1
|
Krishna N, Ramalakshmi NA, Krishnamurthy RG. Comprehensive Bioinformatics Analysis Reveals Molecular Signatures and Potential Caloric Restriction Mimetics with Neuroprotective Effects: Validation in an In Vitro Stroke Model. J Mol Neurosci 2025; 75:32. [PMID: 40080242 DOI: 10.1007/s12031-025-02328-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025]
Abstract
Caloric restriction (CR) is a dietary intervention that reduces calorie intake without inducing malnutrition, demonstrating lifespan-extending effects in preclinical studies and some human trials, along with potential benefits in ameliorating age-related ailments. Caloric restriction mimetics (CRMs) are compounds mimicking CR effects, offering a potential therapeutic avenue for age-related diseases. This study explores the potential protective effects of CR on the brain neocortex (GSE11291) and the identification of CRMs using integrative bioinformatics and systems biology approaches. Our findings indicate that long-term CR activates cellular pathways improving mitochondrial function, enhancing antioxidant capacity, and reducing inflammation, potentially providing neuroprotection. The key signaling pathways enriched in our study include PPAR, mTOR, FoxO, AMPK, and Notch signaling pathways, which are crucial regulators of metabolism, cellular stress response, neuroprotection, and longevity. We identify key signaling molecules and molecular mechanisms associated with CR, including transcription factors, kinase regulators, and microRNAs linked to differentially expressed genes. Furthermore, potential CRMs such as rapamycin, replicating CR-related health benefits, are identified. Additionally, machine learning models were developed to classify small molecules based on their CNS activity and anti-inflammatory properties. As a proof of concept, we have demonstrated the ischemic neuroprotective effects of two top-ranked candidate reference molecules (CRMs) using the oxygen-glucose deprivation (OGD) model, an established in vitro stroke model. However, further investigations are essential to fully elucidate the therapeutic potential of these CRMs. In summary, our study suggests that long-term CR entails protective mechanisms preserving and safeguarding neuronal function, potentially impacting the treatment of age-related neurological diseases. Moreover, our findings contribute to the identification of potential genes and regulatory molecules involved in CR, along with potential CRMs, providing a promising foundation for future research in the field of neurological disorder treatment.
Collapse
Affiliation(s)
- Navami Krishna
- Department of Bioscience and Engineering, National Institute of Technology Calicut, Calicut, Kerala, India, 673601
| | | | | |
Collapse
|
2
|
Ansari MM, Sahu SK, Singh TG, Singh SRJ, Kaur P. Evolving significance of kinase inhibitors in the management of Alzheimer's disease. Eur J Pharmacol 2024; 979:176816. [PMID: 39038637 DOI: 10.1016/j.ejphar.2024.176816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 06/20/2024] [Accepted: 07/17/2024] [Indexed: 07/24/2024]
Abstract
Alzheimer's disease is a neurodegenerative problem with progressive loss of memory and other cognitive function disorders resulting in the imbalance of neurotransmitter activity and signaling progression, which poses the need of the potential therapeutic target to improve the intracellular signaling cascade brought by kinases. Protein kinase plays a significant and multifaceted role in the treatment of Alzheimer's disease, by targeting pathological mechanisms like tau hyperphosphorylation, neuroinflammation, amyloid-beta production and synaptic dysfunction. In this review, we thoroughly explore the essential protein kinases involved in Alzheimer's disease, detailing their physiological roles, regulatory impacts, and the newest inhibitors and compounds that are progressing into clinical trials. All the findings of studies exhibited the promising role of kinase inhibitors in the management of Alzheimer's disease. However, it still poses the need of addressing current challenges and opportunities involved with this disorder for the future perspective of kinase inhibitors in the management of Alzheimer's disease. Further study includes the development of biomarkers, combination therapy, and next-generation kinase inhibitors with increased potency and selectivity for its future prospects.
Collapse
Affiliation(s)
- Md Mustafiz Ansari
- School of Pharmaceutical Sciences, Lovely Professional University, Punjab, India
| | - Sanjeev Kumar Sahu
- School of Pharmaceutical Sciences, Lovely Professional University, Punjab, India
| | | | - Sovia R J Singh
- University Language Centre- Chitkara Business School, Chitkara University, Punjab, India
| | - Paranjeet Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| |
Collapse
|
3
|
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. [PMID: 38931899 PMCID: PMC11207969 DOI: 10.3390/pharmaceutics16060778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/23/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
This study evaluated the probable relevance of a non-covalent conjugate of imatinib with TP10 in the context of a neuroprotective effect in Parkinson's disease. Through the inhibition of c-Abl, which is a non-receptor tyrosine kinase and an indicator of oxidative stress, imatinib has shown promise in preclinical animal models of this disease. The poor distribution of imatinib within the brain tissue triggered experiments in which a conjugate was obtained by mixing the drug with TP10, which is known for exhibiting high translocation activity across the cell membrane. The conjugate was tested on the HT-22 cell line with respect to its impact on MPP+-induced oxidative stress, apoptosis, necrosis, cytotoxicity, and mortality. Additionally, it was checked whether the conjugate activated the ABCB1 protein. The experiments indicated that imatinib+PEG4+TP10 reduced the post-MPP+ oxidative stress, apoptosis, and mortality, and these effects were more prominent than those obtained after the exposition of the HT-22 cells to imatinib alone. Its cytotoxicity was similar to that of imatinib itself. In contrast to imatinib, the conjugate did not activate the ABCB1 protein. These favorable qualities of imatinib+PEG4+TP10 make it a potential candidate for further in vivo research, which would confirm its neuroprotective action in PD-affected brains.
Collapse
Affiliation(s)
- Izabela Rusiecka
- Department of Pharmacology, Medical University of Gdańsk, Dębowa 23, 80-204 Gdańsk, Poland
| | | | | |
Collapse
|
4
|
Kohal R, Bhavana, Kumari P, Sharma AK, Gupta GD, Verma SK. Fyn, Blk, and Lyn kinase inhibitors: A mini-review on medicinal attributes, research progress, and future insights. Bioorg Med Chem Lett 2024; 102:129674. [PMID: 38408513 DOI: 10.1016/j.bmcl.2024.129674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 02/16/2024] [Accepted: 02/22/2024] [Indexed: 02/28/2024]
Abstract
Fyn, Blk, and Lyn are part of a group of proteins called Src family kinases. They are crucial in controlling cell communication and their response to the growth, changes, and immune system. Blocking these proteins with inhibitors can be a way to treat diseases where these proteins are too active. The primary mode of action of these inhibitors is to inhibit the phosphorylation of Fyn, Blk, and Lyn receptors, which in turn affects how signals pass within the cells. This review shows the structural and functional aspects of Fyn, Blk, and Lyn kinases, highlighting the significance of their dysregulation in diseases such as cancer and autoimmune disorders. The discussion encompasses the design strategies, SAR analysis, and chemical characteristics of effective inhibitors, shedding light on their specificity and potency. Furthermore, it explores the progress of clinical trials of these inhibitors, emphasizing their potential therapeutic applications.
Collapse
Affiliation(s)
- Rupali Kohal
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga 142 001, Punjab, India
| | - Bhavana
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga 142 001, Punjab, India
| | - Preety Kumari
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga 142 001, Punjab, India
| | - Arun Kumar Sharma
- Department of Pharmacology, ISF College of Pharmacy, Moga 142 001, Punjab, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, Moga 142 001, Punjab, India
| | - Sant Kumar Verma
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga 142 001, Punjab, India.
| |
Collapse
|
5
|
Du Y, Qiu R, Chen L, Chen Y, Zhong Z, Li P, Fan F, Cheng Y. Identification of serum exosomal metabolomic and proteomic profiles for remote ischemic preconditioning. J Transl Med 2023; 21:241. [PMID: 37009888 PMCID: PMC10069038 DOI: 10.1186/s12967-023-04070-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/18/2023] [Indexed: 04/04/2023] Open
Abstract
BACKGROUND Remote ischemic preconditioning (RIPC) refers to a brief episode of exposure to potential adverse stimulation and prevents injury during subsequent exposure. RIPC has been shown to increase tolerance to ischemic injury and improve cerebral perfusion status. Exosomes have a variety of activities, such as remodeling the extracellular matrix and transmitting signals to other cells. This study aimed to investigate the potential molecular mechanism of RIPC-mediated neuroprotection. METHODS Sixty adult male military personnel participants were divided into the control group (n = 30) and the RIPC group (n = 30). We analyzed the differential metabolites and proteins in the serum exosomes of RIPC participants and control subjects. RESULTS Eighty-seven differentially expressed serum exosomal metabolites were found between the RIPC and control groups, which were enriched in pathways related to tyrosine metabolism, sphingolipid metabolism, serotonergic synapses, and multiple neurodegeneration diseases. In addition, there were 75 differentially expressed exosomal proteins between RIPC participants and controls, which involved the regulation of insulin-like growth factor (IGF) transport, neutrophil degranulation, vesicle-mediated transport, etc. Furthermore, we found differentially expressed theobromine, cyclo gly-pro, hemopexin (HPX), and apolipoprotein A1 (ApoA1), which are associated with neuroprotective benefits in ischemia/reperfusion injury. In addition, five potential metabolite biomarkers, including ethyl salicylate, ethionamide, piperic acid, 2, 6-di-tert-butyl-4-hydroxymethylphenol and zerumbone, that separated RIPC from control individuals were identified. CONCLUSION Our data suggest that serum exosomal metabolites are promising biomarkers for RIPC, and our results provide a rich dataset and framework for future analyses of cerebral ischemia‒reperfusion injury under ischemia/reperfusion conditions.
Collapse
Affiliation(s)
- Yang Du
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China
| | - Rui Qiu
- Institute of National Security, Minzu University of China, Beijing, China
| | - Lei Chen
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China
| | - Yuewen Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Key Laboratory of Translational Research for Brain Diseases, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhifeng Zhong
- Department of High Altitude Operational Medicine, College of High Altitude Military Medicine, Army Medical University, (Third Military Medical University), Chongqing, China
| | - Peng Li
- Department of High Altitude Operational Medicine, College of High Altitude Military Medicine, Army Medical University, (Third Military Medical University), Chongqing, China
| | - Fangcheng Fan
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China.
| | - Yong Cheng
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, China.
- Institute of National Security, Minzu University of China, Beijing, China.
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China.
| |
Collapse
|
6
|
Yan Z, Gu F, Wang Z, Meng J, Tao X, Dai Q, Wang W, Liu M, Wang Z. Safety and efficacy of tyrosine kinase inhibitors for the treatment of multiple sclerosis: A systematic review and meta-analysis from randomized controlled trials. Front Neurol 2022; 13:933123. [PMID: 36226084 PMCID: PMC9548566 DOI: 10.3389/fneur.2022.933123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 09/05/2022] [Indexed: 11/22/2022] Open
Abstract
Background Multiple sclerosis (MS), an autoimmune disease, is characterized by inflammatory demyelinating lesions in the white matter of the central nervous system. Drugs targeting tyrosine kinase, a critical component of immune cell receptor signaling, have been developed to treat MS. However, the exact efficacy and safety of tyrosine kinase inhibitors (TKIs) are still controversial, and comprehensive analysis with a high level of evidence is needed. Methods Medline, Embase, Cochrane Library, and Clinicaltrials.gov for randomized controlled trials (RCTs) evaluating TKIs versus placebo for MS were searched up to April 1st, 2022. The risk ratio (RR) and mean difference (MD) or standard mean difference (SMD) were analyzed using dichotomous outcomes and continuous outcomes, respectively, with a random effect model. Results A total of 1,043 patients derived from four clinical trials were included to investigate the efficacy and safety of TKI therapy for MS. According to our analysis, TKIs decreased the cumulative number of gadolinium-enhancing lesions on T1-weighted MRI with the application of high dose (SMD = −0.61, 95% CI: −0.93 to −0.30, P = 0.0001). Meanwhile, TKIs prevented the expanded disability status scale (EDSS) from rising (MD = −0.10, 95% CI: −0.19 to −0.00, P = 0.046). In terms of MS relapse, TKIs have not revealed an obvious statistical difference compared with placebo (RR = 0.96, 95% CI: 0.55–1.65, P = 0.8755). However, more adverse events seem to occur in the TKIs group, both for adverse events (RR = 1.12, 95% CI: 1.05–1.19, P = 0.0009) and serious adverse events (RR = 1.91, 95% CI: 1.30–2.81, P = 0.001). Conclusion Tyrosine kinase inhibitors have shown promise in treating MS. Generally, TKIs that attain the effective dose demonstrate definite efficacy and have tolerable side effects. More clinical trials and validation are needed, and we anticipate that TKIs will be a viable alternative for MS patients.
Collapse
Affiliation(s)
- Zeya Yan
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Feng Gu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zilan Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiahao Meng
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xinyu Tao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qiling Dai
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wei Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Meirong Liu
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Meirong Liu
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
- Zhong Wang
| |
Collapse
|
7
|
Novel Drugs in a Pipeline for Progressive Multiple Sclerosis. J Clin Med 2022; 11:jcm11123342. [PMID: 35743410 PMCID: PMC9225445 DOI: 10.3390/jcm11123342] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/27/2022] [Accepted: 06/08/2022] [Indexed: 02/01/2023] Open
Abstract
Multiple sclerosis (MS) is a widely known inflammatory, demyelinating disease of the central nervous system. The pathogenesis of progressive multiple sclerosis (PMS) is a complex, multi-level process that causes therapeutic difficulties. Along with variables such as age and duration of the disease, pathogenetic mechanisms change from inflammatory to neurodegenerative processes. Therefore, the efficacy of available anti-inflammatory drugs approved for the treatment of PMS, such as ocrelizumab or siponimod, is limited in time. In search of innovative solutions, several research studies have been conducted to evaluate the effectiveness of drugs with neuroprotective or remyelinating effects in PMS, including biotin, ibudilast, simvastatin, alpha-lipoic acid, clemastine, amiloride, fluoxetine, riluzole, masitinib, opicinumab, and lamotrigine. The current review includes those compounds, which have entered the clinical phase of assessment, and the authors discuss future prospects for successful PMS treatment.
Collapse
|
8
|
Hove VN, Anderson K, Hayden ER, Pasquariello KZ, Gibson AA, Shen S, Qu J, Jin Y, Miecznikowski JC, Hu S, Sprowl JA. Influence of Tyrosine Kinase Inhibition on Organic Anion Transporting Polypeptide 1B3-Mediated Uptake. Mol Pharmacol 2022; 101:381-389. [PMID: 35383108 PMCID: PMC9354029 DOI: 10.1124/molpharm.121.000287] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 03/22/2022] [Indexed: 11/22/2022] Open
Abstract
The organic anion transporting polypeptide family member (OATP) 1B3 is a hepatic uptake transporter that has a broad substrate recognition and plays a significant role in regulating elimination of endogenous biomolecules or xenobiotics. OATP1B3 works in tandem with OATP1B1, with which it shares approximately 80% sequence homology and a high degree of substrate overlap. Despite some substrates being recognized solely by OATP1B3, its ability to compensate for loss of OATP1B1-mediated elimination and recognition by regulatory agencies, little is known about OATP1B3 regulatory factors and how they are involved with drug-drug interaction. It was recently discovered that OATP1B1 function is mediated by the activity of a particular tyrosine kinase that is sensitive to a variety of tyrosine kinase inhibitors (TKIs). This study reports that OATP1B3 is similarly regulated, as at least 50% of its activity is reduced by 20 US Food and Drug Administration -approved TKIs. Nilotinib was assessed as the most potent OATP1B3 inhibitor among the investigated TKIs, which can occur at clinically relevant concentrations and acted predominantly through noncompetitive inhibition without impacting membrane expression. Finally, OATP1B3 function was determined to be sensitive to the knockdown of the Lck/Yes novel tyrosine kinase that is sensitive to nilotinib and has been previously implicated in mediating OATP1B1 activity. Collectively, our findings identify tyrosine kinase activity as a major regulator of OATP1B3 function which is sensitive to kinase inhibition. Given that OATP1B1 is similarly regulated, simultaneous disruption of these transporters can have drastic effects on systemic drug concentrations, which would promote adverse events. SIGNIFICANCE STATEMENT: The organic anion transporting polypeptide family member (OATP) 1B3 is a facilitator of hepatic drug elimination, although much is unknown of how OATP1B3 activity is mediated, or how such regulators contribute to drug-drug interactions. This study reports that OATP1B3 activity is dependent on the Lck/Yes novel tyrosine kinase, which is sensitive to numerous tyrosine kinase inhibitors. These findings provide insight into the occurrence of many clinical drug-drug interactions, and a rationale for future study of tyrosine kinases regulating drug disposition.
Collapse
Affiliation(s)
- Vusumuzi N Hove
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (V.N.H., K.A., E.R.H., K.Z.P., S.S., J.Q., J.A.S.) and Department of Biostatistics (J.C.M.), University at Buffalo, State University of New York, Buffalo, New York and Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio (A.A.G., Y.J., S.H.)
| | - Kenneth Anderson
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (V.N.H., K.A., E.R.H., K.Z.P., S.S., J.Q., J.A.S.) and Department of Biostatistics (J.C.M.), University at Buffalo, State University of New York, Buffalo, New York and Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio (A.A.G., Y.J., S.H.)
| | - Elizabeth R Hayden
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (V.N.H., K.A., E.R.H., K.Z.P., S.S., J.Q., J.A.S.) and Department of Biostatistics (J.C.M.), University at Buffalo, State University of New York, Buffalo, New York and Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio (A.A.G., Y.J., S.H.)
| | - Kyle Z Pasquariello
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (V.N.H., K.A., E.R.H., K.Z.P., S.S., J.Q., J.A.S.) and Department of Biostatistics (J.C.M.), University at Buffalo, State University of New York, Buffalo, New York and Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio (A.A.G., Y.J., S.H.)
| | - Alice A Gibson
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (V.N.H., K.A., E.R.H., K.Z.P., S.S., J.Q., J.A.S.) and Department of Biostatistics (J.C.M.), University at Buffalo, State University of New York, Buffalo, New York and Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio (A.A.G., Y.J., S.H.)
| | - Shichen Shen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (V.N.H., K.A., E.R.H., K.Z.P., S.S., J.Q., J.A.S.) and Department of Biostatistics (J.C.M.), University at Buffalo, State University of New York, Buffalo, New York and Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio (A.A.G., Y.J., S.H.)
| | - Jun Qu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (V.N.H., K.A., E.R.H., K.Z.P., S.S., J.Q., J.A.S.) and Department of Biostatistics (J.C.M.), University at Buffalo, State University of New York, Buffalo, New York and Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio (A.A.G., Y.J., S.H.)
| | - Yan Jin
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (V.N.H., K.A., E.R.H., K.Z.P., S.S., J.Q., J.A.S.) and Department of Biostatistics (J.C.M.), University at Buffalo, State University of New York, Buffalo, New York and Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio (A.A.G., Y.J., S.H.)
| | - Jeffrey C Miecznikowski
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (V.N.H., K.A., E.R.H., K.Z.P., S.S., J.Q., J.A.S.) and Department of Biostatistics (J.C.M.), University at Buffalo, State University of New York, Buffalo, New York and Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio (A.A.G., Y.J., S.H.)
| | - Shuiying Hu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (V.N.H., K.A., E.R.H., K.Z.P., S.S., J.Q., J.A.S.) and Department of Biostatistics (J.C.M.), University at Buffalo, State University of New York, Buffalo, New York and Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio (A.A.G., Y.J., S.H.)
| | - Jason A Sprowl
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences (V.N.H., K.A., E.R.H., K.Z.P., S.S., J.Q., J.A.S.) and Department of Biostatistics (J.C.M.), University at Buffalo, State University of New York, Buffalo, New York and Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio (A.A.G., Y.J., S.H.)
| |
Collapse
|
9
|
Meira Menezes T, Assis C, Lacerda Cintra AJ, Silva dos Santos RC, Martins do Vale WK, Max Gomes Martins R, de Souza Bezerra R, Seabra GDM, Li C, Neves JL. Binding Mechanism between Acetylcholinesterase and Drugs Pazopanib and Lapatinib: Biochemical and Biophysical Studies. ACS Chem Neurosci 2021; 12:4500-4511. [PMID: 34808043 DOI: 10.1021/acschemneuro.1c00521] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Tyrosine kinase inhibitors (TKIs) are antitumor compounds that prevent the phosphorylation of proteins in a biological environment. However, the multitarget performance of TKIs promotes them as possible candidates for drug repositioning. In this work, interaction and inhibition studies through spectroscopic and computational techniques to evaluate the binding effectiveness of lapatinib and pazopanib TKIs to acetylcholinesterase (AChE) are reported. The results indicated potent inhibition at the μM level. The types of inhibition were identified, with pazopanib acting through non-competitive inhibition and lapatinib through acompetitive inhibition. The fluorescence suppression studies indicate a static mechanism for lapatinib-AChE and pazopanib-AChE systems, with a binding constant in the order of 105 M-1. The obtained thermodynamic parameters reveal interactions driven by van der Waals forces and hydrogen bonds in the lapatinib-AChE system (ΔH° and ΔS° < 0). In contrast, the pazopanib-AChE system shows positive ΔH° and ΔS°, characteristic of hydrophobic interactions. The Foster resonance energy transfer study supports the fluorescence studies performed. The 3D fluorescence studies suggest changes in the microenvironment of the tryptophan and tyrosine residues of the protein in contact with lapatinib and pazopanib. The results suggest effective inhibition and moderate interaction of the drugs with AChE, making them interesting for conducting more in-depth repositioning studies as AChE inhibitors.
Collapse
Affiliation(s)
- Thaís Meira Menezes
- Fundamental Chemistry Department, Federal University of Pernambuco, Recife 50670-901, Brazil
| | - Caio Assis
- Department of Biochemistry and Physiology, Federal University of Pernambuco, Recife 50670-901, Brazil
| | | | | | | | - Regildo Max Gomes Martins
- Post-Graduate in Biotechnology Multi-Institutional Program, PPGBIOTEC, Federal University of Amazonas, Manaus 69067-005, Brazil
| | - Ranilson de Souza Bezerra
- Department of Biochemistry and Physiology, Federal University of Pernambuco, Recife 50670-901, Brazil
| | | | - Chenglong Li
- Department of Medicinal Chemistry, University of Florida, Gainesville, Florida 32610, United States
| | - Jorge Luiz Neves
- Fundamental Chemistry Department, Federal University of Pernambuco, Recife 50670-901, Brazil
| |
Collapse
|
10
|
Appunni S, Gupta D, Rubens M, Ramamoorthy V, Singh HN, Swarup V. Deregulated Protein Kinases: Friend and Foe in Ischemic Stroke. Mol Neurobiol 2021; 58:6471-6489. [PMID: 34549335 DOI: 10.1007/s12035-021-02563-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/10/2021] [Indexed: 12/20/2022]
Abstract
Ischemic stroke is the third leading cause of mortality worldwide, but its medical management is still limited to the use of thrombolytics as a lifesaving option. Multiple molecular deregulations of the protein kinase family occur during the period of ischemia/reperfusion. However, experimental studies have shown that alterations in the expression of essential protein kinases and their pharmacological modulation can modify the neuropathological milieu and hasten neurophysiological recovery. This review highlights the role of key protein kinase members and their implications in the evolution of stroke pathophysiology. Activation of ROCK-, MAPK-, and GSK-3β-mediated pathways following neuronal ischemia/reperfusion injury in experimental conditions aggravate the neuropathology and delays recovery. Targeting ROCK, MAPK, and GSK-3β will potentially enhance myelin regeneration, improve blood-brain barrier (BBB) function, and suppress inflammation, which ameliorates neuronal survival. Conversely, protein kinases such as PKA, Akt, PKCα, PKCε, Trk, and PERK salvage neurons post-ischemia by mechanisms including enhanced toxin metabolism, restoring BBB integrity, neurotrophic effects, and apoptosis suppression. Certain protein kinases such as ERK1/2, JNK, and AMPK have favourable and unfavourable effects in salvaging ischemia-injured neurons. Targeting multiple protein kinase-mediated pathways simultaneously may improve neuronal recovery post-ischemia.
Collapse
Affiliation(s)
- Sandeep Appunni
- Department of Biochemistry, Government Medical College, Kozhikode, Kerala, India
| | - Deepika Gupta
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | | | | | - Himanshu Narayan Singh
- Department of Systems Biology, Columbia University Irving Medical Centre, New York City, NY, USA.
| | - Vishnu Swarup
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
11
|
Abstract
We search for ischemic stroke treatment knowing we have failed-intensely and often-to translate mechanistic knowledge into treatments that alleviate our patients' functional impairments. Lessons can be derived from our shared failures that may point to new directions and new strategies. First, the principle criticisms of both preclinical and clinical assessments are summarized. Next, previous efforts to develop single-mechanism treatments are reviewed. Finally, new definitions, novel approaches, and different directions are presented. In previous development efforts, the basic science and preclinical assessment of candidate treatments often lacked rigor and sufficiency; the clinical trials may have lacked power, rigor, or rectitude; or most likely both preclinical and clinical investigations were flawed. Single-target agents directed against specific molecular mechanisms proved unsuccessful. The term neuroprotection should be replaced as it has become ambiguous: protection of the entire neurovascular unit may be called cerebral cytoprotection or cerebroprotection. Success in developing cerebroprotection-either as an adjunct to recanalization or as stand-alone treatment-will require new definitions that recognize the importance of differential vulnerability in the neurovascular unit. Recent focus on pleiotropic multi-target agents that act via multiple mechanisms of action to interrupt ischemia at multiple steps may be more fruitful. Examples of pleiotropic treatments include therapeutic hypothermia and 3K3A-APC (activated protein C). Alternatively, the single-target drug NA-1 triggers multiple downstream signaling events. Renewed commitment to scientific rigor is essential, and funding agencies and journals may enforce quality principles of rigor in preclinical science. Appropriate animal models should be selected that are suited to the purpose of the investigation. Before clinical trials, preclinical assessment could include subjects that are aged, of both sexes, and harbor comorbid conditions such as diabetes or hypertension. With these new definitions, novel approaches, and renewed attention to rigor, the prospect for successful cerebroprotective therapy should improve.
Collapse
Affiliation(s)
- Patrick D Lyden
- Department of Physiology and Neuroscience, Department of Neurology, Zilkha Neurogenetic Institute, Keck School of Medicine of USC, Los Angeles, CA
| |
Collapse
|
12
|
Role of Adaptor Protein Myeloid Differentiation 88 (MyD88) in Post-Subarachnoid Hemorrhage Inflammation: A Systematic Review. Int J Mol Sci 2021; 22:ijms22084185. [PMID: 33919485 PMCID: PMC8073517 DOI: 10.3390/ijms22084185] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 04/12/2021] [Indexed: 12/15/2022] Open
Abstract
Myeloid differentiation 88 (MyD88) is a well-established inflammatory adaptor protein. It is one of the essential downstream proteins of the toll-like receptor 4 (TLR4) signaling pathway. TLRs are pattern recognition receptors that are usually activated by the damage-associated molecular pattern molecules (DAMPs). Sterile inflammation is triggered by the endogenous DAMPs released in response to global cerebral ischemia and from extravasated blood after subarachnoid hemorrhage (SAH). In this review, we highlight the importance of the neuroinflammatory role of the MyD88 in the SAH. We also explore a few possible pharmacological agents that can be used to decrease SAH-associated neuroinflammation by modulating the MyD88 dependent functions. Pharmacological agents such as flavonoids, melatonin, fluoxetine, pentoxifylline and progesterone have been investigated experimentally to reduce the SAH-associated inflammation. Inhibition of the MyD88 not only reduces the expression of pro-inflammatory cytokines, but also potentially inhibits other processes that can augment the SAH associated inflammation. Further investigations are required to translate these findings in the clinical setting.
Collapse
|
13
|
Kang R, Gamdzyk M, Lenahan C, Tang J, Tan S, Zhang JH. The Dual Role of Microglia in Blood-Brain Barrier Dysfunction after Stroke. Curr Neuropharmacol 2020; 18:1237-1249. [PMID: 32469699 PMCID: PMC7770642 DOI: 10.2174/1570159x18666200529150907] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 04/26/2020] [Accepted: 05/25/2020] [Indexed: 12/17/2022] Open
Abstract
It is well-known that stroke is one of the leading causes of death and disability all over the world. After a stroke, the blood-brain barrier subsequently breaks down. The BBB consists of endothelial cells surrounded by astrocytes. Microglia, considered the long-living resident immune cells of the brain, play a vital role in BBB function. M1 microglia worsen BBB disruption, while M2 microglia assist in repairing BBB damage. Microglia can also directly interact with endothelial cells and affect BBB permeability. In this review, we are going to discuss the mechanisms responsible for the dual role of microglia in BBB dysfunction after stroke.
Collapse
Affiliation(s)
- Ruiqing Kang
- Department of Physiology and Pharmacology, Loma Linda University, School of Medicine, Loma Linda, CA, USA,Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Marcin Gamdzyk
- Department of Physiology and Pharmacology, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Cameron Lenahan
- Department of Physiology and Pharmacology, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Sheng Tan
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| |
Collapse
|
14
|
Ureña-Guerrero ME, Castañeda-Cabral JL, Rivera-Cervantes MC, Macias-Velez RJ, Jarero-Basulto JJ, Gudiño-Cabrera G, Beas-Zárate C. Neuroprotective and Neurorestorative Effects of Epo and VEGF: Perspectives for New Therapeutic Approaches to Neurological Diseases. Curr Pharm Des 2020; 26:1263-1276. [PMID: 31942853 DOI: 10.2174/1381612826666200114104342] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 11/27/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Erythropoietin (Epo) and vascular endothelial growth factor (VEGF) are two vasoactive molecules with essential trophic effects for brain development. The expression and secretion of both molecules increase in response to neuronal damage and they exert protective and restorative effects, which may also be accompanied by adverse side effects. OBJECTIVE We review the most relevant evidence on the neuroprotective and neurorestorative effects of Epo and VEGF in three of the most frequent neurological disorders, namely, stroke, epilepsy and Alzheimer's disease, to develop new therapeutic approaches. METHODS Several original scientific manuscripts and reviews that have discussed the evidence in critical way, considering both the beneficial and adverse effects of Epo and VEGF in the selected neurological disorders, were analysed. In addition, throughout this review, we propose several considerations to take into account in the design of therapeutic approaches based on Epo and VEGF signalling. RESULTS Although the three selected disorders are triggered by different mechanisms, they evolve through similar processes: excitotoxicity, oxidative stress, neuroinflammation, neuronal death, glial reactivity and vascular remodelling. Epo and VEGF exert neuroprotective and neurorestorative effects by acting on these processes due to their pleiotropism. In general, the evidence shows that both Epo and VEGF reduce neuronal death but that at the vascular level, their effects are contradictory. CONCLUSION Because the Epo and VEGF signalling pathways are connected in several ways, we conclude that more experimental studies, primarily studies designed to thoroughly assess the functional interactions between Epo and VEGF in the brain under both physiological and pathophysiological conditions, are needed.
Collapse
Affiliation(s)
- Mónica E Ureña-Guerrero
- Departamento de Biologia Celular y Molecular, Centro Universitario de Ciencias Biologicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| | - José L Castañeda-Cabral
- Departamento de Biologia Celular y Molecular, Centro Universitario de Ciencias Biologicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico.,Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados (CINVESTAV sede Sur), IPN, Ciudad de México, México
| | - Martha C Rivera-Cervantes
- Departamento de Biologia Celular y Molecular, Centro Universitario de Ciencias Biologicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| | - Rafael J Macias-Velez
- Departamento de Biologia Celular y Molecular, Centro Universitario de Ciencias Biologicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| | - José J Jarero-Basulto
- Departamento de Biologia Celular y Molecular, Centro Universitario de Ciencias Biologicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| | - Graciela Gudiño-Cabrera
- Departamento de Biologia Celular y Molecular, Centro Universitario de Ciencias Biologicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| | - Carlos Beas-Zárate
- Departamento de Biologia Celular y Molecular, Centro Universitario de Ciencias Biologicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| |
Collapse
|
15
|
Colàs-Campàs L, Farre J, Mauri-Capdevila G, Molina-Seguín J, Aymerich N, Ois Á, Roquer J, Tur S, García-Carreira MDC, Martí-Fàbregas J, Cruz-Culebras A, Segura T, Arque G, Purroy F. Inflammatory Response of Ischemic Tolerance in Circulating Plasma: Preconditioning-Induced by Transient Ischemic Attack (TIA) Phenomena in Acute Ischemia Patients (AIS). Front Neurol 2020; 11:552470. [PMID: 33192985 PMCID: PMC7658473 DOI: 10.3389/fneur.2020.552470] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 09/30/2020] [Indexed: 11/15/2022] Open
Abstract
Introduction: Ischemic tolerance (IT) refers to a state where cells are resistant to the damaging effects caused by periods of ischemia. In a clinical scenario, the IT phenomenon would be activated by a recent transient ischemic attack (TIA) before an ischemic stroke (IS). The characterization of inflammatory protein expression patterns will contribute to improved understanding of IT. Methods: A total of 477 IS patients from nine hospitals, recruited between January 2011 and January 2016, were included in the current study and divided in three groups: 438 (91.9%) patients without previous TIA (group 1), 22 (4.6%) patients who suffered TIA 24 h before IS (group 2), and 17 (3.5%) patients who suffered TIA between 24 h and 7 days prior to IS (group 3). An inflammatory biomarker panel (IL-6, NT-proBNP, hsCRP, hs-Troponin, NSE, and S-100b) on plasma and a cytokine antibody array was performed to achieve the preconditioning signature potentially induced by TIA phenomena. Primary outcome was modified rankin scale (mRs) score at 90 days. Results: Recent previous TIA was associated with better clinical outcome at 90 days (median mRS of group 1: 2.0 [1.0–4.0]; group 2: 2.0 [0.0–3.0]; group 3: 1.0 [0–2.5]; p = 0.086) and smaller brain lesion (group 1: 3.7 [0.7–18.3]; group 2: 0.8 [0.3–8.9]; group 3: 0.6 [0.1–5.5] mL; p = 0.006). All inflammation biomarkers were down regulated in the groups of recent TIA prior to IS compared to those who did not suffer a TIA events. Moreover, a cytokine antibody array revealed 30 differentially expressed proteins between the three groups. Among them, HRG1-alpha (Fold change 74.4 between group 1 and 2; 74.2 between group 1 and 3) and MAC-1 (Fold change 0.05 between group 1 and 2; 0.06 between group 1 and 3) expression levels would better stratify patients with TIA 7 days before IS. These two proteins showed an earlier inflammation profile that was not detectable by the biomarker panel. Conclusion: Inflammatory pathways were activated by transient ischemic attack, however the period of time between this event and a further ischemic stroke could be determined by a protein signature that would contribute to define the role of ischemic tolerance induced by TIA.
Collapse
Affiliation(s)
- Laura Colàs-Campàs
- Clinical Neurosciences Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain
| | - Joan Farre
- Clinical Neurosciences Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain.,Medical Laboratory, Hospital Universitari Arnau de Vilanova, Lleida, Spain
| | - Gerard Mauri-Capdevila
- Clinical Neurosciences Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain.,Stroke Unit, Department of Neurology, Hospital Universitari Arnau de Vilanova, Lleida, Spain
| | - Jessica Molina-Seguín
- Clinical Neurosciences Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain
| | | | | | | | - Silvia Tur
- Hospital Son Espases, Palma de Mallorca, Spain
| | | | | | | | - Tomás Segura
- Complejo Hospitalario Universitario de Albacete, Albacete, Spain
| | - Gloria Arque
- Clinical Neurosciences Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain
| | - Francisco Purroy
- Clinical Neurosciences Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain.,Stroke Unit, Department of Neurology, Hospital Universitari Arnau de Vilanova, Lleida, Spain
| |
Collapse
|
16
|
Paul S, Candelario-Jalil E. Emerging neuroprotective strategies for the treatment of ischemic stroke: An overview of clinical and preclinical studies. Exp Neurol 2020; 335:113518. [PMID: 33144066 DOI: 10.1016/j.expneurol.2020.113518] [Citation(s) in RCA: 418] [Impact Index Per Article: 83.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/20/2020] [Accepted: 10/23/2020] [Indexed: 12/12/2022]
Abstract
Stroke is the leading cause of disability and thesecond leading cause of death worldwide. With the global population aged 65 and over growing faster than all other age groups, the incidence of stroke is also increasing. In addition, there is a shift in the overall stroke burden towards younger age groups, particularly in low and middle-income countries. Stroke in most cases is caused due to an abrupt blockage of an artery (ischemic stroke), but in some instances stroke may be caused due to bleeding into brain tissue when a blood vessel ruptures (hemorrhagic stroke). Although treatment options for stroke are still limited, with the advancement in recanalization therapy using both pharmacological and mechanical thrombolysis some progress has been made in helping patients recover from ischemic stroke. However, there is still a substantial need for the development of therapeutic agents for neuroprotection in acute ischemic stroke to protect the brain from damage prior to and during recanalization, extend the therapeutic time window for intervention and further improve functional outcome. The current review has assessed the past challenges in developing neuroprotective strategies, evaluated the recent advances in clinical trials, discussed the recent initiative by the National Institute of Neurological Disorders and Stroke in USA for the search of novel neuroprotectants (Stroke Preclinical Assessment Network, SPAN) and identified emerging neuroprotectants being currently evaluated in preclinical studies. The underlying molecular mechanism of each of the neuroprotective strategies have also been summarized, which could assist in the development of future strategies for combinational therapy in stroke treatment.
Collapse
Affiliation(s)
- Surojit Paul
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| | - Eduardo Candelario-Jalil
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
17
|
Jeon SG, Lee HJ, Park H, Han KM, Hoe HS. The VEGF inhibitor vatalanib regulates AD pathology in 5xFAD mice. Mol Brain 2020; 13:131. [PMID: 32977842 PMCID: PMC7519542 DOI: 10.1186/s13041-020-00673-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 09/18/2020] [Indexed: 02/08/2023] Open
Abstract
Alzheimer's disease (AD) is a highly prevalent neurodegenerative disease characterized by Aβ accumulation and tau hyperphosphorylation. Epidemiological evidence for a negative correlation between cancer and AD has led to the proposed use of tyrosine kinase inhibitors (TKIs) such as dasatinib and masitinib for AD, with reported beneficial effects in the AD brain. The TKI vatalanib inhibits angiogenesis by inhibiting vascular endothelial growth factor receptor (VEGFR). Although changes in VEGF and VEGFR have been documented in AD, the effect of vatalanib on AD pathology has not been investigated. In this study, the effects of vatalanib on tau phosphorylation and Aβ accumulation in 5xFAD mice, a model of AD, were evaluated by immunohistochemistry. Vatalanib administration significantly reduced tau phosphorylation at AT8 and AT100 by increasing p-GSK-3β (Ser9) in 5xFAD mice. In addition, vatalanib reduced the number and area of Aβ plaques in the cortex in 5xFAD mice. Our results suggest that vatalanib has potential as a regulator of AD pathology.
Collapse
Affiliation(s)
- Seong Gak Jeon
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, Republic of Korea
| | - Hyun-Ju Lee
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, Republic of Korea
| | - HyunHee Park
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, Republic of Korea
| | - Kyung-Min Han
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, Republic of Korea
| | - Hyang-Sook Hoe
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, Republic of Korea.
| |
Collapse
|
18
|
Palomo V, Nozal V, Rojas-Prats E, Gil C, Martinez A. Protein kinase inhibitors for amyotrophic lateral sclerosis therapy. Br J Pharmacol 2020; 178:1316-1335. [PMID: 32737989 DOI: 10.1111/bph.15221] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 07/03/2020] [Accepted: 07/25/2020] [Indexed: 12/14/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder that causes the progressive loss of motoneurons and, unfortunately, there is no effective treatment for this disease. Interconnecting multiple pathological mechanisms are involved in the neuropathology of this disease, including abnormal aggregation of proteins, neuroinflammation and dysregulation of the ubiquitin proteasome system. Such complex mechanisms, together with the lack of reliable animal models of the disease have hampered the development of drugs for this disease. Protein kinases, a key pharmacological target in several diseases, have been linked to ALS as they play a central role in the pathology of many diseases. Therefore several inhibitors are being currently trailed for clinical proof of concept in ALS patients. In this review, we examine the recent literature on protein kinase inhibitors currently in pharmaceutical development for this diseaseas future therapy for AS together with their involvement in the pathobiology of ALS. LINKED ARTICLES: This article is part of a themed issue on Neurochemistry in Japan. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.6/issuetoc.
Collapse
Affiliation(s)
- Valle Palomo
- Centro de Investigaciones Biológicas-CSIC, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Vanesa Nozal
- Centro de Investigaciones Biológicas-CSIC, Madrid, Spain
| | | | - Carmen Gil
- Centro de Investigaciones Biológicas-CSIC, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Ana Martinez
- Centro de Investigaciones Biológicas-CSIC, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, Madrid, Spain
| |
Collapse
|
19
|
Bauzon J, Lee G, Cummings J. Repurposed agents in the Alzheimer's disease drug development pipeline. Alzheimers Res Ther 2020; 12:98. [PMID: 32807237 PMCID: PMC7433208 DOI: 10.1186/s13195-020-00662-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/29/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Treatments are needed to address the growing prevalence of Alzheimer's disease (AD). Clinical trials have failed to produce any AD drugs for Food and Drug Administration (FDA) approval since 2003, and the pharmaceutical development process is both time-consuming and costly. Drug repurposing provides an opportunity to accelerate this process by investigating the AD-related effects of agents approved for other indications. These drugs have known safety profiles, pharmacokinetic characterization, formulations, doses, and manufacturing processes. METHODS We assessed repurposed AD therapies represented in Phase I, Phase II, and Phase III of the current AD pipeline as registered on ClinicalTrials.gov as of February 27, 2020. RESULTS We identified 53 clinical trials involving 58 FDA-approved agents. Seventy-eight percent of the agents in trials had putative disease-modifying mechanisms of action. Of the repurposed drugs in the pipeline 20% are hematologic-oncologic agents, 18% are drugs derived from cardiovascular indications, 14% are agents with psychiatric uses, 12% are drug used to treat diabetes, 10% are neurologic agents, and the remaining 26% of drugs fall under other conditions. Intellectual property strategies utilized in these programs included using the same drug but altering doses, routes of administration, or formulations. Most repurposing trials were supported by Academic Medical Centers and were not funded through the biopharmaceutical industry. We compared our results to a European trial registry and found results similar to those derived from ClinicalTrials.gov. CONCLUSIONS Drug repurposing is a common approach to AD drug development and represents 39% of trials in the current AD pipeline. Therapies from many disease areas provide agents potentially useful in AD. Most of the repurposed agents are generic and a variety of intellectual property strategies have been adopted to enhance their economic value.
Collapse
Affiliation(s)
- Justin Bauzon
- School of Medicine, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, 89154, USA
| | - Garam Lee
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, 89106, USA
| | - Jeffrey Cummings
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, 89106, USA.
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, 89154, USA.
| |
Collapse
|
20
|
Gupta S, Sharma U, Jagannathan NR, Gupta YK. 1 H NMR metabolomic profiling elucidated attenuation of neurometabolic alterations by lercanidipine in MCAo model in rats. J Pharm Pharmacol 2020; 72:816-825. [PMID: 32163186 DOI: 10.1111/jphp.13249] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 02/09/2020] [Indexed: 12/22/2022]
Abstract
OBJECTIVES Stroke is a leading cause of death and disability worldwide with limited therapeutic interventions. The current study explored proton nuclear magnetic resonance spectroscopy (1 H NMR)-based metabolomic approach to elucidate the effect of lercanidipine on neurometabolic alterations in transient model of ischaemic stroke in rats. METHODS In the present investigation, male Wistar rats were subjected to middle cerebral artery occlusion (MCAo) for 2 h followed by reperfusion using intraluminal filament method. Rats were randomly divided into three groups as vehicle-treated sham control, vehicle-treated MCAo control and lercanidipine-treated MCAo. Vehicle or lercanidipine (0.5 mg/kg, i.p.) was administered 120 min post-reperfusion. The rat brain cortex tissues were isolated 24 h post-MCAo and were investigated by 1 H NMR spectroscopy through perchloric extraction method. KEY FINDINGS A total of 23 metabolites were altered significantly after cerebral ischaemic-reperfusion injury in MCAo control as compared to sham control rats. Lercanidipine significantly reduced the levels of valine, alanine, lactate, acetate and tyrosine, while N-acetylaspartate, glutamate, glutamine, aspartate, creatine/phosphocreatine, choline, glycerophosphorylcholine, taurine, myo-inositol and adenosine di-phosphate were elevated as compared to MCAo control. CONCLUSIONS Present study illustrates effect of lercanidipine on neurometabolic alterations which might be mediated through its antioxidant, anti-inflammatory, vasodilatory and anti-apoptotic property in MCAo model of stroke.
Collapse
Affiliation(s)
- Sangeetha Gupta
- Amity Institute of Pharmacy, Amity University Uttar Pradesh, Noida, India
| | - Uma Sharma
- Department of NMR & MRI Facility, All India Institute of Medical Sciences, New Delhi, India
| | | | | |
Collapse
|
21
|
Sn S, Pandurangi J, Murumalla R, Dj V, Garimella L, Acharya A, Rai S, Paul A, Yarreiphang H, Pillai MS, Giridharan M, Clement JP, Alladi PA, Saiyed T, Manjithaya R. Small molecule modulator of aggrephagy regulates neuroinflammation to curb pathogenesis of neurodegeneration. EBioMedicine 2019; 50:260-273. [PMID: 31727601 PMCID: PMC6921191 DOI: 10.1016/j.ebiom.2019.10.036] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 10/13/2019] [Accepted: 10/20/2019] [Indexed: 12/16/2022] Open
Abstract
Background Plethora of efforts fails to yield a single drug to reverse the pathogenesis of Parkinson's disease (PD) and related α-synucleopathies. Methods Using chemical biology, we identified a small molecule inhibitor of c-abl kinase, PD180970 that could potentially clear the toxic protein aggregates. Genetic, molecular, cell biological and immunological assays were performed to understand the mechanism of action. In vivo preclinical disease model of PD was used to assess its neuroprotection efficacy. Findings In this report, we show the ability of a small molecule inhibitor of tyrosine kinases, PD180970, to induce autophagy (cell lines and mice midbrain) in an mTOR-independent manner and ameliorate the α-synuclein mediated toxicity. PD180970 also exerts anti-neuroinflammatory potential by inhibiting the release of proinflammatory cytokines such as IL-6 (interleukin-6) and MCP-1 (monocyte chemoattractant protein-1) through reduction of TLR-4 (toll like receptor-4) mediated NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) activation. In vivo studies show that PD180970 is neuroprotective by degrading the toxic protein oligomers through induction of autophagy and subsiding the microglial activation. Interpretation These protective mechanisms ensure the negation of Parkinson's disease related motor impairments. Fund This work was supported by Wellcome Trust/DBT India Alliance Intermediate Fellowship (500159-Z-09-Z), DST-SERB grant (EMR/2015/001946), DBT (BT/INF/22/SP27679/2018) and JNCASR intramural funds to RM, and SERB, DST (SR/SO/HS/0121/2012) to PAA, and DST-SERB (SB/YS/LS-215/2013) to JPC and BIRAC funding to ETA C-CAMP.
Collapse
Affiliation(s)
- Suresh Sn
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, India; Centre for Brain Research (CBR), IISc, Bangalore, India
| | - Janhavi Pandurangi
- Centre for Cellular and Molecular Platforms (C-CAMP), Bangalore Life Sciences Cluster (BLiSC), Tata Institute of Fundamental Research, Bangalore, India
| | - Ravi Murumalla
- Centre for Cellular and Molecular Platforms (C-CAMP), Bangalore Life Sciences Cluster (BLiSC), Tata Institute of Fundamental Research, Bangalore, India
| | - Vidyadhara Dj
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, India; Yale University, USA
| | - Lakshmi Garimella
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, India
| | - Achyuth Acharya
- Centre for Cellular and Molecular Platforms (C-CAMP), Bangalore Life Sciences Cluster (BLiSC), Tata Institute of Fundamental Research, Bangalore, India
| | - Shashank Rai
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, India; MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Abhik Paul
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, India
| | - Haorei Yarreiphang
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences, Bangalore, India
| | - Malini S Pillai
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, India
| | - Mridhula Giridharan
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, India
| | - James P Clement
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, India
| | - Phalguni Anand Alladi
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences, Bangalore, India
| | - Taslimarif Saiyed
- Centre for Cellular and Molecular Platforms (C-CAMP), Bangalore Life Sciences Cluster (BLiSC), Tata Institute of Fundamental Research, Bangalore, India
| | - Ravi Manjithaya
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, India; Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, India.
| |
Collapse
|
22
|
Beketova TV, Volkov MY, Naryshkin EA, Novoselova TM, Nasonov EL. Imatinib mesylate use in refractory eosinophilic granulomatosis with polyangiitis: a literature review and a case report. Clin Rheumatol 2018; 37:1729-1735. [DOI: 10.1007/s10067-018-4018-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/28/2018] [Accepted: 01/31/2018] [Indexed: 10/17/2022]
|
23
|
Weddell JC, Imoukhuede PI. Integrative meta-modeling identifies endocytic vesicles, late endosome and the nucleus as the cellular compartments primarily directing RTK signaling. Integr Biol (Camb) 2018; 9:464-484. [PMID: 28436498 DOI: 10.1039/c7ib00011a] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Recently, intracellular receptor signaling has been identified as a key component mediating cell responses for various receptor tyrosine kinases (RTKs). However, the extent each endocytic compartment (endocytic vesicle, early endosome, recycling endosome, late endosome, lysosome and nucleus) contributes to receptor signaling has not been quantified. Furthermore, our understanding of endocytosis and receptor signaling is complicated by cell- or receptor-specific endocytosis mechanisms. Therefore, towards understanding the differential endocytic compartment signaling roles, and identifying how to achieve signal transduction control for RTKs, we delineate how endocytosis regulates RTK signaling. We achieve this via a meta-analysis across eight RTKs, integrating computational modeling with experimentally derived cell (compartment volume, trafficking kinetics and pH) and ligand-receptor (ligand/receptor concentration and interaction kinetics) physiology. Our simulations predict the abundance of signaling from eight RTKs, identifying the following hierarchy in RTK signaling: PDGFRβ > IGFR1 > EGFR > PDGFRα > VEGFR1 > VEGFR2 > Tie2 > FGFR1. We find that endocytic vesicles are the primary cell signaling compartment; over 43% of total receptor signaling occurs within the endocytic vesicle compartment for these eight RTKs. Mechanistically, we found that high RTK signaling within endocytic vesicles may be attributed to their low volume (5.3 × 10-19 L) which facilitates an enriched ligand concentration (3.2 μM per ligand molecule within the endocytic vesicle). Under the analyzed physiological conditions, we identified extracellular ligand concentration as the most sensitive parameter to change; hence the most significant one to modify when regulating absolute compartment signaling. We also found that the late endosome and nucleus compartments are important contributors to receptor signaling, where 26% and 18%, respectively, of average receptor signaling occurs across the eight RTKs. Conversely, we found very low membrane-based receptor signaling, exhibiting <1% of the total receptor signaling for these eight RTKs. Moreover, we found that nuclear translocation, mechanistically, requires late endosomal transport; when we blocked receptor trafficking from late endosomes to the nucleus we found a 57% reduction in nuclear translocation. In summary, our research has elucidated the significance of endocytic vesicles, late endosomes and the nucleus in RTK signal propagation.
Collapse
Affiliation(s)
- Jared C Weddell
- Department of Bioengineering, University of Illinois at Urbana-Champaign, 1304 W Springfield Ave., 3233 Digital Computer Laboratory, Urbana, IL 61801, USA.
| | | |
Collapse
|
24
|
Borriello A, Caldarelli I, Bencivenga D, Stampone E, Perrotta S, Oliva A, Della Ragione F. Tyrosine kinase inhibitors and mesenchymal stromal cells: effects on self-renewal, commitment and functions. Oncotarget 2018; 8:5540-5565. [PMID: 27750212 PMCID: PMC5354929 DOI: 10.18632/oncotarget.12649] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 10/04/2016] [Indexed: 12/18/2022] Open
Abstract
The hope of selectively targeting cancer cells by therapy and eradicating definitively malignancies is based on the identification of pathways or metabolisms that clearly distinguish “normal” from “transformed” phenotypes. Some tyrosine kinase activities, specifically unregulated and potently activated in malignant cells, might represent important targets of therapy. Consequently, tyrosine kinase inhibitors (TKIs) might be thought as the “vanguard” of molecularly targeted therapy for human neoplasias. Imatinib and the successive generations of inhibitors of Bcr-Abl1 kinase, represent the major successful examples of TKI use in cancer treatment. Other tyrosine kinases have been selected as targets of therapy, but the efficacy of their inhibition, although evident, is less definite. Two major negative effects exist in this therapeutic strategy and are linked to the specificity of the drugs and to the role of the targeted kinase in non-malignant cells. In this review, we will discuss the data available on the TKIs effects on the metabolism and functions of mesenchymal stromal cells (MSCs). MSCs are widely distributed in human tissues and play key physiological roles; nevertheless, they might be responsible for important pathologies. At present, bone marrow (BM) MSCs have been studied in greater detail, for both embryological origins and functions. The available data are evocative of an unexpected degree of complexity and heterogeneity of BM-MSCs. It is conceivable that this grade of intricacy occurs also in MSCs of other organs. Therefore, in perspective, the negative effects of TKIs on MSCs might represent a critical problem in long-term cancer therapies based on such inhibitors.
Collapse
Affiliation(s)
- Adriana Borriello
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Ilaria Caldarelli
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Debora Bencivenga
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Emanuela Stampone
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Silverio Perrotta
- Department of Woman, Child and of General and Specialized Surgery, Second University of Naples, Naples, Italy
| | - Adriana Oliva
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Fulvio Della Ragione
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| |
Collapse
|
25
|
Cacho-Díaz B, Lorenzana-Mendoza NA, Spínola-Maroño H, Reyes-Soto G, Cantú-Brito C. Comorbidities, Clinical Features, and Prognostic Implications of Cancer Patients with Cerebrovascular Disease. J Stroke Cerebrovasc Dis 2018; 27:365-371. [DOI: 10.1016/j.jstrokecerebrovasdis.2017.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 08/01/2017] [Accepted: 09/07/2017] [Indexed: 12/12/2022] Open
|
26
|
Decrease of cocaine, but not heroin, self-administration and relapse by the tyrosine kinase inhibitor masitinib in male Sprague Dawley rats. Psychopharmacology (Berl) 2018; 235. [PMID: 29520592 PMCID: PMC5920000 DOI: 10.1007/s00213-018-4865-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
RATIONALE Accumulating evidence shows that cocaine, and also heroin, influence several tyrosine kinases, expressed in neurons and in non-neuronal populations such as microglia, astrocytes and mast-cells. Drug-induced activation of mast cells both triggers inflammatory processes in the brain mediated by the glial cells they activate, and facilitates histamine release which may directly influence the dopamine system. Thus, by triggering the activation and degranulation of mast cells dependent on the tyrosine kinase c-kit and Fyn, the latter being also involved in NMDA-dependent synaptic plasticity, cocaine and heroin may indirectly influence the neural mechanisms that mediate their reinforcing properties. Masitinib, a novel tyrosine kinase inhibitor with high selectivity for c-Kit, Fyn and Lyn, may alter the aberrant consequences of the activation of these tyrosine kinases by cocaine and heroin. OBJECTIVE We investigated in rats the effect of a chronic oral treatment with masitinib (20 mg/kg) on the reinforcing and motivational properties of self-administered cocaine (250 μg/infusion) and heroin (40 μg/infusion). METHODS Three different cohorts of rats were trained instrumentally to respond for cocaine, heroin or food under continuous reinforcement. In each group, we assessed the influence of chronic daily treatment with masitinib on the maintenance of instrumental responding and intake and the motivation for the reinforcer. Thus, masitinib and vehicle-treated rats were challenged to adapt to high behavioural demand, to respond under a progressive ratio schedule of reinforcement and to reinstate instrumental responding after extinction and/or abstinence. RESULTS Masitinib selectively decreased cocaine intake, the motivation for cocaine and the subsequent propensity to respond for cocaine under extinction, while having no effect on instrumental responding for heroin or food. CONCLUSION The present findings suggest masitinib, a drug with proven efficacy in CNS disorders, could represent a novel treatment for cocaine addiction provided its influence on the reinforcing and incentive properties of the drug is confirmed.
Collapse
|
27
|
|
28
|
Chatterjee P, Roy D, Rathi N. Epigenetic Drug Repositioning for Alzheimer’s Disease Based on Epigenetic Targets in Human Interactome. J Alzheimers Dis 2017; 61:53-65. [PMID: 29199645 DOI: 10.3233/jad-161104] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
| | - Debjani Roy
- Department of Biophysics, Bose Institute, West Bengal, India
| | - Nitin Rathi
- School of Electrical and Computer Engineering, Purdue University, West Lafayette, IN, USA
| |
Collapse
|