Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Hazard Mater ; 445: 130620, 2023 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-37056004

RESUMEN

The highly effective utilization of uranium resources in global seawater is a viable method to satisfy the rising demands for fueling nuclear energy industry. Herein, inspired by the multi-mechanisms of the marine bacteria for uranium immobilization, CdS nanoparticles are deposited on the cell of marine bacterial strain Bacillus velezensis UUS-1 to create a photosensitized biohybrid system UUS-1/CdS. This system achieves high uranium extraction efficiency using a cascaded strategy, where the bacterial cells guarantee high extraction selectivity and the photosensitive CdS nanoparticles realize cascading photoreduction of high soluble U(VI) to low soluble U(IV) to enhance extraction capacity. As one of the fastest-acting adsorbents in natural seawater, a high extraction capacity for uranium of 7.03 mg g-1 is achieved with an ultrafast extraction speed of 4.69 mg g-1 d-1. The cascaded strategy promisingly improves uranium extraction performance and pioneers a new direction for the design of adsorbents to extract uranium from seawater.


Asunto(s)
Uranio , Agua de Mar
2.
Small ; 19(23): e2207201, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36899444

RESUMEN

Insufficienct T lymphocyte infiltration and unresponsiveness to immune checkpoint blockade therapy are still major difficulties for the clinical treatment of pancreatic ductal adenocarcinoma (PDAC). Although econazole has shown promise in inhibiting PDAC growth, its poor bioavailability and water solubility limit its potential as a clinical therapy for PDAC. Furthermore, the synergistic role of econazole and biliverdin in immune checkpoint blockade therapy in PDAC remains elusive and challenging. Herein, a chemo-phototherapy nanoplatform is designed by which econazole and biliverdin can be co-assembled (defined as FBE NPs), which significantly improve the poor water solubility of econazole and enhance the efficacy of PD-L1 checkpoint blockade therapy against PDAC. Mechanistically, econazole and biliverdin are directly released into the acidic cancer microenvironment, to activate immunogenic cell death via biliverdin-induced PTT/PDT and boost the immunotherapeutic response of PD-L1 blockade. In addition, econazole simultaneously enhances PD-L1 expression to sensitize anti-PD-L1 therapy, leading to suppression of distant tumors, long-term immune memory effects, improved dendritic cell maturation, and tumor infiltration of CD8+ T lymphocytes. The combined FBE NPs and α-PDL1 show synergistic antitumor efficacy. Collectively, FBE NPs show excellent biosafety and antitumor efficacy by combining chemo-phototherapy with PD-L1 blockade, which has promising potential in a precision medicine approach as a PDAC treatment strategy.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Econazol/uso terapéutico , Biliverdina/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Carcinoma Ductal Pancreático/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Inmunoterapia , Agua , Microambiente Tumoral , Línea Celular Tumoral , Neoplasias Pancreáticas
3.
Pharmaceuticals (Basel) ; 15(12)2022 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-36558995

RESUMEN

Polyphenol, one of the major components that exert the therapeutic effect of Chinese herbal medicine (CHM), comprises several categories, including flavonoids, phenolic acids, lignans and stilbenes, and has long been studied in oncology due to its significant efficacy against cancers in vitro and in vivo. Recent evidence has linked this antitumor activity to the role of polyphenols in the modulation of redox homeostasis (e.g., pro/antioxidative effect) in cancer cells. Dysregulation of redox homeostasis could lead to the overproduction of reactive oxygen species (ROS), resulting in oxidative stress, which is essential for many aspects of tumors, such as tumorigenesis, progression, and drug resistance. Thus, investigating the ROS-mediated anticancer properties of polyphenols is beneficial for the discovery and development of novel pharmacologic agents. In this review, we summarized these extensively studied polyphenols and discussed the regulatory mechanisms related to the modulation of redox homeostasis that are involved in their antitumor property. In addition, we discussed novel technologies and strategies that could promote the development of CHM-derived polyphenols to improve their versatile anticancer properties, including the development of novel delivery systems, chemical modification, and combination with other agents.

4.
Phytomed Plus ; 2(3): 100280, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35463625

RESUMEN

Background: The presence of diabetes mellitus (DM) among COVID-19 patients is associated with increased hospitalization, morbidity, and mortality. Evidence has shown that hyperglycemia potentiates SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) infection and plays a central role in severe COVID-19 and diabetes comorbidity. In this review, we explore the therapeutic potentials of herbal medications and natural products in the management of COVID-19 and DM comorbidity and the challenges associated with the preexisting or concurrent use of these substances. Methods: Research papers that were published from January 2016 to December 2021 were retrieved from PubMed, ScienceDirect, and Google Scholar databases. Papers reporting clinical evidence of antidiabetic activities and any available evidence of the anti-COVID-19 potential of ten selected natural products were retrieved and analyzed for discussion in this review. Results: A total of 548 papers (73 clinical trials on the antidiabetic activities of the selected natural products and 475 research and review articles on their anti-COVID-19 potential) were retrieved from the literature search for further analysis. A total of 517 articles (reviews and less relevant research papers) were excluded. A cumulative sum of thirty-one (31) research papers (20 clinical trials and 10 others) met the criteria and have been discussed in this review. Conclusion: The findings of this review suggest that phenolic compounds are the most promising phytochemicals in the management of COVID-19 and DM comorbidity. Curcumin and propolis have shown substantial evidence against COVID-19 and DM in humans and are thus, considered the best potential therapeutic options.

5.
Front Oncol ; 11: 746917, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34722304

RESUMEN

As the main cause of death in the world, cancer is one of the major health threats for humans. In recent years, traditional Chinese medicine has gained great attention in oncology due to the features of multi-targets, multi-pathways, and slight side effects. Moreover, lots of traditional Chinese medicine can exert immunomodulatory effects in vivo. In the tumor microenvironment, tumor cells, immune cells as well as other stromal cells often coexist. With the development of cancer, tumor cells proliferate uncontrollably, metastasize aggressively, and modulate the proportion and status of immune cells to debilitate the antitumor immunity. Reversal of immunosuppressive tumor microenvironment plays an essential role in cancer prevention and therapy. Immunotherapy has become the most promising strategy for cancer therapy. Chinese medicine compounds can stimulate the activation and function of immune cells, such as promoting the maturation of dendritic cells and inducing the differentiation of myeloid-derived suppressor cells to dendritic cells and macrophages. In the present review, we summarize and discuss the effects of Chinese medicine compounds on immune cells in the tumor microenvironment, including innate immune cells (dendritic cells, natural killer cells, macrophages, and myeloid-derived suppressor cells) and adaptive immune cells (CD4+/CD8+ T lymphocytes and regulatory T cells), and the various immunomodulatory roles of Chinese medicine compounds in cancer therapy such as improving tumor-derived inflammation, enhancing the immunity after surgery or chemotherapy, blocking the immune checkpoints, et al., aiming to provide more thoughts for the anti-tumor mechanisms and applications of Chinese medicine compounds in terms of tumor immunity.

6.
Clin Transl Med ; 11(7): e410, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34323416

RESUMEN

BACKGROUND: Liver fibrosis and fibrosis-related hepatocarcinogenesis are a rising cause for morbidity and death worldwide. Although transforming growth factor-ß (TGF-ß) is a critical mediator of chronic liver fibrosis, targeting TGF-ß isoforms and receptors lead to unacceptable side effect. This study was designed to explore the antifibrotic effect of Compound kushen injection (CKI), an approved traditional Chinese medicine formula, via a therapeutic strategy of rebalancing TGF-ß/Smad7 signaling. METHODS: A meta-analysis was performed to evaluate CKI intervention on viral hepatitis-induced fibrosis or cirrhosis in clinical randomized controlled trials (RCTs). Mice were given carbon tetrachloride (CCl4 ) injection or methionine-choline deficient (MCD) diet to induce liver fibrosis, followed by CKI treatment. We examined the expression of TGF-ß/Smad signaling and typical fibrosis-related genes in hepatic stellate cells (HSCs) and fibrotic liver tissues by qRT-PCR, Western blotting, RNA-seq, immunofluorescence, and immunohistochemistry. RESULTS: Based on meta-analysis results, CKI improved the liver function and relieved liver fibrosis among patients. In our preclinical studies by using two mouse models, CKI treatment demonstrated promising antifibrotic effects and postponed hepatocarcinogenesis with improved liver function and histopathologic features. Mechanistically, we found that CKI inhibited HSCs activation by stabilizing the interaction of Smad7/TGF-ßR1 to rebalance Smad2/Smad3 signaling, and subsequently decreased the extracellular matrix formation. Importantly, Smad7 depletion abolished the antifibrotic effect of CKI in vivo and in vitro. Moreover, matrine, oxymatrine, sophocarpine, and oxysophocarpine were identified as material basis responsible for the antifibrosis effect of CKI. CONCLUSIONS: Our results unveil the approach of CKI in rebalancing TGF-ß/Smad7 signaling in HSCs to protect against hepatic fibrosis and hepatocarcinogenesis in both preclinical and clinical studies. Our study suggests that CKI can be a candidate for treatment of hepatic fibrosis and related oncogenesis.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Transducción de Señal/efectos de los fármacos , Proteína smad7/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Tetracloruro de Carbono/toxicidad , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Línea Celular , Medicamentos Herbarios Chinos/uso terapéutico , Células Estrelladas Hepáticas/citología , Células Estrelladas Hepáticas/metabolismo , Humanos , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Masculino , Medicina Tradicional China , Metaanálisis como Asunto , Ratones , Ratones Endogámicos C57BL , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteína smad7/antagonistas & inhibidores , Proteína smad7/genética
7.
J Immunother Cancer ; 8(1)2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32179631

RESUMEN

BACKGROUND: There is an urgent need for effective treatments for hepatocellular carcinoma (HCC). Immunotherapy is promising especially when combined with traditional therapies. This study aimed to investigate the immunomodulatory function of an approved Chinese medicine formula, compound kushen injection (CKI), and its anti-HCC efficiency in combination with low-dose sorafenib. METHODS: Growth of two murine HCC cells was evaluated in an orthotopic model, a subcutaneous model, two postsurgical recurrence model, and a tumor rechallenge model with CKI and low-dose sorafenib combination treatment. In vivo macrophage or CD8+ T cell depletion and in vitro primary cell coculture models were used to determine the regulation of CKI on macrophages and CD8+ T cells. RESULTS: CKI significantly enhanced the anticancer activity of sorafenib at a subclinical dose with no obvious side effects. CKI and sorafenib combination treatment prevented the postsurgical recurrence and rechallenged tumor growth. Further, we showed that CKI activated proinflammatory responses and relieved immunosuppression of tumor-associated macrophages in the HCC microenvironment by triggering tumor necrosis factor receptor superfamily member 1 (TNFR1)-mediated NF-κB and p38 MAPK signaling cascades. CKI-primed macrophages significantly promoted the proliferation and the cytotoxic ability of CD8+ T cells and decreased the exhaustion, which subsequently resulted in apoptosis of HCC cells. CONCLUSIONS: CKI acts on macrophages and CD8+ T cells to reshape the immune microenvironment of HCC, which improves the therapeutic outcomes of low-dose sorafenib and avoids adverse chemotherapy effects. Our study shows that traditional Chinese medicines with immunomodulatory properties can potentiate chemotherapeutic drugs and provide a promising approach for HCC treatment.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Sinergismo Farmacológico , Medicamentos Herbarios Chinos/farmacología , Terapia de Inmunosupresión , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Sorafenib/farmacología , Macrófagos Asociados a Tumores/inmunología , Animales , Antineoplásicos/farmacología , Apoptosis , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Proliferación Celular , Medicamentos Herbarios Chinos/administración & dosificación , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Células Tumorales Cultivadas , Microambiente Tumoral/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Cancer Lett ; 454: 14-25, 2019 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-30910587

RESUMEN

Patients receiving sorafenib treatment for hepatocellular carcinoma (HCC) experience different treatment efficacy. Personalized sorafenib treatment should be achieved through the identification of predictors of therapeutic response. In the current study, we found that high UGT1A9 expression indicated better prognosis for HCC patients treated with sorafenib after surgery. In silico analysis predicted microRNA-200a/-183 as potential regulators of the UGT1A gene family via binding to the shared UGT1A9 3'-UTR. A significant inverse correlation between microRNA-200a/-183 and UGT1A9 mRNA level was observed in a panel of HCC specimens. Direct binding was further demonstrated by luciferase reporter gene vector carrying wild-type or binding site truncated UGT1A9 3'-UTR. MicroRNA-200a/-183 downregulated UGT1A9 expression in a dose-dependent manner and significantly reduced sorafenib ß-D-glucuronide formation in HCC cells. These data indicated that UGT1A9, under epigenetic regulation of microRNA-200a/-183, could predict patients who might benefit from adjuvant sorafenib treatment after surgery.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Glucuronosiltransferasa/genética , Neoplasias Hepáticas/tratamiento farmacológico , MicroARNs/genética , Sorafenib/farmacología , Regiones no Traducidas 3' , Animales , Antineoplásicos/farmacología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Epigénesis Genética , Glucuronosiltransferasa/biosíntesis , Glucuronosiltransferasa/metabolismo , Células HEK293 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Ratones Desnudos , MicroARNs/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , UDP Glucuronosiltransferasa 1A9 , Ensayos Antitumor por Modelo de Xenoinjerto
9.
EBioMedicine ; 22: 58-67, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28754304

RESUMEN

Hepatocellular carcinoma (HCC) is a common malignant tumor in the digestive tract with limited therapeutic choices. Although sorafenib, an orally administered multikinase inhibitor, has produced survival benefits for patients with advanced HCC, favorable clinical outcomes are limited due to individual differences and resistance. The application of immunotherapy, a promising approach for HCC is urgently needed. Macrophage infiltration, mediated by the CCL2/CCR2 axis, is a potential immunotherapeutic target. Here, we report that a natural product from Abies georgei, named 747 and related in structure to kaempferol, exhibits sensitivity and selectivity as a CCR2 antagonist. The specificity of 747 on CCR2 was demonstrated via calcium flux, the binding domain of CCR2 was identified in an extracellular loop by chimera binding assay, and in vivo antagonistic activity of 747 was confirmed through a thioglycollate-induced peritonitis model. In animals, 747 elevated the number of CD8+ T cells in tumors via blocking tumor-infiltrating macrophage-mediated immunosuppression and inhibited orthotopic and subcutaneous tumor growth in a CD8+ T cell-dependent manner. Further, 747 enhanced the therapeutic efficacy of low-dose sorafenib without obvious toxicity, through elevating the numbers of intra-tumoral CD8+ T cells and increasing death of tumor cells. Thus, we have discovered a natural CCR2 antagonist and have provided a new perspective on development of this antagonist for treatment of HCC. In mouse models of HCC, 747 enhanced the tumor immunosuppressive microenvironment and potentiated the therapeutic effect of sorafenib, indicating that the combination of an immunomodulator with a chemotherapeutic drug could be a new approach for treating HCC.


Asunto(s)
Abies/química , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Macrófagos/efectos de los fármacos , Extractos Vegetales/administración & dosificación , Receptores CCR2/antagonistas & inhibidores , Animales , Sitios de Unión , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Células Hep G2 , Humanos , Terapia de Inmunosupresión , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Macrófagos/patología , Ratones , Niacinamida/administración & dosificación , Niacinamida/análogos & derivados , Niacinamida/uso terapéutico , Compuestos de Fenilurea/administración & dosificación , Compuestos de Fenilurea/uso terapéutico , Extractos Vegetales/química , Extractos Vegetales/farmacología , Receptores CCR2/química , Sorafenib , Células THP-1 , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
10.
EBioMedicine ; 14: 44-54, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27939426

RESUMEN

Artemisinin (ARS) and its derivatives, which are clinically used antimalarial agents, have shown antitumor activities. Their therapeutic potencies, however, are limited by their low solubility and poor bioavailability. Here, through a pharmacophore hybridization strategy, we synthesized ARS-drug conjugates, in which the marketed chemotherapeutic agents chlorambucil, melphalan, flutamide, aminoglutethimide, and doxifluridine, were separately bonded to Dihydroartemisinin (DHA) through various linkages. Of these, the artemisinin-melphalan conjugate, ARS4, exhibited most toxicity to human ovarian cancer cells but had low cytotoxicity to normal cells. ARS4 inhibited the growth and proliferation of ovarian cancer cells and resulted in S-phase arrest, apoptosis, and inhibition of migration; these effects were stronger than those of its parent drugs, DHA and melphalan. Furthermore, ARS4 modulated the expression of proteins involved in cell cycle progression, apoptosis, and the epithelial-mesenchymal transition (EMT). Moreover, in mice, ARS4 inhibited growth and intraperitoneal dissemination and metastasis of ovarian cancer cells without observable toxic effects. Our results provide a basis for development of the compound as a chemotherapeutic agent. RESEARCH IN CONTEXT: Artemisinin compounds have recently received attention as anticancer agents because of their clinical safety profiles and broad efficacy. However, their therapeutic potencies are limited by low solubility and poor bioavailability. Here, we report that ARS4, an artemisinin-melphalan conjugate, possesses marked in-vitro and in-vivo antitumor activity against ovarian cancer, the effects of which are stronger than those for its parent drugs, Dihydroartemisinin and melphalan. In mice, ARS4 inhibits localized growth of ovarian cancer cells and intraperitoneal dissemination and metastasis without appreciable host toxicity. Thus, for patients with ovarian cancer, ARS4 is a promising chemotherapeutic agent.


Asunto(s)
Antineoplásicos/farmacología , Artemisininas/farmacología , Neoplasias Ováricas/patología , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Artemisininas/administración & dosificación , Artemisininas/efectos adversos , Artemisininas/química , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Combinación de Medicamentos , Evaluación Preclínica de Medicamentos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Humanos , Ratones , Metástasis de la Neoplasia , Estadificación de Neoplasias , Neoplasias Ováricas/tratamiento farmacológico , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Acta Pharmacol Sin ; 37(11): 1509-1515, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27569392

RESUMEN

AIM: To examine the biological consequences and demographic factors that might affect the pharmacokinetics of vitamin D3 after a single high dose intervention in a young Chinese population with vitamin D insufficiency status. METHODS: A total of 28 young subjects (25 to 35 years old) with vitamin D insufficiency status [serum 25(OH)D <30 ng/mL] was recruited in Shanghai, China. The subjects were orally administered a single high dose of vitamin D3 (300 000 IU). Baseline characteristics and blood samples were collected at d 0, 1, 2, 3, 7, 28, 56, 84 and 112 after the intervention. The blood biomarker levels were determined with standardized methods. RESULTS: The intervention markedly increased the blood 25(OH)D3 levels within the first five days (mean Tmax=5.1±2.1 d) and sustained an optimal circulating level of 25(OH)D3 (≥30 ng/mL) for 56 d. After the intervention, body weight and baseline 25(OH)D3 levels were significantly correlated with circulating 25(OH)D3 levels. No adverse events and no consistently significant changes in serum calcium, creatinine, glucose, parathyroid hormone, vitamin D binding protein, or the urinary calcium/reatinine ratio were observed. However, there was a significant increase in phosphorus after the vitamin D3 intervention. Total cholesterol and triglyceride levels were decreased at the end of the trial. CONCLUSION: The pharmacokinetics of vitamin D after intervention were influenced by baseline 25(OH)D3 levels and the body weight of the subjects. The results suggest that a single high oral vitamin D3 intervention is safe and efficient for improving the vitamin D status of young Chinese people with vitamin D insufficiency.


Asunto(s)
Calcifediol/sangre , Colecalciferol/farmacocinética , Vitaminas/farmacocinética , Administración Oral , Adulto , Factores de Edad , Colecalciferol/administración & dosificación , Femenino , Humanos , Masculino , Factores Sexuales , Factores de Tiempo , Deficiencia de Vitamina D/sangre , Deficiencia de Vitamina D/tratamiento farmacológico , Vitaminas/administración & dosificación
12.
Acta Pharmacol Sin ; 34(8): 1101-7, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23708556

RESUMEN

AIM: To investigate the embryotoxicity of dihydroartemisinin (DHA), the main active metabolite of artemisinin, in zebrafish, and explore the corresponding mechanisms. METHODS: The embryos of wild type and TG (flk1:GFP) transgenic zebrafish were exposed to DHA. Developmental phenotypes of the embryos were observed. Development of blood vessels was directly observed in living embryos of TG (flk1:GFP) transgenic zebrafish under fluorescence microscope. The expression of angiogenesis marker genes vegfa, flk1, and flt1 in the embryos was detected using real-time PCR and RNA in situ hybridization assays. RESULTS: Exposure to DHA (1-10 mg/L) dose-dependently caused abnormal zebrafish embryonic phenotypes in the early developmental stage. Furthermore, exposure to DHA (10 mg/L) resulted in more pronounced embryonic angiogenesis in TG (flk1:GFP) zebrafish line. Exposure to DHA (10 mg/L) significantly increased the mRNA expression of vegfa, flk1, and flt1 in the embryos. Knockdown of the flk1 protein partially blocked the effects of DHA on embryogenesis. CONCLUSION: DHA causes abnormal embryonic phenotypes and promotes angiogenesis in zebrafish early embryonic development, demonstrating the potential embryotoxicity of DHA.


Asunto(s)
Artemisia/toxicidad , Artemisininas/toxicidad , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/embriología , Neovascularización Patológica/inducido químicamente , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente , Embrión no Mamífero/patología , Desarrollo Embrionario/efectos de los fármacos , Desarrollo Embrionario/fisiología , Neovascularización Patológica/patología , Pez Cebra/genética
13.
Curr Cancer Drug Targets ; 13(4): 472-83, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23597199

RESUMEN

Identification of novel chemotherapeutic agents from traditional medicines and elucidation of the molecular basis of their anticancer effects are critical and urgently needed for modern pharmacotherapy. We previously found that analogs of the compounds present in Valeriana jatamansi, a traditional medicine used to treat mental disorders, possess notable antitumor properties; however, the underlying molecular mechanisms have not been fully demonstrated. In this study, we evaluated the anticancer effects of IVHD-valtrate, one of the most active Valeriana jatamansi derivatives, against human ovarian cancer cells in vitro and in vivo. IVHD-valtrate inhibited the growth and proliferation of the A2780 and OVCAR-3 ovarian cancer cell lines in a concentration-dependent manner, while relatively low cytotoxicity to immortalized non-tumorigenic human ovarian surface epithelial cells (IOSE-144) was observed. Treatment with IVHD-valtrate arrested the ovarian cancer cells in the G2/M phase and induced apoptosis, and significantly suppressed the growth of A2780 and OVCAR3 xenograft tumors in a dose-dependent manner. The detailed in vitro and in vivo study on the molecular mechanisms of this compound demonstrated that IVHD-valtrate exposure modulated the expression of numerous molecules involved in cell cycle progression and apoptosis regardless of p53 status, leading to increase the level of p53, Rb, p21, p27 and decrease Mdm2, E2F1, Cyclin B1, Cdc25C and Cdc2. It also down-regulated Bcl-2/Bax and Bcl-2/Bad ratio and enhanced the cleavage of PARP and Caspases. Our preclinical results indicated IVHD-valtrate is a potential therapeutic agent for ovarian cancer, providing a basis for development of the compound as a novel chemotherapeutic agent.


Asunto(s)
Apoptosis/efectos de los fármacos , Biomarcadores de Tumor/metabolismo , Iridoides/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Fitoterapia , Extractos Vegetales/farmacología , Valeriana/química , Animales , Biomarcadores de Tumor/genética , Western Blotting , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Perfilación de la Expresión Génica , Humanos , Técnicas In Vitro , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Estructura Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología
14.
Drug Metab Pharmacokinet ; 26(2): 201-5, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21178302

RESUMEN

Ginseng is one of the most commonly used herbal medicines worldwide. Ginsenosides are believed to be responsible for the therapeutic activities of ginseng; however, co-administration of prescription drugs with ginseng products may give rise to ginseng-drug interactions. Cytochrome P450 enzymes are major phase I enzymes involved in the metabolism of most currently used drugs. Inhibition or induction of P450 enzymes can lead to pharmacokinetic drug interactions. Previous reports on ginseng-drug interactions have been controversial and confusing. In the present study, we examined the effects of thirteen ginsenosides on the expression of CYP1A1, CYP1A2 and CYP3A4 in HepG2 cells. We found that eight ginsenosides and aglycones potently induced CYP1A1 expression, and that structure-activity relationships existed for these effects. Moreover, we discovered that deglycosylated ginsenosides, some of which are putative ginsenoside metabolites, were more potent inducers of CYP1A1, CYP1A2 and CYP3A4 than glycosylated ginsenosides. This finding indicates that ginsenoside metabolites may partially account for ginseng-drug interactions, and that differences in the composition of intestinal bacteria and the extent of deglycosylation of the ginsenosides could be a contributing factor to the inconsistencies observed in previous clinical and pre-clinical studies with regard to ginseng-drug interactions.


Asunto(s)
Citocromo P-450 CYP1A1/efectos de los fármacos , Citocromo P-450 CYP1A2/efectos de los fármacos , Citocromo P-450 CYP3A/efectos de los fármacos , Inducción Enzimática/efectos de los fármacos , Ginsenósidos , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/metabolismo , Citocromo P-450 CYP3A/metabolismo , Ginsenósidos/química , Ginsenósidos/metabolismo , Ginsenósidos/farmacología , Glicosilación , Células Hep G2 , Interacciones de Hierba-Droga , Humanos , Panax/metabolismo , Extractos Vegetales/farmacología , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA