1
|
Islam F, Roy S, Zehravi M, Paul S, Sutradhar H, Yaidikar L, Kumar BR, Dogiparthi LK, Prema S, Nainu F, Rab SO, Doukani K, Emran TB. Polyphenols Targeting MAP Kinase Signaling Pathway in Neurological Diseases: Understanding Molecular Mechanisms and Therapeutic Targets. Mol Neurobiol 2024; 61:2686-2706. [PMID: 37922063 DOI: 10.1007/s12035-023-03706-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/10/2023] [Indexed: 11/05/2023]
Abstract
Polyphenols are a class of secondary metabolic products found in plants that have been extensively studied for how well they regulate biological processes, such as the proliferation of cells, autophagy, and apoptosis. The mitogen-activated protein kinase (MAPK)-mediated signaling cascade is currently identified as a crucial pro-inflammatory pathway that plays a significant role in the development of neuroinflammation. This process has been shown to contribute to the pathogenesis of several neurological conditions, such as Alzheimer's disease (AD), Parkinson's disease (PD), CNS damage, and cerebral ischemia. Getting enough polyphenols through eating habits has resulted in mitigating the effects of oxidative stress (OS) and lowering the susceptibility to associated neurodegenerative disorders, including but not limited to multiple sclerosis (MS), AD, stroke, and PD. Polyphenols possess significant promise in dealing with the root cause of neurological conditions by modulating multiple therapeutic targets simultaneously, thereby attenuating their complicated physiology. Several polyphenolic substances have demonstrated beneficial results in various studies and are presently undergoing clinical investigation to treat neurological diseases (NDs). The objective of this review is to provide a comprehensive summary of the different aspects of the MAPK pathway involved in neurological conditions, along with an appraisal of the progress made in using polyphenols to regulate the MAPK signaling system to facilitate the management of NDs.
Collapse
Affiliation(s)
- Fahadul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Sumon Roy
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy, College of Dentistry & Pharmacy, Buraydah Private Colleges, Buraydah, 51418, Kingdom of Saudi Arabia.
| | - Shyamjit Paul
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Hriday Sutradhar
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Lavanya Yaidikar
- Department of Pharmacology, Seven Hills College of Pharmacy, Tirupati, India
| | - B Raj Kumar
- Department of Pharmaceutical Analysis, Moonray Institute of Pharmaceutical Sciences, Raikal (V), Farooq Nagar (Tlq), Shadnagar (M), R.R Dist., Telangana, 501512, India
| | - Lakshman Kumar Dogiparthi
- Department of Pharmacognosy, MB School of Pharmaceutical Sciences, MBU, Tirupati, Andhra Pradesh, India
| | - S Prema
- Crescent School of Pharmacy, BS Abdur Rahman Crescent Institute of Science and Technology, Vandalur, Chennai, 600048, India
| | - Firzan Nainu
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, 90245, Indonesia
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Koula Doukani
- Faculty of Nature and Life Sciences, University of Ibn Khaldoun-Tiaret, Tiaret, Algeria
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh.
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School & Legorreta Cancer Center, Brown University, Providence, RI, 02912, USA.
| |
Collapse
|
2
|
Alotaibi BS, Abdel-Rahman Mohamed A, Abd-Elhakim YM, Noreldin AE, Elhamouly M, Khamis T, El-Far AH, Alosaimi ME, Dahran N, Alqahtani LS, Nicotra M, El-Gamal M, Di Cerbo A. Exploring the link between pyrethroids exposure and dopaminergic degeneration through morphometric, immunofluorescence, and in-silico approaches: the therapeutic role of chitosan-encapsulated curcumin nanoparticles. Front Pharmacol 2024; 15:1388784. [PMID: 38751787 PMCID: PMC11094265 DOI: 10.3389/fphar.2024.1388784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 04/11/2024] [Indexed: 05/18/2024] Open
Abstract
Introduction: The synthetic pyrethroid derivative fenpropathrin (FNE), a commonly used insecticide, has been associated with various toxic effects in mammals, particularly neurotoxicity. The study addressed the hallmarks of the pathophysiology of Parkinson's disease upon oral exposure to fenpropathrin (FNE), mainly the alteration of dopaminergic markers, oxidative stress, and molecular docking in rat models. In addition, the protective effect of curcumin-encapsulated chitosan nanoparticles (CRM-Chs-NPs) was also assessed. Methods: In a 60-day trial, 40 male Sprague Dawley rats were divided into 4 groups: Control, CRM-Chs-NPs (curcumin-encapsulated chitosan nanoparticles), FNE (15 mg/kg bw), and FNE + CRM-Chs-NPs. Results: FNE exposure induced reactive oxygen species generation, ATP production disruption, activation of inflammatory and apoptotic pathways, mitochondrial function and dynamics impairment, neurotransmitter level perturbation, and mitophagy promotion in rat brains. Molecular docking analysis revealed that FNE interacts with key binding sites of dopamine synthesis and transport proteins. On the other hand, CRM-Chs-NPs mitigated FNE's toxic effects by enhancing mitochondrial dynamics, antioxidant activity, and ATP production and promoting anti-inflammatory and antiapoptotic responses. Conclusion: In summary, FNE appears to induce dopaminergic degeneration through various mechanisms, and CRM-Chs-NPs emerged as a potential therapeutic intervention for protecting the nervous tissue microenvironment.
Collapse
Affiliation(s)
- Badriyah S. Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Amany Abdel-Rahman Mohamed
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Yasmina M. Abd-Elhakim
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Ahmed E. Noreldin
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - Moustafa Elhamouly
- Cytology and Histology Department, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| | - Tarek Khamis
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
- Laboratory of Biotechnology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Ali H. El-Far
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - Manal E. Alosaimi
- Department of Basic Health Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Naief Dahran
- Department of Anatomy, Faculty of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Leena S. Alqahtani
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Mario Nicotra
- School of Biosciences and Veterinary Medicine, University of Camerino, Matelica, Italy
| | - Mohamed El-Gamal
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Department of Biological Sciences, Faculty of Science, New Mansoura University, New Mansoura City, Egypt
| | - Alessandro Di Cerbo
- School of Biosciences and Veterinary Medicine, University of Camerino, Matelica, Italy
| |
Collapse
|
3
|
Qi XJ, Huang CY, Zuo MT, Gong MD, Huang SJ, Tang MH, Liu ZY. Network Pharmacology and Experimental Verification to Unveil the Mechanism of N-Methyl-D-Aspartic Acid Rescue Humantenirine-Induced Excitotoxicity. Metabolites 2023; 13:metabo13020195. [PMID: 36837814 PMCID: PMC9966887 DOI: 10.3390/metabo13020195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/22/2022] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
Gelsemium is a medicinal plant that has been used to treat various diseases, but it is also well-known for its high toxicity. Complex alkaloids are considered the main poisonous components in Gelsemium. However, the toxic mechanism of Gelsemium remains ambiguous. In this work, network pharmacology and experimental verification were combined to systematically explore the specific mechanism of Gelsemium toxicity. The alkaloid compounds and candidate targets of Gelsemium, as well as related targets of excitotoxicity, were collected from public databases. The crucial targets were determined by constructing a protein-protein interaction (PPI) network. Subsequently, Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) were used to explore the bioprocesses and signaling pathways involved in the excitotoxicity corresponding to alkaloids in Gelsemium. Then, the binding affinity between the main poisonous alkaloids and key targets was verified by molecular docking. Finally, animal experiments were conducted to further evaluate the potential mechanisms of Gelsemium toxicity. A total of 85 alkaloids in Gelsemium associated with 214 excitotoxicity-related targets were predicted by network pharmacology. Functional analysis showed that the toxicity of Gelsemium was mainly related to the protein phosphorylation reaction and plasma membrane function. There were also 164 pathways involved in the toxic mechanism, such as the calcium signaling pathway and MAPK signaling pathway. Molecular docking showed that alkaloids have high affinity with core targets, including MAPK3, SRC, MAPK1, NMDAR2B and NMDAR2A. In addition, the difference of binding affinity may be the basis of toxicity differences among different alkaloids. Humantenirine showed significant sex differences, and the LD50 values of female and male mice were 0.071 mg·kg-1 and 0.149 mg·kg-1, respectively. Furthermore, we found that N-methyl-D-aspartic acid (NMDA), a specific NMDA receptor agonist, could significantly increase the survival rate of acute humantenirine-poisoned mice. The results also show that humantenirine could upregulate the phosphorylation level of MAPK3/1 and decrease ATP content and mitochondrial membrane potential in hippocampal tissue, while NMDA could rescue humantenirine-induced excitotoxicity by restoring the function of mitochondria. This study revealed the toxic components and potential toxic mechanism of Gelsemium. These findings provide a theoretical basis for further study of the toxic mechanism of Gelsemium and potential therapeutic strategies for Gelsemium poisoning.
Collapse
Affiliation(s)
- Xue-Jia Qi
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
- Hunan Engineering Technology Research Center of Veterinary Drugs, Hunan Agricultural University, Changsha 410128, China
| | - Chong-Yin Huang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
- Hunan Engineering Technology Research Center of Veterinary Drugs, Hunan Agricultural University, Changsha 410128, China
| | - Meng-Ting Zuo
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
- Hunan Engineering Technology Research Center of Veterinary Drugs, Hunan Agricultural University, Changsha 410128, China
| | - Meng-Die Gong
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
- Hunan Engineering Technology Research Center of Veterinary Drugs, Hunan Agricultural University, Changsha 410128, China
| | - Si-Juan Huang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
- Hunan Engineering Technology Research Center of Veterinary Drugs, Hunan Agricultural University, Changsha 410128, China
| | - Mo-Huan Tang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
- Hunan Engineering Technology Research Center of Veterinary Drugs, Hunan Agricultural University, Changsha 410128, China
| | - Zhao-Ying Liu
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
- Hunan Engineering Technology Research Center of Veterinary Drugs, Hunan Agricultural University, Changsha 410128, China
- Correspondence:
| |
Collapse
|
4
|
Therapeutic Potential of Polyphenols in the Management of Diabetic Neuropathy. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:9940169. [PMID: 34093722 PMCID: PMC8137294 DOI: 10.1155/2021/9940169] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022]
Abstract
Diabetic neuropathy (DN) is a common and serious diabetes-associated complication that primarily takes place because of neuronal dysfunction in patients with diabetes. Use of current therapeutic agents in DN treatment is quite challenging because of their severe adverse effects. Therefore, there is an increased need of identifying new safe and effective therapeutic agents. DN complications are associated with poor glycemic control and metabolic imbalances, primarily oxidative stress (OS) and inflammation. Various mediators and signaling pathways such as glutamate pathway, activation of channels, trophic factors, inflammation, OS, advanced glycation end products, and polyol pathway have a significant contribution to the progression and pathogenesis of DN. It has been indicated that polyphenols have the potential to affect DN pathogenesis and could be used as potential alternative therapy. Several polyphenols including kolaviron, resveratrol, naringenin, quercetin, kaempferol, and curcumin have been administered in patients with DN. Furthermore, chlorogenic acid can provide protection against glutamate neurotoxicity via its hydrolysate, caffeoyl acid group, and caffeic acid through regulating the entry of calcium into neurons. Epigallocatechin-3-gallate treatment can protect motor neurons by regulating the glutamate level. It has been demonstrated that these polyphenols can be promising in combating DN-associated damaging pathways. In this article, we have summarized DN-associated metabolic pathways and clinical manifestations. Finally, we have also focused on the roles of polyphenols in the treatment of DN.
Collapse
|
5
|
Silymarin Inhibits Glutamate Release and Prevents against Kainic Acid-Induced Excitotoxic Injury in Rats. Biomedicines 2020; 8:biomedicines8110486. [PMID: 33182349 PMCID: PMC7695262 DOI: 10.3390/biomedicines8110486] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/06/2020] [Accepted: 11/06/2020] [Indexed: 12/14/2022] Open
Abstract
Silymarin, a polyphenoic flavonoid derived from the seeds of milk thistle (Silybum marianum), exhibits neuroprotective effects. In this study, we used a model of rat cerebrocortical synaptosomes to investigate whether silymarin affects the release of glutamate, an essential neurotransmitter involved in excitotoxicity. Its possible neuroprotective effect on a rat model of kainic acid (KA)-induced excitotoxicity was also investigated. In rat cortical synaptosomes, silymarin reduced glutamate release and calcium elevation evoked by the K+ channel blocker 4-aminopyridine but did not affect glutamate release caused by the Na+ channel activator veratridine or the synaptosomal membrane potential. Decreased glutamate release by silymarin was prevented by removal of extracellular calcium and blocking of N- and P/Q-type Ca2+ channel or extracellular signal-regulated kinase 1/2 (ERK1/2) but not by blocking of intracellular Ca2+ release. Immunoblotting assay results revealed that silymarin reduced 4-aminopyridine-induced phosphorylation of ERK1/2. Moreover, systemic treatment of rats with silymarin (50 or 100 mg/kg) 30 min before systemic KA (15 mg/kg) administration attenuated KA-induced seizures, glutamate concentration elevation, neuronal damage, glial activation, and heat shock protein 70 expression as well as upregulated KA-induced decrease in Akt phosphorylation in the rat hippocampus. Taken together, the present study demonstrated that silymarin depressed synaptosomal glutamate release by suppressing voltage-dependent Ca2+ entry and ERK1/2 activity and effectively prevented KA-induced in vivo excitotoxicity.
Collapse
|
6
|
Naseri R, Farzaei F, Fakhri S, El-Senduny FF, Altouhamy M, Bahramsoltani R, Ebrahimi F, Rahimi R, Farzaei MH. Polyphenols for diabetes associated neuropathy: Pharmacological targets and clinical perspective. Daru 2019; 27:781-798. [PMID: 31352568 PMCID: PMC6895369 DOI: 10.1007/s40199-019-00289-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 07/01/2019] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVES Diabetic neuropathy (DNP) is a widespread and debilitating complication with complex pathophysiology that is caused by neuronal dysfunction in diabetic patients. Conventional therapeutics for DNP are quite challenging due to their serious adverse effects. Hence, there is a need to investigate novel effective and safe options. The novelty of the present study was to provide available therapeutic approaches, emerging molecular mechanisms, signaling pathways and future directions of DNP as well as polyphenols' effect, which accordingly, give new insights for paving the way for novel treatments in DNP. EVIDENCE ACQUISITION A comprehensive review was done in electronic databases including Medline, PubMed, Web of Science, Scopus, national database (Irandoc and SID), and related articles regarding metabolic pathways on the pathogenesis of DNP as well as the polyphenols' effect. The keywords "diabetic neuropathy" and "diabetes mellitus" in the title/abstract and "polyphenol" in the whole text were used. Data were collected from inception until May 2019. RESULTS DNP complications is mostly related to a poor glycemic control and metabolic imbalances mainly inflammation and oxidative stress. Several signaling and molecular pathways play key roles in the pathogenesis and progression of DNP. Among natural entities, polyphenols are suggested as multi-target alternatives affecting most of these pathogenesis mechanisms in DNP. CONCLUSION The findings revealed novel pathogenicity signaling pathways of DNP and affirmed the auspicious role of polyphenols to tackle these destructive pathways in order to prevent, manage, and treat various diseases. Graphical Abstract .
Collapse
Affiliation(s)
- Rozita Naseri
- Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fardous F El-Senduny
- Biochemistry division, Chemistry Department, Faculty of Science, Mansoura University, Mansoura, 35516, Egypt
| | - Miram Altouhamy
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Roodabeh Bahramsoltani
- Department of Pharmacy in Persian Medicine, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran
- PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Farnaz Ebrahimi
- Pharmacy students` research committee, School of pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Roja Rahimi
- Department of Pharmacy in Persian Medicine, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran
- PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
7
|
Luo Y, Tang H, Li H, Zhao R, Huang Q, Liu J. Recent advances in the development of neuroprotective agents and therapeutic targets in the treatment of cerebral ischemia. Eur J Med Chem 2019; 162:132-146. [DOI: 10.1016/j.ejmech.2018.11.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/30/2018] [Accepted: 11/06/2018] [Indexed: 11/25/2022]
|
8
|
Dhir A. Curcumin in epilepsy disorders. Phytother Res 2018; 32:1865-1875. [PMID: 29917276 DOI: 10.1002/ptr.6125] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 03/01/2018] [Accepted: 05/11/2018] [Indexed: 12/20/2022]
Abstract
Curcumin, a principal curcuminoid present in turmeric, has an antioxidant, anti-inflammatory and neuroprotective properties. Preclinical studies have indicated its beneficial effect for the treatment of epilepsy disorders. The molecule has an anti-seizure potential in preclinical studies, including chemical and electrical models of acute and chronic epilepsy. Curcumin also possesses an anti-epileptogenic activity as it reduces spontaneous recurrent seizures severity in a kainate model of temporal lobe epilepsy. The antioxidant and anti-inflammatory nature of curcumin might be responsible for its observed anti-seizure effects; nevertheless, the exact mechanism is not yet clear. The poor availability of curcumin to the brain limits its use in clinics. The application of nanoliposome and liposome technologies has been tested to enhance its brain availability and penetrability. Unfortunately, there are no randomized, double-blinded controlled clinical trials validating the use of curcumin in epilepsy. The present article analyzes different preclinical evidence illustrating the effect of curcumin in seizure models. The review encourages carrying out clinical trials in this important area of research. In conclusion, curcumin might be beneficial in patients with epilepsy disorders, if its bioavailability issues are resolved.
Collapse
Affiliation(s)
- Ashish Dhir
- Department of Neurology, School of Medicine, University of California, Davis, CA, 95817
| |
Collapse
|
9
|
Gullo F, Ceriani M, D'Aloia A, Wanke E, Constanti A, Costa B, Lecchi M. Plant Polyphenols and Exendin-4 Prevent Hyperactivity and TNF-α Release in LPS-Treated In vitro Neuron/Astrocyte/Microglial Networks. Front Neurosci 2017; 11:500. [PMID: 28932183 PMCID: PMC5592223 DOI: 10.3389/fnins.2017.00500] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 08/23/2017] [Indexed: 12/31/2022] Open
Abstract
Increasing evidence supports a decisive role for neuroinflammation in the neurodegenerative process of several central nervous system (CNS) disorders. Microglia are essential mediators of neuroinflammation and can regulate a broad spectrum of cellular responses by releasing reactive oxygen intermediates, nitric oxide, proteases, excitatory amino acids, and cytokines. We have recently shown that also in ex-vivo cortical networks of neurons, astrocytes and microglia, an increased level of tumor necrosis factor-alpha (TNF-α) was detected a few hours after exposure to the bacterial endotoxin lipopolysaccharide (LPS). Simultaneously, an atypical “seizure-like” neuronal network activity was recorded by multi-electrode array (MEA) electrophysiology. These effects were prevented by minocycline, an established anti-inflammatory antibiotic. We show here that the same inhibitory effect against LPS-induced neuroinflammation is exerted also by natural plant compounds, polyphenols, such as curcumin (CU, curcuma longa), crocin (CR, saffron), and resveratrol (RE, grape), as well as by the glucagon like peptide-1 receptor (GLP-1R) agonist exendin-4 (EX-4). The drugs tested also caused per-se early transient (variable) changes of network activity. Since it has been reported that LPS-induced neuroinflammation causes rearrangements of glutamate transporters in astrocytes and microglia, we suggest that neural activity could be putatively increased by an imbalance of glial glutamate transporter activity, leading to prolonged synaptic glutamatergic dysregulation.
Collapse
Affiliation(s)
- Francesca Gullo
- Department of Biotechnology and Biosciences and Milan Center for Neuroscience, University of Milano-BicoccaMilan, Italy
| | - Michela Ceriani
- Department of Biotechnology and Biosciences and Milan Center for Neuroscience, University of Milano-BicoccaMilan, Italy
| | - Alessia D'Aloia
- Department of Biotechnology and Biosciences, University of Milano-BicoccaMilan, Italy
| | - Enzo Wanke
- Department of Biotechnology and Biosciences, University of Milano-BicoccaMilan, Italy
| | - Andrew Constanti
- Department of Pharmacology, School of Pharmacy, University College LondonLondon, United Kingdom
| | - Barbara Costa
- Department of Biotechnology and Biosciences and Milan Center for Neuroscience, University of Milano-BicoccaMilan, Italy
| | - Marzia Lecchi
- Department of Biotechnology and Biosciences and Milan Center for Neuroscience, University of Milano-BicoccaMilan, Italy
| |
Collapse
|
10
|
Chernigovskaya EV, Korotkov AA, Nikitina LS, Dorofeeva NA, Glazova MV. The expression and distribution of seizure-related and synaptic proteins in the insular cortex of rats genetically prone to audiogenic seizures. Neurol Res 2016; 37:1108-17. [PMID: 26923581 DOI: 10.1080/01616412.2015.1114288] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
It is known that perirhinal/insular cortices participate in the transmission of sensory stimuli to the motor cortex, thus coordinating motor activity during seizures. In the present study we analysed seizure-related proteins, such as GABA, glutamate, ERK1/2 and the synaptic proteins in the insular cortex of Krushinsky-Molodkina (KM) rats genetically prone to audiogenic seizures (AGS). We compared seizure-naïve and seizure-experienced KM rats with control Wistar rats in order to distinguish whether seizure-related protein changes are associated with seizure event or representing an inhered pathological abnormality that determines predisposition to AGS. Our data demonstrated an increased level of vesicular glutamate transporter VGLUT2 in naïve and seizure-experienced KM rats, while glutamic acid decarboxylases GAD65 and GAD67 levels were unchanged. Evaluation of the synaptic proteins showed a decrease in SNAP-25 and upregulation of synapsin I phosphorylation in both groups of KM rats in comparison to Wistar rats. However, when phosphorylation level of ERK1/2 in naïve KM rats was significantly increased, several episodes of AGS diminished ERK1/2 activity. Obtained data indicate that changes in ERK1/2 phosphorylation status and glutamate release controlling synaptic proteins in the insular cortex of KM rats could contribute to the AGS susceptibility.
Collapse
Affiliation(s)
- Elena V Chernigovskaya
- 1 Department of Comparative Biochemistry of cellular functions, Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences , Saint-Petersburg, Russia
| | | | | | | | | |
Collapse
|
11
|
Targeting SUMO-1ylation Contrasts Synaptic Dysfunction in a Mouse Model of Alzheimer's Disease. Mol Neurobiol 2016; 54:6609-6623. [PMID: 27738871 DOI: 10.1007/s12035-016-0176-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 09/28/2016] [Indexed: 12/13/2022]
Abstract
Synaptic dysfunction has been recognized as an early feature occurring at the onset of Alzheimer's disease (AD). Compromised neurotransmission leads over time to synaptic loss and these events correlate with the cognitive decline that progressively affects AD patients.Protein SUMOylation (Small Ubiquitin-like MOdifier) is a post-translational modification (PTM) involved in several cellular processes including synaptic transmission.We here demonstrate that cortical synaptosomes prepared from Tg2576 mice of 6 months of age show an increased SUMO-1ylation, which returns back to normal levels at 20 months although synaptic SUMOylation, at this age, resulted more sensible to KCl stimulus. Our previous findings have shown that increased SUMOylation at presynaptic level reduces the KCl-induced glutamate release. Accordingly, Tg2576 mice of 6 and 20 months show a reduced KCl-evoked neurotransmitter (NT) release. In order to target SUMOylation, we developed two cell penetrating HIV Tat-linked peptides, namely TU-1 and TS-1. This strategy allowed us to modulate the SUMO machinery either positively (TU-1) or negatively (TS-1). As expected, Tg2576 synaptosomes treated with TU-1 exhibited a reduced NT release evoked by KCl. On the contrary, TS-1 treatment, which decreased SUMOylation, was able to normalize impaired glutamate release. Notably, an analysis of autopsy human AD brains has shown an increased SUMOylation in both cortical tissue and synaptosomal lysate. Our data indicate that SUMOylation level changes contribute to the development of synaptic alterations typically occurring at the AD onset and that SUMOylation could be a pharmacological target in AD synaptic dysfunction.
Collapse
|
12
|
Single cell amperometry reveals curcuminoids modulate the release of neurotransmitters during exocytosis from PC12 cells. J Electroanal Chem (Lausanne) 2016; 781:30-35. [PMID: 28579928 DOI: 10.1016/j.jelechem.2016.10.025] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We used single cell amperometry to examine whether curcumin and bisdemethoxycurcumin (BDMC), substances that are suggested to affect learning and memory, can modulate monoamine release from PC12 cells. Our results indicate both curcumin and BDMC need long-term treatment (72 h in this study) to influence exocytosis effectively. By analyzing the parameters calculated from single exocytosis events, it can be concluded that curcumin and BDMC affect exocytosis through different mechanisms. Curcumin accelerates the event dynamics with no significant change of the monoamine amount released from single exocytotic events, whereas BDMC attenuates the amount from single exocytotic event with no significant change of the event dynamics. This comparison of the effect of curcumin and BDMC on exocytosis at the single cell level brings insight into their different mechanisms, which might lead to different biological actions. The effect of curcumin and BDMC on the opening and closing of the exocytotic fusion pore were also investigated. These results might be helpful for understanding the improvement of learning and memory and the anti-depression properties of curcuminoids.
Collapse
|
13
|
Li J, Wang P, Zhu Y, Chen Z, Shi T, Lei W, Yu S. Curcumin Inhibits Neuronal Loss in the Retina and Elevates Ca²⁺/Calmodulin-Dependent Protein Kinase II Activity in Diabetic Rats. J Ocul Pharmacol Ther 2015. [PMID: 26207889 DOI: 10.1089/jop.2015.0006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
PURPOSE To determine whether curcumin offers neuroprotection to minimize the apoptosis of neural cells in the retina of diabetic rats. METHODS Streptozotocin (STZ)-induced diabetic rats and control rats were used in this study. A subgroup of STZ-induced diabetic rats were treated with curcumin for 12 weeks. Retinal histology, apoptosis of neural cells in the retina, electroretinograms, and retinal glutamate content were evaluated after 12 weeks. Retinal levels of Ca(2+)/calmodulin-dependent protein kinase II (CaMKII), phospho-CaMKII (p-CaMKII), and cleaved caspase-3 were determined by Western blot analysis. RESULTS The amplitudes a-wave, b-wave, and oscillatory potential were reduced by diabetes, but curcumin treatment suppressed this reduction of amplitudes. Curcumin also prevented cell loss from the outer nuclear, inner nuclear, and ganglion cell layers. Apoptosis of retinal neurons was detected in diabetic rats. The concentration of glutamate in the retina was higher in diabetic rats, but was significantly reduced in the curcumin-treated group. Furthermore, p-CaMKII and cleaved caspase-3 expression were upregulated in the diabetic retina, but reduced in curcumin-treated rats. CONCLUSIONS Curcumin attenuated diabetes-induced apoptosis in retinal neurons by reducing the glutamate level and downregulating CaMKII. Thus, curcumin might be used to prevent neuronal damage in the retina of patients with diabetes mellitus.
Collapse
Affiliation(s)
- Jun Li
- Department of Ophthalmology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical University , Lishui, Zhejiang Province, People's Republic of China
| | - Peipei Wang
- Department of Ophthalmology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical University , Lishui, Zhejiang Province, People's Republic of China
| | - Yanxia Zhu
- Department of Ophthalmology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical University , Lishui, Zhejiang Province, People's Republic of China
| | - Zhen Chen
- Department of Ophthalmology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical University , Lishui, Zhejiang Province, People's Republic of China
| | - Tianyan Shi
- Department of Ophthalmology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical University , Lishui, Zhejiang Province, People's Republic of China
| | - Wensheng Lei
- Department of Ophthalmology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical University , Lishui, Zhejiang Province, People's Republic of China
| | - Songping Yu
- Department of Ophthalmology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical University , Lishui, Zhejiang Province, People's Republic of China
| |
Collapse
|
14
|
Li Y, Li J, Li S, Li Y, Wang X, Liu B, Fu Q, Ma S. Curcumin attenuates glutamate neurotoxicity in the hippocampus by suppression of ER stress-associated TXNIP/NLRP3 inflammasome activation in a manner dependent on AMPK. Toxicol Appl Pharmacol 2015; 286:53-63. [DOI: 10.1016/j.taap.2015.03.010] [Citation(s) in RCA: 150] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 03/05/2015] [Accepted: 03/06/2015] [Indexed: 12/23/2022]
|
15
|
Nisticò R, Florenzano F, Mango D, Ferraina C, Grilli M, Di Prisco S, Nobili A, Saccucci S, D'Amelio M, Morbin M, Marchi M, Mercuri NB, Davis RJ, Pittaluga A, Feligioni M. Presynaptic c-Jun N-terminal Kinase 2 regulates NMDA receptor-dependent glutamate release. Sci Rep 2015; 5:9035. [PMID: 25762148 PMCID: PMC4357012 DOI: 10.1038/srep09035] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 02/13/2015] [Indexed: 12/17/2022] Open
Abstract
Activation of c-Jun N-terminal kinase (JNK) signaling pathway is a critical step for neuronal death occurring in several neurological conditions. JNKs can be activated via receptor tyrosine kinases, cytokine receptors, G-protein coupled receptors and ligand-gated ion channels, including the NMDA glutamate receptors. While JNK has been generally associated with postsynaptic NMDA receptors, its presynaptic role remains largely unexplored. Here, by means of biochemical, morphological and functional approaches, we demonstrate that JNK and its scaffold protein JIP1 are also expressed at the presynaptic level and that the NMDA-evoked glutamate release is controlled by presynaptic JNK-JIP1 interaction. Moreover, using knockout mice for single JNK isoforms, we proved that JNK2 is the essential isoform in mediating this presynaptic event. Overall the present findings unveil a novel JNK2 localization and function, which is likely to play a role in different physiological and pathological conditions.
Collapse
Affiliation(s)
- Robert Nisticò
- 1] Laboratory of Pharmacology of Synaptic Plasticity, EBRI "Rita Levi-Montalcini" Foundation, Rome, 00143, Italy [2] Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, 00185, Italy
| | - Fulvio Florenzano
- Laboratory of Pharmacology of Synaptic Plasticity, EBRI "Rita Levi-Montalcini" Foundation, Rome, 00143, Italy
| | - Dalila Mango
- 1] Laboratory of Pharmacology of Synaptic Plasticity, EBRI "Rita Levi-Montalcini" Foundation, Rome, 00143, Italy [2] Laboratory of Experimental Neurology, IRCCS Fondazione Santa Lucia, Rome, 00143, Italy
| | - Caterina Ferraina
- Laboratory of Pharmacology of Synaptic Plasticity, EBRI "Rita Levi-Montalcini" Foundation, Rome, 00143, Italy
| | - Massimo Grilli
- Department of Pharmacy, Pharmacology and Toxicology Section, University of Genoa, Genoa, 16148, Italy
| | - Silvia Di Prisco
- Department of Pharmacy, Pharmacology and Toxicology Section, University of Genoa, Genoa, 16148, Italy
| | - Annalisa Nobili
- 1] Laboratory of Experimental Neurology, IRCCS Fondazione Santa Lucia, Rome, 00143, Italy [2] University Campus Biomedico, 00100 Rome
| | - Stefania Saccucci
- Neuropathology &Neurology V - IRCCS Foundation C. Besta Milan, 20133, Italy
| | - Marcello D'Amelio
- 1] Laboratory of Experimental Neurology, IRCCS Fondazione Santa Lucia, Rome, 00143, Italy [2] University Campus Biomedico, 00100 Rome
| | - Michela Morbin
- Neuropathology &Neurology V - IRCCS Foundation C. Besta Milan, 20133, Italy
| | - Mario Marchi
- 1] Department of Pharmacy, Pharmacology and Toxicology Section, University of Genoa, Genoa, 16148, Italy [2] Center of Excellence for Biomedical Research, University of Genoa, Genoa, 16132, Italy
| | - Nicola B Mercuri
- Laboratory of Experimental Neurology, IRCCS Fondazione Santa Lucia, Rome, 00143, Italy
| | - Roger J Davis
- Department of Biochemistry and Molecular Biology, Howard Hughes Medical Institute and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, 01605, USA
| | - Anna Pittaluga
- 1] Department of Pharmacy, Pharmacology and Toxicology Section, University of Genoa, Genoa, 16148, Italy [2] Center of Excellence for Biomedical Research, University of Genoa, Genoa, 16132, Italy
| | - Marco Feligioni
- Laboratory of Pharmacology of Synaptic Plasticity, EBRI "Rita Levi-Montalcini" Foundation, Rome, 00143, Italy
| |
Collapse
|
16
|
Chang CH, Chen HX, Yü G, Peng CC, Peng RY. Curcumin-Protected PC12 Cells Against Glutamate-Induced Oxidative Toxicity. Food Technol Biotechnol 2014; 52:468-478. [PMID: 27904320 PMCID: PMC5079148 DOI: 10.17113/ftb.52.04.14.3622] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Accepted: 08/11/2014] [Indexed: 01/23/2023] Open
Abstract
Glutamate is a major excitatory neurotransmitter present in the central nervous system. The glutamate/cystine antiporter system x c- connects the antioxidant defense with neurotransmission and behaviour. Overactivation of ionotropic glutamate receptors induces neuronal death, a pathway called excitotoxicity. Glutamate-induced oxidative stress is a major contributor to neurodegenerative diseases including cerebral ischemia, Alzheimer's and Huntington's disease. Curcuma has a wide spectrum of biological activities regarding neuroprotection and neurocognition. By reducing the oxidative damage, curcumin attenuates a spinal cord ischemia-reperfusion injury, seizures and hippocampal neuronal loss. The rat pheochromocytoma (PC12) cell line exhibits many characteristics useful for the study of the neuroprotection and neurocognition. This investigation was carried out to determine whether the neuroprotective effects of curcumin can be observed via the glutamate-PC12 cell model. Results indicate that glutamate (20 mM) upregulated glutathione peroxidase 1, glutathione disulphide, Ca2+ influx, nitric oxide production, cytochrome c release, Bax/Bcl-2 ratio, caspase-3 activity, lactate dehydrogenase release, reactive oxygen species, H 2 O 2 , and malondialdehyde; and downregulated glutathione, glutathione reductase, superoxide dismutase and catalase, resulting in enhanced cell apoptosis. Curcumin alleviates all these adverse effects. Conclusively, curcumin can effectively protect PC12 cells against the glutamate-induced oxidative toxicity. Its mode of action involves two pathways: the glutathione-dependent nitric oxide-reactive oxygen species pathway and the mitochondria-dependent nitric oxide-reactive oxygen species pathway.
Collapse
Affiliation(s)
- Chi-Huang Chang
- Research Institute of Biotechnology, Hungkuang University, 34 Chung-Chie Rd., Shalu County,
Taichung City 43022, Taiwan
| | - Hua-Xin Chen
- Department of Pharmacy, Kuang-Tieng General Hospital, Shalu County, Taichung City 43302, Taiwan
| | - George Yü
- Research Institute of Biotechnology, Hungkuang University, 34 Chung-Chie Rd., Shalu County,
Taichung City 43022, Taiwan
| | - Chiung-Chi Peng
- Graduate Institute of Clinical Medicine, Taipei Medical University, 250 Wu-Xing St., Taipei 10031, Taiwan
| | - Robert Y. Peng
- Research Institute of Biotechnology, Hungkuang University, 34 Chung-Chie Rd., Shalu County,
Taichung City 43022, Taiwan
- Research Institute of Medical Sciences, Taipei Medical University, 250 Wu-Xing St., Taipei 10031, Taiwan
| |
Collapse
|
17
|
Glazova MV, Nikitina LS, Hudik KA, Kirillova OD, Dorofeeva NA, Korotkov AA, Chernigovskaya EV. Inhibition of ERK1/2 signaling prevents epileptiform behavior in rats prone to audiogenic seizures. J Neurochem 2014; 132:218-29. [PMID: 25351927 DOI: 10.1111/jnc.12982] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 10/14/2014] [Accepted: 10/22/2014] [Indexed: 01/24/2023]
Abstract
It has recently been proposed that extracellular signal-regulated kinases 1 and 2 (ERK1/2) are one of the factors mediating seizure development. We hypothesized that inhibition of ERK1/2 activity could prevent audiogenic seizures by altering GABA and glutamate release mechanisms. Krushinsky-Molodkina rats, genetically prone to audiogenic seizure, were recruited in the experiments. Animals were i.p. injected with an inhibitor of ERK1/2 SL 327 at different doses 60 min before audio stimulation. We demonstrated for the first time that inhibition of ERK1/2 activity by SL 327 injections prevented seizure behavior and this effect was dose-dependent and correlated with ERK1/2 activity. The obtained data also demonstrated unchanged levels of GABA production, and an increase in the level of vesicular glutamate transporter 2. The study of exocytosis protein expression showed that SL 327 treatment leads to downregulation of vesicle-associated membrane protein 2 and synapsin I, and accumulation of synaptosomal-associated protein 25 (SNAP-25). The obtained data indicate that the inhibition of ERK1/2 blocks seizure behavior presumably by altering the exocytosis machinery, and identifies ERK1/2 as a potential target for the development of new strategies for seizure treatment. Extracellular signal-regulated kinases 1 and 2 (ERK1/2) are one of the factors mediating seizure development. Here we report that inhibition of ERK1/2 by SL 327 prevented seizure behavior and this effect was dose-dependent and correlated with ERK1/2 activity. Accumulation of VGLUT2 was associated with differential changing of synaptic proteins VAMP2, SNAP-25 and synapsin I. The obtained data indicate that the inhibition of ERK1/2 alters neurotransmitter release by changing the exocytosis machinery, thus preventing seizures.
Collapse
Affiliation(s)
- Margarita V Glazova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Saint-Petersburg, Russia
| | | | | | | | | | | | | |
Collapse
|