Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
FEBS Lett ; 596(1): 17-28, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34778969

RESUMEN

Phosphodiesterase 5 inhibition (PDE5i) activates cGMP-dependent protein kinase (PKG) and ameliorates heart failure; however, its impact on cardiac mitochondrial regulation has not been fully determined. Here, we investigated the role of the mitochondrial regulator peroxisome proliferator-activated receptor γ co-activator-1α (PGC1α) in the PDE5i-conferred cardioprotection, utilizing PGC1α null mice. In PGC1α+/+ hearts exposed to 7 weeks of pressure overload by transverse aortic constriction, chronic treatment with the PDE5 inhibitor sildenafil improved cardiac function and remodeling, with improved mitochondrial respiration and upregulation of PGC1α mRNA in the myocardium. By contrast, PDE5i-elicited benefits were abrogated in PGC1α-/- hearts. In cultured cardiomyocytes, PKG overexpression induced PGC1α, while inhibition of the transcription factor CREB abrogated the PGC1α induction. Together, these results suggest that the PKG-PGC1α axis plays a pivotal role in the therapeutic efficacy of PDE5i in heart failure.


Asunto(s)
Inhibidores de Fosfodiesterasa 5
2.
Circ Heart Fail ; 14(1): e007300, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33464954

RESUMEN

BACKGROUND: Augmentation of NP (natriuretic peptide) receptor and cyclic guanosine monophosphate (cGMP) signaling has emerged as a therapeutic strategy in heart failure (HF). cGMP-specific PDE9 (phosphodiesterase 9) inhibition increases cGMP signaling and attenuates stress-induced hypertrophic heart disease in preclinical studies. A novel cGMP-specific PDE9 inhibitor, CRD-733, is currently being advanced in human clinical studies. Here, we explore the effects of chronic PDE9 inhibition with CRD-733 in the mouse transverse aortic constriction pressure overload HF model. METHODS: Adult male C57BL/6J mice were subjected to transverse aortic constriction and developed significant left ventricular (LV) hypertrophy after 7 days (P<0.001). Mice then received daily treatment with CRD-733 (600 mg/kg per day; n=10) or vehicle (n=17), alongside sham-operated controls (n=10). RESULTS: CRD-733 treatment reversed existing LV hypertrophy compared with vehicle (P<0.001), significantly improved LV ejection fraction (P=0.009), and attenuated left atrial dilation (P<0.001), as assessed by serial echocardiography. CRD-733 prevented elevations in LV end diastolic pressures (P=0.037) compared with vehicle, while lung weights, a surrogate for pulmonary edema, were reduced to sham levels. Chronic CRD-733 treatment increased plasma cGMP levels compared with vehicle (P<0.001), alongside increased phosphorylation of Ser273 of cardiac myosin binding protein-C, a cGMP-dependent protein kinase I phosphorylation site. CONCLUSIONS: The PDE9 inhibitor, CRD-733, improves key hallmarks of HF including LV hypertrophy, LV dysfunction, left atrial dilation, and pulmonary edema after pressure overload in the mouse transverse aortic constriction HF model. Additionally, elevated plasma cGMP may be used as a biomarker of target engagement. These findings support future investigation into the therapeutic potential of CRD-733 in human HF.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , Insuficiencia Cardíaca/fisiopatología , Corazón/efectos de los fármacos , Hipertrofia Ventricular Izquierda/fisiopatología , Inhibidores de Fosfodiesterasa/farmacología , Volumen Sistólico/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos , Animales , Aorta/cirugía , Proteínas Portadoras/efectos de los fármacos , Proteínas Portadoras/metabolismo , Colágeno/efectos de los fármacos , Colágeno/metabolismo , Constricción Patológica , GMP Cíclico/sangre , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/efectos de los fármacos , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , Fibrosis , Corazón/fisiopatología , Atrios Cardíacos/efectos de los fármacos , Insuficiencia Cardíaca/patología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Hipertrofia Ventricular Izquierda/patología , Pulmón/efectos de los fármacos , Masculino , Ratones , Tamaño de los Órganos , Fosforilación/efectos de los fármacos , Edema Pulmonar/fisiopatología
3.
J Clin Invest ; 124(6): 2464-71, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24837433

RESUMEN

Inhibition of cGMP-specific phosphodiesterase 5 (PDE5) ameliorates pathological cardiac remodeling and has been gaining attention as a potential therapy for heart failure. Despite promising results in males, the efficacy of the PDE5 inhibitor sildenafil in female cardiac pathologies has not been determined and might be affected by estrogen levels, given the hormone's involvement in cGMP synthesis. Here, we determined that the heart-protective effect of sildenafil in female mice depends on the presence of estrogen via a mechanism that involves myocyte eNOS-dependent cGMP synthesis and the cGMP-dependent protein kinase Iα (PKGIα). Sildenafil treatment failed to exert antiremodeling properties in female pathological hearts from Gαq-overexpressing or pressure-overloaded mice after ovary removal; however, estrogen replacement restored the effectiveness of sildenafil in these animals. In females, sildenafil-elicited myocardial PKG activity required estrogen, which stimulated tonic cardiomyocyte cGMP synthesis via an eNOS/soluble guanylate cyclase pathway. In contrast, eNOS activation, cGMP synthesis, and sildenafil efficacy were not estrogen dependent in male hearts. Estrogen and sildenafil had no impact on pressure-overloaded hearts from animals expressing dysfunctional PKGIα, indicating that PKGIα mediates antiremodeling effects. These results support the importance of sex differences in the use of PDE5 inhibitors for treating heart disease and the critical role of estrogen status when these agents are used in females.


Asunto(s)
Estrógenos/metabolismo , Cardiopatías/tratamiento farmacológico , Cardiopatías/metabolismo , Inhibidores de Fosfodiesterasa 5/farmacología , Animales , Cardiotónicos/farmacología , GMP Cíclico/biosíntesis , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , Modelos Animales de Enfermedad , Estradiol/administración & dosificación , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Guanilato Ciclasa/metabolismo , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo III/deficiencia , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Ovariectomía , Piperazinas/farmacología , Purinas/farmacología , Receptores del Factor Natriurético Atrial/deficiencia , Receptores del Factor Natriurético Atrial/genética , Receptores del Factor Natriurético Atrial/metabolismo , Caracteres Sexuales , Citrato de Sildenafil , Sulfonas/farmacología , Resultado del Tratamiento
4.
J Biol Chem ; 288(34): 24972-83, 2013 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-23831687

RESUMEN

NO, via its second messenger cGMP, activates protein kinase GI (PKGI) to induce vascular smooth muscle cell relaxation. The mechanisms by which PKGI kinase activity regulates cardiovascular function remain incompletely understood. Therefore, to identify novel protein kinase G substrates in vascular cells, a λ phage coronary artery smooth muscle cell library was constructed and screened for phosphorylation by PKGI. The screen identified steroid-sensitive gene 1 (SSG1), which harbors several predicted PKGI phosphorylation sites. We observed direct and cGMP-regulated interaction between PKGI and SSG1. In cultured vascular smooth muscle cells, both the NO donor S-nitrosocysteine and atrial natriuretic peptide induced SSG1 phosphorylation, and mutation of SSG1 at each of the two predicted PKGI phosphorylation sites completely abolished its basal phosphorylation by PKGI. We detected high SSG1 expression in cardiovascular tissues. Finally, we found that activation of PKGI with cGMP regulated SSG1 intracellular distribution.


Asunto(s)
Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , GMP Cíclico/metabolismo , Regulación de la Expresión Génica/fisiología , Glicoproteínas/biosíntesis , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas Supresoras de Tumor/biosíntesis , Células Cultivadas , GMP Cíclico/genética , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/genética , Cisteína/análogos & derivados , Cisteína/farmacología , Proteínas de la Matriz Extracelular , Regulación de la Expresión Génica/efectos de los fármacos , Glicoproteínas/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Donantes de Óxido Nítrico/farmacología , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , S-Nitrosotioles/farmacología , Proteínas Supresoras de Tumor/genética
5.
J Gerontol A Biol Sci Med Sci ; 68(11): 1351-5, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23657971

RESUMEN

Hypertensive heart disease causes significant mortality in older patients, yet there is an incomplete understanding of molecular mechanisms that regulate age-dependent hypertensive left ventricular hypertrophy (LVH). Therefore, we tested the hypothesis that the cGMP-dependent protein kinase G I alpha (PKGIα) attenuates hypertensive LVH by evaluating the cardiac phenotype in mice with selective mutations of the PKGIα leucine zipper domain. These leucine zipper mutant (LZM) mice develop basal hypertension. Compared with wild-type controls, 8-month-old adult LZM mice developed increased left ventricular end-diastolic pressure but without frank LVH. In advanced age (15 months), the LZM mice developed overt pathological LVH. These findings reveal a role of PKGIα in normally attenuating hypertensive LVH. Therefore, mutation of the PKGIα LZ domain produces a clinically relevant model for hypertensive heart disease of aging.


Asunto(s)
Proteína Quinasa Dependiente de GMP Cíclico Tipo I/fisiología , Hipertensión/etiología , Hipertrofia Ventricular Izquierda/etiología , Leucina Zippers/fisiología , Factores de Edad , Animales , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/genética , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Mutación , Función Ventricular Izquierda
6.
Int J Cardiol ; 167(5): 2114-9, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22748498

RESUMEN

BACKGROUND AND OBJECTIVE: Nitric oxide (NO) and related nitrovasodilators regulate blood pressure by activation of soluble guanylate cyclase, elevation of cyclic guanosine monophosphate (cGMP), and activation of cGMP-dependent protein kinase (cGPK). Despite the progress of our understanding of the NO/cGMP mediated vasorelaxation, partly through conventional cGPK knock-out mice, the role of cGPK remains unclear. In particular, the downstream target(s) of the kinase are not well defined. We hypothesized that highly selective inhibitors of cGPK delivered in vivo in adult may elucidate the role of the kinase in vasorelaxation and regulation of blood pressure. METHODS AND RESULTS: We have adopted a newly developed method of TAT-mediated protein transduction to study NO/cGMP signaling pathways in mice. In vitro, TAT-cGPK inhibitor peptide blocked autophosphorylation of the kinase. The effect of cGPK inhibition on murine blood pressure (BP) was investigated by continuous infusion of 100 µg of the inhibitor into the internal jugular vein over 72 hours. In 8 animals infused with the inhibitor, the mean BP increased by 38 ± 24/31 ± 30 mm Hg (from 108 ± 14/92 ± 19 to 145 ± 13/123 ± 19 mm Hg) whereas in 8 animals injected with either saline (4) or TAT-green fluorescent protein (4), the BP remained the same (from 117 ± 21/101 ± 26 to 119 ± 22/96 ± 30 mm Hg); P=0.001. Ex vivo, using vascular ring assays, NO-dependent relaxation in murine aortas harvested from animals administered with TAT-cGPK inhibitor was inhibited by 25% (sham 76 ± 11%, inhibitor 51 ± 13%). CONCLUSION: We demonstrated that highly specific peptide inhibitor of cGPK induced adult murine hypertension through inhibition of nitric oxide mediated relaxation.


Asunto(s)
Péptidos de Penetración Celular/administración & dosificación , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/antagonistas & inhibidores , Sistemas de Liberación de Medicamentos/métodos , Endotelio Vascular/efectos de los fármacos , Productos del Gen tat/administración & dosificación , Hipertensión/tratamiento farmacológico , Secuencia de Aminoácidos , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/enzimología , Secuencia de Bases , Péptidos de Penetración Celular/genética , Células Cultivadas , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , Endotelio Vascular/enzimología , Endotelio Vascular/fisiopatología , Femenino , Productos del Gen tat/genética , Hipertensión/enzimología , Hipertensión/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Técnicas de Cultivo de Órganos
7.
J Biol Chem ; 287(49): 41342-51, 2012 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-23066013

RESUMEN

Vascular smooth muscle cell (VSMC) tone is regulated by the state of myosin light chain (MLC) phosphorylation, which is in turn regulated by the balance between MLC kinase and MLC phosphatase (MLCP) activities. RhoA activates Rho kinase, which phosphorylates the regulatory subunit of MLC phosphatase, thereby inhibiting MLC phosphatase activity and increasing contraction and vascular tone. Nitric oxide is an important mediator of VSMC relaxation and vasodilation, which acts by increasing cyclic GMP (cGMP) levels in VSMC, thereby activating cGMP-dependent protein kinase Iα (PKGIα). PKGI is known to phosphorylate Rho kinase, preventing Rho-mediated inhibition of MLC phosphatase, promoting vasorelaxation, although the molecular mechanisms that mediate this are unclear. Here we identify RhoA as a target of activated PKGIα and show further that PKGIα binds directly to RhoA, inhibiting its activation and translocation. In protein pulldown and immunoprecipitation experiments, binding of RhoA and PKGIα was demonstrated via a direct interaction between the amino terminus of RhoA (residues 1-44), containing the switch I domain of RhoA, and the amino terminus of PKGIα (residues 1-59), which includes a leucine zipper heptad repeat motif. Affinity assays using cGMP-immobilized agarose showed that only activated PKGIα binds RhoA, and a leucine zipper mutant PKGIα was unable to bind RhoA even if activated. Furthermore, a catalytically inactive mutant of PKGIα bound RhoA but did not prevent RhoA activation and translocation. Collectively, these results support that RhoA is a PKGIα target and that direct binding of activated PKGIα to RhoA is central to cGMP-mediated inhibition of the VSMC Rho kinase contractile pathway.


Asunto(s)
Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , Regulación de la Expresión Génica , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Células 3T3 , Secuencias de Aminoácidos , Animales , Aorta/metabolismo , Células COS , Chlorocebus aethiops , Células Endoteliales/citología , Humanos , Hipertensión/metabolismo , Ratones , Modelos Biológicos , Unión Proteica , Transducción de Señal , Fracciones Subcelulares/metabolismo , Quinasas Asociadas a rho/metabolismo
8.
Nat Med ; 18(9): 1429-33, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22922412

RESUMEN

Hypertension is a cardiovascular risk factor present in over two-thirds of people over age 60 in North America; elevated blood pressure correlates with increased risk of heart attack, stroke and progression to heart and kidney failure. Current therapies are insufficient to control blood pressure in almost half of these patients. The mineralocorticoid receptor (MR), acting in the kidney, is known to regulate blood pressure through aldosterone binding and stimulation of sodium retention. However, recent studies support the concept that the MR also has extrarenal actions and that defects in sodium handling alone do not fully explain the development of hypertension and associated cardiovascular mortality. We and others have identified functional MR in human vascular smooth muscle cells (SMCs), suggesting that vascular MR might directly regulate blood pressure. Here we show that mice with SMC-specific deficiency of the MR have decreased blood pressure as they age without defects in renal sodium handling or vascular structure. Aged mice lacking MR in SMCs (SMC-MR) have reduced vascular myogenic tone, agonist-dependent contraction and expression and activity of L-type calcium channels. Moreover, SMC-MR contributes to angiotensin II­induced vascular oxidative stress, vascular contraction and hypertension. This study identifies a new role for vascular MR in blood pressure control and in vascular aging and supports the emerging hypothesis that vascular tone contributes directly to systemic blood pressure.


Asunto(s)
Envejecimiento/fisiología , Presión Sanguínea/fisiología , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/metabolismo , Receptores de Mineralocorticoides/metabolismo , Animales , Canales de Calcio Tipo L/metabolismo , Cartilla de ADN/genética , Galactósidos , Corazón/anatomía & histología , Indoles , Ratones , Contracción Muscular/fisiología , Tamaño de los Órganos/fisiología , Reacción en Cadena de la Polimerasa , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Mineralocorticoides/deficiencia , Sodio/metabolismo
9.
Circulation ; 125(22): 2728-38, 2012 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-22592898

RESUMEN

BACKGROUND: Heart failure is a major cause of morbidity and mortality worldwide. The ubiquitously expressed cytokine transforming growth factor-ß1 (TGFß1) promotes cardiac fibrosis, an important component of progressive heart failure. Membrane-associated endoglin is a coreceptor for TGFß1 signaling and has been studied in vascular remodeling and preeclampsia. We hypothesized that reduced endoglin expression may limit cardiac fibrosis in heart failure. METHODS AND RESULTS: We first report that endoglin expression is increased in the left ventricle of human subjects with heart failure and determined that endoglin is required for TGFß1 signaling in human cardiac fibroblasts using neutralizing antibodies and an siRNA approach. We further identified that reduced endoglin expression attenuates cardiac fibrosis, preserves left ventricular function, and improves survival in a mouse model of pressure-overload-induced heart failure. Prior studies have shown that the extracellular domain of endoglin can be cleaved and released into the circulation as soluble endoglin, which disrupts TGFß1 signaling in endothelium. We now demonstrate that soluble endoglin limits TGFß1 signaling and type I collagen synthesis in cardiac fibroblasts and further show that soluble endoglin treatment attenuates cardiac fibrosis in an in vivo model of heart failure. CONCLUSION: Our results identify endoglin as a critical component of TGFß1 signaling in the cardiac fibroblast and show that targeting endoglin attenuates cardiac fibrosis, thereby providing a potentially novel therapeutic approach for individuals with heart failure.


Asunto(s)
Antígenos CD/metabolismo , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/mortalidad , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Miocardio/metabolismo , Miocardio/patología , Receptores de Superficie Celular/metabolismo , Animales , Anticuerpos/farmacología , Colágeno Tipo I/metabolismo , Modelos Animales de Enfermedad , Endoglina , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Insuficiencia Cardíaca/fisiopatología , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Interferente Pequeño/farmacología , Transducción de Señal/fisiología , Tasa de Supervivencia , Factor de Crecimiento Transformador beta1/metabolismo
10.
J Am Heart Assoc ; 1(5): e003731, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23316302

RESUMEN

BACKGROUND: Cyclic GMP (cGMP) signaling attenuates cardiac remodeling, but it is unclear which cGMP effectors mediate these effects and thus might serve as novel therapeutic targets. Therefore, we tested whether the cGMP downstream effector, cGMP-dependent protein kinase G Iα (PKGIα), attenuates pressure overload-induced remodeling in vivo. METHODS AND RESULTS: The effect of transaortic constriction (TAC)-induced left ventricular (LV) pressure overload was examined in mice with selective mutations in the PKGIα leucine zipper interaction domain. Compared with wild-type littermate controls, in response to TAC, these Leucine Zipper Mutant (LZM) mice developed significant LV systolic and diastolic dysfunction by 48 hours (n=6 WT sham, 6 WT TAC, 5 LZM sham, 9 LZM TAC). In response to 7-day TAC, the LZM mice developed increased pathologic hypertrophy compared with controls (n=5 WT sham, 4 LZM sham, 8 WT TAC, 11 LZM TAC). In WT mice, but not in LZM mice, phosphodiesterase 5 (PDE5) inhibition with sildenafil (Sil) significantly inhibited TAC-induced cardiac hypertrophy and LV systolic dysfunction in WT mice, but this was abolished in the LZM mice (n=3 WT sham, 4 LZM sham, 3 WT TAC vehicle, 6 LZM TAC vehicle, 4 WT TAC Sil, 6 LZM TAC Sil). And in response to prolonged, 21-day TAC (n=8 WT sham, 7 LZM sham, 21 WT TAC, 15 LZM TAC), the LZM mice developed markedly accelerated mortality and congestive heart failure. TAC induced activation of JNK, which inhibits cardiac remodeling in vivo, in WT, but not in LZM, hearts, identifying a novel signaling pathway activated by PKGIα in the heart in response to LV pressure overload. CONCLUSIONS: These findings reveal direct roles for PKGIα in attenuating pressure overload-induced remodeling in vivo and as a required effector for the cardioprotective effects of sildenafil.


Asunto(s)
Cardiotónicos/administración & dosificación , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , Insuficiencia Cardíaca/enzimología , Corazón/fisiopatología , Inhibidores de Fosfodiesterasa/administración & dosificación , Piperazinas/administración & dosificación , Sulfonas/administración & dosificación , Disfunción Ventricular Izquierda/enzimología , Remodelación Ventricular/fisiología , Animales , Ecocardiografía , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/fisiopatología , Immunoblotting , Masculino , Ratones , Ratones Endogámicos C57BL , Purinas/administración & dosificación , Transducción de Señal , Citrato de Sildenafil , Disfunción Ventricular Izquierda/tratamiento farmacológico , Disfunción Ventricular Izquierda/fisiopatología , Remodelación Ventricular/efectos de los fármacos
11.
Arterioscler Thromb Vasc Biol ; 31(8): 1871-80, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21617142

RESUMEN

OBJECTIVE: Aldosterone (Aldo) antagonism prevents cardiovascular mortality by unclear mechanisms. Aldo binds to the mineralocorticoid receptor (MR), a ligand-activated transcription factor, which is expressed in human vascular cells. Here we define the early Aldo-regulated vascular transcriptome and investigate the mechanisms of gene regulation by Aldo in the vasculature that may contribute to vascular disease. METHODS AND RESULTS: Gene expression profiling of Aldo-treated mouse aortas identified 72 genes regulated by Aldo. These genes are overrepresented in Gene Ontology categories involved in vascular function and disease. Quantitative reverse transcription-polymerase chain reaction was used to confirm and further explore mechanisms of vascular gene regulation by Aldo. Aldo-regulated vascular gene expression was inhibited by actinomycin D and MR antagonists supporting a transcriptional MR-dependent mechanism. Aldo regulation of a subset of genes was enhanced in the setting of vascular endothelial denudation and blocked by the free radical scavenger Tempol, supporting synergy between Aldo and vascular injury that is oxidative stress dependent. In the aortic arch, a region predisposed to atherosclerosis, the injury-enhanced genes also demonstrated enhanced expression compared with the descending aorta, both at baseline and after Aldo exposure. Furthermore, the clinically beneficial MR antagonist spironolactone inhibited expression of the identified genes in aortic tissue from humans with atherosclerosis. CONCLUSIONS: This study defines the Aldo-regulated vascular transcriptome and characterizes a subset of proatherogenic genes with enhanced Aldo-stimulated, oxidative stress-dependent expression in the setting of vascular injury and in areas predisposed to atherosclerosis. Inhibition of MR regulation of these genes may play a role in the protective effects of Aldo antagonists in patients with vascular disease, and these pathways may provide novel drug targets to prevent atherosclerosis in humans.


Asunto(s)
Aldosterona/farmacología , Aorta/efectos de los fármacos , Aorta/metabolismo , Animales , Aterosclerosis/etiología , Aterosclerosis/genética , Aterosclerosis/metabolismo , Células Cultivadas , Dactinomicina/farmacología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/lesiones , Endotelio Vascular/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Antagonistas de Receptores de Mineralocorticoides , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Estrés Oxidativo/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Espironolactona/farmacología
12.
Arterioscler Thromb Vasc Biol ; 31(2): 443-50, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21071694

RESUMEN

OBJECTIVE: Early recognition of an acute coronary occlusion (ACO) improves clinical outcomes. Soluble fms-like tyrosine kinase-1 (sFLT1) is an endothelium-derived protein induced by hypoxia. We tested whether sFLT1 levels are elevated in ACO. METHODS AND RESULTS: Serum sFLT1 levels were measured by enzyme-linked immunosorbent assay in patients with ST-segment elevations and angiographically confirmed ACO, unstable angina/non ST-segment elevation myocardial infarction, and 2 control groups. To further explore sFLT1 release, a mouse model of ACO and in vitro human coronary artery endothelial cell injury were used. sFLT1 levels were increased in ACO compared with unstable angina/non-ST-elevation myocardial infarction, catheterized controls, or healthy volunteers (200.7±15.5 versus 70.7±44.0 versus 10.2±4.0 versus 11.7±1.7 pg/mL respectively, P<0.001 versus ACO). At presentation, all ACO patients had elevated sFLT1 levels (>15 pg/mL, 99th percentile in controls), whereas 57% had levels of the MB isoform of creatine kinase levels >10 ng/mL (P<0.01) and 85% had ultrasensitive troponin I levels >0.05 ng/mL (P<0.05). Within 60 minutes after symptom onset, sFLT1 was more sensitive than the MB isoform of creatine kinase or ultrasensitive troponin I for ACO (100% versus 20% versus 20% respectively; P≤0.01 for each). Within 60 minutes of ACO in mice, sFLT1 levels were elevated. Hypoxia and thrombin increased sFLT1 levels within 15 minutes in human coronary artery endothelial cells. CONCLUSIONS: sFLT1 levels may be an early indicator of endothelial hypoxia in ACO.


Asunto(s)
Oclusión Coronaria/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Enfermedad Aguda , Anciano , Animales , Estudios de Casos y Controles , Hipoxia de la Célula/fisiología , Células Cultivadas , Creatina Quinasa/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Modelos Animales , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Factores de Tiempo
13.
Arterioscler Thromb Vasc Biol ; 31(1): 50-7, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20966403

RESUMEN

OBJECTIVE: Atherosclerosis encompasses a conspicuously maladaptive inflammatory response that might involve innate immunity. Here, we compared the role of Toll-like receptor 4 (TLR4) with that of TLR2 in intimal foam cell accumulation and inflammation in apolipoprotein E (ApoE) knockout (KO) mice in vivo and determined potential mechanisms of upstream activation and downstream action. METHODS AND RESULTS: We measured lipid accumulation and gene expression in the lesion-prone lesser curvature of the aortic arch. TLR4 deficiency reduced intimal lipid by ≈75% in ApoE KO mice, despite unaltered total serum cholesterol and triglyceride levels, whereas TLR2 deficiency reduced it by ≈45%. TLR4 deficiency prevented the increased interleukin-1α (IL-1α) and monocyte chemoattractant protein-1 mRNA levels seen within lesional tissue, and it also lowered serum IL-1α levels. Smooth muscle cells (SMC) were present within the intima of the lesser curvature of the aortic arch at this early lesion stage, and they enveloped and permeated nascent lesions, which consisted of focal clusters of foam cells. Cholesterol enrichment of SMC in vitro stimulated acyl-coenzyme A:cholesterol acyltransferase-1 mRNA expression, cytoplasmic cholesterol ester accumulation, and monocyte chemoattractant protein-1 mRNA and protein expression in a TLR4-dependent manner. CONCLUSIONS: TLR4 contributes to early-stage intimal foam cell accumulation at lesion-prone aortic sites in ApoE KO mice, as does TLR2 to a lesser extent. Intimal SMC surround and penetrate early lesions, where TLR4 signaling within them may influence lesion progression.


Asunto(s)
Enfermedades de la Aorta/metabolismo , Apolipoproteínas E/deficiencia , Aterosclerosis/metabolismo , Células Espumosas/metabolismo , Transducción de Señal , Receptor Toll-Like 4/metabolismo , Acetil-CoA C-Acetiltransferasa/metabolismo , Animales , Aorta Torácica/metabolismo , Aorta Torácica/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/patología , Quimiocina CCL2/genética , Colesterol/sangre , Modelos Animales de Enfermedad , Femenino , Células Espumosas/patología , Regulación de la Expresión Génica , Mediadores de Inflamación/metabolismo , Interleucina-1alfa/sangre , Interleucina-1alfa/genética , Masculino , Ratones , Ratones Noqueados , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , ARN Mensajero/metabolismo , Factores de Tiempo , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética , Triglicéridos/sangre , Molécula 1 de Adhesión Celular Vascular/genética
14.
Am J Cardiol ; 106(12): 1770-6, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21126621

RESUMEN

Progressive left ventricular (LV) dysfunction induces expression of the cytokine transforming growth factor-ß1. Endoglin (CD105) is a transforming growth factor-ß1 co-receptor that is released into the circulation as soluble endoglin (sEng). The objective of the present study was to assess the serum levels of sEng in patients with heart failure and to identify the predictive value of sEng for detecting elevated left ventricular end-diastolic pressures (LVEDPs). We measured the sEng levels in 82 consecutive patients with suspected LV dysfunction referred for determination of left heart filling pressures using cardiac catheterization. Among these subjects, the sEng levels correlated with the LVEDP (R = 0.689; p <0.0001), irrespective of the LV ejection fraction. Using a receiving operating characteristic curve, the sEng levels predicted an LVEDP of ≥16 mm Hg with an area under the curve of 0.85, exceeding the measured area under the curves for both atrial and brain natriuretic peptide, currently used biomarkers for heart failure diagnosis (atrial natriuretic peptide 0.68 and brain natriuretic peptide 0.65; p <0.01 vs sEng). In 10 subjects receiving medical therapy guided by invasive hemodynamic monitoring for heart failure, decreased a pulmonary capillary wedge pressure was associated with a reduced sEng level (R = 0.75, p = 0.008). Finally, compared to 25 healthy controls, the sEng levels were elevated in subjects with suspected LV dysfunction (3,589 ± 588 vs 4,257 ± 966 pg/ml, respectively, p <0.005) and correlated directly with the New York Heart Association class (R = 0.501, p<0.001). In conclusion, circulating levels of sEng are elevated in patients with increased LVEDP and New York Heart Association class, irrespective of the LV ejection fraction. sEng levels also decreased in association with a reduced cardiac filling pressure after diuresis. These findings have identified circulating sEng as a sensitive measure of elevated left heart filling pressures.


Asunto(s)
Antígenos CD , Insuficiencia Cardíaca/diagnóstico , Función Ventricular Izquierda/fisiología , Presión Ventricular/fisiología , Antígenos CD/administración & dosificación , Antígenos CD/sangre , Cateterismo Cardíaco , Progresión de la Enfermedad , Endoglina , Femenino , Estudios de Seguimiento , Insuficiencia Cardíaca/sangre , Insuficiencia Cardíaca/fisiopatología , Humanos , Inyecciones Intravenosas , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Receptores de Superficie Celular/administración & dosificación , Receptores de Superficie Celular/sangre , Reproducibilidad de los Resultados , Índice de Severidad de la Enfermedad , Volumen Sistólico
15.
J Clin Invest ; 120(11): 3891-900, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20921624

RESUMEN

In clinical trials, aldosterone antagonists reduce cardiovascular ischemia and mortality by unknown mechanisms. Aldosterone is a steroid hormone that signals through renal mineralocorticoid receptors (MRs) to regulate blood pressure. MRs are expressed and regulate gene transcription in human vascular cells, suggesting that aldosterone might have direct vascular effects. Using gene expression profiling, we identify the pro-proliferative VEGF family member placental growth factor (PGF) as an aldosterone-regulated vascular MR target gene in mice and humans. Aldosterone-activated vascular MR stimulated Pgf gene transcription and increased PGF protein expression and secretion in the mouse vasculature. In mouse vessels with endothelial damage and human vessels from patients with atherosclerosis, aldosterone enhanced expression of PGF and its receptor, FMS-like tyrosine kinase 1 (Flt1). In atherosclerotic human vessels, MR antagonists inhibited PGF expression. In vivo, aldosterone infusion augmented vascular remodeling in mouse carotids following wire injury, an effect that was lost in Pgf-/- mice. In summary, we have identified PGF as what we believe to be a novel downstream target of vascular MR that mediates aldosterone augmentation of vascular injury. These findings suggest a non-renal mechanism for the vascular protective effects of aldosterone antagonists in humans and support targeting the vascular aldosterone/MR/PGF/Flt1 pathway as a therapeutic strategy for ischemic cardiovascular disease.


Asunto(s)
Aldosterona/metabolismo , Aldosterona/farmacología , Vasos Sanguíneos , Proteínas Gestacionales/metabolismo , Enfermedades Vasculares/patología , Animales , Aterosclerosis/metabolismo , Aterosclerosis/patología , Vasos Sanguíneos/citología , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/patología , Ensayos Clínicos como Asunto , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Antagonistas de Receptores de Mineralocorticoides/farmacología , Antagonistas de Receptores de Mineralocorticoides/uso terapéutico , Factor de Crecimiento Placentario , Proteínas Gestacionales/genética , Receptores de Mineralocorticoides/genética , Receptores de Mineralocorticoides/metabolismo , Transducción de Señal/efectos de los fármacos , Enfermedades Vasculares/metabolismo , Enfermedades Vasculares/terapia , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
16.
J Am Soc Nephrol ; 21(11): 1847-51, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20847141

RESUMEN

G protein-coupled receptors (GPCRs) have key roles in cardiovascular regulation and are important targets for the treatment of hypertension. GTPase-activating proteins, such as RGS2, modulate downstream signaling by GPCRs. RGS2 displays regulatory selectivity for the Gαq subclass of G proteins, and mice lacking RGS2 develop hypertension through incompletely understood mechanisms. Using total body RGS2-deficient mice, we used a kidney crosstransplantation strategy to examine separately the contributions of RGS2 actions in the kidney from those in extrarenal tissues with regard to BP regulation. Loss of renal RGS2 was sufficient to cause hypertension, whereas the absence of RGS2 from all extrarenal tissues including the peripheral vasculature did not significantly alter BP. Accordingly, these results suggest that RGS2 acts within the kidney to modulate BP and prevent hypertension. These data support a critical role for the renal epithelium and/or vasculature as the final determinants of the intra-arterial pressure in hypertension.


Asunto(s)
Presión Sanguínea/fisiología , Riñón/fisiología , Proteínas RGS/fisiología , Animales , Modelos Animales de Enfermedad , Hipertensión/fisiopatología , Trasplante de Riñón/fisiología , Masculino , Ratones , Ratones Noqueados , Proteínas RGS/genética , Receptores Acoplados a Proteínas G/fisiología
17.
J Clin Invest ; 120(7): 2277-9, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20577045

RESUMEN

Estrogen receptors are best known as ligand-activated transcription factors that regulate vascular cell gene expression. For many years now, a rapid signaling pathway mediated by cell membrane-associated estrogen receptors also has been recognized, but the physiological relevance of this pathway has remained unclear. In this issue of the JCI, Chambliss et al. provide new data to indicate that activation of non-nuclear estrogen receptor signaling regulates processes central to cardiovascular health and disease. These investigators show that an estrogen-dendrimer conjugate (EDC), which activates estrogen receptors but remains non-nuclear, stimulates vascular EC migration in vitro and protects against vascular injury in vivo. They show further that the vascular benefits of EDC in vivo occur selectively in the vasculature, without stimulating the uterus or enhancing growth of breast cancer xenografts. Taken together, these findings indicate that activation of non-nuclear estrogen receptor signaling regulates vascular events of physiological relevance and suggest that translation of these findings into clinically relevant therapeutic interventions is a logical next goal.


Asunto(s)
Receptores de Estrógenos/metabolismo , Transducción de Señal/fisiología , Vasos Sanguíneos/metabolismo , Neoplasias de la Mama/metabolismo , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Estrógenos/metabolismo , Femenino , Expresión Génica , Humanos , Ligandos , Receptores Citoplasmáticos y Nucleares/metabolismo
18.
BMC Genet ; 11: 22, 2010 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-20398283

RESUMEN

BACKGROUND: Sequencing of the human genome has identified numerous chromosome copy number additions and subtractions that include stable partial gene duplications and pseudogenes that when not properly annotated can interfere with genetic analysis. As an example of this problem, an evolutionary chromosome event in the primate ancestral chromosome 18 produced a partial duplication and inversion of rho-associated protein kinase 1 (ROCK1 -18q11.1, 33 exons) in the subtelomeric region of the p arm of chromosome 18 detectable only in humans. ROCK1 and the partial gene copy, which the gene databases also currently call ROCK1, include non-unique single nucleotide polymorphisms (SNPs). RESULTS: Here, we characterize this partial gene copy of the human ROCK1, termed Little ROCK, located at 18p11.32. Little ROCK includes five exons, four of which share 99% identity with the terminal four exons of ROCK1 and one of which is unique to Little ROCK. In human while ROCK1 is expressed in many organs, Little ROCK expression is restricted to vascular smooth muscle cell (VSMC) lines and organs rich in smooth muscle. The single nucleotide polymorphism database (dbSNP) lists multiple variants contained in the region shared by ROCK1 and Little ROCK. Using gene and cDNA sequence analysis we clarified the origins of two non-synonymous SNPs annotated in the genome to actually be fixed differences between the ROCK1 and the Little ROCK gene sequences. Two additional coding SNPs were valid polymorphisms selectively within Little ROCK. Little ROCK-Green Fluorescent fusion proteins were highly unstable and degraded by the ubiquitin-proteasome system in vitro. CONCLUSION: In this report we have characterized Little ROCK (ROCK1P1), a human expressed pseudogene derived from partial duplication of ROCK1. The large number of pseudogenes in the human genome creates significant genetic diversity. Our findings emphasize the importance of taking into consideration pseudogenes in all candidate gene and genome-wide association studies, as well as the need for complete annotation of human pseudogenome.


Asunto(s)
Músculo Liso/metabolismo , Polimorfismo de Nucleótido Simple , Seudogenes , Quinasas Asociadas a rho/genética , Secuencia de Bases , Cromosomas Humanos Par 18 , Duplicación de Gen , Humanos , Datos de Secuencia Molecular , Músculo Liso Vascular/metabolismo , Alineación de Secuencia , Quinasas Asociadas a rho/metabolismo
19.
PLoS One ; 5(1): e8830, 2010 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-20098615

RESUMEN

Bicuspid Aortic Valve (BAV) is a highly heritable congenital heart defect. The low frequency of BAV (1% of general population) limits our ability to perform genome-wide association studies. We present the application of four a priori SNP selection techniques, reducing the multiple-testing penalty by restricting analysis to SNPs relevant to BAV in a genome-wide SNP dataset from a cohort of 68 BAV probands and 830 control subjects. Two knowledge-based approaches, CANDID and STRING, were used to systematically identify BAV genes, and their SNPs, from the published literature, microarray expression studies and a genome scan. We additionally tested Functionally Interpolating SNPs (fitSNPs) present on the array; the fourth consisted of SNPs selected by Random Forests, a machine learning approach. These approaches reduced the multiple testing penalty by lowering the fraction of the genome probed to 0.19% of the total, while increasing the likelihood of studying SNPs within relevant BAV genes and pathways. Three loci were identified by CANDID, STRING, and fitSNPS. A haplotype within the AXIN1-PDIA2 locus (p-value of 2.926x10(-06)) and a haplotype within the Endoglin gene (p-value of 5.881x10(-04)) were found to be strongly associated with BAV. The Random Forests approach identified a SNP on chromosome 3 in association with BAV (p-value 5.061x10(-06)). The results presented here support an important role for genetic variants in BAV and provide support for additional studies in well-powered cohorts. Further, these studies demonstrate that leveraging existing expression and genomic data in the context of GWAS studies can identify biologically relevant genes and pathways associated with a congenital heart defect.


Asunto(s)
Antígenos CD/genética , Válvula Aórtica/anomalías , Redes Reguladoras de Genes , Haplotipos , Cardiopatías Congénitas/genética , Proteína Disulfuro Isomerasas/genética , Receptores de Superficie Celular/genética , Proteínas Represoras/genética , Proteína Axina , Estudios de Casos y Controles , Endoglina , Estudio de Asociación del Genoma Completo , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Polimorfismo de Nucleótido Simple
20.
Nat Rev Cardiol ; 6(8): 532-42, 2009 08.
Artículo en Inglés | MEDLINE | ID: mdl-19564884

RESUMEN

The higher incidence of cardiovascular disease in men than in women of similar age, and the menopause-associated increase in cardiovascular disease in women, has led to speculation that gender-related differences in sex hormones have a key role in the development and evolution of cardiovascular disease. Compelling data have indicated that sex differences in vascular biology are determined not only by gender-related differences in sex steroid levels, but also by gender-specific tissue and cellular differences that mediate sex-specific responses. In this Review, we describe the sex-specific effects of estrogen and testosterone on cardiovascular risk, direct vascular effects of these sex hormones, and how these effects influence development of atherosclerosis. Cardiovascular effects of exogenous hormone administration are also discussed. Importantly, evidence has indicated that estrogens alone or in combination with progestins in postmenopausal women increase cardiovascular risk if started late after menopause, but that it possibly has beneficial cardiovascular effects in younger postmenopausal women, although data on long-term testosterone therapy are lacking. Hormone therapy should not be considered solely for primary prevention or treatment of cardiovascular disease at this time.


Asunto(s)
Aterosclerosis/fisiopatología , Estrógenos/fisiología , Hormonas Esteroides Gonadales/fisiología , Testosterona/fisiología , Factores de Edad , Estrógenos/administración & dosificación , Femenino , Hormonas Esteroides Gonadales/administración & dosificación , Humanos , Masculino , Factores de Riesgo , Factores Sexuales , Testosterona/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA