Skip to main content

Daily Cardiology Research Analysis

3 papers

Three high-impact cardiology studies advance mechanisms and therapeutics across domains. Human cardiac allograft vasculopathy is linked to intragraft plasma cells producing bilirubin‑reactive antibodies, revealing a heme catabolism–immune axis. Disturbed flow plus hypercholesterolemia drives endothelial reprogramming to immune‑like and foam‑like states, while transient mRNA‑based PKM2 overexpression in pigs enhances myocardial repair and function after infarction.

Summary

Three high-impact cardiology studies advance mechanisms and therapeutics across domains. Human cardiac allograft vasculopathy is linked to intragraft plasma cells producing bilirubin‑reactive antibodies, revealing a heme catabolism–immune axis. Disturbed flow plus hypercholesterolemia drives endothelial reprogramming to immune‑like and foam‑like states, while transient mRNA‑based PKM2 overexpression in pigs enhances myocardial repair and function after infarction.

Research Themes

  • Transplant vasculopathy immunopathogenesis (bilirubin-targeting antibodies)
  • Endothelial reprogramming under disturbed flow and hypercholesterolemia
  • mRNA-based cardiac regeneration after myocardial infarction

Selected Articles

1. Dominant intragraft plasma cells targeting bilirubin implicate local heme catabolism in human cardiac allograft vasculopathy.

85.5Level IVCase seriesThe Journal of clinical investigation · 2025PMID: 41289018

Using single-cell profiling and recombinant antibodies from intragraft plasma cells, the authors show that most graft-derived antibodies in human CAV target bilirubin, with corresponding bilirubin accumulation and heme-catabolic enzyme expression in lesions. This identifies local heme catabolism as a likely antigenic driver and a therapeutic target in CAV.

Impact: First mechanistic evidence that bilirubin-specific antibodies dominate intragraft responses in human CAV links metabolism to alloimmune pathology, opening new diagnostic and therapeutic avenues.

Clinical Implications: If validated, targeting heme catabolism (e.g., HO-1/biliverdin reductase pathways) or neutralizing bilirubin-specific antibodies could modify CAV progression and enable biomarker development (bilirubin deposition patterns).

Key Findings

  • 57% (21/37) of graft-derived recombinant antibodies reacted to bilirubin, while none from peripheral blood plasma cells did.
  • Bilirubin deposition localized to lymphocytic aggregates and smooth muscle cell cytoplasm/nuclei in CAV arteries, not seen in healthy hearts.
  • Heme-oxygenase-1 and biliverdin reductases were expressed in graft-infiltrating macrophages with Fe2+ present in hyperplastic arterial media.

Methodological Strengths

  • Single-cell RNA sequencing with immunoglobulin gene profiling to define dominant intragraft plasma cell clones
  • Functional recombinant monoclonal antibody generation and antigen screening, coupled with histologic and tissue iron analyses

Limitations

  • Causality for bilirubin-targeting antibodies in driving CAV progression is not established; interventional validation is lacking
  • Sample size and generalizability across centers and time post-transplant are not fully defined

Future Directions: Test whether inhibiting heme catabolism or neutralizing bilirubin-reactive antibodies alters CAV progression in models and clinical trials; develop noninvasive imaging or circulating biomarkers for bilirubin deposition.

2. Transient overexpression of hPKM2 in porcine cardiomyocytes prevents heart failure after myocardial infarction.

83Level VCase seriesNature communications · 2025PMID: 41285815

A non-viral modified mRNA platform transiently increased PKM2 in porcine cardiomyocytes, augmenting cell-cycle activity, paracrine protection, cardiac function, and reducing fibrosis post-MI in both juvenile and adult animals. The large-animal evidence supports translational feasibility for regenerative therapy to prevent post-infarction heart failure.

Impact: Demonstrates practical, transient mRNA-based myocardial regeneration in a clinically relevant large-animal model, advancing a potential new class of therapy beyond current standards.

Clinical Implications: Supports development of transient mRNA therapeutics targeting cardiomyocyte cell-cycle and metabolism after MI; informs dosing/timing strategies and safety endpoints for first-in-human trials.

Key Findings

  • Modified mRNA delivery of PKM2 increased cardiomyocyte cell-division markers and secretion of protective factors in juvenile pigs.
  • Both juvenile (2 months) and adult (1 month) pigs showed improved cardiac function and reduced fibrosis after PKM2 mRNA treatment.
  • Non-viral, targeted mRNA platform enabled transient expression with regenerative benefit, supporting translational potential.

Methodological Strengths

  • Large-animal (porcine) models including juvenile and adult arms with functional and histologic endpoints
  • Targeted non-viral modified mRNA delivery enabling transient gene expression with translational relevance

Limitations

  • Long-term durability, arrhythmogenic risk, and off-target effects were not fully assessed
  • Human efficacy and safety remain untested; dosing and delivery optimization needed

Future Directions: Define long-term safety/efficacy, dose-response, and delivery route; assess combination with standard post-MI care; initiate phase 1 trials with arrhythmia and fibrosis endpoints.

3. Disturbed flow induces reprogramming of endothelial cells to immune-like and foam cells under hypercholesterolaemia during atherogenesis.

80Level VCase seriesCardiovascular research · 2025PMID: 41288601

Using AAV‑PCSK9 Western diet mice with disturbed flow, single-cell profiling (98,553 cells) and EC-lineage tracing reveal that d-flow plus hypercholesterolemia reprograms endothelial cells into immune-like (EndIT) and foam-like (EndFT) states, beyond inflammation and EndMT. Human plaque datasets and HAEC experiments corroborate that FIRE occurs in human atherosclerosis.

Impact: Introduces and validates a two-hit model in which flow disturbance and hypercholesterolemia jointly drive endothelial transdifferentiation to foam-like cells, redefining cellular origins and targets in atherogenesis.

Clinical Implications: Suggests therapeutic strategies targeting flow-sensitive pathways to prevent endothelial immune/foam transitions; supports patient stratification by hemodynamic risk and lipid status.

Key Findings

  • Atherosclerotic plaques formed only when disturbed flow was combined with hypercholesterolemia, not by either alone.
  • scRNA-seq (98,553 cells) identified endothelial transitions to immune-like (EndIT) and foam-like (EndFT) states in vivo.
  • Lineage tracing and human plaque scRNA-seq reanalysis plus HAEC experiments validated FIRE, EndIT, and EndFT signatures.

Methodological Strengths

  • Multimodal validation: scRNA-seq across 95 mice, EC-lineage tracing, human plaque data reanalysis, and HAEC in vitro studies
  • Physiologic two-hit model integrating hemodynamic disturbance with lipid loading (AAV-PCSK9 + Western diet + partial carotid ligation)

Limitations

  • Causal interventions specifically reversing EndFT/EndIT in vivo are not yet demonstrated
  • Translation to clinical endpoints remains to be tested despite human corroboration

Future Directions: Develop interventions that modulate flow-sensitive signaling to prevent EndIT/EndFT; test in large animals and assess imaging/biomarkers for endothelial phenotypes.