Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Acta Biomater ; 151: 588-599, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-36002126

RESUMEN

Hepatocellular carcinoma (HCC) is a common high-mortality malignancy which still needs efficient treatments. HCC patients undergoing extrahepatic metastases are mostly with unsatisfactory prognosis. Therefore, specific attention has been paid to extrahepatic HCC metastasis. We integrated Sorafenib (Sor) and glucose oxidase (GOx) into a N-acetyl-galactosamine (GalNAc) modified zeolitic imidazolate framework (ZIF-8), designated as SG@GR-ZIF-8, for targeted bimodal therapies of chemotherapy and starvation therapy against HCC. The hepatic delivery of SG@GR-ZIF was mediated by the specific recognition of GalNAc residues with asialoglycoprotein (ASGPR) on the liver cell surface. Sor is a clinically approved anti-proliferation and anti-angiogenesis drug for advanced HCC treatment. GOx can efficiently induce cell death and disturb malignant progression by suppressing glucose supply of cancer cells, which is highly associated with metabolic rewiring in metastasis. The nano-formulation exhibit significant anti-metastatic HCC activity against C5WN1 cells, a liver cancer stem cell-like cell line with tumorigenicity and pulmonary metastasis activity. In a subcutaneous C5WN1-tumor carrying mouse model, SG@GR-ZIF exhibits potent synergistic anti-tumor activity with a tumor inhibition rate of 89% and a prolonged survival status. The growth and pulmonary metastasis of HCC in an orthotopic mouse model of HCC was remarkably suppressed in SG@GR-ZIF treated group. The therapeutic strategy targeting energy supply combined with first-line treatment holds great promise for the future treatment of metastatic HCC. STATEMENT OF SIGNIFICANCE: SG@GR-ZIF, a N-acetyl-galactosamine modified metal-organic framework carrying Sorafenib and glucose oxidase, was fabricated for treating metastatic hepatocellular carcinoma (HCC). Sorafenib is an anti-proliferation and anti-angiogenesis drug for advanced HCC treatment. Glucose oxidase blocks energy demand in HCC metastasis by depleting glucose. C5WN1 was used for therapeutic evaluations as a liver cancer stem cell-like cell line with tumorigenicity and pulmonary metastasis activity. In subcutaneous C5WN1-tumor bearing mice, SG@GR-ZIF exhibited a tumor inhibition rate of 89% and prolonged survival period. In orthotopic C5WN1-tumor carrying mice, the growth and pulmonary metastasis of hepatocarcinoma was remarkably suppressed by SG@GR-ZIF. Together, this study suggests the great potential of synergistic chemo/starvation therapy mediated by SG@GR-ZIF for treating metastatic HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Neoplasias Pulmonares , Estructuras Metalorgánicas , Animales , Asialoglicoproteínas/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Galactosamina/uso terapéutico , Glucosa , Glucosa Oxidasa/química , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Neoplasias Pulmonares/secundario , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Ratones , Células Madre Neoplásicas/patología , Sorafenib
2.
Mol Med ; 19: 62-4, 2013 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-23615965

RESUMEN

The erythropoietin receptor (EpoR) was discovered and described in red blood cells (RBCs), stimulating its proliferation and survival. The target in humans for EpoR agonists drugs appears clear-to treat anemia. However, there is evidence of the pleitropic actions of erythropoietin (Epo). For that reason, rhEpo therapy was suggested as a reliable approach for treating a broad range of pathologies, including heart and cardiovascular diseases, neurodegenerative disorders (Parkinson's and Alzheimer's disease), spinal cord injury, stroke, diabetic retinopathy and rare diseases (Friedreich ataxia). Unfortunately, the side effects of rhEpo are also evident. A new generation of nonhematopoietic EpoR agonists drugs (asialoEpo, Cepo and ARA 290) have been investigated and further developed. These EpoR agonists, without the erythropoietic activity of Epo, while preserving its tissue-protective properties, will provide better outcomes in ongoing clinical trials. Nonhematopoietic EpoR agonists represent safer and more effective surrogates for the treatment of several diseases such as brain and peripheral nerve injury, diabetic complications, renal ischemia, rare diseases, myocardial infarction, chronic heart disease and others.


Asunto(s)
Receptores de Eritropoyetina/agonistas , Animales , Asialoglicoproteínas/farmacología , Asialoglicoproteínas/uso terapéutico , Encefalopatías/tratamiento farmacológico , Enfermedades Cardiovasculares/tratamiento farmacológico , Complicaciones de la Diabetes/tratamiento farmacológico , Eritropoyetina/análogos & derivados , Eritropoyetina/farmacología , Eritropoyetina/uso terapéutico , Humanos , Oligopéptidos/farmacología , Oligopéptidos/uso terapéutico
3.
Stem Cells ; 30(10): 2234-47, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22890889

RESUMEN

Perinatal hypoxia-ischemia (HI) frequently causes white-matter injury, leading to severe neurological deficits and mortality, and only limited therapeutic options exist. The white matter of animal models and human patients with HI-induced brain injury contains increased numbers of oligodendrocyte progenitor cells (OPCs). However, the origin and fates of these OPCs and their potential to repair injured white matter remain unclear. Here, using cell-type- and region-specific genetic labeling methods in a mouse HI model, we characterized the Olig2-expressing OPCs. We found that after HI, Olig2+ cells increased in the posterior part of the subventricular zone (pSVZ) and migrated into the injured white matter. However, their oligodendrocytic differentiation efficiency was severely compromised compared with the OPCs in normal tissue, indicating the need for an intervention to promote their differentiation. Erythropoietin (EPO) treatment is a promising candidate, but it has detrimental effects that preclude its clinical use for brain injury. We found that long-term postinjury treatment with a nonerythropoietic derivative of EPO, asialo-erythropoietin, promoted the maturation of pSVZ-derived OPCs and the recovery of neurological function, without affecting hematopoiesis. These results demonstrate the limitation and potential of endogenous OPCs in the pSVZ as a therapeutic target for treating neonatal white-matter injury.


Asunto(s)
Asialoglicoproteínas/uso terapéutico , Ventrículos Cerebrales/efectos de los fármacos , Eritropoyetina/análogos & derivados , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Oligodendroglía/efectos de los fármacos , Células Madre/efectos de los fármacos , Animales , Animales Recién Nacidos , Asialoglicoproteínas/farmacología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ventrículos Cerebrales/lesiones , Ventrículos Cerebrales/metabolismo , Eritropoyetina/farmacología , Eritropoyetina/uso terapéutico , Expresión Génica/efectos de los fármacos , Humanos , Hipoxia-Isquemia Encefálica/rehabilitación , Ratones , Ratones Endogámicos ICR , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Factor de Transcripción 2 de los Oligodendrocitos , Oligodendroglía/metabolismo , Oligodendroglía/patología , Células Madre/metabolismo , Células Madre/patología
4.
J Control Release ; 160(1): 81-7, 2012 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-22342472

RESUMEN

Cerebral ischemia-reperfusion (I/R) injury induces secondary cerebral damage. As drugs for treating this type of injury have shown poor efficacy and adverse side effects in clinical trials, a novel therapeutic strategy has been long awaited. In this study, we focused on the disruption of the blood-brain barrier after stroke, and applied a liposomal drug delivery system (DDS) designed to enhance the pharmacological effect of the neuroprotectant and to avoid side effects. PEGylated liposomes were injected at varying time after the start of reperfusion in transient middle cerebral artery occlusion (t-MCAO) model rats. The results showed PEGylated liposomes accumulated in the ischemic hemisphere at an early stage after reperfusion and were retained in the lesion for at least 24h after injection. We also investigated the effectiveness of asialo-erythropoietin (AEPO)-modified PEGylated liposomes (AEPO-liposomes) in treating the cerebral I/R injury. AEPO-liposome treatment significantly reduced TTC-defined cerebral legion following cerebral I/R injury, and ameliorated motor function compared with vehicle and AEPO treatment. In conclusion, these results indicate that AEPO-liposomes are a promising liposomal formulation for protecting the brain from I/R injury, and that this liposomal DDS has potential as a novel strategy for the treatment of cerebral I/R injury.


Asunto(s)
Asialoglicoproteínas/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Eritropoyetina/análogos & derivados , Ataque Isquémico Transitorio/tratamiento farmacológico , Fármacos Neuroprotectores/administración & dosificación , Daño por Reperfusión/prevención & control , Animales , Apoptosis/efectos de los fármacos , Asialoglicoproteínas/efectos adversos , Asialoglicoproteínas/farmacocinética , Asialoglicoproteínas/uso terapéutico , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Técnicas de Cultivo de Célula , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Eritropoyetina/administración & dosificación , Eritropoyetina/efectos adversos , Eritropoyetina/farmacocinética , Eritropoyetina/uso terapéutico , Etiquetado Corte-Fin in Situ , Ataque Isquémico Transitorio/complicaciones , Ataque Isquémico Transitorio/metabolismo , Liposomas , Masculino , Fármacos Neuroprotectores/efectos adversos , Fármacos Neuroprotectores/farmacocinética , Fármacos Neuroprotectores/uso terapéutico , Células PC12 , Ratas , Ratas Wistar , Daño por Reperfusión/etiología , Daño por Reperfusión/metabolismo , Distribución Tisular
5.
Circ Heart Fail ; 5(2): 274-85, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22343033

RESUMEN

BACKGROUND: We investigated the effects of asialoerythropoietin (asialoEPO), a nonerythrogenic erythropoietin derivative, on 3 murine models of heart failure with different etiologies. METHODS AND RESULTS: Doxorubicin (15 mg/kg) induced heart failure within 2 weeks (toxic cardiomyopathy). Treatment with asialoEPO (6.9 µg/kg) for 2 weeks thereafter attenuated the associated left ventricular dysfunction and dilatation. In addition, the asialoEPO-treated heart showed less myocardial fibrosis, inflammation, and oxidative damage, and diminished atrophic cardiomyocyte degeneration, which was accompanied by restored expression of GATA-4 and sarcomeric proteins. Mice with large 6-week-old myocardial infarctions exhibited marked left ventricular dysfunction with adverse remodeling (ischemic cardiomyopathy). AsialoEPO treatment for 4 weeks significantly mitigated progression of the dysfunction and remodeling and reduced myocardial fibrosis, inflammation, and oxidative damage. Finally, 25-week-old δ-sarcoglycan-deficient mice (genetic cardiomyopathy) were treated with asialoEPO for 5 weeks. AsialoEPO mitigated the progressive cardiac remodeling and dysfunction through cardiomyocyte hypertrophy, and upregulated expression of GATA-4 and sarcomeric proteins. AsialoEPO appears to act by altering the activity of the downstream erythropoietin receptor signals extracellular signal-regulated protein kinase, Akt, signal transducer, and activator of transcription 3 and 5 in a model-specific manner. CONCLUSIONS: The findings suggest that asialoEPO exerts broad cardioprotective effects through distinct mechanisms depending on the model, which are independent of the erythrogenic action. This compound may be promising for the treatment of heart failure of various etiologies.


Asunto(s)
Asialoglicoproteínas/uso terapéutico , Eritropoyetina/análogos & derivados , Insuficiencia Cardíaca/tratamiento farmacológico , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos , Animales , Asialoglicoproteínas/administración & dosificación , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Eritropoyetina/administración & dosificación , Eritropoyetina/uso terapéutico , Estudios de Seguimiento , Insuficiencia Cardíaca/fisiopatología , Ratones , Ratones Endogámicos C57BL , Resultado del Tratamiento , Función Ventricular Izquierda/fisiología
7.
J Am Coll Cardiol ; 56(23): 1949-58, 2010 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-21109120

RESUMEN

OBJECTIVES: We examined the effect of asialoerythropoietin (asialoEPO), a nonerythrogenic derivative of erythropoietin (EPO), on renal dysfunction-associated heart failure. BACKGROUND: Although EPO is known to exert beneficial effects on cardiac function, the clinical benefits in patients with chronic kidney disease are controversial. It remains to be addressed whether previously reported outcomes were the result of relief of the anemia, adverse effects of EPO, or direct cardiovascular effects. METHODS: Mice underwent 5/6 nephrectomy to cause renal dysfunction. Eight weeks later, when renal dysfunction was established, anemia and cardiac dysfunction and remodeling were apparent. Mice were then assigned to receive saline (control), recombinant human erythropoietin (rhEPO) at 5,000 IU (714 pmol)/kg, or asialoEPO at 714 pmol/kg, twice/week for 4 weeks. RESULTS: Although only rhEPO relieved the nephrectomy-induced anemia, both rhEPO and asialoEPO significantly and similarly mitigated left ventricular dilation and dysfunction. The hearts of rhEPO- or asialoEPO-treated mice showed less hypertrophy, reflecting decreases in cardiomyocyte hypertrophy and degenerative subcellular changes, as well as significant attenuation of fibrosis, leukocyte infiltration, and oxidative deoxyribonucleic acid damage. These phenotypes were accompanied by restored expression of GATA-4, sarcomeric proteins, and vascular endothelial growth factor and decreased inflammatory cytokines and lipid peroxidation. Finally, myocardial activation was observed of extracellular signal-regulated protein kinase and signal transducer and activator of transcription pathways in the treated mice. CONCLUSIONS: EPO receptor signaling exerts direct cardioprotection in an animal model of renal dysfunction-associated heart failure, probably by mitigating degenerative, pro-fibrosis, inflammatory, and oxidative processes but not through relief of anemia.


Asunto(s)
Anemia/complicaciones , Asialoglicoproteínas/uso terapéutico , Eritropoyetina/análogos & derivados , Eritropoyetina/uso terapéutico , Insuficiencia Cardíaca/etiología , Receptores de Eritropoyetina/metabolismo , Insuficiencia Renal/complicaciones , Transducción de Señal , Anemia/tratamiento farmacológico , Anemia/metabolismo , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo , Proteínas Recombinantes , Insuficiencia Renal/tratamiento farmacológico , Insuficiencia Renal/metabolismo
8.
Nephrology (Carlton) ; 15(1): 93-101, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20377776

RESUMEN

AIM: Diabetic patients are at higher risk of failure to recover after acute kidney injury, however, the mechanism and therapeutic strategies remain unclear. Erythropoietin is cytoprotective in a variety of non-haematopoietic cells. The aim of the present study was to clarify the mechanism of diabetes-related acceleration of renal damage after ischaemia-reperfusion injury and to examine the therapeutic potential of asialoerythropoietin, a non-haematopoietic erythropoietin derivative, against ischaemia-reperfusion-induced acute kidney injury in diabetic mice. METHODS: C57BL/6J mice with and without streptozotocin-induced diabetes were subjected to 30 min unilateral renal ischaemia-reperfusion injury at 1 week after induction of diabetes. They were divided into four group: (i) non-diabetic plus ischaemia-reperfusion injury; (ii) non-diabetic plus ischaemia-reperfusion injury plus asialoerythropoietin (3000 IU/kg bodyweight); (iii) diabetic plus ischaemia-reperfusion injury; and (iv) diabetic plus ischemia-reperfusion injury plus asialoerythropoietin. Experiments were conducted at the indicated time periods after ischaemia-reperfusion injury. RESULTS: Ischaemia-reperfusion injury of diabetic kidney resulted in significantly low protein expression levels of bcl-2, an anti-apoptotic molecule, and bone morphogenetic protein-7 (BMP-7), an anti-fibrotic and pro-regenerative factor, compared with non-diabetic kidneys. Diabetic kidney subsequently showed severe damage including increased tubular cell apoptosis, tubulointerstitial fibrosis and decreased tubular proliferation, compared with non-diabetic kidney. Treatment with asialoerythropoietin induced bcl-2 and BMP-7 expression in diabetic kidney and decreased tubular cell apoptosis, tubulointerstitial fibrosis and accelerated tubular proliferation. CONCLUSION: Reduced induction bcl-2 and BMP-7 may play a role in the acceleration of renal damage after ischaemia-reperfusion injury in diabetic kidney. The renoprotective effects of asialoerythropoietin on acute kidney injury may be mediated through the induction of bcl-2 and BMP-7.


Asunto(s)
Lesión Renal Aguda/prevención & control , Asialoglicoproteínas/uso terapéutico , Eritropoyetina/análogos & derivados , Lesión Renal Aguda/etiología , Animales , Diabetes Mellitus Experimental/complicaciones , Eritropoyetina/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Daño por Reperfusión/complicaciones
9.
Surgery ; 143(4): 556-65, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18374054

RESUMEN

OBJECTIVE: To investigate the protective effect of erythropoietin (EPO) and its nonhematopoietic derivative (asialoEPO) against intestinal ischemia/reperfusion (I/R) injury in a rat model. METHODS: The superior mesenteric artery of Wistar rats was clamped for 60 minutes and then released. The rats were divided into 4 groups (n = 15 in each group): sham operation (Sham), vehicle treatment (Vehicle), EPO treatment (EPO), and asialoEPO treatment (AsialoEPO). EPO and asialoEPO were administered subcutaneously at 1000 units/kg for 10 minutes before clamping, 30 minutes after the start of clamping, and just before declamping. This treatment was followed by determination of 72-hour survival rates, serum TNF-alpha and IL-6 levels, histologic evaluation of the small intestine, quantification of the number of apoptotic cells, and analysis of the antiapoptotic molecules Bcl-xL and XIAP by Western blotting. RESULTS: The survival rates at 72 hours after I/R injury in the Sham, Vehicle, EPO, and AsialoEPO groups were 100%, 33%, 75%, and 83%, respectively (P < .05). Blood TNF-alpha and IL-6 were significantly more suppressed in the EPO and AsialoEPO groups than in the Vehicle group at 6 hours after I/R injury. Histologically, injury to villi in the EPO and AsialoEPO groups was significantly less than in the Vehicle group. The number of apoptotic cells in the EPO and AsialoEPO groups was significantly less than in the Vehicle group. Western blotting revealed that EPO and asialoEPO constitutively increased the expression of Bcl-xL. CONCLUSIONS: EPO and asialoEPO exert a strong protective effect against intestinal I/R injury, possibly by inhibiting release of TNF-alpha and IL-6 and decreasing apoptosis.


Asunto(s)
Asialoglicoproteínas/farmacología , Eritropoyetina/análogos & derivados , Eritropoyetina/farmacología , Fármacos Gastrointestinales/farmacología , Enfermedades Intestinales/prevención & control , Intestinos/efectos de los fármacos , Daño por Reperfusión/prevención & control , Animales , Apoptosis/efectos de los fármacos , Asialoglicoproteínas/uso terapéutico , Modelos Animales de Enfermedad , Eritropoyetina/uso terapéutico , Fármacos Gastrointestinales/uso terapéutico , Intestinos/irrigación sanguínea , Masculino , Ratas , Ratas Wistar , Análisis de Supervivencia
10.
J Biomed Sci ; 15(2): 205-13, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18080217

RESUMEN

Ribavirin is a synthetic nucleoside analog that is used for the treatment of hepatitis C virus (HCV) infection. Its primary toxicity is hemolytic anemia, which sometimes necessitates dose reduction or discontinuation of therapy. Selective delivery of ribavirin into liver cells would be desirable to enhance its antiviral activity and avoid systemic side effects. One approach to liver-specific targeting is conjugation of the ribavirin with asialoglycoprotein that is taken up specifically by liver cells. Human uridine-cytidine kinase-1 (UCK-1) was used for ribavirin phosphorylation to its monophosphate form. 1-Ethyl-3-diisopropylaminocarbodiimide (EDC) was used as a coupling agent. The best results were obtained using direct conjugation protocol with a molar ratio of 6.5 ribavirin monophosphate (RMP) molecules per one asialoorosomucoid (AsOR) molecule. Our findings show that ribavirin is a potential substrate of UCK-1, and RMP formed could be chemically coupled to AsOR to form a conjugate for liver specific targeting.


Asunto(s)
Asialoglicoproteínas/química , Sistemas de Liberación de Medicamentos , Hepatitis C/enzimología , Hígado/enzimología , Nucleósido-Fosfato Quinasa/química , Orosomucoide/análogos & derivados , Ribavirina/química , Anemia Hemolítica/inducido químicamente , Anemia Hemolítica/enzimología , Asialoglicoproteínas/uso terapéutico , Hepacivirus , Hepatitis C/complicaciones , Humanos , Hígado/virología , Nucleósido-Fosfato Quinasa/metabolismo , Orosomucoide/química , Orosomucoide/uso terapéutico , Fosforilación , Ribavirina/efectos adversos , Ribavirina/uso terapéutico
11.
Transplantation ; 84(4): 504-10, 2007 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-17713435

RESUMEN

BACKGROUND: The renoprotective effect of erythropoietin (EPO) and the nonhematopoietic EPO, asialoEPO was investigated in a murine ischemia-reperfusion injury (I/R) model. METHODS: I/R was created by clamping the right renal pedicle for 60 min after left nephrectomy. Balb/c mice were divided into four groups (n=15 in each group): sham operation (Sham), vehicle treatment (Vehicle), EPO treatment (EPO), and asialoEPO treatment (AsialoEPO). EPO and asialoEPO were given at a dose of 500 IU/kg 30 min before I/R. Plasma creatinine (Cr), survival, and the number of apoptotic cells were analyzed. Protein expression was analyzed by Western blotting. RESULTS: Plasma Cr level was not significantly different at 6 hr after I/R. At 24 hr after I/R, the Cr (mg/dL) levels in Sham, Vehicle, EPO, and asialoEPO were 0.13+/-0.01, 1.24+/-0.70, 0.24+/-0.08, and 0.25+/-0.13, respectively (P<0.05). The numbers of apoptotic cells in these groups were 0.1+/-0.1, 98.9+/-42.6, 3.3+/-0.7, and 2.9+/-1.6, respectively (P<0.05). Western blotting revealed that in kidney tissue of mice treated with EPO and asialoEPO, p38-MAPK and the proapoptotic molecule Bad was decreased, and the antiapoptotic molecules Bcl-xL and XIAP were increased. Survival rates at 7 days after I/R injury in the Sham, Vehicle, EPO, and AsialoEPO groups were 100%, 21.4%, 23.1%, and 53.8%, respectively (P=0.05). CONCLUSION: EPO and asialoEPO attenuated renal dysfunction caused by I/R in mouse kidney at the same level, but only asialoEPO improved survival.


Asunto(s)
Asialoglicoproteínas/uso terapéutico , Eritropoyetina/análogos & derivados , Riñón/fisiopatología , Daño por Reperfusión/prevención & control , Animales , Apoptosis/efectos de los fármacos , Creatinina/sangre , Modelos Animales de Enfermedad , Eritropoyetina/uso terapéutico , Femenino , Riñón/metabolismo , Riñón/patología , Ratones , Ratones Endogámicos BALB C , Distribución Aleatoria , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Tasa de Supervivencia , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Proteína Letal Asociada a bcl/antagonistas & inhibidores , Proteína bcl-X/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
12.
J Neurochem ; 91(4): 900-10, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15525344

RESUMEN

Recently, erythropoietin (EPO) and the nonerythropoietic derivative asialoEPO have been linked to tissue protection in the nervous system. In this study, we tested their effects in a model of neonatal hypoxia-ischemia (HI) in 7-day-old rats (unilateral carotid ligation and exposure to 7.7% O(2) for 50 min). EPO (10 U/g body weight = 80 ng/g; n = 24), asialoEPO (80 ng/g; n = 23) or vehicle (phosphate-buffered saline with 0.1% human serum albumin; n = 24) was injected intraperitoneally 4 h before HI. Both drugs were protective, as judged by measuring the infarct volumes, neuropathological score and gross morphological score. The infarct volumes were significantly reduced by both EPO (52%) and asialoEPO (55%) treatment, even though the plasma levels of asialoEPO had dropped below the detection limit (1 pm) at the onset of HI, while those of EPO were in the nanomolar range. Thus, a brief trigger by asialoEPO before the insult appears to be sufficient for protection. Proteomics analysis after asialoEPO treatment alone (no HI) revealed at least one differentially up-regulated protein, synaptosome-associated protein of 25 kDa (SNAP-25). Activation (phosphorylation) of ERK was significantly reduced in asialoEPO-treated animals after HI. EPO and the nonerythropoietic asialoEPO both provided significant and equal neuroprotection when administered 4 h prior to HI in 7-day-old rats. The protection might be related to reduced ERK activation and up-regulation of SNAP-25.


Asunto(s)
Asialoglicoproteínas/uso terapéutico , Eritropoyetina/análogos & derivados , Eritropoyetina/uso terapéutico , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Animales , Animales Recién Nacidos , Asialoglicoproteínas/biosíntesis , Asialoglicoproteínas/sangre , Asialoglicoproteínas/farmacocinética , Infarto Encefálico/patología , Infarto Encefálico/prevención & control , Modelos Animales de Enfermedad , Eritropoyetina/biosíntesis , Eritropoyetina/sangre , Eritropoyetina/farmacocinética , Femenino , Hipoxia-Isquemia Encefálica/patología , Masculino , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteómica/métodos , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Proteína 25 Asociada a Sinaptosomas , Resultado del Tratamiento
13.
BMC Neurosci ; 5: 17, 2004 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-15134587

RESUMEN

BACKGROUND: Huntington's disease (HD) is a hereditary neurodegenerative disorder caused by an expanded CAG repeat in the HD gene. Both excitotoxicity and oxidative stress have been proposed to play important roles in the pathogenesis of HD. Since no effective treatment is available, this study was designed to explore the therapeutic potential of erythropoietin (EPO), a cytokine that has been found to prevent excitotoxicity, and to promote neurogenesis. To avoid the side effects of a raised hematocrit, we used asialoerythropoietin (asialoEPO), a neuroprotective variant of EPO that lacks erythropoietic effects in mice. R6/2 transgenic HD mice were treated with this cytokine from five to twelve weeks of age. RESULTS: We provide new evidence that cell proliferation in the dentate gyrus of the R6/2 hippocampus is reduced by 50% compared to wild-type littermate controls. However, we found that the asialoEPO treatment did not affect the progression of motor symptoms, weight loss or the neuropathological changes. Furthermore, cell proliferation was not enhanced. CONCLUSIONS: We conclude that the chosen protocol of asialoEPO treatment is ineffective in the R6/2 model of HD. We suggest that reduced hippocampal cell proliferation may be an important and novel neuropathological feature in R6 HD mice that could be assessed when evaluating potential therapies.


Asunto(s)
Asialoglicoproteínas/uso terapéutico , Eritropoyetina/uso terapéutico , Enfermedad de Huntington/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Animales , Conducta Animal/efectos de los fármacos , División Celular/efectos de los fármacos , División Celular/genética , Citocinas/uso terapéutico , Giro Dentado/efectos de los fármacos , Giro Dentado/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Eritropoyetina/análogos & derivados , Femenino , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Masculino , Ratones , Ratones Transgénicos , Fenotipo , Insuficiencia del Tratamiento , Pérdida de Peso/efectos de los fármacos , Pérdida de Peso/genética
14.
Nat Med ; 5(5): 577-81, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10229237

RESUMEN

Most chronic carriers of hepatitis B virus (HBV) do not respond to interferon (IFN) treatment. This limitation of IFN therapy may be due in part to scant expression of IFN receptor in the liver. Because the asialoglycoprotein (ASGP) receptor is specifically expressed in the liver at high density, the ASGP receptor-binding domain was generated within an N-glycosylated human IFN-beta molecule by the removal of sialic acid to direct this cytokine to the liver. This modified IFN (asialo-IFN-beta) demonstrated greater inhibition of HBV production in ASGP receptor-positive human liver cells transfected with a replication-competent HBV construct than did conventional IFN-alpha or IFN-beta. Furthermore, the enhanced antiviral effect of asialo-IFN-beta was supported by induction of the 2'-5' oligoadenylate synthetase, an indicator of IFN activity, at a level significantly higher than that produced by conventional IFN-beta. Moreover, mouse asialo-IFN-beta profoundly reduced viremia in vivo in HBV-transfected athymic nude mice, in contrast to conventional IFN-beta, which had no substantial effect. These experiments demonstrate that directing IFN to ASGP receptor facilitates its signaling in the liver and augments its antiviral effect, and is therefore useful in overcoming the limited antiviral effect of conventional IFNs.


Asunto(s)
Antivirales/uso terapéutico , Asialoglicoproteínas/uso terapéutico , Hepatitis B/tratamiento farmacológico , Interferón beta/uso terapéutico , Hígado/virología , Receptores de Superficie Celular/metabolismo , 2',5'-Oligoadenilato Sintetasa/biosíntesis , Animales , Receptor de Asialoglicoproteína , Diseño de Fármacos , Inducción Enzimática , Hepatitis B/virología , Virus de la Hepatitis B/efectos de los fármacos , Humanos , Hígado/citología , Hígado/efectos de los fármacos , Ratones , Ratones Desnudos , Receptores de Interferón/metabolismo , Replicación Viral/efectos de los fármacos
15.
J Biol Chem ; 273(9): 5033-6, 1998 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-9478952

RESUMEN

Cholesteryl ester transfer protein (CETP) is the enzyme that facilitates the transfer of cholesteryl ester from high density lipoprotein (HDL) to apolipoprotein B (apoB)-containing lipoproteins. However, the exact role of CETP in the development of atherosclerosis has not been determined. In the present study, we examined the effect of the suppression of increased plasma CETP by intravenous injection with antisense oligodeoxynucleotides (ODNs) against CETP targeted to the liver on the development of atherosclerosis in rabbits fed a cholesterol diet. The ODNs against rabbit CETP were coupled to asialoglycoprotein (ASOR) carrier molecules, which serve as an important method to regulate liver gene expression. Twenty-two male Japanese White rabbits were used in the experiment. Eighteen animals were fed a standard rabbit chow supplemented with 0.3% cholesterol throughout the experiment for 16 weeks. At 8 weeks, they were divided into three groups (six animals in each group), among which the plasma total and HDL cholesterol concentrations did not significantly change. The control group received nothing, the sense group were injected with the sense ODNs complex, and the antisense group were injected with the antisense ODNs complex, respectively, for subsequent 8 weeks. ASOR. poly(L-lysine) ODNs complex were injected via the ear veins twice a week. Four animals were fed a standard rabbit diet for 16 weeks. The total cholesterol concentrations and the CETP mass in the animals injected with antisense ODNs were all significantly decreased in 12 and 16 weeks compared with those injected with sense ODNs and the control animals. The HDL cholesterol concentrations measured by the precipitation assay did not significantly change among the groups fed a cholesterol diet, and triglyceride concentrations did not significantly change in the four groups. However, at the end of the study, when the HDL cholesterol concentrations were measured after the isolation by ultracentrifugation and a column chromotography, they were significantly higher in the animals injected with antisense ODNs than in the animals injected with sense ODNs and in the control animals. A reduction of CETP mRNA and an increase of LDL receptor mRNA in the liver were observed in the animals injected with antisense ODNs compared with those injected with sense ODNs and the control animals. Aortic cholesterol contents and the aortic percentage lesion to total surface area were significantly lower in the animals injected with antisense ODNs than in the animals injected with sense ODNs and in the control animals. These findings showed for the first time that suppression of increased plasma CETP by the injection with antisense ODNs against CETP coupled to ASOR carrier molecules targeted to the liver could thus inhibit the atherosclerosis possibly by decreasing the plasma LDL + very low density lipoprotein (VLDL) cholesterol in cholesterol-fed rabbits.


Asunto(s)
Arteriosclerosis/prevención & control , Proteínas Portadoras/efectos de los fármacos , Colesterol en la Dieta/efectos adversos , Glicoproteínas , Oligonucleótidos Antisentido/uso terapéutico , Animales , Aorta/química , Aorta/patología , Arteriosclerosis/inducido químicamente , Arteriosclerosis/genética , Arteriosclerosis/patología , Asialoglicoproteínas/uso terapéutico , Proteínas Portadoras/sangre , Proteínas Portadoras/genética , Proteínas de Transferencia de Ésteres de Colesterol , LDL-Colesterol/sangre , VLDL-Colesterol/sangre , Portadores de Fármacos/uso terapéutico , Hígado/química , Hígado/efectos de los fármacos , Masculino , ARN Mensajero/análisis , Conejos , Triglicéridos/sangre
16.
Blood Coagul Fibrinolysis ; 8 Suppl 2: S31-8, 1997 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9607111

RESUMEN

The development of non-viral gene therapy has been hampered by an inability to reproducibly manufacture and characterize delivery system components and final formulations. Formation of interpolyelectrolyte complexes as the basis of various gene delivery methods has been approached as the first step towards development of synthetic viruses. We have found that preparation of interpolyelectrolyte complexes from disperse reagents gives a more homogeneous gene delivery vehicle than other methods. Methods which increase homogeneity also result in higher transfection efficiency in vivo. Expression levels of human growth hormone and other reporter proteins in mice confirm the potential of parenteral non-viral gene delivery for some therapeutic applications. Serum is demonstrated to inhibit transfection efficiency in vivo. Our results suggest that further development of methods to manufacture homogeneous disperse non-viral delivery vehicles with stealth characteristics may enhance both the potency and reproducibility of gene transfer in vivo.


Asunto(s)
Técnicas de Transferencia de Gen , Terapia Genética/métodos , Animales , Asialoglicoproteínas/administración & dosificación , Asialoglicoproteínas/uso terapéutico , Centrifugación por Gradiente de Densidad , Regulación de la Expresión Génica , Hormona de Crecimiento Humana/biosíntesis , Hormona de Crecimiento Humana/genética , Inyecciones Intravenosas , Ligandos , Luciferasas/genética , Ratones , Ratones Endogámicos BALB C , Especificidad de Órganos/genética , Orosomucoide/administración & dosificación , Orosomucoide/análogos & derivados , Orosomucoide/uso terapéutico , Polilisina/administración & dosificación , Polilisina/análogos & derivados , Polilisina/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA