5-Methoxytryptophan attenuates postinfarct cardiac injury by controlling oxidative stress and immune activation

https://doi.org/10.1016/j.yjmcc.2021.05.014Get rights and content

Highlights

  • Cardiac 5-MTP biosynthesis is impaired following myocardial infarction (MI).

  • 5-MTP treatment within 24 h of MI improves left ventricular systolic function.

  • 5-MTP administered within 24 h of MI effectively attenuated cardiac inflammation.

  • 5-MTP promotes mitochondrial stabilization and controls redox imbalance.

  • 5-MTP protected against H2O2-induced cardiomyocyte and endothelial cell death.

Abstract

Aims

Myocardial infarction (MI) remains a major cause of heart failure. 5-Methoxytryptophan (5-MTP), a 5-methoxyindole metabolite of L-tryptophan, exerts anti-inflammatory and antifibrotic effects, but MI impairs the biosynthesis of cardiac 5-MTP. Therefore, we evaluated the effect of exogenous 5-MTP administration on rescuing post-MI cardiac injury.

Methods and results

After a detailed pharmacokinetic analysis of 5-MTP, Sprague Dawley rats that had undergone left anterior descending coronary artery ligation received intraperitoneal administration of either 17 mg/kg 5-MTP or saline at 0.5 and 24 h after MI. Cardiac systolic function, infarction size, and fibrosis were evaluated using echocardiography, triphenyltetrazolium chloride staining, and Masson trichrome staining, respectively. Myocardial apoptosis was analyzed by staining for caspase-3 and cardiac troponin I. 5-MTP treatment decreased the infarct area and myocardial apoptosis; attenuated systolic dysfunction and left ventricular dilatation; and reduced cardiomyocyte hypertrophy, myocardial fibrosis, and infarct expansion. Crucially, 5-MTP alleviated oxidative stress by preserving mitochondrial antioxidant enzymes and downregulating reactive oxygen species–generating NADPH oxidase isoforms and endothelin-1. Consequently, 5-MTP-treated MI rat hearts exhibited lower levels of chemokines and cytokines, namely interleukin (IL)-1β, IL-18, IL-6, C-C motif chemokine ligand (CCL)-2, and CCL5, accompanied by reduced infiltration of CD11b+ cells and CD4+ T cells. Notably, 5-MTP protected against H2O2-induced damage in HL-1 cardiomyocytes and human umbilical vein endothelial cells in vitro.

Conclusion

5-MTP prevented post-MI cardiac injury by promoting mitochondrial stabilization and controlling redox imbalance. This cytoprotective effect ameliorated macrophage and T-cell infiltration, thus reducing the infarct size, attenuating fibrosis, and restoring myocardial function.

Introduction

Cardiac remodeling and subsequent heart failure (HF) after myocardial infarction (MI) cause long-term morbidity and mortality [1]. Timely revascularization can reduce myocardial injury and progressive ventricular remodeling [2,3], but this standard therapeutic modality is not available in all medical centers. Moreover, despite epicardial recanalization, myocardial hypoperfusion related to microvascular injury continues to be a challenge [4]. Revascularization may also induce reperfusion injury, which limits the benefit of preserving ventricular function [5]. In the present study, we identified a novel therapeutic strategy to alleviate cardiac injury after nonreperfused MI.

Apoptosis and inflammation following acute MI are critical pathophysiological processes that lead to cardiac remodeling and dysfunction [6]. Apoptosis of cardiomyocytes and nonmyocytes occurs in infarct and peri-infarct zones immediately after MI [7,8]. Excessive apoptosis can lead to progressive myocardial loss, fibrosis, and functional deterioration [9]. Inflammation is essential in the repair process after an acute cardiac insult [10,11], but exaggerated inflammatory responses can cause tissue damage, cardiac remodeling, and fibrosis [10]. At the site of ischemic injury, CXC motif chemokine ligand 2 (CXCL2) and CC motif chemokine ligand 2 (CCL2) play crucial roles in regulating the migration and infiltration of neutrophils and monocytes/macrophages [12,13]. Besides, several proinflammatory cytokines, including tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6, are also expressed to various degrees [14].

The elevated production of reactive oxygen species (ROS) causes oxidative stress. In the infarcted myocardium, a major source of ROS is the infiltrating neutrophils and monocytes involved in the coordinated activation of NADPH oxidase (NOX) and the neutrophil-derived heme enzyme myeloperoxidase (MPO) [15]. Following MI, NOX activation and mitochondrial dysfunction trigger a ROS burst [16]. Three NOX isoforms (NOX1, NOX2, and NOX4) are expressed in heart tissues and contribute to ROS genesis [17]. Moreover, activation of NOX can stimulate endothelin-1 (ET-1) release [18], and inhibition of ET-1 in rats has been reported to confer cardioprotection [19]. Overall, this redox dysregulation is critical in the pathogenesis of HF after MI [20]. Therefore, the development of innovative therapeutic approaches to control inflammation and oxidative stress remains an urgent need.

5-Methoxytrptophan (5-MTP) was first identified as a tryptophan metabolite synthesized by and released from human fibroblasts [21]. Enzymatic catalysis of 5-MTP in human cells involves two enzymes: tryptophan hydroxylase (TPH), which catalyzes the conversion of L-tryptophan to 5-hydroxytryptophan (5-HTP), and hydroxyindole O-methyltransferase, which converts 5-HTP to 5-MTP [22]. Of the two TPH isoforms, only TPH-1, which mediates 5-MTP synthesis, is expressed in human cells [22]. Furthermore, a defect in the TPH-1 gene was demonstrated to cause cardiac dysfunction [23].

The anti-inflammatory properties of 5-MTP have been reported both in vitro and in vivo [24]. Vascular endothelial cells produce and secrete 5-MTP, which acts in a paracrine and autocrine manner to protect endothelial barrier function, control the expression of adhesion molecules, and inhibit leukocyte transmigration [25,26]. Furthermore, 5-MTP inhibits the expression of lipopolysaccharide (LPS)-induced cyclooxygenase-2 and inducible nitric oxide synthase (iNOS) and the release of proinflammatory cytokines and chemokines in macrophages [27]. Mechanistically, 5-MTP exerts anti-inflammatory effects on endothelial cells, macrophages, and vascular smooth muscle cells by blocking p38 and nuclear factor (NF)-κB activation [27,28]. In a carotid artery ligation model, 5-MTP was observed to prevent arterial injury–induced intimal hyperplasia [27]. 5-MTP also ameliorates renal ischemia/reperfusion injury and suppresses bleomycin-induced pulmonary fibrosis through an antifibrotic effect [29,30]. Regarding cardioprotective effects, 5-MTP was found to rescue rat cardiomyocytes from H2O2-induced oxidative damage [31]. Moreover, clinically, 5-MTP appears to be a useful early marker of HF after MI [32]. Therefore, it is reasonable to hypothesize that 5-MTP protects against post-MI injury and dysfunction by controlling inflammation and fibrosis.

In this study, we investigated this hypothesis, and our findings provide evidence in support of it. 5-MTP administration to a rat coronary artery ligation model reduced infarct size, attenuated myocardial inflammation, alleviated cardiac fibrosis, and preserved cardiac function.

Section snippets

Animal surgery

Outbred 6–8-week-old female Sprague Dawley rats (225–300 g, LASCO, Taipei, Taiwan) were anesthetized with 3% isoflurane (Halocarbon Laboratories, North Augusta, South Carolina, USA) and underwent either sham surgery or left anterior descending coronary artery (LAD) ligation for MI induction. We permanently occluded the mid-LAD by using a 6–0 suture (Nylon, NC126, UNIK, Taipei, Taiwan) to close the ligation. To standardize the ligation, we ligated the LAD approximately 3–4 mm beneath the left

5-MTP improved post-MI cardiac function and reduced chamber enlargement

LAD ligation in Sprague Dawley rats induced an acute ST-elevation MI (Fig. S1A, S1B). The cells found positive for 5-MTP through immunohistochemical staining were detectable at various time points after MI (Fig. 1A); among them, 5-MTP was present surrounding CD31+ cells (Fig. 1B). mRNA levels of TPH1 (Fig. 1C), a key enzyme naturally expressed in the normal myocardium for 5-MTP synthesis, and the plasma concentration of 5-MTP (Fig. 1D), decreased at 24 h and 48 h after LAD ligation. These

Discussion

Consistent with a previous report [32], we demonstrated that MI suppressed 5-MTP production by cardiovascular cells and that exogenous administration of 5-MTP rescued the damaged hearts from post-MI injury and cardiac fibrosis in a rat nonreperfused MI model. We provided evidence of geometric alteration accompanied by left ventricular dilation and thinning in the infarct zone 4 weeks after MI. In addition, we demonstrated the pathological changes in the areas distant from the infarct zone,

Conclusions

We demonstrated that 5-MTP administration during acute MI results in a significant reduction in left ventricular injury and the preservation of left ventricular function. The beneficial effect of 5-MTP was accompanied by decreased infarct size, blunted ROS formation, reduced immune cell infiltration, and suppressed proinflammatory cytokines and chemokines in the post-MI acute phase. Moreover, 5-MTP downregulated NOX family and ET-1, which in turn may regulate both hypertrophic and fibrotic

Funding

This work was supported by research grants from the National Health Research Institutes (NHRI-CS-109-PP-14, NHRI-CS-110-PP-14), the Ministry of Science and Technology (MOST-107-2321-B-002-043, MOST-108-2321-B002-019, MOST-109-2326-B-002-005), and National Taiwan University Hospital (UN-109-015, UN-110-025).

Declaration of Competing Interest

Non declared.

Acknowledgments

We thank the Department of Pathology, National Taiwan University Hospital for assisting with histological staining. We also acknowledge the team at the Taipei Medical University (TMU) Core Facility for their assistance with LC/MS; we are particularly grateful to Dr. I-Lin Tsai, Mr. Chun-Chih Jared Liu, and Ms. Yuan-Chin Hsiung for their excellent technical support. This manuscript was edited by Wallace Academic Editing.

References (51)

  • H. Nojiri et al.

    Oxidative stress causes heart failure with impaired mitochondrial respiration

    J. Biol. Chem.

    (2006)
  • A. Hameed et al.

    Immunohistochemical expression of CD68 antigen in human peripheral blood T cells

    Hum. Pathol.

    (1994)
  • N.E. Hahn et al.

    NOX5 expression is increased in intramyocardial blood vessels and cardiomyocytes after acute myocardial infarction in humans

    Am. J. Pathol.

    (2012)
  • M.A. Pfeffer et al.

    Ventricular remodeling after myocardial infarction. Experimental observations and clinical implications

    Circulation

    (1990)
  • M.V. Basalay et al.

    Targeting myocardial ischaemic injury in the absence of reperfusion

    Basic Res. Cardiol.

    (2020)
  • H.M. Piper et al.

    A fresh look at reperfusion injury

    Cardiovasc. Res.

    (1998)
  • E. Teringova et al.

    Apoptosis in ischemic heart disease

    J. Transl. Med.

    (2017)
  • A. Johnson et al.

    Apoptosis and angiogenesis: an evolving mechanism for fibrosis

    FASEB J.

    (2013)
  • N.G. Frangogiannis

    The inflammatory response in myocardial injury, repair, and remodelling

    Nat. Rev. Cardiol.

    (2014)
  • C.J. Zuurbier et al.

    Innate immunity as a target for acute cardioprotection

    Cardiovasc. Res.

    (2019)
  • S.L. Deshmane et al.

    Monocyte chemoattractant protein-1 (MCP-1): an overview

    J. Interf. Cytokine Res.

    (2009)
  • L.Y. Guo et al.

    CXCL2, a new critical factor and therapeutic target for cardiovascular diseases

    Clin. Exp. Hypertens.

    (2020)
  • M. Zhang et al.

    Status of cytokines in ischemia reperfusion induced heart injury

    Cardiovasc. Hematol. Disord. Drug Targets

    (2008)
  • M. El Kazzi et al.

    Neutrophil-mediated cardiac damage after acute myocardial infarction: significance of defining a new target cell type for developing cardioprotective drugs

    Antioxid. Redox Signal.

    (2020)
  • D. Moris et al.

    The role of reactive oxygen species in the pathophysiology of cardiovascular diseases and the clinical significance of myocardial redox

    Ann. Transl. Med.

    (2017)
  • Cited by (11)

    • Mitochondrial interaction of fibrosis-protective 5-methoxy tryptophan enhances collagen uptake by macrophages

      2022, Free Radical Biology and Medicine
      Citation Excerpt :

      These results motivated us to search for other mechanisms by which 5-MTP influences the macrophages. Since 5-MTP has been reported to influence mitochondria [24], and as mitochondrial functions relate to macrophage functions [3,10], we speculated that 5-MTP might affect the mitochondria. 5-MTP is an amphipatic molecule expected to partion in lipid membranes and alter their physicochemical properties such as lateral membrane tension.

    View all citing articles on Scopus
    View full text