1
|
Solmaz M, Erdogan E, Dasdelen D, Mogulkoc R, Vatansev H, Akyurek F, Ozbek H. Comparison of the protective effects of silymarin and thymoquinone in the focal cerebral ischemia-reperfusion rat model. Biotech Histochem 2024:1-18. [PMID: 39508143 DOI: 10.1080/10520295.2024.2421511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024] Open
Abstract
Silymarin and thymoquinone exert neuroprotective effects, although their combined effects in focal cerebral ischemia/reperfusion (I/R) models are unknown. We compared the effect of silymarin and thymoquinone in an I/R rat model. Wistar rats were divided into five groups: SHAM, REP (I/R), SIR (200 mg/kg silymarin+I/R), TIR (3 mg/kg thymoquinone+I/R), and STIR (200 mg/kg silymarin+3-mg thymoquinone+I/R). The rats underwent bilateral carotid artery occlusion for 30 min and neurological assessments 24 h thereafter. Apoptosis was evaluated using anti-caspase-3 and terminal deoxynucleotidyl transferase biotin-dUTP nick end labeling (TUNEL) assays. Astrocyte activation was determined using an anti-GFAP antibody. Total antioxidant status (TAS), total oxidant status (TOS), and trimethylamine N-oxide (TMAO) levels were measured. SHAM and REP rats had the lowest and highest neurological scores, respectively (p = 0.001). REP rats showed greater deterioration than SIR, TIR, and STIR rats. SIR, TIR, and STIR rats had fewer TUNEL and caspase-3-positive cells than REP rats (p<0.05). GFAP expression was higher in REP rats (p<0.05) than in SIR, TIR, and STIR rats (p<0.05). SIR and TIR rats showed higher TAS than REP rats (p<0.05). SIR, TIR, and STIR rats had lower TMAO values than REP and SHAM rats (p<0.05). Silymarin/thymoquinone reduces impairment, apoptosis, and astrocyte activation. Combination therapy reduces TMAO levels.
Collapse
Affiliation(s)
- Merve Solmaz
- Department of Histology and Embryology, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Ender Erdogan
- Department of Histology and Embryology, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Dervis Dasdelen
- Department of Physiology, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Rasim Mogulkoc
- Department of Physiology, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Husamettin Vatansev
- Department of Medical Biochemistry, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Fikret Akyurek
- Department of Medical Biochemistry, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Hanefi Ozbek
- Department of Pharmacology, Faculty of Medicine, Izmir Bakırcay University, Izmir, Turkey
| |
Collapse
|
2
|
Borikar SP, Chitode GV, Tapre DN, Lokwani DK, Jain SP. Empagliflozin ameliorates olfactory bulbectomy-induced depression by mitigating oxidative stress and possible involvement of brain derived neurotrophic factor in diabetic rats. Int J Neurosci 2024:1-17. [PMID: 39392472 DOI: 10.1080/00207454.2024.2414270] [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: 06/29/2024] [Revised: 09/03/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024]
Abstract
Empagliflozin, a sodium-glucose co-transporter 2 (SGLT2) inhibitor, has recently reported to prevent the depression in chronic animal model. The present study aimed to explore the antidepressant potential of empagliflozin using a neuroinflammation-mediated depression involving the olfactory bulbectomy (OBX) model in diabetic rats. A low dose of streptozotocin was injected to induce diabetes in all group of animals. Following the confirmation of hyperglycemia, OBX surgery was performed. Post-surgery, the drug treatments were administered orally for 14 consecutive days. The study evaluated the effects of daily oral administration of empagliflozin at doses of 5 and 10 mg/kg, alongside metformin (200 mg/kg) and clomipramine (50 mg/kg), on OBX-induced behavioral depression in rats. Separate sham and vehicle control groups were also maintained. Behavioral parameters in open field, forced swim test, elevated plus maze and splash test were recorded on 28th day. Results showed that empagliflozin, at the higher dose, significantly enhanced behavioral outcomes, evidenced by increased distance travelled, greater open arm entries, and reduced immobility, alongside a notable reduction in grooming time. Moreover, empagliflozin significantly restored the antioxidants level specifically Glutathione (GSH) and Catalase (CAT) in OBX insulted rat brain and decreased Lipid peroxidase (LPO). Notably, molecular docking study demonstrated a good binding affinity of empagliflozin for Brain-Derived Neurotrophic Factor (BDNF), suggesting that its antidepressant effects may be mediated through the modulation of the BDNF pathway. These findings support the potential therapeutic application of empagliflozin for depression, particularly in cases associated with neuroinflammation and oxidative stress.
Collapse
Affiliation(s)
- Sachin P Borikar
- Department of Pharmacology, Rajarshi Shahu College of Pharmacy, Buldana, India
| | - Gaurav V Chitode
- Department of Pharmacology, Rajarshi Shahu College of Pharmacy, Buldana, India
| | - Deepali N Tapre
- Department of Pharmaceutical Chemistry, Rajarshi Shahu College of Pharmacy, Buldana, India
| | - Deepak K Lokwani
- Department of Pharmaceutical Chemistry, Rajarshi Shahu College of Pharmacy, Buldana, India
| | - Shirish P Jain
- Department of Pharmacology, Rajarshi Shahu College of Pharmacy, Buldana, India
| |
Collapse
|
3
|
Ashique S, Mohanto S, Kumar N, Nag S, Mishra A, Biswas A, Rihan M, Srivastava S, Bhowmick M, Taghizadeh-Hesary F. Unlocking the possibilities of therapeutic potential of silymarin and silibinin against neurodegenerative Diseases-A mechanistic overview. Eur J Pharmacol 2024; 981:176906. [PMID: 39154829 DOI: 10.1016/j.ejphar.2024.176906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 07/28/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
Silymarin, a bioflavonoid derived from the Silybum marianum plant, was discovered in 1960. It contains C25 and has been extensively used as a therapeutic agent against liver-related diseases caused by alcohol addiction, acute viral hepatitis, and toxins-inducing liver failure. Its efficacy stems from its role as a potent anti-oxidant and scavenger of free radicals, employed through various mechanisms. Additionally, silymarin or silybin possesses immunomodulatory characteristics, impacting immune-enhancing and immune-suppressive functions. Recently, silymarin has been recognized as a potential neuroprotective therapy for various neurological conditions, including Parkinson's and Alzheimer's diseases, along with conditions related to cerebral ischemia. Its hepatoprotective qualities, primarily due to its anti-oxidant and tissue-regenerating properties, are well-established. Silymarin also enhances health by modifying processes such as inflammation, β-amyloid accumulation, cellular estrogenic receptor mediation, and apoptotic machinery. While believed to reduce oxidative stress and support neuroprotective mechanisms, these effects represent just one aspect of the compound's multifaceted protective action. This review article further delves into the possibilities of potential therapeutic advancement of silymarin and silibinin for the management of neurodegenerative disorders via mechanics modules.
Collapse
Affiliation(s)
- Sumel Ashique
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India; Department of Pharmaceutics, Bengal College of Pharmaceutical Sciences & Research, Durgapur, 713212, West Bengal, India.
| | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to Be University), Mangalore, Karnataka, 575018, India.
| | - Nitish Kumar
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology (Deemed to Be University), Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh, 201204, India
| | - Sagnik Nag
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia.
| | - Anuradha Mishra
- Amity Institute of Pharmacy, Amity University Lucknow Campus, Uttar Pradesh, 226010, India
| | - Aritra Biswas
- Department of Microbiology, Ramakrishna Mission Vivekananda Centenary College, Rahara Akhil Mukherjee Road, Khardaha, West Bengal, 700118, India; UNESCO Regional Centre for Biotechnology, Department of Biotechnology, Government of India, NCR Biotech Science Cluster, Faridabad, 121001, Haryana, India.
| | - Mohd Rihan
- Department of Pharmacology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab, 160062, India
| | - Shriyansh Srivastava
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, 203201, India; Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), Sector 3 Pushp Vihar, New Delhi, 110017, India
| | - Mithun Bhowmick
- Department of Pharmaceutics, Bengal College of Pharmaceutical Sciences & Research, Durgapur, 713212, West Bengal, India
| | - Farzad Taghizadeh-Hesary
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Aliaghaei A, Meftahi GH. Silymarin ameliorates motor function and averts neuroinflammation-induced cell death in the rat model of Huntington's disease. Brain Res Bull 2024; 216:111039. [PMID: 39089590 DOI: 10.1016/j.brainresbull.2024.111039] [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: 05/18/2024] [Revised: 07/20/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
Huntington's disease (HD) is a scarce neurodegenerative disorder defined by chorea (unusual involuntary movements), behavioral presentations, psychiatric features, and cognitive deterioration. Although the precise pathogenic mechanism behind HD has not yet been identified, the most widely acknowledged pathways include excitotoxicity, mitochondrial malfunction, neuroinflammation, neurochemical imbalance, oxidative stress, and apoptosis HD has no efficient therapy. Current medications have drawbacks. Silymarin, a compound made up of standardized extracts obtained from the seeds of the Silybum marianum and polyphenolic flavonolignan, is utilized in therapeutic settings to treat a variety of experimental disorders in animals. Silymarin's key pharmacological activities include anti-cancer, hepatoprotection, antioxidant, cardioprotection, and anti-inflammatory. It also has no adverse side effects on people or animals. The current study aims to provide Silymarin's neuro-pharmacological activities or therapeutic qualities in HD. In this study, Thirty-six male Sprague-Dawley rats (200-220 g, 8 weeks) at the initial of the study were used. Silymarin solution (100 mg/Kg) was administered by oral gavage for 21 days to ameliorate neural damage in rats injected with 3-nitropropionicacid (3-NP) in a preliminary rat model of HD. The results showed that administration of silymarin to HD rats reduced gliosis, improved motor coordination and muscle activity, and increased striatal volume and the number of neurons and glial cells. Our results suggest that silymarin provides a protective environment for nerve cells and can have beneficial effects against the harmful effects of HD.
Collapse
Affiliation(s)
- Abbas Aliaghaei
- Hearing Disorders Research Center, Loghman‑Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Gholam Hossein Meftahi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Rudrala LC, Challa RR, Subramanyam S, Ayyappa Gouru S, Singh G, Sirisha Mulukuri NVL, Pasala PK, Dintakurthi PSNBK, Gajula S, Rudrapal M. Cerebroprotective Potential of Andrographolide Nanoparticles: In silico and In vivo Investigations. Drug Res (Stuttg) 2024; 74:335-346. [PMID: 38991529 DOI: 10.1055/a-2345-5396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
Ischemic stroke remains the leading cause of death and disability, while the main mechanisms of dominant neurological damage in stroke contain oxidative stress and inflammation. Docking studies revealed a binding energy of - 6.1 kcal/mol for AG, while the co-crystallized ligand (CCl) exhibited a binding energy of - 7.3 kcal/mol with NOS. AG demonstrated favourable hydrogen bond interactions with amino acids ASN A:354 and ARG A:388 and hydrophobic interactions with GLU A:377. Molecular dynamics simulations throughout 100 ns indicated a binding affinity of - 27.65±2.88 kcal/mol for AG, compared to - 18.01±4.02 kcal/mol for CCl. These findings suggest that AG possesses a superior binding affinity for NOS compared to CCl, thus complementing the stability of NOS at the docked site.AG has limited applications owing to its low bioavailability, poor water solubility, and high chemical and metabolic instability.The fabrication method was employed in the preparation of AGNP, SEM analysis confirmed spherical shape with size in 19.4±5 nm and investigated the neuroprotective effect in cerebral stroke rats induced by 30 min of carotid artery occlusion followed by 4 hr reperfusion, evaluated by infarction size, ROS/RNS via GSH, MPO, NO estimationand AchE activity, and monitoring EEG function. Cortex and hippocampal histology were compared between groups. AGNP treatment significantly decreased Infarction size and increased GSH levels (p<0.01**), decreased MPO (p<0.01**), NO (p<0.01**), AchE (p<0.01**), restored to normal EEG amplitude, minimizing unsynchronized polyspikes and histological data revealed that increased pyramidal cell layer thickness and decreased apoptotic neurons in hippocampus, cortex appeared normal neurons with central large vesicular nuclei, containing one or more nucleoli in compared to AG treatment. Based on brain biochemical, histopathology reports AGNP exhibited significant cerebroprotective activity compared to AG on ischemic rats.
Collapse
Affiliation(s)
- Lakshmi Charitha Rudrala
- Department of Pharmacology, SKU College of Pharmaceutical Sciences, S. K. University, Anantapur, India
| | | | - Sibbala Subramanyam
- Department of Pharmaceutical Sciences, School of Biotechnology and Pharmaceutical Sciences, Vignan's Foundation for Science, Technology & Research (Deemed to be University), Guntur, India
| | | | - Gagandeep Singh
- Section of Microbiology, Central Ayurveda Research Institute, Jhansi, Uttar Pradesh, India
| | | | - Praveen Kumar Pasala
- Department of Pharmacology, Raghavendra Institute of Pharmaceutical Education and Research, JNTUA, Anantapur, India
| | | | - Somasekhar Gajula
- Department of Pharmacology, SKU College of Pharmaceutical Sciences, S. K. University, Anantapur, India
| | - Mithun Rudrapal
- Department of Pharmaceutical Sciences, School of Biotechnology and Pharmaceutical Sciences, Vignan's Foundation for Science, Technology & Research (Deemed to be University), Guntur, India
| |
Collapse
|
6
|
Niknahad H, Mobasheri A, Arjmand A, Rafiei E, Alidaee S, Razavi H, Bagheri S, Rezaei H, Sabouri S, Najibi A, Khodaei F, Kashani SMA, Ommati MM, Heidari R. Hepatic encephalopathy complications are diminished by piracetam via the interaction between mitochondrial function, oxidative stress, inflammatory response, and locomotor activity. Heliyon 2023; 9:e20557. [PMID: 37810869 PMCID: PMC10551565 DOI: 10.1016/j.heliyon.2023.e20557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 09/01/2023] [Accepted: 09/28/2023] [Indexed: 10/10/2023] Open
Abstract
Background of the study: Hepatic encephalopathy (HE) is a complication in which brain ammonia (NH4+) levels reach critically high concentrations because of liver failure. HE could lead to a range of neurological complications from locomotor and behavioral disturbances to coma. Several tactics have been established for subsiding blood and brain NH4+. However, there is no precise intervention to mitigate the direct neurological complications of NH4+. Purpose It has been found that oxidative stress, mitochondrial damage, and neuro-inflammation play a fundamental role in NH4+ neurotoxicity. Piracetam is a drug used clinically in neurological complications such as stroke and head trauma. Piracetam could significantly diminish oxidative stress and improve brain mitochondrial function. Research methods In the current study, piracetam (100 and 500 mg/kg, oral) was used in a mice model of HE induced by thioacetamide (TA, 800 mg/kg, single dose, i.p). Results Significant disturbances in animals' locomotor activity, along with increased oxidative stress biomarkers, including reactive oxygen species formation, protein carbonylation, lipid peroxidation, depleted tissue glutathione, and decreased antioxidant capacity, were evident in the brain of TA-treated mice. Meanwhile, mitochondrial permeabilization, mitochondrial depolarization, suppression of dehydrogenases activity, and decreased ATP levels were found in the brain of the TA group. The level of pro-inflammatory cytokines was also significantly high in the brain of HE animals. Conclusion It was found that piracetam significantly enhanced mice's locomotor activity, blunted oxidative stress biomarkers, decreased inflammatory cytokines, and improved mitochondrial indices in hyperammonemic mice. These data suggest piracetam as a neuroprotective agent which could be repurposed for the management of HE.
Collapse
Affiliation(s)
- Hossein Niknahad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mobasheri
- Research Unit of Medical Imaging, Physics, And Technology, Faculty of Medicine, University of Oulu, FI-90014, Oulu, Finland
- University Medical Center Utrecht, Departments of Orthopedics Rheumatology and Clinical Immunology, 3508, GA, Utrecht, the Netherlands
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406, Vilnius, Lithuania
| | - Abdollah Arjmand
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elahe Rafiei
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sepideh Alidaee
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hadi Razavi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sara Bagheri
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Heresh Rezaei
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Samira Sabouri
- Shanxi Key Laboratory of Ecological, Animal Sciences, And Environmental Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Asma Najibi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Forouzan Khodaei
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyyed Mohammad Amin Kashani
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Mehdi Ommati
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Shanxi Key Laboratory of Ecological, Animal Sciences, And Environmental Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
- Henan Key Laboratory of Environmental and Animal Product Safety, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
7
|
Ranjan S, Gautam A. Pharmaceutical prospects of Silymarin for the treatment of neurological patients: an updated insight. Front Neurosci 2023; 17:1159806. [PMID: 37274201 PMCID: PMC10232807 DOI: 10.3389/fnins.2023.1159806] [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: 02/06/2023] [Accepted: 04/17/2023] [Indexed: 06/06/2023] Open
Abstract
Background Silymarin is a polyphenolic flavonoid complex extricated from dried fruits and seeds of the plant Silybum marianum L. Chemically, it is a mixture of flavonolignan complexes consisting of silybin, isosilybin, silychristin, silydianin, a minor quantity of taxifolin, and other polyphenolic compounds, which possess different bio medicinal values. Purpose This review critically looks into the current status, pharmaceutical prospects and limitations of the clinical application of Silymarin for treating neurological disorders. In particular, Silymarin's medicinal properties and molecular mechanisms are focused on providing a better-compiled understanding helpful in its neuro-pharmacological or therapeutic aspects. Methods This review was compiled by the literature search done using three databases, i.e., PubMed (Medline), EMBASE and Science Direct, up to January 2023, using the keywords-Silymarin, neurological disorders, cognitive disorders, Type 2 Diabetes, pharmaceutical prospects and treatment. Then, potentially relevant publications and studies (matching the eligible criteria) were retrieved and selected to explain in this review using PRISMA 2020 (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) study flow chart. Result Since its discovery, it has been widely studied as a hepatoprotective drug for various liver disorders. However, in the last 10-15 years, several research studies have shown its putative neuroprotective nature against various brain disorders, including psychiatric, neurodegenerative, cognitive, metabolic and other neurological disorders. The main underlying neuroprotective mechanisms in preventing and curing such disorders are the antioxidant, anti-inflammatory, anti-apoptotic, pro-neurotrophic and pro-estrogenic nature of the bioactive molecules. Conclusion This review provides a lucid summary of the well-studied neuroprotective effects of Silymarin, its underlying molecular mechanisms and current limitations for its usage during neurological disorders. Finally, we have suggested a future course of action for developing it as a novel herbal drug for the treatment of brain diseases.
Collapse
Affiliation(s)
- Shovit Ranjan
- University Department of Zoology, Kolhan University, Chaibasa, Jharkhand, India
| | - Akash Gautam
- Center for Neural and Cognitive Sciences, University of Hyderabad, Hyderabad, India
| |
Collapse
|
8
|
Singh A, Ujjwal RR, Kumar A, Verma RK, Shukla R. Formulation and Optimization of Silymarin Encapsulated Binary Micelles for Enhanced Amyloid Disaggregation Activity. Drug Dev Ind Pharm 2022; 47:1775-1785. [PMID: 35343354 DOI: 10.1080/03639045.2022.2059498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Purpose-Silymarin (SLY) is natural hydrophobic polyphenol which possess antioxidant and amyloid fibril (Aβ1-42) inhibition activity, but its activity hinders due to low aqueous solubility. In this study, SLY is encapsulated in Binary micelle (SLY-BM) that have been utilized to enhance the Aβ1-42 fibril disaggregation. To enhance the aqueous solubility and SLY payload in micelles were optimized using Box Behnken Design (BBD) to increase the efficiency of Aβ1-42 fibril disaggregation. BBD was employed to investigate the effect of ratio of Solutol HS15: Poloxamer-188, amount of acetone and hydration volume on critical quality attributes (CQA), particle size and entrapment efficiency for SLY-BM. Further SLY-BM was characterized for its physical and drug release properties. The Aβ1-42 fibril disaggregation and antioxidant studies was monitored using spectroscopic and microscopic techniques. BBD optimized the particle size <50 nm with % EE >80% and solubility factor of SLY-BM was enhanced to 460 folds than free SLY. Inhibitory concentration 50% (IC50) value of SLY-BM was (19.67 µg/mL) compared to free SLY (30.06 µg/mL) in diphenylpicrahydrazyl (DPPH) assay. SLY-BM increased the Aβ 1-42 disaggregation compared to free SLY observed via thioflavin -T (ThT) assay, photon correlation spectroscopy (PCS), and Circular dichorism (CD). Further morphological evaluation of Aβ1-42 disaggregation was monitored microscopy which showed SLY-BM disaggregated the fibrils in 48h. According to our findings, we concluded that SLY-BM micelles potential candidates for delivery of neuroprotective agents.
Collapse
Affiliation(s)
- Ajit Singh
- Department of pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER-Raebareli), Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow (UP)-226002, India
| | - Rewati Raman Ujjwal
- Department of pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER-Raebareli), Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow (UP)-226002, India
| | - Ashish Kumar
- Department of pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER-Raebareli), Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow (UP)-226002, India
| | - Rahul K Verma
- Institute of Nano Science and Technology (INST), Phase X, Sector 64. Mohali, Punjab 160062, INDIA
| | - Rahul Shukla
- Department of pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER-Raebareli), Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow (UP)-226002, India
| |
Collapse
|
9
|
Thakare VN, Lakade SH, Mahajan MP, Kulkarni YP, Dhakane VD, Harde MT, Patel BM. Protocatechuic acid attenuates chronic unpredictable mild stress induced-behavioral and biochemical alterations in mice. Eur J Pharmacol 2021; 898:173992. [PMID: 33675783 DOI: 10.1016/j.ejphar.2021.173992] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 02/22/2021] [Accepted: 03/01/2021] [Indexed: 12/28/2022]
Abstract
Amelioration of oxidative stress via promoting the endogenous antioxidant system and enhancement of monoamines in brain were the important underlying antidepressant mechanism of protocatechuic acid (PCA). The aim of the present study is to explore the potential antidepressant mechanism(s) PCA in chronic unpredictable mild stress (CUMS) mice. Mice were subjected to CUMS protocol for 4 weeks, and administered with PCA (100 and 200 mg/kg) and fluoxetine (20 mg/kg) for 24 days (from day 8th to 31st). Behavioral (sucrose preference, immobility time, exploratory behavior), and biochemical alterations such as serum corticosterone, brain derived neurotrophic factor (BDNF), inflammatory cytokines, tumor necrosis factor- α (TNF-α), interleukin-6 (IL-6), and antioxidants parameters were investigated. Experimental findings revealed that CUMS subjected mice exhibited significant impairment in behavioral alterations, such as increased immobility time, impaired preference to the sucrose solution, BDNF levels and, serum corticosterone, cytokines, malondialdehyde (MDA) formation with impaired antioxidants in the hippocampus and cerebral cortex. Administration of PCA to CUMS mice attenuated the immobility time, serum corticosterone, cytokines TNF-α, and IL-6, MDA formation and improved sucrose preference, including restoration of BDNF level. Thus, the present findings demonstrated the antidepressant potential of PCA which is largely achieved probably through maintaining BDNF level, and by modulation of the oxidative stress response, cytokines systems, and antioxidant defense system in mice.
Collapse
Affiliation(s)
- Vishnu N Thakare
- Department of Pharmacology, Sinhgad Institute of Pharmaceutical Sciences, Lonavala, Pune, 410401, India; Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, 382 481, Gujarat, India
| | - Sameer H Lakade
- Department of Pharmacology, Sinhgad Institute of Pharmaceutical Sciences, Lonavala, Pune, 410401, India; RMD Institute of Pharmaceutical Education & Research, Pune, 411019, Maharashtra, India
| | - Moreshwar P Mahajan
- Department of Pharmacology, Sinhgad Institute of Pharmaceutical Sciences, Lonavala, Pune, 410401, India
| | - Yogesh P Kulkarni
- Department of Pharmacology, Sinhgad Institute of Pharmaceutical Sciences, Lonavala, Pune, 410401, India
| | - Valmik D Dhakane
- Research & Development, Astec Life Sciences, Mumbai, 421203, India
| | - Minal T Harde
- Department of Pharmaceutical Chemistry, PES's Modern College of Pharmacy, Nigdi, Pune, 411044, India
| | - Bhoomika M Patel
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, 382 481, Gujarat, India.
| |
Collapse
|
10
|
Marmouzi I, Bouyahya A, Ezzat SM, El Jemli M, Kharbach M. The food plant Silybum marianum (L.) Gaertn.: Phytochemistry, Ethnopharmacology and clinical evidence. JOURNAL OF ETHNOPHARMACOLOGY 2021; 265:113303. [PMID: 32877720 DOI: 10.1016/j.jep.2020.113303] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 08/03/2020] [Accepted: 08/21/2020] [Indexed: 05/26/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Silybum marianum (L.) Gaertn. or Milk thistle is a medicinal plant native to Northern Africa, Southern Europe, Southern Russia and Anatolia. It also grows in South Australia, North and South America. In traditional knowledge, people have used S. marianum for liver disorders such as hepatitis, liver cirrhosis and gallbladder diseases. The main active compound of the plant seeds is silymarin, which is the most commonly used herbal supplement in the United States for liver problems. Nowadays, S. marianum products are available as capsules, powders, and extracts. AIM OF STUDY The aim of our study is to draw a more comprehensive overview of the traditional heritage, pharmacological benefits and chemical fingerprint of S. marianum extracts and metabolites; as well as their metabolism and bioavailability. MATERIALS AND METHODS An extensive literature search has been conducted using relavant keywords and papers with rationale methodology and robust data were selected and discussed. Studies involving S. marianum or its main active ingredients with regards to hepatoprotective, antidiabetic, cardiovascular protection, anticancer and antimicrobial activities as well as the clinical trials performed on the plant, were discussed here. RESULTS S. marianum was subjected to thousands of ethnopharmacological, experimental and clinical investigations. Although, the plant is available for use as a dietary supplement, the FDA did not yet approve its use for cancer therapy. Nowadays, clinical investigations are in progress where a global evidence of its real efficiency is needed. CONCLUSION S. marianum is a worldwide used herb with unlimited number of investigations focusing on its benefits and properties, however, little is known about its clinical efficiency. Moreover, few studies have discussed its metabolism, pharmacokinetics and bioavailability, so that all future studies on S. marianum should focus on such areas.
Collapse
Affiliation(s)
- Ilias Marmouzi
- Laboratory of Pharmacology and Toxicology, Faculty of Medicine and Pharmacy, University Mohammed V in Rabat, Rabat, Morocco
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathology Biology, Faculty of Sciences, Department of Biology, Genomic Center of Human Pathology, Mohammed V University in Rabat, Morocco
| | - Shahira M Ezzat
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Kasr El-Ainy Street, Cairo, 11562, Egypt; Department of Pharmacognosy, Faculty of Pharmacy, October University for Modern Science and Arts (MSA), Giza, 12451, Egypt.
| | - Meryem El Jemli
- Laboratory of Pharmacology and Toxicology, Faculty of Medicine and Pharmacy, University Mohammed V in Rabat, Rabat, Morocco
| | - Mourad Kharbach
- Laboratory of Pharmacology and Toxicology, Faculty of Medicine and Pharmacy, University Mohammed V in Rabat, Rabat, Morocco; Department of Analytical Chemistry, Applied Chemometrics and Molecular Modelling, CePhaR, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, B-1090, Brussels, Belgium
| |
Collapse
|
11
|
Mokhtari Sangdehi SR, Hajizadeh Moghaddam A, Ranjbar M. Anti-apoptotic effect of silymarin-loaded chitosan nanoparticles on hippocampal caspase-3 and Bcl-2 expression following cerebral ischemia/reperfusion injury. Int J Neurosci 2021; 132:1102-1109. [PMID: 33287594 DOI: 10.1080/00207454.2020.1860971] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Cerebral ischemia/reperfusion (I/R) causes memory and learning impairments and apoptosis in the hippocampus. The aim of present study aimed to investigate the anti-apoptotic effects of silymarin-loaded chitosan nanoparticles (SM-CS-NPs) on the expression of Bcl-2 and Caspase-3 genes in hippocampal neurons after I/R injury. MATERIAL AND METHODS SM and SM-CS-NPs were orally administered (15 mg/kg) for 14 days, and then cerebral I/R injury was induced by the bilateral common carotid artery occlusion (BCCAO). One day after I/R induction, memory and learning impairments and various biochemical estimations were assessed. RESULTS Our results indicated that SM-CS-NPs improved I/R-induced memory and learning impairments and oxidative damage in the hippocampal region. The qRT-PCR analysis indicated that SM-CS-NPs pretreatment inhibited I/R-induced neuronal apoptosis by increasing the expression of Bcl-2 and decreasing the expression of Caspase-3 in the hippocampus. CONCLUSION These findings suggest that SM-CS-NPs exert neuroprotective effects, and the neuroprotection is likely to be associated with the regulation of Bcl-2 and Caspase-3, leading to inhibition of apoptotic cell death in hippocampal neurons.
Collapse
Affiliation(s)
| | | | - Mojtaba Ranjbar
- Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran
| |
Collapse
|
12
|
Sharma T, Airao V, Buch P, Vaishnav D, Parmar S. Sesamol protects hippocampal CA1 neurons and reduces neuronal infarction in global model of cerebral ischemia in rats. PHARMANUTRITION 2020. [DOI: 10.1016/j.phanu.2020.100217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
13
|
Haddadi R, Shahidi Z, Eyvari-Brooshghalan S. Silymarin and neurodegenerative diseases: Therapeutic potential and basic molecular mechanisms. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 79:153320. [PMID: 32920285 DOI: 10.1016/j.phymed.2020.153320] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 06/20/2020] [Accepted: 07/01/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Neurodegenerative diseases (NDDs) are primarily characterized by selective neuronal loss in the brain. Alzheimer's disease as the most common NDDs and the most prevalent cause of dementia is characterized by Amyloid-beta deposition, which leads to cognitive and memory impairment. Parkinson's disease is a progressive neurodegenerative disease characterized by the dramatic death of dopaminergic neuronal cells, especially in the SNc and caused alpha-synuclein accumulation in the neurons. Silymarin, an extract from seeds of Silybum marianum, administered mostly for liver disorders and also had anti-oxidant and anti-carcinogenic activities. PURPOSE The present comprehensive review summarizes the beneficial effects of Silymarin in-vivo and in-vitro and even in animal models for these NDDs. METHODS A diagram model for systematic review is utilized for this search. The research is conducted in the following databases: PubMed, Web of Science, Scopus, and Science Direct. RESULTS Based on the inclusion criteria, 83 studies were selected and discussed in this review. CONCLUSION Lastly, we review the latest experimental evidences supporting the potential effects of Silymarin, as a neuroprotective agent in NDDs.
Collapse
Affiliation(s)
- Rasool Haddadi
- Department of Pharmacology and Toxicology, School of Pharmacy, Medicinal plant and natural products Research Center, Hamadan University of Medical Sciences, Hamadan 6517838678, Iran.
| | - Zahra Shahidi
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Shahla Eyvari-Brooshghalan
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
14
|
Rathore P, Arora I, Rastogi S, Akhtar M, Singh S, Samim M. Collagen Nanoparticle-Mediated Brain Silymarin Delivery: An Approach for Treating Cerebral Ischemia and Reperfusion-Induced Brain Injury. Front Neurosci 2020; 14:538404. [PMID: 33192240 PMCID: PMC7649428 DOI: 10.3389/fnins.2020.538404] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 08/25/2020] [Indexed: 12/12/2022] Open
Abstract
Silymarin is a bioactive constituent isolated from milk thistle (Silybum marinum). Since its discovery, silymarin has been considered a gold standard drug in treating ailments related to the liver, resulting from alcohol consumption and viral hepatitis. This hepatoprotective nature of silymarin arises out of antioxidative and tissue-regenerating properties of silymarin. However, several recent studies have established the neuroprotective link of silymarin, too. Thus, the current investigation was aimed at exploring the neuroprotective effect of nanosilymarin (silymarin encapsulated inside collagen-based polymeric nanoparticulate drug delivery system). The study aimed at bringing out the role of nanoparticles in enhancing the therapeutic effect of silymarin against neuronal injury, originating out of oxidative-stress-related brain damages in focal cerebral ischemia. Collagen-based micellar nanoparticles were prepared and stabilized using 3-ethyl carbodiimide-hydrochloride (EDC-Hcl) and malondialdehyde (MDA) as crosslinkers. Nanoparticles were characterized using dynamic light scattering (DLS), transmission electron microscopy (TEM), and Fourier transform infrared (FT-IR) spectroscopy techniques, and the size of nanoparticles was found to be around 48 nm. Male albino Wistar rats were pretreated with three different doses of nanosilymarin of 10, 100, and 1,000 μg/kg b.wt and a dose of free silymarin of 100 mg/kg b.wt intraperitoneally (i.p.) for 7 days. Focal cerebral ischemia was induced using the middle cerebral artery occlusion (MCAO) model on the eighth day for 1 h followed by 24 h reperfusion. The animals were then evaluated for neurobehavioral, infarct analysis, biochemical, histopathological, and immunohistochemical studies. All the above parameters showed remarkable improvement in nanosilymarin-treated groups in comparison to the silymarin-treated group. Nanoparticle encapsulation of drug enhanced neuroprotection by increasing drug bioavailability and targeting. Thus, the present study concluded with satisfactory results, showing the critical role played by nanoparticles in improving the neuroprotection at very low drug doses.
Collapse
Affiliation(s)
- Pankaj Rathore
- Department of Chemistry, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi, India
| | - Indu Arora
- Department of Biomedical Sciences, Shaheed Rajguru College, University of Delhi, New Delhi, India
| | - Shweta Rastogi
- Department of Chemistry, Hansraj College, University of Delhi, New Delhi, India
| | - Mohd Akhtar
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
| | - Shruti Singh
- Department of Botany, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi, India
| | - Mohammed Samim
- Department of Chemistry, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi, India
| |
Collapse
|
15
|
Moghaddam AH, Mokhtari Sangdehi SR, Ranjbar M, Hasantabar V. Preventive effect of silymarin-loaded chitosan nanoparticles against global cerebral ischemia/reperfusion injury in rats. Eur J Pharmacol 2020; 877:173066. [DOI: 10.1016/j.ejphar.2020.173066] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/01/2020] [Accepted: 03/10/2020] [Indexed: 12/29/2022]
|
16
|
Camini FC, Costa DC. Silymarin: not just another antioxidant. J Basic Clin Physiol Pharmacol 2020; 31:/j/jbcpp.2020.31.issue-4/jbcpp-2019-0206/jbcpp-2019-0206.xml. [PMID: 32134732 DOI: 10.1515/jbcpp-2019-0206] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 11/30/2019] [Indexed: 06/10/2023]
Abstract
Silymarin (Silybum marianum; SM), popularly known as milk thistle, is an extract that has been used for many centuries to treat liver diseases. In recent years, several studies have shown that SM is not only just another antioxidant but also a multifunctional compound that exhibits several beneficial properties for use in the treatment and prevention of different types of pathologies and disorders. This review aims at demonstrating the main protective activities of SM in diseases, such as cancer, diabetes, hepatitis, non-alcoholic fatty liver disease, alcoholic liver disease, hepatitis C virus, hepatitis B virus, metabolic syndrome, depression, cardiovascular diseases and thalassemia, in addition to its photoprotective activity in in vitro tests and preclinical studies. Its main functions include antioxidant and anti-inflammatory effects, and it acts as modulator of signaling pathways. It has been suggested that SM presents great multifunctional potential and is capable of achieving promising results in different types of research. However, caution is still needed regarding its indiscriminate use in humans as there are only a few clinical studies relating to the adequate dose and the actual efficacy of this extract in different types of diseases.
Collapse
Affiliation(s)
- Fernanda Caetano Camini
- Laboratory of Metabolic Biochemistry, Post-Graduate Program in Biological Sciences, Nucleus of Research in Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Daniela Caldeira Costa
- Laboratory of Metabolic Biochemistry, Post-Graduate Program in Biological Sciences, Nucleus of Research in Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Minas Gerais, Brazil
- Laboratory of Metabolic Biochemistry, Department of Biological Sciences, Federal University of Ouro Preto, Morro do Cruzeiro University Campus, Ouro Preto, Minas Gerais, Brazil
| |
Collapse
|
17
|
Thakare VN, Patil RR, Suralkar AA, Dhakane VD, Patel BM. Protocatechuic acid attenuate depressive-like behavior in olfactory bulbectomized rat model: behavioral and neurobiochemical investigations. Metab Brain Dis 2019; 34:775-787. [PMID: 30848471 DOI: 10.1007/s11011-019-00401-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 02/25/2019] [Indexed: 12/13/2022]
Abstract
The main objective of the present study is to investigate potential effects of PCA in OBX induced depressive-like behavior in rat model. PCA was administered at a dose of 100 mg/kg and 200 mg/kg, by per oral in OBX and sham operated rats. Behavioral (ambulatory and rearing activity and immobility time), neurochemical [serotonin (5-HT), dopamine (DA), norepinephrine (NE) and brain derived neurotrophic factor (BDNF) expression], biochemical (MDA formation, IL-6, TNF-α and antioxidants) changes in hippocampus and cerebral cortex along with serum corticosterone were investigated. Experimental findings reveals that OBX subjected rats showed alteration in behaviors like, increase in immobility time, ambulatory and rearing behaviors significantly, reduced BDNF level, 5-HT, DA,NE and antioxidant parameters along with increased serum corticosterone, MDA formation, IL-6, and TNF-α in hippocampus and cerebral cortex compared to sham operated rats. Administration of PCA significantly attenuated behavioral and neurobiochemical alterations, thus, its antidepressant-like activity is largely mediated through modulation of neurotransmitter, endocrine and immunologic systems, mainly by improvements of BDNF, 5-HT, DA, NE, reduced MDA, IL-6, and TNF-α in hippocampus and cerebral cortex.
Collapse
Affiliation(s)
- Vishnu N Thakare
- Department of Pharmacology, Sinhgad Institute of Pharmaceutical Sciences, Lonavala, Maharashtra, 410401, India
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Sarkhej-Gandhinagar Highway, Ahmedabad, Gujarat, 382 481, India
| | - Rajesh R Patil
- Department of Pharmacology, Sinhgad Institute of Pharmaceutical Sciences, Lonavala, Maharashtra, 410401, India
| | - Anupama A Suralkar
- Department of Pharmacology, Smt. Kashibai Navale College of Pharmacy, Kondhawa, Pune, Maharashtra, 411048, India
| | - Valmik D Dhakane
- Research and Development, Astec Life Sciences, Mumbai, Maharashtra, India
| | - Bhoomika M Patel
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Sarkhej-Gandhinagar Highway, Ahmedabad, Gujarat, 382 481, India.
| |
Collapse
|
18
|
Ashraf A, Mahmoud PA, Reda H, Mansour S, Helal MH, Michel HE, Nasr M. Silymarin and silymarin nanoparticles guard against chronic unpredictable mild stress induced depressive-like behavior in mice: involvement of neurogenesis and NLRP3 inflammasome. J Psychopharmacol 2019; 33:615-631. [PMID: 30896354 DOI: 10.1177/0269881119836221] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND The neuropathology of depression is quite complex. Thus, treatment failures are frequent with current antidepressants, raising the need for more effective ones. AIMS This study aimed to investigate the influence of silymarin on depressive-like behavior induced by chronic unpredictable mild stress (CUMS) and explore the underlying mechanisms. METHODS Silymarin was formulated as nanostructured lipid carriers (a lipid-based type of nanoparticle with the advantages of physical stability, good release profile, and targeted delivery). Mice were subjected to CUMS paradigm during 14 days. During this period, mice received silymarin (200 mg/kg, p.o.) per se or in its nanoparticle form or fluoxetine (10 mg/kg, p.o.). On the 15th day behavioral and biochemical parameters were analyzed. RESULTS Oral administration of silymarin (200 mg/kg), particularly in its nanoparticulate form, exerted an antidepressant-like effect, comparable with fluoxetine in mice, as demonstrated in the behavioral despair tests. Silymarin also reversed prefrontal cortical and hippocampal CUMS-induced oxidative stress and neuroinflammation. Furthermore, silymarin augmented neurotransmitter levels, enhanced neurogenesis and inhibited nod-like receptor protein 3 inflammasome activation. Silymarin nanoparticles were superior to silymarin in certain parameters probably due to significantly higher brain silybinin (the major active component of silymarin) concentration by 12.46 fold in the group administered silymarin nanoparticles compared with the mice which were administered silymarin per se. CONCLUSIONS The antidepressant-like effect of silymarin can be attributed to its antioxidant and anti-inflammatory effects as well as increased neurogenesis in the prefrontal cortex and hippocampus, which delineates silymarin, especially in nanoparticle form, as a promising strategy for treatment of depression.
Collapse
Affiliation(s)
- Aya Ashraf
- 1 Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | | | - Haidy Reda
- 1 Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Salma Mansour
- 1 Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mehad H Helal
- 1 Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Haidy E Michel
- 2 Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Maha Nasr
- 3 Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
19
|
Grigoruţă M, Vargas-Caraveo A, Vázquez-Mayorga E, Castillo-Michel HA, Díaz-Sánchez ÁG, Reyes-Herrera J, Martínez-Martínez A. Blood mononuclear cells as speculum of emotional stress analyzed by synchrotron infrared spectroscopy and a nootropic drug. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2018; 204:475-483. [PMID: 29966903 DOI: 10.1016/j.saa.2018.06.075] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 06/20/2018] [Accepted: 06/20/2018] [Indexed: 06/08/2023]
Abstract
Chronic psychological stress is an important public health issue which generates behavioral changes, anxiety, immunosuppression and oxidative damage. Piracetam is a cognitive enhancer, at cellular level it protects from oxidative stress. The aim of this study was to evaluate the effect of psychological stress and of piracetam on circulating mononuclear cells by analyzing the biochemical spectrome using Synchrotron Radiation Fourier Transform Infrared Microspectroscopy (SR-μFTIR). Rats were exposed for five days to a stressor (cat odor) under oral administration of piracetam (600 mg/kg). SR-μFTIR analysis showed a decrease in bands associated to the lipids region (2852 cm-1, 2923 cm-1 and 2962 cm-1) and an increase absorption of the amide I band (1654 cm-1) under stress conditions. The principal component analysis showed increase oxidation of lipids (decrease of 3010 cm-1, 2923 cm-1 and 2852 cm-1 bands) as well as proteins denaturation (increase of 1610 cm-1 and 1690 cm-1 bands) under stress. Piracetam provided protection to polyunsaturated lipids (p ≤ 0.001) and lipids/proteins ratio (p ≤ 0.001). Behaviorally, this drug diminished fear and anxiety in stressed animals by the plus maze test (p ≤ 0.002). However, this drug induced oxidative stress in mononuclear cells from unstressed animals and altered their behavior.
Collapse
Affiliation(s)
- Mariana Grigoruţă
- Departamento de Ciencias Químico Biológicas, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez (UACJ), Anillo envolvente Pronaf y Estocolmo s/n, 32310 Cd. Juárez, Mexico
| | | | - Emmanuel Vázquez-Mayorga
- Departamento de Ciencias Químico Biológicas, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez (UACJ), Anillo envolvente Pronaf y Estocolmo s/n, 32310 Cd. Juárez, Mexico
| | | | - Ángel G Díaz-Sánchez
- Departamento de Ciencias Químico Biológicas, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez (UACJ), Anillo envolvente Pronaf y Estocolmo s/n, 32310 Cd. Juárez, Mexico
| | - Juan Reyes-Herrera
- European Synchrotron Radiation Facility (ESRF), B.P. 220, Grenoble, France
| | - Alejandro Martínez-Martínez
- Departamento de Ciencias Químico Biológicas, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez (UACJ), Anillo envolvente Pronaf y Estocolmo s/n, 32310 Cd. Juárez, Mexico; El Colegio de Chihuahua, Calle Partido Díaz 4723 esquina con Anillo Envolvente del PRONAF, colonia Progresista, Ciudad Juárez, Chihuahua C.P. 32310, Mexico.
| |
Collapse
|
20
|
Ullah H, Khan H. Anti-Parkinson Potential of Silymarin: Mechanistic Insight and Therapeutic Standing. Front Pharmacol 2018; 9:422. [PMID: 29755356 PMCID: PMC5934474 DOI: 10.3389/fphar.2018.00422] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 04/11/2018] [Indexed: 02/05/2023] Open
Abstract
Parkinson’s disease (PD) involves aggregation of α-synuclein and progressive loss of dopaminergic neurons. Pathogenesis of PD may also be related to one’s genetic background. PD is most common among geriatric population and approximately 1–2% of population suffers over age 65 years. Currently no successful therapies are in practice for the management of PD and available therapies tend to decrease the symptoms of PD only. Furthermore, these are associated with diverse range of adverse effects profile. The neuroprotective effects of polyphenols are widely studied and documented. Among phytochemicals, silymarin is one of the most widely used flavonoids because of its extensive therapeutic properties and has been indicated in pathological conditions of prostate, CNS, lungs, skin, liver, and pancreas. Silymarin is a mixture of flavonolignans (silybin, isosilybin, and silychristin), small amount of flavonoids (taxifolin), fatty acids, and other polyphenolic compounds extracted from the dried fruit of Silybum marianum and is clinically used for hepatoprotective effects since ancient times. Neuroprotective effects of silymarin have been studied in various models of neurological disorders such as Alzheimer’s disease, PD, and cerebral ischemia. The aim of the present study is to provide a comprehensive review of the recent literature exploring the effects of silymarin administration on the progression of PD. Reducing oxidative stress, inflammatory cytokines, altering cellular apoptosis machinery, and estrogen receptor machinery are mechanisms that are responsible for neuroprotection by silymarin, as discussed in this review. Additionally, because of poor aqueous solubility, the bioavailability of silymarin is low and only 23–47% of silymarin reaches systemic circulation after oral administration. Our primary focus is on the chemical basis of the pharmacology of silymarin in the treatment of PD and its mechanisms and possible therapeutic/clinical status while addressing the bioavailability limitation.
Collapse
Affiliation(s)
- Hammad Ullah
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, Pakistan
| |
Collapse
|
21
|
Barakat W, Fahmy A, Askar M, El-Kannishy S. Effectiveness of arginase inhibitors against experimentally induced stroke. Naunyn Schmiedebergs Arch Pharmacol 2018; 391:603-612. [PMID: 29600431 DOI: 10.1007/s00210-018-1489-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 03/22/2018] [Indexed: 01/28/2023]
Abstract
Stroke is a lethal disease, but it disables more than it kills. Stroke is the second leading cause of death and the most frequent cause of permanent disability in adults worldwide, with 90% of survivors having residual deficits. The pathophysiology of stroke is complex and involves a strong inflammatory response associated with oxidative stress and activation of several proteolytic enzymes. The current study was designed to investigate the effect of arginase inhibitors (L-citruline and L-ornithine) against ischemic stroke induced in rats by middle cerebral artery occlusion (MCAO). MCAO resulted in alteration in rat behavior, brain infarct, and edema associated with disruption of the blood-brain barrier (BBB). This was mediated through overexpression of arginase I and II, inducible NOS (iNOS), malondialdehyde (MDA), advanced glycation end products (AGEs), TNF-α, and IL-1β and downregulation of endothelial nitric oxide synthase (eNOS). Treatment with L-citruline and L-ornithine and the standard neuroprotective drug cerebrolysin ameliorated all the deleterious effects of stroke. These results indicate the possible use of arginase inhibitors in the treatment of stroke after suitable clinical trials are done.
Collapse
Affiliation(s)
- Waleed Barakat
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tabuk University, Tabuk, Kingdom of Saudi Arabia.
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt.
| | - Ahmad Fahmy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Mohamed Askar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Sherif El-Kannishy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tabuk University, Tabuk, Kingdom of Saudi Arabia
- Analytical Toxicology - Emergency Hospital, Faculty of Medicine, University of Mansoura, Mansoura, Egypt
| |
Collapse
|
22
|
Thakare VN, Patil RR, Oswal RJ, Dhakane VD, Aswar MK, Patel BM. Therapeutic potential of silymarin in chronic unpredictable mild stress induced depressive-like behavior in mice. J Psychopharmacol 2018; 32:223-235. [PMID: 29215318 DOI: 10.1177/0269881117742666] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Silymarin, a plant-derived polyphenolic flavonoid of Silybum marianum, elicited significant antidepressant-like activity in an acute restraint stress model of depression. It improved monoamines, mainly 5-hydroxytryptamine (5-HT) levels in the cortex, dopamine (DA) and norepinephrine (NE) in the cerebellum in mice. The present study was undertaken to explore the antidepressant potential of silymarin in chronic unpredictable mild stress (CUMS) induced depressive-like behavior in mice, and to find out its probable mechanism(s) of action, mainly neurogenesis, neuroinflammation, and/or oxidative stress. The mice were subjected to CUMS for 28 days (4 weeks) and administered with silymarin (100 mg/kg and 200 mg/kg), or fluoxetine or vehicle from days 8 to 28 (3 weeks simultaneously). Animals were evaluated for behavioral changes, such as anhedonia by sucrose preference test, behavioral despair by forced swim test, and exploratory behaviors by an open field test. In addition, neurobiochemical alterations, mainly monoamines, 5-HT, NE, DA, neurotrophic factor BDNF, and cytokines, IL-6, TNF-α, oxidant-antioxidant parameters by determining the malondialdehyde formation (an index of lipid peroxidation process), superoxide dismutase (SOD) and catalase (CAT) activity in hippocampus and cerebral cortex along with serum corticosterone were investigated. Our findings reveal that mice subjected to CUMS exhibited lower sucrose preference, increase immobility time without affecting general locomotion of the animals, and reduce BDNF, 5-HT, NE, and DA level, increased serum corticosterone, IL-6 and TNF-α along with an oxidant-antioxidant imbalance in the hippocampus and cerebral cortex. Silymarin significantly reversed the CUMS-induced changes in the hippocampus and cerebral cortex in mice. Thus, the possible mechanism involved in the antidepressant-like activity of silymarin is correlated to the alleviation of monoaminergic, neurogenesis (enhancing 5-HT, NE, and BDNF levels), and attenuation of inflammatory cytokines system and oxidative stress by modulation of corticosterone response, restoration of antioxidant defense system in cerebral cortex and hippocampus.
Collapse
Affiliation(s)
- Vishnu N Thakare
- 1 Department of Pharmacology, Sinhgad Institute of Pharmaceutical Sciences, Lonavala, India.,4 Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, India
| | - Rajesh R Patil
- 1 Department of Pharmacology, Sinhgad Institute of Pharmaceutical Sciences, Lonavala, India
| | - Rajesh J Oswal
- 1 Department of Pharmacology, Sinhgad Institute of Pharmaceutical Sciences, Lonavala, India
| | | | - Manoj K Aswar
- 3 Department of Pharmacology, Sinhgad Institute of Pharmacy, Nerhe, Pune, India
| | - Bhoomika M Patel
- 4 Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, India
| |
Collapse
|
23
|
Thakare VN, Aswar MK, Kulkarni YP, Patil RR, Patel BM. Silymarin ameliorates experimentally induced depressive like behavior in rats: Involvement of hippocampal BDNF signaling, inflammatory cytokines and oxidative stress response. Physiol Behav 2017; 179:401-410. [PMID: 28711395 DOI: 10.1016/j.physbeh.2017.07.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 05/05/2017] [Accepted: 07/06/2017] [Indexed: 12/12/2022]
Abstract
Silymarin is a polyphenolic flavonoid of Silybum marianum, exhibited neuroprotection and antidepressant like activity in acute restraint stressed mice. The main objective of the present study is to investigate possible antidepressant like activity of silymarin in experimentally induced depressive behavior in rats. The depressive behaviors were induced in rats by olfactory bulbectomized (OBX) technique. Wistar rats were administered with silymarin at a dose of 100mg/kg and 200mg/kg, by per oral in OBX and sham operated rats. Behavioral (ambulatory and rearing activity and immobility time), neurochemical [serotonin (5-HT), dopamine (DA), norepinephrine (NE) and brain derived neurotrophic factor (BDNF) level], biochemical (MDA formation, IL-6, TNF-α and antioxidants) changes in hippocampus and cerebral cortex along with serum corticosterone were investigated. Rats subjected to OBX elicited significant increase in immobility time, ambulatory and rearing behaviors, reduced BDNF level, 5-HT, DA, NE and antioxidant parameters along with increased serum corticosterone, MDA formation, IL-6, and TNF-α in hippocampus and cerebral cortex compared to sham operated rats. Administration of with silymarin significantly attenuated immobility time, ambulatory and rearing behaviors, serum corticosterone and improved BDNF expression, 5-HT, DA, NE and antioxidant paradigms in cerebral cortex as well as hippocampus. In addition, silymarin attenuated IL-6, and TNF-α significantly in hippocampus and cerebral cortex in OBX rats. Thus, silymarin exhibits anti-depressant-like activity in OBX rats due to alterations in several neurotransmitters, endocrine and immunologic systems, including BDNF, 5-HT, DA, NE, MDA formation, IL-6, and TNF-α in hippocampus and cerebral cortex as well as serum corticosterone.
Collapse
Affiliation(s)
- Vishnu N Thakare
- Department of Pharmacology, Sinhgad Institute of Pharmaceutical Sciences, Lonavala 410401, Maharashtra, India; Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad 382 481, Gujarat, India
| | - Manoj K Aswar
- Department of Pharmacology, Sinhgad Institute of Pharmacy, Nerhe, Pune, Maharashtra, India
| | - Yogesh P Kulkarni
- Department of Pharmacology, Sinhgad Institute of Pharmaceutical Sciences, Lonavala 410401, Maharashtra, India
| | - Rajesh R Patil
- Department of Pharmacology, Sinhgad Institute of Pharmaceutical Sciences, Lonavala 410401, Maharashtra, India
| | - Bhoomika M Patel
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad 382 481, Gujarat, India.
| |
Collapse
|
24
|
Thakare VN, Dhakane VD, Patel BM. Attenuation of acute restraint stress-induced depressive like behavior and hippocampal alterations with protocatechuic acid treatment in mice. Metab Brain Dis 2017; 32:401-413. [PMID: 27785705 DOI: 10.1007/s11011-016-9922-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 10/19/2016] [Indexed: 01/24/2023]
Abstract
Protocatechuic acid ethyl ester (PCA), a phenolic compound, exhibits neuroprotective effects through improving endogenous antioxidant enzymatic and nonezymatic system. Based on the role of oxidative stress in modulating depressive disorders and the relationship between neuroprotective and antioxidant potential of PCA, we studied if its antidepressant like effect is associated by modulation of cerebral cortex and hippocampal antioxidant alterations. Acute restraint stress (ARS) is known to induce depressive like behavior by neuronal oxidative damage in mice. Swiss albino mice subjected to ARS exhibited an increased immobility time in forced swim test, elevated serum corticosterone and produced oxidative stress dependent alterations in cerebral cortex and hippocampus mainly increased thiobarbituric acid reactive substances and reduced catalase (CAT), superoxide dismutase (SOD) activity. Treatment with PCA was able to prevent stress induced immobility time in forced swim test without altering locomotor activity in mice. Further, PCA treatment attenuated the elevation of serum corticosterone, lipid peroxidation and restored enzymatic antioxidants in cerebral cortex and hippocampus in ARS mice. Altogether, the experimental findings demonstrate the notion that PCA exhibit antidepressant like activity might be related, at least in part, to its capability of modulating antioxidant defense system and oxidative damage induced by ARS in cerebral cortex and hippocampus in mice and thus maintain the pro-/anti-oxidative homeostasis.
Collapse
Affiliation(s)
- Vishnu N Thakare
- Department of Pharmacology, Sinhgad Institute of Pharmaceutical Sciences, Lonavala, Maharashtra, 410401, India
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Sarkhej-Gandhinagar Highway, Ahmedabad, Gujarat, 382 481, India
| | - Valmik D Dhakane
- Research and Development, Astec Life Sciences, Mumbai, Maharashtra, India
| | - Bhoomika M Patel
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Sarkhej-Gandhinagar Highway, Ahmedabad, Gujarat, 382 481, India.
| |
Collapse
|
25
|
Gupta S, Gupta YK. Combination of Zizyphus jujuba and silymarin showed better neuroprotective effect as compared to single agent in MCAo-induced focal cerebral ischemia in rats. JOURNAL OF ETHNOPHARMACOLOGY 2017; 197:118-127. [PMID: 27452658 DOI: 10.1016/j.jep.2016.07.060] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 07/12/2016] [Accepted: 07/21/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditionally, Zizyphus jujuba is used for anticonvulsant, hypnotic-sedative, anxiolytic, tranquilizer, antioxidant and anti-inflammatory properties. Likewise silymarin is popularly used for its potent antioxidant and hepatoprotective effects. Stroke being a multifactorial disease with unsatisfactory treatment outcomes, necessitates development of multimodal therapeutic interventions. Thus, we evaluated the therapeutic benefits of herbal combination of Z. jujuba and silymarin in a focal cerebral ischemia model. AIM OF THE STUDY To evaluate the neuroprotective potential of hydroalcoholic extract of Z. jujuba (HEZJ) fruit and silymarin alone and in combination in middle cerebral artery occlusion (MCAo) model of focal cerebral ischemia in rats. MATERIALS AND METHODS Male Wistar rats were pretreated with HEZJ (100, 250 and 500mg/kg, p.o.) or silymarin (250mg/kg, p.o.) for 3 days prior to induction of MCAo. Neurological deficit score, motor impairment and cerebral infarction were assessed 24h following MCAo. HEZJ (250mg/kg) co-administered with silymarin (250mg/kg) for 3 days prior to induction of MCAo was also evaluated for above parameters and oxidative stress. Malondialdehyde (MDA), nitric oxide (NO) and superoxide dismutase (SOD) levels in the cortex, striatum and hippocampal brain regions were estimated 24h post MCAo. RESULTS Pretreatment with HEZJ and silymarin reduced the neurological deficit score, motor impairment and cerebral infarction volume. HEZJ and silymarin pretreatment also ameliorated the oxidative stress in different brain regions, which was evident from increased SOD levels, decreased MDA and NO levels as compared to MCAo control rats. Interestingly neuroprotective efficacy was potentiated by pretreatment with HEZJ and silymarin combination. CONCLUSION Pretreatment with HEZJ and silymarin combination was observed to have better neuroprotection mediated via amelioration of oxidative stress in the focal cerebral ischemia model.
Collapse
Affiliation(s)
- Sangeetha Gupta
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Yogendra Kumar Gupta
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi 110029, India.
| |
Collapse
|
26
|
Thakare VN, Dhakane VD, Patel BM. Potential antidepressant-like activity of silymarin in the acute restraint stress in mice: Modulation of corticosterone and oxidative stress response in cerebral cortex and hippocampus. Pharmacol Rep 2016; 68:1020-7. [DOI: 10.1016/j.pharep.2016.06.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/02/2016] [Accepted: 06/02/2016] [Indexed: 12/14/2022]
|
27
|
Lan AP, Chen J, Chai ZF, Hu Y. The neurotoxicity of iron, copper and cobalt in Parkinson's disease through ROS-mediated mechanisms. Biometals 2016; 29:665-78. [PMID: 27349232 DOI: 10.1007/s10534-016-9942-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 06/18/2016] [Indexed: 12/14/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease with gradual loss of dopaminergic neurons. Despite extensive research in the past decades, the etiology of PD remains elusive. Nevertheless, multiple lines of evidence suggest that oxidative stress is one of the common causes in the pathogenesis of PD. It has also been suggested that heavy metal-associated oxidative stress may be implicated in the etiology and pathogenesis of PD. Here we review the roles of redox metals, including iron, copper and cobalt, in PD. Iron is a highly reactive element and deregulation of iron homeostasis is accompanied by concomitant oxidation processes in PD. Copper is a key metal in cell division process, and it has been shown to have an important role in neurodegenerative diseases such as PD. Cobalt induces the generation of reactive oxygen species (ROS) and DNA damage in brain tissues.
Collapse
Affiliation(s)
- A P Lan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing, 100049, China
| | - J Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing, 100049, China
| | - Z F Chai
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing, 100049, China.,School of Radiological and Interdisciplinary Sciences, Soochow University, Suzhou, 215123, China
| | - Y Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing, 100049, China.
| |
Collapse
|
28
|
Ferreira EDO, Fernandes MYSD, Lima NMRD, Neves KRT, Carmo MRSD, Lima FAV, Fonteles AA, Menezes APF, Andrade GMD. Neuroinflammatory response to experimental stroke is inhibited by eriodictyol. Behav Brain Res 2016; 312:321-32. [PMID: 27353856 DOI: 10.1016/j.bbr.2016.06.046] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 06/21/2016] [Accepted: 06/24/2016] [Indexed: 11/30/2022]
Abstract
BACKGROUND Cerebral ischemia is a common disease and one of the most common causes of death and disability worldwide. The lack of glucose and oxygen in neuronal tissue leads to a series of inflammatory events, culminating in neuronal death. Eriodictyol is a flavonoid isolated from the Chinese herb Dracocephalum rupestre that has been proven to have anti-inflammatory properties. HYPOTHESIS/PURPOSE Thus, the present study was designed to explore whether eriodictyol has neuroprotective effects against the neuronal damage, motor and memory deficits induced by permanent middle cerebral artery occlusion (pMCAO) in mice. STUDY DESIGN Animals were orally treated with eriodictyol (1, 2 and 4mg/kg) or vehicle (saline) 30min before pMCAO, 2h after, and then once daily for the following five days. METHODS The parameters studied were neuronal viability, brain infarcted area; sensorimotor deficits; exploratory activity; working and aversive memory; myeloperoxidase (MPO) activity; TNFα, iNOS and GFAP immunoreactivity. RESULTS The treatment with eriodictyol prevented neuronal death, reduced infarct area and improved neurological and memory deficits induced by brain ischemia. The increase of MPO activity and TNF-α, iNOS and GFAP expression were also reduced by eriodictyol treatment. CONCLUSION These findings demonstrate that eriodictyol exhibit promising neuroprotection effects against the permanent focal ischemia cerebral injury in the mice experimental model and the underlying mechanisms might be mediated through inhibition of neuroinflammation.
Collapse
Affiliation(s)
- Emerson de Oliveira Ferreira
- Post-Graduate Programme in Medical Sciences, Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceará, Brazil.
| | - Mara Yone Soares Dias Fernandes
- Post-Graduate Programme in Pharmacology, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Brazil.
| | - Neila Maria Rocha de Lima
- Post-Graduate Programme in Medical Sciences, Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceará, Brazil.
| | - Kelly Rose Tavares Neves
- Post-Graduate Programme in Pharmacology, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Brazil.
| | - Marta Regina Santos do Carmo
- Post-Graduate Programme in Pharmacology, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Brazil.
| | - Francisco Arnaldo Viana Lima
- Post-Graduate Programme in Pharmacology, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Brazil.
| | - Analu Aragão Fonteles
- Post-Graduate Programme in Pharmacology, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Brazil.
| | - Ana Paula Fontenele Menezes
- Post-Graduate Programme in Medical Sciences, Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceará, Brazil.
| | - Geanne Matos de Andrade
- Post-Graduate Programme in Medical Sciences, Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceará, Brazil; Post-Graduate Programme in Pharmacology, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Brazil; Institute of Biomedicine of Brazilian Semi-arid, Brazil.
| |
Collapse
|
29
|
The Effect of 3′,4′-Dihydroxyflavonol on Lipid Peroxidation in Rats with Cerebral Ischemia Reperfusion Injury. Neurochem Res 2016; 41:1732-40. [DOI: 10.1007/s11064-016-1889-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 02/10/2016] [Accepted: 03/17/2016] [Indexed: 01/01/2023]
|
30
|
Maiese K. FoxO proteins in the nervous system. Anal Cell Pathol (Amst) 2015; 2015:569392. [PMID: 26171319 PMCID: PMC4478359 DOI: 10.1155/2015/569392] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 05/31/2015] [Indexed: 02/07/2023] Open
Abstract
Acute as well as chronic disorders of the nervous system lead to significant morbidity and mortality for millions of individuals globally. Given the ability to govern stem cell proliferation and differentiated cell survival, mammalian forkhead transcription factors of the forkhead box class O (FoxO) are increasingly being identified as potential targets for disorders of the nervous system, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and auditory neuronal disease. FoxO proteins are present throughout the body, but they are selectively expressed in the nervous system and have diverse biological functions. The forkhead O class transcription factors interface with an array of signal transduction pathways that include protein kinase B (Akt), serum- and glucocorticoid-inducible protein kinase (SgK), IκB kinase (IKK), silent mating type information regulation 2 homolog 1 (S. cerevisiae) (SIRT1), growth factors, and Wnt signaling that can determine the activity and integrity of FoxO proteins. Ultimately, there exists a complex interplay between FoxO proteins and their signal transduction pathways that can significantly impact programmed cell death pathways of apoptosis and autophagy as well as the development of clinical strategies for the treatment of neurodegenerative disorders.
Collapse
|
31
|
New Insights for Oxidative Stress and Diabetes Mellitus. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:875961. [PMID: 26064426 PMCID: PMC4443788 DOI: 10.1155/2015/875961] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 04/15/2015] [Indexed: 12/12/2022]
Abstract
The release of reactive oxygen species (ROS) and the generation of oxidative stress are considered critical factors for the pathogenesis of diabetes mellitus (DM), a disorder that is growing in prevalence and results in significant economic loss. New therapeutic directions that address the detrimental effects of oxidative stress may be especially warranted to develop effective care for the millions of individuals that currently suffer from DM. The mechanistic target of rapamycin (mTOR), silent mating type information regulation 2 homolog 1 (S. cerevisiae) (SIRT1), and Wnt1 inducible signaling pathway protein 1 (WISP1) are especially justified to be considered treatment targets for DM since these pathways can address the complex relationship between stem cells, trophic factors, impaired glucose tolerance, programmed cell death pathways of apoptosis and autophagy, tissue remodeling, cellular energy homeostasis, and vascular biology that greatly impact the biology and disease progression of DM. The translation and development of these pathways into viable therapies will require detailed understanding of their proliferative nature to maximize clinical efficacy and limit adverse effects that have the potential to lead to unintended consequences.
Collapse
|
32
|
Maiese K. Novel applications of trophic factors, Wnt and WISP for neuronal repair and regeneration in metabolic disease. Neural Regen Res 2015; 10:518-28. [PMID: 26170801 PMCID: PMC4424733 DOI: 10.4103/1673-5374.155427] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2015] [Indexed: 12/13/2022] Open
Abstract
Diabetes mellitus affects almost 350 million individuals throughout the globe resulting in significant morbidity and mortality. Of further concern is the growing population of individuals that remain undiagnosed but are susceptible to the detrimental outcomes of this disorder. Diabetes mellitus leads to multiple complications in the central and peripheral nervous systems that include cognitive impairment, retinal disease, neuropsychiatric disease, cerebral ischemia, and peripheral nerve degeneration. Although multiple strategies are being considered, novel targeting of trophic factors, Wnt signaling, Wnt1 inducible signaling pathway protein 1, and stem cell tissue regeneration are considered to be exciting prospects to overcome the cellular mechanisms that lead to neuronal injury in diabetes mellitus involving oxidative stress, apoptosis, and autophagy. Pathways that involve insulin-like growth factor-1, fibroblast growth factor, epidermal growth factor, and erythropoietin can govern glucose homeostasis and are intimately tied to Wnt signaling that involves Wnt1 and Wnt1 inducible signaling pathway protein 1 (CCN4) to foster control over stem cell proliferation, wound repair, cognitive decline, β-cell proliferation, vascular regeneration, and programmed cell death. Ultimately, cellular metabolism through Wnt signaling is driven by primary metabolic pathways of the mechanistic target of rapamycin and AMP activated protein kinase. These pathways offer precise biological control of cellular metabolism, but are exquisitely sensitive to the different components of Wnt signaling. As a result, unexpected clinical outcomes can ensue and therefore demand careful translation of the mechanisms that govern neural repair and regeneration in diabetes mellitus.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, Newark, New Jersey 07101, USA
| |
Collapse
|
33
|
A prolyl-hydroxylase inhibitor, ethyl-3,4-dihydroxybenzoate, induces cell autophagy and apoptosis in esophageal squamous cell carcinoma cells via up-regulation of BNIP3 and N-myc downstream-regulated gene-1. PLoS One 2014; 9:e107204. [PMID: 25232961 PMCID: PMC4169646 DOI: 10.1371/journal.pone.0107204] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Accepted: 08/12/2014] [Indexed: 12/14/2022] Open
Abstract
The protocatechuic acid ethyl ester ethyl-3,4-dihydroxybenzoate is an antioxidant found in the testa of peanut seeds. Previous studies have shown that ethyl-3,4-dihydroxybenzoate can effectively reduce breast cancer cell metastasis by inhibiting prolyl-hydroxylase. In this study, we investigated the cytotoxic effect of ethyl-3,4-dihydroxybenzoate on esophageal squamous cell carcinoma cells in vitro and identified key regulators of ethyl-3,4-dihydroxybenzoate-induced esophageal cancer cell death through transcription expression profiling. Using flow cytometry analysis, we found that ethyl-3,4-dihydroxybenzoate induced S phase accumulation, a loss in mitochondrial membrane permeabilization, and caspase-dependent apoptosis. Moreover, an expression profile analysis identified 46 up- and 9 down-regulated genes in esophageal cancer KYSE 170 cells treated with ethyl-3,4-dihydroxybenzoate. These differentially expressed genes are involved in several signaling pathways associated with cell cycle regulation and cellular metabolism. Consistent with the expression profile results, the transcriptional and protein expression levels of candidate genes NDRG1, BNIP3, AKR1C1, CCNG2 and VEGFA were found to be significantly increased in treated KYSE 170 cells by reverse-transcription PCR and western blot analysis. We also found that protein levels of hypoxia-inducible factor-1α, BNIP3, Beclin and NDRG1 were increased and that enriched expression of BNIP3 and Beclin caused autophagy mediated by microtubule-associated protein 1 light chain 3 in the treated cells. Autophagy and apoptosis were activated together in esophageal cancer cells after exposed to ethyl-3,4-dihydroxybenzoate. Furthermore, knock-down of NDRG1 expression by siRNA significantly attenuated apoptosis in the cancer cells, implying that NDRG1 may be required for ethyl-3,4-dihydroxybenzoate-induced apoptosis. Together, these results suggest that the cytotoxic effects of ethyl-3,4-dihydroxybenzoate were mediated by the up-regulation of NDRG1, BNIP3, Beclin and hypoxia-inducible factor-1α, initiating BNIP3 and Beclin mediated autophagy at an early stage and ultimately resulting in esophageal cancer cell apoptosis.
Collapse
|
34
|
Tabassum R, Vaibhav K, Shrivastava P, Khan A, Ahmed ME, Ashafaq M, Khan MB, Islam F, Safhi MM, Islam F. Perillyl alcohol improves functional and histological outcomes against ischemia-reperfusion injury by attenuation of oxidative stress and repression of COX-2, NOS-2 and NF-κB in middle cerebral artery occlusion rats. Eur J Pharmacol 2014; 747:190-9. [PMID: 25240714 DOI: 10.1016/j.ejphar.2014.09.015] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 09/07/2014] [Accepted: 09/08/2014] [Indexed: 12/26/2022]
Abstract
Perillyl alcohol (PA) is a monoterpene found in essential oils of mints, cherries, citreous fruits and lemon grass, reported to have antioxidant and anti-inflammatory properties. However, the role of PA in stroke is still illusive. Since oxidative stress and inflammation play a pivotal role in ischemia-reperfusion (I-R) injury, this study was designed to elucidate the potential effects of PA against I-R induced pathology in rat׳s brain. Middle cerebral artery occlusion (MCAO) for 2h followed by 22h reperfusion in Wistar male rats (250-280g, 14-16 weeks old) induced the behavioral and histological alterations along with exhausted antioxidant status and enhanced inflammatory mediators. However, PA administration (25, 50 and 100mg/kg b.wt orally once daily for 7 days) prior to MCAO significantly attenuated neurological deficits related to flexion test and spontaneous motor activity, improved grip strength and motor coordination in a dose dependent manner. PA treatment also inhibited oxidative stress in MCAO rats as evident from decreased lipid peroxidation and augmented level of reduced glutathione and restored activities of catalase, glutathione peroxidase, and glutathione reductase and thus, reduced infarct volume and protected the brain histology after I-R injury. Furthermore, PA markedly suppressed the level of proinflammatory cytokines (IL-1β, TNF α and IL-6) and down regulated expressions of cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (NOS-2) and nuclear factor κB (NF-κB) in MCAO group. In conclusion, PA mediates neuroprotection against I-R injury via mitigation of oxidative stress and inflammation and thus, may be a good therapeutic approach in stroke prone patient.
Collapse
Affiliation(s)
- Rizwana Tabassum
- Neurotoxicology Laboratory, Department of Medical Elementology & Toxicology (DST-FIST and UGC-SAP-BSR funded department), Jamia Hamdard (Hamdard University), Hamdard Nagar, New Delhi 110062, India
| | - Kumar Vaibhav
- Neurotoxicology Laboratory, Department of Medical Elementology & Toxicology (DST-FIST and UGC-SAP-BSR funded department), Jamia Hamdard (Hamdard University), Hamdard Nagar, New Delhi 110062, India
| | - Pallavi Shrivastava
- Neurotoxicology Laboratory, Department of Medical Elementology & Toxicology (DST-FIST and UGC-SAP-BSR funded department), Jamia Hamdard (Hamdard University), Hamdard Nagar, New Delhi 110062, India
| | - Andleeb Khan
- Neurotoxicology Laboratory, Department of Medical Elementology & Toxicology (DST-FIST and UGC-SAP-BSR funded department), Jamia Hamdard (Hamdard University), Hamdard Nagar, New Delhi 110062, India
| | - Mohd Ejaz Ahmed
- Neurotoxicology Laboratory, Department of Medical Elementology & Toxicology (DST-FIST and UGC-SAP-BSR funded department), Jamia Hamdard (Hamdard University), Hamdard Nagar, New Delhi 110062, India
| | - Mohammad Ashafaq
- Neurotoxicology Laboratory, Department of Medical Elementology & Toxicology (DST-FIST and UGC-SAP-BSR funded department), Jamia Hamdard (Hamdard University), Hamdard Nagar, New Delhi 110062, India
| | - M Badruzzaman Khan
- Neurotoxicology Laboratory, Department of Medical Elementology & Toxicology (DST-FIST and UGC-SAP-BSR funded department), Jamia Hamdard (Hamdard University), Hamdard Nagar, New Delhi 110062, India
| | - Farah Islam
- Department of Biotechnology, Faculty of Pharmacy, Jamia Hamdard (Hamdard University), Hamdard Nagar, New Delhi 110062, India
| | - Mohammed M Safhi
- Neuroscience and Toxicology Unit, Faculty of Pharmacy, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Fakhrul Islam
- Neurotoxicology Laboratory, Department of Medical Elementology & Toxicology (DST-FIST and UGC-SAP-BSR funded department), Jamia Hamdard (Hamdard University), Hamdard Nagar, New Delhi 110062, India.
| |
Collapse
|
35
|
Demir M, Amanvermez R, Kamalı Polat A, Karabıçak İ, Çınar H, Kesicioğlu T, Polat C. The effect of silymarin on mesenteric ischemia-reperfusion injury. Med Princ Pract 2014; 23:140-4. [PMID: 24356575 PMCID: PMC5586953 DOI: 10.1159/000356860] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 10/21/2013] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE To examine the effect of silymarin (SM), a mixture of flavonoids and polyphenols extracted from Silybum marianum, on mesenteric ischemia-reperfusion (I-R) injury in a rat model. MATERIALS AND METHODS Fifty rats were randomly divided into 5 groups (n = 10). Group 1 was sham operated, while groups 2-5 were subjected to mesenteric I-R lasting 1 h. Group 2 received isotonic sodium chloride, group 3 received SM (100 mg/kg/day) for 7 days before I-R, group 4 received SM for 7 days after I-R, and group 5 received SM for 7 days both before and after I-R. The rats were sacrificed by exsanguination in groups 1-3 at the 24th hour and groups 4 and 5 were sacrificed on the 7th day of reperfusion. Blood and intestinal specimens were taken for biochemical and pathological evaluations. RESULTS Serum superoxide dismutase (SOD) and heat shock protein 70 levels were significantly higher in group 2 (5.24 ± 1.76 U/l and 261.4 ± 16.8 ng/ml) compared to the sham group (2.08 ± 1.76 U/l and 189.9 ± 28.7 ng/ml) (p < 0.001 and p < 0.0001, respectively). However, SOD activity and the extent and severity of the histopathological lesions were significantly less in groups 3 [3.11 ± 1.18 U/l, 1.0 (range 0.0-2.0)], 4 [2.15 ± 0.87 U/l, 1.0 (range 1.0-3.0)], and 5 [1.80 ± 0.61 U/l, 0.5 (range 0.0-2.0)], treated with SM, than in group 2 [5.24 ± 1.76 U/l, 2.0 (range 2.0-3.0)] (p = 0.002, p < 0.001, and p = 0.0001; p < 0.001, p = 0.007, and p = 0.0001, respectively). Also, TNF-α levels were lower in the SM-supplemented groups compared to group 2. Serum thiobarbituric acid-reactive substance concentrations were low in the pre-/posttreatment groups treated with SM compared to group 2. No statistical difference was observed for protein carbonyls between the groups. CONCLUSION Our findings suggest that SM therapy may attenuate the oxidative and intestinal damage induced by I-R injuries.
Collapse
Affiliation(s)
- M. Demir
- Department of General Surgery, Ondokuz Mayıs University, Samsun, Turkey
| | - R. Amanvermez
- Department of Medical Biochemistry, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
- *Dr. Ramazan Amanvermez, Department of Medical Biochemistry, Faculty of Medicine, Ondokuz Mayıs University, TR-55139 Samsun (Turkey), E-Mail
| | - A. Kamalı Polat
- Department of General Surgery, Ondokuz Mayıs University, Samsun, Turkey
| | - İ. Karabıçak
- Department of General Surgery, Ondokuz Mayıs University, Samsun, Turkey
| | - H. Çınar
- Department of General Surgery, Ondokuz Mayıs University, Samsun, Turkey
| | - T. Kesicioğlu
- Department of General Surgery, Ondokuz Mayıs University, Samsun, Turkey
| | - C. Polat
- Department of General Surgery, Ondokuz Mayıs University, Samsun, Turkey
| |
Collapse
|
36
|
Borah A, Paul R, Choudhury S, Choudhury A, Bhuyan B, Das Talukdar A, Dutta Choudhury M, Mohanakumar KP. Neuroprotective potential of silymarin against CNS disorders: insight into the pathways and molecular mechanisms of action. CNS Neurosci Ther 2013; 19:847-53. [PMID: 24118806 PMCID: PMC6493565 DOI: 10.1111/cns.12175] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Revised: 08/07/2013] [Accepted: 08/07/2013] [Indexed: 12/14/2022] Open
Abstract
Silymarin, a C25 containing flavonoid from the plant Silybum marianum, has been the gold standard drug to treat liver disorders associated with alcohol consumption, acute and chronic viral hepatitis, and toxin-induced hepatic failures since its discovery in 1960. Apart from the hepatoprotective nature, which is mainly due to its antioxidant and tissue regenerative properties, Silymarin has recently been reported to be a putative neuroprotective agent against many neurologic diseases including Alzheimer's and Parkinson's diseases, and cerebral ischemia. Although the underlying neuroprotective mechanism of Silymarin is believed to be due to its capacity to inhibit oxidative stress in the brain, it also confers additional advantages by influencing pathways such as β-amyloid aggregation, inflammatory mechanisms, cellular apoptotic machinery, and estrogenic receptor mediation. In this review, we have elucidated the possible neuroprotective effects of Silymarin and the underlying molecular events, and suggested future courses of action for its acceptance as a CNS drug for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Anupom Borah
- Cellular and Molecular Neurobiology LaboratoryDepartment of Life Science and BioinformaticsAssam UniversitySilcharIndia
| | - Rajib Paul
- Cellular and Molecular Neurobiology LaboratoryDepartment of Life Science and BioinformaticsAssam UniversitySilcharIndia
| | - Sabanum Choudhury
- Cellular and Molecular Neurobiology LaboratoryDepartment of Life Science and BioinformaticsAssam UniversitySilcharIndia
| | - Amarendranath Choudhury
- Cellular and Molecular Neurobiology LaboratoryDepartment of Life Science and BioinformaticsAssam UniversitySilcharIndia
| | - Bornalee Bhuyan
- Ethnobotany and Medicinal Plant LaboratoryDepartment of Life Science and BioinformaticsAssam UniversitySilcharIndia
| | - Anupam Das Talukdar
- Ethnobotany and Medicinal Plant LaboratoryDepartment of Life Science and BioinformaticsAssam UniversitySilcharIndia
| | - Manabendra Dutta Choudhury
- Ethnobotany and Medicinal Plant LaboratoryDepartment of Life Science and BioinformaticsAssam UniversitySilcharIndia
- Assam University Biotech Hub (DBT)Assam UniversitySilcharIndia
| | - Kochupurackal P Mohanakumar
- Laboratory of Clinical & Experimental NeuroscienceDivision of Cell Biology & PhysiologyCSIR‐Indian Institute of Chemical BiologyJadavpurIndia
| |
Collapse
|
37
|
Muley MM, Thakare VN, Patil RR, Bafna PA, Naik SR. Amelioration of cognitive, motor and endogenous defense functions with silymarin, piracetam and protocatechuic acid in the cerebral global ischemic rat model. Life Sci 2013; 93:51-57. [DOI: 10.1016/j.lfs.2013.05.020] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 05/06/2013] [Accepted: 05/23/2013] [Indexed: 12/27/2022]
|
38
|
Protective Effect of Silymarin against Acrolein-Induced Cardiotoxicity in Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2012; 2012:352091. [PMID: 23320028 PMCID: PMC3535759 DOI: 10.1155/2012/352091] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 11/12/2012] [Accepted: 11/15/2012] [Indexed: 02/07/2023]
Abstract
Reactive α,β-unsaturated aldehydes such as acrolein (ACR) are major components of environmental pollutants and have been implicated in the neurodegenerative and cardiac diseases. In this study, the protective effect of silymarin (SN) against cardiotoxicity induced by ACR in mice was evaluated. Studies were performed on seven groups of six animals each, including vehicle-control (normal saline + 0.5% w/v methylcellulose), ACR (7.5 mg/kg/day, gavage) for 3 weeks, SN (25, 50 and 100 mg/kg/day, i.p.) plus ACR, vitamin E (Vit E, 100 IU/kg, i.p.) plus ACR, and SN (100 mg/kg, i.p.) groups. Mice received SN 7 days before ACR and daily thereafter throughout the study. Pretreatment with SN attenuated ACR-induced increased levels of malondialdehyde (MDA), serum cardiac troponin I (cTnI), and creatine kinase-MB (CK-MB), as well as histopathological changes in cardiac tissues. Moreover, SN improved glutathione (GSH) content, superoxide dismutase (SOD), and catalase (CAT) activities in heart of ACR-treated mice. Western blot analysis showed that SN pretreatment inhibited apoptosis provoked by ACR through decreasing Bax/Bcl-2 ratio, cytosolic cytochrome c content, and cleaved caspase-3 level in heart. In conclusion, SN may have protective effects against cardiotoxicity of ACR by reducing lipid peroxidation, renewing the activities of antioxidant enzymes, and preventing apoptosis.
Collapse
|
39
|
Maiese K, Chong ZZ, Wang S, Shang YC. Oxidant stress and signal transduction in the nervous system with the PI 3-K, Akt, and mTOR cascade. Int J Mol Sci 2012. [PMID: 23203037 PMCID: PMC3509553 DOI: 10.3390/ijms131113830] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Oxidative stress impacts multiple systems of the body and can lead to some of the most devastating consequences in the nervous system especially during aging. Both acute and chronic neurodegenerative disorders such as diabetes mellitus, cerebral ischemia, trauma, Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, and tuberous sclerosis through programmed cell death pathways of apoptosis and autophagy can be the result of oxidant stress. Novel therapeutic avenues that focus upon the phosphoinositide 3-kinase (PI 3-K), Akt (protein kinase B), and the mammalian target of rapamycin (mTOR) cascade and related pathways offer exciting prospects to address the onset and potential reversal of neurodegenerative disorders. Effective clinical translation of these pathways into robust therapeutic strategies requires intimate knowledge of the complexity of these pathways and the ability of this cascade to influence biological outcome that can vary among disorders of the nervous system.
Collapse
Affiliation(s)
- Kenneth Maiese
- Laboratory of Cellular and Molecular Signaling, Newark, NJ 07101, USA; E-Mails: (Z.Z.C.); (S.W.); (Y.C.S.)
- Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
- New Jersey Health Sciences University, 205 South Orange Avenue, Newark, NJ 07101, USA
- Author to whom correspondence should be addressed: E-Mail:
| | - Zhao Zhong Chong
- Laboratory of Cellular and Molecular Signaling, Newark, NJ 07101, USA; E-Mails: (Z.Z.C.); (S.W.); (Y.C.S.)
- New Jersey Health Sciences University, 205 South Orange Avenue, Newark, NJ 07101, USA
| | - Shaohui Wang
- Laboratory of Cellular and Molecular Signaling, Newark, NJ 07101, USA; E-Mails: (Z.Z.C.); (S.W.); (Y.C.S.)
- New Jersey Health Sciences University, 205 South Orange Avenue, Newark, NJ 07101, USA
| | - Yan Chen Shang
- Laboratory of Cellular and Molecular Signaling, Newark, NJ 07101, USA; E-Mails: (Z.Z.C.); (S.W.); (Y.C.S.)
- New Jersey Health Sciences University, 205 South Orange Avenue, Newark, NJ 07101, USA
| |
Collapse
|