1
|
Aboelnasr FG, George MY, Nasr M, Menze ET. Silymarin nanoparticles counteract cognitive impairment induced by doxorubicin and cyclophosphamide in rats; Insights into mitochondrial dysfunction and Nrf-2/HO-1 axis. Eur J Pharmacol 2024; 988:177217. [PMID: 39706469 DOI: 10.1016/j.ejphar.2024.177217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/16/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Most cancer patients suffer cognitive impairment following chemotherapy, recognized as "chemobrain". Principally, doxorubicin and cyclophosphamide are frequently utilized conjointly for the treatment of several kinds of tumors. Silymarin was reported to possess anti-inflammatory, antioxidant, and neuroprotective impacts. The recent study shed light on the neuroprotective attributes of silymarin against cognitive dysfunction instigated in rats with doxorubicin/cyclophosphamide combination. Unfortunately, silymarin suffers reduced absorption following oral administration. Silymarin was formulated as a nanoemulsion to be administered intranasally. Male rats were allocated into six groups: control, doxorubicin (2 mg/kg, ip) and cyclophosphamide (50 mg/kg, ip), doxorubicin and cyclophosphamide + silymarin (200 mg/kg, oral), doxorubicin and cyclophosphamide + silymarin nanoemulsion (1 mg/kg, intranasal), silymarin (200 mg/kg, oral), and silymarin nanoemulsion (1 mg/kg, intranasal) groups, and treated for 21 days. The amount of silymarin reaching the brain was found to be enhanced following formulated nanoemulsion administration. Doxorubicin and cyclophosphamide caused behavioral, as well as memory deficits indicated by locomotor activity, y maze, and passive avoidance tests. Also, they induced histological alteration in hippocampi and the prefrontal cortices of rats. Besides, chemotherapy caused cognitive impairment assessed by acetylcholinesterase activity elevation. Additionally, caspase-3 augmentation and of nuclear factor erythroid 2-related factor-2 (Nrf-2) and heme oxygenase-1 (HO-1) pathway disturbance were found following chemotherapy treatment. Silymarin treatment opposed such effects via enhancing memory function, preserving brain architecture, and reducing acetylcholinesterase activity and caspase-3 level. Moreover, silymarin treatment improved mitochondrial biogenesis through activation Nrf-2/HO-1 axis. Collectively, silymarin nanoemulsion, at a 200-fold lower dose, can offer an innovative solution for cancer patients globally.
Collapse
Affiliation(s)
- Fatma G Aboelnasr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566, Cairo, Egypt
| | - Mina Y George
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566, Cairo, Egypt.
| | - Maha Nasr
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, 11566, Cairo, Egypt
| | - Esther T Menze
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566, Cairo, Egypt
| |
Collapse
|
2
|
Elesawy WH, El-Sahar AE, Sayed RH, Ashour AM, Alsufyani SE, Arab HH, Kandil EA. Repurposing ezetimibe as a neuroprotective agent in a rotenone-induced Parkinson's disease model in rats: Role of AMPK/SIRT-1/PGC-1α signaling and autophagy. Int Immunopharmacol 2024; 138:112640. [PMID: 38981225 DOI: 10.1016/j.intimp.2024.112640] [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: 05/29/2024] [Revised: 06/30/2024] [Accepted: 07/03/2024] [Indexed: 07/11/2024]
Abstract
As a severe neurological disorder, Parkinson's disease (PD) is distinguished by dopaminergic neuronal degeneration in the substantia nigra (SN), culminating in motor impairments. Several studies have shown that activation of the AMPK/SIRT1/PGC1α pathway contributes to an increase in mitochondrial biogenesis and is a promising candidate for the management of PD. Furthermore, turning on the AMPK/SIRT1/PGC1α pathway causes autophagy activation, which is fundamental for maintaining neuronal homeostasis. Interestingly, ezetimibe is an antihyperlipidemic agent that was recently reported to possess pleiotropic properties in neurology by triggering the phosphorylation and activation of AMPK. Thus, our study aimed to investigate the neuroprotective potential of ezetimibe in rats with rotenone-induced PD by activating AMPK. Adult male Wistar rats received rotenone (1.5 mg/kg, s.c.) every other day for 21 days to induce experimental PD. Rats were treated with ezetimibe (5 mg/kg/day, i.p.) 1 h before rotenone. Ezetimibe ameliorated the motor impairments in open field, rotarod and grip strength tests, restored striatal dopamine and tyrosine hydroxylase in the SN, up-regulated p-AMPK, SIRT1, and PGC1α striatal expression, upsurged the expression of ULK1, beclin1, and LC3II/I, reduced Bax/Bcl2 ratio, and alleviated rotenone-induced histopathological changes in striatum and SN. Our findings also verified the contribution of AMPK activation to the neuroprotective effect of ezetimibe by using the AMPK inhibitor dorsomorphin. Together, this work revealed that ezetimibe exerts a neuroprotective impact in rotenone-induced PD by activating AMPK/SIRT-1/PGC-1α signaling, enhancing autophagy, and attenuating apoptosis. Thus, ezetimibe's activation of AMPK could hold significant therapeutic promise for PD management.
Collapse
Affiliation(s)
- Wessam H Elesawy
- Department of Pharmacology and Toxicology, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST), 6 October, Egypt
| | - Ayman E El-Sahar
- Biology Department, School of Pharmacy, Newgiza University, Giza, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Rabab H Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Ahmed M Ashour
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al Qura University, P.O. Box 13578, Makkah 21955, Saudi Arabia
| | - Shuruq E Alsufyani
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Hany H Arab
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.
| | - Esraa A Kandil
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
3
|
Cai Y, Tong F, Li K, Wang Q, Ding J, Wang X. Cannabinoid receptor 2 agonist AM1241 alleviates epileptic seizures and epilepsy-associated depression via inhibiting neuroinflammation in a pilocarpine-induced chronic epilepsy mouse model. Mol Cell Neurosci 2024; 130:103958. [PMID: 39151841 DOI: 10.1016/j.mcn.2024.103958] [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: 04/14/2024] [Revised: 08/09/2024] [Accepted: 08/11/2024] [Indexed: 08/19/2024] Open
Abstract
Increasing evidence suggests that cannabinoid receptor 2 (CB2R) serves as a promising anti-inflammatory target. While inflammation is known to play crucial roles in the pathogenesis of epilepsy, the involvement of CB2R in epilepsy remains unclear. This study aimed to investigate the effects of a CB2R agonist, AM1241, on epileptic seizures and depressive-like behaviors in a mouse model of chronic epilepsy induced by pilocarpine. A chronic epilepsy mouse model was established by intraperitoneal administration of pilocarpine. The endogenous cannabinoid system (eCBs) in the hippocampus was examined after status epilepticus (SE). Animals were then treated with AM1241 and compared with a vehicle-treated control group. Additionally, the role of the AMPK/NLRP3 signaling pathway was explored using the selective AMPK inhibitor dorsomorphin. Following SE, CB2R expression increased significantly in hippocampal microglia. Administration of AM1241 significantly reduced seizure frequency, immobility time in the tail suspension test, and neuronal loss in the hippocampus. In addition, AM1241 treatment attenuated microglial activation, inhibited pro-inflammatory polarization of microglia, and suppressed NLRP3 inflammasome activation in the hippocampus after SE. Further, the therapeutic effects of AM1241 were abolished by the AMPK inhibitor dorsomorphin. Our findings suggest that CB2R agonist AM1241 may alleviate epileptic seizures and its associated depression by inhibiting neuroinflammation through the AMPK/NLRP3 signaling pathway. These results provide insight into a novel therapeutic approach for epilepsy.
Collapse
Affiliation(s)
- Yiying Cai
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fangchao Tong
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kexian Li
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiang Wang
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Ding
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Xin Wang
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China; Department of the State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
4
|
Yang Y, Ke J, Cao Y, Gao Y, Lin C. Melatonin regulates microglial M1/M2 polarization via AMPKα2-mediated mitophagy in attenuating sepsis-associated encephalopathy. Biomed Pharmacother 2024; 177:117092. [PMID: 38976956 DOI: 10.1016/j.biopha.2024.117092] [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: 04/29/2024] [Revised: 06/23/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND Sepsis-associated encephalopathy (SAE) is a disease characterized by neuroinflammation and cognitive dysfunction caused by systemic infection. Inflammation-induced microglial activation is closely associated with neuroinflammation in SAE. It is widely understood that melatonin has strong anti-inflammatory and immunomodulatory properties beneficial for sepsis-related brain damage. However, the mechanism of melatonin action in SAE has not been fully elucidated. METHODS The SAE cell model and SAE mouse model were induced by lipopolysaccharide (LPS). Behavioral tests were performed to analyze cognitive function. Microglial markers and M1/M2 markers were measured by immunofluorescence. Mitophagy was assessed by western blot, mt-Keima and transmission electron microscopy experiments. Immunoprecipitation and co-immunoprecipitation assays investigated the interactions between AMP-activated protein kinase α2 (AMPKα2) and PTEN-induced putative kinase 1 (PINK1). RESULTS Melatonin suppresses LPS-induced microglia M1 polarization by enhancing mitophagy, thereby attenuating LPS-induced neuroinflammation and behavioral deficits. However, inhibition or knockdown of AMPKα2 can inhibit the enhancement of melatonin on mitophagy, then weaken its promotion of microglia polarization towards M2 phenotype, and eliminate its protective effect on brain function. Furthermore, melatonin enhances mitophagy through activating AMPKα2, promotes PINK1 Ser495 site phosphorylation, and ultimately regulates microglial polarization from M1 to M2. CONCLUSIONS Our findings demonstrate that melatonin facilitates microglia polarization towards M2 phenotype to alleviate LPS-induced neuroinflammation, primarily through AMPKα2-mediated enhancement of mitophagy.
Collapse
Affiliation(s)
- Yang Yang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University; The key Laboratory of Precision Anesthesia & perioperative Organ Protection, Guangzhou, Guangdong, 510515, China.
| | - Jinyong Ke
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University; The key Laboratory of Precision Anesthesia & perioperative Organ Protection, Guangzhou, Guangdong, 510515, China.
| | - Yang Cao
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University; The key Laboratory of Precision Anesthesia & perioperative Organ Protection, Guangzhou, Guangdong, 510515, China.
| | - Yue Gao
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University; The key Laboratory of Precision Anesthesia & perioperative Organ Protection, Guangzhou, Guangdong, 510515, China.
| | - Chunshui Lin
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University; The key Laboratory of Precision Anesthesia & perioperative Organ Protection, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
5
|
Yang H, Shao ZH, Jin X, Chen JW. The critical role of P2XR/PGC-1α signalling pathway in hypoxia-mediated pyroptosis and M1/M2 phenotypic differentiation of mouse microglia. Eur J Neurosci 2024; 60:3629-3642. [PMID: 38697919 DOI: 10.1111/ejn.16363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 01/09/2024] [Accepted: 04/01/2024] [Indexed: 05/05/2024]
Abstract
Microglia are endogenous immune cells in the brain, and their pyroptosis and phenotype dichotomy are proved to play roles in neurodegenerative diseases. We investigated whether and how hypoxia affected pyroptosis and phenotype polarization in mouse microglia. Primary mouse microglia and BV2 microglia were exposed to hypoxia. Pyroptosis and M1/M2 phenotype were assessed by measuring gasdermin D truncation and M1/M2 surface marker expression. Mechanisms including purinergic ionotropic receptor (P2XR), peroxisome proliferator-activated receptor coactivator-1α (PGC-1α) and NOD-like receptor protein 3 (NLRP3) inflammasome were investigated. We reported hypoxia (90% N2, 5% O2 and 5% CO2) induced pyroptosis and promoted M1 phenotype polarization in primary mouse microglia and BV2 microglia, and the effect appeared after 6 h exposure. Although hypoxia (90% N2, 5% O2 and 5% CO2, 6 h) had no effect on P2X1R and P2X7R expression, it increased P2X4R expression and decreased PGC-1α expression. Interestingly, blockade of P2X4R or P2X7R abolished hypoxia-modulated PGC-1α expression, pyroptosis and M1 polarization. PGC-1α overexpression or overactivation alleviated hypoxia-induced pyroptosis and M1 polarization, while PGC-1α knockdown or deactivation promoted pyroptosis and M1 polarization under normoxic situation. Further, hypoxia induced NLRP3 expression and activated caspase-1 and induced the phosphorylation of NF-κB and reduced the phosphorylation of STAT3/6. NLRP3 inhibitor and caspase-1 inhibitor abolished hypoxia-induced pyroptosis, while NF-κB inhibitor and STAT phosphorylation inducer ameliorated hypoxia-induced M1 polarization. In addition, NF-κB activator and STAT3/6 inhibitor caused microglia M1 polarization under normoxic situation. We concluded in cultured mouse microglia, hypoxia may induce pyroptosis via P2XR/PGC-1α/NLRP3/caspase-1 pathway and trigger M1 polarization through P2XR/PGC-1α/NF-κB/STAT3/6 pathway.
Collapse
Affiliation(s)
- Hao Yang
- Department of Critical Care Medicine, Jing'an District Central Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China
| | - Zhen-Hua Shao
- Department of Critical Care Medicine, Jing'an District Central Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China
| | - Xian Jin
- Department of Critical Care Medicine, Jing'an District Central Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China
| | - Jia-Wei Chen
- Department of Critical Care Medicine, Jing'an District Central Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
6
|
George MY, Dabour MS, Rashad E, Zordoky BN. Empagliflozin Alleviates Carfilzomib-Induced Cardiotoxicity in Mice by Modulating Oxidative Stress, Inflammatory Response, Endoplasmic Reticulum Stress, and Autophagy. Antioxidants (Basel) 2024; 13:671. [PMID: 38929110 PMCID: PMC11200801 DOI: 10.3390/antiox13060671] [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: 04/24/2024] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
Carfilzomib is an irreversible proteasome inhibitor used for multiple myeloma patients. However, carfilzomib treatment is associated with cardiovascular complications. Empagliflozin, an Sodium Glucose Co-transporter 2 inhibitor (SGLT-2) inhibitor, is an oral antidiabetic drug with proven antioxidant and anti-inflammatory properties. The aim of the present study was to determine the cardioprotective effects of empagliflozin against carfilzomib-induced cardiotoxicity. C57BL/6 mice were randomly divided into four groups: control, empagliflozin, carfilzomib, and carfilzomib + empagliflozin. Empagliflozin prevented carfilzomib-induced cardiotoxicity by ameliorating histological alterations, CK-MB, and troponin-I. Moreover, it inhibited carfilzomib-induced oxidative damage and inflammation via its action on catalase activity, reduced glutathione levels and superoxide dismutase activity, and reduced nuclear factor-κB (p65) and cytokine levels. Mechanistically, empagliflozin abrogated endoplasmic reticulum stress induced by carfilzomib, as evidenced by the effect on the Glucose Regulated Protein-78 (GRP-78)/Activating Transcription Factor 6 (ATF6)/C/EBP homologous protein (CHOP) axis. Intriguingly, carfilzomib significantly induced autophagy, an effect that was further enhanced by empagliflozin, evidenced by increased LC3B and beclin-1 mRNA expression and reduced p62 expression. The effect of empagliflozin on apoptosis was confirmed by reduced expression of active caspase-3. Importantly, empagliflozin did not alter the cytotoxic effect of carfilzomib on human U266B1 multiple myeloma cells. our findings suggest that empagliflozin may provide a new therapeutic strategy to mitigate carfilzomib-induced cardiotoxicity in multiple myeloma patients.
Collapse
Affiliation(s)
- Mina Y. George
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (M.S.D.); (B.N.Z.)
| | - Mohamed S. Dabour
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (M.S.D.); (B.N.Z.)
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tanta University, Tanta 31111, Egypt
| | - Eman Rashad
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt;
| | - Beshay N. Zordoky
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; (M.S.D.); (B.N.Z.)
| |
Collapse
|
7
|
Elhoseny SM, Saleh NM, Meshali MM. Self-Nanoemulsion Intrigues the Gold Phytopharmaceutical Chrysin: In Vitro Assessment and Intrinsic Analgesic Effect. AAPS PharmSciTech 2024; 25:54. [PMID: 38443653 DOI: 10.1208/s12249-024-02767-0] [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: 11/26/2023] [Accepted: 02/10/2024] [Indexed: 03/07/2024] Open
Abstract
Chrysin is a natural flavonoid with a wide range of bioactivities. Only a few investigations have assessed the analgesic activity of chrysin. The lipophilicity of chrysin reduces its aqueous solubility and bioavailability. Hence, self-nanoemulsifying drug delivery systems (SNEDDS) were designed to overcome this problem. Kollisolv GTA, Tween 80, and Transcutol HP were selected as oil, surfactant, and cosurfactant, respectively. SNEDDS A, B, and C were prepared, loaded with chrysin (0.1%w/w), and extensively evaluated. The optimized formula (B) encompasses 25% Kollisolv GTA, 18.75% Tween 80, and 56.25% Transcutol HP was further assessed. TEM, in vitro release, and biocompatibility towards the normal oral epithelial cell line (OEC) were estimated. Brain targeting and acetic acid-induced writhing in a mouse model were studied. After testing several adsorbents, powdered SNEDDS B was formulated and evaluated. The surfactant/cosurfactant (S/CoS) ratio of 1:3 w/w was appropriate for the preparation of SNEDDS. Formula B exhibited instant self-emulsification, spherical nanoscaled droplets of 155.4 ± 32.02 nm, and a zeta potential of - 12.5 ± 3.40 mV. The in vitro release proved the superiority of formula B over chrysin suspension (56.16 ± 10.23 and 9.26 ± 1.67%, respectively). The biocompatibility of formula B towards OEC was duplicated (5.69 ± 0.03 µg/mL). The nociceptive pain was mitigated by formula B more efficiently than chrysin suspension as the writhing numbers reduced from 8.33 ± 0.96 to 0 after 60 min of oral administration. Aerosil R972 was selected as an adsorbent, and its chemical compatibility was confirmed. In conclusion, our findings prove the therapeutic efficacy of chrysin self-nanoemulsion as a potential targeting platform to combat pain.
Collapse
Affiliation(s)
- Samar Mohamed Elhoseny
- Department of Pharmaceutics, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Noha Mohamed Saleh
- Department of Pharmaceutics, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| | - Mahasen Mohamed Meshali
- Department of Pharmaceutics, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
8
|
Mohammed NN, Tadros MG, George MY. Empagliflozin repurposing in Parkinson's disease; modulation of oxidative stress, neuroinflammation, AMPK/SIRT-1/PGC-1α, and wnt/β-catenin pathways. Inflammopharmacology 2024; 32:777-794. [PMID: 38038781 PMCID: PMC10907444 DOI: 10.1007/s10787-023-01384-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 10/20/2023] [Indexed: 12/02/2023]
Abstract
Parkinson's disease is a neuroprogressive disorder characterized by loss of dopaminergic neurons in substantia nigra pars compacta. Empagliflozin (EMPA), a SGLT-2 inhibitor, is an oral hypoglycemic agent with reported anti-inflammatory and antioxidant effects. The current study aimed to evaluate the neuroprotective effect of EMPA in rotenone-induced Parkinson's disease. Rats were randomly distributed among five groups as follows: control, rotenone (2 mg/kg), rotenone + EMPA (10 mg/kg), rotenone + EMPA (20 mg/kg), and EMPA (20 mg/kg) groups. They were treated for 30 consecutive days. Rotenone reduced locomotor activity and retention time on the rotarod performance test while elongated descent latency time. On the other side, EMPA corrected these behavioral changes. These results were confirmed by histological examination and number of intact neurons. Moreover, rotenone induced alpha-synuclein accumulation, reduced tyrosine hydroxylase expression, dopamine, 3,4-dihydroxyphenylacetic acid, and homovanillic acid concentrations. On the other side, EMPA reversed such effects induced by rotenone. Depending on previous results, EMPA (20 mg/kg) was selected for further mechanistic studies. Rotenone ameliorated superoxide dismutase and catalase activities and enhanced lipid peroxidation, interleukin-1β, and tumor necrosis factor-α levels. By contrast, EMPA opposed rotenone-induced effects on oxidative stress and inflammation. Besides, rotenone reduced the expression of pAMP-activated protein kinase (pAMPK), peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α), and Sirtuin-1 (SIRT-1), as well as abrogated NAD+/NADH ratio. However, EMPA activated the AMPK/SIRT-1/PGC-1α pathway. Moreover, rotenone hindered the wnt/β-catenin pathway by reducing the wnt-3a level and β-catenin expression. On the other side, EMPA triggered activation of the wnt/β-catenin pathway. Collectively, EMPA may provide a promising solution for Parkinson's patients worldwide.
Collapse
Affiliation(s)
- Noha Nabil Mohammed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo Governorate, 11566, Egypt
| | - Mariane G Tadros
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo Governorate, 11566, Egypt
| | - Mina Y George
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo Governorate, 11566, Egypt.
| |
Collapse
|
9
|
Pai MS, Wang KC, Yeh KC, Wang SJ. Stabilization of mitochondrial function by chlorogenic acid protects against kainic acid-induced seizures and neuronal cell death in rats. Eur J Pharmacol 2023; 961:176197. [PMID: 38000721 DOI: 10.1016/j.ejphar.2023.176197] [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: 07/06/2023] [Revised: 10/21/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023]
Abstract
The current study investigated the effect of chlorogenic acid, a polyphenolic compound found in numerous plant products, on a kainic acid-induced seizure rat model and its potential mechanism. Rats were administered chlorogenic acid (10 and 50 mg/kg) intraperitoneally for 30 min before kainic acid (15 mg/kg) intraperitoneal administration. Pretreatment with chlorogenic acid decreased the seizure score, increased the latency to onset of the first seizure, and decreased the mortality rate. Chlorogenic acid pretreatment also resulted in a significant reduction in glutamate elevation and neuronal death in the hippocampus of kainic acid-treated rats. In addition, electron microscopy revealed that kainic acid-induced changes in hippocampal mitochondrial structure were prevented by chlorogenic acid pretreatment. Additionally, the levels of mitochondrial function-related proteins, including sirtuin 3, Complex I, glutamate dehydrogenase 1 and ATP synthase, were increased, and the level of the mitochondrial damage marker cytochrome C was decreased in the hippocampus of chlorogenic acid/kainic acid rats. Furthermore, the expression of mitochondrial biogenesis-related proteins [AMP-activated protein kinase (AMPK), sirtuin1, and peroxisome proliferator-activated receptor γ-coactivator-1α (PGC-1α)] and mitophagy-related proteins [phosphatase and tensin homolog (PTEN)-induced putative kinase 1 (PINK1), Parkin, and microtubule-associated protein 1 light chain 3 (LC3)] was decreased in the hippocampus of kainic acid-treated rats, which was reversed by chlorogenic acid pretreatment. These observations reveal the marked neuroprotective potential of chlorogenic acid against kainic acid-induced neurotoxicity and seizures through prevention of glutamate increase and preservation of AMPK/sirtuin 1/PGC-1α-mediated mitochondrial biogenesis and PINK1/Parkin-induced mitophagy to maintain adequate mitochondrial homeostasis and function.
Collapse
Affiliation(s)
- Ming-Shang Pai
- Department of Psychiatry, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan; School of Medicine, Fu Jen Catholic University, No.510, Zhongzheng Rd., Xinzhuang Dist, New Taipei City, 24205, Taiwan
| | - Kaw-Chen Wang
- School of Medicine, Fu Jen Catholic University, No.510, Zhongzheng Rd., Xinzhuang Dist, New Taipei City, 24205, Taiwan; Department of Neurology, Cardinal Tien Hospital, New Taipei, Taiwan
| | - Kun-Chieh Yeh
- School of Medicine, Fu Jen Catholic University, No.510, Zhongzheng Rd., Xinzhuang Dist, New Taipei City, 24205, Taiwan; Department of Surgery, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan; Department of Surgery, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Su-Jane Wang
- School of Medicine, Fu Jen Catholic University, No.510, Zhongzheng Rd., Xinzhuang Dist, New Taipei City, 24205, Taiwan; Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan City, Taiwan.
| |
Collapse
|
10
|
Rabie O, El-Nashar HAS, George MY, Majrashi TA, Al-Warhi T, Hassan FE, Eldehna WM, Mostafa NM. Phytochemical profiling and neuroprotective activity of Callistemon subulatus leaves against cyclophosphamide-induced chemobrain. Biomed Pharmacother 2023; 167:115596. [PMID: 37797461 DOI: 10.1016/j.biopha.2023.115596] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/07/2023] Open
Abstract
Cyclophosphamide (CPA) is a chemotherapeutic drug used for various types of cancers. However, patients receiving CPA for long periods suffer cognitive impairment associated with difficulties in learning, decreased concentration, and impaired memory. Chemotherapy-induced cognitive impairment, known as chemobrain, has been attributed to enhanced oxidative stress and inflammatory response. The current study aimed to identify the phytoconstituents of Callistemon subulatus extract (CSE) using HPLC-ESI/MS-MS analysis and evaluate its neuroprotective activity against CPA-induced chemobrain in rats. Fourteen compounds were identified following HPLC analysis including, five phlorglucinols, four flavonol glycosides, a triterpene, and a phenolic acid. Forty rats were divided into five groups treated for ten days as follows; group I (control group), group II received CPA (200 mg/kg, i.p.) on the 7th day, groups III and IV received CSE (200 and 400 mg/kg respectively, orally) for ten days and CPA (200 mg/kg, i.p.) on the 7th day, and group V received only CSE (400 mg/kg, orally) for ten days. The administration of CSE effectively ameliorated the deleterious effects of CPA on spatial and short-term memories, as evidenced by behavioral tests, Y-maze and passive avoidance. Such findings were further confirmed by histological examination. In addition, CSE counteracted the effect of CPA on hippocampal acetylcholinesterase (AChE) activity enhancing the level of acetylcholine. Owing to the CSE antioxidant properties, it hindered the CPA-induced redox imbalance, which is represented by decreased catalase and reduced glutathione levels, as well as enhanced lipid peroxidation. Therefore, CSE may be a promising natural candidate for protection against CPA-induced chemobrain in cancer patients.
Collapse
Affiliation(s)
- Omyma Rabie
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo 11566, Egypt
| | - Heba A S El-Nashar
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo 11566, Egypt
| | - Mina Y George
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566 Cairo, Egypt
| | - Taghreed A Majrashi
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Asir 61421, Saudi Arabia
| | - Tarfah Al-Warhi
- Department of Chemistry, College of Science, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Fatma E Hassan
- Department of Physiology, General Medicine Practice Program, Batterjee Medical College, Jeddah 21442, Saudi Arabia; Medical Physiology Department, Kasr Alainy, Faculty of Medicine, Cairo University, Giza 11562, Egypt
| | - Wagdy M Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, P.O. Box 33516, Kafrelsheikh, Egypt.
| | - Nada M Mostafa
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo 11566, Egypt.
| |
Collapse
|
11
|
Gad SR, El-Gogary RI, George MY, Hathout RM. Nose-to-brain delivery of 18β-Glycyrrhetinic acid using optimized lipid nanocapsules: A novel alternative treatment for Alzheimer's disease. Int J Pharm 2023; 645:123387. [PMID: 37678474 DOI: 10.1016/j.ijpharm.2023.123387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/28/2023] [Accepted: 09/05/2023] [Indexed: 09/09/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder and the most relevant form of dementia affecting people worldwide. AD was reported to be associated with increased oxidative stress ending up with neuronal damage. 18β-Glycyrrhetinic acid (GA), triterpenoid aglycone of glycyrrhizin, was reported for its powerful antioxidant activities. However, its high molecular weight and lipophilicity are two major obstacles that limit its use and cause very low brain bioavailability. The aim of the present study was to formulate the GA in lipid nanocapsules (LNCs) for enhanced nose-to-brain delivery, as well as to elucidate its potential neuroprotective effect in AD. The optimized GA-loaded LNCs exhibited nanometric size range, good stability over 6 months, sustained drug release over 24 h and high steady state flux and permeability coefficient across nasal mucosa over 8 h. In-vivo studies were conducted on five groups; control, scopolamine (SCOP)-treated, SCOP + GA-LNCs, SCOP + oral GA suspension, and SCOP + intranasal GA suspension groups. Intranasal administration of GA-LNCs, at a reduced dose of 1 mg/kg, improved scopolamine-induced memory impairment in rats evidenced by behavioral testing, histological examination, and oxidative stress markers; catalase and superoxide dismutase. Collectively, GA-loaded LNCs (with 50 times lower dose) may provide a promising remedy for AD patients worldwide.
Collapse
Affiliation(s)
- Sara R Gad
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Riham I El-Gogary
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Mina Y George
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566 Cairo, Egypt
| | - Rania M Hathout
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt.
| |
Collapse
|
12
|
Zhuo C, Hu S, Chen G, Yang L, Cai Z, Tian H, Jiang D, Chen C, Wang L, Ma X, Li R. Low-dose lithium adjunct to atypical antipsychotic treatment nearly improved cognitive impairment, deteriorated the gray-matter volume, and decreased the interleukin-6 level in drug-naive patients with first schizophrenia symptoms: a follow-up pilot study. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2023; 9:71. [PMID: 37838729 PMCID: PMC10576794 DOI: 10.1038/s41537-023-00400-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 10/02/2023] [Indexed: 10/16/2023]
Abstract
This study was conducted to investigate the effects of long-term low-dose lithium adjunct to antipsychotic agent use on the cognitive performance, whole-brain gray-matter volume (GMV), and interleukin-6 (IL-6) level in drug-naive patients with first-episode schizophrenia, and to examine relationships among these factors. In this double-blind randomized controlled study, 50 drug-naive patients with first-episode schizophrenia each took low-dose (250 mg/day) lithium and placebo (of the same shape and taste) adjunct to antipsychotic agents (mean, 644.70 ± 105.58 and 677.00 ± 143.33 mg/day chlorpromazine equivalent, respectively) for 24 weeks. At baseline and after treatment completion, the MATRICS Consensus Cognitive Battery (MCCB) was used to assess cognitive performance, 3-T magnetic resonance imaging was performed to assess structural brain alterations, and serum IL-6 levels were quantified by immunoassay. Treatment effects were assessed within and between patient groups. Relationships among cognitive performance, whole-brain GMVs, and the IL-6 level were investigated by partial correlation analysis. Relative to baseline, patients in the lithium group showed improved working memory, verbal learning, processing speed, and reasoning/problem solving after 24 weeks of treatment; those in the placebo group showed only improved working memory and verbal learning. The composite MCCB score did not differ significantly between groups. The whole-brain GMV reduction was significantly lesser in the lithium group than in the placebo group (0.46% vs. 1.03%; P < 0.001). The GMV and IL-6 reduction ratios correlated with each other in both groups (r = -0.17, P = 0.025). In the lithium group, the whole-brain GMV reduction ratio correlated with the working memory improvement ratio (r = -0.15, P = 0.030) and processing speed (r = -0.14, P = 0.036); the IL-6 reduction ratio correlated with the working memory (r = -0.21, P = 0.043) and verbal learning (r = -0.30, P = 0.031) improvement ratios. In the placebo group, the whole-brain GMV reduction ratio correlated only with the working memory improvement ratio (r = -0.24, P = 0.019); the IL-6 reduction ratio correlated with the working memory (r = -0.17, P = 0.022) and verbal learning (r = -0.15, P = 0.011) improvement ratios. Both treatments implemented in this study nearly improved the cognitive performance of patients with schizophrenia; relative to placebo, low-dose lithium had slightly greater effects on several aspects of cognition. The patterns of correlation among GMV reduction, IL-6 reduction, and cognitive performance improvement differed between groups.
Collapse
Affiliation(s)
- Chuanjun Zhuo
- Key Laboratory of Sensor Information Processing Abnormalities in Schizophrenia (SIPAS-Lab), Tianjin Fourth Center Hospital, Nankai University Affiliated Tianjin Fourth Center Hospital, Tianjin Medical University Affiliated Tianjin Fourth Center Hospital, Tianjin, 300140, China.
- Department of Psychiatry, Wenzhou Seventh Peoples Hospital, Wenzhou, 325000, China.
- Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin Anding Hospital, Tianjin, 300222, China.
| | - Shuiqing Hu
- Key Laboratory of Sensor Information Processing Abnormalities in Schizophrenia (SIPAS-Lab), Tianjin Fourth Center Hospital, Nankai University Affiliated Tianjin Fourth Center Hospital, Tianjin Medical University Affiliated Tianjin Fourth Center Hospital, Tianjin, 300140, China
| | - Guangdong Chen
- Department of Psychiatry, Wenzhou Seventh Peoples Hospital, Wenzhou, 325000, China
| | - Lei Yang
- Key Laboratory of Sensor Information Processing Abnormalities in Schizophrenia (SIPAS-Lab), Tianjin Fourth Center Hospital, Nankai University Affiliated Tianjin Fourth Center Hospital, Tianjin Medical University Affiliated Tianjin Fourth Center Hospital, Tianjin, 300140, China
| | - Ziyao Cai
- Department of Psychiatry, Wenzhou Seventh Peoples Hospital, Wenzhou, 325000, China
| | - Hongjun Tian
- Key Laboratory of Sensor Information Processing Abnormalities in Schizophrenia (SIPAS-Lab), Tianjin Fourth Center Hospital, Nankai University Affiliated Tianjin Fourth Center Hospital, Tianjin Medical University Affiliated Tianjin Fourth Center Hospital, Tianjin, 300140, China
| | - Deguo Jiang
- Department of Psychiatry, Wenzhou Seventh Peoples Hospital, Wenzhou, 325000, China
| | - Chunmian Chen
- Department of Psychiatry, Wenzhou Seventh Peoples Hospital, Wenzhou, 325000, China
| | - Lina Wang
- Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin Anding Hospital, Tianjin, 300222, China
| | - Xiaoyan Ma
- Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin Anding Hospital, Tianjin, 300222, China
| | - Ranli Li
- Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin Anding Hospital, Tianjin, 300222, China
| |
Collapse
|
13
|
Awad HH, Desouky MA, Zidan A, Bassem M, Qasem A, Farouk M, AlDeab H, Fouad M, Hany C, Basem N, Nader R, Alkalleny A, Reda V, George MY. Neuromodulatory effect of vardenafil on aluminium chloride/D-galactose induced Alzheimer's disease in rats: emphasis on amyloid-beta, p-tau, PI3K/Akt/p53 pathway, endoplasmic reticulum stress, and cellular senescence. Inflammopharmacology 2023; 31:2653-2673. [PMID: 37460908 PMCID: PMC10518298 DOI: 10.1007/s10787-023-01287-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 06/27/2023] [Indexed: 09/26/2023]
Abstract
Dysregulation of protein homeostasis, proteostasis, is a distinctive hallmark of many neurodegenerative disorders and aging. Deleteriously, the accumulation of aberrant proteins in Alzheimer's disease (AD) is accompanied with a marked collapse in proteostasis network. The current study explored the potential therapeutic effect of vardenafil (VAR), a phosphodiesterase-5 inhibitor, in AlCl3/D-galactose (D-gal)-induced AD in rats and its possible underlying mechanisms. The impact of VAR treatment on neurobehavioral function, hippocampal tissue architecture, and the activity of the cholinergic system main enzymes were assessed utilizing VAR at doses of 0.3 mg/kg and 1 mg/kg. Additionally, the expression level of amyloid-beta and phosphorylated tau proteins in the hippocampus were figured out. Accordingly, VAR higher dose was selected to contemplate the possible underlying mechanisms. Intriguingly, VAR elevated the cyclic guanosine monophosphate level in the hippocampus and averted the repressed proteasome activity by AlCl3/D-gal; hence, VAR might alleviate the burden of toxic protein aggregates in AD. In addition, a substantial reduction in the activating transcription factor 6-mediated endoplasmic reticulum stress was demonstrated with VAR treatment. Notably, VAR counteracted the AlCl3/D-gal-induced depletion of nuclear factor erythroid 2-related factor 2 level. Moreover, the anti-senescence activity of VAR was demonstrated via its ability to restore the balance of the redox circuit. The modulation of phosphatidylinositol-3-kinase/protein kinase B/p53 pathway and the reduction of nuclear factor kappa B level, the key regulator of senescence-associated secretory phenotype mediators release, with VAR treatment were also elucidated. Altogether, these findings insinuate the possible therapeutic benefits of VAR in AD management.
Collapse
Affiliation(s)
- Heba H Awad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA University), Cairo, Egypt
| | - Mahmoud A Desouky
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Alaa Zidan
- Drug Design Program, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mariam Bassem
- Drug Design Program, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Amaal Qasem
- Drug Design Program, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mona Farouk
- Drug Design Program, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Haidy AlDeab
- Drug Design Program, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Miral Fouad
- Drug Design Program, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Cherry Hany
- Drug Design Program, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Nada Basem
- Drug Design Program, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Rita Nader
- Drug Design Program, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Ashrakat Alkalleny
- Drug Design Program, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Verina Reda
- Drug Design Program, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mina Y George
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt.
| |
Collapse
|
14
|
Hanna DMF, Youshia J, Fahmy SF, George MY. Nose to brain delivery of naringin-loaded chitosan nanoparticles for potential use in oxaliplatin-induced chemobrain in rats: impact on oxidative stress, cGAS/STING and HMGB1/RAGE/TLR2/MYD88 inflammatory axes. Expert Opin Drug Deliv 2023; 20:1859-1873. [PMID: 37357778 DOI: 10.1080/17425247.2023.2228685] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/26/2023] [Indexed: 06/27/2023]
Abstract
OBJECTIVES Oxaliplatin induces chemobrain in cancer patients/survivors. Nutraceutical naringin has antioxidant and anti-inflammatory properties with low oral bioavailability. Our aim was to formulate naringin in chitosan nanoparticles for nose to brain delivery and assess its neuroprotective effect against oxaliplatin-induced chemobrain in rats. METHODS Naringin chitosan nanoparticles were prepared by ionic gelation. Rats were administered oral naringin (80 mg/kg), intranasal naringin (0.3 mg/kg) or intranasal naringin-loaded chitosan nanoparticles (0.3 mg/kg). Naringin's neuroprotective efficacy was assessed based on behavioral tests, histopathology, and measuring oxidative stress and inflammatory markers. RESULTS Selected nanoparticles formulation showed drug loading of 5%, size of 150 nm and were cationic. Intranasal naringin administration enhanced memory function, inhibited hippocampal acetylcholinesterase activity, and corrected oxaliplatin-induced histological changes. Moreover, it reduced malondialdehyde and elevated reduced glutathione hippocampal levels. Furthermore, it decreased levels of inflammatory markers: NF-kB and TNF-α by 1.25-fold. Upstream to this inflammatory status, intranasal naringin downregulated the hippocampal protein levels of two pathways: cGAS/STING and HMGB1/RAGE/TLR2/MYD88. CONCLUSION Intranasal naringin-loaded chitosan nanoparticles showed superior amelioration of oxaliplatin-induced chemobrain in rats at a dose 267-fold lower to that administered orally. The potential involvement of cGAS/STING and HMGB1/RAGE/TLR2/MYD88 pathways in the mechanistic process of either oxaliplatin-induced chemobrain or naringin-mediated neuroprotection was evidenced.
Collapse
Affiliation(s)
- Diana M F Hanna
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - John Youshia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Sarah Farid Fahmy
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mina Y George
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|