Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 145
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Med Sci Monit ; 30: e944427, 2024 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-38851875

RESUMEN

BACKGROUND Emergence agitation, or delirium, occurs during early recovery from general anesthesia and involves disorientation, excitation, and uncontrolled physical movements. Dexmedetomidine is an alpha agonist that has sedative, anxiolytic, analgesic, and sympatholytic activities and is used as a continuous infusion to prevent emergence agitation. This study aimed to evaluate patients aged 65 years and older undergoing general anesthesia to determine the 90% effective dose (ED90) of dexmedetomidine continuous intraoperative infusion to prevent emergence agitation. MATERIAL AND METHODS We enrolled 44 patients aged 65 years and older undergoing spinal surgery under general anesthesia. Dexmedetomidine administration commenced 30 minutes before surgery completion, with a predetermined infusion dose (µg/kg/h), without a loading dose. The initial dose was 0.2 µg/kg/h, and subsequent step size was ±0.05 µg/kg/h. We tried to find ED90 of dexmedetomidine using the biased-coin design. Vital signs, extubation quality scores, extubation-related complications, and postoperative outcomes were monitored. RESULTS Dexmedetomidine ED90 for smooth emergence in older patients was 0.34 µg/kg/h. Peri-extubation vital signs remained within 20% of baseline values, without requiring pharmacological intervention. No hypoxia, hypoventilation, or post-extubation agitation occurred. In the recovery room, 1 patient briefly exhibited excitement but quickly calmed. Nine patients initially unresponsive in the recovery room fully awoke and were promptly discharged. CONCLUSIONS For older patients who are vulnerable to adverse effects of anesthetics and opioids, dexmedetomidine enables gentle awakening without adverse vital sign changes, respiratory depression, excessive sedation, or emergence agitation (ED90=0.34 µg/kg/h). Further studies should involve a larger patient cohort, considering diverse medical conditions in older individuals.


Asunto(s)
Periodo de Recuperación de la Anestesia , Dexmedetomidina , Hipnóticos y Sedantes , Humanos , Dexmedetomidina/administración & dosificación , Dexmedetomidina/farmacología , Anciano , Masculino , Femenino , Hipnóticos y Sedantes/administración & dosificación , Anestesia General/métodos , Columna Vertebral/cirugía , Anciano de 80 o más Años , Relación Dosis-Respuesta a Droga , Delirio del Despertar/prevención & control
2.
Neurobiol Dis ; 177: 105982, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36592864

RESUMEN

Neuregulin-1 (NRG1) is an epidermal growth factor family member with essential roles in the developing and adult nervous systems. In recent years, establishing evidence has collectively suggested that NRG1 is a new modulator of central nervous system (CNS) injury and disease, with multifaceted roles in neuroprotection, remyelination, neuroinflammation, and other repair mechanisms. NRG1 signaling exerts its effects via the tyrosine kinase receptors ErbB2-ErbB4. The NRG1/ErbB network in CNS pathology and repair has evolved, primarily in recent years. In the present study, we demonstrated that a unilateral microinjection of CoCl2 into the ventral hippocampus (vHPC) induced hypoxic insult and led to anxiety-related behaviors and deficit sociability in mice. NRG1 treatment significantly alleviated the CoCl2-induced increase of hypoxic-related molecules and behavioral abnormalities. Furthermore, NRG1 reduced the CoCl2-induced neuroinflammation and neuronal deficits in the vHPC or primary hippocampal neurons in mice. Collectively, these results suggest that NRG1 ameliorates hypoxia by alleviating synaptic deficits and behavioral abnormalities of the CoCl2-induced vHPC hypoxic model.


Asunto(s)
Neurregulina-1 , Enfermedades Neuroinflamatorias , Ratones , Animales , Neurregulina-1/metabolismo , Hipocampo/metabolismo , Conducta Social , Ansiedad/tratamiento farmacológico
3.
Pharmacol Rev ; 72(3): 639-667, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32554488

RESUMEN

Cancer and organ injury-such as that occurring in the perioperative period, including acute lung injury, myocardial infarction, and acute gut injury-are among the leading causes of death in the United States and impose a significant impact on quality of life. MicroRNAs (miRNAs) have been studied extensively during the last two decades for their role as regulators of gene expression, their translational application as diagnostic markers, and their potential as therapeutic targets for disease treatment. Despite promising preclinical outcomes implicating miRNA targets in disease treatment, only a few miRNAs have reached clinical trials. This likely relates to difficulties in the delivery of miRNA drugs to their targets to achieve efficient inhibition or overexpression. Therefore, understanding how to efficiently deliver miRNAs into diseased tissues and specific cell types in patients is critical. This review summarizes current knowledge on various approaches to deliver therapeutic miRNAs or miRNA inhibitors and highlights current progress in miRNA-based disease therapy that has reached clinical trials. Based on ongoing advances in miRNA delivery, we believe that additional therapeutic approaches to modulate miRNA function will soon enter routine medical treatment of human disease, particularly for cancer or perioperative organ injury. SIGNIFICANCE STATEMENT: MicroRNAs have been studied extensively during the last two decades in cancer and organ injury, including acute lung injury, myocardial infarction, and acute gut injury, for their regulation of gene expression, application as diagnostic markers, and therapeutic potentials. In this review, we specifically emphasize the pros and cons of different delivery approaches to modulate microRNAs, as well as the most recent exciting progress in the field of therapeutic targeting of microRNAs for disease treatment in patients.


Asunto(s)
MicroARNs/genética , Neoplasias/genética , Heridas y Lesiones/genética , Animales , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/genética , Humanos , MicroARNs/biosíntesis , MicroARNs/sangre , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/terapia , Heridas y Lesiones/metabolismo , Heridas y Lesiones/patología , Heridas y Lesiones/terapia
4.
Ann Neurol ; 89(5): 926-941, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33675084

RESUMEN

OBJECTIVE: Brain arteriovenous malformations (bAVMs) are a leading cause of hemorrhagic stroke and neurological deficits in children and young adults, however, no pharmacological intervention is available to treat these patients. Although more than 95% of bAVMs are sporadic without family history, the pathogenesis of sporadic bAVMs is largely unknown, which may account for the lack of therapeutic options. KRAS mutations are frequently observed in cancer, and a recent unprecedented finding of these mutations in human sporadic bAVMs offers a new direction in the bAVM research. Using a novel adeno-associated virus targeting brain endothelium (AAV-BR1), the current study tested if endothelial KRASG12V mutation induces sporadic bAVMs in mice. METHODS: Five-week-old mice were systemically injected with either AAV-BR1-GFP or -KRASG12V . At 8 weeks after the AAV injection, bAVM formation and characteristics were addressed by histological and molecular analyses. The effect of MEK/ERK inhibition on KRASG12V -induced bAVMs was determined by treatment of trametinib, a US Food and Drug Administration (FDA)-approved MEK/ERK inhibitor. RESULTS: The viral-mediated KRASG12V overexpression induced bAVMs, which were composed of a tangled nidus mirroring the distinctive morphology of human bAVMs. The bAVMs were accompanied by focal angiogenesis, intracerebral hemorrhages, altered vascular constituents, neuroinflammation, and impaired sensory/cognitive/motor functions. Finally, we confirmed that bAVM growth was inhibited by trametinib treatment. INTERPRETATION: Our innovative approach using AAV-BR1 confirms that KRAS mutations promote bAVM development via the MEK/ERK pathway, and provides a novel preclinical mouse model of bAVMs which will be useful to develop a therapeutic strategy for patients with bAVM. ANN NEUROL 2021;89:926-941.


Asunto(s)
Endotelio Vascular , Malformaciones Arteriovenosas Intracraneales/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Animales , Cognición , Dependovirus/genética , Encefalitis/genética , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Regulación de la Expresión Génica/genética , Humanos , Malformaciones Arteriovenosas Intracraneales/complicaciones , Malformaciones Arteriovenosas Intracraneales/psicología , Hemorragias Intracraneales/etiología , Hemorragias Intracraneales/genética , Imagen por Resonancia Magnética , Ratones , Mutación/genética , Neovascularización Patológica/etiología , Neovascularización Patológica/genética , Desempeño Psicomotor , Piridonas/farmacología , Pirimidinonas/farmacología
5.
Exp Mol Pathol ; 120: 104622, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33684392

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by neuronal and synaptic loss. The cytoplasmic tail of amyloid precursor protein (APP) undergoes sequential cleavage at a specific intracellular caspase site to generate the cytoplasmic terminal 31 (CT31) fragment. The APP-CT31 fragment is a potent inducer of apoptosis. The cytotoxicity of APP-CT31 in SH-SY5Y cells was evaluated by the lactate dehydrogenase (LDH) assay. TUNEL staining was used to detect apoptotic signals in SH-SY5Y cells and primary cortical neurons. The expression of apoptosis-related proteins, such as p53, PUMA (p53 up-regulated modulator of apoptosis), and cleaved was investigated by immunofluorescence analysis and Western blotting. In this study, we investigated the neuroprotective effect of neuregulin 1 (NRG1) against cytotoxicity induced by APP-CT31. Our data showed that CT31 induced cytotoxicity and apoptosis in SH-SY5Y cells and primary cortical neurons. NRG1 attenuated the neurotoxicity induced by the expression of APP-CT31. We also showed that APP-CT31 altered the expression of p53 and cleaved caspase 3. However, treatment with NRG1 rescued the APP-CT31-induced upregulation of p53 and cleaved caspase 3 expression. The protective effect of NRG1 was abrogated by inhibition of the ErbB4 receptor and Akt. These results indicate an important role of ErbB4/Akt signaling in NRG1-mediated neuroprotection, suggesting that endogenous NRG1/ErbB4 signaling represents a valuable therapeutic target in AD.


Asunto(s)
Precursor de Proteína beta-Amiloide/efectos adversos , Neurregulina-1/metabolismo , Neuroblastoma/prevención & control , Fármacos Neuroprotectores/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor ErbB-4/metabolismo , Apoptosis , Proliferación Celular , Humanos , Neurregulina-1/genética , Neuroblastoma/etiología , Neuroblastoma/patología , Dominios Proteicos , Proteínas Proto-Oncogénicas c-akt/genética , Receptor ErbB-4/genética , Células Tumorales Cultivadas
6.
Med Sci Monit ; 27: e928538, 2021 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-33428608

RESUMEN

BACKGROUND The flexibility of the long flexometallic tube makes insertion of the flexible reinforced laryngeal mask airway (f-LMA) difficult. We compared the usefulness of rigid lightwand-guided f-LMA insertion with standard digital manipulation. MATERIAL AND METHODS Fifty-four patients (aged 19-70 years) were randomly divided into a control group (digital manipulation technique) or the lightwand group (lightwand-guided insertion technique). The insertion profiles, oropharyngeal leak pressure (OLP), peak inspiratory pressure (PIP), expiratory tidal volume, and ventilatory score were measured in patients with neutral, extension, rotation, flexion, and re-neutral head-neck positions in turn. RESULTS The success rate and ease of insertion did not differ between groups, but the insertion time was longer in the lightwand group. The fiberoptic laryngeal view was significantly better in the lightwand group than in the control group. However, the OLP, PIP, expiratory tidal volume, and ventilatory scores were not significantly different between groups according to head-neck positions. The extension posture was associated with a significant negative effect on ventilation, but ventilation returned to initial levels with the other postures. CONCLUSIONS Lightwand-guided f-LMA insertion showed a better fiberoptic laryngeal view than standard digital manipulation, but no improvement in the ventilatory state was observed due to position. Therefore, lightwand-guided insertion could facilitate correct placement of the f-LMA, but it has limited clinical usefulness.


Asunto(s)
Tecnología de Fibra Óptica/métodos , Intubación Intratraqueal/métodos , Adulto , Anciano , Femenino , Tecnología de Fibra Óptica/instrumentación , Humanos , Intubación Intratraqueal/efectos adversos , Intubación Intratraqueal/instrumentación , Laringe/diagnóstico por imagen , Laringe/fisiopatología , Masculino , Persona de Mediana Edad , Posicionamiento del Paciente/métodos , Ventilación Pulmonar , Volumen de Ventilación Pulmonar
7.
Proc Natl Acad Sci U S A ; 115(19): 4927-4932, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29686060

RESUMEN

In the present work, we investigated the role of natural killer (NK) cells in combination therapy with oncolytic virus (OV) and bortezomib, a proteasome inhibitor. NK cells display rapid and potent immunity to metastatic and hematological cancers, and they overcome immunosuppressive effects of tumor microenvironment. We developed a mathematical model to address the question of how the density of NK cells affects the growth of the tumor. We found that the antitumor efficacy increases when the endogenous NKs are depleted and also when exogenous NK cells are injected into the tumor. These predictions were validated by our in vivo and in vitro experiments.


Asunto(s)
Bortezomib/uso terapéutico , Neoplasias Hematológicas , Células Asesinas Naturales/inmunología , Modelos Inmunológicos , Viroterapia Oncolítica , Microambiente Tumoral , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/patología , Neoplasias Hematológicas/terapia , Humanos , Células Asesinas Naturales/patología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Células Vero
8.
FASEB J ; 33(10): 11035-11044, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31287961

RESUMEN

Dendritic cells (DCs) are the most potent professional antigen (Ag)-presenting cells and inducers of T cell-mediated immunity. A previous microarray analysis identified PDZ and LIM domain protein 4 (Pdlim4) as a candidate marker for DC maturation. The aim of this study was to investigate whether Pdlim4 influences DC migration and maturation. Mouse bone marrow-derived DCs were transduced lentivirally with Pdlim4 short hairpin RNA and examined by confocal microscopy, flow cytometry, ELISA, and Western blotting. Pdlim4 was highly induced in LPS-stimulated mature DCs (mDCs). Pdlim4-knockdown mDCs showed reduced expression of molecules associated with Ag presentation and T-cell costimulation, reduced cytokine production, and functional defects in their ability to activate T cells. Moreover, Pdlim4 was necessary for mDC migration via C-C chemokine receptor type 7 (CCR7)-JNK in in vitro Transwell assays. The importance of Pdlim4 in DC migration was confirmed with an in vivo migration model in which C57BL/6 mice were injected with fluorescently labeled DCs in the footpad and migration to the popliteal lymph nodes was assessed by flow cytometry. Moreover, dendrite formation in mDCs was remarkably attenuated under Pdlim4 knockdown. Taken together, these results demonstrate that Pdlim4 is necessary for DC migration via CCR7-JNK, dendrite formation, and subsequent development of functional T-cell responses.-Yoo, J.-Y., Jung, N.-C., Lee, J.-H., Choi, S.-Y., Choi, H.-J., Park, S.-Y., Jang, J.-S., Byun, S.-H., Hwang, S.-U., Noh, K.-E., Park, Y., Lee, J., Song, J.-Y., Seo, H. G., Lee, H. S., Lim, D.-S. Pdlim4 is essential for CCR7-JNK-mediated dendritic cell migration and F-actin-related dendrite formation.


Asunto(s)
Movimiento Celular , Células Dendríticas/metabolismo , Proteínas con Dominio LIM/metabolismo , Proteínas de Microfilamentos/metabolismo , Actinas/metabolismo , Animales , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/fisiología , Proteínas con Dominio LIM/genética , Activación de Linfocitos , MAP Quinasa Quinasa 4/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/genética , Receptores CCR7/metabolismo
9.
Immunity ; 35(5): 819-31, 2011 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-22078798

RESUMEN

Early events in atherosclerosis occur in the aortic intima and involve monocytes that become macrophages. We looked for these cells in the steady state adult mouse aorta, and surprisingly, we found a dominance of dendritic cells (DCs) in the intima. In contrast to aortic adventitial macrophages, CD11c(+)MHC II(hi) DCs were poorly phagocytic but were immune stimulatory. DCs were of two types primarily: classical Flt3-Flt3L signaling-dependent, CD103(+)CD11b(-) DCs and macrophage-colony stimulating factor (M-CSF)-dependent, CD14(+)CD11b(+)DC-SIGN(+) monocyte-derived DCs. Both types expanded during atherosclerosis. By crossing Flt3(-/-) to Ldlr(-/-) atherosclerosis-prone mice, we developed a selective and marked deficiency of classical CD103(+) aortic DCs, and they were associated with exacerbated atherosclerosis without alterations in blood lipids. Concomitantly, the Flt3(-/-)Ldlr(-/-) mice had fewer Foxp3(+) Treg cells and increased inflammatory cytokine mRNAs in the aorta. Therefore, functional DCs are dominant in normal aortic intima and, in contrast to macrophages, CD103(+) classical DCs are associated with atherosclerosis protection.


Asunto(s)
Aterosclerosis/inmunología , Células Dendríticas/inmunología , Transducción de Señal , Tirosina Quinasa 3 Similar a fms/metabolismo , Animales , Antígenos CD/metabolismo , Aorta/efectos de los fármacos , Aorta/inmunología , Aterosclerosis/genética , Aterosclerosis/patología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Regulación de la Expresión Génica/inmunología , Procedimientos de Reducción del Leucocitos , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/inmunología , Proteínas de la Membrana/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/inmunología , Tirosina Quinasa 3 Similar a fms/genética
10.
J Pharmacol Sci ; 137(2): 146-153, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29914798

RESUMEN

The amyloid precursor protein (APP) is a key molecule in Alzheimer's disease. The prevailing view is that APP is initially transported to the plasma membrane as a full-length protein. Its localization at the cell surface can trigger downstream signaling and APP cleavage. Our previous work has shown that Neuregulin 1 (NRG1) has neuroprotective effects in an Alzheimer's disease model. In the present study, we examine whether NRG1 signaling is involved in APP expression and non-amyloidogenic processing in neuronal cells. Here we show that NRG1 increased the cell surface expression of APP without changing the total amount of APP mRNA or protein expression in SH-SY5Y cells and in rat primary cortical neurons. In addition, NRG1 significantly increased the levels of the secreted form of APP, sAPPα, in the conditioned media but did not change the expression of ADAM10 on the cell surface or in the cell lysates. Furthermore, we found that the protein level of NRG1 was reduced in the hippocampus of Alzheimer's disease (AD) patients. Our results demonstrate that NRG1 increased APP expression on the cell surface and sAPPα secretion into the media of neuronal cell cultures. Taken together, these results suggest a role for NRG1 in non-amyloidogenic processing.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Neurregulina-1/fisiología , Neuronas/metabolismo , Transducción de Señal/fisiología , Proteína ADAM10/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Membrana Celular/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Expresión Génica/genética , Proteínas de la Membrana/metabolismo , Neurregulina-1/metabolismo , Fragmentos de Péptidos/metabolismo , Ratas , Ratas Sprague-Dawley , Fracciones Subcelulares/metabolismo
11.
Mol Ther ; 25(7): 1544-1555, 2017 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-28109960

RESUMEN

Targeted inhibition of oncogenic miRNA-21 has been proposed to treat glioblastoma by rescuing tumor suppressors, PTEN and PDCD4. However, systemic delivery of anti-miR-21 sequences requires a robust and efficient delivery platform to successfully inhibit this druggable target. Three-way-junction (3WJ)-based RNA nanoparticles (RNP), artificially derived from pRNA of bacteriophage phi29 DNA packaging motor, was recently shown to target glioblastoma. Here, we report that multi-valent folate (FA)-conjugated 3WJ RNP constructed to harbor anti-miR-21 LNA sequences (FA-3WJ-LNA-miR21) specifically targeted and delivered anti-miR-21 LNA and knocked down miR-21 expression in glioblastoma cells in vitro and in vivo with favorable biodistribution. Systemically injected FA-3WJ-LNA-miR21 RNP efficiently rescued PTEN and PDCD4, resulting in glioblastoma cell apoptosis and tumor growth regression. Overall survival rate was also significantly improved by FA-3WJ-LNA-miR21 RNP. These results are indicative of the clinical benefit of FA-3WJ RNP-based gene therapy for the successful targeted therapy of developing and even recurring glioblastoma.


Asunto(s)
Antagomirs/farmacología , Neoplasias Encefálicas/terapia , Regulación Neoplásica de la Expresión Génica , Glioblastoma/terapia , MicroARNs/antagonistas & inhibidores , Nanopartículas/administración & dosificación , Animales , Antagomirs/química , Antagomirs/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Portadores de Fármacos , Femenino , Receptores de Folato Anclados a GPI/genética , Receptores de Folato Anclados a GPI/metabolismo , Ácido Fólico/química , Ácido Fólico/metabolismo , Glioblastoma/genética , Glioblastoma/mortalidad , Glioblastoma/patología , Humanos , Ratones , Ratones Desnudos , MicroARNs/genética , MicroARNs/metabolismo , Nanopartículas/química , Oligonucleótidos/química , Oligonucleótidos/metabolismo , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Análisis de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
12.
J Gene Med ; 19(3)2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28087981

RESUMEN

BACKGROUND: Malignant gliomas (glioblastomas; GBMs) are extremely aggressive and have a median survival of approximately 15 months. Current treatment modalities, which include surgical resection, radiation and chemotherapy, have done little to prolong the lives of GBM patients. Chondroitin sulfate proteoglycans (CSPG) are critical for cell-cell and cell-extracellular matrix (ECM) interactions and are implicated in glioma growth and invasion. Chondroitinase (Chase) ABC is a bacterial enzyme that cleaves chondroitin sulfate disaccharide chains from CSPGs in the tumor ECM. Wild-type Chase ABC has limited stability and/or activity in mammalian cells; therefore, we created a mutant humanized version (Chase M) with enhanced function in mammalian cells. METHODS: We hypothesized that disruption of cell-cell and cell-ECM interactions by ChaseM and temozolomide (TMZ) will enhance the chemotherapeutic availability and sensitivity of glioma cells. RESULTS: Utilizing primary patient-derived neurospheres, we found that ChaseM decreases glioma neurosphere aggregation in vitro. Furthermore, an oncolytic HSV-1 virus expressing secreted ChaseM (OV-ChaseM) enhanced viral spread and glioma cell killing compared to OV-Control, in vitro. OV-ChaseM plus TMZ combinatorial treatment resulted in a significant synergistic enhancement of glioma cell killing accompanied by an increase in apoptotic cell death. Intracellular flow cytometric analysis revealed a significant reduction in the phosphorylation of the pro-survival AKT protein following OV-ChaseM plus TMZ treatment. Lastly, in nude mice bearing intracranial GBM30 glioma xenografts, intratumoral OV-ChaseM plus TMZ (10 mg/kg by oral gavage) combination therapy resulted in a significant (p < 0.02) enhancement of survival compared to each individual treatment alone. CONCLUSIONS: These data reveal that OV-ChaseM enhances glioma cell viral susceptibility and sensitivity to TMZ.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Condroitina ABC Liasa/genética , Dacarbazina/análogos & derivados , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Glioblastoma/genética , Alelos , Sustitución de Aminoácidos , Animales , Apoptosis/genética , Línea Celular Tumoral , Supervivencia Celular/genética , Chlorocebus aethiops , Condroitina ABC Liasa/metabolismo , Dacarbazina/farmacología , Modelos Animales de Enfermedad , Activación Enzimática , Expresión Génica , Vectores Genéticos/genética , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patología , Herpesvirus Humano 1/genética , Humanos , Ratones , Mutación , Viroterapia Oncolítica , Temozolomida , Transducción Genética , Resultado del Tratamiento , Carga Tumoral , Células Tumorales Cultivadas , Células Vero , Ensayos Antitumor por Modelo de Xenoinjerto
13.
J Fluoresc ; 27(6): 2187-2193, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28791525

RESUMEN

Genetically encoded calcium indicators (GECI) such as GCaMP3 are attracting significant attention as a good option for measuring intracellular calcium levels. Recently, a modified GCaMP3 called dCys-GCaMP3 was developed by replacing two threonine residues with cysteines. dCys-GCaMP3 proved to be a better calcium indicator, but it was not clear how and why the two cysteine residues were able to enhance the protein's calcium sensitivity. The aim of the present study was to investigate the possible roles of these cysteine residues in dCys-GCaMP3. dCys-GCaMP3 (Thr330Cys;Thr364Cys) exhibited enhanced fluorescence intensity compared to the canonical GCaMP3 in calcium imaging experiments. However, substitution of a single residue at position 330 with cysteine (Thr330Cys) also afforded comparable sensitivity to GCaMP3. In contrast, the other single residue substitution at position 364 with cysteine (Thr364Cys) failed to enhance calcium sensitivity, showing that cysteine at position 330 is essential to improve calcium sensitivity. Thr330Cys substitution in the GCaMP3 or "Cys330-GCaMP3" showed significantly reduced background fluorescence, and the fluorescence intensity was proportional to the amount of DNA used to transfect the cells used in the study. The substitute had to be cysteine, because replacement with other amino acids such as alanine, valine, and aspartate did not improve GCaMP3's calcium sensitivity. Cys330-GCaMP3 outperformed a synthetic calcium-specific indicator, Fluo-3, in various calcium imaging experiments. Thus, the present study asserts that substituting the threonine at position 330 in GCaMP3 with cysteine is essential to enhance calcium sensitivity, and suggests that Cys330-GCaMP3 can be used as a potent fluorescent calcium indicator to measure intracellular calcium levels.


Asunto(s)
Calcio/metabolismo , Cisteína/química , Proteínas Fluorescentes Verdes/metabolismo , Imagen Molecular/métodos , Proteínas Recombinantes de Fusión/metabolismo , Sustitución de Aminoácidos , Señalización del Calcio , Calmodulina/química , Calmodulina/metabolismo , Cisteína/genética , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/genética , Células HEK293 , Humanos , Mutagénesis Sitio-Dirigida , Mutación , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Treonina/química , Treonina/genética
14.
PLoS Genet ; 10(10): e1004652, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25340791

RESUMEN

Metastasis is the principal cause of cancer death and occurs through multiple, complex processes that involve the concerted action of many genes. A number of studies have indicated that the Fragile Histidine Triad (FHIT) gene product, FHIT, functions as a tumor suppressor in a variety of common human cancers. Although there are suggestions of a role for FHIT loss in progression of various cancers, a role for such loss in metastasis has not been defined. Here, via in vivo and in vitro assays, we reveal that the enforced expression of FHIT significantly suppresses metastasis, accompanied by inhibition of the epithelial-mesenchymal transition (EMT), a process involved in metastasis through coordinate modulation of EMT-related genes. Specifically, miR-30c, a FHIT-upregulated microRNA, contributes to FHIT function in suppression of EMT and metastasis by directly targeting metastasis genes Metadherin (MTDH), High-mobility group AT-hook 2 (HMGA2), and the mesenchymal markers, Vimentin (VIM) and Fibronectin (FN1), in human lung cancer. Finally, we demonstrate that the expression pattern of FHIT and miR-30c is inversely correlated with that of MTDH and HMGA2 in normal tissue, non-metastatic and metastatic tumors, serving as a potential biomarker for metastasis in lung cancer.


Asunto(s)
Ácido Anhídrido Hidrolasas/genética , Transición Epitelial-Mesenquimal/genética , Neoplasias Pulmonares/genética , MicroARNs/genética , Proteínas de Neoplasias/genética , Ácido Anhídrido Hidrolasas/biosíntesis , Moléculas de Adhesión Celular , Línea Celular Tumoral , Fibronectinas/biosíntesis , Regulación Neoplásica de la Expresión Génica , Proteína HMGA2/biosíntesis , Humanos , Neoplasias Pulmonares/patología , Proteínas de la Membrana , MicroARNs/biosíntesis , Metástasis de la Neoplasia , Proteínas de Neoplasias/biosíntesis , Proteínas de Unión al ARN , Vimentina/biosíntesis
15.
J Anesth ; 31(1): 11-17, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27572548

RESUMEN

BACKGROUND: We designed this study to investigate the effect of dexmedetomidine (1 µg/kg) pretreatment on the median effective dose (ED50) of propofol for facilitating successful laryngeal mask airway (LMA) insertion compared to propofol alone. METHODS: Forty patients were randomized to either the control group (n = 21) or the dexmedetomidine group (n = 19). After infusion of normal saline or dexmedetomidine 1 µg/kg over 10 min, 1 % lidocaine 0.5 mg/kg, followed by propofol 2.5 mg/kg was administered and the laryngeal mask airway was inserted without muscle relaxants. The ED50 of propofol for successful LMA insertion was determined by the modified Dixon's up-and-down method. The ED50 and ED95 were also calculated using an isotonic regression method, based on the pooled adjacent-violators algorithm-adjusted response rate, and the confidential interval (CI) was estimated using a bootstrap approach. RESULTS: The ED50 of propofol for smooth insertion of the LMA was significantly higher in the control group than in the dexmedetomidine group (3.1 ± 0.4 vs 1.9 ± 0.3 mg/kg, P < 0.001). From isotonic regression analysis using a bootstrap approach, the ED50 and ED95 of propofol was 2.9 mg/kg (83 % CI 2.5-3.3 mg/kg) and 3.9 mg/kg (95 % CI 3.5-4.0 mg/kg) in the control group, and 1.8 mg/kg (83 % CI 1.8-2.1 mg/kg) and 2.4 mg/kg (95 % CI 2.0-2.5 mg/kg) in the dexmedetomidine groups, respectively. The apnea time was not significantly different between the two groups. CONCLUSIONS: Pretreatment with dexmedetomidine 1 µg/kg could reduce the propofol requirement by 38 % for facilitating LMA insertion without prolonged respiratory depression and hemodynamic instability.


Asunto(s)
Anestésicos Intravenosos , Dexmedetomidina , Hipnóticos y Sedantes , Máscaras Laríngeas , Propofol , Adulto , Anciano , Anestésicos Intravenosos/administración & dosificación , Anestésicos Locales , Apnea , Dexmedetomidina/administración & dosificación , Femenino , Hemodinámica , Humanos , Hipnóticos y Sedantes/administración & dosificación , Lidocaína , Masculino , Persona de Mediana Edad , Propofol/administración & dosificación
16.
Jpn J Clin Oncol ; 46(2): 138-43, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26685322

RESUMEN

OBJECTIVE: Few reports have described the treatment outcome of osteosarcoma using radiotherapy. We evaluated the efficacy of radiotherapy and gemcitabine and docetaxel chemotherapy for patients with unresectable recurrent or refractory osteosarcoma. METHODS: Data from six patients (five male, one female) who received radiotherapy and gemcitabine and docetaxel chemotherapy at the Korea Cancer Center Hospital were retrospectively reviewed. Tumor response was evaluated according to metabolic changes using (18)F-fluorodeoxy-D-glucose-positron emission tomography. RESULTS: The median age of the patients was 15.0 years (range, 14.0-15.8 years). Two patients had single bone lesions, and four had multiple metastatic bone lesions. Patients received a median 3.5 courses of gemcitabine and docetaxel chemotherapy (range, 2-6 courses). The median dose of radiotherapy was 50.0 Gy (range, 46-84 Gy). There were two complete metabolic responses and one partial metabolic response. The objective response rate was 50.0% (3/6). Responses were maintained for 4.6, 6.1 and 13.7 months, respectively. Patients were followed up for a median of 5.8 months (range, 2.7-84.6 months), and the median progression-free survival after this treatment was 3.6 months (range, 1.1-13.7 months). At the time of analysis, two patients were alive, one was lost to follow-up and three had died. CONCLUSION: Radiotherapy with gemcitabine and docetaxel chemotherapy showed some improvement in cases of refractory tumors or multiple bone metastases. Further studies are needed to investigate the efficacy of newer radiotherapy modalities, as well as to identify new radiosensitizing chemotherapy regimens.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/radioterapia , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/radioterapia , Adolescente , Neoplasias Óseas/mortalidad , Neoplasias Óseas/patología , Niño , Preescolar , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Supervivencia sin Enfermedad , Docetaxel , Esquema de Medicación , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Osteosarcoma/mortalidad , Osteosarcoma/patología , Dosificación Radioterapéutica , Radioterapia Adyuvante , República de Corea , Estudios Retrospectivos , Taxoides/administración & dosificación , Resultado del Tratamiento , Gemcitabina
17.
Int J Cancer ; 137(9): 2253-69, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25944623

RESUMEN

Various ways to inhibit vascular endothelial growth factor (VEGF), a key facilitator in tumor angiogenesis, are being developed to treat cancer. The soluble VEGF decoy receptor (FP3), due to its high affinity to VEGF, is a highly effective and promising strategy to disrupt VEGF signaling pathway. Despite potential advantage and potent therapeutic efficacy, its employment has been limited by very poor in vivo pharmacokinetic properties. To address this challenge, we designed a novel oncolytic adenovirus (Ad) expressing FP3 (RdB/FP3). To demonstrate the VEGF-specific nature of RdB/FP3, replication-incompetent Ad expressing FP3 (dE1/FP3) was also generated. dE1/FP3 was highly effective in reducing VEGF expression and functionally elicited an antiangiogeneic effect. Furthermore, RdB/FP3 exhibited a potent antitumor effect compared with RdB or recombinant FP3. Consistent with these data, RdB/FP3 was shown to greatly decrease VEGF expression level and vessel density and increase apoptosis in both tumor endothelial and tumor cells, verifying potent suppressive effects of RdB/FP3 on VEGF-mediated tumor angiogenesis in vivo. Importantly, the therapeutic mechanism of antitumor effect mediated by RdB/FP3 is associated with prolonged VEGF silencing efficacy and enhanced oncolysis via cancer cell-specific replication of oncolytic Ad. Taken together, RdB/FP3 provides a new promising therapeutic approach in the treatment of cancer and angiogenesis-related diseases.


Asunto(s)
Adenoviridae/genética , Viroterapia Oncolítica , Virus Oncolíticos/genética , Inhibidores de la Angiogénesis/biosíntesis , Inhibidores de la Angiogénesis/genética , Inhibidores de la Angiogénesis/farmacología , Animales , Línea Celular Tumoral , Movimiento Celular , Expresión Génica , Células HEK293 , Humanos , Ratones Desnudos , Trasplante de Neoplasias , Neovascularización Patológica/prevención & control , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/genética
18.
FASEB J ; 28(11): 4779-91, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25059229

RESUMEN

CD137 (4-1BB), a member of the tumor necrosis factor receptor superfamily, has been reported to be expressed in atherosclerotic plaques, and to promote lesion formation. However, the role of CD137 in mediating atherosclerotic plaque stability and the possible underlying molecular and cellular mechanisms are poorly understood. Here, apolipoprotein E-deficient (ApoE(-/-)) and CD137-deficient ApoE(-/-) (ApoE(-/-)CD137(-/-)) mice fed a chow diet for 66 wk were used. CD137 induces plaque instability, which is characterized by increased plaque necrosis, decreased collagen content, decreased vascular smooth muscle cell (VSMC) content, and increased macrophage infiltration. CD137 also increases the infiltration of effector T (Teff) cells into plaque lesion sites, resulting in increased interferon-γ (IFN-γ) expression. Interestingly, Teff-cell-derived IFN-γ inhibits collagen synthesis in atherosclerotic plaques. Furthermore, CD137 activation increases the apoptosis of VSMCs, possibly by decreasing the antiapoptotic regulator, Bcl-2, and subsequently up-regulating cleaved caspase-3. In macrophages, activation of CD137 signaling boosted the oxidized low density lipoprotein-induced expression of matrix metalloproteinase 9 via the p38 mitogen-activated protein kinase and extracellular signal-regulated kinase1/2 signaling pathways. In summary, activation of CD137 signaling decreases the stability of advanced atherosclerotic plaques via its combined effects on Teff cells, VSMCs, and macrophages.


Asunto(s)
Ligando 4-1BB/inmunología , Aterosclerosis/metabolismo , Hiperlipidemias/metabolismo , Macrófagos/metabolismo , Placa Aterosclerótica/metabolismo , Linfocitos T/metabolismo , Animales , Apoptosis/efectos de los fármacos , Aterosclerosis/inmunología , Interferón gamma/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Placa Aterosclerótica/inmunología , Transducción de Señal/inmunología , Linfocitos T/inmunología
19.
Mol Ther ; 22(9): 1678-87, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24895995

RESUMEN

Glioblastoma is a devastating disease, and there is an urgent need to develop novel therapies, such as oncolytic HSV1 (OV) to effectively target tumor cells. OV therapy depends on tumor-specific replication leading to destruction of neoplastic tissues. Host responses that curtail virus replication limit its efficacy in vivo. We have previously shown that cysteine-rich 61 protein (CCN1) activates a type 1 IFN antiviral defense response in glioblastoma cells. Incorporating TCGA data, we found CCN1 expression to be a negative prognostic factor for glioblastoma patients. Based on this, we used neutralizing antibodies against CCN1 to investigate its effect on OV therapy. Use of an anti-CCN1 antibody in mice bearing glioblastomas treated with OV led to enhanced virus expression along with reduced immune cell infiltration. OV-induced CCN1 increases macrophage migration toward infected glioblastoma cells by directly binding macrophages and also by enhancing the proinflammatory activation of macrophages inducing MCP-1 expression in glioblastoma cells. Activation of macrophages by CCN1 also increases viral clearance. Neutralization of integrin αMß2 reversed CCN1-induced macrophage activation and migration, and reduced MCP-1 expression by glioblastoma cells. Our findings reveal that CCN1 plays a novel role in pathogen clearance; increasing macrophage infiltration and activation resulting in increased virus clearance in tumors.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Proteína 61 Rica en Cisteína/genética , Proteína 61 Rica en Cisteína/metabolismo , Glioblastoma/inmunología , Herpesvirus Humano 1/genética , Macrófagos/metabolismo , Animales , Anticuerpos Monoclonales/uso terapéutico , Línea Celular Tumoral , Quimiocina CCL2/metabolismo , Femenino , Vectores Genéticos/administración & dosificación , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Activación de Macrófagos , Ratones , Trasplante de Neoplasias , Virus Oncolíticos/genética
20.
Proc Natl Acad Sci U S A ; 109(14): 5316-21, 2012 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-22431589

RESUMEN

MicroRNAs (miRNAs) are increasingly implicated in regulating cancer initiation and progression. In this study, two miRNAs, miR-25 and -32, are identified as p53-repressed miRNAs by p53-dependent negative regulation of their transcriptional regulators, E2F1 and MYC. However, miR-25 and -32 result in p53 accumulation by directly targeting Mdm2 and TSC1, which are negative regulators of p53 and the mTOR (mammalian target of rapamycin) pathway, respectively, leading to inhibition of cellular proliferation through cell cycle arrest. Thus, there is a recurrent autoregulatory circuit involving expression of p53, E2F1, and MYC to regulate the expression of miR-25 and -32, which are miRNAs that, in turn, control p53 accumulation. Significantly, overexpression of transfected miR-25 and -32 in glioblastoma multiforme cells inhibited growth of the glioblastoma multiforme cells in mouse brain in vivo. The results define miR-25 and -32 as positive regulators of p53, underscoring their role in tumorigenesis in glioblastoma.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , MicroARNs/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Neoplasias Encefálicas/patología , Ciclo Celular , Proliferación Celular , Factor de Transcripción E2F1/fisiología , Glioblastoma/patología , Humanos , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteínas Proto-Oncogénicas c-myc/fisiología , Transcripción Genética , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA