Acessibilidade / Reportar erro

Physical exercise and microRNAs: new frontiers in heart failure

Abstracts

Although the impact of exercise on survival of patients with heart failure has been recently questioned, exercise training improves quality of life, functional capacity, inflammation, endothelial and autonomic function. In recent years, interest has increased regarding a group of small non-protein coding RNAs called microRNAs. Studies have shown that the expression of these molecules changes in several pathological conditions, such as myocardial infarction, myocardial ischemia and heart failure, and when clinical improvement occurs, they seem to normalize. With the potential for practical applicability, markers that may be useful in diagnostic and prognostic assessment of heart failure have been identified, such as miR-423-5p. In addition, results of experimental studies have indicated that there are potential therapeutic effects of microRNAs. MicroRNAs are involved in the regulation of gene expression during fetal development and in adult individuals, increasing or decreasing in the heart in response to physiological stress, injury or hemodynamic overload. Thus, the study of the behavior of these molecules in physical exercise has brought important information about the effects of this therapeutic modality and represents a new era in the understanding of heart failure. This review aims to integrate the evidence on microRNAs in heart failure with greater relevance in the study of physical exercise.

Exercise; microRNAs; heart rate


Embora recentemente tenha sido questionado o impacto do exercício na sobrevida de pacientes com insuficiência cardíaca, o treinamento físico melhora a qualidade de vida, a capacidade funcional, a inflamação, a função autonômica e a função endotelial. Nos últimos anos, vem crescendo o interesse em um grupo de pequenos RNAs não codificadores de proteína chamados microRNAs. Estudos têm demonstrado que a expressão dessas moléculas se modifica em diversas condições patológicas, como a hipertrofia miocárdica, a isquemia miocárdica e a insuficiência cardíaca, e, quando ocorre melhora clínica, elas parecem se normalizar. Com o potencial de aplicabilidade prática, já foram identificados marcadores que poderão ser úteis na avaliação diagnóstica e prognóstica da insuficiência cardíaca, como o miR-423-5p. Além disso, resultados de estudos experimentais indicam haver possíveis efeitos terapêuticos dos microRNAs. Implicados na regulação da expressão genética durante o desenvolvimento fetal e no indivíduo adulto, os microRNAs aumentam ou diminuem no coração em resposta a estresse fisiológico, injúria ou sobrecarga hemodinâmica. Assim, o estudo do comportamento dessas moléculas no exercício físico vem trazendo informações importantes quanto aos efeitos dessa modalidade terapêutica e representa uma nova era no entendimento da insuficiência cardíaca. Esta revisão tem por objetivo integrar as evidências sobre microRNAs na insuficiência cardíaca com maior relevância no estudo do exercício físico.

Exercício; microRNAs; insuficiência cardíaca


REVIEW ARTICLE

Physical exercise and microRNAs: new frontiers in heart failure

Miguel Morita Fernandes-SilvaI, II; Vagner Oliveira CarvalhoI; Guilherme Veiga GuimarãesI; Fernando BacalI; Edimar Alcides BocchiI

IInstituto do Coração do Hospital das Clínicas de São Paulo-INCOR, São Paulo, SP

IIHospital Cardiológico Costantini, Curitiba, PR, Brasil

Correspondence

ABSTRACT

Although the impact of exercise on survival of patients with heart failure has been recently questioned, exercise training improves quality of life, functional capacity, inflammation, endothelial and autonomic function. In recent years, interest has increased regarding a group of small non-protein coding RNAs called microRNAs.

Studies have shown that the expression of these molecules changes in several pathological conditions, such as myocardial infarction, myocardial ischemia and heart failure, and when clinical improvement occurs, they seem to normalize. With the potential for practical applicability, markers that may be useful in diagnostic and prognostic assessment of heart failure have been identified, such as miR-423-5p. In addition, results of experimental studies have indicated that there are potential therapeutic effects of microRNAs.

MicroRNAs are involved in the regulation of gene expression during fetal development and in adult individuals, increasing or decreasing in the heart in response to physiological stress, injury or hemodynamic overload. Thus, the study of the behavior of these molecules in physical exercise has brought important information about the effects of this therapeutic modality and represents a new era in the understanding of heart failure.

This review aims to integrate the evidence on microRNAs in heart failure with greater relevance in the study of physical exercise.

Keywords: Exercise; microRNAs; heart rate / physiopathology.

Introduction

Several advances in the understanding of the physiopathology of heart failure (HF) allowed the development of new therapeutic modalities, or even optimized them, with a consequent increase in survival. Nevertheless, HF is still a condition with high mortality and morbidity, being the leading cause of hospitalization due to cardiovascular disease in Brazil1.

In spite of recent evidence questioning the impact of exercise on survival2, physical training improves quality of life3, functional capacity4, inflammation5, and autonomic4 and endothelial function4. The mechanisms involved in this phenomenon are not yet fully understood. In recent years, interest has increased in the area of research involving aspects related to the body's response to exercise in the genetic scenario, in which microRNAs appear to play a key role.

It has become increasingly evident that microRNAs play a critical role in many biological processes, and research in cancer and cardiovascular diseases have been the main focus in translational medicine. Recent studies have evaluated the expression of several microRNAs in myocardial hypertrophy, myocardial infarction and heart failure, which will improve the understanding of the physiopathology of these clinical conditions, in addition to their promising use in the diagnosis and prognosis of major cardiovascular diseases6,7. Moreover, the possibility of mimicking microRNAs with reduced expression or antagonizing those of which increased expression would be causally linked to a specific pathology, represent a therapeutic potential and new paradigm in the management of heart diseases.

Implicated in the regulation of gene expression during fetal development and in adult individuals8,9, many of these microRNAs increase or decrease in the heart in response to physiological stress, injury or hemodynamic overload10. The study of the behavior of these molecules in physical exercise therefore represents a great potential for new discoveries of this type of therapeutic modality in HF.

This review aims to integrate the evidence for microRNAs in heart failure with greater relevance in the study of physical exercise.

MicroRNAs

In response to external stimuli such as exercise, gene expression can be modulated by different mechanisms, including gene silencing by small RNAs, including the microRNAs (miRNAs)11.

The miRNAs, initially described in 1993 when studying the development of nematodes12, are characterized as a group of small non-protein-coding RNAs, with approximately 19-25 nucleotides in length. Unlike the wide range of RNAs encoded by the human genome, this variety of RNA has been noted for its unique ability to modulate an enormous and complex regulatory network of gene expression13.

It is currently known that, in general, miRNAs are synthesized from specific genes or certain genetic regions that are not associated with protein production (introns)11. The maturation process of miRNAs involves a complex metabolic pathway that begins in the nucleus and extends to the cell cytoplasm (Fig. 1). The first step in the miRNA maturation is the transcription of a long primary miRNA strand (pri-miRNA) from a certain gene14. The pri-miRNA is cleaved by an enzyme complex called Drosha, releasing precursor regions called pre-miRNAs, which will form distinct miRNAs15. In the cytoplasm, after being exported from the nucleus by exportin-516, the pre-miRNA is cleaved by the Dicer enzyme, forming a double-stranded RNA with approximately 22 nucleotides17. The two strands are then separated; however, only one of them will potentially act as a functional miRNA, while the other is generally degraded18.


In its mature form, miRNAs, with the help of an enzyme complex called RNA-Induced Silencing Complex (RISC), binds to the target messenger RNA (mRNA). This binding prevents the ribosomes from accessing the genetic information contained in the mRNA, resulting in decreased protein synthesis of the target gene19,20.

In general, the function of miRNAs is to act as a recognition component for RISC, as they bind to mRNA, identifying it as a target. The complexity of protein expression regulation by miRNA is illustrated by the fact that a single miRNA can regulate hundreds of distinct target genes, and otherwise cooperate to control a single target gene 2,13.

More than 700 human miRNAs have been identified and there seems to be 150 to 200 of them expressed in the heart10. This number is expected to grow even further, due to the recent development of sequencing technologies and computational prediction methods19,20. Although the biological functions of miRNAs are not fully understood, it is believed that 30% to 60% of the protein-coding genes are regulated by miRNAs11.

Studies have shown that although miRNAs are expressed in various cell types, specific miRNAs are synthesized exclusively in certain tissues or cell groups19,20.

MicroRNAs and heart failure

In recent years, it has been shown that many miRNAs are expressed in a specific way in some cardiovascular conditions. Specifically, there is a differential expression in hypertrophy, myocardial ischemia and endothelial dysfunction.

Systolic HF is characterized by ventricular dilatation and remodeling and there seems to be an activation of a fetal gene program that triggers pathological changes in the myocardium, associated with progressive ventricular dysfunction21. A pattern of miRNA expression very similar to that found in fetal hearts has been identified in the heart of patients with advanced-stage HF21. Moreover, in a study evaluating22 patients with dilated, ischemic and valvular cardiomyopathy (aortic stenosis), the expression profile was different according to the etiology of HF, and the miRNA signature was able to predict diagnosis with an accuracy of 70%.

