Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Clin Cancer Res ; 14(12): 4002-9, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18559623

RESUMEN

PURPOSE: The blood-brain tumor barrier (BTB) significantly limits the delivery of chemotherapeutics to brain tumors. Nitric oxide (NO) is involved in the regulation of cerebral vascular permeability. We investigated the effects of NO donors, L-arginine and hydroxyurea, on BTB permeability in 9L gliosarcoma-bearing Fischer rats. EXPERIMENTAL DESIGN: The rats implanted with 9L gliosarcoma were dosed orally with hydroxyurea and L-arginine. BTB permeability, defined by the unidirectional transport constant, Ki, for [14C]sucrose was measured. The expression of neural and endothelial NO synthase (NOS) in tumors and normal brain tissue was examined. Further, the levels of NO, L-citrulline, and cGMP in the tumor and normal brain tissue were measured. RESULTS: Oral administration of l-arginine or hydroxyurea significantly increased BTB permeability when compared with the nontreated control. The selective effects were abolished by iberiotoxin, an antagonist of calcium-dependent potassium (KCa) channel that is a cGMP pathway effector. The expression of endothelial NOS, but not neural NOS, was higher in tumor vessels than in those of normal brain. Moreover, the levels of NO, L-citrulline, a byproduct of NO formation from L-arginine, and cGMP were enhanced in the tumor tissue by oral administration of L-arginine and/or hydroxyurea. CONCLUSIONS: Oral administration of L-arginine or hydroxyurea selectively increased tumor permeability, which is likely mediated by alteration in cGMP levels. The findings suggest that use of oral NO donors may be a strategy to enhance the delivery of chemotherapeutics to malignant brain tumors.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Neoplasias Encefálicas/patología , Permeabilidad Capilar/efectos de los fármacos , Glioma/patología , Donantes de Óxido Nítrico/farmacología , Administración Oral , Animales , Arginina/administración & dosificación , Arginina/farmacología , Barrera Hematoencefálica/fisiología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/fisiopatología , Permeabilidad Capilar/fisiología , Citrulina/metabolismo , GMP Cíclico/metabolismo , Sistemas de Liberación de Medicamentos , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Femenino , Glioma/metabolismo , Glioma/fisiopatología , Hidroxiurea/administración & dosificación , Hidroxiurea/farmacología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , Ratas , Ratas Endogámicas F344 , Células Tumorales Cultivadas
2.
Brain Res ; 1227: 198-206, 2008 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-18602898

RESUMEN

The blood-brain tumor barrier (BTB) significantly limits delivery of effective concentrations of chemotherapeutic drugs to brain tumors. Previous studies suggest that BTB permeability may be modulated via alteration in the activity of potassium channels. In this study, we studied the relationship of BTB permeability increase mediated by potassium channel agonists to channel expression in two rat brain tumor models. Intravenous infusion of KCO912 (K(ATP) agonist), minoxidil sulfate (K(ATP) agonist) or NS1619 (K(Ca) agonist) increased tumor permeability more in the 9L allogeneic brain tumor model than in the syngeneic brain tumor model. Consistently, expression of both K(ATP) and K(Ca) channels in 9L tumors was increased to a significantly greater extent in Wistar rats (allogeneic) as compared to Fischer rats (syngeneic). Furthermore, as a preliminary effort to understand clinical implication of potassium channels in brain tumor treatment, we determined the expression of K(ATP) in surgical specimens. K(ATP) mRNA was detected in glioblastoma multiforme (GBM) from nineteen patients examined, with a wide range of expression levels. Interestingly, in paired GBM tissues from seven patients before and after vaccination therapy, increased levels of K(ATP) were detected in five patients after vaccination that had positive response to chemotherapy after vaccination. The present study indicates that the effects of potassium channel agonists on BTB permeability are different between syngeneic and allogeneic models which have different expression levels of potassium channels. The expression of potassium channels in brain tumors is variable, which may be associated with different tumor permeability to therapeutic agents among patients.


Asunto(s)
Neoplasias Encefálicas/fisiopatología , Permeabilidad Capilar/efectos de los fármacos , Canales KATP/agonistas , Canales de Potasio Calcio-Activados/agonistas , Animales , Bencimidazoles/administración & dosificación , Bencimidazoles/uso terapéutico , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/fisiopatología , Western Blotting , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Línea Celular Tumoral , Femenino , Glioblastoma/patología , Glioblastoma/fisiopatología , Glioblastoma/terapia , Humanos , Inmunohistoquímica , Inyecciones Intravenosas , Canales KATP/genética , Canales KATP/fisiología , Microscopía Confocal , Minoxidil/administración & dosificación , Minoxidil/análogos & derivados , Minoxidil/uso terapéutico , Trasplante de Neoplasias , Neoplasias Experimentales/patología , Neoplasias Experimentales/fisiopatología , Neoplasias Experimentales/terapia , Canales de Potasio Calcio-Activados/genética , Canales de Potasio Calcio-Activados/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas F344 , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Especificidad de la Especie , Vacunación/métodos
3.
Brain Res ; 1230: 290-302, 2008 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-18674521

RESUMEN

The blood-brain tumor barrier (BTB) significantly limits delivery of therapeutic concentrations of chemotherapy to brain tumors. A novel approach to selectively increase drug delivery is pharmacologic modulation of signaling molecules that regulate BTB permeability, such as those in cGMP signaling. Here we show that oral administration of sildenafil (Viagra) and vardenafil (Levitra), inhibitors of cGMP-specific PDE5, selectively increased tumor capillary permeability in 9L gliosarcoma-bearing rats with no significant increase in normal brain capillaries. Tumor-bearing rats treated with the chemotherapy agent, adriamycin, in combination with vardenafil survived significantly longer than rats treated with adriamycin alone. The selective increase in tumor capillary permeability appears to be mediated by a selective increase in tumor cGMP levels and increased vesicular transport through tumor capillaries, and could be attenuated by iberiotoxin, a selective inhibitor for calcium-dependent potassium (K(Ca)) channels, that are effectors in cGMP signaling. The effect by sildenafil could be further increased by simultaneously using another BTB "opener", bradykinin. Collectively, this data demonstrates that oral administration of PDE5 inhibitors selectively increases BTB permeability and enhances anti-tumor efficacy for a chemotherapeutic agent. These findings have significant implications for improving delivery of anti-tumor agents to brain tumors.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Glioma/tratamiento farmacológico , Glioma/metabolismo , Inhibidores de Fosfodiesterasa 5 , Inhibidores de Fosfodiesterasa/farmacología , Animales , Autorradiografía , Presión Sanguínea/efectos de los fármacos , Química Encefálica/efectos de los fármacos , Neoplasias Encefálicas/patología , Capilares/patología , Permeabilidad Capilar/efectos de los fármacos , GMP Cíclico/sangre , GMP Cíclico/metabolismo , Femenino , Glioma/patología , Imidazoles/farmacología , Microscopía Electrónica de Transmisión , Neovascularización Patológica/patología , Piperazinas/farmacología , Purinas/farmacología , Ratas , Ratas Endogámicas F344 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Citrato de Sildenafil , Sacarosa/metabolismo , Sulfonas/farmacología , Análisis de Supervivencia , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/ultraestructura , Triazinas/farmacología , Diclorhidrato de Vardenafil
4.
Clin Cancer Res ; 13(3): 1045-52, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17289901

RESUMEN

PURPOSE: Histone acetylation is one of the main mechanisms involved in regulation of gene expression. During carcinogenesis, tumor-suppressor genes can be silenced by aberrant histone deacetylation. This epigenetic modification has become an important target for tumor therapy. The histone deacetylation inhibitor, suberoylanilide hydroxamic acid (SAHA), can induce growth arrest in transformed cells. The aim of this study is to examine the effects of SAHA on gene expression and growth of glioblastoma multiforme (GBM) cells in vitro and in vivo. EXPERIMENTAL DESIGN: The effect of SAHA on growth of GBM cell lines and explants was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide. Changes of the cell cycle and relative gene expression were detected by fluorescence-activated cell sorting, real-time reverse transcription-PCR, and Western blotting. After glioma cells were implanted in the brains of mice, the ability of SAHA to decrease tumor growth was studied. RESULTS: Proliferation of GBM cell lines and explants were inhibited in vitro by SAHA (ED50, 2x10(-6) to 2x10(-5) mol/L, 5 days). SAHA exposure of human U87 and T98G glioma cell lines, DA66 and JM94 GBM explants, as well as a murine GL26 GBM cell line resulted in an increased accumulation of cells in G2-M of the cell cycle. Many proapoptotic, antiproliferative genes increased in their expression (DR5, TNFalpha, p21WAF1, p27KIP1), and many antiapoptotic, progrowth genes decreased in their levels (CDK2, CDK4, cyclin D1, cyclin D2) as measured by real-time reverse transcription-PCR and/or Western blot after these GBM cells were cultured with SAHA (2.5x10(-6) mol/L, 1 day). Chromatin immunoprecipitation assay found that acetylation of histone 3 on the p21(WAF1) promoter was markedly increased by SAHA. In vivo murine experiments suggested that SAHA (10 mg/kg, i.v., or 100 mg/kg, i.p.) could cross the blood-brain barrier as shown by prominent increased levels of acetyl-H3 and acetyl-H4 in the brain tissue. Furthermore, the drug significantly (P<0.05) inhibited the proliferation of the GL26 glioma cells growing in the brains of mice and increased their survival. CONCLUSIONS: Taken together, SAHA can slow the growth of GBM in vitro and intracranially in vivo. SAHA may be a welcome addition for the treatment of this devastating disease.


Asunto(s)
Antineoplásicos/farmacología , Regulación Neoplásica de la Expresión Génica , Glioma/tratamiento farmacológico , Inhibidores de Histona Desacetilasas , Ácidos Hidroxámicos/farmacología , Animales , Línea Celular Tumoral , Proliferación Celular , Citometría de Flujo , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Trasplante de Neoplasias , Vorinostat
5.
Mol Cancer ; 6: 22, 2007 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-17359538

RESUMEN

BACKGROUND: The blood-brain tumor barrier (BTB) impedes the delivery of therapeutic agents to brain tumors. While adequate delivery of drugs occurs in systemic tumors, the BTB limits delivery of anti-tumor agents into brain metastases. RESULTS: In this study, we examined the function and regulation of calcium-activated potassium (KCa) channels in a rat metastatic brain tumor model. We showed that intravenous infusion of NS1619, a KCa channel agonist, and bradykinin selectively enhanced BTB permeability in brain tumors, but not in normal brain. Iberiotoxin, a KCa channel antagonist, significantly attenuated NS1619-induced BTB permeability increase. We found KCa channels and bradykinin type 2 receptors (B2R) expressed in cultured human metastatic brain tumor cells (CRL-5904, non-small cell lung cancer, metastasized to brain), human brain microvessel endothelial cells (HBMEC) and human lung cancer brain metastasis tissues. Potentiometric assays demonstrated the activity of KCa channels in metastatic brain tumor cells and HBMEC. Furthermore, we detected higher expression of KCa channels in the metastatic brain tumor tissue and tumor capillary endothelia as compared to normal brain tissue. Co-culture of metastatic brain tumor cells and brain microvessel endothelial cells showed an upregulation of KCa channels, which may contribute to the overexpression of KCa channels in tumor microvessels and selectivity of BTB opening. CONCLUSION: These findings suggest that KCa channels in metastatic brain tumors may serve as an effective target for biochemical modulation of BTB permeability to enhance selective delivery of chemotherapeutic drugs to metastatic brain tumors.


Asunto(s)
Barrera Hematoencefálica/patología , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/secundario , Canales de Potasio Calcio-Activados/metabolismo , Animales , Neoplasias Encefálicas/metabolismo , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Células Endoteliales/citología , Células Endoteliales/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/patología , Permeabilidad/efectos de los fármacos , Canales de Potasio Calcio-Activados/agonistas , Canales de Potasio Calcio-Activados/antagonistas & inhibidores , Canales de Potasio Calcio-Activados/genética , Potenciometría , Transporte de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Receptor de Bradiquinina B2/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Oncogene ; 24(3): 344-54, 2005 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-15531918

RESUMEN

The proteasome plays a pivotal role in controlling cell proliferation, apoptosis, and differentiation in a variety of normal and tumor cells. PS-341, a novel boronic acid dipeptide that inhibits 26S proteasome activity, has prominent effects in vitro and in vivo against several solid tumors. We examined its antiproliferation, proapoptotic effects using three human glioblastoma multiforme (GBM) cell lines and five primary GBM explants. PS-341 markedly inhibited proliferation of GBM cell lines and explants in liquid and soft agar culture. These cells developed a G2/M cell cycle arrest with a concomitant decreased percentage of cells in S phase ( approximately 2-fold), associated with an increased expression of p21(WAF1), p27(KIP1), as well as cyclin B1 and decreased levels of CDK2, CDK4, and E2F4. About 35-40% of the cells became apoptotic when exposed to PS-341 (10(-7) M, 24-48 h) as shown by Annexin V analysis; in concert with these findings, immunobloting showed a C-terminal 85 kDa apoptotic fragment of poly ADP-ribose polymerase (PARP), and a decreased level of Bcl2 and Bcl-xl. PS-341 downregulated the expression of Bcl-2 and Bcl-xl in protein levels at an early time of treatment. These changes occurred irrespective of the p53 mutational status of the cells. PS-341 activated JNK/c-Jun signaling in GBM cells, and the JNK inhibitor SP600125 blocked the JNK signaling to reverse partially the PS-341 growth inhibition. PS-341 (10(-7) M, 24 h) decreased nuclear NF-kappaB levels as shown by Western blot, and reduced transcriptional activity of NF-kappaB as measured by reporter assays in these transformed cells. Also, PS-341 enhanced TRAIL (TNF-related apoptosis-inducing ligand) and TNFalpha (tumor necrosis factor alpha) induced cell death and apoptosis (two- to five-fold) in GBM cells. In summary, PS-341 has profound effects on growth and apoptosis of GBM cells, suggesting that PS-341 may be an effective therapy for patients with gliomas.


Asunto(s)
Apoptosis/efectos de los fármacos , Ácidos Borónicos/farmacología , División Celular/efectos de los fármacos , Glioblastoma/patología , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasoma , Pirazinas/farmacología , Bortezomib , Línea Celular Tumoral , Humanos , Modelos Biológicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Atherosclerosis ; 181(2): 261-9, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16039279

RESUMEN

Recent evidence from a double-blind, randomized study showed that treatment with apolipoprotein A-I Milano (ApoA-I Milano) in a complex with phospholipids produced significant regression of the coronary atheroma burden in patients with acute coronary syndromes. We previously showed similar regression of atherosclerosis in an animal model. Here, we examined a viral vector-based gene delivery system as a basis for ApoA-I Milano gene therapy. Comparing levels of expression using combinations of the cytomegalovirus (CMV) promoter in a recombinant serotype 2 adeno-associated virus (rAAV2) linked to ApoA-I Milano or the enhanced green fluorescent protein (EGFP) genes, we found that a promoter construct of two CMV core promoters sharing a CMV enhancer was more active than other combinations or a single CMV promoter. In vivo assessment of this optimal CMV construct using rAAV2 virus particles for intravenous (IV) or intramuscular (IM) routes of delivery produced high circulating levels of ApoA-I Milano protein for extended periods (up to 220 ng/ml at 22 weeks p.i.) by IV delivery while the IM route resulted in a relatively short period of very low-level ApoA-I Milano expression. Since there was no difference in the immune response between the two routes of delivery, we reasoned that tissue tropism might be responsible for this differential gene expression. To explore this possibility, we investigated the effect of different AAV serotypes on ApoA-I Milano gene expression in vivo. It found that rAAV1-mediated expression of ApoA-I Milano was approximately 15- and 9-fold higher than rAAV2 and rAAV5, respectively when IM injection routes were compared while all three AAV serotypes produced substantial levels of ApoA-I Milano expression from IV injection. These studies demonstrate that by modifying the promoter and serotype, increases in the efficiency of AAV-directed transgene expression could be achieved and support the potential of AAV-mediated gene therapy.


Asunto(s)
Apolipoproteína A-I/genética , Enfermedad de la Arteria Coronaria/genética , Enfermedad de la Arteria Coronaria/terapia , Terapia Genética/métodos , Adenoviridae/genética , Animales , Línea Celular , Citomegalovirus/genética , Modelos Animales de Enfermedad , Expresión Génica , Proteínas Fluorescentes Verdes/genética , Humanos , Riñón/citología , Ratones , Ratones Mutantes , Regiones Promotoras Genéticas , Transgenes/genética
8.
Curr Eye Res ; 27(1): 15-24, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12868005

RESUMEN

PURPOSE: We investigated the effects of 35 weeks of a cholesterol diet in apolipoprotein E (apoE)-deficient mice on their ERG response. METHODS: C57BL/6J and apoE-deficient mice were fed regular mouse chow (C57-R and ApoE-R, respectively) or a cholesterol-containing diet (C57-C and ApoE-C, respectively). Retinal function was assessed by dark-adapted electroretinography (ERG). Retina tissue was also analyzed by immunohistochemical staining and nucleic acid array expression analysis performed by gene array technology. RESULTS: ApoE-C mice had diminished a- and b-wave amplitudes (60.7% +/- 8.4% (p < 0.005) and 44.8% +/- 10% (p < 0.005) of control values, respectively). Gene expression profiling revealed upregulation of several pro-apoptotic genes. Furthermore, immunohistochemistry showed increased Bax immunoreactivity. CONCLUSIONS: In the hypercholesterolemic mice, we demonstrated a loss of ERG response and induction of apoptotic activity at the gene and protein levels. Our current and previous findings suggest that cholesterol metabolism plays an important role in retinal function.


Asunto(s)
Apolipoproteínas E/deficiencia , Colesterol en la Dieta/farmacología , Electrorretinografía , Proteínas Proto-Oncogénicas c-bcl-2 , Retina/efectos de los fármacos , Retina/fisiopatología , Animales , Apoptosis/genética , Calbindina 2 , Expresión Génica , Perfilación de la Expresión Génica , Hipercolesterolemia/complicaciones , Hipercolesterolemia/genética , Hipercolesterolemia/fisiopatología , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas/metabolismo , Proteína G de Unión al Calcio S100/metabolismo , Regulación hacia Arriba , Proteína X Asociada a bcl-2
9.
Cancer Res ; 66(17): 8887-96, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16951206

RESUMEN

Dendritic cells (DCs) are potent antigen-presenting cells that play a critical role in priming immune responses to tumor. Interleukin (IL)-23 can act directly on DC to promote immunogenic presentation of tumor peptide in vitro. Here, we evaluated the combination of bone marrow-derived DC and IL-23 on the induction of antitumor immunity in a mouse intracranial glioma model. DCs can be transduced by an adenoviral vector coding single-chain mouse IL-23 to express high levels of bioactive IL-23. Intratumoral implantation of IL-23-expressing DCs produced a protective effect on intracranial tumor-bearing mice. The mice consequently gained systemic immunity against the same tumor rechallenge. The protective effect of IL-23-expressing DCs was comparable with or even better than that of IL-12-expressing DCs. IL-23-transduced DC (DC-IL-23) treatment resulted in robust intratumoral CD8(+) and CD4(+) T-cell infiltration and induced a specific TH1-type response to the tumor in regional lymph nodes and spleen at levels greater than those of nontransduced DCs. Moreover, splenocytes from animals treated with DC-IL-23 showed heightened levels of specific CTL activity. In vivo lymphocyte depletion experiments showed that the antitumor immunity induced by DC-IL-23 was mainly dependent on CD8(+) T cells and that CD4(+) T cells and natural killer cells were also involved. In summary, i.t. injection of DC-IL-23 resulted in significant and effective systemic antitumor immunity in intracranial tumor-bearing mice. These findings suggest a new approach to induce potent tumor-specific immunity to intracranial tumors. This approach may have therapeutic potential for treating human glioma.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/trasplante , Glioma/inmunología , Interleucina-23/inmunología , Melanoma/inmunología , Neoplasias/inmunología , Animales , Células de la Médula Ósea/inmunología , Línea Celular Tumoral , Interleucina-23/genética , Depleción Linfocítica , Ratones
10.
Neurochem Res ; 28(6): 883-91, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12718442

RESUMEN

Studies have shown an intimate relationship between cholesterol and retinal diseases; we examined the effects of cholesterol oxides on cultured cells. Using the rat retinal precursor cell line R28 and the human RPE cell line ARPE-19, we investigated the potential cytotoxicity of cholesterol oxides. Cultured R28 and ARPE-19 cells were treated with either 25-hydroxycholesterol and 7-ketocholesterol (0-50 microg/ml). Cell viability was determined by the WST-1 colorimetric assay. Production of reactive oxygen intermediate (ROI) was assessed by a fluorescent probe-based assay (2',7'-dichlorodihydrofluorescein diacetate [H2DCFDA]). To detect the presence of apoptosis, DNA fragmentation gel analysis and Hoescht nuclear staining were performed. Both cholesterol oxides tested were toxic in a time- and dose-dependent fashion to the two cell lines used in this study. Treatment of R28 cells with either 25-hydroxycholesterol or 7-ketocholesterol at a concentration of 25 micro/ml resulted in greater than 50% loss of cell viability after 24 h. ARPE-19 cells were slightly less affected, with a loss of cell viability of approximately 20% and 40% after 24 h-exposure of 25-hydroxycholesterol and 7-ketocholesterol, respectively. DNA fragmentation and chromatin condensation demonstrated apoptotic events occurring in 7-ketocholesterol-treated cells. The fluorescent assay for ROI production showed that after an hour of exposure to 7-ketocholesterol, R28 cells responded with increased levels of ROIs, whereas no immediate production of ROIs were detected with treated ARPE-19 cells. These in vitro findings provide evidence that cholesterol oxides can directly damage cultured retinal and RPE cells. The oxysterol-induced oxidative stress in these cells may be a factor in the pathology of retinal degenerative diseases.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Hidroxicolesteroles/toxicidad , Cetocolesteroles/toxicidad , Animales , Línea Celular , Relación Dosis-Respuesta a Droga , Humanos , Hidroxiesteroides/toxicidad , Cinética , Ratas , Retina
11.
Biochem Biophys Res Commun ; 295(3): 668-72, 2002 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-12099691

RESUMEN

Age-related macular degeneration (AMD) is the leading cause of blindness in the elderly. We report an association between an Alu polymorphism in the angiotensin-converting enzyme (ACE) gene with the dry/atrophic form of AMD. Using the polymerase chain reaction (PCR) on genomic DNA isolated from patients with AMD (n=173), and an age-matched control population (n=189), we amplified a region polymorphic for an Alu element insertion in the ACE gene. The Alu(+/+) genotype occurred 4.5 times more frequently in the control population than the dry/atrophic AMD patient population, (p=0.004). The predominance of the Alu(+/+) genotype within the unaffected control group represents a protective insertion with respect to the human ocular disease, dry/atrophic AMD. This is the first demonstration of an Alu element insertion exerting protective effects against a known human disease.


Asunto(s)
Elementos Alu/genética , Degeneración Macular/genética , Peptidil-Dipeptidasa A/genética , Polimorfismo Genético , Factores de Edad , Envejecimiento , Alelos , Secuencia de Bases , Genotipo , Humanos , Datos de Secuencia Molecular , Peptidil-Dipeptidasa A/metabolismo , Reacción en Cadena de la Polimerasa , Factores Sexuales , Activador de Tejido Plasminógeno/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA