Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
FASEB J ; 35(5): e21544, 2021 05.
Article in English | MEDLINE | ID: mdl-33819356

ABSTRACT

Serine-rich splicing factor 3 (SRSF3) was recently reported as being necessary to preserve RNA stability via an mTOR mechanism in a cardiac mouse model in adulthood. Here, we demonstrate the link between Srsf3 and mitochondrial integrity in an embryonic cardiomyocyte-specific Srsf3 conditional knockout (cKO) mouse model. Fifteen-day-old Srsf3 cKO mice showed dramatically reduced (below 50%) survival and reduced the left ventricular systolic performance, and histological analysis of these hearts revealed a significant increase in cardiomyocyte size, confirming the severe remodeling induced by Srsf3 deletion. RNA-seq analysis of the hearts of 5-day-old Srsf3 cKO mice revealed early changes in expression levels and alternative splicing of several transcripts related to mitochondrial integrity and oxidative phosphorylation. Likewise, the levels of several protein complexes of the electron transport chain decreased, and mitochondrial complex I-driven respiration of permeabilized cardiac muscle fibers from the left ventricle was impaired. Furthermore, transmission electron microscopy analysis showed disordered mitochondrial length and cristae structure. Together with its indispensable role in the physiological maintenance of mouse hearts, these results highlight the previously unrecognized function of Srsf3 in regulating the mitochondrial integrity.


Subject(s)
Gene Expression Regulation , Mitochondria, Heart/pathology , Myocytes, Cardiac/pathology , Serine-Arginine Splicing Factors/physiology , Alternative Splicing , Animals , Gene Expression Profiling , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Oxidative Phosphorylation , RNA-Seq
2.
Int J Mol Sci ; 23(5)2022 Feb 24.
Article in English | MEDLINE | ID: mdl-35269644

ABSTRACT

Transient receptor potential canonical (TRPC) channels are membrane proteins involved in regulating Ca2+ homeostasis, and whose functions are modulated by G protein-coupled receptors (GPCR). In this study, we developed bioluminescent resonance energy transfer (BRET) biosensors to better study channel conformational changes following receptor activation. For this study, two intramolecular biosensors, GFP10-TRPC7-RLucII and RLucII-TRPC7-GFP10, were constructed and were assessed following the activation of various GPCRs. We first transiently expressed receptors and the biosensors in HEK293 cells, and BRET levels were measured following agonist stimulation of GPCRs. The activation of GPCRs that engage Gαq led to a Gαq-dependent BRET response of the functional TRPC7 biosensor. Focusing on the Angiotensin II type-1 receptor (AT1R), GFP10-TRPC7-RLucII was tested in rat neonatal cardiac fibroblasts, expressing endogenous AT1R and TRPC7. We detected similar BRET responses in these cells, thus validating the use of the biosensor in physiological conditions. Taken together, our results suggest that activation of Gαq-coupled receptors induce conformational changes in a novel and functional TRPC7 BRET biosensor.


Subject(s)
Bioluminescence Resonance Energy Transfer Techniques , Biosensing Techniques , Animals , Bioluminescence Resonance Energy Transfer Techniques/methods , Biosensing Techniques/methods , HEK293 Cells , Humans , Rats , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism
3.
Am J Physiol Heart Circ Physiol ; 320(4): H1646-H1656, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33635165

ABSTRACT

Apelin receptor (APJ) activation by apelin-13 (APLN-13) engages both Gαi proteins and ß-arrestins, stimulating distinct intracellular pathways and triggering physiological responses like enhanced cardiac contractility. Substituting the C-terminal phenylalanine of APLN-13 with α-methyl-l-phenylalanine [(l-α-Me)Phe] or p-benzoyl-l-phenylalanine (Bpa) generates biased analogs inducing APJ functional selectivity toward Gαi proteins. Using these original analogs, we proposed to investigate how the canonical Gαi signaling of APJ regulates the cardiac function and to assess their therapeutic impact in a rat model of isoproterenol-induced myocardial dysfunction. In vivo and ex vivo infusions of either Bpa or (l-α-Me)Phe analogs failed to enhance rats' left ventricular (LV) contractility compared with APLN-13. Inhibition of Gαi with pertussis toxin injection optimized the cardiotropic effect of APLN-13 and revealed the inotropic impact of Bpa. Moreover, both APLN-13 and Bpa efficiently limited the forskolin-induced and PKA-dependent phosphorylation of phospholamban at the Ser16 in neonatal rat ventricular myocytes. However, only Bpa significantly reduced the inotropic effect of forskolin infusion in isolated-perfused heart, highlighting its efficient bias toward Gαi. Compared with APLN-13, Bpa also markedly improved isoproterenol-induced myocardial systolic and diastolic dysfunctions. Bpa prevented cardiac weight increase, normalized both ANP and BNP mRNA expressions, and decreased LV fibrosis in isoproterenol-treated rats. Our results show that APJ-driven Gαi/adenylyl cyclase signaling is functional in cardiomyocytes and acts as negative feedback of the APLN-APJ-dependent inotropic response. Biased APJ signaling toward Gαi over the ß-arrestin pathway offers a promising strategy in the treatment of cardiovascular diseases related to myocardial hypertrophy and high catecholamine levels.NEW & NOTEWORTHY By using more potent Gαi-biased APJ agonists that strongly inhibit cAMP production, these data point to the negative inotropic effect of APJ-mediated Gαi signaling in the heart and highlight the potential protective impact of APJ-dependent Gαi signaling in cardiovascular diseases associated with left ventricular hypertrophy.


Subject(s)
Apelin Receptors/agonists , Apelin/pharmacology , GTP-Binding Protein alpha Subunits/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Myocytes, Cardiac/drug effects , Ventricular Dysfunction, Left/prevention & control , Ventricular Function, Left/drug effects , Adenylyl Cyclases/metabolism , Animals , Apelin/analogs & derivatives , Apelin Receptors/metabolism , Calcium-Binding Proteins/metabolism , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Isolated Heart Preparation , Isoproterenol , Ligands , Male , Myocytes, Cardiac/metabolism , Phosphorylation , Rats, Sprague-Dawley , Signal Transduction , Ventricular Dysfunction, Left/chemically induced , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/physiopathology
4.
Pharmacol Res ; 131: 7-16, 2018 05.
Article in English | MEDLINE | ID: mdl-29530600

ABSTRACT

The apelinergic system is an important player in the regulation of both vascular tone and cardiovascular function, making this physiological system an attractive target for drug development for hypertension, heart failure and ischemic heart disease. Indeed, apelin exerts a positive inotropic effect in humans whilst reducing peripheral vascular resistance. In this study, we investigated the signaling pathways through which apelin exerts its hypotensive action. We synthesized a series of apelin-13 analogs whereby the C-terminal Phe13 residue was replaced by natural or unnatural amino acids. In HEK293 cells expressing APJ, we evaluated the relative efficacy of these compounds to activate Gαi1 and GαoA G-proteins, recruit ß-arrestins 1 and 2 (ßarrs), and inhibit cAMP production. Calculating the transduction ratio for each pathway allowed us to identify several analogs with distinct signaling profiles. Furthermore, we found that these analogs delivered i.v. to Sprague-Dawley rats exerted a wide range of hypotensive responses. Indeed, two compounds lost their ability to lower blood pressure, while other analogs significantly reduced blood pressure as apelin-13. Interestingly, analogs that did not lower blood pressure were less effective at recruiting ßarrs. Finally, using Spearman correlations, we established that the hypotensive response was significantly correlated with ßarr recruitment but not with G protein-dependent signaling. In conclusion, our results demonstrated that the ßarr recruitment potency is involved in the hypotensive efficacy of activated APJ.


Subject(s)
Antihypertensive Agents/pharmacology , Apelin Receptors/metabolism , Blood Pressure/drug effects , Intercellular Signaling Peptides and Proteins/pharmacology , beta-Arrestins/metabolism , Animals , Antihypertensive Agents/chemistry , Cyclic AMP/metabolism , HEK293 Cells , Humans , Hypotension/drug therapy , Hypotension/metabolism , Intercellular Signaling Peptides and Proteins/chemistry , Male , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects
5.
Crit Care ; 22(1): 10, 2018 01 19.
Article in English | MEDLINE | ID: mdl-29347994

ABSTRACT

Catecholamines, in concert with fluid resuscitation, have long been recommended in the management of septic shock. However, not all patients respond positively and controversy surrounding the efficacy-to-safety profile of catecholamines has emerged, trending toward decatecholaminization. Contextually, it is time to re-examine the "maintaining blood pressure" paradigm by identifying safer and life-saving alternatives. We put in perspective the emerging and growing knowledge on a promising alternative avenue: the apelinergic system. This target exhibits invaluable pleiotropic properties, including inodilator activity, cardio-renal protection, and control of fluid homeostasis. Taken together, its effects are expected to be greatly beneficial for patients in septic shock.


Subject(s)
Apelin Receptors/metabolism , Catecholamines/adverse effects , Shock, Septic/drug therapy , Apelin/metabolism , Apelin/pharmacokinetics , Apelin/therapeutic use , Apelin Receptors/drug effects , Homeostasis/physiology , Humans , Multiple Organ Failure/physiopathology , Multiple Organ Failure/prevention & control , Peptide Hormones/metabolism , Peptide Hormones/pharmacokinetics , Peptide Hormones/therapeutic use
6.
Crit Care Med ; 45(4): e391-e398, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27571457

ABSTRACT

OBJECTIVE: Dobutamine is the currently recommended ß-adrenergic inotropic drug for supporting sepsis-induced myocardial dysfunction when cardiac output index remains low after preload correction. Better and safer therapies are nonetheless mandatory because responsiveness to dobutamine is limited with numerous side effects. Apelin-13 is a powerful inotropic candidate that could be considered as an alternative noncatecholaminergic support in the setting of inflammatory cardiovascular dysfunction. DESIGN: Interventional controlled experimental animal study. SETTING: Tertiary care university-based research institute. SUBJECTS: One hundred ninety-eight adult male rats. INTERVENTIONS: Using a rat model of "systemic inflammation-induced cardiac dysfunction" induced by intraperitoneal lipopolysaccharide injection (10 mg/kg), hemodynamic efficacy, cardioprotection, and biomechanics were assessed under IV osmotic pump infusions of apelin-13 (0.25 µg/kg/min) or dobutamine (7.5 µg/kg/min). MEASUREMENTS AND MAIN RESULTS: In this model and in both in vivo and ex vivo studies, apelin-13 compared with dobutamine provoked distinctive effects on cardiac function: 1) optimized cardiac energy-dependent workload with improved cardiac index and lower vascular resistance, 2) upgraded hearts' apelinergic responsiveness, and 3) consecutive downstream advantages, including increased urine output, enhanced plasma volume, reduced weight loss, and substantially improved overall outcomes. In vitro studies confirmed that these apelin-13-driven processes encompassed a significant and rapid reduction in systemic cytokine release with dampening of myocardial inflammation, injury, and apoptosis and resolution of associated molecular pathways. CONCLUSIONS: In this inflammatory cardiovascular dysfunction, apelin-13 infusion delivers distinct and optimized hemodynamic support (including positive fluid balance), along with cardioprotective effects, modulation of circulatory inflammation and extended survival.


Subject(s)
Cardiomyopathies/physiopathology , Cardiotonic Agents/pharmacology , Dobutamine/pharmacology , Intercellular Signaling Peptides and Proteins/pharmacology , Animals , Body Weight/drug effects , Cardiac Output/drug effects , Cardiomyopathies/chemically induced , Cardiomyopathies/prevention & control , Cardiotonic Agents/therapeutic use , Cytokines/blood , Disease Models, Animal , Dobutamine/therapeutic use , Intercellular Signaling Peptides and Proteins/therapeutic use , Lipopolysaccharides , Male , Mitogen-Activated Protein Kinases/metabolism , Myocardium/enzymology , Nitric Oxide Synthase Type II/blood , Peroxidase/metabolism , Phosphorylation/drug effects , Plasma Volume/drug effects , Rats , Survival Rate , Vascular Resistance/drug effects , Water-Electrolyte Balance/drug effects
7.
Crit Care Med ; 45(11): e1139-e1148, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28777197

ABSTRACT

OBJECTIVES: Apelin-13 was recently proposed as an alternative to the recommended ß-adrenergic drugs for supporting endotoxin-induced myocardial dysfunction. Since Apelin-13 signals through its receptor (Apelin peptide jejunum) to exert singular inotropic/vasotropic actions and to optimize body fluid balance, this candidate pathway might benefit septic shock management. Whether the newly discovered ELABELA (ELA), a second endogenous ligand of the Apelin peptide jejunum receptor highly expressed in the kidney, further improves cardio-renal impairment remains unknown. DESIGN, SETTING, AND SUBJECTS: Interventional study in a rat model of septic shock (128 adult males) to assess the effects of ELA and Apelin-13 on vascular and cardio-renal function. Experiments were performed in a tertiary care University-based research institute. INTERVENTIONS: Polymicrobial sepsis-induced cardiac dysfunction was produced by cecal ligation puncture to assess hemodynamic efficacy, cardioprotection, and biomechanics under acute or continuous infusions of the apelinergic agonists ELA or Apelin-13 (39 and 15 µg/kg/hr, respectively) versus normal saline. MEASUREMENTS AND MAIN RESULTS: Apelinergic agonists improved 72-hour survival after sepsis induction, with ELA providing the best clinical outcome after 24 hours. Apelinergic agonist infusion counteracted cecal ligation puncture-induced myocardial dysfunction by improving left ventricular pressure-volume relationship. ELA-treated cecal ligation puncture rats were the only group to 1) display a significant improvement in left ventricular filling as shown by increased E-wave velocity and left ventricular end-diastolic volume, 2) exhibit a higher plasma volume, and 3) limit kidney injury and free-water clearance. These beneficial renal effects were superior to Apelin-13, likely because full-length ELA enabled a distinctive regulation of pituitary vasopressin release. CONCLUSIONS: Activation of the apelinergic system by exogenous ELA or Apelin-13 infusion improves cardiovascular function and survival after cecal ligation puncture-induced sepsis. However, ELA proved better than Apelin-13 by improving fluid homeostasis, cardiovascular hemodynamics recovery, and limiting kidney dysfunction in a vasopressinergic-dependent manner.


Subject(s)
Intercellular Signaling Peptides and Proteins/pharmacology , Peptide Hormones/pharmacology , Shock, Septic/drug therapy , Animals , Biomarkers , Cytokines/immunology , Disease Models, Animal , Echocardiography , Hemodynamics/drug effects , Male , Rats , Real-Time Polymerase Chain Reaction
8.
Org Biomol Chem ; 15(2): 449-458, 2017 Jan 04.
Article in English | MEDLINE | ID: mdl-27924341

ABSTRACT

Apelin is the endogenous ligand for the G protein-coupled receptor APJ and exerts a key role in regulating cardiovascular functions. We report herein a novel series of macrocyclic analogues of apelin-13 in which the N- and C-terminal residues as well as the macrocycle composition were chemically modified to modulate structure-activity relationships on the APJ receptor. To this end, the binding affinity and the ability to engage G protein-dependent and G protein-independent signalling pathways of the resulting analogues were assessed. In this series, the position and the nature of the C-terminal aromatic residue is a determinant for APJ interaction and ß-arrestin recruitment, as previously demonstrated for linear apelin-13 derivatives. We finally discovered compounds 1, 4, 11 and 15, four potent G protein-biased apelin receptor agonists exhibiting affinity in the nanomolar range for APJ. These macrocyclic compounds represent very useful pharmacological tools to explore the therapeutic potential of the apelinergic system.


Subject(s)
Apelin Receptors/agonists , Intercellular Signaling Peptides and Proteins/pharmacology , Macrocyclic Compounds/pharmacology , Animals , Blood Pressure/drug effects , Dose-Response Relationship, Drug , Intercellular Signaling Peptides and Proteins/chemical synthesis , Intercellular Signaling Peptides and Proteins/chemistry , Macrocyclic Compounds/chemical synthesis , Macrocyclic Compounds/chemistry , Male , Molecular Conformation , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
9.
Circ Res ; 112(1): 48-56, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-22993413

ABSTRACT

RATIONALE: Mitogen-activated protein kinases (MAPKs) are activated in the heart by disease-inducing and stress-inducing stimuli, where they participate in hypertrophy, remodeling, contractility, and heart failure. A family of dual-specificity phosphatases (DUSPs) directly inactivates each of the MAPK terminal effectors, potentially serving a cardioprotective role. OBJECTIVE: To determine the role of DUSP1 and DUSP4 in regulating p38 MAPK function in the heart and the effect on disease. METHODS AND RESULTS: Here, we generated mice and mouse embryonic fibroblasts lacking both Dusp1 and Dusp4 genes. Although single nulls showed no molecular effects, combined disruption of Dusp1/4 promoted unrestrained p38 MAPK activity in both mouse embryonic fibroblasts and the heart, with no change in the phosphorylation of c-Jun N-terminal kinases or extracellular signal-regulated kinases at baseline or with stress stimulation. Single disruption of either Dusp1 or Dusp4 did not result in cardiac pathology, although Dusp1/4 double-null mice exhibited cardiomyopathy and increased mortality with aging. Pharmacological inhibition of p38 MAPK with SB731445 ameliorated cardiomyopathy in Dusp1/4 double-null mice, indicating that DUSP1/4 function primarily through p38 MAPK in affecting disease. At the cellular level, unrestrained p38 MAPK activity diminished cardiac contractility and Ca2+ handling, which was acutely reversed with a p38 inhibitory compound. Poor function in Dusp1/4 double-null mice also was partially rescued by phospholamban deletion. CONCLUSIONS: Our data demonstrate that Dusp1 and Dusp4 are cardioprotective genes that play a critical role in the heart by dampening p38 MAPK signaling that would otherwise reduce contractility and induce cardiomyopathy.


Subject(s)
Cardiomyopathies/enzymology , Dual Specificity Phosphatase 1/deficiency , Myocytes, Cardiac/enzymology , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Calcium/metabolism , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Cardiomyopathies/diagnosis , Cardiomyopathies/genetics , Cardiomyopathies/physiopathology , Cardiomyopathies/prevention & control , Cells, Cultured , Disease Models, Animal , Dual Specificity Phosphatase 1/genetics , Enzyme Activation , Fibroblasts/enzymology , Gene Expression Regulation , Hemodynamics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Tyrosine Phosphatases , Time Factors , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
10.
Circ Res ; 110(8): 1077-86, 2012 Apr 13.
Article in English | MEDLINE | ID: mdl-22403241

ABSTRACT

RATIONALE: Both nuclear factors of activated T cells (NFAT) and nuclear factor-κB (NFκB) are Rel homology domain (RHD)-containing transcription factors whose independent activities are critically involved in regulating cardiac hypertrophy and failure. OBJECTIVE: To determine the potential functional interaction between NFAT and NFκB signaling pathways in cardiomyocytes and its role in cardiac hypertrophy and remodeling. METHODS AND RESULTS: We identified a novel transcriptional regulatory mechanism whereby NFκB and NFAT directly interact and synergistically promote transcriptional activation in cardiomyocytes. We show that the p65 subunit of NFκB coimmunoprecipitates with NFAT in cardiomyocytes, and this interaction maps to the RHD within p65. Overexpression of the p65-RHD disrupts the association between endogenous p65 and NFATc1, leading to reduced transcriptional activity. Overexpression of IκB kinase ß (IKKß) or p65-RHD causes nuclear translocation of NFATc1, and expression of a constitutively nuclear NFATc1-SA mutant similarly facilitated p65 nuclear translocation. Combined overexpression of p65 and NFATc1 promotes synergistic activation of NFAT transcriptional activity in cardiomyocytes, whereas inhibition of NFκB with IκBαM or dominant negative IKKß reduces NFAT activity. Importantly, agonist-induced NFAT activation is reduced in p65 null mouse embryonic fibroblasts (MEFs) compared with wild-type MEFs. In vivo, cardiac-specific deletion of p65 using a Cre-loxP system causes a ≈50% reduction in NFAT activity in luciferase reporter mice. Moreover, ablation of p65 in the mouse heart decreases the hypertrophic response after pressure overload stimulation, reduces the degree of pathological remodeling, and preserves contractile function. CONCLUSIONS: Our results suggest a direct interaction between NFAT and NFκB that effectively integrates 2 disparate signaling pathways in promoting cardiac hypertrophy and ventricular remodeling.


Subject(s)
Cardiomegaly/metabolism , Myocardium/metabolism , NFATC Transcription Factors/metabolism , Transcription Factor RelA/metabolism , Ventricular Function, Left , Ventricular Remodeling , Animals , Animals, Newborn , Cardiomegaly/diagnostic imaging , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cardiomegaly/prevention & control , Cells, Cultured , Disease Models, Animal , Echocardiography, Doppler , Gene Expression Regulation , Genes, Reporter , I-kappa B Kinase/metabolism , Immunoprecipitation , Mice , Mice, Knockout , Mice, Transgenic , Myocardial Contraction , Myocardium/pathology , NFATC Transcription Factors/genetics , Protein Binding , Rats , Rats, Sprague-Dawley , Signal Transduction , Transcription Factor RelA/deficiency , Transcription Factor RelA/genetics , Transcriptional Activation
11.
Sci Transl Med ; 16(739): eabn8529, 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38507466

ABSTRACT

Impaired skeletal muscle stem cell (MuSC) function has long been suspected to contribute to the pathogenesis of muscular dystrophy (MD). Here, we showed that defects in the endothelial cell (EC) compartment of the vascular stem cell niche in mouse models of Duchenne MD, laminin α2-related MD, and collagen VI-related myopathy were associated with inefficient mobilization of MuSCs after tissue damage. Using chemoinformatic analysis, we identified the 13-amino acid form of the peptide hormone apelin (AP-13) as a candidate for systemic stimulation of skeletal muscle ECs. Systemic administration of AP-13 using osmotic pumps generated a pro-proliferative EC-rich niche that supported MuSC function through angiocrine factors and markedly improved tissue regeneration and muscle strength in all three dystrophic mouse models. Moreover, EC-specific knockout of the apelin receptor led to regenerative defects that phenocopied key pathological features of MD, including vascular defects, fibrosis, muscle fiber necrosis, impaired MuSC function, and reduced force generation. Together, these studies provide in vivo proof of concept that enhancing endogenous skeletal muscle repair by targeting the vascular niche is a viable therapeutic avenue for MD and characterized AP-13 as a candidate for further study for the systemic treatment of MuSC dysfunction.


Subject(s)
Muscular Dystrophy, Duchenne , Stem Cell Niche , Mice , Animals , Apelin/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Signal Transduction
12.
Front Cardiovasc Med ; 10: 1191891, 2023.
Article in English | MEDLINE | ID: mdl-37636297

ABSTRACT

Introduction: Peripheral arterial disease (PAD) is a major risk factor for lower-extremity amputation in diabetic patients. Unfortunately, previous clinical studies investigating therapeutic angiogenesis using the vascular endothelial growth factor (VEGF) have shown disappointing results in diabetic patients, which evokes the necessity for novel therapeutic agents. The apelinergic system (APJ receptor/apelin) is highly upregulated under hypoxic condition and acts as an activator of angiogenesis. Apelin treatment improves revascularization in nondiabetic models of ischemia, however, its role on angiogenesis in diabetic conditions remains poorly investigated. This study explored the impact of Pyr-apelin-13 in endothelial cell function and diabetic mouse model of hindlimb ischemia. Methods: Nondiabetic and diabetic mice underwent femoral artery ligation to induce limb ischemia. Diabetic mice were implanted subcutaneously with osmotic pumps delivering Pyr-apelin-13 for 28 days. Blood flow reperfusion was measured for 4 weeks post-surgery and exercise willingness was assessed with voluntary wheels. In vitro, bovine aortic endothelial cells (BAECs) were exposed to normal (NG) or high glucose (HG) levels and hypoxia. Cell migration, proliferation and tube formation assays were performed following either VEGF or Pyr-apelin-13 stimulation. Results and Discussion: Following limb ischemia, blood flow reperfusion, functional recovery of the limb and vascular density were improved in diabetic mice receiving Pyr-apelin-13 compared to untreated diabetic mice. In cultured BAECs, exposure to HG concentrations and hypoxia reduced VEGF proangiogenic actions, whereas apelin proangiogenic effects remained unaltered. Pyr-apelin-13 induced its proangiogenic actions through Akt/AMPK/eNOS and RhoA/ROCK signaling pathways under both NG or HG concentrations and hypoxia exposure. Our results identified the apelinergic system as a potential therapeutic target for angiogenic therapy in diabetic patients with PAD.

13.
J Am Heart Assoc ; 12(4): e028056, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36752268

ABSTRACT

Background Deep vein thrombosis (DVT) is the primary cause of pulmonary embolism and the third most life-threatening cardiovascular disease in North America. Post-DVT anticoagulants, such as warfarin, heparin, and direct oral anticoagulants, reduce the incidence of subsequent venous thrombi. However, all currently used anticoagulants affect bleeding time at various degrees, and there is therefore a need for improved therapeutic regimens in DVT. It has recently been shown that mast cells play a crucial role in a DVT murine model. The underlying mechanism involved in the prothrombotic properties of mast cells, however, has yet to be identified. Methods and Results C57BL/6 mice and mouse mast cell protease-4 (mMCP-4) genetically depleted mice (mMCP-4 knockout) were used in 2 mouse models of DVT, partial ligation (stenosis) and ferric chloride-endothelial injury model of the inferior vena cava. Thrombus formation and impact of genetically repressed or pharmacologically (specific inhibitor TY-51469) inhibited mMCP-4 were evaluated by morphometric measurements of thrombi immunochemistry (mouse and human DVT), color Doppler ultrasound, bleeding times, and enzymatic activity assays ex vivo. Recombinant chymases, mMCP-4 (mouse) and CMA-1 (human), were used to characterize the interaction with murine and human plasmin, respectively, by mass spectrometry and enzymatic activity assays. Inhibiting mast cell-generated mMCP-4, genetically or pharmacologically, resolves and prevents venous thrombus formation in both DVT models. Inferior vena cava blood flow obstruction was observed in the stenosis model after 6 hours of ligation, in control- but not in TY-51469-treated mice. In addition, chymase inhibition had no impact on bleeding times of healthy or DVT mice. Furthermore, endogenous chymase limits plasmin activity in thrombi ex vivo. Recombinant mouse or human chymase degrades/inactivates purified plasmin in vitro. Finally, mast cell-containing immunoreactive chymase was identified in human DVT. Conclusions This study identified a major role for mMCP-4, a granule-localized protease of chymase type, in DVT formation. These findings support a novel pharmacological strategy to resolve or prevent DVT without affecting the coagulation cascade through the inhibition of chymase activity.


Subject(s)
Fibrinolysin , Venous Thrombosis , Mice , Humans , Animals , Chymases/metabolism , Bleeding Time , Disease Models, Animal , Constriction, Pathologic , Mice, Inbred C57BL , Venous Thrombosis/prevention & control , Anticoagulants
14.
J Med Chem ; 65(1): 531-551, 2022 01 13.
Article in English | MEDLINE | ID: mdl-34982553

ABSTRACT

We previously reported a series of macrocyclic analogues of [Pyr1]-apelin-13 (Ape13) with increased plasma stability and potent APJ agonist properties. Based on the most promising compound in this series, we synthesized and then evaluated novel macrocyclic compounds of Ape13 to identify agonists with specific pharmacological profiles. These efforts led to the development of analogues 39 and 40, which possess reduced molecular weight (MW 1020 Da vs Ape13, 1534 Da). Interestingly, compound 39 (Ki 0.6 nM), which does not activate the Gα12 signaling pathway while maintaining potency and efficacy similar to Ape13 to activate Gαi1 (EC50 0.8 nM) and ß-arrestin2 recruitment (EC50 31 nM), still exerts cardiac actions. In addition, analogue 40 (Ki 5.6 nM), exhibiting a favorable Gα12-biased signaling and an increased in vivo half-life (t1/2 3.7 h vs <1 min of Ape13), produces a sustained cardiac response up to 6 h after a single subcutaneous bolus injection.


Subject(s)
Apelin/analogs & derivatives , Apelin/pharmacology , GTP-Binding Protein alpha Subunits, G12-G13/drug effects , Heart/drug effects , Signal Transduction/drug effects , Apelin/pharmacokinetics , Apelin Receptors/drug effects , Arrestin/drug effects , HEK293 Cells , Half-Life , Humans , Injections, Subcutaneous , Macrocyclic Compounds/chemical synthesis , Macrocyclic Compounds/pharmacology , Molecular Weight
15.
J Biol Chem ; 285(9): 6716-24, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20037164

ABSTRACT

Calcineurin is a protein phosphatase that is uniquely regulated by sustained increases in intracellular Ca(2+) following signal transduction events. Calcineurin controls cellular proliferation, differentiation, apoptosis, and inducible gene expression following stress and neuroendocrine stimulation. In the adult heart, calcineurin regulates hypertrophic growth of cardiomyocytes in response to pathologic insults that are associated with altered Ca(2+) handling. Here we determined that calcineurin signaling is directly linked to the proper control of cardiac contractility, rhythm, and the expression of Ca(2+)-handling genes in the heart. Our approach involved a cardiomyocyte-specific deletion using a CnB1-LoxP-targeted allele in mice and three different cardiac-expressing Cre alleles/transgenes. Deletion of calcineurin with the Nkx2.5-Cre knock-in allele resulted in lethality at 1 day after birth due to altered right ventricular morphogenesis, reduced ventricular trabeculation, septal defects, and valvular overgrowth. Slightly later deletion of calcineurin with the alpha-myosin heavy chain Cre transgene resulted in lethality in early mid adulthood that was characterized by substantial reductions in cardiac contractility, severe arrhythmia, and reduced myocyte content in the heart. Young calcineurin heart-deleted mice died suddenly after pressure overload stimulation or neuroendocrine agonist infusion, and telemetric monitoring of older mice showed arrhythmia leading to sudden death. Mechanistically, loss of calcineurin reduced expression of key Ca(2+)-handling genes that likely lead to arrhythmia and reduced contractility. Loss of calcineurin also directly impacted cellular proliferation in the postnatal developing heart. These results reveal multiple mechanisms whereby calcineurin regulates cardiac development and myocyte contractility.


Subject(s)
Calcineurin/physiology , Heart/growth & development , Animals , Arrhythmias, Cardiac , Calcineurin/deficiency , Calcineurin/genetics , Calcium , Cell Proliferation , Gene Deletion , Heart/physiology , Mice , Mice, Transgenic , Myocardial Contraction , Protein Subunits
16.
Circulation ; 121(3): 419-25, 2010 Jan 26.
Article in English | MEDLINE | ID: mdl-20065166

ABSTRACT

BACKGROUND: Cardiac cachexia is characterized by an exaggerated loss of skeletal muscle, weakness, and exercise intolerance, although the cause of these effects remains unknown. Here, we hypothesized that the heart functions as an endocrine organ in promoting systemic cachexia by secreting peptide factors such as myostatin. Myostatin is a cytokine of the transforming growth factor-beta superfamily that is known to control muscle wasting. METHODS AND RESULTS: We used a Cre/loxP system to ablate myostatin (Mstn gene) expression in a cell type-specific manner. As expected, elimination of Mstn selectively in skeletal muscle with a myosin light chain 1f (MLC1f)-cre allele induced robust hypertrophy in all skeletal muscle. However, heart-specific deletion of Mstn with an Nkx2.5-cre allele did not alter baseline heart size or secondarily affect skeletal muscle size, but the characteristic wasting and atrophy of skeletal muscle that typify heart failure were not observed in these heart-specific null mice, indicating that myocardial myostatin expression controls muscle atrophy in heart failure. Indeed, myostatin levels in the plasma were significantly increased in wild-type mice subjected to pressure overload-induced cardiac hypertrophy but not in Mstn heart-specific deleted mice. Moreover, cardiac-specific overexpression of myostatin, which increased circulating levels of myostatin by 3- to 4-fold, caused a reduction in weight of the quadriceps, gastrocnemius, soleus, and even the heart itself. Finally, to investigate myostatin as a potential therapeutic target for the treatment of muscle wasting in heart failure, we infused a myostatin blocking antibody (JA-16), which promoted greater maintenance of muscle mass in heart failure. CONCLUSIONS: Myostatin released from cardiomyocytes induces skeletal muscle wasting in heart failure. Targeted inhibition of myostatin in cardiac cachexia might be a therapeutic option in the future.


Subject(s)
Cachexia/prevention & control , Heart Failure/physiopathology , Muscle, Skeletal/pathology , Muscular Atrophy/prevention & control , Myostatin/genetics , Animals , Antibodies/pharmacology , Blotting, Western , Cachexia/etiology , Cachexia/pathology , Gene Expression/physiology , Gene Knock-In Techniques , Heart Failure/complications , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Muscle, Skeletal/metabolism , Muscular Atrophy/etiology , Muscular Atrophy/pathology , Myocytes, Cardiac/metabolism , Myostatin/immunology , Myostatin/metabolism , Organ Specificity , Reverse Transcriptase Polymerase Chain Reaction
17.
Am J Physiol Heart Circ Physiol ; 300(6): H1973-82, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21421824

ABSTRACT

A significant proportion of heart failure patients develop skeletal muscle wasting and cardiac cachexia, which is associated with a very poor prognosis. Recently, myostatin, a cytokine from the transforming growth factor-ß (TGF-ß) family and a known strong inhibitor of skeletal muscle growth, has been identified as a direct mediator of skeletal muscle atrophy in mice with heart failure. Myostatin is mainly expressed in skeletal muscle, although basal expression is also detectable in heart and adipose tissue. During pathological loading of the heart, the myocardium produces and secretes myostatin into the circulation where it inhibits skeletal muscle growth. Thus, genetic elimination of myostatin from the heart reduces skeletal muscle atrophy in mice with heart failure, whereas transgenic overexpression of myostatin in the heart is capable of inducing muscle wasting. In addition to its endocrine action on skeletal muscle, cardiac myostatin production also modestly inhibits cardiomyocyte growth under certain circumstances, as well as induces cardiac fibrosis and alterations in ventricular function. Interestingly, heart failure patients show elevated myostatin levels in their serum. To therapeutically influence skeletal muscle wasting, direct inhibition of myostatin was shown to positively impact skeletal muscle mass in heart failure, suggesting a promising strategy for the treatment of cardiac cachexia in the future.


Subject(s)
Heart Failure/physiopathology , Muscular Atrophy/physiopathology , Myocardium/metabolism , Myostatin/metabolism , Animals , Cachexia/metabolism , Cachexia/physiopathology , Disease Models, Animal , Heart Failure/metabolism , Humans , Mice , Mice, Transgenic , Muscle, Skeletal/physiopathology , Muscular Atrophy/metabolism , Myostatin/deficiency , Myostatin/genetics
18.
J Med Chem ; 64(1): 602-615, 2021 01 14.
Article in English | MEDLINE | ID: mdl-33350824

ABSTRACT

ELABELA (ELA) is the second endogenous ligand of the apelin receptor (APJ). Although apelin-13 and ELA both target APJ, there is limited information on structure-activity relationship (SAR) of ELA. In the present work, we identified the shortest bioactive C-terminal fragment ELA23-32, which possesses high affinity for APJ (Ki 4.6 nM) and produces cardiorenal effects in vivo similar to those of ELA. SAR studies on conserved residues (Leu25, His26, Val29, Pro30, Phe31, Pro32) show that ELA and apelin-13 may interact differently with APJ. His26 and Val29 emerge as important for ELA binding. Docking and binding experiments suggest that Phe31 of ELA may bind to a tight groove distinct from that of Phe13 of Ape13, while the Phe13 pocket may be occupied by Pro32 of ELA. Further characterization of signaling profiles on the Gαi1, Gα12, and ß-arrestin2 pathways reveals the importance of aromatic residue at the Phe31 or Pro32 position for receptor activation.


Subject(s)
Apelin Receptors/agonists , Peptide Hormones/pharmacology , Amino Acid Sequence , Animals , Apelin Receptors/metabolism , Binding Sites , Blood Pressure/drug effects , Computational Biology , Heart/drug effects , Heart/physiology , Humans , Intercellular Signaling Peptides and Proteins/chemistry , Ligands , Male , Peptide Hormones/chemistry , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
19.
Front Pharmacol ; 12: 709467, 2021.
Article in English | MEDLINE | ID: mdl-34385922

ABSTRACT

Objectives: Arterial hypertension, when exacerbated by excessive dietary salt intake, worsens the morbidity and mortality rates associated with cardiovascular and renal diseases. Stimulation of the apelinergic system appears to protect against several circulatory system diseases, but it remains unknown if such beneficial effects are conserved in severe hypertension. Therefore, we aimed at determining whether continuous infusion of apelinergic ligands (i.e., Apelin-13 and Elabela) exerted cardiorenal protective effects in spontaneously hypertensive (SHR) rats receiving high-salt diet. Methods: A combination of echocardiography, binding assay, histology, and biochemical approaches were used to investigate the cardiovascular and renal effects of Apelin-13 or Elabela infusion over 6 weeks in SHR fed with normal-salt or high-salt chow. Results: High-salt intake upregulated the cardiac and renal expression of APJ receptor in SHR. Importantly, Elabela was more effective than Apelin-13 in reducing high blood pressure, cardiovascular and renal dysfunctions, fibrosis and hypertrophy in high-salt fed SHR. Unlike Apelin-13, the beneficial effects of Elabela were associated with a counter-regulatory role of the ACE/ACE2/neprilysin axis of the renin-angiotensin-aldosterone system (RAAS) in heart and kidneys of salt-loaded SHR. Interestingly, Elabela also displayed higher affinity for APJ in the presence of high salt concentration and better resistance to RAAS enzymes known to cleave Apelin-13. Conclusion: These findings highlight the protective action of the apelinergic system against salt-induced severe hypertension and cardiorenal failure. As compared with Apelin-13, Elabela displays superior pharmacodynamic and pharmacokinetic properties that warrant further investigation of its therapeutic use in cardiovascular and kidney diseases.

20.
Sci Rep ; 11(1): 22770, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34815457

ABSTRACT

Sepsis is a prevalent life-threatening condition related to a systemic infection, and with unresolved issues including refractory septic shock and organ failures. Endogenously released catecholamines are often inefficient to maintain blood pressure, and low reactivity to exogenous catecholamines with risk of sympathetic overstimulation is well documented in septic shock. In this context, apelinergics are efficient and safe inotrope and vasoregulator in rodents. However, their utility in a larger animal model as well as the limitations with regards to the enzymatic breakdown during sepsis, need to be investigated. The therapeutic potential and degradation of apelinergics in sepsis were tested experimentally and in a cohort of patients. (1) 36 sheep with or without fecal peritonitis-induced septic shock (a large animal experimental design aimed to mimic the human septic shock paradigm) were evaluated for hemodynamic and renal responsiveness to incremental doses of two dominant apelinergics: apelin-13 (APLN-13) or Elabela (ELA), and (2) 52 subjects (33 patients with sepsis/septic shock and 19 healthy volunteers) were investigated for early levels of endogenous apelinergics in the blood, the related enzymatic degradation profile, and data regarding sepsis outcome. APLN-13 was the only one apelinergic which efficiently improved hemodynamics in both healthy and septic sheep. Endogenous apelinergic levels early rose, and specific enzymatic breakdown activities potentially threatened endogenous apelin system reactivity and negatively impacted the outcome in human sepsis. Short-term exogenous APLN-13 infusion is helpful in stabilizing cardiorenal functions in ovine septic shock; however, this ability might be impaired by specific enzymatic systems triggered during the early time course of human sepsis. Strategies to improve resistance of APLN-13 to degradation and/or to overcome sepsis-induced enzymatic breakdown environment should guide future works.


Subject(s)
Apelin/metabolism , Enzymes/metabolism , Hemodynamics , Pancreatic Elastase/metabolism , Proteolysis , Shock, Septic/pathology , Aged , Animals , Apelin/genetics , Case-Control Studies , Catecholamines/metabolism , Feces , Female , Follow-Up Studies , Humans , Male , Middle Aged , Pancreatic Elastase/genetics , Peritonitis/complications , Prognosis , Prospective Studies , Sheep , Shock, Septic/etiology , Shock, Septic/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL