune function and immunosuppression in mice were also observed in this study. In addition, there were interactions between immune cells in the peripheral blood, spleen, and liver to regulate the immune balance of the body and activate immunity via T-cell activation. CONCLUSION: In summary, oral and intravenous arsenic agents are beneficial in improving peripheral hemogram and immunity in mice.


Asunto(s)
Trióxido de Arsénico , Arsenicales , Modelos Animales de Enfermedad , Síndromes Mielodisplásicos , Animales , Trióxido de Arsénico/administración & dosificación , Trióxido de Arsénico/farmacología , Arsenicales/farmacología , Arsenicales/administración & dosificación , Ratones , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/inmunología , Sulfuros/farmacología , Sulfuros/administración & dosificación , Disulfuros/farmacología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Bazo/efectos de los fármacos , Bazo/inmunología
18.
Talanta ; 276: 126240, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38754186

RESUMEN

Arsenic has been an element of great interest among scientists for many years as it is a widespread metalloid in our ecosystem. Arsenic is mostly recognized with negative connotations due to its toxicity. Surely, most of us know that a long time ago, arsenic trioxide was used in medicine to treat, mainly, skin diseases. However, not everyone knows about its very wide and promising use in the treatment of cancer. Initially, in the seventies, it was used to treat leukemia, but new technological possibilities and the development of nanotechnology have made it possible to use arsenic trioxide for the treatment of solid tumours. The most toxic arsenic compound - arsenic trioxide - as the basis of anticancer drugs in which they function as a component of nanoparticles is used in the fight against various types of cancer. This review aims to present the current solutions in various cancer treatment using arsenic compounds with different binding motifs and methods of preparation to create targeted nanoparticles, nanodiamonds, nanohybrids, nanodrugs, or nanovehicles.


Asunto(s)
Antineoplásicos , Nanopartículas , Neoplasias , Animales , Humanos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Arsénico/farmacología , Arsénico/uso terapéutico , Trióxido de Arsénico/farmacología , Trióxido de Arsénico/uso terapéutico , Nanopartículas/química , Neoplasias/tratamiento farmacológico
19.
Int J Phytoremediation ; 26(10): 1683-1690, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38712857

RESUMEN

The studies showed the effectiveness of green-synthesized SiO2NPs in mitigating the toxicity of Arsenic. Density Functional Theory (DFT) is a computational method used to determine electronic structure, energy gap, and toxicity prediction. Experimentally, silicon nanoparticles of 0 (S0) and 100% v/v (S100) were applied to the surface of the soil. 150 mL of Arsenic trioxide was applied twice at a rate of 0 (As0) and 3.2 g/mL (As3.2) at an interval of three weeks. Green synthesized SiO2NPs possessed a higher chemical potential (µ) and electrophilicity index; consequently, charges could be transferred and easily polarized. The highest occupied molecular orbital (HOMO) and lowest unoccupied molecular orbital (LUMO) levels of the green synthesized SiO2NPs enable them to donate electrons and complex with arsenic, reducing their bioavailability and toxicity. Evidence from the studies further showed that SiO2NPs had buffered the soil acidity and electric conductivity, posing a high binding site and reactivity with exchangeable cations and micronutrients due to their smaller energy gap. Furthermore, the catalytic activities of the soil enzymes dehydrogenase (DHA) and peroxidase (POD) were greatly increased, which enhanced the electrostatic interaction between the SiO2NPs and As.


Asunto(s)
Arsénico , Nanopartículas , Dióxido de Silicio , Contaminantes del Suelo , Contaminantes del Suelo/metabolismo , Dióxido de Silicio/química , Arsénico/metabolismo , Suelo/química , Tecnología Química Verde , Trióxido de Arsénico , Restauración y Remediación Ambiental/métodos
20.
Leg Med (Tokyo) ; 69: 102458, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38781725

RESUMEN

Arsenic trioxide (ATO), one of the oldest and most frequently used poisons, is well-known in forensic science for inducing hepatotoxicity. The regulation of peroxisomal antioxidative enzyme catalase (CAT) involves intricate mechanisms at both transcriptional and post-transcriptional levels. However, the molecular mechanisms underlying the regulation of CAT gene expression in hepatic cells remain elusive. Furthermore, the regulation of CAT gene expression evident in animals administered with ATO in vivo is not well-explored, although several studies have revealed ATO-induced reductions in CAT enzymatic activity in rat livers. In this study, we revealed ATO-dependent reductions in CAT gene expression in both rat liver and Huh-7 human hepatoma cells. Our results indicate that the decline in CAT enzymatic activity can be attributed, at least in part, to the downregulation of its gene expression. The ATO-induced reduction in CAT expression was concurrent with the reduction in peroxisome proliferator-activated receptor-gamma (PPARγ) coactivator (PGC)-1α and inactivation of PPARγ, both considered as positive regulators of CAT gene expression. Moreover, antioxidant N-acetylcysteine (NAC) demonstrated the capability to alleviate the downregulation of CAT gene expression both in vivo and in vitro. Additionally, NAC played a role in alleviating ATO-induced hepatotoxicity, potentially by mitigating the transcriptional downregulation of the CAT gene. Altogether, these results indicate that ATO exerts toxicity by inhibiting the antioxidant defense mechanism, which may be useful for forensic diagnosis of arsenic poisoning and clinical treatment of mitigating ATO-induced hepatotoxicity.


Asunto(s)
Acetilcisteína , Trióxido de Arsénico , Catalasa , Hígado , Óxidos , Trióxido de Arsénico/farmacología , Acetilcisteína/farmacología , Animales , Catalasa/metabolismo , Catalasa/genética , Ratas , Hígado/metabolismo , Hígado/efectos de los fármacos , Masculino , Arsenicales , Humanos , Expresión Génica/efectos de los fármacos , Antioxidantes/farmacología , Antioxidantes/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA