Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Paula A. da Costa Martins is active.

Publication


Featured researches published by Paula A. da Costa Martins.


Circulation | 2008

Conditional Dicer Gene Deletion in the Postnatal Myocardium Provokes Spontaneous Cardiac Remodeling

Paula A. da Costa Martins; Meriem Bourajjaj; Monika M. Gladka; Mara Kortland; Ralph J. van Oort; Yigal M. Pinto; Jeffery D. Molkentin; Leon J. De Windt

Background— Dicer, an RNAse III endonuclease critical for processing of pre-microRNAs (miRNAs) into mature 22-nucleotide miRNAs, has proven a useful target to dissect the significance of miRNAs biogenesis in mammalian biology. Methods and Results— To circumvent the embryonic lethality associated with germline null mutations for Dicer, we triggered conditional Dicer loss through the use of a tamoxifen-inducible Cre recombinase in the postnatal murine myocardium. Targeted Dicer deletion in 3-week-old mice provoked premature death within 1 week accompanied by mild ventricular remodeling and dramatic atrial enlargement. In the adult myocardium, loss of Dicer induced rapid and dramatic biventricular enlargement, accompanied by myocyte hypertrophy, myofiber disarray, ventricular fibrosis, and strong induction of fetal gene transcripts. Comparative miRNA profiling revealed a set of miRNAs that imply causality between miRNA depletion and spontaneous cardiac remodeling. Conclusions— Overall, these results indicate that modifications in miRNA biogenesis affect both juvenile and adult myocardial morphology and function.


Nature Cell Biology | 2010

MicroRNA-199b targets the nuclear kinase Dyrk1a in an auto-amplification loop promoting calcineurin/NFAT signalling

Paula A. da Costa Martins; Kanita Salic; Monika M. Gladka; Anne-Sophie Armand; Stefanos Leptidis; Hamid el Azzouzi; Arne Hansen; Christina J. Coenen-De Roo; Marti F.A. Bierhuizen; Roel van der Nagel; Joyce van Kuik; Roel A. de Weger; Alain de Bruin; Gianluigi Condorelli; Maria L. Arbonés; Thomas Eschenhagen; Leon J. De Windt

MicroRNAs (miRs) are a class of single-stranded, non-coding RNAs of about 22 nucleotides in length. Increasing evidence implicates miRs in myocardial disease processes. Here we show that miR-199b is a direct calcineurin/NFAT target gene that increases in expression in mouse and human heart failure, and targets the nuclear NFAT kinase dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 1a (Dyrk1a), constituting a pathogenic feed forward mechanism that affects calcineurin-responsive gene expression. Mutant mice overexpressing miR-199b, or haploinsufficient for Dyrk1a, are sensitized to calcineurin/NFAT signalling or pressure overload and show stress-induced cardiomegaly through reduced Dyrk1a expression. In vivo inhibition of miR-199b by a specific antagomir normalized Dyrk1a expression, reduced nuclear NFAT activity and caused marked inhibition and even reversal of hypertrophy and fibrosis in mouse models of heart failure. Our results reveal that microRNAs affect cardiac cellular signalling and gene expression, and implicate miR-199b as a therapeutic target in heart failure.


Journal of the American College of Cardiology | 2014

Circulating miR-29a, Among Other Up-Regulated MicroRNAs, Is the Only Biomarker for Both Hypertrophy and Fibrosis in Patients With Hypertrophic Cardiomyopathy

Roberta Roncarati; Chiara Viviani Anselmi; Maria Angela Losi; Laura Papa; Elena Cavarretta; Paula A. da Costa Martins; Carla Contaldi; Gloria Saccani Jotti; Anna Franzone; Laura Galastri; Michael V.G. Latronico; Massimo Imbriaco; Giovanni Esposito; Leon J. De Windt; Sandro Betocchi; Gianluigi Condorelli

OBJECTIVES The purpose of this paper was to determine whether microRNAs (miRNAs) involved in myocardial remodeling were differentially expressed in the blood of hypertrophic cardiomyopathy (HCM) patients, and whether circulating miRNAs correlated with the degree of left ventricular hypertrophy and fibrosis. BACKGROUND miRNAs-small, noncoding ribonucleic acids (RNAs) that regulate gene expression by inhibiting RNA translation-modulate cellular function. Myocardial miRNAs modulate processes such as cardiomyocyte (CM) hypertrophy, excitation-contraction coupling, and apoptosis; non-CM-specific miRNAs regulate myocardial vascularization and fibrosis. Recently, the possibility that circulating miRNAs may be biomarkers of cardiovascular disease has been raised. METHODS Forty-one HCM patients were characterized with conventional transthoracic echocardiography and cardiac magnetic resonance. Peripheral plasma levels of 21 miRNAs were assessed by quantitative real-time polymerase chain reaction and were compared with levels in a control group of 41 age- and sex-matched blood donors. RESULTS Twelve miRNAs (miR-27a, -199a-5p, -26a, -145, -133a, -143, -199a-3p, -126-3p, -29a, -155, -30a, and -21) were significantly increased in HCM plasma. However, only 3 miRNAs (miR-199a-5p, -27a, and -29a) correlated with hypertrophy; more importantly, only miR-29a correlated also with fibrosis. CONCLUSIONS Our data suggest that cardiac remodeling associated with HCM determines a significant release of miRNAs into the bloodstream: the circulating levels of both cardiac- and non-cardiac-specific miRNAs are significantly increased in the plasma of HCM patients. However, correlation with left ventricular hypertrophy parameters holds true for only a few miRNAs (i.e., miR-199a-5p, -27a, and -29a), whereas only miR-29a is significantly associated with both hypertrophy and fibrosis, identifying it as a potential biomarker for myocardial remodeling assessment in HCM.


Journal of Biological Chemistry | 2008

NFATc2 is a necessary mediator of calcineurin-dependent cardiac hypertrophy and heart failure.

Meriem Bourajjaj; Anne-Sophie Armand; Paula A. da Costa Martins; Bart Weijts; Roel van der Nagel; Sylvia Heeneman; Xander H.T. Wehrens; Leon J. De Windt

One major intracellular signaling pathway involved in heart failure employs the phosphatase calcineurin and its downstream transcriptional effector nuclear factor of activated T-cells (NFAT). In vivo evidence for the involvement of NFAT factors in heart failure development is still ill defined. Here we reveal that nfatc2 transcripts outnumber those from other nfat genes in the unstimulated heart by severalfold. Transgenic mice with activated calcineurin in the postnatal myocardium crossbred with nfatc2-null mice revealed a significant abrogation of calcineurin-provoked cardiac growth, indicating that NFATc2 plays an important role downstream of calcineurin and validates the original hypothesis that calcineurin mediates myocyte hypertrophy through activation of NFAT transcription factors. In the absence of NFATc2, a clear protection against the geometrical, functional, and molecular deterioration of the myocardium following biomechanical stress was also evident. In contrast, physiological cardiac enlargement in response to voluntary exercise training was not affected in nfatc2-null mice. Combined, these results reveal a major role for the NFATc2 transcription factor in pathological cardiac remodeling and heart failure.


Cell Metabolism | 2013

The hypoxia-inducible microRNA cluster miR-199a∼214 targets myocardial PPARδ and impairs mitochondrial fatty acid oxidation.

Hamid el Azzouzi; Stefanos Leptidis; Ellen Dirkx; Joris Hoeks; Bianca van Bree; Karl Brand; Elizabeth A. McClellan; Ella M. Poels; Judith C. Sluimer; Maarten M.G. van den Hoogenhof; Anne-Sophie Armand; Xiaoke Yin; Sarah R. Langley; Meriem Bourajjaj; Servé Olieslagers; Jaya Krishnan; Marc Vooijs; Hiroki Kurihara; Andrew Stubbs; Yigal M. Pinto; Wilhelm Krek; Manuel Mayr; Paula A. da Costa Martins; Patrick Schrauwen; Leon J. De Windt

Peroxisome proliferator-activated receptor δ (PPARδ) is a critical regulator of energy metabolism in the heart. Here, we propose a mechanism that integrates two deleterious characteristics of heart failure, hypoxia and a metabolic shift toward glycolysis, involving the microRNA cluster miR-199a∼214 and PPARδ. We demonstrate that under hemodynamic stress, cardiac hypoxia activates DNM3os, a noncoding transcript that harbors the microRNA cluster miR-199a∼214, which shares PPARδ as common target. To address the significance of miR-199a∼214 induction and concomitant PPARδ repression, we performed antagomir-based silencing of both microRNAs and subjected mice to biomechanical stress to induce heart failure. Remarkably, antagomir-treated animals displayed improved cardiac function and restored mitochondrial fatty acid oxidation. Taken together, our data suggest a mechanism whereby miR-199a∼214 actively represses cardiac PPARδ expression, facilitating a metabolic shift from predominant reliance on fatty acid utilization in the healthy myocardium toward increased reliance on glucose metabolism at the onset of heart failure.


Biochimica et Biophysica Acta | 2013

Regulation of fetal gene expression in heart failure

Ellen Dirkx; Paula A. da Costa Martins; Leon J. De Windt

During the processes leading to adverse cardiac remodeling and heart failure, cardiomyocytes react to neurohumoral stimuli and biomechanical stress by activating pathways that induce pathological hypertrophy. The gene expression patterns and molecular changes observed during cardiac hypertrophic remodeling bare resemblance to those observed during fetal cardiac development. The re-activation of fetal genes in the adult failing heart is a complex biological process that involves transcriptional, posttranscriptional and epigenetic regulation of the cardiac genome. In this review, the mechanistic actions of transcription factors, microRNAs and chromatin remodeling processes in regulating fetal gene expression in heart failure are discussed.


Cardiovascular Research | 2012

MicroRNAs in control of cardiac hypertrophy

Paula A. da Costa Martins; Leon J. De Windt

MicroRNAs refer to a subfamily of small non-coding RNA species that are designed to influence gene expression in nearly all cell types studied to date. Studies from the past decade have demonstrated that microRNAs are atypically expressed in the cardiovascular system under specific pathological conditions. Gain- and loss-of-function studies using in vitro and in vivo models have revealed distinct roles for specific microRNAs in cardiovascular development, physiological functions, and cardiac pathological conditions. In this review, the current relevant findings on the role of microRNAs in cardiac hypertrophic growth are updated, the target genes of these microRNAs are summarized, and the future of microRNAs as potential therapeutic targets is discussed.


Nature Cell Biology | 2013

Nfat and miR-25 cooperate to reactivate the transcription factor Hand2 in heart failure

Ellen Dirkx; Monika M. Gladka; Leonne E. Philippen; Anne-Sophie Armand; Virginie Kinet; Stefanos Leptidis; Hamid el Azzouzi; Kanita Salic; Meriem Bourajjaj; Gustavo J. Silva; Servé Olieslagers; Roel van der Nagel; Roel A. de Weger; Nicole Bitsch; Natasja Kisters; Sandrine Seyen; Yuka Morikawa; Christophe Chanoine; Stephane Heymans; Paul G.A. Volders; Thomas Thum; Stefanie Dimmeler; Peter Cserjesi; Thomas Eschenhagen; Paula A. da Costa Martins; Leon J. De Windt

Although aberrant reactivation of embryonic gene programs is intricately linked to pathological heart disease, the transcription factors driving these gene programs remain ill-defined. Here we report that increased calcineurin/Nfat signalling and decreased miR-25 expression integrate to re-express the basic helix-loop-helix (bHLH) transcription factor dHAND (also known as Hand2) in the diseased human and mouse myocardium. In line, mutant mice overexpressing Hand2 in otherwise healthy heart muscle cells developed a phenotype of pathological hypertrophy. Conversely, conditional gene-targeted Hand2 mice demonstrated a marked resistance to pressure-overload-induced hypertrophy, fibrosis, ventricular dysfunction and induction of a fetal gene program. Furthermore, in vivo inhibition of miR-25 by a specific antagomir evoked spontaneous cardiac dysfunction and sensitized the murine myocardium to heart failure in a Hand2-dependent manner. Our results reveal that signalling cascades integrate with microRNAs to induce the expression of the bHLH transcription factor Hand2 in the postnatal mammalian myocardium with impact on embryonic gene programs in heart failure.


Current Hypertension Reports | 2012

Circulating miRNAs: reflecting or affecting cardiovascular disease?

Vanessa van Empel; Leon J. De Windt; Paula A. da Costa Martins

MicroRNAs are a class of small, noncoding RNAs encoded by the metazoan genome that regulate protein expression. A collection of studies point to vital roles for microRNAs in the onset and development of cardiovascular diseases. So far, microRNAs have been considered as important intracellular mediators in maintaining proper cardiac function and hemostasis, and have been proposed as potential therapeutic targets in cardiovascular disease. The recent discovery that microRNAs circulate in a stable form in many body fluids, including blood, suggests that circulating microRNAs can serve as a new generation of biomarkers for cardiovascular diseases. In this review, we summarize the findings of studies focusing on circulating microRNAs present in human blood cells or plasma/serum, where they potentially could serve as diagnostic or prognostic markers for a variety of cardiovascular pathologies, including acute myocardial infarction, heart failure, coronary artery disease, stroke, diabetes and hypertension. The significance and limitations of microRNAs as the new biomarker generation for cardiovascular disease are also discussed.MicroRNAs are a class of small, noncoding RNAs encoded by the metazoan genome that regulate protein expression. A collection of studies point to vital roles for microRNAs in the onset and development of cardiovascular diseases. So far, microRNAs have been considered as important intracellular mediators in maintaining proper cardiac function and hemostasis, and have been proposed as potential therapeutic targets in cardiovascular disease. The recent discovery that microRNAs circulate in a stable form in many body fluids, including blood, suggests that circulating microRNAs can serve as a new generation of biomarkers for cardiovascular diseases. In this review, we summarize the findings of studies focusing on circulating microRNAs present in human blood cells or plasma/serum, where they potentially could serve as diagnostic or prognostic markers for a variety of cardiovascular pathologies, including acute myocardial infarction, heart failure, coronary artery disease, stroke, diabetes and hypertension. The significance and limitations of microRNAs as the new biomarker generation for cardiovascular disease are also discussed.


PLOS ONE | 2013

A Deep Sequencing Approach to Uncover the miRNOME in the Human Heart

Stefanos Leptidis; Hamid el Azzouzi; Sjoukje I. Lok; Roel A. de Weger; Serv Olieslagers; Natasja Kisters; Gustavo J. Silva; Stephane Heymans; Edwin Cuppen; Eugene Berezikov; Leon J. De Windt; Paula A. da Costa Martins

MicroRNAs (miRNAs) are a class of non-coding RNAs of ∼22 nucleotides in length, and constitute a novel class of gene regulators by imperfect base-pairing to the 3′UTR of protein encoding messenger RNAs. Growing evidence indicates that miRNAs are implicated in several pathological processes in myocardial disease. The past years, we have witnessed several profiling attempts using high-density oligonucleotide array-based approaches to identify the complete miRNA content (miRNOME) in the healthy and diseased mammalian heart. These efforts have demonstrated that the failing heart displays differential expression of several dozens of miRNAs. While the total number of experimentally validated human miRNAs is roughly two thousand, the number of expressed miRNAs in the human myocardium remains elusive. Our objective was to perform an unbiased assay to identify the miRNOME of the human heart, both under physiological and pathophysiological conditions. We used deep sequencing and bioinformatics to annotate and quantify microRNA expression in healthy and diseased human heart (heart failure secondary to hypertrophic or dilated cardiomyopathy). Our results indicate that the human heart expresses >800 miRNAs, the majority of which not being annotated nor described so far and some of which being unique to primate species. Furthermore, >250 miRNAs show differential and etiology-dependent expression in human dilated cardiomyopathy (DCM) or hypertrophic cardiomyopathy (HCM). The human cardiac miRNOME still possesses a large number of miRNAs that remain virtually unexplored. The current study provides a starting point for a more comprehensive understanding of the role of miRNAs in regulating human heart disease.

Collaboration


Dive into the Paula A. da Costa Martins's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge