Next Article in Journal
In Vitro Antiviral and Anti-Inflammatory Activities of N-Acetylglucosamine: Development of an Alternative and Safe Approach to Fight Viral Respiratory Infections
Next Article in Special Issue
The Liver in Heart Failure: From Biomarkers to Clinical Risk
Previous Article in Journal
Respiratory Health Effects of In Vivo Sub-Chronic Diesel and Biodiesel Exhaust Exposure
Previous Article in Special Issue
New Insight in Cardiorenal Syndrome: From Biomarkers to Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Natriuretic Peptides: It Is Time for Guided Therapeutic Strategies Based on Their Molecular Mechanisms

1
Department of Clinical and Molecular Medicine, Sapienza University of Rome, Sant’Andrea Hospital, Via di Grottarossa 1035, 00189 Rome, RM, Italy
2
IRCCS Neuromed, Via Atinense 18, 86077 Pozzilli, IS, Italy
3
IRCCS San Raffaele Cassino, Via G. Di Biasio 1, 03043 Cassino, FR, Italy
4
IRCCS San Raffaele Roma, Via della Pisana 235, 00163 Rome, RM, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(6), 5131; https://doi.org/10.3390/ijms24065131
Submission received: 10 February 2023 / Revised: 2 March 2023 / Accepted: 6 March 2023 / Published: 7 March 2023

Abstract

:
Natriuretic peptides (NPs) are the principal expression products of the endocrine function of the heart. They exert several beneficial effects, mostly mediated through guanylate cyclase-A coupled receptors, including natriuresis, diuresis, vasorelaxation, blood volume and blood pressure reduction, and regulation of electrolyte homeostasis. As a result of their biological functions, NPs counterbalance neurohormonal dysregulation in heart failure and other cardiovascular diseases. NPs have been also validated as diagnostic and prognostic biomarkers in cardiovascular diseases such as atrial fibrillation, coronary artery disease, and valvular heart disease, as well as in the presence of left ventricular hypertrophy and severe cardiac remodeling. Serial measurements of their levels may be used to contribute to more accurate risk stratification by identifying patients who are more likely to experience death from cardiovascular causes, heart failure, and cardiac hospitalizations and to guide tailored pharmacological and non-pharmacological strategies with the aim to improve clinical outcomes. On these premises, multiple therapeutic strategies based on the biological properties of NPs have been attempted to develop new targeted cardiovascular therapies. Apart from the introduction of the class of angiotensin receptor/neprilysin inhibitors to the current management of heart failure, novel promising molecules including M-atrial natriuretic peptide (a novel atrial NP-based compound) have been tested for the treatment of human hypertension with promising results. Moreover, different therapeutic strategies based on the molecular mechanisms involved in NP regulation and function are under development for the management of heart failure, hypertension, and other cardiovascular conditions.

1. Regulation of Natriuretic Peptide Synthesis and Secretion

Natriuretic peptides (NPs) are the principal expression products of the hormonal function of the heart.
The main components of the NP family are atrial natriuretic peptides (ANPs), brain or B-type natriuretic peptide (BNPs), and C-type natriuretic peptides (CNPs). ANPs and BNPs are mostly synthesized in the heart whereas CNPs are produced by endothelial cells [1,2,3,4].
NPs exert relevant physiological cardiovascular and renal effects, which include natriuresis, diuresis, vasorelaxation, blood volume and blood pressure reduction, trans-capillary fluid shift, increase in glomerular filtration rate, and inhibition of the renin-angiotensin-aldosterone system (RAAS) [1,2,3,4].
Apart from the endocrine functions, NPs exert several autocrine/paracrine effects, including lipolysis, reduction in inflammation and of oxidative stress, stimulation of autophagy, and anti-fibrotic and anti-hypertrophic actions which play a role in cardiovascular pathophysiology [5,6].
The production of ANPs and BNPs by cardiomyocytes occurs mostly as a consequence of pressure and volume overload and entails regulated secretion deriving from stored granules and a constitutive secretion of newly synthesized hormones [7,8,9,10]. At least three distinct mechanisms are involved in the stimulated secretion of NPs: stretch-activated Goα-coupled secretion, Gqα-coupled secretion, and cytokine-promoted secretion. Acute mechanical stretch, which occurs within hours, produces a short-term burst of regulated secretion of NPs, without evident effects on their biosynthesis. In subacute (days) and chronic (weeks) haemodynamic load conditions circulating NPs derive from both stored and newly synthesized molecule [7,8,9,10].
In fact, the prohormones proANP and proBNP are stored in secretory granules, are enzymatically cleaved by proprotein convertases (corin and furin), and are processed to form the N-terminal peptides NT-proANP and NT-proBNP and the biologically active COOH-terminal peptides ANP and BNP [11]. The main circulating form of ANP is the COOH-terminal peptide (αANP), while BNPs and NT-proBNPs circulate in the plasma along with unprocessed proBNPs and other inactive fragments [12,13]. In normal conditions, ANPs and BNPs are mostly produced within the atria and, more specifically, within the auricular appendage [14]. Studies performed with electron microscopy have shown that secretory granules are not displayed in ventricular cardiomyocytes from normal hearts, but they may be detected in samples of ventricular myocardium collected during surgery or endocardial biopsies in patients with cardiac disease [15]. These data suggest that NP synthesis and secretion are differently regulated in atrial cardiomyocytes as compared to ventricular cardiomyocytes and that pathological conditions leading to increased wall stress stimulate the pre-programmed expression of fetal genes in ventricular cardiomyocytes [15]. It has been also demonstrated that additional ventricular BNP expression occurs in clinically overt HF, suggesting that in this condition the atrial secretion might not be sufficient to counteract the activation of vasoconstrictive and sodium retaining neurohormonal systems, such as the renin-angiotensin-aldosterone and sympathetic nervous systems [16]. These findings were confirmed in an advanced HF setting of heart transplantation, further supporting the hypothesis that ventricular production mostly occurs under advanced pathological conditions and is related to ventricular remodeling and other abnormalities associated with hypertrophy, fibrosis, inflammation, and hypoxia [17,18].
Besides the mechanical triggering stimulus occurring in conditions of volume and pressure overload, different neurohormonal stimuli, including angiotensin II, endothelin-1, arginine vasopressin, adrenergic agonists, growth factors and cytokines (tumor necrosis factor and interleukins −1 and 6), glucocorticoids, thyroid hormones, sex steroids, and the incretin glucagon-like peptide-1 (GLP1) [19,20], may promote increased secretion of NPs. The neurohormonal stimuli regulate the expression of both NPPA and NPPB, the genes encoding ANP and BNP, respectively, through the transcriptional factors RAS/c-Raf-1 and the phosphoinositide signaling pathways which stimulate the p38-mitogen-activated protein kinase (MAPK) [21,22,23].
Epigenetic regulation, consisting in histone acetylation and methylation, also plays a role in the synthesis of NPs [24]. Pressure and volume overload have been related to the enhanced acetylation of H3 and H4 histones and to the demethylation of H3K9 at NPPA and NPPB. In these conditions, the expression of three histone methylation-related molecules H3K4me3, H3K9me2, and H3K9me3 is reduced [25,26,27]. The NPPA expression is also modulated by known miRNAs, such as miR-425, miR-155, and miR-105 [28,29,30]. Indeed, subjects carrying the minor G allele at rs5068, which does not bind miR-425, show higher ANP levels resulting in a lower risk of CVD, obesity, and hypertension [31,32,33]. Moreover, the inhibition of the NPPA-AS1 natural antisense transcript, which reduces the NPPA transcription, increases both ANP levels and cGMP resulting in BP reduction [34] (Figure 1).
The orange boxes represent the regulatory mechanisms of gene expression and the secretion of NPs, including new potential strategies such as epigenetic regulation, microRNA modulation, and NPPA AS1 inhibition.
The light-blue boxes illustrate NP-based potential therapeutic strategies.
ANP, atrial natriuretic peptide; ARNi, angiotensin receptor/neprilysin inhibitors; BNP, brain natriuretic peptide; CNP, C-type natriuretic peptide; miR, microRNA; NEP, neprilysin; NPR-A, type A natriuretic peptide receptor; NPR-B, type B natriuretic peptide receptor; NPR-C, type C natriuretic peptide receptor; NPs, natriuretic peptides

2. Natriuretic Peptide Receptors

The biological actions of NPs are mediated by three receptors: type A (NPR-A), type B (NPR-B), and type C (NPR-C) NP receptors [35]. NPR-A is the main receptor for ANP and BNP, with ANP showing the greatest affinity to NPR-A, followed by BNP and CNP [35]. CNP has the highest affinity to NPR-B, followed by ANP and BNP. BNP has the highest affinity to NPR-C, followed by CNP and ANP. NPR-A and NPR-B are expressed in many tissues, including the heart, vessels, kidneys, brain, lungs, adrenal glands, and adipose tissue. NPR-A and NPR-B are composed of three domains: an extracellular domain that binds NPs, a short transmembrane domain, an intracellular domain that contains a protein kinase homology domain, a helical hinge region, and a guanylyl cyclase domain [36]. In the absence of stimuli, the protein kinase domain of NPR-A is phosphorylated, whereas when a ligand binding occurs, phosphates are removed resulting in receptor desensitization. The hinge region is involved in the oligomerization of NP receptors and in the activation of the catalytic domain [36].
Differently from NPR-A and NPR-B, NPR-C is characterized by a cell membrane dimeric conformation and does not have guanylyl cyclase activity. NPR-C internalizes NPs leading to lysosome degradation and to their clearance [37]. Neprilysin is the other major catalytic pathway involved in the degradation of NPs and other biologically active peptides, including bradykinins, substance P, enkephalins, and adrenomedullin [38].
The NPs binding to NPR-A and NPR-B cause the intracellular formation of cyclic guanylate monophosphate (cGMP) whose intracellular targets are cGMP-dependent protein kinases (PKGs), cGMP-gated ion channels, and cGMP-regulated cyclic nucleotide phosphodiesterases [39,40].
PKG phosphorylates Ca2+-activated K+ channels and inositol trisphosphate receptor-associated cGMP kinase substrates, resulting in a reduction in the cytosolic concentration of Ca2+ and hence the relaxation of vascular smooth muscle cells [41]. In addition to these biological mechanisms, endothelium derived CNP contributes to maintaining endothelial function and integrity and possibly to the chronic regulation of vascular tone, promoting vasorelaxation in isolated resistance arteries and BP reduction [42,43]. PKG1α plays an important role in the inhibition of inflammation and leukocyte recruitment, platelet aggregation, smooth muscle proliferation, vasoconstriction, fibrosis, coronary microvascular impairment, and hypertrophy through different downstream mechanisms, including SR Ca2+-ATPase (SERCA)-mediated increased re-uptake of Ca2+ into the sarcoplasmic reticulum and the blockade of mitochondrial permeability transition pore [44,45].
NPR-B also plays a role in maintaining normal heart rate and sino-atrial node function by modulating ion channel function via a cGMP/phosphodiesterase-3 (PDE3)/cyclic adenosine monophosphate (cAMP) signaling mechanism [46].
NPs exert anti-hypertrophic and anti-fibrotic effects through the modulation of several pathways: calcineurin/NFAT (nuclear factor of activated T-cells), the sodium exchanger NHE-1 (sodium-hydrogen antiporter 1), and the TGFβ1 (transforming growth factor β1)/Smad signaling pathways. NPs reduce vascular inflammation by inhibiting the TNFα-induced expression of adhesion molecules, such as E-selectin and ICAM-1. NPs also inhibit the pathway of RAC1 (Ras-related C3 botulinum toxin substrate)/p38/MAPK (mitogen activated protein kinase), which is involved in the activation of Akt and mTOR and its downstream target p70S6K, the Ras/MEK (mitogen-activated protein kinase)/ERK (extracellular-signal-regulated kinase) signaling. These biological pathways lead to an increased expression of c-fos mRNA, JNK (c-JUN N-terminal kinase), and c-Src, which stimulate JAK2 (Janus kinase 2)-STAT3 (signal transducer and activator of transcription 3) through PKC (protein kinase C) and PyK2 (proline-rich tyrosine kinase 2) [47,48,49] (Figure 2).
Consistently, the ablation of NPPA or NPR1 (the gene encoding NPR-A) as well as NP genetic variants leading to reduced peptide expression have been associated with cardiac hypertrophy and impaired endothelial cell viability and proliferation [50,51,52,53,54]. A long-term proBNP peptide gene delivery exerted an anti-hypertrophic effect and improved both systolic and diastolic function in the spontaneously hypertensive rat [55]. An NPPA mutation was associated with the enhanced expression and function of a cardiac potassium channel and to mitochondrial electron transport chain dysfunction, resulting in an electrophysiological substrate for atrial fibrillation [56,57]. Moreover, ANPs were recently discovered to regulate cardiac autophagy, reducing the cellular quote of damaged cells and organelles, and finally protecting the heart from ischemic insult [58].
Resistance to the biological effects of NPs can be attributed to different mechanisms acting at pre-receptor, receptor, and post-receptor levels. The impaired post-translational processing of proANP and proBNP was associated with NP resistance in HF, as well as the reduced corin and furin levels and the glycosylation of proBNP near the cleavage site of the enzyme, resulting in the resistance to proteolysis [59]. An increased expression of the clearance receptor NPR-C was associated with a reduction in the circulating levels of active NPs. Different gene polymorphisms of NP receptors may contribute to the development of CVD [60]. Moreover, the increased metabolism of cGMP due to the enhanced activity of PDE may also contribute to the increased resistance of NPs. Although the substitution of exogenous NO by nitrates and the inhibition of cGMP degradation by PDE inhibitors have shown some potential benefits in the treatment of HF, a long-term effect has not been demonstrated [61].

3. The Key Role of Natriuretic Peptides as Clinical Biomarkers in Cardiovascular Diseases

BNP and NT-proBNP represent widely recommended biomarkers for the diagnosis of acute decompensated HF and also have an important prognostic role since their increase tightly reflect HF worsening and predict outcomes in HF patients. According to current international guidelines, cut-off values for BNP and NT-proBNP are in routine use for ruling in or ruling out HF [62], since they represent a more stable and reproducible parameter in the clinical work-up.
The role of NPs as biomarkers to guide the clinical behavior in HF has resulted in an improvement in clinical outcomes, though they did not specifically address the impact on the intensity of care. In fact, in the recent COACH (Comparison of Outcomes and Access to Care for Heart Failure) trial, the use of a point-of-care tool for risk stratification in the emergency department to support clinical decision-making and rapid outpatient follow-up was associated with an improvement of 30-day and 20-month clinical outcomes [63]. In this approach, the use of NPs as routine biomarkers most likely played a role [64]. In addition to BNPs and NT-proBNPs, different studies have shown that ANPs and MR-proANPs may representuseful tools for the diagnosis of HF and are associated with increased all-cause and cardiovascular mortality, although their diagnostic values as biomarkers have not been extensively investigated [1,65]. In HF patients enrolled in the Atherosclerosis Risk in Community study, NT-proBNP level >125 pg/mL reflected an increased risk for HF events and death, supporting the utility of serial NT-proBNP measurements to improve risk stratification [66].
In addition to being well-established markers of HF, plasma levels of NPs are also risk markers for different CVD such as atrial fibrillation (AF), coronary artery disease, and valvular heart disease, as well as in the presence of left ventricular hypertrophy [67,68] and severe cardiac remodeling [69]. Higher levels of BNPs and NT-proBNPs were detected in subjects with grade-2 hypertension compared to normotensives, corresponding to an increased risk of organ damage and hypertension-related sequelae [70]. On the other hand, a deficiency of NPs was reported in the early stage of hypertension, and it was proposed as a contributory pathogenetic mechanism [71,72]. NP levels are increased in >75% of patients with AF and were associated with an increased risk of stroke, systemic embolism, and cardiac-related death, thus enhancing the prognostic role of the CHA2DS2-VASc score [73]. NPs are sensitive markers of both clinically evident and subclinical myocardial ischemia, being significantly more expressed in human coronary explants of advanced atherosclerotic lesions compared to early atherosclerotic lesions [74]. NP levels were able to predict future cardiovascular events in subjects with previous myocardial infarction [75]. In a 10-year follow-up study performed in patients with stable coronary artery disease, NT-proBNP levels within the highest quartile were associated with a 50% survival rate and with an increased risk of myocardial infarction cardiovascular, and all-cause death [76].
In valvular heart disease, NPs associated with the severity of aortic stenosis, LV chamber size, wall thickness, wall stress, LVEF, left atrial size, and right ventricular pressure are useful markers for monitoring the evolution from asymptomatic to symptomatic disease and finally to HF [77]. In mitral regurgitation NPs contribute to assess the progression of the valve disease, even before the development of hemodynamic consequences in a preclinical stage of myocardial damage. They may also contribute to more accurate risk stratification by identifying patients who are more likely to experience death from cardiovascular causes, HF, and cardiac hospitalizations, thus requiring surgical management rather than a conservative approach [78].
In patients with pulmonary arterial hypertension, BNP levels showed a direct correlation with the mean pulmonary artery pressure, namely with a substantial increase in mortality when accompanied by an enlarged right atrium measured by echocardiography [79,80,81,82]. NPs predict mortality in patients with end-stage renal disease [83].

4. Therapeutic Implications of the Regulation of Natriuretic Peptide Levels and Activity

Over the last few years, the regulation of NP levels and activity have established broad applications in clinical practice due to the development of a new therapeutic strategy, namely the angiotensin receptor neprilysin inhibitor (ARNi): the single pill sacubitril/valsartan aimed at combining the effect of reduced enzymatic degradation of NPs with consequent increased NP levels and reduced angiotensin II receptor binding and related signal transduction pathways. Due to the higher affinity of neprilysin for ANP and CNP than for BNP, neprilysin inhibition increases the ANP carboxyterminal level. The parallel decrease of the circulating amino-terminal peptides observed upon ARNi treatment mirrors the cardiac function improvement [38].
In the proof-of-concept study PARADIGM-HF, which enrolled 8442 patients with HFrEF (EF ≤ 35%, ≤40% before the amendment) and NYHA class II to IV, sacubitril/valsartan reduced the risk of the composite primary outcome of cardiovascular death or hospitalization for HF by 20% compared with enalapril. Sacubitril/valsartan also lowered the risk of cardiovascular death by 20% and HF hospitalization by 21% compared to enalapril [84]. The benefits of sacubitril/valsartan were independent from LVEF, age, renal function, BP, HF etiology (ischemic, idiopathic, hypertensive, or other non-ischemic causes such as infective/viral, alcoholic, valvular, drug-related, and peripartum-related), previous hospitalizations, signs of congestion, background pharmacological therapy, glycemic status, and geographic origin [84]. The improvement of cardiovascular outcomes in the group treated with sacubitril/valsartan was mirrored by a reduction in the NT-proBNP level. In the group who received sacubitril/valsartan, the BNP level increased at 30 days after randomization but tended to be lower at the 8-month follow-up compared to the group treated with enalapril. Otherwise, the NT-proBNP levels were reduced at 30 days and 8 months in the active group [85,86].
The efficacy of sacubitril/valsartan was also demonstrated in acute subsets. The TITRATION study showed that initiation of sacubitril/valsartan and subsequent up-titration in both hospitalized and outpatient HFrEF patients with LVEF ≤ 35% had a similar tolerability profile without significant differences in the occurrence of adverse events such as hyperkalemia, angioedema, renal dysfunction, or hypotension [87]. In the PIONEER-HF study conducted in patients hospitalized for acute HF, treatment with sacubitril/valsartan produced a 29% greater reduction in the NT-proBNP plasma concentration compared with enalapril (percent change, −46.7% vs. −25.3%), without significant differences in the risk of adverse events. Moreover, patients treated with sacubitril/valsartan experienced a lower incidence of all-cause mortality, rehospitalization for HF, left ventricular assist device implantation, or listing for cardiac transplant and of the composite of cardiovascular death or rehospitalization for HF [88]. In the TRANSITION trial, the target dose of sacubitril 97 mg and valsartan 103 mg twice daily after 10 weeks was achieved in a comparable percentage of hospitalized patients who started the treatment ≥12-h pre-discharge or between days 1–14 post-discharge, with a similar incidence of discontinuation due to adverse events [89].
In the PARAGON-HF trial, the efficacy of sacubitril/valsartan compared to valsartan alone was investigated in 4822 patients with LVEF of 45% or higher. Although a trend towards a reduction in the composite endpoint of cardiovascular death and total hospitalizations for HF was observed in the sacubitril/valsartan arm compared to valsartan (Rate ratio 0.87; 95%CI 0.75–1.01, p = 0.06), statistical significance was not reached [90].
More recently, Solomon and colleagues performed a pooled analysis of these two trials with the aim to evaluate the efficacy of sacubitril/valsartan across the EF spectrum. In this cohort of >13,000 subjects, sacubitril/valsartan was associated with a significant reduction in the combined endpoint of cardiovascular mortality and first hospitalization for HF (−16%), in all-cause mortality (−12%), total HF hospitalizations and cardiovascular mortality (−18%), and total HF hospitalizations (−19%) [91].
On the basis of this evidence, the European Guidelines recommend sacubitril/valsartan as first-line treatment in HFrEF and suggest considering its use in HF with mildly reduced EF (40–49%) [62,92,93].
In the PARADISE-MI (Prospective ARNI vs. ACE Inhibitor Trial to Determine Superiority in Reducing Heart Failure Events after Myocardial Infarction) study, sacubitril/valsartan did not produce significant benefits compared to ramipril on the composite outcome of cardiovascular death, HF hospitalizations, or outpatient development of HF in patients with acute myocardial infarction [94]. However, a superiority of sacubitril/valsartan was reported in a recent posthoc analysis of this study including only investigator-identified events [95].
Different studies have hypothesized and proven that the benefits of sacubitril/valsartan might be related to reverse cardiac remodeling. In the PROVE-HF study, the reduction in NT-proBNP levels was associated with the improvement of cardiac systolic and diastolic function consisting in the increase in LVEF and in the decrease of LV end-diastolic volume index (LVEDVI), LV end-systolic volume index (LVESVI), left atrial volume index (LAVI), and early diastolic filling–early diastolic annular velocity (E/eʹ) [96]. In the EVALUATE-HF study, sacubitril/valsartan reduced the remodeling indices (LVEDVI, LVESVI, LAVI, and E/eʹ) compared to enalapril, although without significant effects on LVEF, longitudinal strain, or aortic characteristic impedance [97].
An anti-fibrotic effect of sacubitril/valsartan was also demonstrated, with a reduction in the levels of different markers of tissue fibrosis such as ST2, tissue inhibitor of metalloproteinases-1 (TIMP-1), N-terminal pro-peptide of type I procollagen, and matrix metalloproteinases. In this context, the reduction in TIMP-1 levels is associated with a lower risk of cardiovascular events [98].
This large body of data may support the role of serial measurements of NP levels to guide the titration of long-term medical therapy in HF patients with the aim of reducing HF hospitalizations and improving clinical outcomes. NP activity might be enhanced by their interaction with other post-receptor pathways. In experimental models of HF, soluble guanylate cyclase (sGC) stimulators normalized endothelial function and improved sensitivity to NO, pulmonary arterial pressure, and pulmonary capillary wedge pressure, producing an increase in cardiac output [99]. Both stimulation and activation were developed as mechanisms directly acting on sGC. Soluble GC stimulation may occur by sensitizing sGC to lower levels of NO by stabilizing the nitrosyl-heme complex, which results in a long-lasting active configuration of the enzyme, or by increasing sGC activity through NO-independent binding to the enzyme [100,101]. Vericiguat, a sGC stimulator, was developed and tested in HfrEF. The VICTORIA (the Vericiguat Global Study in Subjects with Heart Failure with Reduced Ejection Fraction) study, a phase 3, randomized, double-blind, placebo-controlled trial, investigated the effects of vericiguat on top of standard therapy (target dose 10 mg) in a population of 5050 patients with worsening HF, LVEF < 45%, and New York Heart Association class II–IV, who experienced hospitalization for HF within 6 months before randomization or needed intravenous diuretic therapy, without hospitalization, within the previous 3 months 93. Compared to the placebo, vericiguat significantly reduced the primary composite outcome of death from cardiovascular causes or first hospitalization for HF (−10%), the secondary endpoints of total hospitalizations for HF (−9%), and death from any cause or first hospitalization for HF (−10%) [102]. The enhancement of cardiovascular protective effects of NPs linked to cGMP activation could be enhanced in the long term with sGC stimulation although this needs to be tested and proven [96]. Based on these results, vericiguat was introduced as a therapeutic tool for resistant HF in the most recent European guidelines on HF management [62].

5. Novel NP-Based Therapeutic Strategies

Besides ARNi, different NP based therapeutic strategies were developed in the last few decades [103,104]. In the 1990s, recombinant human ANP (carperitide) and synthetic human BNP (nesiritide) were approved in Japan and the United States, respectively, for intravenous infusion in acute decompensated HF [105]. However, the phase III ASCEND-HF trial did not demonstrate significant benefits on cardiovascular outcomes in patients treated with nesiritide who otherwise developed severe hypotension [106]. Based on these results, the Food and Drug Administration recommended the discontinuation of the manufacture of nesiritide in 2018.
Cenderitide, designed as CNP added to the C-terminal tail of dendroaspis natriuretic peptide (DNP), was designed to stimulate both NPR-A and NPR-B and is under investigation to evaluate its effects on natriuresis, diuresis, cardiovascular unloading, and cardiac remodeling [107].
More recently, mutant-ANP (MANP) was shown to stimulate cGMP activity in human cardiomyocytes and endothelial cells and to produce sustained BP reduction in animal models and in the first human study without significant adverse effects. MANP is more resistant to neprilysin and insulin-induced proteolytic degradation compared to ANP, therefore showing more prolonged diuretic, natriuretic, and antihypertensive effects [108,109,110]. MANP also inhibits aldosterone synthesis via PDE2 by a reduction in intracellular Ca2+ levels [111].
Another fragment derived from NT-proANP, the proANP 31–67 peptide, has shown cardiorenal protective actions in a preclinical model of hypertension-mediated organ damage with cardiac hypertrophy and renal damage [112].
The therapeutic efficacy of BNP was proven in the context of experimental congestive heart failure where the coadministration of BNP and furosemide maximized natriuretic and diuretic responses, preserved renal function, and inhibited aldosterone activation [113]. Therefore, several attempts were performed to obtain peptides derived from BNP to be used in clinical practice. One of them, called CRRL269, is characterized by the ring structure of BNP and the C-terminal and N-terminal tails of urodilatin, one of the precursors of ANP, as well as an additional four amino acids on the N-terminal end. In cellular models, CRRL269 demonstrated a cGMP production equivalent to BNP and urodilatin in HEK cells overexpressing NPR-A [114]. NPA7, a peptide that replaces the N-terminal tail of BNP with Ang1–7 combining the stimulation of NPR-A and Mas receptors, is currently under investigation [115].
Additional designer peptides are represented by ASBNP.1, derived from an alternative spliced transcript form of BNP [116] and by the conjugated ANP consisting of ANP-Fc (ANP fused to the Fc domain of IgG), ANP-HAS (recombinant human ANP and human serum albumin), and CNP/ANP [117].
Sangaralingham and colleagues have recently identified the NPR-A(GC-A) positive allosteric modulator (PAM) MCUF-651 which could enhance the binding affinity of both ANP and BNP to the receptor with a dose-dependent augmentation of cGMP production [118].
Finally, innovative strategies are under development with the aim to increase the circulating NP level through genetic/epigenetic modulation and, particularly, through RNA-based targets including miR-425 and miR-155 [28,29,30].
Future studies conducted in large patient cohorts with long follow-ups will validate these novel compounds for the prevention and clinical management of hypertension, its related target organ damage, and HF. Moreover, the potential efficacy and safety of these molecules need to be investigated in combination with other drugs currently used in clinical practice.

6. Conclusions

NPs exert different endocrine effects on both cardiovascular and renal systems counterbalancing the overactivation of the renin-angiotensin-aldosterone and sympathetic nervous systems in HF and other CVD. Moreover, NPs exert anti-hypertrophic and anti-fibrotic autocrine effects through complex signaling pathways, therefore contributing in a significant manner to cardiovascular remodeling.
NPs represent an important diagnostic and prognostic tool to be used as a biomarker in the clinical management of HF and several other cardiovascular conditions. Therapeutic strategies based on NPs’ biological properties and on their underlying molecular mechanisms have been investigated or are currently under development. The clinical applications of the discoveries related to the NP system as well as the novel therapeutic strategies developed represent an excellent example of successful translational medicine and may prompt other innovative steps towards more targeted treatment of HF and other major cardiovascular disease conditions.

Author Contributions

G.G. contributed to writing—original draft preparation and data curation and S.R., C.A. and M.V. contributed to conceiving the paper, writing—review and editing, and supervision. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

There are no new data associated with this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Volpe, M.; Gallo, G.; Rubattu, S. Endocrine functions of the heart: From bench to bedside. Eur. Heart J. 2023, 44, 643–655. [Google Scholar] [CrossRef] [PubMed]
  2. Volpe, M.; Rubattu, S.; Burnett, J., Jr. Natriuretic peptides in cardiovascular diseases: Current use and perspectives. Eur. Heart J. 2014, 35, 419–425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Goetze, J.P.; Bruneau, B.G.; Ramos, H.R.; Ogawa, T.; de Bold, M.K.; de Bold, A.J. Cardiac natriuretic peptides. Nat. Rev. Cardiol. 2020, 17, 698–717. [Google Scholar] [CrossRef] [PubMed]
  4. Levin, E.R.; Gardner, D.G.; Samson, W.K. Natriuretic peptides. N. Engl. J. Med. 1998, 339, 321–328. [Google Scholar]
  5. Pandey, K.N. Biology of natriuretic peptides and their receptors. Peptides 2005, 26, 901–932. [Google Scholar] [CrossRef]
  6. Forte, M.; Madonna, M.; Schiavon, S.; Valenti, V.; Versaci, F.; Zoccai, G.B.; Frati, G.; Sciarretta, S. Cardiovascular Pleiotropic Effects of Natriuretic Peptides. Int. J. Mol. Sci. 2019, 20, 3874. [Google Scholar] [CrossRef] [Green Version]
  7. Ogawa, T.; Vatta, M.; Bruneau, B.G.; de Bold, A.J. Characterization of natriuretic peptide production by adult heart atria. Am. J. Physiol. 1999, 276, H1977–H1986. [Google Scholar] [CrossRef]
  8. Lugnier, C.; Meyer, A.; Charloux, A.; Andrès, E.; Gény, B.; Talha, S. The Endocrine Function of the Heart: Physiology and Involvements of Natriuretic Peptides and Cyclic Nucleotide Phosphodiesterases in Heart Failure. J. Clin. Med. 2019, 8, 1746. [Google Scholar] [CrossRef] [Green Version]
  9. Mangat, H.; de Bold, A.J. Stretch-induced atrial natriuretic factor release utilizes a rapidly depleting pool of newly synthesized hormone. Endocrinology 1993, 133, 1398–1403. [Google Scholar] [CrossRef]
  10. Dietz, J.R. Mechanisms of atrial natriuretic peptide secretion from the atrium. Cardiovasc. Res. 2005, 68, 8–17. [Google Scholar] [CrossRef] [Green Version]
  11. Semenov, A.G.; Tamm, N.N.; Seferian, K.R.; Postnikov, A.B.; Karpova, N.S.; Serebryanaya, D.V.; Koshkina, E.V.; Krasnoselsky, M.I.; Katrukha, A.G. Processing of pro-B-type natriuretic peptide: Furin and corin as candidate convertases. Clin. Chem. 2010, 56, 1166–1176. [Google Scholar] [CrossRef] [Green Version]
  12. Gupta, D.K.; Daniels, L.B.; Cheng, S.; deFilippi, C.R.; Criqui, M.H.; Maisel, A.S.; Lima, J.A.; Bahrami, H.; Greenland, P.; Cushman, M.; et al. Differences in Natriuretic Peptide Levels by Race/Ethnicity (From the Multi-Ethnic Study of Atherosclerosis). Am. J. Cardiol. 2017, 120, 1008–1015. [Google Scholar] [CrossRef]
  13. Patel, N.; Russell, G.K.; Musunuru, K.; Gutierrez, O.M.; Halade, G.; Kain, V.; Lv, W.; Prabhu, S.D.; Margulies, K.B.; Cappola, T.P.; et al. Race, Natriuretic Peptides, and High-Carbohydrate Challenge: A Clinical Trial. Circ. Res. 2019, 125, 957–968. [Google Scholar] [CrossRef]
  14. Luchner, A.; Stevens, T.L.; Borgeson, D.D.; Redfield, M.; Wei, C.M.; Porter, J.G.; Burnett, J.C., Jr. Differential atrial and ventricular expression of myocardial BNP during evolution of heart failure. Am. J. Physiol. 1998, 274, H1684–H1689. [Google Scholar] [CrossRef]
  15. Frustaci, A.; Russo, M.A.; Chimenti, C. Diagnostic contribution of left ventricular endomyocardial biopsy in patients with clinical phenotype of hypertrophic cardiomyopathy. Hum. Pathol. 2013, 44, 133–141. [Google Scholar] [CrossRef]
  16. Murakami, Y.; Shimada, T.; Inoue, S.; Shimizu, H.; Ohta, Y.; Katoh, H.; Nakamura, K.; Ishibashi, Y. New insights into the mechanism of the elevation of plasma brain natriuretic polypeptide levels in patients with left ventricular hypertrophy. Can. J. Cardiol. 2002, 18, 1294–1300. [Google Scholar]
  17. Ogawa, T.; Veinot, J.P.; Davies, R.A.; Haddad, H.; Smith, S.J.; Masters, R.G. Neuroendocrine profiling of humans receiving cardiac allografts. J. Heart Lung Transplant. 2005, 24, 1046–1054. [Google Scholar] [CrossRef]
  18. Masters, R.G.; Davies, R.A.; Veinot, J.P.; Hendry, P.J.; Smith, S.J.; de Bold, A.J. Discoordinate modulation of natriuretic peptides during acute cardiac allograft rejection in humans. Circulation 1999, 100, 287–291. [Google Scholar] [CrossRef] [Green Version]
  19. Kim, M.; Platt, M.J.; Shibasaki, T.; Quaggin, S.E.; Backx, P.H.; Seino, S.; Simpson, J.A.; Drucker, D.J. GLP-1 receptor activation and Epac2 link atrial natriuretic peptide secretion to control of blood pressure. Nat. Med. 2013, 19, 567–575. [Google Scholar] [CrossRef]
  20. Fish-Trotter, H.; Ferguson, J.F.; Patel, N.; Arora, P.; Allen, N.B.; Bachmann, K.N. Inflammation and Circulating Natriuretic Peptide Levels. Circ. Heart Fail. 2020, 13, e006570. [Google Scholar] [CrossRef]
  21. Richards, A.M. The renin-angiotensin-aldosterone system and the cardiac natriuretic peptides. Heart 1996, 76 (Suppl. 3), 36–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Undank, S.; Kaiser, J.; Sikimic, J.; Düfer, M.; Krippeit-Drews, P.; Drews, G. Atrial Natriuretic Peptide Affects Stimulus-Secretion Coupling of Pancreatic β-Cells. Diabetes 2017, 66, 2840–2848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Clerk, A.; Pham, F.H.; Fuller, S.J.; Sahai, E.; Aktories, K.; Marais, R.; Marshall, C.; Sugden, P.H. Regulation of mitogen-activated protein kinases in cardiac myocytes through the small G protein Rac1. Mol. Cell. Biol. 2001, 21, 1173–1184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Mahmoud, S.A.; Poizat, C. Epigenetics and chromatin remodeling in adult cardiomyopathy. J. Pathol. 2013, 231, 147–157. [Google Scholar] [CrossRef] [Green Version]
  25. Hohl, M.; Wagner, M.; Reil, J.C.; Müller, S.A.; Tauchnitz, M.; Zimmer, A.M.; Lehmann, L.; Thiel, G.; Böhm, M.; Backs, J.; et al. HDAC4 controls histone methylation in response to elevated cardiac load. J. Clin. Investig. 2013, 123, 1359–1370. [Google Scholar] [CrossRef] [Green Version]
  26. Rosales, W.; Lizcano, F. The Histone Demethylase JMJD2A Modulates the Induction of Hypertrophy Markers in iPSC-Derived Cardiomyocytes. Front. Genet. 2018, 9, 9–14. [Google Scholar] [CrossRef] [Green Version]
  27. Ito, E.; Miyagawa, S.; Fukushima, S.; Yoshikawa, Y.; Saito, S.; Saito, T.; Harada, A.; Takeda, M.; Kashiyama, N.; Nakamura, Y.; et al. Histone Modification Is Correlated with Reverse Left Ventricular Remodeling in Nonischemic Dilated Cardiomyopathy. Ann. Thorac. Surg. 2017, 104, 1531–1539. [Google Scholar] [CrossRef] [Green Version]
  28. Arora, S.; Rana, R.; Chhabra, A.; Jaiswal, A.; Rani, V. miRNA-transcription factor interactions: A combinatorial regulation of gene expression. Mol. Genet. Genom. 2013, 288, 77–87. [Google Scholar] [CrossRef]
  29. Arora, P.; Wu, C.; Hamid, T.; Arora, G.; Agha, O.; Allen, K. Acute Metabolic Influences on the Natriuretic Peptide System in Humans. J. Am. Coll. Cardiol. 2016, 67, 804–812. [Google Scholar] [CrossRef]
  30. Wu, C.; Arora, P.; Agha, O.; Hurst, L.A.; Allen, K.; Nathan, D.I.; Hu, D.; Jiramongkolchai, P.; Smith, J.G.; Melander, O.; et al. Novel MicroRNA Regulators of Atrial Natriuretic Peptide Production. Mol. Cell. Biol. 2016, 36, 1977–1987. [Google Scholar] [CrossRef] [Green Version]
  31. Newton-Cheh, C.; Larson, M.G.; Vasan, R.S.; Levy, D.; Bloch, K.D.; Surti, A.; Guiducci, C.; Kathiresan, S.; Benjamin, E.; Struck, J.; et al. Association of common variants in NPPA and NPPB with circulating natriuretic peptides and blood pressure. Nat. Genet. 2009, 41, 348–353. [Google Scholar] [CrossRef] [Green Version]
  32. Cannone, V.; Boerrigter, G.; Cataliotti, A.; Costello-Boerrigter, L.C.; Olson, T.M.; McKie, P.M.; Heublein, D.M.; Lahr, B.D.; Bailey, K.R.; Averna, M.; et al. A genetic variant of the atrial natriuretic peptide gene is associated with cardiometabolic protection in the general community. J. Am. Coll. Cardiol. 2011, 58, 629–633. [Google Scholar] [CrossRef] [Green Version]
  33. Cannone, V.; Cefalu’, A.B.; Noto, D.; Scott, C.G.; Bailey, K.R.; Cavera, G.; Pagano, M.; Sapienza, M.; Averna, M.R.; Burnett, J.C., Jr. The atrial natriuretic peptide genetic variant rs5068 is associated with a favorable cardiometabolic phenotype in a Mediterranean population. Diabetes Care 2013, 36, 2850–2856. [Google Scholar] [CrossRef] [Green Version]
  34. Celik, S.; Sadegh, M.K.; Morley, M.; Roselli, C.; Ellinor, P.T.; Cappola, T.; Smith, J.G.; Gidlof, O. Antisense regulation of atrial natriuretic peptide expression. JCI Insight 2019, 4, e130978. [Google Scholar] [CrossRef]
  35. Pandey, K.N. Molecular Signaling Mechanisms and Function of Natriuretic Peptide Receptor-A in the Pathophysiology of Cardiovascular Homeostasis. Front. Physiol. 2021, 12, 693099. [Google Scholar] [CrossRef]
  36. Airhart, N.; Yang, Y.F.; Roberts, C.T., Jr.; Silberbach, M. Atrial natriuretic peptide induces natriuretic peptide receptor-cGMP-dependent protein kinase interaction. J. Biol. Chem. 2003, 278, 38693–38698. [Google Scholar] [CrossRef] [Green Version]
  37. Moyes, A.J.; Khambata, R.S.; Villar, I.; Bubb, K.J.; Baliga, R.S.; Lumsden, N.G.; Xiao, F.; Gane, P.; Rebstock, A.-S.; Worthington, R.J.; et al. Endothelial C-type natriuretic peptide maintains vascular homeostasis. J. Clin. Investig. 2014, 124, 4039–4051. [Google Scholar] [CrossRef]
  38. Bayes-Genis, A.; Barallat, J.; Richards, A.M. A Test in Context: Neprilysin: Function, Inhibition, and Biomarker. J. Am. Coll. Cardiol. 2016, 68, 639–653. [Google Scholar] [CrossRef]
  39. Supaporn, T.; Sandberg, S.M.; Borgeson, D.D.; Heublein, D.M.; Luchner, A.; Wei, C.M.; Dousa, T.P.; Burnett, J.C., Jr. Blunted cGMP response to agonists and enhanced glomerular cyclic 3′,5′-nucleotide phosphodiesterase activities in experimental congestive heart failure. Kidney Int. 1996, 50, 1718–1725. [Google Scholar] [CrossRef] [Green Version]
  40. Francis, S.H.; Busch, J.L.; Corbin, J.D.; Sibley, D. cGMP-dependent protein kinases and cGMP phosphodiesterases in nitric oxide and cGMP action. Pharmacol. Rev. 2010, 62, 525–563. [Google Scholar] [CrossRef]
  41. Inserte, J.; Garcia-Dorado, D. The cGMP/PKG pathway as a common mediator of cardioprotection: Translatability and mechanism. Br. J. Pharmacol. 2015, 172, 1996–2009. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Nakao, K.; Kuwahara, K.; Nishikimi, T.; Nakagawa, Y.; Kinoshita, H.; Minami, T. Endothelium-Derived C-Type Natriuretic Peptide Contributes to Blood Pressure Regulation by Maintaining Endothelial Integrity. Hypertension 2017, 69, 286–296. [Google Scholar] [CrossRef] [PubMed]
  43. Kavurma, M.M.; Bursill, C.; Stanley, C.P.; Passam, F.; Cartland, S.P.; Patel, S.; Loa, J.; Figtree, G.A.; Golledge, J.; Aitken, S.; et al. Endothelial cell dysfunction: Implications for the pathogenesis of peripheral artery disease. Front. Cardiovasc. Med. 2022, 9, 1054576. [Google Scholar] [CrossRef] [PubMed]
  44. Greene, S.J.; Gheorghiade, M.; Borlaug, B.A.; Pieske, B.; Vaduganathan, M.; Burnett, J.C., Jr.; Roessig, L.; Stasch, J.P.; Solomon, S.D.; Paulus, W.J.; et al. The cGMP signaling pathway as a therapeutic target in heart failure with preserved ejection fraction. J. Am. Heart Assoc. 2013, 2, e000536. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Inserte, J.; Hernando, V.; Ruiz-Meana, M.; Poncelas-Nozal, M.; Fernandez, C.; Agullo, L.; Sartorio, C.; Vilardosa, U.; Garcia-Dorado, D. Delayed phospholamban phosphorylation in post-conditioned heart favours Ca2+ normalization and contributes to protection. Cardiovasc. Res. 2014, 103, 542–553. [Google Scholar] [CrossRef] [Green Version]
  46. Dorey, T.W.; Mackasey, M.; Jansen, H.J.; McRae, M.D.; Bohne, L.J.; Liu, Y.; Belke, D.D.; Atkinson, L.; Rose, R.A. Natriuretic peptide receptor B maintains heart rate and sinoatrial node function via cyclic GMP-mediated signalling. Cardiovasc. Res. 2022, 118, 1917–1931. [Google Scholar] [CrossRef]
  47. Calvieri, C.; Rubattu, S.; Volpe, M. Molecular mechanisms underlying cardiac antihypertrophic and antifibrotic effects of natriuretic peptides. J. Mol. Med. 2012, 90, 5–13. [Google Scholar] [CrossRef]
  48. Kiemer, A.K.; Weber, N.C.; Vollmar, A.M. Induction of IkappaB: Atrial natriuretic peptide as a regulator of the NF-kappaB pathway. Biochem. Biophys. Res. Commun. 2002, 295, 1068–1076. [Google Scholar] [CrossRef]
  49. Rubattu, S.; Gallo, G.; Volpe, M. A Contemporary View of Natriuretic Peptides in the SARS-CoV-2 Era. Front. Physiol. 2021, 12, 643721. [Google Scholar] [CrossRef]
  50. John, S.W.; Krege, J.H.; Oliver, P.M.; Hagaman, J.R.; Hodgin, J.B.; Pang, S.C.; Smithies, O. Genetic decreases in atrial natriuretic peptide and salt-sensitive hypertension. Science 1995, 267, 679–681. [Google Scholar] [CrossRef]
  51. Scott, N.J.; Ellmers, L.J.; Lainchbury, J.G.; Maeda, N.; Smithies, O.; Richards, A.M.; Cameron, V.A. Influence of natriuretic peptide receptor-1 on survival and cardiac hypertrophy during development. Biochim. Biophys. Acta 2009, 1792, 1175–1184. [Google Scholar] [CrossRef] [Green Version]
  52. Rubattu, S.; Bigatti, G.; Evangelista, A.; Lanzani, C.; Stanzione, R.; Zagato, L. Association of atrial natriuretic peptide and type a natriuretic peptide receptor gene polymorphisms with left ventricular mass in human essential hypertension. J. Am. Coll. Cardiol. 2006, 48, 499–505. [Google Scholar] [CrossRef] [Green Version]
  53. Tamura, N.; Ogawa, Y.; Chusho, H.; Nakamura, K.; Nakao, K.; Suda, M.; Kasahara, M.; Hashimoto, R.; Katsuura, G.; Mukoyama, M.; et al. Cardiac fibrosis in mice lacking brain natriuretic peptide. Proc. Natl. Acad. Sci. USA 2000, 97, 4239–4244. [Google Scholar] [CrossRef] [Green Version]
  54. Rubattu, S.; Stanzione, R.; Cotugno, M.; Bianchi, F.; Marchitti, S.; Forte, M. Epigenetic control of natriuretic peptides: Implications for health and disease. Cell. Mol. Life Sci. 2020, 77, 5121–5130. [Google Scholar] [CrossRef]
  55. Cataliotti, A.; Tonne, J.M.; Bellavia, D.; Martin, F.L.; Oehler, E.A.; Harders, G.E.; Campbell, J.M.; Peng, K.W.; Russell, S.J.; Malatino, L.S.; et al. Long-term cardiac pro-B-type natriuretic peptide gene delivery prevents the development of hypertensive heart disease in spontaneously hypertensive rats. Circulation 2011, 123, 1297–1305. [Google Scholar] [CrossRef]
  56. Ly, O.T.; Chen, H.; Brown, G.E.; Hong, L.; Wang, X.; Han, Y.D.; Pavel, M.A.; Sridhar, A.; Maienschein-Cline, M.; Chalazan, B.; et al. Mutant ANP induces mitochondrial and ion channel remodeling in a human iPSC-derived atrial fibrillation model. JCI Insight 2022, 7, e155640. [Google Scholar] [CrossRef]
  57. Abraham, R.L.; Yang, T.; Blair, M.; Roden, D.M.; Darbar, D. Augmented potassium current is a shared phenotype for two genetic defects associated with familial atrial fibrillation. J. Mol. Cell. Cardiol. 2010, 48, 181–190. [Google Scholar] [CrossRef] [Green Version]
  58. Forte, M.; Marchitti, S.; Di Nonno, F.; Stanzione, R.; Schirone, L.; Cotugno, M.; Bianchi, F.; Schiavon, S.; Raffa, S.; Ranieri, D.; et al. NPPA/atrial natriuretic peptide is an extracellular modulator of autophagy in the heart. Autophagy 2022. Online ahead of print. [Google Scholar] [CrossRef]
  59. Nakagawa, Y.; Nishikimi, T.; Kuwahara, K.; Fujishima, A.; Oka, S.; Tsutamoto, T.; Kinoshita, H.; Nakao, K.; Cho, K.; Inazumi, H.; et al. MiR30-GALNT1/2 Axis-Mediated Glycosylation Contributes to the Increased Secretion of Inactive Human Prohormone for Brain Natriuretic Peptide (proBNP) from Failing Hearts. J. Am. Heart Assoc. 2017, 6, e003601. [Google Scholar] [CrossRef] [Green Version]
  60. Rose, R.A.; Giles, W.R. Natriuretic peptide C receptor signalling in the heart and vasculature. J. Physiol. 2008, 586, 353–366. [Google Scholar] [CrossRef]
  61. Preedy, M.E.J. Cardiac Cyclic Nucleotide Phosphodiesterases: Roles and Therapeutic Potential in Heart Failure. Cardiovasc. Drugs Ther. 2020, 34, 401–417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. McDonagh, T.A.; Metra, M.; Adamo, M.; Gardner, R.S.; Baumbach, A.; Böhm, M.; Burri, H.; Butler, J.; Čelutkienė, J.; Chioncel, O.; et al. 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure. Eur. Heart J. 2021, 42, 3599–3726. [Google Scholar] [CrossRef] [PubMed]
  63. Lee, D.S.; Straus, S.E.; Farkouh, M.E.; Austin, P.C.; Taljaard, M.; Chong, A.; Fahim, C.; Poon, S.; Cram, P.; Smith, S.; et al. Trial of an Intervention to Improve Acute Heart Failure Outcomes. N. Engl. J. Med. 2023, 388, 22–32. [Google Scholar] [CrossRef] [PubMed]
  64. Pedicino, D.; Volpe, M. Improving care for heart failure patients by COACHing clinicians to use decision-support tools. Eur. Heart J. 2023, in press. [Google Scholar]
  65. Miller, W.L.; Hartman, K.A.; Grill, D.E.; Struck, J.; Bergmann, A.; Jaffe, A.S. Serial measurements of midregion proANP and copeptin in ambulatory patients with heart failure: Incremental prognostic value of novel biomarkers in heart failure. Heart 2012, 98, 389–394. [Google Scholar] [CrossRef] [Green Version]
  66. Jia, X.; Al Rifai, M.; Hoogeveen, R.; Echouffo-Tcheugui, J.B.; Shah, A.M.; Ndumele, C.E.; Virani, S.S.; Bozkurt, B.; Selvin, E.; Ballantyne, C.M.; et al. Association of Long-term Change in N-Terminal Pro-B-Type Natriuretic Peptide with Incident Heart Failure and Death. JAMA Cardiol. 2023. Online ahead of print. [Google Scholar] [CrossRef]
  67. Enia, G.; Mallamaci, F.; Benedetto, F.A.; Panuccio, V.; Parlongo, S.; Cutrupi, S.; Giacone, G.; Cottini, E.; Tripepi, G.; Malatino, L.S.; et al. Long-term CAPD patients are volume expanded and display more severe left ventricular hypertrophy than haemodialysis patients. Nephrol. Dial. Transplant. 2001, 16, 1459–1464. [Google Scholar] [CrossRef] [Green Version]
  68. Cataliotti, A.; Malatino, L.S.; Jougasaki, M.; Zoccali, C.; Castellino, P.; Giacone, G.; Bellanuova, I.; Tripepi, R.; Seminara, G.; Parlongo, S.; et al. Circulating natriuretic peptide concentrations in patients with end-stage renal disease: Role of brain natriuretic peptide as a biomarker for ventricular remodeling. Mayo Clin. Proc. 2001, 76, 1111–1119. [Google Scholar] [CrossRef]
  69. Mallamaci, F.; Zoccali, C.; Tripepi, G.; Benedetto, F.A.; Parlongo, S.; Cataliotti, A.; Cutrupi, S.; Giacone, G.; Bellanuova, I.; Stancanelli, B.; et al. The Cardiovascular Risk Extended Evaluation. Diagnostic potential of cardiac natriuretic peptides in dialysis patients. Kidney Int. 2001, 59, 1559–1566. [Google Scholar] [CrossRef] [Green Version]
  70. Paget, V.; Legedz, L.; Gaudebout, N.; Girerd, N.; Bricca, G.; Milon, H.; Vincent, M.; Lantelme, P. N-terminal pro-brain natriuretic peptide: A powerful predictor of mortality in hypertension. Hypertension 2011, 57, 702–709. [Google Scholar] [CrossRef] [Green Version]
  71. Belluardo, P.; Cataliotti, A.; Bonaiuto, L.; Giuffrè, E.; Maugeri, E.; Noto, P.; Orlando, G.; Raspa, G.; Piazza, B.; Babuin, L.; et al. Lack of activation of molecular forms of the BNP system in human grade 1 hypertension and relationship to cardiac hypertrophy. Am. J. Physiol. Heart Circ. Physiol. 2006, 291, H1529–H1535. [Google Scholar] [CrossRef] [Green Version]
  72. Macheret, F.; Heublein, D.; Costello-Boerrigter, L.C.; Boerrigter, G.; McKie, P.; Bellavia, D.; Mangiafico, S.; Ikeda, Y.; Bailey, K.; Scott, C.G.; et al. Human hypertension is characterized by a lack of activation of the antihypertensive cardiac hormones ANP and BNP. J. Am. Coll. Cardiol. 2012, 60, 1558–1565. [Google Scholar] [CrossRef] [Green Version]
  73. Hijazi, Z.; Lindahl, B.; Oldgren, J.; Andersson, U.; Lindbäck, J.; Granger, C.B.; Alexander, J.H.; Gersh, B.J.; Hanna, M.; Harjola, V.P.; et al. Repeated Measurements of Cardiac Biomarkers in Atrial Fibrillation and Validation of the ABC Stroke Score Over Time. J. Am. Heart Assoc. 2017, 6, e004851. [Google Scholar] [CrossRef] [Green Version]
  74. Barbato, E.; Rubattu, S.; Bartunek, J.; Berni, A.; Sarno, G.; Vanderheyden, M.; Delrue, L.; Zardi, D.; Pace, B.; De Bruyne, B.; et al. Human coronary atherosclerosis modulates cardiac natriuretic peptide release. Atherosclerosis 2009, 206, 258–264. [Google Scholar] [CrossRef]
  75. Barbato, E.; Bartunek, J.; Marchitti, S.; Mangiacapra, F.; Stanzione, R.; Delrue, L.; Cotugno, M.; Di Castro, S.; De Bruyne, B.; Wijns, W.; et al. NT-proANP circulating level is a prognostic marker in stable ischemic heart disease. Int. J. Cardiol. 2012, 155, 311–312. [Google Scholar] [CrossRef]
  76. Stewart, R.A.H.; Kirby, A.; White, H.D.; Marschner, S.L.; West, M.; Thompson, P.L.; Sullivan, D.; Janus, E.; Hunt, D.; Kritharides, L.; et al. B-Type Natriuretic Peptide and Long-Term Cardiovascular Mortality in Patients with Coronary Heart Disease. J. Am. Heart Assoc. 2022, 11, e024616. [Google Scholar] [CrossRef]
  77. Gallo, G.; Presta, V.; Volpe, M.; Rubattu, S. Molecular and clinical implications of natriuretic peptides in aortic valve stenosis. J. Mol. Cell. Cardiol. 2019, 129, 266–271. [Google Scholar] [CrossRef]
  78. Gallo, G.; Forte, M.; Stanzione, R.; Cotugno, M.; Bianchi, F.; Marchitti, S.; Berni, A.; Volpe, M.; Rubattu, S. Functional Role of Natriuretic Peptides in Risk Assessment and Prognosis of Patients with Mitral Regurgitation. J. Clin. Med. 2020, 9, 1348. [Google Scholar] [CrossRef]
  79. Helgeson, S.A.; Imam, J.S.; Moss, J.E.; Hodge, D.O.; Burger, C.D. Comparison of Brain Natriuretic Peptide Levels to Simultaneously Obtained Right Heart Hemodynamics in Stable Outpatients with Pulmonary Arterial Hypertension. Diseases 2018, 6, 33. [Google Scholar] [CrossRef] [Green Version]
  80. Benza, R.L.; Miller, D.P.; Gomberg-Maitland, M.; Frantz, R.P.; Foreman, A.J.; Coffey, C.S.; Frost, A.; Barst, R.J.; Badesch, D.B.; Elliott, C.G.; et al. Predicting survival in pulmonary arterial hypertension: Insights from the Registry to Evaluate Early and Long-Term Pulmonary Arterial Hypertension Disease Management (REVEAL). Circulation 2010, 122, 164–172. [Google Scholar] [CrossRef] [Green Version]
  81. Romaniello, A.; Rubattu, S.; Gigante, A.; Simonelli, F.; Grimaldi, M.C.; D’Angelo, A.; Alunni, D.; Sada, L.; Gasperini, M.L.; Marchitti, S.; et al. Atrial natriuretic peptide predicts disease progression and digital ulcers development in systemic sclerosis patients. J. Cardiovasc. Med. 2019, 20, 771–779. [Google Scholar] [CrossRef] [PubMed]
  82. Romaniello, A.; Rubattu, S.; Vaiarello, V.; Gigante, A.; Volpe, M.; Rosato, E. Circulating NT-proANP level is a predictor of mortality for systemic sclerosis: A retrospective study of an Italian cohort. Expert Rev. Clin. Immunol. 2021, 17, 661–666. [Google Scholar] [CrossRef] [PubMed]
  83. Zoccali, C.; Mallamaci, F.; Benedetto, F.A.; Tripepi, G.; Parlongo, S.; Cataliotti, A.; Cutrupi, S.; Giacone, G.; Bellanuova, I.; Cottini, E.; et al. Cardiac natriuretic peptides are related to left ventricular mass and function and predict mortality in dialysis patients. J. Am. Soc. Nephrol. 2001, 12, 1508–1515. [Google Scholar] [CrossRef] [PubMed]
  84. McMurray, J.J.; Packer, M.; Desai, A.S.; Gong, J.; Lefkowitz, M.P.; Rizkala, A.R.; Rouleau , J.L.; Shi , V.C.; Solomon , S.D.; Swedberg , K.; et al. PARADIGM-HF Investigators and Committees. Angiotensin-neprilysin inhibition versus enalapril in heart failure. N. Engl. J. Med. 2014, 371, 993–1004. [Google Scholar] [CrossRef] [Green Version]
  85. Zile, M.R.; Claggett, B.L.; Prescott, M.F.; McMurray, J.J.; Packer, M.; Rouleau, J.L.; Swedberg, K.; Desai, A.S.; Gong, J.; Shi, V.C.; et al. Prognostic Implications of Changes in N-Terminal Pro-B-Type Natriuretic Peptide in Patients with Heart Failure. J. Am. Coll. Cardiol. 2016, 68, 2425–2436. [Google Scholar] [CrossRef] [Green Version]
  86. Myhre, P.L.; Vaduganathan, M.; Claggett, B.; Packer, M.; Desai, A.S.; Rouleau, J.L.; Zile, M.R.; Swedberg, K.; Lefkowitz, M.; Shi, V.; et al. B-Type Natriuretic Peptide during Treatment with Sacubitril/Valsartan: The PARADIGM-HF Trial. J. Am. Coll. Cardiol. 2019, 73, 1264–1272. [Google Scholar] [CrossRef]
  87. Senni, M.; Wachter, R.; Witte, K.K.; Straburzynska-Migaj, E.; Belohlavek, J.; Fonseca, C.; Mueller, C.; Lonn, E.; Chakrabarti, A.; Bao, W.; et al. TRANSITION Investigators. Initiation of sacubitril/valsartan shortly after hospitalisation for acutely decompensated heart failure in patients with newly diagnosed (de novo) heart failure: A subgroup analysis of the TRANSITION study. Eur. J. Heart Fail. 2020, 22, 303–312. [Google Scholar] [CrossRef]
  88. Berg, D.D.; Samsky, M.D.; Velazquez, E.J.; Duffy, C.I.; Gurmu, Y.; Braunwald, E.; Morrow, D.A.; DeVore, A.D. 2 Efficacy and Safety of Sacubitril/Valsartan in High-Risk Patients in the PIONEER-HF Trial. Circ. Heart Fail. 2021, 14, e007034. [Google Scholar] [CrossRef]
  89. Senni, M.; McMurray, J.J.; Wachter, R.; McIntyre, H.F.; Reyes, A.; Majercak, I.; Andreka, P.; Shehova-Yankova, N.; Anand, I.; Yilmaz, M.B.; et al. Initiating sacubitril/valsartan (LCZ696) in heart failure: Results of TITRATION, a double-blind, randomized comparison of two uptitration regimens. Eur. J. Heart Fail. 2016, 18, 1193–1202. [Google Scholar] [CrossRef] [Green Version]
  90. Solomon, S.D.; McMurray, J.J.V.; Anand, I.S.; Ge, J.; Lam, C.S.P.; Maggioni, A.P. PARAGON-HF Investigators and Committees. Angiotensin-Neprilysin Inhibition in Heart Failure with Preserved Ejection Fraction. N. Engl. J. Med. 2019, 381, 1609–1620. [Google Scholar] [CrossRef] [Green Version]
  91. Solomon, S.D.; Vaduganathan, M.; LClaggett, B.; Packer, M.; Zile, M.; Swedberg, K.; Rouleau, J.; Pfeffer, M.A.; Desai, A.; Lund, L.H.; et al. Sacubitril/Valsartan Across the Spectrum of Ejection Fraction in Heart Failure. Circulation 2020, 141, 352–361. [Google Scholar] [CrossRef]
  92. Volpe, M.; Bauersachs, J.; Bayés-Genís, A.; Butler, J.; Cohen-Solal, A.; Gallo, G.; Deichl, A.S.; Khan, M.S.; Battistoni, A.; Pieske, B.; et al. Sacubitril/valsartan for the management of heart failure: A perspective viewpoint on current evidence. Int. J. Cardiol. 2021, 327, 138–145. [Google Scholar] [CrossRef]
  93. Gallo, G.; Volpe, M.; Battistoni, A.; Russo, D.; Tocci, G.; Musumeci, M.B. Sacubitril/Valsartan as a Therapeutic Tool Across the Range of Heart Failure Phenotypes and Ejection Fraction Spectrum. Front. Physiol. 2021, 12, 652163. [Google Scholar] [CrossRef]
  94. Pfeffer, M.A.; Claggett, B.; Lewis, E.F.; Granger, C.B.; Køber, L.; Maggioni, A.P.; Mann, D.L.; McMurray, J.J.; Rouleau, J.L.; Solomon, S.D.; et al. PARADISE-MI Investigators and Committees. Angiotensin Receptor-Neprilysin Inhibition in Acute Myocardial Infarction. N. Engl. J. Med. 2021, 385, 1845–1855. [Google Scholar] [CrossRef]
  95. Berwanger, O.; Pfeffer, M.; Claggett, B.; Jering, K.S.; Maggioni, A.P.; Steg, P.G.; Mehran, R.; Lewis, E.F.; Zhou, Y.; van der Meer, P.; et al. Sacubitril/valsartan versus ramipril for patients with acute myocardial infarction: Win-ratio analysis of the PARADISE-MI trial. Eur. J. Heart Fail. 2022, 24, 1918–1927. [Google Scholar] [CrossRef]
  96. Januzzi, J.L.; Prescott, M.F.; Butler, J.; Felker, G.M.; Maisel, A.S.; McCague, K.; Camacho, A.; Pina, I.L.; Rocha, R.A.; Shah, A.M.; et al. PROVE-Hf Investigators. Association of change in N-terminal pro-B-type natriuretic peptide following initiation of sacubitril-valsartan treatment with cardiac structure and function in patients with heart failure with reduced ejection fraction. JAMA 2019, 322, 1085–1095. [Google Scholar] [CrossRef]
  97. Desai, A.S.; Solomon, S.D.; Shah, A.M.; Claggett, B.L.; Fang, J.C.; Izzo, J.; McCague, K.; Abbas, C.A.; Rocha, R.; Mitchell, G.F.; et al. Effect of Sacubitril-Valsartan vs. Enalapril on Aortic Stiffness in Patients with Heart Failure and Reduced Ejection Fraction: A Randomized Clinical Trial. JAMA 2019, 322, 1077–1084. [Google Scholar] [CrossRef]
  98. Zile, M.R.; O’Meara, E.; Claggett, B.; Prescott, M.F.; Solomon, S.D.; Swedberg, K.; Packer, M.; McMurray, J.J.V.; Shi, V.; Lefkowitz, M.; et al. Effects of Sacubitril/Valsartan on Biomarkers of Extracellular Matrix Regulation in Patients with HFrEF. J. Am. Coll. Cardiol. 2019, 73, 795–806. [Google Scholar] [CrossRef]
  99. Stasch, J.P.; Becker, E.M.; Alonso-Alija, C.; Apeler, H.; Dembowsky, K.; Feurer, A.; Gerzer, R.; Minuth, T.; Perzborn, E.; Pleiss, U.; et al. NO independent regulatory site on soluble guanylate cyclase. Nature 2001, 410, 212–215. [Google Scholar] [CrossRef]
  100. Schmidt, P.; Schramm, M.; Schroder, H.; Stasch, J.P. Mechanisms of nitric oxide independent activation of soluble guanylyl cyclase. Eur. J. Pharmacol. 2003, 468, 167–174. [Google Scholar] [CrossRef]
  101. Boerrigter, G.; Costello-Boerrigter, L.C.; Cataliotti, A.; Lapp, H.; Stasch, J.P.; Burnett, J.C., Jr. Targeting heme-oxidized soluble guanylate cyclase in experimental heart failure. Hypertension 2007, 49, 1128–1133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Armstrong, P.W.; Pieske, B.; Anstrom, K.J.; Ezekowitz, J.; Hernandez, A.F.; Butler, J.; Lam, C.S.P.; Ponikowski, P.; Voors, A.A.; Jia, G.; et al. Vericiguat in Patients with Heart Failure and Reduced Ejection Fraction. N. Engl. J. Med. 2020, 382, 1883–1893. [Google Scholar] [CrossRef] [PubMed]
  103. Gallo, G.; Rubattu, S.; Volpe, M. Targeting Cyclic Guanylate Monophosphate in Resistant Hypertension and Heart Failure: Are Sacubitril/Valsartan and Vericiguat Synergistic and Effective in Both Conditions? High Blood Press. Cardiovasc. Prev. 2021, 28, 541–545. [Google Scholar] [CrossRef] [PubMed]
  104. Rubattu, S.; Gallo, G. The Natriuretic Peptides for Hypertension Treatment. High Blood Press. Cardiovasc. Prev. 2022, 29, 15–21. [Google Scholar] [CrossRef]
  105. Tanaka, T.D.; Sawano, M.; Ramani, R.; Friedman, M.; Kohsaka, S. Acute heart failure management in the USA and Japan: Overview of practice patterns and review of evidence. ESC Heart Fail. 2018, 5, 931–947. [Google Scholar] [CrossRef]
  106. Ezekowitz, J.A.; Hernandez, A.F.; Starling, R.C.; Yancy, C.W.; Massie, B.; Hill, J.A.; Krum, H.; Diaz, R.; Ponikowski, P.; Metra, M.; et al. Standardizing care for acute decompensated heart failure in a large megatrial: The approach for the Acute Studies of Clinical Effectiveness of Nesiritide in Subjects with Decompensated Heart Failure (ASCEND-HF). Am. Heart J. 2009, 157, 219–228. [Google Scholar] [CrossRef]
  107. Lee, C.Y.; Huntley, B.K.; McCormick, D.J.; Ichiki, T.; Sangaralingham, S.J.; Lisy, O.; Burnett, J.C., Jr. Cenderitide: Structural requirements for the creation of a novel dual particulate guanylyl cyclase receptor agonist with renal-enhancing in vivo and ex vivo actions. Eur. Heart J. Cardiovasc. Pharmacother. 2016, 2, 98–105. [Google Scholar] [CrossRef] [Green Version]
  108. Chen, H.H.; Wan, S.H.; Iyer, S.R.; Cannone, V.; Sangaralingham, S.J.; Nuetel, J. First-in-Human Study of MANP: A Novel ANP (Atrial Natriuretic Peptide) Analog in Human Hypertension. Hypertension 2021, 78, 1859–1867. [Google Scholar] [CrossRef]
  109. Cannone, V.; Burnett, J.C., Jr. Natriuretic Peptides and Blood Pressure Homeostasis: Implications for MANP, a Novel Guanylyl Cyclase a Receptor Activator for Hypertension. Front. Physiol. 2022, 12, 815796. [Google Scholar] [CrossRef]
  110. Volpe, M.; Gallo, G.; Rubattu, S. Novel ANP (Atrial Natriuretic Peptide)-Based Therapy for Hypertension: The Promising Role of a Disease Mechanism Targeted Approach. Hypertension 2021, 78, 1868–1870. [Google Scholar] [CrossRef]
  111. Chen, Y.; Iyer, S.R.; Nikolaev, V.O.; Naro, F.; Pellegrini, M.; Cardarelli, S.; Ma, X.; Lee, H.C.; Burnett, J.C., Jr. MANP Activation of the cGMP Inhibits Aldosterone via PDE2 and CYP11B2 in H295R Cells and in Mice. Hypertension 2022, 79, 1702–1712. [Google Scholar] [CrossRef] [PubMed]
  112. Altara, R.; da Silva, G.J.J.; Frisk, M.; Spelta, F.; Zouein, F.A.; Louch, W.E.; Booz, G.W.; Cataliotti, A. Cardioprotective Effects of the Novel Compound Vastiras in a Preclinical Model of End-Organ Damage. Hypertension 2020, 75, 1195–1204. [Google Scholar] [CrossRef] [PubMed]
  113. Cataliotti, A.; Boerrigter, G.; Costello-Boerrigter, L.C.; Schirger, J.A.; Tsuruda, T.; Heublein, D.M.; Chen, H.H.; Malatino, L.S.; Burnett, J.C., Jr. Brain natriuretic peptide enhances renal actions of furosemide and suppresses furosemide-induced aldosterone activation in experimental heart failure. Circulation 2004, 109, 1680–1685. [Google Scholar] [CrossRef] [Green Version]
  114. Chen, Y.; Harty, G.J.; Huntley, B.K.; Iyer, S.R.; Heublein, D.M.; Harders, G.E.; Meems, L.; Pan, S.; Sangaralingham, S.J.; Ichiki, T.; et al. CRRL269: A novel designer and renal-enhancing pGC-A peptide activator. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2018, 314, R407–R414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Meems, L.M.G.; Andersen, I.A.; Pan, S.; Harty, G.; Chen, Y.; Zheng, Y.; Harders, G.E.; Ichiki, T.; Heublein, D.M.; Iyer, S.R.; et al. Design, Synthesis, and Actions of an Innovative Bispecific Designer Peptide. Hypertension 2019, 73, 900–909. [Google Scholar] [CrossRef]
  116. Pan, S.; Chen, H.H.; Dickey, D.M.; Boerrigter, G.; Lee, C.; Kleppe, L.S.; Hall, J.L.; Lerman, A.; Redfield, M.M.; Potter, L.R.; et al. Biodesign of a renal-protective peptide based on alternative splicing of B-type natriuretic peptide. Proc. Natl. Acad. Sci. USA 2009, 106, 11282–11287. [Google Scholar] [CrossRef] [Green Version]
  117. Mezo, A.R.; McDonnell, K.A.; Low, S.C.; Song, J.; Reidy, T.J.; Lu, Q.; Amari, J.V.; Hoehn, T.; Peters, R.T.; Dumont, J.; et al. Atrial natriuretic peptide-Fc, ANP-Fc, fusion proteins: Semisynthesis, in vitro activity and pharmacokinetics in rats. Bioconjugate Chem. 2012, 23, 518–526. [Google Scholar] [CrossRef]
  118. Sangaralingham, S.J.; Whig, K.; Peddibhotla, S.; Kirby, R.J.; Sessions, H.E.; Maloney, P.R.; Hershberger, P.M.; Mose-Yates, H.; Hood, B.L.; Vasile, S.; et al. Discovery of small molecule guanylyl cyclase A receptor positive allosteric modulators. Proc. Natl. Acad. Sci. USA 2021, 118, e2109386118. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of the natriuretic peptide system from secretion to biological actions. Legend to Figure 1. NPs are produced by cardiomyocytes upon hemodynamic and neuroendocrine stimuli. Cardiomyocytes store prohormones, namely proANP and proBNP, that are then processed to the biologically active peptides ANP and BNP. NPs act on target cells through specific cGMP-coupled receptors.
Figure 1. Schematic representation of the natriuretic peptide system from secretion to biological actions. Legend to Figure 1. NPs are produced by cardiomyocytes upon hemodynamic and neuroendocrine stimuli. Cardiomyocytes store prohormones, namely proANP and proBNP, that are then processed to the biologically active peptides ANP and BNP. NPs act on target cells through specific cGMP-coupled receptors.
Ijms 24 05131 g001
Figure 2. Anti-hypertrophic and anti-fibrotic effects of natriuretic peptides. Legend to Figure 2: ANPs and BNPs antagonize calcineurin/NFAT, NHE-1, TGF-β1, and angiotensin II signaling. ANP, atrial natriuretic peptide; cGMP, cyclic guanosine monophosphate; ERK, extracellular-signal-regulated kinase; JAK, Janus kinase; JNK, c-JUN N-terminal kinase; MAPK, p38 mitogen-activated protein kinase; MEK, mitogen-activated protein kinase; NFAT, nuclear factor of activated T cells; NHE, Na+/H+ exchanger; NPR-A, type A natriuretic peptide receptor; NP, natriuretic peptide; p70S6K, ribosomal protein S6 kinase beta-1; PKG, protein kinase G; PyK2, proline-rich tyrosine kinase 2; Rac1, Ras-related C3 botulinum toxin substrate; Smad, suppressor of mothers against decapentaplegic; STAT3 signal transducer and activator of transcription 3; TGF, tumor growth factor.
Figure 2. Anti-hypertrophic and anti-fibrotic effects of natriuretic peptides. Legend to Figure 2: ANPs and BNPs antagonize calcineurin/NFAT, NHE-1, TGF-β1, and angiotensin II signaling. ANP, atrial natriuretic peptide; cGMP, cyclic guanosine monophosphate; ERK, extracellular-signal-regulated kinase; JAK, Janus kinase; JNK, c-JUN N-terminal kinase; MAPK, p38 mitogen-activated protein kinase; MEK, mitogen-activated protein kinase; NFAT, nuclear factor of activated T cells; NHE, Na+/H+ exchanger; NPR-A, type A natriuretic peptide receptor; NP, natriuretic peptide; p70S6K, ribosomal protein S6 kinase beta-1; PKG, protein kinase G; PyK2, proline-rich tyrosine kinase 2; Rac1, Ras-related C3 botulinum toxin substrate; Smad, suppressor of mothers against decapentaplegic; STAT3 signal transducer and activator of transcription 3; TGF, tumor growth factor.
Ijms 24 05131 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Gallo, G.; Rubattu, S.; Autore, C.; Volpe, M. Natriuretic Peptides: It Is Time for Guided Therapeutic Strategies Based on Their Molecular Mechanisms. Int. J. Mol. Sci. 2023, 24, 5131. https://doi.org/10.3390/ijms24065131

AMA Style

Gallo G, Rubattu S, Autore C, Volpe M. Natriuretic Peptides: It Is Time for Guided Therapeutic Strategies Based on Their Molecular Mechanisms. International Journal of Molecular Sciences. 2023; 24(6):5131. https://doi.org/10.3390/ijms24065131

Chicago/Turabian Style

Gallo, Giovanna, Speranza Rubattu, Camillo Autore, and Massimo Volpe. 2023. "Natriuretic Peptides: It Is Time for Guided Therapeutic Strategies Based on Their Molecular Mechanisms" International Journal of Molecular Sciences 24, no. 6: 5131. https://doi.org/10.3390/ijms24065131

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop