Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Toxicol Appl Pharmacol ; 475: 116655, 2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37579951

RESUMEN

Hepatic fibrosis is the pathological repair response of the liver to chronic injury; hepatic stellate cell (HSC) activation is the central link in the pathogenesis of hepatic fibrosis. Previously, we showed that kinetin, a plant cytokinin hormone, has a protective effect on CCl4-induced liver injury in mice. However, the role of kinetin in liver fibrosis remains unclear. We aimed to study these protective effects and to determine the mechanisms by which kinetin mediates HSC activation and apoptosis. For this purpose, the human HSC line LX-2 was treated with 10 ng/ml transforming growth factor-ß1 (TGF-ß1) for 24 h to stimulate activation. We found that treatment with kinetin at the sub-cytotoxic dose of 40 µg/ml for 48 h reduced the expression of the HSC activation marker α-SMA and inhibited the secretion of extracellular matrix proteins. In addition, kinetin was found to inhibit the proliferation and migration of LX-2 cells. We found that kinetin induced apoptosis in LX-2 cells by increasing the level of cleaved-caspase 3 and the Bax-to-Bcl-2 ratio. Interestingly, these effect were not observed in quiescent HSCs, suggesting that they are activation-dependent. Further study showed that kinetin attenuates activation and promotes apoptosis of LX-2 cells in vitro in part by suppressing the TGF-ß1/Smad signaling pathway.


Asunto(s)
Células Estrelladas Hepáticas , Factor de Crecimiento Transformador beta1 , Humanos , Ratones , Animales , Factor de Crecimiento Transformador beta1/metabolismo , Cinetina/metabolismo , Cinetina/farmacología , Cinetina/uso terapéutico , Cirrosis Hepática/metabolismo , Transducción de Señal , Apoptosis
2.
Int J Biol Sci ; 16(13): 2382-2391, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32760206

RESUMEN

COVID-19 is a public health emergency that has rapidly spread to over 200 countries and regions, and no effective treatment has been established to date. Severe and critical cases have been associated with higher mortality due to acute respiratory distress syndrome (ARDS) and cytokine storm. Based on the novelty and recent emergence of COVID-19, no effective treatment regimen has been identified, thus prompting clinicians to engage in drug repurposing to address the immediate therapeutic need. This study focused on the molecular target angiotensin-converting enzyme 2 (ACE2) of SARS-CoV-2 and screened a group of ACE2 agonists by bioinformatics. Glucocorticoids are a type of ACE2 activator. We verified the efficacy of nine chemicals on regulating ACE2 expression in human GES-1, an upper digestive tract epithelial cell line, and THP-1, a human monocyte cell line, and found that several glucocorticoids imparted activating effects on ACE2 in both cell lines. The drugs triciribine and kinetin riboside activate ACE2 expression or inhibit IL-6 production in macrophages to some extent. In addition, we compared the efficacies of several glucocorticoids. Hydrocortisone showed the strongest effect on ACE2 activation, followed by prednisolone, dexamethasone, and methylprednisolone. We retrospectively analyzed the therapeutic efficacy of nine severe or critical patients from a cohort of 90 COVID-19 cases, who received medium to small doses of glucocorticoids from our integrated medical team in Wuhan. Seven out of nine patients revealed significant improvement in clinical parameters and chest CT images. This study provides experimental and clinical evidence that medium-to-low-dose glucocorticoids may play a protective role in the respiratory and digestive systems by activating ACE2 and suppressing cytokine storm.


Asunto(s)
Infecciones por Coronavirus/tratamiento farmacológico , Glucocorticoides/uso terapéutico , Interleucina-6/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/tratamiento farmacológico , Adenosina/uso terapéutico , Adulto , Anciano , Enzima Convertidora de Angiotensina 2 , Antivirales/uso terapéutico , Betacoronavirus , COVID-19 , Línea Celular , Línea Celular Tumoral , Infecciones por Coronavirus/metabolismo , Citocinas/metabolismo , Células Epiteliales/virología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Hidrocortisona/uso terapéutico , Cinetina/uso terapéutico , Macrófagos/efectos de los fármacos , Masculino , Persona de Mediana Edad , Monocitos/virología , Pandemias , Neumonía Viral/metabolismo , Estudios Retrospectivos , Ribonucleósidos/uso terapéutico , SARS-CoV-2 , Transcriptoma , Tratamiento Farmacológico de COVID-19
3.
Ageing Res Rev ; 55: 100958, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31479763

RESUMEN

It has been more than 60 years since the discovery of kinetin, the first known member of a group of plant hormones called cytokinins. In this review we summarize the health-promoting activity of kinetin in animal systems, ranging from cells cultured in vitro through invertebrates to mammals. Kinetin has been shown to modulate aging, to delay age-related physiological decline and to protect against some neurodegenerative diseases. We also review studies on its mechanism of action, as well as point out gaps in our current knowledge.


Asunto(s)
Envejecimiento , Envejecimiento Saludable , Cinetina/uso terapéutico , Animales , Citocininas , Humanos , Cinetina/farmacología
4.
Am J Hum Genet ; 104(4): 638-650, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30905397

RESUMEN

Familial dysautonomia (FD) is a recessive neurodegenerative disease caused by a splice mutation in Elongator complex protein 1 (ELP1, also known as IKBKAP); this mutation leads to variable skipping of exon 20 and to a drastic reduction of ELP1 in the nervous system. Clinically, many of the debilitating aspects of the disease are related to a progressive loss of proprioception; this loss leads to severe gait ataxia, spinal deformities, and respiratory insufficiency due to neuromuscular incoordination. There is currently no effective treatment for FD, and the disease is ultimately fatal. The development of a drug that targets the underlying molecular defect provides hope that the drastic peripheral neurodegeneration characteristic of FD can be halted. We demonstrate herein that the FD mouse TgFD9;IkbkapΔ20/flox recapitulates the proprioceptive impairment observed in individuals with FD, and we provide the in vivo evidence that postnatal correction, promoted by the small molecule kinetin, of the mutant ELP1 splicing can rescue neurological phenotypes in FD. Daily administration of kinetin starting at birth improves sensory-motor coordination and prevents the onset of spinal abnormalities by stopping the loss of proprioceptive neurons. These phenotypic improvements correlate with increased amounts of full-length ELP1 mRNA and protein in multiple tissues, including in the peripheral nervous system (PNS). Our results show that postnatal correction of the underlying ELP1 splicing defect can rescue devastating disease phenotypes and is therefore a viable therapeutic approach for persons with FD.


Asunto(s)
Disautonomía Familiar/terapia , Cinetina/uso terapéutico , Propiocepción , Empalme del ARN , Factores de Elongación Transcripcional/genética , Alelos , Animales , Conducta Animal , Línea Celular , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Disautonomía Familiar/genética , Exones , Fibroblastos , Genotipo , Humanos , Intrones , Cinetina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación , Neuronas/metabolismo , Fenotipo
5.
Blood ; 119(5): 1200-7, 2012 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-22160482

RESUMEN

Gene regulatory networks that govern hematopoietic stem cells (HSCs) and leukemia-initiating cells (L-ICs) are deeply entangled. Thus, the discovery of compounds that target L-ICs while sparing HSC is an attractive but difficult endeavor. Presently, most screening approaches fail to counter-screen compounds against normal hematopoietic stem/progenitor cells (HSPCs). Here, we present a multistep in vitro and in vivo approach to identify compounds that can target L-ICs in acute myeloid leukemia (AML). A high-throughput screen of 4000 compounds on novel leukemia cell lines derived from human experimental leukemogenesis models yielded 80 hits, of which 10 were less toxic to HSPC. We characterized a single compound, kinetin riboside (KR), on AML L-ICs and HSPCs. KR demonstrated comparable efficacy to standard therapies against blast cells in 63 primary leukemias. In vitro, KR targeted the L-IC-enriched CD34(+)CD38(-) AML fraction, while sparing HSPC-enriched fractions, although these effects were mitigated on HSC assayed in vivo. KR eliminated L-ICs in 2 of 4 primary AML samples when assayed in vivo and highlights the importance of in vivo L-IC and HSC assays to measure function. Overall, we provide a novel approach to screen large drug libraries for the discovery of anti-L-IC compounds for human leukemias.


Asunto(s)
Adenosina/uso terapéutico , Ensayos Analíticos de Alto Rendimiento/métodos , Cinetina/uso terapéutico , Leucemia/tratamiento farmacológico , Leucemia/patología , Células Madre Neoplásicas/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/análisis , Adenosina/análisis , Adenosina/aislamiento & purificación , Adenosina/farmacología , Animales , Antineoplásicos/análisis , Antineoplásicos/aislamiento & purificación , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Humanos , Cinetina/análisis , Cinetina/aislamiento & purificación , Cinetina/farmacología , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Células Madre Neoplásicas/patología , Resultado del Tratamiento , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Cosmet Dermatol ; 9(3): 218-25, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20883295

RESUMEN

New developments in the realm of skin rejuvenation such as phytotherapy are at an astounding increasing pace in the cosmeceutical market. Yet, many of these products that are classified as cosmeceuticals are tested less vigorously and do not have to be approved by the Food and Drug Administration to establish efficacy and safety. Thus, as clinicians, we must ask the question, "Is there science-based evidence to validate the mechanism of these new treatments?" We assessed the top anti-aging creams currently on the market specifically evaluating their botanical ingredients. Some of the most common botanicals that are hot off the market are: Rosmarinus officinalis, Vitis vinifera (grape seed extract), Citronellol, Limonene, Oenothera biennis (evening primrose), Glycyrrhiza glabra (licorice extract), Aframomum angustifolium seed extract, Diosgenin (wild yam), N6 furfuryladenine (kinetin), and Ergothioneine. Through researching each of these botanical ingredients, we have concluded that randomized controlled trials are still needed in this area, but there is promise in some of these ingredients and science to validate them.


Asunto(s)
Fitoterapia/métodos , Extractos Vegetales/uso terapéutico , Envejecimiento de la Piel/efectos de los fármacos , Monoterpenos Acíclicos , Antioxidantes/uso terapéutico , Ciclohexenos/uso terapéutico , Diosgenina , Ergotioneína/uso terapéutico , Extracto de Semillas de Uva/uso terapéutico , Humanos , Cinetina/uso terapéutico , Limoneno , Monoterpenos/uso terapéutico , Oenothera , Preparaciones de Plantas/uso terapéutico , Rosa , Terpenos/uso terapéutico , Zingiberaceae
7.
Invest New Drugs ; 28(3): 299-305, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19390782

RESUMEN

The effects of the cyclin-dependent kinase inhibitors roscovitine and olomoucine on DNA synthesis rate during normal rat brain development were studied by using short time (90 min) incubation. Both purine analogues at 100 microM concentration decreased the DNA synthesis of rat cerebral cortex in an age-dependent manner. The maximum inhibitory effect (approximately 90% for roscovitine, approximately 60% for olomoucine) occurred in rats of 2-13 days postnatal age. In adult rats (> 60 days postnatal age), the effect of both purine analogues was low. Roscovitine even at 200 microM concentration did not inhibit the fraction of DNA synthesis insensitive to hydroxyurea (unscheduled DNA synthesis (UDS)). In addition, in the RG2 rat glioma model, roscovitine produced a strong inhibition of DNA synthesis in glioma cells when compared to adult normal tissue. Since in adult rat brain more than 60% of DNA synthesis is related to DNA repair, usually measured as UDS, our results indicate that roscovitine strongly blocks ongoing DNA synthesis connected with replicative processes.


Asunto(s)
Anticarcinógenos/uso terapéutico , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , ADN/biosíntesis , Glioma/tratamiento farmacológico , Inhibidores de la Síntesis del Ácido Nucleico/uso terapéutico , Purinas/uso terapéutico , Factores de Edad , Animales , Animales Recién Nacidos , Anticarcinógenos/farmacología , Línea Celular Tumoral , ADN/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Técnicas In Vitro , Cinetina/farmacología , Cinetina/uso terapéutico , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Purinas/farmacología , Ratas , Ratas Sprague-Dawley , Roscovitina
8.
Nature ; 461(7262): 402-6, 2009 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-19693009

RESUMEN

The isolation of human induced pluripotent stem cells (iPSCs) offers a new strategy for modelling human disease. Recent studies have reported the derivation and differentiation of disease-specific human iPSCs. However, a key challenge in the field is the demonstration of disease-related phenotypes and the ability to model pathogenesis and treatment of disease in iPSCs. Familial dysautonomia (FD) is a rare but fatal peripheral neuropathy, caused by a point mutation in the IKBKAP gene involved in transcriptional elongation. The disease is characterized by the depletion of autonomic and sensory neurons. The specificity to the peripheral nervous system and the mechanism of neuron loss in FD are poorly understood owing to the lack of an appropriate model system. Here we report the derivation of patient-specific FD-iPSCs and the directed differentiation into cells of all three germ layers including peripheral neurons. Gene expression analysis in purified FD-iPSC-derived lineages demonstrates tissue-specific mis-splicing of IKBKAP in vitro. Patient-specific neural crest precursors express particularly low levels of normal IKBKAP transcript, suggesting a mechanism for disease specificity. FD pathogenesis is further characterized by transcriptome analysis and cell-based assays revealing marked defects in neurogenic differentiation and migration behaviour. Furthermore, we use FD-iPSCs for validating the potency of candidate drugs in reversing aberrant splicing and ameliorating neuronal differentiation and migration. Our study illustrates the promise of iPSC technology for gaining new insights into human disease pathogenesis and treatment.


Asunto(s)
Disautonomía Familiar/patología , Disautonomía Familiar/terapia , Modelos Biológicos , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/trasplante , Adolescente , Empalme Alternativo/efectos de los fármacos , Empalme Alternativo/genética , Animales , Proteínas Portadoras/genética , Desdiferenciación Celular , Diferenciación Celular , Linaje de la Célula , Movimiento Celular , Células Cultivadas , Niño , Disautonomía Familiar/tratamiento farmacológico , Disautonomía Familiar/genética , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Humanos , Cinetina/farmacología , Cinetina/uso terapéutico , Masculino , Ratones , Cresta Neural/citología , Cresta Neural/efectos de los fármacos , Especificidad de Órganos , Fenotipo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos , Factores de Elongación Transcripcional
9.
Brain Res ; 1264: 85-97, 2009 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-19368812

RESUMEN

The finding of nuclear translocation of cathepsin L and its ability to process the CDP/Cux transcription factor uncovers an important role of cathepsin L in control of cell cycle progression. As the expression of certain cell cycle regulators is associated with nigral neuronal death, the present study was sought to investigate if nuclear translocation of cathepsin L and expression of certain cyclins were induced in DA neurons by 6-hydroxydopamine (6-OHDA). The neuroprotective effects of the cell cycle inhibitor olomoucine against 6-OHDA-induced death of nigral neurons were examined. Using immunocytochemistry and real-time PCR we demonstrated that cyclin D1, cyclin B1 and proliferating cell nuclear antigen (PCNA) were aberrantly expressed in some dopaminergic neurons after 6-OHDA infusion. The nuclear translocation of cathepsin L and up-regulation of LC3, a protein involved in autophagy, were observed in nigral DA neurons. Olomoucine, a cyclin dependent kinase (CDK) inhibitor, reduced contralateral rotations and the loss of TH-positive neurons in substantia nigra induced by lesion with 6-OHDA. Pretreatment of rats or primary DA neurons with olomoucine resulted in a partial blockade of nuclear translocation of cathepsin L. Olomoucine also increased the expression of punctate LC3 immunoreactivity, indicating activation of autophagy. These findings suggest that olomoucine may exert neuroprotective effects through inhibiting cathepsin L nuclear translocation and activating autophagy.


Asunto(s)
Autofagia/efectos de los fármacos , Catepsinas/metabolismo , Cisteína Endopeptidasas/metabolismo , Cinetina/farmacología , Enfermedad de Parkinson Secundaria/tratamiento farmacológico , Transporte de Proteínas/efectos de los fármacos , Sustancia Negra/efectos de los fármacos , Análisis de Varianza , Animales , Catepsina L , Recuento de Células , Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Células Cultivadas , Ciclina B/metabolismo , Ciclina D1/metabolismo , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Técnica del Anticuerpo Fluorescente , Cinetina/uso terapéutico , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oxidopamina/toxicidad , Enfermedad de Parkinson Secundaria/inducido químicamente , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sustancia Negra/metabolismo , Sustancia Negra/patología , Factores de Tiempo , Tirosina 3-Monooxigenasa/metabolismo , Regulación hacia Arriba/efectos de los fármacos
10.
Eur Neuropsychopharmacol ; 18(8): 578-88, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18406108

RESUMEN

The aim of the present study was to evaluate whether the activation of Cdk5, a protein that has been suggested to participate in higher cognitive functions, is required for the onset of a sensitized anxiety-related behavior induced by stress. The exposure to restraint enhanced both Cdk5 expression in certain subareas of the septohippocampal system, principally in the lateral septum (LS) and septal Cdk5 kinase activity in rats. Behaviorally, restrained wild type mice showed a behavior indicative of enhanced anxiety in the elevated plus maze (EPM). In contrast, unstressed mice and stressed knockout mice, which lacked the p35 protein, the natural activator of Cdk5, displayed similar anxiety-like behavior in the EPM. Finally, the intra-LS infusion of olomoucine - a Cdk5 inhibitor - blocked the enhanced anxiety in the EPM induced by prior stress in rats. All these data provide evidence that septal Cdk5 is required in the emergence of a sensitized emotional process induced by stress.


Asunto(s)
Ansiedad/patología , Quinasa 5 Dependiente de la Ciclina/metabolismo , Regulación Enzimológica de la Expresión Génica/fisiología , Tabique del Cerebro/enzimología , Estrés Psicológico/complicaciones , Análisis de Varianza , Animales , Ansiedad/etiología , Ansiedad/prevención & control , Conducta Animal , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Cinetina/farmacología , Cinetina/uso terapéutico , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratas , Ratas Wistar
11.
J Cosmet Dermatol ; 6(4): 243-9, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18047609

RESUMEN

BACKGROUND: Kinetin and niacinamide are used in the cosmetic industry as anti-aging agents. Neither the interactive/additive effects of these compounds nor the anti-aging efficacy on Asian skin has been studied. Objective To assess the clinical anti-aging effects and efficacy differences between kinetin plus niacinamide and niacinamide alone vs. vehicle placebo in an Asian cohort. METHODS: Fifty-two Taiwanese subjects were enrolled in a randomized, double-blind, placebo-controlled, split-face comparative study. Group 1 subjects were treated with kinetin 0.03% plus niacinamide 4%, whereas group 2 subjects received niacinamide 4%. The treatment formulation was applied on one side of the face, whereas a placebo was applied on the other for a period of 12 weeks. We used noninvasive biometrological instruments to evaluate a variety of skin parameters at baseline and at weeks 4, 8, and 12. RESULTS: Persistent and significant reductions in spot, pore, wrinkle, and evenness counts were found at weeks 8 and 12 in group 1. A significant increase in corneal hydration status was also evident at week 12, whereas persistent decreases in erythema index were apparent at 8 and 12 weeks. In group 2, significant reductions in pore and evenness counts at week 8 and wrinkle counts at week 12 were noted. CONCLUSION: We found kinetin and niacinamide exert a synergistic anti-aging effect. Our data suggest that these compounds have multiactive, multifunctional, and pluripotent effects on skin. They are also both promising to be included in the cutaneous anti-aging cosmeceuticals in the future.


Asunto(s)
Fármacos Dermatológicos/uso terapéutico , Cinetina/uso terapéutico , Niacinamida/uso terapéutico , Envejecimiento de la Piel , Administración Cutánea , Adulto , Pueblo Asiatico , Fármacos Dermatológicos/administración & dosificación , Método Doble Ciego , Quimioterapia Combinada , Femenino , Humanos , Cinetina/administración & dosificación , Masculino , Persona de Mediana Edad , Niacinamida/administración & dosificación , Índice de Severidad de la Enfermedad , Taiwán , Resultado del Tratamiento
12.
Clin Exp Dermatol ; 32(6): 693-5, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17868391

RESUMEN

Many patients with rosacea are unable to tolerate extended treatment periods with topical agents because of the unusually high skin sensitivity that often accompanies rosacea. Kinetin (N(6)-furfuryladenine) is a plant cytokinin that reportedly helps restore skin barrier function and may be useful to ameliorate the signs and symptoms of rosacea. The purpose of this open-label study was to determine the tolerance and efficacy of twice-daily application of kinetin 0.1% lotion for improving the signs and symptoms of mild to moderate facial rosacea. Subjects applied kinetin 0.1% lotion twice daily to the face, with daily use of a sunscreen of sun protection factor 30. Subjects were evaluated at baseline and at 4-week intervals for 12 weeks to assess efficacy and tolerance. Results of this study suggest that kinetin 0.1% lotion is a well-tolerated moisturizing lotion option for subjects with mild to moderate inflammatory rosacea.


Asunto(s)
Fármacos Dermatológicos/uso terapéutico , Cinetina/uso terapéutico , Rosácea/tratamiento farmacológico , Administración Cutánea , Adulto , Anciano , Fármacos Dermatológicos/efectos adversos , Esquema de Medicación , Emolientes/uso terapéutico , Femenino , Estudios de Seguimiento , Humanos , Cinetina/efectos adversos , Masculino , Persona de Mediana Edad , Rosácea/patología , Índice de Severidad de la Enfermedad , Resultado del Tratamiento
14.
Brain Res ; 1154: 206-14, 2007 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-17482149

RESUMEN

Microglial activation/proliferation and reactive astrogliosis are commonly observed and have been considered to be closely relevant pathological processes during spinal cord injury (SCI). However, the molecular mechanisms underlying this microglial-astroglial interaction are still poorly understood. We showed recently that the continuous injection of the cell cycle inhibitor olomoucine not only markedly suppressed microglial proliferation and associated release of pro-inflammatory cytokines, but also attenuated astroglial scar formation and the lesion cavity and mitigated the functional deficits in rat SCI animal model. In this study, we asked whether microglial activation/proliferation plays an initial role and also necessary in maintaining astrogliosis in SCI model. Our results showed that traumatic induced microglial activation/proliferation precedes astrogliosis, and the up-regulated GFAP expression at both mRNA and protein levels was temporally posterior to the microglial activation. Furthermore, when the cell cycle inhibitor olomoucine was administered only once 1 h post-SCI that should selectively suppress microglial proliferation, the subsequent SCI induced increase in GFAP expression at 1, 2 and 4 weeks was significantly attenuated, suggesting that microglial activation/proliferation played an important role for the later onset astrogliosis after SCI. Consistent with the results that microglial proliferation always precedes astroglial proliferation and there is at present no evidence of other astroglial precursors, which as always does not mean that they will not be uncovered by further searching, and in view of the fact that microglial-derived pro-inflammatory cytokines promote astrogliosis as we reported recently, these findings together suggest that by release of cytokines and other soluble products, the early onset microglial activation/proliferation can significantly influence the subsequent development of reactive astrogliosis and glial scar formation in SCI animal model.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/uso terapéutico , Gliosis/tratamiento farmacológico , Cinetina/uso terapéutico , Microglía/efectos de los fármacos , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/patología , Análisis de Varianza , Animales , Antígeno CD11b/metabolismo , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Gliosis/etiología , Antígeno Ki-67/metabolismo , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Traumatismos de la Médula Espinal/complicaciones , Factores de Tiempo
15.
J Mol Med (Berl) ; 85(2): 149-61, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17206408

RESUMEN

Mutations that affect the splicing of pre-mRNA are a major cause of human disease. Familial dysautonomia (FD) is a recessive neurodegenerative disease caused by a T to C transition at base pair 6 of IKBKAP intron 20. This mutation results in variable tissue-specific skipping of exon 20. Previously, we reported that the plant cytokinin kinetin dramatically increases exon 20 inclusion in RNA isolated from cultured FD cells. The goal of the current study was to investigate the nature of the FD splicing defect and the mechanism by which kinetin improves exon inclusion, as such knowledge will facilitate the development of future therapeutics aimed at regulating mRNA splicing. In this study, we demonstrate that treatment of FD lymphoblast cell lines with kinetin increases IKBKAP mRNA and IKAP protein to normal levels. Using a series of minigene constructs, we show that deletion of a region at the end of IKBKAP exon 20 disrupts the ability of kinetin to improve exon inclusion, pinpointing a kinetin responsive sequence element. We next performed a screen of endogenously expressed genes with multiple isoforms resulting from exon skipping events and show that kinetin's ability to improve exon inclusion is not limited to IKBKAP. Lastly, we highlight the potential of kinetin for the treatment of other human splicing disorders by showing correction of a splicing defect in neurofibromatosis.


Asunto(s)
Proteínas Portadoras/genética , Disautonomía Familiar/tratamiento farmacológico , Cinetina/uso terapéutico , Empalme del ARN/efectos de los fármacos , Proteínas Portadoras/análisis , Proteínas Portadoras/efectos de los fármacos , Línea Celular Tumoral , Exones/efectos de los fármacos , Humanos , Cinetina/farmacología , Neurofibromatosis/tratamiento farmacológico , Neurofibromatosis/genética , ARN Mensajero/análisis , ARN Mensajero/efectos de los fármacos , Factores de Elongación Transcripcional
17.
Brain Res ; 1135(1): 177-85, 2007 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-17188663

RESUMEN

The spinal cord is well known to undergo inflammatory reactions in response to traumatic injury. Activation and proliferation of microglial cells, with associated proinflammatory cytokines expression, plays an important role in the secondary damage following spinal cord injury. It is likely that microglial cells are at the center of injury cascade and are targets for treatments of CNS traumatic diseases. Recently, we have demonstrated that the cell cycle inhibitor olomoucine attenuates astroglial proliferation and glial scar formation, decreases lesion cavity and mitigates functional deficits after spinal cord injury (SCI) in rats [Tian, D.S., Yu, Z.Y., Xie, M.J., Bu, B.T., Witte, O.W., Wang, W., 2006. Suppression of astroglial scar formation and enhanced axonal regeneration associated with functional recovery in a spinal cord injury rat model by the cell cycle inhibitor olomoucine. J. Neurosci. Res. 84, 1053-1063]. Whether neuroprotective effects of cell cycle inhibition are involved in attenuation of microglial induced inflammation awaits to be elucidated. In the present study, we sought to determine the influence of olomoucine on microglial proliferation with associated inflammatory response after spinal cord injury. Tissue edema formation, microglial response and neuronal cell death were quantified in rats subjected to spinal cord hemisection. Microglial proliferation and neuronal apoptosis were observed by immunofluorescence. Level of the proinflammatory cytokine interleukin-1beta (IL-1beta) expression in the injured cord was determined by Western blot analysis. Our results showed that the cell cycle inhibitor olomoucine, administered at 1 h post injury, significantly suppressed microglial proliferation and produced a remarkable reduction of tissue edema formation. In the olomoucine-treated group, a significant reduction of activated and/or proliferated microglial induced IL-1beta expression was observed 24 h after SCI. Moreover, olomoucine evidently attenuated the number of apoptotic neurons after SCI. Our findings suggest that modulation of microglial proliferation with associated proinflammatory cytokine expression may be a mechanism of cell cycle inhibition-mediated neuroprotections in the CNS trauma.


Asunto(s)
Ciclo Celular/fisiología , Microglía/patología , Neuronas/patología , Traumatismos de la Médula Espinal/complicaciones , Traumatismos de la Médula Espinal/patología , Análisis de Varianza , Animales , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/etiología , Edema Encefálico/patología , Antígeno CD11b/metabolismo , Ciclo Celular/efectos de los fármacos , Muerte Celular , Proliferación Celular/efectos de los fármacos , Citocinas/metabolismo , Fragmentación del ADN , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Femenino , Inmunohistoquímica/métodos , Antígeno Ki-67/metabolismo , Cinetina/farmacología , Cinetina/uso terapéutico , Fosfopiruvato Hidratasa/metabolismo , Ratas , Ratas Sprague-Dawley
18.
Mol Neurobiol ; 34(1): 27-50, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17003520

RESUMEN

Cyclin-dependent kinases (Cdks) play a key role in orchestrating the coordination of cell cycle progression in proliferating cells. The escape from the proper control of the cell cycle by the upregulation of cyclins or aberrant activation of Cdks leads to malignant transformation. In quiescent cells and/or terminally differentiated cells, the expression pattern and activity of Cdks is altered. In postmitotic neurons, expression of mitotic kinases is downregulated, whereas Cdk5 expression becomes upregulated. Similarly to other Cdks, free Cdk5 displays no enzymatic activity and requires complex formation with a specific regulatory subunit. Two activators of Cdk5 have been identified. p35 and its isoform p39 bind to, and thereby activate, Cdk5. Unlike mitotic kinases, Cdk5 does not require activating phosphorylation within the T-loop. Because p35 is a short-lived protein, the p35/Cdk5 complexes are unstable. The stability of the p35 protein is regulated by its Cdk5-mediated phosphorylation of p35. Activated p35/Cdk5 kinase phosphorylates numerous physiological targets. The proper phosphorylation of the most important substrates, such as tau protein and neurofilament H, is essential for the correct regulation of the cytoskeletal organization, thereby regulating cell adhesion, motility, and synaptic plasticity. Moreover, Cdk5 regulates the activity of the p53 tumor suppressor via phosphorylation. p53 is upregulated in multiple neuronal death paradigms, including hypoxia, ischemia, and excitotoxicity, and plays a key role in the induction of apoptosis. On the other hand, an abnormally high expression and elevated activity of Cdk5 was observed in neurodegenerative diseases, suggesting the application of Cdk inhibitors for their therapy. Considering the action of some Cdk inhibitors on the expression and activity of the p53 protein, their therapeutic efficacy must be carefully evaluated.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/metabolismo , Enfermedades Neurodegenerativas , Inhibidores de Proteínas Quinasas , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis/fisiología , Isquemia Encefálica , Ciclo Celular/fisiología , Humanos , Cinetina/química , Cinetina/uso terapéutico , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , Neuronas/citología , Neuronas/fisiología , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Purinas/química , Purinas/uso terapéutico , Roscovitina
19.
J Neurosci Res ; 84(5): 1053-63, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16862564

RESUMEN

It is well established that axons of the adult mammalian CNS are capable of regrowing only a limited amount after injury. Astrocytes are believed to play a crucial role in the failure to regenerate, producing multiple inhibitory proteoglycans, such as chondroitin sulphate proteoglycans (CSPGs). After spinal cord injury (SCI), astrocytes become hypertrophic and proliferative and form a dense network of astroglial processes at the site of lesion constituting a physical and biochemical barrier. Down-regulations of astroglial proliferation and inhibitory CSPG production might facilitate axonal regeneration. Recent reports indicated that aberrant activation of cell cycle machinery contributed to overproliferation and apoptosis of cells in various insults. In the present study, we sought to determine whether a cell cycle inhibitior, olomoucine, would decrease neuronal cell death, limit astroglial proliferation and production of inhibitory CSPGs, and eventually enhance the functional compensation after SCI in rats. Our results showed that up-regulations of cell cycle components were closely associated with neuronal cell death and astroglial proliferation as well as the production of CSPGs after SCI. Meanwhile, administration of olomoucine, a selective cell cycle kinase (CDK) inhibitor, has remarkably reduced the up-regulated cell cycle proteins and then decreased neuronal cell death, astroglial proliferation, and accumulation of CSPGs. More importantly, the treatment with olomoucine has also increased expression of growth-associated proteins-43, reduced cavity formation, and improved functional deficits. We consider that suppressing astroglial cell cycle in acute SCIs is beneficial to axonal growth. In the future, therapeutic strategies can be designed to achieve efficient axonal regeneration and functional compensation after traumatic CNS injury.


Asunto(s)
Astrocitos/efectos de los fármacos , Inhibidores Enzimáticos/uso terapéutico , Cinetina/uso terapéutico , Regeneración Nerviosa/efectos de los fármacos , Recuperación de la Función/efectos de los fármacos , Traumatismos de la Médula Espinal/tratamiento farmacológico , Análisis de Varianza , Animales , Conducta Animal , Western Blotting/métodos , Recuento de Células/métodos , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Cicatriz/tratamiento farmacológico , Cicatriz/patología , Modelos Animales de Enfermedad , Femenino , Inmunohistoquímica/métodos , Etiquetado Corte-Fin in Situ/métodos , Proteínas del Tejido Nervioso/metabolismo , Ratas , Ratas Sprague-Dawley , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/fisiopatología , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos
20.
Zhonghua Yi Xue Za Zhi ; 86(13): 901-5, 2006 Apr 04.
Artículo en Chino | MEDLINE | ID: mdl-16759516

RESUMEN

OBJECTIVE: To investigate the effects of olomoucine, a cyclin dependent protein kinase (CDK) inhibitor, on the microenvironment of axonal regeneration after spinal cord injury (SCI). METHODS: Forty-five SD rats were randomly divided into 3 equal groups: SCI group undergoing SCI by hemisection technique and peritoneal injection of dimethyl sulfoxide (DMSO) solution 30 min after the SCI, SCI + olomoucine (SCI + Olo) group undergoing SCI by hemisection technique and peritoneal injection of olomoucine solution 30 min after the SCI, and sham operation group undergoing sham operation and peritoneal injection of DMSO solution 30 min after the operation. Three days after the operation the injured spinal cord segments of 5 rats from each group were taken out. Western blotting was used to detect the expression of the cell cycle related proteins, cyclin A, cyclin B, cyclin E, and proliferating cell nuclear antigen (PCNA). Immunofluorescence (IF) staining was used to detect the expression of glial fibrillary acidic protein (GFAP), growth associated protein-43 (GAP-43) and chondroitin sulphate proteoglycan (CSPG). Four weeks after the operation specimens of the injured spinal cord segment 15 mm in length were taken out from 5 rats in each group to undergo histological examination. The locomotion function of the hindlimbs was determined by modified Gale combined behavioral scoring (SBS) 1 day and 1, 2, 4, 6, and 8 weeks after the operation. RESULTS: Western blotting 3 days after the operation showed that the expressions of cyclin A, cyclin B, cyclin E, and PCNA were very weak in the sham operation group, were significantly increased in the SCI group, and were significantly down-regulated in the SCI + Olo group compared with those of the SCI group. IF staining showed that the number of astrocytes was small and the expressions of GFAP, CSPG, and GAP-43 were weak in the sham operation group; in the SCI group the astrocytic proliferation and glial scar was obvious, and the expressions of GFAP, CSPG, and GAP-43 were significantly increased compared with those of the sham operation group (all P < 0.05); and the astrocytic proliferation was significantly weaker and no obvious glial scar could be seen, and the expressions of GFAP and CSPG were weaker in the SCI + Olo group in comparison with the SCI group, however, the GAP-43 expression of the sham operation group was significantly increased compared with that of the sham operation group (P < 0.05). The hindlimbs of the SCI + Olo group and sham operation group were paralyzed without significant difference in the CBS values between these 2 groups, however, two weeks after the operation, the locomotion function scores at different time points of the SCI + Olo group were all significantly improved in comparison with that of the SCI group (all P < 0.05). CONCLUSION: Olomoucine promotes the recovery of the locomotion function of the paralyzed hindlimbs, probably through microenvironmental improvement of axonal regeneration by inhibiting the glial scar formation and CSPG secretion as well as upregulating the GAP-43 expression.


Asunto(s)
Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Cinetina/farmacología , Regeneración Nerviosa/efectos de los fármacos , Traumatismos de la Médula Espinal/fisiopatología , Animales , Axones/efectos de los fármacos , Axones/fisiología , Western Blotting , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/biosíntesis , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Fluoroinmunoensayo , Proteína GAP-43/biosíntesis , Proteína Ácida Fibrilar de la Glía/biosíntesis , Cinetina/uso terapéutico , Antígeno Nuclear de Célula en Proliferación/biosíntesis , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Traumatismos de la Médula Espinal/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA