Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Sensors (Basel) ; 21(11)2021 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-34073449

RESUMEN

Wi-Fi round-trip timing (RTT) was applied to indoor positioning systems based on distance estimation. RTT has a higher reception instability than the received signal strength indicator (RSSI)-based fingerprint in non-line-of-sight (NLOS) environments with many obstacles, resulting in large positioning errors due to multipath fading. To solve these problems, in this paper, we propose high-precision RTT-based indoor positioning system using an RTT compensation distance network (RCDN) and a region proposal network (RPN). The proposed method consists of a CNN-based RCDN for improving the prediction accuracy and learning rate of the received distances and a recurrent neural network-based RPN for real-time positioning, implemented in an end-to-end manner. The proposed RCDN collects and corrects a stable and reliable distance prediction value from each RTT transmitter by applying a scanning step to increase the reception rate of the TOF-based RTT with unstable reception. In addition, the user location is derived using the fingerprint-based location determination method through the RPN in which division processing is applied to the distances of the RTT corrected in the RCDN using the characteristics of the fast-sampling period.

2.
Biomacromolecules ; 16(1): 87-96, 2015 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-25400213

RESUMEN

Adenovirus (Ad) vectors show promise as cancer gene therapy delivery vehicles, but immunogenic safety concerns and coxsackie and adenovirus receptor (CAR)-dependency have limited their use. Alternately, biocompatible and bioreducible nonviral vectors, including arginine-grafted cationic polymers, have been shown to deliver nucleic acids through a cell penetration peptide (CPP) and protein transduction domain (PTD) effect. We utilized the advantages of both viral and nonviral vectors to develop a hybrid gene delivery vehicle by coating Ad with mPEG-PEI-g-Arg-S-S-Arg-g-PEI-mPEG (Ad/PPSA). Characterization of Ad/PPSA particle size and zeta potential showed an overall size and cationic charge increase in a polymer concentration-dependent manner. Ad/PPSA also showed a marked transduction efficiency increase in both CAR-negative and -positive cells compared to naked Ad. Competition assays demonstrated that Ad/PPSA produced higher transgene expression levels than naked Ad and achieved CAR-independent transduction. Oncolytic Ad (DWP418)/PPSA was able to overcome the nonspecificity of polymer-only therapies by demonstrating cancer-specific killing effects. Furthermore, the DWP418/PPSA nanocomplex elicited a 2.24-fold greater antitumor efficacy than naked Ad in vivo. This was supported by immunohistochemical confirmation of Ad E1As accumulation in MCF7 xenografted tumors. Lastly, intravenous injection of DWP418/PPSA elicited less innate immune response compared to naked Ad, evaluated by interleukin-6 cytokine release into the serum. The increased antitumor effect and improved vector targeting to both CAR-negative and -positive cells make DWP418/PPSA a promising tool for cancer gene therapy.


Asunto(s)
Adenoviridae/química , Antineoplásicos/química , Materiales Biocompatibles/química , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , Virus Oncolíticos/química , Polímeros/química , Animales , Antineoplásicos/administración & dosificación , Materiales Biocompatibles/administración & dosificación , Femenino , Células HEK293 , Humanos , Células MCF-7 , Ratones , Ratones Desnudos , Viroterapia Oncolítica/métodos , Polímeros/administración & dosificación , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
3.
Adv Healthc Mater ; : e2400240, 2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-39081097

RESUMEN

Necroptosis, a cell death mechanism with the characteristics of both apoptosis and necrosis, is proposed as a promising therapeutic approach for cancer therapy. Induction of necroptosis for cancer therapy may be possible through the regulation of the expression of a key factor gene receptor-interacting protein kinase-3 (RIPK3) via in vitro transcription (IVT) mRNA delivery. However, mRNA is susceptible to degradation and has a low delivery efficiency, which highlights the requirement of a proper delivery vehicle for intracellular delivery. Therefore, a new mRNA delivery system based on the nanostructured silica nanoparticles, termed mRNA-protective nanocage (mPN) has been developed. High-efficiency expression of RIPK3 and induction of necroptosis is achieved through delivery of RIPK3 IVT mRNA with mPN in vitro and in vivo models. Importantly, the mPN carrying RIPK3 mRNA distributed locally in tumors upon intravascular injection, and successfully induced necroptosis and immune cell infiltration, a hallmark of necroptosis. the suppression of tumor growth in a murine cancer model, demonstrating the synergistic effect of RIPK3 mRNA- and immune cell-mediated therapy is also observed. These findings suggest the potential for anticancer therapy through necroptosis induction and provide a strategy for the development of mRNA-based nanomedicine.

4.
Cancer Res Commun ; 4(7): 1748-1764, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38916448

RESUMEN

Immune checkpoint inhibitors are effective first-line therapy for solid cancers. However, low response rate and acquired resistance over time has led to the need for additional therapeutic options. Here, we evaluated synergistic antitumor efficacy of EGFR × MET targeting bispecific antibody, amivantamab with PD-L1 immunotherapy, pembrolizumab in head and neck squamous cell carcinoma (HNSCC) and lung squamous cell carcinoma tumor-bearing humanized patient-derived xenograft (PDX) models. We demonstrated that pembrolizumab or amivantamab alone was ineffective and that combination treatment induced a significant reduction of tumor growth in both models (P < 0.0001 and P < 0.01, respectively). It appeared that combination of amivantamab and pembrolizumab significantly enhanced infiltration of granzyme B-producing CD8 T cells was in the TME of HNSCC PDX (P < 0.01) and enhanced neoantigen-associated central memory CD8 T cells in circulating immune cells. Analysis of single-cell RNA transcriptomics suggested that the tumor cells dramatically upregulated EGFR and MET in response to PD-L1 immunotherapy, potentially creating a metabolic state fit for tumor persistence in the tumor microenvironment (TME) and rendered pembrolizumab ineffective. We demonstrated that EGFRHIGHMETHIGH subcluster displayed an increased expression of genes implicated in production of lactate [SLC16A3 and lactate dehydrogenase A (LDHA)] compared to the EGFRLOWMETLOW cluster. Accumulation of lactate in the TME has been associated with immunosuppression by hindering the infiltration of tumor killing CD8 T and NK cells. This study proved that amivantamab reduced glycolytic markers in the EGFRHIGHMETHIGH subcluster including SLC16A3 and LDHA and highlighted remodeling of the TME by combination treatment, providing rationale for additional therapy of amivantamab with PD-1 immunotherapy. SIGNIFICANCE: Amivantamab in synergy with pembrolizumab effectively eradicated EGFRHIGHMETHIGH tumor subcluster in the tumor microenvironment of head and neck squamous cell carcinoma and overcame resistance against anti-PD-1 immunotherapy.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Neoplasias Pulmonares , Carcinoma de Células Escamosas de Cabeza y Cuello , Microambiente Tumoral , Humanos , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales Humanizados/administración & dosificación , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Carcinoma de Células Escamosas de Cabeza y Cuello/inmunología , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Animales , Ratones , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/patología , Neoplasias de Cabeza y Cuello/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Antígeno B7-H1/metabolismo , Línea Celular Tumoral
5.
Carcinogenesis ; 34(7): 1543-50, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23508637

RESUMEN

Genomic analyses have revealed the enormous heterogeneity in essentially all cancer types. However, the identification of precise subtypes, which are biologically informative and clinically useful, remains a challenge. The application of integrative analysis of multilayered genomic profiles to define the chromosomal regions of genomic copy number alterations with concomitant transcriptional deregulation is posited to provide a promising strategy to identify driver targets. In this study, we performed an integrative analysis of the DNA copy numbers and gene expression profiles of hepatocellular carcinoma (HCC). By comparing DNA copy numbers between HCC subtypes based on gene expression pattern, we revealed the DNA copy number alteration with concordant gene expression changes at 6p21-p24 particularly in the HCC subtype of aggressive phenotype without expressing stemness genes. Among the genes at 6p21-p24, we identified IER3 as a potential driver. The clinical utility of IER3 copy numbers was demonstrated by validating its clinical correlation with independent cohorts. In addition, short hairpin RNA-mediated knock-down experiment revealed the functional relevance of IER3 in liver cancer progression. In conclusion, our results suggest that genomic copy number alterations with transcriptional deregulation at 6p21-p24 identify an aggressive HCC phenotype and a novel functional biomarker.


Asunto(s)
Carcinoma Hepatocelular/genética , Cromosomas Humanos Par 6/genética , Variaciones en el Número de Copia de ADN , Regulación Neoplásica de la Expresión Génica , Transcripción Genética , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/patología , Proliferación Celular , Hibridación Genómica Comparativa , Progresión de la Enfermedad , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Fenotipo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transcriptoma
6.
Bioengineering (Basel) ; 10(5)2023 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-37237691

RESUMEN

Neurodegenerative diseases (NDDs), which are chronic and progressive diseases, are a growing health concern. Among the therapeutic methods, stem-cell-based therapy is an attractive approach to NDD treatment owing to stem cells' characteristics such as their angiogenic ability, anti-inflammatory, paracrine, and anti-apoptotic effects, and homing ability to the damaged brain region. Human bone-marrow-derived mesenchymal stem cells (hBM-MSCs) are attractive NDD therapeutic agents owing to their widespread availability, easy attainability and in vitro manipulation and the lack of ethical issues. Ex vivo hBM-MSC expansion before transplantation is essential because of the low cell numbers in bone marrow aspirates. However, hBM-MSC quality decreases over time after detachment from culture dishes, and the ability of hBM-MSCs to differentiate after detachment from culture dishes remains poorly understood. Conventional analysis of hBM-MSCs characteristics before transplantation into the brain has several limitations. However, omics analyses provide more comprehensive molecular profiling of multifactorial biological systems. Omics and machine learning approaches can handle big data and provide more detailed characterization of hBM-MSCs. Here, we provide a brief review on the application of hBM-MSCs in the treatment of NDDs and an overview of integrated omics analysis of the quality and differentiation ability of hBM-MSCs detached from culture dishes for successful stem cell therapy.

7.
Biomater Sci ; 10(15): 4293-4308, 2022 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-35766864

RESUMEN

Oncolytic virotherapy is a highly promising and novel treatment modality for cancer. Several clinical trials with oncolytic viruses have illustrated that the potent antitumor efficacy of these viruses may rely on the efficient induction of antitumor immune response. In contrast, antiviral immune response is attributed to adverse side defects and diminishing therapeutic efficacy. In the present report, we generated a nanohybrid complex incorporating immune stimulatory oncolytic adenovirus (oAd) co-expressing decorin (DCN) and interleukin (IL)-12 with a bioreducible nanomaterial composed of PEI-Arg-mPEG-S-S-mPEG-Arg-PEI blocks (PAPS), ultimately aiming to modulate both antitumor and antiviral immune responses to be favorable toward oncolytic virotherapy. The transduction efficacy of the PAPS-incorporated nanohybrid vector (Ad/PAPS) was significantly higher than that of a complex using our previously reported polymer PPSA (Ad/PPSA) regardless of the cellular coxsackievirus and adenovirus receptor expression level of cancer cells. oAd complexed with PAPS (oAd/PAPS) also elicited a more potent cancer cell killing effect, antitumor efficacy, and metastasis inhibition than naked oAd or oAd complexed with PPSA (oAd/PPSA) through a higher level of therapeutic transgenes (DCN and IL-12), viral replication, and more efficient infiltration of T cells into tumor tissues. Notably, oAd/PAPS induced the highest level of antitumor immune response while the antiviral immune response was mediated at a significantly lower level than those of naked oAd. Adaptive immune response against the virus was also significantly attenuated in the oAd/PAPS group. oAd/PAPS treatment also led to the highest level of antitumor central memory T cells and the lowest level of immunosuppressive regulatory T cells in the spleen. Collectively, our findings illustrate that oAd/PAPS can simultaneously regulate both antitumor and antiviral immune responses to be more favorable to oncolytic virotherapy, leading to improved gene expression, viral replication, and growth inhibition of both primary and metastatic tumors.


Asunto(s)
Adenoviridae , Viroterapia Oncolítica , Inmunidad Adaptativa , Adenoviridae/genética , Adenoviridae/metabolismo , Antivirales , Línea Celular Tumoral , Interleucina-12/metabolismo , Polímeros/metabolismo
8.
Theranostics ; 12(8): 3676-3689, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35664056

RESUMEN

Understanding cancer heterogeneity is essential to finding diverse genetic mutations in metastatic cancers. Thus, it is critical to isolate all types of CTCs to identify accurate cancer information from patients. Moreover, full automation robustly capturing the full spectrum of CTCs is an urgent need for CTC diagnosis to be routine clinical practice. Methods: Here we report the full capture of heterogeneous CTC populations using fully automated, negative depletion-based continuous centrifugal microfluidics (CCM). Results: The CCM system demonstrated high performance (recovery rates exceeding 90% and WBC depletion rate of 99.9%) across a wide range of phenotypes (EpCAM(+), EpCAM(-), small-, large-sized, and cluster) and cancers (lung, breast, and bladder). Applied in 30 lung adenocarcinoma patients harboring epidermal growth factor receptor (EGFR) mutations, the system isolated diverse phenotypes of CTCs in marker expression and size, implying the importance of unbiased isolation. Genetic analyses of intra-patient samples comparing cell-free DNA with CCM-isolated CTCs yielded perfect concordance, and CTC enumeration using our technique was correlated with clinical progression as well as response to EGFR inhibitors. Conclusion: Our system also introduces technical advances which assure rapid, reliable, and reproducible results, thus enabling a more comprehensive application of robust CTC analysis in clinical practice.


Asunto(s)
Células Neoplásicas Circulantes , Automatización , Línea Celular Tumoral , Separación Celular/métodos , Molécula de Adhesión Celular Epitelial/genética , Receptores ErbB/genética , Humanos , Microfluídica/métodos , Células Neoplásicas Circulantes/metabolismo
9.
Stem Cells ; 28(10): 1816-28, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20979137

RESUMEN

Neuronal precursor cells (NPCs) are temporally regulated and have the ability to proliferate and differentiate into mature neurons, oligodendrocytes, and astrocytes in the presence of growth factors (GFs). In the present study, the role of the Jak pathway in brain development was investigated in NPCs derived from neurosphere cultures using Jak2 and Jak3 small interfering RNAs and specific inhibitors. Jak2 inhibition profoundly decreased NPC proliferation, preventing further differentiation into neurons and glial cells. However, Jak3 inhibition induced neuronal differentiation accompanied by neurite growth. This phenomenon was due to the Jak3 inhibition-mediated induction of neurogenin (Ngn)2 and NeuroD in NPCs. Jak3 inhibition induced NPCs to differentiate into scattered neurons and increased the expression of Tuj1, microtubule associated protein 2 (MAP2), Olig2, and neuroglial protein (NG)2, but decreased glial fibrillary acidic protein (GFAP) expression, with predominant neurogenesis/polydendrogenesis compared with astrogliogenesis. Therefore, Jak2 may be important for NPC proliferation and maintenance, whereas knocking-down of Jak3 signaling is essential for NPC differentiation into neurons and oligodendrocytes but does not lead to astrocyte differentiation. These results suggest that NPC proliferation and differentiation are differentially regulated by the Jak pathway.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Janus Quinasa 3/metabolismo , Neuronas/citología , Neuronas/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , Western Blotting , Encéfalo/embriología , Encéfalo/metabolismo , Diferenciación Celular/genética , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Inmunohistoquímica , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Janus Quinasa 3/antagonistas & inhibidores , Janus Quinasa 3/genética , Ratones , Neurogénesis/efectos de los fármacos , Neurogénesis/genética , Neuronas/efectos de los fármacos , Quinazolinas/farmacología , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/efectos de los fármacos , Tirfostinos/farmacología
10.
J Neurosci ; 29(18): 5974-84, 2009 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-19420263

RESUMEN

Neural-cadherin (N-cadherin), a member of the classical cadherin family of transmembrane glycoproteins, mediates cellular recognition and cell-cell adhesion through calcium-dependent homophilic interactions and plays important roles in the development and maintenance of the nervous system. Metalloproteinase is known to cleave N-cadherin, which is further cleaved by gamma-secretase. The intracellular domain of N-cadherin interacts with beta-catenin, and beta-catenin stability is critical for cell-cell adhesion and cell survival. In the present study, we showed that N-cadherin is cleaved specifically by calpain, resulting in the generation of a novel 110 kDa fragment. The cleavage occurred in ischemic brain lesions and in vitro neural cells in the presence of NMDA and ionomycin, and was restored by calpain inhibitors but not matrix metalloproteinase or gamma-secretase inhibitors. Calpain directly cleaved N-cadherin in in vitro calpain assays, and calpain inhibitors prevented its cleavage in a dose-dependent manner. Using N-cadherin deletion mutants, we found that calpain cleavage sites exist in at least four regions of the cytoplasmic domain. Treatment with NMDA induced neuronal death, and it suppressed the expression of surface N-cadherin and the N-cadherin/beta-catenin interaction, effects that were prevented by calpain inhibitor. Furthermore, calpain-mediated N-cadherin cleavage significantly affected cell-cell adhesion, AKT signaling, the N-cadherin/beta-catenin interaction and the Wnt target gene expressions through the accumulation of nuclear beta-catenin.


Asunto(s)
Lesiones Encefálicas/metabolismo , Cadherinas/metabolismo , Calpaína/metabolismo , Animales , Animales Recién Nacidos , Biotinilación/métodos , Lesiones Encefálicas/patología , Cadherinas/genética , Calcio/metabolismo , Calpaína/farmacología , Células Cultivadas , Modelos Animales de Enfermedad , Maleato de Dizocilpina/farmacología , Embrión de Mamíferos , Endocitosis/efectos de los fármacos , Endocitosis/fisiología , Inhibidores Enzimáticos/farmacología , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/genética , Técnicas In Vitro , Ionomicina/farmacología , Ionóforos/farmacología , Ratones , Modelos Biológicos , Mutación/genética , N-Metilaspartato/farmacología , Neuronas/efectos de los fármacos , Estructura Terciaria de Proteína/efectos de los fármacos , Estructura Terciaria de Proteína/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Fracciones Subcelulares/efectos de los fármacos , Transfección/métodos , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
11.
Cancer Discov ; 10(8): 1194-1209, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32414908

RESUMEN

EGFR exon 20 insertion driver mutations (Exon20ins) in non-small cell lung cancer (NSCLC) are insensitive to EGFR tyrosine kinase inhibitors (TKI). Amivantamab (JNJ-61186372), a bispecific antibody targeting EGFR-MET, has shown preclinical activity in TKI-sensitive EGFR-mutated NSCLC models and in an ongoing first-in-human study in patients with advanced NSCLC. However, the activity of amivantamab in Exon20ins-driven tumors has not yet been described. Ba/F3 cells and patient-derived cells/organoids/xenograft models harboring diverse Exon20ins were used to characterize the antitumor mechanism of amivantamab. Amivantamab inhibited proliferation by effectively downmodulating EGFR-MET levels and inducing immune-directed antitumor activity with increased IFNγ secretion in various models. Importantly, in vivo efficacy of amivantamab was superior to cetuximab or poziotinib, an experimental Exon20ins-targeted TKI. Amivantamab produced robust tumor responses in two Exon20ins patients, highlighting the important translational nature of this preclinical work. These findings provide mechanistic insight into the activity of amivantamab and support its continued clinical development in Exon20ins patients, an area of high unmet medical need. SIGNIFICANCE: Currently, there are no approved targeted therapies for EGFR Exon20ins-driven NSCLC. Preclinical data shown here, together with promising clinical activity in an ongoing phase I study, strongly support further clinical investigation of amivantamab in EGFR Exon20ins-driven NSCLC.This article is highlighted in the In This Issue feature, p. 1079.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Animales , Anticuerpos Biespecíficos/farmacología , Antineoplásicos Inmunológicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Exones , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Mutación , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/metabolismo
12.
Eur J Nutr ; 48(4): 235-42, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19234663

RESUMEN

BACKGROUND: Although there is growing awareness of the beneficial potential of onion intake to lower the risk of cardiovascular disease, there is little information about the effect of onion on ischemic heart injury, one of the most common cardiovascular diseases. AIM OF THE STUDY: This study investigates the effect of the methanol-soluble extract of onion on ischemic injury in heart-derived H9c2 cells in vitro and in rat hearts in vivo. The underlying mechanism is also investigated. METHODS: To evaluate the effect of onion on ischemia-induced cell death, LDH release and TUNEL-positivity were assessed in H9c2 cells, and the infarct size was measured in a myocardial infarct model. To investigate the mechanism of the cardioprotection by onion, the reactive oxygen species (ROS) level and the mitochondrial membrane potential (DeltaPsi(m)) were measured using an imaging technique; the caspase-3 activity was assayed, and Western blotting was performed to examine cytochrome c release in H9c2 cells. RESULTS: The methanolic extract of onion had a preventive effect on ischemia/hypoxia-induced apoptotic death in H9c2 cells in vitro and in rat heart in vivo. The onion extract (0.05 g/ml) inhibited the elevation of the ROS, mitochondrial membrane depolarization, cytochrome c release and caspase-3 activation during hypoxia in H9c2 cells. In the in vivo rat myocardial infarction model, onion extract (10 g/kg) significantly reduced the infarct size, the apoptotic cell death of the heart and the plasma MDA level. CONCLUSION: In conclusion, the results of this study suggest that the methanolic extract of onion attenuates ischemia/hypoxia-induced apoptosis in heart-derived H9c2 cells in vitro and in rat hearts in vivo, through, at least in part, an antioxidant effect.


Asunto(s)
Apoptosis/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Isquemia/tratamiento farmacológico , Cebollas/química , Extractos Vegetales/farmacología , Animales , Antioxidantes/farmacología , Western Blotting , Caspasa 3/metabolismo , Vasos Coronarios , Citocromos c/metabolismo , Humanos , Etiquetado Corte-Fin in Situ , Malondialdehído/sangre , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo
13.
J Chem Phys ; 131(14): 144702, 2009 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-19831459

RESUMEN

We have examined the diffusion and agglomeration of Au adatoms on the H-terminated Si(111)-(1x1) surface using periodic slab density functional theory calculations. We find that a single Au atom favorably resides atop a surface Si atom by breaking an original identical withSi-H bond while the H atom is bonded to the Au atom in the vertical direction, leading to the identical withSi-Au-H state. Starting from the most favorable on-top (T) site, a Au adatom is predicted to undergo diffusion by moving in and out of the T site without disrupting surface Si-H bonds. The predicted overall activation energy for the Au diffusion is 0.5 eV. Our calculations show that Au agglomeration leads to libration of H atoms from the Au/Si interface, while the H atoms are weakly bound to Au clusters and subsequently undergo associative H(2) desorption with no significant barrier. Based on charge density analysis we also discuss bonding mechanisms for Au on H-terminated Si(111)-(1x1). Our findings are as a whole consistent with experimental results available in literature.

14.
Biomaterials ; 197: 51-59, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30640137

RESUMEN

Mitochondria are the primary organelle of regulating apoptosis, and intracellular calcium ions are a key component of pro-apoptosis induction. Herein, we report an artificial apoptosis-inducing polypeptide that destabilizes the mitochondrial membrane and transports calcium ions into the cytosol, thereby synergistically creating severe oxidative conditions. The oxidative stress highly activates an apoptotic signaling cascade, and also inhibits cell migration and invasion in vitro and in vivo. The suggested strategy for simultaneous mitochondrial disruption and perturbed calcium homeostasis demonstrates the applicability of polypeptide-based therapeutics as potent apoptosis-inducers in cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Membranas Mitocondriales/efectos de los fármacos , Péptidos/farmacología , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Desarrollo de Medicamentos , Humanos , Ratones , Membranas Mitocondriales/metabolismo , Membranas Mitocondriales/patología , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Estrés Oxidativo/efectos de los fármacos , Péptidos/química , Péptidos/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo
15.
Adv Sci (Weinh) ; 6(14): 1801995, 2019 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-31380199

RESUMEN

Perturbation of potassium homeostasis can affect various cell functions and lead to the onset of programmed cell death. Although ionophores have been intensively used as an ion homeostasis disturber, the mechanisms of cell death are unclear and the bioapplicability is limited. In this study, helical polypeptide-based potassium ionophores are developed to induce endoplasmic reticulum (ER) stress-mediated apoptosis. The polypeptide-based potassium ionophores disturb ion homeostasis and then induce prolonged ER stress in the cells. The ER stress results in oxidative environments that accelerate the activation of mitochondria-dependent apoptosis. Moreover, ER stress-mediated apoptosis is triggered in a tumor-bearing mouse model that suppresses tumor proliferation. This study provides the first evidence showing that helical polypeptide-based potassium ionophores trigger ER stress-mediated apoptosis by perturbation of potassium homeostasis.

16.
Brain Res ; 1193: 153-61, 2008 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-18178179

RESUMEN

COX-2 and prostaglandins (PGs) might play important roles in epilepsy. In kainic acid-induced seizures, the brain largely increases PGD(2), first from COX-1 and later COX-2-induced PGF(2alpha). Pre-treatment with COX-2 inhibitors such as indomethacin, nimesulide, and celecoxib is known to aggravate kainic acid (KA)-induced seizure activity. However it is not known whether the proconvulsant effect of those non-steroidal anti-inflammatory drugs (NSAIDs) is due to changes in endogenous prostaglandins (PGs), or what types of PGs are involved. The purpose of this study was to determine the effect of intracisternally administered PGs on KA-induced seizures aggravated by pre- or post-treatment with COX-2 inhibitors. Systemic KA injection (10 mg/kg i.p.) in mice evoked mild seizure activity within 15 min. PGs were administrated intracisternally 20 min prior to KA administration. COX inhibitors (indomethacin, nimesulide, and ketoprofen, 10 mg/kg i.p.) were injected 1 h before or 15 min after KA. An additional COX-2 inhibitor, celecoxib, was administered orally. Intracisternally administered PGF(2alpha) (700 ng), but not PGD(2) (700 ng) or PGE(2) (700 ng) completely alleviated KA-induced seizures potentiated by COX-2 inhibitors, and also reduced KA-induced hippocampal neuronal death aggravated by indomethacin. PGF(2alpha) alone did not affect KA-induced seizures. However, an FP receptor antagonist, AL 8810 (10 or 50 ng) which is an 11beta-fluoro analogue of PGF(2alpha) potentiated KA-induced seizure activity dose-dependently. In summary, pre- or post-treatment with COX-2 inhibitors aggravates KA-induced seizures, which suggests to change the endogenous PGF(2alpha). Seizure-induced PGF(2alpha) might act as an endogenous anticonvulsant through FP receptors.


Asunto(s)
Inhibidores de la Ciclooxigenasa/administración & dosificación , Dinoprost/metabolismo , Dinoprost/uso terapéutico , Receptores de Prostaglandina/metabolismo , Convulsiones/metabolismo , Convulsiones/prevención & control , Análisis de Varianza , Animales , Supervivencia Celular/efectos de los fármacos , Dinoprost/administración & dosificación , Dinoprost/análogos & derivados , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Electroencefalografía/métodos , Ácido Kaínico , Masculino , Ratones , Ratones Endogámicos ICR , Receptores de Prostaglandina/antagonistas & inhibidores , Convulsiones/inducido químicamente , Convulsiones/patología
17.
Eur J Pharmacol ; 578(1): 11-8, 2008 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-17961543

RESUMEN

Cadmium (Cd), an ubiquitous heavy metal, is known to be accumulated outside of the blood-brain barrier. In this study, we investigated whether Cd has cytotoxicity in mouse brain microvascular endothelial cells (bEnd.3). Results from the cell viability assay showed that Cd caused a remarkable decrease in cell viability in a dose-dependent manner. The cell death induced by Cd appeared to involve apoptosis, based on our results from annexin V staining, electron microscopy and TUNEL staining. And the cell death induced by Cd was inhibited by caspase inhibitor ZVAD-fmk. To further investigate the mechanism of the Cd-induced cell death, we examined the effects of selective inhibitors for mitogen activated protein kinase (MAPK) pathways on the cell death. The Cd-induced cell death was significantly inhibited by p38 MAPK inhibitor SB202190, but not by either, c-Jun N-terminal kinase (JNK) inhibitor SP600125 or extracellular signal-regulated kinase (ERK) inhibitor U0126. Phosphorylations of p38 MAPK, JNK and ERK were stimulated by treatment with CdCl(2). In summary, our results suggest that Cd can induce apoptotic cell death, at least in part, through the p38 MAPK pathway in brain microvascular endothelial cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Cloruro de Cadmio/toxicidad , Endotelio Vascular/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Animales , Anexina A5/metabolismo , Barrera Hematoencefálica/metabolismo , Encéfalo/irrigación sanguínea , Cloruro de Cadmio/administración & dosificación , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Etiquetado Corte-Fin in Situ , Ratones , Microcirculación/citología , Microcirculación/efectos de los fármacos , Microcirculación/metabolismo , Microscopía Electrónica , Fosforilación/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
Neuroscience ; 340: 487-500, 2017 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-27845178

RESUMEN

Deregulation of glutamate homeostasis is associated with degenerative neurological disorders. Glutamate dehydrogenase (GDH) is important for glutamate metabolism and plays a central role in expanding the pool of tricarboxylic acid (TCA) cycle intermediate alpha-ketoglutarate (α-KG), which improves overall bioenergetics. Under high energy demand, maintenance of ATP production results in functionally active mitochondria. Here, we tested whether the modulation of GDH activity can rescue ischemia/reperfusion-induced neuronal death in an in vivo mouse model of middle artery occlusion and an in vitro oxygen/glucose depletion model. Iodoacetate, an inhibitor of glycolysis, was also used in a model of energy failure, remarkably depleting ATP and α-KG. To stimulate GDH activity, the GDH activator 2-aminobicyclo-(2,2,1)-heptane-2-carboxylic acid and potential activator beta-lapachone were used. The GDH activators restored α-KG and ATP levels in the injury models and provided potent neuroprotection. We also found that beta-lapachone increased glutamate utilization, accompanied by a reduction in extracellular glutamate. Thus, our hypothesis that mitochondrial GDH activators increase α-KG production as an alternative energy source for use in the TCA cycle under energy-depleted conditions was confirmed. Our results suggest that increasing GDH-mediated glutamate oxidation represents a new therapeutic intervention for neurodegenerative disorders, including stoke.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Glutamato Deshidrogenasa/metabolismo , Naftoquinonas/farmacología , Fármacos Neuroprotectores/farmacología , Daño por Reperfusión/tratamiento farmacológico , Animales , Astrocitos/efectos de los fármacos , Astrocitos/enzimología , Astrocitos/patología , Encéfalo/enzimología , Encéfalo/patología , Isquemia Encefálica/enzimología , Isquemia Encefálica/patología , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media , Masculino , Ratones Endogámicos ICR , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Neuronas/efectos de los fármacos , Neuronas/enzimología , Neuronas/patología , Distribución Aleatoria , Daño por Reperfusión/enzimología , Daño por Reperfusión/patología
19.
Neuroreport ; 28(14): 929-935, 2017 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-28817455

RESUMEN

During spinal cord development, endogenous progenitors expressing nestin can migrate into the target and differentiate into neurons and other glial cells. Microglial cells can also be derived from nestin progenitor cells, even in the adult brain. Knockdown of Jak kinase 3 (Jak3) signaling can increase neurogenesis with longer neurite outgrowth in cortical progenitor cells. This study investigated the effect of Jak3 signaling on differentiation from nestin progenitor cells using E13.5 spinal progenitor cell cultures. In growth factors-enriched culture, developing neurons could not survive after several days and also a significant proportion of nestin-expressing cells transformed into ameboid Iba1 microglial cells, which increased exponentially after 5 days. This microgliogenesis was predominantly regulated by Jak3 signaling, which was confirmed by transcription factors responsible for microgliogenesis, and microglial migration and phagocytosis, such as Pu.1, Irf8, and Runx1. Jak3 inhibition also significantly increased the Tuj1 growing neurites with little microglial activation. These results indicated that neuronal and microglial cell differentiation was regulated primarily by Jak3 signaling and the developing neurons and neurite outgrowth might also be regulated by Jak3-dependent microglial activity.


Asunto(s)
Diferenciación Celular/fisiología , Janus Quinasa 3/metabolismo , Microglía/metabolismo , Nestina/metabolismo , Células-Madre Neurales/metabolismo , Animales , Proteínas de Unión al Calcio/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Péptidos y Proteínas de Señalización Intercelular/administración & dosificación , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Janus Quinasa 3/antagonistas & inhibidores , Janus Quinasa 3/genética , Ratones Endogámicos ICR , Proteínas de Microfilamentos/metabolismo , Microglía/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Médula Espinal/efectos de los fármacos , Médula Espinal/embriología , Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/metabolismo
20.
J Control Release ; 264: 24-33, 2017 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-28778477

RESUMEN

Artificial cationic helical peptides possess an enhanced cell-penetrating property. However, their cell-penetrability is not converted by cellular environmental changes resulting in nonspecific uptake. In this study, pH-sensitive anion-donating groups were added to a helical polypeptide to simultaneously achieve tumor targeting and pro-apoptotic activity. The mitochondria-destabilizing helical polypeptide undergoing pH-dependent conformational transitions selectively targeted cancer cells consequently disrupting mitochondrial membranes and subsequently inducing apoptosis. This work presents a promising peptide therapeutic system for cancer therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Péptidos/uso terapéutico , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Humanos , Concentración de Iones de Hidrógeno , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones Endogámicos BALB C , Ratones Desnudos , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Péptidos/química , Conformación Proteica , Especies Reactivas de Oxígeno/metabolismo , Carga Tumoral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA