Betulinic Acid Hydroxamate is Neuroprotective and Induces Protein Phosphatase 2A-Dependent HIF-1α Stabilization and Post-transcriptional Dephosphorylation of Prolyl Hydrolase 2

Elsevier BV - Tập 18 - Trang 1849-1861 - 2021
María E. Prados1, Alejandro Correa-Sáez2,3,4, Juan D. Unciti-Broceta1, Martín Garrido-Rodríguez2,3,4, Carla Jimenez-Jimenez2,3,4, Massimiliano Mazzone5, Alberto Minassi6, Giovanni Appendino6, Marco A. Calzado2,3,4, Eduardo Muñoz2,3,4
1Emerald Health Biotechnology, Cordoba, Spain
2Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain
3Department of Cellular Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
4Hospital Universitario Hospital Reina Sofia, Cordoba, Spain
5Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB-KULeuven, Leuven, Belgium
6Department of Drug Science, University of Piemonte Orientale, Novara, Italy

Tóm tắt

Huntington’s disease (HD) is a neurodegenerative disorder characterized by unwanted choreatic movements, behavioral and psychiatric disturbances, and dementia. The activation of the hypoxic response pathway through the pharmacological inhibition of hypoxia-inducing factor (HIF) prolyl-hydroxylases (PHDs) is a promising approach for neurodegenerative diseases, including HD. Herein, we have studied the mechanism of action of the compound Betulinic acid hydroxamate (BAH), a hypoximimetic derivative of betulinic acid, and its efficacy against striatal neurodegeneration using complementary approaches. Firstly, we showed the molecular mechanisms through which BAH modifies the activity of the PHD2 prolyl hydroxylase, thus directly affecting HIF-1α stability. BAH treatment reduces PHD2 phosphorylation on Ser-125 residue, responsible for the control of its hydrolase activity. HIF activation by BAH is inhibited by okadaic acid and LB-100 indicating that a protein phosphatase 2A (PP2A) is implicated in the mechanism of action of BAH. Furthermore, in striatal cells bearing a mutated form of the huntingtin protein, BAH stabilized HIF-1α protein, induced Vegf and Bnip3 gene expression and protected against mitochondrial toxin-induced cytotoxicity. Pharmacokinetic analyses showed that BAH has a good brain penetrability and experiments performed in a mouse model of striatal neurodegeneration induced by 3-nitropropionic acid showed that BAH improved the clinical symptoms. In addition, BAH also prevented neuronal loss, decreased reactive astrogliosis and microglial activation, inhibited the upregulation of proinflammatory markers, and improved antioxidant defenses in the brain. Taken together, our results show BAH’s ability to activate the PP2A/PHD2/HIF pathway, which may have important implications in the treatment of HD and perhaps other neurodegenerative diseases.

Tài liệu tham khảo

Piotr, R. and B.-K. Teresa, Natural Triterpenoids and their Derivatives with Pharmacological Activity Against Neurodegenerative Disorders. Mini Rev Org Chem, 2014. 11(3): p. 307-315. Anna Hordyjewska, A.O., Anna Horecka & Jacek Kurzepa Betulin and betulinic acid: triterpenoids derivatives with a powerful biological potential. Phytochemistry Reviews, 2019. 18: p. 929–951. McMillin, M., et al., TGR5 signaling reduces neuroinflammation during hepatic encephalopathy. J Neurochem, 2015. 135(3): p. 565-76. Jiao, S., et al., Betulinic acid protects against cerebral ischemia/reperfusion injury by activating the PI3K/Akt signaling pathway. Biomed Pharmacother, 2016. 84: p. 1533-1537. Wang, D., et al., Betulinic acid protects the neuronal damage in new born rats from isoflurane-induced apoptosis in the developing brain by blocking FASL-FAS signaling pathway. Biomed Pharmacother, 2017. 95: p. 1631-1635. Kaundal, M., et al., Betulinic acid, a natural PDE inhibitor restores hippocampal cAMP/cGMP and BDNF, improve cerebral blood flow and recover memory deficits in permanent BCCAO induced vascular dementia in rats. Eur J Pharmacol, 2018. 832: p. 56-66. Kaundal, M., R. Deshmukh, and M. Akhtar, Protective effect of betulinic acid against intracerebroventricular streptozotocin induced cognitive impairment and neuronal damage in rats: Possible neurotransmitters and neuroinflammatory mechanism. Pharmacol Rep, 2018. 70(3): p. 540-548. Li, C., et al., Inhibitory Effects of Betulinic Acid on LPS-Induced Neuroinflammation Involve M2 Microglial Polarization via CaMKKbeta-Dependent AMPK Activation. Front Mol Neurosci, 2018. 11: p. 98. Xu, S.H., et al., New 30-norlupane derivatives through chemical-microbial semi-synthesis of betulinic acid and their neuroprotective effect. Bioorg Med Chem Lett, 2020. 30(17): p. 127407. Minassi, A., et al., Triterpenoid Hydroxamates as HIF Prolyl Hydrolase Inhibitors. J Nat Prod, 2018. 81(10): p. 2235-2243. Prados, M.E., et al., Betulinic acid hydroxamate prevents colonic inflammation and fibrosis in murine models of inflammatory bowel disease. Acta Pharmacol Sin, 2020. Choudhry, H. and A.L. Harris, Advances in Hypoxia-Inducible Factor Biology. Cell Metab, 2018. 27(2): p. 281-298. Di Conza, G., et al., The mTOR and PP2A Pathways Regulate PHD2 Phosphorylation to Fine-Tune HIF1alpha Levels and Colorectal Cancer Cell Survival under Hypoxia. Cell Rep, 2017. 18(7): p. 1699-1712. Ehling, M., et al., B55alpha/PP2A Limits Endothelial Cell Apoptosis During Vascular Remodeling: A Complementary Approach To Disrupt Pathological Vessels? Circ Res, 2020. 127(6): p. 707-723. Caterino, M., et al., Huntingtin protein: A new option for fixing the Huntington's disease countdown clock. Neuropharmacology, 2018. 135: p. 126-138. Rebec, G.V., Corticostriatal network dysfunction in Huntington's disease: Deficits in neural processing, glutamate transport, and ascorbate release. CNS Neurosci Ther, 2018. 24(4): p. 281-291. Benarroch, E.E., Hypoxia-induced mediators and neurologic disease. Neurology, 2009. 73(7): p. 560-5. Speer, R.E., et al., Hypoxia-inducible factor prolyl hydroxylases as targets for neuroprotection by "antioxidant" metal chelators: From ferroptosis to stroke. Free Radic Biol Med, 2013. 62: p. 26-36. Yang, Y.T., T.C. Ju, and D.I. Yang, Induction of hypoxia inducible factor-1 attenuates metabolic insults induced by 3-nitropropionic acid in rat C6 glioma cells. J Neurochem, 2005. 93(3): p. 513-25. Niatsetskaya, Z., et al., HIF prolyl hydroxylase inhibitors prevent neuronal death induced by mitochondrial toxins: therapeutic implications for Huntington's disease and Alzheimer's disease. Antioxid Redox Signal, 2010. 12(4): p. 435-43. Li, H.S., et al., HIF-1alpha protects against oxidative stress by directly targeting mitochondria. Redox Biol, 2019. 25: p. 101109. Chi, O.Z., et al., Effects of erythropoietin on blood-brain barrier disruption in focal cerebral ischemia. Pharmacology, 2008. 82(1): p. 38-42. Bhattarai, D., X. Xu, and K. Lee, Hypoxia-inducible factor-1 (HIF-1) inhibitors from the last decade (2007 to 2016): A "structure-activity relationship" perspective. Med Res Rev, 2018. 38(4): p. 1404-1442. Trettel, F., et al., Dominant phenotypes produced by the HD mutation in STHdh(Q111) striatal cells. Hum Mol Genet, 2000. 9(19): p. 2799-809. Morrugares, R., et al., Phosphorylation-dependent regulation of the NOTCH1 intracellular domain by dual-specificity tyrosine-regulated kinase 2. Cell Mol Life Sci, 2020. 77(13): p. 2621-2639. Chiva, C., et al., QCloud: A cloud-based quality control system for mass spectrometry-based proteomics laboratories. PLoS One, 2018. 13(1): p. e0189209. Perkins, D.N., et al., Probability-based protein identification by searching sequence databases using mass spectrometry data. Electrophoresis, 1999. 20(18): p. 3551-67. Beer, L.A., et al., Efficient Quantitative Comparisons of Plasma Proteomes Using Label-Free Analysis with MaxQuant. Methods Mol Biol, 2017. 1619: p. 339-352. Teo, G., et al., SAINTexpress: improvements and additional features in Significance Analysis of INTeractome software. J Proteomics, 2014. 100: p. 37-43. Calzado, M.A., et al., An inducible autoregulatory loop between HIPK2 and Siah2 at the apex of the hypoxic response. Nat Cell Biol, 2009. 11(1): p. 85-91. Brouillet, E., et al., Partial inhibition of brain succinate dehydrogenase by 3-nitropropionic acid is sufficient to initiate striatal degeneration in rat. J Neurochem, 1998. 70(2): p. 794-805. Fernagut, P.O., et al., Subacute systemic 3-nitropropionic acid intoxication induces a distinct motor disorder in adult C57Bl/6 mice: behavioural and histopathological characterisation. Neuroscience, 2002. 114(4): p. 1005-17. Ito, D., et al., Enhanced expression of Iba1, ionized calcium-binding adapter molecule 1, after transient focal cerebral ischemia in rat brain. Stroke, 2001. 32(5): p. 1208-15. Chen, S.D., et al., More Insight into BDNF against Neurodegeneration: Anti-Apoptosis, Anti-Oxidation, and Suppression of Autophagy. Int J Mol Sci, 2017. 18(3). Sharp, F.R., et al., Hypoxic preconditioning protects against ischemic brain injury. NeuroRx, 2004. 1(1): p. 26-35. Kim, J.W., et al., HIF-1-mediated expression of pyruvate dehydrogenase kinase: a metabolic switch required for cellular adaptation to hypoxia. Cell Metab, 2006. 3(3): p. 177-85. Deepak Bhatia, M.S.A., Qiwen Shi and Shahrzad Movafagh, Hypoxia and its Emerging Therapeutics in Neurodegenerative, Inflammatory and Renal Diseases, in Hypoxia and Human Diseases, C.Z. Jing Zheng, Editor 2017: Intechopen. Awan, M.U., et al., Neuroprotective role of BNIP3 under oxidative stress through autophagy in neuroblastoma cells. Mol Biol Rep, 2014. 41(9): p. 5729-34. Clark, A.R. and M. Ohlmeyer, Protein phosphatase 2A as a therapeutic target in inflammation and neurodegeneration. Pharmacol Ther, 2019. 201: p. 181-201. Chang, K.H., et al., Plasma inflammatory biomarkers for Huntington's disease patients and mouse model. Brain Behav Immun, 2015. 44: p. 121-7. Dobrogowska, D.H., et al., Increased blood-brain barrier permeability and endothelial abnormalities induced by vascular endothelial growth factor. J Neurocytol, 1998. 27(3): p. 163-73. Sondell, M., G. Lundborg, and M. Kanje, Vascular endothelial growth factor has neurotrophic activity and stimulates axonal outgrowth, enhancing cell survival and Schwann cell proliferation in the peripheral nervous system. J Neurosci, 1999. 19(14): p. 5731-40. Jin, K.L., X.O. Mao, and D.A. Greenberg, Vascular endothelial growth factor rescues HN33 neural cells from death induced by serum withdrawal. J Mol Neurosci, 2000. 14(3): p. 197-203. Ellison, S.M., et al., Dose-dependent neuroprotection of VEGF(1)(6)(5) in Huntington's disease striatum. Mol Ther, 2013. 21(10): p. 1862-75. Bjorkqvist, M., et al., A novel pathogenic pathway of immune activation detectable before clinical onset in Huntington's disease. J Exp Med, 2008. 205(8): p. 1869-77. Palpagama, T.H., et al., The Role of Microglia and Astrocytes in Huntington's Disease. Front Mol Neurosci, 2019. 12: p. 258. Ben Haim, L., et al., Elusive roles for reactive astrocytes in neurodegenerative diseases. Front Cell Neurosci, 2015. 9: p. 278. Franco, R. and D. Fernandez-Suarez, Alternatively activated microglia and macrophages in the central nervous system. Prog Neurobiol, 2015. 131: p. 65-86. Zuccato, C. and E. Cattaneo, Role of brain-derived neurotrophic factor in Huntington's disease. Prog Neurobiol, 2007. 81(5-6): p. 294-330. Ciammola, A., et al., Low brain-derived neurotrophic factor (BDNF) levels in serum of Huntington's disease patients. Am J Med Genet B Neuropsychiatr Genet, 2007. 144B(4): p. 574-7. Islam, M.T., Oxidative stress and mitochondrial dysfunction-linked neurodegenerative disorders. Neurol Res, 2017. 39(1): p. 73-82. Damiano, M., et al., Mitochondria in Huntington's disease. Biochim Biophys Acta, 2010. 1802(1): p. 52-61.