CSF MicroRNAs Reveal Impairment of Angiogenesis and Autophagy in Parkinson Disease

Alan J Fowler, Jaeil Ahn, Michaeline Hebron, Timothy Chiu, Reem Ayoub, Sanjana Mulki, Habtom Ressom, Yasar Torres-Yaghi, Barbara Wilmarth, Fernando L Pagan, Charbel Moussa, Alan J Fowler, Jaeil Ahn, Michaeline Hebron, Timothy Chiu, Reem Ayoub, Sanjana Mulki, Habtom Ressom, Yasar Torres-Yaghi, Barbara Wilmarth, Fernando L Pagan, Charbel Moussa

Abstract

Background and objectives: We assessed longitudinal changes in CSF microRNAs (miRNAs) in patients with moderately severe Parkinson disease.

Methods: We used next-generation whole-genome miRNA sequencing to determine CSF miRNA expression in 75 patients with Parkinson disease after single random ascending doses of nilotinib and longitudinal miRNA expression after daily nilotinib, 150 and 300 mg, vs placebo for 1 year.

Results: Significant changes in the expression of miRNAs that control genes and pathways that regulate angiogenesis, autophagy, and the blood-brain-barrier components, primarily collagen, were observed over 1 year, suggesting impairment of these pathways in Parkinson progression in these patients. Different miRNAs that indicate activation of genes associated with autophagy flux and clearance and angiogenesis were significantly altered in the nilotinib, 300 mg vs 150 mg, or placebo group, and these changes correlated with clinical outcomes. No changes were observed in miRNAs after a single dose of nilotinib vs placebo.

Discussion: This study suggests vascular and autophagy defects in Parkinson progression. Nilotinib, 300 mg, reverses these effects via alteration of miRNA expression, suggesting epigenomic changes that may underlie long-term disease-modifying effects.

Trial registration information: Clinical trial registration number: NCT02954978.

Copyright © 2021 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the American Academy of Neurology.

Figures

Figure 1. Cross-sectional Analysis of miRNA After…
Figure 1. Cross-sectional Analysis of miRNA After a Single Dose of Nilotinib
(A) Schematic of the clinical trial design. This schematic was created using BioRender.com. (B) Principal component analysis of miRNA expression in the single-dose study for treatment vs untreated effects with placebo- (red) and nilotinib- (blue) treated populations and (C) placebo (red), 150 mg (blue), 200 mg (green), 300 mg (purple), and 400 mg (orange) nilotinib-treated populations. Principal component analysis classified 63 participants into groups using 2,559 miRNAs. PC1 and PC2 are depicted.
Figure 2. Longitudinal MicroRNA Expression
Figure 2. Longitudinal MicroRNA Expression
(A) Placebo, 150 mg nilotinib, and 300 mg nilotinib volcano plot of longitudinal miRNA expression in patients with Parkinson disease who underwent placebo treatment. Expression data are plotted as -log10 (p-value) (y-axis) vs log (fold-change) (x-axis). Light colors indicate p < 0.05 for the Wald test, and dark colors indicate that miRNA was also p < 0.05, according to mixed-effects analysis of variance (ME-ANOVA) with the Kruskal-Wallis test. Blue indicates a decrease in expression, and red indicates an increase in expression. The number of miRNAs that meet both the Wald test and ME-ANOVA with the Kruskal-Wallis test (p < 0.05) is shown in the top corners. (B) Heatmap of fold-regulation (FR = FC-1) for miRNAs (p < 0.05) that were in common between the Wald test and Kruskal-Wallis test. ME-ANOVA *p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001. miRNA that displays an effect of treatment (expression directionality for nilotinib is opposite to placebo) is outlined by dotted line and shown to the right of the heatmap. Tyrosine kinase, autophagy, and angiogenesis predicted gene targets were determined via Ingenuity Pathway Analysis software (Qiagen) with confidence filtering to include only highly predicted gene targets. Gene targets with additional experimental confirmation are indicated by ‡. Dashed lines indicate decreased miRNA expression leading to disinhibition of the gene targets, whereas bold line inhibition indicates increased inhibition of the gene targets.
Figure 3. Pathway and GO Functional Analysis…
Figure 3. Pathway and GO Functional Analysis Demonstrate Angiogenesis and BBB Breakdown Associated With Placebo and Inflammation and Autophagy Associated With 300 mg Nilotinib
(A) KEGG (light blue), Reactome (pink), and WikiPathway (light purple) enrichment analysis for placebo differentially expressed miRNAs (DEMs). Enriched pathways were determined using the g:Profiler online tool with Benjamini-Hochberg multiple testing for significance (p < 0.05). (B) Gene ontology (GO) molecular function (green), biological process (orange), and cellular component (purple) enrichment analysis for placebo DEMs. (C) KEGG (light blue), Reactome (pink), and WikiPathway (light purple) enrichment analysis for placebo DEMs. Enriched pathways were determined using the g:Profiler online tool with Benjamini-Hochberg multiple testing for significance (p < 0.05). (D) GO molecular function (green), biological process (orange), and cellular component (purple) enrichment analysis for placebo DEMs. The bubble plot x-axis represents the –log10(Padj), and the size of the bubble corresponds to how many input genes intersect with the associated pathway or GO term.
Figure 4. Correlating Longitudinal miRNA Expression to…
Figure 4. Correlating Longitudinal miRNA Expression to UPDRS II + III
(A) The sum of Unified Parkinson's Disease Rating Score (UPDRS) II (motor) and III (activities of daily living) and at 0, 12, and 27 months ±SD. The difference in scores is shown below in the table. (B) Heatmap of the correlation between 12-month and longitudinal changes in UPDRS II-III and longitudinal changes in miRNAs were computed using Spearman rank correlations. Placebo group miRNAs that highly correlate with UPDRS II + III (r > 0.5, p < 0.05) are outlined by a dotted line and are shown to the right. Twenty-seven–month correlation when placebo was transitioned to nilotinib, 150 or 300 mg, for the second year (right). (C) KEGG, Reactome, and WikiPathway enrichment analysis of gene regulation by miRNAs that highly correlate with UPDRS II + III in placebo.
Figure 5. Summary of Mechanism for miRNA…
Figure 5. Summary of Mechanism for miRNA in Parkinson Disease and Response to DDR1 Inhibition
(A) Discoidin domain receptor1 (DDR1) is overactivated, and several miRNAs, including hsa-miR-30b-5p, hsa-miR-7162-5p, and hsa-miR-4252, regulate several key Parkinson disease-associated pathologies, including blood-brain barrier (BBB) degradation, neuroinflammation, and impaired early autophagy. These pathologies may then promote DDR1 activity, creating a negative feedback loop. When DDR1 is inhibited, via nilotinib, 300 mg, we find that the miRNA differentially expressed miRNAs (DEMs) in placebo are reversed. In addition, we find that miRNAs, including hsa-miR-15a-5p, hsa-miR-5195, and hsa-miR182-5p, positively affect BBB maintenance, transport of glucose across the BBB, and bioenergetics, and restore autophagic flux. This schematic was created using BioRender.com.

References

    1. Fowler AJ, Hebron M, Balaraman K, et al. . Discoidin Domain Receptor 1 is a therapeutic target for neurodegenerative diseases. Hum Mol Genet. 2020;29(17):2882-2898.
    1. Lonskaya I, Hebron ML, Algarzae NK, Desforges N, Moussa CE. Decreased parkin solubility is associated with impairment of autophagy in the nigrostriatum of sporadic Parkinson's disease. Neuroscience. 2013;232:90-105.
    1. Lonskaya I, Shekoyan AR, Hebron ML, Desforges N, Algarzae NK, Moussa CE. Diminished parkin solubility and co-localization with intraneuronal amyloid-beta are associated with autophagic defects in Alzheimer's disease. J Alzheimers Dis. 2013;33(1):231-247.
    1. Hebron M, Peyton M, Liu X, et al. . Discoidin domain receptor inhibition reduces neuropathology and attenuates inflammation in neurodegeneration models. J Neuroimmunol. 2017;311(1):1-9.
    1. Vogel W, Brakebusch C, Fassler R, Alves F, Ruggiero F, Pawson T. Discoidin domain receptor 1 is activated independently of beta(1) integrin. J Biol Chem. 2000;275(8):5779-5784.
    1. Zhu M, Xing D, Lu Z, et al. . DDR1 may play a key role in destruction of the blood-brain barrier after cerebral ischemia-reperfusion. Neurosci Res. 2015;96:14-19.
    1. Fowler AJ, Hebron M, Missner AA, et al. . Multikinase Abl/DDR/Src inhibition produces optimal effects for tyrosine kinase inhibition in neurodegeneration. Drugs R D 2019;19(2):149-166.
    1. Hebron ML, Lonskaya I, Moussa CE. Tyrosine kinase inhibition facilitates autophagic SNCA/alpha-synuclein clearance. Autophagy. 2013;9(8):1249-1250.
    1. Hebron ML, Lonskaya I, Moussa CE. Nilotinib reverses loss of dopamine neurons and improves motor behavior via autophagic degradation of alpha-synuclein in Parkinson's disease models. Hum Mol Genet. 2013;22(16):3315-3328.
    1. Lonskaya I, Desforges NM, Hebron ML, Moussa CE. Ubiquitination increases parkin activity to promote autophagic alpha-synuclein clearance. Plos One. 2013;8(12):e83914.
    1. Lonskaya I, Hebron M, Desforges NM, Franjie A, Moussa CE. Tyrosine kinase inhibition increases functional parkin-Beclin-1 interaction and enhances amyloid clearance and cognitive performance. EMBO Mol Med. 2013;5(8):1247-1262.
    1. Lonskaya I, Hebron M, Desforges NM, Schachter JB, Moussa CE. Nilotinib-induced autophagic changes increase endogenous parkin level and ubiquitination, leading to amyloid clearance. J Mol Med. 2014;92(4):373-386.
    1. Hebron ML, Javidnia M, Moussa CE. Tau clearance improves astrocytic function and brain glutamate-glutamine cycle. J Neurol Sci. 2018;391:90-99.
    1. Pagan F, Hebron M, Valadez EH, et al. . Nilotinib effects in Parkinson's disease and dementia with Lewy bodies. J Parkinsons Dis. 2016;6(3):503-517.
    1. Pagan FL, Hebron ML, Wilmarth B, et al. . Nilotinib effects on safety, tolerability, and potential biomarkers in Parkinson disease: a phase 2 randomized clinical trial. JAMA Neurol. 2019;77(3):309-317.
    1. Pagan FL, Hebron ML, Wilmarth B, et al. . Pharmacokinetics and pharmacodynamics of a single dose Nilotinib in individuals with Parkinson's disease. Pharmacol Res Perspect. 2019;7(2):e00470.
    1. Deremer DL, Ustun C, Natarajan K. Nilotinib: a second-generation tyrosine kinase inhibitor for the treatment of chronic myelogenous leukemia. Clin Ther. 2008;30(11):1956-1975.
    1. Jeitany M, Leroy C, Tosti P, et al. . Inhibition of DDR1-BCR signalling by nilotinib as a new therapeutic strategy for metastatic colorectal cancer. EMBO Mol Med. 2018;10(4):e7918.
    1. Day E, Waters B, Spiegel K, et al. . Inhibition of collagen-induced discoidin domain receptor 1 and 2 activation by imatinib, nilotinib and dasatinib. Eur J Pharmacol. 2008;599(1-3):44-53.
    1. Manley PW, Drueckes P, Fendrich G, et al. . Extended kinase profile and properties of the protein kinase inhibitor nilotinib. Biochim Biophys Acta. 2010;1804(3):445-453.
    1. Pagan FL, Wilmarth B, Torres-Yaghi Y, et al. . Long-term safety and clinical effects of nilotinib in Parkinson's disease. Mov Disord. 2021;36(3):740-749.
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15(12):550.
    1. Tarazona S, Furio-Tari P, Turra D, et al. . Data quality aware analysis of differential expression in RNA-seq with NOISeq R/Bioc package. Nucleic Acids Res. 2015;43(21):e140.
    1. Mi H, Muruganujan A, Huang X, et al. . Protocol Update for large-scale genome and gene function analysis with the PANTHER classification system (v.14.0). Nat Protoc. 2019;14(3):703-721.
    1. Orr M, Liu P. Sample size estimation while controlling false discovery rate for microarray experiments using ssize.fdr package. R J. 2009;1:47-53.
    1. Chou CH, Shrestha S, Yang CD, et al. . miRTarBase update 2018: a resource for experimentally validated microRNA-target interactions. Nucleic Acids Res. 2018;46(D1):D296-D302.
    1. Klionsky DJ, Abdelmohsen K, Abe A, et al. . Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition). Autophagy. 2016;12(1):1-222.
    1. Fowler AJ, Moussa CE. Activating autophagy as a therapeutic strategy for Parkinson's disease. CNS Drugs. 2018;32(1):1-11.
    1. Eskelinen EL. Roles of LAMP-1 and LAMP-2 in lysosome biogenesis and autophagy. Mol Aspects Med. 2006;27(5-6):495-502.
    1. Saftig P, Beertsen W, Eskelinen EL. LAMP-2: a control step for phagosome and autophagosome maturation. Autophagy. 2008;4(4):510-512.
    1. Liu X, Hebron M, Shi W, Lonskaya I, Moussa CE. Ubiquitin specific protease-13 independently regulates parkin ubiquitination and alpha-synuclein clearance in alpha-synucleinopathies. Hum Mol Genet. 2019;28(4):548-560.
    1. Ngai D, Lino M, Rothenberg KE, Simmons CA, Fernandez-Gonzalez R, Bendeck MP. DDR1 (discoidin domain receptor-1)-RhoA (ras homolog family member A) axis senses matrix stiffness to promote vascular calcification. Arterioscler Thromb Vasc Biol. 2020;40(7):1763-1776.
    1. Jing H, Song J, Zheng J. Discoidin domain receptor 1: new star in cancer-targeted therapy and its complex role in breast carcinoma. Oncol Lett. 2018;15(3):3403-3408.
    1. Munoz DG, Woulfe JM. Angiogenesis: a new paradigm for Parkinson disease with practical and pathogenic implications. Neurology. 2015;85(21):1826-1827.
    1. Vogel W, Gish GD, Alves F, Pawson T. The discoidin domain receptor tyrosine kinases are activated by collagen. Mol Cel. 1997;1(1):13-23.
    1. Shrivastava A, Radziejewski C, Campbell E, et al. . An orphan receptor tyrosine kinase family whose members serve as nonintegrin collagen receptors. Mol Cel. 1997;1(1):25-34.
    1. Ratzka A, Baron O, Grothe C. FGF-2 deficiency does not influence FGF ligand and receptor expression during development of the nigrostriatal system. PLoS One. 2011;6(8):e23564.
    1. Timmer M, Cesnulevicius K, Winkler C, et al. . Fibroblast growth factor (FGF)-2 and FGF receptor 3 are required for the development of the substantia nigra, and FGF-2 plays a crucial role for the rescue of dopaminergic neurons after 6-hydroxydopamine lesion. J Neurosci. 2007;27(3):459-471.
    1. Barcia C, Bautista V, Sanchez-Bahillo A, et al. . Changes in vascularization in substantia nigra pars compacta of monkeys rendered parkinsonian. J Neural Transm (Vienna). 2005;112(9):1237-1248.
    1. Turner RS, Hebron ML, Lawler A, et al. . Nilotinib effects on safety, tolerability, and biomarkers in Alzheimer's disease. Ann Neurol. 2020;88(1):183-194.
    1. Desai Bradaric B, Patel A, Schneider JA, Carvey PM, Hendey B. Evidence for angiogenesis in Parkinson's disease, incidental Lewy body disease, and progressive supranuclear palsy. J Neural Transm (Vienna). 2012;119(1):59-71.
    1. Franco C, Hou G, Ahmad PJ, et al. . Discoidin domain receptor 1 (ddr1) deletion decreases atherosclerosis by accelerating matrix accumulation and reducing inflammation in low-density lipoprotein receptor-deficient mice. Circ Res. 2008;102(10):1202-1211.
    1. Janelidze S, Lindqvist D, Francardo V, et al. . Increased CSF biomarkers of angiogenesis in Parkinson disease. Neurology. 2015;85(21):1834-1842.
    1. Raz L, Knoefel J, Bhaskar K. The neuropathology and cerebrovascular mechanisms of dementia. J Cereb Blood Flow Metab. 2016;36(1):172-186.
    1. Rosenberg GA. Matrix metalloproteinases and their multiple roles in neurodegenerative diseases. Lancet Neurol. 2009;8(2):205-216.
    1. Oh S, Seo M, Choi JS, Joo CK, Lee SK. MiR-199a/b-5p inhibits lymphangiogenesis by targeting discoidin domain receptor 1 in corneal injury. Mol Cell. 2018;41(2):93-102.
    1. Stamatovic SM, Keep RF, Andjelkovic AV. Brain endothelial cell-cell junctions: how to “open” the blood brain barrier. Curr Neuropharmacol. 2008;6(3):179-192.
    1. Leitinger B, Saltel F. Discoidin domain receptors: multitaskers for physiological and pathological processes. Cell Adh Migr. 2018;12(4):398-399.
    1. Roberts ME, Magowan L, Hall IP, Johnson SR. Discoidin domain receptor 1 regulates bronchial epithelial repair and matrix metalloproteinase production. Eur Respir J. 2011;37(6):1482-1493.
    1. Dorison A, Dussaule JC, Chatziantoniou C. The role of discoidin domain receptor 1 in inflammation, fibrosis and renal disease. Nephron. 2017;137(3):212-220.

Source: PubMed

3
구독하다