[HTML][HTML] Transcriptome analysis of human heart failure reveals dysregulated cell adhesion in dilated cardiomyopathy and activated immune pathways in ischemic …

ME Sweet, A Cocciolo, D Slavov, KL Jones, JR Sweet… - BMC genomics, 2018 - Springer
ME Sweet, A Cocciolo, D Slavov, KL Jones, JR Sweet, SL Graw, TB Reece, AV Ambardekar
BMC genomics, 2018Springer
Background Current heart failure (HF) treatment is based on targeting symptoms and left
ventricle dysfunction severity, relying on a common HF pathway paradigm to justify common
treatments for HF patients. This common strategy may belie an incomplete understanding of
heterogeneous underlying mechanisms and could be a barrier to more precise treatments.
We hypothesized we could use RNA-sequencing (RNA-seq) in human heart tissue to
delineate HF etiology-specific gene expression signatures. Results RNA-seq from 64 human …
Background
Current heart failure (HF) treatment is based on targeting symptoms and left ventricle dysfunction severity, relying on a common HF pathway paradigm to justify common treatments for HF patients. This common strategy may belie an incomplete understanding of heterogeneous underlying mechanisms and could be a barrier to more precise treatments. We hypothesized we could use RNA-sequencing (RNA-seq) in human heart tissue to delineate HF etiology-specific gene expression signatures.
Results
RNA-seq from 64 human left ventricular samples: 37 dilated (DCM), 13 ischemic (ICM), and 14 non-failing (NF). Using a multi-analytic approach including covariate adjustment for age and sex, differentially expressed genes (DEGs) were identified characterizing HF and disease-specific expression. Pathway analysis investigated enrichment for biologically relevant pathways and functions. DCM vs NF and ICM vs NF had shared HF-DEGs that were enriched for the fetal gene program and mitochondrial dysfunction. DCM-specific DEGs were enriched for cell-cell and cell-matrix adhesion pathways. ICM-specific DEGs were enriched for cytoskeletal and immune pathway activation. Using the ICM and DCM DEG signatures from our data we were able to correctly classify the phenotypes of 24/31 ICM and 32/36 DCM samples from publicly available replication datasets.
Conclusions
Our results demonstrate the commonality of mitochondrial dysfunction in end-stage HF but more importantly reveal key etiology-specific signatures. Dysfunctional cell-cell and cell-matrix adhesion signatures typified DCM whereas signals related to immune and fibrotic responses were seen in ICM. These findings suggest that transcriptome signatures may distinguish end-stage heart failure, shedding light on underlying biological differences between ICM and DCM.
Springer