Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Cell Biochem ; 118(2): 382-396, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27404023

RESUMEN

Apolipoprotein A-I (ApoA-I) is a key component of high density lipoproteins which possess anti-atherosclerotic and anti-inflammatory properties. Insulin is a crucial mediator of the glucose and lipid metabolism that has been implicated in atherosclerotic and inflammatory processes. Important mediators of insulin signaling such as Liver X Receptors (LXRs) and Forkhead Box A2 (FOXA2) are known to regulate apoA-I expression in liver. Forkhead Box O1 (FOXO1) is a well-known target of insulin signaling and a key mediator of oxidative stress response. Low doses of insulin were shown to activate apoA-I expression in human hepatoma HepG2 cells. However, the detailed mechanisms for these processes are still unknown. We studied the possible involvement of FOXO1, FOXA2, LXRα, and LXRß transcription factors in the insulin-mediated regulation of apoA-I expression. Treatment of HepG2 cells with high doses of insulin (48 h, 100 nM) suppresses apoA-I gene expression. siRNAs against FOXO1, FOXA2, LXRß, or LXRα abrogated this effect. FOXO1 forms a complex with LXRß and insulin treatment impairs FOXO1/LXRß complex binding to hepatic enhancer and triggers its nuclear export. Insulin as well as LXR ligand TO901317 enhance the interaction between FOXA2, LXRα, and hepatic enhancer. These data suggest that high doses of insulin downregulate apoA-I gene expression in HepG2 cells through redistribution of FOXO1/LXRß complex, FOXA2, and LXRα on hepatic enhancer of apoA-I gene. J. Cell. Biochem. 118: 382-396, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Apolipoproteína A-I/biosíntesis , Carcinoma Hepatocelular/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Proteína Forkhead Box O1/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Insulina/farmacología , Neoplasias Hepáticas/metabolismo , Receptores X del Hígado/metabolismo , Proteínas de Neoplasias/metabolismo , Carcinoma Hepatocelular/patología , Células Hep G2 , Humanos , Hidrocarburos Fluorados/farmacología , Neoplasias Hepáticas/patología , Sulfonamidas/farmacología
2.
J Cell Biochem ; 117(9): 2010-22, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26813964

RESUMEN

Apolipoprotein A-I (ApoA-I) is the main anti-atherogenic component of human high-density lipoproteins (HDL). ApoA-I gene expression is regulated by several nuclear receptors, which are the sensors for metabolic changes during development of cardiovascular diseases. Activation of nuclear receptor PPARγ has been shown to impact lipid metabolism as well as inflammation. Here, we have shown that synthetic PPARγ agonist GW1929 decreases both ApoA-I mRNA and protein levels in HepG2 cells and the effect of GW1929 on apoA-I gene transcription depends on PPARγ. PPARγ binds to the sites A and C within the hepatic enhancer of apoA-I gene and the negative regulation of apoA-I gene transcription by PPARγ appears to be realized via the site C (-134 to -119). Ligand activation of PPARγ leads to an increase of LXRß and a decrease of PPARα binding to the apoA-I gene hepatic enhancer in HepG2 cells. GW1929 abolishes the TNFα-mediated decrease of ApoA-I mRNA expression in both HepG2 and Caco-2 cells but does not block TNFα-mediated inhibition of ApoA-I protein secretion by HepG2 cells. These data demonstrate that complex of PPARγ with GW1929 is a negative regulator involved in the control of ApoA-I expression and secretion in human hepatocyte- and enterocyte-like cells. J. Cell. Biochem. 117: 2010-2022, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Apolipoproteína A-I/biosíntesis , Regulación hacia Abajo , Elementos de Facilitación Genéticos , Enterocitos/metabolismo , Hepatocitos/metabolismo , PPAR gamma/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Apolipoproteína A-I/genética , Benzofenonas/farmacología , Células CACO-2 , Células Hep G2 , Humanos , Receptores X del Hígado/genética , Receptores X del Hígado/metabolismo , PPAR alfa/genética , PPAR alfa/metabolismo , PPAR gamma/genética , Factor de Necrosis Tumoral alfa/genética , Tirosina/análogos & derivados , Tirosina/farmacología
3.
J Biol Chem ; 288(3): 1726-38, 2013 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-23168409

RESUMEN

Complement C3 is a pivotal component of three cascades of complement activation. The liver is the main source of C3 in circulation and expression and secretion of C3 by hepatocytes is increased during acute inflammation. However, the mechanism of the regulation of the C3 gene in hepatocytes is not well elucidated. We showed that the C3 gene is the direct target for peroxisome proliferator-activated receptor α (PPARα) in human hepatoma HepG2 cells and mouse liver. Using PPARα siRNA and synthetic PPARα agonist WY-14643 and antagonist MK886 we showed that activation of PPARα results in up-regulation of C3 gene expression and protein secretion by HepG2 cells. The PPAR response element (PPRE), which is able to bind PPARα in vitro and in vivo, was found in the human C3 promoter. PPRE is conserved between human and mouse, and WY-14643 stimulates mouse C3 expression in the liver. TNFα increases C3 gene via NF-κB and, to a lesser extent, MEK1/2 signaling pathways, whereas TNFα-mediated stimulation of C3 protein secretion depends on activation of MEK1/2, p38, and JNK in HepG2 cells. Activation of PPARα abolishes TNFα-mediated up-regulation of C3 gene expression and protein secretion due to interference with NF-κB via PPRE-dependent mechanism in HepG2 cells. TNFα decreases PPARα protein content via NF-κB and MEK1/2 signaling pathways and inhibits PPARα binding with the human C3 promoter in HepG2 cells. These results suggest novel mechanism controlling C3 expression in hepatocytes during acute phase inflammation and demonstrate a crosstalk between PPARα and TNFα in the regulation of complement system.


Asunto(s)
Complemento C3/genética , Hepatocitos/metabolismo , Inflamación/genética , PPAR alfa/genética , Factor de Necrosis Tumoral alfa/farmacología , Animales , Complemento C3/agonistas , Complemento C3/antagonistas & inhibidores , Complemento C3/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Células Hep G2 , Hepatocitos/efectos de los fármacos , Hepatocitos/inmunología , Humanos , Inflamación/inducido químicamente , Inflamación/inmunología , Inflamación/metabolismo , Lipopolisacáridos , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/genética , MAP Quinasa Quinasa 2/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/genética , FN-kappa B/metabolismo , PPAR alfa/inmunología , Proliferadores de Peroxisomas/farmacología , Unión Proteica , Pirimidinas/farmacología , Elementos de Respuesta , Transducción de Señal , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
4.
J Biol Chem ; 287(8): 5954-68, 2012 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-22194611

RESUMEN

Complement C3 is a pivotal component of three cascades of complement activation. C3 is expressed in human atherosclerotic lesions and is involved in atherogenesis. However, the mechanism of C3 accumulation in atherosclerotic lesions is not well elucidated. We show that acetylated low density lipoprotein and oxidized low density lipoprotein (oxLDL) increase C3 gene expression and protein secretion by human macrophages. Modified LDL (mLDL)-mediated activation of C3 expression mainly depends on liver X receptor (LXR) and partly on Toll-like receptor 4 (TLR4), whereas C3 secretion is increased due to TLR4 activation by mLDL. LXR agonist TO901317 stimulates C3 gene expression in human monocyte-macrophage cells but not in human hepatoma (HepG2) cells. We find LXR-responsive element inside of the promoter region of the human C3 gene, which binds to LXRß in macrophages but not in HepG2 cells. We show that C3 expression and secretion is decreased in IL-4-treated (M2) and increased in IFNγ/LPS-stimulated (M1) human macrophages as compared with resting macrophages. LXR agonist TO901317 potentiates LPS-induced C3 gene expression and protein secretion in macrophages, whereas oxLDL differently modulates LPS-mediated regulation of C3 in M1 or M2 macrophages. Treatment of human macrophages with anaphylatoxin C3a results in stimulation of C3 transcription and secretion as well as increased oxLDL accumulation and augmented oxLDL-mediated up-regulation of the C3 gene. These data provide a novel mechanism of C3 gene regulation in macrophages and suggest new aspects of cross-talk between mLDL, C3, C3a, and TLR4 during development of atherosclerotic lesions.


Asunto(s)
Complemento C3/genética , Complemento C3/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Lipoproteínas LDL/farmacología , Macrófagos/efectos de los fármacos , Receptores Nucleares Huérfanos/metabolismo , Receptor Toll-Like 4/metabolismo , Secuencia de Bases , Transporte Biológico , Complemento C3/biosíntesis , Células Hep G2 , Humanos , Lipoproteínas LDL/metabolismo , Receptores X del Hígado , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Macrófagos/citología , Macrófagos/metabolismo , FN-kappa B/metabolismo , Receptores Nucleares Huérfanos/agonistas , Regiones Promotoras Genéticas/genética , Transcripción Genética/efectos de los fármacos
5.
FASEB J ; 26(5): 2019-30, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22271762

RESUMEN

Apolipoprotein A-I (ApoA-I) is the main functional protein component of human high-density lipoproteins. ApoA-I shows various anti-inflammatory and atheroprotective properties toward macrophages; however, endogenous apoA-I expression has not been investigated in macrophages. We have shown that endogenous apoA-I gene is expressed in human macrophages at both mRNA and protein levels. Endogenous ApoA-I is localized in intracellular vesicles and at the external side of the plasma membrane in association with ATP-binding cassette transporter A1 (ABCA1) and lipid rafts in macrophages. We have shown that endogenous ApoA-I stabilizes ABCA1, moreover, down-regulation of ApoA-I by siRNA results in an increase of Toll-like receptor 4 (TLR4) mRNA and membrane surface protein expression, as well as an enhancement of bacterial lipopolysaccharide (LPS)-induced expression of tumor necrosis factor-α (TNF-α), interleukin 1ß (IL-1ß), and inducible nitric oxide synthase (NOS2) genes in human macrophages. TNF-α stimulates ApoA-I expression and secretion (1.2±0.2 vs. 4.3±0.9 ng/mg total protein) in macrophages. Obtained results suggest that endogenous ApoA-I has anti-inflammatory properties, presumably due to ABCA1 stabilization in macrophages; these results elucidate the cell type-specific mechanism of the TNF-α-mediated regulation of apoA-I gene expression in monocytes and macrophages.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Apolipoproteína A-I/fisiología , Macrófagos/metabolismo , Transducción de Señal , Receptor Toll-Like 4/metabolismo , Transportador 1 de Casete de Unión a ATP , Animales , Apolipoproteína A-I/genética , Secuencia de Bases , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
6.
Biochem Biophys Res Commun ; 402(3): 477-82, 2010 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-20951680

RESUMEN

Synthesis of ABCA1 protein in liver is necessary for high-density lipoproteins (HDL) formation in mammals. Nuclear receptor PPARγ is known as activator of ABCA1 expression, but details of PPARγ-mediated regulation of ABCA1 at both transcriptional and post-transcriptional levels in hepatocytes have not still been well elucidated. In this study we have shown, that PPARγ activates ABCA1 gene transcription in human hepatoma cells HepG2 through increasing of LXRß binding with promoter region of ABCA1 gene. Treatment of HepG2 cells with PPARγ agonist GW1929 leads to dissociation of LXRß from ABCA1/LXRß complex and to nuclear translocation of this nuclear receptor resulting in reduction of ABCA1 protein level 24h after treatment. Inhibition of protein kinases MEK1/2 abolishes PPARγ-mediated dissociation of LXRß from ABCA1/LXRß complex, but does not block PPARγ-dependent down-regulation of ABCA1 protein in HepG2 cells. These data suggest that PPARγ may be important for regulation of the level of hepatic ABCA1 protein and indicate the new interplays between PPARγ, LXRß and MEK1/2 in regulation of ABCA1 mRNA and protein expression.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Hígado/metabolismo , PPAR gamma/metabolismo , Activación Transcripcional , Transportador 1 de Casete de Unión a ATP , Benzofenonas/farmacología , Núcleo Celular/metabolismo , Regulación hacia Abajo , Células Hep G2 , Humanos , Receptores X del Hígado , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/antagonistas & inhibidores , MAP Quinasa Quinasa 2/metabolismo , Receptores Nucleares Huérfanos/genética , Receptores Nucleares Huérfanos/metabolismo , PPAR gamma/agonistas , Regiones Promotoras Genéticas , Biosíntesis de Proteínas , Transcripción Genética , Tirosina/análogos & derivados , Tirosina/farmacología
7.
Biochem Biophys Res Commun ; 398(2): 224-30, 2010 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-20599735

RESUMEN

Human apolipoprotein A-I (ApoA-I) is a major structural and functional protein component of high-density lipoproteins. The expression of the apolipoprotein A-I gene (apoA-I) in hepatocytes is repressed by pro-inflammatory cytokines such as IL-1beta and TNFalpha. Recently, two novel additional (alternative) promoters for human apoA-I gene have been identified. Nothing is known about the role of alternative promoters in TNFalpha-mediated downregulation of apoA-I gene. In this article we report for the first time about the different effects of TNFalpha on two alternative promoters of human apoA-I gene. Stimulation of HepG2 cells by TNFalpha leads to activation of the distal alternative apoA-I promoter and downregulation of the proximal alternative and the canonical apoA-I promoters. This effect is mediated by weakening of the promoter competition within human apoA-I 5'-regulatory region (apoA-I promoter switching) in the cells treated by TNFalpha. The MEK1/2-ERK1/2 cascade and nuclear receptors PPARalpha and LXRs are important for TNFalpha-mediated apoA-I promoter switching.


Asunto(s)
Apolipoproteína A-I/genética , Regulación de la Expresión Génica , Hepatocitos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Línea Celular Tumoral , Humanos , Receptores X del Hígado , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Receptores Nucleares Huérfanos/metabolismo , PPAR alfa/metabolismo , Regiones Promotoras Genéticas
8.
FEBS J ; 274(18): 4848-62, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17714511

RESUMEN

A series of relatively short (GCC)(n) triplet repeats (n = 3-30) located within regulatory regions of many mammalian genes may be considered as putative cis-acting transcriptional elements (GCC-elements). Fragile X-mental retardation syndrome is caused by an expansion of (GCC)(n) triplet repeats within the 5'-untranslated region of the human fragile X-mental retardation 1 (FMR1) gene. The present study aimed to characterize a novel human (GCC)(n)-binding protein and investigate its possible role in the regulation of the FMR1 gene. A novel human (GCC)(n)-binding protein, p56, was isolated and identified as a Krüppel-like transcription factor, ZF5, by MALDI-TOF analysis. The capacity of ZF5 to specifically interact with (GCC)(n) triplet repeats was confirmed by the electrophoretic mobility shift assay with purified recombinant ZF5 protein. In cotransfection experiments, ZF5 overexpression repressed activity of the GCC-element containing mouse ribosomal protein L32 gene promoter. Moreover, RNA interference assay results showed that endogenous ZF5 acts as a repressor of the human FMR1 gene. Thus, these data identify a new class of ZF5 targets, a subset of genes containing GCC-elements in their regulatory regions, and raise the question of whether transcription factor ZF5 is implicated in the pathogenesis of fragile X syndrome.


Asunto(s)
Regulación hacia Abajo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas Represoras/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Línea Celular Tumoral , Humanos , Factores de Transcripción de Tipo Kruppel/química , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/aislamiento & purificación , Peso Molecular , Proteínas Nucleares/aislamiento & purificación , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Secuencias Repetitivas de Ácidos Nucleicos/genética , Proteínas Represoras/química , Proteínas Represoras/genética , Proteínas Represoras/aislamiento & purificación , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
9.
Cell Stress Chaperones ; 22(1): 123-134, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27896567

RESUMEN

Reactive oxygen species damage various cell components including DNA, proteins, and lipids, and these impairments could be a reason for severe human diseases including atherosclerosis. Forkhead box O1 (FOXO1), an important metabolic transcription factor, upregulates antioxidant and proapoptotic genes during oxidative stress. Apolipoprotein A-I (ApoA-I) forms high density lipoprotein (HDL) particles that are responsible for cholesterol transfer from peripheral tissues to liver for removal in bile in vertebrates. The main sources for plasma ApoA-I in mammals are liver and jejunum. Hepatic apoA-I transcription depends on a multitude of metabolic transcription factors. We demonstrate that ApoA-I synthesis and secretion are decreased during H2O2-induced oxidative stress in human hepatoma cell line HepG2. Here, we first show that FOXO1 binds to site B of apoA-I hepatic enhancer and downregulates apoA-I gene activity in HepG2 cells. Moreover, FOXO1 and LXRα transcription factors participate in H2O2-triggered downregulation of apoA-I gene together with Src, JNK, p38, and AMPK kinase cascades. Mutations of sites B or C as well as the administration of siRNAs against FOXO1 or LXRα to HepG2 cells abolished the hydrogen peroxide-mediated suppression of apoA-I gene.


Asunto(s)
Proteína Forkhead Box O1/metabolismo , Peróxido de Hidrógeno/toxicidad , Receptores X del Hígado/metabolismo , Estrés Oxidativo/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Apolipoproteína A-I/genética , Apolipoproteína A-I/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Proteína Forkhead Box O1/antagonistas & inhibidores , Proteína Forkhead Box O1/genética , Células Hep G2 , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Receptores X del Hígado/antagonistas & inhibidores , Receptores X del Hígado/genética , Inhibidores de Proteínas Quinasas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo
10.
Gene ; 524(2): 187-92, 2013 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-23628799

RESUMEN

Complement C3 is involved in various protective and regulatory mechanisms of immune system. Recently it was established that C3 expression is regulated by nuclear receptors. Hepatic nuclear factor 4α (HNF4α) is a nuclear receptor critical for hepatic development and metabolism. We have shown that HNF4α is a positive regulator of C3 gene expression, realizing its effects through binding to two HNF4-response elements within the C3 promoter in HepG2 cells. TNFα is a well established positive regulator of C3 expression in hepatocytes during acute phase of inflammation. TNFα decreases the amount of HNF4α protein in HepG2 cells through NF-κB and MEK1/2 pathways thereby leading to a decrease in HNF4α bound to the C3 promoter. TNFα and HNF4α act in a synergetic way resulting in the potent activation of C3 transcription. These results suggest a novel mechanism of C3 regulation during acute phase response in HepG2 cells and display the mechanism of interaction of TNFα-induced pathways and HNF4α in transcriptional regulation of C3 gene.


Asunto(s)
Complemento C3/genética , Regulación de la Expresión Génica , Factor Nuclear 4 del Hepatocito/genética , Factor de Necrosis Tumoral alfa/inmunología , Complemento C3/inmunología , Complemento C3/metabolismo , Células Hep G2 , Factor Nuclear 4 del Hepatocito/inmunología , Factor Nuclear 4 del Hepatocito/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/inmunología , Hepatocitos/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas , FN-kappa B/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Mapeo de Interacción de Proteínas , Activación Transcripcional , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba
11.
J Biol Chem ; 278(43): 42625-36, 2003 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-12882958

RESUMEN

Arginine-rich peptides, penetratins, as part of a number of cellular and viral proteins, can penetrate across plasma membrane directly, without participation of endocytosis. We show that one of penetratins, the basic domain 47-57 of human immunodeficiency virus, type 1, transcription factor Tat (Tat peptide), is able to interact with plasmid DNA electrostatically. These interactions result in formation of polyelectrolytic complexes at various negative/positive charge ratios of plasmid DNA and Tat peptide. Plasmid DNA is capable of binding to Tat peptide up to 1.7-fold excess of the complex positive charge. The DNA-Tat complexes can be used for delivery of plasmid DNA into mammalian cells. Transfection efficacy of cultured cells by DNA-Tat complexes is stimulated by free Tat peptide, most likely because it protects DNA-Tat complexes from disruption by anionic proteoglycans of cellular surface. Our data strongly argue in favor of the endocytosis-dependent mechanism of DNA-Tat complex uptake by mammalian cells similarly to internalization of complexes of plasmid DNA with other polycationic carriers. Moreover, different cell lines use different endocytosis-mediated pathways for DNA-Tat complex internalization. Intravenous injections to mice of DNA-Tat complexes in comparison with injections of naked DNA showed an inhibitory effect of DNA-Tat complex positive charge on expression of transferred gene. A low level of foreign gene expression in the liver of mice injected intravenously with positively charged DNA-Tat complexes is accounted for by inactivation of DNA-Tat complexes in the bloodstream due to their interactions with serum albumin. These data should be taken into account in an attempt to develop versatile gene delivery systems based on penetratin application for human disease therapy.


Asunto(s)
Endocitosis , Productos del Gen tat/farmacocinética , Técnicas de Transferencia de Gen , Secuencia de Aminoácidos , Aminoácidos Básicos , Animales , Apolipoproteína A-I/genética , Proteínas Portadoras , Línea Celular , Péptidos de Penetración Celular , Sistemas de Liberación de Medicamentos , Productos del Gen tat/química , Productos del Gen tat/metabolismo , Terapia Genética/métodos , Vectores Genéticos/metabolismo , Vectores Genéticos/farmacocinética , Humanos , Cinética , Ratones , Plásmidos/metabolismo , Plásmidos/farmacocinética , Unión Proteica , Azida Sódica/farmacología , beta-Galactosidasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA