Monocyte biomarkers define sargramostim treatment outcomes for Parkinson's disease

Mai M Abdelmoaty, Jatin Machhi, Pravin Yeapuri, Farah Shahjin, Vikas Kumar, Katherine E Olson, R Lee Mosley, Howard E Gendelman, Mai M Abdelmoaty, Jatin Machhi, Pravin Yeapuri, Farah Shahjin, Vikas Kumar, Katherine E Olson, R Lee Mosley, Howard E Gendelman

Abstract

Background: Dysregulation of innate and adaptive immunity heralds both the development and progression of Parkinson's disease (PD). Deficits in innate immunity in PD are defined by impairments in monocyte activation, function, and pro-inflammatory secretory factors. Each influences disease pathobiology.

Methods and results: To define monocyte biomarkers associated with immune transformative therapy for PD, changes in gene and protein expression were evaluated before and during treatment with recombinant human granulocyte-macrophage colony-stimulating factor (GM-CSF, sargramostim, Leukine® ). Monocytes were recovered after leukapheresis and isolation by centrifugal elutriation, before and 2 and 6 months after initiation of treatment. Transcriptome and proteome biomarkers were scored against clinical motor functions. Pathway enrichments from single cell-RNA sequencing and proteomic analyses from sargramostim-treated PD patients demonstrate a neuroprotective signature, including, but not limited to, antioxidant, anti-inflammatory, and autophagy genes and proteins (LRRK2, HMOX1, TLR2, TLR8, RELA, ATG7, and GABARAPL2).

Conclusions: This monocyte profile provides an "early" and unique biomarker strategy to track clinical immune-based interventions, but requiring validation in larger case studies.

Keywords: GM-CSF; Parkinson's disease; biomarkers; monocytes; proteomics; scRNA-seq.

Conflict of interest statement

The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported.

© 2022 The Authors. Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

Figures

FIGURE 1
FIGURE 1
Pathway enrichment of differentially expressed proteins in monocytes of PD patients at 2 months after sargramostim treatment. (A) Gene ontology (GO)‐term functional enrichment by five categories (immune response, biological process, cellular component, KEGG, and Reactome) performed using Cytoscape in conjunction with the plug‐in ClueGO. (B) Canonical pathway enrichment analysis performed using IPA (Qiagen). Black arrow points to the state of canonical pathways illustrated in Figure 1B; orange colour (activation), blue colour (inhibition), and grey colour (no activity pattern)
FIGURE 2
FIGURE 2
Pathway enrichment of differentially expressed proteins/genes in monocytes at 6 months after sargramostim treatment. (A) GO‐term functional enrichment by five categories (immune response, biological process, cellular component, KEGG, and Reactome) was performed using Cytoscape in conjunction with the plug‐in ClueGO. (B) Canonical pathway enrichment analysis was performed using IPA (Qiagen). Black arrows point to the state of canonical pathways illustrated in Figure (B); orange colour (activation), blue colour (inhibition), and grey colour (no activity pattern)
FIGURE 3
FIGURE 3
Gene and protein expression of potential biomarkers in monocytes at 2 and 6 months after sargramostim treatment. The ddPCR assay was performed to determine the gene expression of LRRK2, HMOX1, TLR2, TLR8, RELA, ATG7, and GABARAPL2 at 2 (A) and 6 (B) months after starting the sargramostim treatment compared to baseline. Gene expression was normalized to HPRT1, and the ddPCR assay was performed four times (n = 4 technical replicates). Western blot analysis was performed to determine the protein expression of β‐actin, LRRK2, HMOX1, TLR2, TLR8, RELA, ATG7, and GABARAPL2 at 2 (C) and 6 (D) months after starting the sargramostim treatment compared to baseline. Protein expression was normalized to β‐actin and densitometric quantification is shown. Western blot analysis was done thrice (n = 3 technical replicates). Data represent mean ± SD. Horizontal line in each image represents baseline expression; values above the line indicate upregulation, while values below the line indicate downregulation
FIGURE 4
FIGURE 4
Prediction of UPDRS III score by gene expression of potential biomarkers. (A) Correlation between genetic expression of LRRK2, HMOX1, TLR2, and ATG7 and change in UPDRS III score. (B) Correlation between genetic expression of LRRK2, TLR2, and ATG7 and raw UPDRS III score. (C) Multiple linear regression analysis of effect of genetic expression of LRRK2, HMOX1, TLR2, TLR8, and ATG7 on change in UPDRS III score. (D) Multiple linear regression analysis of effect of genetic expression of LRRK2, HMOX1, TLR2, TLR8, and ATG7 on raw UPDRS III score. (A,B) r = Pearson product‐moment correlation coefficient. (C,D) r = regression coefficient. p ≤ 0.05 was considered significant
FIGURE 5
FIGURE 5
Prediction of UPDRS III score by protein expression of potential biomarkers. (A) Correlation between protein expression of LRRK2, RELA, and ATG7 and change in UPDRS III score. (B) Correlation between protein expression of LRRK2, TLR2, and ATG7 and raw UPDRS III score. (C) Multiple linear regression analysis of effect of protein expression of LRRK2, HMOX1, RELA, and GABARAPL2 on change in UPDRS III score. (D) Multiple linear regression analysis of effect of protein expression of TLR2, TLR8, and ATG7 on raw UPDRS III score. (A,B) r = Pearson product‐moment correlation coefficient. (C,D) r = regression coefficient. p ≤ 0.05 was considered significant

References

    1. Jain S. Multi‐organ autonomic dysfunction in Parkinson disease. Parkinsonism Relat Disord. 2011;17(2):77‐83.
    1. Houser MC, Tansey MG. The gut‐brain axis: is intestinal inflammation a silent driver of Parkinson's disease pathogenesis? NPJ Parkinsons Dis. 2017. ; 3:3.
    1. Nissen SK, Shrivastava K, Schulte C, et al. Alterations in blood monocyte functions in Parkinson's disease. Mov Disord 2019;34(11):1711‐1721.
    1. Brochard V, Combadiere B, Prigent A, et al. Infiltration of CD4+ lymphocytes into the brain contributes to neurodegeneration in a mouse model of Parkinson disease. J Clin Invest. 2009;119(1):182‐192.
    1. Rocha EM, De Miranda B, Sanders LH. Alpha‐synuclein: pathology, mitochondrial dysfunction and neuroinflammation in Parkinson's disease. Neurobiol Dis. 2018;109(Pt B):249‐257.
    1. Jankovic J, Tan EK. Parkinson's disease: etiopathogenesis and treatment. J Neurol Neurosurg Psychiatry. 2020;91(8):795‐808.
    1. Harms AS, Thome AD, Yan Z, et al. Peripheral monocyte entry is required for alpha‐synuclein induced inflammation and neurodegeneration in a model of Parkinson disease. Exp Neurol. 2018;300:179‐187.
    1. Waschbisch A, Schroder S, Schraudner D, et al. Pivotal role for CD16+ monocytes in immune surveillance of the central nervous system. J Immunol. 2016;196(4):1558‐1567.
    1. Thome AD, Atassi F, Wang J, et al. Ex vivo expansion of dysfunctional regulatory T lymphocytes restores suppressive function in Parkinson's disease. NPJ Parkinsons Dis. 2021;7(1):41.
    1. Machhi J, Kevadiya BD, Muhammad IK, et al. Harnessing regulatory T cell neuroprotective activities for treatment of neurodegenerative disorders. Mol Neurodegener. 2020;15(1):32.
    1. Schabitz WR, Kruger C, Pitzer C, et al. A neuroprotective function for the hematopoietic protein granulocyte‐macrophage colony stimulating factor (GM‐CSF). J Cereb Blood Flow Metab. 2008;28(1):29‐43.
    1. Schutt CR, Gendelman HE, Mosley RL. Tolerogenic bone marrow‐derived dendritic cells induce neuroprotective regulatory T cells in a model of Parkinson's disease. Mol Neurodegener. 2018;13(1):26.
    1. Gendelman HE, Zhang Y, Santamaria P, et al. Evaluation of the safety and immunomodulatory effects of sargramostim in a randomized, double‐blind phase 1 clinical Parkinson's disease trial. NPJ Parkinsons Dis. 2017;3:10.
    1. Olson KE, Namminga KL, Lu Y, et al. Safety, tolerability, and immune‐biomarker profiling for year‐long sargramostim treatment of Parkinson's disease. EBioMedicine. 2021;67:103380.
    1. Kosloski LM, Kosmacek EA, Olson KE, Mosley RL, Gendelman HE. GM‐CSF induces neuroprotective and anti‐inflammatory responses in 1‐methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine intoxicated mice. J Neuroimmunol 2013;265(1‐2):1‐10.
    1. Olson KE, Namminga KL, Schwab AD, et al. Neuroprotective activities of long‐acting granulocyte‐macrophage colony‐stimulating factor (mPDM608) in 1‐methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine‐intoxicated mice. Neurotherapeutics. 2020;17(4):1861‐1877.
    1. Mosley RL, Lu Y, Olson KE, et al. A synthetic agonist to vasoactive intestinal peptide receptor‐2 induces regulatory T cell neuroprotective activities in models of Parkinson's disease. Front Cell Neurosci. 2019;13:421.
    1. Zheng SG, Wang JH, Koss MN, Quismorio F, Jr. , Gray JD, Horwitz DA. CD4+ and CD8+ regulatory T cells generated ex vivo with IL‐2 and TGF‐beta suppress a stimulatory graft‐versus‐host disease with a lupus‐like syndrome. J Immunol. 2004;172(3):1531‐1539.
    1. Cook AD, Louis C, Robinson MJ, Saleh R, Sleeman MA, Hamilton JA. Granulocyte macrophage colony‐stimulating factor receptor alpha expression and its targeting in antigen‐induced arthritis and inflammation. Arthritis Res Ther. 2016;18(1):287.
    1. Hamilton JA, Cook AD, Tak PP. Anti‐colony‐stimulating factor therapies for inflammatory and autoimmune diseases. Nat Rev Drug Discov. 2016;16(1):53‐70.
    1. Potter H, Woodcock JH, Boyd TD, et al. Safety and efficacy of sargramostim (GM‐CSF) in the treatment of Alzheimer's disease. Alzheimers Dement (N Y) 2021;7(1):e12158.
    1. Lotankar S, Prabhavalkar KS, Bhatt LK. Biomarkers for Parkinson's disease: recent advancement. Neurosci Bull. 2017;33(5):585‐597.
    1. Arainga M, Guo D, Wiederin J, Ciborowski P, McMillan J, Gendelman HE. Opposing regulation of endolysosomal pathways by long‐acting nanoformulated antiretroviral therapy and HIV‐1 in human macrophages. Retrovirology 2015;12:5.
    1. Gao L, Kumar V, Vellichirammal NN, et al. Functional, proteomic and bioinformatic analyses of Nrf2‐ and Keap1‐ null skeletal muscle. J Physiol. 2020;598(23):5427‐5451.
    1. Benjamini Y, Drai D, Elmer G, Kafkafi N, Golani I. Controlling the false discovery rate in behavior genetics research. Behav Brain Res. 2001;125(1‐2):279‐284.
    1. Bindea G, Mlecnik B, Hackl H, et al. ClueGO: a Cytoscape plug‐in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics 2009;25(8):1091‐1093.
    1. Gaurav R, Mikuls TR, Thiele GM, et al. High‐throughput analysis of lung immune cells in a combined murine model of agriculture dust‐triggered airway inflammation with rheumatoid arthritis. PLoS One 2021;16(2):e0240707.
    1. Benjamini Y, Krieger AM, Yekutieli D. Adaptive linear step‐up procedures that control the false discovery rate. Biometrika 2006;93(3):491‐507.
    1. Xu E, Boddu R, Abdelmotilib HA, et al. Pathological alpha‐synuclein recruits LRRK2 expressing pro‐inflammatory monocytes to the brain. Mol Neurodegener. 2022;17(1):7.
    1. Grozdanov V, Bousset L, Hoffmeister M, et al. Increased immune activation by pathologic alpha‐synuclein in Parkinson's disease. Ann Neurol 2019;86(4):593‐606.
    1. Lee IH. Mechanisms and disease implications of sirtuin‐mediated autophagic regulation. Exp Mol Med. 2019;51(9):1‐11.
    1. Clarke CJ, Hales A, Hunt A, Foxwell BM. IL‐10‐mediated suppression of TNF‐alpha production is independent of its ability to inhibit NF kappa B activity. Eur J Immunol. 1998;28(5):1719‐1726.
    1. Dickensheets HL, Donnelly RP. IFN‐gamma and IL‐10 inhibit induction of IL‐1 receptor type I and type II gene expression by IL‐4 and IL‐13 in human monocytes. J Immunol. 1997;159(12):6226‐33.
    1. Nibbs RJ, Graham GJ. Immune regulation by atypical chemokine receptors. Nat Rev Immunol. 2013;13(11):815‐829.
    1. Li RY, Guan J, Zhou S. Boosting scRNA‐seq data clustering by cluster‐aware feature weighting. BMC Bioinformatics. 2021;22(suppl 6):130.
    1. Beyer M, Mallmann MR, Xue J, et al. High‐resolution transcriptome of human macrophages. PLoS One 2012;7(9):e45466.
    1. Alonso MN, Gregorio JG, Davidson MG, Gonzalez JC, Engleman EG. Depletion of inflammatory dendritic cells with anti‐CD209 conjugated to saporin toxin. Immunol Res. 2014;58(2‐3):374‐377.
    1. Xiong J. Atg7 in development and disease: panacea or Pandora's box? Protein Cell. 2015;6(10):722‐734.
    1. Szalai P, Hagen LK, Saetre F, et al. Autophagic bulk sequestration of cytosolic cargo is independent of LC3, but requires GABARAPs. Exp Cell Res. 2015;333(1):21‐38.
    1. Yamada Y, Suzuki NN, Hanada T, et al. The crystal structure of Atg3, an autophagy‐related ubiquitin carrier protein (E2) enzyme that mediates Atg8 lipidation. J Biol Chem. 2007;282(11):8036‐8043.
    1. Collaborators, G. B. D. N. Global, regional, and national burden of neurological disorders, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019;18(5):459‐480.
    1. Ganguly U, Singh S, Pal S, et al. Alpha‐synuclein as a biomarker of Parkinson's disease: good, but not good enough. Front Aging Neurosci. 2021;13:702639.
    1. Verber NS, Shepheard SR, Sassani M, et al. Biomarkers in motor neuron disease: a state of the art review. Front Neurol. 2019;10:291.
    1. Niethammer M, Feigin A, Eidelberg D. Functional neuroimaging in Parkinson's disease. Cold Spring Harb Perspect Med. 2012;2(5):a009274.
    1. Rahmim A, Zaidi H. PET versus SPECT: strengths, limitations and challenges. Nucl Med Commun. 2008;29(3):193‐207.
    1. Schlachetzki JCM, Prots I, Tao J, et al. Author correction: a monocyte gene expression signature in the early clinical course of Parkinson's disease. Sci Rep. 2020;10(1):6261.
    1. Gao L, Tang H, Nie K, et al. Cerebrospinal fluid alpha‐synuclein as a biomarker for Parkinson's disease diagnosis: a systematic review and meta‐analysis. Int J Neurosci. 2015;125(9):645‐654.
    1. van Dijk KD, Bidinosti M, Weiss A, Raijmakers P, Berendse HW, van de Berg WD. Reduced alpha‐synuclein levels in cerebrospinal fluid in Parkinson's disease are unrelated to clinical and imaging measures of disease severity. Eur J Neurol. 2014;21(3):388‐394.
    1. Gustafsson G, Loov C, Persson E, et al. Secretion and uptake of alpha‐synuclein via extracellular vesicles in cultured cells. Cell Mol Neurobiol 2018;38(8):1539‐1550.
    1. Schaeffer E, Kluge A, Bottner M, et al. Alpha synuclein connects the gut‐brain axis in Parkinson's disease patients ‐ a view on clinical aspects, cellular pathology and analytical methodology. Front Cell Dev Biol. 2020;8:573696.
    1. Campo F, Carletti R, Fusconi M, et al. Alpha‐synuclein in salivary gland as biomarker for Parkinson's disease. Rev Neurosci 2019;30(5):455‐462.
    1. Kim JY, Illigens BM, McCormick MP, Wang N, Gibbons CH. Alpha‐synuclein in skin nerve fibers as a biomarker for alpha‐synucleinopathies. J Clin Neurol. 2019;15(2):135‐142.
    1. Ortuno‐Lizaran I, Beach TG, Serrano GE, Walker DG, Adler CH, Cuenca N. Phosphorylated alpha‐synuclein in the retina is a biomarker of Parkinson's disease pathology severity. Mov Disord. 2018;33(8):1315‐1324.
    1. Gaetani L, Blennow K, Calabresi P, Di Filippo M, Parnetti L, Zetterberg H. Neurofilament light chain as a biomarker in neurological disorders. J Neurol Neurosurg Psychiatry. 2019;90(8):870‐881.
    1. Parnetti L, Gaetani L, Eusebi P, et al. CSF and blood biomarkers for Parkinson's disease. Lancet Neurol. 2019;18(6):573‐586.
    1. Xicoy H, Penuelas N, Vila M, Laguna A. Autophagic‐ and lysosomal‐related biomarkers for Parkinson's disease: lights and shadows. Cells 2019;8(11):1317.
    1. Bliederhaeuser C, Grozdanov V, Speidel A, et al. Age‐dependent defects of alpha‐synuclein oligomer uptake in microglia and monocytes. Acta Neuropathol. 2016;131(3):379‐391.
    1. Codolo G, Plotegher N, Pozzobon T, et al. Triggering of inflammasome by aggregated alpha‐synuclein, an inflammatory response in synucleinopathies. PLoS One. 2013;8(1):e55375.
    1. Sulzer D, Alcalay RN, Garretti F, et al. T cells from patients with Parkinson's disease recognize alpha‐synuclein peptides. Nature 2017;546(7660):656‐661.
    1. Kannarkat GT, Cook DA, Lee JK, et al. Common genetic variant association with altered HLA expression, synergy with pyrethroid exposure, and risk for Parkinson's disease: an observational and case‐control study. NPJ Parkinsons Dis. 2015;1:15002.
    1. da Silva DJ, Borges AF, Souza PO, et al. Decreased toll‐like receptor 2 and toll‐like receptor 7/8‐induced cytokines in Parkinson's disease patients. Neuroimmunomodulation 2016;23(1):58‐66.
    1. Qin XY, Zhang SP, Cao C, Loh YP, Cheng Y. Aberrations in peripheral inflammatory cytokine levels in Parkinson disease: a systematic review and meta‐analysis. JAMA Neurol. 2016;73(11):1316‐1324.
    1. Tansey MG, Romero‐Ramos M. Immune system responses in Parkinson's disease: early and dynamic. Eur J Neurosci. 2019;49(3):364‐383.
    1. Easley‐Neal C, Foreman O, Sharma N, Zarrin AA, Weimer RM. CSF1R ligands IL‐34 and CSF1 are differentially required for microglia development and maintenance in white and gray matter brain regions. Front Immunol. 2019;10:2199.
    1. Esen N, Kielian T. Effects of low dose GM‐CSF on microglial inflammatory profiles to diverse pathogen‐associated molecular patterns (PAMPs). J Neuroinflammation. 2007;4:10.
    1. Ushach I, Zlotnik A. Biological role of granulocyte macrophage colony‐stimulating factor (GM‐CSF) and macrophage colony‐stimulating factor (M‐CSF) on cells of the myeloid lineage. J Leukoc Biol. 2016;100(3):481‐489.
    1. Bergman O, Ben‐Shachar D. Mitochondrial oxidative phosphorylation system (OXPHOS) deficits in schizophrenia: possible interactions with cellular processes. Can J Psychiatry. 2016;61(8):457‐469.
    1. Hammond ME, Lapointe GR, Feucht PH, et al. IL‐8 induces neutrophil chemotaxis predominantly via type I IL‐8 receptors. J Immunol. 1995;155(3):1428‐1433.
    1. Yilmaz R, Strafella AP, Bernard A, et al. Serum inflammatory profile for the discrimination of clinical subtypes in Parkinson's disease. Front Neurol. 2018;9:1123.
    1. Schipper HM, Song W. A heme oxygenase‐1 transducer model of degenerative and developmental brain disorders. Int J Mol Sci. 2015;16(3):5400‐5419.
    1. Schipper HM, Song W, Tavitian A, Cressatti M. The sinister face of heme oxygenase‐1 in brain aging and disease. Prog Neurobiol. 2019;172:40‐70.
    1. Schipper HM. Brain iron deposition and the free radical‐mitochondrial theory of ageing. Ageing Res Rev. 2004;3(3):265‐301.
    1. Hou L, Bao X, Zang C, et al. Integrin CD11b mediates alpha‐synuclein‐induced activation of NADPH oxidase through a Rho‐dependent pathway. Redox Biol. 2018;14:600‐608.
    1. Kim C, Ho DH, Suk JE, et al. Neuron‐released oligomeric alpha‐synuclein is an endogenous agonist of TLR2 for paracrine activation of microglia. Nat Commun. 2013;4:1562.
    1. Fellner L, Irschick R, Schanda K, et al. Toll‐like receptor 4 is required for alpha‐synuclein dependent activation of microglia and astroglia. Glia. 2013;61(3):349‐360.
    1. Schwab AD, Thurston MJ, Machhi J, et al. Immunotherapy for Parkinson's disease. Neurobiol Dis. 2020;137:104760.
    1. Braza F, Brouard S, Chadban S, Goldstein DR. Role of TLRs and DAMPs in allograft inflammation and transplant outcomes. Nat Rev Nephrol. 2016;12(5):281‐290.
    1. Campolo M, Filippone A, Biondo C, et al. TLR7/8 in the pathogenesis of Parkinson's disease. Int J Mol Sci. 2020;21(24):9384.
    1. Roh JS, Sohn DH. Damage‐associated molecular patterns in inflammatory diseases. Immune Netw. 2018;18(4):e27.
    1. Ahmed AU, Sarvestani ST, Gantier MP, Williams BR, Hannigan GE. Integrin‐linked kinase modulates lipopolysaccharide‐ and Helicobacter pylori‐induced nuclear factor kappaB‐activated tumor necrosis factor‐alpha production via regulation of p65 serine 536 phosphorylation. J Biol Chem. 2014;289(40):27776‐27793.
    1. Dasgupta S, Jana M, Liu X, Pahan K. Role of very‐late antigen‐4 (VLA‐4) in myelin basic protein‐primed T cell contact‐induced expression of proinflammatory cytokines in microglial cells. J Biol Chem. 2003;278(25):22424‐22431.
    1. Liu X, Jana M, Dasgupta S, et al. Human immunodeficiency virus type 1 (HIV‐1) tat induces nitric‐oxide synthase in human astroglia. J Biol Chem. 2002;277(42):39312‐39319.
    1. Rothwarf DM, Zandi E, Natoli G, Karin M. IKK‐gamma is an essential regulatory subunit of the IkappaB kinase complex. Nature 1998;395(6699):297‐300.
    1. Kirkley KS, Popichak KA, Hammond SL, Davies C, Hunt L, Tjalkens RB. Genetic suppression of IKK2/NF‐kappaB in astrocytes inhibits neuroinflammation and reduces neuronal loss in the MPTP‐probenecid model of Parkinson's disease. Neurobiol Dis. 2019;127:193‐209.
    1. Lim KL, Tan JM. Role of the ubiquitin proteasome system in Parkinson's disease. BMC Biochem. 2007;8(suppl 1):S13.
    1. Hou X, Watzlawik JO, Fiesel FC, Springer W. Autophagy in Parkinson's disease. J Mol Biol. 2020;432(8):2651‐2672.
    1. Xilouri M, Brekk OR, Stefanis L. Autophagy and alpha‐synuclein: relevance to Parkinson's disease and related synucleopathies. Mov Disord. 2016;31(2):178‐192.
    1. Ryan BJ, Hoek S, Fon EA, Wade‐Martins R. Mitochondrial dysfunction and mitophagy in Parkinson's: from familial to sporadic disease. Trends Biochem Sci 2015;40(4):200‐210.
    1. Thomas MP, Chartrand K, Reynolds A, Vitvitsky V, Banerjee R, Gendelman HE. Ion channel blockade attenuates aggregated alpha synuclein induction of microglial reactive oxygen species: relevance for the pathogenesis of Parkinson's disease. J Neurochem. 2007;100(2):503‐519.
    1. Kocaturk NM, Gozuacik D. Crosstalk between mammalian autophagy and the ubiquitin‐proteasome system. Front Cell Dev Biol. 2018;6:128.
    1. Lee IH, Cao L, Mostoslavsky R, et al. A role for the NAD‐dependent deacetylase Sirt1 in the regulation of autophagy. Proc Natl Acad Sci U S A. 2008;105(9):3374‐3379.
    1. Sasai M, Sakaguchi N, Ma JS, et al. Essential role for GABARAP autophagy proteins in interferon‐inducible GTPase‐mediated host defense. Nat Immunol. 2017;18(8):899‐910.
    1. Rideout HJ, Stefanis L. The neurobiology of LRRK2 and its role in the pathogenesis of Parkinson's disease. Neurochem Res. 2014;39(3):576‐592.
    1. Wang D. Discrepancy between mRNA and protein abundance: insight from information retrieval process in computers. Comput Biol Chem. 2008;32(6):462‐468.
    1. Liu Y, Beyer A, Aebersold R. On the dependency of cellular protein levels on mRNA abundance. Cell 2016;165(3):535‐550.

Source: PubMed

3
Iratkozz fel