1
|
Waheeb TS, Abdulkader MA, Ghareeb DA, Moustafa ME. Neuroprotective efficacy of berberine and caffeine against rotenone-induced neuroinflammatory and oxidative disturbances associated with Parkinson's disease via inhibiting α-synuclein aggregation and boosting dopamine release. Inflammopharmacology 2025; 33:2129-2150. [PMID: 40057928 PMCID: PMC11991993 DOI: 10.1007/s10787-025-01661-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 01/12/2025] [Indexed: 04/13/2025]
Abstract
Parkinson's disease (PD) is characterized by motor impairment, glial-mediated inflammation, redox imbalance, and α-synuclein (α-syn) aggregation. Conventional therapies relieve early PD symptoms, but they do not repair dopaminergic neurons. Berberine (BBR) and caffeine (CAF), both natural alkaloids, exhibited neuroprotective effects in many neurodegenerative disorders. Consequently, we hypothesized that the combination of BBR and CAF therapies would offer protection against PD-related impairments in the rotenone (ROT)-induced rat model when compared to the commercial drug, metformin (MTF). Our results showed that the combined administration of BBR (25 mg/kg/day) and CAF (2.5 mg/kg/day) for four weeks prevented motor deficits, weight reduction, dopamine (DA) depletion, and monoamine oxidase (MAO) activity in ROT-induced rats in comparison with monotherapy of BBR and CAF along with MTF. This combination produced a notable neuroprotective effect by reducing tumor necrosis factor (TNF)-α and interleukin-16 (IL-6) in midbrain of rats. BBR and CAF combinations markedly normalized tyrosine hydroxylase (TH) levels and decreased total α-syn and α-syn-pser129 aggregation and increased protein phosphatase 2A (PP2A) levels. Histological analysis indicated that damaged neurons exhibited significant amelioration with the co-administration of BBR and CAF. The molecular docking results indicated that both BBR and CAF had notable binding affinity for the protein pocket surrounding the α-syn, PP2A, and TH in comparison to MTF. They are predicted to serve as effective inhibitors of enzyme-mediated phosphorylation of α-syn-pser129. Conclusively, combined BBR and CAF administration presents a novel strategy for neuroprotection by blocking the initial events in PD incidence, demonstrating considerable anti-oxidative and anti-inflammatory benefits relative to MTF.
Collapse
Affiliation(s)
- Tasnim S Waheeb
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, 21511, Egypt.
| | - Mohammad A Abdulkader
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, 21511, Egypt
| | - Doaa A Ghareeb
- Bio-Screening and Preclinical Trial Lab, Biochemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
- Center of Excellence for Drug Preclinical Studies (CE-DPS), Pharmaceutical and Fermentation Industry Development Center, City of Scientific Research and Technological Applications (SRTA-City), New Borg El Arab, Alexandria, Egypt
- Research Projects Unit, Pharos University in Alexandria, Canal El Mahmoudia Street, Beside Green Plaza Complex, Alexandria, 21648, Egypt
| | - Mohamed E Moustafa
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, 21511, Egypt
| |
Collapse
|
2
|
Soni D, Garg Y, Upadhayay S, Bhatia A, Basir B, Singh SK, Dua K, Kumar P. Auranofin-loaded chitosan-lipid hybrid nanoparticle protects against rotenone model of Parkinson's disease via modulation of GSK-3β/ Nrf2/HO-1 signaling. Eur J Pharmacol 2025; 998:177523. [PMID: 40107337 DOI: 10.1016/j.ejphar.2025.177523] [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: 12/09/2024] [Revised: 03/15/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025]
Abstract
Auranofin (AUF) is a gold-based compound that has demonstrated a wide range of biological effects, such as anti-inflammatory and antibacterial effects. However, the neuronal use of AUF is restricted due to its low bioavailability. Thus, to improve blood brain barrier (BBB) penetration and investigate its antiparkinsonian impact, the researchers developed AUF-loaded hybrid nanoparticles (AUFHNPs). This research delves into the neuroprotective potential of AUFHNPs against rotenone-induced Parkinson's disease (PD). The MTT assay, Acridine orange/Ethidium bromide (AO/EB) staining, RT-PCR, and Western blot analysis were performed on SH-SY5Y lines. Also, AUFHNPs were prepared and characterized. For the in-vivo study, AUF, its NPs and rotenone were administered for 28 days, and behavioral parameters were performed on day 27 and 28. On the 29th day, animals were sacrificed, and brains were isolated for biochemical assessment, apoptotic and inflammatory markers evaluation, histopathology, and molecular examination. In-vitro results showed that AUF significantly restored cell viability and reduced apoptosis. Spherical-shaped NPs were observed under FE-SEM/TEM analysis. Administration of AUFHNPs in rats significantly restored motor activity and neuronal morphological changes by phosphorylating GSK-3β to increase the expression of Nrf2/HO-1. This study concludes that developing AUFHNPs increases AUF's bioavailability in the brain and exerts neuroprotection via modulating GSK-3β/Nrf2/HO-1 pathways.
Collapse
Affiliation(s)
- Divya Soni
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Yogesh Garg
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab, India
| | - Shubham Upadhayay
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Amit Bhatia
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab, India
| | - Bushra Basir
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia; School of Medical and Life Sciences, Sunway University, 47500, Sunway City, Malaysia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative, Medicine, University of Technology Sydney, Ultimo, NSW, Australia
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab, India.
| |
Collapse
|
3
|
Sharma V, Sharma P, Singh TG. Leukotriene signaling in neurodegeneration: implications for treatment strategies. Inflammopharmacology 2024; 32:3571-3584. [PMID: 39167313 DOI: 10.1007/s10787-024-01557-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 08/11/2024] [Indexed: 08/23/2024]
Abstract
Leukotrienes (LTs) are a group of substances that cause inflammation. They are produced by the enzyme 5-lipoxygenase (5-LOX) from arachidonic acid. Cysteinyl LTs are a group of lipid molecules that have a prominent role in inflammatory signaling in the allergic diseases. Although they are traditionally known for their role in allergic disease, current advancements in bio-medical research have shed light on the involvement of these inflammatory mediators in diseases such as in the inflammation related to central nervous system (CNS) disorders. Among the CNS diseases, LTs, along with 5-LOX and their receptors, have been shown to be associated with multiple sclerosis (MS), Alzheimer's disease (AD), and Parkinson's disease (PD). Through a comprehensive review of current research and experimentation, this investigation provides an insight on the biosynthesis, receptors, and biological effects of LTs in the body. Furthermore, implications of leukotriene signaling in CNS and its intricate role in neurodegeneration are also studied. Through the revelation of these insights, our aim is to establish a foundation for the development of enhanced and focused therapeutic approaches in the continuous endeavor to combat neurodegeneration. Furthermore, the pharmacological inhibition of leukotriene signaling with selective inhibitors offers promising prospects for future interventions and treatments for neurodegenerative diseases.
Collapse
Affiliation(s)
- Veerta Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Prateek Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| |
Collapse
|
4
|
Scorziello A, Sirabella R, Sisalli MJ, Tufano M, Giaccio L, D’Apolito E, Castellano L, Annunziato L. Mitochondrial Dysfunction in Parkinson's Disease: A Contribution to Cognitive Impairment? Int J Mol Sci 2024; 25:11490. [PMID: 39519043 PMCID: PMC11546611 DOI: 10.3390/ijms252111490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/05/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Among the non-motor symptoms associated with Parkinson's disease (PD), cognitive impairment is one of the most common and disabling. It can occur either early or late during the disease, and it is heterogeneous in terms of its clinical manifestations, such as Subjective Cognitive Dysfunction (SCD), Mild Cognitive Impairment (MCI), and Parkinson's Disease Dementia (PDD). The aim of the present review is to delve deeper into the molecular mechanisms underlying cognitive decline in PD. This is extremely important to delineate the guidelines for the differential diagnosis and prognosis of the dysfunction, to identify the molecular and neuronal mechanisms involved, and to plan therapeutic strategies that can halt cognitive impairment progression. Specifically, the present review will discuss the pathogenetic mechanisms involved in the progression of cognitive impairment in PD, with attention to mitochondria and their contribution to synaptic dysfunction and neuronal deterioration in the brain regions responsible for non-motor manifestations of the disease.
Collapse
Affiliation(s)
- Antonella Scorziello
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.S.); (M.T.); (L.G.); (E.D.); (L.C.)
| | - Rossana Sirabella
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.S.); (M.T.); (L.G.); (E.D.); (L.C.)
| | - Maria Josè Sisalli
- Department of Translational Medicine, Federico II University of Naples, 80138 Napoli, Italy;
| | - Michele Tufano
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.S.); (M.T.); (L.G.); (E.D.); (L.C.)
| | - Lucia Giaccio
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.S.); (M.T.); (L.G.); (E.D.); (L.C.)
| | - Elena D’Apolito
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.S.); (M.T.); (L.G.); (E.D.); (L.C.)
| | - Lorenzo Castellano
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.S.); (M.T.); (L.G.); (E.D.); (L.C.)
| | | |
Collapse
|
5
|
Rafeeq M, Al-Abbasi FA, Afzal M, Moglad E, Al-Qahtani SD, Alzrea SI, Almalki NAR, Imam F, Sayyed N, Kazmi I. 6-Shogaol Abrogates Parkinson's Disease in Rotenone-Induced Rodents: Based on In Silico Study and Inhibiting TNF-α/NF-κB/IL-1β/MAO-B. Pharmaceuticals (Basel) 2024; 17:1348. [PMID: 39458989 PMCID: PMC11510247 DOI: 10.3390/ph17101348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: 6-Shogaol is a comparatively innovative anti-Parkinson's remedy with antioxidant and anti-inflammatory characteristics. This investigation intended to determine the role of 6-shogaol in the Parkinson's disease (PD) paradigm in rotenone-induced rats. Methods: Thirty male Wistar rats (10-12 weeks old; 180 ± 20 g) were divided into five groups. Animals with rotenone-induced experimental PD were subsequently treated with 6-shogaol-10 at 20 mg/kg for 28 days. After the experimental duration, behavioural investigations were performed, i.e., open field test, forced swim test, rotarod test, and catalepsy test. Biochemical assessments like AChE, GSH, CAT, SOD, MDA, nitrite, ceruloplasmin, proinflammatory markers such as IL-1β, NF-κB, TNF-α, and catecholamines markers (DA, GABA, and MAO-B) were determined. The docking procedure was conducted using the AutoDock Vina docking protocol. Furthermore, histopathology was performed. Results: Rotenone significantly increased the level of MAO-B, oxidative, nitrative, and pro-inflammatory markers. However, there was a decline in ceruloplasmin, dopamine, and endogenous antioxidants. Treatment with 6-shogaol (10 and 20 mg/kg) considerably sustained the elevation of oxidative stress and inflammatory indicators and decreased AChE activity and dopamine levels. In the histology of the brain, 6-shogaol improved the neuronal structure and reduced the degeneration of neurons. Based on the binding energy values, compound 6-shogaol demonstrates a favourable binding affinity to AChE, MAO-B, DA, and GABA with respective binding energies of -8.214, -8.133, -7.396 and -6.189 kcal/mol. Conclusions: In this study, 6-shogaol exhibited neuroprotective properties against PD, which could be employed as a prospective medication for PD.
Collapse
Affiliation(s)
- Misbahuddin Rafeeq
- Department of Pharmacology Faculty of Medicine, Rabigh King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Fahad A. Al-Abbasi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, Jeddah 21442, Saudi Arabia;
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam bin Abdulaziz University, Alkharj 11942, Saudi Arabia;
| | - Salwa D. Al-Qahtani
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah 11952, Saudi Arabia
| | - Sami I. Alzrea
- Department of Pharmacology, College of Pharmacy, Jouf University, Aljouf, Sakaka 72341, Saudi Arabia;
| | - Naif A. R. Almalki
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Faisal Imam
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Nadeem Sayyed
- School of Pharmacy, Glocal University, Saharanpur 247121, India
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
6
|
Puri S, Kirad S, Muzaffar-Ur-Rehman M, Mandal SK, Sharma PK, Sankaranarayanan M, Deepa PR. Lipogenic stearoyl-CoA desaturase-1 (SCD1) targeted virtual screening for chemical inhibitors: molecular docking / dynamics simulation and in vitro assessment of anti-NAFLD efficacy. RSC Adv 2024; 14:31797-31808. [PMID: 39380655 PMCID: PMC11459445 DOI: 10.1039/d4ra06037g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/01/2024] [Indexed: 10/10/2024] Open
Abstract
Amidst rising global prevalence of metabolic syndrome, the associated risk of non-alcoholic fatty liver disease (NAFLD) is also rapidly increasing. The pathogenesis of NAFLD starts with fat accumulation and progresses through inflammation and fibrotic sequel, often involving complex molecular mechanisms involving de novo lipogenesis. Stearoyl-CoA desaturase 1 (SCD1) enzyme, expressed in liver and adipose tissue, converts saturated fatty acids to monounsaturated fatty acids (MUFAs), contributing to triglyceride and cholesterol ester formation. In this study, potential SCD1 inhibitors were screened using the ZINC database of curated medically-approved drugs by virtual screening, molecular docking, and molecular dynamics simulations. The top-scoring five ligands with strong binding affinity against SCD1 were ZINC000003831151 > ZINC000001540998 > ZINC000003830713 > ZINC000000897251 > ZINC000002005305, which showed stable protein-ligand complexation and favorable pharmacokinetic attributes. The top ligand, Montelukast, was experimentally validated for its pharmacological efficacy in an in vitro cell culture model of steatosis (NAFLD). Montelukast showed a dose-dependent decrease in hepatic fat accumulation, reduced levels of free radicals, and lowered oxidative stress (P < 0.05). These outcomes suggest Montelukast to be a potential SCD1 inhibitor, with anti-NAFLD efficacy. These findings open new avenues for therapeutic development of the top 5 ligands in metabolic disorders involving SCD1.
Collapse
Affiliation(s)
- Sonakshi Puri
- Biochemistry and Enzyme Biotechnology Lab, Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Pilani Campus Pilani-333031 Rajasthan India +91 1596 255881
| | - Shivani Kirad
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus Pilani-333031 Rajasthan India +91-1596-255717
| | - Mohammed Muzaffar-Ur-Rehman
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus Pilani-333031 Rajasthan India +91-1596-255717
| | - Sumit Kumar Mandal
- Biochemistry and Enzyme Biotechnology Lab, Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Pilani Campus Pilani-333031 Rajasthan India +91 1596 255881
| | - Pankaj Kumar Sharma
- Biochemistry and Enzyme Biotechnology Lab, Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Pilani Campus Pilani-333031 Rajasthan India +91 1596 255881
| | - Murugesan Sankaranarayanan
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus Pilani-333031 Rajasthan India +91-1596-255717
| | - P R Deepa
- Biochemistry and Enzyme Biotechnology Lab, Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Pilani Campus Pilani-333031 Rajasthan India +91 1596 255881
| |
Collapse
|
7
|
Sood R, Anoopkumar-Dukie S, Rudrawar S, Hall S. Neuromodulatory effects of leukotriene receptor antagonists: A comprehensive review. Eur J Pharmacol 2024; 978:176755. [PMID: 38909933 DOI: 10.1016/j.ejphar.2024.176755] [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: 02/19/2024] [Revised: 06/09/2024] [Accepted: 06/16/2024] [Indexed: 06/25/2024]
Abstract
Cysteinyl leukotrienes (CysLTs) are central to the pathophysiology of asthma and various inflammatory disorders. Leukotriene receptor antagonists (LTRAs) effectively treat respiratory conditions by targeting cysteinyl leukotriene receptors, CysLT1 and CysLT2 subtypes. This review explores the multifaceted effects of LTs, extending beyond bronchoconstriction. CysLT receptors are not only present in the respiratory system but are also crucial in neuronal signaling pathways. LTRAs modulate these receptors, influencing downstream signaling, calcium levels, inflammation, and oxidative stress (OS) within neurons hinting at broader implications. Recent studies identify novel molecular targets, sparking interest in repurposing LTRAs for therapeutic use. Clinical trials are investigating their potential in neuroinflammation control, particularly in Alzheimer's disease (AD) and Parkinson's diseases (PD). However, montelukast, a long-standing LTRA since 1998, raises concerns due to neuropsychiatric adverse drug reactions (ADRs). Despite widespread use, understanding montelukast's metabolism and underlying ADR mechanisms remains limited. This review comprehensively examines LTRAs' diverse biological effects, emphasizing non-bronchoconstrictive activities. It also analyses plausible mechanisms behind LTRAs' neuronal effects, offering insights into their potential as neurodegenerative disease modulators. The aim is to inform clinicians, researchers, and pharmaceutical developers about LTRAs' expanding roles, particularly in neuroinflammation control and their promising repurposing for neurodegenerative disease management.
Collapse
Affiliation(s)
- Radhika Sood
- School of Pharmacy and Medical Sciences, Griffith University, Queensland, 4222, Australia
| | | | - Santosh Rudrawar
- School of Pharmacy and Medical Sciences, Griffith University, Queensland, 4222, Australia; Institute for Glycomics, Griffith University, Queensland, 4222, Australia
| | - Susan Hall
- School of Pharmacy and Medical Sciences, Griffith University, Queensland, 4222, Australia.
| |
Collapse
|
8
|
Zhang X, Liu H, Xiu X, Cheng J, Li T, Wang P, Men L, Qiu J, Jin Y, Zhao J. Exosomal GDNF from Bone Marrow Mesenchymal Stem Cells Moderates Neuropathic Pain in a Rat Model of Chronic Constriction Injury. Neuromolecular Med 2024; 26:34. [PMID: 39167282 DOI: 10.1007/s12017-024-08800-6] [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: 06/21/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024]
Abstract
Both of exosomes derived from mesenchymal stem cells (MSCs) and glial cell line-derived neurotrophic factor (GDNF) show potential for the treatment of neuropathic pain. Here, the analgesic effects of exosomes derived from bone marrow MSCs (BMSCs) were investigated. BMSCs-derived exosomes were isolated and characterized. Chronic constriction injury (CCI) was constructed to induce neuropathic pain in rats, which were then treated with exosomes. Pain behaviors were evaluated by measuring paw withdrawal thresholds and latency. The changes of key proteins, including cytokines, were explored using Western blot and ELISA. Administration of BMSCs-derived exosomes alleviated neuropathic pain, as demonstrated by the decrease of thermal hyperalgesia and mechanical allodynia, as well as the reduced secretion of pro-inflammatory cytokines in CCI rats. These effects were comparable to the treatment of GDNF alone. Mechanically, the exosomes suppressed the CCI-induced activation of TLR2/MyD88/NF-κB signaling pathway, while GDNF knockdown impaired their analgesic effects on CCI rat. BMSCs-derived exosomes may alleviate CCI-induced neuropathic pain and inflammation in rats by transporting GDNF.
Collapse
Affiliation(s)
- Xuelei Zhang
- Graduate School, Hebei University of Chinese Medicine, No. 3 Xingyuan Road, Luquan District, Shijiazhuang, 050200, Hebei, China.
- Department of Hand Microsurgery, The Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Cangzhou, 061000, Hebei, China.
- Hebei Key Laboratory of Intergraded Traditional and Western Medicine in Osteoarthrosis Research (Preparing), Cangzhou, 061000, Hebei, China.
| | - Huan Liu
- Graduate School, Hebei University of Chinese Medicine, No. 3 Xingyuan Road, Luquan District, Shijiazhuang, 050200, Hebei, China
| | - Xiaolei Xiu
- Department of Hand Microsurgery, The Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Cangzhou, 061000, Hebei, China
| | - Jibo Cheng
- Chengde Medical University, Anyuan Road, Chengde, 067000, Hebei, China
| | - Tong Li
- Department of Hand Microsurgery, The Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Cangzhou, 061000, Hebei, China
| | - Ping Wang
- Department of Hand Microsurgery, The Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Cangzhou, 061000, Hebei, China
| | - Lili Men
- Department of Hand Microsurgery, The Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Cangzhou, 061000, Hebei, China
| | - Junru Qiu
- Department of Hand Microsurgery, The Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Cangzhou, 061000, Hebei, China
| | - Yanyan Jin
- Department of Hand Microsurgery, The Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Cangzhou, 061000, Hebei, China
| | - Jianyong Zhao
- Department of Hand Microsurgery, The Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Cangzhou, 061000, Hebei, China.
| |
Collapse
|
9
|
Tseng HC, Wang MH, Fang CH, Lin YW, Soung HS. Neuroprotective Potentials of Berberine in Rotenone-Induced Parkinson's Disease-like Motor Symptoms in Rats. Brain Sci 2024; 14:596. [PMID: 38928596 PMCID: PMC11201892 DOI: 10.3390/brainsci14060596] [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: 05/07/2024] [Revised: 05/24/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Rotenone (RTN) induces neurotoxicity and motor dysfunction in rats, mirroring the pathophysiological traits of Parkinson's disease (PD), including striatal oxidative stress, mitochondrial dysfunction, and changes in neural structure. This makes RTN a valuable model for PD research. Berberine (BBR), an isoquinoline alkaloid recognized for its antioxidative, anti-inflammatory, and neuroprotective properties, was evaluated for its ability to counteract RTN-induced impairments. Rats received subcutaneous RTN at 0.5 mg/kg for 21 days, resulting in weight loss and significant motor deficits assessed through open-field, bar catalepsy, beam-crossing, rotarod, and grip strength tests. BBR, administered orally at 30 or 100 mg/kg doses, one hour prior to RTN exposure for the same duration, effectively mitigated many of the RTN-induced motor impairments. Furthermore, BBR treatment reduced RTN-induced nitric oxide (NO) and lipid peroxidation (LPO) levels, bolstered antioxidative capacity, enhanced mitochondrial enzyme activities (e.g., succinate dehydrogenase (SDH), ATPase, and the electron transport chain (ETC)), and diminished striatal neuroinflammation and apoptosis markers. Notably, the co-administration of trigonelline (TGN), an inhibitor of the nuclear factor erythroid-2-related factor 2 (Nrf2) pathway, significantly attenuated BBR's protective effects, indicating that BBR's neuroprotective actions are mediated via the Nrf2 pathway. These results underscore BBR's potential in ameliorating motor impairments akin to PD, suggesting its promise in potentially delaying or managing PD symptoms. Further research is warranted to translate these preclinical findings into clinical settings, enhancing our comprehension of BBR's therapeutic prospects in PD.
Collapse
Affiliation(s)
- Hsiang-Chien Tseng
- Department of Anesthesiology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 11101, Taiwan
- School of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan
| | - Mao-Hsien Wang
- Department of Anesthesia, En Chu Kon Hospital, Sanshia District, New Taipei City 23702, Taiwan
| | - Chih-Hsiang Fang
- Department of Orthopedics, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Yi-Wen Lin
- Institute of Biomedical Engineering, National Taiwan University, Taipei 10051, Taiwan
| | - Hung-Sheng Soung
- Department of Psychiatry, Yuan-Shan Branch of Taipei Veteran General Hospital, No. 386, Rongguang Rd., Neicheng, Yuanshan Township, Yilan 26604, Taiwan
- Department of Biomedical Engineering, National Defense Medical Center, Taipei 11490, Taiwan
| |
Collapse
|
10
|
Manuel AM, Gottlieb A, Freeman L, Zhao Z. Montelukast as a repurposable additive drug for standard-efficacy multiple sclerosis treatment: Emulating clinical trials with retrospective administrative health claims data. Mult Scler 2024; 30:696-706. [PMID: 38660773 PMCID: PMC11073911 DOI: 10.1177/13524585241240398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
BACKGROUND Effective and safe treatment options for multiple sclerosis (MS) are still needed. Montelukast, a leukotriene receptor antagonist (LTRA) currently indicated for asthma or allergic rhinitis, may provide an additional therapeutic approach. OBJECTIVE The study aimed to evaluate the effects of montelukast on the relapses of people with MS (pwMS). METHODS In this retrospective case-control study, two independent longitudinal claims datasets were used to emulate randomized clinical trials (RCTs). We identified pwMS aged 18-65 years, on MS disease-modifying therapies concomitantly, in de-identified claims from Optum's Clinformatics® Data Mart (CDM) and IQVIA PharMetrics® Plus for Academics. Cases included 483 pwMS on montelukast and with medication adherence in CDM and 208 in PharMetrics Plus for Academics. We randomly sampled controls from 35,330 pwMS without montelukast prescriptions in CDM and 10,128 in PharMetrics Plus for Academics. Relapses were measured over a 2-year period through inpatient hospitalization and corticosteroid claims. A doubly robust causal inference model estimated the effects of montelukast, adjusting for confounders and censored patients. RESULTS pwMS treated with montelukast demonstrated a statistically significant 23.6% reduction in relapses compared to non-users in 67.3% of emulated RCTs. CONCLUSION Real-world evidence suggested that montelukast reduces MS relapses, warranting future clinical trials and further research on LTRAs' potential mechanism in MS.
Collapse
Affiliation(s)
- Astrid M Manuel
- Center for Precision Health, McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, TX
| | - Assaf Gottlieb
- Center for Precision Health, McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, TX
| | - Leorah Freeman
- Neurology Department, Dell Medical School, The University of Texas at Austin, TX
| | - Zhongming Zhao
- Center for Precision Health, McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, TX
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, TX
| |
Collapse
|
11
|
Farid HA, Sayed RH, El-Shamarka MES, Abdel-Salam OME, El Sayed NS. PI3K/AKT signaling activation by roflumilast ameliorates rotenone-induced Parkinson's disease in rats. Inflammopharmacology 2024; 32:1421-1437. [PMID: 37541971 PMCID: PMC11006765 DOI: 10.1007/s10787-023-01305-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 07/20/2023] [Indexed: 08/06/2023]
Abstract
Parkinson's disease (PD) is the second most common progressive age-related neurodegenerative disorder. Paramount evidence shed light on the role of PI3K/AKT signaling activation in the treatment of neurodegenerative disorders. PI3K/AKT signaling can be activated via cAMP-dependent pathways achieved by phosphodiesterase 4 (PDE4) inhibition. Roflumilast is a well-known PDE4 inhibitor that is currently used in the treatment of chronic obstructive pulmonary disease. Furthermore, roflumilast has been proposed as a favorable candidate for the treatment of neurological disorders. The current study aimed to unravel the neuroprotective role of roflumilast in the rotenone model of PD in rats. Ninety male rats were allocated into six groups as follows: control, rotenone (1.5 mg/kg/48 h, s.c.), L-dopa (22.5 mg/kg, p.o), and roflumilast (0.2, 0.4 or 0.8 mg/kg, p.o). All treatments were administrated for 21 days 1 h after rotenone injection. Rats treated with roflumilast showed an improvement in motor activity and coordination as well as preservation of dopaminergic neurons in the striatum. Moreover, roflumilast increased cAMP level and activated the PI3K/AKT axis via stimulation of CREB/BDNF/TrkB and SIRT1/PTP1B/IGF1 signaling cascades. Roflumilast also caused an upsurge in mTOR and Nrf2, halted GSK-3β and NF-ĸB, and suppressed FoxO1 and caspase-3. Our study revealed that roflumilast exerted neuroprotective effects in rotenone-induced neurotoxicity in rats. These neuroprotective effects were mediated via the crosstalk between CREB/BDNF/TrkB and SIRT1/PTP1B/IGF1 signaling pathways which activates PI3K/AKT trajectory. Therefore, PDE4 inhibition is likely to offer a reliable persuasive avenue in curing PD via PI3K/AKT signaling activation.
Collapse
Affiliation(s)
- Heba A Farid
- Department of Narcotics, Ergogenic Aids and Poisons, National Research Centre, Cairo, Egypt
| | - Rabab H Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini St., Cairo, 11562, Egypt.
| | | | - Omar M E Abdel-Salam
- Department of Narcotics, Ergogenic Aids and Poisons, National Research Centre, Cairo, Egypt
| | - Nesrine S El Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini St., Cairo, 11562, Egypt
| |
Collapse
|
12
|
Wang X, Gan W, Kang M, Lv C, Zhao Z, Wu Y, Zhang X, Wang R. Asthma aggravates alzheimer's disease by up-regulating NF- κB signaling pathway through LTD4. Brain Res 2024; 1825:148711. [PMID: 38092296 DOI: 10.1016/j.brainres.2023.148711] [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: 08/22/2023] [Revised: 11/17/2023] [Accepted: 12/09/2023] [Indexed: 12/25/2023]
Abstract
Clinical studies have shown that asthma is a risk factor for dementia or Alzheimer's disease (AD). To investigate whether asthma aggravates AD in APP/PS1 mice and explore the potential mechanisms, an asthma model was established using six-month-old APP/PS1 mice, and montelukast was used as a therapeutic agent in APP/PS1 mice with asthma. The Morris water maze test showed that asthma aggravates spatial learning and memory abilities. Asthma also upregulates the NF-κB inflammatory pathway in APP/PS1 mice and promotes the expression of beta-site amyloid precursor protein cleaving enzyme 1 (BACE1), amyloid-β (Aβ) deposition, neuronal damage, synaptic plasticity deficiency, activation of microglia and astrocytes. The level of LTD4 and its receptor CysLT1R in the hippocampus of APP/PS1 mice after the asthma modeling was established was higher than that in APP/PS1 mice, suggesting that asthma may affect the pathology of AD through LTD4 and its receptor Cys-LT1R. Montelukast ameliorates these pathological changes and cognitive impairment. These results suggest that asthma aggravates AD pathology and cognitive impairment of APP/PS1 mice via upregulation of the NF-κB inflammatory pathway, and montelukast ameliorates these pathological changes.
Collapse
Affiliation(s)
- Xiaozhen Wang
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing Geriatric Medical Research Center, Key Laboratory for Neurodegenerative Disease of Ministry of Education, Beijing, PR China
| | - Wenjing Gan
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing Geriatric Medical Research Center, Key Laboratory for Neurodegenerative Disease of Ministry of Education, Beijing, PR China
| | - Meimei Kang
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing Geriatric Medical Research Center, Key Laboratory for Neurodegenerative Disease of Ministry of Education, Beijing, PR China
| | - Caizhen Lv
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing Geriatric Medical Research Center, Key Laboratory for Neurodegenerative Disease of Ministry of Education, Beijing, PR China
| | - Zhiwei Zhao
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing Geriatric Medical Research Center, Key Laboratory for Neurodegenerative Disease of Ministry of Education, Beijing, PR China
| | - Yanchuan Wu
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing Geriatric Medical Research Center, Key Laboratory for Neurodegenerative Disease of Ministry of Education, Beijing, PR China
| | - Xu Zhang
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing Geriatric Medical Research Center, Key Laboratory for Neurodegenerative Disease of Ministry of Education, Beijing, PR China
| | - Rong Wang
- Central Laboratory, Xuanwu Hospital, Capital Medical University, Beijing Geriatric Medical Research Center, Key Laboratory for Neurodegenerative Disease of Ministry of Education, Beijing, PR China; Beijing Institute for Brain Disorders, Beijing, PR China; National Clinical Research Center for Geriatric Disorders, Beijing, PR China.
| |
Collapse
|
13
|
Bano A, Garg A, Mumtaz, Nidhi, Ashif Khan M. The anti-seizure potential of cysteine leukotriene receptor antagonists: A systematic review of animal studies. Epilepsy Res 2024; 200:107305. [PMID: 38325237 DOI: 10.1016/j.eplepsyres.2024.107305] [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: 08/21/2023] [Revised: 12/12/2023] [Accepted: 01/12/2024] [Indexed: 02/09/2024]
Abstract
BACKGROUND Emerging literature has suggested the antiepileptic activity of cysteine leukotriene receptor (CysLTR) antagonists in experimental animals of epilepsy. Leukotrienes are substances that cause inflammation and affect brain activity, blood flow, oxidation, and inflammation in the brain. These processes are related to epilepsy and its complications. CysLTR antagonists are drugs that prevent leukotrienes from working. They may be useful for treating epilepsy, especially for people who do not respond to other drugs. Therefore, the current study aims to systematically review the potential anti-seizure effect of CysLTR antagonists in experimental studies. METHOD We systematically reviewed the online databases using online databases such as Google Scholar, science direct, and PubMed until December 2022 to identify experimental studies assessing the anti-seizure activity of CysLTR antagonists. The Systematic Review Centre for Laboratory Animal Experimentation (SYRCLE) was used to evaluate the risk of bias (RoB) of the included studies. RESULTS Initially we identified 3823 studies. After screening using inclusion and exclusion criteria, 8 studies were finally included in the current study. All included studies, reported that CysLTR antagonists reduced the intensity of seizures in animal models of epilepsy. CONCLUSION In conclusion, CysLTR antagonists could be a potential therapeutic approach for the treatment of epilepsy. However, further preclinical and clinical studies are required to confirm their efficacy, safety, and mechanism of anti-seizure activity.
Collapse
Affiliation(s)
- Aysha Bano
- Department of Translational and Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard New Delhi, 110062, India
| | - Aakriti Garg
- Department of Translational and Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard New Delhi, 110062, India; Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mumtaz
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Nidhi
- Department of Translational and Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard New Delhi, 110062, India
| | - Mohd Ashif Khan
- Department of Translational and Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard New Delhi, 110062, India.
| |
Collapse
|
14
|
Xue J, Tao K, Wang W, Wang X. What Can Inflammation Tell Us about Therapeutic Strategies for Parkinson's Disease? Int J Mol Sci 2024; 25:1641. [PMID: 38338925 PMCID: PMC10855787 DOI: 10.3390/ijms25031641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder with a complicated etiology and pathogenesis. α-Synuclein aggregation, dopaminergic (DA) neuron loss, mitochondrial injury, oxidative stress, and inflammation are involved in the process of PD. Neuroinflammation has been recognized as a key element in the initiation and progression of PD. In this review, we summarize the inflammatory response and pathogenic mechanisms of PD. Additionally, we describe the potential anti-inflammatory therapies, including nod-like receptor pyrin domain containing protein 3 (NLRP3) inflammasome inhibition, nuclear factor κB (NF-κB) inhibition, microglia inhibition, astrocyte inhibition, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibition, the peroxisome proliferator-activated receptor γ (PPARγ) agonist, targeting the mitogen-activated protein kinase (MAPK) pathway, targeting the adenosine monophosphate-activated protein kinase (AMPK)-dependent pathway, targeting α-synuclein, targeting miRNA, acupuncture, and exercise. The review focuses on inflammation and will help in designing new prevention strategies for PD.
Collapse
Affiliation(s)
- Jinsong Xue
- School of Biology, Food and Environment, Hefei University, Hefei 230601, China; (K.T.); (W.W.)
| | | | | | - Xiaofei Wang
- School of Biology, Food and Environment, Hefei University, Hefei 230601, China; (K.T.); (W.W.)
| |
Collapse
|
15
|
Hu K, Zhu S, Wu F, Zhang Y, Li M, Yuan L, Huang W, Zhang Y, Wang J, Ren J, Yang H. Aureusidin ameliorates 6-OHDA-induced neurotoxicity via activating Nrf2/HO-1 signaling pathway and preventing mitochondria-dependent apoptosis pathway in SH-SY5Y cells and Caenorhabditis elegans. Chem Biol Interact 2024; 387:110824. [PMID: 38056806 DOI: 10.1016/j.cbi.2023.110824] [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: 10/08/2023] [Revised: 11/22/2023] [Accepted: 11/30/2023] [Indexed: 12/08/2023]
Abstract
Movement disorder Parkinson's disease (PD) is the second most common neurodegenerative disease in the world after Alzheimer's disease, which severely affects the quality of patients' lives and imposes an increasingly heavy socioeconomic burden. Aureusidin is a kind of natural flavonoid compound with anti-inflammatory and anti-oxidant activities, while its pharmacological action and mechanism are rarely reported in PD. This study aimed to explore the neuroprotective effects and potential mechanisms of Aureusidin in PD. The present study demonstrated that Aureusidin protected SH-SY5Y cells from cell damage induced by 6-hydroxydopamine (6-OHDA) via inhibiting the mitochondria-dependent apoptosis and activating the Nrf2/HO-1 antioxidant signaling pathway. Additionally, Aureusidin diminished dopaminergic (DA) neuron degeneration induced by 6-OHDA and reduced the aggregation toxicity of α-synuclein (α-Syn) in Caenorhabditis elegans (C. elegans.) In conclusion, Aureusidin showed a neuroprotective effect in the 6-OHDA-induced PD model via activating Nrf2/HO-1 signaling pathway and prevented mitochondria-dependent apoptosis pathway, and these findings suggested that Aureusidin may be an effective drug for the treatment of PD.
Collapse
Affiliation(s)
- Kun Hu
- School of Pharmacy, Changzhou University, Changzhou, China
| | - Susu Zhu
- School of Pharmacy, Changzhou University, Changzhou, China
| | - Fanyu Wu
- School of Pharmacy, Changzhou University, Changzhou, China
| | - Yongzhen Zhang
- School of Pharmacy, Changzhou University, Changzhou, China
| | - Minyue Li
- School of Pharmacy, Changzhou University, Changzhou, China
| | - Ling Yuan
- School of Pharmacy, Changzhou University, Changzhou, China
| | - Wenjing Huang
- School of Pharmacy, Changzhou University, Changzhou, China
| | - Yichi Zhang
- School of Pharmacy, Changzhou University, Changzhou, China
| | - Jie Wang
- School of Pharmacy, Changzhou University, Changzhou, China
| | - Jie Ren
- School of Pharmacy, Changzhou University, Changzhou, China.
| | - Hao Yang
- Department of Pharmacy, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China.
| |
Collapse
|
16
|
Patel B, Greenland JC, Williams-Gray CH. Clinical Trial Highlights: Anti-Inflammatory and Immunomodulatory Agents. JOURNAL OF PARKINSON'S DISEASE 2024; 14:1283-1300. [PMID: 39331111 PMCID: PMC11492043 DOI: 10.3233/jpd-240353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/13/2024] [Indexed: 09/28/2024]
Abstract
Inflammation and immune dysregulation have been linked to the pathogenesis and progression of Parkinson's disease (PD), and represent an attractive target for therapeutic intervention, given the potential for repurposing of existing anti-inflammatory and immunomodulatory agents. Despite the fact that initial studies of drugs with secondary anti-inflammatory effects did not yield positive results, agents specifically targeting immune and inflammatory pathways may hold more promise. This article will briefly review the evidence base for targeting the immune system and neuroinflammation in PD, and discuss in detail the recently completed and currently active trials of primary anti-inflammatory/immunomodulatory drugs in PD.
Collapse
Affiliation(s)
- Bina Patel
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK
| | - Julia C. Greenland
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK
| | - Caroline H. Williams-Gray
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK
| |
Collapse
|
17
|
Yerraguravagari B, Penchikala NP, Kolusu AS, Ganesh GS, Konduri P, Nemmani KVS, Samudrala PK. Montelukast Ameliorates Scopolamine-induced Alzheimer's Disease: Role on Cholinergic Neurotransmission, Antioxidant Defence System, Neuroinflammation and Expression of BDNF. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:1040-1055. [PMID: 37779395 DOI: 10.2174/0118715273258337230925040049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 10/03/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is an overwhelming neurodegenerative disease with progressive loss of memory. AD is characterized by the deposition of the senile plaques mainly composed of β-amyloid (Aβ) fragment, BDNF decline, Cholinergic system overactivity and neuroinflammation. Montelukast (MTK), a leukotriene receptor antagonist, showed astounding neuroprotective effects in a variety of neurodegenerative disorders. OBJECTIVE This study aims to investigate the ameliorative effects of Montelukast in the scopolamineinduced Alzheimer's disease (AD) model in rats and evaluate its activity against neuroinflammation. METHODS Thirty rats were split into five groups: Control group (1 mL/kg normal saline, i.p.), Montelukast perse (10 mg/kg, i.p.), Disease group treated with Scopolamine (3 mg/kg, i.p.), Donepezil group (3 mg/kg, i.p.), Montelukast treatment group (10 mg/kg, i.p.) and behavioural and biochemical tests were carried out to assess the neuro protective effect. RESULTS Scopolamine treatment led to a significant reduction in learning and memory and an elevation in cholinesterase levels when compared with the control group (p < 0.01). Additionally, elevated oxidative stress and Amyloid-β levels were associated with enhanced neuroinflammation (p < 0.05, p < 0.01). Furthermore, the decline in neurotrophic factor BDNF is also observed when compared with the normal control group (p < 0.01). Montelukast pre-treatment significantly attenuated learning and memory impairment and cholinesterase levels. Besides, Montelukast and standard drug donepezil administration significantly suppressed the oxidative stress markers (p < 0.01), Amyloid-β levels, neuroinflammatory mediators (p < 0.05) and caused a significant increase in BDNF levels (p < 0.05). CONCLUSION Montelukast bestowed ameliorative effects in scopolamine-induced AD animal models as per the previous studies via attenuation of memory impairment, cholinesterase neurotransmission, oxidative stress, Amyloid-β levels, neuroinflammatory mediators and enhanced BDNF levels.
Collapse
Affiliation(s)
- Bhavana Yerraguravagari
- Department of Pharmacology, Shri Vishnu College of Pharmacy (SVCP) - Vishnupur, Bhimavaram - 534202, West Godavari, Andhra Pradesh, India
| | - Naga Pavani Penchikala
- Department of Pharmacology, Shri Vishnu College of Pharmacy (SVCP) - Vishnupur, Bhimavaram - 534202, West Godavari, Andhra Pradesh, India
| | - Aravinda Sai Kolusu
- Department of Pharmacology, Shri Vishnu College of Pharmacy (SVCP) - Vishnupur, Bhimavaram - 534202, West Godavari, Andhra Pradesh, India
| | - Grandhi Sandeep Ganesh
- Department of Pharmacology, Shri Vishnu College of Pharmacy (SVCP) - Vishnupur, Bhimavaram - 534202, West Godavari, Andhra Pradesh, India
| | - Prasad Konduri
- Department of Pharmacology, Shri Vishnu College of Pharmacy (SVCP) - Vishnupur, Bhimavaram - 534202, West Godavari, Andhra Pradesh, India
| | - Kumar V S Nemmani
- Department of Pharmacology, Shri Vishnu College of Pharmacy (SVCP) - Vishnupur, Bhimavaram - 534202, West Godavari, Andhra Pradesh, India
| | - Pavan Kumar Samudrala
- Department of Pharmacology, Shri Vishnu College of Pharmacy (SVCP) - Vishnupur, Bhimavaram - 534202, West Godavari, Andhra Pradesh, India
| |
Collapse
|
18
|
Umukoro S, Ajayi AM, Ben-Azu B, Ademola AP, Areelu J, Orji C, Okubena O. Jobelyn® improves motor dysfunctions induced by haloperidol in mice via neuroprotective mechanisms relating to modulation of cAMP response-element binding protein and mitogen-activated protein kinase. Metab Brain Dis 2023; 38:2269-2280. [PMID: 37347426 DOI: 10.1007/s11011-023-01253-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/08/2023] [Indexed: 06/23/2023]
Abstract
The clinical efficacy of haloperidol in the treatment of psychosis has been limited by its tendency to cause parkinsonian-like motor disturbances such as bradykinesia, muscle rigidity and postural instability. Oxidative stress-evoked neuroinflammation has been implicated as the key neuropathological mechanism by which haloperidol induces loss of dopaminergic neurons and motor dysfunctions. This study was therefore designed to evaluate the effect of Jobelyn® (JB), an antioxidant supplement, on haloperidol-induced motor dysfunctions and underlying molecular mechanisms in male Swiss mice. The animals were distributed into 5 groups (n = 8), and treated orally with distilled water (control), haloperidol (1 mg/kg) alone or in combination with each dose of JB (10, 20 and 40 mg/kg), daily for 14 days. Thereafter, changes in motor functions were evaluated on day 14. Brain biomarkers of oxidative stress, proinflammatory cytokines (tumor necrosis factor-alpha and interleukin-6), cAMP response-element binding protein (CREB), mitogen-activated protein kinase (MAPK) and histomorphological changes were also investigated. Haloperidol induces postural instability, catalepsy and impaired locomotor activity, which were ameliorated by JB. Jobelyn® attenuated haloperidol-induced elevated brain levels of MDA, nitrite, proinflammatory cytokines and also boosted neuronal antioxidant profiles (GSH and catalase) of mice. It also restored the deregulated brain activities of CREB and MAPK, and reduced the histomorphological distortions as well as loss of viable neuronal cells in the striatum and prefrontal cortex of haloperidol-treated mice. These findings suggest possible benefits of JB as adjunctive remedy in mitigating parkinsonian-like adverse effects of haloperidol through modulation of CREB/MAPK activities and oxidative/inflammatory pathways.
Collapse
Affiliation(s)
- Solomon Umukoro
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College of Medicine, University of Ibadan, Sango-Ojo Road, Ibadan, Oyo State, Nigeria.
| | - Abayomi Mayowa Ajayi
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College of Medicine, University of Ibadan, Sango-Ojo Road, Ibadan, Oyo State, Nigeria
| | - Benneth Ben-Azu
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College of Medicine, University of Ibadan, Sango-Ojo Road, Ibadan, Oyo State, Nigeria
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta University, Abraka, Nigeria
| | - Adeleke Pual Ademola
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College of Medicine, University of Ibadan, Sango-Ojo Road, Ibadan, Oyo State, Nigeria
| | - Jacob Areelu
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College of Medicine, University of Ibadan, Sango-Ojo Road, Ibadan, Oyo State, Nigeria
| | - Chika Orji
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College of Medicine, University of Ibadan, Sango-Ojo Road, Ibadan, Oyo State, Nigeria
| | | |
Collapse
|
19
|
Huang ZP, Liu SF, Zhuang JL, Li LY, Li MM, Huang YL, Chen YH, Chen XR, Lin S, Ye LC, Chen CN. Role of microglial metabolic reprogramming in Parkinson's disease. Biochem Pharmacol 2023; 213:115619. [PMID: 37211170 DOI: 10.1016/j.bcp.2023.115619] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/16/2023] [Accepted: 05/16/2023] [Indexed: 05/23/2023]
Abstract
Parkinson's disease (PD) is a common age-related neurodegenerative disorder characterized by damage to nigrostriatal dopaminergic neurons. Key pathogenic mechanisms underlying PD include alpha-synuclein misfolding and aggregation, impaired protein clearance, mitochondrial dysfunction, oxidative stress, and neuroinflammation. However, to date, no study has confirmed the specific pathogenesis of PD. Similarly, current PD treatment methods still have shortcomings. Although some emerging therapies have proved effective for PD, the specific mechanism still needs further clarification. Metabolic reprogramming, a term first proposed by Warburg, is applied to the metabolic energy characteristics of tumor cells. Microglia have similar metabolic characteristics. Pro-inflammatory M1 type and anti-inflammatory M2 type are the two types of activated microglia, which exhibit different metabolic patterns in glucose, lipid, amino acid, and iron metabolism. Additionally, mitochondrial dysfunction may be involved in microglial metabolic reprogramming by activating various signaling mechanisms. Functional changes in microglia resulting from metabolic reprogramming can cause changes in the brain microenvironment, thus playing an important role in neuroinflammation or tissue repair. The involvement of microglial metabolic reprogramming in PD pathogenesis has been confirmed. Neuroinflammation and dopaminergic neuronal death can effectively be reduced by inhibiting certain metabolic pathways in M1 microglia or reverting M1 cells to the M2 phenotype. This review summarizes the relationship between microglial metabolic reprogramming and PD and provides strategies for PD treatment.
Collapse
Affiliation(s)
- Zheng-Ping Huang
- Department of Neurology, Second Affiliated Hospital, Fujian Medical University, Quanzhou, Fujian Province 362000, China
| | - Shu-Fen Liu
- Department of Neurology, Second Affiliated Hospital, Fujian Medical University, Quanzhou, Fujian Province 362000, China
| | - Jian-Long Zhuang
- Prenatal Diagnosis Center, Quanzhou Women's and Children's Hospital, Quanzhou, China
| | - Lin-Yi Li
- Department of Neurology, Second Affiliated Hospital, Fujian Medical University, Quanzhou, Fujian Province 362000, China
| | - Mi-Mi Li
- Department of Neurology, Second Affiliated Hospital, Fujian Medical University, Quanzhou, Fujian Province 362000, China
| | - Ya-Li Huang
- Department of Neurology, Second Affiliated Hospital, Fujian Medical University, Quanzhou, Fujian Province 362000, China
| | - Yan-Hong Chen
- Department of Neurology, Shishi General Hospital, Quanzhou, Fujian Province 362000, China
| | - Xiang-Rong Chen
- Department of Neurosurgery, Second Affiliated Hospital, Second Clinical Medical College, Fujian Medical University, Quanzhou, China
| | - Shu Lin
- Center of Neurological and Metabolic Research, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province 362000, China; Group of Neuroendocrinology, Garvan Institute of Medical Research, 384 Victoria St, Sydney, Australia.
| | - Li-Chao Ye
- Department of Neurology, Second Affiliated Hospital, Fujian Medical University, Quanzhou, Fujian Province 362000, China.
| | - Chun-Nuan Chen
- Department of Neurology, Second Affiliated Hospital, Fujian Medical University, Quanzhou, Fujian Province 362000, China.
| |
Collapse
|
20
|
Wang H, Li J, Zhang H, Wang M, Xiao L, Wang Y, Cheng Q. Regulation of microglia polarization after cerebral ischemia. Front Cell Neurosci 2023; 17:1182621. [PMID: 37361996 PMCID: PMC10285223 DOI: 10.3389/fncel.2023.1182621] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
Stroke ranks second as a leading cause of death and permanent disability globally. Microglia, innate immune cells in the brain, respond rapidly to ischemic injury, triggering a robust and persistent neuroinflammatory reaction throughout the disease's progression. Neuroinflammation plays a critical role in the mechanism of secondary injury in ischemic stroke and is a significant controllable factor. Microglia activation takes on two general phenotypes: the pro-inflammatory M1 type and the anti-inflammatory M2 type, although the reality is more complex. The regulation of microglia phenotype is crucial to controlling the neuroinflammatory response. This review summarized the key molecules and mechanisms of microglia polarization, function, and phenotypic transformation following cerebral ischemia, with a focus on the influence of autophagy on microglia polarization. The goal is to provide a reference for the development of new targets for the treatment for ischemic stroke treatment based on the regulation of microglia polarization.
Collapse
Affiliation(s)
- Hao Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Jingjing Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Han Zhang
- School of Medicine, Nantong University, Nantong, China
| | - Mengyao Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Lifang Xiao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Yitong Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Qiong Cheng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| |
Collapse
|
21
|
Liu B, Svenningsson P, Ludvigsson JF, Lundholm C, Wallin J, Larsson H, Sjölander A, Williams DM, Pedersen NL, Wirdefeldt K. β2-Adrenoreceptor Agonists, Montelukast, and Parkinson Disease Risk. Ann Neurol 2023; 93:1023-1028. [PMID: 36897287 PMCID: PMC11497350 DOI: 10.1002/ana.26638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 02/11/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023]
Abstract
OBJECTIVE This study was undertaken to examine the association between montelukast use, β2-adrenoreceptor (β2AR) agonist use, and later Parkinson disease (PD). METHODS We ascertained use of β2AR agonists (430,885 individuals) and montelukast (23,315 individuals) from July 1, 2005 to June 30, 2007, and followed 5,186,886 PD-free individuals from July 1, 2007 to December 31, 2013 for incident PD diagnosis. We estimated hazard ratios and 95% confidence intervals using Cox regressions. RESULTS We observed 16,383 PD cases during on average 6.1 years of follow-up. Overall, use of β2AR agonists and montelukast were not related to PD incidence. A 38% lower PD incidence was noted among high-dose montelukast users when restricted to PD registered as the primary diagnosis. INTERPRETATION Overall, our data do not support inverse associations between β2AR agonists, montelukast, and PD. The prospect of lower PD incidence with high-dose montelukast exposure warrants further investigation, especially with adjustment for high-quality data on smoking. ANN NEUROL 2023;93:1023-1028.
