Elsevier

International Journal of Cardiology

Volume 341, 15 October 2021, Pages 24-30
International Journal of Cardiology

Platelet derived growth factor-A (Pdgf-a) gene transfer modulates scar composition and improves left ventricular function after myocardial infarction

https://doi.org/10.1016/j.ijcard.2021.07.021Get rights and content

Highlights

  • Recombinant AAV9 Pdgf-a gene therapy upregulates PDGF-A in the rat heart.

  • Pdgf-a gene therapy led to improved left ventricular function post myocardial infarction.

  • PDGF-A upregulation increased neovascularization in the border zone and Collagen type III alpha 1 in the scar.

  • This study highlights post-infarct scar modulation as a target for cardiac reparative therapies.

Abstract

Background: Novel therapies that can limit or reverse damage caused by myocardial infarction (MI) could ease the increasing burden of heart failure. In this regard Platelet Derived Growth Factor (PDGF) has been previously shown to contribute to cardiac repair after MI. Here, we use a rodent model of MI and recombinant adeno-associated virus 9 (rAAV9)-mediated gene transfer to overexpress Pdgf-a in the injured heart and assess its therapeutic potential.

Methods and results: Sprague Dawley rats underwent temporary occlusion of the left anterior descending coronary artery, followed immediately by systemic delivery of 1 × 10^11 vector genomes of either rAAV9 Pdgf-a or rAAV9 Empty vector (control). At day 28 post-MI echocardiography showed significantly improved left ventricular (LV) function (fractional shortening) after rAAV9 Pdgf-a (0.394 ± 0.019%) treatment vs control (0.304 ± 0.018%). Immunohistochemical analysis demonstrated significantly increased capillary and arteriolar density in the infarct border zone of rAAV9 Pdgf-a treated hearts together with a significant reduction in infarct scar size (rAAV9 Pdgf-a 6.09 ± 0.94% vs Empty 12.45 ± 0.92%). Western blot and qPCR analyses confirmed overexpression of PDGF-A and showed upregulation of smooth muscle alpha actin (Acta2), collagen type III alpha 1 (Col3a1) and lysyl oxidase (Lox) genes in rAAV9 Pdgf-a treated infarcts.

Conclusion: Overexpression of Pdgf-a in the post-MI heart can modulate scar composition and improve LV function. Our study highlights the potential of rAAV gene transfer of Pdgf-a as a cardio-reparative therapy.

Introduction

Heart failure (HF) is a progressive and debilitating condition, affecting over 37 million people worldwide and costing more than US$100 billion each year [1,2]. The adult myocardium has very limited self-regenerative capacity; previous studies have suggested a small proportion of existing cardiomyocytes can re-enter the cell cycle to proliferate, with a turnover rate estimated at 0.5% – 2% per year [3]. This is insufficient to replace the hundreds of millions of cardiomyocytes lost during and immediately following MI. As a result, the left ventricle undergoes adverse remodelling. Depending on infarct size, the bulk of the infarcted myocardium is replaced by collagen-rich scar tissue. Numerous cytokines and growth factors regulate the activation of cardiac interstitial fibroblasts and therefore extracellular matrix deposition and scar formation during post-infarct wound healing [4].

Platelet derived growth factor (PDGF) is a potent mitogen and chemotactic agent for cardiac fibroblasts. There are multiple PDGF isoforms (A, B, C, and D), which combine to form biologically active dimers. PDGF dimers bind tyrosine kinase receptors, platelet derived growth factor receptor alpha (PDGFRα) and beta (PDGFRβ) to initiate cell signalling [[5], [6], [7]]. We have previously identified a PDGFRα+ cardiac progenitor cell population and have reported on therapeutic effects of transplanting these cells [[8], [9], [10], [11]]. These PDGFRα+/CD90+/CD31- cells demonstrate a clonogenic progenitor phenotype and give rise to cardiac interstitial fibroblasts, vascular smooth muscle and endothelial cells.

PDGF and its receptors have complex timing and context-dependent roles in cardiac development, fibrosis and post-infarct remodelling. Although adverse pro-fibrotic effects in the normal heart after transgenic overexpression of PDGF isoforms have been reported [12], PDGFRα and -β signalling are also known to mediate effective and beneficial wound healing in the infarcted heart, orchestrating collagen deposition and neovessel formation [13]. Intramyocardial and systemic treatment with PDGF-AB enhances angiogenesis and reduces infarct size in small animal models [14,15]. Furthermore, our group recently demonstrated that systemic administration of recombinant human PDGF-AB improves survival and cardiac function in a pre-clinical, large animal model of MI [16]. Mechanistically, these benefits were mediated by increasing collagen anisotropy and neovasculogenesis.

Gene transfer via recombinant adeno-associated virus (rAAV) is a powerful method to overexpress a specific gene directly. Adeno-associated viruses have minimal immunogenicity compared to other vector types and can achieve persistent overexpression with minimal DNA integration in the host genome [17,18]. Here, we use rAAV9 vector-mediated gene transfer to investigate the therapeutic potential of Pdgf-a overexpression in a rat model of MI. We demonstrate that Pdgf-a upregulation in the heart reduces scar size, modulates scar collagen composition and increases vascularity in the peri-infarct region leading to improved post-infarct left ventricular (LV) function. This supports overexpression of PDGF-A after MI as a potential cardiac therapeutic.

Section snippets

Myocardial infarction (ischemia-reperfusion) model and rAAV9 Pdgf-a delivery

Adult male Sprague Dawley rats (8–12 weeks of age) were subjected to myocardial ischemia/reperfusion (I/R) injury by occlusion of the left anterior descending coronary artery (LAD) for 90 min [19]. rAAV9 Pdgf-a (1 × 10^11 vector copies per animal) was injected via the tail vein under anaesthesia, immediately following I/R. The control group was injected with the same dose of rAAV9 Empty. Two experimental time points were set for tissue harvesting; day 14 and day 28 post-I/R. All animal

rAAV9 Pdgf-a induces overexpression of PDGF-A and improves cardiac function after myocardial infarction

After confirming rAAV9 Pdgf-a transduction efficacy and Pdgf-a transgene functionality in vitro (Supplementary Fig. 1) we then evaluated rAAV9 cardiac transduction after systemic administration (tail-vein injection) in rats with induced myocardial infarction (MI). At 7 days post-MI, we quantified rAAV9 Pdgf-a and rAAV9 Empty vector copy numbers in the heart, liver, lung, and kidney (Fig. 1a). Vector copy numbers were detected in all organs, but were significantly higher in the heart and liver.

Discussion

This study demonstrates that systemic administration of rAAV9 Pdgf-a after induction of MI: 1) significantly improves LV function 28 days post-I/R; 2) increases revascularization of the peri-infarct border zone with de novo arterioles and capillaries, and 3) results in upregulation of COL3A1 in the infarct zone.

Recombinant AAV is the most commonly used gene chaperoning system in clinical trials for gene therapy [23]. As AAV serotype 9 (AAV9) has high transduction efficiency for rat myocardium,

Conclusion

This study shows that systemic gene delivery of Pdgf-a using the rAAV9 vector increases neovascularization and modulates scar collagen composition in the infarcted rodent heart, which is accompanied by an improvement in LV systolic function. This highlights the extracellular matrix as a potential target for therapies to improve ventricular function in the post-infarct heart.

Disclosures

The authors declare that there is no conflict of interest regarding the publication of this article.

Funding acknowledgement

This work was supported by the National Health and Medical Research Council [APP1100046]; the National Heart Foundation, Australia [100463]; and a Stem Cells Australia Project Grant.

Declaration of Competing Interest

None.

References (46)

  • A.V. Shinde et al.

    Fibroblasts in myocardial infarction: a role in inflammation and repair

    J. Mol. Cell. Cardiol.

    (2014)
  • S.Y. Lim et al.

    Adjuvant role of macrophages in stem cell-induced cardiac repair in rats

    Exp. Mol. Med.

    (2018)
  • M.A. Horn et al.

    Aging and the cardiac collagen matrix: novel mediators of fibrotic remodelling

    J. Mol. Cell. Cardiol.

    (2016)
  • E.J. Miller et al.

    Quantitation of type I, III, and V collagens in human tissue samples by high-performance liquid chromatography of selected cyanogen bromide peptides

    Anal. Biochem.

    (1991)
  • N.C. Avery et al.

    The effects of the Maillard reaction on the physical properties and cell interactions of collagen

    Pathol. Biol. (Paris)

    (2006)
  • H.N. Lovvorn et al.

    Relative distribution and crosslinking of collagen distinguish fetal from adult sheep wound repair

    J. Pediatr. Surg.

    (1999)
  • B. Ziaeian et al.

    Epidemiology and aetiology of heart failure

    Nat. Rev. Cardiol.

    (2016)
  • W.J. Richardson et al.

    Physiological implications of myocardial scar structure

    Compr. Physiol.

    (2015)
  • X. Yang et al.

    PDGF signalling controls the migration of mesoderm cells during chick gastrulation by regulating N-cadherin expression

    Development

    (2008)
  • J. Yu et al.

    Platelet-derived growth factor signaling and human cancer

    J. Biochem. Mol. Biol.

    (2003)
  • J.J. Chong et al.

    Progenitor cells identified by PDGFR-alpha expression in the developing and diseased human heart

    Stem Cells Dev.

    (2013)
  • T.Y.L. Le et al.

    Enhanced cardiac repair by telomerase reverse transcriptase over-expression in human cardiac mesenchymal stromal cells

    Sci. Rep.

    (2019)
  • T.Y.L. Le et al.

    Platelet-derived growth factor receptor-alpha expressing cardiac progenitor cells can be derived from previously cryopreserved human heart samples

    Stem Cells Dev.

    (2018)
  • Cited by (0)

    1

    FNR, ZEC and MO contributed equally to this work.

    2

    Masahito Ogawa (Present Address): Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Centre, Osaka, Japan.

    View full text