Abstract

The recruitment and subsequent polarization of inflammatory monocytes/macrophages in the perivascular regions of pulmonary arteries is a key feature of pulmonary hypertension (PH). However, the mechanisms driving macrophage polarization within the adventitial microenvironment during PH progression remain unclear. We previously established that reciprocal interactions between fibroblasts and macrophages are essential in driving the activated phenotype of both cell types although the signals involved in these interactions remain undefined. We sought to test the hypothesis that adventitial fibroblasts produce a complex array of metabolites and proteins that coordinately direct metabolomic and transcriptomic re-programming of naïve macrophages to recapitulate the pathophysiologic phenotype observed in PH. Media conditioned by pulmonary artery adventitial fibroblasts isolated from pulmonary hypertensive (PH-CM) or age-matched control (CO-CM) calves were used to activate bone marrow derived macrophages. RNA-Seq and mass spectrometry-based metabolomics analyses were performed. Fibroblast conditioned medium from patients with idiopathic pulmonary arterial hypertension or controls were used to validate transcriptional findings. The microenvironment was targeted in vitro using a fibroblast-macrophage co-culture system and in vivo in a mouse model of hypoxia-induced PH. Both CO-CM and PH-CM actively, yet distinctly regulated macrophage transcriptomic and metabolomic profiles. Network integration revealed coordinated rewiring of pro-inflammatory and pro-remodeling gene regulation in concert with altered mitochondrial and intermediary metabolism in response to PH-CM. Pro-inflammation and metabolism are key regulators of macrophage phenotype in vitro, and are closely related to in vivo flow sorted lung interstitial/perivascular macrophages from hypoxic mice. Metabolic changes are accompanied by increased free NADH levels and increased expression of a metabolic sensor and transcriptional co-repressor, C-terminal binding protein 1 (CtBP1), a mechanism shared with adventitial PH-fibroblasts. Targeting the microenvironment created by both cell types with the CtBP1 inhibitor MTOB, inhibited macrophage pro-inflammatory and metabolic re-programming both in vitro and in vivo. In conclusion, coordinated transcriptional and metabolic reprogramming is a critical mechanism regulating macrophage polarization in response to the complex adventitial microenvironment in PH. Targeting the adventitial microenvironment can return activated macrophages toward quiescence and attenuate pathological remodeling that drives PH progression.

Highlights

  • Pulmonary Hypertension (PH) is a progressive and devastating disease characterized by both vascular inflammation and structural remodeling [1,2,3,4,5,6,7]

  • We found that even though IL6, lactate, or IL6+lactate can stimulate IL1b, Vegf, Arg1, Asl and Ass1 gene expression in BMDM, the level of the induction is far less than that of complete PH-Conditioned media (CM) (Supplemental Figure 1)

  • We treated naïve mouse BMDMs with cell culture media conditioned by adventitial fibroblasts isolated from either pulmonary hypertensive (PH-CM) or age-matched control neonatal calves (CO-CM), and performed RNA Sequencing (RNA-Seq) analysis

Read more

Summary

Introduction

Pulmonary Hypertension (PH) is a progressive and devastating disease characterized by both vascular inflammation and structural remodeling [1,2,3,4,5,6,7]. Recruitment of circulating monocyte/macrophages to the pulmonary arterial wall, especially the adventitial compartment, is a prerequisite for vascular remodeling and development of PH [1, 4, 10,11,12,13,14,15,16,17]. It is logical to believe that the functional phenotype of resident or recruited macrophages changes dynamically in response to local environmental and metabolic cues. These considerations are likely to be especially important in diseases of chronic and progressive inflammatory-fibrotic remodeling such as PH. Little is known of the specific metabolic and transcriptomic re-programming of pulmonary artery adventitial macrophages under homeostatic or pathologic conditions

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call