Theranostics 2024; 14(7):2856-2880. doi:10.7150/thno.88223 This issue Cite

Research Paper

Activation of the hypoxia-inducible factor pathway protects against acute ischemic stroke by reprogramming central carbon metabolism

Sarah Madai1, Pinar Kilic1, Rolf M. Schmidt2, Carlos Bas-Orth2,*, Thomas Korff1, Michael Büttner3, Glynis Klinke3, Gernot Poschet3, Hugo H. Marti1, Reiner Kunze1✉

1. Institute of Physiology and Pathophysiology, Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, Germany.
2. Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany.
3. Metabolomics Core Technology Platform, Centre for Organismal Studies, Heidelberg University, Heidelberg, Germany.
* Present address: Institute of Clinical Neuroanatomy, Goethe University Frankfurt, Frankfurt, Germany

Citation:
Madai S, Kilic P, Schmidt RM, Bas-Orth C, Korff T, Büttner M, Klinke G, Poschet G, Marti HH, Kunze R. Activation of the hypoxia-inducible factor pathway protects against acute ischemic stroke by reprogramming central carbon metabolism. Theranostics 2024; 14(7):2856-2880. doi:10.7150/thno.88223. https://www.thno.org/v14p2856.htm
Other styles

File import instruction

Abstract

Graphic abstract

Cell metabolism reprogramming to sustain energy production, while reducing oxygen and energy consuming processes is crucially important for the adaptation to hypoxia/ischemia. Adaptive metabolic rewiring is controlled by hypoxia-inducible factors (HIFs). Accumulating experimental evidence indicates that timely activation of HIF in brain-resident cells improves the outcome from acute ischemic stroke. However, the underlying molecular mechanisms are still incompletely understood. Thus, we investigated whether HIF-dependent metabolic reprogramming affects the vulnerability of brain-resident cells towards ischemic stress.

Methods: We used genetic and pharmacological approaches to activate HIF in the murine brain in vivo and in primary neurons and astrocytes in vitro. Numerous metabolomic approaches and molecular biological techniques were applied to elucidate potential HIF-dependent effects on the central carbon metabolism of brain cells. In animal and cell models of ischemic stroke, we analysed whether HIF-dependent metabolic reprogramming influences the susceptibility to ischemic injury.

Results: Neuron-specific gene ablation of prolyl-4-hydroxylase domain 2 (PHD2) protein, negatively regulating the protein stability of HIF-α in an oxygen dependent manner, reduced brain injury and functional impairment of mice after acute stroke in a HIF-dependent manner. Accordingly, PHD2 deficient neurons showed an improved tolerance towards ischemic stress in vitro, which was accompanied by enhanced HIF-1-mediated glycolytic lactate production through pyruvate dehydrogenase kinase-mediated inhibition of the pyruvate dehydrogenase. Systemic treatment of mice with roxadustat, a low-molecular weight pan-PHD inhibitor, not only increased the abundance of numerous metabolites of the central carbon and amino acid metabolism in murine brain, but also ameliorated cerebral tissue damage and sensorimotor dysfunction after acute ischemic stroke. In neurons and astrocytes roxadustat provoked a HIF-1-dependent glucose metabolism reprogramming including elevation of glucose uptake, glycogen synthesis, glycolytic capacity, lactate production and lactate release, which enhanced the ischemic tolerance of astrocytes, but not neurons. We found that strong activation of HIF-1 in neurons by non-selective inhibition of all PHD isoenzymes caused a HIF-1-dependent upregulation of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 redirecting glucose-6-phosphate from pentose phosphate pathway (PPP) to the glycolysis pathway. This was accompanied by a reduction of NADPH production in the PPP, which further decreased the low intrinsic antioxidant reserve of neurons, making them more susceptible to ischemic stress. Nonetheless, in organotypic hippocampal cultures with preserved neuronal-glial interactions roxadustat decreased the neuronal susceptibility to ischemic stress, which was largely prevented by restricting glycolytic energy production through lactate transport blockade.

Conclusion: Collectively, our results indicate that HIF-1-mediated metabolic reprogramming alleviates the intrinsic vulnerability of brain-resident cells to ischemic stress.

Keywords: HIF, PHD, Roxadustat, ischemic stroke, metabolic reprogramming, aerobic glycolysis


Citation styles

APA
Madai, S., Kilic, P., Schmidt, R.M., Bas-Orth, C., Korff, T., Büttner, M., Klinke, G., Poschet, G., Marti, H.H., Kunze, R. (2024). Activation of the hypoxia-inducible factor pathway protects against acute ischemic stroke by reprogramming central carbon metabolism. Theranostics, 14(7), 2856-2880. https://doi.org/10.7150/thno.88223.

ACS
Madai, S.; Kilic, P.; Schmidt, R.M.; Bas-Orth, C.; Korff, T.; Büttner, M.; Klinke, G.; Poschet, G.; Marti, H.H.; Kunze, R. Activation of the hypoxia-inducible factor pathway protects against acute ischemic stroke by reprogramming central carbon metabolism. Theranostics 2024, 14 (7), 2856-2880. DOI: 10.7150/thno.88223.

NLM
Madai S, Kilic P, Schmidt RM, Bas-Orth C, Korff T, Büttner M, Klinke G, Poschet G, Marti HH, Kunze R. Activation of the hypoxia-inducible factor pathway protects against acute ischemic stroke by reprogramming central carbon metabolism. Theranostics 2024; 14(7):2856-2880. doi:10.7150/thno.88223. https://www.thno.org/v14p2856.htm

CSE
Madai S, Kilic P, Schmidt RM, Bas-Orth C, Korff T, Büttner M, Klinke G, Poschet G, Marti HH, Kunze R. 2024. Activation of the hypoxia-inducible factor pathway protects against acute ischemic stroke by reprogramming central carbon metabolism. Theranostics. 14(7):2856-2880.

This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
Popup Image