Original article
Taurine depletion caused by knocking out the taurine transporter gene leads to cardiomyopathy with cardiac atrophy

https://doi.org/10.1016/j.yjmcc.2008.03.001Get rights and content

Abstract

The sulfur-containing β-amino acid, taurine, is the most abundant free amino acid in cardiac and skeletal muscle. Although its physiological function has not been established, it is thought to play an important role in ion movement, calcium handling, osmoregulation and cytoprotection. To begin examining the physiological function of taurine, we generated taurine transporter− (TauT−) knockout mice (TauTKO), which exhibited a deficiency in myocardial and skeletal muscle taurine content compared with their wild-type littermates. The TauTKO heart underwent ventricular remodeling, characterized by reductions in ventricular wall thickness and cardiac atrophy accompanied with the smaller cardiomyocytes. Associated with the structural changes in the heart was a reduction in cardiac output and increased expression of heart cardiac failure (fetal) marker genes, such as ANP, BNP and β-MHC. Moreover, ultrastructural damage to the myofilaments and mitochondria was observed. Further, the skeletal muscle of the TauTKO mice also exhibited decreased cell volume, structural defects and a reduction of exercise endurance capacity. Importantly, the expression of Hsp70, ATA2 and S100A4, which are upregulated by osmotic stress, was elevated in both heart and skeletal muscle of the TauTKO mice. Taurine depletion causes cardiomyocyte atrophy, mitochondrial and myofiber damage and cardiac dysfunction, effects likely related to the actions of taurine. Our data suggest that multiple actions of taurine, including osmoregulation, regulation of mitochondrial protein expression and inhibition of apoptosis, collectively ensure proper maintenance of cardiac and skeletal muscular structure and function.

Introduction

The sulfur-containing amino acid, taurine (2-ethanesulfonic acid), is the most abundant free amino acid in mammalian tissue, reaching concentrations as high as 5–20 μmol/g wet wt [1], [2]. Since the capacity to synthesize taurine in most tissues including the heart and skeletal muscle is limited, maintenance of the large intracellular taurine pool depends upon uptake of the amino acid from the blood. This transport process requires the accumulation of taurine against a substantial concentration gradient, as the concentration of taurine is 100 fold less in the plasma (20–100 μM) than in the tissues (5–20 μmol/g wet wt.) [1], [2]. Taurine uptake is mediated by an ubiquitous Na+ and Cl dependent transporter, which uses the Na+ gradient across the cell membrane to drive taurine accumulation [3], [4], [5]. The expression of the taurine transporter (TauT; SLC6a6) is regulated by osmotic stress and transcription factors, such as NFAT5, MEF-2 and p53 [4], [6], [7], which control intracellular taurine content.

Accumulating evidences appear that taurine plays cytoprotective roles in the hearts. Oral supplementation of taurine is effective to animal model and human patients with congestive heart failure and cardiomyopathy [8], [9], [10], [11]. Although the essential role of taurine in heart has not been clarified, taurine exerts several actions that could potentially benefit the diseased heart. First, it modulates ion transport and regulates intracellular calcium levels [12], [13], [14]. Maintenance of Ca2+ homeostasis is of paramount importance in the heart because either reductions in [Ca2+]i or impaired Ca2+ sensitivity of the myofibrils can lead to the development of heart failure. Second, it possesses antioxidant [11], [12], [13] and anti-apoptotic activity [15], [16], which would be expected to limit ventricular remodeling. Finally, taurine is a key osmoregulator in the heart [13], an action that should limit damaging osmotic imbalances that develop in conditions, such as ischemia. Interestingly, taurine content is altered in various pathological states [10], [17]. Based on these findings, it has been suggested that severe reductions in the size of the intracellular taurine pool, as occurs in ischemia, may contribute to cell shrinkage and the development of pathological lesions. On the other hand, increases in taurine levels in conditions, such as failing and hypertrophic heart. Taken together, it is hypothesized that taurine would play a critical role in compensatory adaptation against the pathophysiological loads associated with the development of myocardial hypertrophy and/or heart failure.

In a small number of species (fox and cat), taurine levels can be dramatically diminished merely by reductions in dietary taurine content [18], [19]. After prolonged exposure to the taurine depletion condition, the nutritionally deprived animals develop cardiomyopathy as well as retinopathy or immune deficiency [18], [20], [21]. In contrast to fox and cat, the size of the intracellular taurine pool of most animal species remains fairly constant even with significant reductions in dietary taurine content. This occurs because a decline in plasma taurine levels is accompanied by enhanced cellular retention of taurine [22]. Despite resistance to depletion, tissue taurine levels can be decreased by treatment of these animals with a taurine transport inhibitor, such β-alanine or guanidinoethane sulfonate, which interferes with taurine uptake by the tissues [10], [23], [24]. However, treatment of these animals with a taurine transport inhibitor does not generally cause sufficient taurine deficiency to promote the development of severe, overt pathology. This has been attributed to the limited capacity of the taurine transport inhibitors to cause severe taurine deficiency. However, resistant species, such as rodents, also exhibit a greater capacity to synthesize taurine than cats or fox. Therefore, a more effective means of producing taurine deficiency in the rodent is the formation of TauT null animals. The present study shows that depletion of taurine in the TauT null mouse is associated with the development of a cardiomyopathy.

Section snippets

Generation of TauT-null Mice

All animal experiments were performed in accordance with protocols approved by the Animal Care and Use Committee of Graduate School of Pharmaceutical Sciences, Osaka University. A clone of the murine TauT gene including exon 2 to exon 5 was isolated from the 129SVJ murine genomic library in bacterial artificial clone, as identified by PCR with specific primers for the TauT gene (Table 1). The targeting vector was designed to replace the Stu I–Xba I fragment including the end of exon 2 to exon 4

Generation of TauT-null mice

A targeting construct was generated to replace exons 2–4 of the TauT gene with a cassette containing a neomycin-resistance gene (see Methods and Fig. 1A). Germline transmission of the mutant allele was confirmed by Southern blotting (Fig. 1B) and PCR. Disruption of the TauT gene in heart resulted in an truncated transcript that included exon 5 but not exon 2–4; in other tissues reverse transcript-PCR was used to evaluate knockout of the TauT (Fig. 1C). As expected, cellular taurine uptake

Discussion

The present study demonstrates that the downregulation of the taut gene leads to cardiac dysfunction, ventricular remodeling, upregulation of cardiac failure marker genes, loss of body weight and a decrease in exercise capacity. At the cellular level, it was shown that myocytes from the heart and skeletal muscle of TauTKO mice lost cell volume, develop mitochondrial defects and undergo myofibrillar disruption. These findings are consistent with the actions of taurine as an osmoregulator,

Funding sources

This study was supported in part by a Grants-in-Aid from the Ministry of Health, Labour and Welfare and from the Ministry of Education, Science, Sports and Culture of Japan. This study was also partly granted by Taisho Pharmaceutical Ltd. and The Nakatomi Foundation.

Acknowledgments

We thank Ms. Yasuko Murao for her excellent secretary work, Mr. Eizi Oiki (Osaka University) for his technical support for electron microscopic analyses, and Dr. Nishiya and Dr. Nakata (Osaka City University) for their technical support for echocardiographic analyses.

References (53)

  • M. Tanaka et al.

    Establishment and assessment of a rat model of fatigue

    Neurosci Lett

    (2003)
  • C.J. Rivard et al.

    Expression of the calcium-binding protein S100A4 is markedly up-regulated by osmotic stress and is involved in the renal osmoadaptive response

    J Biol Chem

    (2007)
  • L.K. Russell et al.

    Mouse models of mitochondrial dysfunction and heart failure

    J Mol Cell Cardiol

    (2005)
  • R.A. Chapman et al.

    Taurine and the heart

    Cardiovasc Res

    (1993)
  • S. Uchida et al.

    Taurine behaves as an osmolyte in Madin–Darby canine kidney cells. Protection by polarized, regulated transport of taurine

    J Clin Invest

    (1991)
  • Y. Uozumi et al.

    Myogenic differentiation induces taurine transporter in association with taurine-mediated cytoprotection in skeletal muscles

    Biochem J

    (2006)
  • T. Ito et al.

    Expression of taurine transporter is regulated through the TonE (tonicity-responsive element)/TonEBP (TonE-binding protein) pathway and contributes to cytoprotection in HepG2 cells

    Biochem J

    (2004)
  • J. Azuma et al.

    Therapy of congestive heart failure with orally administered taurine

    Clin Ther

    (1983)
  • J. Azuma et al.

    Therapeutic effect of taurine in congestive heart failure: a double-blind crossover trial

    Clin Cardiol

    (1985)
  • G.Y. Oudit et al.

    Taurine supplementation reduces oxidative stress and improves cardiovascular function in an iron-overload murine model

    Circulation

    (2004)
  • R.J. Huxtable

    Physiological actions of taurine

    Physiol Rev

    (1992)
  • S. Schaffer et al.

    Role of osmoregulation in the actions of taurine

    Amino Acids

    (2000)
  • T. Takatani et al.

    Taurine inhibits apoptosis by preventing formation of the Apaf-1/caspase-9 apoptosome

    Am J Physiol Cell Physiol

    (2004)
  • R. Huxtable et al.

    Taurine concentrations in congestive heart failure

    Science

    (1974)
  • P.D. Pion et al.

    Myocardial failure in cats associated with low plasma taurine: a reversible cardiomyopathy

    Science

    (1987)
  • G.Y. Wen et al.

    Tapetum disorganization in taurine-depleted cats

    Invest Ophthalmol Vis Sci

    (1979)
  • Cited by (168)

    • Taurine and deferiprone against Al-linked apoptosis in rat hippocampus

      2023, Journal of Trace Elements in Medicine and Biology
      Citation Excerpt :

      Under the influence of factors such as BCL2, BAX, caspases, and β-amyloid [6], Al accumulation may result in harmful neurodegenerative diseases. Taurine, one of the most abundant free amino acids in mammals [7], plays a fundamental role in promoting normal growth and development by modulating cellular calcium balance, maintaining nerve conduction, regulating cellular osmotic pressure, and protecting against oxidative stress. In addition, studies have reported that taurine can alleviate the toxic effects of heavy metals, such as copper [8], zinc [9], lead [10], chromium [11], and cadmium [12].

    View all citing articles on Scopus
    View full text