Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Mol Endocrinol ; 7(10): 1284-96, 1993 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8264660

RESUMEN

During left ventricular hypertrophy, brain natriuretic peptide (BNP) and atrial natriuretic factor (ANF) mRNA levels increase, possibly due to stretch-induced activation of protein kinase C. Phorbol ester treatment of primary cultures of neonatal rat ventricular cardiocytes represents an in vitro model of hypertrophic cell growth and has previously been shown to stimulate ANF synthesis and secretion. Using this model, we studied the synthesis and secretion of BNP to determine whether its regulation in cardiac cells is similar to ANF. Addition of 10(-7) M phorbol 12-myristate 13-acetate (PMA) resulted in a 3- to 4-fold increase in immunoreactive BNP (irBNP) secretion 24-48 h after treatment. Over a concentration range of 10(-8)-10(-6) M, PMA increased irBNP secretion to equivalent levels. Another phorbol ester agonist, phorbol 12,13-didecanoate, stimulated irBNP secretion, while the inactive analog 4 alpha-phorbol 12,13-didecanoate had no effect. Inhibition of protein kinase C (PKC) with 10(-8) M staurosporine decreased basal secretion of irBNP 60% and prevented PMA induction of irBNP, whereas both stimulated and basal secretion of ANF were minimally affected. BNP mRNA increased 6-fold by 3 h of PMA treatment and remained elevated above control levels for 48 h. Staurosporine prevented the increase in BNP mRNA. To determine whether PKC or a PKC-dependent pathway was involved in persistent stimulation of BNP and ANF in cells chronically treated with PMA, ventricular cardiocytes were treated with PMA for 24 h, followed by PMA plus 10(-8) M staurosporine for 24 h. BNP mRNA was reduced to control levels, while ANF mRNA was reduced by an average of 20%. To test whether mRNA stability was involved in the differential effect of chronic phorbol ester treatment, cardiocytes were treated with the protein synthesis inhibitor cycloheximide (20 micrograms/ml). BNP mRNA levels were stimulated as early as 30 min after treatment, but ANF mRNA remained unaffected. Cycloheximide also potentiated PMA's effect on BNP mRNA after 1.5, 9.5, and 24 h of treatment. To test whether a transcriptional mechanism was involved in the stimulation of BNP mRNA by PMA, cells were treated with the inhibitor actinomycin D (5 micrograms/ml) for 24 h in the presence of PMA. Actinomycin D reduced the stimulatory effect of PMA on BNP mRNA.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Animales Recién Nacidos , Factor Natriurético Atrial/metabolismo , Miocardio/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Acetato de Tetradecanoilforbol/farmacología , Alcaloides/farmacología , Animales , Células Cultivadas , Dactinomicina/farmacología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Cinética , Péptido Natriurético Encefálico , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Ésteres del Forbol/farmacología , Proteína Quinasa C/metabolismo , Inhibidores de la Síntesis de la Proteína/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Estaurosporina , Transcripción Genética/efectos de los fármacos
2.
Mol Endocrinol ; 5(9): 1311-22, 1991 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-1837591

RESUMEN

Expression of the human atrial natriuretic peptide (hANP)gene is controlled by a series of positive and negative cis-acting regulatory elements present in the 5' flanking sequences (5'FS) of the gene. Positive elements located between -1150 and -222, relative to the transcription start site, appear to be responsible for the major portion of ANP gene expression in neonatal rat cardiac atrial cells. While neonatal ventricular cardiocytes, at a qualitative level, seem to employ regulatory signals similar to their atrial counterparts, they do so with reduced efficiency. Expression of the hANP gene in nonmyocardial cells is limited by the presence of silencer elements in the distal (-2593 to -1150) and proximal (-222 to the CAP site) 5'FS. Further characterization of a 64-base pair cardiac-specific element (-410 to -332), described previously, revealed that a core sequence of 40 base pairs is required for functional activity. This core sequence includes a previously defined DNAse-I footprint region flanked by two GC-rich segments arranged in an inverted repeat-like array. These findings suggest that the disparity in atrial vs. ventricular cardiocyte expression of the ANP gene reflects differences that are largely quantitative in nature, while differences in myocardial vs. nonmyocardial cells result from fundamental qualitative differences in the way these cells recognize and use the regulatory elements present within the 5'FS.


Asunto(s)
Factor Natriurético Atrial/genética , Regulación Neoplásica de la Expresión Génica , Miocardio/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Animales , Animales Recién Nacidos , Secuencia de Bases , Cloranfenicol O-Acetiltransferasa/genética , Análisis Mutacional de ADN , Fibroblastos , Atrios Cardíacos , Ventrículos Cardíacos , Humanos , Datos de Secuencia Molecular , Plásmidos/genética , Ratas , Proteínas Recombinantes de Fusión , Homología de Secuencia de Ácido Nucleico
3.
Endocrinology ; 128(1): 5-12, 1991 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-1702703

RESUMEN

To identify factors that directly regulate the synthesis and secretion of atrial natriuretic peptide (ANP) in neuronal cells, we have developed a neuron-enriched primary culture system from fetal rat brains. A number of factors proved of importance in maintaining adequate levels of ANP secretion in such cultures: 1) cultures derived from diencephalon produced much more ANP than cultures derived from diencephalon produced with the distribution of ANP-containing cells in the rat brain; 2) brains from rats at gestational day 17 proved a better source of ANP-secreting cells than brains from rats at gestational day 16; 3) the presence of serum was required in the latter stages of the culture period to allow expression of the ANP gene; and 4) the cultures secreted more ANP when maintained at 39 C vs. 37 C. ANP mRNA transcripts in the neuron-enriched primary cultures were analyzed by S1 nuclease protection and shown to have a transcription start site similar to that employed by rat atrium and fetal hypothalamus in vivo. Dexamethasone and T3, in contrast to their stimulatory effect on ANP production in cardiocyte cultures, suppressed both the release of immunoreactive ANP and the levels of ANP mRNA in the neuron-enriched primary cultures. The cultures incorporated [35S]cysteine into immunoprecipitable ANP. HPLC analysis of 35S-labeled products in the medium revealed that, unlike neonatal cardiocyte cultures, the majority of secreted immunoreactive ANP migrated with the processed form(s) of ANP rather than the prohormone.


Asunto(s)
Factor Natriurético Atrial/genética , Encéfalo/metabolismo , Neuronas/metabolismo , Animales , Animales Recién Nacidos , Astrocitos/metabolismo , Factor Natriurético Atrial/biosíntesis , Factor Natriurético Atrial/aislamiento & purificación , Células Cultivadas , Dexametasona/farmacología , Feto , Inmunohistoquímica , Cinética , Miocardio/metabolismo , Neuronas/efectos de los fármacos , ARN/genética , ARN/aislamiento & purificación , ARN Mensajero/genética , Ratas , Transcripción Genética , Triyodotironina/farmacología
4.
Hypertension ; 35(1 Pt 2): 249-54, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10642306

RESUMEN

We have previously shown that the regulation by interleukin-1beta (IL-1beta) of inducible nitric oxide synthase (iNOS) involves phospholipase A(2) (PLA(2)) metabolites in neonatal ventricular myocytes. Based on studies in which ONO-RS-082 is used to inhibit secretory PLA(2) and methyl arachidonyl fluorophosphonate is used to inhibit cytosolic PLA(2), our data suggest that a secretory PLA(2) metabolite was involved in the regulation by IL-1beta of iNOS. In addition, a third PLA(2) isoform, which is Ca(2+) independent (iPLA(2)), has also been detected in cardiac myocytes and shown to be regulated by cytokines. We tested whether iPLA(2) metabolites are involved in the regulation by IL-1beta of iNOS with the use of bromoenol lactone (BEL), a specific and irreversible inhibitor of iPLA(2). For this, we measured IL-1beta-stimulated nitrite (NOx) production with use of the Griess reagent, prostaglandin E(2) (PGE(2)) production with use of an enzyme immunoassay, and arachidonic acid release in the presence and absence of BEL. We also detected iNOS and iPLA(2) proteins by Western blotting. Treatment with IL-1beta (5 ng/mL) for 24 hours stimulated NOx production by 8-fold and iNOS protein levels by at least 10-fold. In addition, arachidonic acid release was increased by 1.6-fold and PGE(2) production was increased by 300-fold. When neonatal ventricular myocytes were treated with 10 micromol/L BEL, both IL-1beta-stimulated PGE(2) production and arachidonic acid release were inhibited. BEL inhibited IL-1beta-stimulated NOx production and iNOS protein by 88% and 93%, respectively. Lysophosphatidic acid, but not arachidonic acid or lysophosphatidylcholine, stimulated iNOS expression. Our results indicate that an iPLA(2) metabolite, perhaps lysophosphatidic acid, may be involved in the IL-1beta-signaling pathway, regulating the synthesis of iNOS.


Asunto(s)
Fibras Musculares Esqueléticas/enzimología , Miocardio/enzimología , Óxido Nítrico Sintasa/metabolismo , Fosfolipasas A/metabolismo , Animales , Animales Recién Nacidos , Ácido Araquidónico/farmacología , Calcio/metabolismo , Células Cultivadas , Dinoprostona/metabolismo , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/enzimología , Interleucina-1/farmacología , Lisofosfatidilcolinas/farmacología , Lisofosfolípidos/farmacología , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/efectos de los fármacos , Miocardio/citología , Naftalenos/farmacología , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico Sintasa de Tipo II , Inhibidores de Fosfodiesterasa/farmacología , Fosfolipasas A/antagonistas & inhibidores , Pironas/farmacología , Ratas , Ratas Sprague-Dawley
5.
Hypertension ; 35(1 Pt 2): 292-6, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10642313

RESUMEN

We have shown that interleukin-1beta (IL-1beta) activates the human brain natriuretic peptide (hBNP) promoter via a transcriptional mechanism. Others have reported that changes in intracellular calcium (Ca(2+)) mediate the action of IL-1beta. We questioned whether Ca(2+) and Ca(2+)-dependent pathways mediate IL-1beta regulation of the hBNP promoter in cardiac myocytes. The hBNP promoter (-1818 to +100) coupled to a luciferase cDNA reporter gene was transferred into neonatal cardiac myocytes. Cells were then treated with agents that modify Ca(2+) levels or inhibit Ca(2+)-dependent kinases, and luciferase activity was measured as an index of hBNP promoter activity. The Ca(2+) ionophore A23187 increased hBNP promoter activity; however, neither EGTA nor nifedipine reduced IL-1beta-stimulated promoter activity. Long-term treatment with thapsigargin, which depletes intracellular Ca(2+) stores, decreased basal promoter activity and blocked the effect of IL-1beta. Inhibition of protein kinase C completely blocked IL-1beta-stimulated hBNP promoter activity, whereas inhibition of Ca(2+)/calmodulin-dependent kinase II decreased promoter activity by 40%. In contrast, inhibition of the Ca(2+)-regulated phosphatase calcineurin by cyclosporin A had no effect. These data suggest that (1) Ca(2+) activates the hBNP promoter; (2) release of Ca(2+) from intracellular stores is important to IL-1beta regulation of the hBNP promoter, but transport via voltage-sensitive Ca(2+) channels is not; (3) protein kinase C and Ca(2+)/calmodulin-dependent kinase II mediate the action of IL-1beta; and (4) the phosphatase calcineurin is not involved in IL-1beta regulation of the hBNP promoter. Thus, Ca(2+) and Ca(2+)-dependent pathways are critical to IL-1beta regulation of the hBNP promoter.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Interleucina-1/farmacología , Péptido Natriurético Encefálico/genética , Regiones Promotoras Genéticas/fisiología , Animales , Calcineurina/metabolismo , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Cardiomegalia/enzimología , Células Cultivadas , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/enzimología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Miocardio/citología , Miocardio/enzimología , Proteína Quinasa C/metabolismo , Ratas , Ratas Sprague-Dawley , Transfección
6.
Hypertension ; 31(1 Pt 2): 218-24, 1998 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9453306

RESUMEN

The proinflammatory cytokine interleukin-1beta (IL) stimulates inducible nitric oxide synthase (iNOS) mRNA, protein, and nitric oxide (NO) production in neonatal ventricular myocytes (NVM). In other types of cells, IL also activates phospholipase A2 (PLA2), which liberates arachidonic acid for the pathways involved in eicosanoid production, and induces the cyclooxygenase-2 (COX-2) isoform, which increases prostanoid production. Since NO has been shown to directly stimulate COX activity and the resulting prostanoids to modulate IL induction of iNOS, we questioned whether PLA2 and/or COX products are involved in IL regulation of iNOS and NO production in NVM. We first found that IL induced COX-2 mRNA and protein, resulting in approximately 200-fold and 15-fold increases in PGE2 and 6-keto-PGF1alpha (the stable metabolite of PGI2), respectively. IL-stimulated prostanoid production was inhibited by the COX-2-specific inhibitor NS-398, as well as the nonspecific COX inhibitor indomethacin (INDO). We next studied the involvement of the PLA2 inhibitor ONO-RS-082 (ONO) and the COX inhibitor INDO in IL regulation of iNOS. Pretreatment with ONO blocked IL-stimulated NO production and iNOS protein, suggesting that PLA2 products are involved in regulation of iNOS synthesis. Unlike ONO, the COX inhibitor INDO had little effect on IL-stimulated NO. In addition to the COX pathway, arachidonic acid (AA) is also metabolized by the lipoxygenase (LO) pathway. The LO inhibitor nordihydroguaiaretic acid (NDGA) decreased IL-stimulated NO and iNOS synthesis. These data suggest that: (1) IL upregulates COX-2 expression and prostanoid production in NVM; and (2) AA metabolites other than COX products, possibly products of the LO pathway, are involved in IL regulation of iNOS.


Asunto(s)
Interleucina-1/farmacología , Isoenzimas/biosíntesis , Miocardio/enzimología , Óxido Nítrico Sintasa/biosíntesis , Fosfolipasas A/metabolismo , Prostaglandina-Endoperóxido Sintasas/biosíntesis , 6-Cetoprostaglandina F1 alfa/biosíntesis , Animales , Animales Recién Nacidos , Células Cultivadas , Ciclooxigenasa 2 , Dinoprostona/biosíntesis , Inducción Enzimática/efectos de los fármacos , Epoprostenol/biosíntesis , Indometacina/farmacología , Masoprocol/farmacología , Miocardio/citología , Óxido Nítrico Sintasa de Tipo II , Fosfolipasas A2 , Prostaglandinas/biosíntesis , Ratas , Ratas Sprague-Dawley , Transcripción Genética
7.
Hypertension ; 33(1 Pt 2): 276-82, 1999 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9931117

RESUMEN

The genes encoding inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2, also known as prostaglandin-endoperoxide synthase-2) are induced in many types of cells in response to proinflammatory cytokines. We have previously shown that interleukin-1beta (IL) stimulates iNOS and COX-2 mRNA in cardiac myocytes. Because IL has been shown to activate mitogen-activated protein kinase (MAPK) signaling pathways in many different cells, we tested whether the p42/44 and p38 MAPK pathways were involved in IL stimulation of iNOS and COX-2, using a specific inhibitor of p42/44 activation, PD98059 (PD), and the p38 inhibitor SB205380 (SB). Nitrites were measured using the Griess reagent, prostaglandin PGE2 by an enzyme immunoassay, iNOS and COX-2 protein by Western blot analysis, and iNOS mRNA by Northern blot analysis. Tested separately, the p38 kinase and MAPK inhibitors partially reduced IL stimulation of nitrite, iNOS protein, and iNOS mRNA; used together, they completely abolished the effect of IL. SB and PD inhibited IL-stimulated COX-2 protein by 60% and 80%, respectively, and IL-stimulated COX-2 protein was totally prevented by the combination of inhibitors. PGE2 production was inhibited more than 99% by either drug alone, suggesting a posttranslational effect on enzyme activity. To test whether this posttranslational effect involved the cytosolic phospholipase A2 (cPLA2) isoform, Western blots were probed for cPLA2 protein. Results indicated that IL stimulated cPLA2 activity and synthesis, which was inhibited by SB but not PD. These data indicate that (1) IL induction of iNOS synthesis depends on both the p42/44 and p38 signaling pathways, acting primarily at the level of transcriptional regulation; and (2) IL regulation of COX-2 synthesis involves the p42/44 and p38 signaling pathways, with an additional level of regulation occurring posttranslationally, perhaps at the level of activation of the cPLA2 isoform, which may be involved in intracellular signaling, as well as regulation of arachidonic acid release for COX-2 activity.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Regulación Enzimológica de la Expresión Génica , Corazón/fisiología , Interleucina-1/fisiología , Isoenzimas/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos , Óxido Nítrico Sintasa/genética , Prostaglandina-Endoperóxido Sintasas/genética , Transducción de Señal/fisiología , Animales , Animales Recién Nacidos , Células Cultivadas , Ciclooxigenasa 2 , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Corazón/efectos de los fármacos , Ventrículos Cardíacos , Imidazoles/farmacología , Interleucina-1/farmacología , Proteína Quinasa 3 Activada por Mitógenos , Miocardio/citología , Óxido Nítrico Sintasa de Tipo II , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Transcripción Genética/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos
8.
Hypertension ; 33(1 Pt 2): 283-9, 1999 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9931118

RESUMEN

Because both the brain natriuretic peptide (BNP) gene and the cytokine interleukin-1beta (IL-1beta) are induced in the infarcted myocardium, localized production of IL-1beta may regulate the BNP gene. We tested whether (1) IL-1beta regulates the human BNP promoter, (2) cis elements in the proximal promoter respond to IL-1beta, and (3) mitogen-activated protein kinase (MAPK) signaling pathways [p42/44, c-jun (JNK) and p38 kinase] are involved. We transferred the hBNP promoter coupled to a luciferase reporter gene or constructs with mutations in the proximal promoter GATA and M-CAT elements into neonatal rat ventricular myocytes and treated the cells with IL-1beta for 24 hours. IL-1beta-stimulated hBNP luciferase activity was eliminated by pretreatment with the transcription inhibitor actinomycin D. Both the p38 kinase inhibitor SB205380 (SB) and cotransfection of a dominant-negative mutant of p38 kinase reduced IL-1beta stimulation of the hBNP promoter. Dominant-negative mutants of Ras and Rac inhibited IL-1beta-stimulated hBNP luciferase activity by 64% and 90%, respectively. Constitutively active forms of Rac and MKK6, the immediate upstream activator of p38, were stimulatory; however, only the effect of MKK6 was inhibited by SB. Neither the p42/44 nor the JNK pathway was involved in the action of IL-1beta. Both IL-1beta and MKK6 activation of the hBNP promoter were partially reduced when the promoter contained a mutated M-CAT element. In summary, (1) IL-1beta is a transcriptional activator of the hBNP promoter; (2) IL-1beta acts through a Ras-dependent pathway not coupled to activation of p42/44 MAPK or JNK; (3) IL-1beta acts through a Rac-dependent pathway, but the downstream effector is not known; and (4) IL-1beta activation of p38 kinase is partially involved in regulation of the hBNP promoter, targeting the proximal M-CAT element.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Regulación de la Expresión Génica , Interleucina-1/fisiología , Proteínas Quinasas Activadas por Mitógenos , Miocardio/metabolismo , Péptido Natriurético Encefálico/genética , Regiones Promotoras Genéticas , Transducción de Señal , Proteínas ras/metabolismo , Animales , Animales Recién Nacidos , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Células Cultivadas , Dactinomicina/farmacología , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Proteínas Activadoras de GTPasa , Regulación de la Expresión Génica/efectos de los fármacos , Corazón/fisiología , Ventrículos Cardíacos , Humanos , Imidazoles/farmacología , Interleucina-1/farmacología , Luciferasas/biosíntesis , Luciferasas/genética , MAP Quinasa Quinasa 6 , Miocardio/citología , Proteínas/metabolismo , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/metabolismo , Transcripción Genética/efectos de los fármacos , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos , Proteínas Activadoras de ras GTPasa
9.
Hypertension ; 27(3 Pt 2): 709-14, 1996 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-8613229

RESUMEN

Cytokines and endotoxin stimulate inducible NO synthase (iNOS) in different types of cells; however, little is known about regulatory mechanisms. Using the Griess reagent for nitric levels, Western blots for iNOS protein, Northern blots for iNOS mRNA, and transient transfection studies to monitor transcription, we determined potential mechanisms involved in interleukin-1beta stimulation of iNOS in cultured neonatal ventricular myocytes. When myocytes were treated with interleukin-1beta (5 ng/mL), nitrite levels increased, and this effect was inhibited 80% by the specific iNOS inhibitor aminoguanidine. Neither interferon gamma nor tumor necrosis factor-alpha alone stimulated nitrite production. Bacterial endotoxin alone stimulated nitrites and potentiated the effect of interleukin. To determine whether a tyrosine kinase-mediated signaling pathway was involved in interleukin action, we used the inhibitor genistein, which blocked interleukin-stimulated nitrites, iNOS protein, and iNOS mRNA. To determine the effect of activation of protein kinase C, we treated cells with the phorbol ester phorbol 12-myristate 13-acetate (PMA). PMA decreased both interleukin-stimulated nitrites and iNOS protein by 40%. To determine the involvement of cyclic nucleotides, cells were treated with either dibutyryl cAMP or cGMP. cAMP (1 mmol/L) stimulated iNOS mRNA, protein, and nitrite production, whereas cGMP had no effect. To test for a direct effect of interleukin on transcription of the iNOS gene, we transfected the full-length mouse iNOS 5' regulatory sequences (-1592 to +160) coupled to a luciferase reporter gene (-1592iNOSLuc). Interleukin stimulated luciferase activity 1.8 +/- 0.2-fold. To determine whether interleukin also affects iNOS mRNA stability, interleukin-stimulated iNOS mRNA was allowed to decay in the presence of the transcription inhibitor actinomycin D. iNOS mRNA t1/2 (approximately 1 hour) was not affected by interleukin. Thus, our data suggest that (1) interleukin-1beta is the primary cytokine in myocyte iNOS regulation and acts predominantly at the transcriptional level; (2) interleukin stimulation of iNOS mRNA and protein is coupled to a tyrosine kinase-mediated signaling pathway; and (3) protein kinase C and cAMP can modify interleukin signaling by decreasing and increasing iNOS, respectively.


Asunto(s)
Interleucina-1/farmacología , Miocardio/enzimología , Óxido Nítrico Sintasa/biosíntesis , Animales , Células Cultivadas , Regulación Enzimológica de la Expresión Génica , Genisteína , Isoflavonas/farmacología , Lipopolisacáridos/farmacología , Óxido Nítrico Sintasa/genética , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Transcripción Genética/efectos de los fármacos
10.
Hypertension ; 25(3): 421-30, 1995 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-7875768

RESUMEN

Using cultured neonatal ventricular myocytes, we investigated whether nitric oxide (NO) directly influences myocyte growth. Treatment of myocytes with phenylephrine stimulated growth, as indicated by increases in atrial natriuretic factor, brain natriuretic peptide (BNP) mRNA and BNP secretion, activator protein 1 activity (activation of early-response genes), and total cellular protein content. NO was stimulated by treatment of myocytes with interleukin-1 beta (IL-1 beta) or was generated by the NO donor nitroglycerin, and its effects on total protein content and BNP secretion were measured. Treatment of cardiocytes with 3.4 nmol/L IL-1 beta for 24 hours stimulated NO (nitrite) production by threefold, which resulted from an increase in the inducible isoform of NO synthase mRNA. Dexamethasone inhibited IL-1 beta induction of nitrite production, whereas the protein kinase C inhibitor staurosporine had no effect. IL-1 beta had no effect on either basal or phenylephrine-stimulated protein content but inhibited phenylephrine-stimulated BNP secretion. Nitroglycerin (10(-7) to 10(-3) mol/L) dose-dependently increased NO production; however, only the highest dose (10(-3) mol/L) reduced basal and phenylephrine-stimulated total protein content and BNP secretion. cGMP, a second messenger of NO, had no effect on either basal or phenylephrine-stimulated BNP secretion or total protein content. In conclusion, our data indicate that BNP mRNA is stimulated by phenylephrine as shown previously for atrial natriuretic factor. Although both BNP and total protein content are increased by phenylephrine, these effects are not inhibited by NO. However, IL-1 beta inhibits phenylephrine-stimulated BNP secretion but not total protein content, suggesting that regulation of BNP secretion can be dissociated from total protein synthesis during myocyte growth.


Asunto(s)
Corazón/efectos de los fármacos , Interleucina-1/farmacología , Óxido Nítrico/farmacología , Animales , Factor Natriurético Atrial/genética , Factor Neurotrófico Derivado del Encéfalo , Bucladesina/farmacología , Células Cultivadas , GMP Cíclico/fisiología , Miocardio/citología , Proteínas del Tejido Nervioso/genética , Nitroglicerina/farmacología , Fenilefrina/farmacología , Proteínas/metabolismo , ARN Mensajero/metabolismo , Ratas , Factor de Transcripción AP-1/metabolismo
11.
J Clin Endocrinol Metab ; 69(4): 729-37, 1989 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-2528567

RESUMEN

Expression of the human atrial natriuretic peptide (hANP) gene was examined in tissues of 19 to 28-week-old human fetuses. As expected the fetal atria express the hANP gene at a high level, accruing substantial quantities of ANP immunoreactivity and ANP mRNA. The neonatal ventricle also expresses the hANP gene at a significant level. ANP mRNA levels in the ventricle were, on the average, about 20% of those in the right atria. ANP immunoreactivity in ventricle was less that 5% of that in the right atria, suggesting important differences in the way these tissues synthesize and store the ANP peptide. Much lower levels of hANP transcripts were also detected in the lung and aortic arch. Analysis of the 5' termini of cardiac hANP transcripts using three independent techniques suggests the presence of two transcription start sites. A major start site is located approximately 28 basepairs downstream from the primary TATA sequence. A second minor start site is positioned about 110 basepairs further upstream. Immunocytochemistry and in situ hybridization analysis indicate that the hANP gene is expressed in a homogeneous distribution throughout the atrial myocardium. Diffuse low level expression is also present within the ventricular myocardium. In addition, there are scattered foci of increased expression in the ventricle which tend to be concentrated in the subendocardium of that organ. These findings indicate that the developing human ventricles as well as the atria possess the capacity to express the hANP gene at a substantial level and suggest that the ventricle may contribute significantly to the circulating pool of plasma ANP.


Asunto(s)
Factor Natriurético Atrial/genética , Genes , Corazón/embriología , ARN Mensajero/genética , Transcripción Genética , Factor Natriurético Atrial/análisis , Feto , Humanos , Inmunohistoquímica , Miocardio/citología , Hibridación de Ácido Nucleico , ARN Mensajero/análisis
12.
Hypertension ; 27(3 Pt 2): 715-22, 1996 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-8613230

RESUMEN

Brain natriuretic peptide (BNP) is a cardiac hormone constitutively expressed in the adult heart. To identify the cis-acting elements involved in regulation of the human BNP gene, we subcloned the full-length promoter (-1818 to +100) and deletions thereof upstream from a luciferase reporter gene and transiently transfected them into primary cultures of neonatal rat atrial and ventricular myocytes and myocardial fibroblasts. Luciferase activity of the full-length construct was higher in ventricular (39064 +/- 8488 relative light units, N=11) and atrial (11225 +/- 1907, N=17) myocytes than myocardial fibroblasts (329 +/- 113, n=5). Maximal promoter activity in ventricular and atrial myocytes was maintained by sequences positioned between -1818 and -1283 relative to the transcription start site. Deletion to -1175 resulted in a decrease, whereas further deletion to -500 effected an increase in reporter activity in both cell types. In ventricular and atrial myocytes, deletion from -500 to -40 reduced luciferase activity 20-fold and 2-fold, respectively, whereas in myocardial fibroblasts, deletion to -40 upregulated the BNP promoter 2-fold. Of note, deleting 16 bp between -127 and -111 reduced luciferase activity 7-fold and 4-fold in ventricular and atrial myocytes, respectively, but had essentially no effect on luciferase activity in fibroblasts. Placement of sequences lying between -127 and -40 upstream from a heterologous thymidine kinase promoter resulted in reporter expression that was 7.4-fold greater than the vector alone in ventricular myocytes, approximately 2-fold greater in atrial myocytes, and equivalent to the vector alone in fibroblasts. For study of activity of the human BNP promoter in adult myocytes, either 408 or 97 bp of 5' flanking sequence coupled to the luciferase reporter gene was injected into the apex of adult male Sprague-Dawley rat hearts. After 7 days, luciferase activity in the injected myocardium was 9.8-fold higher for the longer construct (70683 +/- 14744 versus 7223 +/- 3920, n=4, P < .01), consistent with our in vitro data. These data indicate that (1) the full-length human BNP promoter is more active in ventricular versus atrial myocytes and essentially inactive in fibroblasts, (2) the distal BNP promoter contains both positive and negative regulatory elements, (3) a region of the proximal BNP promoter located between -127 and -40 confers tissue specificity, and (4) the BNP promoter is active after injection into the adult rat heart.


Asunto(s)
Regulación de la Expresión Génica , Miocardio/metabolismo , Proteínas del Tejido Nervioso/genética , Animales , Secuencia de Bases , Células Cultivadas , Técnicas de Transferencia de Gen , Humanos , Datos de Secuencia Molecular , Péptido Natriurético Encefálico , Proteínas del Tejido Nervioso/biosíntesis , Especificidad de Órganos , Regiones Promotoras Genéticas/genética , Ratas , Ratas Sprague-Dawley
13.
Hypertension ; 15(1): 20-8, 1990 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-1688546

RESUMEN

Primary cultures of neonatal rat cardiocytes were exposed for 24 hours to culture media containing 0-2.0 mM extracellular calcium. Both atrial natriuretic factor (ANF) messenger RNA (mRNA) and ANF secretion were increased approximately threefold in the presence of 2.0 mM CaCl2 (vs. Ca2(+)-free medium). When cardiocytes were treated with the calcium channel blockers diltiazem, nifedipine, or verapamil, both ANF synthesis and secretion fell to 25-40% of control values. The choice of transcription start site on the ANF gene was not altered by the calcium channel blockers. When exogenous calcium was added to cardiocytes treated with verapamil, secretion of ANF was partially restored to control levels. High-performance liquid chromatographic analysis of medium from cardiocytes exposed to varying extracellular calcium concentrations or treated with calcium channel blockers for 24 hours revealed that the majority of secreted immunoreactivity (60-70%) migrated with pro-ANF (17 kDa) and that none of the various experimental manipulations significantly changed the ratio of pro-ANF to ANF in the media. These results were confirmed by immunoprecipitation analysis of the culture medium from the individual treatment groups. Treatment of cardiocytes for 24 hours with either the calcium ionophore A23187 or the phorbol ester 12-O-tetradecanoylphorbol 13-acetate increased ANF secretion. The combined use of these agents resulted in stimulation of both ANF secretion and ANF mRNA accumulation.


Asunto(s)
Factor Natriurético Atrial/genética , Calcio/fisiología , Espacio Extracelular/fisiología , Regulación de la Expresión Génica/fisiología , Animales , Calcimicina/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Células Cultivadas , Cromatografía Líquida de Alta Presión , Medios de Cultivo , Miocardio/citología , Miocardio/metabolismo , Hibridación de Ácido Nucleico , Pruebas de Precipitina , ARN , ARN Complementario , Acetato de Tetradecanoilforbol/farmacología
14.
Brain Res Mol Brain Res ; 11(3-4): 355-8, 1991 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-1661827

RESUMEN

There is increasing evidence that some proteases and protease inhibitors are produced within the central nervous system. It has been proposed that the balance between these two classes of proteins may be an important modulator of brain cell growth and differentiation. Here we report that antithrombin III (ATIII) is produced in brain and primary astroglial cultures. In addition, we show that human astroglial cultures contain prothrombin mRNA, and secrete a thrombin-like protein that makes complexes with antithrombin III.


Asunto(s)
Antitrombina III/genética , Astrocitos/fisiología , Encéfalo/fisiología , Trombina/genética , Animales , Northern Blotting , Línea Celular , Células Cultivadas , Glioma , Humanos , Hígado/fisiología , Especificidad de Órganos , Ratas
15.
Am J Hypertens ; 12(2 Pt 1): 174-82, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10090345

RESUMEN

The inducible nitric oxide synthase isoform (iNOS) is upregulated by cytokines and endotoxins in many types of cells, including cardiac myocytes. Nitric oxide (NO) induced by cytokines can be cytotoxic, and has been implicated in the pathophysiology of myocardial infarction, cardiomyopathy, and septic shock. To examine the role of iNOS in the ischemic myocardium, we studied: 1) the time course of expression of iNOS mRNA after myocardial infarction (MI) in male Sprague-Dawley rat hearts and expression of iNOS protein in the infarcted region; 2) whether hypoxia in vitro is a potential mediator of the induction of iNOS mRNA; and 3) whether inhibition of iNOS by two different selective inhibitors (aminoguanidine and S-methylisothiourea sulfate) in vivo influences infarct size. Myocardial infarction was induced by ligation of the left anterior descending coronary artery (LAD), and tissue was collected at selected times thereafter from both ligated and sham-operated rats. iNOS mRNA was induced in the infarcted region of the left ventricle for 7 days; iNOS protein was also detected in the infarcted area. We next tested whether hypoxia would induce iNOS in vitro. In cultured neonatal ventricular myocytes, iNOS mRNA was slightly induced by 6 to 24 h of hypoxia; however, iNOS protein was only detected when the cytokine interleukin-1beta was present. To study whether iNOS activity contributed to myocardial damage (eg, infarct size), we administered the first dose of the NOS inhibitors 24 h before LAD occlusion and then a second dose after surgery. Inhibition of iNOS activity with aminoguanidine reduced infarct size by 20% but had no effect on infiltration by neutrophils, whereas the more selective inhibitor S-methylisothiourea sulfate reduced infarct size by 41%. These data suggest that NO derived from the iNOS isoform contributes to some of the myocardial injury following MI, possibly by causing myocardial cell death in areas bordering the ischemic region of the heart.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Guanidinas/farmacología , Corazón/efectos de los fármacos , Isotiuronio/análogos & derivados , Infarto del Miocardio/prevención & control , Óxido Nítrico Sintasa/antagonistas & inhibidores , Animales , Animales Recién Nacidos , Northern Blotting , Western Blotting , Muerte Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Estudios de Seguimiento , Interleucina-1/farmacología , Isotiuronio/farmacología , Masculino , Infarto del Miocardio/enzimología , Infarto del Miocardio/patología , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , ARN Mensajero/biosíntesis , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
16.
Am J Physiol Heart Circ Physiol ; 279(2): H719-25, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10924071

RESUMEN

Phorbol-12-myristate- 13-acetate (PMA) has been shown to induce hypertrophy of cardiac myocytes. The prostaglandin endoperoxide H synthase isoform 2 (cyclooxygenase-2, COX-2) has been associated with enhanced growth and/or proliferation of several types of cells. Thus we studied whether PMA induces COX-2 and prostanoid products PGE(2) and PGF(2alpha) in neonatal ventricular myocytes and whether endogenous COX-2 products participate in their growth. In addition, we examined whether PMA affects interleukin-1beta (IL-1beta) stimulation of COX-2 and PGE(2) production. PMA (0.1 micromol/l) stimulated growth, as indicated by a 1.6-fold increase in [(3)H]leucine incorporation. PMA increased COX-2 protein levels 2. 8-fold, PGE(2) 3.7-fold, and PGF(2alpha) 2.9-fold. Inhibition of either p38 kinase or protein kinase C (PKC) prevented PMA-stimulated COX-2. Inhibition of COX-2 with either indomethacin or NS-398 had no effect on PMA-stimulated [(3)H]leucine incorporation. Exogenous administration of PGF(2alpha), but not PGE(2), stimulated protein synthesis. Treatment with IL-1beta (5 ng/ml) increased COX-2 protein levels 42-fold, whereas cotreatment with IL-1beta and PMA stimulated COX-2 protein only 32-fold. IL-1beta did not affect control or PMA-stimulated protein synthesis. These findings indicate that: 1) PMA, acting through PKC and p38 kinase, enhances COX-2 expression, but chronic treatment with PMA partially inhibits IL-1beta stimulation of COX-2; and 2) exogenous PGF(2alpha) is involved in neonatal ventricular myocyte growth but endogenous COX-2 products are not.


Asunto(s)
Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Isoenzimas/biosíntesis , Isoenzimas/genética , Miocardio/enzimología , Prostaglandina-Endoperóxido Sintasas/biosíntesis , Prostaglandina-Endoperóxido Sintasas/genética , Prostaglandinas/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Animales , Animales Recién Nacidos , División Celular/efectos de los fármacos , Células Cultivadas , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa/farmacología , Dinoprost/metabolismo , Dinoprostona/metabolismo , Indometacina/farmacología , Interleucina-1/farmacología , Isoenzimas/metabolismo , Leucina/metabolismo , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Miocardio/citología , Nitrobencenos/farmacología , Prostaglandina-Endoperóxido Sintasas/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Sulfonamidas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos
17.
Hypertension ; 37(2 Pt 2): 478-84, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11230322

RESUMEN

Brain natriuretic peptide (BNP) gene expression accompanies cardiac hypertrophy and heart failure. The vasoconstrictor endothelin-1 (ET) may be involved in the development of these diseases. ET has also been shown to activate phospholipase A(2) (PLA(2)), and the resulting metabolites are important second messengers. We studied how ET and PLA(2) metabolites regulate BNP gene expression. The human BNP (hBNP) promoter (from -1818 to +100) coupled to a luciferase reporter gene was transferred into neonatal ventricular myocytes (NVMs), and luciferase activity was measured as an index of promoter activity. ET induced BNP mRNA in NVMs as assessed by Northern blot. It also stimulated the hBNP promoter, an effect completely inhibited by actinomycin D. To test the involvement of different PLA(2) isoforms, transfected cells were treated with various PLA(2) inhibitors before stimulation with ET. Only Ca(2+)-independent PLA(2) blockade prevented ET-stimulated hBNP promoter activity. The PLA(2) metabolite lysophosphatidic acid (LPA) also activated the hBNP promoter, but arachidonic acid itself did not. ET regulation of the hBNP promoter is pertussis toxin-sensitive. The nonreceptor tyrosine kinase Src and the small GTPase Rac mediate the effects of both ET and LPA in stimulation of the hBNP promoter. We studied the involvement of cis elements in ET-stimulated hBNP promoter activity. Deletion of BNP promoter sequences from -1818 to -408 and from -408 to -40 reduced the effect of ET by 60% and 80%, respectively. Moreover, ET-stimulated luciferase activity was reduced by 50% when the proximal GATA element was mutated. These data suggest that (1) ET activates the hBNP promoter through a transcriptional mechanism; (2) LPA, perhaps generated by iPLA(2), is involved in the effect of ET; (3) Src and Rac mediate ET and LPA stimulation of the hBNP promoter; and (4) ET regulation of the hBNP promoter targets both distal and proximal cis elements.


Asunto(s)
Encéfalo/efectos de los fármacos , Endotelina-1/farmacología , Miocardio/metabolismo , Péptido Natriurético Encefálico/biosíntesis , Ácidos Fosfatidicos/farmacología , Animales , Encéfalo/metabolismo , Enfermedades Cardiovasculares/metabolismo , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Luciferasas/genética , Péptido Natriurético Encefálico/genética , Fosfolipasas A/antagonistas & inhibidores , Fosfolipasas A/metabolismo , Regiones Promotoras Genéticas , Proteínas/farmacología , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Transfección , Disfunción Ventricular Izquierda/metabolismo
18.
Biochemistry ; 25(8): 2094-101, 1986 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-3707935

RESUMEN

Gel-exclusion high-performance liquid chromatography (HPLC) has been used to separate the untransformed from the transformed glucocorticoid receptor (GC-R) extracted from mouse AtT-20 cells. With 200 mM potassium phosphate as the eluent, an efficient separation of the forms of the GC-R is attained in 15-20 min. The untransformed cytosolic GC-R elutes from the column with a Stokes radius (Rs) of 8.2-8.6 nm, as do the molybdate-stabilized GC-R, the purified untransformed GC-R, and the cross-linked cytosolic GC-R. GC-R transformed in vitro by either ammonium sulfate precipitation, KCl treatment, or G-25 chromatography elutes with an Rs of 5.7-6 nm. Also, GC-R extracted from the nucleus with either 0.3 M KCl or 2 mM sodium tungstate, or purified by two cycles of DNA-cellulose chromatography, has an Rs of 5.5-6.3 nm. The data are identical either in the presence or in the absence of 20 mM Na2MoO4, suggesting that molybdate is not causing aggregation to produce a larger Rs value than that of the native receptor. Vertical tube rotor sucrose gradient ultracentrifugation of cytosol produces three forms of the GC-R: 9.1 S, 5.2 S, and 3.8 S. Sequential analysis of the GC-R forms by HPLC and vertical tube rotor ultracentrifugation and vice versa allows for the hydrodynamic determination of molecular weight within a very short time period (2-3 h total).(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Receptores de Glucocorticoides/aislamiento & purificación , Animales , Línea Celular , Centrifugación por Gradiente de Densidad/métodos , Cromatografía en Gel/métodos , Cromatografía Líquida de Alta Presión/métodos , Citosol/metabolismo , Dexametasona/metabolismo , Ratones , Peso Molecular , Neoplasias Hipofisarias/metabolismo , Conformación Proteica , Receptores de Glucocorticoides/metabolismo
19.
Am J Physiol Endocrinol Metab ; 278(6): E1115-23, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10827015

RESUMEN

Brain natriuretic peptide (BNP) gene expression and chronic activation of the sympathetic nervous system are characteristics of the development of heart failure. We studied the role of the beta-adrenergic signaling pathway in regulation of the human BNP (hBNP) promoter. An hBNP promoter (-1818 to +100) coupled to a luciferase reporter gene was transferred into neonatal cardiac myocytes, and luciferase activity was measured as an index of promoter activity. Isoproterenol (ISO), forskolin, and cAMP stimulated the promoter, and the beta(2)-antagonist ICI 118,551 abrogated the effect of ISO. In contrast, the protein kinase A (PKA) inhibitor H-89 failed to block the action of cAMP and ISO. Pertussis toxin (PT), which inactivates Galpha(i), inhibited ISO- and cAMP-stimulated hBNP promoter activity. The Src tyrosine kinase inhibitor PP1 and a dominant-negative mutant of the small G protein Rac also abolished the effect of ISO and cAMP. Finally, we studied the involvement of M-CAT-like binding sites in basal and inducible regulation of the hBNP promoter. Mutation of these elements decreased basal and cAMP-induced activity. These data suggest that beta-adrenergic regulation of hBNP is PKA independent, involves a Galpha(i)-activated pathway, and targets regulatory elements in the proximal BNP promoter.


Asunto(s)
Agonistas Adrenérgicos beta/farmacología , AMP Cíclico/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Isoproterenol/farmacología , Péptido Natriurético Encefálico/genética , Antagonistas Adrenérgicos beta/farmacología , Animales , Animales Recién Nacidos , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Inhibidores Enzimáticos/farmacología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Técnicas de Transferencia de Gen , Humanos , Luciferasas/genética , Miocardio/metabolismo , Regiones Promotoras Genéticas , Ratas , Ratas Sprague-Dawley , Receptores Adrenérgicos beta/fisiología
20.
Am J Physiol Heart Circ Physiol ; 280(1): H368-76, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11123253

RESUMEN

Studies have shown that brain natriuretic peptide (BNP) gene expression is rapidly induced in the infarcted heart and that plasma BNP levels reflect the degree of left ventricular dysfunction. Our previous in vitro work using transiently transfected neonatal rat cardiac myocytes has shown that the human BNP (hBNP) promoter, in particular a region extending from -127 to -40 relative to the start site of transcription, is more active in cardiac myocytes than in fibroblasts. To study tissue-specific and transcriptional regulation of the hBNP gene in vivo, we generated transgenic mice containing the proximal hBNP promoter (-408 to +100) coupled to a luciferase reporter gene. In four lines of transgenic mice, luciferase activity was approximately 33- to 100-fold higher in the heart than in other tissues, including the whole brain. To test whether the transgene responded to a pathophysiological stimulus, we induced infarction by coronary artery ligation. Luciferase activity was fivefold higher in the infarcted region of the left ventricle at 48 h than in sham-operated animals and remained elevated for 4 wk. Endogenous BNP mRNA was similarly increased in the infarcted hearts of a separate group of mice. We conclude that 1) the proximal 408-bp region of the hBNP promoter confers cardiac-specific expression and 2) myocardial infarction activates the proximal hBNP promoter in vivo. These data suggest that we have a valid model for the study of basal and inducible regulation of the hBNP gene in vivo.


Asunto(s)
Péptido Natriurético Encefálico/genética , Regiones Promotoras Genéticas , Animales , Presión Sanguínea , Northern Blotting , Southern Blotting , Corazón , Humanos , Inmunohistoquímica , Luciferasas/biosíntesis , Luciferasas/genética , Ratones , Ratones Transgénicos , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Péptido Natriurético Encefálico/biosíntesis , Transgenes , Ultrasonografía
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda