• No results found

University of Groningen Circulating microRNAs in heart failure Vegter, Eline Lizet

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Circulating microRNAs in heart failure Vegter, Eline Lizet"

Copied!
15
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

University of Groningen

Circulating microRNAs in heart failure

Vegter, Eline Lizet

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date: 2017

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Vegter, E. L. (2017). Circulating microRNAs in heart failure. Rijksuniversiteit Groningen.

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

Chapter 1

Introduction and aims

(3)
(4)

1

HEART fAiLuRE

Heart failure is a major public health concern and affects 1-2% of the adult population.1

The European Society of Cardiology defined heart failure as a clinical syndrome charac-terized by typical symptoms (such as dyspnea and fatigue) and signs (pulmonary rales, ankle oedema, elevated jugular venous pressure), accompanied by structural and/or functional cardiac abnormality, resulting in a reduced cardiac output and/or elevated intracardiac pressures at rest or during stress. The aetiology of heart failure is diverse, including cardiac abnormalities caused by for example valve defects, congenital heart disease and myocarditis, as well as arrhythmias and systemic diseases such as hyper-tension, diabetes and anaemia. Although it is likely that multiple factors contribute to the development of heart failure, ischaemic heart disease is known as the most com-mon cause of heart failure in the Western world.2 Ischaemic heart disease results from a

reduced blood supply from the coronary arteries to the myocardium, most often caused by atherosclerosis. This in turn leads to a mismatch between oxygen demand and sup-ply, which eventually can lead to ventricular remodeling and impaired cardiac function. Acute heart failure is characterized by the acute onset of severe signs and symptoms of heart failure requiring immediate treatment, while physicians generally speak of chronic heart failure when patients are stable and treated for previously diagnosed heart failure. Heart failure can be further categorized in 3 main groups based on left ven-tricular ejection fraction (LVEF); heart failure with a reduced LVEF (<40%), also known as HFrEF, heart failure with a mid-range LVEF of 40-49% (HFmrEF) and heart failure with a preserved LVEF of ≥50% (HFpEF).1 These 3 groups represent patients with different

underlying characteristics and do not respond in a similar way to therapy.3

In the last decades several advancements in heart failure treatment have been made, such as the development of neurohormonal antagonists (beta-blockers, ACE inhibi-tors, angiotensin II receptor blockers and mineralocorticoid receptor antagonists), and more recently the new drug sacubitril/valsartan, an angiotensin receptor-neprilysin inhibitor.1,4 Besides medication, device therapy including the implantable

cardioverter-defibrillator (ICD) and cardiac resynchronization therapy (CRT) has shown to improve survival in selected patient groups.1 Despite these much-needed improvements, the

prognosis of heart failure is poor. Five-year mortality in heart failure ranges between 41-60%,2 while more recent data on 1-year mortality report 7-17%.5 When a patient is

hospitalized with acute heart failure, the 5-year mortality rate increases to more than 75%.6

As the prognosis of heart failure patients clearly remains unsatisfactory, new treatment strategies are needed. In search for new drug targets, it is important to understand the underlying pathophysiological mechanisms in heart failure. Several processes have

(5)

12

Chapter 1

been discovered with crucial involvement in the development and progression of the disease, for example the neurohormonal adaptation system; the keystone of the presently available heart failure pharmacotherapy. These drugs are mainly focused on targeting cell-surface or intracellular mineralocorticoid receptors, eventually attenuat-ing cardiac remodelattenuat-ing and cardiac dysfunction.7,8 Other approaches focusing on

intra-cellular molecular signalling pathways may lead to additional benefit ideally resulting in reduced mortality and rehospitalisation rates. Currently there is a rapidly growing interest in the genetic and epigenetic background of heart failure patients, providing detailed information on the cellular level. Gene therapy can interfere with these mo-lecular processes by modulating the production and/or activity of important regulatory proteins in heart failure. Pharmacogenomics may therefore be a promising new area of interest and may eventually emerge as new treatment strategy in heart failure.

GENE ExPREssioN ANd EPiGENETiCs

A large part of the human body consists of proteins and they play a central role in the function of important (patho)physiological processes. Proteins are molecules made of amino acids and the information to produce these proteins lies within portions of the DNA (deoxyribonucleic acid), called genes. DNA consists of two strands which are both formed of nucleotides. These nucleotides contain the nucleobase adenine (A), thymine (T), cytosine (C) or guanine (G), deoxyribose (a sugar) and a phosphate group, and form sequences holding the genes containing the biologically important information for the production of proteins.9 The nucleobases of the 2 strands join together according to

base complementary (A with T and C with G). DNA gets transcribed into RNA (ribonucleic acid) and the mature transcript (messenger RNA, or mRNA) leaves the nucleus. This mRNA can then be translated into amino acids to form proteins.10 This process from

gene to gene product (most often proteins) is called gene expression. Gene expression of specific genes can by quantified by measuring the mRNA transcripts in a particular cell type or tissue and can give an impression of the regulation of important (patho) physiological processes.

Gene expression can be regulated and this may have important consequences for pro-tein synthesis and subsequently the development of diseases. These gene expression modifying mechanisms are all collectively known as epigenetics. The Oxford dictionary defines epigenetics as “the study of changes in organisms caused by modification of gene expression rather than alteration of the genetic code itself”, or in other words; a change in phenotype without a change in genotype. At the moment, several different gene expression modifying mechanisms have been described including DNA methyla-tion, chromatin remodeling, histone modifications and non-coding RNA mechanisms.11

(6)

1

Non-coding RNAs are transcribed from DNA, but are not translated into proteins. This

is the large majority of all transcripts, as only 2% is translated into proteins. In the early days, these non-coding transcripts were regarded to as non-functional, however it is now clear that non-coding RNAs have several functions in regulatory mechanisms of gene expression and function. Non-coding RNAs can be divided into short non-coding RNAs (< 30 nucleotides) and long non-coding RNAs (>200 nucleotides).

MiCRoRNAs

The microRNAs (miRNAs) belong to the short non-coding RNAs and were first described in the roundworm Caenorhabditis elegans by Lee et al.12 and Wightman et al.,13 in 1993.

They showed that the gene lin-4 was not coding for proteins but instead produced a short RNA of 22 nucleotides long, which was later found to be capable of binding to lin-14 and thereby reducing the production of the LIN-14 protein. In 2001, several more miRNAs were discovered in C. elegans and could also be identified in other invertebrates, vertebrates and humans.14-16 These milestone papers paved the way for several years of

continuously increasing miRNA research (Figure 1). So far, 2588 mature miRNAs have been discovered in humans (www.mirbase.org).

In the nucleus, primary miRNAs (pri-miRNAs) are transcribed from DNA. These long stemloop structures with single stranded RNA extensions on both ends are then cleaved by processing proteins DGR8 and Drosha, resulting in precursors hairpin miRNAs (pre-miRNAs). The pre-miRNAs leave the nucleus and are further processed by Dicer in the cytoplasm of the cell. This results in the loss of the hairpin structure and a mature miRNA is formed. These mature miRNAs can then be cleaved by helicase, creating 2 separate

0 2000 4000 6000 8000 10000 12000 2001 2002 2003 2004 2005 2006 2007 2008 2009 2010 2011 2012 2013 2014 2015 2016 Cou nts Year

figure 1. Number of PubMed publications containing the search term “microRNA” from the year 2001 to

(7)

14

Chapter 1

miRNA strands. The active miRNA strand binds to an Ago protein which can be loaded into a RNA-induced silencing complex (RISC). This complex is capable of binding to the complementary 3’-untranslated region of the mRNA strand, and with perfect comple-mentary the mRNA transcript will degrade. With incomplete binding the mRNA will not degrade but repressed translationally.17 Figure 1 in Chapter 2 graphically presents the

miRNA biogenesis and function. Thus, by targeting the complementary mRNA, miRNAs are able to regulate gene expression at the post-transcriptional level.

The current understanding is that miRNAs function intracellularly, and hence in tissue. Dysregulation of miRNAs have been linked to specific diseases and underly-ing pathways. Of all mature miRNAs, a small minority was found to be organ specific, such as miR-122 (which is only expressed in the liver) or miR-9 (expression limited to the brain).18 MiRNAs with high specificity for the heart are miR-208, miR-133, miR-1 and

miR-499, which are all associated with cardiac development and disease.19 For example,

deletion of the mature miR-1-2 sequence in mouse embryonic stem cells resulted in ventricular septum abnormalities, rapid heart dilatation, rhythm disturbances and ventricular dysfunction.20 MiR-208a was shown to be required for normal cardiac

con-duction and contraction, while upregulated levels were found to be related to cardiac hypertrophy, fibrosis and a disturbed electrical conduction in mice.21 As such, methods

were developed to alter miRNA expression in order to inhibit or stimulate their gene expression-modifying function. Several pre-clinical studies were published to date and have shown promising results in the cardiovascular field, although no clinical trials have entered the arena yet.22

Circulating microRNAs

Although miRNAs have a gene silencing function within cells, in 2008, researchers dis-covered extracellular miRNAs in blood23 and later also in other body fluids such as urine,

saliva and cerebrospinal fluid.24 In this thesis, we specifically focus on miRNAs in blood

plasma. These miRNAs are protected from early degradation by RNases in the circulation since they are either bound to proteins or encapsulated by exosomes, apoptotic bodies or other microparticles. Even extreme conditions such as boiling and long-term storage do not affect circulating miRNA levels.25 As it appeared that miRNAs can be transported

through the circulation,26 the hypothesis that miRNAs may exert paracrine functions led

to a plethora of studies investigating this possibility. Indeed, several studies showed that miRNAs are able to change gene expression and thereby protein synthesis in tar-get cells in controlled, experimental settings.27,28 Besides their function as cell-to-cell

communicators, the stability and relative straightforward measurability prompted the investigation of circulating miRNAs as biomarkers of disease, also in heart failure.

Biomarkers can reflect biological processes in health and disease, and are therefore helpful tools in the management of disease. In heart failure, the biomarkers N-terminal

(8)

1

pro b-type natriuretic peptide (NT-proBNP) and b-type natriuretic peptide (BNP) can

help establishing the diagnosis of heart failure and are currently the gold standard.1

Additionally, several other, novel biomarkers such as soluble ST2, growth differentiation factor 15 (GDF-15) and galectin-3 have been described to have a role in heart failure and mainly reported as prognosticators of survival.29 However, biomarkers may also be

used to evaluate therapy and can provide insight into underlying disease mechanisms. Circulating miRNAs could add value to the already available heart failure biomarkers and have been detected in blood and plasma of patients with cardiovascular disease.

Distinct circulating miRNA patterns were reported for patients with myocardial infarc-tion in comparison to control subjects, and also in cardiac arrhythmias and hyperten-sion.30 In heart failure, most studies focused on differential miRNA expression in chronic

heart failure patients compared to healthy subjects,31-33 while only few research groups

directed their attention to acute heart failure. In these relatively small studies, circu-lating miRNAs capable of differentiating patients presenting at the emergency room with dyspnea owing to heart failure and breathlessness caused by other causes were described.34,35 Some groups tried to use circulating miRNAs to distinguish HFpEF from

HFrEF, and identified specific differential miRNA patterns, although very few overlap was found between these studies.35-37 Because of the lack of robust circulating miRNA

studies in acute heart failure, we focused on the investigation of a circulating miRNA profile in acute heart failure patients. In addition, the association between circulating miRNAs and outcome in acute heart failure has not been described before.

Although the proper identification of the most differentially expressed circulating miRNAs in heart failure is the first step, understanding the findings is a crucial next stage in the study of circulating miRNAs. These miRNAs may represent several clinical factors and disease processes in heart failure, however, information on this topic is limited. Increasing our knowledge concerning the potential meaning of these miRNAs in the circulation of heart failure patients is therefore one of the primary aims of this thesis.

AiMs ANd ouTLiNE of THE THEsis

The main aims of this thesis are as follows:

1) Discover differentially expressed circulating miRNAs in acute heart failure patients 2) Provide additional information regarding clinical factors and disease processes

these circulating miRNAs may reflect

(9)

16

Chapter 1

In Chapter 2 we discuss the current knowledge of miRNAs in heart failure, specifically focusing on circulating miRNAs and their release and uptake from the circulation, their role in paracrine cell signalling and biomarker potential. Further, we describe miRNAs with important functions in cardiac hypertrophy and fibrosis and discuss the possibili-ties of inhibiting or overexpressing these miRNAs in the context of future drug targets.

Although some studies identified a pattern of differentially expressed circulating miRNAs in heart failure, most studies were conducted in a small number of chronic heart failure patients and few validated their findings in independent patient cohorts. Therefore, we aimed in Chapter 3 to identify a miRNA signature in several different heart failure cohorts representing different stages of acute and chronic heart failure.

While the identification of these differentially expressed miRNAs in plasma is the first step in the study of heart failure-related circulating miRNAs, not much is known regarding their origin, role and function in the circulation of heart failure patients. In the next chapters, we try to provide more background on our previous findings described in Chapter 3, in which we show that the most differentially expressed circulating miRNAs in heart failure are downregulated. We explore several different hypotheses in Chapter 4, 5 and 6, as presented in Figure 2. In Chapter 4 we investigate the previously identified miRNAs in relation to other, well-known biomarkers. We also sought to find associations between these circulating miRNAs, their potential targets and relevant cardiac-related pathways. In Chapter 5 we focus on another import aspect of acute heart failure, namely dilution and fluid overload. We explored whether volume status was associated to cir-culating miRNA levels by relating changes in haemoconcentration to changes in miRNA levels in patients with acute heart failure. We also investigated the hypothesis that these heart failure-related miRNAs may be associated to vascular processes. In general, the current concept is that miRNAs in the circulation mainly derive from blood cells. Further,

Downregulated circulating miRNAs in heart failure Volume status Cardiac related

pathways dysfunction Vascular

(10)

1

in heart failure it has been shown that the most abundantly expressed miRNAs in the

circulation of do not originate from the heart, but from blood and endothelial cells. Therefore, we aimed in Chapter 6 to measure the heart failure-related set of miRNAs in another cohort of heart failure patients and investigated the potential relation with vascular dysfunction as reflected in heart failure patients with atherosclerotic disease. Further, we assessed the associations between circulating miRNA levels and biomarkers reflecting atherosclerosis-related processes including angiogenesis, inflammation and endothelial dysfunction.

In order to gain insight into the potential function of these circulating miRNAs, proper animal models suitable for further mechanistic follow-up studies are needed. However, very few circulating miRNAs identified in human heart failure have been measured in animal models of heart failure. In Chapter 7, we report about 3 mechanistically differ-ent roddiffer-ent heart failure models in which we investigate our set of previously described heart failure-related circulating miRNAs.

Finally, in Chapter 8 we discuss the main findings and conclusions and place our re-sults in perspective. Moreover, we describe the future directions and new developments in non-coding RNA research.

(11)

18

Chapter 1

REfERENCEs

1. Ponikowski P, Voors AA, Anker SD, Bueno H, Cleland JG, Coats AJ, Falk V, Gonzalez-Juanatey JR, Harjola VP, Jankowska EA, Jessup M, Linde C, Nihoyannopoulos P, Parissis JT, Pieske B, Riley JP, Rosano GM, Ruilope LM, Ruschitzka F, Rutten FH, van der Meer P, Authors/Task Force Members. 2016 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: The Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC)Developed with the special contribution of the Heart Failure Association (HFA) of the ESC. Eur Heart J 2016; 37: 2129-2200.

2. Mosterd A, Hoes AW. Clinical epidemiology of heart failure. Heart 2007; 93: 1137-1146.

3. Paulus WJ, van Ballegoij JJ. Treatment of heart failure with normal ejection fraction: an inconve-nient truth! J Am Coll Cardiol 2010; 55: 526-537.

4. McMurray JJ, Packer M, Desai AS, Gong J, Lefkowitz MP, Rizkala AR, Rouleau JL, Shi VC, Solomon SD, Swedberg K, Zile MR, PARADIGM-HF Investigators and Committees. Angiotensin-neprilysin inhibition versus enalapril in heart failure. N Engl J Med 2014; 371: 993-1004.

5. Maggioni AP, Dahlstrom U, Filippatos G, Chioncel O, Crespo Leiro M, Drozdz J, Fruhwald F, Gulles-tad L, Logeart D, Fabbri G, Urso R, Metra M, Parissis J, Persson H, Ponikowski P, Rauchhaus M, Voors AA, Nielsen OW, Zannad F, Tavazzi L, Heart Failure Association of the European Society of Cardiology (HFA). EURObservational Research Programme: regional differences and 1-year follow-up results of the Heart Failure Pilot Survey (ESC-HF Pilot). Eur J Heart Fail 2013; 15: 808-817. 6. Goldberg RJ, Ciampa J, Lessard D, Meyer TE, Spencer FA. Long-term survival after heart failure: a

contemporary population-based perspective. Arch Intern Med 2007; 167: 490-496.

7. Iwaz JA, Lee E, Aramin H, Romero D, Iqbal N, Kawahara M, Khusro F, Knight B, Patel MV, Sharma S, Maisel AS. New Targets in the Drug Treatment of Heart Failure. Drugs 2016; 76: 187-201.

8. Kaye DM, Krum H. Drug discovery for heart failure: a new era or the end of the pipeline? Nat Rev

Drug Discov 2007; 6: 127-139.

9. Watson JD, Crick FH. Molecular structure of nucleic acids: a structure for deoxyribose nucleic acid. J.D. Watson and F.H.C. Crick. Published in Nature, number 4356 April 25, 1953. Nature 1974; 248: 765.

10. Alberts B, Bray D, Hopkin K. From DNA to protein: how cells read the genome. Essential Cell

Biol-ogy. 3rd ed. New York (NY): Garland Science; 2009. p. 231-260.

11. Gibney ER, Nolan CM. Epigenetics and gene expression. Heredity (Edinb) 2010; 105: 4-13.

12. Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993; 75: 843-854.

13. Wightman B, Ha I, Ruvkun G. Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell 1993; 75: 855-862.

14. Lagos-Quintana M, Rauhut R, Lendeckel W, Tuschl T. Identification of novel genes coding for small expressed RNAs. Science 2001; 294: 853-858.

15. Lau NC, Lim LP, Weinstein EG, Bartel DP. An abundant class of tiny RNAs with probable regulatory roles in Caenorhabditis elegans. Science 2001; 294: 858-862.

16. Lee RC, Ambros V. An extensive class of small RNAs in Caenorhabditis elegans. Science 2001; 294: 862-864.

17. Ha M, Kim VN. Regulation of microRNA biogenesis. Nat Rev Mol Cell Biol 2014; 15: 509-524. 18. Ludwig N, Leidinger P, Becker K, Backes C, Fehlmann T, Pallasch C, Rheinheimer S, Meder B,

Stahler C, Meese E, Keller A. Distribution of miRNA expression across human tissues. Nucleic Acids

(12)

1

19. Chistiakov DA, Orekhov AN, Bobryshev YV. Cardiac-specific miRNA in cardiogenesis, heart func-tion, and cardiac pathology (with focus on myocardial infarction). J Mol Cell Cardiol 2016; 94: 107-121.

20. Zhao Y, Ransom JF, Li A, Vedantham V, von Drehle M, Muth AN, Tsuchihashi T, McManus MT, Schwartz RJ, Srivastava D. Dysregulation of cardiogenesis, cardiac conduction, and cell cycle in mice lacking miRNA-1-2. Cell 2007; 129: 303-317.

21. Callis TE, Pandya K, Seok HY, Tang RH, Tatsuguchi M, Huang ZP, Chen JF, Deng Z, Gunn B, Shu-mate J, Willis MS, Selzman CH, Wang DZ. MicroRNA-208a is a regulator of cardiac hypertrophy and conduction in mice. J Clin Invest 2009; 119: 2772-2786.

22. Duygu B, de Windt LJ, da Costa Martins PA. Targeting microRNAs in heart failure. Trends

Cardio-vasc Med 2016; 26: 99-110.

23. Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, Peterson A, Note-boom J, O’Briant KC, Allen A, Lin DW, Urban N, Drescher CW, Knudsen BS, Stirewalt DL, Gentle-man R, Vessella RL, Nelson PS, Martin DB, Tewari M. Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci U S A 2008; 105: 10513-10518.

24. Weber JA, Baxter DH, Zhang S, Huang DY, Huang KH, Lee MJ, Galas DJ, Wang K. The microRNA spectrum in 12 body fluids. Clin Chem 2010; 56: 1733-1741.

25. Chen X, Ba Y, Ma L, Cai X, Yin Y, Wang K, Guo J, Zhang Y, Chen J, Guo X, Li Q, Li X, Wang W, Zhang Y, Wang J, Jiang X, Xiang Y, Xu C, Zheng P, Zhang J, Li R, Zhang H, Shang X, Gong T, Ning G, Wang J, Zen K, Zhang J, Zhang CY. Characterization of microRNAs in serum: a novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res 2008; 18: 997-1006.

26. Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol 2007; 9: 654-659.

27. Pegtel DM, Cosmopoulos K, Thorley-Lawson DA, van Eijndhoven MA, Hopmans ES, Lindenberg JL, de Gruijl TD, Wurdinger T, Middeldorp JM. Functional delivery of viral miRNAs via exosomes. Proc

Natl Acad Sci U S A 2010; 107: 6328-6333.

28. Bang C, Batkai S, Dangwal S, Gupta SK, Foinquinos A, Holzmann A, Just A, Remke J, Zimmer K, Zeug A, Ponimaskin E, Schmiedl A, Yin X, Mayr M, Halder R, Fischer A, Engelhardt S, Wei Y, Schober A, Fiedler J, Thum T. Cardiac fibroblast-derived microRNA passenger strand-enriched exosomes mediate cardiomyocyte hypertrophy. J Clin Invest 2014; 124: 2136-2146.

29. Schmitter D, Cotter G, Voors AA. Clinical use of novel biomarkers in heart failure: towards person-alized medicine. Heart Fail Rev 2014; 19: 369-381.

30. Romaine SP, Tomaszewski M, Condorelli G, Samani NJ. MicroRNAs in cardiovascular disease: an introduction for clinicians. Heart 2015; 101: 921-928.

31. Vogel B, Keller A, Frese KS, Leidinger P, Sedaghat-Hamedani F, Kayvanpour E, Kloos W, Backe C, Thanaraj A, Brefort T, Beier M, Hardt S, Meese E, Katus HA, Meder B. Multivariate miRNA signa-tures as biomarkers for non-ischaemic systolic heart failure. Eur Heart J 2013; 34: 2812-2822. 32. Voellenkle C, van Rooij J, Cappuzzello C, Greco S, Arcelli D, Di Vito L, Melillo G, Rigolini R, Costa E,

Crea F, Capogrossi MC, Napolitano M, Martelli F. MicroRNA signatures in peripheral blood mono-nuclear cells of chronic heart failure patients. Physiol Genomics 2010; 42: 420-426.

33. Corsten MF, Dennert R, Jochems S, Kuznetsova T, Devaux Y, Hofstra L, Wagner DR, Staessen JA, Heymans S, Schroen B. Circulating MicroRNA-208b and MicroRNA-499 reflect myocardial damage in cardiovascular disease. Circ Cardiovasc Genet 2010; 3: 499-506.

34. Tijsen AJ, Creemers EE, Moerland PD, de Windt LJ, van der Wal AC, Kok WE, Pinto YM. MiR423-5p as a circulating biomarker for heart failure. Circ Res 2010; 106: 1035-1039.

(13)

20

Chapter 1

35. Ellis KL, Cameron VA, Troughton RW, Frampton CM, Ellmers LJ, Richards AM. Circulating microR-NAs as candidate markers to distinguish heart failure in breathless patients. Eur J Heart Fail 2013; 15: 1138-1147.

36. Wong LL, Armugam A, Sepramaniam S, Karolina DS, Lim KY, Lim JY, Chong JP, Ng JY, Chen YT, Chan MM, Chen Z, Yeo PS, Ng TP, Ling LH, Sim D, Leong KT, Ong HY, Jaufeerally F, Wong R, Chai P, Low AF, Lam CS, Jeyaseelan K, Richards AM. Circulating microRNAs in heart failure with reduced and preserved left ventricular ejection fraction. Eur J Heart Fail 2015; 17: 393-404.

37. Watson CJ, Gupta SK, O’Connell E, Thum S, Glezeva N, Fendrich J, Gallagher J, Ledwidge M, Grote-Levi L, McDonald K, Thum T. MicroRNA signatures differentiate preserved from reduced ejection fraction heart failure. Eur J Heart Fail 2015; 17: 405-415.

(14)
(15)

Referenties

GERELATEERDE DOCUMENTEN

To probe our hypothesis that cardiac secreted factors of the failing heart could be re- sponsible, we conducted a literature search from databases from myocardial

Nevertheless, a large number of HF trials exist, and in the event that specific drugs counteract risk factors associated with cancer develop- ment, or when they attenuate

Deze studies laten zien dat biomarkers bruikbaar zijn in de klinische praktijk, voor zowel het inschatten van de kans op een heropname als voor het identificeren van patiënten met

deposition in WT mice with UUO compared to galectin-3 KO counterparts, and immunohistological staining for α-SMA was markedly reduced in galectin-3-KO mice.26 In chronic

Financial support by the Dutch Heart Foundation for the publication of this thesis is gratefully acknowledged. The research described in this thesis was supported by a grant from

Although there is increasing interest in circulating miRNAs in heart failure, there are still major uncertainties about their origin and function in the circulation. Some speculate

(AHF) patients at admission (Wroclaw, validation study) compared with healthy controls (continued) miRNAs fold change P-value miRNAs fold change P-value. (AHf vs. controls)

The majority of correlations between circulating AHF-specific miRNAs were related to biomarkers predictive for a worse clinical outcome in a subgroup of worsening heart failure