Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Genome Res ; 26(9): 1233-44, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27516619

RESUMEN

Long noncoding RNAs (lncRNAs), a recently discovered class of cellular RNAs, play important roles in the regulation of many cellular developmental processes. Although lncRNAs have been systematically identified in various systems, most of them have not been functionally characterized in vivo in animal models. In this study, we identified 128 testis-specific Drosophila lncRNAs and knocked out 105 of them using an optimized three-component CRISPR/Cas9 system. Among the lncRNA knockouts, 33 (31%) exhibited a partial or complete loss of male fertility, accompanied by visual developmental defects in late spermatogenesis. In addition, six knockouts were fully or partially rescued by transgenes in a trans configuration, indicating that those lncRNAs primarily work in trans Furthermore, gene expression profiles for five lncRNA mutants revealed that testis-specific lncRNAs regulate global gene expression, orchestrating late male germ cell differentiation. Compared with coding genes, the testis-specific lncRNAs evolved much faster. Moreover, lncRNAs of greater functional importance exhibited higher sequence conservation, suggesting that they are under constant evolutionary selection. Collectively, our results reveal critical functions of rapidly evolving testis-specific lncRNAs in late Drosophila spermatogenesis.


Asunto(s)
Secuencia Conservada/genética , ARN Largo no Codificante/genética , Espermatogénesis/genética , Testículo/crecimiento & desarrollo , Animales , Sistemas CRISPR-Cas , Drosophila/genética , Drosophila/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/crecimiento & desarrollo , Infertilidad Masculina/genética , Infertilidad Masculina/patología , Masculino
2.
Cell Biochem Funct ; 37(1): 21-30, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30604499

RESUMEN

Currently, a new technology termed PROTAC, proteolysis targeting chimera, has been developed for inducing the protein degradation by a targeting molecule. This technology takes advantage of a moiety of targeted protein and a moiety of recognizing E3 ubiquitin ligase and produces a hybrid molecule to specifically knock down a targeted protein. During the first decade, three pedigreed groups worked on the development of this technology. To date, this technology has been extended by different groups, aiming to develop new drugs against different diseases including cancers. This review summarizes the contributions of the groups for the development of PROTAC. SIGNIFICANCE OF THE STUDY: This review summarized the development of the PROTAC technology for readers and also presented the author's opinions on the application of the technology in tumor therapy.


Asunto(s)
Desarrollo de Medicamentos/métodos , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Proteínas Recombinantes de Fusión/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Humanos
3.
Cell Biochem Funct ; 35(7): 420-425, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28940415

RESUMEN

Mesenchymal stromal cells (MSCs) have been extensively investigated as a potential antiinflammatory treatment in many inflammatory-related diseases; however, it remains unclear whether MSCs could be used to treat acute allergic rhinitis. A rat model of allergic rhinitis was treated with MSCs. The effect of MSCs on the inflammation of allergic rhinitis was evaluated by sneezing, nose rubbing, the pathology of the nasal mucosa, and the expression of interleukin 4, tumour necrosis factor alpha, and immunoglobulin E in the serum of rats. Also, the population of MSCs isolated from umbilical cords of humans was evaluated to determine if they could inhibit the symptoms and inflammation of acute allergic rhinitis in a rat model. We observed that this population of cells inhibited sneezing, nose rubbing, and changes in the pathology of the nasal mucosa. Intriguingly, we observed that MSCs reduced the expression of interleukin 4, tumour necrosis factor alpha, and immunoglobulin E in the serum. Furthermore, MSCs reduced the expression of histamine and the recruitment of macrophages in the nasal mucosa of allergic rhinitis rats. We reasoned that the effect of MSCs on allergic rhinitis might be through its regulation of the secretion of related cytokines from macrophages during the process of acute allergic rhinitis. This work suggested that MSCs from the umbilical cords of humans could be used as a positive clinical therapy for the human disease.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Rinitis Alérgica/terapia , Enfermedad Aguda , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Histamina/sangre , Humanos , Inmunoglobulina E/sangre , Interleucina-4/sangre , Macrófagos/citología , Macrófagos/inmunología , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Mucosa Nasal/citología , Mucosa Nasal/metabolismo , Mucosa Nasal/patología , Ratas , Ratas Sprague-Dawley , Rinitis Alérgica/metabolismo , Rinitis Alérgica/patología , Trasplante Heterólogo , Factor de Necrosis Tumoral alfa/sangre , Cordón Umbilical/citología
4.
Science ; 384(6702): eadh5548, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38900896

RESUMEN

The molecular mechanisms that regulate breast cancer cell (BCC) metastasis and proliferation within the leptomeninges (LM) are poorly understood, which limits the development of effective therapies. In this work, we show that BCCs in mice can invade the LM by abluminal migration along blood vessels that connect vertebral or calvarial bone marrow and meninges, bypassing the blood-brain barrier. This process is dependent on BCC engagement with vascular basement membrane laminin through expression of the neuronal pathfinding molecule integrin α6. Once in the LM, BCCs colocalize with perivascular meningeal macrophages and induce their expression of the prosurvival neurotrophin glial-derived neurotrophic factor (GDNF). Intrathecal GDNF blockade, macrophage-specific GDNF ablation, or deletion of the GDNF receptor neural cell adhesion molecule (NCAM) from BCCs inhibits breast cancer growth within the LM. These data suggest integrin α6 and the GDNF signaling axis as new therapeutic targets against breast cancer LM metastasis.


Asunto(s)
Neoplasias Óseas , Neoplasias de la Mama , Integrina alfa6 , Neoplasias Meníngeas , Meninges , Vías Nerviosas , Animales , Femenino , Humanos , Ratones , Membrana Basal/metabolismo , Neoplasias Óseas/secundario , Neoplasias Óseas/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/genética , Línea Celular Tumoral , Movimiento Celular , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Integrina alfa6/metabolismo , Laminina/metabolismo , Macrófagos/metabolismo , Neoplasias Meníngeas/metabolismo , Neoplasias Meníngeas/secundario , Meninges/patología , Invasividad Neoplásica , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Moléculas de Adhesión de Célula Nerviosa/genética , Transducción de Señal , Vías Nerviosas/metabolismo , Ratones SCID , Ratones Noqueados
5.
Oncogene ; 40(4): 705-716, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33239754

RESUMEN

CREPT and p15RS, also named RPRD1B and RPRD1A, are RPRD (regulation of nuclear pre-mRNA-domain-containing) proteins containing C-terminal domain (CTD)-interacting domain (CID), which mediates the binding to the CTD of Rpb1, the largest subunit of RNA polymerase II (RNAPII). CREPT and p15RS are highly conserved, with a common yeast orthologue Rtt103. Intriguingly, human CREPT and p15RS possess opposite functions in the regulation of cell proliferation and tumorigenesis. While p15RS inhibits cell proliferation, CREPT promotes cell cycle and tumor growth. Aberrant expression of both CREPT and p15RS was found in numerous types of cancers. At the molecular level, both CREPT and p15RS were reported to regulate gene transcription by interacting with RNAPII. However, CREPT also exerts a key function in the processes linked to DNA damage repairs. In this review, we summarized the recent studies regarding the biological roles of CREPT and p15RS, as well as the molecular mechanisms underlying their activities. Fully revealing the mechanisms of CREPT and p15RS functions will not only provide new insights into understanding gene transcription and maintenance of DNA stability in tumors, but also promote new approach development for tumor diagnosis and therapy.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Proteínas de Neoplasias/fisiología , Neoplasias/etiología , Proteínas Represoras/fisiología , Ciclo Celular , Reparación del ADN , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias/genética , ARN Polimerasa II/metabolismo , Transcripción Genética
6.
Front Oncol ; 11: 653902, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33928038

RESUMEN

Hepatocellular carcinoma (HCC) as a main type of primary liver cancers has become one of the most deadly tumors because of its high morbidity and poor prognosis. Fucoidan is a family of natural, heparin-like sulfated polysaccharides extracted from brown algae. It is not only a widely used dietary supplement, but also participates in many biological activities, such as anti-oxidation, anti-inflammation and anti-tumor. However, the mechanism of fucoidan induced inhibition of HCC is elusive. In our study, we demonstrated that fucoidan contributes to inhibiting cell proliferation in vivo and in vitro, restraining cell motility and invasion and inducing cell cycle arrest and apoptosis. According to High-Throughput sequencing of long-non-coding RNA (lncRNA) in MHCC-97H cells treated with 0.5 mg/mL fucoidan, we found that 56 and 49 lncRNAs were correspondingly up- and down-regulated. LINC00261, which was related to the progression of tumor, was highly expressed in fucoidan treated MHCC-97H cells. Moreover, knocking down LINC00261 promoted cell proliferation by promoting the expression level of miR-522-3p, which further decreased the expression level of downstream SFRP2. Taken together, our results verified that fucoidan effectively inhibits the progression of HCC via causing lncRNA LINC00261 overexpression.

7.
Front Oncol ; 11: 652567, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34235076

RESUMEN

Non-small cell lung cancer (NSCLC) has the highest morbidity and mortality among all carcinomas. However, it is difficult to diagnose in the early stage, and current therapeutic efficacy is not ideal. Although numerous studies have revealed that Ailanthone (Aila), a natural product, can inhibit multiple cancers by reducing cell proliferation and invasion and inducing apoptosis, the mechanism by which Aila represses NSCLC progression in a time-dependent manner remains unclear. In this study, we observed that most long noncoding RNAs (lncRNAs) were either notably up- or downregulated in NSCLC cells after treatment with Aila. Moreover, alterations in lncRNA expression induced by Aila were crucial for the initiation and metastasis of NSCLC. Furthermore, in our research, expression of DUXAP8 was significantly downregulated in NSCLC cells after treatment with Aila and regulated expression levels of EGR1. In conclusion, our findings demonstrate that Aila is a potent natural suppressor of NSCLC by modulating expression of DUXAP8 and EGR1.

8.
ACS Appl Mater Interfaces ; 13(39): 46334-46342, 2021 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-34549583

RESUMEN

Copper is vital for various life processes, whereas severely toxic at excess level. Intracellular copper homeostasis is strictly controlled by a set of transporters and chaperones encoded by the copper homeostasis genes. Increasing evidence has shown that copper is usually overloaded in multiple malignancies, including pancreatic cancer, which has an extremely poor prognosis. Recently, silencing the SLC31A1 gene, which encodes a major transmembrane copper transporter (CTR1), has been demonstrated to be an effective means for reducing the malignant degree of pancreatic cancer by downregulating the cellular copper levels. Herein, we utilized tetrahedral framework nucleic acids (tFNAs) as vehicles to overcome the biological barriers for delivering small molecular RNAs and efficiently transferred two kinds of CTR1 mRNA-targeted RNA therapeutics, siCTR1 or miR-124, into PANC-1 cells. Both therapeutic tFNAs, termed t-siCTR1 and t-miR-124, prevented copper intake more effective than the free RNA therapeutics via efficiently suppressing the expression of CTR1, thereby significantly attenuating the progression of PANC-1 cells. In this study, therapeutic tFNAs are constructed to target metal ion transporters for the first time, which may provide an effective strategy for future treatment of other metal metabolism disorders.


Asunto(s)
Antineoplásicos/uso terapéutico , Cobre/metabolismo , ADN/química , Portadores de Fármacos/química , Neoplasias Pancreáticas/tratamiento farmacológico , ARN sin Sentido/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Transportador de Cobre 1/metabolismo , Células HEK293 , Humanos , MicroARNs/uso terapéutico , Mitocondrias/efectos de los fármacos , Conformación de Ácido Nucleico , Neoplasias Pancreáticas/metabolismo , ARN Interferente Pequeño/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo
9.
Front Oncol ; 11: 653061, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33996574

RESUMEN

Pancreatic cancer is a common malignant tumor with a high incidence and mortality rate. The prognosis of patients with pancreatic cancer is considerably poor due to the lack of effective treatment in clinically. Despite numerous studies have revealed that baicalein, a natural product, is responsible for suppressing multiple cancer cells proliferation, motility and invasion. The mechanism by which baicalein restraining pancreatic cancer progression remains unclear. In this study, we firstly verified that baicalein plays a critical role in inhibiting pancreatic tumorigenesis in vitro and in vivo. Then we analyzed the alteration of microRNAs (miRNAs) expression levels in Panc-1 cells incubated with DMSO, 50 and 100 µM baicalein by High-Throughput sequencing. Intriguingly, we observed that 20 and 39 miRNAs were accordingly up- and down-regulated through comparing Panc-1 cells exposed to 100 µM baicalein with the control group. Quantitative PCR analysis confirmed that miR-139-3p was the most up-regulated miRNA after baicalein treatment, while miR-196b-5p was the most down-regulated miRNA. Further studies showed that miR-139-3p induced, miR-196b-5p inhibited the apoptosis of Panc-1 cells via targeting NOB1 and ING5 respectively. In conclusion, we demonstrated that baicalein is a potent inhibitor against pancreatic cancer by modulating the expression of miR-139-3p or miR-196b-5p.

10.
ACS Appl Mater Interfaces ; 13(25): 29416-29423, 2021 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-34148345

RESUMEN

DNA self-assembled nanostructures have been considered as effective vehicles for biomolecule delivery because of their excellent biocompatibility, cellular permeability, noncytotoxicity, and small size. Here, we report an efficient antiviral strategy with self-assembled tetrahedral framework nucleic acids (tFNAs) delivering small interfering RNA (t-siRNA) to silence classical swine fever virus (CSFV) gene in porcine host cells. In this study, two previously reported siRNAs, C3 and C6, specifically targeting the CSFV genome were selected and modified on tFNAs, respectively, and termed t-C3 and t-C6. Results indicate that t-C3 and t-C6 can inhibit the viral proliferation of CSFV in kidney derived porcine cells, PK-15, effectively and that inhibition was markedly stronger than free siRNA-C3 or siRNA-C6 only. In addition, the DNA nanostructure also has high cargo-carrying capacity, allowing to deliver multiple functional groups. To improve the antiviral ability of tFNAs, a dual-targeting DNA nanostructure t-C3-C6 was constructed and used to silence the CSFV gene in porcine host cells. This study found that t-C3-C6 can inhibit the viral release and replication, exhibiting outstanding anti-CSFV capabilities. Therefore, these dual-targeting tFNAs have great potential in virus therapy. This strategy not only provides a novel method to inhibit CSFV replication in porcine cells but also verifies that tFNAs are effective tools for delivery of antiviral elements, which have great application potential.


Asunto(s)
Antivirales , Virus de la Fiebre Porcina Clásica/efectos de los fármacos , Portadores de Fármacos , Nanoestructuras/química , ARN Interferente Pequeño , Animales , Antivirales/química , Antivirales/metabolismo , Antivirales/farmacología , Línea Celular , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Ácidos Nucleicos/química , Ácidos Nucleicos/metabolismo , ARN Interferente Pequeño/química , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/farmacología , Porcinos , Replicación Viral/efectos de los fármacos
11.
Theranostics ; 10(8): 3708-3721, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32206117

RESUMEN

Cancers remain a threat to human health due to the lack of effective therapeutic strategies. Great effort has been devoted to the discovery of drug targets to treat cancers, but novel oncoproteins still need to be unveiled for efficient therapy. Methods: We show that CREPT is highly expressed in pancreatic cancer and is associated with poor disease-free survival. CREPT overexpression promotes but CREPT deletion blocks colony formation and proliferation of pancreatic cancer cells. To provide a proof of concept for CREPT as a new target for the inhibition of pancreatic cancer, we designed a cell-permeable peptide-based proteolysis targeting chimera (PROTAC), named PRTC, based on the homodimerized leucine-zipper-like motif in the C-terminus domain of CREPT to induce its degradation in vivo. Results: PRTC has high affinity for CREPT, with Kd = 0.34 +/- 0.11 µM and is able to permeate into cells because of the attached membrane-transportable peptide RRRRK. PRTC effectively induces CREPT degradation in a proteasome-dependent manner. Intriguingly, PRTC inhibits colony formation, cell proliferation, and motility in pancreatic cancer cells and ultimately impairs xenograft tumor growth, comparable to the effect of CREPT deletion. Conclusions: PRTC-induced degradation of CREPT leads to inhibition of tumor growth, which is promising for the development of new drugs against pancreatic cancer. In addition, using an interacting motif based on the dimerized structure of proteins may be a new way to design a PROTAC aiming at degrading any protein without known interacting small molecules or peptides.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Péptidos/uso terapéutico , Proteolisis , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos
12.
Cell Death Dis ; 9(12): 1172, 2018 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-30518842

RESUMEN

Gastric cancer, like most of other cancers, has an uncontrolled cell cycle regulated by cyclins and cyclin-dependent kinases (CDKs). In this study, we reported that gastric cancer cells showed an accelerated G2/M transition promoted by CREPT/RPRD1B and Aurora kinase B (Aurora B). We found that CREPT/RPRD1B and Aurora B were coordinately expressed during the cell cycle in gastric cancer cells. Deletion of CREPT/RPRD1B disturbed the cell progression and extended the length of cell cycle, leading to a significant accumulation of mitotic cells. Mechanistically, we revealed that CREPT/RPRD1B interacted with Aurora B to regulate the expression of Cyclin B1 in gastric cancer cells. Interestingly, Aurora B phosphorylates S145 in a well-conserved motif of CREPT/RPRD1B. We proposed that phosphorylation of CREPT/RPRD1B by Aurora B is required for promoting the transcription of Cyclin B1, which is critical for the regulation of gastric tumorigenesis. Our study provides a mechanism by which gastric tumor cells maintain their high proliferation rate via coordination of Aurora B and CREPT/RPRD1B on the expression of Cyclin B1. Targeting the interaction of Aurora B and CREPT/RPRD1B might be a strategy for anti-gastric cancer therapy in the future.


Asunto(s)
Aurora Quinasa B/genética , Proteínas de Ciclo Celular/genética , Ciclina B1/genética , Puntos de Control de la Fase G2 del Ciclo Celular/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/genética , Neoplasias Gástricas/genética , Animales , Aurora Quinasa B/metabolismo , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Ciclina B1/metabolismo , Humanos , Metástasis Linfática , Ratones , Ratones Desnudos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Estadificación de Neoplasias , Fosforilación , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Nat Genet ; 48(12): 1508-1516, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27798626

RESUMEN

Skin integrity is essential for protection from external stress and trauma. Defects in structural proteins such as keratins cause skin fragility, epitomized by epidermolysis bullosa (EB), a life-threatening disorder. Here we show that dominant mutations of KLHL24, encoding a cullin 3-RBX1 ubiquitin ligase substrate receptor, cause EB. We have identified start-codon mutations in the KLHL24 gene in five patients with EB. These mutations lead to truncated KLHL24 protein lacking the initial 28 amino acids (KLHL24-ΔN28). KLHL24-ΔN28 is more stable than its wild-type counterpart owing to abolished autoubiquitination. We have further identified keratin 14 (KRT14) as a KLHL24 substrate and found that KLHL24-ΔN28 induces excessive ubiquitination and degradation of KRT14. Using a knock-in mouse model, we have confirmed that the Klhl24 mutations lead to stabilized Klhl24-ΔN28 and cause Krt14 degradation. Our findings identify a new disease-causing mechanism due to dysregulation of autoubiquitination and open new avenues for the treatment of related disorders.


Asunto(s)
Epidermólisis Ampollosa/genética , Queratina-14/metabolismo , Mutación/genética , Proteínas Represoras/genética , Adulto , Animales , Estudios de Casos y Controles , Preescolar , Femenino , Genotipo , Humanos , Recién Nacido , Masculino , Ratones , Linaje , Fenotipo , Proteolisis , Piel/metabolismo , Ubiquitina/metabolismo , Ubiquitinación
15.
Bioresour Technol ; 179: 8-12, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25514396

RESUMEN

In this study, a novel algal biofilm membrane photobioreactor (BMPBR) equipped with solid carriers and submerged membrane module was developed for attached growth of Chlorella vulgaris and secondary effluent treatment. The volumetric microalgae production achieved in BMPBR was 0.072 g L(-1) d(-1), which was 1.44-fold larger than that in suspended growth membrane photobioreactor (MPBR). Furthermore, 72.4% of the total produced algal biomass was immobilized as algal biofilm in BMPBR. Advanced nutrients removal from secondary effluent was achieved both in BMPBR and MPBR, with average reduction of about 85% for PO4(3-)-P in the stable stage. Additionally, BMPBR showed better nitrogen removal performance than MPBR due to its higher algal biomass productivity. Moreover, with the filtration effect of the submerged membrane module in the reactor, suspended microalgae could be completely isolated from the effluent and a low average SS concentration of 0.28 mg L(-1) was achieved in the effluent of BMPBR.


Asunto(s)
Biopelículas , Membranas Artificiales , Microalgas/crecimiento & desarrollo , Nitrógeno/aislamiento & purificación , Fósforo/aislamiento & purificación , Fotobiorreactores/microbiología , Eliminación de Residuos Líquidos , Purificación del Agua/instrumentación , Biodegradación Ambiental , Biomasa , Chlorella vulgaris/crecimiento & desarrollo , Clorofila/metabolismo
16.
Anal Chim Acta ; 804: 143-50, 2013 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-24267075

RESUMEN

A rapid, green and effective miniaturized sample preparation and analytical technique, i.e. ionic liquids-assisted microwave distillation coupled with headspace single-drop microextraction (ILAMD-HS-SDME) followed by gas chromatography-mass spectrometry (GC-MS) was developed for the analysis of essential oil (EO) in Fructus forsythiae. In this work, ionic liquids (ILs) were not only used as the absorption medium of microwave irradiation but also as the destruction agent of plant cell walls. 1-Ethyl-3-methylimidazolium acetate ([C2mim]OAc) was chosen as the optimal ILs. Moreover, n-heptadecane (2.0 µL) was selected as the appropriate suspended solvent for the extraction and concentration of EO. Extraction conditions of the proposed method were optimized using the relative peak area of EO constituents as the index, and the optimal operational parameters were obtained as follows: irradiation power (300 W), sample mass (0.7 g), mass ratio of ILs to sample (2.4), temperature (78°C) and time (3.4 min). In comparison to previous reports, the proposed method was faster and required smaller sample amount but could equally monitor all EO constituents with no significant differences.


Asunto(s)
Medicamentos Herbarios Chinos/química , Cromatografía de Gases y Espectrometría de Masas/métodos , Microondas , Aceites Volátiles/análisis , Microextracción en Fase Líquida , Microscopía Electrónica de Rastreo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA