scholarly journals A Murine Model with JAK2V617F Expression in Both Hematopoietic Cells and Vascular Endothelial Cells Recapitulates the Key Features of Human Myeloproliferative Neoplasm

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1467-1467
Author(s):  
Haotian Zhang ◽  
Amar Yeware ◽  
Sandy Lee ◽  
Huichun Zhan

Abstract Introduction Although murine models have provided unequivocal evidence that the JAK2V617F mutation is able to cause myeloproliferative neoplasms (MPNs), there is significant heterogeneity in disease phenotypes between different murine models and none has been able to recapitulate both the myeloproliferative phenotype and cardiovascular pathology seen in patients with MPNs. In addition, these murine models were mostly followed for less than 3-9 months and how aging affects MPN disease progression has not been studied. Endothelial cells (ECs) are an essential component of the hematopoietic niche, and they have been shown to express the JAK2V617F mutation in patients with MPNs. In this study, we investigated how MPN progresses in the JAK2V617F-bearing vascular niche during aging. Methods JAK2V617F Flip-Flop (FF1) mice (which carry a Cre-inducible human JAK2V617F gene driven by the human JAK2 promoter) were crossed with Tie2-cre mice to express JAK2V617F specifically in all hematopoietic cells (including HSPCs) and vascular ECs (Tie2FF1). Results The Tie2FF1 mice developed essential thrombocythemia to primary myelofibrosisdisease transformation with extramedullary splenic hematopoiesis during 18-month follow up. No evidence of leukemia transformation was observed in the Tie2FF1 mice. (Figure 1) Hematopoietic colony formation assays, flow cytometry analysis, and in vitro culture experiments revealed that there was a loss of both HSPC number and HSPC function in the marrow of old Tie2FF1 mice during aging, mimicking the advanced phases of myelofibrosis. In contrast, the spleen of old Tie2FF1 mice was able to maintain the expansion of JAK2V617F mutant hematopoiesis during aging and MPN disease progression. (Figure 2) These differences between marrow and spleen hematopoiesis in the old Tie2FF1 mice prompted us to investigate how aging affects the JAK2V617F mutant hematopoiesis differently in the marrow and spleen. We found that, although the JAK2V617F mutant HSCs (Lin -cKit +Sca1 +CD150 +CD48 -) from old Tie2FF1 mice were more proliferative than wild-type HSCs in both the marrow and spleen, mutant marrow HSCs were more apoptotic and senescent than wild-type HSCs in the marrow while mutant spleen HSCs were relatively protected in the spleen. Examination of the hematopoietic vascular niche revealed that marrow ECs (CD45 -CD31 +) were significantly decreased in old Tie2FF1 mice compared to age-matched control mice; in contrast, spleen ECs were significantly expanded and less senescent in old Tie2FF1 mice compared to control mice. Therefore, the different vascular niche function of the marrow and spleen could contribute to the decreased marrow hematopoiesis and expanded splenic hematopoiesis we have observed in the Tie2FF1 mice during aging. (Figure 3) Previously, we reported that the Tie2FF1 mice developed spontaneous heart failure with thrombosis, vasculopathy, and cardiomyopathy at 20wk of age. Here, we followed the cardiovascular function of Tie2FF1 mice during aging. At 18mo of age, the Tie2FF1 mice continued to demonstrate a phenotype of dilated cardiomyopathy with a moderate but significant decrease in left ventricular ejection fraction and an increase in left ventricular volume and mass compared to age-matched control mice. Histology examination revealed spontaneous thrombosis in the right ventricle, pulmonary arteries, both main (epicardial) coronary arteries and scattered coronary arterioles (microvessels) in the old Tie2FF1 mice, while age-matched Tie2-cre control mice had no evidence of spontaneous thrombosis in their heart or lungs. Despite these cardiovascular dysfunctions, there was no difference in body weight nor was there any increased incidence of sudden death between the old Tie2FF1 mice and control mice. These findings suggested that there was a persistent but compensated cardiomyopathy and heart failure in the Tie2FF1 mice during aging. (Figure 4) Conclusion Compared to other MPN murine models reported so far, the Tie2FF1 mice is the first MPN murine model that faithfully recapitulated almost all the key features of the human MPN diseases. Considering the presence of the JAK2V617F mutation in microvascular ECs isolated from patients with MPNs and the recapitulation of all the key features of human MPN diseasesby the Tie2FF1 mice, the roles of endothelial dysfunction in the hematologic and cardiovascular pathogenesis of MPN shall be further investigated. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 7-7
Author(s):  
Melissa Castiglione ◽  
Christopher Mazzeo ◽  
Ya-Ping Jiang ◽  
Juei-Suei Chen ◽  
Wei Yin ◽  
...  

Introduction Cardiovascular complications are the leading cause of morbidity and mortality in patients with myeloproliferative neoplasms (MPNs). The acquired kinase mutation JAK2V617F plays a central role in these disorders. Many studies have established that a significant number of vascular endothelial cells (ECs) in MPN patients carry the mutation.In this study, we tested the hypothesis that the JAK2V617F mutation alters vascular endothelial function to promote cardiovascular complications in patients with MPNs. Methods JAK2V617F Flip-Flop (FF1) mice and Tie2-Cre mice were crossed to generate the Tie2FF1 mice, in which human JAK2V617F is expressed specifically in hematopoietic cells and vascular ECs, so as to model the human diseases in which both the hematopoietic stem cells and ECs harbor the mutation. All mice were fed a standard chow diet. Results As we previously reported, Tie2FF1 mice develop a myeloproliferative phenotype with leukocytosis, thrombocytosis, significant splenomegaly, and greatly increased numbers of hematopoietic stem cells by 8 wk of age. We observed an increased incidence of sudden death during performance of minor procedures (e.g. submandibular bleeding) in Tie2FF1 mice, especially after 20 wk of age. Transthoracic echocardiography revealed significant decreases in left ventricular ejection fraction and fractional shortening, and increases in left ventricular end-diastolic volume and end-systolic volume in 20-22 wk old Tie2FF1 mice compared to age-matched Tie2-Cre controls, indicative of cardiac dysfunction. Pathological evaluation revealed increased heart size and heart weight, and evidence of pulmonary congestion in Tie2FF1 mice compared to controls, consistent with the diagnosis of congestive heart failure. No atherosclerotic lesions or myocardium infarctions were observed in Tie2FF1 mice. (Figure 1) No significant cardiac dysfunction was observed in Tie2FF1 mice at 10-12 wk of age, suggesting that the heart failure is apparently not present from birth. Next,we generated a chimeric murine model with JAK2V617F-mutant blood cells and wild-type vascular ECs by transplanting Tie2FF1 marrow cells into wild-type recipients. The transplantation of wild-type marrow cells into wild-type recipients served as a control. Recipients of Tie2FF1 marrow developed myeloproliferation and there was no difference in blood cell counts between these mice and the primary Tie2FF1 mice. However, serial echocardiography evaluation did not reveal any evidence of cardiac dysfunction in recipients of Tie2FF1 marrow. In addition, Ly-6Chighmonocytes, which are important participant at various stages of cardiovascular disease development, were significantly increased in primary Tie2FF1 micecompared to Tie2-Cre controls, but not in wild-type recipients of Tie2FF1 marrow. These observations suggest that JAK2V617F-mutant blood cells alone are not sufficient to generate the spontaneous heart failure phenotype we have observed in Tie2FF1 mice; JAK2V617F-mutant ECs are required, as cardiac dysfunction and inflammatory monocytes are only generated when the ECs bear the JAK2V617F mutation. (Figure 2) To begin to understand the roles of JAK2V617F-mutant ECs in the development of cardiovascular dysfunction in Tie2FF1 mice, we found that the expression levels of Kruppel-like factors 2 (KLF2) and 4 (KLF4), two important regulators of vascular homeostasis that contribute to an anti-adhesive, anti-thrombotic, and anti-inflammatory endothelial phenotype, as well as thrombomodulin and eNOS, two downstream targets of KLF2/4 signaling, were significantly down-regulated in JAK2V617F-mutant ECs compared to wild-type ECs. In addition, cell surface adhesion molecules PECAM and E-selection were significantly up-regulated in JAK2V617F-mutant ECs compared to wild-type ECs. Both PECAM and E-selectin were further up-regulated in flow sheared (60dyn/cm2) JAK2V617F-mutant ECs compared to un-sheared ECs, while their levels did not change in wild-type ECs. These results indicate that JAK2V617F-mutant ECs display a pro-adhesive phenotype that can contribute to the cardiovascular dysfunction we observed in Tie2FF1 mice. (Figure 3) Conclusions In summary, our findings indicate that the JAK2V617F mutation can alter vascular endothelial function to promote cardiovascular complications in a murine model of MPN. Disclosures No relevant conflicts of interest to declare.


Author(s):  
Mortimer Korf-Klingebiel ◽  
Marc R. Reboll ◽  
Felix Polten ◽  
Natalie Weber ◽  
Felix Jäckle ◽  
...  

Background: Inflammation contributes to the pathogenesis of heart failure, but there is limited understanding of inflammation's potential benefits. Inflammatory cells secrete myeloid-derived growth factor (MYDGF) to promote tissue repair after acute myocardial infarction. We hypothesized that MYDGF has a role in cardiac adaptation to persistent pressure overload. Methods: We defined the cellular sources and function of MYDGF in wild-type, Mydgf -deficient ( Mydgf -/- ), and Mydgf bone marrow-chimeric or bone marrow-conditional transgenic mice with pressure overload-induced heart failure after transverse aortic constriction surgery. We measured MYDGF plasma concentrations by targeted liquid chromatography-mass spectrometry. We identified MYDGF signaling targets by phosphoproteomics and substrate-based kinase activity inference. We recorded Ca 2+ transients and sarcomere contractions in isolated cardiomyocytes. Additionally, we explored the therapeutic potential of recombinant MYDGF. Results: MYDGF protein abundance increased in the left ventricular (LV) myocardium and in blood plasma of pressure-overloaded mice. Patients with severe aortic stenosis also had elevated MYDGF plasma concentrations, which declined after transcatheter aortic valve implantation. Monocytes and macrophages emerged as the main MYDGF sources in the pressure-overloaded murine heart. While Mydgf -/- mice had no apparent phenotype at baseline, they developed more severe LV hypertrophy and contractile dysfunction during pressure overload than wild-type mice. Conversely, conditional transgenic overexpression of MYDGF in bone marrow-derived inflammatory cells attenuated pressure overload-induced hypertrophy and dysfunction. Mechanistically, MYDGF inhibited G protein coupled receptor agonist-induced hypertrophy and augmented sarco/endoplasmic reticulum Ca 2+ ATPase 2a (SERCA2a) expression in cultured neonatal rat cardiomyocytes by enhancing PIM1 serine/threonine kinase expression and activity. Along this line, cardiomyocytes from pressure-overloaded Mydgf -/- mice displayed reduced PIM1 and SERCA2a expression, greater hypertrophy, and impaired Ca 2+ cycling and sarcomere function compared to cardiomyocytes from pressure-overloaded wild-type mice. Transplanting Mydgf -/- mice with wild-type bone marrow cells augmented cardiac PIM1 and SERCA2a levels and ameliorated pressure overload-induced hypertrophy and dysfunction. Pressure-overloaded Mydgf -/- mice were similarly rescued by adenoviral Serca2a gene transfer. Treating pressure-overloaded wild-type mice subcutaneously with recombinant MYDGF enhanced SERCA2a expression, attenuated LV hypertrophy and dysfunction, and improved survival. Conclusions: These findings establish a MYDGF-based adaptive crosstalk between inflammatory cells and cardiomyocytes that protects against pressure overload-induced heart failure.


2011 ◽  
Vol 17 (3) ◽  
pp. 253-263 ◽  
Author(s):  
Ellina Cheskis Feiner ◽  
Paul Chung ◽  
Jean Francois Jasmin ◽  
Jin Zhang ◽  
Diana Whitaker-Menezes ◽  
...  

Circulation ◽  
2008 ◽  
Vol 118 (suppl_18) ◽  
Author(s):  
Ashish Shukla ◽  
S Wamique Yusuf ◽  
Iyad Daher ◽  
Daniel Lenihan ◽  
Jb Durand

Beta blockers (BB) and ACE inhibitors (ACE) are recommended for management of left ventricular (LV)dysfunction including patients with chemotherapy-induced cardiomyopathy (CCM). There is a scarcity of data for the long term requirement of these medications in cancer patients, and the effect of their withdrawal after improvement of LV function is unknown. We report a cohort study involving patients with CCM on BB and ACE who underwent withdrawal of these medications and their outcome. Methods: In a cohort of patients with CCM stabilized on BB (commonly carvedilol) and ACE, sixteen patients were identified that had therapy withdrawn and then presented with acute heart failure. Patients resumed BB and ACE therapy and outcomes of LV function and survival were evaluated. The change in LVEF by ECHO was compared using paired t-test, with a matched control population with t-test and survivability compared and calculated using Kaplan-Meier estimates and log rank test. Results: A total of 48 patients, case (n=16) and matched control (n=32), on maximal tolerated doses of ACE+BB, mean EF of 49.62% Upon withdrawal mean EF decreased to 30.62% (p<0.0001). Nine patients died within six months of discontinuation of therapy, three of sudden cardiac death. Reinstitution of carvedilol+ACE improved LVEF to a mean of 45% (p<0.0001). Discussion: This study demonstrates acute deterioration in cardiac function and survival upon withdrawal of BB and ACE in patients with CCM, and a mortality of 56.25% for case patients and 28.12% for control patients. These data indicate that BB and ACE are pivotal in cancer patients with CCM and necessitates their indefinite use.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2795-2795
Author(s):  
Selcuk Sozer ◽  
Isabel M. Fiel ◽  
Thomas Schiano ◽  
Faye Feller ◽  
John Mascarenhas ◽  
...  

Abstract Post-natal vasculogenesis has been reported to be derived from a hierarchy of circulating endothelial progenitor cells (EPC). Some of these EPC are of myeloid origin while others have a more robust proliferative potential and are solely of endothelial cell (EC) origin. A number of groups have hypothesized that EC dysfunction might contribute to the hypercoagulable state associated with polycythemia vera (PV) by orchestrating the recruitment of blood elements to sites of injury or by regulating vascular tone. The JAK2V617F mutation is present in &gt;95% of patients with PV. In addition, some individuals with normal blood counts who develop splanchnic vein thrombosis, including Budd-Chiari syndrome (BCS) and portal vein thrombosis (PVT) have been reported to have JAK2V617F positive hematopoiesis (45% and 34%, respectively) indicating that this thrombotic tendency might precede the development of PV. We explored whether this activating mutation is present in EC in the vessels of patients with BCS. We tested this hypothesis by studying EC in venules of liver biopsy specimens of patients with BCS with (n=2) or without PV (n=1) and PVT (n=2) without PV using laser capture microdissection (LCM) followed by nested PCR or RT-PCR in order to determine if EC were JAK2V617F positive and were of hematopoietic or EC origin. EC from the hepatic venules of BCS and PVT patients were captured by LCM from hematoxylin and eosin stained sections of archival formalin-fixed paraffin-embedded liver biopsy tissue specimens. EC were identified by their fusiform nuclei and their location along the lining of the hepatic venules. Hepatocytes were identified by their morphology as having round centrally placed nuclei and abundant cytoplasm with a trabecular arrangement. At least 10 EC and 10 hepatocytes from each biopsy specimen were captured and DNA or RNA was extracted. The JAK2V617F mutation was detected using nested allele-specific PCR. The hepatocytes of each patient contained exclusively wild type JAK2 while the EC of the two BCS patients with PV were homozygous for the JAK2V617F. The EC of the other BCS patient and 2 PVT patients who did not have PV contained exclusively wild type JAK2. The EC identity of these cells in the PV patients was confirmed by the presence of the EC transcripts (VE-Cadherin and VEGF-R2) and the absence of hematopoietic cell transcripts (CD45 and CD14). These findings indicate that hepatic venule EC in BCS patients with PV are JAK2V617F positive. The presence of JAK2V617F in both endothelial cells and hematopoietic cells belonging to such patients suggests that PV originates in an adult hemangioblast like cell in such patients. Further studies are required to understand the consequence of JAK2 mutation in EC as related to their propensity to thrombosis in PV.


2004 ◽  
Vol 286 (3) ◽  
pp. H1146-H1153 ◽  
Author(s):  
Jo El J. Schultz ◽  
Betty J. Glascock ◽  
Sandra A. Witt ◽  
Michelle L. Nieman ◽  
Kalpana J. Nattamai ◽  
...  

We recently developed a mouse model with a single functional allele of Serca2 ( Serca2+/–) that shows impaired cardiac contractility and relaxation without overt heart disease. The goal of this study was to test the hypothesis that chronic reduction in sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA)2 levels in combination with an increased hemodynamic load will result in an accelerated pathway to heart failure. Age-matched wild-type and Serca2+/– mice were subjected to 10 wk of pressure overload via transverse aortic coarctation surgery. Cardiac hypertrophy and heart failure were assessed by echocardiography, gravimetry/histology, hemodynamics, and Western blotting analyses. Our results showed that ∼64% of coarcted Serca2+/– mice were in heart failure compared with 0% of coarcted wild-type mice ( P < 0.05). Overall, morbidity and mortality were greatly increased in Serca2+/– mice under pressure overload. Echocardiography assessment revealed a significant increase in left ventricular (LV) mass, and LV hypertrophy in coarcted Serca2+/– mice converted from a concentric to an eccentric pattern, similar to that seen in human heart failure. Coarcted Serca2+/– mice had decreased contractile/systolic and relaxation/diastolic performance and/or function compared with coarcted wild-type mice ( P < 0.05), despite a similar duration and degree of pressure overload. SERCA2a protein levels were significantly reduced (>50%) in coarcted Serca2+/– mice compared with noncoarcted and coarcted wild-type mice. Our findings suggest that reduction in SERCA2 levels in combination with an increased hemodynamic load results in an accelerated pathway to heart failure.


Endocrinology ◽  
2010 ◽  
Vol 151 (4) ◽  
pp. 1837-1845 ◽  
Author(s):  
Jérôme Thireau ◽  
Franck Aimond ◽  
Denise Poisson ◽  
BeiLi Zhang ◽  
Patrick Bruneval ◽  
...  

Neurohormonal imbalance is a key determinant of the progression of heart failure (HF), which results in an elevated risk of mortality. A better understanding of mechanisms involved may influence treatment strategies. The incidence and prevalence of HF are lower in women. We explored sexual dimorphism in the progression of HF using a mice model of neurohormonal-dependent HF. Male and female mice overexpressing the human β2-adrenergic receptor (TG4 strain) develop HF. We compared TG4 animals with age-matched wild-type controls. Cardiac function was studied in vivo by echocardiography and electrocardiography. Histological studies were performed. Conduction parameters were assessed by intracardiac electrophysiological exploration, as was the occurrence of spontaneous and inducible arrhythmias. The patch-clamp technique was used to determine the cellular electrophysiological profile. The role of hormonal status in HF progression was investigated by surgical gonadectomy. High mortality rate was observed in TG4 mice with a dramatic difference between males and females. Male TG4 mice exhibited intraventricular conduction abnormalities, as measured by infrahisian interval and QRS durations potentially determining reentrant circuits and increasing susceptibility to arrhythmia. The severity of HF was correlated with the degree of fibrosis, which was modulated by the gonadal hormones. Action potentials recorded from male and female left ventricular cardiomyocytes were indistinguishable, although both sexes exhibited delayed repolarization when compared with their wild-type counterparts. In conclusion, female TG4 mice were better protected than males against cardiac remodeling and rhythm disorders. A link between fibrosis, conduction time, and mortality was established in relation with sex hormones.


Author(s):  
Zhiyu Dai ◽  
Jianding Cheng ◽  
Bin Liu ◽  
Dan Yi ◽  
Anlin Feng ◽  
...  

Background Cardiac hypertrophy and fibrosis are common adaptive responses to injury and stress, eventually leading to heart failure. Hypoxia signaling is important to the (patho)physiological process of cardiac remodeling. However, the role of endothelial PHD2 (prolyl‐4 hydroxylase 2)/hypoxia inducible factor (HIF) signaling in the pathogenesis of cardiac hypertrophy and heart failure remains elusive. Methods and Results Mice with Egln1 Tie2Cre ( Tie2 ‐Cre‐mediated deletion of Egln1 [encoding PHD2]) exhibited left ventricular hypertrophy evident by increased thickness of anterior and posterior wall and left ventricular mass, as well as cardiac fibrosis. Tamoxifen‐induced endothelial Egln1 deletion in adult mice also induced left ventricular hypertrophy and fibrosis. Additionally, we observed a marked decrease of PHD2 expression in heart tissues and cardiovascular endothelial cells from patients with cardiomyopathy. Moreover, genetic ablation of Hif2a but not Hif1a in Egln1 Tie2Cre mice normalized cardiac size and function. RNA sequencing analysis also demonstrated HIF‐2α as a critical mediator of signaling related to cardiac hypertrophy and fibrosis. Pharmacological inhibition of HIF‐2α attenuated cardiac hypertrophy and fibrosis in Egln1 Tie2Cre mice. Conclusions The present study defines for the first time an unexpected role of endothelial PHD2 deficiency in inducing cardiac hypertrophy and fibrosis in an HIF‐2α–dependent manner. PHD2 was markedly decreased in cardiovascular endothelial cells in patients with cardiomyopathy. Thus, targeting PHD2/HIF‐2α signaling may represent a novel therapeutic approach for the treatment of pathological cardiac hypertrophy and failure.


Hypertension ◽  
2021 ◽  
Vol 78 (Suppl_1) ◽  
Author(s):  
Chandramohan Ramasamy ◽  
Umadevi Subramanian ◽  
Kailash N Pandey

The cardiac hormones, atrial and brain natriuretic peptides (ANP and BNP) bind to natriuretic peptide receptor-A (NPRA), which synthesizes the second messenger cGMP. The objective of this study was to determine the underlying mechanisms that regulate the development of cardiac hypertrophy, fibrosis, and congestive heart failure (CHF) in Npr1 (encoding NPRA) gene-knockout mice. The Npr1 null mutant ( Npr1 -/- , 0-copy), heterozygous ( Npr1 +/- , 1-copy), and wild-type ( Npr1 +/+ , 2-copy) mice were orally administered with transforming growth factor-β1 receptor I (TGF-β1R1) antagonist, GW788388 (2 mg/kg/day) by oral gavage for 28 days. The left ventricular end-diastolic dimension (LVEDD), left ventricular end-systolic dimension (LVEDS), posterior wall thickness (PWT), and percent fractional shortening (FS) were analyzed by echocardiography. The heart was isolated and used for the analysis of fibrotic markers using quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and Western blot methods. The heart weight-to-body weight (HW/BW) ratio, LVEDD, LVEDS and PWT were significantly (p<0.005) increased in Npr1 -/- and Npr1 +/- mice than wild-type Npr1 +/+ mice. The FS was greatly reduced in Npr1 -/- and Npr1 +/- mice compared with Npr1 +/+ mice. The Npr1 -/- null mutant (0-copy) mice showed 52% increase in HW/BW ratio and 6-fold induction of cardiac fibrosis as compared with 2-copy control mice. The cardiac expression of fibrotic markers including collagen-1a (COL-1a; 3.5-fold), connective tissue growth factor (CTGF; 5-fold), α-smooth muscle actin (α-SMA; 4-fold), TGF-β1RI (4-fold), TGF-β1RII (3.5-fold), and SMAD-2/3 proteins (3-to-5 fold) were significantly increased in Npr1 -/- and Npr1 +/- mutant mice compared with age-matched Npr1 +/+ animals. The treatment with TGF-β1R1 antagonist, significantly (p<0.001) prevented the cardiac hypertrophy, fibrosis, CHF, and down-regulated the expression of fibrotic markers and SMAD proteins in mutant mice. The LVEDD, LVEDS, and FS were significantly (p<0.001) improved in the drug treated Npr1 -/- mice. The present results indicate that the cardiac hypertrophy, fibrosis, and CHF in Npr1 mutant mice is regulated through the TGF-β1-mediated SMAD-dependent signaling pathway.


2017 ◽  
Vol 121 (suppl_1) ◽  
Author(s):  
Olivia Ziegler ◽  
Ravi Shah ◽  
Kirsty Danielson ◽  
Chun Yang Xiao ◽  
Dustin Rabideau ◽  
...  

Introduction: Left ventricular reverse remodeling (LVRR) in heart failure (HF) is linked to improved patient outcomes. Current strategies to identify individuals who respond favorably to therapy in HF are limited. Though circulating microRNAs (miRs) show epigenetic regulation of LVRR in vivo , there is little clinical data linking plasma-circulating miRs to LVRR. Here we employ a point-of-care microRNA assay, Firefly, in silico analyses, and murine models of HF to characterize profiles of circulating miRs that prognosticate LVRR in patients with HF undergoing guideline directed therapy. Hypothesis: Profiles of miRs in circulating plasma will prognosticate LVRR in patients with HF better than the current clinical model alone and may play a functional role in LVRR. Methods: Plasma from 64 patients from the PROTECT study who had serial echocardiography and available plasma at pre-randomization study visit were run on Abcam’s Firefly platform to assay levels of 51 miRs, selected on prior sequencing efforts (for discovery) and published roles in CVD. miR levels were subject to PC analysis to predict LVRR. Candidate miRs were then validated in a murine model of transverse aortic constriction-induced heart failure (TAC-HF) and their function assessed in cardiomyocyte culture systems. Results: Principle component analysis revealed 4 PCs accounting for 62.4% of the observed variation without significant association with extant markers of HF. When PC2 miR levels were added to the clinical model prognostic ability for LVRR improved substantially (AIC 79.5, (OR=6.84, 95% CI 1.81-25.80, P= 0.005). In silico analysis was then applied to generate networks of mRNAs regulated by PC2 miRs. In the murine TAC-HF model, miRs 423, 212, 221, 193b were differentially regulated, as were the predicted targets of these miRs. These miRs, particularly in combination, appeared to be regulators of cardiac hypertrophy in vitro . Conclusions: Circulating miR profiles in HF improve prognosticative ability for patients who will undergo LVRR over the clinical model alone. Additionally these miRs and their targets are dynamically regulated in murine models of HF, suggesting they may have functional and potential therapeutic utility in addition to their prognostic power.


Sign in / Sign up

Export Citation Format

Share Document