Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 328, 2024 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-38184609

RESUMEN

Membrane-camouflaged nanomedicines often suffer from reduced efficacy caused by membrane protein disintegration and spatial disorder caused by separation and reassembly of membrane fragments during the coating process. Here we show that intracellularly gelated macrophages (GMs) preserve cell membrane structures, including protein content, integration and fluidity, as well as the membrane lipid order. Consequently, in our testing GMs act as cellular sponges to efficiently neutralize various inflammatory cytokines via receptor-ligand interactions, and serve as immune cell-like carriers to selectively bind inflammatory cells in culture medium, even under a flow condition. In a rat model of collagen-induced arthritis, GMs alleviate the joint injury, and suppress the overall arthritis severity. Upon intravenous injection, GMs efficiently accumulate in the inflammatory lungs of acute pneumonia mice for anti-inflammatory therapy. Conveniently, GMs are amenable to lyophilization and can be stored at ambient temperatures for at least 1 month without loss of integrity and bio-activity. This intracellular gelation technology provides a universal platform for targeted inflammation neutralization treatment.


Asunto(s)
Artritis Experimental , Ratas , Ratones , Animales , Artritis Experimental/tratamiento farmacológico , Medios de Cultivo , Citocinas , Liofilización , Macrófagos
2.
Bio Protoc ; 13(12): e4695, 2023 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-37397790

RESUMEN

Cell-based carrier exhibits inherent advantages as the next generation of drug delivery system, namely high biocompatibility and physiological function. Current cell-based carriers are constructed via direct payload internalization or conjugation between cell and payload. However, the cells involved in these strategies must be firstly extracted from the body and the cell-based carrier must be prepared in vitro. Herein, we synthesize bacteria-mimetic gold nanoparticles (GNPs) for the construction of cell-based carrier in mice. Both ß-cyclodextrin (ß-CD)-modified GNPs and adamantane (ADA)-modified GNPs are coated by E. coli outer membrane vesicles (OMVs). The E. coli OMVs induce the phagocytosis of GNPs by circulating immune cells, leading to intracellular degradation of OMVs and subsequent supramolecular self-assembly of GNPs driven by ß-CD-ADA host-guest interactions. In vivo construction of cell-based carrier based on bacteria-mimetic GNPs avoids the immunogenicity induced by allogeneic cells and restriction by the number of separated cells. Due to the inflammatory tropism, endogenous immune cells carry the intracellular GNP aggregates to the tumor tissues in vivo. Graphical overview Collect the outer membrane vesicles (OMVs) of E. coli by gradient centrifugation (a) and coat on gold nanoparticles (GNP) surface (b) to prepare OMV-coated cyclodextrin (CD)-GNPs and OMV-coated adamantane (ADA)-GNPs (c) via ultrasonic method.

3.
Adv Mater ; 35(20): e2211626, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36905923

RESUMEN

Immune cells exhibit great potential as carriers of nanomedicine, attributed to their high tolerance to internalized nanomaterials and targeted accumulation in inflammatory tissues. However, the premature efflux of internalized nanomedicine during systemic delivery and slow infiltration into inflammatory tissues have limited their translational applications. Herein, a motorized cell platform as a nanomedicine carrier for highly efficient accumulation and infiltration in the inflammatory lungs and effective treatment of acute pneumonia are reported. ß-Cyclodextrin and adamantane respectively modified manganese dioxide nanoparticles are intracellularly self-assembled into large aggregates mediated via host-guest interactions, to effectively inhibit the efflux of nanoparticles, catalytically consume/deplete H2 O2 to alleviate inflammation, and generate O2 to propel macrophage movement for rapid tissue infiltration. With curcumin loaded into MnO2 nanoparticles, macrophages carry the intracellular nano-assemblies rapidly into the inflammatory lungs via chemotaxis-guided, self-propelled movement, for effective treatment of acute pneumonia via immunoregulation induced by curcumin and the aggregates.


Asunto(s)
Curcumina , Neumonía , Curcumina/farmacología , Curcumina/uso terapéutico , Nanopartículas , Neumonía/tratamiento farmacológico , Quimiotaxis , Macrófagos
4.
J Control Release ; 349: 2-15, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35779655

RESUMEN

Current pharmacological treatments of atherosclerosis often target either cholesterol control or inflammation management, to inhibit atherosclerotic progression, but cannot lead to direct plaque lysis and atherosclerotic regression, partly due to the poor accumulation of medicine in the atherosclerotic plaques. Due to enhanced macrophage recruitment during atheromatous plaque progression, a macrophage-liposome conjugate was facilely constructed for targeted anti-atherosclerosis therapy via synergistic plaque lysis and inflammation alleviation. Endogenous macrophage is utilized as drug-transporting cell, upon membrane-modification with a ß-cyclodextrin (ß-CD) derivative to form ß-CD decorated macrophage (CD-MP). Adamantane (ADA) modified quercetin (QT)-loaded liposome (QT-NP), can be conjugated to CD-MP via host-guest interactions between ß-CD and ADA to form macrophage-liposome conjugate (MP-QT-NP). Thus, macrophage carries liposome "hand-in-hand" to significantly increase the accumulation of anchored QT-NP in the aorta plaque in response to the plaque inflammation. In addition to anti-inflammation effects of QT, MP-QT-NP efficiently regresses atherosclerotic plaques from both murine aorta and human carotid arteries via CD-MP mediated cholesterol efflux, due to the binding of cholesterol by excess membrane ß-CD. Transcriptome analysis of atherosclerotic murine aorta and human carotid tissues reveal that MP-QT-NP may activate NRF2 pathway to inhibit plaque inflammation, and simultaneously upregulate liver X receptor to promote cholesterol efflux.


Asunto(s)
Adamantano , Aterosclerosis , Ciclodextrinas , Placa Aterosclerótica , beta-Ciclodextrinas , Adamantano/metabolismo , Adamantano/farmacología , Adamantano/uso terapéutico , Animales , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/metabolismo , Colesterol/metabolismo , Ciclodextrinas/farmacología , Humanos , Inflamación/metabolismo , Liposomas/metabolismo , Receptores X del Hígado , Macrófagos , Ratones , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/farmacología , Factor 2 Relacionado con NF-E2/uso terapéutico , Placa Aterosclerótica/tratamiento farmacológico , Placa Aterosclerótica/metabolismo , Quercetina/farmacología , Quercetina/uso terapéutico , beta-Ciclodextrinas/uso terapéutico
5.
Sci Adv ; 8(19): eabn1805, 2022 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-35544569

RESUMEN

Cell-based drug carriers are mostly prepared in vitro, which may negatively affect the physiological functions of cells, and induce possible immune rejections when applied to different individuals. In addition, the immunosuppressive tumor microenvironment limits immune cell-mediated delivery. Here, we report an in vivo strategy to construct cell-based nanomedicine carriers, where bacteria-mimetic gold nanoparticles (GNPs) are intravenously injected, selectively phagocytosed by phagocytic immune cells, and subsequently self-assemble into sizable intracellular aggregates via host-guest interactions. The intracellular aggregates minimize exocytosis of GNPs from immune cells and activate the photothermal property via plasmonic coupling effects. Phagocytic immune cells carry the intracellular GNP aggregates to melanoma tissue via inflammatory tropism. Moreover, an initial photothermal treatment (PTT) of the tumor induces tumor damage that subsequently provides positive feedback to recruit more immune cell-based carriers for enhanced targeting efficiency. The optimized secondary PTT notably improves antitumor immunotherapy, further strengthened by immune checkpoint blockade.


Asunto(s)
Melanoma , Nanopartículas del Metal , Nanopartículas , Neoplasias , Bacterias , Línea Celular Tumoral , Oro , Humanos , Melanoma/tratamiento farmacológico , Nanomedicina , Microambiente Tumoral
6.
Chin Chem Lett ; 32(10): 3019-3022, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33840982

RESUMEN

The wide-spreading SARS-CoV-2 virus has put the world into boiling water for more than a year, however pharmacological therapies to act effectively against coronavirus disease 2019 (COVID-19) remain elusive. Chloroquine (CQ), an antimalarial drug, was found to exhibit promising antiviral activity in vitro and in vivo at a high dosage, thus CQ was approved by the FDA for the emergency use authorization (EUA) in the fight against COVID-19 in the US, but later was revoked the EUA status due to the severe clinical toxicity. Herein, we show that supramolecular formulation of CQ by a macrocyclic host, curcurbit[7]uril (CB[7]), reduced its non-specific toxicity and improved its antiviral activity against coronavirus, working in synergy with CB[7]. CB[7] was found to form 1:1 host-guest complexes with CQ, with a binding constant of ∼104 L/mol. The CQ-CB[7] formulation decreased the cytotoxicity of CQ against Vero E6 and L-02 cell lines. In particular, the cytotoxicity of CQ (60 µmol/L) against both Vero E6 cell line and L-02 cell lines was completely inhibited in the presence of 300 µmol/L and 600 µmol/L CB[7], respectively. Furthermore, the CB[7] alone showed astonishing antiviral activity in SARS-CoV-2 infected Vero E6 cells and mouse hepatitis virus strain A59 (MHV-A59) infected N2A cells, and synergistically improved the antiviral activity of CQ-CB[7], suggesting that CB[7]-based CQ formulation has a great potential as a safe and effective antiviral agent against SARS-CoV-2 and other coronavirus.

7.
Biomed Pharmacother ; 132: 110749, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33017766

RESUMEN

BACKGROUND: Clofazimine (CFZ), a riminophenazine, is now commonly used in the treatment of multidrug-resistant tuberculosis. However, its use may be potentially associated with cardiac dysfunction in some individuals. In this study, the zebrafish heart, by merit of its developmental and genetic characteristics being in homology with that of human, was chosen as an animal model for evaluation of such dysfunction. METHODS: Morphological and physiological parameters were used to assess cardiac dysfunction. Transcriptome analysis was performed, followed by validation with real-time quantitative PCR, for delineation of the relevant genomics. RESULTS: Exposure of 2 dpf zebrafish to 4 mg/L CFZ for 2 days, adversely affected cardiac functions including significant decreases in HR, SV, CO, and FS, with observable pathophysiological developments of pericardial effusion and blood accumulation in the heart, in comparison with the control group. In addition, genes which respond to xenobiotic stimulus, related to oxygen transport, glutathione metabolism and extracellular matrix -receptor interactions, were significantly enriched among the differentially up-regulated genes. Antioxidant response element motif was enriched in the 5000 base pair upstream regions of the differentially expressed genes. Co-administration of N-acetylcysteine was shown to protect zebrafish against the development of CFZ-induced cardiac dysfunction. CONCLUSIONS: This study suggests an important role of oxidative stress as a major pathogenetic mechanism of riminophenazine-induced cardiac dysfunction.


Asunto(s)
Antituberculosos/toxicidad , Clofazimina/toxicidad , Cardiopatías/inducido químicamente , Estrés Oxidativo/efectos de los fármacos , Acetilcisteína/farmacología , Animales , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Cardiopatías/fisiopatología , Cardiopatías/prevención & control , Pez Cebra
8.
ACS Appl Mater Interfaces ; 12(23): 25604-25615, 2020 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-32406668

RESUMEN

In spite of the rapid emergence of numerous nanoparticles (NPs) for biomedical applications, it is often challenging to precisely control, or effectively tame, the bioactivity/toxicity of NPs, thereby exhibiting limited applications in biomedical areas. Herein, we report the construction of hyaluronic acid (HA)-laminated, otherwise toxic methylviologen (MV), NPs via ternary host-guest complexation among cucurbit[8]uril, trans-azobenzene-conjugated HA, and MV-functionalized polylactic acid NPs (MV-NPs). The high, nonspecific toxicity of MV-NPs was effectively shielded (turned off) by HA lamination, as demonstrated in cells, zebrafish, and mouse models. The supramolecular host-guest interaction-mediated HA coating offered several HA-MV-NP modalities, including hyaluronidase locally and photoirradiation remotely, to precisely remove HA lamination on demand, thereby endowing materials with the capability of selective decoating-induced activation (DIA) for applications as a user-friendly herbicide, a selective antibacterial agent, or an anticancer nanomedicine. This work offers facile supramolecular coating and DIA strategies to effectively tame and precisely control the bioactivity and toxicity of functional nanomaterials for diverse applications.


Asunto(s)
Antibacterianos/uso terapéutico , Antineoplásicos/uso terapéutico , Nanopartículas/uso terapéutico , Neoplasias/tratamiento farmacológico , Paraquat/uso terapéutico , Infecciones Estafilocócicas/tratamiento farmacológico , Animales , Antibacterianos/química , Antibacterianos/toxicidad , Antineoplásicos/química , Antineoplásicos/toxicidad , Hidrocarburos Aromáticos con Puentes/química , Hidrocarburos Aromáticos con Puentes/toxicidad , Línea Celular Tumoral , Escherichia coli/efectos de los fármacos , Femenino , Fluoruros/química , Fluoruros/efectos de la radiación , Gadolinio/química , Gadolinio/efectos de la radiación , Ácido Hialurónico/química , Ácido Hialurónico/toxicidad , Imidazoles/química , Imidazoles/toxicidad , Rayos Infrarrojos , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Nanopartículas/química , Nanopartículas/efectos de la radiación , Nanopartículas/toxicidad , Paraquat/química , Paraquat/toxicidad , Poliésteres/química , Poliésteres/toxicidad , Staphylococcus aureus/efectos de los fármacos , Tulio/química , Tulio/efectos de la radiación , Iterbio/química , Iterbio/efectos de la radiación , Pez Cebra
9.
Nat Commun ; 11(1): 2622, 2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32457361

RESUMEN

Vascular disease remains the leading cause of death and disability, the etiology of which often involves atherosclerosis. The current treatment of atherosclerosis by pharmacotherapy has limited therapeutic efficacy. Here we report a biomimetic drug delivery system derived from macrophage membrane coated ROS-responsive nanoparticles (NPs). The macrophage membrane not only avoids the clearance of NPs from the reticuloendothelial system, but also leads NPs to the inflammatory tissues, where the ROS-responsiveness of NPs enables specific payload release. Moreover, the macrophage membrane sequesters proinflammatory cytokines to suppress local inflammation. The synergistic effects of pharmacotherapy and inflammatory cytokines sequestration from such a biomimetic drug delivery system lead to improved therapeutic efficacy in atherosclerosis. Comparison to macrophage internalized with ROS-responsive NPs, as a live-cell based drug delivery system for treatment of atherosclerosis, suggests that cell membrane coated drug delivery approach is likely more suitable for dealing with an inflammatory disease than the live-cell approach.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Citocinas/metabolismo , Sistemas de Liberación de Medicamentos , Macrófagos/metabolismo , Nanopartículas/metabolismo , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Aterosclerosis/metabolismo , Atorvastatina/uso terapéutico , Materiales Biomiméticos , Membrana Celular/metabolismo , Liberación de Fármacos , Femenino , Ratones , Células RAW 264.7 , Especies Reactivas de Oxígeno/metabolismo , Resultado del Tratamiento
10.
Mol Ecol Resour ; 19(4): 944-956, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30735609

RESUMEN

Marine mammals are important models for studying convergent evolution and aquatic adaption, and thus reference genomes of marine mammals can provide evolutionary insights. Here, we present the first chromosome-level marine mammal genome assembly based on the data generated by the BGISEQ-500 platform, for a stranded female sperm whale (Physeter macrocephalus). Using this reference genome, we performed chromosome evolution analysis of the sperm whale, including constructing ancestral chromosomes, identifying chromosome rearrangement events and comparing with cattle chromosomes, which provides a resource for exploring marine mammal adaptation and speciation. We detected a high proportion of long interspersed nuclear elements and expanded gene families, and contraction of major histocompatibility complex region genes which were specific to sperm whale. Using comparisons with sheep and cattle, we analysed positively selected genes to identify gene pathways that may be related to adaptation to the marine environment. Further, we identified possible convergent evolution in aquatic mammals by testing for positively selected genes across three orders of marine mammals. In addition, we used publicly available resequencing data to confirm a rapid decline in global population size in the Pliocene to Pleistocene transition. This study sheds light on the chromosome evolution and genetic mechanisms underpinning sperm whale adaptations, providing valuable resources for future comparative genomics.


Asunto(s)
Organismos Acuáticos/genética , Ecosistema , Evolución Molecular , Genoma , Cachalote/genética , Adaptación Biológica , Animales , Bovinos , Femenino , Ovinos
11.
Front Pharmacol ; 9: 614, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29988625

RESUMEN

Microglia-mediated neuroinflammatory responses are inevitable and important pathological processes in several kinds of disorder of the central nervous system (CNS). Therefore, alleviating activated microglia-induced inflammatory process might be a valuable therapeutic approach to neuroinflammation-related diseases. In the present study, we investigated BHDPC, a novel neuroprotectant discovered in our previous study that had anti-inflammatory effects under neuroinflammatory conditions. First, we found that BHDPC could inhibit neuroinflammatory responses and promote microglial M2 phenotype polarization in both lipopolysaccharide (LPS)-activated BV-2 microglia l cells. Furthermore, BHDPC provided protective actions against neuroinflammation-induced neurotoxicity in HT22 mouse hippocampal cells co-cultured with activated BV-2 microglia. Further experiments demonstrated that BHDPC could suppress LPS-induced activation of transcription factor nuclear factor kappa B (NF-κB) via interfering with the degradation of the inhibitor of kappa B (IκB) and phosphorylation of IκB, the IκB kinase (IKK). Moreover, we also found that BHDPC could induce phosphorylation of cAMP-dependent protein kinase A (PKA) and cAMP-response element-binding protein (CREB) in BV-2 microglial cells. Also, using the PKA-specific inhibitor, we found that BHDPC-induced CREB phosphorylation was dependent on PKA, which also contributed to BHDPC-mediated anti-inflammation and neuroprotection.

12.
Nat Commun ; 9(1): 2967, 2018 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-30054483

RESUMEN

Paraquat, as one of the most widely used herbicides globally, is highly toxic to humans, and chronic exposure and acute ingestion leads to high morbidity and mortality rates. Here, we report user-friendly, photo-responsive paraquat-loaded supramolecular vesicles, prepared via one-pot self-assembly of amphiphilic, ternary host-guest complexes between cucurbit[8]uril, paraquat, and an azobenzene derivative. In this vesicle formulation, paraquat is only released upon UV or sunlight irradiation that converts the azobenzene derivative from its trans- to its cis- form, which in turn dissociates the ternary host-guest complexations and the vesicles. The cytotoxicity evaluation of this vesicle formulation of paraquat on in vitro cell models, in vivo zebrafish models, and mouse models demonstrates an enhanced safety profile. Additionally, the PQ-loaded vesicles' herbicidal activity against a model of invasive weed is nearly identical to that of free paraquat under natural sunlight. This study provides a safe yet effective herbicide formulation.


Asunto(s)
Herbicidas/metabolismo , Herbicidas/efectos de la radiación , Herbicidas/toxicidad , Animales , Compuestos Azo/química , Compuestos Azo/metabolismo , Compuestos Azo/efectos de la radiación , Compuestos Azo/toxicidad , Células COS , Chlorocebus aethiops , Femenino , Herbicidas/química , Humanos , Riñón/efectos de los fármacos , Hígado/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Paraquat/química , Paraquat/metabolismo , Paraquat/efectos de la radiación , Paraquat/toxicidad , Pez Cebra
13.
Nanoscale ; 10(22): 10333-10336, 2018 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-29717743

RESUMEN

A synthetic nanoreceptor, cucurbit[7]uril (CB[7]), fully encapsulated pentylenetetrazol (PTZ), a seizure-inducing model drug. As a consequence of the encapsulation, the development of PTZ induced convulsion behaviors in both larval zebrafish and mouse models were dramatically alleviated, suggesting that CB[7] holds great neuroprotection potential against neurotoxic drugs for clinical applications.


Asunto(s)
Hidrocarburos Aromáticos con Puentes/farmacología , Imidazoles/farmacología , Fármacos Neuroprotectores/farmacología , Convulsiones/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Larva , Ratones , Ratones Endogámicos C57BL , Estructura Molecular , Pentilenotetrazol/efectos adversos , Convulsiones/inducido químicamente , Pez Cebra
14.
J Org Chem ; 83(8): 4882-4887, 2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29616817

RESUMEN

This is the first time that cucurbit[7]uril and cucurbit[8]uril have been demonstrated to serve as synthetic receptors for a halonium guest species, diphenyleneiodonium, modulating its bioactivities and alleviating its cardiotoxicity, which further expands the onium family of guest molecules for the cucurbit[ n]uril family and provides new insights for halonium-cucurbit[ n]uril host-guest chemistry and its potential applications in pharmaceutical chemistry.


Asunto(s)
Hidrocarburos Aromáticos con Puentes/química , Hidrocarburos Aromáticos con Puentes/farmacología , Cardiotónicos/química , Cardiotónicos/farmacología , Halógenos/química , Imidazoles/química , Imidazoles/farmacología , Animales , Cápsulas , Relación Dosis-Respuesta a Droga , Corazón/efectos de los fármacos , Ratones , Células RAW 264.7 , Relación Estructura-Actividad , Pez Cebra
15.
Front Mol Neurosci ; 11: 98, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29666569

RESUMEN

In response to the microenvironment, microglia may polarize into either an M1 pro-inflammatory phenotype, exacerbating neurotoxicity, or an M2 anti-inflammatory phenotype, conferring neuroprotection. Betulinic acid (BA) is a naturally pentacyclic triterpenoid with considerable anti-inflammatory properties. Here, we aim to investigate the potential effects of BA on microglial phenotype polarization and to reveal the underlying mechanisms of action. First, we confirmed that BA promoted M2 polarization and inhibited M1 polarization in lipopolysaccharide (LPS)-stimulated BV-2 microglial cells. Then, we demonstrated that the effect of BA on microglial polarization was dependent on AMP-activated protein kinase (AMPK) activation, as evidenced by the fact that both AMPK inhibitor compound C and AMPK siRNA abolished the M2 polarization promoted by BA. Moreover, we found that calmodulin-dependent protein kinase kinase ß (CaMKKß), but not liver kinase B1, was the upstream kinase required for BA-mediated AMPK activation and microglial M2 polarization, via the use of both the CaMKKß inhibitor STO-609 and CaMKKß siRNA. Finally, BA enhanced AMPK phosphorylation and promoted M2 microglial polarization in the cerebral cortex of LPS-injected mice brains, which was attenuated by pre-administration of the AMPK inhibitor. This study demonstrated that BA promoted M2 polarization of microglia, thus conferring anti-neuroinflammatory effects via CaMKKß-dependent AMPK activation.

16.
Chem Asian J ; 13(1): 41-45, 2018 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-29181896

RESUMEN

The teratogenicity of the pesticide nereistoxin (NTX) and its derivative thiocyclam (THI) towards aquatic life was dramatically constrained by a synthetic nanoreceptor, cucurbit[7]uril, through selective encapsulation of the pesticides (KCB[7]-NTX of 3.24(±0.31)×106 m-1 and KCB[7]-THI of 7.46(±0.10)×105 m-1 ), as evidenced by the rate of hatchability, morphology development, and tyrosinase activity of zebrafish larvae incubated with the pesticides (3-300 µm) in the absence and in the presence of 300 µm cucurbit[7]uril, demonstrating the significant potential of the nanoreceptor in managing ecological pollution of these pesticides.


Asunto(s)
Hidrocarburos Aromáticos con Puentes/farmacología , Compuestos Heterocíclicos con 1 Anillo/antagonistas & inhibidores , Compuestos Heterocíclicos con 1 Anillo/toxicidad , Imidazoles/farmacología , Toxinas Marinas/antagonistas & inhibidores , Toxinas Marinas/toxicidad , Teratógenos/toxicidad , Contaminantes Químicos del Agua/antagonistas & inhibidores , Animales , Hidrocarburos Aromáticos con Puentes/química , Relación Dosis-Respuesta a Droga , Compuestos Heterocíclicos con 1 Anillo/química , Imidazoles/química , Larva/efectos de los fármacos , Toxinas Marinas/química , Estructura Molecular , Relación Estructura-Actividad , Teratógenos/química , Contaminantes Químicos del Agua/química , Contaminantes Químicos del Agua/toxicidad , Pez Cebra
17.
Food Chem Toxicol ; 119: 425-429, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29258954

RESUMEN

Bedaquiline (BDQ) is a newly approved anti-tuberculosis drug in treating multidrug-resistant tuberculosis. However, it has very poor aqueous solubility and several case reports have proposed that BDQ has potential risk of cardiotoxicity to patients. In this present study, we have explored into employing host-guest interactions between a synthetic receptor, cucurbit[7]uril (CB[7]), and BDQ aiming to improve the solubility and reduce the inherent cardiotoxicity of BDQ. HPLC-UV test on the solubility of BDQ in the absence and in the presence of increasing concentrations of CB[7] suggested a host-dependent guest-solubility enhancements. Cardiovascular studies using an in vivo zebrafish model demonstrated that the cardiotoxicity of BDQ was indeed alleviated upon its complexations by the synthetic receptor. Furthermore, our in vitro antibacterial studies suggested that CB[7] formulated BDQ preserved its antimycobacterial efficacy against Mycobacterium smegmatis. Therefore, CB[7] may become a suitable pharmaceutical excipient in formulating BDQ for improving its physiochemical properties (such as solubility), and for alleviating its side effects (such as cardiotoxicity), while the antimycobacterial efficacy of BDQ may be well maintained.


Asunto(s)
Antituberculosos/uso terapéutico , Diarilquinolinas/toxicidad , Corazón/efectos de los fármacos , Tuberculosis Resistente a Múltiples Medicamentos/tratamiento farmacológico , Animales , Animales Modificados Genéticamente , Hidrocarburos Aromáticos con Puentes/farmacología , Cromatografía Líquida de Alta Presión , Imidazoles/farmacología , Pruebas de Sensibilidad Microbiana , Mycobacterium smegmatis/efectos de los fármacos , Solubilidad , Pez Cebra/embriología
18.
Mol Ecol Resour ; 18(2): 281-295, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29131534

RESUMEN

Anthozoans (e.g., corals, anemones) are an ecologically important and diverse group of marine metazoans that occur from shallow to deep waters worldwide. However, our understanding of the evolutionary relationships among the ~7,500 species within this class is hindered by the lack of phylogenetically informative markers that can be reliably sequenced across a diversity of taxa. We designed and tested 16,306 RNA baits to capture 720 ultraconserved element loci and 1,071 exon loci. Library preparation and target enrichment were performed on 33 taxa from all orders within the class Anthozoa. Following Illumina sequencing and Trinity assembly, we recovered 1,774 of 1,791 targeted loci. The mean number of loci recovered from each species was 638 ± 222, with more loci recovered from octocorals (783 ± 138 loci) than hexacorals (475 ± 187 loci). Parsimony informative sites ranged from 26 to 49% for alignments at differing hierarchical taxonomic levels (e.g., Anthozoa, Octocorallia, Hexacorallia). The per cent of variable sites within each of three genera (Acropora, Alcyonium, and Sinularia) for which multiple species were sequenced ranged from 4.7% to 30%. Maximum-likelihood analyses recovered highly resolved trees with topologies matching those supported by other studies, including the monophyly of the order Scleractinia. Our results demonstrate the utility of this target-enrichment approach to resolve phylogenetic relationships from relatively old to recent divergences. Redesigning the baits with improved affinities to capture loci within each subclass will provide a valuable toolset to address systematic questions, further our understanding of the timing of diversifications and help resolve long-standing controversial relationships in the class Anthozoa.


Asunto(s)
Antozoos/clasificación , Antozoos/genética , Genética de Población/métodos , Técnicas de Genotipaje/métodos , Animales
19.
Food Chem Toxicol ; 112: 421-426, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29248475

RESUMEN

In order to develop a novel strategy to alleviate the inherent hepatotoxicity of antidepressant trazodone (TZ), Cucurbit[7]uril (CB[7]) was adopted as pharmaceutical excipients and was studied for its capability to reduce the hepatotoxicity of TZ via supramolecular encapsulation. CB[7] was found to form strong 1:1 host-guest complexes with TZ and its metabolite m-chlorophenyl piperazine (mCPP), with binding constants of 1.50 (±0.13) × 106 M-1 and 6.90 (±0.49) × 105 M-1, respectively. The supramolecular complexations were examined by 1H NMR and UV-visible spectroscopic titrations, ESI-MS and ITC. In the presence of 0.5 mM CB[7], the IC50 values of TZ and mCPP on a human normal liver cell line L02 were increased from 215.5 ±â€¯3.3 µM to 544.1 ±â€¯51.2 µM, and from 166.8 ±â€¯3.8 µM to 241.7 ±â€¯6.8 µM, respectively. Evaluation on a zebrafish model demonstrated that CB[7] (0.1 mM) significantly alleviated the TZ induced liver toxicity, as shown by the level of liver degeneration, liver size and yolk sac retention. Our study may provide a supramolecular strategy to alleviate the hepatotoxicity induced by TZ and its metabolite mCPP, and this strategy may be extendable to other drugs that have inherent hepatotoxicity or other adverse effects.


Asunto(s)
Antidepresivos de Segunda Generación/toxicidad , Hígado/efectos de los fármacos , Trazodona/toxicidad , Animales , Hidrocarburos Aromáticos con Puentes/química , Calorimetría , Línea Celular , Excipientes/química , Humanos , Imidazoles/química , Concentración 50 Inhibidora , Larva/efectos de los fármacos , Modelos Animales , Espectroscopía de Protones por Resonancia Magnética , Espectrometría de Masa por Ionización de Electrospray , Espectrofotometría Ultravioleta , Saco Vitelino , Pez Cebra/embriología
20.
Org Biomol Chem ; 15(38): 8046-8053, 2017 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-28795750

RESUMEN

Small-molecule kinase inhibitors (SMKIs) have been widely used in the treatment of a variety of cancers due to their clinically demonstrated efficacy. However, the use of some SMKIs, such as sorafenib (SO), has been plagued by their cardiotoxicity that has been frequently observed in treated patients. Herein we report that the encapsulation of SO by a synthetic receptor cucurbit[7]uril (CB[7]) alleviated the inherent cardiotoxicity of SO, as demonstrated in an in vivo zebrafish model. Moreover, the anti-cancer activity of SO was well preserved, upon its encapsulation by CB[7], as demonstrated by both in vitro and in vivo cancer/angiogenesis models. This discovery may provide new insights into a novel supramolecular formulation of SMKIs for the management of their side-effects.


Asunto(s)
Antineoplásicos/efectos adversos , Antineoplásicos/farmacología , Hidrocarburos Aromáticos con Puentes/química , Cardiotoxicidad , Imidazoles/química , Niacinamida/análogos & derivados , Compuestos de Fenilurea/efectos adversos , Compuestos de Fenilurea/química , Animales , Antineoplásicos/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Embrión no Mamífero/efectos de los fármacos , Humanos , Sustancias Macromoleculares , Espectroscopía de Resonancia Magnética , Estructura Molecular , Neovascularización Fisiológica/efectos de los fármacos , Niacinamida/efectos adversos , Niacinamida/química , Sorafenib , Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...