Experimental and clinical tests of FDA-approved kinase inhibitors for the treatment of neurological disorders (update 2024)
Sections
Open Access Review
Experimental and clinical tests of FDA-approved kinase inhibitors for the treatment of neurological disorders (update 2024)

Affiliation:

1Department of Neurology, University of California, Davis, CA 95616, USA

Hassan Aliashrafzadeh
1

Affiliation:

1Department of Neurology, University of California, Davis, CA 95616, USA

Dewey Liu
1

Affiliation:

1Department of Neurology, University of California, Davis, CA 95616, USA

Samantha De Alba
1

Affiliation:

2Ayub Medical College, Abbottabad 22020, Pakistan

Imad Akbar
2

Affiliation:

1Department of Neurology, University of California, Davis, CA 95616, USA

Austin Lui
1

Affiliation:

1Department of Neurology, University of California, Davis, CA 95616, USA

Jordan Vanleuven
1

Affiliation:

1Department of Neurology, University of California, Davis, CA 95616, USA

3Department of Neurological Surgery, University of California, Davis, CA 95616, USA

Ryan Martin
1,3

Affiliation:

4Institute for Psychedelics and Neurotherapeutics, University of California, Davis, CA 95616, USA

5Department of Chemistry, University of California, Davis, CA 95616, USA

ORCID: https://orcid.org/0000-0001-5659-0036

Zhang Wang
4,5

Affiliation:

1Department of Neurology, University of California, Davis, CA 95616, USA

6Mirnova Therapeutics, Inc., Davis, CA 95618, USA

Email: djliu@mirnovatherapeutics.com

ORCID: https://orcid.org/0000-0002-3934-9651

Da Zhi Liu
1,6*

Explor Drug Sci. 2025;3:1008116 DOl: https://doi.org/10.37349/eds.2025.1008116

Received: February 05, 2025 Accepted: May 27, 2025 Published: July 01, 2025

Academic Editor: Fernando Albericio, University of KwaZulu-Natal, South Africa, Universidad de Barcelona, Spain

The article belongs to the special issue Leveraging the FDA-Approved Kinase Inhibitors to Treat Neurological Disorders

Abstract

Since our previous summary of the 74 FDA-approved kinase inhibitors in clinical and preclinical trials for non-cancerous neurological treatment, the US FDA has approved 13 additional kinase inhibitors since early 2022. This update incorporates new evidence for the now 87 FDA-approved kinase inhibitors in clinical and preclinical trials for the treatment of non-cancerous neurological disorders. By the end of October 2024, nearly all 87 FDA-approved kinase inhibitors have been tested in various animal models of non-cancerous neurological disorders, with twenty entered into clinical trials and six used for off-label treatments of neurological conditions in humans. Considering the challenges posed by intellectual property (IP), legal considerations, and limited blood-brain barrier (BBB) permeability, which may restrict some FDA-approved kinase inhibitors from effectively targeting the central nervous system (CNS), we further discuss the feasibility of designing novel proprietary analogs with enhanced BBB penetration to improve their therapeutic potential in neurological disorders. The new drugs typically retain full IP rights and remain costly; while repurposing kinase inhibitors may provide effective and affordable treatments for non-cancerous neurological disorders.

Keywords

Kinase inhibitors, aberrant cell cycle, clinical trials, preclinical experiments, neurological disorders

Introduction

We developed an original concept of “Aberrant Cell Cycle Disease (ACCD)”, revealing that two major types of diseases (cancers and neurological diseases) share the same mechanism of “aberrant cell cycle re-entry” [1]. The aberrant cell cycle is implicated in both the unlimited proliferation of tumor cells in cancers and the post-mitotic death of mature neurons in non-cancerous neurological disorders [13]. Considering that aberrant cell cycle re-entry is manifested as oncogenic kinase activation and/or tumor suppressor activation, the cell cycle arrest (e.g., inhibiting oncogenic kinase, elevating tumor suppressors) that is widely practiced for cancer treatment can be leveraged for the treatment of non-cancerous neurological disorders.

Given human genome encodes ~500 protein kinases and a single kinase often has tens of artificial/synthetic inhibitors, there are likely thousands of synthetic kinase inhibitors. Among those, the FDA has approved a total of 87 kinase inhibitors, mostly for cancer therapy by the end of October 2024, with 74 approved by 2021 and 13 thereafter (i.e., abrocitinib, capivasertib, others) (Supplementary material). Unlike cancer drug development, the advancement of neurologically effective drugs for human use has seen limited success, despite neurological disorders being life-threatening and impacting millions of people worldwide each year [46]. Utilizing the ACCD concept, we previously reviewed the 74 kinase inhibitors that the FDA had approved by 2021 [2], summarizing their clinical and preclinical applications for neurological disorders and assessing their potential for repurposing in non-cancerous neurological treatments.

In this update, we incorporate new evidence of using the FDA-approved kinase inhibitors for non-cancerous neurological treatment. In addition to intellectual property (IP) and legal matters, the poor blood-brain barrier (BBB) permeability of some kinase inhibitors limits the repurposed use for neurological treatment, except for acute brain injuries [e.g., traumatic brain injury (TBI), intracerebral hemorrhage (ICH), subarachnoid hemorrhage (SAH)] that are characterized by immediate BBB disruption. To overcome these barriers, this review also discusses the strategy of structure modification of FDA-approved kinase inhibitors to synthesize new compounds of full IP and improved BBB permeability for the treatment of non-cancerous neurological disorders. We searched each individual kinase inhibitor with each subtype of neurological disorder in the databases (https://pubmed.ncbi.nlm.nih.gov; https://clinicaltrials.gov) for published preclinical experiments, clinical trials, and off-label use of the 87 FDA-approved kinase inhibitors for neurological treatments. Of note, non-FDA-approved kinase inhibitors and neurological cancers (e.g., glioblastoma, brain metastasis from peripheral cancers, neuroblastoma) are beyond the scope of this review.

Kinase, kinase inhibitors, and FDA-approved kinase inhibitors and indications

Kinases, existing universally in various species, have predominantly been thought as oncogenes involving tumorigenesis for many decades [7, 8], in spite of the opposite evidence that shows several kinases [e.g., protein kinase C (PKC), mitogen-activated protein kinase kinase 4 (MAPKK4/MEK4), and death-associated protein kinase 3 (DAPK3)] function as tumor suppressors [7]. Kinases catalyze various reactions of phosphorylation where the high-energy molecules (e.g., ATP) donate phosphoryl groups to substrate molecules [9, 10], with approximately 13,000 human proteins having phosphorylation sites [11]. The transfer of the phosphoryl group from one molecule to another is a fundamental process in many aspects of metabolism, gene regulation, signal transduction, and others [12]. Regarding target substrates, human protein kinases are classified into three types: serine-threonine kinases (STKs), tyrosine kinases (TKs), and dual specificity kinases (STKs/TKs). According to the presence or absence of transmembrane receptor structure, TKs are further divided into receptor TKs (RTKs) and non-RTKs (NRTKs).

There are an estimated thousands of synthetic kinase inhibitors, as the human genome encodes ~500 protein kinases while a single kinase often has tens of artificial/synthetic inhibitors. For example, Src kinase has an increasing list of its synthetic inhibitors: dasatinib, bosutinib, saracatinib, WH-4-023, ENMD-2076, PP1, PP2, pelitinib, ponatinib, PP121, tirbanibulin, SU6656, repotrectinib, UM-164, apatinib, eCF506, elzovantinib, DGY-06-116, 1-NM-PP1, TPX-0046, TL02-59, other.

As for the non-FDA-approved kinase inhibitors, it was reported that bruton TK (BTK) inhibitors (i.e., evobrutinib, fenebrutinib, orelabrutinib, remibrutinib, tolebrutinib) had been tested in clinical trials for treatment of multiple sclerosis (MS) [13]. In addition, a number of non-FDA-approved kinase inhibitors have shown efficacy for the treatment of neurological disorders in experimental models, such as Src family kinase (SFK) inhibitors (PP2, saracatinib) [1418], rho-associated protein kinase (ROCK) inhibitor (Y-27632) [1921], extracellular signal-regulated kinase (ERK) inhibitor (PD98059) [22], cyclin-dependent kinases (CDKs) inhibitors (flavopiridol, roscovitine) [2327], and others. Therefore, many kinase inhibitors (whether FDA-approved or not) can be used to treat both cancers and neurological disorders, which in turn strongly supports the ACCD concept that cancers and neurological disorders share similar underlying pathophysiological mechanisms.

Clinical trials of FDA-approved kinase inhibitors for neurological treatment

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 treatments (Supplementary material). Acalabrutinib, a BTK inhibitor, is currently undergoing investigation in a phase II clinical trial (NCT05065554) to assess the safety and efficacy of its combination with rituximab or other CD20 antibodies for individuals suffering from neuropathies associated with immunoglobulin M monoclonal gammopathy of undetermined significance (IgM-MGUS) or Waldenstrom macroglobulinemia (WM). Alpelisib, an inhibitor of PI3K, is in an ongoing phase II clinical trial (NCT05577754) in pediatric and adult patients with megalencephaly-capillary malformation polymicrogyria syndrome (MCAP).

Baricitinib, classified as a JAK inhibitor, has finished a phase II trial (NCT03921554) and is presently being evaluated in a phase II/III trial (NCT04517253) focused on determining the safety and efficacy of baricitinib for Aicardi-Goutieres syndrome (AGS), a genetic disorder that results in profound mental and physical disabilities in infants. This drug was studied in a phase I/II trial (NCT05792462) assessing the efficacy and safety of baricitinib in neuromyelitis optica spectrum disorders (NMOSD). In another phase III trial (NCT06631287) in patients with long COVID, baricitinib was studied to determine if it can improve neurocognitive function, measures of physical function, quality of life, post-exertional malaise, the effect of breathlessness on daily activities, post-COVID-19 symptom burden, and biomarkers of inflammation and viral measures. The effect of baricitinib administration on the outcome of participants with moderate and severe traumatic ICH/contusions was studied in a phase II clinical trial (NCT06065046). In another trial (NCT04378621), the anti-inflammatory effect of baricitinib was investigated on neuropsychiatric symptoms and morphological changes in the brain caused by rheumatoid arthritis (RA). Other ongoing trials include phase II/III trials (NCT06548802) assessing the efficacy and safety of baricitinib in patients with pulmonary injury after ICH, and a phase II study (NCT05452564) evaluating the efficacy and safety of baricitinib for the reduction of human immunodeficiency virus (HIV) in the central nervous system (CNS). In addition, the role of this drug in CNS mechanisms of anhedonia and psychomotor slowing in depressed people with HIV was investigated in a phase II trial (NCT05849038). Lastly, a phase I/II trial (NCT05189106) assessed the safety of baricitinib in Alzheimer’s disease (AD) and amyotrophic lateral sclerosis (ALS). There is also a case report describing positive effects of baricitinib for the treatment of ocular myasthenia gravis (MG) in a 58-year-old woman who received baricitinib for alopecia areata [28]. The effects of baricitinib, empagliflozin, linagliptin, and telmisartan on cardiovascular autonomic neuropathy in type 1 diabetes were studied in a randomized, crossover trial [29]. The drug bosutinib, known for its inhibitory effects on Src and BCR-ABL, has been the focus of multiple clinical trials addressing various neurodegenerative disorders. Among these trials is a phase I study (NCT04744532) evaluating its application in ALS, along with another ongoing phase I trial (NCT02921477) that investigates its potential benefits for dementia and mild cognitive impairment. Moreover, a completed phase II trial (NCT03888222) has specifically targeted Lewy body dementia (LBD).

In a phase II trial (NCT04079179), Cobimetinib, known for its role as a MAPKK/MEK inhibitor, has been researched for its safety and efficacy in addressing histiocytic disorders that could potentially lead to neurodegeneration. Dasatinib, which acts as an inhibitor of BCR-ABL, Kit, Src, EphA2, EGFR, and PDGFR, has been evaluated in combination with quercetin, a flavonoid recognized for its anti-inflammatory and antioxidant characteristics, in multiple completed trials (NCT04063124 phase I and II, NCT05422885 phase I and II) and is currently under investigation in ongoing trials (phase I and II in NCT04785300, phase II in NCT04685590) for the management of mild cognitive impairment and AD [30, 31]. Extensive clinical trials have investigated Everolimus, an agent that acts as an inhibitor of both FKBP and mTOR, in relation to different types of acute brain injuries, as well as neurodegenerative and neurodevelopmental disorders. Such instances include the completion of NCT03198949 (a phase II trial) of Everolimus for epilepsy and focal cortical dysplasia (FCD), and another completed phase I/II trial (NCT02991807) of Everolimus for hamartoma tumor syndrome. A phase II trial (NCT01997255) that is presently underway is assessing the safety and efficacy of Everolimus for patients experiencing seizures linked to Sturge-Weber syndrome. A total of twelve studies, which include both finished and current trials, are examining the safety and efficacy of Everolimus for patients suffering from tuberous sclerosis complex (TSC), which is commonly connected with FCD, cognitive disabilities, autism, intractable seizures, self-injurious behavior, and a range of other neurocognitive challenges (NCT01070316, NCT01713946, NCT02451696, NCT01929642, NCT01289912, NCT02962414, NCT01730209, and NCT01954693), and a terminated phase II clinical trial study (NCT00857259) evaluated the safety profile and therapeutic efficacy of Everolimus, used alone or alongside ranibizumab, in patients affected by neo-vascular age-related macular degeneration. Imatinib, which acts as an inhibitor of Kit, BCR-ABL, and PDGFR, has been studied in various acute brain injury conditions as well as neurodegenerative diseases. A phase III clinical trial (NCT03639922) evaluated the addition of Imatinib to standard acute stroke treatment, focusing on its potential to enhance functional recovery. Furthermore, a phase II clinical trial (NCT02363361) is examining the safety, absorption, and tolerability of Imatinib in individuals with cervical spinal cord injury (SCI). A separate phase II clinical trial (NCT03674099) is currently evaluating Imatinib as an innovative treatment for MS, assessing its effectiveness in comparison to methylprednisolone, which is the standard treatment for relapses of MS. Additionally, there was an intended phase I clinical trial (NCT00403156) that aimed to explore the application of Imatinib in the treatment of choroidal neovascularization; however, this study has been retracted. Nilotinib functions as a kinase inhibitor, targeting the activities of PDGFR, BCR-ABL, and DDR1. It has undergone investigation into various neurodegenerative disorders through clinical trials. Currently, a phase II trial (NCT04002674) is exploring Nilotinib’s effects on patients with dementia with Lewy bodies, focusing on pharmacokinetics, tolerability, biomarkers, and safety. Also, a phase I trial (NCT03764215) involved administering Nilotinib to individuals diagnosed with Huntington’s disease (HD). The study evaluated biomarkers, including levels of phosphorylated tau, alongside functional outcomes. Currently, a phase III clinical trial (NCT05143528) is underway to assess the safety and effectiveness of Nilotinib in early AD patients, utilizing two distinct dosages. A phase II clinical trial (NCT02947893) investigated the effectiveness of Nilotinib in individuals diagnosed with AD. The study specifically measured the impact of Nilotinib on cell death using various cell markers. Additionally, the concentrations of amyloid in the brain were evaluated through positron emission tomography (PET) imaging. Furthermore, there are three trials, both completed and ongoing, that focus on the effects of Nilotinib in patients with Parkinson’s disease (PD) (NCT03205488, NCT02954978, and NCT02281474). Pazopanib, an inhibitor of PDGFR α/β, FGFR 1/3, VEGFR 1/2/3, Itk, Lck, Fms, and Kit, has been tested in multiple completed or terminated trials (NCT01154062, NCT00659555, NCT00612456, NCT00463320, NCT01072214, NCT01134055, NCT01051700, NCT01362348, and NCT00733304) for macular degeneration. Pirtobrutinib, a BTK inhibitor, was studied in a phase II clinical trial to assess its efficacy and safety in participants with MS (NCT06104683), though the trial has been withdrawn. Regorafenib, a VEGF kinase inhibitor, successfully passed phase I trial for neovascular age-related macular degeneration. The phase IIa trial study (NCT02222207) was terminated as its results did not meet the effectiveness of the current standard of care [32].

Sirolimus, which functions as an mTOR inhibitor and was first used as an immunosuppressant in kidney transplants, has been incorporated into various clinical trials targeting neurological and psychiatric conditions, including epilepsy (NCT03486366, NCT03646240), cerebral aneurysms (NCT04141020), age-related macular degeneration (NCT02732899, NCT01445548, NCT00766649, NCT01675947, NCT00712491, NCT00304954, NCT02357342, and NCT00766337), AD (NCT04629495, NCT06022068, and NCT04200911), ALS (NCT03359538), frontotemporal dementia (NCT04408625), MS (NCT00095329), PD (NCT04127578), refractory epilepsy (NCT05613166) multiple system atrophy (MSA) [33], (NCT03589976), diabetic retinopathy (NCT00711490), diabetic macular edema (NCT00401115 and NCT00656643), Sturge-Weber syndrome (NCT03047980, NCT02080624), TSC (NCT05104983, NCT04595513, NCT02061397, NCT01929642, and NCT00490789), Lysosomal diseases (NCT03952637), Leigh syndrome (LS) (NCT03747328), Gaucher disease type 2 (NCT04411654), alcohol use disorder (NCT03732248), smoking cessation (NCT04161144), depression (NCT02487485), stroke prevention (NCT04948749), myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) (NCT06257420), communicating hydrocephalus secondary to ICH (NCT06563817), post-traumatic stress disorder (PTSD) (NCT01449955), surgically refractory epilepsy (NCT03646240), extracranial vertebral artery stenosis (NCT05885932), ischemic stroke (IS) (NCT02578069), intracranial atherosclerotic stenosis (NCT05719883, NCT06614972, and NCT04949880), and mild cognitive impairment (NCT04200911).

Overall, in the context of clinical trials, four of them have successfully completed phase I (NCT00401115, NCT03732248, NCT03646240, and NCT04200911). Fourteen trials have advanced to complete phases I/II or II (NCT01445548, NCT02357342, NCT02732899, NCT00766649, NCT00304954, NCT02080624, NCT01929642, NCT00656643, NCT04161144, NCT02487485, NCT02061397, NCT00711490, NCT03359538, and NCT04595513). One trial has successfully concluded in phase II/III (NCT03047980), six trials have been withdrawn or terminated during phases I/II (NCT00095329, NCT01675947, NCT03589976, NCT00712491, NCT00766337, and NCT03747328), two trials are currently in phase II with an unknown status (NCT00490789, NCT05613166). As a TK inhibitor, sunitinib has been evaluated in two completed phase I/II trials targeting neovascular age-related macular degeneration (NCT03249740) and diabetic macular edema associated with retinal vein occlusion (NCT04085341). Temsirolimus, known for being a prodrug of sirolimus, inhibits the mTOR signaling pathway, has a completed phase II trial for the treatment of relapsing-remitting MS (NCT00228397). Tofacitinib, known for its role as a Janus kinase enzyme inhibitor, has been involved in multiple clinical trials. one of which is an ongoing phase I trial for MG patients (NCT04431895). An early phase I clinical trial (NCT06689982) explored the potential of tofacitinib in treating ALS and also a terminated phase I/II clinical trial of tofacitinib focused on treatment-resistant depression (NCT04141904). Another completed phase II trial focused on the application of tofacitinib in individuals with Down syndrome (DS) to address various skin conditions, including alopecia areata, atopic dermatitis/eczema, and psoriasis (NCT04246372). Trametinib, a MEK inhibitor, was used in a terminated phase I/II clinical trial for ALS (NCT04326283). Upadacitinib, a JAK1 selective inhibitor, is currently going through a phase III trial investigating its use in the treatment of giant cell arteritis (NCT03725202), comparing the efficacy of upadacitinib plus corticosteroids versus corticosteroids alone. Zanubrutinib, a BTK inhibitor, is being tested in an ongoing phase II trial (NCT05356858) for the treatment of NMOSD, a medical condition characterized by the immune system’s attack on the optic nerves and spinal cord. The clinical trial demonstrated that BTK activity is increased in both B cells and microglia, indicating that this medication plays a role in improving the pathology associated with NMOSD [34].

Off-label use of FDA-approved kinase inhibitors for neurological treatment

Although the FDA has not approved any kinase inhibitors for the treatment of neurological disorders, several FDA-approved kinase inhibitors have been used off-label. For example, Everolimus has been documented for the treatment of patients with TSC-associated refractory seizures [35], epilepsy [36], hemimegalencephaly-related epilepsy [37], pediatric TSC-related epilepsy [38], and FCD [39]. Ibrutinib has been used for the treatment of patients with white matter injury from graft-versus-host disease (GVHD) [40], anti-myelin-associated glycoprotein neuropathy [41], and MS [42], while Imatinib has been used for the treatment of patients with BBB malfunction in DiGeorge syndrome [43], and in IS [44]. With mixed results, ruxolitinib and baricitinib were recently used in the treatment of AGS, an immune-mediated neurological disease refractory to conventional immunosuppression [45]. Tofacitinib has also been reported in the treatment of refractory autoimmune encephalitis, again with mixed results [46].

Preclinical trials of FDA-approved kinase inhibitors for neurological treatment

A total of 20 FDA-approved kinase inhibitors have undergone evaluation in clinical trials focused on neurological disorders; however, there exists a significant number of preclinical investigations examining the impact of these FDA-approved kinase inhibitors on neurological conditions. Research has been conducted on abemaciclib in preclinical settings to explore its potential for treating motor neuron degeneration [47], PTSD [48], and AD [49, 50]. Abrocitinib, an inhibitor of JAK1, JAK2, JAK3, and Tyk2, has been investigated for its neuroinflammation and neuroprotective effects on TBI [51]. In preclinical studies, Afatinib has been assessed for its potential to treat neuroinflammation resulting from a lack of oxygen and glucose [52], MS [53], nicotine dependence [54], TSC [54], and cortical brain injuries [55]. The efficacy of Axitinib has been investigated as a possible treatment option for AD [56], IS [57, 58], and diabetes-induced neuropathy [59]. The potential of Alectinib as a therapeutic intervention for binge drinking has been explored [60, 61] and IS [62]. Preclinical studies have investigated the use of baricitinib as a potential therapeutic option for neurocognitive disorders resulting from HIV infection [63], encephalitis [64, 65], MS [65], hypersensitivity in DS [66], SCI [67], Hutchinson-Gilford progeria [68], AD [6971], AGS [7275], ALS [76], neuropathic pain [77], type 1 diabetes mellitus peripheral neuropathy and bone denervation [78] and chronic itching induced by frontal cortex neurons [79]. Research conducted in a preclinical setting suggests that Binimetinib could be a viable treatment for specific variants of AD [80]. The efficacy of Bosutinib has been investigated as a possible treatment option for ICH [81], cerebral ischemia [82], PD [8386], AD [87], transactive response DNA binding protein 43 (TDP-43) pathology [88], stress-activated protein kinase interacting protein 1 (SIN1)-mediated neurotoxicity [89], botulinum neurotoxins [90], LBD [91, 92], and ALS [9395]. Cabozantinib has undergone evaluation for its potential use in treating Rett syndrome (RTT) [96], and AD [9799]. Crizotinib has undergone evaluation for its potential use in treating PD [100], AD [101], persistent pain [102], Toxoplasma gondii [103], and the condition known as craniosynostosis, which is associated with Saethre-Chotzen syndrome, involves the early closure of sutures in the skull, potentially resulting in abnormal head shapes and neurological complications [104].

Research has been conducted on Dabrafenib to assess its viability as a treatment of IS [105], SCI [106], PD [107, 108], ataxia caused by neurohistiocytosis of the cerebellum [109], and brain arteriovenous malformation (BAVM) [110]. Dasatinib has been tested for the potential treatment of lipopolysaccharide (LPS)-induced neuroinflammation [111], kainic acid-induced neuroinflammation [112], glaucoma [113], tau‐associated pathology [114], MS [115], ALS [116118], PD [117], cognitive dysfunction [119, 120], obesity-induced anxiety [121], chronic unpredictable stress that can induce cognitive deficits [122], fetal alcohol syndrome [123], botulinum neurotoxins [90], AD [124], endotoxemia [125], and IS [126]. In combination with quercetin, Dasatinib has been reported for treatment of tauopathy [127], seizure [128, 129], cognitive deficits [130, 131], TBI [132], AD [133137], TBI [138], motor neuron disease [118], and brain aging models [139143]. Deucravacitinib, a selective Tyk2 inhibitor, has been proven to inhibit monocyte activation [144], which may be useful in treating neuroinflammation. Research has been conducted on Duvelisib to assess its effectiveness in addressing peripheral neuropathy caused by Paclitaxel [145]. Erlotinib has undergone evaluation for its potential use in treating nerve fiber damage [146], intracranial aneurysm formation [147], ALS [148], diabetic peripheral neuropathy [149, 150], and memory loss [151]. The efficacy of Everolimus has been investigated as a possible treatment for encephalopathy of prematurity [152], atherosclerosis-associated brain hypoxia [153], IS [154156], AD [157, 158], HD [159, 160], vascular dementia [161], LPS-induced neuroinflammation [162], insulin-dysfunction-related cognitive dysfunction [163], glutamate-induced neurotoxicity [164], Guillain-Barre’ syndrome [165], MS [166], TSC-associated autism-like social deficits [167, 168], Lafora disease [169], TSC-related epileptic encephalopathy [170173], ICH [174], white matter injury [175], and hyperthermia-induced seizures [176] and neuroinflammation associated with seizures [177, 178].

Fedratinib has been evaluated for the potential treatment of IS [179], ICH [180], Wernicke’s encephalopathy [181, 182], and AD [183]. Fostamatinib was recently examined in experimental models for the treatment of schizophrenia [184] and ALS [185]. As a highly selective inhibitor, Fruquintinib specifically targets the TKs VEGFR 1, VEGFR 2, and VEGFR 3, has been carried out to assess the potential treatment options for cerebral amyloid angiopathy (CAA) [186]. The efficacy of gefitinib has been investigated as a possible treatment for SCIs [187], amyloid-β-induced memory loss [151], schizophrenia [188], Streptococcus pneumoniae meningitis [189], Toxoplasma gondii (that is capable of inducing symptoms that are characteristic of congenital neurological conditions and meningoencephalitis) [103, 190], IS [191], AD [192], and epilepsy [193].

The efficacy of Ibrutinib has been assessed in relation to the treatment of IS [194, 195], SCI [196, 197], age-related cognitive deterioration [198], AD [98, 199, 200], LPS-induced neuroinflammation [201], anxiogenic behavior [202], depression [203, 204]. Cocaine use disorder [205], SAH [206], and ALS [207]. Imatinib has been tested for the potential treatment of SAH [208213], ICH [214218], cerebral small vessel disease [219], TBI-induced seizure [220], seizures [220222], TBI [223], IS [224226], AD [227244], PD [245250], prion diseases [251254], ALS [255], HD [256], cerebral malaria [257], Hypoxic ventilatory depression (that is characterized by a diminished ventilatory response triggered by hypoxemia, or low oxygen levels in the blood) [258], Niemann-Pick type C disease [259], Niemann-Pick type A disease [260], Gaucher disease [261], simian HIV encephalitis [262], morphine tolerance [263], BBB dysfunction in chronic cerebral hypoperfusion [264], and glutamate-induced oxidative injury [265]. Infigratinib has been evaluated as a prospective treatment option for diabetic retinopathy [266] and MS [267].

Lapatinib has been assessed for the potential treatment of epilepsy [268], organophosphate induced axonal damage in the spinal cord (it highlights the potential for neurotoxic effects that can compromise motor function and overall neural health) [269], AD [270, 271], PD [272], and BBB disruption in SARS-CoV-2 [273]. The efficacy of Lorlatinib has been investigated as a possible treatment option for persistent pain [102]. Midostaurin has been assessed for the potential treatment of traumatic SCI [274] and AD [275]. Neratinib has been tested for the potential treatment of AD [276, 277]. Netarsudil has been tested for the potential treatment of optic nerve degeneration [278]. Nilotinib has been tested for the potential treatment of Epileptic seizures [279], tauopathies [83, 84, 280], alpha-synucleinopathies [84, 281283], TDP-43 pathology [85, 88], Beta-amyloid pathology [280], AD [69, 284286], PD [287289], chorea-acanthocytosis [290, 291], Niemann-Pick type A disease [260], LPS-induced cognitive impairment and neuroinflammation [292].

Pacritinib, an inhibitor of JAK1, JAK2, JAK3, and Tyk2, has been used to explore the potential interventions for RTT [293], HIV-associated neurological disorders, and also neurodegenerative diseases [294]. The efficacy of Palbociclib has been investigated as a possible therapeutic option for Spinal Muscular Atrophy (SMA) [295], amyloid beta-peptide pathology [296], and PD [297]. Research has been conducted on pazopanib to assess its efficacy in the treatment of tauopathy [298], neurodegeneration-induced memory and cognitive deficits [299], osteoarthritis pain [300], PD [301, 302], and AD [303]. Pexidartinib has been tested for the potential treatment of ICH [304306], SAH [307], obesity-related cerebrovascular dysfunction [308], cognitive decline due to brain damage [309], tauopathy [310], AD [311, 312], HD [313], MS [314316], spinocerebellar ataxia type 1 (SCA1) that is a genetic, neurodegenerative disorder that primarily affects the cerebellum [317], DS (also known as trisomy 21) [318], peripheral nerve injury induced mechanical hypersensitivity (also known as allodynia or hyperesthesia) [319], cocaine addiction [320], PD [321323], psychiatric disorders like schizophrenia [324], cerebellar ataxias [325], LS [326, 327], opioid use disorder [328], psychiatric disorders such as depression [329], TBI [330], and neuropathic pain [331]. Ponatinib has been assessed for the potential treatment of IS [332], epilepsy [333], and cerebral cavernous malformation (abnormal blood vessel clusters within the brain may result in various neurological manifestations, including seizures, headaches, and the potential for stroke) [334]. Regorafenib has been assessed for the potential treatment of AD [335]. The efficacy of Ruxolitinib has been investigated as a possible treatment option for PD [336], MS [337339], DS [340], dysfunction of the BBB induced by cytokines [341], HIV-related neurocognitive disorders [342], depressive-like behaviors and cognitive impairments [343], TBI [344, 345], IS [346], SCI [347, 348], AD with TDP-43 inclusions [70], and COVID-19 induced peripheral and CNS disease [349].

Selumetinib has been tested for the potential treatment of frontotemporal lobar degeneration [350], obsessive-compulsive disorder [351], acrolein-induced neurotoxicity [352], and ICH [353]. Sirolimus has been assessed for the potential treatment of IS [154, 354388], TBI [389396], SAH [397403], SCI [404415], germinal matrix hemorrhage (GMH) [416, 417], ICH [418420], seizure-induced memory deficits [421424], seizure in LS [425], spinal cord ischemia [426, 427], preganglionic cervical root transection [428], optic nerve crush [429], alveolar nerve transection [430], ischemic retinal disease [431], MS [432442], PD [443446], cerebral palsy (CP) [447], prion disease [448450], AD [451455], vascular dementia (that is a type of cognitive impairment that occurs due to reduced blood flow to the brain) [456], diabetes-related pathology that mimics AD [457], diabetes-induced neuropathology [458], HD [459464], macular degeneration [465], optic nerve degeneration [466], neurodegeneration of retina [467], cadmium-induced neurodegeneration [468471], spiral ganglion neurons degeneration [472], tauopathy [473, 474], synucleinopathy [475477], MG [478, 479], iron-induced cognitive impairments [480], cognitive impairments resulting from intermittent hypoxia [481], cognitive impairments resulting from cannabinoid exposure [482], diabetic perioperative neurocognitive disorders [483], ethanol-induced neurodegeneration [484], neurodegeneration associated with aging [485], methylmercury-induced neurotoxicity [486], TDP-43 proteinopathy [487], ALS [488], autism spectrum disorder (ASD) [489496], autism-associated behavioral disorders [497], Krabbe disease [498], trisomy 21 [499502], intellectual disability [503], fetal alcohol spectrum disorders [504506], autism linked to TS [507510], TSC [511514], TSC-associated neuropsychiatric disorders (TAND) [515], neurodevelopmental defects in TSC [516, 517], cognitive deficits in TSC [518, 519], Koolen-de Vries syndrome (that is a hereditary condition marked by delays in development, cognitive impairments, and unique facial characteristics) [520], FCD [521], Schaaf-Yang syndrome [522], cerebral malaria [523525], neuropathic pain [526], seizure-induced anxiety [527], anxiety disorders [528530], obesity-induced anxiety and depression [531], diabetes mellitus-related cognitive deficits [532534], nicotine addiction [535], alcohol-related disorders [516, 536, 537]. Herpes simplex virus encephalitis [538], mania [539], epilepsy [172, 540543], epilepsy-induced anxiety and depression [544], porcine hemagglutinating encephalomyelitis virus [545], photochemical damage in retinal photoreceptor cells [546], multisystem proteinopathy [547], N-methyl-D-aspartate (NMDA)-induced retinal damage [548550], adverse optineurin phenotypes [551], hydrocephalus [552], sleep disorders [553], depression [554], drug-seeking behavior [555557], Inflammation of the nervous system resulting from aging [558], mitochondrial encephalopathy [559], TANC2 mutation-induced neuropsychiatric disorders [560], impact of general anesthesia on neurodevelopmental disorders in individuals with fragile-X syndrome [561]. The behavioral manifestations of depression and anxiety induced by H. pylori infection [562], epileptic brain injury [563], age-related hearing loss [564], MSA [565], brain hypervascularization [566], sepsis-induced cognitive impairment [567], polyhydramnios, megalencephaly, and symptomatic epilepsy syndrome [568].

FCD type II induced epileptic seizures [569]. Sorafenib has been tested for the potential treatment of SAH [570], IS [571], SCI [572], AD [97, 573, 574], PD [575], MS [576, 577], rabies [578], Rift Valley fever (RVF) virus [579], alphaviruses [580], and picornavirus enterovirus 71 [581]. Sunitinib has been tested for the potential treatment of TBI [582], seizure [583], AD [584587], RTT [96], cognitive impairment associated with HIV [588, 589], Dengue virus [590], rabies [591], and optic nerve injury [592].

Temsirolimus has been assessed for the potential treatment of SCI [410], PD [593, 594], tauopathy [595, 596], AD [597], SCA3 [598], nicotine withdrawal-associated cognitive deficits [599], and X-linked adrenoleukodystrophy (a genetic disorder primarily affecting the nervous system and adrenal glands) [600]. Tofacitinib has been evaluated for the potential treatment for IS [601], AD [602], MS [603, 604], PD [605], ALS [606], Venezuelan equine encephalitis virus (VEEV) [607], SCI [608], neuropsychiatric lupus (NPSLE) [609]. Trametinib has been tested for the potential treatment of TBI [610], aneurysmal SAH [611], BAVMs [612], ALS [613, 614], AD [615, 616], demyelinating disease [617]. Vandetanib has been tested for the potential treatment of GMH [618]. Vemurafenib has been tested for the treatment of laminin-α2-related congenital muscular dystrophy (LAMA2-CMD), which is a neuromuscular disease (LAMA2-CMD) [619].

Structure modification for new chemical entities with improved BBB penetration

While repurposing FDA-approved kinase inhibitors for CNS disorders accelerates the clinical trials, early kinase inhibitors, which were often not developed to treat CNS disorders, exhibit limited selectivity and off-target side effects. Therefore, developing new BBB-penetrating chemical entities such as kinase inhibitors will provide a more targeted treatment of kinase-related CNS disorders. Because of the unique structure and function of the BBB, such as the tight junction and efflux pumps, small-molecule CNS drugs usually possess different structural features from non-CNS drugs from the medicinal chemistry perspective. For example, other than using Lipinski’s guidelines to design oral small-molecule drugs, Pfizer chemists have provided a more focused CNS multiparameter optimization (MPO) score to guide BBB-penetrating small-molecule discovery [620, 621]. The CNS MPO score evaluates a small molecule’s properties of cLogP (preferably < 3), cLogD (< 2), molecular weight (< 360), pKa (< 8), topological polar surface area (between 40 to 90), and the number of hydrogen-bond donors (< 2). Usually, a CNS MPO score of four or above indicates a higher probability of BBB penetration. It is a common strategy in drug discovery to improve the physiological properties of existing non-BBB-penetrating drugs by structural modifications so that the new chemical entities possess better BBB permeability. In this way, a therapeutic concentration can be reached in the CNS while the plasma concentration is maintained relatively low. In the next paragraph, we highlight some recent developments in CNS-penetrating kinase inhibitors that originate from FDA-approved drugs.

Erlotinib, a first-generation EGFR inhibitor with a low BBB penetration and an unbound brain-to-plasma partition coefficient (Kp,uu,brain) of 0.05, was approved in the US to treat non-small cell lung cancer and pancreatic cancer. Structural optimization of erlotinib led to the discovery of JCN037 which has substantially improved BBB penetration with a Kp,uu,brain of 1.30 [622]. The structural modifications include rigidification of the molecular architecture by shortening the water-exposed ether chain of erlotinib as well as replacing the alkyne group with a bromine atom to improve the BBB-penetration. Ibrutinib, the first FDA-approved BTK inhibitor for the treatment of multiple cancers, such as leukemia and lymphoma, cannot accumulate in the CNS due to drug efflux by P-glycoprotein (P-gp) [623]. However, structural modifications have resulted in a new molecule, tolebrutinib [624], which can effectively penetrate the BBB. The major structural difference between Ibrutinib and tolebrutinib is the hinge-binding group: Ibrutinib utilizes a pyrimidine substructure, and tolebrutinib employs a more lipophilic pyridine substructure. Additionally, the FDA-approved BRAFV600E inhibitor vemurafenib cannot cross the BBB, but after removing the flexible 4-chlorophenyl group and reducing the molecular weight, a new BRAFV600E inhibitor, PF-07284890 (ARRY-461), was developed with improved BBB penetration [625]. Its phase Ia/b clinical trial (NCT04543188) was terminated recently due to an internal business decision, but not major safety concerns or requests from any regulatory authorities. Lastly, AZD0156, an ataxia-telangiectasia mutated (ATM) kinase inhibitor, shows an efflux ratio of 23.7 in MDCKII-MDR1-BCRP assays and is unlikely to penetrate the BBB. By increasing the lipophilicity of AZD0156, a new compound, AZD1390, exhibited substantially increased BBB penetration with an efflux ratio of 1.8 in MDCKII-MDR1-BCRP assays, unlikely to engage the efflux pump [626].

Conclusions

By the end of October 2024, nearly all 87 FDA-approved kinase inhibitors have been tested in various animal models of non-cancerous neurological disorders, with twenty entered into clinical trials and six used for off-label treatments for non-cancerous neurological disorders in humans. More kinase inhibitors are expected to enter the pipeline of clinical trials for neurological indications. As compared to the new drugs that typically retain full IP rights and remain costly, repurposing kinase inhibitors, if proven successful, could offer effective and affordable treatments for non-cancerous neurological disorders.

Abbreviations

ACCD: Aberrant Cell Cycle Disease

AD: Alzheimer’s disease

AGS: Aicardi-Goutieres syndrome

ALS: amyotrophic lateral sclerosis

BAVM: brain arteriovenous malformation

BBB: blood-brain barrier

BTK: bruton tyrosine kinase

CNS: central nervous system

DS: Down syndrome

FCD: focal cortical dysplasia

GMH: germinal matrix hemorrhage

HD: Huntington’s disease

HIV: human immunodeficiency virus

ICH: intracerebral hemorrhage

IP: intellectual property

IS: ischemic stroke

Kp,uu,brain: unbound brain-to-plasma partition coefficient

LAMA2-CMD: laminin-α2-related congenital muscular dystrophy

LBD: Lewy body dementia

LPS: lipopolysaccharide

LS: Leigh syndrome

MAPKK4/MEK4: mitogen-activated protein kinase kinase 4

MG: myasthenia gravis

MPO: multiparameter optimization

MS: multiple sclerosis

MSA: multiple system atrophy

NMOSD: neuromyelitis optica spectrum disorders

PD: Parkinson’s disease

PTSD: post-traumatic stress disorder

RTKs: receptor tyrosine kinases

RTT: Rett syndrome

SAH: subarachnoid hemorrhage

SCA1: spinocerebellar ataxia type 1

SCI: spinal cord injury

STKs: serine-threonine kinases

TBI: traumatic brain injury

TDP-43: transactive response DNA binding protein 43

TKs: tyrosine kinases

TSC: tuberous sclerosis complex

Supplementary materials

Other supplementary material for this article is available at: https://www.explorationpub.com/uploads/Article/file/1008116_sup_1.xlsx.

Declarations

Author contributions

HA, DL, SDA, IA, AL, JV, RM, and ZW: Writing—review & editing. DZL Conceptualization, Writing—review & editing, Funding acquisition. All authors have read and agreed to the published version of the manuscript.

Conflicts of interest

Da Zhi Liu who is the Editorial Board Member of Exploration of Drug Science and Guest Editor of Leveraging the FDA-Approved Kinase Inhibitors to Treat Neurological Disorders had no involvement in the decision-making or the review process of this manuscript. The other authors declare no conflicts of interest.

Ethical approval

Not applicable.

Consent to participate

Not applicable.

Consent to publication

Not applicable.

Availability of data and materials

Not applicable.

Funding

We acknowledge the support of NIH/NINDS grants [R01NS089901] and [R01NS114061] to DZL. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Copyright

© The Author(s) 2025.

Publisher’s note

Open Exploration maintains a neutral stance on jurisdictional claims in published institutional affiliations and maps. All opinions expressed in this article are the personal views of the author(s) and do not represent the stance of the editorial team or the publisher.

References

Liu DZ, Ander BP. Cell cycle inhibition without disruption of neurogenesis is a strategy for treatment of aberrant cell cycle diseases: an update. ScientificWorldJournal. 2012;2012:491737. [DOI] [PubMed] [PMC]
Lui A, Vanleuven J, Perekopskiy D, Liu D, Xu D, Alzayat O, et al. FDA-Approved Kinase Inhibitors in Preclinical and Clinical Trials for Neurological Disorders. Pharmaceuticals (Basel). 2022;15:1546. [DOI] [PubMed] [PMC]
Lui A, Do T, Alzayat O, Yu N, Phyu S, Santuya HJ, et al. Tumor Suppressor MicroRNAs in Clinical and Preclinical Trials for Neurological Disorders. Pharmaceuticals (Basel). 2024;17:426. [DOI] [PubMed] [PMC]
Dewan MC, Rattani A, Gupta S, Baticulon RE, Hung YC, Punchak M, et al. Estimating the global incidence of traumatic brain injury. J Neurosurg. 2018;130:108097. [DOI] [PubMed]
Feigin VL, Brainin M, Norrving B, Martins S, Sacco RL, Hacke W, et al. World Stroke Organization (WSO): Global Stroke Fact Sheet 2022. Int J Stroke. 2022;17:1829. [DOI] [PubMed]
Corriveau RA, Bosetti F, Emr M, Gladman JT, Koenig JI, Moy CS, et al. The Science of Vascular Contributions to Cognitive Impairment and Dementia (VCID): A Framework for Advancing Research Priorities in the Cerebrovascular Biology of Cognitive Decline. Cell Mol Neurobiol. 2016;36:2818. [DOI] [PubMed] [PMC]
An E, Brognard J. Orange is the new black: Kinases are the new master regulators of tumor suppression. IUBMB Life. 2019;71:73848. [DOI] [PubMed] [PMC]
Du Z, Lovly CM. Mechanisms of receptor tyrosine kinase activation in cancer. Mol Cancer. 2018;17:58. [DOI] [PubMed] [PMC]
Berndt N, Karim RM, Schönbrunn E. Advances of small molecule targeting of kinases. Curr Opin Chem Biol. 2017;39:12632. [DOI] [PubMed] [PMC]
Fabbro D, Cowan-Jacob SW, Moebitz H. Ten things you should know about protein kinases: IUPHAR Review 14. Br J Pharmacol. 2015;172:2675700. [DOI] [PubMed] [PMC]
Vlastaridis P, Kyriakidou P, Chaliotis A, Van de Peer Y, Oliver SG, Amoutzias GD. Estimating the total number of phosphoproteins and phosphorylation sites in eukaryotic proteomes. Gigascience. 2017;6:111. [DOI] [PubMed] [PMC]
Matte A, Tari LW, Delbaere LT. How do kinases transfer phosphoryl groups? Structure. 1998;6:4139. [DOI] [PubMed]
Krämer J, Bar-Or A, Turner TJ, Wiendl H. Bruton tyrosine kinase inhibitors for multiple sclerosis. Nat Rev Neurol. 2023;19:289304. [DOI] [PubMed] [PMC]
Liu DZ, Cheng XY, Ander BP, Xu H, Davis RR, Gregg JP, et al. Src kinase inhibition decreases thrombin-induced injury and cell cycle re-entry in striatal neurons. Neurobiol Dis. 2008;30:20111. [DOI] [PubMed] [PMC]
Liu DZ, Ander BP, Xu H, Shen Y, Kaur P, Deng W, et al. Blood-brain barrier breakdown and repair by Src after thrombin-induced injury. Ann Neurol. 2010;67:52633. [DOI] [PubMed] [PMC]
Liu DZ, Sharp FR, Van KC, Ander BP, Ghiasvand R, Zhan X, et al. Inhibition of SRC family kinases protects hippocampal neurons and improves cognitive function after traumatic brain injury. J Neurotrauma. 2014;31:126876. [DOI] [PubMed] [PMC]
Liu DZ, Waldau B, Ander BP, Zhan X, Stamova B, Jickling GC, et al. Inhibition of Src family kinases improves cognitive function after intraventricular hemorrhage or intraventricular thrombin. J Cereb Blood Flow Metab. 2017;37:235967. [DOI] [PubMed] [PMC]
Sharma S, Carlson S, Gregory-Flores A, Hinojo-Perez A, Olson A, Thippeswamy T. Mechanisms of disease-modifying effect of saracatinib (AZD0530), a Src/Fyn tyrosine kinase inhibitor, in the rat kainate model of temporal lobe epilepsy. Neurobiol Dis. 2021;156:105410. [DOI] [PubMed] [PMC]
Ye Z, Izadi A, Gurkoff GG, Rickerl K, Sharp FR, Ander BP, et al. Combined Inhibition of Fyn and c-Src Protects Hippocampal Neurons and Improves Spatial Memory via ROCK after Traumatic Brain Injury. J Neurotrauma. 2022;39:5209. [DOI] [PubMed] [PMC]
Jeon BT, Jeong EA, Park SY, Son H, Shin HJ, Lee DH, et al. The Rho-kinase (ROCK) inhibitor Y-27632 protects against excitotoxicity-induced neuronal death in vivo and in vitro. Neurotox Res. 2013;23:23848. [DOI] [PubMed]
Kubo T, Yamashita T. Rho-ROCK inhibitors for the treatment of CNS injury. Recent Pat CNS Drug Discov. 2007;2:1739. [DOI] [PubMed]
Ohnishi M, Katsuki H, Fujimoto S, Takagi M, Kume T, Akaike A. Involvement of thrombin and mitogen-activated protein kinase pathways in hemorrhagic brain injury. Exp Neurol. 2007;206:4352. [DOI] [PubMed]
Di Giovanni S, Movsesyan V, Ahmed F, Cernak I, Schinelli S, Stoica B, et al. Cell cycle inhibition provides neuroprotection and reduces glial proliferation and scar formation after traumatic brain injury. Proc Natl Acad Sci U S A. 2005;102:83338. [DOI] [PubMed] [PMC]
Hilton GD, Stoica BA, Byrnes KR, Faden AI. Roscovitine reduces neuronal loss, glial activation, and neurologic deficits after brain trauma. J Cereb Blood Flow Metab. 2008;28:184559. [DOI] [PubMed] [PMC]
Kabadi SV, Stoica BA, Hanscom M, Loane DJ, Kharebava G, Murray Ii MG, et al. CR8, a selective and potent CDK inhibitor, provides neuroprotection in experimental traumatic brain injury. Neurotherapeutics. 2012;9:40521. [DOI] [PubMed] [PMC]
Aubrecht TG, Faden AI, Sabirzhanov B, Glaser EP, Roelofs BA, Polster BM, et al. Comparing effects of CDK inhibition and E2F1/2 ablation on neuronal cell death pathways in vitro and after traumatic brain injury. Cell Death Dis. 2018;9:1121. [DOI] [PubMed] [PMC]
Osuga H, Osuga S, Wang F, Fetni R, Hogan MJ, Slack RS, et al. Cyclin-dependent kinases as a therapeutic target for stroke. Proc Natl Acad Sci U S A. 2000;97:102549. [DOI] [PubMed] [PMC]
Iguchi M, Honjo J, Yamamoto T, Kanai K. Baricitinib as a treatment for myasthenia gravis: a case report. Neuromuscul Disord. 2024;41:568. [DOI] [PubMed]
Laursen JC, Rotbain Curovic V, Kroonen MYAM, Jongs N, Zobel EH, Hansen TW, et al. Effects of baricitinib, empagliflozin, linagliptin and telmisartan on cardiovascular autonomic neuropathy in type 1 diabetes: An exploratory, randomized, open-label, crossover trial. Diabetes Obes Metab. 2023;25:30647. [DOI] [PubMed]
Gonzales MM, Garbarino VR, Marques Zilli E, Petersen RC, Kirkland JL, Tchkonia T, et al. Senolytic Therapy to Modulate the Progression of Alzheimer’s Disease (SToMP-AD): A Pilot Clinical Trial. J Prev Alzheimers Dis. 2022;9:229. [DOI] [PubMed] [PMC]
Gonzales MM, Garbarino VR, Kautz TF, Palavicini JP, Lopez-Cruzan M, Dehkordi SK, et al. Senolytic therapy in mild Alzheimer’s disease: a phase 1 feasibility trial. Nat Med. 2023;29:24818. [DOI] [PubMed] [PMC]
Joussen AM, Wolf S, Kaiser PK, Boyer D, Schmelter T, Sandbrink R, 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;85:34755. [DOI] [PubMed] [PMC]
Palma JA, Martinez J, Millar Vernetti P, Ma T, Perez MA, Zhong J, et al. mTOR Inhibition with Sirolimus in Multiple System Atrophy: A Randomized, Double-Blind, Placebo-Controlled Futility Trial and 1-Year Biomarker Longitudinal Analysis. Mov Disord. 2022;37:77889. [DOI] [PubMed] [PMC]
Liu Y, Huang Z, Zhang TX, Han B, Yang G, Jia D, et al. Bruton’s tyrosine kinase-bearing B cells and microglia in neuromyelitis optica spectrum disorder. J Neuroinflammation. 2023;20:309. [DOI] [PubMed] [PMC]
Specchio N, Nabbout R, Aronica E, Auvin S, Benvenuto A, de Palma L, et al. Updated clinical recommendations for the management of tuberous sclerosis complex associated epilepsy. Eur J Paediatr Neurol. 2023;47:2534. [DOI] [PubMed]
Tong J, Ji T, Liu T, Liu J, Chen Y, Li Z, et al. Efficacy and safety of six new antiseizure medications for adjunctive treatment of focal epilepsy and epileptic syndrome: A systematic review and network meta-analysis. Epilepsy Behav. 2024;152:109653. [DOI] [PubMed]
Edmonds B, Ngo JP, Groves A, Reyes B, Gott RA, Chia DJ, et al. Multi-disciplinary team approach for pediatric hemimegalencephaly: Insights from a single institutional case series. Epilepsia Open. 2024;9:25107. [DOI] [PubMed] [PMC]
Pearsson K, Björk Werner J, Lundgren J, Gränse L, Karlsson E, Källén K, et al. Childhood tuberous sclerosis complex in southern Sweden: a paradigm shift in diagnosis and treatment. BMC Pediatr. 2023;23:329. [DOI] [PubMed] [PMC]
Leitner DF, Kanshin E, Askenazi M, Siu Y, Friedman D, Devore S, et al. Pilot study evaluating everolimus molecular mechanisms in tuberous sclerosis complex and focal cortical dysplasia. PLoS One. 2022;17:e0268597. [DOI] [PubMed] [PMC]
Butera S, Tavarozzi R, Brunello L, Rivela P, Sofia A, Viero L, et al. The black swan: a case of central nervous system graft-versus-host disease. J Basic Clin Physiol Pharmacol. 2023;34:805809. [DOI] [PubMed]
Castellani F, Visentin A, Schirinzi E, Salvalaggio A, Cacciavillani M, Candiotto C, et al. Mutational Profile in 75 Patients With Anti-Myelin-Associated Glycoprotein Neuropathy: Clinical and Hematologic Therapy Response and Hints on New Therapeutic Targets. Neurol Neuroimmunol Neuroinflamm. 2023;10:e200122. [DOI] [PubMed] [PMC]
Shirani A, Saez-Calveras N, Antel JP, Yaqubi M, Moore W, Brewster AL, et al. Exploring the association of disease-modifying therapies for multiple sclerosis and BTK inhibitors with epilepsy. Ther Adv Neurol Disord. 2024;17:17562864241276204. [DOI] [PubMed] [PMC]
Li Y, Sun Z, Zhu H, Sun Y, Shteyman DB, Markx S, et al. Inhibition of Abl Kinase by Imatinib Can Rescue the Compromised Barrier Function of 22q11.2DS Patient-iPSC-Derived Blood-Brain Barriers. Cells. 2023;12:422. [DOI] [PubMed] [PMC]
Vos EM, Geraedts VJ, van der Lugt A, Dippel DWJ, Wermer MJH, Hofmeijer J, et al. Systematic Review - Combining Neuroprotection With Reperfusion in Acute Ischemic Stroke. Front Neurol. 2022;13:840892. [DOI] [PubMed] [PMC]
Frémond ML, Hully M, Fournier B, Barrois R, Lévy R, Aubart M, et al. JAK Inhibition in Aicardi-Goutières Syndrome: a Monocentric Multidisciplinary Real-World Approach Study. J Clin Immunol. 2023;43:143647. [DOI] [PubMed] [PMC]
Jang Y, Lee WJ, Lee HS, Chu K, Lee SK, Lee ST. Tofacitinib treatment for refractory autoimmune encephalitis. Epilepsia. 2021;62:e539. [DOI] [PubMed]
Wang F, Li S, Wang TY, Lopez GA, Antoshechkin I, Chou TF. P97/VCP ATPase inhibitors can rescue p97 mutation-linked motor neuron degeneration. Brain Commun. 2022;4:fcac176. [DOI] [PubMed] [PMC]
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;602:1428. [DOI] [PubMed]
Liu T, Hou K, Li J, Han T, Liu S, Wei J. Alzheimer’s Disease and Aging Association: Identification and Validation of Related Genes. J Prev Alzheimers Dis. 2024;11:196213. [DOI] [PubMed]
Lee HJ, Hoe HS. Inhibition of CDK4/6 regulates AD pathology, neuroinflammation and cognitive function through DYRK1A/STAT3 signaling. Pharmacol Res. 2023;190:106725. [DOI] [PubMed]
Li T, Li L, Peng R, Hao H, Zhang H, Gao Y, et al. Abrocitinib Attenuates Microglia-Mediated Neuroinflammation after Traumatic Brain Injury via Inhibiting the JAK1/STAT1/NF-κB Pathway. Cells. 2022;11:3588. [DOI] [PubMed] [PMC]
Chen YJ, Hsu CC, Shiao YJ, Wang HT, Lo YL, Lin AMY. Anti-inflammatory effect of afatinib (an EGFR-TKI) on OGD-induced neuroinflammation. Sci Rep. 2019;9:2516. [DOI] [PubMed] [PMC]
Linnerbauer M, Lößlein L, Vandrey O, Tsaktanis T, Beer A, Naumann UJ, et al. Intranasal delivery of a small-molecule ErbB inhibitor promotes recovery from acute and late-stage CNS inflammation. JCI Insight. 2022;7:e154824. [DOI] [PubMed] [PMC]
Schachenhofer J, Gruber VE, Fehrer SV, Haider C, Glatter S, Liszewska E, et al. Targeting the EGFR pathway: An alternative strategy for the treatment of tuberous sclerosis complex? Neuropathol Appl Neurobiol. 2024;50:e12974. [DOI] [PubMed]
Gómez-Oliva R, Geribaldi-Doldán N, Domínguez-García S, Pardillo-Díaz R, Martínez-Ortega S, Oliva-Montero JM, et al. Targeting epidermal growth factor receptor to recruit newly generated neuroblasts in cortical brain injuries. J Transl Med. 2023;21:867. [DOI] [PubMed] [PMC]
Singh CSB, Choi KB, Munro L, Wang HY, Pfeifer CG, Jefferies WA. Reversing pathology in a preclinical model of Alzheimer’s disease by hacking cerebrovascular neoangiogenesis with advanced cancer therapeutics. EBioMedicine. 2021;71:103503. [DOI] [PubMed] [PMC]
Wang S, Guo Y, Cao RQ, Zhu YM, Qiao SG, Du HP, et al. VEGFD/VEGFR3 signaling contributes to the dysfunction of the astrocyte IL-3/microglia IL-3Rα cross-talk and drives neuroinflammation in mouse ischemic stroke. Acta Pharmacol Sin. 2025;46:292307. [DOI] [PubMed]
Wang K, Zhou W, Wen L, Jin X, Meng T, Li S, et al. The protective effects of Axitinib on blood-brain barrier dysfunction and ischemia-reperfusion injury in acute ischemic stroke. Exp Neurol. 2024;379:114870. [DOI] [PubMed]
Königs V, Pierre S, Schicht M, Welss J, Hahnefeld L, Rimola V, et al. GPR40 Activation Abolishes Diabetes-Induced Painful Neuropathy by Suppressing VEGF-A Expression. Diabetes. 2022;71:77487. [DOI] [PubMed]
Dutton JW 3rd, Chen H, You C, Brodie MS, Lasek AW. Anaplastic lymphoma kinase regulates binge-like drinking and dopamine receptor sensitivity in the ventral tegmental area. Addict Biol. 2017;22:66578. [DOI] [PubMed] [PMC]
Hamada K, Ferguson LB, Mayfield RD, Krishnan HR, Maienschein-Cline M, Lasek AW. 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:e12729. [DOI] [PubMed] [PMC]
Hu Y, Chang L, Zhu Y, Geng X, Liu Z, Wang R, et al. Inhibition of Anaplastic Lymphoma Kinase Protects From Ischemic Stroke. Stroke. 2024;55:107585. [DOI] [PubMed]
Gavegnano C, Haile WB, Hurwitz S, Tao S, Jiang Y, Schinazi RF, et al. Baricitinib reverses HIV-associated neurocognitive disorders in a SCID mouse model and reservoir seeding in vitro. J Neuroinflammation. 2019;16:182. [DOI] [PubMed] [PMC]
Nakamura J, Yanagida M, Saito K, Kamata Y, Nagashima T, Iwamoto M, et al. Epstein-Barr virus encephalitis in a patient with rheumatoid arthritis. Mod Rheumatol Case Rep. 2022;6:1602. [DOI] [PubMed]
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. [DOI] [PubMed] [PMC]
Tuttle KD, Waugh KA, Araya P, Minter R, Orlicky DJ, Ludwig M, et al. JAK1 Inhibition Blocks Lethal Immune Hypersensitivity in a Mouse Model of Down Syndrome. Cell Rep. 2020;33:108407. [DOI] [PubMed] [PMC]
Zheng XQ, Huang JF, Lin JL, Zhu YX, Wang MQ, Guo ML, et al. 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. [DOI] [PubMed]
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. [DOI] [PubMed] [PMC]
Agarwal K, Katare DP, Jakhmola-Mani R. Foresee Novel Targets for Alzheimer’s Disease by Investigating Repurposed Drugs. CNS Neurol Disord Drug Targets. 2023;22:120931. [DOI] [PubMed]
König LE, Rodriguez S, Hug C, Daneshvari S, Chung A, Bradshaw GA, et al. TYK2 as a novel therapeutic target in Alzheimer’s Disease with TDP-43 inclusions. BioRxiv 595773 [Preprint]. 2024 [cited 2025 May 26]. Available from: https://www.biorxiv.org/content/10.1101/2024.06.04.595773v1
Hindam MO, Ahmed LA, El Sayed NS, Khattab M, Sallam NA. Repositioning of baricitinib for management of memory impairment in ovariectomized/D-galactose treated rats: A potential role of JAK2/STAT3-PI3K/AKT/mTOR signaling pathway. Life Sci. 2024;351:122838. [DOI] [PubMed]
Bernal-Bermúdez B, Martínez-López A, Martínez-Morcillo FJ, Tyrkalska SD, Martínez-Menchón T, Mesa-Del-Castillo P, et al. A zebrafish model of Ifih1-driven Aicardi-Goutières syndrome reproduces the interferon signature and the exacerbated inflammation of patients. Front Immunol. 2023;14:1294766. [DOI] [PubMed] [PMC]
Železnik M, Soltirovska Šalamon A, Debeljak M, Goropevšek A, Šuštar N, Ključevšek D, et al. Case report: Pneumocystis jirovecii pneumonia in a severe case of Aicardi-Goutières syndrome with an IFIH1 gain-of-function mutation mimicking combined immunodeficiency. Front Immunol. 2023;13:1033513. [DOI] [PubMed] [PMC]
Jafarpour S, Suddock J, Hawes D, Santoro JD. Neuropathologic Impacts of JAK Inhibitor Treatment in Aicardi-Goutières Syndrome. J Clin Immunol. 2024;44:68. [DOI] [PubMed]
Ryckmans C, Donge M, Marchèse A, Mastouri M, Thomee C, Stouffs K, et al. TREX-1 related Aicardi-Goutières syndrome improved by Janus kinase inhibitor. Am J Med Genet A. 2024;194:e63510. [DOI] [PubMed]
Richardson PJ, Smith DP, de Giorgio A, Snetkov X, Almond-Thynne J, Cronin S, et al. Janus kinase inhibitors are potential therapeutics for amyotrophic lateral sclerosis. Transl Neurodegener. 2023;12:47. [DOI] [PubMed] [PMC]
Makabe K, Okada H, Tachibana N, Ishikura H, Ito N, Tanaka M, et al. Baricitinib ameliorates inflammatory and neuropathic pain in collagen antibody-induced arthritis mice by modulating the IL-6/JAK/STAT3 pathway and CSF-1 expression in dorsal root ganglion neurons. Arthritis Res Ther. 2024;26:121. [DOI] [PubMed] [PMC]
Acosta-González RI, Hernández-Jiménez AY, Ramírez-Quintanilla LY, Torres-Rodríguez HF, Vargas Muñoz VM, Jiménez-Andrade JM. Effect of 28 days treatment of baricitinib on mechanical allodynia, osteopenia, and loss of nerve fibers in an experimental model of type-1 diabetes mellitus. Pharmacol Rep. 2024;76:107988. [DOI] [PubMed]
Wohlrab J, Stintzing D, Schultz L, Jügelt K, Schroeder OH. Influence of Janus Kinase Inhibitors on the Neuronal Activity as a Proof-of-Concept Model for Itch. Skin Pharmacol Physiol. 2022;35:94101. [DOI] [PubMed] [PMC]
Schapansky J, Grinberg YY, Osiecki DM, Freeman EA, Walker SG, Karran E, et al. MEK1/2 activity modulates TREM2 cell surface recruitment. J Biol Chem. 2021;296:100218. [DOI] [PubMed] [PMC]
Ma L, Manaenko A, Ou YB, Shao AW, Yang SX, Zhang JH. Bosutinib Attenuates Inflammation via Inhibiting Salt-Inducible Kinases in Experimental Model of Intracerebral Hemorrhage on Mice. Stroke. 2017;48:310816. [DOI] [PubMed] [PMC]
Liang S, Pong K, Gonzales C, Chen Y, Ling HP, Mark RJ, et al. Neuroprotective profile of novel SRC kinase inhibitors in rodent models of cerebral ischemia. J Pharmacol Exp Ther. 2009;331:82735. [DOI] [PubMed]
Hebron ML, Javidnia M, Moussa CE. Tau clearance improves astrocytic function and brain glutamate-glutamine cycle. J Neurol Sci. 2018;391:909. [DOI] [PubMed]
Hebron ML, Lonskaya I, Olopade P, Selby ST, Pagan F, Moussa CE. Tyrosine Kinase Inhibition Regulates Early Systemic Immune Changes and Modulates the Neuroimmune Response in α-Synucleinopathy. J Clin Cell Immunol. 2014;5:259. [DOI] [PubMed] [PMC]
Wenqiang C, Lonskaya I, Hebron ML, Ibrahim Z, Olszewski RT, Neale JH, et al. Parkin-mediated reduction of nuclear and soluble TDP-43 reverses behavioral decline in symptomatic mice. Hum Mol Genet. 2014;23:49609. [DOI] [PubMed]
Lonskaya I, Hebron ML, Desforges NM, Franjie A, Moussa CE. Tyrosine kinase inhibition increases functional parkin-Beclin-1 interaction and enhances amyloid clearance and cognitive performance. EMBO Mol Med. 2013;5:124762. [DOI] [PubMed] [PMC]
Lonskaya I, Hebron ML, Selby ST, Turner RS, Moussa CE. Nilotinib and bosutinib modulate pre-plaque alterations of blood immune markers and neuro-inflammation in Alzheimer’s disease models. Neuroscience. 2015;304:31627. [DOI] [PubMed]
Heyburn L, Hebron ML, Smith J, Winston C, Bechara J, Li Z, et al. Tyrosine kinase inhibition reverses TDP-43 effects on synaptic protein expression, astrocytic function and amino acid dis-homeostasis. J Neurochem. 2016;139:61023. [DOI] [PubMed]
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. [DOI] [PubMed]
Kiris E, Burnett JC, Nuss JE, Wanner LM, Peyser BD, Du HT, 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;27:38498. [DOI] [PubMed] [PMC]
Khan SA, Khan S, Kausar H, Shah R, Luitel A, Gautam S, et al. Insights into the management of Lewy body dementia: a scoping review. Ann Med Surg (Lond). 2024;86:93042. [DOI] [PubMed] [PMC]
Skylar-Scott IA, Sha SJ. Lewy Body Dementia: An Overview of Promising Therapeutics. Curr Neurol Neurosci Rep. 2023;23:58192. [DOI] [PubMed]
Ito D, Morimoto S, Takahashi S, Okada K, Nakahara J, Okano H. Maiden voyage: induced pluripotent stem cell-based drug screening for amyotrophic lateral sclerosis. Brain. 2023;146:139. [DOI] [PubMed]
Noguchi Y, Asano H, Masuda R, Teshigawara Y, Go M, Kimura M, et al. Relationship between Anaplastic Lymphoma Kinase Inhibitors and Epileptic Seizure Disorder: A Post-Marketing Surveillance Study. Oncology. 2024;102:9961003. [DOI] [PubMed]
Imamura K, Izumi Y, Nagai M, Nishiyama K, Watanabe Y, Hanajima R, et al. Safety and tolerability of bosutinib in patients with amyotrophic lateral sclerosis (iDReAM study): A multicentre, open-label, dose-escalation phase 1 trial. EClinicalMedicine. 2022;53:101707. [DOI] [PubMed] [PMC]
Tang X, Drotar J, Li K, Clairmont CD, Brumm AS, Sullins AJ, 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. [DOI] [PubMed] [PMC]
Tucker Edmister S, Del Rosario Hernández T, Ibrahim R, Brown CA, Gore SV, Kakodkar R, et al. Novel use of FDA-approved drugs identified by cluster analysis of behavioral profiles. Sci Rep. 2022;12:6120. [DOI] [PubMed] [PMC]
Lee HJ, Hwang JW, Park JH, Jeong YJ, Jang JY, Hoe HS. Profiling tyrosine kinase inhibitors as AD therapeutics in a mouse model of AD. Mol Brain. 2023;16:63. [DOI] [PubMed] [PMC]
Del Rosario Hernandez T, Joshi NR, Gore SV, Kreiling JA, Creton R. Combining supervised and unsupervised analyses to quantify behavioral phenotypes and validate therapeutic efficacy in a triple transgenic mouse model of Alzheimer’s disease. Biomed Pharmacother. 2024;181:117718. [DOI] [PubMed] [PMC]
Bolz SN, Salentin S, Jennings G, Haupt VJ, Sterneckert J, Schroeder M. Structural binding site comparisons reveal Crizotinib as a novel LRRK2 inhibitor. Comput Struct Biotechnol J. 2021;19:367481. [DOI] [PubMed] [PMC]
Lim JW, Kim SK, Choi SY, Kim DH, Gadhe CG, Lee HN, et al. Identification of crizotinib derivatives as potent SHIP2 inhibitors for the treatment of Alzheimer’s disease. Eur J Med Chem. 2018;157:40522. [DOI] [PubMed]
Defaye M, Iftinca MC, Gadotti VM, Basso L, Abdullah NS, Cuménal M, et al. The neuronal tyrosine kinase receptor ligand ALKAL2 mediates persistent pain. J Clin Invest. 2022;132:e154317. [DOI] [PubMed] [PMC]
Yang Z, Ahn HJ, Nam HW. Gefitinib inhibits the growth of Toxoplasma gondii in HeLa cells. Korean J Parasitol. 2014;52:43941. [DOI] [PubMed] [PMC]
Camp E, Anderson PJ, Zannettino ACW, Glackin CA, 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;233:732032. [DOI] [PubMed]
Cruz SA, Qin Z, Stewart AFR, Chen HH. Dabrafenib, an inhibitor of RIP3 kinase-dependent necroptosis, reduces ischemic brain injury. Neural Regen Res. 2018;13:2526. [DOI] [PubMed] [PMC]
Sugaya T, Kanno H, Matsuda M, Handa K, Tateda S, Murakami T, et al. B-RAFV600E Inhibitor Dabrafenib Attenuates RIPK3-Mediated Necroptosis and Promotes Functional Recovery after Spinal Cord Injury. Cells. 2019;8:1582. [DOI] [PubMed] [PMC]
Uenaka T, Satake W, Cha PC, Hayakawa H, Baba K, Jiang S, 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;27:397485. [DOI] [PubMed] [PMC]
Okamoto T. Parkinson’s Disease: amantadine, zonisamide, dabrafenib. Brain Nerve. 2019;71:9539. Japanese. [DOI] [PubMed]
Elkouzi A, Rauschkolb P, Grogg KL, Gilchrist JM. Neurohistiocytosis of the Cerebellum: A Rare Cause of Ataxia. Mov Disord Clin Pract. 2015;3:1259. [DOI] [PubMed] [PMC]
Tu T, Yu J, Jiang C, Zhang S, Li J, Ren J, et al. Somatic BrafV600E mutation in the cerebral endothelium induces brain arteriovenous malformations. Angiogenesis. 2024;27:44160. [DOI] [PubMed]
Saminathan H, Charli A, Luo J, Panicker N, Gordon R, Hostetter JM, et al. Fyn kinase mediates pro-inflammatory response in a mouse model of endotoxemia: Relevance to translational research. Eur J Pharmacol. 2020;881:173259. [DOI] [PubMed] [PMC]
Gangoso E, Talaverón R, Jaraíz-Rodríguez M, Domínguez-Prieto M, Ezan P, Koulakoff A, et al. A c-Src Inhibitor Peptide Based on Connexin43 Exerts Neuroprotective Effects through the Inhibition of Glial Hemichannel Activity. Front Mol Neurosci. 2017;10:418. [DOI] [PubMed] [PMC]
El-Nimri NW, Moore SM, Zangwill LM, Proudfoot JA, Weinreb RN, Skowronska-Krawczyk D, et al. Evaluating the neuroprotective impact of senolytic drugs on human vision. Sci Rep. 2020;10:21752. [DOI] [PubMed] [PMC]
Musi N, Valentine JM, Sickora KR, Baeuerle E, Thompson CS, Shen Q, et al. Tau protein aggregation is associated with cellular senescence in the brain. Aging Cell. 2018;17:e12840. [DOI] [PubMed] [PMC]
Azizi G, Goudarzvand M, Afraei S, Sedaghat R, Mirshafiey A. Therapeutic effects of dasatinib in mouse model of multiple sclerosis. Immunopharmacol Immunotoxicol. 2015;37:28794. [DOI] [PubMed]
Katsumata R, Ishigaki S, Katsuno M, Kawai K, Sone J, Huang Z, et al. c-Abl inhibition delays motor neuron degeneration in the G93A mouse, an animal model of amyotrophic lateral sclerosis. PLoS One. 2012;7:e46185. [DOI] [PubMed] [PMC]
Lawana V, Singh N, Sarkar S, Charli A, Jin H, Anantharam V, et al. Involvement of c-Abl Kinase in Microglial Activation of NLRP3 Inflammasome and Impairment in Autolysosomal System. J Neuroimmune Pharmacol. 2017;12:62460. [DOI] [PubMed] [PMC]
Torres P, Anerillas C, Ramírez-Núñez O, Fernàndez A, Encinas M, Povedano M, et al. A motor neuron disease mouse model reveals a non-canonical profile of senescence biomarkers. Dis Model Mech. 2022;15:dmm049059. [DOI] [PubMed] [PMC]
Fang Y, Medina D, Stockwell R, McFadden S, Quinn K, Peck MR, et al. Sexual dimorphic metabolic and cognitive responses of C57BL/6 mice to Fisetin or Dasatinib and quercetin cocktail oral treatment. Geroscience. 2023;45:283550. [DOI] [PubMed] [PMC]
Budamagunta V, Kumar A, Rani A, Bean L, Manohar-Sindhu S, Yang Y, et al. Effect of peripheral cellular senescence on brain aging and cognitive decline. Aging Cell. 2023;22:e13817. [DOI] [PubMed] [PMC]
Ogrodnik M, Zhu Y, Langhi LGP, Tchkonia T, Krüger P, Fielder E, et al. Obesity-Induced Cellular Senescence Drives Anxiety and Impairs Neurogenesis. Cell Metab. 2019;29:106177. [DOI] [PubMed] [PMC]
Lin YF, Wang LY, Chen CS, Li CC, Hsiao YH. Cellular senescence as a driver of cognitive decline triggered by chronic unpredictable stress. Neurobiol Stress. 2021;15:100341. [DOI] [PubMed] [PMC]
Wang D, Howell BW, Olson EC. Maternal Ethanol Exposure Acutely Elevates Src Family Kinase Activity in the Fetal Cortex. Mol Neurobiol. 2021;58:521023. [DOI] [PubMed] [PMC]
Cuní-López C, Stewart R, Oikari LE, Nguyen TH, Roberts TL, Sun Y, et al. Advanced patient-specific microglia cell models for pre-clinical studies in Alzheimer’s disease. J Neuroinflammation. 2024;21:50. [DOI] [PubMed] [PMC]
Mohammad AR, Hassan ES, Majeed SA. PI3K/AKT and STAT3 pathways mediate the neuroprotective effect of dasatinib from acute cerebral injury in endotoxemic mice. Res Pharm Sci. 2024;19:6472. [DOI] [PubMed] [PMC]
Zhang X, Guo J, Liu J, Liu J, Li Z, Chen J, et al. Exosomal Src from hypoxic vascular smooth muscle cells exacerbates ischemic brain injury by promoting M1 microglial polarization. Neurochem Int. 2024;179:105819. [DOI] [PubMed]
Yao M, Wei Z, Nielsen JS, Ouyang Y, Kakazu A, Wang H, et al. Senolytic therapy preserves blood-brain barrier integrity and promotes microglia homeostasis in a tauopathy model. Neurobiol Dis. 2024;202:106711. [DOI] [PubMed] [PMC]
Ribierre T, Bacq A, Donneger F, Doladilhe M, Maletic M, Roussel D, et al. Targeting pathological cells with senolytic drugs reduces seizures in neurodevelopmental mTOR-related epilepsy. Nat Neurosci. 2024;27:112536. [DOI] [PubMed] [PMC]
Khan T, Hussain AI, Casilli TP, Frayser L, Cho M, Williams G, et al. Prophylactic senolytic treatment in aged mice reduces seizure severity and improves survival from Status Epilepticus. Aging Cell. 2024;23:e14239. [DOI] [PubMed] [PMC]
Wang C, Kang Y, Liu P, Liu W, Chen W, Hayashi T, et al. Combined use of dasatinib and quercetin alleviates overtraining-induced deficits in learning and memory through eliminating senescent cells and reducing apoptotic cells in rat hippocampus. Behav Brain Res. 2023;440:114260. [DOI] [PubMed]
Lin X, Zhang K, Li C, Liu K, Sun Y, Wu W, et al. Combination of Dasatinib and Quercetin alleviates heat stress-induced cognitive deficits in aged and young adult male mice. Eur J Pharmacol. 2024;974:176631. [DOI] [PubMed]
Wang J, Lu Y, Carr C, Dhandapani KM, Brann DW. Senolytic therapy is neuroprotective and improves functional outcome long-term after traumatic brain injury in mice. Front Neurosci. 2023;17:1227705. [DOI] [PubMed] [PMC]
Orr ME. A Need for Refined Senescence Biomarkers and Measures of Senolytics in the Brain. J Alzheimers Dis. 2024;98:4115. [DOI] [PubMed] [PMC]
Fang Y, Peck MR, Quinn K, Chapman JE, Medina D, McFadden SA, et al. Senolytic intervention improves cognition, metabolism, and adiposity in female APPNL-F/NL-F mice. Geroscience. 2025;47:112338. [DOI] [PubMed] [PMC]
Riordan R, Rong W, Yu Z, Ross G, Valerio J, Dimas-Muñoz J, et al. Effect of Nrf2 loss on senescence and cognition of tau-based P301S mice. Geroscience. 2023;45:145169. [DOI] [PubMed] [PMC]
Ng PY, Zhang C, Li H, Baker DJ. Senescence Targeting Methods Impact Alzheimer’s Disease Features in 3xTg Mice. J Alzheimers Dis. 2024;97:175163. [DOI] [PubMed] [PMC]
Krzystyniak A, Wesierska M, Petrazzo G, Gadecka A, Dudkowska M, Bielak-Zmijewska A, et al. 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;14:57295. [DOI] [PubMed] [PMC]
Van Houcke J, Mariën V, Zandecki C, Ayana R, Pepermans E, Boonen K, et al. A short dasatinib and quercetin treatment is sufficient to reinstate potent adult neuroregenesis in the aged killifish. NPJ Regen Med. 2023;8:31. [DOI] [PubMed] [PMC]
Riessland M, Ximerakis M, Jarjour AA, Zhang B, Orr ME. Therapeutic targeting of senescent cells in the CNS. Nat Rev Drug Discov. 2024;23:81737. [DOI] [PubMed] [PMC]
Ya J, Bayraktutan U. Senolytics and Senomorphics Targeting p38MAPK/NF-κB Pathway Protect Endothelial Cells from Oxidative Stress-Mediated Premature Senescence. Cells. 2024;13:1292. [DOI] [PubMed] [PMC]
Ya J, Kadir RRA, Bayraktutan U. Delay of endothelial cell senescence protects cerebral barrier against age-related dysfunction: role of senolytics and senomorphics. Tissue Barriers. 2023;11:2103353. [DOI] [PubMed] [PMC]
Ota H, Kodama A. Dasatinib plus quercetin attenuates some frailty characteristics in SAMP10 mice. Sci Rep. 2022;12:2425. [DOI] [PubMed] [PMC]
Faria OW, de Aguiar MSS, de Mello JE, Alvez FL, Luduvico KP, Garcia DN, et al. Senolytics prevent age-associated changes in female mice brain. Neurosci Lett. 2024;826:137730. [DOI] [PubMed]
Johnson B, Cheng L, Koenitzer J, Catlett IM, Schafer P. Nonclinical evaluations of deucravacitinib and Janus kinase inhibitors in homeostatic and inflammatory pathways. Front Immunol. 2024;15:1437512. [DOI] [PubMed] [PMC]
Adamek P, Heles M, Bhattacharyya A, Pontearso M, Slepicka J, Palecek J. Dual PI3Kδ/γ Inhibitor Duvelisib Prevents Development of Neuropathic Pain in Model of Paclitaxel-Induced Peripheral Neuropathy. J Neurosci. 2022;42:186481. [DOI] [PubMed] [PMC]
Koprivica V, Cho KS, Park JB, Yiu G, Atwal J, Gore B, et al. EGFR activation mediates inhibition of axon regeneration by myelin and chondroitin sulfate proteoglycans. Science. 2005;310:10610. [DOI] [PubMed]
Luo Y, Tang H, Zhang Z, Zhao R, Wang C, Hou W, et al. Pharmacological inhibition of epidermal growth factor receptor attenuates intracranial aneurysm formation by modulating the phenotype of vascular smooth muscle cells. CNS Neurosci Ther. 2022;28:6476. [DOI] [PubMed] [PMC]
Le Pichon CE, Dominguez SL, Solanoy H, Ngu H, Lewin-Koh N, Chen M, et al. EGFR inhibitor erlotinib delays disease progression but does not extend survival in the SOD1 mouse model of ALS. PLoS One. 2013;8:e62342. [DOI] [PubMed] [PMC]
Pan P, Dobrowsky RT. 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. [DOI] [PubMed] [PMC]
McGuire JF, Rouen S, Siegfreid E, Wright DE, Dobrowsky RT. Caveolin-1 and altered neuregulin signaling contribute to the pathophysiological progression of diabetic peripheral neuropathy. Diabetes. 2009;58:267786. [DOI] [PubMed] [PMC]
Wang L, Chiang HC, Wu W, Liang B, Xie Z, Yao X, et al. Epidermal growth factor receptor is a preferred target for treating amyloid-β-induced memory loss. Proc Natl Acad Sci U S A. 2012;109:167438. [DOI] [PubMed] [PMC]
Lechpammer M, Tran YP, Wintermark P, Martínez-Cerdeño V, Krishnan VV, Ahmed W, et al. Upregulation of cystathionine β-synthase and p70S6K/S6 in neonatal hypoxic ischemic brain injury. Brain Pathol. 2017;27:44958. [DOI] [PubMed] [PMC]
Kurdi A, Roth L, Van der Veken B, Van Dam D, De Deyn PP, De Doncker M, et al. Everolimus depletes plaque macrophages, abolishes intraplaque neovascularization and improves survival in mice with advanced atherosclerosis. Vascul Pharmacol. 2019;113:706. [DOI] [PubMed]
Li D, Wang C, Yao Y, Chen L, Liu G, Zhang R, et al. mTORC1 pathway disruption ameliorates brain inflammation following stroke via a shift in microglia phenotype from M1 type to M2 type. FASEB J. 2016;30:338899. [DOI] [PubMed]
Suvanish Kumar VS, Pretorius E, Rajanikant GK. The Synergistic Combination of Everolimus and Paroxetine Exerts Post-ischemic Neuroprotection In Vitro. Cell Mol Neurobiol. 2018;38:138397. [DOI] [PubMed] [PMC]
Forouzanfar F, Ebrahimi PR, Sadeghnia HR. Neuroprotection of Everolimus Against Focal Cerebral Ischemia-Reperfusion Injury in Rats. J Stroke Cerebrovasc Dis. 2022;31:106576. [DOI] [PubMed]
Cassano T, Magini A, Giovagnoli S, Polchi A, Calcagnini S, Pace L, et al. Early intrathecal infusion of everolimus restores cognitive function and mood in a murine model of Alzheimer’s disease. Exp Neurol. 2019;311:88105. [DOI] [PubMed]
Fanoudi S, Hosseini M, Alavi MS, Boroushaki MT, Hosseini A, Sadeghnia HR. Everolimus, a mammalian target of rapamycin inhibitor, ameliorated streptozotocin-induced learning and memory deficits via neurochemical alterations in male rats. EXCLI J. 2018;17:9991017. [DOI] [PubMed] [PMC]
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;119:398407. [DOI] [PubMed]
Fox JH, Connor T, Chopra V, Dorsey K, Kama JA, Bleckmann D, 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. [DOI] [PubMed] [PMC]
Chen L, Zhang Y, Li D, Zhang N, Liu R, Han B, et al. Everolimus (RAD001) ameliorates vascular cognitive impairment by regulating microglial function via the mTORC1 signaling pathway. J Neuroimmunol. 2016;299:16471. [DOI] [PubMed]
Dello Russo C, Lisi L, Tringali G, Navarra P. Involvement of mTOR kinase in cytokine-dependent microglial activation and cell proliferation. Biochem Pharmacol. 2009;78:124251. [DOI] [PubMed]
Bansal S, Agrawal M, Mahendiratta S, Kumar S, Arora S, Joshi R, et al. Everolimus: A potential therapeutic agent targeting PI3K/Akt pathway in brain insulin system dysfunction and associated neurobehavioral deficits. Fundam Clin Pharmacol. 2021;35:101831. [DOI] [PubMed]
Alavi MS, Fanoudi S, Hosseini A, Jalili-Nik M, Bagheri A, Sadeghnia HR. Everolimus attenuates glutamate-induced PC12 cells death. Int J Neurosci. 2023;133:45766. [DOI] [PubMed]
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;280:10614. [DOI] [PubMed]
Hoepner R, Bagnoud M, Pistor M, Salmen A, Briner M, Synn H, et al. Vitamin D increases glucocorticoid efficacy via inhibition of mTORC1 in experimental models of multiple sclerosis. Acta Neuropathol. 2019;138:44356. [DOI] [PubMed] [PMC]
Schneider M, de Vries PJ, 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;267:45563. [DOI] [PubMed]
Petrasek T, Vojtechova I, Klovrza O, Tuckova K, Vejmola C, Rak 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. [DOI] [PubMed] [PMC]
Mishra N, Wang P, Goldsmith D, Zhao X, Xue Y, Christians U, et al. Everolimus does not prevent Lafora body formation in murine Lafora disease. Neurol Genet. 2017;3:e127. [DOI] [PubMed] [PMC]
Nguyen LH, Leiser SC, Song D, Brunner D, Roberds SL, Wong M, et al. Inhibition of MEK-ERK signaling reduces seizures in two mouse models of tuberous sclerosis complex. Epilepsy Res. 2022;181:106890. [DOI] [PubMed] [PMC]
Aronica E, Specchio N, Luinenburg MJ, Curatolo P. Epileptogenesis in tuberous sclerosis complex-related developmental and epileptic encephalopathy. Brain. 2023;146:2694710. [DOI] [PubMed] [PMC]
Zhao W, Xie C, Zhang X, Liu J, Liu J, Xia Z. Advances in the mTOR signaling pathway and its inhibitor rapamycin in epilepsy. Brain Behav. 2023;13:e2995. [DOI] [PubMed] [PMC]
Bonazzi S, Gray A, Thomsen NM, Biag J, Labbe-Giguere N, Keaney EP, et al. Identification of Brain-Penetrant ATP-Competitive mTOR Inhibitors for CNS Syndromes. J Med Chem. 2023;66:9095119. [DOI] [PubMed]
Shirzad S, Vafaee F, Forouzanfar F. The Neuroprotective Effects and Probable Mechanisms of Everolimus in a Rat Model of Intracerebral Hemorrhage. Cell Mol Neurobiol. 2023;43:421930. [DOI] [PubMed] [PMC]
Qiu X, Zhou R, Su X, Ying J, Qu Y, Mu D. Pleiotrophin ameliorates white matter injury of neonatal rats by activating the mTOR/YY1/Id4 signaling pathway. FASEB J. 2023;37:e23082. [DOI] [PubMed]
Tsai CW, Ho SY, Chen IC, Chang KC, Chen HJ, Tsai FC, et al. Abnormal increased mTOR signaling regulates seizure threshold in Dravet syndrome. Neuropharmacology. 2025;262:110166. [DOI] [PubMed]
Huang XY, Hu QP, Shi HY, Zheng YY, Hu RR, 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. [DOI] [PubMed]
Yang MT, Lin YC, Ho WH, Liu CL, Lee WT. Everolimus is better than rapamycin in attenuating neuroinflammation in kainic acid-induced seizures. J Neuroinflammation. 2017;14:15. [DOI] [PubMed] [PMC]
Tao P, Jing Z, Shou-Hong G, Shu-Juan P, Jian-Peng J, Wen-Quan L, et al. Effects of leptin on norepinephrine in acute ischemic stroke. Pharmazie. 2019;74:47780. [DOI] [PubMed]
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;16:891900. [DOI] [PubMed] [PMC]
Zhang Q, Zhang Y, Diamond S, Boer J, Harris JJ, Li Y, 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;42:165662. [DOI] [PubMed]
Hazell AS, Afadlal S, Cheresh DA, Azar A. Treatment of rats with the JAK-2 inhibitor fedratinib does not lead to experimental Wernicke’s encephalopathy. Neurosci Lett. 2017;642:1637. [DOI] [PubMed]
Zhou Y, Li C, Li D, Zheng Y, Wang J. IL-5 blocks apoptosis and tau hyperphosphorylation induced by Aβ25-35 peptide in PC12 cells. J Physiol Biochem. 2017;73:25966. [DOI] [PubMed]
Feng Y, Shen J, He J, Lu M. Schizophrenia and cell senescence candidate genes screening, machine learning, diagnostic models, and drug prediction. Front Psychiatry. 2023;14:1105987. [DOI] [PubMed] [PMC]
Duan QQ, Wang H, Su WM, Gu XJ, Shen XF, Jiang Z, et al. TBK1, a prioritized drug repurposing target for amyotrophic lateral sclerosis: evidence from druggable genome Mendelian randomization and pharmacological verification in vitro. BMC Med. 2024;22:96. [DOI] [PubMed] [PMC]
Zhou G, Xiang T, Xu Y, He B, Wu L, Zhu G, et al. Fruquintinib/HMPL-013 ameliorates cognitive impairments and pathology in a mouse model of cerebral amyloid angiopathy (CAA). Eur J Pharmacol. 2023;939:175446. [DOI] [PubMed]
Xue W, Zhao Y, Xiao Z, Wu X, Ma D, Han J, et al. Epidermal growth factor receptor-extracellular-regulated kinase blockade upregulates TRIM32 signaling cascade and promotes neurogenesis after spinal cord injury. Stem Cells. 2020;38:11833. [DOI] [PubMed]
Mizuno M, Sotoyama H, Namba H, Shibuya M, Eda T, Wang R, et al. ErbB inhibitors ameliorate behavioral impairments of an animal model for schizophrenia: implication of their dopamine-modulatory actions. Transl Psychiatry. 2013;3:e252. [DOI] [PubMed] [PMC]
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;671:439. [DOI] [PubMed]
Lopez Corcino Y, Gonzalez Ferrer S, Mantilla LE, Trikeriotis S, Yu JS, Kim S, et al. 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. [DOI] [PubMed] [PMC]
Li D, Lian L, Huang L, Gamdzyk M, Huang Y, Doycheva D, et al. Delayed recanalization reduced neuronal apoptosis and neurological deficits by enhancing liver-derived trefoil factor 3-mediated neuroprotection via LINGO2/EGFR/Src signaling pathway after middle cerebral artery occlusion in rats. Exp Neurol. 2024;371:114607. [DOI] [PubMed] [PMC]
Dhamodharan J, Sekhar G, Muthuraman A. Epidermal Growth Factor Receptor Kinase Inhibitor Ameliorates β-Amyloid Oligomer-Induced Alzheimer Disease in Swiss Albino Mice. Molecules. 2022;27:5182. [DOI] [PubMed] [PMC]
Pastor-Alonso O, Durá I, Bernardo-Castro S, Varea E, Muro-García T, Martín-Suárez S, et al. HB-EGF activates EGFR to induce reactive neural stem cells in the mouse hippocampus after seizures. Life Sci Alliance. 2024;7:e202201840. [DOI] [PubMed] [PMC]
Ito M, Shichita T, Okada M, Komine R, Noguchi Y, Yoshimura A, et al. Bruton’s tyrosine kinase is essential for NLRP3 inflammasome activation and contributes to ischaemic brain injury. Nat Commun. 2015;6:7360. [DOI] [PubMed] [PMC]
Jin L, Mo Y, Yue EL, Liu Y, Liu KY. Ibrutinib ameliorates cerebral ischemia/reperfusion injury through autophagy activation and PI3K/Akt/mTOR signaling pathway in diabetic mice. Bioengineered. 2021;12:743245. [DOI] [PubMed] [PMC]
Yu CG, Bondada V, Iqbal H, Moore KL, Gensel JC, Bondada S, et al. Inhibition of Bruton Tyrosine Kinase Reduces Neuroimmune Cascade and Promotes Recovery after Spinal Cord Injury. Int J Mol Sci. 2021;23:355. [DOI] [PubMed] [PMC]
Torabi S, Anjamrooz SH, 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;54:35060. [DOI] [PubMed] [PMC]
Ekpenyong-Akiba AE, Poblocka M, Althubiti M, Rada M, Jurk D, Germano S, et al. Amelioration of age-related brain function decline by Bruton’s tyrosine kinase inhibition. Aging Cell. 2020;19:e13079. [DOI] [PubMed] [PMC]
Lee HJ, Jeon SG, Kim J, Kang RJ, Kim SM, Han KM, et al. Ibrutinib modulates Aβ/tau pathology, neuroinflammation, and cognitive function in mouse models of Alzheimer’s disease. Aging Cell. 2021;20:e13332. [DOI] [PubMed] [PMC]
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;14:44861. [DOI] [PubMed] [PMC]
Nam HY, Nam JH, Yoon G, Lee JY, Nam Y, Kang HJ, et al. Ibrutinib suppresses LPS-induced neuroinflammatory responses in BV2 microglial cells and wild-type mice. J Neuroinflammation. 2018;15:271. [DOI] [PubMed] [PMC]
Ghosh S, Mohammed Z, Singh I. Bruton’s tyrosine kinase drives neuroinflammation and anxiogenic behavior in mouse models of stress. J Neuroinflammation. 2021;18:289. [DOI] [PubMed] [PMC]
Li W, Ali T, He K, Liu Z, Shah FA, Ren Q, et al. Ibrutinib alleviates LPS-induced neuroinflammation and synaptic defects in a mouse model of depression. Brain Behav Immun. 2021;92:1024. [DOI] [PubMed]
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;170:6573. [DOI] [PubMed]
Huggett SB, Hatfield JS, Walters JD, McGeary JE, Welsh JW, Mackay TFC, et al. Ibrutinib as a potential therapeutic for cocaine use disorder. Transl Psychiatry. 2021;11:623. [DOI] [PubMed] [PMC]
Liu C, Yao K, Tian Q, Guo Y, Wang G, He P, et al. CXCR4-BTK axis mediate pyroptosis and lipid peroxidation in early brain injury after subarachnoid hemorrhage via NLRP3 inflammasome and NF-κB pathway. Redox Biol. 2023;68:102960. [DOI] [PubMed] [PMC]
Zheng C, Li W, Ali T, Peng Z, Liu J, Pan Z, et al. Ibrutinib Delays ALS Installation and Increases Survival of SOD1G93A Mice by Modulating PI3K/mTOR/Akt Signaling. J Neuroimmune Pharmacol. 2023;18:38396. [DOI] [PubMed]
Zhan Y, Krafft PR, Lekic T, Ma Q, Souvenir R, Zhang JH, et al. Imatinib preserves blood-brain barrier integrity following experimental subarachnoid hemorrhage in rats. J Neurosci Res. 2015;93:94103. [DOI] [PubMed] [PMC]
Shiba M, Fujimoto M, Kawakita F, Imanaka-Yoshida K, Yoshida T, Kanamaru K, et al. Effects of tenascin-C on early brain injury after subarachnoid hemorrhage in rats. Acta Neurochir Suppl. 2015;120:6973. [DOI] [PubMed]
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;25:22038. [DOI] [PubMed]
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;5:23847. [DOI] [PubMed]
Shiba M, Suzuki H, Fujimoto M, Shimojo N, Imanaka-Yoshida K, Yoshida T, et al. Imatinib mesylate prevents cerebral vasospasm after subarachnoid hemorrhage via inhibiting tenascin-C expression in rats. Neurobiol Dis. 2012;46:1729. [DOI] [PubMed]
Shiba M, Suzuki H, Fujimoto M, Shimojo N, Imanaka-Yoshida K, Yoshida T, et al. Role of platelet-derived growth factor in cerebral vasospasm after subarachnoid hemorrhage in rats. Acta Neurochir Suppl. 2013;115:21923. [DOI] [PubMed]
Ma Q, Huang B, Khatibi N, Rolland W 2nd, Suzuki H, Zhang JH, et al. PDGFR-α inhibition preserves blood-brain barrier after intracerebral hemorrhage. Ann Neurol. 2011;70:92031. [DOI] [PubMed] [PMC]
Yang P, Manaenko A, Xu F, Miao L, Wang G, Hu X, et al. Role of PDGF-D and PDGFR-β in neuroinflammation in experimental ICH mice model. Exp Neurol. 2016;283:15764. [DOI] [PubMed] [PMC]
Yang P, Wu J, Miao L, Manaenko A, Matei N, Zhang Y, 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:e390402. [DOI] [PubMed] [PMC]
Pearce WJ, Doan C, Carreon D, Kim D, Durrant LM, Manaenko A, et al. Imatinib attenuates cerebrovascular injury and phenotypic transformation after intracerebral hemorrhage in rats. Am J Physiol Regul Integr Comp Physiol. 2016;311:R1093104. [DOI] [PubMed] [PMC]
Ding J, Lian J, Wang J, Yang S, Li H, Shen H, et al. The role of Tenascin C in intracerebral hemorrhage-induced secondary brain injury in rats via induction of neuronal cell death and neuroinflammation. J Chem Neuroanat. 2022;125:102147. [DOI] [PubMed]
Sun Z, Gao C, Gao D, Sun R, Li W, Wang F, 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. [DOI] [PubMed] [PMC]
Sakai K, Takata F, Yamanaka G, Yasunaga M, Hashiguchi K, Tominaga K, et al. 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;145:15565. [DOI] [PubMed]
Klement W, Blaquiere M, Zub E, deBock F, Boux F, Barbier E, et al. A pericyte-glia scarring develops at the leaky capillaries in the hippocampus during seizure activity. Epilepsia. 2019;60:1399411. [DOI] [PubMed]
Singh S, Singh TG. Imatinib Attenuates Pentylenetetrazole Kindled and Pilocarpine Induced Recurrent Spontaneous Seizures in Mice. Neurochem Res. 2023;48:41834. [DOI] [PubMed]
Su EJ, Fredriksson L, Kanzawa M, Moore S, Folestad E, Stevenson TK, et al. Imatinib treatment reduces brain injury in a murine model of traumatic brain injury. Front Cell Neurosci. 2015;9:385. [DOI] [PubMed] [PMC]
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;24:118. [DOI] [PubMed] [PMC]
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;6:3949. [DOI] [PubMed]
Torrente D, Su EJ, Fredriksson L, Warnock M, Bushart D, Mann KM, et al. Compartmentalized Actions of the Plasminogen Activator Inhibitors, PAI-1 and Nsp, in Ischemic Stroke. Transl Stroke Res. 2022;13:80115. [DOI] [PubMed] [PMC]
Gardner LE, White JD, Eimerbrink MJ, Boehm GW, Chumley MJ. Imatinib methanesulfonate reduces hyperphosphorylation of tau following repeated peripheral exposure to lipopolysaccharide. Neuroscience. 2016;331:727. [DOI] [PubMed]
Weintraub MK, Bisson CM, Nouri JN, Vinson BT, Eimerbrink MJ, Kranjac D, et al. Imatinib methanesulfonate reduces hippocampal amyloid-beta and restores cognitive function following repeated endotoxin exposure. Brain Behav Immun. 2013;33:248. [DOI] [PubMed]
Sun W, Netzer WJ, Sinha A, Gindinova K, Chang E, Sinha SC. Development of Gleevec Analogues for Reducing Production of β-Amyloid Peptides through Shifting β-Cleavage of Amyloid Precursor Proteins. J Med Chem. 2019;62:312234. [DOI] [PubMed]
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 Alzheimers Dis. 2011;27:51120. [DOI] [PubMed]
Sutcliffe JG, Hedlund PB, Thomas EA, Bloom FE, Hilbush BS. Peripheral reduction of β-amyloid is sufficient to reduce brain β-amyloid: implications for Alzheimer’s disease. J Neurosci Res. 2011;89:80814. [DOI] [PubMed]
Hussain I, Fabrègue J, Anderes L, Ousson S, Borlat F, Eligert V, 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;288:252131. [DOI] [PubMed] [PMC]
Netzer WJ, Bettayeb K, Sinha SC, Flajolet M, Greengard P, Bustos V. Gleevec shifts APP processing from a β-cleavage to a nonamyloidogenic cleavage. Proc Natl Acad Sci U S A. 2017;114:138994. [DOI] [PubMed] [PMC]
Estrada LD, Chamorro D, Yañez MJ, Gonzalez M, Leal N, von Bernhardi R, et al. Reduction of Blood Amyloid-β Oligomers in Alzheimer’s Disease Transgenic Mice by c-Abl Kinase Inhibition. J Alzheimers Dis. 2016;54:1193205. [DOI] [PubMed]
Netzer WJ, Dou F, Cai D, Veach D, Jean S, Li Y, et al. Gleevec inhibits β-amyloid production but not Notch cleavage. Proc Natl Acad Sci U S A. 2003;100:124449. [DOI] [PubMed] [PMC]
Chu J, Lauretti E, Craige CP, 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 Alzheimers Dis. 2014;41:72937. [DOI] [PubMed]
Alvarez AR, Sandoval PC, Leal NR, Castro PU, Kosik KS. Activation of the neuronal c-Abl tyrosine kinase by amyloid-β-peptide and reactive oxygen species. Neurobiol Dis. 2004;17:32636. [DOI] [PubMed]
Cancino GI, Toledo EM, Leal NR, Hernandez DE, Yévenes LF, Inestrosa NC, et al. STI571 prevents apoptosis, tau phosphorylation and behavioural impairments induced by Alzheimer’s β-amyloid deposits. Brain. 2008;131:242542. [DOI] [PubMed]
Kerridge C, Belyaev ND, Nalivaeva NN, Turner AJ. The Aβ-clearance protein transthyretin, like neprilysin, is epigenetically regulated by the amyloid precursor protein intracellular domain. J Neurochem. 2014;130:41931. [DOI] [PubMed]
He G, Luo W, Li P, Remmers C, Netzer WJ, Hendrick J, et al. Gamma-secretase activating protein is a therapeutic target for Alzheimer’s disease. Nature. 2010;467:958. [DOI] [PubMed] [PMC]
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;41:43148. [DOI] [PubMed] [PMC]
Lee HJ, Hoe HS. Repurposing ibrutinib: therapeutic effects and implications for translational approaches in Alzheimer’s disease. Neural Regen Res. 2023;18:21945. [DOI] [PubMed] [PMC]
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;94:14917. [DOI] [PubMed]
Netzer WJ, Sinha A, Ghias M, Chang E, Gindinova K, Mui E, et al. Stretching the structural envelope of Imatinib to reduce β-amyloid production by modulating both β- and γ-secretase cleavages of APP. Front Chem. 2024;12:1381205. [DOI] [PubMed] [PMC]
Ren Y, Chen J, Wu X, Gui C, Mao K, Zou F, et al. Role of c-Abl-GSK3β Signaling in MPP+-Induced Autophagy-Lysosomal Dysfunction. Toxicol Sci. 2018;165:23243. [DOI] [PubMed]
Wu R, Chen H, Ma J, He Q, Huang Q, Liu Q, et al. c-Abl-p38α signaling plays an important role in MPTP-induced neuronal death. Cell Death Differ. 2016;23:54252. [DOI] [PubMed] [PMC]
Imam SZ, Zhou Q, Yamamoto A, Valente AJ, Ali SF, Bains M, et al. Novel regulation of parkin function through c-Abl-mediated tyrosine phosphorylation: implications for Parkinson’s disease. J Neurosci. 2011;31:15763. [DOI] [PubMed] [PMC]
Yamamura Y, Morigaki R, Kasahara J, Yokoyama H, Tanabe A, Okita S, et al. Dopamine signaling negatively regulates striatal phosphorylation of Cdk5 at tyrosine 15 in mice. Front Cell Neurosci. 2013;7:12. [DOI] [PubMed] [PMC]
Marín T, Valls C, Jerez C, Huerta T, Elgueta D, Vidal RL, et al. The c-Abl/p73 pathway induces neurodegeneration in a Parkinson’s disease model. IBRO Neurosci Rep. 2022;13:37887. [DOI] [PubMed] [PMC]
Rusiecka I, Gągało I, Kocić I. Neuroprotective Activity of a Non-Covalent Imatinib+TP10 Conjugate in HT-22 Neuronal Cells In Vitro. Pharmaceutics. 2024;16:778. [DOI] [PubMed] [PMC]
Pan Y, Sun L, Wang J, Fu W, Fu Y, Wang J, et al. STI571 protects neuronal cells from neurotoxic prion protein fragment-induced apoptosis. Neuropharmacology. 2015;93:1918. [DOI] [PubMed]
Aguib Y, Heiseke A, Gilch S, Riemer C, Baier M, Schätzl HM, et al. Autophagy induction by trehalose counter-acts cellular prion infection. Autophagy. 2009;5:3619. [DOI] [PubMed]
Yun SW, Ertmer A, Flechsig E, Gilch S, Riederer P, Gerlach M, et al. The tyrosine kinase inhibitor Imatinib mesylate delays prion neuroinvasion by inhibiting prion propagation in the periphery. J Neurovirol. 2007;13:32837. [DOI] [PubMed]
Ertmer A, Gilch S, Yun SW, Flechsig E, Klebl B, Stein-Gerlach M, et al. The tyrosine kinase inhibitor STI571 induces cellular clearance of PrPSc in prion-infected cells. J Biol Chem. 2004;279:4191827. [DOI] [PubMed]
Rojas F, Gonzalez D, Cortes N, Ampuero E, Hernández DE, Fritz E, 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. [DOI] [PubMed] [PMC]
Kegel KB, Sapp E, Alexander J, Reeves P, Bleckmann D, Sobin L, et al. Huntingtin cleavage product A forms in neurons and is reduced by gamma-secretase inhibitors. Mol Neurodegener. 2010;5:58. [DOI] [PubMed] [PMC]
Nacer A, Movila A, Baer K, Mikolajczak SA, Kappe SH, Frevert U. Neuroimmunological blood brain barrier opening in experimental cerebral malaria. PLoS Pathog. 2012;8:e1002982. [DOI] [PubMed] [PMC]
Vlasic V, Simakajornboon N, Gozal E, Gozal D. PDGF-β receptor expression in the dorsocaudal brainstem parallels hypoxic ventilatory depression in the developing rat. Pediatr Res. 2001;50:23641. [DOI] [PubMed]
Yáñez MJ, Belbin O, Estrada LD, Leal N, Contreras PS, Lleó A, et al. c-Abl links APP-BACE1 interaction promoting APP amyloidogenic processing in Niemann-Pick type C disease. Biochim Biophys Acta. 2016;1862:215867. [DOI] [PubMed]
Marín T, Dulcey AE, Campos F, de la Fuente C, Acuña M, Castro J, 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. [DOI] [PubMed] [PMC]
Yañez MJ, Campos F, Marín T, Klein AD, Futerman AH, Alvarez AR, et al. c-Abl activates RIPK3 signaling in Gaucher disease. Biochim Biophys Acta Mol Basis Dis. 2021;1867:166089. [DOI] [PubMed]
Potula R, Dhillion N, Sui Y, Zien CA, Funa K, Pinson D, et al. Association of platelet-derived growth factor-B chain with simian human immunodeficiency virus encephalitis. Am J Pathol. 2004;165:81524. [DOI] [PubMed] [PMC]
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;158:37390. [DOI] [PubMed]
Shang J, Li W, Zhang H, Wang W, Liu N, Gao D, et al. C-kit controls blood-brain barrier permeability by regulating caveolae-mediated transcytosis after chronic cerebral hypoperfusion. Biomed Pharmacother. 2024;170:115778. [DOI] [PubMed]
Pokharel S, Gliyazova NS, Dandepally SR, Williams AL, Ibeanu GC. Neuroprotective effects of an in vitro BBB permeable phenoxythiophene sulfonamide small molecule in glutamate-induced oxidative injury. Exp Ther Med. 2022;23:79. [DOI] [PubMed] [PMC]
Rezzola S, Guerra J, Krishna Chandran AM, Loda A, Cancarini A, Sacristani P, et al. VEGF-Independent Activation of Müller Cells by the Vitreous from Proliferative Diabetic Retinopathy Patients. Int J Mol Sci. 2021;22:2179. [DOI] [PubMed] [PMC]
Rajendran R, Rajendran V, Böttiger G, Stadelmann C, Shirvanchi K, von Au L, et al. The small molecule fibroblast growth factor receptor inhibitor infigratinib exerts anti-inflammatory effects and remyelination in a model of multiple sclerosis. Br J Pharmacol. 2023;180:29893007. [DOI] [PubMed]
Jia JN, Yin XX, Li Q, Guan QW, Yang N, Chen KN, et al. Neuroprotective Effects of the Anti-cancer Drug Lapatinib Against Epileptic Seizures via Suppressing Glutathione Peroxidase 4-Dependent Ferroptosis. Front Pharmacol. 2020;11:601572. [DOI] [PubMed] [PMC]
Xu HY, Sun YJ, Sun YY, Wu YJ, Xu MY, Chen LP, et al. Lapatinib alleviates TOCP-induced axonal damage in the spinal cord of mouse. Neuropharmacology. 2021;189:108535. [DOI] [PubMed]
Mansour HM, Fawzy HM, El-Khatib AS, Khattab MM. 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. [DOI] [PubMed]
Mansour HM, Fawzy HM, El-Khatib AS, Khattab MM. Inhibition of mitochondrial pyruvate carrier 1 by lapatinib ditosylate mitigates Alzheimer’s-like disease in D-galactose/ovariectomized rats. Neurochem Int. 2021;150:105178. [DOI] [PubMed]
Mansour HM, Mohamed AF, Khattab MM, El-Khatib AS. Unveiling the therapeutic prospects of EGFR inhibition in rotenone-mediated parkinsonism in rats: Modulation of dopamine D3 receptor. Brain Res. 2024;1834:148893. [DOI] [PubMed]
Saul S, Karim M, Ghita L, Huang PT, Chiu W, Durán V, et al. Anticancer pan-ErbB inhibitors reduce inflammation and tissue injury and exert broad-spectrum antiviral effects. J Clin Invest. 2023;133:e169510. [DOI] [PubMed] [PMC]
Zavvarian MM, Hong J, Khazaei M, Chio JCT, Wang J, Badner A, et al. The Protein Kinase Inhibitor Midostaurin Improves Functional Neurological Recovery and Attenuates Inflammatory Changes Following Traumatic Cervical Spinal Cord Injury. Biomolecules. 2021;11:972. [DOI] [PubMed] [PMC]
Esteban-Martos A, Brokate-Llanos AM, Real LM, Melgar-Locatelli S, de Rojas I, Castro-Zavala A, et al. A Functional Pipeline of Genome-Wide Association Data Leads to Midostaurin as a Repurposed Drug for Alzheimer’s Disease. Int J Mol Sci. 2023;24:12079. [DOI] [PubMed] [PMC]
Dent P, Booth L, Roberts JL, Poklepovic A, Cridebring D, Reiman EM. Inhibition of heat shock proteins increases autophagosome formation, and reduces the expression of APP, Tau, SOD1 G93A and TDP-43. Aging (Albany NY). 2021;13:17097117. [DOI] [PubMed] [PMC]
Dent P, Booth L, Roberts JL, Poklepovic A, Martinez J, Cridebring D, et al. AR12 increases BAG3 expression which is essential for Tau and APP degradation via LC3-associated phagocytosis and macroautophagy. Aging (Albany NY). 2022;14:822142. [DOI] [PubMed] [PMC]
Kitaoka Y, Sase K, Tsukahara C, Fujita N, Arizono I, Kogo J, et al. Axonal Protection by Netarsudil, a ROCK Inhibitor, Is Linked to an AMPK-Autophagy Pathway in TNF-Induced Optic Nerve Degeneration. Invest Ophthalmol Vis Sci. 2022;63:4. [DOI] [PubMed] [PMC]
Attia GM, Elmansy RA, Elsaed WM. Neuroprotective effect of nilotinib on pentylenetetrazol-induced epilepsy in adult rat hippocampus: involvement of oxidative stress, autophagy, inflammation, and apoptosis. Folia Neuropathol. 2019;57:14660. [DOI] [PubMed]
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. [DOI] [PubMed] [PMC]
Liu X, Hebron M, Shi W, Lonskaya I, Moussa CE. Ubiquitin specific protease-13 independently regulates parkin ubiquitination and alpha-synuclein clearance in alpha-synucleinopathies. Hum Mol Genet. 2019;28:54860. [DOI] [PubMed]
Mahul-Mellier AL, Fauvet B, Gysbers A, Dikiy I, Oueslati A, Georgeon S, 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;23:285879. [DOI] [PubMed] [PMC]
Imberdis T, Negri J, Ramalingam N, Terry-Kantor E, Ho GPH, Fanning S, 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 U S A. 2019;116:207609. [DOI] [PubMed] [PMC]
Adlimoghaddam A, Odero GG, Glazner G, Turner RS, Albensi BC. Nilotinib Improves Bioenergetic Profiling in Brain Astroglia in the 3xTg Mouse Model of Alzheimer’s Disease. Aging Dis. 2021;12:44165. [DOI] [PubMed] [PMC]
La Barbera L, Vedele F, Nobili A, Krashia P, Spoleti E, Latagliata EC, et al. Nilotinib restores memory function by preventing dopaminergic neuron degeneration in a mouse model of Alzheimer’s Disease. Prog Neurobiol. 2021;202:102031. [DOI] [PubMed]
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;17:127880. [DOI] [PubMed] [PMC]
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. [DOI] [PubMed]
Kuo YC, Tsai HC, Rajesh R. Glutathione Liposomes Carrying Ceftriaxone, FK506, and Nilotinib to Control Overexpressed Dopamine Markers and Apoptotic Factors in Neurons. ACS Biomater Sci Eng. 2021;7:324255. [DOI] [PubMed]
Kim H, Shin JY, Jo A, Kim JH, Park S, Choi JY, et al. Parkin interacting substrate phosphorylation by c-Abl drives dopaminergic neurodegeneration. Brain. 2021;144:367491. [DOI] [PubMed] [PMC]
Peikert K, Federti E, Matte A, Constantin G, Pietronigro EC, Fabene PF, et al. Therapeutic targeting of Lyn kinase to treat chorea-acanthocytosis. Acta Neuropathol Commun. 2021;9:81. [DOI] [PubMed] [PMC]
Federti E, Matte A, Riccardi V, Peikert K, Alper SL, Danek A, et al. Adaptative Up-Regulation of PRX2 and PRX5 Expression Characterizes Brain from a Mouse Model of Chorea-Acanthocytosis. Antioxidants (Basel). 2021;11:76. [DOI] [PubMed] [PMC]
Kim J, Lee HJ, Park JH, Cha BY, Hoe HS. Nilotinib modulates LPS-induced cognitive impairment and neuroinflammatory responses by regulating P38/STAT3 signaling. J Neuroinflammation. 2022;19:187. [DOI] [PubMed] [PMC]
Unterman I, Bloch I, Cazacu S, Kazimirsky G, Ben-Zeev B, Berman BP, et al. Expanding the MECP2 network using comparative genomics reveals potential therapeutic targets for Rett syndrome. Elife. 2021;10:e67085. [DOI] [PubMed] [PMC]
Campbell GR, Rawat P, Teodorof-Diedrich C, Spector SA. IRAK1 inhibition blocks the HIV-1 RNA mediated pro-inflammatory cytokine response from microglia. J Gen Virol. 2023;104:001858. [DOI] [PubMed] [PMC]
Hor JH, Soh ES, Tan LY, Lim VJW, Santosa MM, Winanto, et al. Cell cycle inhibitors protect motor neurons in an organoid model of Spinal Muscular Atrophy. Cell Death Dis. 2018;9:1100. [DOI] [PubMed] [PMC]
Chao AC, Chen CH, Chang SH, Huang CT, Hwang WC, Yang DI. Id1 and Sonic Hedgehog Mediate Cell Cycle Reentry and Apoptosis Induced by Amyloid Beta-Peptide in Post-mitotic Cortical Neurons. Mol Neurobiol. 2019;56:46589. [DOI] [PubMed]
Esteras N, Alquézar C, Bartolomé F, de la Encarnación A, Bermejo-Pareja F, Molina JA, et al. G1/S Cell Cycle Checkpoint Dysfunction in Lymphoblasts from Sporadic Parkinson’s Disease Patients. Mol Neurobiol. 2015;52:38698. [DOI] [PubMed]
Javidnia M, Hebron ML, Xin Y, Kinney NG, Moussa CE. Pazopanib Reduces Phosphorylated Tau Levels and Alters Astrocytes in a Mouse Model of Tauopathy. J Alzheimers Dis. 2017;60:46181. [DOI] [PubMed] [PMC]
Yang Y, Li G, Zhao D, Yu H, Zheng X, Peng X, 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;6:281221. [DOI] [PubMed] [PMC]
Ma K, Pham T, Wang J, O-Sullivan I, DiCamillo A, Du S, et al. Nanoparticle-based inhibition of vascular endothelial growth factor receptors alleviates osteoarthritis pain and cartilage damage. Sci Adv. 2024;10:eadi5501. [DOI] [PubMed] [PMC]
Mansour HM, Mohamed AF, Khattab MM, El-Khatib AS. Pazopanib ameliorates rotenone-induced Parkinsonism in rats by suppressing multiple regulated cell death mechanisms. Food Chem Toxicol. 2023;181:114069. [DOI] [PubMed]
Sun HY, Wu J, Wang R, Zhang S, Xu H, Kaznacheyeva Е, et al. Pazopanib alleviates neuroinflammation and protects dopaminergic neurons in LPS-stimulated mouse model by inhibiting MEK4-JNK-AP-1 pathway. Acta Pharmacol Sin. 2023;44:113548. [DOI] [PubMed] [PMC]
Abdelhady R, Younis NS, Ali O, Shehata S, Sayed RH, Nadeem RI. Cognitive enhancing effects of pazopanib in D‑galactose/ovariectomized Alzheimer’s rat model: insights into the role of RIPK1/RIPK3/MLKL necroptosis signaling pathway. Inflammopharmacology. 2023;31:271929. [DOI] [PubMed] [PMC]
Li M, Li Z, Ren H, Jin WN, Wood K, Liu Q, et al. Colony stimulating factor 1 receptor inhibition eliminates microglia and attenuates brain injury after intracerebral hemorrhage. J Cereb Blood Flow Metab. 2017;37:238395. [DOI] [PubMed] [PMC]
Shi E, Shi K, Qiu S, Sheth KN, Lawton MT, Ducruet AF. Chronic inflammation, cognitive impairment, and distal brain region alteration following intracerebral hemorrhage. FASEB J. 2019;33:961626. [DOI] [PubMed]
Li X, Gao X, Zhang W, Liu M, Han Z, Li M, et al. Microglial replacement in the aged brain restricts neuroinflammation following intracerebral hemorrhage. Cell Death Dis. 2022;13:33. [DOI] [PubMed] [PMC]
Heinz R, Brandenburg S, Nieminen-Kelhä M, Kremenetskaia I, Boehm-Sturm P, Vajkoczy P, et al. Microglia as target for anti-inflammatory approaches to prevent secondary brain injury after subarachnoid hemorrhage (SAH). J Neuroinflammation. 2021;18:36. [DOI] [PubMed] [PMC]
Shen Q, Zhang G. Depletion of microglia mitigates cerebrovascular dysfunction in diet-induced obesity mice. Am J Physiol Endocrinol Metab. 2021;321:E36775. [DOI] [PubMed]
Rice RA, Spangenberg EE, Yamate-Morgan H, Lee RJ, Arora RP, Hernandez MX, et al. Elimination of Microglia Improves Functional Outcomes Following Extensive Neuronal Loss in the Hippocampus. J Neurosci. 2015;35:997789. [DOI] [PubMed] [PMC]
Shi Y, Manis M, Long J, Wang K, Sullivan PM, Remolina Serrano J, et al. Microglia drive APOE-dependent neurodegeneration in a tauopathy mouse model. J Exp Med. 2019;216:254661. [DOI] [PubMed] [PMC]
Sosna J, Philipp S, Albay R 3rd, Reyes-Ruiz JM, Baglietto-Vargas D, LaFerla FM, et al. 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. [DOI] [PubMed] [PMC]
Son Y, Jeong YJ, Shin NR, Oh SJ, Nam KR, Choi HD, et al. 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. [DOI] [PubMed] [PMC]
Crapser JD, Ochaba J, Soni N, Reidling JC, Thompson LM, Green KN. Microglial depletion prevents extracellular matrix changes and striatal volume reduction in a model of Huntington’s disease. Brain. 2020;143:26688. [DOI] [PubMed] [PMC]
Tahmasebi F, Pasbakhsh P, Mortezaee K, Madadi S, Barati S, Kashani IR. Effect of the CSF1R inhibitor PLX3397 on remyelination of corpus callosum in a cuprizone-induced demyelination mouse model. J Cell Biochem. 2019;120:1057686. [DOI] [PubMed]
Groh J, Klein D, Berve K, West BL, Martini R. Targeting microglia attenuates neuroinflammation-related neural damage in mice carrying human PLP1 mutations. Glia. 2019;67:27790. [DOI] [PubMed]
Tahmasebi F, Pasbakhsh P, Barati S, Madadi S, Kashani IR. The effect of microglial ablation and mesenchymal stem cell transplantation on a cuprizone-induced demyelination model. J Cell Physiol. 2021;236:355264. [DOI] [PubMed]
Qu W, Johnson A, Kim JH, Lukowicz A, Svedberg D, Cvetanovic M. Inhibition of colony-stimulating factor 1 receptor early in disease ameliorates motor deficits in SCA1 mice. J Neuroinflammation. 2017;14:107. [DOI] [PubMed] [PMC]
Pinto B, Morelli G, Rastogi M, Savardi A, Fumagalli A, Petretto A, et al. Rescuing Over-activated Microglia Restores Cognitive Performance in Juvenile Animals of the Dp(16) Mouse Model of Down Syndrome. Neuron. 2020;108:887904. [DOI] [PubMed] [PMC]
Ueta Y, Miyata M. Brainstem local microglia induce whisker map plasticity in the thalamus after peripheral nerve injury. Cell Rep. 2021;34:108823. [DOI] [PubMed]
da Silva MCM, Gomes GF, de Barros Fernandes H, da Silva AM, Teixeira AL, Moreira FA, et al. Inhibition of CSF1R, a receptor involved in microglia viability, alters behavioral and molecular changes induced by cocaine. Sci Rep. 2021;11:15989. [DOI] [PubMed] [PMC]
Zhang D, Li S, Hou L, Jing L, Ruan Z, Peng B, et al. Microglial activation contributes to cognitive impairments in rotenone-induced mouse Parkinson’s disease model. J Neuroinflammation. 2021;18:4. [DOI] [PubMed] [PMC]
Stoll AC, Kemp CJ, Patterson JR, Kubik M, Kuhn N, Benskey M, et al. Microglial depletion does not impact alpha-synuclein aggregation or nigrostriatal degeneration in the rat preformed fibril model. Res Sq 2890683 [Preprint]. 2023 [cited 2025 May 26]. Available from: https://www.researchsquare.com/article/rs-2890683/v1
Stoll AC, Kemp CJ, Patterson JR, Kubik M, Kuhn N, Benskey M, et al. Alpha-synuclein inclusion responsive microglia are resistant to CSF1R inhibition. J Neuroinflammation. 2024;21:108. [DOI] [PubMed] [PMC]
Ni RJ, Wang YY, Gao TH, Wang QR, Wei JX, Zhao LS, et al. Depletion of microglia with PLX3397 attenuates MK-801-induced hyperactivity associated with regulating inflammation-related genes in the brain. Zool Res. 2023;44:54355. [DOI] [PubMed] [PMC]
Xie ST, Fan WC, Zhao XS, Ma XY, Li ZL, Zhao YR, et al. Proinflammatory activation of microglia in the cerebellum hyperexcites Purkinje cells to trigger ataxia. Pharmacol Res. 2023;191:106773. [DOI] [PubMed]
Spencer KA, Mulholland M, Snell J, Howe M, James K, Hanaford AR, et al. Volatile anaesthetic toxicity in the genetic mitochondrial disease Leigh syndrome. Br J Anaesth. 2023;131:83246. [DOI] [PubMed] [PMC]
Daneshgar N, Leidinger MR, Le S, Hefti M, Prigione A, Dai DF. Activated microglia and neuroinflammation as a pathogenic mechanism in Leigh syndrome. Front Neurosci. 2023;16:1068498. [DOI] [PubMed] [PMC]
El Jordi O, Fischer KD, Meyer TB, Atwood BK, Oblak AL, Pan RW, et al. Microglial knockdown does not affect acute withdrawal but delays analgesic tolerance from oxycodone in male and female C57BL/6J mice. Adv Drug Alcohol Res. 2022;2:10848. [DOI] [PubMed] [PMC]
Wang L, Wang X, Deng L, Zhang H, He B, Cao W, et al. Pexidartinib (PLX3397) through restoring hippocampal synaptic plasticity ameliorates social isolation-induced mood disorders. Int Immunopharmacol. 2022;113:109436. [DOI] [PubMed] [PMC]
Wang Y, Wernersbach I, Strehle J, Li S, Appel D, Klein M, et al. Early posttraumatic CSF1R inhibition via PLX3397 leads to time- and sex-dependent effects on inflammation and neuronal maintenance after traumatic brain injury in mice. Brain Behav Immun. 2022;106:4966. [DOI] [PubMed]
Saika F, Fukazawa Y, Hatano Y, Kishioka S, Hino Y, Hino S, et al. Sexually dimorphic effects of pexidartinib on nerve injury-induced neuropathic pain in mice. Glia. 2024;72:140217. [DOI] [PubMed]
Tian J, Guo S, Chen H, Peng JJ, Jia MM, Li NS, et al. Combination of Emricasan with Ponatinib Synergistically Reduces Ischemia/Reperfusion Injury in Rat Brain Through Simultaneous Prevention of Apoptosis and Necroptosis. Transl Stroke Res. 2018;9:38292. [DOI] [PubMed]
Zhang Y, Zhang M, Zhu W, Pan X, Wang Q, Gao X, et al. Role of Elevated Thrombospondin-1 in Kainic Acid-Induced Status Epilepticus. Neurosci Bull. 2020;36:26376. [DOI] [PubMed] [PMC]
Choi JP, Wang R, Yang X, Wang X, Wang L, Ting KK, et al. Ponatinib (AP24534) inhibits MEKK3-KLF signaling and prevents formation and progression of cerebral cavernous malformations. Sci Adv. 2018;4:eaau0731. [DOI] [PubMed] [PMC]
Han KM, Kang RJ, Jeon H, Lee HJ, Lee JS, Park H, et al. Regorafenib Regulates AD Pathology, Neuroinflammation, and Dendritic Spinogenesis in Cells and a Mouse Model of AD. Cells. 2020;9:1655. [DOI] [PubMed] [PMC]
Maher P, Conti B. Deciphering the pathways that protect from IL-13-mediated potentiation of oxidative stress-induced dopaminergic nerve cell death. Cytokine. 2018;103:11420. [DOI] [PubMed] [PMC]
Hosseini A, Gharibi T, Mohammadzadeh A, Ebrahimi-Kalan A, Jadidi-Niaragh F, Babaloo Z, et al. Ruxolitinib attenuates experimental autoimmune encephalomyelitis (EAE) development as animal models of multiple sclerosis (MS). Life Sci. 2021;276:119395. [DOI] [PubMed]
Yu X, Lv J, Wu J, Chen Y, Chen F, Wang L. The autoimmune encephalitis-related cytokine TSLP in the brain primes neuroinflammation by activating the JAK2-NLRP3 axis. Clin Exp Immunol. 2022;207:11322. [DOI] [PubMed] [PMC]
Sharma S, Risen S, Gilberto VS, Boland S, Chatterjee A, Moreno JA, et al. Targeted-Neuroinflammation Mitigation Using Inflammasome-Inhibiting Nanoligomers is Therapeutic in an Experimental Autoimmune Encephalomyelitis Mouse Model. ACS Chem Neurosci. 2024;15:1596608. [DOI] [PubMed]
Sullivan KD, Lewis HC, Hill AA, Pandey A, Jackson LP, Cabral JM, et al. Trisomy 21 consistently activates the interferon response. Elife. 2016;5:e16220. [DOI] [PubMed] [PMC]
Takata F, Dohgu S, Sakaguchi S, Sakai K, Yamanaka G, Iwao T, et al. Oncostatin-M-Reactive Pericytes Aggravate Blood-Brain Barrier Dysfunction by Activating JAK/STAT3 Signaling In Vitro. Neuroscience. 2019;422:1220. [DOI] [PubMed]
Haile WB, Gavegnano C, Tao S, Jiang Y, Schinazi RF, Tyor WR. The Janus kinase inhibitor ruxolitinib reduces HIV replication in human macrophages and ameliorates HIV encephalitis in a murine model. Neurobiol Dis. 2016;92:13743. [DOI] [PubMed] [PMC]
Zhang J, He H, Qiao Y, Zhou T, He H, Yi S, et al. Priming of microglia with IFN-γ impairs adult hippocampal neurogenesis and leads to depression-like behaviors and cognitive defects. Glia. 2020;68:267492. [DOI] [PubMed]
Chen X, Gao C, Yan Y, Cheng Z, Chen G, Rui T, et al. Ruxolitinib exerts neuroprotection via repressing ferroptosis in a mouse model of traumatic brain injury. Exp Neurol. 2021;342:113762. [DOI] [PubMed]
Peng J, Gao C, Chen X, Wang T, Luo C, Zhang M, et al. Ruxolitinib, a promising therapeutic candidate for traumatic brain injury through maintaining the homeostasis of cathepsin B. Exp Neurol. 2023;363:114347. [DOI] [PubMed]
Zhu H, Jian Z, Zhong Y, Ye Y, Zhang Y, Hu X, et al. Janus Kinase Inhibition Ameliorates Ischemic Stroke Injury and Neuroinflammation Through Reducing NLRP3 Inflammasome Activation via JAK2/STAT3 Pathway Inhibition. Front Immunol. 2021;12:714943. [DOI] [PubMed] [PMC]
Qian ZY, Kong RY, Zhang S, Wang BY, Chang J, Cao J, et al. Ruxolitinib attenuates secondary injury after traumatic spinal cord injury. Neural Regen Res. 2022;17:202935. [DOI] [PubMed] [PMC]
Cao J, Yu X, Liu J, Fu J, Wang B, Wu C, et al. Ruxolitinib improves the inflammatory microenvironment, restores glutamate homeostasis, and promotes functional recovery after spinal cord injury. Neural Regen Res. 2024;19:2499512. [DOI] [PubMed] [PMC]
Passos V, Henkel LM, Wang J, Zapatero-Belinchón FJ, Möller R, Sun G, et al. Innate immune response to SARS-CoV-2 infection contributes to neuronal damage in human iPSC-derived peripheral neurons. J Med Virol. 2024;96:e29455. [DOI] [PubMed]
Alquezar C, Esteras N, de la Encarnación A, Moreno F, López de Munain A, Martín-Requero Á. Increasing progranulin levels and blockade of the ERK1/2 pathway: upstream and downstream strategies for the treatment of progranulin deficient frontotemporal dementia. Eur Neuropsychopharmacol. 2015;25:386403. [DOI] [PubMed]
Ullrich M, Weber M, Post AM, Popp S, Grein J, Zechner M, et al. OCD-like behavior is caused by dysfunction of thalamo-amygdala circuits and upregulated TrkB/ERK-MAPK signaling as a result of SPRED2 deficiency. Mol Psychiatry. 2018;23:44458. [DOI] [PubMed] [PMC]
Huang HJ, Wang HT, Yeh TY, Lin BW, Shiao YJ, Lo YL, et al. Neuroprotective effect of selumetinib on acrolein-induced neurotoxicity. Sci Rep. 2021;11:12497. [DOI] [PubMed] [PMC]
Fei X, Dou YN, Wang L, Wu X, Huan Y, Wu S, et al. Homer1 promotes the conversion of A1 astrocytes to A2 astrocytes and improves the recovery of transgenic mice after intracerebral hemorrhage. J Neuroinflammation. 2022;19:67. [DOI] [PubMed] [PMC]
Talebi A, Rahnema M, Bigdeli MR. Effect of intravenous injection of antagomiR-1 on brain ischemia. Mol Biol Rep. 2019;46:114955. [DOI] [PubMed]
Guo W, Feng G, Miao Y, Liu G, Xu C. Rapamycin alleviates brain edema after focal cerebral ischemia reperfusion in rats. Immunopharmacol Immunotoxicol. 2014;36:21123. [DOI] [PubMed]
Wang J, Lin X, Mu Z, Shen F, Zhang L, Xie Q, et al. Rapamycin Increases Collateral Circulation in Rodent Brain after Focal Ischemia as detected by Multiple Modality Dynamic Imaging. Theranostics. 2019;9:492334. [DOI] [PubMed] [PMC]
Chauhan A, Sharma U, Jagannathan NR, Gupta YK. Rapamycin ameliorates brain metabolites alterations after transient focal ischemia in rats. Eur J Pharmacol. 2015;757:2833. [DOI] [PubMed]
Chi OZ, Kiss GK, Mellender SJ, Liu X, Weiss HR. Rapamycin decreased blood-brain barrier permeability in control but not in diabetic rats in early cerebral ischemia. Neurosci Lett. 2017;654:1722. [DOI] [PubMed]
Liu P, Yang X, Hei C, Meli Y, Niu J, Sun T, et al. Rapamycin Reduced Ischemic Brain Damage in Diabetic Animals Is Associated with Suppressions of mTOR and ERK1/2 Signaling. Int J Biol Sci. 2016;12:103240. [DOI] [PubMed] [PMC]
Liang G, Niu Y, Li Y, Wei A, Dong J, Zeng L. Rapamycin treatment starting at 24 h after cerebral ischemia/reperfusion exhibits protective effect on brain injury in rats. Zhejiang Da Xue Xue Bao Yi Xue Ban. 2018;47:44349. Chinese. [DOI] [PubMed] [PMC]
Yang X, Hei C, Liu P, Song Y, Thomas T, Tshimanga S, et al. Inhibition of mTOR Pathway by Rapamycin Reduces Brain Damage in Rats Subjected to Transient Forebrain Ischemia. Int J Biol Sci. 2015;11:142435. [DOI] [PubMed] [PMC]
Park JH, Ahn JH, Song M, Kim H, Park CW, Park YE, et al. A 2-Min Transient Ischemia Confers Cerebral Ischemic Tolerance in Non-Obese Gerbils, but Results in Neuronal Death in Obese Gerbils by Increasing Abnormal mTOR Activation-Mediated Oxidative Stress and Neuroinflammation. Cells. 2019;8:1126. [DOI] [PubMed] [PMC]
Mehta RI, Tsymbalyuk N, Ivanova S, Stokum JA, Woo K, Gerzanich V, et al. α-Endosulfine (ARPP-19e) Expression in a Rat Model of Stroke. J Neuropathol Exp Neurol. 2017;76:898907. [DOI] [PubMed] [PMC]
Carloni S, Buonocore G, Balduini W. Protective role of autophagy in neonatal hypoxia-ischemia induced brain injury. Neurobiol Dis. 2008;32:32939. [DOI] [PubMed]
Hei C, Liu P, Yang X, Niu J, Li PA. Inhibition of mTOR signaling Confers Protection against Cerebral Ischemic Injury in Acute Hyperglycemic Rats. Int J Biol Sci. 2017;13:87887. [DOI] [PubMed] [PMC]
Zhang B, Wu M, Liu L, Zhu Y, Kai J, Zeng L. Inhibiting mammalian target of rapamycin signaling pathway improves cognitive function in mice with chronic cerebral ischemia. Zhejiang Da Xue Xue Bao Yi Xue Ban. 2017;46:40512. Chinese. [DOI] [PubMed] [PMC]
Li Q, Zhang T, Wang J, Zhang Z, Zhai Y, Yang GY, et al. Rapamycin attenuates mitochondrial dysfunction via activation of mitophagy in experimental ischemic stroke. Biochem Biophys Res Commun. 2014;444:1828. [DOI] [PubMed]
Ghiglieri V, Pendolino V, Bagetta V, Sgobio C, Calabresi P, Picconi B. mTOR inhibitor rapamycin suppresses striatal post-ischemic LTP. Exp Neurol. 2010;226:32831. [DOI] [PubMed]
Fletcher L, Evans TM, Watts LT, Jimenez DF, Digicaylioglu M. Rapamycin treatment improves neuron viability in an in vitro model of stroke. PLoS One. 2013;8:e68281. [DOI] [PubMed] [PMC]
Xia DY, Li W, Qian HR, Yao S, Liu JG, Qi XK. Ischemia preconditioning is neuroprotective in a rat cerebral ischemic injury model through autophagy activation and apoptosis inhibition. Braz J Med Biol Res. 2013;46:5808. [DOI] [PubMed] [PMC]
Su J, Zhang T, Wang K, Zhu T, Li X. Autophagy activation contributes to the neuroprotection of remote ischemic perconditioning against focal cerebral ischemia in rats. Neurochem Res. 2014;39:206877. [DOI] [PubMed]
Sheng R, Zhang LS, Han R, Liu XQ, Gao B, Qin ZH. Autophagy activation is associated with neuroprotection in a rat model of focal cerebral ischemic preconditioning. Autophagy. 2010;6:48294. [DOI] [PubMed]
Yin L, Ye S, Chen Z, Zeng Y. Rapamycin preconditioning attenuates transient focal cerebral ischemia/reperfusion injury in mice. Int J Neurosci. 2012;122:74856. [DOI] [PubMed]
Sheng R, Liu XQ, Zhang LS, Gao B, Han R, Wu YQ, et al. Autophagy regulates endoplasmic reticulum stress in ischemic preconditioning. Autophagy. 2012;8:31025. [DOI] [PubMed]
Fan T, Huang Z, Chen L, Wang W, Zhang B, Xu Y, et al. Associations between autophagy, the ubiquitin-proteasome system and endoplasmic reticulum stress in hypoxia-deoxygenation or ischemia-reperfusion. Eur J Pharmacol. 2016;791:15767. [DOI] [PubMed]
Carloni S, Girelli S, Scopa C, Buonocore G, Longini M, Balduini W. Activation of autophagy and Akt/CREB signaling play an equivalent role in the neuroprotective effect of rapamycin in neonatal hypoxia-ischemia. Autophagy. 2010;6:36677. [DOI] [PubMed]
Hwang JY, Gertner M, Pontarelli F, Court-Vazquez B, Bennett MV, Ofengeim D, et al. Global ischemia induces lysosomal-mediated degradation of mTOR and activation of autophagy in hippocampal neurons destined to die. Cell Death Differ. 2017;24:31729. [DOI] [PubMed] [PMC]
XXie L, Sun F, Wang J, Mao X, Xie L, Yang SH, et al. mTOR signaling inhibition modulates macrophage/microglia-mediated neuroinflammation and secondary injury via regulatory T cells after focal ischemia. J Immunol. 2014;192:600919. [DOI] [PubMed] [PMC]
Lu Y, Li C, Chen Q, Liu P, Guo Q, Zhang Y, et al. Microthrombus-Targeting Micelles for Neurovascular Remodeling and Enhanced Microcirculatory Perfusion in Acute Ischemic Stroke. Adv Mater. 2019;31:e1808361. [DOI] [PubMed]
QQi H, Su FY, Wan S, Chen Y, Cheng YQ, Liu AJ. The antiaging activity and cerebral protection of rapamycin at micro-doses. CNS Neurosci Ther. 2014;20:9918. [DOI] [PubMed] [PMC]
Chauhan A, Sharma U, Jagannathan NR, Reeta KH, Gupta YK. Rapamycin protects against middle cerebral artery occlusion induced focal cerebral ischemia in rats. Behav Brain Res. 2011;225:6039. [DOI] [PubMed]
Beard DJ, Li Z, Schneider AM, Couch Y, Cipolla MJ, Buchan AM. Rapamycin Induces an eNOS (Endothelial Nitric Oxide Synthase) Dependent Increase in Brain Collateral Perfusion in Wistar and Spontaneously Hypertensive Rats. Stroke. 2020;51:283443. [DOI] [PubMed] [PMC]
Wang CY, Kim HH, Hiroi Y, Sawada N, Salomone S, Benjamin LE, et al. Obesity increases vascular senescence and susceptibility to ischemic injury through chronic activation of Akt and mTOR. Sci Signal. 2009;2:ra11. [DOI] [PubMed] [PMC]
Moradpour S, Aliaghaei A, Bigdeli M. Effect of Sertoli Cell Transplant and Rapamycin Pretreatment on Middle Cerebral Artery Occlusion-Induced Brain Ischemia in a Rat Model. Exp Clin Transplant. 2021;19:120411. [DOI] [PubMed]
Wang Y, Wang Y, Li S, Cui Y, Liang X, Shan J, et al. Functionalized nanoparticles with monocyte membranes and rapamycin achieve synergistic chemoimmunotherapy for reperfusion-induced injury in ischemic stroke. J Nanobiotechnology. 2021;19:331. [DOI] [PubMed] [PMC]
Zhang Y, Li D, Gao H, Zhao H, Zhang S, Li T. Rapamycin Alleviates Neuronal Injury and Modulates Microglial Activation After Cerebral Ischemia. Mol Neurobiol. 2024;61:5699717. [DOI] [PubMed]
Villa-González M, Rubio M, Martín-López G, Mallavibarrena PR, Vallés-Saiz L, Vivien D, et al. Pharmacological inhibition of mTORC1 reduces neural death and damage volume after MCAO by modulating microglial reactivity. Biol Direct. 2024;19:26. [DOI] [PubMed] [PMC]
Hei C, Zhou Y, Zhang C, Gao F, Cao M, Yuan S, et al. Rapamycin ameliorates brain damage and maintains mitochondrial dynamic balance in diabetic rats subjected to middle cerebral artery occlusion. Metab Brain Dis. 2023;38:40918. [DOI] [PubMed]
Nikolaeva I, Crowell B, Valenziano J, Meaney D, D’Arcangelo G. Beneficial Effects of Early mTORC1 Inhibition after Traumatic Brain Injury. J Neurotrauma. 2016;33:18393. [DOI] [PubMed] [PMC]
Song Q, Xie D, Pan S, Xu W. Rapamycin protects neurons from brain contusion-induced inflammatory reaction via modulation of microglial activation. Mol Med Rep. 2015;12:720310. [DOI] [PubMed] [PMC]
Fan YY, Nan F, Guo BL, Liao Y, Zhang MS, Guo J, et al. Effects of long-term rapamycin treatment on glial scar formation after cryogenic traumatic brain injury in mice. Neurosci Lett. 2018;678:6875. [DOI] [PubMed]
Chen Y, Meng J, Xu Q, Long T, Bi F, Chang C, et al. Rapamycin improves the neuroprotection effect of inhibition of NLRP3 inflammasome activation after TBI. Brain Res. 2019;1710:16372. [DOI] [PubMed]
Wang C, Hu Z, Zou Y, Xiang M, Jiang Y, Botchway BOA, et al. The post-therapeutic effect of rapamycin in mild traumatic brain-injured rats ensuing in the upregulation of autophagy and mitophagy. Cell Biol Int. 2017;41:103947. [DOI] [PubMed]
Erlich S, Alexandrovich A, Shohami E, Pinkas-Kramarski R. Rapamycin is a neuroprotective treatment for traumatic brain injury. Neurobiol Dis. 2007;26:8693. [DOI] [PubMed]
Ding K, Wang H, Wu Y, Zhang L, Xu J, Li T, et al. Rapamycin protects against apoptotic neuronal death and improves neurologic function after traumatic brain injury in mice via modulation of the mTOR-p53-Bax axis. J Surg Res. 2015;194:23947. [DOI] [PubMed]
Campolo M, Casili G, Lanza M, Filippone A, Cordaro M, Ardizzone A, et al. The inhibition of mammalian target of rapamycin (mTOR) in improving inflammatory response after traumatic brain injury. J Cell Mol Med. 2021;25:785566. [DOI] [PubMed] [PMC]
Sasaki K, Yamamoto S, Mutoh T, Tsuru Y, Taki Y, Kawashima R. Rapamycin protects against early brain injury independent of cerebral blood flow changes in a mouse model of subarachnoid haemorrhage. Clin Exp Pharmacol Physiol. 2018;45:85962. [DOI] [PubMed]
Zhao H, Ji Z, Tang D, Yan C, Zhao W, Gao C. Role of autophagy in early brain injury after subarachnoid hemorrhage in rats. Mol Biol Rep. 2013;40:81927. [DOI] [PubMed]
You W, Wang Z, Li H, Shen H, Xu X, Jia G, et al. Inhibition of mammalian target of rapamycin attenuates early brain injury through modulating microglial polarization after experimental subarachnoid hemorrhage in rats. J Neurol Sci. 2016;367:22431. [DOI] [PubMed]
Li J, Lu J, Mi Y, Shi Z, Chen C, Riley J, et al. Voltage-dependent anion channels (VDACs) promote mitophagy to protect neuron from death in an early brain injury following a subarachnoid hemorrhage in rats. Brain Res. 2014;1573:7483. [DOI] [PubMed]
Yamamoto S, Mutoh T, Sasaki K, Mutoh T, Taki Y. Central action of rapamycin on early ischemic injury and related cardiac depression following experimental subarachnoid hemorrhage. Brain Res Bull. 2019;144:8591. [DOI] [PubMed]
Jing CH, Wang L, Liu PP, Wu C, Ruan D, Chen G. Autophagy activation is associated with neuroprotection against apoptosis via a mitochondrial pathway in a rat model of subarachnoid hemorrhage. Neuroscience. 2012;213:14453. [DOI] [PubMed]
Zhang W, Khatibi NH, Yamaguchi-Okada M, Yan J, Chen C, Hu Q, et al. Mammalian target of rapamycin (mTOR) inhibition reduces cerebral vasospasm following a subarachnoid hemorrhage injury in canines. Exp Neurol. 2012;233:799806. [DOI] [PubMed]
Zhang X, Qin C, Jing Y, Yang D, Liu C, Gao F, et al. Therapeutic effects of rapamycin and surgical decompression in a rabbit spinal cord injury model. Cell Death Dis. 2020;11:567. [DOI] [PubMed] [PMC]
Liu D, Jia S, Sun D, Wang SY, Meng FC, Guo WC. Rapamycin repairs damaged nerve cells and neurological function in rats with spinal cord injury through ERK signaling pathway. J Biol Regul Homeost Agents. 2020;34:86573. [DOI] [PubMed]
Wang ZY, Lin JH, Muharram A, Liu WG. Beclin-1-mediated autophagy protects spinal cord neurons against mechanical injury-induced apoptosis. Apoptosis. 2014;19:93345. [PubMed]
Song Y, Xue H, Liu TT, Liu JM, Chen D. Rapamycin plays a neuroprotective effect after spinal cord injury via anti-inflammatory effects. J Biochem Mol Toxicol. 2015;29:2934. [DOI] [PubMed]
Tang P, Hou H, Zhang L, Lan X, Mao Z, Liu D, et al. Autophagy reduces neuronal damage and promotes locomotor recovery via inhibition of apoptosis after spinal cord injury in rats. Mol Neurobiol. 2014;49:27687. [DOI] [PubMed]
Goldshmit Y, Kanner S, Zacs M, Frisca F, Pinto AR, Currie PD, et al. Rapamycin increases neuronal survival, reduces inflammation and astrocyte proliferation after spinal cord injury. Mol Cell Neurosci. 2015;68:8291. [DOI] [PubMed]
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;54:241527. [DOI] [PubMed]
Chen HC, Fong TH, Hsu PW, Chiu WT. Multifaceted effects of rapamycin on functional recovery after spinal cord injury in rats through autophagy promotion, anti-inflammation, and neuroprotection. J Surg Res. 2013;179:e20310. [DOI] [PubMed]
Sekiguchi A, Kanno H, Ozawa H, Yamaya S, Itoi E. Rapamycin promotes autophagy and reduces neural tissue damage and locomotor impairment after spinal cord injury in mice. J Neurotrauma. 2012;29:94656. [DOI] [PubMed]
Hakim JS, Rodysill BR, Chen BK, Schmeichel AM, Yaszemski MJ, Windebank AJ, et al. Combinatorial tissue engineering partially restores function after spinal cord injury. J Tissue Eng Regen Med. 2019;13:85773. [DOI] [PubMed] [PMC]
Xiao X, Chen XY, Dong YH, Dong HR, Zhou LN, Ding YQ, et al. Pre-treatment of rapamycin transformed M2 microglia alleviates traumatic cervical spinal cord injury via AIM2 signaling pathway in vitro and in vivo. Int Immunopharmacol. 2023;121:110394. [DOI] [PubMed]
Shen K, Li X, Huang G, Yuan Z, Xie B, Chen T, et al. High rapamycin-loaded hollow mesoporous Prussian blue nanozyme targets lesion area of spinal cord injury to recover locomotor function. Biomaterials. 2023;303:122358. [DOI] [PubMed]
Lekic T, Krafft PR, Klebe D, Flores J, Rolland WB, Tang J, et al. PAR-1, -4, and the mTOR Pathway Following Germinal Matrix Hemorrhage. Acta Neurochir Suppl. 2016;121:2136. [DOI] [PubMed]
Lekic T, Klebe D, McBride DW, Manaenko A, Rolland WB, Flores JJ, et al. Protease-activated receptor 1 and 4 signal inhibition reduces preterm neonatal hemorrhagic brain injury. Stroke. 2015;46:17103. [DOI] [PubMed] [PMC]
Wang JP, Zhang MY. Role for Target of Rapamycin (mTOR) Signal Pathway in Regulating Neuronal Injury after Intracerebral Hemorrhage. Cell Physiol Biochem. 2017;41:14553. [DOI] [PubMed]
Lu Q, Gao L, Huang L, Ruan L, Yang J, Huang W, et al. Inhibition of mammalian target of rapamycin improves neurobehavioral deficit and modulates immune response after intracerebral hemorrhage in rat. J Neuroinflammation. 2014;11:44. [DOI] [PubMed] [PMC]
Li D, Liu F, Yang T, Jin T, Zhang H, Luo X, et al. Rapamycin protects against neuronal death and improves neurological function with modulation of microglia after experimental intracerebral hemorrhage in rats. Cell Mol Biol (Noisy-le-grand). 2016;62:6775. [PubMed]
Brewster AL, Lugo JN, Patil VV, Lee WL, Qian Y, Vanegas F, et al. Rapamycin reverses status epilepticus-induced memory deficits and dendritic damage. PLoS One. 2013;8:e57808. [DOI] [PubMed] [PMC]
Zhang H, Xie Y, Weng L, Zhang Y, Shi Q, Chen T, et al. Rapamycin improves learning and memory ability in ICR mice with pilocarpine-induced temporal lobe epilepsy. Zhejiang Da Xue Xue Bao Yi Xue Ban. 2013;42:6028. Chinese. [PubMed]
Raffo E, Coppola A, Ono T, Briggs SW, Galanopoulou AS. A pulse rapamycin therapy for infantile spasms and associated cognitive decline. Neurobiol Dis. 2011;43:3229. [DOI] [PubMed] [PMC]
Aghaie F, Shemshaki A, Rajabi M, Khatami P, Hosseini A. Rapamycin alleviates memory deficit against pentylenetetrazole-induced neural toxicity in Wistar male rats. Mol Biol Rep. 2021;48:508391. [DOI] [PubMed]
Bornstein R, James K, Stokes J, Park KY, Kayser EB, Snell J, et al. Differential effects of mTOR inhibition and dietary ketosis in a mouse model of subacute necrotizing encephalomyelopathy. Neurobiol Dis. 2022;163:105594. [DOI] [PubMed] [PMC]
Codeluppi S, Svensson CI, Hefferan MP, Valencia F, Silldorff MD, Oshiro M, et al. The Rheb-mTOR pathway is upregulated in reactive astrocytes of the injured spinal cord. J Neurosci. 2009;29:1093104. [DOI] [PubMed] [PMC]
Fang B, Li XQ, Bao NR, Tan WF, Chen FS, Pi XL, et al. Role of autophagy in the bimodal stage after spinal cord ischemia reperfusion injury in rats. Neuroscience. 2016;328:10716. [DOI] [PubMed]
Chang KT, Lin YL, Lin CT, Hong CJ, Cheng YH, Tsai MJ, et al. Neuroprotection in the Acute Stage Enables Functional Recovery Following Repair of Chronic Cervical Root Transection After a 3-Week Delay. Neurosurgery. 2020;87:82332. [DOI] [PubMed]
Oku H, Morishita S, Horie T, Kida T, Mimura M, Kojima S, et al. P7C3 Suppresses Neuroinflammation and Protects Retinal Ganglion Cells of Rats from Optic Nerve Crush. Invest Ophthalmol Vis Sci. 2017;58:487788. [DOI] [PubMed]
Inada T, Sato H, Hayashi Y, Hitomi S, Furukawa A, Ando M, et al. Rapamycin Accelerates Axon Regeneration Through Schwann Cell-mediated Autophagy Following Inferior Alveolar Nerve Transection in Rats. Neuroscience. 2021;468:4352. [DOI] [PubMed]
Li N, Wang F, Zhang Q, Jin M, Lu Y, Chen S, et al. Rapamycin mediates mTOR signaling in reactive astrocytes and reduces retinal ganglion cell loss. Exp Eye Res. 2018;176:109. [DOI] [PubMed]
Xu L, Zhang C, Jiang N, He D, Bai Y, Xin Y. Rapamycin combined with MCC950 to treat multiple sclerosis in experimental autoimmune encephalomyelitis. J Cell Biochem. 2019;120:51608. [DOI] [PubMed]
Lisi L, Navarra P, Cirocchi R, Sharp A, Stigliano E, Feinstein DL, et al. Rapamycin reduces clinical signs and neuropathic pain in a chronic model of experimental autoimmune encephalomyelitis. J Neuroimmunol. 2012;243:4351. [DOI] [PubMed]
Feng X, Hou H, Zou Y, Guo L. Defective autophagy is associated with neuronal injury in a mouse model of multiple sclerosis. Bosn J Basic Med Sci. 2017;17:95103. [DOI] [PubMed] [PMC]
Hou H, Cao R, Quan M, Sun Y, Sun H, Zhang J, et al. Rapamycin and fingolimod modulate Treg/Th17 cells in experimental autoimmune encephalomyelitis by regulating the Akt-mTOR and MAPK/ERK pathways. J Neuroimmunol. 2018;324:2634. [DOI] [PubMed]
Li Z, Nie L, Chen L, Sun Y, Guo L. Rapamycin alleviates inflammation by up-regulating TGF-β/Smad signaling in a mouse model of autoimmune encephalomyelitis. Nan Fang Yi Ke Da Xue Xue Bao. 2019;39:3542. Chinese. [DOI] [PubMed] [PMC]
Li Z, Nie L, Chen L, Sun Y, Li G. Rapamycin relieves inflammation of experimental autoimmune encephalomyelitis by altering the balance of Treg/Th17 in a mouse model. Neurosci Lett. 2019;705:3945. [DOI] [PubMed]
Hou H, Miao J, Cao R, Han M, Sun Y, Liu X, et al. Rapamycin Ameliorates Experimental Autoimmune Encephalomyelitis by Suppressing the mTOR-STAT3 Pathway. Neurochem Res. 2017;42:283140. [DOI] [PubMed]
Li Z, Chen L, Niu X, Liu J, Ping M, Li R, et al. Immunomodulatory synergy by combining atorvastatin and rapamycin in the treatment of experimental autoimmune encephalomyelitis (EAE). J Neuroimmunol. 2012;250:917. [DOI] [PubMed]
Esposito M, Ruffini F, Bellone M, Gagliani N, Battaglia M, Martino G, et al. Rapamycin inhibits relapsing experimental autoimmune encephalomyelitis by both effector and regulatory T cells modulation. J Neuroimmunol. 2010;220:5263. [DOI] [PubMed]
Donia M, Mangano K, Amoroso A, Mazzarino MC, Imbesi R, Castrogiovanni P, et al. Treatment with rapamycin ameliorates clinical and histological signs of protracted relapsing experimental allergic encephalomyelitis in Dark Agouti rats and induces expansion of peripheral CD4+CD25+Foxp3+ regulatory T cells. J Autoimmun. 2009;33:13540. [DOI] [PubMed]
Borim PA, Mimura LAN, Zorzella-Pezavento SFG, Polonio CM, Peron JPS, Sartori A, et al. Effect of Rapamycin on MOG-Reactive Immune Cells and Lipopolysaccharide-Activated Microglia: An In Vitro Approach for Screening New Therapies for Multiple Sclerosis. J Interferon Cytokine Res. 2022;42:15360. [DOI] [PubMed]
Zhang G, Yin L, Luo Z, Chen X, He Y, Yu X, et al. Effects and potential mechanisms of rapamycin on MPTP-induced acute Parkinson’s disease in mice. Ann Palliat Med. 2021;10:288997. [DOI] [PubMed]
Currim F, Singh J, Shinde A, Gohel D, Roy M, Singh K, et al. Exosome Release Is Modulated by the Mitochondrial-Lysosomal Crosstalk in Parkinson’s Disease Stress Conditions. Mol Neurobiol. 2021;58:181933. [DOI] [PubMed]
Pupyshev AB, Tenditnik MV, Ovsyukova MV, Akopyan AA, Dubrovina NI, Tikhonova MA. Restoration of Parkinson’s Disease-Like Deficits by Activating Autophagy through mTOR-Dependent and mTOR-Independent Mechanisms in Pharmacological and Transgenic Models of Parkinson’s Disease in Mice. Bull Exp Biol Med. 2021;171:42530. [DOI] [PubMed]
Guo Q, Wang B, Wang X, Smith WW, Zhu Y, Liu Z. Activation of Nrf2 in Astrocytes Suppressed PD-Like Phenotypes via Antioxidant and Autophagy Pathways in Rat and Drosophila Models. Cells. 2021;10:1850. [DOI] [PubMed] [PMC]
Srivastava IN, Shperdheja J, Baybis M, Ferguson T, Crino PB. mTOR pathway inhibition prevents neuroinflammation and neuronal death in a mouse model of cerebral palsy. Neurobiol Dis. 2016;85:14454. [DOI] [PubMed]
Abdulrahman BA, Tahir W, Doh-Ura K, Gilch S, Schatzl HM. Combining autophagy stimulators and cellulose ethers for therapy against prion disease. Prion. 2019;13:18596. [DOI] [PubMed] [PMC]
Cortes CJ, Qin K, Cook J, Solanki A, Mastrianni JA. Rapamycin delays disease onset and prevents PrP plaque deposition in a mouse model of Gerstmann-Sträussler-Scheinker disease. J Neurosci. 2012;32:12396405. [DOI] [PubMed] [PMC]
Ishibashi D, Homma T, Nakagaki T, Fuse T, Sano K, Takatsuki H, et al. Strain-Dependent Effect of Macroautophagy on Abnormally Folded Prion Protein Degradation in Infected Neuronal Cells. PLoS One. 2015;10:e0137958. [DOI] [PubMed] [PMC]
Wang H, Fu J, Xu X, Yang Z, Zhang T. Rapamycin Activates Mitophagy and Alleviates Cognitive and Synaptic Plasticity Deficits in a Mouse Model of Alzheimer’s Disease. J Gerontol A Biol Sci Med Sci. 2021;76:170713. [DOI] [PubMed]
Pupyshev AB, Belichenko VM, Tenditnik MV, Bashirzade AA, Dubrovina NI, Ovsyukova MV, et al. Combined induction of mTOR-dependent and mTOR-independent pathways of autophagy activation as an experimental therapy for Alzheimer’s disease-like pathology in a mouse model. Pharmacol Biochem Behav. 2022;217:173406. [DOI] [PubMed]
Van Skike CE, Hussong SA, Hernandez SF, Banh AQ, DeRosa N, Galvan V. mTOR Attenuation with Rapamycin Reverses Neurovascular Uncoupling and Memory Deficits in Mice Modeling Alzheimer’s Disease. J Neurosci. 2021;41:430520. [DOI] [PubMed] [PMC]
Kakoty V, C SK, Yang CH, Kumari S, Dubey SK, Taliyan R. Neuroprotective Effect of Lentivirus-Mediated FGF21 Gene Delivery in Experimental Alzheimer’s Disease is Augmented when Concerted with Rapamycin. Mol Neurobiol. 2022;59:265977. [DOI] [PubMed]
Lai C, Chen Z, Ding Y, Chen Q, Su S, Liu H, et al. Rapamycin Attenuated Zinc-Induced Tau Phosphorylation and Oxidative Stress in Rats: Involvement of Dual mTOR/p70S6K and Nrf2/HO-1 Pathways. Front Immunol. 2022;13:782434. [DOI] [PubMed] [PMC]
Zheng G, Wang L, Li X, Niu X, Xu G, Lv P. Rapamycin alleviates cognitive impairment in murine vascular dementia: The enhancement of mitophagy by PI3K/AKT/mTOR axis. Tissue Cell. 2021;69:101481. [DOI] [PubMed]
Ding Y, Liu H, Cen M, Tao Y, Lai C, Tang Z. Rapamycin Ameliorates Cognitive Impairments and Alzheimer’s Disease-Like Pathology with Restoring Mitochondrial Abnormality in the Hippocampus of Streptozotocin-Induced Diabetic Mice. Neurochem Res. 2021;46:26575. [DOI] [PubMed]
Jiang T, Zhang W, Wang Y, Zhang T, Wang H, Yang Z. Rapamycin Pretreatment Attenuates High Glucose-induced Alteration of Synaptic Transmission in Hippocampal Dentate Gyrus Neurons. Neuroscience. 2022;490:18292. [DOI] [PubMed]
Ravikumar B, Vacher C, Berger Z, Davies JE, Luo S, Oroz LG, et al. Inhibition of mTOR induces autophagy and reduces toxicity of polyglutamine expansions in fly and mouse models of Huntington disease. Nat Genet. 2004;36:58595. [DOI] [PubMed]
King MA, Hands S, Hafiz F, Mizushima N, Tolkovsky AM, Wyttenbach A. Rapamycin inhibits polyglutamine aggregation independently of autophagy by reducing protein synthesis. Mol Pharmacol. 2008;73:105263. [DOI] [PubMed]
Pereira GJ, Tressoldi N, Hirata H, Bincoletto C, Smaili SS. Autophagy as a neuroprotective mechanism against 3-nitropropionic acid-induced murine astrocyte cell death. Neurochem Res. 2013;38:241826. [DOI] [PubMed]
Sarkar S, Davies JE, Huang Z, Tunnacliffe A, Rubinsztein DC. Trehalose, a novel mTOR-independent autophagy enhancer, accelerates the clearance of mutant huntingtin and α-synuclein. J Biol Chem. 2007;282:564152. [DOI] [PubMed]
Chen LL, Wu JC, Wang LH, Wang J, Qin ZH, Difiglia M, et al. Rapamycin prevents the mutant huntingtin-suppressed GLT-1 expression in cultured astrocytes. Acta Pharmacol Sin. 2012;33:38592. [DOI] [PubMed] [PMC]
Sarkar S, Krishna G, Imarisio S, Saiki S, O’Kane CJ, Rubinsztein DC. A rational mechanism for combination treatment of Huntington’s disease using lithium and rapamycin. Hum Mol Genet. 2008;17:1708. [DOI] [PubMed]
Kolosova NG, Muraleva NA, Zhdankina AA, Stefanova NA, Fursova AZ, Blagosklonny MV. Prevention of age-related macular degeneration-like retinopathy by rapamycin in rats. Am J Pathol. 2012;181:4727. [DOI] [PubMed]
Kitaoka Y, Munemasa Y, Kojima K, Hirano A, Ueno S, Takagi H. Axonal protection by Nmnat3 overexpression with involvement of autophagy in optic nerve degeneration. Cell Death Dis. 2013;4:e860. [DOI] [PubMed] [PMC]
Russo R, Varano GP, Adornetto A, Nazio F, Tettamanti G, Girardello R, et al. Rapamycin and fasting sustain autophagy response activated by ischemia/reperfusion injury and promote retinal ganglion cell survival. Cell Death Dis. 2018;9:981. [DOI] [PubMed] [PMC]
Xu C, Zhang H, Liu C, Zhu Y, Wang X, Gao W, et al. Rapamycin inhibits Erk1/2-mediated neuronal apoptosis caused by cadmium. Oncotarget. 2015;6:2145267. [DOI] [PubMed] [PMC]
Wang T, Yuan Y, Zou H, Yang J, Zhao S, Ma Y, et al. The ER stress regulator Bip mediates cadmium-induced autophagy and neuronal senescence. Sci Rep. 2016;6:38091. [DOI] [PubMed] [PMC]
Xu C, Liu C, Liu L, Zhang R, Zhang H, Chen S, et al. Rapamycin prevents cadmium-induced neuronal cell death via targeting both mTORC1 and mTORC2 pathways. Neuropharmacology. 2015;97:3545. [DOI] [PubMed] [PMC]
Xu C, Wang X, Zhu Y, Dong X, Liu C, Zhang H, et al. Rapamycin ameliorates cadmium-induced activation of MAPK pathway and neuronal apoptosis by preventing mitochondrial ROS inactivation of PP2A. Neuropharmacology. 2016;105:27084. [DOI] [PubMed] [PMC]
Guo S, Xu N, Chen P, Liu Y, Qi X, Liu S, et al. Rapamycin Protects Spiral Ganglion Neurons from Gentamicin-Induced Degeneration In Vitro. J Assoc Res Otolaryngol. 2019;20:47587. [DOI] [PubMed] [PMC]
Caccamo A, Magrì A, Medina DX, Wisely EV, López-Aranda MF, Silva AJ, et al. mTOR regulates tau phosphorylation and degradation: implications for Alzheimer’s disease and other tauopathies. Aging Cell. 2013;12:37080. [DOI] [PubMed] [PMC]
Ozcelik S, Fraser G, Castets P, Schaeffer V, Skachokova Z, Breu K, et al. Rapamycin attenuates the progression of tau pathology in P301S tau transgenic mice. PLoS One. 2013;8:e62459. [DOI] [PubMed] [PMC]
Cullen V, Sardi SP, Ng J, Xu YH, Sun Y, Tomlinson JJ, et al. Acid β-glucosidase mutants linked to Gaucher disease, Parkinson disease, and Lewy body dementia alter α-synuclein processing. Ann Neurol. 2011;69:94053. [DOI] [PubMed]
Hoffmann AC, Minakaki G, Menges S, Salvi R, Savitskiy S, Kazman A, et al. Extracellular aggregated alpha synuclein primarily triggers lysosomal dysfunction in neural cells prevented by trehalose. Sci Rep. 2019;9:544. [DOI] [PubMed] [PMC]
Rockenstein E, Ostroff G, Dikengil F, Rus F, Mante M, Florio J, et al. Combined Active Humoral and Cellular Immunization Approaches for the Treatment of Synucleinopathies. J Neurosci. 2018;38:100014. [DOI] [PubMed] [PMC]
Jing F, Yang F, Cui F, Chen Z, Ling L, Huang X. Rapamycin alleviates inflammation and muscle weakness, while altering the Treg/Th17 balance in a rat model of myasthenia gravis. Biosci Rep. 2017;37:BSR20170767. [DOI] [PubMed] [PMC]
Gao X, Wen Y, Wang Z, Wang G, Guo J, Yu L, et al. Rapamycin alleviates the symptoms of experimental autoimmune myasthenia gravis rats by down-regulating Th17 cell/regulatory T cell ratio. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi. 2021;37:2430. Chinese. [PubMed]
Uberti VH, de Freitas BS, Molz P, Bromberg E, Schröder N. Iron Overload Impairs Autophagy: Effects of Rapamycin in Ameliorating Iron-Related Memory Deficits. Mol Neurobiol. 2020;57:104454. [DOI] [PubMed]
Zhang CQ, Yi S, Chen BB, Cui PP, Wang Y, Li YZ. mTOR/NF-κB signaling pathway protects hippocampal neurons from injury induced by intermittent hypoxia in rats. Int J Neurosci. 2021;131:9941003. [DOI] [PubMed]
Sabran-Cohen T, Bright U, Mizrachi Zer-Aviv T, Akirav I. Rapamycin prevents the long-term impairing effects of adolescence Δ-9-tetrahydrocannabinol on memory and plasticity in male rats. Eur J Neurosci. 2021;54:610422. [DOI] [PubMed]
Chen X, Gao F, Lin C, Chen A, Deng J, Chen P, et al. mTOR-mediated autophagy in the hippocampus is involved in perioperative neurocognitive disorders in diabetic rats. CNS Neurosci Ther. 2022;28:54053. [DOI] [PubMed] [PMC]
Chen G, Ke Z, Xu M, Liao M, Wang X, Qi Y, et al. Autophagy is a protective response to ethanol neurotoxicity. Autophagy. 2012;8:157789. [DOI] [PubMed] [PMC]
Yang N, Liu X, Niu X, Wang X, Jiang R, Yuan N, et al. Activation of Autophagy Ameliorates Age-Related Neurogenesis Decline and Neurodysfunction in Adult Mice. Stem Cell Rev Rep. 2022;18:62641. [DOI] [PubMed]
Ni L, Wei Y, Pan J, Li X, Xu B, Deng Y, et al. The effects of mTOR or Vps34-mediated autophagy on methylmercury-induced neuronal apoptosis in rat cerebral cortex. Food Chem Toxicol. 2021;155:112386. [DOI] [PubMed]
Caccamo A, Majumder S, Deng JJ, Bai Y, Thornton FB, Oddo S. Rapamycin rescues TDP-43 mislocalization and the associated low molecular mass neurofilament instability. J Biol Chem. 2009;284:2741624. [DOI] [PubMed] [PMC]
Chennampally P, Sayed-Zahid A, Soundararajan P, Sharp J, Cox GA, Collins SD, et al. A microfluidic approach to rescue ALS motor neuron degeneration using rapamycin. Sci Rep. 2021;11:18168. [DOI] [PubMed] [PMC]
Tsai PT, Rudolph S, Guo C, Ellegood J, Gibson JM, Schaeffer SM, et al. Sensitive Periods for Cerebellar-Mediated Autistic-like Behaviors. Cell Rep. 2018;25:35767.e4. [DOI] [PubMed] [PMC]
Kotajima-Murakami H, Kobayashi T, Kashii H, Sato A, Hagino Y, Tanaka M, et al. Effects of rapamycin on social interaction deficits and gene expression in mice exposed to valproic acid in utero. Mol Brain. 2019;12:3. [DOI] [PubMed] [PMC]
McMahon JJ, Yu W, Yang J, Feng H, Helm M, McMahon E, et al. Seizure-dependent mTOR activation in 5-HT neurons promotes autism-like behaviors in mice. Neurobiol Dis. 2015;73:296306. [DOI] [PubMed] [PMC]
Burket JA, Benson AD, Tang AH, Deutsch SI. Rapamycin improves sociability in the BTBR T+Itpr3tf/J mouse model of autism spectrum disorders. Brain Res Bull. 2014;100:705. [DOI] [PubMed] [PMC]
Xing X, Zhang J, Wu K, Cao B, Li X, Jiang F, et al. Suppression of Akt-mTOR pathway rescued the social behavior in Cntnap2-deficient mice. Sci Rep. 2019;9:3041. [DOI] [PubMed] [PMC]
Wu J, de Theije CG, da Silva SL, van der Horst H, Reinders MT, Broersen LM, et al. mTOR plays an important role in cow’s milk allergy-associated behavioral and immunological deficits. Neuropharmacology. 2015;97:22032. [DOI] [PubMed]
Xie J, Han Q, Wei Z, Wang Y, Wang S, Chen M. Phenanthrene induces autism-like behavior by promoting oxidative stress and mTOR pathway activation. Toxicology. 2021;461:152910. [DOI] [PubMed]
Iezzi D, Curti L, Ranieri G, Gerace E, Costa A, Ilari A, et al. Acute rapamycin rescues the hyperexcitable phenotype of accumbal medium spiny neurons in the valproic acid rat model of autism spectrum disorder. Pharmacol Res. 2022;183:106401. [DOI] [PubMed]
Amegandjin CA, Choudhury M, Jadhav V, Carriço JN, Quintal A, Berryer M, et al. Sensitive period for rescuing parvalbumin interneurons connectivity and social behavior deficits caused by TSC1 loss. Nat Commun. 2021;12:3653. [DOI] [PubMed] [PMC]
Del Grosso A, Angella L, Tonazzini I, Moscardini A, Giordano N, Caleo M, et al. Dysregulated autophagy as a new aspect of the molecular pathogenesis of Krabbe disease. Neurobiol Dis. 2019;129:195207. [DOI] [PubMed]
Di Domenico F, Tramutola A, Barone E, Lanzillotta C, Defever O, Arena A, et al. Restoration of aberrant mTOR signaling by intranasal rapamycin reduces oxidative damage: Focus on HNE-modified proteins in a mouse model of down syndrome. Redox Biol. 2019;23:101162. [DOI] [PubMed] [PMC]
Andrade-Talavera Y, Benito I, Casañas JJ, Rodríguez-Moreno A, Montesinos ML. Rapamycin restores BDNF-LTP and the persistence of long-term memory in a model of Down’s syndrome. Neurobiol Dis. 2015;82:51625. [DOI] [PubMed]
Troca-Marín JA, Alves-Sampaio A, Montesinos ML. An increase in basal BDNF provokes hyperactivation of the Akt-mammalian target of rapamycin pathway and deregulation of local dendritic translation in a mouse model of Down’s syndrome. J Neurosci. 2011;31:944555. [DOI] [PubMed] [PMC]
Urbano-Gámez JD, Casañas JJ, Benito I, Montesinos ML. Prenatal treatment with rapamycin restores enhanced hippocampal mGluR-LTD and mushroom spine size in a Down’s syndrome mouse model. Mol Brain. 2021;14:84. [DOI] [PubMed] [PMC]
Reijnders MRF, Kousi M, van Woerden GM, Klein M, Bralten J, Mancini GMS, et al. Variation in a range of mTOR-related genes associates with intracranial volume and intellectual disability. Nat Commun. 2017;8:1052. [DOI] [PubMed] [PMC]
Lopatynska-Mazurek M, Antolak A, Grochecki P, Gibula-Tarlowska E, Bodzon-Kulakowska A, Listos J, et al. Rapamycin Improves Spatial Learning Deficits, Vulnerability to Alcohol Addiction and Altered Expression of the GluN2B Subunit of the NMDA Receptor in Adult Rats Exposed to Ethanol during the Neonatal Period. Biomolecules. 2021;11:650. [DOI] [PubMed] [PMC]
Lopatynska-Mazurek M, Pankowska A, Gibula-Tarlowska E, Pietura R, Kotlinska JH. Rapamycin Improves Recognition Memory and Normalizes Amino-Acids and Amines Levels in the Hippocampal Dentate Gyrus in Adult Rats Exposed to Ethanol during the Neonatal Period. Biomolecules. 2021;11:362. [DOI] [PubMed] [PMC]
Lopatynska-Mazurek M, Komsta L, Gibula-Tarlowska E, Kotlinska JH. Aversive Learning Deficits and Depressive-Like Behaviors Are Accompanied by an Increase in Oxidative Stress in a Rat Model of Fetal Alcohol Spectrum Disorders: The Protective Effect of Rapamycin. Int J Mol Sci. 2021;22:7083. [DOI] [PubMed] [PMC]
Chi OZ, Wu CC, Liu X, Rah KH, Jacinto E, Weiss HR. Restoration of Normal Cerebral Oxygen Consumption with Rapamycin Treatment in a Rat Model of Autism-Tuberous Sclerosis. Neuromolecular Med. 2015;17:30513. [DOI] [PubMed] [PMC]
TTang G, Gudsnuk K, Kuo SH, Cotrina ML, Rosoklija G, Sosunov A, et al. Loss of mTOR-dependent macroautophagy causes autistic-like synaptic pruning deficits. Neuron. 2014;83:113143. [DOI] [PubMed] [PMC]
Tsai PT, Hull C, Chu Y, Greene-Colozzi E, Sadowski AR, Leech JM, et al. Autistic-like behaviour and cerebellar dysfunction in Purkinje cell Tsc1 mutant mice. Nature. 2012;488:64751. [DOI] [PubMed] [PMC]
Sato A, Kasai S, Kobayashi T, Takamatsu Y, Hino O, Ikeda K, et al. Rapamycin reverses impaired social interaction in mouse models of tuberous sclerosis complex. Nat Commun. 2012;3:1292. [DOI] [PubMed] [PMC]
Carson RP, Van Nielen DL, Winzenburger PA, Ess KC. Neuronal and glia abnormalities in Tsc1-deficient forebrain and partial rescue by rapamycin. Neurobiol Dis. 2012;45:36980. [DOI] [PubMed] [PMC]
Meikle L, Pollizzi K, Egnor A, Kramvis I, Lane H, Sahin M, et al. Response of a neuronal model of tuberous sclerosis to mammalian target of rapamycin (mTOR) inhibitors: effects on mTORC1 and Akt signaling lead to improved survival and function. J Neurosci. 2008;28:542232. [DOI] [PubMed] [PMC]
Sundberg M, Tochitsky I, Buchholz DE, Winden K, Kujala V, Kapur K, et al. Purkinje cells derived from TSC patients display hypoexcitability and synaptic deficits associated with reduced FMRP levels and reversed by rapamycin. Mol Psychiatry. 2018;23:216783. [DOI] [PubMed] [PMC]
Karalis V, Caval-Holme F, Bateup HS. Raptor downregulation rescues neuronal phenotypes in mouse models of Tuberous Sclerosis Complex. Nat Commun. 2022;13:4665. [DOI] [PubMed] [PMC]
Koike-Kumagai M, Fujimoto M, Wataya-Kaneda M. Sirolimus relieves seizures and neuropsychiatric symptoms via changes of microglial polarity in tuberous sclerosis complex model mice. Neuropharmacology. 2022;218:109203. [DOI] [PubMed]
Way SW, Rozas NS, Wu HC, McKenna J 3rd, Reith RM, Hashmi SS, et al. The differential effects of prenatal and/or postnatal rapamycin on neurodevelopmental defects and cognition in a neuroglial mouse model of tuberous sclerosis complex. Hum Mol Genet. 2012;21:322636. [DOI] [PubMed] [PMC]
Martin P, Wagh V, Reis SA, Erdin S, Beauchamp RL, Shaikh G, et al. TSC patient-derived isogenic neural progenitor cells reveal altered early neurodevelopmental phenotypes and rapamycin-induced MNK-eIF4E signaling. Mol Autism. 2020;11:2. [DOI] [PubMed] [PMC]
Ehninger D, Han S, Shilyansky C, Zhou Y, Li W, Kwiatkowski DJ, et al. Reversal of learning deficits in a Tsc2+/- mouse model of tuberous sclerosis. Nat Med. 2008;14:8438. [DOI] [PubMed] [PMC]
Cambiaghi M, Cursi M, Magri L, Castoldi V, Comi G, Minicucci F, et al. Behavioural and EEG effects of chronic rapamycin treatment in a mouse model of tuberous sclerosis complex. Neuropharmacology. 2013;67:17. [DOI] [PubMed]
Linda K, Lewerissa EI, Verboven AHA, Gabriele M, Frega M, Klein Gunnewiek TM, et al. Imbalanced autophagy causes synaptic deficits in a human model for neurodevelopmental disorders. Autophagy. 2022;18:42342. [DOI] [PubMed] [PMC]
Zhong S, Zhao Z, Xie W, Cai Y, Zhang Y, Ding J, et al. GABAergic Interneuron and Neurotransmission Are mTOR-Dependently Disturbed in Experimental Focal Cortical Dysplasia. Mol Neurobiol. 2021;58:15669. [DOI] [PubMed]
Crutcher E, Pal R, Naini F, Zhang P, Laugsch M, Kim J, et al. mTOR and autophagy pathways are dysregulated in murine and human models of Schaaf-Yang syndrome. Sci Rep. 2019;9:15935. [DOI] [PubMed] [PMC]
Gordon EB, Hart GT, Tran TM, Waisberg M, Akkaya M, Skinner J, et al. Inhibiting the Mammalian target of rapamycin blocks the development of experimental cerebral malaria. mBio. 2015;6:e00725. [DOI] [PubMed] [PMC]
Mejia P, Treviño-Villarreal JH, Reynolds JS, De Niz M, Thompson A, Marti M, et al. A single rapamycin dose protects against late-stage experimental cerebral malaria via modulation of host immunity, endothelial activation and parasite sequestration. Malar J. 2017;16:455. [DOI] [PubMed] [PMC]
Mejia P, Treviño-Villarreal JH, Hine C, Harputlugil E, Lang S, Calay E, et al. Dietary restriction protects against experimental cerebral malaria via leptin modulation and T-cell mTORC1 suppression. Nat Commun. 2015;6:6050. [DOI] [PubMed] [PMC]
Hu J, Chen X, Cheng J, Kong F, Xia H, Wu J. Mammalian target of rapamycin signaling pathway is involved in synaptic plasticity of the spinal dorsal horn and neuropathic pain in rats by regulating autophagy. Neuroreport. 2021;32:92535. [DOI] [PubMed]
Liu F, Wu M, Kai J, Dong J, Zhang B, Liu L, et al. Effectiveness of low dose of rapamycin in preventing seizure-induced anxiety-like behaviour, cognitive impairment, and defects in neurogenesis in developing rats. Int J Neurosci. 2020;130:918. [DOI] [PubMed]
Blundell J, Kouser M, Powell CM. Systemic inhibition of mammalian target of rapamycin inhibits fear memory reconsolidation. Neurobiol Learn Mem. 2008;90:2835. [DOI] [PubMed] [PMC]
Fifield K, Hebert M, Angel R, Adamec R, Blundell J. Inhibition of mTOR kinase via rapamycin blocks persistent predator stress-induced hyperarousal. Behav Brain Res. 2013;256:45763. [DOI] [PubMed]
Levin N, Kritman M, Maroun M, Akirav I. Differential roles of the infralimbic and prelimbic areas of the prefrontal cortex in reconsolidation of a traumatic memory. Eur Neuropsychopharmacol. 2017;27:90012. [DOI] [PubMed]
Li Y, Cheng Y, Zhou Y, Du H, Zhang C, Zhao Z, et al. High fat diet-induced obesity leads to depressive and anxiety-like behaviors in mice via AMPK/mTOR-mediated autophagy. Exp Neurol. 2022;348:113949. [DOI] [PubMed]
Wang S, Zhou SL, Min FY, Ma JJ, Shi XJ, Bereczki E, et al. mTOR-mediated hyperphosphorylation of tau in the hippocampus is involved in cognitive deficits in streptozotocin-induced diabetic mice. Metab Brain Dis. 2014;29:72936. [DOI] [PubMed]
Sun Q, Wei LL, Zhang M, Li TX, Yang C, Deng SP, et al. Rapamycin inhibits activation of AMPK-mTOR signaling pathway-induced Alzheimer’s disease lesion in hippocampus of rats with type 2 diabetes mellitus. Int J Neurosci. 2019;129:17988. [DOI] [PubMed]
Xu T, Liu J, Li XR, Yu Y, Luo X, Zheng X, et al. The mTOR/NF-κB Pathway Mediates Neuroinflammation and Synaptic Plasticity in Diabetic Encephalopathy. Mol Neurobiol. 2021;58:384862. [DOI] [PubMed]
Gao Y, Peng S, Wen Q, Zheng C, Lin J, Tan Y, et al. The mammalian target of rapamycin pathway in the basolateral amygdala is critical for nicotine-induced behavioural sensitization. Int J Neuropsychopharmacol. 2014;17:188194. [DOI] [PubMed]
Zhou Y, Liang Y, Kreek MJ. mTORC1 pathway is involved in the kappa opioid receptor activation-induced increase in excessive alcohol drinking in mice. Pharmacol Biochem Behav. 2020;195:172954. [DOI] [PubMed] [PMC]
Barak S, Liu F, Ben Hamida S, Yowell QV, Neasta J, Kharazia V, et al. Disruption of alcohol-related memories by mTORC1 inhibition prevents relapse. Nat Neurosci. 2013;16:11117. [DOI] [PubMed] [PMC]
Canivet C, Menasria R, Rhéaume C, Piret J, Boivin G. Valacyclovir combined with artesunate or rapamycin improves the outcome of herpes simplex virus encephalitis in mice compared to antiviral therapy alone. Antiviral Res. 2015;123:10513. [DOI] [PubMed]
Kara NZ, Flaisher-Grinberg S, Anderson GW, Agam G, Einat H. Mood-stabilizing effects of rapamycin and its analog temsirolimus: relevance to autophagy. Behav Pharmacol. 2018;29:37984. [DOI] [PubMed]
Akman O, Briggs SW, Mowrey WB, Moshé SL, Galanopoulou AS. Antiepileptogenic effects of rapamycin in a model of infantile spasms due to structural lesions. Epilepsia. 2021;62:198599. [DOI] [PubMed] [PMC]
Ishida S, Zhao D, Sawada Y, Hiraoka Y, Mashimo T, Tanaka K. Dorsal telencephalon-specific Nprl2- and Nprl3-knockout mice: novel mouse models for GATORopathy. Hum Mol Genet. 2022;31:151930. [DOI] [PubMed] [PMC]
Xie P, Zhu S, Zhou H, Fang R, Zhuang J, Wen J, et al. Rapamycin Plays an Anti-Epileptic Role by Restoring Blood-Brain Barrier Dysfunction, Balancing T Cell Subsets and Inhibiting Neuronal Apoptosis. Discov Med. 2023;35:104351. [PubMed]
Liu L, Liu Z, Zeng C, Xu Y, He L, Fang Q, et al. Dynorphin/KOR inhibits neuronal autophagy by activating mTOR signaling pathway to prevent acute seizure epilepsy. Cell Biol Int. 2022;46:181424. [DOI] [PubMed]
Aghaie F, Moradifar F, Hosseini A. Rapamycin attenuates depression and anxiety-like behaviors through modulation of the NLRP3 pathway in pentylenetetrazole-kindled male Wistar rats. Fundam Clin Pharmacol. 2021;35:104554. [DOI] [PubMed]
Ding N, Zhao K, Lan Y, Li Z, Lv X, Su J, et al. Induction of Atypical Autophagy by Porcine Hemagglutinating Encephalomyelitis Virus Contributes to Viral Replication. Front Cell Infect Microbiol. 2017;7:56. [DOI] [PubMed] [PMC]
Li GY, Fan B, Jiao YY. Rapamycin attenuates visible light-induced injury in retinal photoreceptor cells via inhibiting endoplasmic reticulum stress. Brain Res. 2014;1563:112. [DOI] [PubMed]
Nalbandian A, Llewellyn KJ, Nguyen C, Yazdi PG, Kimonis VE. Rapamycin and chloroquine: the in vitro and in vivo effects of autophagy-modifying drugs show promising results in valosin containing protein multisystem proteinopathy. PLoS One. 2015;10:e0122888. [DOI] [PubMed] [PMC]
Hayashi I, Aoki Y, Ushikubo H, Asano D, Mori A, Sakamoto K, et al. Protective effects of PF-4708671 against N-methyl-d-aspartic acid-induced retinal damage in rats. Fundam Clin Pharmacol. 2016;30:52936. [DOI] [PubMed]
Ichikawa A, Nakahara T, Kurauchi Y, Mori A, Sakamoto K, Ishii K. Rapamycin prevents N-methyl-D-aspartate-induced retinal damage through an ERK-dependent mechanism in rats. J Neurosci Res. 2014;92:692702. [DOI] [PubMed]
Aoki Y, Nakahara T, Asano D, Ushikubo H, Mori A, Sakamoto K, et al. Preventive effects of rapamycin on inflammation and capillary degeneration in a rat model of NMDA-induced retinal injury. Biol Pharm Bull. 2015;38:3214. [DOI] [PubMed]
Ying H, Turturro S, Nguyen T, Shen X, Zelkha R, Johnson EC, et al. Induction of autophagy in rats upon overexpression of wild-type and mutant optineurin gene. BMC Cell Biol. 2015;16:14. [DOI] [PubMed] [PMC]
Foerster P, Daclin M, Asm S, Faucourt M, Boletta A, Genovesio A, et al. mTORC1 signaling and primary cilia are required for brain ventricle morphogenesis. Development. 2017;144:20110. [DOI] [PubMed] [PMC]
Metaxakis A, Tain LS, Grönke S, Hendrich O, Hinze Y, Birras U, et al. Lowered insulin signalling ameliorates age-related sleep fragmentation in Drosophila. PLoS Biol. 2014;12:e1001824. [DOI] [PubMed] [PMC]
Cleary C, Linde JA, Hiscock KM, Hadas I, Belmaker RH, Agam G, et al. Antidepressive-like effects of rapamycin in animal models: Implications for mTOR inhibition as a new target for treatment of affective disorders. Brain Res Bull. 2008;76:46973. [DOI] [PubMed]
James MH, Quinn RK, Ong LK, Levi EM, Charnley JL, Smith DW, et al. mTORC1 inhibition in the nucleus accumbens ‘protects’ against the expression of drug seeking and ‘relapse’ and is associated with reductions in GluA1 AMPAR and CAMKIIα levels. Neuropsychopharmacology. 2014;39:1694702. [DOI] [PubMed] [PMC]
James MH, Quinn RK, Ong LK, Levi EM, Smith DW, Dickson PW, et al. Rapamycin reduces motivated responding for cocaine and alters GluA1 expression in the ventral but not dorsal striatum. Eur J Pharmacol. 2016;784:14754. [DOI] [PubMed]
Bailey J, Ma D, Szumlinski KK. Rapamycin attenuates the expression of cocaine-induced place preference and behavioral sensitization. Addict Biol. 2012;17:24858. [DOI] [PubMed] [PMC]
Towner RA, Gulej R, Zalles M, Saunders D, Smith N, Lerner M, et al. Rapamycin restores brain vasculature, metabolism, and blood-brain barrier in an inflammaging model. Geroscience. 2021;43:56378. [DOI] [PubMed] [PMC]
Felici R, Buonvicino D, Muzzi M, Cavone L, Guasti D, Lapucci A, et al. Post onset, oral rapamycin treatment delays development of mitochondrial encephalopathy only at supramaximal doses. Neuropharmacology. 2017;117:7484. [DOI] [PubMed]
Kim SG, Lee S, Kim Y, Park J, Woo D, Kim D, et al. Tanc2-mediated mTOR inhibition balances mTORC1/2 signaling in the developing mouse brain and human neurons. Nat Commun. 2021;12:2695. [DOI] [PubMed] [PMC]
Wen J, Xu J, Mathena RP, Choi JH, Mintz CD. Early Isoflurane Exposure Impairs Synaptic Development in Fmr1 KO Mice via the mTOR Pathway. Neurochem Res. 2021;46:157788. [DOI] [PubMed]
Tian J, Wang Z, Ren Y, Jiang Y, Zhao Y, Li M, et al. Rapamycin Attenuates Anxiety and Depressive Behavior Induced by Helicobacter pylori in Association with Reduced Circulating Levels of Ghrelin. Neural Plast. 2022;2022:2847672. [DOI] [PubMed] [PMC]
Zhao Y, Zhao W, Han Y. Inhibition of mTORC2 improves brain injury in epileptic rats by promoting chaperone-mediated autophagy. Epilepsy Res. 2023;193:107161. [DOI] [PubMed]
Liu H, Li F, Li X, Wu Q, Dai C. Rapamycin ameliorates age-related hearing loss in C57BL/6J mice by enhancing autophagy in the SGNs. Neurosci Lett. 2022;772:136493. [DOI] [PubMed]
Pucha KA, Ma TC, York W, Kang UJ, Kaufmann H, Kapogiannis D, et al. Neuron-derived extracellular vesicles to examine brain mTOR target engagement with sirolimus in patients with multiple system atrophy. Parkinsonism Relat Disord. 2023;115:105821. [DOI] [PubMed] [PMC]
Dusing M, LaSarge CL, White A, Jerow LG, Gross C, Danzer SC. Neurovascular Development in Pten and Tsc2 Mouse Mutants. eNeuro. 2023;10:ENEURO.034022.2023. [DOI] [PubMed] [PMC]
Liu W, Guo J, Mu J, Tian L, Zhou D. Rapamycin Protects Sepsis-Induced Cognitive Impairment in Mouse Hippocampus by Enhancing Autophagy. Cell Mol Neurobiol. 2017;37:1195205. [DOI] [PubMed] [PMC]
Parker WE, Orlova KA, Parker WH, Birnbaum JF, Krymskaya VP, Goncharov DA, et al. Rapamycin prevents seizures after depletion of STRADA in a rare neurodevelopmental disorder. Sci Transl Med. 2013;5:182ra53. [DOI] [PubMed] [PMC]
Kato M, Kada A, Shiraishi H, Tohyama J, Nakagawa E, Takahashi Y, et al. Sirolimus for epileptic seizures associated with focal cortical dysplasia type II. Ann Clin Transl Neurol. 2022;9:18192. [DOI] [PubMed] [PMC]
Zhang J, Xu X, Zhou D, Li H, You W, Wang Z, et al. 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;52:152739. [DOI] [PubMed]
Hsieh CH, Lin YJ, Chen WL, Huang YC, Chang CW, Cheng FC, et al. HIF-1α triggers long-lasting glutamate excitotoxicity via system xc- in cerebral ischaemia-reperfusion. J Pathol. 2017;241:33749. [DOI] [PubMed]
Wang T, Li B, Wang Z, Wang X, Xia Z, Ning G, et al. Sorafenib promotes sensory conduction function recovery via miR-142-3p/AC9/cAMP axis post dorsal column injury. Neuropharmacology. 2019;148:34757. [DOI] [PubMed]
Echeverria V, Burgess S, Gamble-George J, Zeitlin R, Lin X, Cao C, et al. Sorafenib inhibits nuclear factor kappa B, decreases inducible nitric oxide synthase and cyclooxygenase-2 expression, and restores working memory in APPswe mice. Neuroscience. 2009;162:122031. [DOI] [PubMed]
Kim J, Park JH, Park SK, Hoe HS. Sorafenib Modulates the LPS- and Aβ-Induced Neuroinflammatory Response in Cells, Wild-Type Mice, and 5xFAD Mice. Front Immunol. 2021;12:684344. [DOI] [PubMed] [PMC]
Liu Z, Hamamichi S, Lee BD, Yang D, Ray A, Caldwell GA, et al. Inhibitors of LRRK2 kinase attenuate neurodegeneration and Parkinson-like phenotypes in Caenorhabditis elegans and Drosophila Parkinson’s disease models. Hum Mol Genet. 2011;20:393342. [DOI] [PubMed] [PMC]
Moawad EY. Induction of multiple sclerosis and response to tyrosine kinase inhibitors. Indian J Clin Biochem. 2014;29:4915. [DOI] [PubMed] [PMC]
Crespo O, Kang SC, Daneman R, Lindstrom TM, Ho PP, Sobel RA, et al. Tyrosine kinase inhibitors ameliorate autoimmune encephalomyelitis in a mouse model of multiple sclerosis. J Clin Immunol. 2011;31:101020. [DOI] [PubMed] [PMC]
Martina BEE, Smreczak M, Orlowska A, Marzec A, Trebas P, Roose JM, et al. Combination drug treatment prolongs survival of experimentally infected mice with silver-haired bat rabies virus. Vaccine. 2019;37:473642. [DOI] [PubMed]
Brahms A, Mudhasani R, Pinkham C, Kota K, Nasar F, Zamani R, et al. Sorafenib Impedes Rift Valley Fever Virus Egress by Inhibiting Valosin-Containing Protein Function in the Cellular Secretory Pathway. J Virol. 2017;91:e0096817. [DOI] [PubMed] [PMC]
Lundberg L, Brahms A, Hooper I, Carey B, Lin SC, Dahal B, et al. Repurposed FDA-Approved drug sorafenib reduces replication of Venezuelan equine encephalitis virus and other alphaviruses. Antiviral Res. 2018;157:5767. [DOI] [PubMed]
Gao M, Duan H, Liu J, Zhang H, Wang X, Zhu M, et al. The multi-targeted kinase inhibitor sorafenib inhibits enterovirus 71 replication by regulating IRES-dependent translation of viral proteins. Antiviral Res. 2014;106:805. [DOI] [PubMed]
Welsbie DS, Ziogas NK, Xu L, Kim BJ, Ge Y, Patel AK, 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. [DOI] [PubMed] [PMC]
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;49:316. [DOI] [PubMed] [PMC]
Lee JC, Kim HY, Lee S, Shin J, Kim HV, Kim K, 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;59:11491500. [DOI] [PubMed]
Grammas P, Martinez J, Sanchez A, Yin X, Riley J, Gay D, et al. A new paradigm for the treatment of Alzheimer’s disease: targeting vascular activation. J Alzheimers Dis. 2014;40:61930. [DOI] [PubMed]
Son SM, Jung ES, Shin HJ, Byun J, Mook-Jung I. Aβ-induced formation of autophagosomes is mediated by RAGE-CaMKKβ-AMPK signaling. Neurobiol Aging. 2012;33:1006.e1123. [DOI] [PubMed]
Zhao P, Cheng P, Wang J, Zhu G, Wang X. Shenqi Yizhi prescription prevents AβO-induced memory impairment in mice by regulating the contractility of brain pericytes. Phytomedicine. 2024;129:155639. [DOI] [PubMed]
Wrasidlo W, Crews LA, Tsigelny IF, Stocking E, Kouznetsova VL, Price D, 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;171:575773. [DOI] [PubMed] [PMC]
Fields JA, Metcalf J, Overk C, Adame A, Spencer B, Wrasidlo W, et al. The anticancer drug sunitinib promotes autophagyand protects from neurotoxicity in an HIV-1 Tat model of neurodegeneration. J Neurovirol. 2017;23:290303. [DOI] [PubMed] [PMC]
Pu SY, Xiao F, Schor S, Bekerman E, Zanini F, Barouch-Bentov R, et al. Feasibility and biological rationale of repurposing sunitinib and erlotinib for dengue treatment. Antiviral Res. 2018;155:6775. [DOI] [PubMed] [PMC]
Luo J, Zhang Y, Wang Y, Liu Q, Chen L, Zhang B, et al. Rhabdovirus Infection Is Dependent on Serine/Threonine Kinase AP2-Associated Kinase 1. Life (Basel). 2020;10:170. [DOI] [PubMed] [PMC]
Hsueh HT, Chou RT, Rai U, Kolodziejski P, Liyanage W, Pejavar J, et al. Engineered peptide-drug conjugate provides sustained protection of retinal ganglion cells with topical administration in rats. J Control Release. 2023;362:37180. [DOI] [PubMed] [PMC]
Siracusa R, Paterniti I, Cordaro M, Crupi R, Bruschetta G, Campolo M, et al. Neuroprotective Effects of Temsirolimus in Animal Models of Parkinson’s Disease. Mol Neurobiol. 2018;55:240319. [DOI] [PubMed]
Decressac M, Björklund A. mTOR inhibition alleviates L-DOPA-induced dyskinesia in parkinsonian rats. J Parkinsons Dis. 2013;3:137. [DOI] [PubMed]
Jiang T, Yu JT, Zhu XC, Zhang QQ, Cao L, Wang HF, et al. Temsirolimus attenuates tauopathy in vitro and in vivo by targeting tau hyperphosphorylation and autophagic clearance. Neuropharmacology. 2014;85:12130. [DOI] [PubMed]
Frederick C, Ando K, Leroy K, Héraud C, Suain V, Buée L, et al. Rapamycin ester analog CCI-779/Temsirolimus alleviates tau pathology and improves motor deficit in mutant tau transgenic mice. J Alzheimers Dis. 2015;44:114556. [DOI] [PubMed]
Jiang T, Yu JT, Zhu XC, Tan MS, Wang HF, Cao L, 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;81:5463. [DOI] [PubMed]
Menzies FM, Huebener J, Renna M, Bonin M, Riess O, Rubinsztein DC. Autophagy induction reduces mutant ataxin-3 levels and toxicity in a mouse model of spinocerebellar ataxia type 3. Brain. 2010;133:93104. [DOI] [PubMed] [PMC]
Saravia R, Flores Á, Plaza-Zabala A, Busquets-Garcia A, Pastor A, de la Torre R, et al. CB1 Cannabinoid Receptors Mediate Cognitive Deficits and Structural Plasticity Changes During Nicotine Withdrawal. Biol Psychiatry. 2017;81:62534. [DOI] [PubMed]
Launay N, Aguado C, Fourcade S, Ruiz M, Grau L, Riera J, et al. Autophagy induction halts axonal degeneration in a mouse model of X-adrenoleukodystrophy. Acta Neuropathol. 2015;129:399415. [DOI] [PubMed] [PMC]
Konoeda F, Shichita T, Yoshida H, Sugiyama Y, Muto G, Hasegawa E, et al. Therapeutic effect of IL-12/23 and their signaling pathway blockade on brain ischemia model. Biochem Biophys Res Commun. 2010;402:5006. [DOI] [PubMed]
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 (Sao Paulo). 2021;76:e2348. [DOI] [PubMed] [PMC]
Zhou Y, Leng X, Luo S, Su Z, Luo X, Guo H, et al. Tolerogenic Dendritic Cells Generated with Tofacitinib Ameliorate Experimental Autoimmune Encephalomyelitis through Modulation of Th17/Treg Balance. J Immunol Res. 2016;2016:5021537. [DOI] [PubMed] [PMC]
Günaydın C, Önger ME, Avcı B, Bozkurt A, Terzi M, Bilge SS. Tofacitinib enhances remyelination and improves myelin integrity in cuprizone-induced mice. Immunopharmacol Immunotoxicol. 2021;43:7908. [DOI] [PubMed]
Alshammari A, Alharbi M, Albekairi NA, Albekairi TH, Alharbi OO, Yeapuri P, et al. Protective Effect of CP690550 in MPTP-Induced Parkinson’s Like Behavioural, Biochemical and Histological Alterations in Mice. Neurotox Res. 2022;40:56472. [DOI] [PubMed]
Figueroa-Romero C, Monteagudo A, Murdock BJ, Famie JP, Webber-Davis IF, Piecuch CE, et al. Tofacitinib Suppresses Natural Killer Cells In Vitro and In Vivo: Implications for Amyotrophic Lateral Sclerosis. Front Immunol. 2022;13:773288. [DOI] [PubMed] [PMC]
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. [DOI] [PubMed] [PMC]
Ma H, Wang C, Han L, Kong F, Liu Z, Zhang B, et al. Tofacitinib Promotes Functional Recovery after Spinal Cord Injury by Regulating Microglial Polarization via JAK/STAT Signaling Pathway. Int J Biol Sci. 2023;19:486582. [DOI] [PubMed] [PMC]
Zhou Y, Chen L, Zheng X, Fang Q, Qian Y, Xu T, et al. Microglia orchestrate synaptic and neuronal stripping: Implication in neuropsychiatric lupus. J Cell Mol Med. 2024;28:e18190. [DOI] [PubMed] [PMC]
Huang Y, Li Q, Tian H, Yao X, Bakina O, Zhang H, et al. MEK inhibitor trametinib attenuates neuroinflammation and cognitive deficits following traumatic brain injury in mice. Am J Transl Res. 2020;12:635165. [PubMed] [PMC]
Christensen ST, Haanes KA, Spray S, Grell AS, Warfvinge K, Edvinsson L, et al. Pre-clinical effects of highly potent MEK1/2 inhibitors on rat cerebral vasculature after organ culture and subarachnoid haemorrhage. Clin Sci (Lond). 2019;133:1797811. [DOI] [PubMed]
Park ES, Kim S, Huang S, Yoo JY, Körbelin J, Lee TJ, et al. Selective Endothelial Hyperactivation of Oncogenic KRAS Induces Brain Arteriovenous Malformations in Mice. Ann Neurol. 2021;89:92641. [DOI] [PubMed]
Caldi Gomes L, Hänzelmann S, Hausmann F, Khatri R, Oller S, Parvaz M, et al. Multiomic ALS signatures highlight subclusters and sex differences suggesting the MAPK pathway as therapeutic target. Nat Commun. 2024;15:4893. [DOI] [PubMed] [PMC]
Yue W, Deng X, Wang Z, Jiang M, Hu R, Duan Y, et al. Inhibition of the MEK/ERK pathway suppresses immune overactivation and mitigates TDP-43 toxicity in a Drosophila model of ALS. Immun Ageing. 2023;20:27. [DOI] [PubMed] [PMC]
Kim MY, Kim MJ, Lee C, Lee J, Kim SS, Hong S, et al. Trametinib activates endogenous neurogenesis and recovers neuropathology in a model of Alzheimer’s disease. Exp Mol Med. 2023;55:217789. [DOI] [PubMed] [PMC]
Chun YS, Kim MY, Lee SY, Kim MJ, Hong TJ, Jeon JK, et al. MEK1/2 inhibition rescues neurodegeneration by TFEB-mediated activation of autophagic lysosomal function in a model of Alzheimer’s Disease. Mol Psychiatry. 2022;27:477080. [DOI] [PubMed] [PMC]
Yang Y, Suo N, Cui SH, Wu X, Ren XY, Liu Y, et al. Trametinib, an anti-tumor drug, promotes oligodendrocytes generation and myelin formation. Acta Pharmacol Sin. 2024;45:252739. [DOI] [PubMed]
Ballabh P, Xu H, Hu F, Braun A, Smith K, Rivera A, et al. Angiogenic inhibition reduces germinal matrix hemorrhage. Nat Med. 2007;13:47785. [DOI] [PubMed]
Oliveira-Santos A, Dagda M, Wittmann J, Smalley R, Burkin DJ. Vemurafenib improves muscle histopathology in a mouse model of LAMA2-related congenital muscular dystrophy. Dis Model Mech. 2023;16:dmm049916. [DOI] [PubMed] [PMC]
Wager TT, Hou X, Verhoest PR, Villalobos A. Central Nervous System Multiparameter Optimization Desirability: Application in Drug Discovery. ACS Chem Neurosci. 2016;7:76775. [DOI] [PubMed]
Wager TT, Hou X, Verhoest PR, Villalobos A. Moving beyond rules: the development of a central nervous system multiparameter optimization (CNS MPO) approach to enable alignment of druglike properties. ACS Chem Neurosci. 2010;1:43549. [DOI] [PubMed] [PMC]
Tsang JE, Urner LM, Kim G, Chow K, Baufeld L, Faull K, et al. Development of a Potent Brain-Penetrant EGFR Tyrosine Kinase Inhibitor against Malignant Brain Tumors. ACS Med Chem Lett. 2020;11:1799809. [DOI] [PubMed] [PMC]
van Hoppe S, Rood JJM, Buil L, Wagenaar E, Sparidans RW, Beijnen JH, et al. P-Glycoprotein (MDR1/ABCB1) Restricts Brain Penetration of the Bruton’s Tyrosine Kinase Inhibitor Ibrutinib, While Cytochrome P450-3A (CYP3A) Limits Its Oral Bioavailability. Mol Pharm. 2018;15:512434. [DOI] [PubMed]
Reich DS, Arnold DL, Vermersch P, Bar-Or A, Fox RJ, Matta A, et al.; Tolebrutinib Phase 2b Study Group. Safety and efficacy of tolebrutinib, an oral brain-penetrant BTK inhibitor, in relapsing multiple sclerosis: a phase 2b, randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2021;20:72938. [DOI] [PubMed] [PMC]
Ren L, Moreno D, Baer BR, Barbour P, Bettendorf T, Bouhana K, Brown K, et al. Identification of the Clinical Candidate PF-07284890 (ARRY-461), a Highly Potent and Brain Penetrant BRAF Inhibitor for the Treatment of Cancer. J Med Chem. 2024;67:1301932. [DOI] [PubMed]
Pike KG, Hunt TA, Barlaam B, Benstead D, Cadogan E, Chen K, et al. Identification of Novel, Selective Ataxia-Telangiectasia Mutated Kinase Inhibitors with the Ability to Penetrate the Blood-Brain Barrier: The Discovery of AZD1390. J Med Chem. 2024;67:3090111. [DOI] [PubMed]
Cite this Article
Export Citation
Aliashrafzadeh H, Liu D, De Alba S, Akbar I, Lui A, Vanleuven J, et al. Experimental and clinical tests of FDA-approved kinase inhibitors for the treatment of neurological disorders (update 2024). Explor Drug Sci. 2025;3:1008116. https://doi.org/10.37349/eds.2025.1008116
Article Metrics

View: 43

Download: 10

Times Cited: 0