BOLA (BolA Family Member 3) deficiency controls endothelial metabolism and glycine homeostasis in pulmonary hypertension

Q Yu, YY Tai, Y Tang, J Zhao, V Negi, MK Culley… - Circulation, 2019 - Am Heart Assoc
Q Yu, YY Tai, Y Tang, J Zhao, V Negi, MK Culley, J Pilli, W Sun, K Brugger, J Mayr, R Saggar…
Circulation, 2019Am Heart Assoc
Background: Deficiencies of iron-sulfur (Fe-S) clusters, metal complexes that control redox
state and mitochondrial metabolism, have been linked to pulmonary hypertension (PH), a
deadly vascular disease with poorly defined molecular origins. BOLA3 (BolA Family Member
3) regulates Fe-S biogenesis, and mutations in BOLA3 result in multiple mitochondrial
dysfunction syndrome, a fatal disorder associated with PH. The mechanistic role of BOLA3 in
PH remains undefined. Methods: In vitro assessment of BOLA3 regulation and gain-and loss …
Background
Deficiencies of iron-sulfur (Fe-S) clusters, metal complexes that control redox state and mitochondrial metabolism, have been linked to pulmonary hypertension (PH), a deadly vascular disease with poorly defined molecular origins. BOLA3 (BolA Family Member 3) regulates Fe-S biogenesis, and mutations in BOLA3 result in multiple mitochondrial dysfunction syndrome, a fatal disorder associated with PH. The mechanistic role of BOLA3 in PH remains undefined.
Methods
In vitro assessment of BOLA3 regulation and gain- and loss-of-function assays were performed in human pulmonary artery endothelial cells using siRNA and lentiviral vectors expressing the mitochondrial isoform of BOLA3. Polymeric nanoparticle 7C1 was used for lung endothelium-specific delivery of BOLA3 siRNA oligonucleotides in mice. Overexpression of pulmonary vascular BOLA3 was performed by orotracheal transgene delivery of adeno-associated virus in mouse models of PH.
Results
In cultured hypoxic pulmonary artery endothelial cells, lung from human patients with Group 1 and 3 PH, and multiple rodent models of PH, endothelial BOLA3 expression was downregulated, which involved hypoxia inducible factor-2α–dependent transcriptional repression via histone deacetylase 1–mediated histone deacetylation. In vitro gain- and loss-of-function studies demonstrated that BOLA3 regulated Fe-S integrity, thus modulating lipoate-containing 2-oxoacid dehydrogenases with consequent control over glycolysis and mitochondrial respiration. In contexts of siRNA knockdown and naturally occurring human genetic mutation, cellular BOLA3 deficiency downregulated the glycine cleavage system protein H, thus bolstering intracellular glycine content. In the setting of these alterations of oxidative metabolism and glycine levels, BOLA3 deficiency increased endothelial proliferation, survival, and vasoconstriction while decreasing angiogenic potential. In vivo, pharmacological knockdown of endothelial BOLA3 and targeted overexpression of BOLA3 in mice demonstrated that BOLA3 deficiency promotes histological and hemodynamic manifestations of PH. Notably, the therapeutic effects of BOLA3 expression were reversed by exogenous glycine supplementation.
Conclusions
BOLA3 acts as a crucial lynchpin connecting Fe-S–dependent oxidative respiration and glycine homeostasis with endothelial metabolic reprogramming critical to PH pathogenesis. These results provide a molecular explanation for the clinical associations linking PH with hyperglycinemic syndromes and mitochondrial disorders. These findings also identify novel metabolic targets, including those involved in epigenetics, Fe-S biogenesis, and glycine biology, for diagnostic and therapeutic development.
Am Heart Assoc