Skip to main content

Abstract

The heart lodges in the mediastinum. It is usually situated in the middle of the thorax with its largest part in the left side; its apex points toward in the inferior and left direction. However, sometimes it resides on the right thoracic region (dextrocardia situs inversus totalis).

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 169.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 219.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 219.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Notes

  1. 1.

    A.k.a. regulatory myosin light chain MyL9.

  2. 2.

    The homeobox gene NKX2-5 (or NKX2.5) is also called cardiac-specific homeobox CSx. The homeobox gene set includes many members (NKX2-1–NKX2-6). Homeodomain protein NKx2-1 corresponds to thyroid transcription factor-1. It is also expressed in lungs. Factor NKx2-5 is also produced in the primitive pharyngeal endoderm that secretes the heart inducer.

  3. 3.

    Homeobox protein Isl1 as well as Pdx1 of endocrine cells of the gastric mucosa that control the production of gastrointestinal hormones were discovered in pancreatic islets.

  4. 4.

    Damaged heart components (valves, vessels, nodal conduction tissue, and myocardium) must be regenerated. The mobilization of endogenous progenitors and differentiation of exogenous stem cells participate in tissue repair. Adult stem cells are genetically equivalent to embryonic cells. Differential gene expression results from reversible epigenetic changes gradually imposed on the genome during development. However, adult cells have a reprogramming capacity toward an undifferentiated embryonic state, as differentiation is reversible.

  5. 5.

    During epithelial-to-mesenchymal transformation, epicardial and endocardial (endothelial) cells transform into mesenchymal cells that generate different tissues. Epithelial-to-mesenchymal transformation involves fibroblast growth factor, epidermal growth factor, vascular endothelial growth factor, transforming growth factor-β (Vol. 2 –Chap. 3. Growth Factors), and transcription factor WT1 encoded by the Wilms tumor-suppressor Wt1 gene.

  6. 6.

    Alias ZFPM stands for zinc finger protein multitype. A.k.a. zinc finger protein ZnF89b and Friend of GATA protein-2 (FOG2).

  7. 7.

    Neuregulin-1 is involved in the development of the nervous system and heart. It acts via receptors of the HER set. It has numerous isoforms produced by alternative splicing. Isoform NRg1 type-1 (heregulin), -2 (glial growth factor GGF2), and -3 (sensory and motor neuron-derived factor [SMDF]) are synthesized in excitatory and inhibitory neurons and astrocytes. Neuregulin-1 is a cardioactive growth factor released by endothelial cells that ensures cardiac development as well as structural maintenance and functional integrity. Effectors of the NRg1–HER axis include cardiac-specific myosin light chain kinase (cMLCK), protein phosphatase-1 (PP1), sarcoplasmic reticulum Ca2 +  ATPase SERCA2, and focal adhesion kinase (FAK).

  8. 8.

    Glial cell-derived neurotrophic factor and neurturin preferentially bind to GFRα1 and GFRα2, respectively.

  9. 9.

    A.k.a. GFRα2 and ReTL2.

  10. 10.

    A.k.a. GFRα1 and ReTL1. Coreceptor GDNFRα1 binds to GDNF tightly to form a membrane-associated complex that then interacts with ReT. On the other hand, GDNFRα2 forms a high-affinity complex with GDNF or Nrtn only in the presence of ReT [496].

  11. 11.

    Chondromodulin-1 is also called leukocyte-derived chemotaxin LeCT1.

  12. 12.

    The high-mobility group of non-histone chromatin proteins regulates the transcription of many genes by chromatin remodeling and proteic complex formation on promoter and enhancer regions. The HMG family of nuclear proteins includes 3 categories: HMGA, HMGB, and HMGN. Proteins of the HMGA category are ubiquitous and abound during embryogenesis. The HMGA category comprises 4 members: 3 splice variants (HMGA1a–HMGA1c) produced from the HMGA1 gene transcript and HMGA2 encoded by HMGA2 gene.

  13. 13.

    The E2F family includes 9 members encoded by 8 genes (E2F1–E2F2, E2F3a–E2F3b, and E2F4–E2F8). Three members are activators (E2F1–E2F3a), 6 are suppressors (E2F3b–E2F8). In most quiescent cells, E2F4 is primarily nuclear. It localizes to the cytoplasm during entry into the S phase. Nuclear E2F4 is mainly bound to Retinoblastoma-like protein-2 (or P130) in G0 phase and Retinoblastoma protein and Retinoblastoma-like protein-1 (or P107) during the G1–S transition.

  14. 14.

    Shortly after early heart formation, GATA4 is downregulated in the anterior part of the heart field.

  15. 15.

    A.k.a. stretch-responsive skeletal muscle protein and Chisel in mice.

  16. 16.

    Molecule LDOPA is a precursor to dopamine, noradrenaline, and adrenaline. Dopamine can be converted to noradrenaline by dopamine β-hydroxylase and adrenaline in the nervous system and adrenal glands.

  17. 17.

    Neuregulin-1 carries out diverse functions in different cell types. It operates in the differentiation of neurons, thereby acting as a neuronal differentiation factor, accumulation of acetylcholine receptors in skeletal muscle, and proliferation of glial cells, hence serving as a glial growth factor. Neuregulin-1 is encoded by a single gene (NRG1). It has numerous isoforms produced by alternative splicing. Isoform-1 (heregulin), -2 (glial growth factor GGF2), and -3 (sensory and motor neuron-derived factor [SMDF]) are processed as transmembrane proteins that contain EGF-like domains. Heregulin and glial growth factor GGF2 that possess a heparin-binding domain (HBD) are cleaved by adamlysins into soluble forms that act as paracrine factors. Sensory and motor neuron-derived factor is also cleaved by adamlysins; its EGF motif-containing extracellular domain (SMDFECD) remains attached to the transmembrane domain through the cysteine-rich domain (CRD) to signal as a juxtacrine messengers. Neuregulin-1 acts via receptor protein Tyr kinases HER2 to HER4.

  18. 18.

    Receptor HER4 also binds NRg2 to NRg4, HBEGF, and epiregulin. Receptor HER4 has multiple isoforms generated by alternative splicing [515]. Alternatively spliced variants can be categorized into juxtamembrane domain variant-A and -B (HER4JMa and HER4JMb) and cytoplasmic tail variant-1 and -2. Variant HER4Cyt1 is produced predominantly in the heart and breast. Variant HER4Cyt2), synthesized mainly in the nervous system and kidney, lacks a binding site for P85PI3K subunit, thus being unable to stimulate PI3K, but remaining functional, as it binds neuregulin-1.

  19. 19.

    Daughters of cardiomyocytes needs to have electromechanical connections to cooperate electrically and mechanically with the surrounding myocardium.

  20. 20.

    A.k.a. ankyrin repeat domain-containing protein-6 (AnkRD6).

  21. 21.

    In mice, Wnt11 is synthesized in or in close proximity to the precardiac mesoderm, and later in the myocardium of the primitive heart tube, thereby overlapping the first and secondary heart fields. At later stages, Wnt11 as well as Wnt5a are expressed in the outflow tract.

  22. 22.

    A.k.a. 60-kDa Brm-associated factors (BAF60a–BAF60c) encoded by 3 genes (Baf60A–Baf60C). Both SMARCd3a (BAF60C1) and SMARCd3b (BAF60C2) isoforms are widely expressed [520]. These isoforms bind to several nuclear receptors and transcription factors. A variant of the BAF complex in embryonic stem cells (esBAF) interacts with octamer-binding transcription factor Oct4 and sex-determining region Y (SRY)-box transcription factor Sox2 [521]. Specialized chromatin-remodeling complexes exist in neural progenitors (npBAF) and neurons (nBAF), in addition to cardiac BAF complexes (cBAF) [521].

  23. 23.

    A.k.a. ATP-dependent helicase brahma homolog Brg1.

  24. 24.

    A.k.a. transcriptional enhancer factor TEF1.

  25. 25.

    A.k.a. plakoglobin.

  26. 26.

    Regulator of muscular development MEF2, in cooperation with MyoD, activates transcription of miR1-2 and miR133a-1 in skeletal myocytes.

  27. 27.

    A.k.a. Wolf-Hirschhorn syndrome candidate WHSC2.

  28. 28.

    The βMHC level rises during cardiac diseases when αMHC is downregulated.

  29. 29.

    A.k.a. mediator complex subunit MED13, mediator of RNA polymerase-2 transcription subunit-13, 250-kDa component of activator-recruited cofactor ARC250, 250-kDa vitamin-D3 receptor-interacting protein complex component DRIP250, and 240-kDa thyroid hormone receptor-associated protein complex TRAP240.

  30. 30.

    Heart failure is a defect in proper blood filling and/or ejection. This progressive disorder is initiated by myocardial injury, most commonly caused by coronary artery disease, hypertension, and genetic factors.

  31. 31.

    A.k.a. RNA-binding domain (RBD) and RNP domain.

  32. 32.

    For example, induced pluripotent stem cell factor Lin28 is a repressor of microRNA processing and a post-transcriptional regulator of a mRNA set.

  33. 33.

    RNA editing is a type of RNA modification that results from the deamination of adenosine to inosine catalyzed by the ADAR proteins. Target transcripts localize mainly in the nervous system, where they generate ion channels and G-protein-coupled receptors, such as glutamate and serotonin receptors [536].

  34. 34.

    Polyadenylation of mRNAs using polynucleotide adenylate transferase (a.k.a. adenosine triphosphate (ATP):ribonucleic acid adenylate transferase, RNA adenylase, polyadenylate synthase, poly(A) polymerase, and poly(A) hydrolase) that has 2 substrates, ATP and RNA, and 2 products, diphosphate and RNA with an extra adenosine nucleotide at its 3end. Polyadenylation of mRNAs strongly influences their nuclear transport, translation efficiency, and stability. Cleavage and polyadenylation-specific factor (CPSF) is a ribonucleoproteic complex (with CPSF1–CPSF4, CPSF2L–CPSF4L, CPSF6–CPSF7, and nudix, or CPSF25 possible subunits) required in polyadenylation [536]. In cooperation with nuclear polyadenylate-binding protein (PABPn1), it activates polyadenylate polymerase.

  35. 35.

    RNA-binding protein hnRNPa1 is involved in pre-mRNA splicing and nuclear export; hnRNPa2b1 in splicing and mRNA transfer; hnRNPc in pre-mRNA packaging, splicing, stability, and nuclear retention; hnRNPd in mRNA stability; hnRNPf in splicing; hnRNPh in splicing and polyadenylation; hnRNPk in transcription, pre-mRNA splicing, translation, and regulation, and mRNA stability; hnRNPl in mRNA export and stability; and hnRNPu in nuclear retention [537].

  36. 36.

    Cleavage of ELAV1 generates 2 fragments: ELAV1 cleavage products ELAV1CP1 and ELAV1CP2. The former tethers to transportin-2, thereby allowing non-cleaved ELAV1 to lodge in the cytoplasm and promoting myogenesis.

  37. 37.

    RMB24 has 2 known variants (Rbm24a and Rbm24b).

  38. 38.

    Epicardial cells migrate from the proepicardium, an outgrowth of the septum transversum, and spread over the heart surface.

  39. 39.

    Among markers that identify cardiac stem cells (VEGFR2, also called KDR and Flk1, and transcription factors Islet-1 and NKx2-5), stem cell factor receptor (SCFR or KIT) is also expressed during various stages of cell lineage commitment in germ, mast, stellate, epithelial, endothelial, and smooth muscle cells [557].

  40. 40.

    Protein Wnt2 induces cardiac differentiation from mesoderm, whereas Wnt4 acts via the β-catenin-independent pathway that inhibits the β-catenin-dependent pathway for cardiac specification of mesoderm.

  41. 41.

    Factor WT1 is expressed in proepicardium and epicardium, but not in myocardium.

  42. 42.

    FOG2(\(-/-\)) hearts are characterized by a thin ventricular myocardium, common atrioventricular canal, tetralogy of Fallot, and absence of coronary vasculature. Transgenic re-expression of FOG2 in cardiomyocytes leads to coronary vessel development.

  43. 43.

    Angiotensin-2 favors the production of collagen-1 and -3.

  44. 44.

    The pars membranacea septi correspond to the upper part of the interventricular septum.

  45. 45.

    Second messenger cGMP inhibits TGFβ1-induced phosphorylation of SMAD3, hence precluding myofibroblast transformation and proliferation as well as synthesis of extracellular matrix proteins.

  46. 46.

    Periostin is secreted primarily by osteoblasts and fibroblasts. It is produced in the bone and, to a lesser extent, in the lung, kidney, and heart valves under normal conditions. However, periostin expression rises in heart failure.

  47. 47.

    Interstitial cells of Cajal are considered to be pacemakers of the motility of the gastrointestinal tract, where they have been discovered. Peculiar interstitial cells have also been detected in the urinary tract, blood vessels, pancreas, male and female reproductive tracts, mammary glands, placenta, and heart.

  48. 48.

    Adiponectin is also called ACRP30, AdipoQ, or gelatin-binding protein-28. It is involved in the regulation of lipid and glucose metabolism, as well as in differentiation of adipocytes. Its plasma concentration ranges from 3 to 30 μg/ml and accounts for 0.01% of total plasma protein. Adiponectin expression and subsequent release from adipocytes are stimulated by activated peroxisome proliferator-activated receptor PPARγ.

  49. 49.

    Circulating leptin concentration is related to body mass and adiposity. Leptin plasma level ranges from 5 to 15 ng/ml in non-obese individuals to reach more than 100 ng/ml in obese subjects. Leptin is convected by blood mainly bound to plasma proteins, whereas it flows primarily in free form in obesity.

  50. 50.

    Fatty acid oxidation accounts for about 70% of ATP generated under normoxic conditions, whereas carbohydrates become the major substrate in anaerobic conditions.

  51. 51.

    In addition to the valve cross curvature of the open leaflet, the Arantian nodule induces axial curvature due to stiffer medial thickening.

  52. 52.

    Collagen bundles run from commissure to commissure, spreading out near the cusp belly and combining again toward the opposite commissure.

  53. 53.

    Many types of connexins aggregate to form gap junctions between adjacent cells. They form homo- or heteromeric hemichannels and homotypic, heterotypic, or heteromeric channels with different conductance, permeability, and gating properties. Connexin type expression varies according to the heart site. Cells that express human connexin Cx31.9 exhibit much faster transport than cells expressing other connexin types [590].

  54. 54.

    Thyroid hormones regulate the metabolic rate, energy expenditure, and cardiac contractility.

  55. 55.

    MicroRNA-208A is encoded by an intron of the cardiac-specific α-myosin heavy-chain gene.

  56. 56.

    Transcription factors of the FOXO family that are effectors of the PI3Kc1α–PKB axis regulate the promoter activity of several K +  channel genes, such as KCNJ8 (KIR6.1), KCNJ11 (KIR6.2), and AbcC8 (SUR1), as well as KCND2 (KV4.2), KCNB1 (KV2.1), KCNK3 (TASK1), and KCNIP2 (KChIP2) [600]. Furthermore, glycogen synthase kinase GSK3β, another mediator of PI3Kc1α, also controls the activity of several transcription factors, such as NFAT, GATA4, myocardin, MyC, Jun, and β-catenin, many of which may bind to promoters of several K +  channel subunit genes.

  57. 57.

    During development, HCN4 location becomes restricted to the dorsal wall of the right atria, then to the junction of the right atrial appendage and the superior vena cava, where the sinoatrial node appears [605]. HCN4 channels are highly expressed in the adult sinoatrial node.

  58. 58.

    In adult mice, HCN4 channel does not contribute to cardiac pacemaking, but prevents sinus pauses during and after adrenergic stimulation stress. In humans, the role of HCN4 channels is different, as the cardiac frequency is higher in mice than in humans.

  59. 59.

    The activity of both adenosine- and acetylcholine-sensitive K +  channels involves GTP-binding proteins. Both ATP and Mg2 +  are cofactors of acetylcholine-sensitive K +  channels.

  60. 60.

    Sarcoplasmic reticulum calcium channel respond to increase in calcium ion concentration in the subspace caused by the local sarcolemmal influx.

  61. 61.

    Pacemaker cells lacking hyperpolarization-activated inward Na +  current have a lower frequency.

  62. 62.

    Channel protein conformations are influenced by the membrane potential, so that voltage-gated ion channels are only permeable over a narrow range of membrane potentials. On the other hand, ligand-gated ion channels require specific chemical activators.

  63. 63.

    The expression of sarcoplasmic reticulum Ca2 +  ATPase is greater in epicardial than midmyocardial and endocardial myocytes [625]. Consequently, epicardial myocytes exhibit a quicker decay of intracellular Ca2 +  content than endocardial myocytes. Transmural expression of Na + –Ca2 +  exchanger does not significantly vary [625]. Regional differences in ryanodine channel expression and/or its binding protein might exist.

  64. 64.

    Myosin isoforms do not have similar ATPase and contractile activities [626].

  65. 65.

    Loss in repolarization heterogeneity can lead to polymorphic ventricular tachycardia.

  66. 66.

    The so-called dyad is constituted by a T-tubule and a single terminal cisterna.

  67. 67.

    About 10% of the calcium needed for cardiac contraction enters from the extracellular space. This trigger calcium induces release of the remainder from the sarcoplasmic reticulum.

  68. 68.

    Two calsequestrin isoforms exist, one in the skeletal muscle, the second in the myocardium [458].

  69. 69.

    Phosphorylation of phospholamban by cAMP-dependent (PKA) or calmodulin-dependent (CamK2) protein kinases relieves the inhibition of SERCA by phospholamban.

  70. 70.

    A.k.a. exchange proteins directly activated by cAMP EPAC1 and EPAC2, respectively.

  71. 71.

    Multifunctional proteins of the S100 category are diversified according to the structure, ion-binding property (Ca2 + , Zn2 + , or Cu2 + ), spatial distribution in the intra- or extracellular space, and ability to homo- and heterodimerize [640]. Several S100 proteins (S100a4, S100a8–S100a9, S100a12–S100a13, and S100b) are secreted and have cytokine-like functions, as they interact with the receptor for advanced glycation end (RAGE) products of the immunoglobulin superfamily. Protein S100a4 has angiogenic effects [641].

  72. 72.

    Adenovirus-mediated S100A1 gene delivery normalizes S100a1 protein expression in a postinfarction rat heart failure model and reverses contractile dysfunction of failing myocardium. Moreover, S100A1 gene transfer decreases elevated [Na + ]i to levels detected in non-failing cardiomyocytes and restores energy supply in failing cardiomyocytes.

  73. 73.

    Dephosphorylated phospholamban binds to SERCA2 and suppresses SERCA2 activity. PKA-mediated phosphorylation of phospholamban unbinds it from SERCA2 to relieve SERCA2 inhibition.

  74. 74.

    CaV1.2 channels are responsible for the major calcium current in cardiomyocytes which triggers release of stored calcium from the sarcoplasmic reticulum for contraction.

  75. 75.

    β2-Adrenergic receptor-mediated phosphorylation depends on intracellular calcium concentration that must be increased. β2-Adrenergic receptors do not significantly participate in the calcium flux at basal calcium levels [645]. β2-Adrenergic receptors associated with adenylate cyclases, AKAP15, and calcium channels in the cardiomyocyte sarcolemma stimulate a small set of CaV1.2 calcium channels. Protein kinase-C and calcium–calmodulin-dependent protein kinase-2 can also phosphorylate voltage-gated calcium channels to increase the calcium influx.

  76. 76.

    These cardiac L-type voltage-gated Ca2 +  channels outside of junctional complexes do not strongly intervene in excitation–contraction coupling but regulate cellular functions.

  77. 77.

    Caveolae (Vol. 1 – Chap. 9. Intracellular Transport) are tiny invaginations of the plasma membrane with cholesterol, sphingolipids, and caveolin. Many proteins involved in cellular Ca2 +  activity are located in caveolae. They include IP3 receptor, Na + –Ca2 +  exchanger, Ca2 +  ATPase, members of the transient receptor potential family. Caveolin-3, CaV1.2 channel, β2-adrenergic receptor (but not β1-adrenoceptor), Gαs and Gαi proteins, adenylate cyclase, protein kinase-A, and protein phosphatase-2 form signaling complexes in caveolae of cardiomyocytes.

  78. 78.

    Synthesis of cAMP can greatly exceed requirements for PKA activation.

  79. 79.

    β1-Adrenergic receptors are downregulated in hypertrophic and ischemic cardiomyopathy.

  80. 80.

    Inactive PKA binds to cAMP, then dissociates into cAMP-bound dimer and active PKAs. PKC family members are activated via phospholipase-C. PKCα, PKCβ1/2, and PKCγ require a phospholipid, Ca2 + , and DAG for activation; PKCδ, PKCε, PKCη, and PKCθ needs DAG but not Ca2 + ; PKCζ and PKCλ do not have Ca2 + - and DAG-binding sites.

  81. 81.

    Kinases GRK2 and GRK5 are expressed in cardiomyocytes.

  82. 82.

    Among the 9 known isoforms (ACase1–ACase9), adenylate cyclase-5 is specifically expressed in cardiomyocytes; adenylate cyclase-6 is expressed in other cardiac cells. Other isoforms are expressed in the heart to a lesser extent. The fine-tuning in ACase regulation controls the adrenergic signal transmission rate. Elevated intracellular calcium levels during sustained activity inhibit adenylate cyclases ACase5 and ACase6.

  83. 83.

    Activity of PDE1 depends on the calcium–calmodulin complex, the quantity of which increases concomitantly with elevation in cytoplasmic calcium level. Subtype PDE2 is stimulated by cAMP messenger. Subtype PDE3 is inhibited by cAMP agent. Subtype PDE4 is insensitive to cAMP mediator.

References

Chap. 6. Heart Wall

  1. Forouhar AS, Liebling M, Hickerson A, Nasiraei-Moghaddam A, Tsai HJ, Hove JR, Fraser SE, Dickinson ME, Gharib M (2006) The embryonic vertebrate heart tube is a dynamic suction pump. Science 312:751–753

    ADS  Google Scholar 

  2. Brand T (2003) Heart development: molecular insights into cardiac specification and early morphogenesis. Developmental Biology 258:1–19

    Google Scholar 

  3. Red-Horse K, Ueno H, Weissman IL, Krasnow MA (2010) Coronary arteries form by developmental reprogramming of venous cells. Nature 464:549–553

    ADS  Google Scholar 

  4. Armstrong EJ, Bischoff J (2004) Heart valve development: endothelial cell signaling and differentiation. Circulation Research 95:459–470

    Google Scholar 

  5. Brand T (2010) Exciting news: catecholamines in induction and regionalization of the heart. Cardiovascular Research 88:1–2

    ADS  Google Scholar 

  6. Bu L, Jiang X, Martin-Puig S, Caron L, Zhu S, Shao Y, Roberts DJ, Huang PL, Domian IJ, Chien KR (2009) Human ISL1 heart progenitors generate diverse multipotent cardiovascular cell lineages. Nature 460:113–117

    ADS  Google Scholar 

  7. Gupta V, Poss KD (2012) Clonally dominant cardiomyocytes direct heart morphogenesis. Nature 484:479–484

    ADS  Google Scholar 

  8. Eisenberg LM, Markwald RR (1995) Molecular regulation of atrioventricular valvuloseptal morphogenesis. Circulation Research 77:1–6

    Google Scholar 

  9. Katsuragi N, Morishita R, Nakamura N, Ochiai T, Taniyama Y, Hasegawa Y, Kawashima K, Kaneda Y, Ogihara T, Sugimura K (2004) Periostin as a novel factor responsible for ventricular dilation. Circulation 110:1806–1813

    Google Scholar 

  10. Kim J, Wu Q, Zhang Y, Wiens KM, Huang Y, Rubin N, Shimada H, Handin RI, Chao MY, Tuan TL, Starnes VA, Lien CL (2010) PDGF signaling is required for epicardial function and blood vessel formation in regenerating zebrafish hearts. Proceedings of the National Academy of Sciences of the United States of America 107:17206–17210

    ADS  Google Scholar 

  11. Kamei M, Saunders WB, Bayless KJ, Dye L, Davis GE, Weinstein BM (2006) Endothelial tubes assemble from intracellular vacuoles in vivo. Nature 442:453–456

    ADS  Google Scholar 

  12. Bhattacharya S, Macdonald ST, Farthing CR (2006) Molecular mechanisms controlling the coupled development of myocardium and coronary vasculature. Clinical Science (London) 111:35–46

    Google Scholar 

  13. Saito D, Takase1 Y, Murai H, Takahashi Y (2012) The dorsal aorta initiates a molecular cascade that instructs sympatho-adrenal specification. Science 336:1578–1581

    Google Scholar 

  14. Mabe AM, Hoover DB (2009) Structural and functional cardiac cholinergic deficits in adult neurturin knockout mice. Cardiovascular Research 82:93–99

    Google Scholar 

  15. Sanicola M, Hession C, Worley D, Carmillo P, Ehrenfels C, Walus L, Robinson S, Jaworski G, Wei H, Tizard R, Whitty A, Pepinsky RB, Cate RL (1997) Glial cell line-derived neurotrophic factor-dependent RET activation can be mediated by two different cell-surface accessory proteins. Proceedings of the National Academy of Sciences of the United States of America 94:6238–6243

    ADS  Google Scholar 

  16. Johnson EN, Lee YM, Sander TL, Rabkin E, Schoen FJ, Kaushal S, Bischoff J (2003) NFATc1 mediates vascular endothelial growth factor-induced proliferation of human pulmonary valve endothelial cells. Journal of Biological Chemistry 278:1686–1692

    Google Scholar 

  17. Nakajima Y, Yamagishi T, Hokari S, Nakamura H (2000) Mechanisms involved in valvuloseptal endocardial cushion formation in early cardiogenesis: roles of transforming growth factor (TGF)-β and bone morphogenetic protein (BMP). Anatomical Record 258:119–127

    Google Scholar 

  18. Yoshioka M, Yuasa S, Matsumura K, Kimura K, Shiomi T, Kimura N, Shukunami C, Okada Y, Mukai M, Shin H, Yozu R, Sata M, Ogawa S, Hiraki Y, Fukuda K (2006) Chondromodulin-I maintains cardiac valvular function by preventing angiogenesis. Nature – Medicine 12:1151–1159

    Google Scholar 

  19. Murry CE, Lee RT (2009) Turnover after the fallout. Science 324:47–48

    ADS  Google Scholar 

  20. Olivetti G, Cigola E, Maestri R, Corradi D, Lagastra C, Gambert SR, Anversa P (1996) Aging, cardiac hypertrophy and ischemic cardiomyopathy do not affect the proportion of mononucleated and multinucleated myocytes in the human heart. Journal of Molecular and Cellular Cardiology 28:1463–1477

    Google Scholar 

  21. Bergmann O, Bhardwaj RD, Bernard S, Zdunek S, Barnab-Heider F, Walsh S, Zupicich J, Alkass K, Buchholz BA, Druid H, Jovinge S, Frisn J (2009) Evidence for cardiomyocyte renewal in humans. Science 324:98–102

    ADS  Google Scholar 

  22. Huang J, Min Lu M, Cheng L, Yuan LJ, Zhu X, Stout AL, Chen M, Li J, Parmacek MS (2009) Myocardin is required for cardiomyocyte survival and maintenance of heart function. Proceedings of the National Academy of Sciences of the United States of America 106:18734–18739

    ADS  Google Scholar 

  23. Taurin S, Sandbo N, Yau DM, Sethakorn N, Kach J, Dulin NO (2009) Phosphorylation of myocardin by extracellular signal-regulated kinase. Journal of Biological Chemistry 284:33789–33794

    Google Scholar 

  24. Muraski JA, Rota M, Misao Y, Fransioli J, Cottage C, Gude N, Esposito G, Delucchi F, Arcarese M, Alvarez R, Siddiqi S, Emmanuel GN, Wu W, Fischer K, Martindale JJ, Glembotski CC, Leri A, Kajstura J, Magnuson N, Berns A, Beretta RM, Houser SR, Schaefer EM, Anversa P, Sussman MA (2007) Pim-1 regulates cardiomyocyte survival downstream of Akt. Nature – Medicine 13:1467–1475

    Google Scholar 

  25. Lage K, Møllgd̈ K, Greenway S, Wakimoto H, Gorham JM, Workman CT, Bendsen E, Hansen NT, Rigina O, Roque FS, Wiese C, Christoffels VM, Roberts AE, Smoot LB, Pu WT, Donahoe PK, Tommerup N, Brunak S, Seidman CE, Seidman JG, Larsen LA (2010) Dissecting spatio-temporal protein networks driving human heart development and related disorders. Molecular Systems Biology 6:381

    Google Scholar 

  26. Ferdous A, Caprioli A, Iacovino M, Martin CM, Morris J, Richardson JA, Latif S, Hammer RE, Harvey RP, Olson EN, Kyba M, Garry DJ (2009) Nkx2-5 transactivates the Ets-related protein 71 gene and specifies an endothelial/endocardial fate in the developing embryo. Proceedings of the National Academy of Sciences of the United States of America 106:814–819

    ADS  Google Scholar 

  27. Monzen K, Ito Y, Naito AT, Kasai H, Hiroi Y, Hayashi D, Shiojima I, Yamazaki T, Miyazono K, Asashima M, Nagai R, Komuro I (2008) A crucial role of a high mobility group protein HMGA2 in cardiogenesis. Nature – Cell Biology 10:567–574

    Google Scholar 

  28. Bushdid PB, Osinska H, Waclaw RR, Molkentin JD, Yutzey KE (2003) NFATc3 and NFATc4 are required for cardiac development and mitochondrial function. Circulation Research 92:1305–1313

    Google Scholar 

  29. van Amerongen MJ, Diehl F, Novoyatleva T, Patra C, Engel FB (2010) E2F4 is required for cardiomyocyte proliferation. Cardiovascular Research 86:92–102

    Google Scholar 

  30. López-Sánchez C, Bártulos O, Martnez-Campos E, Gañán C, Valenciano AI, García-Martnez V, De Pablo F, Hernández-Sánchez C (2010) Tyrosine hydroxylase is expressed during early heart development and is required for cardiac chamber formation. Cardiovascular Research 88:111–120

    Google Scholar 

  31. Ilagan R, Abu-Issa R, Brown D, Yang YP, Jiao K, Schwartz RJ, Klingensmith J, Meyers EN (2006) Fgf8 is required for anterior heart field development. Development 133:2435–2445

    Google Scholar 

  32. Park EJ, Ogden LA, Talbot A, Evans S, Cai CL, Black BL, Frank DU, Moon AM (2006) Required, tissue-specific roles for Fgf8 in outflow tract formation and remodeling. Development 133:2419–2433

    Google Scholar 

  33. Wadugu B, Kühn B (2012) The role of neuregulin/ErbB2/ErbB4 signaling in the heart with special focus on effects on cardiomyocyte proliferation. American Journal of Physiology – Heart and Circulatory Physiology 302:H2139–H2147

    Google Scholar 

  34. Elenius K, Choi CJ, Paul S, Santiestevan E, Nishi E, Klagsbrun M (1999) Characterization of a naturally occurring ErbB4 isoform that does not bind or activate phosphatidyl inositol 3-kinase. Oncogene 18:2607–2615

    Google Scholar 

  35. Novoyatleva T, Diehl F, van Amerongen MJ, Patra C, Ferrazzi F, Bellazzi R, Engel FB (2010) TWEAK is a positive regulator of cardiomyocyte proliferation. Cardiovascular Research 85:681–690

    Google Scholar 

  36. Nagy II, Railo A, Rapila R, Hast T, Sormunen R, Tavi P, Räsänen J, Vainio SJ (2009) Wnt-11 signalling controls ventricular myocardium development by patterning N-cadherin and β-catenin expression. Circulation Research 85:100–109

    Google Scholar 

  37. Schwarz-Romond T, Asbrand C, Bakkers J, Kühl M, Schaeffer HJ, Huelsken J, Behrens J, Hammerschmidt M, Birchmeier W (2002) The ankyrin repeat protein Diversin recruits Casein kinase Iε to the beta-catenin degradation complex and acts in both canonical Wnt and Wnt/JNK signaling. Genes and Development 16:2073–2084

    Google Scholar 

  38. Moeller H, Jenny A, Schaeffer HJ, Schwarz-Romond T, Mlodzik M, Hammerschmidt M, Birchmeier W (2006) Diversin regulates heart formation and gastrulation movements in development. Proceedings of the National Academy of Sciences of the United States of America 103:15900–15905

    ADS  Google Scholar 

  39. Debril MB, Gelman L, Fayard E, Annicotte JS, Rocchi S, Auwerx J (2004) Transcription factors and nuclear receptors interact with the SWI/SNF complex through the BAF60c subunit. Journal of Biological Chemistry 279:16677–16686

    Google Scholar 

  40. Liang FS, Crabtree GR (2009) Developmental biology: the early heart remodelled. Nature 459:654–655

    ADS  Google Scholar 

  41. Takeuchi JK, Bruneau BG (2009) Directed transdifferentiation of mouse mesoderm to heart tissue by defined factors. Nature 459:708–711

    ADS  Google Scholar 

  42. Hang CT, Yang J, Han P, Cheng HL, Shang C, Ashley E, Zhou B, Chang CP (2010) Chromatin regulation by Brg1 underlies heart muscle development and disease. Nature 466:62–67

    ADS  Google Scholar 

  43. Stankunas K, Hang CT, Tsun ZY, Chen H, Lee NV, Wu JI, Shang C, Bayle JH, Shou W, Iruela-Arispe ML, Chang CP (2008) Endocardial Brg1 represses ADAMTS1 to maintain the microenvironment for myocardial morphogenesis. Developmental Cell 14:298–311

    Google Scholar 

  44. Sasse P, Malan D, Fleischmann M, Roell W, Gustafsson E, Bostani T, Fan Y, Kolbe T, Breitbach M, Addicks K, Welz A, Brem G, Hescheler J, Aszodi A, Costell M, Bloch W, Fleischmann BK (2008) Perlecan is critical for heart stability. Cardiovascular Research 80:435–444

    Google Scholar 

  45. Yang B, Lu Y, Wang Z (2008) Control of cardiac excitability by microRNAs. Cardiovascular Research 79:571–580

    Google Scholar 

  46. Yang B, Lin H, Xiao J, Lu Y, Luo X, Li B, Zhang Y, Xu C, Bai Y, Wang H, Chen G, Wang Z (2007) The muscle-specific microRNA miR-1 regulates cardiac arrhythmogenic potential by targeting GJA1 and KCNJ2. Nature – Medicine 13:486–491

    Google Scholar 

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

    Google Scholar 

  48. Care A, Catalucci D, Felicetti F, Bonci D, Addario A, Gallo P, Bang ML, Segnalini P, Gu Y, Dalton ND, Elia L, Latronico MV, Hoydal M, Autore C, Russo MA, Dorn GW, Ellingsen O, Ruiz-Lozano P, Peterson KL, Croce CM, Peschle C, Condorelli G (2007) MicroRNA-133 controls cardiac hypertrophy. Nature – Medicine 13:613–618

    Google Scholar 

  49. van Rooij E, Sutherland LB, Qi X, Richardson JA, Hill J, Olson EN (2007) Control of stress-dependent cardiac growth and gene expression by a MicroRNA. Science 316:575–579

    ADS  Google Scholar 

  50. Thum T, Catalucci D, Bauersachs J (2008) MicroRNAs: novel regulators in cardiac development and disease. Cardiovascular Research 79:562–570

    Google Scholar 

  51. Morton SU, Scherz PJ, Cordes KR, Ivey KN, Stainier DY, Srivastava D (2008) MicroRNA-138 modulates cardiac patterning during embryonic development. Proceedings of the National Academy of Sciences of the United States of America 105:17830–17835

    ADS  Google Scholar 

  52. Thum T, Gross C, Fiedler J, Fischer T, Kissler S, Bussen M, Galuppo P, Just S, Rottbauer W, Frantz S, Castoldi M, Soutschek J, Koteliansky V, Rosenwald A, Basson MA, Licht JD, Pena JT, Rouhanifard SH, Muckenthaler MU, Tuschl T, Martin GR, Bauersachs J, Engelhardt S (2008) MicroRNA-21 contributes to myocardial disease by stimulating MAP kinase signalling in fibroblasts. Nature 456:980–984

    ADS  Google Scholar 

  53. Roy S, Khanna S, Hussain SR, Biswas S, Azad A, Rink C, Gnyawali S, Shilo S, Nuovo GJ, Sen CK (2009) MicroRNA expression in response to murine myocardial infarction: miR-21 regulates fibroblast metalloprotease-2 via phosphatase and tensin homologue. Cardiovascular Research 82:21–29

    Google Scholar 

  54. Lunde BM, Moore C, Varani G (2007) RNA-binding proteins: modular design for efficient function. Nature Reviews – Molecular Cell Biology 8:479–490

    Google Scholar 

  55. Glisovic T, Bachorik JL, Yong J, Dreyfuss G (2008) RNA-binding proteins and post-transcriptional gene regulation. FEBS Letters 582:1977–1986

    Google Scholar 

  56. Dreyfuss G, Kim VN, Kataoka N (2002) Messenger-RNA-binding proteins and the messages they carry. Nature Reviews – Molecular Cell Biology 3:195–205

    Google Scholar 

  57. Lobbardi R, Lambert G, Zhao J, Geisler R, Kim HR, Rosa FM (2011) Fine-tuning of Hh signaling by the RNA-binding protein Quaking to control muscle development. Development 138:1783–1794

    Google Scholar 

  58. Beauchamp P, Nassif C, Hillock S, van der Giessen K, von Roretz C, Jasmin BJ, Gallouzi IE (2010) The cleavage of HuR interferes with its transportin-2-mediated nuclear import and promotes muscle fiber formation. Cell Death and Differentiation 17:1588–1599

    Google Scholar 

  59. Gerber WV, Yatskievych TA, Antin PB, Correia KM, Conlon RA, Krieg PA (1999) The RNA-binding protein gene, hermes, is expressed at high levels in the developing heart. Mechanisms of Development 80:77–86

    Google Scholar 

  60. Guo W, Schafer S, Greaser ML, Radke MH, Liss M, Govindarajan T, Maatz H, Schulz H, Li S, Parrish AM, Dauksaite V, Vakeel P, Klaassen S, Gerull B, Thierfelder L, Regitz-Zagrosek V, Hacker TA, Saupe KW, Dec GW, Ellinor PT, MacRae CA, Spallek B, Fischer R, Perrot A, Ozcelik C, Saar K, Hubner N, Gotthardt M (2012) RBM20, a gene for hereditary cardiomyopathy, regulates titin splicing. Nature – Medicine 18:766–773

    Google Scholar 

  61. Poon KL, Tan KT, Wei YY, Ng CP, Colman A, Korzh V, Xu XQ (2012) RNA-binding protein RBM24 is required for sarcomere assembly and heart contractility. Cardiovascular Research 94:418–427

    Google Scholar 

  62. Buckingham M, Meilhac S, Zaffran S (2005) Building the mammalian heart from two sources of myocardial cells. Nature Reviews – Genetics 6:826–837

    Google Scholar 

  63. Gaetani R, Ledda M, Barile L, Chimenti I, De Carlo F, Forte E, Ionta V, Giuliani L, D’Emilia E, Frati G, Miraldi F, Pozzi D, Messina E, Grimaldi S, Giacomello A, Lisi A (2009) Differentiation of human adult cardiac stem cells exposed to extremely low-frequency electromagnetic fields. Cardiovascular Research 82:411–420

    Google Scholar 

  64. Moretti A, Caron L, Nakano A, Lam JT, Bernshausen A, Chen Y, Qyang Y, Bu L, Sasaki M, Martin-Puig S, Sun Y, Evans SM, Laugwitz KL, Chien KR (2006) Multipotent embryonic isl1+ progenitor cells lead to cardiac, smooth muscle, and endothelial cell diversification. Cell 127:1151–1165

    Google Scholar 

  65. Kattman SJ, Huber TL, Keller GM (2006) Multipotent flk-1 + cardiovascular progenitor cells give rise to the cardiomyocyte, endothelial, and vascular smooth muscle lineages. Developmental Cell 11:723–732

    Google Scholar 

  66. Wu SM, Fujiwara Y, Cibulsky SM, Clapham DE, LienCL, Schultheiss TM, Orkin SH (2006) Developmental origin of a bipotential myocardial and smooth muscle cell precursor in the mmmalian heart. Cell 127:1137–1150

    Google Scholar 

  67. Kwon C, Qian L, Cheng P, Nigam V, Arnold J, Srivastava D (2009) A regulatory pathway involving Notch1/β-catenin/Isl1 determines cardiac progenitor cell fate. Nature – Cell Biology 11:951–957

    Google Scholar 

  68. Lian X, Hsiao C, Wilson G, Zhu K, Hazeltine LB, Azarin SM, Raval KK, Zhang J, Kamp TJ, Palecek SP (2012) Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling. Proceedings of the National Academy of Sciences of the United States of America 109:E1848–E1857

    ADS  Google Scholar 

  69. Tang JM, Wang JN, Zhang L, Zheng F, Yang JY, Kong X, Guo LY, Chen L, Huang YZ, Wan Y, Chen SY (2011) VEGF/SDF-1 promotes cardiac stem cell mobilization and myocardial repair in the infarcted heart. Cardiovascular Research 91:402–411

    Google Scholar 

  70. Bearzi C, Leri A, Lo Monaco F, Rota M, Gonzalez A, Hosoda T, Pepe M, Qanud K, Ojaimi C, Bardelli S, D’Amario D, D’Alessandro DA, Michler RE, Dimmeler S, Zeiher AM, Urbanek K, Hintze TH, Kajstura J, Anversa P (2009) Identification of a coronary vascular progenitor cell in the human heart. Proceedings of the National Academy of Sciences of the United States of America 106:15885–15890

    ADS  Google Scholar 

  71. Boni A, Urbanek K, Nascimbene A, Hosoda T, Zheng H, Delucchi F, Amano K, Gonzalez A, Vitale S, Ojaimi C, Rizzi R, Bolli R, Yutzey KE, Rota M, Kajstura J, Anversa P, Leri A (2008) Notch1 regulates the fate of cardiac progenitor cells. Proceedings of the National Academy of Sciences of the United States of America 105:15529–15534

    ADS  Google Scholar 

  72. Chen VC, Stull R, Joo D, Cheng X, Keller G (2008) Notch signaling respecifies the hemangioblast to a cardiac fate. Nature – Biotechnology 26:1169–1178

    Google Scholar 

  73. Cai CL, Martin JC, Sun Y, Cui L, Wang L, Ouyang K, Yang L, Bu L, Liang X, Zhang X, Stallcup WB, Denton CP, McCulloch A, Chen J, Evans SM (2008) A myocardial lineage derives from Tbx18 epicardial cells. Nature 454:104–108

    ADS  Google Scholar 

  74. Zhou B, Ma Q, Rajagopal S, Wu SM, Domian I, Rivera-Feliciano J, Jiang D, von Gise A, Ikeda S, Chien KR, Pu WT (2008) Epicardial progenitors contribute to the cardiomyocyte lineage in the developing heart. Nature 454:109–113

    ADS  Google Scholar 

  75. Beltrami AP, Barlucchi L, Torella D, Baker M, Limana F, Chimenti S, Kasahara H, Rota M, Musso E, Urbanek K, Leri A, Kajstura J, Nadal-Ginard B, Anversa P (2003) Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell 114:763–776

    Google Scholar 

  76. Tallini YN, Greene KS, Craven M, Spealman A, Breitbach M, Smith J, Fisher PJ, Steffey M, Hesse M, Doran RM, Woods A, Singh B, Yen A, Fleischmann BK, Kotlikoff MI (2009) c-Kit Expression identifies cardiovascular precursors in the neonatal heart. Proceedings of the National Academy of Sciences of the United States of America 106:1808–1813

    ADS  Google Scholar 

  77. Tevosian SG, Deconinck AE, Tanaka M, Schinke M, Litovsky SH, Izumo S, Fujiwara Y, Orkin SH (2000) FOG-2, a cofactor for GATA transcription factors, is essential for heart morphogenesis and development of coronary vessels from epicardium. Cell 101:729–739

    Google Scholar 

  78. Smart N, Risebro CA, Melville AAD, Moses K, Schwartz RJ, Chien KR, Riley PR (2007) Thymosin bold beta4 induces adult epicardial progenitor mobilization and neovascularization. Nature 445:177–182

    ADS  Google Scholar 

  79. Liu Y, Asakura M, Inoue H, Nakamura T, Sano M, Niu Z, Chen M, Schwartz RJ, Schneider MD (2007) Sox17 is essential for the specification of cardiac mesoderm in embryonic stem cells. Proceedings of the National Academy of Sciences of the United States of America 104:3859–3864

    ADS  Google Scholar 

  80. David R, Brenner C, Stieber J, Schwarz F, Brunner S, Vollmer M, Mentele E, Mller-Hcker J, Kitajima S, Lickert H, Rupp R, Franz WM (2008) MesP1 drives vertebrate cardiovascular differentiation through Dkk-1-mediated blockade of Wnt-signalling. Nature – Cell Biology 10:338–345

    Google Scholar 

  81. Kwon C, Arnold J, Hsiao EC, Taketo MM, Conklin BR, Srivastava C (2007) Canonical Wnt signaling is a positive regulator of mammalian cardiac progenitors. Proceedings of the National Academy of Sciences of the United States of America 104:10894–10899

    ADS  Google Scholar 

  82. Naito AT, Shiojima I, Akazawa H, Hidaka K, Morisaki T, Kikuchi A, Komuro I (2006) Developmental stage-specific biphasic roles of Wnt/beta-catenin signaling in cardiomyogenesis and hematopoiesis. Proceedings of the National Academy of Sciences of the United States of America 103:19812–19817

    ADS  Google Scholar 

  83. Zamora M, Mnner J, Ruiz-Lozano P (2007) Epicardium-derived progenitor cells require β-catenin for coronary artery formation. Proceedings of the National Academy of Sciences of the United States of America 104:18109–18114

    ADS  Google Scholar 

  84. Chang DF, Belaguli NS, Chang J, Schwartz RJ (2007) LIM-only protein, CRP2, switched on smooth muscle gene activity in adult cardiac myocytes. Proceedings of the National Academy of Sciences of the United States of America 104:157–162

    ADS  Google Scholar 

  85. Parker KK, Ingber DE (2007) Extracellular matrix, mechanotransduction and structural hierarchies in heart tissue engineering. Philosophical Transactions of the Royal Society – London – B Biological Sciences 362:1267–1279

    Google Scholar 

  86. Weber KT (2000) Fibrosis and hypertensive heart disease. Current Opinion in Cardiology 15:264–272

    Google Scholar 

  87. Ongstad EL, Gourdie RG (2012) Myocyte–fibroblast electrical coupling: the basis of a stable relationship? Cardiovascular Research 93:215–217

    Google Scholar 

  88. He K, Shi X, Zhang X, Dang S, Ma X, Liu F, Xu M, Lv Z, Han D, Fang X, Zhang Y (2011) Long-distance intercellular connectivity between cardiomyocytes and cardiofibroblasts mediated by membrane nanotubes. Cardiovascular Research 92:39–47

    Google Scholar 

  89. Nguyen TP, Xie Y, Garfinkel A, Qu Z, Weiss JN (2012) Arrhythmogenic consequences of myofibroblast-myocyte coupling. Cardiovascular Research 93:242–251

    Google Scholar 

  90. Glenn DJ, Rahmutula D, Nishimoto M, Liang F, Gardner DG (2009) Atrial natriuretic peptide suppresses endothelin gene expression and proliferation in cardiac fibroblasts through a GATA4-dependent mechanism. Cardiovascular Research 84:209–217

    Google Scholar 

  91. Jankowski M (2009) GATA4, a new regulator of cardiac fibroblasts, is sensitive to natriuretic peptides. Cardiovascular Research 84:176–177

    Google Scholar 

  92. Heineke J, Auger-Messier M, Xu J, Oka T, Sargent MA, York A, Klevitsky R, Vaikunth S, Duncan SA, Aronow BJ, Robbins J, Crombleholme TM, Molkentin JD (2007) Cardiomyocyte GATA4 functions as a stress-responsive regulator of angiogenesis in the murine heart. Journal of Clinical Investigation 117:3198–3210

    Google Scholar 

  93. Ieda M, Tsuchihashi T, Ivey KN, Ross RS, Hong TT, Shaw RM, Srivastava D (2009) Cardiac fibroblasts regulate myocardial proliferation through β1 integrin signaling. Developmental Cell 16:233–244

    Google Scholar 

  94. Li L, Fan D, Wang C, Wang JY, Cui XB, Wu D, Zhou Y, Wu LL (2011) Angiotensin II increases periostin expression via Ras/p38 MAPK/CREB and ERK1/2/TGF-β1 pathways in cardiac fibroblasts. Cardiovascular Research 91:80–89

    Google Scholar 

  95. LM Hinescu ME, Popescu LM (2005) Interstitial Cajal-like cells (ICLC) in human atrial myocardium. Journal of Cellular and Molecular Medicine 9:972–975

    Google Scholar 

  96. Gherghiceanu M, Hinescu ME, Andrei F, Mandache E, Macarie CE, Faussone-Pellegrini MS, Popescu LM (2008) Interstitial Cajal-like cells (ICLC) in myocardial sleeves of human pulmonary veins. Journal of Cellular and Molecular Medicine 12:1777–1781

    Google Scholar 

  97. Gherghiceanu M, Manole CG, Popescu LM (2010) Telocytes in endocardium: electron microscope evidence. Journal of Cellular and Molecular Medicine 14:2330–2334

    Google Scholar 

  98. Popescu LM, Gherghiceanu M (2010) Cardiomyocyte precursors and telocytes in epicardial stem cells niche. Journal of Cellular and Molecular Medicine (http://telocytes.com/Telocyte/Telocytes_in_the_heart_files/Cardiomyocyteprecursorsandtelocytesinepicardialstemcellsniche-Electronmicroscopeimages.pdf)

  99. Iacobellis G, Corradi D, Sharma AM (2005) Epicardial adipose tissue: anatomic, biomolecular and clinical relationships with the heart. Nature – Clinical Practice – Cardiovascular Medicine 2:536–543.

    Google Scholar 

  100. Kadowaki T, Yamauchi T (2005) Adiponectin and adiponectin receptors. Endocrine Reviews 26:439–451

    Google Scholar 

  101. Cui XB, Wang C, Li L, Fan D, Zhou Y, Wu D, Cui QH, Fu FY, Wu LL (2012) Insulin decreases myocardial adiponectin receptor 1 expression via PI3K/Akt and FoxO1 pathway. Cardiovascular Research 93:69–78

    Google Scholar 

  102. Shibata R, Sato K, Pimentel DR, Takemura Y, Kihara S, Ohashi K, Funahashi T, Ouchi N, Walsch K (2005) Adiponectin protects against myocardial ischemia-reperfusion injury through AMPK- and COX-2-dependent mechanisms. Nature – Medicine 11:1096–1103

    Google Scholar 

  103. Karmazyn M, Purdham DM, Rajapurohitam V, Zeidan A (2008) Signalling mechanisms underlying the metabolic and other effects of adipokines on the heart. Cardiovascular Research 79:279–286

    Google Scholar 

  104. Barrenechea GR, Le Tallec P, Valentin F (2002) New wall laws for the unsteady incompressible Navier–Stokes equations on rough domains. Mathematical Modelling and Numerical Analysis 36:177–203

    MATH  Google Scholar 

  105. Sacks MS, Smith DB, Hiester ED (1998) The aortic valve microstructure: effects of transvalvular pressure. Journal of Biomedical Materials Research 41:131–141.

    Google Scholar 

  106. Mendelson K, Schoen F (2006) Heart valve tissue engineering: concepts, approaches, progress, and challenges. Annals of Biomedical Engineering 34:1799–1819

    Google Scholar 

  107. Peskin CS, McQueen DM (1994) Mechanical equilibrium determines the fractal fiber architecture of aortic heart valve leaflets. American Journal of Physiology – Heart and Circulatory Physiology 266:319–328

    Google Scholar 

  108. Peskin CS (1989) Fiber architecture of the left ventricular wall: an asymptotic analysis. Communications on Pure and Applied Mathematics 42:79–113

    MathSciNet  MATH  Google Scholar 

  109. Bukauskas FF, Kreuzberg MM, Rackauskas M, Bukauskiene A, Bennett MLV, Verselis VK, Willecke K (2006) Properties of mouse connexin 30.2 and human connexin 31.9 hemichannels: Implications for atrioventricular conduction in the heart. Proceedings of the National Academy of Sciences of the United States of America 103:9726–9731

    ADS  Google Scholar 

  110. Gros D, Thveniau-Ruissy M, Bernard M, Calmels T, Kober F, Söhl G, Willecke K, Nargeot J, Jongsma HJ, Mangoni ME (2009) Connexin 30 is expressed in the mouse sino-atrial node and modulates heart rate. Cardiovascular Research 85:45–55

    Google Scholar 

  111. Fedorov VV, Glukhov AV, Chang R (2012) Conduction barriers and pathways of the sinoatrial pacemaker complex: their role in normal rhythm and atrial arrhythmias. American Journal of Physiology – Heart and Circulatory Physiology 302:H1773–H1783

    Google Scholar 

  112. Robinson RB (2011) The long and short of calcium-dependent automaticity in the sinoatrial node. American Journal of Physiology – Heart and Circulatory Physiology 300:H31–32

    Google Scholar 

  113. Choi BR, Salama G (1998) Optical mapping of atrioventricular node reveals a conduction barrier between atrial and nodal cells. American Journal of Physiology – Heart and Circulatory Physiology 275:H1905–H1909

    Google Scholar 

  114. Stuyvers BD, Dun W, Matkovich S, Sorrentino V, Boyden PA, ter Keurs HE (2005) Ca2 +  sparks and waves in canine Purkinje cells: a triple layered system of Ca2+ activation. Circulation Research 97:35–43

    Google Scholar 

  115. Nonidez JF (1943) The structure and innervation of the conductive system of the heart of the dog and rhesus monkey, as seen with a silver impregnation technique. American Heart Journal 26:577–597

    Google Scholar 

  116. Grueter CE, van Rooij E, Johnson BA, DeLeon SM, Sutherland LB, Qi X, Gautron L, Elmquist JK, Bassel-Duby R, Olson EN (2012) A cardiac microRNA governs systemic energy homeostasis by regulation of MED13. Cell 149:671–683

    Google Scholar 

  117. Zhang M, Wang Y, Jiang M, Zankov DP, Chowdhury S, Kasirajan V, Tseng GN (2011) KCNE2 protein is more abundant in ventricles than in atria and can accelerate hERG protein degradation in a phosphorylation-dependent manner. American Journal of Physiology – Heart and Circulatory Physiology 302:H910–H922

    Google Scholar 

  118. Rozanski GJ (2012) Physiological remodelling of potassium channels in the heart. Cardiovascular Research 93:218–219

    Google Scholar 

  119. Yang KC, Jay PY, McMullen JR, Nerbonne JM (2012) Enhanced cardiac PI3Kα signalling mitigates arrhythmogenic electrical remodelling in pathological hypertrophy and heart failure. Cardiovascular Research 93:252–262

    Google Scholar 

  120. Irisawa H, Brown HF, Giles W (1993) Cardiac pacemaker in the sinoatrial node, Physiological Reviews 73:197–227

    Google Scholar 

  121. Demir SS, Clark JW, Murphey CR, Giles WR (1994) A mathematical model of a rabbit sinoatrial node cell. American Journal of Physiology 266:C832–C852.

    Google Scholar 

  122. Courtemanche M, Ramirez RJ, Nattel S (1998) Ionic mechanisms underlying human atrial action potential properties: insights from a mathematical model. American Journal of Physiology – Heart and Circulatory Physiology 275:H301–H321.

    Google Scholar 

  123. Djabella K, Sorine M (2006) A differential model of controlled cardiac pacemaker cell, SIAM Conference on the Life Sciences, Raleigh, USA (http://hal.inria.fr/docs/00/06/46/22/PDF/Djabella_Sorine.pdf )

  124. Garcia-Frigola C, Shi Y, Evans SM (2003) Expression of the hyperpolarization-activated cyclic nucleotide-gated cation channel HCN4 during mouse heart development. Gene Expression Patterns 3:777–783

    Google Scholar 

  125. Harzheim D, Pfeiffer KH, Fabritz L, Kremmer E, Buch T, Waisman A, Kirchhof P, Kaupp UB, Seifert R (2008) Cardiac pacemaker function of HCN4 channels in mice is confined to embryonic development and requires cyclic AMP. EMBO Journal 27:692–703

    Google Scholar 

  126. Wu Y, Gao Z, Chen B, Koval OM, Singh MV, Guan X, Hund TJ, Kutschke W, Sarma S, Grumbach IM, Wehrens XH, Mohler PJ, Song LS, Anderson ME (2009) Calmodulin kinase II is required for fight or flight sinoatrial node physiology. Proceedings of the National Academy of Sciences of the United States of America 106:5972–5977

    ADS  Google Scholar 

  127. Tingley WG, Pawlikowska L, Zaroff JG, Kim T, Nguyen T, Young SG, Vranizan K, Kwok PY, Whooley MA, Conklin BR (2007) Gene-trapped mouse embryonic stem cell-derived cardiac myocytes and human genetics implicate AKAP10 in heart rhythm regulation. Proceedings of the National Academy of Sciences of the United States of America 104:8461-8466

    ADS  Google Scholar 

  128. Vinogradova TM, Maltsev VA, Bogdanov KY, Lyashkov AE, Lakatta EG (2005) Rhythmic Ca2 +  oscillations drive sinoatrial nodal cell pacemaker function to make the heart tick. Annals of the New York Academy of Sciences 1047:138–156

    ADS  Google Scholar 

  129. Maltsev VA, Lakatta EG (2010) A novel quantitative explanation for the autonomic modulation of cardiac pacemaker cell automaticity via a dynamic system of sarcolemmal and intracellular proteins. American Journal of Physiology – Heart and Circulatory Physiology 298:H2010–H2023

    Google Scholar 

  130. Lyashkov AE, Vinogradova TM, Zahanich I, Li Y, Younes A, Nuss HB, Spurgeon HA, Maltsev VA, Lakatta EG (2009) Cholinergic receptor signaling modulates spontaneous firing of sinoatrial nodal cells via integrated effects on PKA-dependent Ca2 +  cycling and KACh . American Journal of Physiology – Heart and Circulatory Physiology 297:H949–H959

    Google Scholar 

  131. ten Tusscher KHWJ, Noble D, Noble PJ, Panfilov AV (2004) A model for human ventricular tissue. American Journal of Physiology – Heart and Circulatory Physiology 286:H1573–H1589

    Google Scholar 

  132. Beeler GW, Reuter H (1977) Reconstruction of the action potential of ventricular myocardial fibres. Journal of Physiology 268:177-210

    Google Scholar 

  133. Luo CH, Rudy Y (1994) A dynamic model of the cardiac ventricular action potential. I. Simulations of ionic currents and concentration changes. Circulation Research 74:1071–1096

    Google Scholar 

  134. Luo CH, Rudy Y (1994) A dynamic model of the cardiac ventricular action potential. II. Afterdepolarizations, triggered activity, and potentiation. Circulation Research 74:1097–1113

    Google Scholar 

  135. Noble D, Varghese A, Kohl P, Noble P (1998) Improved guinea-pig ventricular cell model incorporating a diadic space, IKr and IKs, and length- and tension-dependent processes. Canadian Journal of Cardiology 14:123–134

    Google Scholar 

  136. Priebe L, Beuckelmann DJ (1998) Simulation study of cellular electric properties in heart failure. Circulation Research 82:1206–1223

    Google Scholar 

  137. Sipido KR (1998) Efficiency of L-type Ca2 +  current compared with reverse mode Na/Ca exchange or T-type Ca2 +  current as trigger for Ca2 +  release from the sarcoplasmic reticulum. Annals of the New York Academy of Sciences 853:357–360

    ADS  Google Scholar 

  138. Strom M, Wan X, Poelzing S, Ficker E, Rosenbaum DS (2010) Gap junction heterogeneity as mechanism for electrophysiologically distinct properties across the ventricular wall. American Journal of Physiology – Heart and Circulatory Physiology 298:H787–H794

    Google Scholar 

  139. Casini S, Verkerk AO, van Borren MMGJ, van Ginneken ACG, Veldkamp MW, de Bakker JMT, Tan HL (2009) Intracellular calcium modulation of voltage-gated sodium channels in ventricular myocytes. Cardiovascular Research 81:72–81

    Google Scholar 

  140. Wang LJ, Sobie EA (2008) Mathematical model of the neonatal mouse ventricular action potential. American Journal of Physiology – Heart and Circulatory Physiology 294:H2565–H2575

    Google Scholar 

  141. Malmivuo J, Plonsey R (2006) Bioelectromagnetism; Principles and Applications of Bioelectric and Biomagnetic Fields, University of Technology, Finland and Department of Biomedical Engineering, Duke University, Durham, NC, http://butler.cc.tut.fi/malmivuo/bem/bembook, Ragnar Granit Institute, Tampere

  142. Lipsius SL, Huser J, Blatter LA (2001) Intracellular Ca2 +  release sparks atrial pacemaker activity. News in Physiological Sciences 16:101–106

    Google Scholar 

  143. Liu DW, Antzelevitch C (1995) Characteristics of the delayed rectifier current (IKr and IKs) in canine ventricular epicardial, midmyocardial and endocardial myocytes: a weaker IKs contributes to the longer action potential of the M cell. Circulation Research 76:351–365

    Google Scholar 

  144. Laurita KR, Katra R, Wible B, Wan X, Koo MH (2003) Transmural heterogeneity of calcium handling in canine. Circulation Research 92:668–675

    Google Scholar 

  145. Joseph T, Coirault C, Lecarpentier Y (2000) Species-dependent changes in mechano-energetics of isolated cardiac muscle during hypoxia. Basic Research in Cardiology 95:378–384

    Google Scholar 

  146. Fedida D, Giles WR (1991) Regional variations in action potentials and transient outward current in myocytes isolated from rabbit left ventricle. Journal of Physiology (London) 442:191–209

    Google Scholar 

  147. Sah R, Ramirez RJ, Backx PH (2002) Modulation of Ca2 +  release in cardiac myocytes by changes in repolarization rate: role of phase-1 action potential repolarization in excitation-contraction coupling. Circulation Research 90:165–173

    Google Scholar 

  148. Drouin E, Charpentier F, Gauthier C, Chevallier JC, Laurent K, Michaud JL, Le Marec H (1993) Evidence for the presence of M cells in the human ventricle. Pacing and Clinical Electrophysiology (PACE) 16:876

    Google Scholar 

  149. Antzelevitch C (2001) Heterogeneity of cellular repolarization in LQTS: the role of M cells. European Heart Journal Supplements 3:K2–K16

    Google Scholar 

  150. Zygmunt AC, Goodrow RJ, Antzelevitch C (2000) INa-Ca contributes to electrical heterogeneity within the canine ventricle. American Journal of Physiology – Heart and Circulatory Physiology 278:H1671–H1678

    Google Scholar 

  151. Zygmunt AC, Eddlestone GT, Thomas GP, Nesterenko VV, Antzelevitch C (2001) Larger late sodium conductance in M cells contributes to electrical heterogeneity in canine ventricle. American Journal of Physiology – Heart and Circulatory Physiology 281:H689–H697

    Google Scholar 

  152. Cordeiro JM, Greene L, Heilmann C, Antzelevitch D, Antzelevitch C (2004) Transmural heterogeneity of calcium activity and mechanical function in the canine left ventricle. American Journal of Physiology – Heart and Circulatory Physiology 286:H1471–H1479

    Google Scholar 

  153. Janssen PM (2010) Myocardial contraction-relaxation coupling. American Journal of Physiology – Heart and Circulatory Physiology 299:H1741–H1749

    Google Scholar 

  154. McCain ML, Lee H, Aratyn-Schaus Y, Kléber AG, Parker KK (2012) Cooperative coupling of cell-matrix and cell-cell adhesions in cardiac muscle. Proceedings of the National Academy of Sciences of the United States of America 109:9881–9886

    ADS  Google Scholar 

  155. Bers DM (2000) Calcium fluxes involved in control of cardiac myocyte contraction. Circulation Research 87:275–281

    Google Scholar 

  156. Babu GJ, Bhupathy P, Timofeyev V, Petrashevskaya NN, Reiser PJ, Chiamvimonvat N, Periasamy M (2007) Ablation of sarcolipin enhances sarcoplasmic reticulum calcium transport and atrial contractility. Proceedings of the National Academy of Sciences of the United States of America 104:17867–17872

    ADS  Google Scholar 

  157. Tuncay E, Bilginoglu A, Sozmen NN, Zeydanli EN, Ugur M, Vassort G, Turan B (2011) Intracellular free zinc during cardiac excitation-contraction cycle: calcium and redox dependencies. Cardiovascular Research 89:634–642

    Google Scholar 

  158. Cazorla O, Lucas A, Poirier F, Lacampagne A, Lezoualc’h F (2009) The cAMP binding protein Epac regulates cardiac myofilament function. Proceedings of the National Academy of Sciences of the United States of America 106:14144–14149

    ADS  Google Scholar 

  159. Marenholz I, Heizmann CW, Fritz G (2004) S100 proteins in mouse and man: from evolution to function and pathology. Biochemical and Biophysical Research Communications 322:1111–1122

    Google Scholar 

  160. Ambartsumian N, Klingelhofer J, Grigorian M, Christensen C, Kriajevska M, Tulchinsky E, Georgiev G, Berezin V, Bock E, Rygaard J, Cao R, Cao Y, Lukanidin E (2001) The metastasis-associated Mts1(S100A4) protein could act as an angiogenic factor. Oncogene 20:4685–4695

    Google Scholar 

  161. Ehlermann P, Remppis A, Guddat O, Weimann J, Schnabel PA, Motsch J, Heizmann CW, Katus HA (2000) Right ventricular upregulation of the Ca2 +  binding protein S100A1 in chronic pulmonary hypertension. Biochimica et Biophysica Acta 1500:249–255

    Google Scholar 

  162. Remppis A, Greten T, Schafer BW, Hunziker P, Erne P, Katus HA, Heizmann CW (1996) Altered expression of the Ca2 + -binding protein S100A1 in human cardiomyopathy. Biochimica et Biophysica Acta 1313:253–257

    Google Scholar 

  163. Most P, Pleger ST, Volkers M, Heidt B, Boerries M, Weichenhan D, Loffler E, Janssen PM, Eckhart AD, Martini J, Williams ML, Katus HA, Remppis A, Koch WJ (2004) Cardiac adenoviral S100A1 gene delivery rescues failing myocardium. Journal of Clinical Investigation 114:1550–1563

    Google Scholar 

  164. Hulme JT, Westenbroek RE, Scheuer T, Catterall WA (2006) Phosphorylation of serine 1928 in the distal C-terminal domain of cardiac CaV1.2 channels during beta1-adrenergic regulation. Proceedings of the National Academy of Sciences of the United States of America 103:16574–16579

    ADS  Google Scholar 

  165. Hulme JT, Yarov-Yarovoy V, Lin TWC, Scheuer T, Catterall WA (2006) Autoinhibitory control of the CaV1.2 channel by its proteolytically processed distal C-terminal domain. Journal of Physiology 576:87–102

    Google Scholar 

  166. Lygren B, Carlson CR, Santamaria K, Lissandron V, McSorley T, Litzenberg J, Lorenz D, Wiesner B, Rosenthal W, Zaccolo M, Taskn K, Klussmann E (2007) AKAP complex regulates Ca2 +  re-uptake into heart sarcoplasmic reticulum. EMBO reports 8:1061–1067

    Google Scholar 

  167. Aprigliano O, Rybin VO, Pak E, Robinson RB, Steinberg SF (1997) β1- and β2-adrenergic receptors exhibit differing susceptibility to muscarinic accentuated antagonism. American Journal of Physiology – Heart and Circulatory Physiology 272:2726–2735

    Google Scholar 

  168. Saucerman JJ, Zhang J, Martin JC, Peng LX, Stenbit AE, Tsien RY, McCulloch AD (2006) Systems analysis of PKA-mediated phosphorylation gradients in live cardiac myocytes. Proceedings of the National Academy of Sciences of the United States of America 103:12923–12928

    ADS  Google Scholar 

  169. Zaugg M, Schaub MC (2004) Cellular mechanisms in sympatho-modulation of the heart. British Journal of Anaesthesia 93:34–52

    Google Scholar 

  170. Potter EK, Smith-White MA (2004) Galanin modulates cholinergic neurotransmission in the heart. Neuropeptides 39:345–348

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Rights and permissions

Reprints and permissions

Copyright information

© 2013 Springer Science+Business Media New York

About this chapter

Cite this chapter

Thiriet, M. (2013). Heart Wall. In: Tissue Functioning and Remodeling in the Circulatory and Ventilatory Systems. Biomathematical and Biomechanical Modeling of the Circulatory and Ventilatory Systems, vol 5. Springer, New York, NY. https://doi.org/10.1007/978-1-4614-5966-8_6

Download citation

  • DOI: https://doi.org/10.1007/978-1-4614-5966-8_6

  • Published:

  • Publisher Name: Springer, New York, NY

  • Print ISBN: 978-1-4614-5965-1

  • Online ISBN: 978-1-4614-5966-8

  • eBook Packages: Physics and AstronomyPhysics and Astronomy (R0)

Publish with us

Policies and ethics