Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
EBioMedicine ; 99: 104944, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38176215

RESUMEN

BACKGROUND: Severe fever with thrombocytopenia syndrome virus (SFTSV) is an emerging tick-borne pathogen that causes severe hemorrhagic fever in humans, but no FDA-approved specific antivirals or vaccines are available to treat or prevent SFTS. METHODS: The plasmids construction and transfection were performed to generate the recombinant SFTSV harboring the nanoluciferase gene (SFTSV-Nluc). Immunostaining plaque assay was performed to measure viral titers, and DNA electrophoresis and Sanger sequencing were performed to evaluate the genetic stability. Luciferase assay and quantitative RT-PCR were performed to evaluate the efficacy of antivirals in vitro. Bioluminescence imaging, titration of virus from excised organs, hematology, and histopathology and immunohistochemistry were performed to evaluate the efficacy of antivirals in vivo. FINDINGS: SFTSV-Nluc exhibited high genetic stability and replication kinetics similar to those of wild-type virus (SFTSVwt), then a rapid high-throughput screening system for identifying inhibitors to treat SFTS was developed, and a nucleoside analog, 4-FlU, was identified to effectively inhibit SFTSV in vitro. SFTSV-Nluc mimicked the replication characteristics and localization of SFTSVwt in counterpart model mice. Bioluminescence imaging of SFTSV-Nluc allowed real-time visualization and quantification of SFTSV replication in the mice. 4-FlU was demonstrated to inhibit the replication of SFTSV with more efficiency than T-705 and without obvious adverse effect in vivo. INTERPRETATION: The high-throughput screening system based on SFTSV-Nluc for use in vitro and in vivo revealed that a safe and effective antiviral nucleoside analog, 4-FlU, may be a basis for the strategic treatment of SFTSV and other bunyavirus infections, paving the way for the discovery of antivirals. FUNDING: This work was supported by grants from the National Key Research and Development Plan of China (2021YFC2300700 to L. Zhang, 2022YFC2303300 to L. Zhang), Strategic Priority Research Program of Chinese Academy of Sciences (XDB0490000 to L. Zhang), National Natural Science Foundation of China (31970165 to L. Zhang, U22A20379 to G. Xiao), the Science and Technology Commission of Shanghai Municipality (21S11903100 to Y. Xie), Hubei Natural Science Foundation for Distinguished Young Scholars (2022CFA099 to L. Zhang).


Asunto(s)
Phlebovirus , Síndrome de Trombocitopenia Febril Grave , Humanos , Animales , Ratones , Phlebovirus/genética , Nucleósidos , China , Antivirales/farmacología , Antivirales/uso terapéutico , Fiebre
2.
Int J Biol Sci ; 19(8): 2366-2381, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37215991

RESUMEN

Due to drug resistance, the clinical response to cisplatin (CDDP) from patients with liver cancer is unsatisfactory. The alleviation or overcoming of CDDP resistance is an urgent problem to be solved in clinics. Tumor cells rapidly change signal pathways to mediate drug resistance under drug exposure. Here, multiple phosphor-kinase assays were performed and c-Jun N-terminal kinase (JNK) was activated in liver cancer cells treated with CDDP. The high activity of the JNK promotes poor progression and mediates cisplatin resistance in liver cancer, leading to a poor prognosis of liver cancer. Mechanistically, the highly activated JNK phosphorylated c-Jun and ATF2 formed a heterodimer to upregulate the expression of Galectin-1, leading to promoting cisplatin resistance in liver cancer. Importantly, we simulated the clinical evolution of drug resistance in liver cancer by continuous CDDP administration in vivo. In vivo bioluminescence imaging showed the activity of JNK gradually increased during this process. Moreover, the inhibition of JNK activity by small molecular or genetic inhibitors enhanced DNA damage and overcame CDDP resistance in vitro and in vivo. Collectively, our results underline that the high activity of JNK/c-Jun-ATF2/Galectin-1 mediates cisplatin resistance in liver cancer and provides an optional scheme for dynamic monitoring of molecular activity in vivo.


Asunto(s)
Antineoplásicos , Neoplasias Hepáticas , Humanos , Factor de Transcripción Activador 2/genética , Factor de Transcripción Activador 2/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis , Línea Celular Tumoral , Cisplatino/farmacología , Cisplatino/uso terapéutico , Resistencia a Antineoplásicos/genética , Galectina 1/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética
3.
Methods Mol Biol ; 2524: 347-352, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35821486

RESUMEN

In vivo bioluminescence imaging (BLI) methods enable the longitudinal and semi-quantitative monitoring of viral replication dynamics in small animal models and, thus, are useful for examining viral pathogenesis and the effect of antiviral drugs. Here, we describe an in vivo BLI method to evaluate the efficacy of antiviral drugs against rabies virus (RABV) infection in mice. We exemplify mice inoculated with recombinant RABV expressing red firefly luciferase and administered orally with the antiviral drug, favipiravir. For the imaging, mice are intraperitoneally administered with D-luciferin and placed in the dark chamber of an imaging system. The BL images are captured using a highly sensitive charge-coupled device camera. Image data are processed and analyzed using image analysis software.


Asunto(s)
Virus de la Rabia , Rabia , Animales , Antivirales/uso terapéutico , Diagnóstico por Imagen/métodos , Mediciones Luminiscentes/métodos , Ratones , Rabia/tratamiento farmacológico
4.
Int J Mol Sci ; 23(10)2022 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-35628522

RESUMEN

The sole currently approved malaria vaccine targets the circumsporozoite protein-the protein that densely coats the surface of sporozoites, the parasite stage deposited in the skin of the mammalian host by infected mosquitoes. However, this vaccine only confers moderate protection against clinical diseases in children, impelling a continuous search for novel candidates. In this work, we studied the importance of the membrane-associated erythrocyte binding-like protein (MAEBL) for infection by Plasmodium sporozoites. Using transgenic parasites and live imaging in mice, we show that the absence of MAEBL reduces Plasmodium berghei hemolymph sporozoite infectivity to mice. Moreover, we found that maebl knockout (maebl-) sporozoites display reduced adhesion, including to cultured hepatocytes, which could contribute to the defects in multiple biological processes, such as in gliding motility, hepatocyte wounding, and invasion. The maebl- defective phenotypes in mosquito salivary gland and liver infection were reverted by genetic complementation. Using a parasite line expressing a C-terminal myc-tagged MAEBL, we found that MAEBL levels peak in midgut and hemolymph parasites but drop after sporozoite entry into the salivary glands, where the labeling was found to be heterogeneous among sporozoites. MAEBL was found associated, not only with micronemes, but also with the surface of mature sporozoites. Overall, our data provide further insight into the role of MAEBL in sporozoite infectivity and may contribute to the design of future immune interventions.


Asunto(s)
Plasmodium berghei , Proteínas Protozoarias , Receptores de Superficie Celular , Animales , Culicidae , Eritrocitos/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Plasmodium berghei/genética , Plasmodium berghei/patogenicidad , Proteínas Protozoarias/metabolismo , Receptores de Superficie Celular/metabolismo , Esporozoítos/metabolismo
5.
Cancers (Basel) ; 13(19)2021 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-34638483

RESUMEN

Endogenous acetaldehyde production from the metabolism of ingested alcohol exposes hematopoietic progenitor cells to increased genotoxic risk. To develop possible therapeutic strategies to prevent or reverse alcohol abuse effects, it would be critical to determine the temporal progression of acute ethanol toxicity on progenitor cell numbers and proliferative status. We followed the variation of the cell proliferation rate in bone marrow and spleen in response to acute ethanol intoxication in the MITO-Luc mouse, in which NF-Y-dependent cell proliferation can be assessed in vivo by non-invasive bioluminescent imaging. One week after ethanol administration, bioluminescent signals in bone marrow and spleen decreased below the level corresponding to physiological proliferation, and they progressively resumed to pre-treatment values in approximately 4 weeks. Boosting acetaldehyde catabolism by administration of an aldehyde dehydrogenase activity activator or administration of polyphenols with antioxidant activity partially restored bone marrow cells' physiological proliferation. These results indicate that in this mouse model, bioluminescent alteration reflects the reduction of the physiological proliferation rate of bone marrow progenitor cells due to the toxic effect of aldehydes generated by alcohol oxidation. In summary, this study presents a novel view of the impact of acute alcohol intake on bone marrow cell proliferation in vivo.

6.
Arch Microbiol ; 203(7): 4737-4742, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34184097

RESUMEN

In vivo bioluminescence imaging (BLI) offers a unique opportunity to analyze ongoing bacterial infections qualitatively and quantitatively in intact animals over time, leading to a reduction in the number of animals needed for a study. Since accurate determination of the bacterial burden plays an essential role in microbiological research, the present study aimed to evaluate the ability to quantify bacteria by non-invasive BLI technique in comparison to standard spread plate method and reverse transcription quantitative PCR (RT-qPCR). For this purpose, BALB/c mice were intranasally infected with 1 × 105 CFU of bioluminescent Streptococcus pneumoniae A66.1. At day 1 post-infection, the presence of S. pneumoniae in lungs was demonstrated by spread plate method and RT-qPCR, but not by in vivo BLI. However, on the second day p.i., the bioluminescent signal was already detectable, and the photon flux values positively correlated with CFU counts and RT-qPCR data within days 2-6. Though in vivo BLI is valuable research tool allowing the continuous monitoring and quantification of pneumococcal infection in living mice, it should be kept in mind that early in the infection, depending on the infective dose, the bioluminescent signal may be below the detection limit.


Asunto(s)
Fenómenos Fisiológicos Bacterianos , Animales , Bacterias/genética , Técnicas Bacteriológicas/normas , Mediciones Luminiscentes/normas , Ratones , Ratones Endogámicos BALB C , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/normas
7.
Front Pharmacol ; 10: 1337, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31798449

RESUMEN

Inhibitors of phosphodiesterase 4 (PDE4) are potent anti-inflammatory agents, inhibiting the production of inflammatory mediators through the elevation of intracellular cAMP concentrations. We studied the activity of a novel PDE4 inhibitor, CHF6001, both in vitro in human cells and in vivo, using bioluminescence imaging (BLI) in mice lung inflammation. Mice transiently transfected with the luciferase gene under the control of an NF-κB responsive element (NF-κB-luc) have been used to assess the in vivo anti-inflammatory activity of CHF6001 in lipopolysaccharide (LPS)-induced lung inflammation. BLI as well as inflammatory cells and the concentrations of pro-inflammatory cytokines were monitored in bronchoalveolar lavage fluids (BALF) while testing in vitro its ability to affect the production of leukotriene B4 (LTB4), measured by LC/MS/MS, by LPS/LPS/N-formyl--methionyl--leucyl-phenylalanine (fMLP)-activated human blood. CHF6001 inhibited the production of LTB4 in LPS/fMLP-activated human blood at sub-nanomolar concentrations. LPS-induced an increase of BLI signal in NF-κB-luc mice, and CHF6001 administered by dry powder inhalation decreased in parallel luciferase signal, cell airway infiltration, and pro-inflammatory cytokine concentrations in BALF. The results obtained provide in vitro and in vivo evidence of the anti-inflammatory activity of the potent PDE4 inhibitor CHF6001, showing that with a topical administration that closely mimics inhalation in humans, it efficiently disrupts the NF-κB activation associated with LPS challenge, an effect that may be relevant for the prevention of exacerbation episodes in chronic obstructive pulmonary disease subjects.

8.
Antiviral Res ; 172: 104641, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31672666

RESUMEN

Rabies virus (RABV) is a highly neurotropic virus and the causative agent of rabies, an encephalitis with an almost 100% case-fatality rate that remains incurable after the onset of symptoms. Favipiravir (T-705), a broad-spectrum antiviral drug against RNA viruses, has been shown to be effective against RABV in vitro but ineffective in vivo. We hypothesized that favipiravir is effective in infected mice when RABV replicates in the peripheral tissues/nerves but not after virus neuroinvasion. We attempted to clarify this point in this study using in vivo bioluminescence imaging. We generated a recombinant RABV from the field isolate 1088, which expressed red firefly luciferase (1088/RFLuc). This allowed semiquantitative detection and monitoring of primary replication at the inoculation site and viral spread in the central nervous system (CNS) in the same mice. Bioluminescence imaging revealed that favipiravir (300 mg/kg/day) treatment commencing 1 h after intramuscular inoculation of RABV efficiently suppressed viral replication at the inoculation site and the subsequent replication in the CNS. However, virus replication in the CNS was not inhibited when the treatment began 2 days after inoculation. We also found that higher doses (600 or 900 mg/kg/day) of favipiravir could suppress viral replication in the CNS even when administration started 2 days after inoculation. These results support our hypothesis and suggest that a highly effective drug-delivery system into the CNS and/or the enhancement of favipiravir conversion to its active form are required to improve favipiravir treatment of rabies. Furthermore, the bioluminescence imaging system established in this study will facilitate the development of treatment for symptomatic rabies.


Asunto(s)
Amidas , Sistema Nervioso Central/virología , Pirazinas , Virus de la Rabia/efectos de los fármacos , Rabia/tratamiento farmacológico , Amidas/administración & dosificación , Amidas/farmacología , Animales , Antivirales/administración & dosificación , Antivirales/farmacología , Línea Celular , Diagnóstico por Imagen/métodos , Mediciones Luminiscentes , Ratones , Pirazinas/administración & dosificación , Pirazinas/farmacología , Carga Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos
9.
Methods Mol Biol ; 1947: 361-376, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30969428

RESUMEN

Engineered G protein-coupled receptors (DREADDs, designer receptors exclusively activated by designer drugs) are convenient tools for specific activation of GPCR signaling in many cell types. DREADDs have been utilized as research tools to study numerous cellular and physiologic processes, including regulation of neuronal activity, behavior, and metabolism. Mice with random insertion transgenes and adeno-associated viruses have been widely used to express DREADDs in individual cell types. We recently created and characterized ROSA26-GsDREADD knock-in mice to allow Cre recombinase-dependent expression of a Gαs-coupled DREADD (GsD) fused to GFP in distinct cell populations in vivo. These animals also harbor a CREB-activated luciferase reporter gene for analysis of CREB activity by in vivo imaging, ex vivo imaging, or biochemical reporter assays. In this chapter, we provide detailed methods for breeding GsD animals, inducing GsD expression, stimulating GsD activity, and measuring basal and stimulated CREB reporter bioluminescence in tissues in vivo, ex vivo, and in vitro. These animals are available from our laboratory for non-profit research.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Genes Reporteros , Procesamiento de Imagen Asistido por Computador/métodos , Mediciones Luminiscentes/métodos , Moduladores del Transporte de Membrana/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Animales , Clozapina/análogos & derivados , Clozapina/farmacología , Integrasas/metabolismo , Ratones , Especificidad de Órganos , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores Acoplados a Proteínas G/genética , Transducción de Señal
10.
BMC Microbiol ; 18(1): 171, 2018 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-30376820

RESUMEN

BACKGROUND: Lactic acid bacteria (LAB) are major inhabitants and part of the normal microflora of the gastrointestinal tract (GIT) of humans and animals. Despite substantial evidence supporting the beneficial properties of LAB, only a few studies have addressed the migration and colonization of probiotic bacteria in the GIT. The reason for this is mostly due to the limitations, or lack of, efficient reporter systems. Here we describe the development and application of a non-invasive in vivo bioluminescence reporter system to study, in real-time, the spatial and temporal persistence of Lactobacillus plantarum 423 and Enterococcus mundtii ST4SA in the intestinal tract of mice. RESULTS: This study reports on the application of the firefly luciferase gene (ffluc) from Photinus pyralis to develop luciferase-expressing L. plantarum 423 and E. mundtii ST4SA, using a Lactococcus lactis NICE system on a high copy number plasmid (pNZ8048) and strong constitutive lactate dehydrogenase gene promoters (Pldh and STldh). The reporter system was used for in vivo and ex vivo monitoring of both probiotic LAB strains in the GIT of mice after single and multiple oral administrations. Enterococcus mundtii ST4SA reached the large intestine 45 min after gavage, while L. plantarum 423 reached the cecum/colon after 90 min. Both strains predominantly colonized the cecum and colon after five consecutive daily administrations. Enterococcus mundtii ST4SA persisted in faeces at higher numbers and for more days compared to L. plantarum 423. CONCLUSIONS: Our findings demonstrate the efficiency of a high-copy number vector, constitutive promoters and bioluminescence imaging to study the colonization and persistence of L. plantarum 423 and E. mundtii ST4SA in the murine GIT. The system allowed us to differentiate between intestinal transit times of the two strains in the digestive tract. This is the first report of bioluminescence imaging of a luciferase-expressing E. mundtii strain to study colonization dynamics in the murine model. The bioluminescence system developed in this study may be used to study the in vivo colonization dynamics of other probiotic LAB.


Asunto(s)
Enterococcus/fisiología , Intestinos/microbiología , Microscopía Intravital , Lactobacillus plantarum/fisiología , Mediciones Luminiscentes , Animales , Enterococcus/genética , Femenino , Tracto Gastrointestinal/microbiología , Lactobacillus plantarum/genética , Luciferasas/genética , Ratones , Ratones Endogámicos BALB C , Probióticos , Análisis Espacial
11.
Methods Mol Biol ; 1836: 431-459, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30151586

RESUMEN

To streamline standard virological assays, we developed bioluminescent replication-competent influenza reporter viruses that mimic their parental counterparts. These reporter viruses provide a rapid and quantitative readout of viral infection and replication. Moreover, they permit real-time in vivo measures of viral load, tissue distribution, and transmission in the same cohort of animals over the entire course of infection-measurements that were not previously possible. Here we provide detailed protocols using bioluminescent reporter viruses for in vivo imaging in mice and ferrets. We also describe cell culture-based techniques using reporter viruses for quantification of viral titers and performing microneutralization assays. The ease, speed, and adaptability of these approaches have the potential to accelerate multiple areas of influenza virus research.


Asunto(s)
Genes Reporteros , Mediciones Luminiscentes , Infecciones por Orthomyxoviridae/diagnóstico , Infecciones por Orthomyxoviridae/virología , Orthomyxoviridae/fisiología , Replicación Viral , Animales , Línea Celular , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Femenino , Hurones , Humanos , Mediciones Luminiscentes/métodos , Masculino , Ratones , Pruebas de Neutralización , Carga Viral
12.
Cell Physiol Biochem ; 46(2): 829-846, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29627822

RESUMEN

BACKGROUND/AIMS: Gambogic acid (GA), the main active compound of Gamboge hanburyi, has been reported to be a potential novel antitumor drug. Whether GA inhibits putative cancer stem cells (CSCs), which are considered to be the major cause of cancer treatment failure, remains largely unknown. This study investigated whether GA inhibits the CSCs of colorectal cancer (CRC) and its possible mechanisms. METHODS: We performed CCK8 and tumor sphere formation assays, percentage analysis of both side population and CD133+CD44+ cells, and the detection of stem cells markers, in order to assess the role of GA in inhibiting the stem celllike features of CRC. An mRNA microarray was performed to identify the downstream gene affected by GA and rescue assays were performed to further clarify whether the downstream gene is involved in the GA induced decrease of the stem cell-like CRC population. CRC cells were engineered with a CSC detector vector encoding GFP and luciferase (Luc) under the control of the Nanog promoter, which were utilized to investigate the effect of GA on putative CSC in human tumor xenograft-bearing mice using in vivo bioluminescence imaging. RESULTS: Our results showed that GA significantly reduced tumor sphere formation and the percentages of side population and CD133+CD44+ cells, while also decreasing the expression of stemness and EMT-associated markers in CRC cells in vitro. GA killed stem-like CRC cells by upregulating the expression of ZFP36, which is dependent on the inactivation of the EGFR/ ERK signaling pathway. GFP+ cells harboring the PNanog-GFP-T2A-Luc transgene exhibited CSC characteristics. The in vivo results showed that GA significantly inhibited tumor growth in nude mice, accompanied by a remarkable reduction in the putative CSC number, based on whole-body bioluminescence imaging. CONCLUSION: These findings suggest that GA significantly inhibits putative CSCs of CRC both in vitro and in vivo by inhibiting the activation of the EGFR/ ERK/ZFP36 signaling pathway and may be an effective drug candidate for anticancer therapies.


Asunto(s)
Apoptosis/efectos de los fármacos , Tristetraprolina/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Xantonas/toxicidad , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibronectinas/genética , Fibronectinas/metabolismo , Células HCT116 , Humanos , Ratones , Ratones Desnudos , Proteína Homeótica Nanog/genética , Proteína Homeótica Nanog/metabolismo , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal/efectos de los fármacos , Trasplante Heterólogo , Tristetraprolina/genética , Xantonas/uso terapéutico
13.
Microbes Infect ; 20(3): 166-175, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29180033

RESUMEN

Yersinia pestis has evolved from Yersinia pseudotuberculosis serotype O:1b. A typical Y. pestis contains three plasmids: pCD1, pMT1 and pPCP1. However, some isolates only harbor pCD1 (pCD1+-mutant). Y. pestis and Y. pseudotuberculosis share a common plasmid (pCD1 or pYV), but little is known about whether Y. pseudotuberculosis exhibited plague-inducing potential before it was evolved into Y. pestis. Here, the luxCDABE::Tn5::kan was integrated into the chromosome of the pCD1+-mutant, Y. pseudotuberculosis or Escherichia coli K12 to construct stable bioluminescent strains for investigation of their dissemination in mice by bioluminescence imaging technology. After subcutaneous infection, the pCD1+-mutant entered the lymph nodes, followed by the liver and spleen, and, subsequently, the lungs, causing pathological changes in these organs. Y. pseudotuberculosis entered the lymph nodes, but not the liver, spleen and lungs. It also resided in the lymph nodes for several days, but did not cause lymphadenitis or pathological lesions. By contrast, E. coli K12-lux was not isolatable from mouse lymph nodes, liver, spleen and lungs. These results indicate that the pCD1+-mutant can cause typical bubonic and pneumonic plague-like diseases, and Y. pestis has inherited lymphoid tissue tropism from its ancestor rather than acquiring these properties independently.


Asunto(s)
Rastreo Celular , Mediciones Luminiscentes , Peste/microbiología , Yersinia pestis/fisiología , Yersinia pseudotuberculosis/fisiología , Animales , Modelos Animales de Enfermedad , Escherichia coli/genética , Escherichia coli/crecimiento & desarrollo , Escherichia coli/patogenicidad , Hígado/microbiología , Hígado/patología , Pulmón/microbiología , Pulmón/patología , Tejido Linfoide/microbiología , Tejido Linfoide/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Peste/patología , Plásmidos/genética , Bazo/microbiología , Bazo/patología , Tropismo Viral , Virulencia , Yersinia pestis/genética , Yersinia pestis/crecimiento & desarrollo , Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/crecimiento & desarrollo
14.
Neoplasma ; 64(5): 718-724, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28592123

RESUMEN

The study aimed to investigate the effects of sorafenib on growth of hepatoma with hypoxia and angiogenesis. Green fluorescent protein (GFP)-expressing SMMC-7721 hepatoma cells were established via transfection with a lentiviral vector carrying GFP. GFP-labelled cells were treated with CoCl2 for 24 h with or without pretreatment of sorafenib for 30 min, and then injected subcutaneously into nude mice to induce hepatoma xenografts. This study used 16 BALB/c nude mice, which were divided into 4 groups: control group (GFP-labelled cells), CoCl2 group (GFP-labelled cells treated with CoCl2), CoCl2 plus sorafenib group (GFP-labelled cells pretreated with sorafenib and then treated with CoCl2) and CoCl2 plus sorafenib (cell+i.g.) group (GFP-labelled cells pretreated with sorafenib and then treated with CoCl2). After injection, CoCl2 plus sorafenib (cell+i.g.) group received intragastrical administration of sorafenib daily for 40 days. Tumor volume and weight were measured for each mouse. Cy5.5-annexinV and in vivo bioluminescence imaging were used for in vivo detection of cell apoptosis in tumor. Vascular endothelial growth factor (VEGF) and CD 34 were detected by immunohistochemistry. The results showed that, under the hypoxia condition induced by CoCl2, sorafenib pretreatment combined with intragastric administration of sorafenib more obviously suppressed tumor growth and decreased VEGF expression and MVD compared to sorafenib pretreatment alone, and induced cell apoptosis as well. Sorafenib pretreatment combined with intragastric administration is more effective than sorafenib pretreatment alone in the therapy of hepatoma.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Hipoxia/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Sorafenib/farmacología , Animales , Antígenos CD34 , Línea Celular Tumoral , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Factor A de Crecimiento Endotelial Vascular , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Sensors (Basel) ; 17(6)2017 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-28587284

RESUMEN

Since articular cartilage has a limited regeneration potential, for developing biological therapies for cartilage regeneration it is important to study the mechanisms underlying chondrogenesis of stem cells. Bioluminescence assays can visualize a wide range of biological phenomena such as gene expression, signaling, metabolism, development, cellular movements, and molecular interactions by using visible light and thus contribute substantially to elucidation of their biological functions. This article gives a concise review to introduce basic principles of bioluminescence assays and applications of the technology to visualize the processes of chondrogenesis and cartilage regeneration. Applications of bioluminescence assays have been highlighted in the methods of real-time monitoring of gene expression and intracellular levels of biomolecules and noninvasive cell tracking within animal models. This review suggests that bioluminescence assays can be applied towards a visual understanding of chondrogenesis and cartilage regeneration.


Asunto(s)
Condrogénesis , Animales , Cartílago Articular , Diferenciación Celular , Condrocitos , Humanos , Células Madre Mesenquimatosas , Regeneración , Células Madre
16.
Curr Pharm Des ; 23(13): 1963-1973, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28025948

RESUMEN

BACKGROUND: In vivo bioluminescence imaging has been used extensively for screening assays and for qualitative determination of localization of cells, in particular in cancer studies. OBJECTIVE: In this review we show the potential of this noninvasive molecular imaging modality to investigate gene activity, dynamic processes, and translational disease processes, all under true in vivo conditions with the specific focus on brain. RESULTS: We demonstrate a range of applications of bioluminescence imaging in basic and translational neuroscience. Here, emphasis is on the contribution of bioluminescence imaging of the brain to the elucidation of cellular and genetic mechanisms, understanding of dynamic processes, and to the discussion of disease characterization and therapeutic strategies.


Asunto(s)
Encéfalo/diagnóstico por imagen , Mediciones Luminiscentes , Imagen Molecular , Neurociencias/métodos , Investigación Biomédica Traslacional/métodos , Animales , Encéfalo/metabolismo , Encéfalo/patología , Humanos
17.
React Oxyg Species (Apex) ; 4(12): 382-388, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29732415

RESUMEN

The nuclear factor kappaB (NF-κB) is a redox-sensitive transcription factor that plays a critical role in inflammation among other biological functions. This ROS Protocol article describes an in vivo bioluminescence imaging assay for assessing NF-κB activation using the commercially available transgenic mice carrying NF-κB response element-luciferase reporter gene (NF-κB-RE-Luc). Using the highly sensitive Berthold NightOwl LB981 in vivo bioluminescence imaging system, we are able to visualize the NF-κB activation in live mice under basal conditions, suggesting constitutive activation of NF-κB as a part of its fundamental biology. Treatment of mice with lipopolysaccharides (LPS) results in a drastic increase in bioluminescence, proving the validity of the model in assessing inflammatory stress. Treatment of mice with 3H-1,2-dithiole-3-thione (D3T), an activator of nuclear factor E-2 related factor 2 (Nrf2), led to a significant reduction in both basal and LPS-induced activation of NF-κB in the live mice, suggesting a value of this model in assessing drug efficacy in suppressing NF-κB activation and inflammatory stress. The protocols of this valuable model are detailed in this article along with a discussion of its potential use in studying disease conditions involving inflammatory and oxidative stress mechanisms and in assessing therapeutic modalities targeting the NF-κB signaling for disease intervention.

18.
Int J Mol Sci ; 18(1)2016 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-28036021

RESUMEN

Increased levels of unconjugated bilirubin are neurotoxic, but the mechanism leading to neurological damage has not been completely elucidated. Innovative strategies of investigation are needed to more precisely define this pathological process. By longitudinal in vivo bioluminescence imaging, we noninvasively visualized the brain response to hyperbilirubinemia in the MITO-Luc mouse, in which light emission is restricted to the regions of active cell proliferation. We assessed that acute hyperbilirubinemia promotes bioluminescence in the brain region, indicating an increment in the cell proliferation rate. Immunohistochemical detection in brain sections of cells positive for both luciferase and the microglial marker allograft inflammatory factor 1 suggests proliferation of microglial cells. In addition, we demonstrated that brain induction of bioluminescence was altered by pharmacological displacement of bilirubin from its albumin binding sites and by modulation of the blood-brain barrier permeability, all pivotal factors in the development of bilirubin-induced neurologic dysfunction. We also determined that treatment with minocycline, an antibiotic with anti-inflammatory and neuroprotective properties, or administration of bevacizumab, an anti-vascular endothelial growth factor antibody, blunts bilirubin-induced bioluminescence. Overall the study supports the use of the MITO-Luc mouse as a valuable tool for the rapid response monitoring of drugs aiming at preventing acute bilirubin-induced neurological dysfunction.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Hiperbilirrubinemia/diagnóstico por imagen , Mediciones Luminiscentes/métodos , Imagen Óptica/métodos , Animales , Bevacizumab/farmacología , Barrera Hematoencefálica/diagnóstico por imagen , Barrera Hematoencefálica/efectos de los fármacos , Femenino , Luciferasas/genética , Luciferasas/metabolismo , Masculino , Ratones , Minociclina/farmacología
19.
BMC Cancer ; 16(1): 723, 2016 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-27609092

RESUMEN

BACKGROUND: The primary cause of treatment failure in medulloblastomas (MB) is the development of leptomeningeal dissemination (seeding). For translational research on MB seeding, one of the major challenges is the development of reliable experimental models that simulate the seeding and growth characteristics of MBs. To overcome this obstacle, we improved an experimental mouse model by intracisternal inoculation of human MB cells and monitoring with in vivo live images. METHODS: Human MB cells (UW426, D283 and MED8A) were transfected with a firefly luciferase gene and a Thy1.1 (CD90.1) marker linked with IRES under the control of the CMV promoter in a retroviral DNA backbone (effLuc). The MB-effLuc cells were injected into the cisterna magna using an intrathecal catheter, and bioluminescence images were captured. We performed histopathological analysis to confirm the extent of tumor seeding. RESULTS: The luciferase activity of MB-effLuc cells displayed a gradually increasing pattern, which correlated with a quantitative luminometric assay. Live imaging showed that the MB-effLuc cells were diffusely distributed in the cervical spinal cord and the lumbosacral area. All mice injected with UW426-effLuc, D283-effLuc and MED8A-effLuc died within 51 days. The median survival was 22, 41 and 12 days after injection of 1.2 × 10(6) UW426-effLuc, D283-effLuc and MED8A-effLuc cells, respectively. The histopathological studies revealed that the MB-effLuc cells spread extensively and diffusely along the leptomeninges of the brain and spinal cord, forming tumor cell-coated layers. The tumor cells in the subarachnoid space expressed a human nuclei marker and Ki-67. Compared with the intracerebellar injection method in which the subfrontal area and distal spinal cord were spared by tumor cell seeding in some mice, the intracisternal injection model more closely resembled the widespread leptomeningeal seeding observed in MB patients. CONCLUSION: The results and described method are valuable resources for further translational research to overcome MB seeding.


Asunto(s)
Neoplasias Cerebelosas/metabolismo , Meduloblastoma/metabolismo , Neoplasias Meníngeas/secundario , Animales , Línea Celular Tumoral , Neoplasias Cerebelosas/patología , Femenino , Genes Reporteros , Humanos , Luminiscencia , Meduloblastoma/patología , Neoplasias Meníngeas/metabolismo , Ratones , Microscopía Electrónica de Rastreo , Trasplante de Neoplasias
20.
J Transl Med ; 14(1): 226, 2016 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-27468800

RESUMEN

BACKGROUND: Experimentally, lung inflammation in laboratory animals is usually detected by the presence of inflammatory markers, such as immune cells and cytokines, in the bronchoalveolar lavage fluid (BALF) of sacrificed animals. This method, although extensively used, is time, money and animal life consuming, especially when applied to genetically modified animals. Thus a new and more convenient approach, based on in vivo imaging analysis, has been set up to evaluate the inflammatory response in the lung of CFTR-deficient (CF) mice, a murine model of cystic fibrosis. METHODS: Wild type (WT) and CF mice were stimulated with P. aeruginosa LPS, TNF-alpha and culture supernatant derived from P. aeruginosa (strain VR1). Lung inflammation was detected by measuring bioluminescence in vivo in mice transiently transgenized with a luciferase reporter gene under the control of a bovine IL-8 gene promoter. RESULTS: Differences in bioluminescence (BLI) signal were revealed by comparing the two types of mice after intratracheal challenge with pro-inflammatory stimuli. BLI increased at 4 h after stimulation with TNF-alpha and at 24 h after administration of LPS and VR1 supernatant in CF mice with respect to untreated animals. The BLI signal was significantly more intense and lasted for longer times in CF animals when compared to WT mice. Analysis of BALF markers: leukocytes, cytokines and histology revealed no significant differences between CF and WT mice. CONCLUSIONS: In vivo gene delivery technology and non-invasive bioluminescent imaging has been successfully adapted to CFTR-deficient mice. Activation of bIL-8 transgene promoter can be monitored by non-invasive BLI imaging in the lung of the same animal and compared longitudinally in both CF or WT mice, after challenge with pro-inflammatory stimuli. The combination of these technologies and the use of CF mice offer the unique opportunity of evaluating the impact of therapies aimed to control inflammation in a CF background.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/deficiencia , Neumonía/metabolismo , Neumonía/patología , Animales , Líquido del Lavado Bronquioalveolar , Fibrosis Quística , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Citocinas , Femenino , Procesamiento de Imagen Asistido por Computador , Ratones Endogámicos C57BL , Ratones Endogámicos CFTR
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...