Four families of miRNAs are highly expressed in the heart: miR-1, miR-133, miR-208 and miR-499. The miR-1 family represents 40% of all miRNAs expressed in the heart, whereas the miR-208 is the only one known as cardio-specific. Table 1 contains some miRNAs and their possible biological effects on the adult heart.

The miR-1 is reduced in myocardial hypertrophy, and in vitro23 and in vivo24 studies indicate that there is a causal relationship, in which the reduction of this miRNA is a necessary condition for cell mass increasing25. An experiment in rats26 showed that one of the earliest changes observed after pressure overload to the heart is the reduction in miR-1, even before the increase in cardiac mass. The miR-1 is reduced in the hypertrophic ventricle of patients with acromegaly26 and aortic stenosis22 with preserved ejection fraction.

On the other hand, the expression of miR-1 shows different findings in HF. While some authors22,27,28 observed a reduction in miR-1 in ischemic and non-ischemic dilated cardiomyopathy, others observed an increase21,29. Han et al25 suggest the possibility that miR-1 is reduced in hypertrophy, but returns to normal or above normal when the condition progresses to HF.

The family of miR-208 is restricted to the heart and consists of the miRs-208a and -208b, which are encoded in introns of genes of myosin heavy chains (MHC) α and β, respectively30,31. The interactions of these miRNAs with the MHC genes are involved in the development of hypertrophy in adults.

Other miRNAs also show a differentiated expression in hypertrophy and myocardial ischemia. Some examples are shown in Table 2.

Changing in miRNA expression in response to heart functional alterations have also been demonstrated. An interesting study29 evaluated the profiles of mRNA and miRNA in 10 hearts treated with left ventricular assistance device (LVAD), 17 hearts of individuals with HF not treated with LVAD and 11 controls (no HF). In the hearts of individuals with HF without LVAD, a significant increase in the expression of 28 miRNAs was observed, while in those treated with LVAD, 20 of these miRNAs were completely normalized, and the other eight tended to normalization. Experimental studies32 have shown that a phenotype can be reversed through the inhibition of a specific miRNA that has an increased expression in this condition. This can be accomplished by administering an anti-miRNA oligonucleotide, which acts as a competitive inhibitor of that miRNA and is called antagomiR. Recently, Montgomery et al33 demonstrated that administration of an anti-miR-208a antagomiR was able to prevent ventricular hypertrophy, cardiac remodeling and mortality in Dahl rats due to HF (sensitive to salt) subjected to a hypersodic diet.

The study strongly suggests that the effects of antagomiR were due to reduced levels of miR-208a. This result not only establishes a causal correlation of this miRNA with the mechanism of hypertrophy, but also brings a very promising therapeutic use to reverse remodeling. However, in adult rats, the profile miR-208a/α-MHC is predominant, and when pressure overload occurs, there is a shift in the production from α to β-MHC30,31. In humans, the role of this mechanism in hypertrophy remains to be clarified, as this species is not able to reverse the MHC isoforms, and the β-MHC is the predominant form, together with the miR-208b31. In addition, experimental models must be viewed with caution when extrapolating data, as they are artificial and may not accurately mimic the clinical conditions observed in humans10.

MicroRNA as biomarker in heart failure

For a widespread use of miRNA as a biomarker in heart failure, its determination must be simple and reliable, preferably dispensing with the need to perform an invasive procedure such as a biopsy. Although the research of miRNAs in tissues and cells have been carried out for some time, only recently it was found the existence of a series of these miRNAs in circulating blood. Mitchell et al34 demonstrated in patients with prostate cancer that tissue-specific circulating miRNAs could be detected in plasma. The mechanisms by which they are released are uncertain, but it has been postulated to be through the secretion within microvesicles called exosomes35. It is unknown whether this occurs with other miRNAs, but characteristics such as specificity and stability in plasma make it very promising as a marker of tissue injury36.

The miRs-208a, -208b and -499 miRNAs are specific to cardiomyocytes and were evaluated in the diagnosis of conditions that cause myocardial injury. There was an increase of more than 1,000-fold in acute myocardial infarction6,37-39 of miRs-208b and-499. In this condition, they reflect tissue injury and cell death. In addition, miR-208a showed diagnostic accuracy that was similar to troponin, with the advantage of being detected earlier in plasma6.

In order to determine which could be used as biomarkers in HF, Tijsen et al40 compared the expression profile of miRNAs in the plasma of 12 healthy subjects with 12 patients admitted for acute HF. Among 108 miRNAs that showed significant difference between the groups, 16 were selected and validated in a larger group, including 30 patients with HF, 20 who had dyspnea due to other causes rather than the HF, and a third group of 39 healthy individuals. In this validation cohort, an elevated level of mir-423-5p was strongly associated with the diagnosis of HF in a logistic regression model including age and sex. The mechanisms by which there is an increase in this marker in patients with HF remains unknown. Whether this is due to cell damage and subsequent release of that in plasma, or is related to a specific secretory pathway of a given cell, it remains to be clarified41. In another study7, plasma concentrations of miR-126 showed an inverse correlation with the levels of brain natriuretic peptide (BNP). However, the patient sample was heterogeneous and the criteria for HF were not well established. More studies are needed to define the role of these miRNAs as biomarkers in HF.

MicroRNA and exercise

Physical exercise, pregnancy and the individual's growth are stimuli for physiological increase of the heart. In the mid-nineteenth century, descriptions of the so-called "athlete's heart" had deleterious connotations8. More recently, the multiple benefits of physical training on survival and the harmful effects of a sedentary lifestyle made this therapeutic modality required to treat patients with heart disease, including those with systolic ventricular dysfunction, who have already shown a pathological increasing in ventricular volumes and masses. Its benefits regarding functional capacity can be explained by effects on endothelial function, peripheral vascular resistance and changes in the structure of skeletal muscle, although significant improvement in left ventricular ejection fraction have not been demonstrated.

The study of microRNAs may generate hypotheses about the mechanisms by which exercise affects the pathophysiology of HF. Interestingly, the signaling pathways leading to hypertrophy due to physiological stimuli such as exercise, are different from those that cause pathological hypertrophy and thus may trigger different expressions in miRNAs8. Ischemic preconditioning and its relationship with the miRNA is already being evaluated in experimental studies42.

When analyzing individual responses to physical training, doubts are raised concerning the reasons for the variability of their results. There is evidence that physiological adaptability to exercise has a parallel with gene expression43. However, the complexity of the genetic interaction limits the identification of individual genes that may explain this variability44. Moreover, its expression changes according to the stimulus that the body receives and it is in this aspect that the study of miRNAs has a promising role.

The skeletal muscle is also an organ with high plasticity, capable of altering the phenotype in response to mechanical overload45. Experimental studies have identified changes in the skeletal muscle profile of specific miRNAs in aerobic and resistance exercises46 (Table 3).

There are several miRNAs present in skeletal muscle, and miRs-1, -133a, -133b and -206 comprise 25% of them, often being referred to as "myomirs"47. In healthy subjects, these four myomirs decrease significantly after 12 weeks of aerobic training48, indicating that they adjust quickly to the level of physical activity. Interestingly, these levels return to baseline after 14 days of its discontinuation. When evaluating their response to a single exercise session, an increased expression of miR-1 and 133a was observed only before the training period.

With respect to resistance exercise, the gain in muscle mass is also highly variable among individuals. This variability is also accompanied by differences in the behavior of miRNAs. A study47 that analyzed biopsies of the vastus lateralis muscle of 56 men undergoing resistance training for 12 weeks found that the expression of miR-378 decreased and that of miR-478 increased, both significantly, only in those who responded little to the training. In addition, there was a strong correlation between the variation in the expression of miR-378 with the gain in lean body mass. With respect to myomirs, resistance exercise did not change miRs-1 and -133 in this study.

With respect to the levels of miRNAs in circulating blood, Baggish et al49 studied the behavior of miRNAs that were previously implicated in angiogenesis (miR-20a, miR-210, miR-221, miR-222, miR-328), inflammation (miR-21, miR-146a), cardiac and skeletal muscle contractility (miR-21, miR-133a) and muscular adaptation to hypoxia and ischemia (miR-21, miR-146a, and miR-210). The results of this and other studies involving the behavior of miRNAs in exercise in humans are summarized in Table 4.

The study of miRNAs can also help to understand the effects of exercise on immune response, of which mechanisms remain unclear. For instance, based on the analysis of cytokines, it was observed that both pro-inflammatory and anti-inflammatory pathways are activated after strenuous exercise50. Studying the behavior of neutrophils in peripheral blood51 after a workout, it was possible to observe changes in the expression profile of miRNAs, suggesting that these contribute to changes in gene expression of immunocompetent cells52.

By the time of publication of this review, we are not aware of studies that have evaluated the behavior of miRNAs in response to physical training specifically in patients with HF.

Perspectives

Much has been discovered on the effects of exercise training on HF. In addition to improving functional capacity and survival, other evidence has shown positive results of this therapy, such as an increasing in circulating endothelial progenitor cells53, anti-inflammatory effects and effects on the endothelium, and changing in levels of interleukin-1β54, -654,55, -1056, tumor necrosis factor-α and its receptors 1 and 2 and C-reactive protein56, inducible nitric oxide synthase (iNOS)54, soluble intracellular adhesion molecule (sICAM)55, adiponectin57,58, brain natriuretic peptide (BNP)59 and others (Fig. 2). Therefore, a promising tool emerges for diagnostic and prognostic evaluation of treatment in HF, which will increase the knowledge of the effects of physical exercise in these patients, going deeper into the molecular mechanisms, with the advantage of reflecting more directly the genetic signaling pathways, linking environmental stimuli to the hereditary aspects of the individual. MicroRNAs represent a new era in knowledge of heart failure and the possible effects that exercise has on it.


Potential Conflict of Interest

No potential conflict of interest relevant to this article was reported.

Sources of Funding

This study was funded by Fundação de Apoio à Pesquisa do Estado de São Paulo (FAPESP).

Study Association

This article is part of the thesis of doctoral submitted by Miguel Morita Fernandes da Silva, from Universidade de São Paulo.

References

  • 1
    Ministério da Saúde. Dados do Sistema de Informações Hospitalares do Sistema Único de Saúde - DATASUS - SIH / SUS. [Acesso em 2011 jul 15]. Disponível em: http://tabnet.datasus.gov.br/cgi/tabcgi.exe?sih/cnv/niuf.def, 2011.
  • 2. O'Connor CM, Whellan DJ, Lee KL, Keteyian SJ, Cooper LS, Ellis SJ, et al. Efficacy and safety of exercise training in patients with chronic heart failure: HF-ACTION randomized controlled trial. JAMA. 2009;301(14):1439-50.
  • 3. Flynn KE, Pina IL, Whellan DJ, Lin L, Blumenthal JA, Ellis SJ, et al. Effects of exercise training on health status in patients with chronic heart failure: HF-ACTION randomized controlled trial. JAMA. 2009;301(14):1451-9.
  • 4. Downing J, Balady GJ. The role of exercise training in heart failure. J Am Coll Cardiol. 2011;58(6):561-9.
  • 5. Niebauer J. Effects of exercise training on inflammatory markers in patients with heart failure. Heart Fail Rev. 2008;13(1):39-49.
  • 6. Wang GK, Zhu JQ, Zhang JT, Li Q, Li Y, He J, et al. Circulating microRNA: a novel potential biomarker for early diagnosis of acute myocardial infarction in humans. Eur Heart J. 2010;31(6):659-66.
  • 7. Fukushima Y, Nakanishi M, Nonogi H, Goto Y, Iwai N. Assessment of plasma miRNAs in congestive heart failure. Circ J. 2011;75(2):336-40.
  • 8. Hill JA, Olson EN. Cardiac plasticity. N Engl J Med. 2008;358(13):1370-80.
  • 9. Olson EN. Gene regulatory networks in the evolution and development of the heart. Science. 2006;313(5795):1922-7.
  • 10. Condorelli G, Latronico MV, Dorn GW 2nd microRNAs in heart disease: putative novel therapeutic targets? Eur Heart J. 2010;31(6):649-58.
  • 11. Rodriguez A, Griffiths-Jones S, Ashurst JL, Bradley A. Identification of mammalian microRNA host genes and transcription units. Genome Res. 2004;14(10A):1902-10.
  • 12. Miranda KC, Huynh T, Tay Y, Ang YS, Tam WL, Thomson AM, et al. A pattern-based method for the identification of MicroRNA binding sites and their corresponding heteroduplexes. Cell. 2006;126(6):1203-17.
  • 13. Lewis BP, Burge CB, Bartel DP. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 2005;120(1):15-20.
  • 14. Lee Y, Kim M, Han J, Yeom KH, Lee S, Baek SH, et al. MicroRNA genes are transcribed by RNA polymerase II. EMBO J. 2004;23(20):4051-60.
  • 15. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116(2):281-97.
  • 16. Lund E, Guttinger S, Calado A, Dahlberg JE, Kutay U. Nuclear export of microRNA precursors. Science. 2004;303(5654):95-8.
  • 17. Hutvagner G, McLachlan J, Pasquinelli AE, Balint E, Tuschl T, Zamore PD. A cellular function for the RNA-interference enzyme Dicer in the maturation of the let-7 small temporal RNA. Science. 2001;293(5531):834-8.
  • 18. Schwarz DS, Hutvagner G, Du T, Xu Z, Aronin N, Zamore PD. Asymmetry in the assembly of the RNAi enzyme complex. Cell. 2003;115(2):199-208.
  • 19. Kloosterman WP, Plasterk RH. The diverse functions of microRNAs in animal development and disease. Dev Cell. 2006;11(4):441-50.
  • 20. Beuvink I, Kolb FA, Budach W, Garnier A, Lange J, Natt F, et al. A novel microarray approach reveals new tissue-specific signatures of known and predicted mammalian microRNAs. Nucleic Acids Res. 2007;35(7):e52.
  • 21. Thum T, Galuppo P, Wolf C, Fiedler J, Kneitz S, van Laake LW, et al. MicroRNAs in the human heart: a clue to fetal gene reprogramming in heart failure. Circulation. 2007;116(3):258-67.
  • 22. Ikeda S, Kong SW, Lu J, Bisping E, Zhang H, Allen PD, et al. Altered microRNA expression in human heart disease. Physiol Genomics. 2007;31(3):367-73.
  • 23. Elia L, Contu R, Quintavalle M, Varrone F, Chimenti C, Russo MA, et al. Reciprocal regulation of microRNA-1 and insulin-like growth factor-1 signal transduction cascade in cardiac and skeletal muscle in physiological and pathological conditions. Circulation. 2009;120(23):2377-85.
  • 24. Care A, Catalucci D, Felicetti F, Bonci D, Addario A, Gallo P, et al. MicroRNA-133 controls cardiac hypertrophy. Nat Med. 2007;13(5):613-8.
  • 25. Han M, Toli J, Abdellatif M. MicroRNAs in the cardiovascular system. Curr Opin Cardiol. 2011;26(3):181-9.
  • 26. Sayed D, Hong C, Chen IY, Lypowy J, Abdellatif M. MicroRNAs play an essential role in the development of cardiac hypertrophy. Circ Res. 2007;100(3):416-24.
  • 27. Sucharov C, Bristow MR, Port JD. miRNA expression in the failing human heart: functional correlates. J Mol Cell Cardiol. 2008;45(2):185-92.
  • 28. Naga Prasad SV, Duan ZH, Gupta MK, Surampudi VS, Volinia S, Calin GA, et al. Unique microRNA profile in end-stage heart failure indicates alterations in specific cardiovascular signaling networks. J Biol Chem. 2009;284(40):27487-99.
  • 29. Matkovich SJ, Van Booven DJ, Youker KA, Torre-Amione G, Diwan A, Eschenbacher WH, et al. Reciprocal regulation of myocardial microRNAs and messenger RNA in human cardiomyopathy and reversal of the microRNA signature by biomechanical support. Circulation. 2009;119(9):1263-71.
  • 30. Schroen B, Heymans S. MicroRNAs and beyond: the heart reveals its treasures. Hypertension. 2009;54(6):1189-94.
  • 31. Dorn GW 2nd MicroRNAs in cardiac disease. Transl Res. 2011;157(4):226-35.
  • 32. da Costa Martins PA, Salic K, Gladka MM, Armand AS, Leptidis S, el Azzouzi H, et al. MicroRNA-199b targets the nuclear kinase Dyrk1a in an auto-amplification loop promoting calcineurin/NFAT signalling. Nat Cell Biol. 2010;12(12):1220-7.
  • 33. Montgomery RL, Hullinger TG, Semus HM, Dickinson BA, Seto AG, Lynch JM, et al. Therapeutic inhibition of miR-208a improves cardiac function and survival during heart failure. Circulation. 2011;124(14):1537-47.
  • 34. Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci U S A. 2008;105(30):10513-8.
  • 35. Hunter MP, Ismail N, Zhang X, Aguda BD, Lee EJ, Yu L, et al. Detection of microRNA expression in human peripheral blood microvesicles. PLoS One. 2008;3(11):e3694.
  • 36. Laterza OF, Lim L, Garrett-Engele PW, Vlasakova K, Muniappa N, Tanaka WK, et al. Plasma MicroRNAs as sensitive and specific biomarkers of tissue injury. Clin Chem. 2009;55(11):1977-83.
  • 37. Corsten MF, Dennert R, Jochems S, Kuznetsova T, Devaux Y, Hofstra L, et al. Circulating MicroRNA-208b and MicroRNA-499 reflect myocardial damage in cardiovascular disease. Circ Cardiovasc Genet. 2010;3(6):499-506.
  • 38. D'Alessandra Y, Devanna P, Limana F, Straino S, Di Carlo A, Brambilla PG, et al. Circulating microRNAs are new and sensitive biomarkers of myocardial infarction. Eur Heart J. 2010;31(22):2765-73.
  • 39. Adachi T, Nakanishi M, Otsuka Y, Nishimura K, Hirokawa G, Goto Y, et al. Plasma microRNA 499 as a biomarker of acute myocardial infarction. Clin Chem. 2010;56(7):1183-5.
  • 40. Tijsen AJ, Creemers EE, Moerland PD, de Windt LJ, van der Wal AC, Kok WE, et al. MiR423-5p as a circulating biomarker for heart failure. Circ Res. 2010;106(6):1035-9.
  • 41. Elton TS, Khan M, Terentyev D. MicroRNAs in cardiovascular disease. F1000 Med Rep. 2011;3:10-4.
  • 42. Kim HW, Haider HK, Jiang S, Ashraf M. Ischemic preconditioning augments survival of stem cells via miR-210 expression by targeting caspase-8-associated protein 2. J Biol Chem. 2009;284(48):33161-8.
  • 43. Timmons JA, Jansson E, Fischer H, Gustafsson T, Greenhaff PL, Ridden J, et al. Modulation of extracellular matrix genes reflects the magnitude of physiological adaptation to aerobic exercise training in humans. BMC Biol. 2005;3:19.
  • 44. Keller P, Vollaard N, Babraj J, Ball D, Sewell DA, Timmons JA. Using systems biology to define the essential biological networks responsible for adaptation to endurance exercise training. Biochem Soc Trans. 2007;35(Pt 5):1306-9.
  • 45. Hoppeler H, Fluck M. Normal mammalian skeletal muscle and its phenotypic plasticity. J Exp Biol. 2002;205(Pt 15):2143-52.
  • 46. Guller I, Russell AP. MicroRNAs in skeletal muscle: their role and regulation in development, disease and function. J Physiol. 2010;588(Pt 21):4075-87.
  • 47. Davidsen PK, Gallagher IJ, Hartman JW, Tarnopolsky MA, Dela F, Helge JW, et al. High responders to resistance exercise training demonstrate differential regulation of skeletal muscle microRNA expression. J Appl Physiol. 2011;110(2):309-17.
  • 48. Nielsen S, Scheele C, Yfanti C, Akerstrom T, Nielsen AR, Pedersen BK, et al. Muscle specific microRNAs are regulated by endurance exercise in human skeletal muscle. J Physiol. 2010;588(Pt 20):4029-37.
  • 49. Baggish AL, Hale A, Weiner RB, Lewis GD, Systrom D, Wang F, et al. Dynamic regulation of circulating microRNA during acute exhaustive exercise and sustained aerobic exercise training. J Physiol. 2011;589(Pt 16):3983-94.
  • 50. Ostrowski K, Rohde T, Asp S, Schjerling P, Pedersen BK. Pro- and anti-inflammatory cytokine balance in strenuous exercise in humans. J Physiol. 1999;515 (Pt 1):287-91.
  • 51. Radom-Aizik S, Zaldivar F Jr, Leu SY, Galassetti P, Cooper DM. Effects of 30 min of aerobic exercise on gene expression in human neutrophils. J Appl Physiol. 2008;104(1):236-43.
  • 52. Wessner B, Gryadunov-Masutti L, Tschan H, Bachl N, Roth E. Is there a role for microRNAs in exercise immunology? A synopsis of current literature and future developments. Exerc Immunol Rev. 2010;16:22-39.
  • 53. Sarto P, Balducci E, Balconi G, Fiordaliso F, Merlo L, Tuzzato G, et al. Effects of exercise training on endothelial progenitor cells in patients with chronic heart failure. J Card Fail. 2007;13(9):701-8.
  • 54. Gielen S, Adams V, Mobius-Winkler S, Linke A, Erbs S, Yu J, et al. Anti-inflammatory effects of exercise training in the skeletal muscle of patients with chronic heart failure. J Am Coll Cardiol. 2003;42(5):861-8.
  • 55. Niebauer J, Clark AL, Webb-Peploe KM, Coats AJ. Exercise training in chronic heart failure: effects on pro-inflammatory markers. Eur J Heart Fail. 2005;7(2):189-93.
  • 56. Batista ML Jr, Lopes RD, Seelaender MC, Lopes AC. Anti-inflammatory effect of physical training in heart failure: role of TNF-alpha and IL-10. Arq Bras Cardiol. 2009;93(6):643-51.
  • 57. Ferraz A, Bocchi E, Meneghelo R, Carvalhais C, Doi A, Guimaraes G, et al. Adiponectin is reduced by exercise training in chronic heart failure patients: a prospective randomized controlled study [abstract]. Eur Heart J. 2009;30 (Suppl 1):74.
  • 58. Van Berendoncks AM, Beckers P, Hoymans VY, Possemiers N, Wuytss FL, Vrints CJ, et al. Exercise training reduces circulating adiponectin levels in patients with chronic heart failure. Clin Sci (Lond). 2010;118(4):281-9.
  • 59. Maria Sarullo F, Gristina T, Brusca I, Milia S, Raimondi R, Sajeva M, et al. Effect of physical training on exercise capacity, gas exchange and N-terminal pro-brain natriuretic peptide levels in patients with chronic heart failure. Eur J Cardiovasc Prev Rehabil. 2006;13(5):812-7.
  • 60. Drummond MJ, McCarthy JJ, Fry CS, Esser KA, Rasmussen BB. Aging differentially affects human skeletal muscle microRNA expression at rest and after an anabolic stimulus of resistance exercise and essential amino acids. Am J Physiol Endocrinol Metab. 2008;295(6):E1333-40.
  • 61. Ringholm S, Bienso RS, Kiilerich K, Guadalupe-Grau A, Aachmann-Andersen NJ, Saltin B, et al. Bed rest reduces metabolic protein content and abolishes exercise-induced mRNA responses in human skeletal muscle. Am J Physiol Endocrinol Metab. 2011;301(4):E649-58.
  • 62. Fernandes T, Hashimoto NY, Magalhaes FC, Fernandes FB, Casarini DE, Carmona AK, et al. Aerobic exercise training-induced left ventricular hypertrophy involves regulatory microRNAs, decreased angiotensin-converting enzyme-angiotensin II, and synergistic regulation of angiotensin-converting enzyme 2-angiotensin (1-7). Hypertension. 2011;58(2):182-9.
  • 63. Aoi W, Naito Y, Mizushima K, Takanami Y, Kawai Y, Ichikawa H, et al. The microRNA miR-696 regulates PGC-1{alpha} in mouse skeletal muscle in response to physical activity. Am J Physiol Endocrinol Metab. 2010;298(4):E799-806.
  • 64. McCarthy JJ, Esser KA. MicroRNA-1 and microRNA-133a expression are decreased during skeletal muscle hypertrophy. J Appl Physiol. 2007;102(1):306-13.
  • 65. Safdar A, Abadi A, Akhtar M, Hettinga BP, Tarnopolsky MA. miRNA in the regulation of skeletal muscle adaptation to acute endurance exercise in C57Bl/6J male mice. PLoS One. 2009;4(5):e5610.
  • 66. Bauersachs J, Thum T. Biogenesis and regulation of cardiovascular microRNAs. Circ Res. 2011;109(3):334-47.
  • 67. Bonauer A, Carmona G, Iwasaki M, Mione M, Koyanagi M, Fischer A, et al. MicroRNA-92a controls angiogenesis and functional recovery of ischemic tissues in mice. Science. 2009;324(5935):1710-3.
  • 68. Harris TA, Yamakuchi M, Ferlito M, Mendell JT, Lowenstein CJ. MicroRNA-126 regulates endothelial expression of vascular cell adhesion molecule 1. Proc Natl Acad Sci U S A. 2008;105(5):1516-21.
  • 69. Li Q, Song XW, Zou J, Wang GK, Kremneva E, Li XQ, et al. Attenuation of microRNA-1 derepresses the cytoskeleton regulatory protein twinfilin-1 to provoke cardiac hypertrophy. J Cell Sci. 2010;123(Pt 14):2444-52.
  • Endereço para correspondência:
    Miguel Morita Fernandes da Silva
    Avenida Enéas de Carvalho Aguiar, 44, Bloco I, 1° Andar, Bairro Cerqueira Cesar
    CEP 05403-000, São Paulo, SP - Brasil
    E-mail:
  • Publication Dates

    • Publication in this collection
      31 July 2012
    • Date of issue
      May 2012

    History

    • Received
      29 Sept 2011
    • Accepted
      03 Nov 2011
    • Reviewed
      25 Oct 2011
    Sociedade Brasileira de Cardiologia - SBC Avenida Marechal Câmara, 160, sala: 330, Centro, CEP: 20020-907, (21) 3478-2700 - Rio de Janeiro - RJ - Brazil, Fax: +55 21 3478-2770 - São Paulo - SP - Brazil
    E-mail: revista@cardiol.br