Novel Cardiokine GDF3 Predicts Adverse Fibrotic Remodeling After Myocardial Infarction

Circulation. 2023 Feb 7;147(6):498-511. doi: 10.1161/CIRCULATIONAHA.121.056272. Epub 2022 Dec 9.

Abstract

Background: Myocardial infarction (MI) induces a repair response that ultimately generates a stable fibrotic scar. Although the scar prevents cardiac rupture, an excessive profibrotic response impairs optimal recovery by promoting the development of noncontractile fibrotic areas. The mechanisms that lead to cardiac fibrosis are diverse and incompletely characterized. We explored whether the expansion of cardiac fibroblasts after MI can be regulated through a paracrine action of cardiac stromal cells.

Methods: We performed a bioinformatic secretome analysis of cardiac stromal PW1+ cells isolated from normal and post-MI mouse hearts to identify novel secreted proteins. Functional assays were used to screen secreted proteins that promote fibroblast proliferation. The expressions of candidates were subsequently analyzed in mouse and human hearts and plasmas. The relationship between levels of circulating protein candidates and adverse post-MI cardiac remodeling was examined in a cohort of 80 patients with a first ST-segment-elevation MI and serial cardiac magnetic resonance imaging evaluations.

Results: Cardiac stromal PW1+ cells undergo a change in paracrine behavior after MI, and the conditioned media from these cells induced a significant increase in the proliferation of fibroblasts. We identified a total of 12 candidates as secreted proteins overexpressed by cardiac PW1+ cells after MI. Among these factors, GDF3 (growth differentiation factor 3), a member of the TGF-β (transforming growth factor-β) family, was markedly upregulated in the ischemic hearts. Conditioned media specifically enriched with GDF3 induced fibroblast proliferation at a high level by stimulation of activin-receptor-like kinases. In line with the secretory nature of this protein, we next found that GDF3 can be detected in mice and human plasma samples, with a significant increase in the days after MI. In humans, higher GDF3 circulating levels (measured in the plasma at day 4 after MI) were significantly associated with an increased risk of adverse remodeling 6 months after MI (adjusted odds ratio, 1.76 [1.03-3.00]; P=0.037), including lower left ventricular ejection fraction and a higher proportion of akinetic segments.

Conclusions: Our findings define a mechanism for the profibrotic action of cardiac stromal cells through secreted cardiokines, such as GDF3, a candidate marker of adverse fibrotic remodeling after MI.

Registration: URL: https://www.

Clinicaltrials: gov; Unique identifier: NCT01113268.

Keywords: biomarker; cardiac remodeling; fibrosis; growth differentiation factor; myocardial infarction.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Cicatrix / pathology
  • Culture Media, Conditioned / metabolism
  • Culture Media, Conditioned / pharmacology
  • Disease Models, Animal
  • Fibrosis
  • Growth Differentiation Factor 3 / metabolism
  • Humans
  • Mice
  • Myocardial Infarction*
  • Myocardium* / metabolism
  • Stroke Volume
  • Transforming Growth Factor beta / metabolism
  • Ventricular Function, Left
  • Ventricular Remodeling

Substances

  • Culture Media, Conditioned
  • Growth Differentiation Factor 3
  • Transforming Growth Factor beta
  • GDF3 protein, human
  • Gdf3 protein, mouse

Associated data

  • ClinicalTrials.gov/NCT01113268