Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-31192163

RESUMEN

Antimicrobial peptides, also known as host defense peptides, have recently emerged as a promising new category of therapeutic agents for the treatment of infectious diseases. This study evaluated the preclinical in vitro, ex vivo, and in vivo antimicrobial activity, as well as the potential to cause skin irritation, of human kininogen-derived antimicrobial peptide DPK-060 in different formulations designed for topical delivery. We found that DPK-060 formulated in acetate buffer or poloxamer gel caused a marked reduction of bacterial counts of Staphylococcus aureus in vitro (minimum microbicidal concentration <5 µg/ml). We also found that DPK-060 in poloxamer gel significantly suppressed microbial survival in an ex vivo wound infection model using pig skin and in an in vivo mouse model of surgical site infection (≥99 or ≥94% reduction in bacterial counts was achieved with 1% DPK-060 at 4 h post-treatment, respectively). Encapsulation of DPK-060 in different types of lipid nanocapsules or cubosomes did not improve the bactericidal potential of the peptide under the applied test conditions. No reduction in cell viability was observed in response to administration of DPK-060 in any of the formulations tested. In conclusion, the present study confirms that DPK-060 has the potential to be an effective and safe drug candidate for the topical treatment of microbial infections; however, adsorption of the peptide to nanocarriers failed to show any additional benefits.


Asunto(s)
Administración Tópica , Antibacterianos/uso terapéutico , Péptidos Catiónicos Antimicrobianos/uso terapéutico , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/efectos de los fármacos , Animales , Antibacterianos/administración & dosificación , Péptidos Catiónicos Antimicrobianos/administración & dosificación , Modelos Animales de Enfermedad , Femenino , Lípidos/química , Ratones , Pruebas de Sensibilidad Microbiana , Nanocápsulas , Poloxámero/uso terapéutico , Proteínas Serina-Treonina Quinasas/administración & dosificación , Proteínas Serina-Treonina Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/uso terapéutico , Pruebas de Irritación de la Piel , Infecciones Estafilocócicas/microbiología , Infecciones Cutáneas Estafilocócicas/tratamiento farmacológico , Porcinos
2.
Transgenic Res ; 27(2): 179-191, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29446008

RESUMEN

Genetic engineering approaches offer an alternative method to the conventional breeding of Citrus sp. 'W. Murcott' mandarin (a hybrid of 'Murcott' and an unknown pollen parent) is one of the most commercially important cultivars grown in many regions around the world. Transformation of 'W. Murcott' mandarin was achieved by direct DNA uptake using a protoplast transformation system. DNA construct (pAO3), encoding Green Fluorescent Protein (GFP) and the cDNA of Xa21, a Xanthomonas resistance gene from rice, was used to transform protoplasts of 'W. Murcott' mandarin. Following citrus protoplast culture and regeneration, transformed micro calli were microscopically designated via GFP expression, physically isolated from non-transformed tissue, and cultured on somatic embryogenesis induction medium. More than 150 transgenic embryos were recovered and from them, ten transgenic lines were regenerated and cultured on rooting medium for shoot elongation. Transgenic shoots were micrografted and established in the greenhouse with 3-5 replicates per line. The insertion of Xa21 and GFP was confirmed by PCR and southern blot analysis. GFP expression was verified by fluorescence microscopy and western blot analysis revealed expression of Xa21 although it was variable among transgenic lines, as shown by RT-qPCR. Transgenic plants challenged with the citrus canker pathogen by syringe inoculation showed a reduction in lesion number and bacterial populations within lesions compared to non-transgenic control plants. Transgenic 'W. Murcott' mandarin lines with improved canker resistance via protoplast transformation from embryogenic callus with the Xa21 gene from rice are being evaluated under field conditions to validate the level of resistance.


Asunto(s)
Citrus/genética , Oryza/genética , Proteínas de Plantas/genética , Plantas Modificadas Genéticamente/genética , Proteínas Serina-Treonina Quinasas/genética , Citrus/crecimiento & desarrollo , Citrus/microbiología , Resistencia a la Enfermedad/genética , Oryza/microbiología , Enfermedades de las Plantas/genética , Enfermedades de las Plantas/microbiología , Proteínas de Plantas/administración & dosificación , Plantas Modificadas Genéticamente/efectos de los fármacos , Plantas Modificadas Genéticamente/microbiología , Proteínas Serina-Treonina Quinasas/administración & dosificación , Protoplastos/microbiología
3.
Nanomedicine ; 14(3): 991-1003, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29339188

RESUMEN

In order to enhance the penetration of small interference RNA against the polo-like kinase I (siPLK1) across BBB to treat glioblastoma (GBM), transferrin (Tf) modified magnetic nanoparticle (Tf-PEG-PLL/MNP@siPLK1) was prepared. The in vitro experiments indicated that Tf-PEG-PLL/MNP@siPLK1 enhanced the cellular uptake of siPLK1, which resulted in an increase of gene silencing effect and cytotoxicity of Tf-PEG-PLL/MNP@siPLK1 on U87 cells. Besides, Tf-PEG-PLL/MNP@siPLK1 significantly inhibited the growth of U87 glioblastoma spheroids and markedly increased the BBB penetration efficiency of siPLK1 with the application of external magnetic field in in-vitro BBB model. The in vivo experiments indicated that siPLK1 selectively accumulated in the brain tissue, and markedly reduced tumor volume and prolonged the survival time of GBM-bearing mice after Tf-PEG-PLL/MNP@siPLK1 was injected to GBM-bearing mice via tail vein. The above data indicated that magnet and transferrin co-modified nanoparticle enhanced siPLK1 penetration across BBB and increased its anti GBM activity in vivo.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Proteínas de Ciclo Celular/antagonistas & inhibidores , Silenciador del Gen , Glioblastoma/terapia , Nanopartículas de Magnetita/administración & dosificación , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , ARN Interferente Pequeño/genética , Transferrina/química , Animales , Barrera Hematoencefálica/patología , Ciclo Celular , Proteínas de Ciclo Celular/administración & dosificación , Proteínas de Ciclo Celular/genética , Supervivencia Celular , Endocitosis , Glioblastoma/genética , Glioblastoma/patología , Nanopartículas de Magnetita/química , Ratones , Tamaño de la Partícula , Proteínas Serina-Treonina Quinasas/administración & dosificación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/administración & dosificación , Proteínas Proto-Oncogénicas/genética , Quinasa Tipo Polo 1
4.
Biomaterials ; 112: 176-191, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27768972

RESUMEN

During mammalian embryonic development, neurons polarize to create distinct cellular compartments of axon and dendrite that inherently differ in form and function, providing the foundation for directional signaling in the nervous system. Polarization results from spatio-temporal segregation of specific proteins' activities to discrete regions of the neuron to dictate axonal vs. dendritic fate. We aim to manipulate axon formation by directed subcellular localization of crucial intracellular protein function. Here we report critical steps toward the development of a nanotechnology for localized subcellular introduction and retention of an intracellular kinase, LKB1, crucial regulator of axon formation. This nanotechnology will spatially manipulate LKB1-linked biomagnetic nanocomplexes (LKB1-NCs) in developing rodent neurons in culture and in vivo. We created a supramolecular assembly for LKB1 rapid neuronal uptake and prolonged cytoplasmic stability. LKB1-NCs retained kinase activity and phosphorylated downstream targets. NCs were successfully delivered to cultured embryonic hippocampal neurons, and were stable in the cytoplasm for 2 days, sufficient time for axon formation. Importantly, LKB1-NCs promoted axon formation in these neurons, representing unique proof of concept for the sufficiency of intracellular protein function in dictating a central developmental event. Lastly, we established NC delivery into cortical progenitors in live rat embryonic brain in utero. Our nanotechnology provides a viable platform for spatial manipulation of intracellular protein-activity, to dictate central events during neuronal development.


Asunto(s)
Orientación del Axón/fisiología , Preparaciones de Acción Retardada/administración & dosificación , Nanopartículas de Magnetita/administración & dosificación , Nanocápsulas/administración & dosificación , Neuronas/fisiología , Proteínas Serina-Treonina Quinasas/administración & dosificación , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Orientación del Axón/efectos de los fármacos , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/efectos de la radiación , Campos Magnéticos , Nanopartículas de Magnetita/química , Nanopartículas de Magnetita/efectos de la radiación , Micelas , Nanocápsulas/química , Nanocápsulas/efectos de la radiación , Neurogénesis/efectos de los fármacos , Neurogénesis/fisiología , Neuronas/citología , Neuronas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
5.
Physiol Rep ; 4(21)2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27905294

RESUMEN

Glucocorticoids (GC) are a frontline therapy for numerous acute and chronic diseases because of their demonstrated efficacy at reducing systemic inflammation. An unintended side effect of GC therapy is the stimulation of skeletal muscle atrophy. Pathophysiological mechanisms responsible for GC-induced skeletal muscle atrophy have been extensively investigated, and the ability to treat patients with GC without unintended muscle atrophy has yet to be realized. We have reported that a single, standard-of-care dose of Methylprednisolone increases in vivo expression of NF-κB-inducing kinase (NIK), an important upstream regulatory kinase controlling NF-κB activation, along with other key muscle catabolic regulators such as Atrogin-1 and MuRF1 that induce skeletal muscle proteolysis. Here, we provide experimental evidence that overexpressing NIK by intramuscular injection of recombinant human NIK via adenoviral vector in mouse tibialis anterior muscle induces a 30% decrease in the average fiber cross-sectional area that is associated with increases in mRNA expression of skeletal muscle atrophy biomarkers MuRF1, Atrogin-1, myostatin and Gadd45. A single injection of GC induced NIK mRNA and protein within 2 h, with the increased NIK localized to nuclear and sarcolemmal locations within muscle fibers. Daily GC injections induced skeletal muscle fore limb weakness as early as 3 days with similar atrophy of muscle fibers as observed with NIK overexpression. NIK overexpression in primary human skeletal muscle myotubes increased skeletal muscle atrophy biomarkers, while NIK knockdown significantly attenuated GC-induced increases in NIK and Atrogin-1. These results suggest that NIK may be a novel, previously unrecognized mediator of GC-induced skeletal muscle atrophy.


Asunto(s)
Glucocorticoides/farmacología , Músculo Esquelético/enzimología , Atrofia Muscular/inducido químicamente , Atrofia Muscular/metabolismo , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Glucocorticoides/administración & dosificación , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/efectos de los fármacos , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Atrofia Muscular/patología , Proteínas Serina-Treonina Quinasas/administración & dosificación , ARN Mensajero/genética , Proteínas Ligasas SKP Cullina F-box/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Quinasa de Factor Nuclear kappa B
6.
Int J Nanomedicine ; 11: 5277-5286, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27822032

RESUMEN

To explore a novel combination of chemotherapy, gene therapy, and thermotherapy for osteosarcoma, a targeted heat-sensitive co-delivery system based on bacterial magnetosomes (BMs) was developed. The optimal culture conditions of magnetotactic bacteria (MTB) AMB-1 and characterization of BMs were achieved. A recombinant eukaryotic plasmid heat shock protein 70-polo-like kinase 1-short hairpin RNA (pHSP70-Plk1-shRNA) under transcriptional control of a thermosensitive promoter (human HSP70 promoter) was constructed for gene therapy. Doxorubicin (DOX) and pHSP70-Plk1-shRNA were included in the targeted thermosensitive co-delivery system, and in vitro DOX release activity, targeted gene silencing efficiency and in vitro antitumor efficacy were investigated. The results showed that the optimal culture conditions of MTB AMB-1 are an oxygen concentration of 4.0%, a pH value of 7.0, 20 µmol/L of ferrous sulfate, 800 mg/L of sodium nitrate, and 200 mg/L of succinic acid. The temperature of BMs reached 43°C within 3 minutes and could be maintained for 30 minutes by adjusting the magnitude of the alternating magnetic field (AMF). The diameters of BMs, BM-DOX, BM-recombinant eukaryotic plasmid pHSP70-Plk1-shRNA (shPlk1), and BM-DOX-shPlk1 were 43.7±4.6, 79.2±5.4, 88.9±7.8, and 133.5±11.4 nm, respectively. The zeta potentials of BMs, BM-DOX, BM-shPlk1, and BM-DOX-shPlk1 were -29.4±6.9, -9.5±5.6, -16.7±4.8, and -10.3±3.1 mV, respectively. Besides, the system exhibited good release behavior. DOX release rate from BM-DOX-shPlk1 was 54% after incubation with phosphate-buffered saline at 43°C and 37% after incubation with 50% fetal bovine serum, which was significantly higher than that at 37°C (P<0.05). In addition, the expressions of Plk1 mRNA and protein were significantly suppressed in cells treated with BM-DOX-shPlk1 following hyperthermia treatment under the influence of an AMF compared to other groups (P<0.05). Furthermore, evaluation of the effect of in vitro antitumor revealed that BM-DOX-shPlk1 following hyperthermia treatment under the influence of an AMF was significantly more effective than others in tumor inhibition. In conclusion, the new heat-sensitive co-delivery system represents a promising approach for the treatment of cancer.


Asunto(s)
Neoplasias Óseas/metabolismo , Doxorrubicina/administración & dosificación , Terapia Genética/métodos , Magnetosomas/química , Osteosarcoma/metabolismo , ARN Interferente Pequeño/genética , Apoptosis , Bacterias/metabolismo , Proteínas de Ciclo Celular/administración & dosificación , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral/efectos de los fármacos , Proliferación Celular , Citometría de Flujo , Silenciador del Gen/efectos de los fármacos , Proteínas del Choque Térmico HSP72/metabolismo , Humanos , Hipertermia Inducida/métodos , Oxígeno/química , Plásmidos/metabolismo , Proteínas Serina-Treonina Quinasas/administración & dosificación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/administración & dosificación , Proteínas Proto-Oncogénicas/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Recombinantes/química , Temperatura , Quinasa Tipo Polo 1
7.
Parasitol Res ; 114(12): 4553-63, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26337271

RESUMEN

Toxoplasmosis is one of the most common parasitic infections worldwide. An effective vaccine against human and animal toxoplasmosis is still needed to control this parasitosis. The polymorphic rhoptry proteins, ROP5 and ROP18, secreted by Toxoplasma gondii during the invasion of the host cell have been recently considered as promising vaccine antigens, as they appear to be the major determinants of T. gondii virulence in mice. The goal of this study was to evaluate their immunogenic and immunoprotective activity after their administration (separately or both recombinant proteins together) with the poly I:C as an adjuvant. Immunization of BALB/c and C3H/HeOuJ mice generated both cellular and humoral specific immune responses with some predominance of IgG1 antibodies. The spleen cells derived from vaccinated animals reacted to the parasite's native antigens. Furthermore, the immunization led to a partial protection against acute and chronic toxoplasmosis. These findings confirm the previous assumptions about ROP5 and ROP18 antigens as valuable components of a subunit vaccine against toxoplasmosis.


Asunto(s)
Proteínas Serina-Treonina Quinasas/inmunología , Toxoplasma/inmunología , Toxoplasmosis/prevención & control , Vacunas/inmunología , Animales , Anticuerpos Antiprotozoarios/inmunología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Proteínas Serina-Treonina Quinasas/administración & dosificación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Protozoarias , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Bazo/inmunología , Toxoplasma/genética , Toxoplasmosis/inmunología , Toxoplasmosis/parasitología , Vacunación , Vacunas/administración & dosificación , Vacunas/genética , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/genética , Vacunas de Subunidad/inmunología
8.
J Neurol Sci ; 358(1-2): 226-35, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26365288

RESUMEN

Oxidative stress is considered a major factor in various neuronal diseases including ischemia-reperfusion injury. Proviral Integration Moloney 2 (PIM2) proteins, one of the families of PIM kinases, play crucial roles in cell survival. However, the functions of PIM2 protein against ischemia are not understood. Therefore, the protective effects of PIM2 against oxidative stress-induced hippocampal HT22 cell death and brain ischemic injury were evaluated using Tat-PIM2, a cell permeable fusion protein. Tat-PIM2 protein transduced into hippocampal HT22 cells. Low doses of transduced Tat-PIM2 protein protected against oxidative stress-induced cell death including DNA damage and markedly inhibited the activation of mitogen activated protein kinase (MAPKs), NF-κB and the expression levels of Bax protein. Furthermore, Tat-PIM2 protein transduced into the CA1 region of the hippocampus and significantly prevented neuronal cell death in an ischemic insult animal model. These results indicated that low doses of Tat-PIM2 protein protects against oxidative stress-induced neuronal cell death, suggesting low doses of Tat-PIM2 protein provides a potential therapeutic agent against oxidative stress-induced neuronal diseases including ischemia.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Productos del Gen tat/administración & dosificación , Hipocampo/efectos de los fármacos , Neuronas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/administración & dosificación , Proteínas Proto-Oncogénicas/administración & dosificación , Animales , Línea Celular , Gerbillinae , Hipocampo/metabolismo , Ratones , Neuronas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción Genética
9.
Invest New Drugs ; 33(2): 409-22, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25529193

RESUMEN

Purpose BI 831266 is a potent, selective, low-molecular-weight inhibitor of Aurora kinase B. This trial aimed to determine the maximum tolerated dose (MTD) of BI 831266 in patients with advanced solid tumors (NCT00756223; EudraCT 2008-001631-36; 1257.1). Methods BI 831266 (4-130 mg) was administered over 24 h on days 1 and 15 of a 4-week schedule. A modified 3 + 3 dose-escalation design was utilized to evaluate the MTD. Safety, pharmacokinetics, pharmacodynamics, objective response rate, progression-free survival (PFS) and exploratory biomarkers were secondary endpoints. Results Twenty-five patients received BI 831266. The most frequent tumor type was colorectal cancer (48%). One patient (130 mg) experienced a dose-limiting toxicity of grade 3 febrile neutropenia. The trial was prematurely terminated (sponsor decision) without further dose-escalation. The most frequent treatment-related adverse events (AEs) were fatigue (20%), neutropenia, alopecia (16% each), anemia, dry skin, and nausea (12% each). Treatment-related grade ≥3 AEs were neutropenia (12%), anemia (8%), and febrile neutropenia (4%); 15 patients experienced serious AEs. High variability in the pharmacokinetic profiles precluded definitive pharmacokinetic conclusions. Exploratory biomarker determination revealed consistency with the mode of action as an Aurora kinase B inhibitor. One patient (4%; 32 mg) with cervical cancer demonstrated a confirmed partial response (duration 141 days, PFS 414 days). Four patients had stable disease. Conclusion The MTD of BI 831266 was not reached because of early trial termination. BI 831266 demonstrated a generally manageable safety profile and signs of antitumor activity in some patients' solid tumors.


Asunto(s)
Antineoplásicos/farmacología , Aurora Quinasa B/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/farmacología , Adulto , Anciano , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Supervivencia sin Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/farmacocinética , Proteínas Serina-Treonina Quinasas/administración & dosificación , Proteínas Serina-Treonina Quinasas/efectos adversos , Proteínas Serina-Treonina Quinasas/farmacocinética
10.
Antioxid Redox Signal ; 21(14): 1960-73, 2014 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-23901824

RESUMEN

AIMS: We have shown that autophagy and mitophagy are required for preconditioning. While statin's cardioprotective effects are well known, the role of autophagy/mitophagy in statin-mediated cardioprotection is not. In this study, we used HL-1 cardiomyocytes and mice subjected to ischemia/reperfusion to elucidate the mechanism of statin-mediated cardioprotection. RESULTS: HL-1 cardiomyocytes exposed to simvastatin for 24 h exhibited diminished protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signaling, increased activation of unc-51-like kinase 1, and upregulation of autophagy and mitophagy. Similar findings were obtained in hearts of mice given simvastatin. Mevalonate abolished simvastatin's effects on Akt/mTOR signaling and autophagy induction in HL-1 cells, indicating that the effects are mediated through inhibition of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase. Simvastatin-treated HL-1 cells exhibited mitochondrial translocation of Parkin and p62/SQSTM1, fission, and mitophagy. Because Parkin is required for mitophagy and is expressed in heart, we investigated the effect of simvastatin on infarct size in Parkin knockout mice. Simvastatin reduced infarct size in wild-type mice but showed no benefit in Parkin knockout mice. Inhibition of HMG-CoA reductase limits mevalonate availability for both cholesterol and coenzyme Q10 (CoQ) biosynthesis. CoQ supplementation had no effect on statin-induced Akt/mTOR dephosphorylation or macroautophagy in HL-1 cells, but it potently blocked mitophagy. Importantly, CoQ supplementation abolished statin-mediated cardioprotection in vivo. INNOVATION AND CONCLUSION: Acute simvastatin treatment suppresses mTOR signaling and triggers Parkin-dependent mitophagy, the latter which is required for cardioprotection. Coadministration of CoQ with simvastatin impairs mitophagy and cardioprotection. These results raise the concern that CoQ may interfere with anti-ischemic benefits of statins mediated through stimulation of mitophagy.


Asunto(s)
Cardiotónicos/administración & dosificación , Mitofagia/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Simvastatina/administración & dosificación , Animales , Autofagia/efectos de los fármacos , Homólogo de la Proteína 1 Relacionada con la Autofagia , Línea Celular , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Ratones , Mitofagia/genética , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteínas Serina-Treonina Quinasas/administración & dosificación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Proteínas Proto-Oncogénicas c-akt/genética , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/patología , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/biosíntesis , Serina-Treonina Quinasas TOR/genética
11.
Exp Parasitol ; 135(2): 234-9, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23896123

RESUMEN

Toxoplasma gondii, the etiological agent of toxoplasmosis, is an obligate intracellular protozoan parasite that infects a variety of mammals including humans. In an attempt to find new antigen-adjuvant combinations that enhance the immunogenicity of antigen candidates for toxoplasma vaccines, we analyzed the potent protection in mice immunized with recombinant protein ROP18 when co-administered with ginsenoside Re, a most important component isolated from Panax ginseng. All immunized mice produced specific anti-rROP18 immunoglobulins, with high levels of IgG antibody and a mixed IgG1/IgG2a response, with predominance of IgG1 production. The cellular and humoral immune responses were associated with the production of IFN-γ and IL-4 cytokines respectively. Vaccinated mice displayed a significantly increased survival time compared with control mice which died within 6 days of challenge with RH strain. Our data demonstrate that by addition of ginsenoside Re, the rROP18 triggered a stronger humoral and cellular response against T. gondii, and that Re is a promising vaccine adjuvant against toxoplasmosis, deserves further evaluation and development.


Asunto(s)
Ginsenósidos/inmunología , Proteínas Serina-Treonina Quinasas/inmunología , Vacunas Antiprotozoos/inmunología , Toxoplasma/inmunología , Toxoplasmosis/prevención & control , Animales , Anticuerpos Antiprotozoarios/biosíntesis , Anticuerpos Antiprotozoarios/sangre , Western Blotting , Citocinas/análisis , Ensayo de Inmunoadsorción Enzimática , Femenino , Ginsenósidos/administración & dosificación , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/sangre , Inyecciones Subcutáneas , Activación de Linfocitos , Ratones , Ratones Endogámicos ICR , Proteínas Serina-Treonina Quinasas/administración & dosificación , Proteínas Protozoarias , Vacunas Antiprotozoos/administración & dosificación , Distribución Aleatoria , Proteínas Recombinantes/inmunología , Organismos Libres de Patógenos Específicos , Bazo/citología , Bazo/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología
12.
PLoS One ; 7(2): e31279, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22348065

RESUMEN

AIMS: Integrin-linked kinase (ILK) is a multifunctional kinase linking the extracellular matrix to intracellular signaling pathways, whose activation in the heart gives rise to a number of functional consequences. The aim of this study is to demonstrate the therapeutic and survival benefit of cardiac ILK overexpression in a rat model of dilated cardiomyopathy. METHODS AND RESULTS: The dilated cardiomyopathy model was generated in rats by intraperitoneal administration of six equal doses of doxorubicin over a 2 week period. Five weeks after the first injection, echocardiographic analysis demonstrated impaired cardiac function and, at that point, recombinant adenoviral vector harboring ILK cDNA or vehicle was injected into the myocardium, and the rats re-studied 4 weeks later. Compared with vehicle injection, ILK treatment ameliorated inflammatory cell infiltration and cardiomyocyte degeneration, as well as left ventricular dilation and dysfunction. ILK treatment was also associated with a reduction in apoptosis and an increase in proliferation of cardiomyocytes, as well as decreased oxidative stress and autophagic vacuole accumulation. Importantly, mortality was lower in rats following ILK treatment than in those following vehicle injection. In cultured neonatal rat cardiomyocytes, we also found that ILK overexpression protected against doxorubicin-induced apoptosis, giving rise to an increase in their proliferation. CONCLUSIONS: These data demonstrate for the first time that ILK gene therapy improves cardiac function and survival in a model of dilated cardiomyopathy, and this may be mediated through suppression of inflammation, prevention of ventricular remodeling, inhibition of cardiomyocyte apoptosis and autophagy, and stimulation of cardiomyocyte proliferation.


Asunto(s)
Cardiomiopatía Dilatada/terapia , Terapia Genética/métodos , Corazón/fisiología , Proteínas Serina-Treonina Quinasas/administración & dosificación , Animales , Cardiomiopatía Dilatada/mortalidad , Cardiomiopatía Dilatada/patología , Modelos Animales de Enfermedad , Inflamación/tratamiento farmacológico , Mortalidad , Miocitos Cardíacos/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/uso terapéutico , Ratas , Resultado del Tratamiento , Remodelación Ventricular/efectos de los fármacos
13.
Int J Radiat Oncol Biol Phys ; 84(1): 257-65, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22285666

RESUMEN

PURPOSE: Patients treated with radiotherapy for head-and-neck cancer invariably suffer its deleterious side effect, xerostomia. Salivary hypofunction ensuing from the irreversible destruction of glands is the most common and debilitating oral complication affecting patients undergoing regional radiotherapy. Given that the current management of xerostomia is palliative and ineffective, efforts are now directed toward preventive measures to preserve gland function. The human homolog of Tousled protein, TLK1B, facilitates chromatin remodeling at DNA repair sites and improves cell survival against ionizing radiation (IR). Therefore, we wanted to determine whether a direct transfer of TLK1B protein to rat salivary glands could protect against IR-induced salivary hypofunction. METHODS: The cell-permeable TAT-TLK1B fusion protein was generated. Rat acinar cell line and rat salivary glands were pretreated with TAT peptide or TAT-TLK1B before IR. The acinar cell survival in vitro and salivary function in vivo were assessed after radiation. RESULTS: We demonstrated that rat acinar cells transduced with TAT-TLK1B were more resistant to radiation (D0 = 4.13 ± 1.0 Gy; α/ß = 0 Gy) compared with cells transduced with the TAT peptide (D0 = 4.91 ± 1.0 Gy; α/ß = 20.2 Gy). Correspondingly, retroductal instillation of TAT-TLK1B in rat submandibular glands better preserved salivary flow after IR (89%) compared with animals pretreated with Opti-MEM or TAT peptide (31% and 39%, respectively; p < 0.01). CONCLUSIONS: The results demonstrate that a direct transfer of TLK1B protein to the salivary glands effectively attenuates radiation-mediated gland dysfunction. Prophylactic TLK1B-protein therapy could benefit patients undergoing radiotherapy for head-and-neck cancer.


Asunto(s)
Células Acinares/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/administración & dosificación , Tolerancia a Radiación/efectos de los fármacos , Proteínas Recombinantes de Fusión/administración & dosificación , Glándulas Salivales/efectos de la radiación , Xerostomía/prevención & control , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/administración & dosificación , Células Acinares/metabolismo , Células Acinares/patología , Células Acinares/efectos de la radiación , Animales , Línea Celular , Supervivencia Celular/efectos de la radiación , Femenino , Neoplasias de Cabeza y Cuello/radioterapia , Luciferasas/metabolismo , Proteínas Serina-Treonina Quinasas/aislamiento & purificación , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Glándulas Salivales/metabolismo , Glándulas Salivales/patología , Salivación/efectos de los fármacos , Salivación/efectos de la radiación , Glándula Submandibular/efectos de los fármacos , Glándula Submandibular/metabolismo , Glándula Submandibular/patología , Glándula Submandibular/efectos de la radiación , beta-Galactosidasa/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/aislamiento & purificación , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo
14.
Gene Ther ; 19(12): 1141-9, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22189415

RESUMEN

We previously demonstrated that an artificial protein, TAT-FNK, has antiapoptotic effects against cochlear hair cell (HC) damage caused by ototoxic agents when applied systemically. To examine the feasibility of topical protein therapy for inner ear disorders, we investigated whether gelatin sponge soaked with TAT-FNK and placed on the guinea pig round window membrane (RWM) could deliver the protein to the cochlea and attenuate aminoglycoside (AG)-induced cochlear damage in vivo. First, we found that the immunoreactivity of TAT-myc-FNK was distributed throughout the cochlea. The immunoreactivity was observed from 1-24 h after application. When Tat-FNK was applied 1 h before ototoxic insult (a combination of kanamycin sulfate and ethacrynic acid), auditory brainstem response threshold shifts and the extent of HC death were significantly attenuated. When cochlear organotypic cultures prepared from P5 rats were treated with kanamycin, TAT-FNK significantly reduced the extent of caspase-9 activation and HC death. These findings indicate that TAT-FNK topically applied on the RWM can enter the cochlea by diffusion and effectively prevent AG-induced apoptosis of cochlear HCs by suppressing the mitochondrial caspase-9 pathway.


Asunto(s)
Aminoglicósidos/toxicidad , Apoptosis/efectos de los fármacos , Cóclea/efectos de los fármacos , Productos del Gen tat/farmacología , Enfermedades del Laberinto/prevención & control , Proteínas Serina-Treonina Quinasas/farmacología , Proteínas Recombinantes de Fusión/administración & dosificación , Administración Tópica , Animales , Caspasa 9/metabolismo , Cóclea/metabolismo , Ácido Etacrínico/farmacología , Ácido Etacrínico/toxicidad , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Productos del Gen tat/administración & dosificación , Cobayas , Células Ciliadas Auditivas/efectos de los fármacos , Kanamicina/farmacología , Enfermedades del Laberinto/inducido químicamente , Fármacos Neuroprotectores , Proteínas Serina-Treonina Quinasas/administración & dosificación , Ratas , Proteínas Recombinantes de Fusión/farmacología , Ventana Redonda , Proteínas Supresoras de Tumor
15.
Gene Ther ; 18(3): 275-82, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21048794

RESUMEN

Oral complications of salivary hypofunction often afflict cancer patients undergoing radiotherapy for head and neck cancers. Dry mouth or xerostomia is an undesirable consequence of radiotherapy that compromises normal oral functions in addition to causing odynophagia and increasing the patient's risk of oral infections and dental caries. Radiation-induced xerostomia is irreversible, and palliative measures to provide symptomatic relief remain the mainstay of treatment. Previously, we identified a splice variant of a cellular kinase, Tousled-like kinase 1B (TLK1B), which when overexpressed protects normal epithelial cells against ionizing radiation (IR)-induced cell death. To address the need to protect salivary glands in patients undergoing regional radiotherapy, we investigated whether preemptive expression of TLK1B in salivary glands protects against IR. In stably-derived salivary cell lines in vitro, TLK1B expression increased cell survival after IR. Cells expressing exogenous TLK1B were less radiosensitive (A5-TLK1B, α/ß=0.67 Gy; ParC5-TLK1B, α/ß=4.3 Gy) compared to control cells (A5-BK, α/ß=1.7 Gy; ParC5-BK, α/ß=32.7 Gy). Using a recombinant adenovirus serotype 5 viral vector for TLK1B gene transfer into rat submandibular salivary glands in vivo, we demonstrated that TLK1B protects the saliva-secreting acinar cells and better preserves salivary gland function against IR relative to control glands. After a single fraction of 16 Gy, the decline in salivary function at 8 weeks was less pronounced in TLK1B-treated animals (40%) as compared to saline-treated controls (67%). Histopathological analysis demonstrated increase in acinar atrophy, decrease in acinar cell number, and increase in inflammatory infiltrate and fibrosis in irradiated control tissues relative to TLK1B-treated glands. These results show the radioprotective benefits of TLK1B and implicate its usefulness in the management of regional radiotherapy-induced xerostomia.


Asunto(s)
Proteínas Serina-Treonina Quinasas/administración & dosificación , Radioterapia/efectos adversos , Glándulas Salivales/efectos de la radiación , Xerostomía/prevención & control , Adenoviridae , Animales , Línea Celular , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes , Humanos , Inmunohistoquímica , Técnicas In Vitro , Ratas , Glándulas Salivales/metabolismo , Xerostomía/etiología
16.
Coron Artery Dis ; 21(6): 369-77, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20613497

RESUMEN

OBJECTIVE: Cardiac fibroblasts (CFs) regulate myocardial fibrosis and remodeling through proliferation and differentiation. Transforming growth factor-beta1 (TGF-beta1) plays a critical role in the development of myocardial fibrosis after myocardial infarction (MI). The aim of this study was to investigate the effects of inhibiting TGF-beta1 action on myofibroblast differentiation and cardiac function after MI. METHODS: CFs were cultured and treated, respectively with PBS, TGF-beta1, soluble TGF-beta1 receptor II (sTbetaRII), and TGF-beta1 plus sTbetaRII. Proliferation CFs were measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Myofibroblast differentiation was examined by alpha-smooth muscle actin immunostaining. Expression of P-Smad2 and Smad2/3 was determined by immunostaining and western blot analysis. Four days after ligation of left anterior descending coronary artery, sTbetaRII was injected into injured heart. Two weeks after sTbetaRII administration, myofibroblast differentiation was measured with alpha-smooth muscle actin immunostaining. Four weeks after sTbetaRII administration, cardiac function was evaluated by hemodynamic measurements. Weight parameters, infarct size, and collagen fiber were detected with an earlier experimental method. RESULTS: Compared with TGF-beta1, TGF-beta1 plus sTbetaRII significantly decreased cell proliferation, myofibroblast differentiation, and expression of P-Smad2 in CFs (P<0.05). Two weeks after sTbetaRII administration, myofibroblast differentiation in MI rats treated with sTbetaRII was reduced compared with MI group (P<0.05). Four weeks after sTbetaRII administration, MI rats that received sTbetaRII showed significantly higher cardiac function and lower in weight parameters, infarct size, and collagen fiber than that of MI group (P<0.05). CONCLUSION: sTbetaRII could inhibit TGF-beta1-induced myofibroblast differentiation, alleviate myocardial fibrosis and remodeling, and improve ischemic cardiac function after MI.


Asunto(s)
Fármacos Cardiovasculares/administración & dosificación , Diferenciación Celular/efectos de los fármacos , Infarto del Miocardio/tratamiento farmacológico , Miocardio/metabolismo , Miofibroblastos/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/administración & dosificación , Receptores de Factores de Crecimiento Transformadores beta/administración & dosificación , Factor de Crecimiento Transformador beta1/metabolismo , Actinas/metabolismo , Animales , Animales Recién Nacidos , Western Blotting , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Fibrosis , Inmunohistoquímica , Microinyecciones , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/patología , Miofibroblastos/metabolismo , Miofibroblastos/patología , Fosforilación , Ratas , Ratas Sprague-Dawley , Receptor Tipo II de Factor de Crecimiento Transformador beta , Recuperación de la Función , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factores de Tiempo , Función Ventricular Izquierda , Presión Ventricular , Remodelación Ventricular
17.
J Immunol ; 184(9): 5130-40, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20348421

RESUMEN

Patients receiving hematopoietic stem cell transplantation or bone marrow transplantation (BMT) as therapy for various malignancies or autoimmune diseases have an increased risk for infectious complications posttransplant, especially in the lung. We have used BMT in mice and murine gammaherpesvirus, gammaHV-68, to study the efficacy of adaptive immune responses post-BMT. Five weeks posttransplant, mice have fully reconstituted their hematopoietic lineages in both the lung and periphery. When challenged with virus, however, BMT mice have a reduced ability to clear lytic virus from the lung. Defective viral control in BMT mice is not related to impaired leukocyte recruitment or defective APC function. Rather, BMT mice are characterized by defective CD4 cell proliferation, skewing of effector CD4 T cells from a Th1 to a Th17 phenotype, and an immunosuppressive lung environment at the time of infection that includes overexpression of TGF-beta1 and PGE(2) and increased numbers of regulatory T cells. Neither indomethacin treatment to block PG synthesis nor anti-CD25 depletion of regulatory T cells improved antiviral host defense post-BMT. Transplanting mice with transgenic bone marrow expressing a dominant-negative TGF-betaRII under the permissive CD4 promoter created mice in which effector CD4 and CD8 cells were unresponsive to TGF-beta1. Mice with TGF-beta1-nonresponsive effector T cells had restored antiviral immunity and improved Th1 responses post-BMT. Thus, our results indicate that overexpression of TGF-beta1 following myeloablative conditioning post-BMT results in impaired effector T cell responses to viral infection.


Asunto(s)
Trasplante de Médula Ósea/inmunología , Infecciones por Herpesviridae/inmunología , Neumonía Viral/inmunología , Rhadinovirus/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta1/efectos adversos , Factor de Crecimiento Transformador beta1/biosíntesis , Infecciones Tumorales por Virus/inmunología , Animales , Trasplante de Médula Ósea/efectos adversos , Diferenciación Celular/inmunología , Proliferación Celular , Inhibidores de Crecimiento/efectos adversos , Inhibidores de Crecimiento/biosíntesis , Inhibidores de Crecimiento/genética , Infecciones por Herpesviridae/terapia , Infecciones por Herpesviridae/virología , Interleucina-17/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Neumonía Viral/terapia , Neumonía Viral/virología , Proteínas Serina-Treonina Quinasas/administración & dosificación , Proteínas Serina-Treonina Quinasas/uso terapéutico , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/administración & dosificación , Receptores de Factores de Crecimiento Transformadores beta/uso terapéutico , Linfocitos T Reguladores/patología , Linfocitos T Reguladores/virología , Células TH1/inmunología , Células TH1/patología , Células TH1/virología , Factor de Crecimiento Transformador beta1/genética , Acondicionamiento Pretrasplante/métodos , Infecciones Tumorales por Virus/terapia , Infecciones Tumorales por Virus/virología
18.
Clin Exp Pharmacol Physiol ; 35(11): 1288-93, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18505441

RESUMEN

1. In the present study, we examined the effects of inhibiting transforming growth factor (TGF)-beta in a mouse model of diabetic nephropathy. 2. An adenovirus harbouring the gene encoding soluble TGF-beta type II receptor (Ad.CAG-sTbetaRII), a competitive inhibitor of TGF-beta, was injected into hindlimb muscles (systemic delivery) of mice 5 weeks after the induction of diabetes with streptozotocin. The control group was injected with an adenovirus encoding the LacZ gene (Ad-LacZ). 3. Five weeks after administration, anti-TGF-beta gene therapy was found to have had no effect on renal function, albuminuria or glucose metabolism in mice with diabetic nephropathy. Nonetheless, this gene therapy did significantly reduce fibrosis in both glomeruli and renal tubules. These effects were accompanied by attenuation of the increased expression of alpha-smooth muscle actin normally seen in kidneys of diabetic mice and better preservation of glomerular cell numbers, although the thickness of the glomerular capillary basement membrane was unchanged. The plasma concentration of soluble TGF-beta type II receptor peaked on Day 7 after treatment, but was undetectable by Day 14. Moreover, a second treatment with Ad.CAG-sTbetaRII failed to prolong the interval of gene product expression in the blood. 4. The present anti-TGF-beta gene therapy showed a significant antifibrotic effect in a model of diabetic nephropathy, but failed to improve renal function. The inadequacy of the observed effect is likely due to the relatively short interval of gene product expression. This problem will have to be overcome if gene therapies for slowly progressing diseases, like diabetic nephropathy, are to be realised.


Asunto(s)
Adenoviridae/genética , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/terapia , Modelos Animales de Enfermedad , Vectores Genéticos/administración & dosificación , Proteínas Serina-Treonina Quinasas/administración & dosificación , Proteínas Serina-Treonina Quinasas/genética , Receptores de Factores de Crecimiento Transformadores beta/administración & dosificación , Receptores de Factores de Crecimiento Transformadores beta/genética , Animales , Pollos , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/terapia , Nefropatías Diabéticas/patología , Femenino , Terapia Genética/métodos , Vectores Genéticos/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Conejos , Receptor Tipo II de Factor de Crecimiento Transformador beta , Solubilidad , Factor de Crecimiento Transformador beta/antagonistas & inhibidores
19.
Blood Cells Mol Dis ; 40(1): 119-21, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-17988908

RESUMEN

Trib2 is a member of the Trib family of serine/threonine kinase-like proteins (Trib1, Trib2, Trib3). Mice reconstituted with hematopoietic stem cells (HSC) retrovirally expressing Trib2 uniformly developed fatal transplantable acute myelogenous leukemia (AML). Trib2-induced AML was clonal and we sought to identify cooperating genes in Trib2-induced AML. Using Splinkerette PCR techniques, we identified proviral insertion near HoxA9 in a Trib2 monoclonal tumor, which resulted in greatly elevated HoxA9 expression. Mice reconstituted with HSC cotransduced with HoxA9 and Trib2 had accelerated onset of AML compared to either gene alone. These data identify Trib2 and HoxA9 as cooperating genes in AML.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Proteínas de Homeodominio/farmacología , Péptidos y Proteínas de Señalización Intracelular/farmacología , Leucemia Mieloide Aguda/etiología , Proteínas Serina-Treonina Quinasas/farmacología , Animales , Células Madre Hematopoyéticas/metabolismo , Proteínas de Homeodominio/administración & dosificación , Péptidos y Proteínas de Señalización Intracelular/administración & dosificación , Leucemia Mieloide Aguda/patología , Ratones , Proteínas Oncogénicas , Proteínas Serina-Treonina Quinasas/administración & dosificación , Transducción Genética
20.
Eur J Neurosci ; 26(12): 3411-20, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18052980

RESUMEN

Previously, we have established that a product of the doublecortin-like kinase (DCLK) gene, DCLK-short, is cleaved by caspases during serum deprivation. Subsequently, the N-terminal cleavage product of DCLK-short facilitates apoptosis in the neuroblastoma cell line NG108. As this N-terminal cleavage product is highly homologous to calcium/calmodulin-dependent protein kinase-related peptide (CARP), another DCLK gene splice variant, we aimed to determine the possible apoptotic properties of CARP in vivo and in vitro. We report highly specific CARP expression in apoptotic granule cells in the rat dentate gyrus after adrenalectomy relative to healthy granule cells. CARP is significantly upregulated in the suprapyramidal blade of the dentate gyrus, with varying levels of upregulation, depending on the extent of adrenalectomy-induced apoptosis. Similar to the caspase-cleaved N-terminus of DCLK-short, CARP overexpression itself facilitated apoptosis in serum-deprived NG108 cells. Furthermore, CARP facilitated polymerization of tubulin in vitro and was capable of interacting with growth factor receptor-bound protein 2, an intracellular protein involved in vesicle trafficking. Together, our data demonstrate a facilitating role for CARP in the apoptotic process in granule cell populations sensitive to adrenalectomy, and suggest that this proapoptotic effect is mediated by increasing the stability of the microtubule cytoskeleton.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Apoptosis/fisiología , Giro Dentado/fisiología , Neuronas/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Adrenalectomía , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Giro Dentado/citología , Proteína Doblecortina , Quinasas Similares a Doblecortina , Interacciones Farmacológicas , Proteína Adaptadora GRB2/farmacología , Técnicas In Vitro , Masculino , Microinyecciones , Neuronas/efectos de los fármacos , Polímeros/metabolismo , Proteínas Serina-Treonina Quinasas/administración & dosificación , Proteínas Serina-Treonina Quinasas/farmacología , Ratas , Ratas Wistar , Proteínas Recombinantes/farmacología , Tubulina (Proteína)/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...