Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 276
Filtrar
1.
Int J Nanomedicine ; 19: 7831-7850, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39105099

RESUMEN

Purpose: Compared with traditional photothermal therapy (PTT, >50°C), mild PTT (≤45°C) is a promising strategy for tumor therapy with fewer adverse effects. Unfortunately, its anti-tumor efficacy is hampered by thermoresistance induced by overexpression of heat shock proteins (HSPs). In our previous study, we found bufalin (BU) is a glycolysis inhibitor that depletes HSPs, which is expected to overcome thermotolerance of tumor cells. In this study, BU-loaded multifunctional nanoparticles (NPs) were developed for enhancing the mild PTT of colorectal cancer (CRC). Methods: Fe3O4 NPs coated with the polydopamine (PDA) shell modified with polyethylene glycol (PEG) and cyclic arginine-glycyl-aspartic peptide (cRGD) for loading BU (Fe3O4@PDA-PEG-cRGD/BU NPs) were developed. The thermal variations in Fe3O4@PDA-PEG-cRGD/BU NPs solution under different conditions were measured. Glycolysis inhibition was evaluated by measuring the glucose uptake, extracellular lactate, and intracellular adenosine triphosphate (ATP) levels. The cellular cytotoxicity of Fe3O4@PDA-PEG-cRGD/BU NPs was analyzed using a cell counting kit-8 assay, Calcein-AM/PI double staining, and flow cytometry in HCT116 cells. The magnetic resonance imaging (MRI) performance and anti-tumor therapeutic efficacy of Fe3O4@PDA-PEG-cRGD/BU NPs were evaluated in HCT116-tumor bearing mice. Results: Fe3O4@PDA-PEG-cRGD/BU NPs had an average diameter of 260.4±3.5 nm, the zeta potential of -23.8±1.6 mV, the drug loading rate of 1.1%, which had good thermal stability, photothermal conversion efficiencies and MRI performance. In addition, the released BU not only killed tumor cells but also interfered with glycolysis by targeting the steroid receptor coactivator 3 (SRC-3)/HIF-1α pathway, preventing intracellular ATP synthesis, and combating HSP-dependent tumor thermoresistance, ultimately strengthening the thermal sensitivity toward mild PTT both in vitro and in vivo. Conclusion: This study provides a highly effective strategy for enhancing the therapeutic effects of mild PTT toward tumors.


Asunto(s)
Bufanólidos , Neoplasias Colorrectales , Glucólisis , Subunidad alfa del Factor 1 Inducible por Hipoxia , Terapia Fototérmica , Animales , Bufanólidos/farmacología , Bufanólidos/química , Bufanólidos/farmacocinética , Humanos , Glucólisis/efectos de los fármacos , Neoplasias Colorrectales/terapia , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/tratamiento farmacológico , Terapia Fototérmica/métodos , Ratones , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Indoles/química , Indoles/farmacología , Polietilenglicoles/química , Polímeros/química , Ratones Endogámicos BALB C , Línea Celular Tumoral , Ratones Desnudos , Células HCT116 , Nanopartículas de Magnetita/química , Nanopartículas/química , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Agric Food Chem ; 72(31): 17377-17391, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39051843

RESUMEN

Bufadienolides (BDs) are a class of naturally occurring toxins present in amphibian toads. Serving as the chemical weapons, they exist not only in the adult toads but also in toad eggs. Guided by mass spectrometry (MS)-based component analysis and feature-based molecular networking (FBMN), 30 bufadienolide-fatty acid conjugates (BDFs) were isolated from the fertilized eggs of toad Bufo gargrizans, including 25 previously undescribed compounds (1-25). Their chemical structures were elucidated by extensive spectroscopic analysis, chemical methods, and GC-MS. The toxicities of all BDFs and their corresponding free BDs were assessed using the zebrafish model. The structure-toxicity relationship analysis showed that the modification of BDs by hydroxy fatty acids can cause a significant increase of the toxicity. Furthermore, all the isolated compounds were evaluated for their antiproliferative activities in pancreatic cancer cell lines ASPC-1 and PANC10.05. The structure-activity relationship (SAR) analysis revealed that BDFs with hellebrigenin as the bufogenin moiety (6 and 7) exhibited the most potent antiproliferative effect. Further investigation into their functional mechanism demonstrated that 6 and 7 induced apoptosis in pancreatic cancer cells PANC10.05 and significantly suppressed the expression of the apoptosis-related gene c-MYC. In addition, 6 and 7 effectively inhibited the expression of the PI3K/Akt/mTOR pathway in PANC10.05. Moreover, we assessed the efficacy of 6 and 7 on cancer cells from various tissues and observed their broad-spectrum antiproliferative activity.


Asunto(s)
Bufanólidos , Bufonidae , Proliferación Celular , Ácidos Grasos , Pez Cebra , Animales , Bufanólidos/química , Bufanólidos/farmacología , Bufanólidos/toxicidad , Bufanólidos/aislamiento & purificación , Proliferación Celular/efectos de los fármacos , Humanos , Línea Celular Tumoral , Ácidos Grasos/química , Ácidos Grasos/farmacología , Ácidos Grasos/toxicidad , Relación Estructura-Actividad , Cigoto/efectos de los fármacos , Cigoto/química , Estructura Molecular
3.
Int J Nanomedicine ; 19: 7273-7305, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39050871

RESUMEN

Dried toad skin (TS) and toad venom (TV) are the dried skin of the Bufo bufo gargarizans Cantor and the Bufo melanostictus Schneider, which remove the internal organs and the white secretions of the skin and retroauricular glands. Since 2005, cinobufacini preparations have been approved by the State Food and Drug Administration for use as adjuvant therapies in the treatment of various advanced cancers. Meanwhile, bufalenolides has been identified as the main component of TS/TV, exhibiting antitumor activity, inducing apoptosis of cancer cells and inhibiting cancer cell proliferation or metastasis through a variety of signaling pathways. However, clinical agents frequently face limitations such as inherent toxicity at high concentrations and insufficient tumor targeting. Additionally, the development and utilization of these active ingredients are hindered by poor water solubility, low bioavailability, and rapid clearance from the bloodstream. To address these challenges, the design of a targeted drug delivery system (TDDS) aims to enhance drug bioavailability, improve targeting within the body, increase drug efficacy, and reduce adverse reactions. This article reviews the TDDS for TS/TV, and their active components, including passive, active, and stimuli-responsive TDDS, to provide a reference for advancing their clinical development and use.


Asunto(s)
Venenos de Anfibios , Bufanólidos , Piel , Animales , Venenos de Anfibios/química , Venenos de Anfibios/farmacología , Venenos de Anfibios/farmacocinética , Humanos , Piel/efectos de los fármacos , Piel/química , Bufanólidos/química , Bufanólidos/farmacología , Bufanólidos/farmacocinética , Bufanólidos/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Bufo bufo , Bufonidae , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/farmacocinética , Neoplasias/tratamiento farmacológico , Disponibilidad Biológica
4.
J Sep Sci ; 47(11): e2400145, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38822225

RESUMEN

In this study, a combination approach involving macroporous resin (MR) column chromatography and gradient countercurrent chromatography (CCC) was employed to enrich and purify bufadienolides from the roots and rhizomes of Helleborus thibetanus Franch. Initially, a D101 MR-packed column chromatography was utilized for fractionation and enrichment of the bufadienolides, which were effectively eluted from the column using a 60% ethanol solution. CCC was subsequently introduced to separate the enriched product using the ethyl acetate/n-butanol/water (EBuWat, 4:1:5, v/v) and EBuWat (5:0:5, v/v) solvent systems in a gradient elution mode. As results, five bufadienolides, including 6.1 mg of hellebrigenin-3-O-ß-D-glucoside (1), 2.2 mg of tigencaoside A (2), 8.3 mg of deglucohellebrin (3), 3.5 mg of 14 ß-hydroxy-3ß-[ß-D-glucopyranosyl-(1→6)-(ß-D-glucopyranosyl)oxy]-5α-bufa-20,22-dienolide (4), and 3.0 mg of 14ß-hydroxy-3ß-[(ß-D-glucopyranosyl)oxy]-5α-bufa-20,22-dienolide (5), were effectively separated from 300 mg of the enriched product. The respective high-performance liquid chromatography purities were as follows: 95.2%, 75.8%, 85.7%, 82.3%, and 92.8%. This study provides valuable insights for the efficient enrichment and separation of bufadienolides from Helleborus thibetanus Franch.


Asunto(s)
Bufanólidos , Distribución en Contracorriente , Helleborus , Distribución en Contracorriente/métodos , Bufanólidos/química , Bufanólidos/aislamiento & purificación , Helleborus/química , Porosidad , Resinas Sintéticas/química , Cromatografía Líquida de Alta Presión , Raíces de Plantas/química
5.
ACS Chem Biol ; 19(5): 1169-1179, 2024 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-38624108

RESUMEN

Bufadienolides are a class of steroids with a distinctive α-pyrone ring at C17, mostly produced by toads and consisting of over 100 orthologues. They exhibit potent cardiotonic and antitumor activities and are active ingredients of the traditional Chinese medicine Chansu and Cinobufacini. Direct extraction from toads is costly, and chemical synthesis is difficult, limiting the accessibility of active bufadienolides with diverse modifications and trace content. In this work, based on the transcriptome and genome analyses, using a yeast-based screening platform, we obtained eight cytochrome P450 (CYP) enzymes from toads, which catalyze the hydroxylation of bufalin and resibufogenin at different sites. Moreover, a reported fungal CYP enzyme Sth10 was found functioning in the modification of bufalin and resibufogenin at multiple sites. A total of 15 bufadienolides were produced and structurally identified, of which six were first discovered. All of the compounds were effective in inhibiting the proliferation of tumor cells, especially 19-hydroxy-bufalin (2) and 1ß-hydroxy-bufalin (3), which were generated from bufalin hydroxylation catalyzed by CYP46A35. The catalytic efficiency of CYP46A35 was improved about six times and its substrate diversity was expanded to progesterone and testosterone, the common precursors for steroid drugs, achieving their efficient and site-specific hydroxylation. These findings elucidate the key modification process in the synthesis of bufadienolides by toads and provide an effective way for the synthesis of unavailable bufadienolides with site-specific modification and active potentials.


Asunto(s)
Bufanólidos , Sistema Enzimático del Citocromo P-450 , Bufanólidos/química , Bufanólidos/metabolismo , Bufanólidos/farmacología , Sistema Enzimático del Citocromo P-450/metabolismo , Animales , Humanos , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/metabolismo , Hidroxilación , Línea Celular Tumoral , Bufonidae/metabolismo , Proliferación Celular/efectos de los fármacos
6.
Drug Des Devel Ther ; 18: 1321-1338, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38681206

RESUMEN

Purpose: Cinobufotalin injection has obvious curative effects on liver cancer patients with less toxicity and fewer side effects than other therapeutic approaches. However, the core ingredients and mechanism underlying these anti-liver cancer effects have not been fully clarified due to its complex composition. Methods: Multidimensional network analysis was used to screen the core ingredients, key targets and pathways underlying the therapeutic effects of cinobufotalin injection on liver cancer, and in vitro and in vivo experiments were performed to confirm the findings. Results: By construction of ingredient networks and integrated analysis, eight core ingredients and ten key targets were finally identified in cinobufotalin injection, and all of the core ingredients are tightly linked with the key targets, and these key targets are highly associated with the cell cycle-related pathways, supporting that both cinobufotalin injection and its core ingredients exert anti-liver cancer roles by blocking cell cycle-related pathways. Moreover, in vitro and in vivo experiments confirmed that either cinobufotalin injection or one of its core ingredients, cinobufagin, significantly inhibited cell proliferation, colony formation, cell cycle progression and xenograft tumor growth, and the key target molecules involved in the cell cycle pathway such as CDK1, CDK4, CCNB1, CHEK1 and CCNE1, exhibit consistent changes in expression after treatment with cinobufotalin injection or cinobufagin. Interestingly, some key targets CDK1, CDK4, PLK1, CHEK1, TTK were predicted to bind with multiple of core ingredients of cinobufotalin injection, and the affinity between one of the critical ingredients cinobufagin and key target CDK1 was further confirmed by SPR assay. Conclusion: Cinobufotalin injection was confirmed to includes eight core ingredients, and they play therapeutic effects in liver cancer by blocking cell cycle-related pathways, which provides important insights for the mechanism of cinobufotalin injection antagonizing liver cancer and the development of novel small molecule anti-cancer drugs.


Asunto(s)
Antineoplásicos , Bufanólidos , Proliferación Celular , Neoplasias Hepáticas , Bufanólidos/farmacología , Bufanólidos/química , Bufanólidos/administración & dosificación , Humanos , Animales , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/metabolismo , Proliferación Celular/efectos de los fármacos , Ratones , Antineoplásicos/farmacología , Antineoplásicos/química , Ensayos de Selección de Medicamentos Antitumorales , Neoplasias Hepáticas Experimentales/tratamiento farmacológico , Neoplasias Hepáticas Experimentales/patología , Neoplasias Hepáticas Experimentales/metabolismo , Ratones Endogámicos BALB C , Ciclo Celular/efectos de los fármacos , Ratones Desnudos , Relación Dosis-Respuesta a Droga , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Neoplasias Experimentales/metabolismo , Células Tumorales Cultivadas , Relación Estructura-Actividad , Estructura Molecular , Inyecciones
7.
J Liposome Res ; 34(3): 489-506, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38269490

RESUMEN

Breast cancer stem cells (BCSCs) play a key role in therapeutic resistance in breast cancer treatments and disease recurrence. This study aimed to develop a combination therapy loaded with pH-sensitive liposomes to kill both BCSCs and the okbulk cancer cells using trastuzumab-sensitive and resistant human epidermal growth factor receptor 2 positive (HER2+) breast cancer cell models. The anti-BCSCs effect and cytotoxicity of all-trans retinoic acid, salinomycin, and bufalin alone or in combination with doxorubicin were compared in HER2+ cell line BT-474 and a validated trastuzumab-resistant cell line, BT-474R. The most potent anti-BCSC agent was selected and loaded into a pH-sensitive liposome system. The effects of the liposomal combination on BCSCs and bulk cancer cells were assessed. Compared with BT-474, the aldehyde dehydrogenase positive BCSC population was elevated in BT-474R (3.9 vs. 23.1%). Bufalin was the most potent agent and suppressed tumorigenesis of BCSCs by ∼50%, and showed strong synergism with doxorubicin in both BT-474 and BT-474R cell lines. The liposomal combination of bufalin and doxorubicin significantly reduced the BCSC population size by 85%, and inhibited both tumorigenesis and self-renewal, although it had little effect on the migration and invasiveness. The cytotoxicity against the bulk cancer cells was also enhanced by the liposomal combination than either formulation alone in both cell lines (p < 0.001). The liposomal bufalin and doxorubicin combination therapy may effectively target both BCSCs and bulk cancer cells for a better outcome in trastuzumab-resistant HER2+ breast cancer.


Asunto(s)
Neoplasias de la Mama , Bufanólidos , Doxorrubicina , Resistencia a Antineoplásicos , Liposomas , Células Madre Neoplásicas , Trastuzumab , Humanos , Doxorrubicina/farmacología , Doxorrubicina/administración & dosificación , Bufanólidos/farmacología , Bufanólidos/administración & dosificación , Bufanólidos/química , Células Madre Neoplásicas/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Liposomas/química , Femenino , Trastuzumab/farmacología , Trastuzumab/administración & dosificación , Línea Celular Tumoral , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Receptor ErbB-2/metabolismo , Supervivencia Celular/efectos de los fármacos
8.
Toxicon ; 238: 107591, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38160738

RESUMEN

Bufadienolides are digitalis-like aglycones mainly found in skin secretions of toads. Among their biological properties, the mechanisms of antiproliferative action on tumor cells remain unclear for many compounds, including against leukemia cells. Herein, it was evaluated the mechanisms involved in the antiproliferative and genotoxic actions of hellebrigenin on tumor cell lines and in silico capacity to inhibit the human topoisomerase IIa enzyme. Firstly, its cytotoxic action was investigated by colorimetric assays in human tumor and peripheral blood mononuclear cells (PBMC). Next, biochemical and morphological studies were detailed by light microscopy (trypan blue dye exclusion), immunocytochemistry (BrdU uptake), flow cytometry and DNA/chromosomal damages (Cometa and aberrations). Finally, computational modelling was used to search for topoisomerase inhibition. Hellebrigenin reduced proliferation, BrdU incorporation, viability, and membrane integrity of HL-60 leukemia cells. Additionally, it increased G2/M arrest, internucleosomal DNA fragmentation, mitochondrial depolarization, and phosphatidylserine externalization in a concentration-dependent manner. In contrast to doxorubicin, hellebrigenin did not cause DNA strand breaks in HL-60 cell line and lymphocytes, and it interacts with ATPase domain residues of human topoisomerase IIa, generating a complex of hydrophobic and van der Waals interactions and hydrogen bonds. So, hellebrigenin presented potent anti-leukemic activity at concentrations as low as 0.06 µM, a value comparable to the clinical anticancer agent doxorubicin, and caused biochemical changes suggestive of apoptosis without genotoxic/clastogenic-related action, but it probably triggers catalytic inhibition of topoisomerase II. These findings also emphasize toad steroid toxins as promising lead antineoplasic compounds with relatively low cytotoxic action on human normal cells.


Asunto(s)
Antineoplásicos , Bufanólidos , Leucemia , Humanos , Leucocitos Mononucleares , Bromodesoxiuridina/farmacología , Daño del ADN , Antineoplásicos/farmacología , Bufanólidos/química , Células HL-60 , Apoptosis , ADN/farmacología , Doxorrubicina/farmacología
9.
Drug Dev Res ; 84(5): 815-838, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37154099

RESUMEN

Bufadienolides, naturally found in toad venoms having steroid-like structures, reveal antiproliferative effects at low doses. However, their application as anticancer drugs is strongly prevented by their Na+ /K+ -ATPase binding activities. Although several kinds of research were dedicated to moderating their Na+ /K+ -ATPase binding activity, still deeper fundamental knowledge is required to bring these findings into medical practice. In this work, we reviewed data related to anticancer activity of bufadienolides such as bufalin, arenobufagin, bufotalin, gamabufotalin, cinobufotalin, and cinobufagin and their derivatives. Bufotoxins, derivatives of bufadienolides containing polar molecules mainly belonging to argininyl residues, are reviewed as well. The established structures of bufotoxins have been compiled into a one-page figure to review their structures. We also highlighted advances in the structure-modification of the structure of compounds in this class. Drug delivery approaches to target these compounds to tumor cells were discussed in one section. The issues related to extraction, identification, and quantification are separated into another section.


Asunto(s)
Venenos de Anfibios , Antineoplásicos , Bufanólidos , Bufanólidos/farmacología , Bufanólidos/química , Bufanólidos/metabolismo , Antineoplásicos/farmacología , Venenos de Anfibios/farmacología , Venenos de Anfibios/química , Adenosina Trifosfatasas
10.
Toxicol In Vitro ; 89: 105566, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36738868

RESUMEN

A series of bufadienolides were isolated from the Bufo viridis toad venom, and their cytotoxic activities against three human cancer cell lines (HeLa, HT-29, MCF7) and a non-cancer cell line (L-O2) were explored using the MTT assay in vitro. All of nine compounds exhibited cytotoxic activities against the three cancer cell lines, with compound D4 exhibiting potent cytotoxic activity against HeLa cells and was better than positive control. Herein, we further evaluated the effect of compound D4 on HeLa cells. The results revealed that compound D4 has excellent cytotoxic effect on HeLa cells by inhibiting cell colony formation and migration, promoting cell apoptosis, increasing reactive oxygen species (ROS) levels and arresting of HeLa cells in S and G2/M phases. These findings encourage further work on the chemistry and bioactivity of the Bufo viridis toad venom.


Asunto(s)
Venenos de Anfibios , Antineoplásicos , Bufanólidos , Neoplasias , Animales , Humanos , Células HeLa , Línea Celular Tumoral , Bufanólidos/toxicidad , Bufanólidos/química , Venenos de Anfibios/farmacología , Venenos de Anfibios/química , Bufonidae , Antineoplásicos/farmacología , Puntos de Control del Ciclo Celular , Apoptosis
11.
Chem Biodivers ; 20(2): e202200911, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36627123

RESUMEN

Arenobufagin, one of the bufadienolides isolated from traditional Chinese medicine Chan'su, exhibits potent antitumor activity. However, serious toxicity and small therapeutic window limits its drug development. In the present study, to our knowledge, novel 3,11-bispeptide ester arenobufagin derivatives have been firstly designed and synthesized on the base of our previous discovery of active 3-monopeptide ester derivative. The in vitro antiproliferative activity evaluation revealed that the moiety at C3 and C11 hydroxy had an important influence on cytotoxic activity and selectivity. Compound ZM350 notably inhibited tumor growth by 58.8 % at a dose 10 mg/kg in an A549 nude mice xenograft model. Therefore, compound ZM350 also presented a concentration-dependent apoptosis induction and low inhibitory effect against both hERG potassium channel and Cav1.2 calcium channel. Our study suggests that novel 3,11-bispeptide ester derivatives will be a potential benefit to further antitumor agent development of arenobufagin.


Asunto(s)
Antineoplásicos , Bufanólidos , Animales , Ratones , Humanos , Línea Celular Tumoral , Cardiotoxicidad/tratamiento farmacológico , Ratones Desnudos , Antineoplásicos/farmacología , Bufanólidos/química , Apoptosis , Ensayos de Selección de Medicamentos Antitumorales , Proliferación Celular
12.
Molecules ; 27(19)2022 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-36235123

RESUMEN

Cardiotonic steroids (CTS) were first documented by ancient Egyptians more than 3000 years ago. Cardiotonic steroids are a group of steroid hormones that circulate in the blood of amphibians and toads and can also be extracted from natural products such as plants, herbs, and marines. It is well known that cardiotonic steroids reveal effects against congestive heart failure and atrial fibrillation; therefore, the term "cardiotonic" has been coined. Cardiotonic steroids are divided into two distinct groups: cardenolides (plant-derived) and bufadienolides (mainly of animal origin). Cardenolides have an unsaturated five-membered lactone ring attached to the steroid nucleus at position 17; bufadienolides have a doubly unsaturated six-membered lactone ring. Cancer is a leading cause of mortality in humans all over the world. In 2040, the global cancer load is expected to be 28.4 million cases, which would be a 47% increase from 2020. Moreover, viruses and inflammations also have a very nebative impact on human health and lead to mortality. In the current review, we focus on the chemistry, antiviral and anti-cancer activities of cardiotonic steroids from the naturally derived (toads) venom to combat these chronic devastating health problems. The databases of different research engines (Google Scholar, PubMed, Science Direct, and Sci-Finder) were screened using different combinations of the following terms: "cardiotonic steroids", "anti-inflammatory", "antiviral", "anticancer", "toad venom", "bufadienolides", and "poison chemical composition". Various cardiotonic steroids were isolated from diverse toad species and exhibited superior anti-inflammatory, anticancer, and antiviral activities in in vivo and in vitro models such as marinobufagenin, gammabufotalin, resibufogenin, and bufalin. These steroids are especially difficult to identify. However, several compounds and their bioactivities were identified by using different molecular and biotechnological techniques. Biotechnology is a new tool to fully or partially generate upscaled quantities of natural products, which are otherwise only available at trace amounts in organisms.


Asunto(s)
Productos Biológicos , Bufanólidos , Glicósidos Cardíacos , Venenos , Animales , Antivirales , Bufanólidos/química , Bufonidae , Cardenólidos/química , Glicósidos Cardíacos/farmacología , Hormonas , Humanos , Lactonas
13.
Chembiochem ; 23(22): e202200473, 2022 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-36125775

RESUMEN

Bufadienolides are toxic components widely found in amphibious toads that exhibit a wide range of biological activities. Guided by UPLC-QTOF-MS analysis, several 3-epi-bufadienolides with unique structures were isolated from the bile of the Asiatic toad, Bufo gargarizans. However, the enzymatic machinery of this epimerization in toads and its significance in chemical ecology remains poorly understood. Herein, we firstly compared the toxicities of two typical bufadienolides, bufalin (featuring a 14ß-hydroxyl) and resibufogenin (containing a 14, 15-epoxy group), with their corresponding 3-epi isomers in a zebrafish model. The results of the toxicology assays showed that the ratio of maximum non-toxic concentrations of these two pairs of compounds are 256 and 96 times, respectively, thereby indicating that 3-hydroxyl epimerization leads to a significant decrease in toxicity. Aiming to investigate the biotransformation of 3-epi bufadienolides in toads, we applied liver lysate to transform bufalin and found that it could stereoselectively catalyze the conversion of bufalin into its 3α-hydroxyl epimer. Following this, we cloned and characterized a short-chain dehydrogenase/reductase, HSE-1, from the toad liver cDNA library and verified its 3(ß→α)-hydroxysteroid epimerization activity. To the best of our knowledge, this is the first hydroxyl epimerase identified from amphibians that regulates the toxicity of animal-derived natural products.


Asunto(s)
Bufanólidos , Deshidrogenasas-Reductasas de Cadena Corta , Animales , Bufo bufo/metabolismo , Deshidrogenasas-Reductasas de Cadena Corta/metabolismo , Pez Cebra , Bufanólidos/toxicidad , Bufanólidos/química , Bufanólidos/metabolismo , Catálisis
14.
Phytochemistry ; 198: 113153, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35240134

RESUMEN

From the leaves of Kenyan medicinal plant Bersama abyssinica Subspecies abyssinica, four previously undescribed compounds namely, three bufadienolides, 10ß-formylpaulliniogenin B, 10ß-formylpaulliniogenin A and 1ß-acetoxy-3ß,5ß-dihydroxy-15-methoxy-16,19-dioxobufa-14(15),20,22-trienolide, and a phenolic compound 2,6,4'-trihydroxybenzophenone-4-O-(6‴-cinnamoyl)-ß-D-glucoside were isolated together with four known compounds. The structural elucidation of the compounds was based on 1D and 2D NMR spectroscopy and HRMS data analyses. The relative configurations were defined by NOESY correlations. Cytotoxic activities on L929 and KB3.1 cell lines of the isolated compounds were investigated using MTT assay. The 1ß-acetoxy-3ß,5ß-dihydroxy-15-methoxy-16,19-dioxobufa-14(15),20,22-trienolide showed significant cytotoxic activity against KB3.1 cell lines with IC50 of 3.9 ± 0.99 µM.


Asunto(s)
Antineoplásicos , Bufanólidos , Magnoliopsida , Plantas Medicinales , Bufanólidos/análisis , Bufanólidos/química , Línea Celular Tumoral , Kenia , Magnoliopsida/química , Hojas de la Planta/química
15.
Comput Math Methods Med ; 2022: 3197402, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35069780

RESUMEN

OBJECTIVE: To explore the active compounds and targets of cinobufotalin (huachansu) compared with the osteosarcoma genes to obtain the potential therapeutic targets and pharmacological mechanisms of action of cinobufotalin on osteosarcoma through network pharmacology. METHODS: The composition of cinobufotalin was searched by literature retrieval, and the target was selected from the CTD and TCMSP databases. The osteosarcoma genes, found from the GeneCards, OMIM, and other databases, were compared with the cinobufotalin targets to obtain potential therapeutic targets. The protein-protein interaction (PPI) network of potential therapeutic targets, constructed through the STRING database, was inputted into Cytoscape software to calculate the hub genes, using the NetworkAnalyzer. The hub genes were inputted into the Kaplan-Meier Plotter online database for exploring the survival curve. Functional enrichment analysis was identified using the DAVID database. RESULTS: 28 main active compounds of cinobufotalin were explored, including bufalin, adenosine, oleic acid, and cinobufagin. 128 potential therapeutic targets on osteosarcoma are confirmed among 184 therapeutic targets form cinobufotalin. The hub genes included TP53, ACTB, AKT1, MYC, CASP3, JUN, TNF, VEGFA, HSP90AA1, and STAT3. Among the hub genes, TP53, ACTB, MYC, TNF, VEGFA, and STAT3 affect the patient survival prognosis of sarcoma. Through function enrichment analysis, it is found that the main mechanisms of cinobufotalin on osteosarcoma include promoting sarcoma apoptosis, regulating the cell cycle, and inhibiting proliferation and differentiation. CONCLUSION: The possible mechanisms of cinobufotalin against osteosarcoma are preliminarily predicted through network pharmacology, and further experiments are needed to prove these predictions.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Óseas/tratamiento farmacológico , Bufanólidos/farmacología , Osteosarcoma/tratamiento farmacológico , Antineoplásicos/química , Biomarcadores de Tumor/genética , Neoplasias Óseas/genética , Bufanólidos/química , Biología Computacional , Bases de Datos de Compuestos Químicos , Bases de Datos Farmacéuticas , Redes Reguladoras de Genes/efectos de los fármacos , Humanos , Medicina Tradicional China , Farmacología en Red , Osteosarcoma/genética , Mapas de Interacción de Proteínas/efectos de los fármacos , Mapas de Interacción de Proteínas/genética
16.
J Biochem Mol Toxicol ; 35(10): e22862, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34309954

RESUMEN

Cinobufagin, a bufadienolide of toad venom of Bufo bufo gargarizans, is used as a cardiotonic, central nervous system (CNS) respiratory agent, as well as an analgesic and anesthetic. However, several research showed that bufadienolide has a few side effects on the CNS, such as breathlessness or coma. Although cinobufagin was shown to display pharmacological effects in various models, the toxic effect of cinobufagin is elusive in brain cell models. The aim of this study was to explore whether cinobufagin affected viability, Ca2+ homeostasis, and reactive oxygen species (ROS) production in Gibco® Human Astrocyte (GHA) and HCN-2 neuronal cell line. In GHA cells but not in HCN-2 cells, cinobufagin (20-60 µM) induced [Ca2+ ]i rises. In terms of cell viability, chelation of cytosolic Ca2+ with 1,2-bis(2-aminophenoxy)ethane-N,N,N'N'-tetraacetic acid reduced cinobufagin-induced cytotoxicity in GHA cells. In GHA cells, cinobufagin-induced Ca2+ entry was inhibited by 2-aminoethoxydiphenyl borate or SKF96365. In a Ca2+ -free medium, treatment with thapsigargin or U73122 abolished cinobufagin-evoked [Ca2+ ]i rises. Furthermore, treatment with N-acetylcysteine reversed ROS production and cytotoxicity in cinobufagin-treated GHA cells. Together, in GHA cells but not in HCN-2 cells cinobufagin caused cytotoxicity that was linked to preceding [Ca2+ ]i rises by Ca2+ influx via store-operated Ca2+ entry and phospholipase C-dependent Ca2+ release from the endoplasmic reticulum. Moreover, cinobufagin induced ROS-associated cytotoxicity.


Asunto(s)
Venenos de Anfibios/química , Astrocitos/metabolismo , Encéfalo/metabolismo , Bufanólidos/farmacología , Señalización del Calcio/efectos de los fármacos , Homeostasis/efectos de los fármacos , Neuronas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Astrocitos/efectos de los fármacos , Encéfalo/patología , Bufanólidos/química , Bufanólidos/aislamiento & purificación , Bufo bufo , Calcio/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Citosol/metabolismo , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Humanos , Neuronas/efectos de los fármacos , Tapsigargina/farmacología , Fosfolipasas de Tipo C/metabolismo
17.
Int J Nanomedicine ; 16: 3581-3598, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34079251

RESUMEN

BACKGROUND: BF211, a derivative of bufalin (BF), shows significantly improved solubility and potent antitumor efficiency compared to BF. Unfortunately, the unwanted toxicity such as cardiotoxicity caused by unspecific distribution has hindered its clinical use. METHODS: PEGylated BF211 liposomes (BF211@Lipo) were designed and optimizely prepared based on the pre-prescription research. In vitro and in vivo cardiotoxicity was evaluated. In vivo pharmacokinetics and biodistribution of BF211@Lipo were investigated. In vivo antitumor activity and toxicity were evaluated in HepG2 cell xenograft models. The rapid-release triggered by Poloxamer 188 (P188) was assessed in vitro and in vivo. RESULTS: The optimized BF211@Lipo displayed a spherical morphology with a size of (164.6 ± 10.3) nm and a high encapsulation efficiency of (93.24 ± 2.15) %. The in vivo concentration-time curves of BF211 loaded in liposomes showed a prolonged half-life in plasma and increased tumor accumulation. No obvious abnormality in electrocardiograms was observed in guinea pigs even at 9 mg/kg. Moreover, to improve the efficient release of BF211@Lipo, a surfactant-assisted rapid-release strategy was developed, and the release-promoting mechanism was revealed by the fluorescence resonance energy transfer (FRET) and fluorescence nanoparticle tracking analysis (fl-NTA) technology. Sequential injection of BF211@Lipo and P188 could ignite the "cold" liposomes locally in tumor regions, facilitating the burst release of BF211 and enhancing the therapeutic index. CONCLUSION: Our progressive efforts that begin with preparation technology and dosage regimen enable BF211 to like a drug, providing a promising nano platform to deliver the cardiac glycosides and alleviate the side effects by decreasing unspecific biodistribution.


Asunto(s)
Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Bufanólidos/administración & dosificación , Bufanólidos/farmacología , Corazón/efectos de los fármacos , Tensoactivos/química , Animales , Antineoplásicos/química , Antineoplásicos/toxicidad , Bufanólidos/química , Bufanólidos/toxicidad , Cobayas , Células Hep G2 , Humanos , Liposomas , Nanopartículas/química , Poloxámero/química , Solubilidad , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Sci Rep ; 11(1): 12527, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34131171

RESUMEN

Chemical cues produced by late-stage embryos of the cane toad (Rhinella marina) attract older conspecific larvae, which are highly cannibalistic and can consume an entire clutch. To clarify the molecular basis of this attraction response, we presented captive tadpoles with components present in toad eggs. As previously reported, attractivity arises from the distinctive toxins (bufadienolides) produced by cane toads, with some toxins (e.g., bufagenins) much stronger attractants than others (e.g., bufotoxins). Extracts of frozen toad parotoid glands (rich in bufagenins) were more attractive than were fresh MeOH extracts of the parotoid secretion (rich in bufotoxins), and purified marinobufagin was more effective than marinobufotoxin. Cardenolide aglycones (e.g., digitoxigenin) were active attractors, whereas C-3 glycosides (e.g., digoxin, oubain) were far less effective. A structure-activity relationship study revealed that tadpole attractant potency strongly correlated with Na+/K+ ATPase inhibitory activity, suggesting that tadpoles monitor and rapidly react to perturbations to Na+/K+ ATPase activity.


Asunto(s)
Bufanólidos/química , Bufo marinus/fisiología , Canibalismo , Toxinas Biológicas/química , Animales , Larva/efectos de los fármacos , Larva/fisiología
19.
J Mater Chem B ; 9(18): 3838-3855, 2021 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-33908580

RESUMEN

Although drug combination has proved to be an efficient strategy for clinic gastric cancer therapy, how to further improve their bioavailability and reduce the side effects are still challenges due to the low solubility and untargeted ability of drugs. Recently, newly emerging nanotechnology has provided an alternative for constructing new drug delivery systems with high targeting ability and solubility. In this study, a pH-responsive liposome (Liposome-PEO, LP) loaded with apatinib (AP) and cinobufagin (CS-1) was used for combinational therapy against gastric cancer after coating with a hybrid membrane (R/C). The results indicated that the constructed nanocomplex LP-R/C@AC not only efficiently killed tumor cells in vitro by inducing apoptosis, autophagy, and pyroptosis, but also significantly inhibited tumor invasion and metastasis via the VEGFR2/STAT3 pathway. Moreover, it showed stronger anti-tumor activity in gastric cancer-bearing mouse models, as compared to the sole drugs. A naturally-derived hybrid cell membrane coating bestowed nanocomplexes with enhanced biointerfacing including prolonged circulation time and targeting ability.


Asunto(s)
Antineoplásicos/farmacología , Liposomas/química , Nanopartículas/química , Transducción de Señal/efectos de los fármacos , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Materiales Biocompatibles/química , Bufanólidos/química , Bufanólidos/farmacología , Bufanólidos/uso terapéutico , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Humanos , Ratones , Ratones Desnudos , Nanopartículas/metabolismo , Piridinas/química , Piridinas/farmacología , Piridinas/uso terapéutico , Factor de Transcripción STAT3/metabolismo , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/patología , Distribución Tisular , Trasplante Heterólogo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
20.
Int J Biol Macromol ; 175: 67-78, 2021 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-33548318

RESUMEN

Bufadienolides are the main active ingredients of Venenum Bufonis, which is a widely used traditional Chinese medicine secreted from parotoid gland and skin glands of Bufo bufo gargarizans. According to the transcriptome analysis, "cholesterol-bile acid-bufadienolidies pathway" was proposed as animal-derived bufadienolides biosynthesis pathway by us previously. In this pathway 3ß-hydroxysteroid dehydrogenase (3ßHSD) and steroid 5ß-reductase (SRD5ß) might be the key enzymes to convert the A/B ring to cis-configuration. Therefore, as the second report of our group, here we report the cloning of the full length of SRD5ß cDNA of B. bufo gargarizans (Bbg-SRD5ß) from the parotoid gland of B. bufo gargarizans for the first time, and site-directed mutagenesis was used to explored the character of Bbg-SRD5ß. Bbg-SRD5ß had an open reading frame of 981 bp and encoded 326 amino acids residues. The expression conditions of the recombinant Bbg-SRD5ß in E. coli BL21 (DE3) harbored with pCold-Bbg-SRD5ß was optimized as induction for 10 h at 15 °C with 0.1 mM IPTG. With NADPH as a cofactor, Bbg-SRD5ß can reduce the Δ4,5 double bonds of progesterone to generate dihydroprogesterone õwithout substrate inhibition effect. The catalytic rate of mutant type Bbg-SRD5ß-Y132G was 1.8 times higher than that of wild type Bbg-SRD5ß. Although Bbg-SRD5ß was almost unable to reduce the progesterone to dihydroprogesterone after mutation of V309, the affinity of enzyme with NADPH changed significantly. Bbg-SRD5ß is the key enzymes to convert the A/B ring of steroid to cis-configuration, and V309 is a key site affecting the binding affinity of enzyme with NADPH, and the mutation of Y132 can adjust the catalytic rate of Bbg-SRD5ß.


Asunto(s)
Venenos de Anfibios/química , Bufo bufo/metabolismo , Oxidorreductasas/aislamiento & purificación , Secuencia de Aminoácidos , Venenos de Anfibios/metabolismo , Animales , Bufanólidos/química , Bufanólidos/metabolismo , Bufonidae/metabolismo , Clonación Molecular/métodos , ADN Complementario/metabolismo , Sistemas de Lectura Abierta , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Esteroides/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA