Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Circ Res ; 124(10): 1462-1472, 2019 05 10.
Article in English | MEDLINE | ID: mdl-30929579

ABSTRACT

RATIONALE: Acute kidney injury (AKI) has a high prevalence and mortality in critically ill patients. It is also a powerful risk factor for heart failure incidence driven by hemodynamic changes and neurohormonal activation. However, no drugs have been approved by the Food and Drug Administration. Endogenous pGC-A (particulate guanylyl cyclase A receptor) activators were reported to preserve renal function and improve mortality in AKI patients, although hypotension accompanied by pGC-A activators have limited their therapeutic potential. OBJECTIVE: We investigated the therapeutic potential of a nonhypotensive pGC-A activator/designer natriuretic peptide, CRRL269, in a short-term, large animal model of ischemia-induced AKI and also investigated the potential of uCNP (urinary C-type natriuretic peptide) as a biomarker for AKI. METHODS AND RESULTS: We first showed that CRRL269 stimulated cGMP generation, suppressed plasma angiotensin II, and reduced cardiac filling pressures without lowering blood pressure in the AKI canine model. We also demonstrated that CRRL269 preserved glomerular filtration rate, increased renal blood flow, and promoted diuresis and natriuresis. Further, CRRL269 reduced kidney injury and apoptosis as evidenced by ex vivo histology and tissue apoptosis analysis. We also showed, compared with native pGC-A activators, that CRRL269 is a more potent inhibitor of apoptosis in renal cells and induced less decreases in intracellular Ca2+ concentration in vascular smooth muscle cells. The renal antiapoptotic effects were at least mediated by cGMP/PKG pathway. Further, CRRL269 inhibited proapoptotic genes expression using a polymerase chain reaction gene array. Additionally, we demonstrated that AKI increased uCNP levels. CONCLUSIONS: Our study supports developing CRRL269 as a novel renocardiac protective agent for AKI treatment.


Subject(s)
Acute Kidney Injury/drug therapy , Acute Kidney Injury/urine , Natriuretic Peptide, C-Type/urine , Natriuretic Peptides/therapeutic use , Renal Agents/therapeutic use , Acute Kidney Injury/chemically induced , Acute Kidney Injury/prevention & control , Angiotensin II/blood , Animals , Apoptosis/drug effects , Biomarkers/urine , Blood Pressure/physiology , Cyclic GMP/biosynthesis , Diuresis/drug effects , Dogs , Glomerular Filtration Rate/drug effects , Male , Natriuresis/drug effects , Natriuretic Peptides/pharmacology , Receptors, Atrial Natriuretic Factor/analysis , Receptors, Atrial Natriuretic Factor/drug effects , Renal Circulation/drug effects
2.
Hypertension ; 73(4): 900-909, 2019 04.
Article in English | MEDLINE | ID: mdl-30798663

ABSTRACT

Despite optimal current therapies, cardiovascular disease remains the leading cause for death worldwide. Importantly, advances in peptide engineering have accelerated the development of innovative therapeutics for diverse human disease states. Additionally, the advancement of bispecific therapeutics targeting >1 signaling pathway represents a highly innovative strategy for the treatment of cardiovascular disease. We, therefore, engineered a novel, designer peptide, which simultaneously targets the pGC-A (particulate guanylyl cyclase A) receptor and the MasR (Mas receptor), potentially representing an attractive cardiorenoprotective therapeutic for cardiovascular disease. We engineered a novel, bispecific receptor activator, NPA7, that represents the fusion of a 22-amino acid sequence of BNP (B-type natriuretic peptide; an endogenous ligand of pGC-A) with Ang 1-7 (angiotensin 1-7)-the 7-amino acid endogenous activator of MasR. We assessed NPA7's dual receptor activating actions in vitro (second messenger production and receptor interaction). Further, we performed an intravenous peptide infusion comparison study in normal canines to study its biological actions in vivo, including in the presence of an MasR antagonist. Our in vivo and in vitro studies demonstrate the successful synthesis of NPA7 as a bispecific receptor activator targeting pGC-A and MasR. In normal canines, NPA7 possesses enhanced natriuretic, diuretic, systemic, and renal vasorelaxing and cardiac unloading properties. Importantly, NPA7's actions are superior to that of the individual native pGC-A or MasR ligands. These studies advance NPA7 as a novel, bispecific designer peptide with potential cardiorenal therapeutic benefit for the treatment of cardiovascular disease, such as hypertension and heart failure.


Subject(s)
Blood Pressure/drug effects , Drug Design , Hypertension/drug therapy , Oligopeptides/pharmacology , Vascular Resistance/drug effects , Animals , Disease Models, Animal , Dogs , Humans , Hypertension/metabolism , Hypertension/physiopathology , Kidney/drug effects , Kidney/metabolism , Male , Proto-Oncogene Mas
3.
Am J Physiol Regul Integr Comp Physiol ; 314(3): R407-R414, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29187381

ABSTRACT

The natriuretic peptides (NPs) B-type NP (BNP) and urodilatin (URO) exert renal protective properties via the particulate guanylyl cyclase A receptor (pGC-A). As a potential renal-enhancing strategy, we engineered a novel designer peptide that we call CRRL269. CRRL269 was investigated in human cell lines and in normal canines to define potential cardiorenal enhancing actions. The mechanism of its cardiorenal selective properties was also investigated. In vitro NP receptor activity was quantified with guanosine 3',5'-cyclic monophosphate generation. In vivo effects were determined in normal canine acute infusion studies. We observed that CRRL269 demonstrated enhanced pGC-A activity in renal compared with nonrenal cell lines. CRRL269 exerted enhanced resistance to neprilysin compared with URO. Importantly, CRRL269 exhibited significant and greater increases in urinary sodium excretion and diuresis, with less blood pressure reduction, than BNP or URO in normal canines. CRRL269 retained potent renin-angiotensin-aldosterone system (RAAS) suppressing properties shared by URO and BNP. Also, CRRL269 exerted less arterial relaxation and higher cAMP cardiomyocytes generation than BNP. CRRL269 possessed superior renal and pGC-A activating properties compared with BNP or URO in vitro. CRRL269 exerted enhanced renal actions while suppressing RAAS in vivo and with less hypotension compared with URO or BNP. Together, our study suggests that CRRL269 is a promising innovative renal-enhancing drug, with favorable protective actions targeting cardiorenal disease states through the pGC-A receptor.


Subject(s)
Diuresis/drug effects , Diuretics/pharmacology , Drug Design , Kidney/drug effects , Natriuretic Peptide, Brain/pharmacology , Oligopeptides/pharmacology , Receptors, Atrial Natriuretic Factor/agonists , Animals , Atrial Natriuretic Factor/pharmacology , Blood Pressure/drug effects , Cyclic GMP/metabolism , Diuretics/chemical synthesis , Dogs , Dose-Response Relationship, Drug , Drug Stability , HEK293 Cells , Humans , Kidney/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Natriuresis/drug effects , Natriuretic Peptide, Brain/chemistry , Neprilysin/pharmacology , Oligopeptides/chemistry , Peptide Fragments/pharmacology , Receptors, Atrial Natriuretic Factor/metabolism , Renin-Angiotensin System/drug effects , Second Messenger Systems/drug effects , Vasodilation/drug effects , Vasodilator Agents/chemical synthesis , Vasodilator Agents/pharmacology
4.
Physiol Rep ; 5(9)2017 May.
Article in English | MEDLINE | ID: mdl-28507167

ABSTRACT

Heart failure (HF) is a major health problem with worsening outcomes when renal impairment is present. Therapeutics for early phase HF may be effective for cardiorenal protection, however the detailed characteristics of the kidney in early-stage HF (ES-HF), and therefore treatment for potential renal protection, are poorly defined. We sought to determine the gene and protein expression profiles of specific maladaptive pathways of ES-HF in the kidney and heart. Experimental canine ES-HF, characterized by de-novo HF with atrial remodeling but not ventricular fibrosis, was induced by right ventricular pacing for 10 days. Kidney cortex (KC), medulla (KM), left ventricle (LV), and left atrial (LA) tissues from ES-HF versus normal canines (n = 4 of each) were analyzed using RT-PCR microarrays and protein assays to assess genes and proteins related to inflammation, renal injury, apoptosis, and fibrosis. ES-HF was characterized by increased circulating natriuretic peptides and components of the renin-angiotensin-aldosterone system and decreased sodium and water excretion with mild renal injury and up-regulation of CNP and renin genes in the kidney. Compared to normals, widespread genes, especially genes of the inflammatory pathways, were up-regulated in KC similar to increases seen in LA Protein expressions related to inflammatory cytokines were also augmented in the KC Gene and protein changes were less prominent in the LV and KM The ES-HF displayed mild renal injury with widespread gene changes and increased inflammatory cytokines. These changes may provide important clues into the pathophysiology of ES-HF and for therapeutic molecular targets in the kidney of ES-HF.


Subject(s)
Atrial Remodeling , Heart Failure/metabolism , Kidney/metabolism , Metabolic Networks and Pathways/genetics , Animals , Apoptosis/genetics , Cytokines/genetics , Cytokines/metabolism , Dogs , Heart Failure/pathology , Male , Myocardium/metabolism , Natriuretic Peptides/blood , Renin/genetics , Renin/metabolism , Water-Electrolyte Balance/genetics
5.
Am J Physiol Regul Integr Comp Physiol ; 304(2): R102-9, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23152112

ABSTRACT

In heart failure (HF), the cardiac hormone natriuretic peptides (NPs) atrial (ANP), B-type (BNP), and C-type (CNP) play a key role to protect cardiac remodeling. The proprotein convertases corin and furin process their respective pro-NPs into active NPs. Here we define in a canine model of HF furin and corin gene and protein expression in normal and failing left atrium (LA) or ventricle (LV) testing the hypothesis that the NP proproteins convertases production is altered in experimental HF. Experimental canine HF was produced by rapid right ventricular pacing for 10 days. NPs, furin, and corin mRNA expression were determined by quantitative RT-PCR. Protein concentration or expression was determined by immunostaining, radioimmunoassay, or Western blot. Furin and corin proteins were present in normal canine LA and LV myocardium and vasculature and in smooth muscle cells. In normal canines, expression of NPs was dominant in the atrium compared with the ventricle. In experimental early stage HF characterized with marked atrial fibrosis, ANP, BNP, and CNP mRNA, and protein concentrations were higher in HF LA but not HF LV compared with normals. In LA, corin mRNA and protein expressions in HF were lower, whereas furin mRNA and protein expressions were higher than normals. NPs and furin expressions were augmented in the atrium in experimental early stage HF and, conversely, corin mRNA and protein expressions were decreased with atrial remodeling. Selective changes of these NP convertases may have significance in the regulation of pro-NP processing and atrial remodeling in early stage HF.


Subject(s)
Furin/metabolism , Heart Failure/enzymology , Heart Ventricles/enzymology , Myocardium/enzymology , Natriuretic Peptides/metabolism , Protein Precursors/metabolism , Serine Endopeptidases/metabolism , Animals , Blotting, Western , Cardiac Pacing, Artificial , Disease Models, Animal , Dogs , Fibrosis , Furin/genetics , Heart Atria/enzymology , Heart Atria/pathology , Heart Failure/genetics , Heart Failure/pathology , Heart Ventricles/pathology , Humans , Immunohistochemistry , Male , Myocardium/pathology , Natriuretic Peptides/genetics , Protein Precursors/genetics , RNA, Messenger/metabolism , Radioimmunoassay , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Serine Endopeptidases/genetics , Time Factors , Ventricular Remodeling
6.
Circ Heart Fail ; 3(3): 412-9, 2010 May.
Article in English | MEDLINE | ID: mdl-20176717

ABSTRACT

BACKGROUND: Hemodynamic and neurohumoral function can affect the efficacy of diuretic therapy in congestive heart failure. Arginine vasopressin increases water reabsorption through the V(2) receptor in the collecting duct, whereas B-type natriuretic peptide (BNP) decreases sodium reabsorption in the collecting duct. We hypothesized that combining BNP to the V(2)-receptor antagonist tolvaptan (TLV) would enhance renal excretory function by augmenting sodium excretion together with aquaresis without adversely affecting renal hemodynamics in experimental congestive heart failure. METHODS AND RESULTS: Congestive heart failure was induced in 3 groups (n=6 per group) of dogs by tachypacing. A acute experiment was done after 10 days. After baseline measurements, study groups received a 0.1 mg/kg IV bolus of TLV alone (TLV), TLV in combination with BNP (TLV+BNP; 50 ng/[kg . min]), or BNP alone (BNP). Mean arterial pressure increased with TLV, remained unchanged with TLV+BNP, and decreased with BNP (+5+/-1mm Hg versus -1+/-1 mm Hg versus -15+/-1 mm Hg; P<0.05). Renal blood flow and glomerular filtration rate were preserved with all regimens. Urine flow increased in all 3 groups but significantly more so with TLV+BNP (TLV: +0.4+/-0.1 mL/min versus TLV+BNP: +2.4+/-0.5 mL/min versus BNP: +0.8+/-0.3 mL/min; P<0.05). Only TLV+BNP and BNP were natriuretic (P<0.05), whereas only TLV and TLV+BNP increased electrolyte-free water excretion (P<0.05). Compared with TLV alone, TLV+BNP prevented an increase in aldosterone (P<0.05). CONCLUSIONS: Coadministration of TLV and BNP in experimental HF resulted in a beneficial profile of renal, neurohumoral, and hemodynamic actions, specifically potent diuresis with natriuresis, neutral effect on mean arterial pressure, and lack of aldosterone activation.


Subject(s)
Benzazepines/administration & dosage , Heart Failure/metabolism , Heart Failure/physiopathology , Natriuretic Agents/administration & dosage , Natriuretic Peptide, Brain/administration & dosage , Receptors, Vasopressin/administration & dosage , Aldosterone/blood , Animals , Blood Pressure/drug effects , Disease Models, Animal , Dogs , Drug Therapy, Combination , Glomerular Filtration Rate/drug effects , Heart Failure/drug therapy , Male , Renal Circulation/drug effects , Tolvaptan , Water-Electrolyte Balance/drug effects
7.
Am J Physiol Regul Integr Comp Physiol ; 296(6): R1744-50, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19386989

ABSTRACT

32-amino acid B-type natriuretic peptide (BNP 1-32) plays an important role in cardiovascular homeostasis. Recently, it was reported that BNP 1-32 is cleaved by the metalloprotease meprin A to BNP 8-32, the bioactivity of which is undefined. We hypothesized that BNP 8-32 has reduced vasodilating and natriuretic bioactivity compared with BNP 1-32 in vivo. Human BNP 8-32 and BNP 1-32 were compared in a crossover study in eight anesthetized normal canines. After a preinfusion clearance, BNP 1-32 was infused at 30 ng.kg(-1) x min(-1) for 45 min followed by a 60-min washout and a second preinfusion clearance. Then, equimolar BNP 8-32 was infused. In half of the studies, the peptide sequence was reversed. Changes with peptides from the respective preinfusion clearance to infusion clearance were compared with paired tests. Mean arterial pressure was reduced by both BNP 8-32 and BNP 1-32 (-8 +/- 3 vs. -6 +/- 2 mmHg, P = 0.48). Changes in right atrial pressure, pulmonary capillary wedge pressure, heart rate, cardiac output, and glomerular filtration rate were similar. However, urinary sodium excretion increased less with BNP 8-32 than with BNP 1-32 (+171 +/- 24 vs. +433 +/- 43 muEq/min; P = 0.008), as did urinary potassium excretion, urine flow, and renal blood flow. While BNP 8-32 has similar vasodilating actions as BNP 1-32, its diuretic and natriuretic actions are reduced, suggesting a role for meprin A in the regulation of BNP 1-32 bioactivity in the kidney. Meprin A inhibition may be a potential strategy to increase the bioactivity of endogenous and exogenous BNP 1-32 in cardiovascular diseases.


Subject(s)
Cardiovascular System/metabolism , Diuresis , Hemodynamics , Kidney/metabolism , Metalloendopeptidases/metabolism , Natriuresis , Natriuretic Peptide, Brain/metabolism , Peptide Fragments/metabolism , Amino Acid Sequence , Animals , Biomarkers/blood , Cells, Cultured , Cyclic GMP/metabolism , Dogs , Endothelial Cells/metabolism , Humans , Infusions, Intravenous , Male , Molecular Sequence Data , Natriuretic Peptide, Brain/administration & dosage , Natriuretic Peptide, Brain/chemistry , Peptide Fragments/administration & dosage , Peptide Fragments/chemistry
8.
Hypertension ; 50(3): 481-8, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17635858

ABSTRACT

Unlike healthy subjects, overt congestive heart failure cannot "escape" the sodium- and water-retaining actions of mineralocorticoid excess. It is undefined whether escape occurs in asymptomatic left ventricular dysfunction (ALVD), which is characterized by preserved sodium homeostasis, natriuretic peptide activation, and normal circulating aldosterone. We hypothesized that, in ALVD, mineralocorticoid excess with exogenous deoxycorticosterone acetate (DOCA) would overwhelm renal compensatory mechanisms, resulting in sodium and water retention, and promote renal and cardiac collagen deposition. ALVD was induced in 2 groups (N=5 each) of dogs by tachypacing at 180 bpm. Urine was collected daily and blood drawn at baseline and days 2, 5, 8, and 11. One group served as control (ALVD), and the other received DOCA (ALVD+DOCA) starting at day 2 of pacing. Urine flow and sodium excretion were unchanged in the ALVD group. In ALVD+DOCA, urine flow and sodium excretion decreased on the first 2 days DOCA was given but normalized starting day 4. Urine flow and urinary cGMP excretion increased in ALVD+DOCA after DOCA escape. Plasma atrial natriuretic peptide, B-type natriuretic peptide, and cGMP increased equally in both groups. There were no differences in cardiorenal and hemodynamic parameters in an acute study on day 11. Although renal collagen area fraction was similar, left ventricular collagen area fraction in ALVD+DOCA was significantly higher than in ALVD (3.3+/-0.4% versus 2.0+/-0.2%; P=0.012). We conclude that ALVD can escape the sodium- and water-retaining effects of mineralocorticoid excess. Despite renal escape, increased left ventricular collagen deposition suggests that the heart but not the kidney failed to escape the tissue effects of DOCA.


Subject(s)
Desoxycorticosterone/pharmacology , Kidney/metabolism , Myocardium/metabolism , Receptors, Mineralocorticoid/metabolism , Ventricular Dysfunction, Left/metabolism , Animals , Body Water/metabolism , Collagen/metabolism , Cyclic GMP/urine , Diuresis , Dogs , Male , Natriuresis , Sodium/metabolism , Ventricular Dysfunction, Left/urine
9.
Am J Physiol Regul Integr Comp Physiol ; 292(2): R897-901, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17068158

ABSTRACT

Brain natriuretic peptide (BNP 1-32) plays an important physiologic role in cardiorenal homeostasis. Recently, it has been reported that BNP 1-32 is rapidly cleaved by the ubiquitous enzyme dipeptidyl peptidase IV to BNP 3-32, which lacks the two NH2-terminal amino acids of BNP 1-32. The bioactivity of BNP 3-32 in cardiorenal regulation is unknown. We hypothesized that BNP 3-32 has reduced vasodilating and natriuretic bioactivity compared with BNP 1-32 in vivo. Synthetic human BNP 3-32 and BNP 1-32 were administered to eight anesthetized normal canines. After baseline measurements, BNP 1-32 at 30 ng x kg(-1) x min(-1) was administered, followed by a washout, a postinfusion clearance, and a clearance with an equimolar dose of BNP 3-32. In four studies, the sequence of BNP 1-32 and BNP 3-32 infusion was reversed. Peptides were compared by analyzing the changes from the respective preinfusion clearance to the respective infusion clearance. *P < 0.05 between peptides. BNP 3-32, unlike BNP 1-32, did not decrease mean arterial pressure (0 +/- 1 vs. -7 +/- 2* mmHg, respectively) and did not increase renal blood flow (+12 +/- 10 vs. +52 +/- 10* ml/min). Effects on heart rate and cardiac output were similar. Urinary sodium excretion increased 128 +/- 18 microeq/min with BNP 3-32 and 338 +/- 40* microeq/min with BNP 1-32. Urine flow increased 1.1 +/- 0.2 ml/min with BNP 3-32 and 2.8 +/- 0.4* ml/min with BNP 1-32. Plasma BNP immunoreactivity was lower with BNP 3-32, suggesting accelerated degradation. In this study, BNP 3-32 showed reduced natriuresis and diuresis and a lack of vasodilating actions compared with BNP 1-32.


Subject(s)
Heart/drug effects , Kidney/drug effects , Natriuretic Peptide, Brain/pharmacology , Animals , Blood Pressure/drug effects , Cardiac Output/drug effects , Dipeptidyl Peptidase 4/chemistry , Diuresis/drug effects , Dogs , Heart Rate/drug effects , Hormones/blood , Kidney/metabolism , Kidney Function Tests , Male , Natriuretic Peptide, Brain/chemical synthesis , Neurotransmitter Agents/physiology , Renal Circulation/drug effects , Urodynamics/physiology , Vascular Resistance/drug effects , Vasodilation/drug effects , Water-Electrolyte Balance/drug effects , Water-Electrolyte Balance/physiology
10.
Circulation ; 112(6): 836-40, 2005 Aug 09.
Article in English | MEDLINE | ID: mdl-16061734

ABSTRACT

BACKGROUND: The objective of this study was to address the feasibility and the biological activity of orally administered human brain natriuretic peptide (hBNP). Proprietary technology has been developed in which short, amphiphilic oligomers are covalently attached to peptides. The conjugated peptides are intended to have an improved pharmacokinetic profile and to enable oral administration. We hypothesized that novel oral conjugated hBNP (CONJ-hBNP) increases plasma hBNP, activates cGMP, and reduces mean arterial pressure (MAP). METHODS AND RESULTS: This randomized crossover-designed study tested the biological activity of oral CONJ-hBNP compared with oral native hBNP in normal conscious dogs. Measurements of MAP, plasma hBNP, and cGMP were made at baseline (BL) and repeated at 10, 30, 60, 120, 180, and 240 minutes after oral administration. Plasma hBNP was not detectable in dogs at BL. Plasma hBNP was detected after native hBNP and CONJ-HBNP administration. However, plasma hBNP concentration was significantly higher after CONJ-hBNP than after native hBNP administration (P=0.0374 between groups). Plasma cGMP increased after CONJ-hBNP for 60 minutes (from 10.8+/-3 to 36.8+/-26 pmol/mL; P<0.05), whereas it did not change after native hBNP (P=0.001 between groups). MAP decreased at 10 minutes and remained decreased for 60 minutes after CONJ-hBNP (from 113+/-8 to 101+/-12 mm Hg after 10 minutes to 97.5+/-10 mm Hg after 30 minutes to 99+/-13 mm Hg after 60 minutes) while remaining unchanged after native hBNP (P=0.0387 between groups). CONCLUSIONS: This study reports for the first time that novel conjugated oral BNP activates cGMP and significantly reduces MAP, thus implying an efficacious coupling of CONJ-hBNP to the natriuretic receptor-A. These data advance a new concept of orally administered chronic BNP therapy for cardiovascular diseases.


Subject(s)
Blood Pressure/drug effects , Cyclic GMP/blood , Natriuretic Peptide, Brain/pharmacology , Administration, Oral , Animals , Cross-Over Studies , Dogs , Humans , Hypotension/chemically induced , Male , Models, Animal , Natriuretic Peptide, Brain/administration & dosage , Natriuretic Peptide, Brain/blood
11.
Circulation ; 107(5): 686-9, 2003 Feb 11.
Article in English | MEDLINE | ID: mdl-12578869

ABSTRACT

BACKGROUND: BAY 41-2272 is a recently introduced novel orally available agent that directly stimulates soluble guanylate cyclase (sGC) and sensitizes it to its physiological stimulator, nitric oxide. To date, its therapeutic actions in congestive heart failure (CHF) remain undefined. We characterized the cardiorenal actions of intravenous BAY 41-2272 in a canine model of CHF and compared it to nitroglycerin (NTG). METHODS AND RESULTS: CHF was induced by rapid ventricular pacing for 10 days. Cardiorenal and humoral function were assessed at baseline and with administration of 2 doses of BAY 41-2272 (2 and 10 micro g x kg(-1) x min(-1); n=8) or NTG (1 and 5 micro g x kg(-1) x min(-1); n=6). Administration of 10 micro g x kg(-1) x min(-1) BAY 41-2272 reduced mean arterial pressure (113+/-8 to 94+/-6 mm Hg; P<0.05), pulmonary artery pressure (29+/-2 to 25+/-2 mm Hg; P<0.05), and pulmonary capillary wedge pressure (25+/-2 to 20+/-2 mm Hg; P<0.05). Cardiac output (2.1+/-0.2 to 2.3+/-0.2 L/min; P<0.05) and renal blood flow (131+/-17 to 162+/-18 mL/min; P<0.05) increased. Glomerular filtration rate was maintained. There were no changes in plasma renin activity, angiotensin II, or aldosterone. NTG mediated similar hemodynamic changes and additionally decreased right atrial pressure and pulmonary vascular resistance. CONCLUSION: The new sGC stimulator BAY 41-2272 potently unloaded the heart, increased cardiac output, and preserved glomerular filtration rate without activation of the renin-angiotensin-aldosterone system in experimental CHF. These beneficial properties make direct sGC stimulation with BAY 41-2272 a promising new strategy for the treatment of cardiovascular diseases such as CHF.


Subject(s)
Heart Failure/drug therapy , Heart/drug effects , Kidney/drug effects , Pyrazoles/pharmacology , Pyridines/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Aldosterone/blood , Angiotensin II/blood , Animals , Cardiac Output/drug effects , Cardiac Pacing, Artificial , Disease Models, Animal , Dogs , Drug Evaluation, Preclinical , Glomerular Filtration Rate/drug effects , Guanylate Cyclase , Heart Function Tests/drug effects , Hemodynamics/drug effects , Injections, Intravenous , Kidney Function Tests , Male , Nitroglycerin/pharmacology , Pyrazoles/therapeutic use , Pyridines/therapeutic use , Receptors, Cytoplasmic and Nuclear/drug effects , Renin/blood , Renin-Angiotensin System/drug effects , Soluble Guanylyl Cyclase
12.
Circulation ; 105(8): 999-1003, 2002 Feb 26.
Article in English | MEDLINE | ID: mdl-11864932

ABSTRACT

BACKGROUND: A hallmark of congestive heart failure (CHF) is the elevation of the cardiac natriuretic peptides (NPs), which have natriuretic, renin-inhibiting, vasodilating, and lusitropic properties. We have reported that chronic subcutaneous (SQ) administration of brain natriuretic peptide (BNP) in experimental CHF improves cardiorenal function. Vasopeptidase inhibitors (VPIs) are single molecules that simultaneously inhibit both neutral endopeptidase 24.1 (NEP) and ACE. We hypothesized that acute VPI administration would potentiate the cardiorenal actions of SQ BNP in experimental CHF. METHODS AND RESULTS: We determined the cardiorenal and humoral responses to acute VPI alone with omapatrilat (OMA) (1 micromol/kg IV bolus) (n=6), acute low-dose SQ BNP (5 microg/kg) alone (n=5), acute VPI plus low-dose SQ BNP (n=5), and acute high-dose SQ BNP (25 microg/kg) alone in 4 groups of anesthetized dogs with experimental CHF produced by ventricular pacing for 10 days. Plasma BNP was greater with VPI+low-dose SQ BNP compared with VPI alone or low-dose SQ BNP alone and was similar to high-dose SQ BNP alone. Urinary BNP excretion was greatest with VPI+SQ BNP. Urinary sodium excretion was also highest with VPI+SQ BNP, with the greatest increase in glomerular filtration rate. VPI+SQ BNP resulted in a greater increase in cardiac output and reduction in cardiac filling pressures as compared with low-dose SQ BNP, high-dose SQ BNP, or VPI alone. CONCLUSIONS: This study reports that acute VPI potentiates the cardiorenal actions of SQ BNP in experimental CHF. This study advances the concept that protein therapy with BNP together with vasopeptide inhibition represents a novel therapeutic strategy in CHF to maximize the beneficial properties of the natriuretic peptide system.


Subject(s)
Atrial Natriuretic Factor/metabolism , Heart Failure/drug therapy , Heart Failure/physiopathology , Natriuretic Peptide, Brain/administration & dosage , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Animals , Cardiac Pacing, Artificial , Cyclic GMP/blood , Cyclic GMP/urine , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Glomerular Filtration Rate/drug effects , Hemodynamics/drug effects , Injections, Subcutaneous , Kidney/drug effects , Kidney/physiopathology , Male , Natriuretic Peptide, Brain/blood , Natriuretic Peptide, Brain/urine , Neprilysin/antagonists & inhibitors , Peptidyl-Dipeptidase A/metabolism , Pyridines/administration & dosage , Sodium/urine , Thiazepines/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...