Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
JACC Heart Fail ; 12(1): 100-113, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37921801

RESUMEN

BACKGROUND: Intracellular second messenger cyclic guanosine monophosphate (cGMP) mediates bioactivity of the natriuretic peptides and nitric oxide, and is key to circulatory homeostasis and protection against cardiovascular disease. Inhibition of cGMP-degrading phosphodiesterases (PDEs) PDE5 and PDE9 are emerging as pharmacological targets in heart failure (HF). OBJECTIVES: The present study investigated dual enhancement of cGMP in experimental HF by combining inhibition of PDE-5 (P5-I) and PDE-9 (P9-I). METHODS: Eight sheep with pacing-induced HF received on separate days intravenous P5-I (sildenafil), P9-I (PF-04749982), P5-I+P9-I, and vehicle control, in counterbalanced order. RESULTS: Compared with control, separate P5-I and P9-I significantly increased circulating cGMP concentrations in association with reductions in mean arterial pressure (MAP), left atrial pressure (LAP), and pulmonary arterial pressure (PAP), with effects of P5-I on cGMP, MAP, and PAP greater than those of P9-I. Only P5-I decreased pulmonary vascular resistance. Combination P5-I+P9-I further reduced MAP, LAP, and PAP relative to inhibition of either phosphodiesterase alone. P9-I and, especially, P5-I elevated urinary cGMP levels relative to control. However, whereas inhibition of either enzyme increased urine creatinine excretion and clearance, only P9-I induced a significant diuresis and natriuresis. Combined P5-I+P9-I further elevated urine cGMP with concomitant increases in urine volume, sodium and creatinine excretion, and clearance similar to P9-I alone, despite the greater MAP reductions induced by combination treatment. CONCLUSIONS: Combined P5-I+P9-I amalgamated the superior renal effects of P9-I and pulmonary effects of P5-1, while concurrently further reducing cardiac preload and afterload. These findings support combination P5-I+P9-I as a therapeutic strategy in HF.


Asunto(s)
Insuficiencia Cardíaca , Humanos , Animales , Ovinos , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/uso terapéutico , Insuficiencia Cardíaca/tratamiento farmacológico , Creatinina , Inhibidores de Fosfodiesterasa/uso terapéutico , Inhibidores de Fosfodiesterasa/farmacología , GMP Cíclico
2.
Int J Mol Sci ; 24(7)2023 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-37047281

RESUMEN

Mass spectrometry is a powerful technique for investigating renal pathologies and identifying biomarkers, and efficient protein extraction from kidney tissue is essential for bottom-up proteomic analyses. Detergent-based strategies aid cell lysis and protein solubilization but are poorly compatible with downstream protein digestion and liquid chromatography-coupled mass spectrometry, requiring additional purification and buffer-exchange steps. This study compares two well-established detergent-based methods for protein extraction (in-solution sodium deoxycholate (SDC); suspension trapping (S-Trap)) with the recently developed sample preparation by easy extraction and digestion (SPEED) method, which uses strong acid for denaturation. We compared the quantitative performance of each method using label-free mass spectrometry in both sheep kidney cortical tissue and plasma. In kidney tissue, SPEED quantified the most unique proteins (SPEED 1250; S-Trap 1202; SDC 1197). In plasma, S-Trap produced the most unique protein quantifications (S-Trap 150; SDC 148; SPEED 137). Protein quantifications were reproducible across biological replicates in both tissue (R2 = 0.85-0.90) and plasma (SPEED R2 = 0.84; SDC R2 = 0.76, S-Trap R2 = 0.65). Our data suggest SPEED as the optimal method for proteomic preparation in kidney tissue and S-Trap or SPEED as the optimal method for plasma, depending on whether a higher number of protein quantifications or greater reproducibility is desired.


Asunto(s)
Detergentes , Espectrometría de Masas en Tándem , Animales , Ovinos , Detergentes/química , Espectrometría de Masas en Tándem/métodos , Proteómica/métodos , Reproducibilidad de los Resultados , Proteínas
3.
JACC Heart Fail ; 11(2): 227-239, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36752488

RESUMEN

BACKGROUND: The natriuretic peptides (NPs) are potent natriuretic/diuretic and vasodilatory factors, and augmentation of their levels or signaling via inhibition of the enzymes neprilysin (NEP) and phosphodiesterase 9 (PDE9), respectively, has beneficial actions in heart failure (HF). OBJECTIVES: The authors investigated dual enhancement of NP bioactivity by combining PDE9 inhibition and NEP inhibition in HF using an ovine model. METHODS: Eight sheep with pacing-induced HF received on 4 separate days intravenous PDE9 inhibition (PF-04749982), NEP inhibition (SCH-32615), PDE9 inhibition + NEP inhibition (PI+NI), and vehicle control treatment. RESULTS: Compared with the control treatment, NEP inhibition significantly increased plasma NP concentrations with a corresponding rise in second messenger cyclic guanosine monophosphate (cGMP), whereas PDE9 inhibition increased circulating cGMP with a negligible effect on NP levels. Combined PI+NI elevated plasma NPs to an extent comparable to that seen with NEP inhibition alone but further increased cGMP, resulting in a rise in the cGMP-to-NP ratio. All active treatments reduced mean arterial pressure, left atrial pressure, pulmonary arterial pressure, and peripheral resistance, with combined PI+NI further reducing mean arterial pressure and left atrial pressure relative to either inhibitor separately. Active treatments increased urine volume and sodium, potassium and creatinine excretion, and creatinine clearance, in association with rises in urine cGMP levels. PI+NI induced a significantly greater natriuresis and increase in urinary cGMP relative to either inhibitor singly. CONCLUSIONS: The present study demonstrates for the first time that combined PI+NI has additional beneficial hemodynamic and renal effects when compared with either PDE9 inhibition or NEP inhibition alone. The superior efficacy of this 2-pronged augmentation of NP bioactivity supports PI+NI as a potential therapeutic strategy for HF.


Asunto(s)
Insuficiencia Cardíaca , Animales , Ovinos , Humanos , Neprilisina , Hidrolasas Diéster Fosfóricas/uso terapéutico , Creatinina , Factor Natriurético Atrial , Péptidos Natriuréticos , Vasodilatadores/uso terapéutico , GMP Cíclico , Diuréticos/uso terapéutico
4.
Int J Mol Sci ; 23(2)2022 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-35055195

RESUMEN

One-quarter of patients with acute decompensated heart failure (ADHF) experience acute kidney injury (AKI)-an abrupt reduction or loss of kidney function associated with increased long-term mortality. There is a critical need to identify early and real-time markers of AKI in ADHF; however, to date, no protein biomarkers have exhibited sufficient diagnostic or prognostic performance for widespread clinical uptake. We aimed to identify novel protein biomarkers of AKI associated with ADHF by quantifying changes in protein abundance in the kidneys that occur during ADHF development and recovery in an ovine model. Relative quantitative protein profiling was performed using sequential window acquisition of all theoretical fragment ion spectra-mass spectrometry (SWATH-MS) in kidney cortices from control sheep (n = 5), sheep with established rapid-pacing-induced ADHF (n = 8), and sheep after ~4 weeks recovery from ADHF (n = 7). Of the 790 proteins quantified, we identified 17 candidate kidney injury markers in ADHF, 1 potential kidney marker of ADHF recovery, and 2 potential markers of long-term renal impairment (differential abundance between groups of 1.2-2.6-fold, adjusted p < 0.05). Among these 20 candidate protein markers of kidney injury were 6 candidates supported by existing evidence and 14 novel candidates not previously implicated in AKI. Proteins of differential abundance were enriched in pro-inflammatory signalling pathways: glycoprotein VI (activated during ADHF development; adjusted p < 0.01) and acute phase response (repressed during recovery from ADHF; adjusted p < 0.01). New biomarkers for the early detection of AKI in ADHF may help us to evaluate effective treatment strategies to prevent mortality and improve outcomes for patients.


Asunto(s)
Lesión Renal Aguda/diagnóstico , Biomarcadores/metabolismo , Insuficiencia Cardíaca/metabolismo , Proteómica/métodos , Lesión Renal Aguda/sangre , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/orina , Animales , Biomarcadores/sangre , Biomarcadores/orina , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/sangre , Insuficiencia Cardíaca/complicaciones , Insuficiencia Cardíaca/orina , Humanos , Glicoproteínas de Membrana Plaquetaria/metabolismo , Glicoproteínas de Membrana Plaquetaria/orina , Pronóstico , Ovinos
5.
J Am Heart Assoc ; 10(18): e021312, 2021 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-34533033

RESUMEN

BACKGROUND Acute decompensated heart failure (ADHF) is associated with deterioration in renal function-an important risk factor for poor outcomes. Whether ADHF results in permanent kidney damage/dysfunction is unknown. METHODS AND RESULTS We investigated for the first time the renal responses to the development of, and recovery from, ADHF using an ovine model. ADHF development induced pronounced hemodynamic changes, neurohormonal activation, and decline in renal function, including decreased urine, sodium and urea excretion, and creatinine clearance. Following ADHF recovery (25 days), creatinine clearance reductions persisted. Kidney biopsies taken during ADHF and following recovery showed widespread mesangial cell prominence, early mild acute tubular injury, and medullary/interstitial fibrosis. Renal transcriptomes identified altered expression of 270 genes following ADHF development and 631 genes following recovery. A total of 47 genes remained altered post-recovery. Pathway analysis suggested gene expression changes, driven by a network of inflammatory cytokines centered on IL-1ß (interleukin 1ß), lead to repression of reno-protective eNOS (endothelial nitric oxide synthase) signaling during ADHF development, and following recovery, activation of glomerulosclerosis and reno-protective pathways and repression of proinflammatory/fibrotic pathways. A total of 31 dysregulated genes encoding proteins detectable in urine, serum, and plasma identified potential candidate markers for kidney repair (including CNGA3 [cyclic nucleotide gated channel subunit alpha 3] and OIT3 [oncoprotein induced transcript 3]) or long-term renal impairment in ADHF (including ACTG2 [actin gamma 2, smooth muscle] and ANGPTL4 [angiopoietin like 4]). CONCLUSIONS In an ovine model, we provide the first direct evidence that an episode of ADHF leads to an immediate decline in kidney function that failed to fully resolve after ≈4 weeks and is associated with persistent functional/structural kidney injury. We identified molecular pathways underlying kidney injury and repair in ADHF and highlighted 31 novel candidate biomarkers for acute kidney injury in this setting.


Asunto(s)
Lesión Renal Aguda , Insuficiencia Cardíaca , Lesión Renal Aguda/genética , Animales , Biomarcadores , Creatinina , Insuficiencia Cardíaca/genética , Riñón/fisiología , Ovinos , Oveja Doméstica , Transcriptoma
6.
Cardiovasc Res ; 117(2): 508-519, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-32167565

RESUMEN

AIMS: Management of acute decompensated heart failure (ADHF) requires disparate treatments depending on the state of systemic/peripheral perfusion and the presence/absence of expanded body-fluid volumes. There is an unmet need for therapeutics that differentially treat each aspect. Atrial natriuretic peptide (ANP) plays an important role in blood pressure and volume regulation. We investigate for the first time the integrated haemodynamic, endocrine and renal effects of human ANP analogues, modified for exclusive vasodilatory (ANP-DRD) or diuretic (ANP-DGD) activities, in normal health and experimental ADHF. METHODS AND RESULTS: We compared the effects of incremental infusions of ANP analogues ANP-DRD and ANP-DGD with native ANP, in normal (n = 8) and ADHF (n = 8) sheep. ANP-DRD administration increased plasma cyclic guanosine monophosphate (cGMP) in association with dose-dependent reductions in arterial pressure in normal and heart failure (HF) sheep similarly to ANP responses. In contrast to ANP, which in HF produced a diuresis/natriuresis, this analogue was without significant renal effect. Conversely, ANP-DGD induced marked stepwise increases in urinary cGMP, urine volume, and sodium excretion in HF comparable to ANP, but without accompanying vasodilatory effects. All peptides increased packed cell volume relative to control in both states, and in HF, decreased left atrial pressure. In response to ANP-DRD-induced blood pressure reductions, plasma renin activity rose compared to control only during the high dose in normals, and not at all in HF-suggesting relative renin inhibition, with no increase in aldosterone in either state, whereas renin and aldosterone were both significantly reduced by ANP-DGD in HF. CONCLUSION: These ANP analogues exhibit distinct vasodilatory (ANP-DRD) and diuretic/natriuretic (ANP-DGD) activities, and therefore have the potential to provide precision therapy for ADHF patients with differing pathophysiological derangement of pressure-volume homeostasis.


Asunto(s)
Factor Natriurético Atrial/farmacología , Diuresis/efectos de los fármacos , Diuréticos/farmacología , Insuficiencia Cardíaca/tratamiento farmacológico , Hemodinámica/efectos de los fármacos , Riñón/efectos de los fármacos , Vasodilatadores/farmacología , Función Ventricular Izquierda/efectos de los fármacos , Aldosterona/sangre , Animales , Factor Natriurético Atrial/sangre , GMP Cíclico/sangre , Modelos Animales de Enfermedad , Endotelina-1/sangre , Femenino , Insuficiencia Cardíaca/sangre , Insuficiencia Cardíaca/fisiopatología , Riñón/metabolismo , Riñón/fisiopatología , Natriuresis/efectos de los fármacos , Péptido Natriurético Encefálico/sangre , Renina/sangre , Oveja Doméstica , Vasodilatación/efectos de los fármacos
7.
Animals (Basel) ; 10(10)2020 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-33080942

RESUMEN

Heart failure (HF) is the final common end point of multiple metabolic and cardiovascular diseases and imposes a significant health care burden worldwide. Despite significant improvements in clinical management and outcomes, morbidity and mortality remain high and there remains an indisputable need for improved treatment options. The pathophysiology of HF is complex and covers a spectrum of clinical presentations from HF with reduced ejection fraction (HFrEF) (≤40% EF) through to HF with preserved EF (HFpEF), with HFpEF patients demonstrating a reduced ability of the heart to relax despite an EF maintained above 50%. Prior to the last decade, the majority of clinical trials and animal models addressed HFrEF. Despite growing efforts recently to understand underlying mechanisms of HFpEF and find effective therapies for its treatment, clinical trials in patients with HFpEF have failed to demonstrate improvements in mortality. A significant obstacle to therapeutic innovation in HFpEF is the absence of preclinical models including large animal models which, unlike rodents, permit detailed instrumentation and extensive imaging and sampling protocols. Although several large animal models of HFpEF have been reported, none fulfil all the features present in human disease and few demonstrate progression to frank decompensated HF. This review summarizes well-established models of HFrEF in pigs, dogs and sheep and discusses attempts to date to model HFpEF in these species.

8.
J Am Coll Cardiol ; 74(7): 889-901, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31416533

RESUMEN

BACKGROUND: Phosphodiesterase-9 (PDE9) reduces natriuretic peptide (NP) signaling and may be involved in the pathophysiology of heart failure (HF). OBJECTIVES: This study investigated for the first time the integrated hemodynamic, endocrine, and renal effects of phosphodiesterase-9 inhibition (PDE9-I). METHODS: A total of 8 normal sheep and 8 sheep with pacing-induced HF received incremental intravenous boluses of PDE9-I (30, 100, and 300 mg PF-04749982 at 1-h intervals). RESULTS: PDE9-I dose-dependently increased plasma cyclic guanosine monophosphate (cGMP) in normal sheep (p < 0.05) while concurrently reducing circulating atrial natriuretic peptide levels (p < 0.01). Similar trends were evident in HF, resulting in significant elevations in the cGMP/NP ratio in both states (p < 0.001 and p < 0.05, respectively). PDE9-I also produced progressive falls in arterial pressure (HF: p < 0.001), atrial pressure (Normal: p < 0.001; HF: p < 0.001), and peripheral resistance (HF: p < 0.001), and transiently increased cardiac output at the top dose (Normal: p < 0.05; HF: p < 0.001). Inhibition of PDE9 had a negligible effect on circulating hormones at the lower doses, but post-high dose, acutely increased renin activity (Normal: p < 0.001; HF: p < 0.05), vasopressin (Normal: p < 0.001; HF: p < 0.01), and cyclic adenosine monophosphate (HF: p < 0.001). Plasma aldosterone increased briefly after high-dose PDE9-I in normal sheep, and fell following the top dose in HF. PDE9-I dose-dependently increased urinary cGMP in both states (both p < 0.001). In HF, this was associated with increases in urine volume (p < 0.01), sodium excretion (p < 0.01), and creatinine clearance (p < 0.001). CONCLUSIONS: PDE9-I improves NP efficacy in conjunction with beneficial hemodynamic and renal effects in experimental HF. These results support a role for PDE9 in HF pathophysiology and suggest its inhibition may constitute a novel therapeutic approach to this disease.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , Insuficiencia Cardíaca/tratamiento farmacológico , Inhibidores de Fosfodiesterasa/administración & dosificación , Adenosina Monofosfato/sangre , Aldosterona/sangre , Animales , Factor Natriurético Atrial/sangre , Presión Atrial/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Gasto Cardíaco/efectos de los fármacos , Creatinina/orina , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Guanosina Monofosfato/sangre , Guanosina Monofosfato/orina , Renina/sangre , Ovinos , Sodio/orina , Orina , Resistencia Vascular/efectos de los fármacos , Vasopresinas/sangre
9.
In Vivo ; 32(5): 1071-1080, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30150428

RESUMEN

BACKGROUND/AIM: Patients with breast cancer and metabolic syndrome have poorer outcomes. We aimed to develop and characterise an apolipoprotein E-null/aromatase knockout (ApoE-/-/ArKO) mouse model of breast cancer with metabolic syndrome to aid research of the mechanisms behind poor prognosis. MATERIALS AND METHODS: Wild-type, ApoE-/- and ApoE-/-/ArKO mice were orthotopically implanted with EO771 murine breast cancer cells. Tumour growth was monitored and tumours investigated for pathological features such as cancer-associated adipocytes, hypoxia and cancer cell proliferation. RESULTS: Tumours from ApoE-/-/ArKO mice were significantly more proliferative than those from wild-type mice (p=0.003), and exhibited reduced expression of insulin-like growth factor binding protein-5 (p=0.002). However, ApoE-/-/ArKO mice also had a reduced rate of metastasis compared to wild-type and ApoE-/- mice. Tumour hypoxia and the number of cancer-associated adipocytes did not differ. CONCLUSION: The ApoE-/-/ArKO model with EO771 breast cancer provides a novel mouse model to investigate the effects of metabolic syndrome on aspects of breast tumour biology.


Asunto(s)
Neoplasias de la Mama/complicaciones , Modelos Animales de Enfermedad , Síndrome Metabólico/complicaciones , Síndrome Metabólico/metabolismo , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aromatasa/deficiencia , Aromatasa/genética , Biomarcadores , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Femenino , Hipoxia/genética , Hipoxia/metabolismo , Inmunohistoquímica , Síndrome Metabólico/genética , Ratones , Ratones Noqueados
10.
Nutrients ; 9(7)2017 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-28686204

RESUMEN

There is extensive evidence that walnut consumption is protective against cardiovascular disease and diabetes in the healthy population, but the beneficial effects of walnut consumption in individuals with the metabolic syndrome (MetS) remain uncertain. We compared a range of cardio-metabolic traits and related tissue gene expression associated with 21 weeks of dietary walnut supplementation in a mouse model of MetS (MetS-Tg) and wild-type (WT) mice (n = 10 per genotype per diet, equal males and females). Compared to standard diet, walnuts did not significantly alter food consumption or body weight trajectory of either MetS-Tg or WT mice. In MetS-Tg mice, walnuts were associated with reductions in oral glucose area under the curve (gAUC, standard diet 1455 ± 54, walnut 1146 ± 91, p = 0.006) and mean arterial blood pressure (MAP, standard diet 100.6 ± 1.9, walnut 73.2 ± 1.8 mmHg, p < 0.001), with neutral effects on gAUC and MAP in WT mice. However, in MetS-Tg mice, walnuts were also associated with trends for higher plasma cholesterol (standard diet 4.73 ± 0.18, walnut 7.03 ± 1.99 mmol/L, p = 0.140) and triglyceride levels (standard diet 2.4 ± 0.5, walnut 5.4 ± 1.6 mmol/L, p = 0.061), despite lowering cholesterol and having no effect on triglycerides in WT mice. Moreover, in both MetS-Tg and WT mice, walnuts were associated with significantly increased liver expression of genes associated with metabolism (Fabp1, Insr), cell stress (Atf6, Ddit3, Eif2ak3), fibrosis (Hgf, Sp1, Timp1) and inflammation (Tnf, Ptpn22, Pparg). In conclusion, dietary walnuts were associated with modest favourable effects in WT mice, but a combination of beneficial and adverse effects in MetS-Tg mice, and up-regulation of hepatic pro-fibrotic and pro-inflammatory genes in both mouse strains.


Asunto(s)
Juglans/química , Síndrome Metabólico/fisiopatología , Nueces/química , Extractos Vegetales/administración & dosificación , Extractos Vegetales/efectos adversos , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aromatasa/deficiencia , Aromatasa/genética , Presión Sanguínea/efectos de los fármacos , Cruzamientos Genéticos , Suplementos Dietéticos , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Lípidos/sangre , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Síndrome Metabólico/tratamiento farmacológico , Síndrome Metabólico/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos/genética
11.
J Cardiovasc Pharmacol ; 65(3): 269-75, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25658462

RESUMEN

The impact of chronic urocortin 2 (Ucn2) treatment after myocardial infarction (MI) has not previously been investigated. In this study, we examined the effects of 30-day Ucn2 administration (415 µg·kg·d SC per day) in mice post-MI. Compared with surgical sham + vehicle controls (n = 10), MI + vehicle animals (n = 10) after 30 days demonstrated decreased ejection fraction (75.6 ± 1.2 vs. 43.6% ± 0.8%, P < 0.001) and fractional shortening (38.20 ± 0.83 vs. 18.4% ± 0.54%, P < 0.001) in association with increased heart weight-to-body weight ratio (4.57 ± 0.25 vs. 5.29 ± 0.18, P < 0.01), left ventricular (LV) mass (91 ± 7 vs. 126 ± 8 mg, P < 0.01), LV internal diameters at both systole (1.91 ± 0.14 vs. 3.45 ± 0.09 mm, P < 0.001) and diastole (3.14 ± 0.15 vs. 4.25 ± 0.10 mm, P < 0.001), LV end systolic volumes (0.02 ± 0.01 vs. 0.11 ± 0.01 mL, P < 0.001), and ventricular collagen 1 and ß-myosin heavy chain gene expression. Compared with MI + vehicle mice, MI + Ucn2 animals (n = 10) exhibited significantly reduced infarct size (4.00 ± 0.39 vs. 1.83 ± 0.44 mm, P < 0.01), heart weight-to-body weight ratio (4.75 ± 0.19, P = 0.06), LV mass (101 ± 6 mg, P < 0.01), LV internal diameters (systole 2.61 ± 0.09 mm, P < 0.001; diastole 3.78 ± 0.09 mm, P < 0.001), and end systolic volumes (0.14 ± 0.02 mL, P < 0.01) in conjunction with improved ejection fraction (65.2% ± 0.9%, P < 0.001) and fractional shortening (18.4 ± 0.5 vs. 30.5% ± 0.5%, P < 0.001). Ucn2 treatment also decreased collagen 1 and ß-myosin heavy chain expression. In conclusion, chronic Ucn2 treatment significantly improves cardiovascular function and attenuates cardiac injury and remodeling in experimental MI.


Asunto(s)
Cardiotónicos/farmacología , Hipertrofia Ventricular Izquierda/prevención & control , Infarto del Miocardio/tratamiento farmacológico , Urocortinas/farmacología , Disfunción Ventricular Izquierda/prevención & control , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos , Animales , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Regulación de la Expresión Génica , Hemodinámica/efectos de los fármacos , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/metabolismo , Hipertrofia Ventricular Izquierda/patología , Hipertrofia Ventricular Izquierda/fisiopatología , Masculino , Ratones Endogámicos C57BL , Contracción Miocárdica/efectos de los fármacos , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Miocardio/patología , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , ARN Mensajero/metabolismo , Recuperación de la Función , Volumen Sistólico/efectos de los fármacos , Factores de Tiempo , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/metabolismo , Disfunción Ventricular Izquierda/patología , Disfunción Ventricular Izquierda/fisiopatología
12.
Am J Physiol Endocrinol Metab ; 302(5): E576-84, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22185842

RESUMEN

The aim of this study was to create a comprehensive mouse model of the metabolic syndrome by crossing aromatase-deficient (ArKO) mice with apolipoprotein E-deficient (ApoE(-/-)) mice. Successive crossbreeding of ArKO with ApoE(-/-)-deficient mice generated double knockout, MetS-Tg mice. The phenotypic characteristics of the MetS-Tg mice were assessed at 3, 6, and 12 mo of age and compared with age- and sex-matched wild-type (WT) controls. Blood pressure and heart rate were recorded by a noninvasive, computerized tail-cuff system. Oral glucose and intraperitoneal insulin tolerance tests were performed. Serum cholesterol levels were measured by a combined quantitative colorimetric assay. Plasma adiponectin, C-reactive protein (CRP), insulin, interleukin-6 (IL-6), leptin, resistin, and tumor necrosis factor-α (TNF-α) were measured by multiplexed ELISA. MetS-Tg mice displayed significantly increased body weight, central obesity, and elevated blood pressure at all three ages compared with WT mice. Elevated serum cholesterol was associated with higher triglycerides and LDL/VLDL cholesterol particles and was accompanied by a decrease in HDL and histological evidence of fatty liver. MetS-Tg mice of all ages showed impaired glucose tolerance. At 12 mo, MetS-Tg mice had elevated plasma levels of CRP, IL-6, leptin, and TNF-α, but resistin levels were largely unchanged. We now report that this combination of gene knockouts produces a novel strain of mice that display the diverse clinical features of the metabolic syndrome, including central obesity, progressive hypertension, an adverse serum lipid profile, fatty liver, glucose intolerance, insulin resistance, and evidence of an inflammatory state.


Asunto(s)
Apolipoproteínas E/fisiología , Aromatasa/fisiología , Modelos Animales de Enfermedad , Síndrome Metabólico/fisiopatología , Tejido Adiposo Blanco/inmunología , Tejido Adiposo Blanco/patología , Animales , Aorta/inmunología , Aorta/patología , Apolipoproteínas E/genética , Aromatasa/genética , Aterosclerosis/etiología , Proteína C-Reactiva/análisis , Cruzamientos Genéticos , Citocinas/sangre , Hígado Graso/etiología , Femenino , Intolerancia a la Glucosa/etiología , Hiperlipidemias/etiología , Hipertensión/etiología , Resistencia a la Insulina , Hígado/inmunología , Hígado/patología , Masculino , Síndrome Metabólico/sangre , Síndrome Metabólico/inmunología , Síndrome Metabólico/patología , Ratones , Ratones Noqueados , Obesidad Abdominal/etiología
13.
Biochim Biophys Acta ; 1792(12): 1175-84, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19782130

RESUMEN

The heart adapts to an increased workload through the activation of a hypertrophic response within the cardiac ventricles. This response is characterized by both an increase in the size of the individual cardiomyocytes and an induction of a panel of genes normally expressed in the embryonic and neonatal ventricle, such as atrial natriuretic peptide (ANP). ANP and brain natriuretic peptide (BNP) exert their biological actions through activation of the natriuretic peptide receptor-1 (Npr1). The current study examined mice lacking Npr1 (Npr1(-/-)) activity and investigated the effects of the absence of Npr1 signaling during cardiac development on embryo viability, cardiac structure and gene and protein expression. Npr1(-/-)embryos were collected at embryonic day (ED) 12.5, 15.5 and neonatal day 1 (ND 1). Npr1(-/-)embryos occurred at the expected Mendelian frequency at ED 12.5, but knockout numbers were significantly decreased at ED 15.5 and ND 1. There was no indication of cardiac structural abnormalities in surviving embryos. However, Npr1(-/-)embryos exhibited cardiac enlargement (without fibrosis) from ED 15.5 as well as significantly increased ANP mRNA and protein expression compared to wild-type (WT) mice, but no concomitant increase in expression of the hypertrophy-related transcription factors, Mef2A, Mef2C, GATA-4, GATA-6 or serum response factor (SRF). However, there was a significant decrease in Connexin-43 (Cx43) gene and protein expression at mid-gestation in Npr1(-/-)embryos. Our findings suggest that the mechanism by which natriuretic peptide signaling influences cardiac development in Npr1(-/-) mice is distinct from that seen during the development of pathological cardiac hypertrophy and fibrosis. The decreased viability of Npr1(-/-)embryos may result from a combination of cardiomegaly and dysregulated Cx43 protein affecting cardiac contractility.


Asunto(s)
Cardiomegalia/mortalidad , Cardiomegalia/patología , Embrión de Mamíferos/citología , Corazón/embriología , Receptores del Factor Natriurético Atrial/fisiología , Animales , Factor Natriurético Atrial/genética , Factor Natriurético Atrial/metabolismo , Presión Sanguínea , Western Blotting , Cardiomegalia/metabolismo , Conexina 43/metabolismo , Embrión de Mamíferos/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Corazón/fisiología , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones Noqueados , Péptido Natriurético Encefálico/genética , Péptido Natriurético Encefálico/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tasa de Supervivencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
14.
Endocrinology ; 149(11): 5828-34, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18653707

RESUMEN

After myocardial infarction (MI), the heart may undergo progressive ventricular remodeling, resulting in a deterioration of cardiac function. TGF-beta is a key cytokine that both initiates and terminates tissue repair, and its sustained production underlies the development of tissue fibrosis, particularly after MI. We investigated the effects of a novel orally active specific inhibitor of the TGF-beta receptor 1 (SD-208) in an experimental model of MI. Mice underwent ligation of the left coronary artery to induce MI and were subsequently treated for 30 d after infarction with either SD-208 or a vehicle control. Blockade of TGF-beta signaling reduced mean arterial pressure in all groups. SD-208 treatment after MI resulted in a trend for reduced ventricular and renal gene expression of TGF-beta-activated kinase-1 (a downstream modulator of TGF-beta signaling) and a significant decrease in collagen 1, in association with a marked decrease in cardiac mass. Post-MI SD-208 treatment significantly reduced circulating levels of plasma renin activity as well as down-regulating the components of the cardiac and renal renin-angiotensin system (angiotensinogen, angiotensin converting enzyme, and angiotensin II type I receptor). Our findings indicate that blockade of the TGF-beta signaling pathway results in significant amelioration of deleterious cardiac remodeling after infarction.


Asunto(s)
Infarto del Miocardio/fisiopatología , Pteridinas/farmacología , Sistema Renina-Angiotensina/efectos de los fármacos , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Remodelación Ventricular/efectos de los fármacos , Animales , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/patología , Regulación hacia Abajo/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Regulación de la Expresión Génica/efectos de los fármacos , Corazón/efectos de los fármacos , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Miocardio/patología , Pteridinas/uso terapéutico , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Renina/sangre , Renina/metabolismo , Sistema Renina-Angiotensina/fisiología , Factor de Crecimiento Transformador beta/fisiología
15.
J Mol Endocrinol ; 38(1-2): 245-57, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17293444

RESUMEN

The natriuretic peptides, atrial (ANP) and brain natriuretic peptide (BNP) are known to suppress cardiac hypertrophy and fibrosis. Both ANP and BNP exert their bioactivities through the Npr1 receptor, and Npr1 knockout mice (Npr1-/-) exhibit marked cardiac hypertrophy and fibrosis. In this study, we investigated which genes within the hypertrophic and fibrotic pathways are influenced by the lack of Npr1 signalling. cDNA microarray and quantitative real-time PCR (RT-PCR) analyses were performed on cardiac ventricles from Npr1-/-mice. Gene expression at early and late stages during development of hypertrophy was investigated in male and female Npr1-/-mice at 8 weeks and 6 months of age. Heart weight to body weight ratios (HW:BW) were maximally increased in 8-week males (P<0 x 01), whilst HW:BW in females continued to increase progressively up to 6 months (P<0 x 01). This was despite blood pressure being similarly elevated at both the ages in male and female knockout when compared with wild-type (WT) mice (P<0 x 001). Microarray analysis identified altered gene expression at the earliest steps in the hypertrophy-signalling cascade in Npr1-/- mice, particularly calcium-calmodulin signalling and ion channels, with subsequent changes in the expression of intracellular messengers including protein kinases and transcription factors. Real-time PCR analysis confirmed significant differences in gene expression of ANP, BNP, calmodulin 1, histone deacetylase 7a (HDAC7a), protein kinase C (PKC)iota, (GATA) 4, collagen 1, phospholamban and transforming growth factor-beta1 in Npr1-/- mice when compared with WT (P<0 x 05). The present study implicates the calmodulin-CaMK-Hdac-Mef2 and PKC-MAPK-GATA4 pathways in Npr1 mediation of cardiac hypertrophy.


Asunto(s)
Cardiomegalia/genética , Fibrosis/genética , Regulación de la Expresión Génica/fisiología , Guanilato Ciclasa/fisiología , Miocardio/patología , Receptores del Factor Natriurético Atrial/fisiología , Animales , Cardiomegalia/metabolismo , Fibrosis/metabolismo , Ratones , Ratones Noqueados , Miocardio/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA