Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Biotechnol Biofuels Bioprod ; 16(1): 87, 2023 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-37218014

RESUMEN

BACKGROUND: Lignocellulose is the most abundant renewable resource in the world and has attracted widespread attention. It can be hydrolyzed into sugars with the help of cellulases and hemicellulases that are secreted by filamentous fungi. Several studies have revealed that the Ras small GTPase superfamily regulates important cellular physiological processes, including synthesis of metabolites, sporulation, and cell growth and differentiation. However, it remains unknown how and to what extent Ras small GTPases participate in cellulase production. RESULTS: In this study, we found that the putative Ras small GTPase RSR1 negatively regulated the expression of cellulases and xylanases. Deletion of rsr1 (∆rsr1) significantly increased cellulase production and decreased the expression levels of ACY1-cAMP-protein kinase A (PKA) signaling pathway genes and the concentration of intracellular cyclic adenosine monophosphate (cAMP). Loss of acy1 based on ∆rsr1 (∆rsr1∆acy1) could further increase cellulase production and the expression levels of cellulase genes, while overexpression of acy1 based on ∆rsr1 (∆rsr1-OEacy1) significantly reduced cellulase production and transcriptional levels of cellulase genes. In addition, our results revealed that RSR1 negatively controlled cellulase production via the ACY1-cAMP-PKA pathway. Transcriptome analysis revealed significantly increased expression of three G-protein coupled receptors (GPCRs; tre62462, tre58767, and tre53238) and approximately two-fold higher expression of ACE3 and XYR1, which transcriptionally activated cellulases with the loss of rsr1. ∆rsr1∆ tre62462 exhibited a decrease in cellulase activity compared to ∆rsr1, while that of ∆rsr1∆tre58767 and ∆rsr1∆tre53238 showed a remarkable improvement compared to ∆rsr1. These findings revealed that GPCRs on the membrane may sense extracellular signals and transmit them to rsr1 and then to ACY1-cAMP-PKA, thereby negatively controlling the expression of the cellulase activators ACE3 and XYR1. These data indicate the crucial role of Ras small GTPases in regulating cellulase gene expression. CONCLUSIONS: Here, we demonstrate that some GPCRs and Ras small GTPases play key roles in the regulation of cellulase genes in Trichoderma reesei. Understanding the roles of these components in the regulation of cellulase gene transcription and the signaling processes in T. reesei can lay the groundwork for understanding and transforming other filamentous fungi.

2.
Bioresour Bioprocess ; 9(1): 96, 2022 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-38647894

RESUMEN

Trichoderma reesei RUT-C30 is a well-known high-yielding cellulase-producing fungal strain that converts lignocellulose into cellulosic sugar for resource regeneration. Calcium is a ubiquitous secondary messenger that regulates growth and cellulase production in T. reesei. We serendipitously found that adding Sr2+ to the medium significantly increased cellulase activity in the T. reesei RUT-C30 strain and upregulated the expression of cellulase-related genes. Further studies showed that Sr2+ supplementation increased the cytosolic calcium concentration and activated the calcium-responsive signal transduction pathway of Ca2+-calcineurin-responsive zinc finger transcription factor 1 (CRZ1). Using the plasma membrane Ca2+ channel blocker, LaCl3, we demonstrated that Sr2+ induces cellulase production via the calcium signaling pathway. Supplementation with the corresponding concentrations of Sr2+ also inhibited colony growth. Sr2+ supplementation led to an increase in intracellular reactive oxygen species (ROS) and upregulated the transcriptional levels of intracellular superoxide dismutase (sod1) and catalase (cat1). We further demonstrated that ROS content was detrimental to cellulase production, which was alleviated by the ROS scavenger N-acetyl cysteine (NAC). This study demonstrated for the first time that Sr2+ supplementation stimulates cellulase production and upregulates cellulase genes via the calcium signaling transduction pathway. Sr2+ leads to an increase in intracellular ROS, which is detrimental to cellulase production and can be alleviated by the ROS scavenger NAC. Our results provide insights into the mechanistic study of cellulase synthesis and the discovery of novel inducers of cellulase.

3.
Protein Expr Purif ; 188: 105970, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34500070

RESUMEN

HarpinEa protein can stimulate plants to produce defense responses to resist the attack of pathogens, improve plant immune resistance, and promote plant growth. This has extremely high application value in agriculture. To efficiently express soluble HarpinEa protein, in this study, we expressed HarpinEa protein with a 6× His-tag in Escherichia coli BL21 (DE3). Because of the low level of expression of HarpinEa protein in E. coli, three rounds of synonymous codon optimization were performed on the +53 bp of the translation initiation region (TIR) of HarpinEa. Soluble HarpinEa protein after optimization accounted for 50.3% of the total soluble cellular protein expressed. After purification using a Ni Bestarose Fast Flow column, the purity of HarpinEa protein exceeded 95%, and the yield reached 227.5 mg/L of culture medium. The purified HarpinEa protein was sensitive to proteases and exhibited thermal stability. It triggered visible hypersensitive responses after being injected into tobacco leaves for 48 h. Plants treated with HarpinEa showed obvious growth-promoting and resistance-improving performance. Thus, the use of TIR synonymous codon optimization successfully achieved the economical, efficient, and soluble production of HarpinEa protein.


Asunto(s)
Codón , Nicotiana/genética , Iniciación de la Cadena Peptídica Traduccional , Proteínas de Plantas/genética , Mutación Silenciosa , Triticum/genética , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Clonación Molecular , Secuencia Conservada , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Conformación de Ácido Nucleico , Reguladores del Crecimiento de las Plantas/biosíntesis , Reguladores del Crecimiento de las Plantas/genética , Reguladores del Crecimiento de las Plantas/farmacología , Hojas de la Planta/efectos de los fármacos , Hojas de la Planta/genética , Hojas de la Planta/crecimiento & desarrollo , Hojas de la Planta/metabolismo , Proteínas de Plantas/biosíntesis , Proteínas de Plantas/farmacología , Estabilidad Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Solubilidad , Nicotiana/efectos de los fármacos , Nicotiana/crecimiento & desarrollo , Nicotiana/metabolismo , Triticum/efectos de los fármacos , Triticum/crecimiento & desarrollo , Triticum/metabolismo
4.
Nanotechnology ; 30(5): 055601, 2019 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-30511654

RESUMEN

Novel type of multifunctional polymeric micelles (PMs) designated as HM-PMss/CTX micelles were developed in the present study for tumor-targeted and glutathione (GSH)-responsive delivery of cabazitaxel (CTX). The surface of the vehicles was modified with piloting molecules (HM-3 peptide), which targets α v ß 3 integrin overexpressed on cancer cells, and the micelle core was cross-linked by GSH-disintegrable disulfide linkages for controlled drug release. HM-PMss/CTX micelles were prepared using a mixture of two functionalized amphiphilic block copolymers and found to physically encapsulate CTX with excellent entrapment efficiency (93.94 ± 4.19%), drug-loading capacity (8.39 ± 2.28%), and a narrow size distribution. In vitro release profiles showed that CTX remained stably entrapped in the micelles in a release medium without GSH or with GSH of low concentration, while undergoing a rapid release in a highly reductive environment. Cellular uptake experiments showed that the conjugation of the targeting peptide, containing an arginine-glycine-aspartate sequence, enhanced the cellular uptake of HM-PMss/CTX micelles via α v ß 3 integrin-mediated endocytosis. In vitro cell viability measurements revealed that blank micelles were biocompatible, while HM-PMss/CTX micelles, owing to their tumor-targeting ability and GSH sensitivity, effectively inhibited the proliferation of MDA-MB-231 breast cancer cells. These results indicate that HM-PMss/CTX micelles could be a promising platform for future intelligent drug delivery in cancer therapy.


Asunto(s)
Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacología , Portadores de Fármacos/química , Glutatión/metabolismo , Neoplasias/tratamiento farmacológico , Taxoides/química , Taxoides/farmacología , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos/métodos , Liberación de Fármacos/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Micelas
5.
Sci Rep ; 8(1): 15766, 2018 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-30361617

RESUMEN

Inclusion body (IB) formation generates substantial bio-waste in the pharmaceutical industry and remains a major challenge for heterologous protein expression. Although chaperones can be co-expressed to improve soluble protein yield, their contribution to IB processing in vivo has not been thoroughly studied. Here, a GroEL-GroES co-expressing strain and a deficient strain were constructed to study the in vivo recovery of recombinant human tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). The interaction between GroEL/ES and TRAIL was simulated by molecular docking and identified by co-immunoprecipitation. The in vitro cytotoxicity of TRAIL IBs before and after in vivo recovery was subsequently determined by MTT assay. Additionally, IB structures were measured by Fourier transform infrared (FT-IR) spectroscopy and fluorescence spectroscopy. The results showed that after in vivo refolding, IBs retained lower levels of anti-tumor activity and fewer native-like ß-sheet structures. Fewer recoverable polypeptides were trapped in IBs after GroEL/ES co-expression and refolding in vivo. Therefore, GroEL/ES mediated the in vivo recovery of TRAIL IBs in Escherichia coli. These results may identify potential uses for IBs and provide additional insight into the detailed mechanisms of in vivo protein recovery.


Asunto(s)
Chaperonina 10/metabolismo , Chaperonina 60/metabolismo , Escherichia coli/metabolismo , Cuerpos de Inclusión/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Muerte Celular , Línea Celular Tumoral , Humanos , Simulación del Acoplamiento Molecular , Desnaturalización Proteica , Proteínas Recombinantes/metabolismo , Espectrometría de Fluorescencia
6.
J Biomater Sci Polym Ed ; 29(14): 1729-1744, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29851539

RESUMEN

Combination therapy of two or more drugs has gradually become of outmost importance in cancer treatment. Cabazitaxel (CTX) is a taxoid drug and tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of TNF superfamily. In this study, we prepared TRAIL-modified and CTX-loaded polymer micelle (TRAIL-M-CTX). This nanoparticle was self-assembled from biodegradable amphiphilic copolymers, monomethoxyl poly(ethylene glycol)-b-poly(DL-lactide) (mPEG-PLA) and COOH-PEG-PLA, via a nanoprecipitation method and were modified with the TRAIL protein, resulting in a particle size of 39.75 ± 0.17 nm in diameter and a drug encapsulation efficiency of 95.52 ± 1.69%. The successful coupling was confirmed by 1H NMR, FTIR spectroscopy, and DLS article size measurement. Pharmacodynamic analysis in two human cancer cell lines with different TRAIL sensitivities showed that TRAIL-M-CTX has a significantly better anticancer efficacy than the individual CTX and TRAIL protein. Importantly, TRAIL-M-CTX showed synergistic effects against TRAIL-insensitive cells (MCF-7). A study of cellular uptake implied that the modified micelles were internalized into MCF-7 cells more effectively than unmodified micelles, owing to the coupled TRAIL protein. A cell cycle assay of MCF-7 cells revealed that TRAIL-M-CTX significantly increased the sub-G1 population compared with CTX or TRIAL, thus, facilitating cancer cell apoptosis. These results suggest that TRAIL-M-CTX micelles have potential as a cancer chemotherapy formulation.


Asunto(s)
Antineoplásicos/administración & dosificación , Nanocápsulas/química , Poliésteres/química , Polietilenglicoles/química , Ligando Inductor de Apoptosis Relacionado con TNF/administración & dosificación , Taxoides/administración & dosificación , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Permeabilidad de la Membrana Celular , Supervivencia Celular/efectos de los fármacos , Liberación de Fármacos , Sinergismo Farmacológico , Humanos , Células MCF-7 , Micelas , Ligando Inductor de Apoptosis Relacionado con TNF/química , Taxoides/química
7.
Protein Expr Purif ; 148: 16-23, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29555311

RESUMEN

Escherichia coli is an essential host for large-scale expression of heterologous polypeptides. However, further applications are limited by the formation of potential protein aggregates. In this work, we developed a novel on-column tag removal and purification system based on Fh8 hydrophobic interaction chromatography purification and ΔI-CM self-cleavage to obtain soluble tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). We evaluated several methods to improve TRAIL solubility and finally demonstrated that the Fh8 tag was a powerful solubility enhancer. Finally, we replaced the tobacco etch virus (TEV) protease site with a ΔI-CM self-cleavage intein to simplify the purification process. The released soluble TRAIL purity and yield reached 98.4% and 82.1 mg/L in shake flasks, respectively. Thus, the Fh8-ΔI-CM system enhanced target protein solubility by Fh8, enabled on-column tag removal and purification based on Fh8 calcium-binding properties and ΔI-CM self-cleavage properties, and promoted the release of highly active protein with high yield and purity. Overall, our findings suggest that this Fh8-ΔI-CM system could be used as a novel solubility-inducing and purification fusion tag for protein production in E. coli.


Asunto(s)
Inteínas/genética , Proteínas de Microfilamentos/química , Ligando Inductor de Apoptosis Relacionado con TNF/química , Ligando Inductor de Apoptosis Relacionado con TNF/aislamiento & purificación , Sitios de Unión , Escherichia coli/química , Escherichia coli/genética , Péptido Hidrolasas/química , Potyvirus/química , Potyvirus/genética , Agregado de Proteínas/genética , Solubilidad , Ligando Inductor de Apoptosis Relacionado con TNF/genética
8.
Invest New Drugs ; 36(2): 269-277, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28868573

RESUMEN

Background Polymeric micellar paclitaxel (PM-paclitaxel) is a novel Cremophor EL-free, nanoparticle-encapsulated paclitaxel formulation administered through intravenous injection. The primary objective of this phase I trial was to determine the first cycle dose-limiting toxicities (DLTs) and maximum tolerated dose (MTD) of PM-paclitaxel. Secondary objectives included the evaluation of the safety, antitumor activity, and pharmacokinetic (PK) profile of PM-paclitaxel in patients with advanced malignancies. Methods The PM-paclitaxel dose was escalated from 175 mg/m2 (level 1) to 435 mg/m2 (level 5). PM-paclitaxel was intravenously administered to patients for 3 h without premedication on day 1 of a 21-day cycle. Results Eighteen patients with confirmed advanced malignancies received PM-paclitaxel. DLT included grade 4 neutropenia (four patients) and grade 3 numbness (one patient), which occurred in one of the six patients who received 300 mg/m2 (level 3) PM-paclitaxel and all three patients who were treated with 435 mg/m2 PM-paclitaxel. Thus, the MTD of PM-paclitaxel was determined as 390 mg/m2 (level 4). Acute hypersensitive reactions were not observed. Partial response was observed in six of 18 patients (33.3%), three of whom had prior exposure to paclitaxel chemotherapy. The peak concentration and area under the curve values of paclitaxel increased with increasing dosage, indicating that PM-paclitaxel exhibits linear PKs. Conclusions PM-paclitaxel showed high MTD without additional toxicity, and exhibited desirable antitumor activity. The recommended dose of PM paclitaxel for phase II study is 300 mg/m2.


Asunto(s)
Composición de Medicamentos , Micelas , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Paclitaxel/administración & dosificación , Paclitaxel/farmacocinética , Polímeros/química , Adulto , Anciano , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/efectos adversos , Antineoplásicos Fitogénicos/farmacocinética , Antineoplásicos Fitogénicos/uso terapéutico , Área Bajo la Curva , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Paclitaxel/efectos adversos , Paclitaxel/uso terapéutico , Supervivencia sin Progresión
9.
Sci Rep ; 7(1): 3461, 2017 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-28615636

RESUMEN

The EstPS1 gene, which encodes a novel carboxylesterase of Pseudomonas synxantha PS1 isolated from oil well-produced water, was cloned and sequenced. EstPS1 has an open reading frame of 1923 bp and encodes the 640-amino acid carboxylesterase (EstPS1), which contains an autotransporter (AT) domain (357-640 amino acids). Homology analysis revealed that EstPS1 shared the highest identity (88%) with EstA from Pseudomonas fluorescens A506 (NCBI database) and belonged to the carboxylesterase family (EC 3.1.1.1). The optimum pH and temperature of recombinant EstPS1 were found to be 8.0 and 60 °C, respectively. EstPS1 showed high thermostability, and the half-lives (T1/2 thermal inactivation) at 60, 70, 80, 90, and 100 °C were 14 h, 2 h, 31 min, 10 min, and 1 min, respectively. To understand the role of the AT domain in carboxylesterase, AT domain-truncated carboxylesterase (EstPS1ΔAT) was generated. EstPS1ΔAT showed a clearly decreased secretion rate, owing to the AT domain strongly improved secretory expression in the heterogeneous system. EstPS1 degraded various pyrethroid pesticides, and hydrolysis efficiencies were dependent on the pyrethroid molecular structure. EstPS1 degraded all the tested pyrethroid pesticides and hydrolysed the p-nitrophenyl esters of medium-short-chain fatty acids, indicating that EstPS1 is an esterase with broad specificity.


Asunto(s)
Biodegradación Ambiental , Carboxilesterasa/metabolismo , Plaguicidas/metabolismo , Piretrinas/metabolismo , Secuencia de Aminoácidos , Carboxilesterasa/química , Carboxilesterasa/genética , Secuencia Conservada , Perfilación de la Expresión Génica , Biblioteca de Genes , Modelos Moleculares , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , Pseudomonas/genética , Pseudomonas/metabolismo , Relación Estructura-Actividad , Temperatura
10.
Mol Pharm ; 14(8): 2585-2594, 2017 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-28199114

RESUMEN

Curcumin has high potential in suppressing many types of cancer and overcoming multidrug resistance in a multifaceted manner by targeting diverse molecular targets. However, the rather low systemic bioavailability resulted from its poor solubility in water and fast metabolism/excretion in vivo has hampered its applications in cancer therapy. To increase the aqueous solubility of curcumin while retaining the stability in blood circulation, here we report curcumin-loaded copolymer micelles with excellent in vitro and in vivo stability and antitumor efficacy. The two copolymers used for comparison were methoxy-poly(ethylene glycol)-block-poly(ε-caprolactone) (mPEG-PCL) and N-(tert-butoxycarbonyl)-l-phenylalanine end-capped mPEG-PCL (mPEG-PCL-Phe(Boc)). In vitro cytotoxicity evaluation against human pancreatic SW1990 cell line showed that the delivery of curcumin in mPEG-PCL-Phe(Boc) micelles to cancer cells was efficient and dosage-dependent. The pharmacokinetics in ICR mice indicated that intravenous (i.v.) administration of curcumin/mPEG-PCL-Phe(Boc) micelles could retain curcumin in plasma much better than curcumin/mPEG-PCL micelles. Biodistribution results in Sprague-Dawley rats also showed higher uptake and slower elimination of curcumin into liver, lung, kidney, and brain, and lower uptake into heart and spleen of mPEG-PCL-Phe(Boc) micelles, as compared with mPEG-PCL micelles. Further in vivo efficacy evaluation in multidrug-resistant human erythroleukemia K562/ADR xenograft model revealed that i.v. administration of curcumin-loaded mPEG-PCL-Phe(Boc) micelles significantly delayed tumor growth, which was attributed to the improved stability of curcumin in the bloodstream and increased systemic bioavailability. The mPEG-PCL-Phe(Boc) micellar system is promising in overcoming the key challenge of curcumin's to promote its applications in cancer therapy.


Asunto(s)
Curcumina/química , Leucemia Eritroblástica Aguda/tratamiento farmacológico , Polímeros/química , Animales , Línea Celular Tumoral , Curcumina/uso terapéutico , Humanos , Lactonas/química , Ratones , Poliésteres/química , Polietilenglicoles/química , Ratas , Ratas Sprague-Dawley
11.
Appl Microbiol Biotechnol ; 100(14): 6403-6413, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27063011

RESUMEN

Immunotoxins are a new class of antibody-targeted therapy in clinical development. Traditional immunotoxins that are constructed from the toxins of plants or bacteria need to be internalized to the cytoplasm and thus have limited antitumor efficacy. In the present study, we combined a recently reported sea anemone cytolysin Gigantoxin-4 with an anti-HER2/neu single-chain variable fragment 4D5 scFv to construct a novel immunotoxin. We fused a SUMO tag to the N-terminus of Gigantoxin-4-4D5 scFv and it was successfully expressed in Escherichia coli strain BL21 (DE3) in a soluble form. After purification, the purity of Gigantoxin-4-4D5 scFv reached 96 % and the yield was 14.3 mg/L. Our results demonstrated that Gigantoxin-4-4D5 scFv exerted a highly cytotoxic effect on the HER2/neu-positive ovarian carcinoma SK-OV-3 cell line. And the hemolytic activity was weaker, making it safe for normal cells. The results of immunofluorescence analysis showed that this novel immunotoxin could specifically bind to SK-OV-3 cells with no recognition of human embryonic kidney 293 cells. Scanning electron microscope observations and extracellular lactate dehydrogenase activity indicated that it could induce necrosis in SK-OV-3 cells by disrupting the cell membrane. Moreover, it could also mediate apoptosis of SK-OV-3 cells.


Asunto(s)
Venenos de Cnidarios/farmacología , Inmunotoxinas/farmacología , Neoplasias Ováricas/patología , Anticuerpos de Cadena Única/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Escherichia coli/genética , Escherichia coli/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Células HEK293 , Humanos , Inmunotoxinas/genética , Neoplasias Ováricas/tratamiento farmacológico , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología
12.
J Biomater Sci Polym Ed ; 27(7): 626-42, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26914063

RESUMEN

Cabazitaxel (CTX) is a second-generation semisynthetic taxane that demonstrates antitumor activity superior to docetaxel. However, the low aqueous solubility of CTX has hampered its use as a therapeutic agent. In this work, CTX-loaded N-t-butoxycarbonyl-L-phenylalanine end-capped monomethyl poly (ethylene glycol)-block-poly (D,L-lactide) (mPEG-PLA-Phe(Boc)/CTX) micelles were prepared to improve the solubility of CTX while retaining its superior stability before accessing the tumor site. The mPEG-PLA-Phe(Boc)/CTX micelles showed excellent stability in vitro compared with mPEG-PLA/CTX micelles. When stored at 25 °C, the mPEG-PLA/CTX micelles tended to aggregate within 1 h, whereas the mPEG-PLA-Phe(Boc)/CTX micelles were uniformly transparent even after three weeks. Dilution of mPEG-PLA/CTX micelles widened their size distribution and decreased the encapsulation efficiency, while significant change was not found in mPEG-PLA-Phe(Boc)/CTX micelles, even when diluted 1000-fold. Pharmacokinetic results in Sprague-Dawley rats indicated that, compared with Jevtana(®), intravenous administration of mPEG-PLA-Phe(Boc)/CTX micelles stably retained the CTX in plasma with 26.03-fold larger of the area under the time-concentration curve, 2.13-fold longer of the half-life, and 9.99-fold higher of the maximum concentration. In conclusion, mPEG-PLA-Phe(Boc) micelle may be a potential nanocarrier not only to improve the solubility of CTX but also to prolong the blood circulation time, which results in improved biological activity.


Asunto(s)
Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Micelas , Poliésteres/química , Poliésteres/farmacocinética , Polietilenglicoles/química , Polietilenglicoles/farmacocinética , Taxoides/química , Animales , Tiempo de Circulación Sanguínea , Línea Celular Tumoral , Portadores de Fármacos/síntesis química , Portadores de Fármacos/toxicidad , Liberación de Fármacos , Estabilidad de Medicamentos , Humanos , Masculino , Poliésteres/síntesis química , Poliésteres/toxicidad , Polietilenglicoles/síntesis química , Polietilenglicoles/toxicidad , Ratas , Ratas Sprague-Dawley
13.
Protein Expr Purif ; 125: 68-73, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26358405

RESUMEN

VAS-TRAIL is a bifunctional fusion protein that combines anti-angiogenic activity with tumor-selective apoptotic activity for enhanced anti-tumor efficacy. VAS-TRAIL is expressed as inclusion body in Escherichia coli, but protein refolding is difficult to achieve and results in low yields of bioactive protein. In this study, we describe an efficient method for VAS-TRAIL refolding. The solubilization of aggregated VAS-TRAIL was achieved by a triple agent solution, which consists of an alkaline solution (pH 11.5) containing 0.4M l-arginine and 2M urea. The solubilized protein showed high purity and preserved secondary structure according to fluorescence properties. VAS-TRAIL refolding was performed through stepwise dialysis and resulted in more than 50% recovery of the soluble protein. The function of l-arginine was additive with alkaline pH, as shown by the significant improvement in refolding yield (≈30%) by l-arginine-containing solubilization solutions compared with alkaline solubilization solutions without l-arginine. The refolded VAS-TRAIL also showed ß-sheet structures and the propensity for oligomerization. Bioassays showed that the refolded fusion protein exhibited the expected activities, including its apoptotic activities toward tumor and endothelial cells, which proposed its promising therapeutic potential.


Asunto(s)
Calreticulina/química , Calreticulina/farmacología , Cuerpos de Inclusión/química , Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacología , Replegamiento Proteico , Ligando Inductor de Apoptosis Relacionado con TNF/química , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Apoptosis/efectos de los fármacos , Arginina/química , Calreticulina/aislamiento & purificación , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cromatografía en Gel , Cromatografía por Intercambio Iónico , Escherichia coli , Humanos , Fragmentos de Péptidos/aislamiento & purificación , Estructura Secundaria de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/farmacología , Solubilidad , Soluciones , Ligando Inductor de Apoptosis Relacionado con TNF/aislamiento & purificación , Urea/química
14.
J Biotechnol ; 209: 16-22, 2015 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-26072465

RESUMEN

A bifunctional fusion protein, VAS-TRAIL, was designed for superior therapeutic efficacy by combining anti-angiogenesis activity with tumor-selective apoptosis activity. The protein was expressed as inclusion body (IB) in Escherichia coli. To enhance refolding yield and bioactivity, four fusions were constructed with different linkers (no linker, flexible linker, rigid linker, and helix-forming linker). A novel linker selection strategy based on IB conformational quality and activity was applied to predict the suitable linker. The conformational quality and activity of VAS-TRAIL IBs were analyzed by ATR-FTIR and cytotoxicity assay, respectively. Results demonstrated that aggregated VRT (fusion with rigid linker) contained the highest native-like ß structure content and retained part of the expected activity, namely, cytotoxicity activity on tumor cells. This finding suggested that the rigid linker was the most suitable candidate. Further results of in vitro refolding and subsequent circular dichroism and activity assay of four refolded fusions were significantly correlated with the predictions. Refolding of VRT yielded more soluble proteins containing the expected secondary structure and the highest bioactivity compared with that of other fusions. Our research may offer an efficient method for the high-throughput design of aggregated-prone therapeutic fusion protein.


Asunto(s)
Antineoplásicos/farmacología , Calreticulina/genética , Cuerpos de Inclusión/metabolismo , Fragmentos de Péptidos/genética , Proteínas Recombinantes de Fusión/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Calreticulina/metabolismo , Calreticulina/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Dicroismo Circular , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Mutagénesis Insercional , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Replegamiento Proteico , Estructura Secundaria de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología
15.
Mol Cell Biochem ; 315(1-2): 25-30, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18483760

RESUMEN

Serratia marcescens utilizes two types of quorum-sensing signal molecules: N-acyl homoserine lactones and furanosyl borate diester (AI-2). S-adenosylmethionine synthetase (METK), S-adenosylhomocysteine nucleosidase (PFS), and S-ribosylhomocysteinase (LUXS) are three key enzymes in the biosynthetic pathway leading to AI-2 production. The sequence of luxS gene was published at NCBI (Accession number: EF164926). So in this study, Serratia marcescens metK and pfs genes were successfully cloned with inverse PCR. The results show that the ORF lengths of metK and pfs are 1155 and 702 bp, and encode proteins of 384 and 233 residues. Their molecular weights and isoelectric points are 41.85 kD and 5.50; 27.67 kD and 5.56, which are acidic proteins judging from the calculated pI values. Expression products of two genes with pET28a((+)) system exhibited molecular weights in sodium dodecyl sulfate-polyacrylamide gel electrophoresis analysis comparable with a theoretical estimation. The sequences of these two genes were conferred China patent application numbers CN 200710048016.X and CN 200710048015.5, respectively.


Asunto(s)
Boratos/metabolismo , Compuestos Bicíclicos Heterocíclicos con Puentes/metabolismo , Ésteres/metabolismo , Genes Bacterianos , Percepción de Quorum , Serratia marcescens/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Clonación Molecular , Electroforesis en Gel de Agar , Electroforesis en Gel de Poliacrilamida , Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Datos de Secuencia Molecular
16.
Biotechnol Prog ; 23(1): 286-92, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17269700

RESUMEN

TNF-related apoptosis-inducing ligand (TRAIL/Apo2L) was produced mainly as inclusion bodies (IBs) by recombinant Escherichia coli with a temperature-inducible expression system. The yield of TRAIL type 2 IBs at higher preinduction specific growth rate (mu = 0.15 h-1) was higher than that of TRAIL type 1 IBs at lower preinduction specific growth rate (mu = 0.05 h-1). With the same optimized refolding protocols, two types of IBs exhibited different refolding features. Refolded type 1 IBs had higher recovery of more than 80% compared with type 2 IBs (57-63%). By the measurements of fluorescence and CD spectroscopy, type 1 TRAIL IBs dissolved by urea appeared to be a closer secondary structure to the native TRAIL than type 2. Furthermore, with trypsin treatment, the striking decrease in stability of type 1 IBs against protease digestion cannot be attributed to their small size particles observed by scanning electron microscope and probably depend on different protein structure properties between the two IBs. Different properties of inclusion bodies were mainly influenced by different physiological states of the cells just prior to the induction.


Asunto(s)
Escherichia coli/metabolismo , Cuerpos de Inclusión/química , Cuerpos de Inclusión/ultraestructura , Ingeniería de Proteínas/métodos , Ligando Inductor de Apoptosis Relacionado con TNF/química , Ligando Inductor de Apoptosis Relacionado con TNF/ultraestructura , Proliferación Celular , Escherichia coli/genética , Conformación Proteica , Pliegue de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
17.
Biotechnol Lett ; 28(15): 1215-9, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16799759

RESUMEN

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (Apo2L/TRAIL) is a new member of the TNF superfamily. In this work, the key role of Zn(2+) in high-level expression of soluble TRAIL was confirmed. The yield of soluble TRAIL reached 1.6 g l(-1) using a novel, two-stage Zn(2+) feeding strategy, and the accumulation of TRAIL inclusion bodies decreased. Furthermore, the purified TRAIL showed stronger cytotoxicity activity against human pancreatic 1990 tumor cells as the molar ratio of Zn(2+) to TRAIL monomer was 2 in purified TRAIL solution.


Asunto(s)
Medios de Cultivo/farmacología , Proteínas Recombinantes/biosíntesis , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Zinc/farmacología , Apoptosis/fisiología , Reactores Biológicos/microbiología , Medios de Cultivo/química , Escherichia coli/metabolismo , Solubilidad , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología
18.
Appl Microbiol Biotechnol ; 72(5): 1033-8, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16575568

RESUMEN

TRAIL was a tumor-specific protein in development as a novel anticancer therapeutic agent. Generally, when expressed in recombinant Escherichia coli, TRAIL protein was prone to form inclusion bodies. In this study, coexpression of human TRAIL protein and protein isoaspartate methyltranferase (PIMT) from E. coli on plasmid pBV-TRAIL-PCM in E. coli C600 was investigated to overcome the difficulties in soluble expression. The results showed that this PIMT coexpression strategy exerted a positive effect on the TRAIL protein expression in recombinant E. coli, which led to a mean increase in the intracellular concentration of soluble and total protein of TRAIL by 1.57-fold and 1.33-fold, respectively. At the same time, results also suggested that PIMT was a prospective partner for soluble expression of TRAIL protein.


Asunto(s)
Proteína D-Aspartato-L-Isoaspartato Metiltransferasa/biosíntesis , Ligando Inductor de Apoptosis Relacionado con TNF/biosíntesis , Clonación Molecular , Escherichia coli/genética , Escherichia coli/crecimiento & desarrollo , Escherichia coli/metabolismo , Fermentación , Proteína D-Aspartato-L-Isoaspartato Metiltransferasa/genética , Ligando Inductor de Apoptosis Relacionado con TNF/genética
19.
Appl Microbiol Biotechnol ; 71(2): 184-91, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16215715

RESUMEN

Different nutrient-feeding cultures were carried out in producing recombinant protein of truncated tumor necrosis factor related apoptosis-inducing ligand (TRAIL) (114-281 amino acids of TRAIL) in Escherichia coli strain C600/pBV-TRAIL. The effects of preinduction specific growth rate, postinduction carbon source (glucose and glycerol), and feeding strategies were investigated. The higher preinduction specific growth rate (mu = 0.22 h(-1)) contributed to the increase in the TRAIL production, at which TRAIL was accumulated in bacterial cells as 7.2% of total cellular protein, corresponding to 1.99 g l(-1) in contrast with 5.1% (1.29 g l(-1)) at preinduction specific growth rate (mu = 0.1 h(-1)) during high-cell-density culture. Glycerol was superior to glucose as the postinduction carbon source for TRAIL production. Under similar culture conditions, the final concentration of TRAIL was produced 1.59-fold more when glycerol was used as postinduction carbon source than when glucose was used. At the same time, the results showed that it is efficient to adopt the pH-stat feeding strategy at postinduction for the overproduction of TRAIL. The TRAIL production was increased up to 4.51 g l(-1), approximately 16.1% of total cellular protein. The mechanisms behind the preinduction specific growth rate effect on the expression level may be ascribed to the leakage secretion of acetate.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/biosíntesis , Biotecnología/métodos , Técnicas de Cultivo de Célula/métodos , Escherichia coli/metabolismo , Glicoproteínas de Membrana/biosíntesis , Proteínas Recombinantes/biosíntesis , Factor de Necrosis Tumoral alfa/biosíntesis , Acetatos/metabolismo , Reactores Biológicos/microbiología , Electroforesis en Gel de Poliacrilamida , Glucosa/metabolismo , Glicerol/metabolismo , Proteínas Recombinantes/análisis , Ligando Inductor de Apoptosis Relacionado con TNF
20.
Biochem Pharmacol ; 69(3): 407-14, 2005 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-15652232

RESUMEN

Prodigiosin, a bacterial metabolite, was reported to have immunosuppressive and anticancer activities. In this study, we investigated novel functions of prodigiosin about anti-metastasis and anti-invasion. Prodigiosin dose-dependently inhibited 95-D cells' migration and invasion according to wound healing assay and the Transwell assay. The inhibitive effect could reach about 50% when cells were treated with 5 microM prodigiosin for 12 h. In animal experiment, intraperitoneal administration of 5 mg kg(-1) prodigiosin decreased the number of metastatic nodules by 53% and elevated the survival rate of mice about one-fold comparing with control group. Results of cell aggregation and adhesion assay showed that prodigiosin could promote cell aggregation and simultaneously inhibit cell from adhering to extracellular matrix (ECM). In addition, prodigiosin suppressed RhoA gene expression, hence, decreased protein level of RhoA in 95-D cells, according to RT-PCR assay and Western blot assay. Gel zymogram assay revealed that prodigiosin could suppress the activity of matrix metalloproteinase-2 (MMP-2). These results demonstrate that prodigiosin effectively inhibit tumor metastasis in vitro and in vivo. The action mechanisms of prodigiosin are associated with the promotion of cell aggregation and the inhibition of various steps in cell invasive process, which include the inhibition of cell adhesion and mobility in a RhoA-dependent way and the suppression of MMP-2 ability.


Asunto(s)
Metástasis de la Neoplasia/prevención & control , Prodigiosina/farmacología , Animales , Adhesión Celular/efectos de los fármacos , Agregación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Invasividad Neoplásica , ARN Mensajero/análisis , Proteína de Unión al GTP rhoA/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA