Review article
S100A1 in cardiovascular health and disease: Closing the gap between basic science and clinical therapy

https://doi.org/10.1016/j.yjmcc.2009.06.003Get rights and content

Abstract

Calcium (Ca2+) signaling plays a major role in a wide range of physiological functions including control and regulation of cardiac and skeletal muscle performance and vascular tone [1], [2]. As all Ca2+ signals require proteins to relay intracellular Ca2+ oscillations downstream to different signaling networks, a specific toolkit of Ca2+-sensor proteins involving members of the EF-hand S100 Ca2+ binding protein superfamily maintains the integrity of the Ca2+ signaling in a variety of cardiac and vascular cells, transmitting the message with great precision and in a temporally and spatially coordinated manner [3], [4], [5], [6]. Indeed, the possibility that S100 proteins might contribute to heart and vascular diseases was first suggested by the discovery of distinctive patterns of S100 expression in healthy and diseased hearts and vasculature from humans and animal heart failure (HF) models [7], [8], [9], [10], [11], [12], [13], [14], [15], [16], [17], [18]. Based on more elaborate genetic studies in mice and strategies to manipulate S100 protein expression in human cardiac, skeletal muscle and vascular cells, it is now apparent that the integrity of distinct S100 protein isoforms in striated muscle and vascular cells such as S100A1, S100A4, S100A6, S100A8/A9 or S100B is a basic requirement for normal cardiovascular and muscular development and function; loss of integrity would naturally lead to profound deregulation of the implicated Ca2+ signaling systems with detrimental consequences to cardiac, skeletal muscle, and vascular function [7], [8], [9], [10], [11], [12], [13], [14], [15], [16], [17], [18], [19], [20]. The brief debate and discussion here are confined by design to the biological actions and pathophysiological relevance of the EF-hand Ca2+-sensor protein S100A1 in the heart, vasculature and skeletal muscle with a particular focus on current translational therapeutic strategies [4], [21], [23]. By virtue of its ability to modulate the activity of numerous key effector proteins that are essentially involved in the control of Ca2+ and NO homeostasis in cardiac, skeletal muscle and vascular cells, S100A1 has been proven to play a critical role both in cardiac performance, blood pressure regulation and skeletal muscle function [4,21,23]. Given that deregulated S100A1 expression in cardiomyocytes and endothelial cells has recently been linked to heart failure and hypertension [4,21,23], it is arguably a molecular target of considerable clinical interest as S100A1 targeted therapies have already been successfully investigated in preclinical translational studies.

Section snippets

S100A1 genomic organization

S100A1 is a member of the multigenic S100 protein family constituting by far the largest subgroup within the EF-hand Ca2+ binding protein superfamily [6], [24]. These proteins were first discovered in 1965 by B.W. Moore and characterized as “S100”, accounting for their solubility in a 100%-saturated solution with ammonium sulphate [24]. Since then S100 proteins have emerged as regulators of fundamental cellular and molecular functions, including cell proliferation, differentiation, survival and

S100A1 expression in cardiomyocytes

S100A1 is preferentially expressed in human cardiac muscle but also found in skeletal muscle, brain and kidney (most recent expression profiles are accessible through http://www.genecards.org/cgi-bin/carddisp.pl?gene=S100A1&search=S100A1), albeit at much lower concentrations. Comparative analyses revealed a similar S100A1 expression pattern in rodents and larger mammals [34], [35], [36], [37], [38]. Heizmann's group showed that S100A1 mRNA and protein levels steadily increase in the developing

S100A1 in cardiac disease

Chronically dysfunctional human myocardium is characterized by progressively diminished S100A1 mRNA and protein levels that inversely correlate with the severity of the disease [67]. Given the same pathological plasticity in chronically diseased rodent, rabbit and pig hearts [13], [49], [58], [59], [60], [68], decreased S100A1 expression seems to be a common pathognomonic molecular signature of failing myocardium. Abnormal in vivo cardiac S100A1 expression has been recapitulated in vitro by

Conclusions and future perspective

In recent years, S100A1 has clearly emerged as a critical regulator of Ca2+ handling and NO homeostasis in cardiac and vascular cells reflected by numerous reports highlighting its physiological and pathophysiological relevance in the cardiovascular system (reviewed in [4], [21]). However, S100A1 appears to have a likewise significant role in skeletal muscle function although its pathophysiological and therapeutic relevance in congenital and HF-associated skeletal muscle disorders have not been

Acknowledgements

This article was supported by grants from the NIH (R01 HL92130 to P. Most), DFG (562/1-1 to P. Most and S.T. Pleger) and BMBF (01GU0527 to P. Most).

References (77)

  • D. Osterloh et al.

    Hydrophobic residues in the C-terminal region of S100A1 are essential for target protein binding but not for dimerization

    Cell Calcium

    (1998)
  • L. Zhukova et al.

    Redox modifications of the C-terminal cysteine residue cause structural changes in S100A1 and S100B proteins

    Biochem. Biophys. Acta

    (2004)
  • R. Kiewitz et al.

    Transcriptional regulation of S100A1 and expression during mouse heart development

    Biochim. Biophys. Acta

    (2000)
  • D.B. Zimmer et al.

    Isolation of a rat S100 alpha cDNA and distribution of its mRNA in rat tissues

    Brain Res. Bull.

    (1991)
  • K. Kato et al.

    S100ao (alpha alpha) protein is mainly located in the heart and striated muscles

    Biochim. Biophys. Acta

    (1985)
  • S. Kettlewell et al.

    S100A1 increases the gain of excitation–contraction coupling in isolated rabbit ventricular cardiomyocytes

    J. Mol. Cell. Cardiol.

    (2005)
  • R. Kiewitz et al.

    Ca(2+)-dependent interaction of S100A1 with the sarcoplasmic reticulum Ca(2+)-ATPase2a and phospholamban in the human heart

    Biochem. Biophys. Res. Commun.

    (2003)
  • P. Most et al.

    Transgenic overexpression of the Ca2+ binding protein S100A1 in the heart leads to increased in vivo myocardial contractile performance

    J. Biol. Chem.

    (2003)
  • M. Volkers et al.

    S100A1 decreases calcium spark frequency and alters their spatial characteristics in permeabilized adult ventricular cardiomyocytes

    Cell Calcium

    (2007)
  • B.L. Prosser et al.

    S100A1 binds to the calmodulin-binding site of ryanodine receptor and modulates skeletal muscle excitation–contraction coupling

    J. Biol. Chem.

    (2008)
  • N.T. Wright et al.

    S100A1 and calmodulin compete for the same binding site on ryanodine receptor

    J. Biol. Chem.

    (2008)
  • R.S. Balaban

    Cardiac energy metabolism homeostasis: role of cytosolic calcium

    J. Mol. Cell. Cardiol.

    (2002)
  • R. Yamasaki et al.

    Titin–actin interaction in mouse myocardium: passive tension modulation and its regulation by calcium/s100a1

    Biophys. J.

    (2001)
  • S.T. Pleger et al.

    S100A1 gene therapy preserves in vivo cardiac function after myocardial infarction

    Mol. Therapy

    (2005)
  • A.A. Polyakov et al.

    Interaction of isoforms of S100 protein with smooth muscle caldesmon

    FEBS Let.

    (1998)
  • P. Most et al.

    The C-terminus (aa 75–94) and the linker region (aa 42–54) of the Ca2+ binding protein S100A1 differentially enhance sarcoplasmic Ca2+ release in murine skinned skeletal muscle fibres

    J. Biol. Chem.

    (2003)
  • H. Zhang et al.

    A noncontiguous, intersubunit binding site for calmodulin on the skeletal muscle Ca2+ release channel

    J. Biol. Chem.

    (2003)
  • A. Remppis et al.

    Altered expression of the Ca(2+)-binding protein S100A1 in human cardiomyopathy

    Biochim. Biophys. Acta

    (1996)
  • P. Ehlermann et al.

    Right ventricular upregulation of the Ca(2+) binding protein S100A1 in chronic pulmonary hypertension

    Biochim. Biophys. Acta

    (2000)
  • P. Most et al.

    Extracellular S100A1 protein inhibits apoptosis in ventricular cardiomyocytes via activation of the extracellular-regulated kinase (ERK1/2) pathway

    J. Biol. Chem.

    (2003)
  • R. Donato

    S100: a multigenic family of calcium-modulated proteins of the EF-hand type with intracellular and extracellular functional roles

    Int. J. Biochem. Cell. Biol.

    (2001)
  • Q.K. Tran et al.

    Calcium signalling in endothelial cells

    Cardiovasc. Res.

    (2000 Oct)
  • D.M. Bers

    Calcium cycling and signaling in cardiac myocytes

    Ann. Rev. Physiol.

    (2008)
  • R. Donato

    Intracellular and extracellular roles of S100 proteins

    Microsc. Res. Tech.

    (2003)
  • P. Most et al.

    S100A1: a novel inotropic regulator of cardiac performance. Transition from molecular physiology to pathophysiological relevance

    Am. J. Physiol.

    (2007)
  • L. Santamaria-Kisiel et al.

    Calcium-dependent and -independent interactions of the S100 protein family

    Biochem. J.

    (2006)
  • J.H. Boyd et al.

    S100A8 and S100A9 mediate endotoxin-induced cardiomyocyte dysfunction via the receptor for advanced glycation end products

    Circ. Res.

    (2008)
  • P. Ehlermann et al.

    Increased proinflammatory endothelial response to S100A8/A9 after preactivation through advanced glycation end products

    Cardiovasc. Diabet.

    (2006)
  • Cited by (61)

    • Intracavernosal Adeno-Associated Virus-Mediated S100A1 Gene Transfer Enhances Erectile Function in Diabetic Rats by Promoting Cavernous Angiogenesis via VEGF-A/VEGFR2 Signaling

      2019, Journal of Sexual Medicine
      Citation Excerpt :

      Furthermore, our data suggested that administration of exogenous S100A1 could activate pro-angiogenic factors, augment endothelial content, and eventually partially ameliorate erectile function in rats with streptozotocin-induced diabetes.34 Although the role of S100A1 with respect to cardiovascular pathophysiology has been described,10–12 its role in ED is unknown. Our previous work noted that APO-positive rats had only mild to moderate ED, whereas APO-negative rats’ sexual function was severely impaired.19

    View all citing articles on Scopus
    View full text