You're using an outdated browser. Please upgrade to a modern browser for the best experience.
FDA-Approved Kinase Inhibitors in Trials for Neurological Disorders: Comparison
Please note this is a comparison between Version 2 by Catherine Yang and Version 1 by Austin Lui.

The United States Food and Drug Administration (US FDA) has so far approved 74 kinase inhibitors, with numerous other kinase inhibitors in clinical trials, mostly for the treatment of cancers. In contrast, there are dire unmet needs of FDA-approved drugs for neurological treatments, such as Alzheimer’s disease (AD), intracerebral hemorrhage (ICH), ischemic stroke (IS), traumatic brain injury (TBI), and others.

  • aberrant cell cycle disease
  • cancers
  • neurological disorders
  • kinase inhibitors

1. FDA-Approved Kinase Inhibitors in Clinical Trials for Neurological Disorders

Many of the FDA-approved kinase inhibitors have been tested in clinical and preclinical trials for neurological disorders, though none of them have been approved by the FDA for neurological treatment. In terms of clinical trials, Baricitinib, a JAK inhibitor, is being studied in a phase II trial (NCT03921554) along with a phase II and III trial (NCT04517253), for efficacy and safety in Aicardi–Goutieres Syndrome, an inherited encephalopathy that affects infants and usually results in severe mental and physical disabilities.
Bosutinib, an inhibitor of Src and Bcr-Abl, is and has been investigated in clinical trials for different neurodegenerative disorders. There is a phase I trial (NCT04744532) studying the safety and tolerability of bosutinib for amyotrophic lateral sclerosis (ALS), a progressive neurological disease leading to loss of muscle control. The safety, tolerability, and clinical outcomes of bosutinib on patients with dementia with Lewy bodies has also been studied in a completed phase 2 trial (NCT03888222). In preclinical studies, it has been found that bosutinib reduces levels of alpha-synuclein, tau, and beta-amyloid in the CNS, and improves motor and cognitive behavior in animal models [38,39,40][1][2][3]. Bosutinib was also found to promote autophagy and clear protein aggregates in neurons [41,42][4][5]. There is also an ongoing phase I trial (NCT02921477) studying the safety and tolerability of bosutinib for mild cognitive impairment (MCI) and dementia.
Cobimetinib, an MEK inhibitor, has been studied in a phase 2 trial (NCT04079179) studying its safety and efficacy in histiocytic disorders, which can lead to neurodegeneration. Dasatinib, an inhibitor of Src, Bcr-Abl, Kit, EGFR, PDGFR, and EPH (EphA2), has been studied in multiple clinical trials examining its effectiveness in treating AD and mild cognitive impairment. Particularly, in four clinical trials (NCT04063124—phase I and II, NCT04785300—phase I and II, NCT04685590- phase II, NCT05422885—phase I/II), the safety, feasibility, and efficacy of dasatinib and quercetin, a flavonoid known to have antioxidant and anti-inflammatory effects, are being assessed.
Everolimus, an inhibitor of mTOR and FKBP, has been extensively evaluated in clinical trials studying different acute brain injury disorders, neurodegenerative disorders, and neurodevelopmental disorders. A phase II trial (NCT03198949) studying the safety and anti-epileptic efficacy of everolimus in patients with Epilepsy and focal cortical dysplasia II, who have failed more than two antiepileptic drugs and surgery, has been recently completed. Everolimus has been shown in animal models to protect seizure-induced brain injury and reduce neuroinflammation associated with seizures [43,44][6][7]. A phase II trial (NCT00857259) evaluating the safety and efficacy of everolimus with or without ranibizumab in patients with neovascular age-related macular degeneration, a neurodegenerative disorder that results in a loss of central vision, is currently in progress. Additionally, in a phase I and II trial (NCT02991807), researchers studied whether everolimus can improve neurocognitive outcomes in patients with hamartoma tumor syndrome caused by a PTEN germline mutation. There are also multiple studies (NCT02962414, NCT01730209, NCT01070316, NCT01713946, NCT02451696, NCT01954693, NCT01929642, and NCT012899-12) evaluating the safety and efficacy of everolimus in patients with tuberous sclerosis complex, which is often associated with refractory seizures, cognitive disabilities, autism, focal cortical dysplasia, other neurocognitive problems, and self-injury. Lastly, there is a phase II trial that studied the safety and efficacy of everolimus in patients with seizures who have Sturge–Weber syndrome, a rare disease in which tumors form in the brain (NCT01997255).
Imatinib, a Bcr-Abl, Kit and PDGFR inhibitor, has been studied in several acute brain injury and neurodegenerative disorders. In a phase III trial (NCT03639922), imatinib was studied in ischemic stroke patients to determine whether there was improvement in functional outcomes. Imatinib, administered for 6 days, was added to conventional stroke therapy and started within 8 days of the onset of stroke. Additionally, in a phase II trial (NCT02363361), the safety, uptake, and tolerability of imatinib is being studied in patients with cervical SCI. A phase II trial (NCT03674099) is currently testing imatinib as a novel therapy for multiple sclerosis, comparing its efficacy to methylprednisolone, the standard of care drug for multiple sclerosis relapses. Lastly, imatinib had been studied in a phase I trial (NCT00403156), examining choroidal neovascularization, although this study has been withdrawn.
Nilotinib is a kinase inhibitor that inhibits the activity of Bcr-Abl, PDGFR and DDR1. It has been studied in several neurodegenerative diseases in clinical trials. In a phase I study, nilotinib (NCT03764215) was administered to patients with Huntington disease. Biomarkers, such as phosphorylated tau levels, and functional outcomes were assessed. In an ongoing phase II study (NCT04002674), the use of nilotinib in patients with dementia with Lewy bodies is being studied, particularly on the pharmacokinetics, tolerability, biomarkers, and safety of use. In a phase II study (NCT02947893), the efficacy of nilotinib in AD was studied. Specifically, the effects of nilotinib on cell death was detected with cell markers, and the amyloid concentrations in the brain were assessed with PET scans. Also, a recent phase III clinical trial (NCT05143528) is currently studying the safety and efficacy of nilotinib in patients with early AD using two different dosages. There are also three studies that examined the effects of nilotinib in patients with Parkinson’s disease (NCT02954978, NCT02281474, NCT03205488). In a phase II trial (NCT03932669), the efficacy and adverse events of nilotinib are being studied in patients with spinocerebellar ataxia. In particular, improvement in daily living performance and cerebellar functions are being assessed.
Pazopanib inhibits the activities of VEGFR 1/2/3, PDGFR α/β, FGFR 1/3, Kit, Lck, Fms, and Itk. In terms of neurological disease settings, it is studied mainly in macular degeneration (NCT00659555, NCT01154062, NCT00612456, NCT01072214, NCT00463320, NCT01362348, NCT01134055, NCT01051700, and NCT00733304). Regorafenib, a VEGF kinase inhibitor, was studied in neovascular age-related macular degeneration. After successfully passing phase I clinical trials, (NCT02222207), phase IIa trials were terminated after the results were less effective than the current gold standard treatment [45][8].
Sirolimus, an mTOR inhibitor initially used as an immunosuppressant in kidney transplants, has been repurposed in a multitude of neurological and psychiatric clinical trials, including cerebral aneurysms (NCT04141020), epilepsy (NCT03646240), Alzheimer’s disease (NCT04629495 and NCT04200911), frontotemporal dementia (NCT04408625), amyotrophic lateral sclerosis (NCT03359538), Parkinson’s disease (NCT04127578), age-related macular degeneration (NCT01445548, NCT00766649, NCT00712491, NCT02357342, NCT02732899, NCT00766337, and NCT00304954), multiple sclerosis (NCT00095329), geographic atrophy associated with age-related macular degeneration (NCT01675947), multiple system atrophy (NCT03589976), Sturge–Weber syndrome (NCT03047980 and NCT02080624), lysosomal diseases (NCT03952637), Leigh syndrome (NCT03747328), tuberous sclerosis complex (NCT04595513, NCT01929642, and NCT05104983), Gaucher disease type 2 (NCT04411654), diabetic retinopathy (NCT00711490), diabetic macular edema (NCT00656643 and NCT00401115), alcohol use disorder (NCT03732248), smoking cessation (NCT04161144), depression (NCT02487485), and stroke prevention (NCT04948749).

2. FDA-Approved Kinase Inhibitors in Preclinical Trials for Neurological Disorders

While there have been 16 FDA-approved kinase inhibitors in clinical trials for neurological disorders, there are numerous preclinical studies of FDA-approved kinase inhibitors evaluating their effects on neurological disorders. Abemaciclib has been studied in preclinical models for the treatment of motor neuron degeneration [46][9] and post-traumatic stress disorder [47][10]. Afatinib has been tested in preclinical models for the treatment of oxygen/glucose deprivation-induced neuroinflammation [48][11], multiple sclerosis [49][12], autoimmune CNS inflammation [49][12], and nicotine dependance [50][13]. Axitinib has been tested for treatment of AD [51][14]. Alectinib has been tested for the potential treatment of binge drinking [52,53][15][16]. Baricitinib has been tested in preclinical models for the potential treatment of neurocognitive disorders induced by HIV [54][17], encephalitis [55[18][19],56], multiple sclerosis [56][19], hypersensitivity in Down syndrome [57][20], acute spinal cord injury [58][21], Hutchinson–Gilford progeria [59][22], and AD [60][23]. Binimetinib has been shown in a preclinical study to be a potential treatment for some forms of AD [61][24]. Bosutinib has been tested for the potential treatment of intracerebral hemorrhage [62][25], cerebral ischemia [63][26], α-synucleinopathies and tauopathies in neurodegeneration [40[3][4],41], Parkinson’s disease [38[1][5][27],42,64], TDP-43 pathology [65][28], SIN1-mediated neurotoxicity [66][29], and botulinum neurotoxins [67][30]. Cabozantinib has been tested for the potential treatment of Rett syndrome [68][31] and AD [69][32].
Crizotinib has been tested for the potential treatment of Parkinson’s disease [70][33], AD [71][34], persistent pain [72][35], Toxoplasma gondii (can result in symptoms of congenital neurological and meningoencephalitis) [73][36], and craniosynostosis associated with Saethre–Chotzen syndrome [74][37]. Dabrafenib has been tested for the potential treatment of ischemic brain injury [75][38], spinal cord injury [76][39], Parkinson’s diseases [77[40][41],78], and ataxia caused by neurohistiocytosis of the cerebellum [79][42]. Dasatinib has been tested for the potential treatment of traumatic brain injury [80][43], lipopolysaccharide-induced neuroinflammation [81][44], kainic acid-induced neuroinflammation [82][45], glaucoma [83][46], tau-associated pathology [84][47], multiple sclerosis [85][48], amyotrophic lateral sclerosis [86[49][50][51],87,88], Parkinson’s disease [87][50], age-related blood brain barrier dysfunction [89][52], age-related cognitive dysfunction [89[52][53],90], obesity-induced anxiety [91][54], chronic unpredictable stress-induced cognitive deficits [92][55], fetal alcohol syndrome [93][56], and botulinum neurotoxins [67][30]. Erlotinib has been tested for the potential treatment of nerve fiber injury [94][57], intracranial aneurysm formation [95][58], amyotrophic lateral sclerosis [96][59], diabetic peripheral neuropathy [97[60][61],98], and amyloid-β-induced memory loss [99][62].
Everolimus has been tested for the potential treatment of encephalopathy of prematurity [100][63], atherosclerosis-associated brain hypoxia [101][64], ischemic stroke [102[65][66][67],103,104], Alzheimer’s disease [105[68][69],106], Huntington disease [107[70][71],108], vascular dementia [109][72], lipopolysaccharide-induced neuroinflammation [110][73], insulin dysfunction-related cognitive dysfunction [111][74], glutamate-induced neurotoxicity [112][75], Guillain–Barre syndrome [113][76], multiple sclerosis [114][77], tuberous sclerosis complex-associated autism-like social deficits [115[78][79],116], and Lafora disease [117][80]. Fedratinib has been tested for the potential treatment of ischemic stroke [118][81], intracerebral hemorrhage [119][82], Wernicke’s encephalopathy [120[83][84],121], and Alzheimer’s disease [122][85]. Gefitinib has been tested for the potential treatment of spinal cord injury [123][86], amyloid-β-induced memory loss [99][62], schizophrenia [124][87], Streptococcus pneumoniae meningitis [125][88], and Toxoplasma gondii (can result in symptoms of congenital neurological and meningoencephalitis) [73,126][36][89].
Ibrutinib has been tested for the potential treatment of ischemic stroke [127[90][91],128], spinal cord injury [129,130][92][93], age-related cognitive deterioration [131][94], Alzheimer’s disease [132[95][96],133], lipopolysaccharide-induced neuroinflammation [134][97], anxiogenic behavior [135][98], depression [136,137][99][100], and cocaine use disorder [138][101]. Imatinib has been tested for the potential treatment of subarachnoid hemorrhage [139[102][103][104][105][106][107],140,141,142,143,144], intracerebral hemorrhage [145[108][109][110][111],146,147,148], cerebral small vessel disease [149][112], traumatic brain injury-induced seizures [150][113], seizures [150[113][114],151], traumatic brain injury [152][115], ischemia reperfusion-induced cerebral injury [153[116][117],154], Alzheimer’s disease [155[118][119][120][121][122][123][124][125][126][127][128][129][130][131][132][133],156,157,158,159,160,161,162,163,164,165,166,167,168,169,170], Parkinson’s disease [171[134][135][136][137],172,173,174], prion diseases [175[138][139][140][141],176,177,178], amyotrophic lateral sclerosis [179][142], Huntington’s diseases [180][143], cerebral malaria [181][144], hypoxic ventilatory depression [182][145], Niemann–Pick type C disease [183][146], Niemann–Pick type A disease [184][147], Gaucher disease [185][148], simian human immunodeficiency virus encephalitis [186][149], and morphine tolerance [187][150]. Lapatinib has been tested for the potential treatment of epileptic seizures [188][151], organophosphate-induced axonal damage in spinal cord [189][152], and Alzheimer’s disease [190,191][153][154]. Lorlatinib has been tested for the potential treatment of persistent pain [72][35]. Midostaurin has been tested for the potential treatment of traumatic spinal cord injury [192][155]. Neratinib has been tested for the potential treatment of AD [193][156].
Nilotinib has been tested for the potential treatment of epileptic seizures [194][157], tauopathies [40[3][4][158],41,195], alpha-synucleinopathies [40[3][5][159][160][161],42,196,197,198], TDP-43 pathology [64,65][27][28], beta-amyloid pathology [195][158], AD [60,199,200[23][162][163][164],201], Parkinson’s disease [202[165][166][167],203,204], chorea-acanthocytosis [205[168][169],206], and Niemann–Pick type A disease [184][147]. Palbociclib has been tested for the potential treatment of spinal muscular atrophy [207][170], amyloid beta-peptide pathology [208][171], and Parkinson’s disease [209][172]. Pazopanib has been tested for the potential treatment of tauopathy [210][173] and neurodegeneration-induced memory and cognitive deficits [211][174]. Pexidartinib has been tested for the potential treatment of intracerebral hemorrhage [212[175][176],213], subarachnoid hemorrhage [214][177], obesity-related cerebrovascular dysfunction [215][178], cognitive decline due to brain damage [216][179], tauopathy [217][180], AD [218[181][182],219], Huntington’s disease [220][183], multiple sclerosis [221[184][185][186],222,223], spinocerebellar ataxia type 1 [224][187], Down syndrome [225][188], peripheral nerve injury-induced mechanical hypersensitivity [226][189], cocaine addiction [227][190], and Parkinson’s disease [228][191]. Ponatinib has been tested for the potential treatment of ischemic stroke [229][192], epilepsy [230][193], and cerebral cavernous malformation [231][194].
Sorafenib has been tested for the potential treatment for subarachnoid hemorrhage [450][195], ischemic stroke [451][196], spinal cord injury [452][197], AD [69,453[32][198][199],454], Parkinson’s disease [455][200], multiple sclerosis [456[201][202],457], rabies [458][203], Rift Valley fever virus [459][204], alphaviruses [460][205], and Picornavirus enterovirus 71 [461][206]. Sunitinib has been tested for the potential treatment for traumatic brain injury [462][207], seizure [463][208], AD [464[209][210][211],465,466], Rett syndrome [68][31], cognitive impairment associated with HIV [467[212][213],468], dengue virus [469][214], and rabies [470][215]. Temsirolimus has been tested for the potential treatment for spinal cord injury [300][216], Parkinson’s disease [471[217][218],472], tauopathy [473[219][220],474], AD [475][221], spinocerebellar ataxia type 3 [476][222], nicotine withdrawal-associated cognitive deficits [477][223], and X-linked adrenoleukodystrophy [478][224]. Tofacitinib has been tested for the potential treatment for ischemic stroke [479][225], AD [480][226], multiple sclerosis [481,482][227][228], Parkinson’s disease [483][229], amyotrophic lateral sclerosis [484][230], and Venezuelan equine encephalitis virus [485][231]. Trametinib has been tested for the potential treatment for traumatic brain injury [486][232], aneurysmal subarachnoid hemorrhage [487][233], and brain arteriovenous malformations [488][234]. Vandetanib has been tested for the potential treatment for germinal matrix hemorrhage [489][235]. Lastly, Infigratinib has also been tested as a potential treatment for diabetic retinopathy [490][236].

References

  1. Lonskaya, I.; Hebron, M.L.; Desforges, N.M.; Franjie, A.; Moussa, C.E. Tyrosine kinase inhibition increases functional parkin-Beclin-1 interaction and enhances amyloid clearance and cognitive performance. EMBO Mol. Med. 2013, 5, 1247–1262.
  2. Lonskaya, I.; Hebron, M.L.; Selby, S.T.; Turner, R.S.; Moussa, C.E. Nilotinib and bosutinib modulate pre-plaque alterations of blood immune markers and neuro-inflammation in Alzheimer’s disease models. Neuroscience 2015, 304, 316–327.
  3. Hebron, M.L.; Lonskaya, I.; Olopade, P.; Selby, S.T.; Pagan, F.; Moussa, C.E. Tyrosine Kinase Inhibition Regulates Early Systemic Immune Changes and Modulates the Neuroimmune Response in α-Synucleinopathy. J. Clin. Cell Immunol. 2014, 5, 259.
  4. Hebron, M.L.; Javidnia, M.; Moussa, C.E. Tau clearance improves astrocytic function and brain glutamate-glutamine cycle. J. Neurol. Sci. 2018, 391, 90–99.
  5. Lonskaya, I.; Desforges, N.M.; Hebron, M.L.; Moussa, C.E. Ubiquitination increases parkin activity to promote autophagic α-synuclein clearance. PLoS ONE 2013, 8, e83914.
  6. Huang, X.Y.; Hu, Q.P.; Shi, H.Y.; Zheng, Y.Y.; Hu, R.R.; Guo, Q. Everolimus inhibits PI3K/Akt/mTOR and NF-kB/IL-6 signaling and protects seizure-induced brain injury in rats. J. Chem. Neuroanat. 2021, 114, 101960.
  7. Yang, M.T.; Lin, Y.C.; Ho, W.H.; Liu, C.L.; Lee, W.T. Everolimus is better than rapamycin in attenuating neuroinflammation in kainic acid-induced seizures. J. Neuroinflamm. 2017, 14, 15.
  8. Joussen, A.M.; Wolf, S.; Kaiser, P.K.; Boyer, D.; Schmelter, T.; Sandbrink, R.; Zeitz, O.; Deeg, G.; Richter, A.; Zimmermann, T.; et al. The Developing Regorafenib Eye drops for neovascular Age-related Macular degeneration (DREAM) study: An open-label phase II trial. Br. J. Clin. Pharmacol. 2019, 85, 347–355.
  9. Wang, F.; Li, S.; Wang, T.Y.; Lopez, G.A.; Antoshechkin, I.; Chou, T.F. P97/VCP ATPase inhibitors can rescue p97 mutation-linked motor neuron degeneration. Brain Commun. 2022, 4, fcac176.
  10. Wang, X.; Ma, L.; Li, J.; Kong, F. Activated cell-cycle CDK4/CyclinD1-pRB-E2F1 signaling pathway is involved in the apoptosis of dorsal raphe nucleus in the rat model of PTSD. Biochem. Biophys. Res. Commun. 2022, 602, 142–148.
  11. Chen, Y.J.; Hsu, C.C.; Shiao, Y.J.; Wang, H.T.; Lo, Y.L.; Lin, A.M.Y. Anti-inflammatory effect of afatinib (an EGFR-TKI) on OGD-induced neuroinflammation. Sci. Rep. 2019, 9, 2516.
  12. Linnerbauer, M.; Lößlein, L.; Vandrey, O.; Tsaktanis, T.; Beer, A.; Naumann, U.J.; Panier, F.; Beyer, T.; Nirschl, L.; Kuramatsu, J.B.; et al. Intranasal delivery of a small-molecule ErbB inhibitor promotes recovery from acute and late-stage CNS inflammation. JCI Insight 2022, 7, e154824.
  13. Turner, J.R.; Ray, R.; Lee, B.; Everett, L.; Xiang, J.; Jepson, C.; Kaestner, K.H.; Lerman, C.; Blendy, J.A. Evidence from mouse and man for a role of neuregulin 3 in nicotine dependence. Mol. Psychiatry 2014, 19, 801–810.
  14. Singh, C.S.B.; Choi, K.B.; Munro, L.; Wang, H.Y.; Pfeifer, C.G.; Jefferies, W.A. Reversing pathology in a preclinical model of Alzheimer’s disease by hacking cerebrovascular neoangiogenesis with advanced cancer therapeutics. EBioMedicine 2021, 71, 103503.
  15. Dutton, J.W., 3rd; Chen, H.; You, C.; Brodie, M.S.; Lasek, A.W. Anaplastic lymphoma kinase regulates binge-like drinking and dopamine receptor sensitivity in the ventral tegmental area. Addict. Biol. 2017, 22, 665–678.
  16. Hamada, K.; Ferguson, L.B.; Mayfield, R.D.; Krishnan, H.R.; Maienschein-Cline, M.; Lasek, A.W. Binge-like ethanol drinking activates anaplastic lymphoma kinase signaling and increases the expression of STAT3 target genes in the mouse hippocampus and prefrontal cortex. Genes Brain Behav. 2021, 20, e12729.
  17. Gavegnano, C.; Haile, W.B.; Hurwitz, S.; Tao, S.; Jiang, Y.; Schinazi, R.F.; Tyor, W.R. Baricitinib reverses HIV-associated neurocognitive disorders in a SCID mouse model and reservoir seeding in vitro. J. Neuroinflamm. 2019, 16, 182.
  18. Nakamura, J.; Yanagida, M.; Saito, K.; Kamata, Y.; Nagashima, T.; Iwamoto, M.; Sato, T.; Sato, K. Epstein-Barr Virus Encephalitis in a Patient with Rheumatoid Arthritis. Mod. Rheumatol. Case Rep. 2021, 6, 160–162.
  19. Dang, C.; Lu, Y.; Chen, X.; Li, Q. Baricitinib Ameliorates Experimental Autoimmune Encephalomyelitis by Modulating the Janus Kinase/Signal Transducer and Activator of Transcription Signaling Pathway. Front. Immunol. 2021, 12, 650708.
  20. Tuttle, K.D.; Waugh, K.A.; Araya, P.; Minter, R.; Orlicky, D.J.; Ludwig, M.; Andrysik, Z.; Burchill, M.A.; Tamburini, B.A.J.; Sempeck, C.; et al. JAK1 Inhibition Blocks Lethal Immune Hypersensitivity in a Mouse Model of Down Syndrome. Cell Rep. 2020, 33, 108407.
  21. Zheng, X.Q.; Huang, J.F.; Lin, J.L.; Zhu, Y.X.; Wang, M.Q.; Guo, M.L.; Zan, X.J.; Wu, A.M. Controlled release of baricitinib from a thermos-responsive hydrogel system inhibits inflammation by suppressing JAK2/STAT3 pathway in acute spinal cord injury. Colloids Surf. B Biointerfaces 2021, 199, 111532.
  22. Liu, C.; Arnold, R.; Henriques, G.; Djabali, K. Inhibition of JAK-STAT Signaling with Baricitinib Reduces Inflammation and Improves Cellular Homeostasis in Progeria Cells. Cells 2019, 8, 1276.
  23. Agarwal, K.; Katare, D.P.; Jakhmola-Mani, R. Foresee novel targets for Alzheimer’s disease by investigating repurposed drugs. CNS Neurol. Disord. Drug Targets, 2022; online ahead of print.
  24. Schapansky, J.; Grinberg, Y.Y.; Osiecki, D.M.; Freeman, E.A.; Walker, S.G.; Karran, E.; Gopalakrishnan, S.M.; Talanian, R.V. MEK1/2 activity modulates TREM2 cell surface recruitment. J. Biol. Chem. 2021, 296, 100218.
  25. Ma, L.; Manaenko, A.; Ou, Y.B.; Shao, A.W.; Yang, S.X.; Zhang, J.H. Bosutinib Attenuates Inflammation via Inhibiting Salt-Inducible Kinases in Experimental Model of Intracerebral Hemorrhage on Mice. Stroke 2017, 48, 3108–3116.
  26. Liang, S.; Pong, K.; Gonzales, C.; Chen, Y.; Ling, H.P.; Mark, R.J.; Boschelli, F.; Boschelli, D.H.; Ye, F.; Barrios Sosa, A.C.; et al. Neuroprotective profile of novel SRC kinase inhibitors in rodent models of cerebral ischemia. J. Pharmacol. Exp. Ther. 2009, 331, 827–835.
  27. Wenqiang, C.; Lonskaya, I.; Hebron, M.L.; Ibrahim, Z.; Olszewski, R.T.; Neale, J.H.; Moussa, C.E. Parkin-mediated reduction of nuclear and soluble TDP-43 reverses behavioral decline in symptomatic mice. Hum. Mol. Genet. 2014, 23, 4960–4969.
  28. Heyburn, L.; Hebron, M.L.; Smith, J.; Winston, C.; Bechara, J.; Li, Z.; Lonskaya, I.; Burns, M.P.; Harris, B.T.; Moussa, C.E. Tyrosine kinase inhibition reverses TDP-43 effects on synaptic protein expression, astrocytic function and amino acid dis-homeostasis. J. Neurochem. 2016, 139, 610–623.
  29. Yilmaz, S.; Alkan, T.; Ballar Kirmizibayrak, P. A new underlying mechanism for the neuroprotective effect of bosutinib: Reverting toxicity-induced PARylation in SIN1-mediated neurotoxicity. J. Biochem. Mol. Toxicol. 2021, 35, e22915.
  30. Kiris, E.; Burnett, J.C.; Nuss, J.E.; Wanner, L.M.; Peyser, B.D.; Du, H.T.; Gomba, G.Y.; Kota, K.P.; Panchal, R.G.; Gussio, R.; et al. SRC family kinase inhibitors antagonize the toxicity of multiple serotypes of botulinum neurotoxin in human embryonic stem cell-derived motor neurons. Neurotox. Res. 2015, 27, 384–398.
  31. Tang, X.; Drotar, J.; Li, K.; Clairmont, C.D.; Brumm, A.S.; Sullins, A.J.; Wu, H.; Liu, X.S.; Wang, J.; Gray, N.S.; et al. Pharmacological enhancement of KCC2 gene expression exerts therapeutic effects on human Rett syndrome neurons and Mecp2 mutant mice. Sci. Transl. Med. 2019, 11, eaau0164.
  32. Tucker Edmister, S.; Del Rosario Hernández, T.; Ibrahim, R.; Brown, C.A.; Gore, S.V.; Kakodkar, R.; Kreiling, J.A.; Creton, R. Novel use of FDA-approved drugs identified by cluster analysis of behavioral profiles. Sci. Rep. 2022, 12, 6120.
  33. Bolz, S.N.; Salentin, S.; Jennings, G.; Haupt, V.J.; Sterneckert, J.; Schroeder, M. Structural binding site comparisons reveal Crizotinib as a novel LRRK2 inhibitor. Comput. Struct. Biotechnol. J. 2021, 19, 3674–3681.
  34. Lim, J.W.; Kim, S.K.; Choi, S.Y.; Kim, D.H.; Gadhe, C.G.; Lee, H.N.; Kim, H.J.; Kim, J.; Cho, S.J.; Hwang, H.; et al. Identification of crizotinib derivatives as potent SHIP2 inhibitors for the treatment of Alzheimer’s disease. Eur. J. Med. Chem. 2018, 157, 405–422.
  35. Defaye, M.; Iftinca, M.C.; Gadotti, V.M.; Basso, L.; Abdullah, N.S.; Cuménal, M.; Agosti, F.; Hassan, A.; Flynn, R.; Martin, J.; et al. The neuronal tyrosine kinase receptor ligand ALKAL2 mediates persistent pain. J. Clin. Invest. 2022, 132, e154317.
  36. Yang, Z.; Ahn, H.J.; Nam, H.W. Gefitinib inhibits the growth of Toxoplasma gondii in HeLa cells. Korean J. Parasitol. 2014, 52, 439–441.
  37. Camp, E.; Anderson, P.J.; Zannettino, A.C.W.; Glackin, C.A.; Gronthos, S. Tyrosine kinase receptor c-ros-oncogene 1 inhibition alleviates aberrant bone formation of TWIST-1 haploinsufficient calvarial cells from Saethre-Chotzen syndrome patients. J. Cell Physiol. 2018, 233, 7320–7332.
  38. Cruz, S.A.; Qin, Z.; Stewart, A.F.R.; Chen, H.H. Dabrafenib, an inhibitor of RIP3 kinase-dependent necroptosis, reduces ischemic brain injury. Neural Regen. Res. 2018, 13, 252–256.
  39. Sugaya, T.; Kanno, H.; Matsuda, M.; Handa, K.; Tateda, S.; Murakami, T.; Ozawa, H.; Itoi, E. B-RAF(V600E) Inhibitor Dabrafenib Attenuates RIPK3-Mediated Necroptosis and Promotes Functional Recovery after Spinal Cord Injury. Cells 2019, 8, 1582.
  40. Uenaka, T.; Satake, W.; Cha, P.C.; Hayakawa, H.; Baba, K.; Jiang, S.; Kobayashi, K.; Kanagawa, M.; Okada, Y.; Mochizuki, H.; et al. In silico drug screening by using genome-wide association study data repurposed dabrafenib, an anti-melanoma drug, for Parkinson’s disease. Hum. Mol. Genet. 2018, 27, 3974–3985.
  41. Okamoto, T. Parkinson’s Disease: Amantadine, zonisamide, dabrafenib. Brain Nerve 2019, 71, 953–959.
  42. Elkouzi, A.; Rauschkolb, P.; Grogg, K.L.; Gilchrist, J.M. Neurohistiocytosis of the Cerebellum: A Rare Cause of Ataxia. Mov. Disord. Clin. Pract. 2016, 3, 125–129.
  43. Yu, W.; Parakramaweera, R.; Teng, S.; Gowda, M.; Sharad, Y.; Thakker-Varia, S.; Alder, J.; Sesti, F. Oxidation of KCNB1 Potassium Channels Causes Neurotoxicity and Cognitive Impairment in a Mouse Model of Traumatic Brain Injury. J. Neurosci. 2016, 36, 11084–11096.
  44. Saminathan, H.; Charli, A.; Luo, J.; Panicker, N.; Gordon, R.; Hostetter, J.M.; Jin, H.; Anantharam, V.; Kanthasamy, A.G.; Kanthasamy, A. Fyn kinase mediates pro-inflammatory response in a mouse model of endotoxemia: Relevance to translational research. Eur. J. Pharmacol. 2020, 881, 173259.
  45. Gangoso, E.; Talaverón, R.; Jaraíz-Rodríguez, M.; Domínguez-Prieto, M.; Ezan, P.; Koulakoff, A.; Medina, J.M.; Giaume, C.; Tabernero, A. A c-Src Inhibitor Peptide Based on Connexin43 Exerts Neuroprotective Effects through the Inhibition of Glial Hemichannel Activity. Front. Mol. Neurosci. 2017, 10, 418.
  46. El-Nimri, N.W.; Moore, S.M.; Zangwill, L.M.; Proudfoot, J.A.; Weinreb, R.N.; Skowronska-Krawczyk, D.; Baxter, S.L. Evaluating the neuroprotective impact of senolytic drugs on human vision. Sci. Rep. 2020, 10, 21752.
  47. Musi, N.; Valentine, J.M.; Sickora, K.R.; Baeuerle, E.; Thompson, C.S.; Shen, Q.; Orr, M.E. Tau protein aggregation is associated with cellular senescence in the brain. Aging Cell 2018, 17, e12840.
  48. Azizi, G.; Goudarzvand, M.; Afraei, S.; Sedaghat, R.; Mirshafiey, A. Therapeutic effects of dasatinib in mouse model of multiple sclerosis. Immunopharmacol. Immunotoxicol. 2015, 37, 287–294.
  49. Katsumata, R.; Ishigaki, S.; Katsuno, M.; Kawai, K.; Sone, J.; Huang, Z.; Adachi, H.; Tanaka, F.; Urano, F.; Sobue, G. c-Abl inhibition delays motor neuron degeneration in the G93A mouse, an animal model of amyotrophic lateral sclerosis. PLoS ONE 2012, 7, e46185.
  50. Lawana, V.; Singh, N.; Sarkar, S.; Charli, A.; Jin, H.; Anantharam, V.; Kanthasamy, A.G.; Kanthasamy, A. Involvement of c-Abl Kinase in Microglial Activation of NLRP3 Inflammasome and Impairment in Autolysosomal System. J. Neuroimmune Pharmacol. 2017, 12, 624–660.
  51. Torres, P.; Anerillas, C.; Ramírez-Núñez, O.; Fernàndez, A.; Encinas, M.; Povedano, M.; Andrés-Benito, P.; Ferrer, I.; Ayala, V.; Pamplona, R.; et al. A motor neuron disease mouse model reveals a non-canonical profile of senescence biomarkers. Dis. Models Mech. 2022, 15, dmm049059.
  52. Ya, J.; Kadir, R.R.A.; Bayraktutan, U. Delay of endothelial cell senescence protects cerebral barrier against age-related dysfunction: Role of senolytics and senomorphics. Tissue Barriers, 2022; online ahead of print.
  53. Krzystyniak, A.; Wesierska, M.; Petrazzo, G.; Gadecka, A.; Dudkowska, M.; Bielak-Zmijewska, A.; Mosieniak, G.; Figiel, I.; Wlodarczyk, J.; Sikora, E. Combination of dasatinib and quercetin improves cognitive abilities in aged male Wistar rats, alleviates inflammation and changes hippocampal synaptic plasticity and histone H3 methylation profile. Aging Albany NY 2022, 14, 572–595.
  54. Ogrodnik, M.; Zhu, Y.; Langhi, L.G.P.; Tchkonia, T.; Krüger, P.; Fielder, E.; Victorelli, S.; Ruswhandi, R.A.; Giorgadze, N.; Pirtskhalava, T.; et al. Obesity-Induced Cellular Senescence Drives Anxiety and Impairs Neurogenesis. Cell Metab. 2019, 29, 1061–1077.e8.
  55. Lin, Y.F.; Wang, L.Y.; Chen, C.S.; Li, C.C.; Hsiao, Y.H. Cellular senescence as a driver of cognitive decline triggered by chronic unpredictable stress. Neurobiol. Stress 2021, 15, 100341.
  56. Wang, D.; Howell, B.W.; Olson, E.C. Maternal Ethanol Exposure Acutely Elevates Src Family Kinase Activity in the Fetal Cortex. Mol. Neurobiol. 2021, 58, 5210–5223.
  57. Koprivica, V.; Cho, K.S.; Park, J.B.; Yiu, G.; Atwal, J.; Gore, B.; Kim, J.A.; Lin, E.; Tessier-Lavigne, M.; Chen, D.F.; et al. EGFR activation mediates inhibition of axon regeneration by myelin and chondroitin sulfate proteoglycans. Science 2005, 310, 106–110.
  58. Luo, Y.; Tang, H.; Zhang, Z.; Zhao, R.; Wang, C.; Hou, W.; Huang, Q.; Liu, J. Pharmacological inhibition of epidermal growth factor receptor attenuates intracranial aneurysm formation by modulating the phenotype of vascular smooth muscle cells. CNS Neurosci. Ther. 2022, 28, 64–76.
  59. LePichon, C.E.; Dominguez, S.L.; Solanoy, H.; Ngu, H.; Lewin-Koh, N.; Chen, M.; Eastham-Anderson, J.; Watts, R.; Scearce-Levie, K. EGFR inhibitor erlotinib delays disease progression but does not extend survival in the SOD1 mouse model of ALS. PLoS ONE 2013, 8, e62342.
  60. Pan, P.; Dobrowsky, R.T. Differential expression of neuregulin-1 isoforms and downregulation of erbin are associated with Erb B2 receptor activation in diabetic peripheral neuropathy. Acta Neuropathol. Commun. 2013, 1, 39.
  61. McGuire, J.F.; Rouen, S.; Siegfreid, E.; Wright, D.E.; Dobrowsky, R.T. Caveolin-1 and altered neuregulin signaling contribute to the pathophysiological progression of diabetic peripheral neuropathy. Diabetes 2009, 58, 2677–2686.
  62. Wang, L.; Chiang, H.C.; Wu, W.; Liang, B.; Xie, Z.; Yao, X.; Ma, W.; Du, S.; Zhong, Y. Epidermal growth factor receptor is a preferred target for treating amyloid-β-induced memory loss. Proc. Natl. Acad. Sci. USA 2012, 109, 16743–16748.
  63. Lechpammer, M.; Tran, Y.P.; Wintermark, P.; Martínez-Cerdeño, V.; Krishnan, V.V.; Ahmed, W.; Berman, R.F.; Jensen, F.E.; Nudler, E.; Zagzag, D. Upregulation of cystathionine β-synthase and p70S6K/S6 in neonatal hypoxic ischemic brain injury. Brain Pathol. 2017, 27, 449–458.
  64. Kurdi, A.; Roth, L.; Van der Veken, B.; Van Dam, D.; De Deyn, P.P.; De Doncker, M.; Neels, H.; De Meyer, G.R.Y.; Martinet, W. Everolimus depletes plaque macrophages, abolishes intraplaque neovascularization and improves survival in mice with advanced atherosclerosis. Vasc. Pharmacol. 2019, 113, 70–76.
  65. Li, D.; Wang, C.; Yao, Y.; Chen, L.; Liu, G.; Zhang, R.; Liu, Q.; Shi, F.D.; Hao, J. mTORC1 pathway disruption ameliorates brain inflammation following stroke via a shift in microglia phenotype from M1 type to M2 type. FASEB J. 2016, 30, 3388–3399.
  66. Suvanish Kumar, V.S.; Pretorius, E.; Rajanikant, G.K. The Synergistic Combination of Everolimus and Paroxetine Exerts Post-ischemic Neuroprotection In Vitro. Cell Mol. Neurobiol. 2018, 38, 1383–1397.
  67. Forouzanfar, F.; Ebrahimi, P.R.; Sadeghnia, H.R. Neuroprotection of Everolimus Against Focal Cerebral Ischemia-Reperfusion Injury in Rats. J. Stroke Cerebrovasc. Dis. 2022, 31, 106576.
  68. Cassano, T.; Magini, A.; Giovagnoli, S.; Polchi, A.; Calcagnini, S.; Pace, L.; Lavecchia, M.A.; Scuderi, C.; Bronzuoli, M.R.; Ruggeri, L.; et al. Early intrathecal infusion of everolimus restores cognitive function and mood in a murine model of Alzheimer’s disease. Exp. Neurol. 2019, 311, 88–105.
  69. Fanoudi, S.; Hosseini, M.; Alavi, M.S.; Boroushaki, M.T.; Hosseini, A.; Sadeghnia, H.R. Everolimus, a mammalian target of rapamycin inhibitor, ameliorated streptozotocin-induced learning and memory deficits via neurochemical alterations in male rats. EXCLI J. 2018, 17, 999–1017.
  70. Roscic, A.; Baldo, B.; Crochemore, C.; Marcellin, D.; Paganetti, P. Induction of autophagy with catalytic mTOR inhibitors reduces huntingtin aggregates in a neuronal cell model. J. Neurochem. 2011, 119, 398–407.
  71. Fox, J.H.; Connor, T.; Chopra, V.; Dorsey, K.; Kama, J.A.; Bleckmann, D.; Betschart, C.; Hoyer, D.; Frentzel, S.; Difiglia, M.; et al. The mTOR kinase inhibitor Everolimus decreases S6 kinase phosphorylation but fails to reduce mutant huntingtin levels in brain and is not neuroprotective in the R6/2 mouse model of Huntington’s disease. Mol. Neurodegener. 2010, 5, 26.
  72. Chen, L.; Zhang, Y.; Li, D.; Zhang, N.; Liu, R.; Han, B.; Wei, C.; Liu, H.; Xu, X.; Hao, J. Everolimus (RAD001) ameliorates vascular cognitive impairment by regulating microglial function via the mTORC1 signaling pathway. J. Neuroimmunol. 2016, 299, 164–171.
  73. Dello Russo, C.; Lisi, L.; Tringali, G.; Navarra, P. Involvement of mTOR kinase in cytokine-dependent microglial activation and cell proliferation. Biochem. Pharmacol. 2009, 78, 1242–1251.
  74. Bansal, S.; Agrawal, M.; Mahendiratta, S.; Kumar, S.; Arora, S.; Joshi, R.; Prajapat, M.; Sarma, P.; Prakash, A.; Chopra, K.; et al. Everolimus: A potential therapeutic agent targeting PI3K/Akt pathway in brain insulin system dysfunction and associated neurobehavioral deficits. Fundam. Clin. Pharmacol. 2021, 35, 1018–1031.
  75. Alavi, M.S.; Fanoudi, S.; Hosseini, A.; Jalili-Nik, M.; Bagheri, A.; Sadeghnia, H.R. Everolimus attenuates glutamate-induced PC12 cells death. Int. J. Neurosci. 2021, 17, 1–12.
  76. Han, R.; Gao, J.; Zhai, H.; Xiao, J.; Ding, Y.; Hao, J. RAD001 (everolimus) attenuates experimental autoimmune neuritis by inhibiting the mTOR pathway, elevating Akt activity and polarizing M2 macrophages. Exp. Neurol. 2016, 280, 106–114.
  77. Hoepner, R.; Bagnoud, M.; Pistor, M.; Salmen, A.; Briner, M.; Synn, H.; Schrewe, L.; Guse, K.; Ahmadi, F.; Demir, S.; et al. Vitamin D increases glucocorticoid efficacy via inhibition of mTORC1 in experimental models of multiple sclerosis. Acta Neuropathol. 2019, 138, 443–456.
  78. Schneider, M.; de Vries, P.J.; Schönig, K.; Rößner, V.; Waltereit, R. mTOR inhibitor reverses autistic-like social deficit behaviours in adult rats with both Tsc2 haploinsufficiency and developmental status epilepticus. Eur. Arch. Psychiatry Clin. Neurosci. 2017, 267, 455–463.
  79. Petrasek, T.; Vojtechova, I.; Klovrza, O.; Tuckova, K.; Vejmola, C.; Rak, J.; Sulakova, A.; Kaping, D.; Bernhardt, N.; de Vries, P.J.; et al. mTOR inhibitor improves autistic-like behaviors related to Tsc2 haploinsufficiency but not following developmental status epilepticus. J. Neurodev. Disord. 2021, 13, 14.
  80. Mishra, N.; Wang, P.; Goldsmith, D.; Zhao, X.; Xue, Y.; Christians, U.; Minassian, B.A. Everolimus does not prevent Lafora body formation in murine Lafora disease. Neurol. Genet. 2017, 3, e127.
  81. Tao, P.; Jing, Z.; Shou-Hong, G.; Shu-Juan, P.; Jian-Peng, J.; Wen-Quan, L.; Wan-Sheng, C. Effects of leptin on norepinephrine in acute ischemic stroke. Pharmazie 2019, 74, 477–480.
  82. Yue, X.; Liu, L.; Yan, H.; Gui, Y.; Zhao, J.; Zhang, P. Intracerebral Hemorrhage Induced Brain Injury Is Mediated by the Interleukin-12 Receptor in Rats. Neuropsychiatr. Dis. Treat. 2020, 16, 891–900.
  83. Zhang, Q.; Zhang, Y.; Diamond, S.; Boer, J.; Harris, J.J.; Li, Y.; Rupar, M.; Behshad, E.; Gardiner, C.; Collier, P.; et al. The Janus kinase 2 inhibitor fedratinib inhibits thiamine uptake: A putative mechanism for the onset of Wernicke’s encephalopathy. Drug Metab. Dispos. 2014, 42, 1656–1662.
  84. Hazell, A.S.; Afadlal, S.; Cheresh, D.A.; Azar, A. Treatment of rats with the JAK-2 inhibitor fedratinib does not lead to experimental Wernicke’s encephalopathy. Neurosci. Lett. 2017, 642, 163–167.
  85. Zhou, Y.; Li, C.; Li, D.; Zheng, Y.; Wang, J. IL-5 blocks apoptosis and tau hyperphosphorylation induced by Aβ(25-35) peptide in PC12 cells. J. Physiol. Biochem. 2017, 73, 259–266.
  86. Xue, W.; Zhao, Y.; Xiao, Z.; Wu, X.; Ma, D.; Han, J.; Li, X.; Xue, X.; Yang, Y.; Fang, Y.; et al. Epidermal growth factor receptor-extracellular-regulated kinase blockade upregulates TRIM32 signaling cascade and promotes neurogenesis after spinal cord injury. Stem Cells 2020, 38, 118–133.
  87. Mizuno, M.; Sotoyama, H.; Namba, H.; Shibuya, M.; Eda, T.; Wang, R.; Okubo, T.; Nagata, K.; Iwakura, Y.; Nawa, H. ErbB inhibitors ameliorate behavioral impairments of an animal model for schizophrenia: Implication of their dopamine-modulatory actions. Transl. Psychiatry 2013, 3, e252.
  88. Zheng, Y.; Shang, F.; An, L.; Zhao, H.; Liu, X. NOD2-RIP2 contributes to the inflammatory responses of mice in vivo to Streptococcus pneumoniae. Neurosci. Lett. 2018, 671, 43–49.
  89. Lopez Corcino, Y.; Gonzalez Ferrer, S.; Mantilla, L.E.; Trikeriotis, S.; Yu, J.S.; Kim, S.; Hansen, S.; Portillo, J.C.; Subauste, C.S. Toxoplasma gondii induces prolonged host epidermal growth factor receptor signalling to prevent parasite elimination by autophagy: Perspectives for in vivo control of the parasite. Cell Microbiol. 2019, 21, e13084.
  90. Ito, M.; Shichita, T.; Okada, M.; Komine, R.; Noguchi, Y.; Yoshimura, A.; Morita, R. Bruton’s tyrosine kinase is essential for NLRP3 inflammasome activation and contributes to ischaemic brain injury. Nat. Commun. 2015, 6, 7360.
  91. Jin, L.; Mo, Y.; Yue, E.L.; Liu, Y.; Liu, K.Y. Ibrutinib ameliorates cerebral ischemia/reperfusion injury through autophagy activation and PI3K/Akt/mTOR signaling pathway in diabetic mice. Bioengineered 2021, 12, 7432–7445.
  92. Yu, C.G.; Bondada, V.; Iqbal, H.; Moore, K.L.; Gensel, J.C.; Bondada, S.; Geddes, J.W. Inhibition of Bruton Tyrosine Kinase Reduces Neuroimmune Cascade and Promotes Recovery after Spinal Cord Injury. Int. J. Mol. Sci. 2021, 23, 355.
  93. Torabi, S.; Anjamrooz, S.H.; Zeraatpisheh, Z.; Aligholi, H.; Azari, H. Ibrutinib reduces neutrophil infiltration, preserves neural tissue and enhances locomotor recovery in mouse contusion model of spinal cord injury. Anat. Cell Biol. 2021, 54, 350–360.
  94. Ekpenyong-Akiba, A.E.; Poblocka, M.; Althubiti, M.; Rada, M.; Jurk, D.; Germano, S.; Kocsis-Fodor, G.; Shi, Y.; Canales, J.J.; Macip, S. Amelioration of age-related brain function decline by Bruton’s tyrosine kinase inhibition. Aging Cell 2020, 19, e13079.
  95. Lee, H.J.; Jeon, S.G.; Kim, J.; Kang, R.J.; Kim, S.M.; Han, K.M.; Park, H.; Kim, K.T.; Sung, Y.M.; Nam, H.Y.; et al. Ibrutinib modulates Aβ/tau pathology, neuroinflammation, and cognitive function in mouse models of Alzheimer’s disease. Aging Cell 2021, 20, e13332.
  96. Keaney, J.; Gasser, J.; Gillet, G.; Scholz, D.; Kadiu, I. Inhibition of Bruton’s Tyrosine Kinase Modulates Microglial Phagocytosis: Therapeutic Implications for Alzheimer’s Disease. J. Neuroimmune Pharmacol. 2019, 14, 448–461.
  97. Nam, H.Y.; Nam, J.H.; Yoon, G.; Lee, J.Y.; Nam, Y.; Kang, H.J.; Cho, H.J.; Kim, J.; Hoe, H.S. Ibrutinib suppresses LPS-induced neuroinflammatory responses in BV2 microglial cells and wild-type mice. J. Neuroinflamm. 2018, 15, 271.
  98. Ghosh, S.; Mohammed, Z.; Singh, I. Bruton’s tyrosine kinase drives neuroinflammation and anxiogenic behavior in mouse models of stress. J. Neuroinflamm. 2021, 18, 289.
  99. Li, W.; Ali, T.; He, K.; Liu, Z.; Shah, F.A.; Ren, Q.; Liu, Y.; Jiang, A.; Li, S. Ibrutinib alleviates LPS-induced neuroinflammation and synaptic defects in a mouse model of depression. Brain Behav. Immun. 2021, 92, 10–24.
  100. Zheng, M.; Li, K.; Chen, T.; Liu, S.; He, L. Geniposide protects depression through BTK/JAK2/STAT1 signaling pathway in lipopolysaccharide-induced depressive mice. Brain Res. Bull. 2021, 170, 65–73.
  101. Huggett, S.B.; Hatfield, J.S.; Walters, J.D.; McGeary, J.E.; Welsh, J.W.; Mackay, T.F.C.; Anholt, R.R.H.; Palmer, R.H.C. Ibrutinib as a potential therapeutic for cocaine use disorder. Transl. Psychiatry 2021, 11, 623.
  102. Zhan, Y.; Krafft, P.R.; Lekic, T.; Ma, Q.; Souvenir, R.; Zhang, J.H.; Tang, J. Imatinib preserves blood-brain barrier integrity following experimental subarachnoid hemorrhage in rats. J. Neurosci. Res. 2015, 93, 94–103.
  103. Shiba, M.; Fujimoto, M.; Kawakita, F.; Imanaka-Yoshida, K.; Yoshida, T.; Kanamaru, K.; Taki, W.; Suzuki, H. Effects of tenascin-C on early brain injury after subarachnoid hemorrhage in rats. Acta Neurochir. Suppl. 2015, 120, 69–73.
  104. Changlong, Z.; Guangwei, Z.; Xuenong, H.; Xiaohui, X.; Xiaochuan, S.; Yanfeng, X. The Role of Platelet-Derived Growth Factor Receptor in Early Brain Injury Following Subarachnoid Hemorrhage. J. Stroke Cerebrovasc. Dis. 2016, 25, 2203–2208.
  105. Shiba, M.; Fujimoto, M.; Imanaka-Yoshida, K.; Yoshida, T.; Taki, W.; Suzuki, H. Tenascin-C causes neuronal apoptosis after subarachnoid hemorrhage in rats. Transl. Stroke Res. 2014, 5, 238–247.
  106. Shiba, M.; Suzuki, H.; Fujimoto, M.; Shimojo, N.; Imanaka-Yoshida, K.; Yoshida, T.; Kanamaru, K.; Matsushima, S.; Taki, W. Imatinib mesylate prevents cerebral vasospasm after subarachnoid hemorrhage via inhibiting tenascin-C expression in rats. Neurobiol. Dis. 2012, 46, 172–179.
  107. Shiba, M.; Suzuki, H.; Fujimoto, M.; Shimojo, N.; Imanaka-Yoshida, K.; Yoshida, T.; Kanamaru, K.; Matsushima, S.; Taki, W. Role of platelet-derived growth factor in cerebral vasospasm after subarachnoid hemorrhage in rats. Acta Neurochir. Suppl. 2013, 115, 219–223.
  108. Ma, Q.; Huang, B.; Khatibi, N.; Rolland, W., 2nd; Suzuki, H.; Zhang, J.H.; Tang, J. PDGFR-α inhibition preserves blood-brain barrier after intracerebral hemorrhage. Ann. Neurol. 2011, 70, 920–931.
  109. Yang, P.; Manaenko, A.; Xu, F.; Miao, L.; Wang, G.; Hu, X.; Guo, Z.N.; Hu, Q.; Hartman, R.E.; Pearce, W.J.; et al. Role of PDGF-D and PDGFR-β in neuroinflammation in experimental ICH mice model. Exp. Neurol. 2016, 283, 157–164.
  110. Yang, P.; Wu, J.; Miao, L.; Manaenko, A.; Matei, N.; Zhang, Y.; Xu, L.; Pearce, W.J.; Hartman, R.E.; Obenaus, A.; et al. Platelet-Derived Growth Factor Receptor-β Regulates Vascular Smooth Muscle Cell Phenotypic Transformation and Neuroinflammation After Intracerebral Hemorrhage in Mice. Crit. Care Med. 2016, 44, e390–e402.
  111. Pearce, W.J.; Doan, C.; Carreon, D.; Kim, D.; Durrant, L.M.; Manaenko, A.; McCoy, L.; Obenaus, A.; Zhang, J.H.; Tang, J. Imatinib attenuates cerebrovascular injury and phenotypic transformation after intracerebral hemorrhage in rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2016, 311, R1093–R1104.
  112. Sun, Z.; Gao, C.; Gao, D.; Sun, R.; Li, W.; Wang, F.; Wang, Y.; Cao, H.; Zhou, G.; Zhang, J.; et al. Reduction in pericyte coverage leads to blood-brain barrier dysfunction via endothelial transcytosis following chronic cerebral hypoperfusion. Fluids Barriers CNS 2021, 18, 21.
  113. Sakai, K.; Takata, F.; Yamanaka, G.; Yasunaga, M.; Hashiguchi, K.; Tominaga, K.; Itoh, K.; Kataoka, Y.; Yamauchi, A.; Dohgu, S. Reactive pericytes in early phase are involved in glial activation and late-onset hypersusceptibility to pilocarpine-induced seizures in traumatic brain injury model mice. J. Pharmacol. Sci. 2021, 145, 155–165.
  114. Klement, W.; Blaquiere, M.; Zub, E.; deBock, F.; Boux, F.; Barbier, E.; Audinat, E.; Lerner-Natoli, M.; Marchi, N. A pericyte-glia scarring develops at the leaky capillaries in the hippocampus during seizure activity. Epilepsia 2019, 60, 1399–1411.
  115. Su, E.J.; Fredriksson, L.; Kanzawa, M.; Moore, S.; Folestad, E.; Stevenson, T.K.; Nilsson, I.; Sashindranath, M.; Schielke, G.P.; Warnock, M.; et al. Imatinib treatment reduces brain injury in a murine model of traumatic brain injury. Front. Cell Neurosci. 2015, 9, 385.
  116. Wang, J.; Bai, T.; Wang, N.; Li, H.; Guo, X. Neuroprotective potential of imatinib in global ischemia-reperfusion-induced cerebral injury: Possible role of Janus-activated kinase 2/signal transducer and activator of transcription 3 and connexin 43. Korean J. Physiol. Pharmacol. 2020, 24, 11–18.
  117. Merali, Z.; Leung, J.; Mikulis, D.; Silver, F.; Kassner, A. Longitudinal assessment of imatinib’s effect on the blood-brain barrier after ischemia/reperfusion injury with permeability MRI. Transl. Stroke Res. 2015, 6, 39–49.
  118. Gardner, L.E.; White, J.D.; Eimerbrink, M.J.; Boehm, G.W.; Chumley, M.J. Imatinib methanesulfonate reduces hyperphosphorylation of tau following repeated peripheral exposure to lipopolysaccharide. Neuroscience 2016, 331, 72–77.
  119. Weintraub, M.K.; Bisson, C.M.; Nouri, J.N.; Vinson, B.T.; Eimerbrink, M.J.; Kranjac, D.; Boehm, G.W.; Chumley, M.J. Imatinib methanesulfonate reduces hippocampal amyloid-β and restores cognitive function following repeated endotoxin exposure. Brain Behav. Immun. 2013, 33, 24–28.
  120. Sun, W.; Netzer, W.J.; Sinha, A.; Gindinova, K.; Chang, E.; Sinha, S.C. Development of Gleevec Analogues for Reducing Production of β-Amyloid Peptides through Shifting β-Cleavage of Amyloid Precursor Proteins. J. Med. Chem. 2019, 62, 3122–3134.
  121. Bauer, C.; Pardossi-Piquard, R.; Dunys, J.; Roy, M.; Checler, F. γ-Secretase-mediated regulation of neprilysin: Influence of cell density and aging and modulation by imatinib. J. Alzheimer’s Dis. 2011, 27, 511–520.
  122. Sutcliffe, J.G.; Hedlund, P.B.; Thomas, E.A.; Bloom, F.E.; Hilbush, B.S. Peripheral reduction of β-amyloid is sufficient to reduce brain β-amyloid: Implications for Alzheimer’s disease. J. Neurosci. Res. 2011, 89, 808–814.
  123. Hussain, I.; Fabrègue, J.; Anderes, L.; Ousson, S.; Borlat, F.; Eligert, V.; Berger, S.; Dimitrov, M.; Alattia, J.R.; Fraering, P.C.; et al. The role of γ-secretase activating protein (GSAP) and imatinib in the regulation of γ-secretase activity and amyloid-β generation. J. Biol. Chem. 2013, 288, 2521–2531.
  124. Netzer, W.J.; Bettayeb, K.; Sinha, S.C.; Flajolet, M.; Greengard, P.; Bustos, V. Gleevec shifts APP processing from a β-cleavage to a nonamyloidogenic cleavage. Proc. Natl. Acad. Sci. USA 2017, 114, 1389–1394.
  125. Estrada, L.D.; Chamorro, D.; Yañez, M.J.; Gonzalez, M.; Leal, N.; von Bernhardi, R.; Dulcey, A.E.; Marugan, J.; Ferrer, M.; Soto, C.; et al. Reduction of Blood Amyloid-β Oligomers in Alzheimer’s Disease Transgenic Mice by c-Abl Kinase Inhibition. J. Alzheimer’s Dis. 2016, 54, 1193–1205.
  126. Netzer, W.J.; Dou, F.; Cai, D.; Veach, D.; Jean, S.; Li, Y.; Bornmann, W.G.; Clarkson, B.; Xu, H.; Greengard, P. Gleevec inhibits beta-amyloid production but not Notch cleavage. Proc. Natl. Acad. Sci. USA 2003, 100, 12444–12449.
  127. Chu, J.; Lauretti, E.; Craige, C.P.; Praticò, D. Pharmacological modulation of GSAP reduces amyloid-β levels and tau phosphorylation in a mouse model of Alzheimer’s disease with plaques and tangles. J. Alzheimer’s Dis. 2014, 41, 729–737.
  128. Alvarez, A.R.; Sandoval, P.C.; Leal, N.R.; Castro, P.U.; Kosik, K.S. Activation of the neuronal c-Abl tyrosine kinase by amyloid-beta-peptide and reactive oxygen species. Neurobiol. Dis. 2004, 17, 326–336.
  129. Cancino, G.I.; Toledo, E.M.; Leal, N.R.; Hernandez, D.E.; Yévenes, L.F.; Inestrosa, N.C.; Alvarez, A.R. STI571 prevents apoptosis, tau phosphorylation and behavioural impairments induced by Alzheimer’s beta-amyloid deposits. Brain 2008, 131, 2425–2442.
  130. Kerridge, C.; Belyaev, N.D.; Nalivaeva, N.N.; Turner, A.J. The Aβ-clearance protein transthyretin, like neprilysin, is epigenetically regulated by the amyloid precursor protein intracellular domain. J. Neurochem. 2014, 130, 419–431.
  131. He, G.; Luo, W.; Li, P.; Remmers, C.; Netzer, W.J.; Hendrick, J.; Bettayeb, K.; Flajolet, M.; Gorelick, F.; Wennogle, L.P.; et al. Gamma-secretase activating protein is a therapeutic target for Alzheimer’s disease. Nature 2010, 467, 95–98.
  132. Reichenstein, M.; Borovok, N.; Sheinin, A.; Brider, T.; Michaelevski, I. Abelson Kinases Mediate the Depression of Spontaneous Synaptic Activity Induced by Amyloid Beta 1-42 Peptides. Cell Mol. Neurobiol. 2021, 41, 431–448.
  133. Peng, Q.; Zhang, M.; Shi, G. High-Performance Extended-Gate Field-Effect Transistor for Kinase Sensing in Aβ Accumulation of Alzheimer’s Disease. Anal. Chem. 2022, 94, 1491–1497.
  134. Ren, Y.; Chen, J.; Wu, X.; Gui, C.; Mao, K.; Zou, F.; Li, W. Role of c-Abl-GSK3β Signaling in MPP+-Induced Autophagy-Lysosomal Dysfunction. Toxicol. Sci. 2018, 165, 232–243.
  135. Wu, R.; Chen, H.; Ma, J.; He, Q.; Huang, Q.; Liu, Q.; Li, M.; Yuan, Z. c-Abl-p38α signaling plays an important role in MPTP-induced neuronal death. Cell Death Differ. 2016, 23, 542–552.
  136. Imam, S.Z.; Zhou, Q.; Yamamoto, A.; Valente, A.J.; Ali, S.F.; Bains, M.; Roberts, J.L.; Kahle, P.J.; Clark, R.A.; Li, S. Novel regulation of parkin function through c-Abl-mediated tyrosine phosphorylation: Implications for Parkinson’s disease. J. Neurosci. 2011, 31, 157–163.
  137. Yamamura, Y.; Morigaki, R.; Kasahara, J.; Yokoyama, H.; Tanabe, A.; Okita, S.; Koizumi, H.; Nagahiro, S.; Kaji, R.; Goto, S. Dopamine signaling negatively regulates striatal phosphorylation of Cdk5 at tyrosine 15 in mice. Front. Cell Neurosci. 2013, 7, 12.
  138. Pan, Y.; Sun, L.; Wang, J.; Fu, W.; Fu, Y.; Wang, J.; Tong, Y.; Pan, B. STI571 protects neuronal cells from neurotoxic prion protein fragment-induced apoptosis. Neuropharmacology 2015, 93, 191–198.
  139. Aguib, Y.; Heiseke, A.; Gilch, S.; Riemer, C.; Baier, M.; Schätzl, H.M.; Ertmer, A. Autophagy induction by trehalose counteracts cellular prion infection. Autophagy 2009, 5, 361–369.
  140. Yun, S.W.; Ertmer, A.; Flechsig, E.; Gilch, S.; Riederer, P.; Gerlach, M.; Schätzl, H.M.; Klein, M.A. The tyrosine kinase inhibitor imatinib mesylate delays prion neuroinvasion by inhibiting prion propagation in the periphery. J. Neurovirol. 2007, 13, 328–337.
  141. Ertmer, A.; Gilch, S.; Yun, S.W.; Flechsig, E.; Klebl, B.; Stein-Gerlach, M.; Klein, M.A.; Schätzl, H.M. The tyrosine kinase inhibitor STI571 induces cellular clearance of PrPSc in prion-infected cells. J. Biol. Chem. 2004, 279, 41918–41927.
  142. Rojas, F.; Gonzalez, D.; Cortes, N.; Ampuero, E.; Hernández, D.E.; Fritz, E.; Abarzua, S.; Martinez, A.; Elorza, A.A.; Alvarez, A.; et al. Reactive oxygen species trigger motoneuron death in non-cell-autonomous models of ALS through activation of c-Abl signaling. Front. Cell Neurosci. 2015, 9, 203.
  143. Kegel, K.B.; Sapp, E.; Alexander, J.; Reeves, P.; Bleckmann, D.; Sobin, L.; Masso, N.; Valencia, A.; Jeong, H.; Krainc, D.; et al. Huntingtin cleavage product A forms in neurons and is reduced by gamma-secretase inhibitors. Mol. Neurodegener. 2010, 5, 58.
  144. Nacer, A.; Movila, A.; Baer, K.; Mikolajczak, S.A.; Kappe, S.H.; Frevert, U. Neuroimmunological blood brain barrier opening in experimental cerebral malaria. PLoS Pathog 2012, 8, e1002982.
  145. Vlasic, V.; Simakajornboon, N.; Gozal, E.; Gozal, D. PDGF-beta receptor expression in the dorsocaudal brainstem parallels hypoxic ventilatory depression in the developing rat. Pediatr. Res. 2001, 50, 236–241.
  146. Yáñez, M.J.; Belbin, O.; Estrada, L.D.; Leal, N.; Contreras, P.S.; Lleó, A.; Burgos, P.V.; Zanlungo, S.; Alvarez, A.R. c-Abl links APP-BACE1 interaction promoting APP amyloidogenic processing in Niemann-Pick type C disease. Biochim. Biophys. Acta 2016, 1862, 2158–2167.
  147. Marín, T.; Dulcey, A.E.; Campos, F.; de la Fuente, C.; Acuña, M.; Castro, J.; Pinto, C.; Yañez, M.J.; Cortez, C.; McGrath, D.W.; et al. c-Abl Activation Linked to Autophagy-Lysosomal Dysfunction Contributes to Neurological Impairment in Niemann-Pick Type A Disease. Front. Cell Dev. Biol. 2022, 10, 844297.
  148. Yañez, M.J.; Campos, F.; Marín, T.; Klein, A.D.; Futerman, A.H.; Alvarez, A.R.; Zanlungo, S. c-Abl activates RIPK3 signaling in Gaucher disease. Biochim. Biophys. Acta Mol. Basis Dis. 2021, 1867, 166089.
  149. Potula, R.; Dhillion, N.; Sui, Y.; Zien, C.A.; Funa, K.; Pinson, D.; Mayo, M.S.; Singh, D.K.; Narayan, O.; Buch, S. Association of platelet-derived growth factor-B chain with simian human immunodeficiency virus encephalitis. Am. J. Pathol. 2004, 165, 815–824.
  150. Jia, X.; Zhang, A.; Li, Z.; Peng, X.; Tian, X.; Gao, F. Activation of spinal PDGFRβ in microglia promotes neuronal autophagy via p38 MAPK pathway in morphine-tolerant rats. J. Neurochem. 2021, 158, 373–390.
  151. Jia, J.N.; Yin, X.X.; Li, Q.; Guan, Q.W.; Yang, N.; Chen, K.N.; Zhou, H.H.; Mao, X.Y. Neuroprotective Effects of the Anti-cancer Drug Lapatinib Against Epileptic Seizures via Suppressing Glutathione Peroxidase 4-Dependent Ferroptosis. Front. Pharmacol. 2020, 11, 601572.
  152. Xu, H.Y.; Sun, Y.J.; Sun, Y.Y.; Wu, Y.J.; Xu, M.Y.; Chen, L.P.; Zhu, L. Lapatinib alleviates TOCP-induced axonal damage in the spinal cord of mouse. Neuropharmacology 2021, 189, 108535.
  153. Mansour, H.M.; Fawzy, H.M.; El-Khatib, A.S.; Khattab, M.M. Lapatinib ditosylate rescues memory impairment in D-galactose/ovariectomized rats: Potential repositioning of an anti-cancer drug for the treatment of Alzheimer’s disease. Exp. Neurol. 2021, 341, 113697.
  154. Mansour, H.M.; Fawzy, H.M.; El-Khatib, A.S.; Khattab, M.M. Inhibition of mitochondrial pyruvate carrier 1 by lapatinib ditosylate mitigates Alzheimer’s-like disease in D-galactose/ovariectomized rats. Neurochem. Int. 2021, 150, 105178.
  155. Zavvarian, M.M.; Hong, J.; Khazaei, M.; Chio, J.C.T.; Wang, J.; Badner, A.; Fehlings, M.G. The Protein Kinase Inhibitor Midostaurin Improves Functional Neurological Recovery and Attenuates Inflammatory Changes Following Traumatic Cervical Spinal Cord Injury. Biomolecules 2021, 11, 972.
  156. Dent, P.; Booth, L.; Roberts, J.L.; Poklepovic, A.; Cridebring, D.; Reiman, E.M. Inhibition of heat shock proteins increases autophagosome formation, and reduces the expression of APP, Tau, SOD1 G93A and TDP-43. Aging Albany NY 2021, 13, 17097–17117.
  157. Attia, G.M.; Elmansy, R.A.; Elsaed, W.M. Neuroprotective effect of nilotinib on pentylenetetrazol-induced epilepsy in adult rat hippocampus: Involvement of oxidative stress, autophagy, inflammation, and apoptosis. Folia Neuropathol. 2019, 57, 146–160.
  158. Lonskaya, I.; Hebron, M.; Chen, W.; Schachter, J.; Moussa, C. Tau deletion impairs intracellular β-amyloid-42 clearance and leads to more extracellular plaque deposition in gene transfer models. Mol. Neurodegener. 2014, 9, 46.
  159. Liu, X.; Hebron, M.; Shi, W.; Lonskaya, I.; Moussa, C.E. Ubiquitin specific protease-13 independently regulates parkin ubiquitination and alpha-synuclein clearance in alpha-synucleinopathies. Hum. Mol. Genet. 2019, 28, 548–560.
  160. Mahul-Mellier, A.L.; Fauvet, B.; Gysbers, A.; Dikiy, I.; Oueslati, A.; Georgeon, S.; Lamontanara, A.J.; Bisquertt, A.; Eliezer, D.; Masliah, E.; et al. c-Abl phosphorylates α-synuclein and regulates its degradation: Implication for α-synuclein clearance and contribution to the pathogenesis of Parkinson’s disease. Hum. Mol. Genet. 2014, 23, 2858–2879.
  161. Imberdis, T.; Negri, J.; Ramalingam, N.; Terry-Kantor, E.; Ho, G.P.H.; Fanning, S.; Stirtz, G.; Kim, T.E.; Levy, O.A.; Young-Pearse, T.L.; et al. Cell models of lipid-rich α-synuclein aggregation validate known modifiers of α-synuclein biology and identify stearoyl-CoA desaturase. Proc. Natl. Acad. Sci. USA 2019, 116, 20760–20769.
  162. Adlimoghaddam, A.; Odero, G.G.; Glazner, G.; Turner, R.S.; Albensi, B.C. Nilotinib Improves Bioenergetic Profiling in Brain Astroglia in the 3xTg Mouse Model of Alzheimer’s Disease. Aging Dis. 2021, 12, 441–465.
  163. La Barbera, L.; Vedele, F.; Nobili, A.; Krashia, P.; Spoleti, E.; Latagliata, E.C.; Cutuli, D.; Cauzzi, E.; Marino, R.; Viscomi, M.T.; et al. Nilotinib restores memory function by preventing dopaminergic neuron degeneration in a mouse model of Alzheimer’s Disease. Prog. Neurobiol. 2021, 202, 102031.
  164. Nobili, A.; La Barbera, L.; D’Amelio, M. Targeting autophagy as a therapeutic strategy to prevent dopamine neuron loss in early stages of Alzheimer disease. Autophagy 2021, 17, 1278–1280.
  165. Wu, J.; Xu, X.; Zheng, L.; Mo, J.; Jin, X.; Bao, Y. Nilotinib inhibits microglia-mediated neuroinflammation to protect against dopaminergic neuronal death in Parkinson’s disease models. Int. Immunopharmacol. 2021, 99, 108025.
  166. Kuo, Y.C.; Tsai, H.C.; Rajesh, R. Glutathione Liposomes Carrying Ceftriaxone, FK506, and Nilotinib to Control Overexpressed Dopamine Markers and Apoptotic Factors in Neurons. ACS Biomater. Sci. Eng. 2021, 7, 3242–3255.
  167. Kim, H.; Shin, J.Y.; Jo, A.; Kim, J.H.; Park, S.; Choi, J.Y.; Kang, H.C.; Dawson, V.L.; Dawson, T.M.; Shin, J.H.; et al. Parkin interacting substrate phosphorylation by c-Abl drives dopaminergic neurodegeneration. Brain 2021, 144, 3674–3691.
  168. Peikert, K.; Federti, E.; Matte, A.; Constantin, G.; Pietronigro, E.C.; Fabene, P.F.; Defilippi, P.; Turco, E.; Del Gallo, F.; Pucci, P.; et al. Therapeutic targeting of Lyn kinase to treat chorea-acanthocytosis. Acta Neuropathol. Commun. 2021, 9, 81.
  169. Federti, E.; Matte, A.; Riccardi, V.; Peikert, K.; Alper, S.L.; Danek, A.; Walker, R.H.; Siciliano, A.; Iatcenko, I.; Hermann, A.; et al. Adaptative Up-Regulation of PRX2 and PRX5 Expression Characterizes Brain from a Mouse Model of Chorea-Acanthocytosis. Antioxidants 2021, 11, 76.
  170. Hor, J.H.; Soh, E.S.; Tan, L.Y.; Lim, V.J.W.; Santosa, M.M.; Winanto; Ho, B.X.; Fan, Y.; Soh, B.S.; Ng, S.Y. Cell cycle inhibitors protect motor neurons in an organoid model of Spinal Muscular Atrophy. Cell Death Dis. 2018, 9, 1100.
  171. Chao, A.C.; Chen, C.H.; Chang, S.H.; Huang, C.T.; Hwang, W.C.; Yang, D.I. Id1 and Sonic Hedgehog Mediate Cell Cycle Reentry and Apoptosis Induced by Amyloid Beta-Peptide in Post-mitotic Cortical Neurons. Mol. Neurobiol. 2019, 56, 465–489.
  172. Esteras, N.; Alquézar, C.; Bartolomé, F.; de la Encarnación, A.; Bermejo-Pareja, F.; Molina, J.A.; Martín-Requero, Á. G1/S Cell Cycle Checkpoint Dysfunction in Lymphoblasts from Sporadic Parkinson’s Disease Patients. Mol. Neurobiol. 2015, 52, 386–398.
  173. Javidnia, M.; Hebron, M.L.; Xin, Y.; Kinney, N.G.; Moussa, C.E. Pazopanib Reduces Phosphorylated Tau Levels and Alters Astrocytes in a Mouse Model of Tauopathy. J. Alzheimer’s Dis. 2017, 60, 461–481.
  174. Yang, Y.; Li, G.; Zhao, D.; Yu, H.; Zheng, X.; Peng, X.; Zhang, X.; Fu, T.; Hu, X.; Niu, M.; et al. Computational discovery and experimental verification of tyrosine kinase inhibitor pazopanib for the reversal of memory and cognitive deficits in rat model neurodegeneration. Chem. Sci. 2015, 6, 2812–2821.
  175. Li, M.; Li, Z.; Ren, H.; Jin, W.N.; Wood, K.; Liu, Q.; Sheth, K.N.; Shi, F.D. Colony stimulating factor 1 receptor inhibition eliminates microglia and attenuates brain injury after intracerebral hemorrhage. J. Cereb. Blood Flow Metab. 2017, 37, 2383–2395.
  176. Shi, E.; Shi, K.; Qiu, S.; Sheth, K.N.; Lawton, M.T.; Ducruet, A.F. Chronic inflammation, cognitive impairment, and distal brain region alteration following intracerebral hemorrhage. FASEB J. 2019, 33, 9616–9626.
  177. Heinz, R.; Brandenburg, S.; Nieminen-Kelhä, M.; Kremenetskaia, I.; Boehm-Sturm, P.; Vajkoczy, P.; Schneider, U.C. Microglia as target for anti-inflammatory approaches to prevent secondary brain injury after subarachnoid hemorrhage (SAH). J. Neuroinflamm. 2021, 18, 36.
  178. Shen, Q.; Zhang, G. Depletion of microglia mitigates cerebrovascular dysfunction in diet-induced obesity mice. Am. J. Physiol. Endocrinol. Metab. 2021, 321, E367–E375.
  179. Rice, R.A.; Spangenberg, E.E.; Yamate-Morgan, H.; Lee, R.J.; Arora, R.P.; Hernandez, M.X.; Tenner, A.J.; West, B.L.; Green, K.N. Elimination of Microglia Improves Functional Outcomes Following Extensive Neuronal Loss in the Hippocampus. J. Neurosci. 2015, 35, 9977–9989.
  180. Shi, Y.; Manis, M.; Long, J.; Wang, K.; Sullivan, P.M.; Remolina Serrano, J.; Hoyle, R.; Holtzman, D.M. Microglia drive APOE-dependent neurodegeneration in a tauopathy mouse model. J. Exp. Med. 2019, 216, 2546–2561.
  181. Sosna, J.; Philipp, S.; Albay, R., 3rd; Reyes-Ruiz, J.M.; Baglietto-Vargas, D.; LaFerla, F.M.; Glabe, C.G. Early long-term administration of the CSF1R inhibitor PLX3397 ablates microglia and reduces accumulation of intraneuronal amyloid, neuritic plaque deposition and pre-fibrillar oligomers in 5XFAD mouse model of Alzheimer’s disease. Mol. Neurodegener. 2018, 13, 11.
  182. Son, Y.; Jeong, Y.J.; Shin, N.R.; Oh, S.J.; Nam, K.R.; Choi, H.D.; Choi, J.Y.; Lee, H.J. Inhibition of Colony-Stimulating Factor 1 Receptor by PLX3397 Prevents Amyloid Beta Pathology and Rescues Dopaminergic Signaling in Aging 5xFAD Mice. Int. J. Mol. Sci. 2020, 21, 5553.
  183. Crapser, J.D.; Ochaba, J.; Soni, N.; Reidling, J.C.; Thompson, L.M.; Green, K.N. Microglial depletion prevents extracellular matrix changes and striatal volume reduction in a model of Huntington’s disease. Brain 2020, 143, 266–288.
  184. Tahmasebi, F.; Pasbakhsh, P.; Mortezaee, K.; Madadi, S.; Barati, S.; Kashani, I.R. Effect of the CSF1R inhibitor PLX3397 on remyelination of corpus callosum in a cuprizone-induced demyelination mouse model. J. Cell Biochem. 2019, 120, 10576–10586.
  185. Groh, J.; Klein, D.; Berve, K.; West, B.L.; Martini, R. Targeting microglia attenuates neuroinflammation-related neural damage in mice carrying human PLP1 mutations. Glia 2019, 67, 277–290.
  186. Tahmasebi, F.; Pasbakhsh, P.; Barati, S.; Madadi, S.; Kashani, I.R. The effect of microglial ablation and mesenchymal stem cell transplantation on a cuprizone-induced demyelination model. J. Cell Physiol. 2021, 236, 3552–3564.
  187. Qu, W.; Johnson, A.; Kim, J.H.; Lukowicz, A.; Svedberg, D.; Cvetanovic, M. Inhibition of colony-stimulating factor 1 receptor early in disease ameliorates motor deficits in SCA1 mice. J. Neuroinflamm. 2017, 14, 107.
  188. Pinto, B.; Morelli, G.; Rastogi, M.; Savardi, A.; Fumagalli, A.; Petretto, A.; Bartolucci, M.; Varea, E.; Catelani, T.; Contestabile, A.; et al. Rescuing Over-activated Microglia Restores Cognitive Performance in Juvenile Animals of the Dp(16) Mouse Model of Down Syndrome. Neuron 2020, 108, 887–904.e12.
  189. Ueta, Y.; Miyata, M. Brainstem local microglia induce whisker map plasticity in the thalamus after peripheral nerve injury. Cell Rep. 2021, 34, 108823.
  190. da Silva, M.C.M.; Gomes, G.F.; de Barros Fernandes, H.; da Silva, A.M.; Teixeira, A.L.; Moreira, F.A.; de Miranda, A.S.; de Oliveira, A.C.P. Inhibition of CSF1R, a receptor involved in microglia viability, alters behavioral and molecular changes induced by cocaine. Sci. Rep. 2021, 11, 15989.
  191. Zhang, D.; Li, S.; Hou, L.; Jing, L.; Ruan, Z.; Peng, B.; Zhang, X.; Hong, J.S.; Zhao, J.; Wang, Q. Microglial activation contributes to cognitive impairments in rotenone-induced mouse Parkinson’s disease model. J. Neuroinflamm. 2021, 18, 4.
  192. Tian, J.; Guo, S.; Chen, H.; Peng, J.J.; Jia, M.M.; Li, N.S.; Zhang, X.J.; Yang, J.; Luo, X.J.; Peng, J. Combination of Emricasan with Ponatinib Synergistically Reduces Ischemia/Reperfusion Injury in Rat Brain Through Simultaneous Prevention of Apoptosis and Necroptosis. Transl. Stroke Res. 2018, 9, 382–392.
  193. Zhang, Y.; Zhang, M.; Zhu, W.; Pan, X.; Wang, Q.; Gao, X.; Wang, C.; Zhang, X.; Liu, Y.; Li, S.; et al. Role of Elevated Thrombospondin-1 in Kainic Acid-Induced Status Epilepticus. Neurosci. Bull. 2020, 36, 263–276.
  194. Choi, J.P.; Wang, R.; Yang, X.; Wang, X.; Wang, L.; Ting, K.K.; Foley, M.; Cogger, V.; Yang, Z.; Liu, F.; et al. Ponatinib (AP24534) inhibits MEKK3-KLF signaling and prevents formation and progression of cerebral cavernous malformations. Sci. Adv. 2018, 4, eaau0731.
  195. Zhang, J.; Xu, X.; Zhou, D.; Li, H.; You, W.; Wang, Z.; Chen, G. Possible Role of Raf-1 Kinase in the Development of Cerebral Vasospasm and Early Brain Injury After Experimental Subarachnoid Hemorrhage in Rats. Mol. Neurobiol. 2015, 52, 1527–1539.
  196. Hsieh, C.H.; Lin, Y.J.; Chen, W.L.; Huang, Y.C.; Chang, C.W.; Cheng, F.C.; Liu, R.S.; Shyu, W.C. HIF-1α triggers long-lasting glutamate excitotoxicity via system x(c)(-) in cerebral ischaemia-reperfusion. J. Pathol. 2017, 241, 337–349.
  197. Wang, T.; Li, B.; Wang, Z.; Wang, X.; Xia, Z.; Ning, G.; Wang, X.; Zhang, Y.; Cui, L.; Yu, M.; et al. Sorafenib promotes sensory conduction function recovery via miR-142-3p/AC9/cAMP axis post dorsal column injury. Neuropharmacology 2019, 148, 347–357.
  198. Echeverria, V.; Burgess, S.; Gamble-George, J.; Zeitlin, R.; Lin, X.; Cao, C.; Arendash, G.W. Sorafenib inhibits nuclear factor kappa B, decreases inducible nitric oxide synthase and cyclooxygenase-2 expression, and restores working memory in APPswe mice. Neuroscience 2009, 162, 1220–1231.
  199. Kim, J.; Park, J.H.; Park, S.K.; Hoe, H.S. Sorafenib Modulates the LPS- and Aβ-Induced Neuroinflammatory Response in Cells, Wild-Type Mice, and 5xFAD Mice. Front. Immunol. 2021, 12, 684344.
  200. Liu, Z.; Hamamichi, S.; Lee, B.D.; Yang, D.; Ray, A.; Caldwell, G.A.; Caldwell, K.A.; Dawson, T.M.; Smith, W.W.; Dawson, V.L. Inhibitors of LRRK2 kinase attenuate neurodegeneration and Parkinson-like phenotypes in Caenorhabditis elegans and Drosophila Parkinson’s disease models. Hum. Mol. Genet. 2011, 20, 3933–3942.
  201. Moawad, E.Y. Induction of multiple sclerosis and response to tyrosine kinase inhibitors. Indian J. Clin. Biochem. 2014, 29, 491–495.
  202. Crespo, O.; Kang, S.C.; Daneman, R.; Lindstrom, T.M.; Ho, P.P.; Sobel, R.A.; Steinman, L.; Robinson, W.H. Tyrosine kinase inhibitors ameliorate autoimmune encephalomyelitis in a mouse model of multiple sclerosis. J. Clin. Immunol. 2011, 31, 1010–1020.
  203. Martina, B.E.E.; Smreczak, M.; Orlowska, A.; Marzec, A.; Trebas, P.; Roose, J.M.; Zmudzinski, J.; Gerhauser, I.; Wohlsein, P.; Baumgärtner, W.; et al. Combination drug treatment prolongs survival of experimentally infected mice with silver-haired bat rabies virus. Vaccine 2019, 37, 4736–4742.
  204. Brahms, A.; Mudhasani, R.; Pinkham, C.; Kota, K.; Nasar, F.; Zamani, R.; Bavari, S.; Kehn-Hall, K. Sorafenib Impedes Rift Valley Fever Virus Egress by Inhibiting Valosin-Containing Protein Function in the Cellular Secretory Pathway. J. Virol. 2017, 91, e00968-17.
  205. Lundberg, L.; Brahms, A.; Hooper, I.; Carey, B.; Lin, S.C.; Dahal, B.; Narayanan, A.; Kehn-Hall, K. Repurposed FDA-Approved drug sorafenib reduces replication of Venezuelan equine encephalitis virus and other alphaviruses. Antivir. Res. 2018, 157, 57–67.
  206. Gao, M.; Duan, H.; Liu, J.; Zhang, H.; Wang, X.; Zhu, M.; Guo, J.; Zhao, Z.; Meng, L.; Peng, Y. The multi-targeted kinase inhibitor sorafenib inhibits enterovirus 71 replication by regulating IRES-dependent translation of viral proteins. Antiviral Res. 2014, 106, 80–85.
  207. Welsbie, D.S.; Ziogas, N.K.; Xu, L.; Kim, B.J.; Ge, Y.; Patel, A.K.; Ryu, J.; Lehar, M.; Alexandris, A.S.; Stewart, N.; et al. Targeted disruption of dual leucine zipper kinase and leucine zipper kinase promotes neuronal survival in a model of diffuse traumatic brain injury. Mol. Neurodegener. 2019, 14, 44.
  208. Benini, R.; Roth, R.; Khoja, Z.; Avoli, M.; Wintermark, P. Does angiogenesis play a role in the establishment of mesial temporal lobe epilepsy? Int. J. Dev. Neurosci. 2016, 49, 31–36.
  209. Lee, J.C.; Kim, H.Y.; Lee, S.; Shin, J.; Kim, H.V.; Kim, K.; Baek, S.; Lee, D.; Jeon, H.; Kim, D.; et al. Discovery of Chemicals to Either Clear or Indicate Amyloid Aggregates by Targeting Memory-Impairing Anti-Parallel Aβ Dimers. Angew. Chem. Int. Ed. Engl. 2020, 59, 11491–11500.
  210. Grammas, P.; Martinez, J.; Sanchez, A.; Yin, X.; Riley, J.; Gay, D.; Desobry, K.; Tripathy, D.; Luo, J.; Evola, M.; et al. A new paradigm for the treatment of Alzheimer’s disease: Targeting vascular activation. J. Alzheimer’s Dis. 2014, 40, 619–630.
  211. Son, S.M.; Jung, E.S.; Shin, H.J.; Byun, J.; Mook-Jung, I. Aβ-induced formation of autophagosomes is mediated by RAGE-CaMKKβ-AMPK signaling. Neurobiol. Aging 2012, 33, 1006.e11–1006.e23.
  212. Wrasidlo, W.; Crews, L.A.; Tsigelny, I.F.; Stocking, E.; Kouznetsova, V.L.; Price, D.; Paulino, A.; Gonzales, T.; Overk, C.R.; Patrick, C.; et al. Neuroprotective effects of the anti-cancer drug sunitinib in models of HIV neurotoxicity suggests potential for the treatment of neurodegenerative disorders. Br. J. Pharmacol. 2014, 171, 5757–5773.
  213. Fields, J.A.; Metcalf, J.; Overk, C.; Adame, A.; Spencer, B.; Wrasidlo, W.; Florio, J.; Rockenstein, E.; He, J.J.; Masliah, E. The anticancer drug sunitinib promotes autophagyand protects from neurotoxicity in an HIV-1 Tat model of neurodegeneration. J. Neurovirol. 2017, 23, 290–303.
  214. Pu, S.Y.; Xiao, F.; Schor, S.; Bekerman, E.; Zanini, F.; Barouch-Bentov, R.; Nagamine, C.M.; Einav, S. Feasibility and biological rationale of repurposing sunitinib and erlotinib for dengue treatment. Antiviral Res. 2018, 155, 67–75.
  215. Luo, J.; Zhang, Y.; Wang, Y.; Liu, Q.; Chen, L.; Zhang, B.; Luo, Y.; Huang, S.; Guo, X. Rhabdovirus Infection Is Dependent on Serine/Threonine Kinase AP2-Associated Kinase 1. Life 2020, 10, 170.
  216. Cordaro, M.; Paterniti, I.; Siracusa, R.; Impellizzeri, D.; Esposito, E.; Cuzzocrea, S. KU0063794, a Dual mTORC1 and mTORC2 Inhibitor, Reduces Neural Tissue Damage and Locomotor Impairment After Spinal Cord Injury in Mice. Mol. Neurobiol. 2017, 54, 2415–2427.
  217. Siracusa, R.; Paterniti, I.; Cordaro, M.; Crupi, R.; Bruschetta, G.; Campolo, M.; Cuzzocrea, S.; Esposito, E. Neuroprotective Effects of Temsirolimus in Animal Models of Parkinson’s Disease. Mol. Neurobiol. 2018, 55, 2403–2419.
  218. Decressac, M.; Björklund, A. mTOR inhibition alleviates L-DOPA-induced dyskinesia in parkinsonian rats. J. Parkinson’s Dis. 2013, 3, 13–17.
  219. Jiang, T.; Yu, J.T.; Zhu, X.C.; Zhang, Q.Q.; Cao, L.; Wang, H.F.; Tan, M.S.; Gao, Q.; Qin, H.; Zhang, Y.D.; et al. Temsirolimus attenuates tauopathy in vitro and in vivo by targeting tau hyperphosphorylation and autophagic clearance. Neuropharmacology 2014, 85, 121–130.
  220. Frederick, C.; Ando, K.; Leroy, K.; Héraud, C.; Suain, V.; Buée, L.; Brion, J.P. Rapamycin ester analog CCI-779/Temsirolimus alleviates tau pathology and improves motor deficit in mutant tau transgenic mice. J. Alzheimer’s Dis. 2015, 44, 1145–1156.
  221. Jiang, T.; Yu, J.T.; Zhu, X.C.; Tan, M.S.; Wang, H.F.; Cao, L.; Zhang, Q.Q.; Shi, J.Q.; Gao, L.; Qin, H.; et al. Temsirolimus promotes autophagic clearance of amyloid-β and provides protective effects in cellular and animal models of Alzheimer’s disease. Pharmacol. Res. 2014, 81, 54–63.
  222. Menzies, F.M.; Huebener, J.; Renna, M.; Bonin, M.; Riess, O.; Rubinsztein, D.C. Autophagy induction reduces mutant ataxin-3 levels and toxicity in a mouse model of spinocerebellar ataxia type 3. Brain 2010, 133, 93–104.
  223. Saravia, R.; Flores, Á.; Plaza-Zabala, A.; Busquets-Garcia, A.; Pastor, A.; de la Torre, R.; Di Marzo, V.; Marsicano, G.; Ozaita, A.; Maldonado, R.; et al. CB(1) Cannabinoid Receptors Mediate Cognitive Deficits and Structural Plasticity Changes During Nicotine Withdrawal. Biol. Psychiatry 2017, 81, 625–634.
  224. Launay, N.; Aguado, C.; Fourcade, S.; Ruiz, M.; Grau, L.; Riera, J.; Guilera, C.; Giròs, M.; Ferrer, I.; Knecht, E.; et al. Autophagy induction halts axonal degeneration in a mouse model of X-adrenoleukodystrophy. Acta Neuropathol. 2015, 129, 399–415.
  225. Konoeda, F.; Shichita, T.; Yoshida, H.; Sugiyama, Y.; Muto, G.; Hasegawa, E.; Morita, R.; Suzuki, N.; Yoshimura, A. Therapeutic effect of IL-12/23 and their signaling pathway blockade on brain ischemia model. Biochem. Biophys. Res. Commun. 2010, 402, 500–506.
  226. Yang, L.; Liu, Y.; Wang, Y.; Li, J.; Liu, N. Azeliragon ameliorates Alzheimer’s disease via the Janus tyrosine kinase and signal transducer and activator of transcription signaling pathway. Clinics 2021, 76, e2348.
  227. Zhou, Y.; Leng, X.; Luo, S.; Su, Z.; Luo, X.; Guo, H.; Mo, C.; Zou, Q.; Liu, Y.; Wang, Y. Tolerogenic Dendritic Cells Generated with Tofacitinib Ameliorate Experimental Autoimmune Encephalomyelitis through Modulation of Th17/Treg Balance. J. Immunol. Res. 2016, 2016, 5021537.
  228. Günaydın, C.; Önger, M.E.; Avcı, B.; Bozkurt, A.; Terzi, M.; Bilge, S.S. Tofacitinib enhances remyelination and improves myelin integrity in cuprizone-induced mice. Immunopharmacol. Immunotoxicol. 2021, 43, 790–798.
  229. Alshammari, A.; Alharbi, M.; Albekairi, N.A.; Albekairi, T.H.; Alharbi, O.O.; Yeapuri, P.; Singh, S. Protective Effect of CP690550 in MPTP-Induced Parkinson’s Like Behavioural, Biochemical and Histological Alterations in Mice. Neurotox. Res. 2022, 40, 564–572.
  230. Figueroa-Romero, C.; Monteagudo, A.; Murdock, B.J.; Famie, J.P.; Webber-Davis, I.F.; Piecuch, C.E.; Teener, S.J.; Pacut, C.; Goutman, S.A.; Feldman, E.L. Tofacitinib Suppresses Natural Killer Cells In Vitro and In Vivo: Implications for Amyotrophic Lateral Sclerosis. Front. Immunol. 2022, 13, 773288.
  231. Risner, K.; Ahmed, A.; Bakovic, A.; Kortchak, S.; Bhalla, N.; Narayanan, A. Efficacy of FDA-Approved Anti-Inflammatory Drugs Against Venezuelan Equine Encephalitis Virus Infection. Viruses 2019, 11, 1151.
  232. Huang, Y.; Li, Q.; Tian, H.; Yao, X.; Bakina, O.; Zhang, H.; Lei, T.; Hu, F. MEK inhibitor trametinib attenuates neuroinflammation and cognitive deficits following traumatic brain injury in mice. Am. J. Transl. Res. 2020, 12, 6351–6365.
  233. Christensen, S.T.; Haanes, K.A.; Spray, S.; Grell, A.S.; Warfvinge, K.; Edvinsson, L.; Johansson, S.E. Pre-clinical effects of highly potent MEK1/2 inhibitors on rat cerebral vasculature after organ culture and subarachnoid haemorrhage. Clin. Sci. 2019, 133, 1797–1811.
  234. Park, E.S.; Kim, S.; Huang, S.; Yoo, J.Y.; Körbelin, J.; Lee, T.J.; Kaur, B.; Dash, P.K.; Chen, P.R.; Kim, E. Selective Endothelial Hyperactivation of Oncogenic KRAS Induces Brain Arteriovenous Malformations in Mice. Ann. Neurol. 2021, 89, 926–941.
  235. Ballabh, P.; Xu, H.; Hu, F.; Braun, A.; Smith, K.; Rivera, A.; Lou, N.; Ungvari, Z.; Goldman, S.A.; Csiszar, A.; et al. Angiogenic inhibition reduces germinal matrix hemorrhage. Nat. Med. 2007, 13, 477–485.
  236. Rezzola, S.; Guerra, J.; Krishna Chandran, A.M.; Loda, A.; Cancarini, A.; Sacristani, P.; Semeraro, F.; Presta, M. VEGF-Independent Activation of Müller Cells by the Vitreous from Proliferative Diabetic Retinopathy Patients. Int. J. Mol. Sci. 2021, 22, 2179.
More
Academic Video Service