Collapse
Affiliation(s)
- Bojing Liu
- Department of Medical Epidemiology and BiostatisticsKarolinska InstituteStockholmSweden
| | - Per Svenningsson
- Department of Clinical NeuroscienceKarolinska InstituteStockholmSweden
| | - Jonas F. Ludvigsson
- Department of Medical Epidemiology and BiostatisticsKarolinska InstituteStockholmSweden
- Department of PediatricsÖrebro University HospitalÖrebroSweden
| | - Cecilia Lundholm
- Department of Medical Epidemiology and BiostatisticsKarolinska InstituteStockholmSweden
| | - Johan Wallin
- Department of Clinical NeuroscienceKarolinska InstituteStockholmSweden
| | - Henrik Larsson
- Department of Medical Epidemiology and BiostatisticsKarolinska InstituteStockholmSweden
- School of Medical SciencesÖrebro UniversityÖrebroSweden
| | - Arvid Sjölander
- Department of Medical Epidemiology and BiostatisticsKarolinska InstituteStockholmSweden
| | - Dylan M. Williams
- Department of Medical Epidemiology and BiostatisticsKarolinska InstituteStockholmSweden
- MRC Unit for Lifelong Health and Ageing at UCLUniversity College LondonLondonUK
| | - Nancy L. Pedersen
- Department of Medical Epidemiology and BiostatisticsKarolinska InstituteStockholmSweden
- Department of PsychologyUniversity of Southern CaliforniaLos AngelesCAUSA
| | - Karin Wirdefeldt
- Department of Medical Epidemiology and BiostatisticsKarolinska InstituteStockholmSweden
- Department of Clinical NeuroscienceKarolinska InstituteStockholmSweden
| |
Collapse
|
22
|
Khalaf MM, El-Sayed MM, Kandeil MA, Ahmed S. A novel protective modality against rotenone-induced Parkinson's disease: A pre-clinical study with dulaglutide. Int Immunopharmacol 2023; 119:110170. [PMID: 37075673 DOI: 10.1016/j.intimp.2023.110170] [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: 11/25/2022] [Revised: 03/29/2023] [Accepted: 04/06/2023] [Indexed: 04/21/2023]
Abstract
Parkinson's disease (PD) drugs treat symptoms without inhibiting progression. In recent years, finding novel therapeutic medications that can halt disease progression has become crucial. Research on antidiabetic medicines is valuable in these investigations because of the parallels between the two disorders. Using Rotenone (ROT), a frequently used PD model, the possible neuroprotective benefits of Dulaglutide (DUL), an extended-acting glucagon-like peptide-1 agonist, were considered. Twenty-four rats were randomly assigned to 4 groups to complete this experiment (n = 6). 0.2 ml of the vehicle (1 ml of dimethyl sulfoxide (DMSO) diluted in sunflower oil) was administered to the standard control group subcutaneously with a 48-hour pause. The second group was administered ROT 2.5 mg/kg SC every 48 h for 20 days as a positive control group. The third and fourth groups were administered one dose of DUL each week (0.05 and 0.1 mg/kg SC, respectively) to their regimens. The mice received ROT (2.5 mg/kg SC) every 48 h for 20 days after receiving DUL for the initial dose (96 h later). The current study focused on the DUL's ability to preserve usual behavioral function, enhance antioxidant and anti-inflammatory pathways, inhibit alpha-synuclein (α-syn), and increase parkin levels. It is concluded that DUL acts as an antioxidant and an anti-inflammatory to protect against ROT-induced PD. However, more studies are required to support this finding.
Collapse
Affiliation(s)
- Marwa M Khalaf
- Pharmacology & Toxicology Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt.
| | - Mahmoud M El-Sayed
- Pharmacology & Toxicology Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt.
| | - Mohamed A Kandeil
- Biochemistry Department, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef 62514, Egypt.
| | - Sanaa Ahmed
- Pharmacology Department, Faculty of Medicine, Sohag University, Sohag 82524, Egypt.
| |
Collapse
|
23
|
Luo BL, Zhang ZZ, Chen J, Liu X, Zhang YM, Yang QG, Chen GH. Effects of gestational inflammation on age-related cognitive decline and hippocampal Gdnf-GFRα1 levels in F1 and F2 generations of CD-1 Mice. BMC Neurosci 2023; 24:26. [PMID: 37055728 PMCID: PMC10103445 DOI: 10.1186/s12868-023-00793-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/23/2023] [Indexed: 04/15/2023] Open
Abstract
BACKGROUND It has been reported that age-associated cognitive decline (AACD) accelerated by maternal lipopolysaccharide (LPS) insult during late pregnancy can be transmitted to the second generation in a sex-specificity manner. In turn, recent studies indicated that glial cell line-derived neurotrophic factor (GDNF) and its cognate receptor (GFRα1) are critical for normal cognitive function. Based on this evidence, we aimed to explore whether Gdnf-GFRα1 expression contributes to cognitive decline in the F1 and F2 generations of mouse dams exposed to lipopolysaccharide (LPS) during late gestation, and to evaluate also the potential interference effect of pro-inflammatory cytokines. METHODS During gestational days 15-17, pregnant CD-1 mice (8-10 weeks old) received a daily intraperitoneal injection of LPS (50 μg/kg) or saline (control). In utero LPS-exposed F1 generation mice were selectively mated to produce F2 generation mice. In F1 and F2 mice aged 3 and 15 months, the Morris water maze (MWM) was used to evaluated the spatial learning and memory ability, the western blotting and RT-PCR were used for analyses of hippocampal Gdnf and GFRα1 expression, and ELISA was used to analyse IL-1β, IL-6 and TNF-α levels in serum. RESULTS Middle-aged F1 offspring from LPS-treated mothers exhibited longer swimming latency and distance during the learning phase, lower percentage swimming time and distance in targe quadrant during memory phase, and lower hippocampal levels of Gdnf and GFRα1 gene products compared to age-matched controls. Similarly, the middle-aged F2 offspring from the Parents-LPS group had longer swimming latency and distance in the learning phase, and lower percentage swimming time and distance in memory phase than the F2-CON group. Moreover, the 3-month-old Parents-LPS and 15-month-old Parents- and Father-LPS groups had lower GDNF and GFRα1 protein and mRNAs levels compared to the age-matched F2-CON group. Furthermore, hippocampal levels of Gdnf and GFRα1 were correlated with impaired cognitive performance in the Morris water maze after controlling for circulating pro-inflammatory cytokine levels. CONCLUSIONS Our findings indicate that accelerated AACD by maternal LPS exposure can be transmitted across at least two generations through declined Gdnf and GFRα1 expression, mainly via paternal linage.
Collapse
Affiliation(s)
- Bao-Ling Luo
- Department of Neurology (Sleep Disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, 238000, Anhui, People's Republic of China
| | - Zhe-Zhe Zhang
- Department of Neurology (Sleep Disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, 238000, Anhui, People's Republic of China
| | - Jing Chen
- Department of Neurology (Sleep Disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, 238000, Anhui, People's Republic of China
| | - Xue Liu
- Department of Geriatrics, the First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, Anhui, People's Republic of China
| | - Yue-Ming Zhang
- Department of Neurology (Sleep Disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, 238000, Anhui, People's Republic of China
| | - Qi-Gang Yang
- Department of Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, People's Republic of China.
| | - Gui-Hai Chen
- Department of Neurology (Sleep Disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, 238000, Anhui, People's Republic of China.
| |
Collapse
|
24
|
Ling Y, Upadhyaya P, Chen L, Jiang X, Kim Y. Emulate randomized clinical trials using heterogeneous treatment effect estimation for personalized treatments: Methodology review and benchmark. J Biomed Inform 2023; 137:104256. [PMID: 36455806 PMCID: PMC9845190 DOI: 10.1016/j.jbi.2022.104256] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/28/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022]
Abstract
Big data and (deep) machine learning have been ambitious tools in digital medicine, but these tools focus mainly on association. Intervention in medicine is about the causal effects. The average treatment effect has long been studied as a measure of causal effect, assuming that all populations have the same effect size. However, no "one-size-fits-all" treatment seems to work in some complex diseases. Treatment effects may vary by patient. Estimating heterogeneous treatment effects (HTE) may have a high impact on developing personalized treatment. Lots of advanced machine learning models for estimating HTE have emerged in recent years, but there has been limited translational research into the real-world healthcare domain. To fill the gap, we reviewed and compared eleven recent HTE estimation methodologies, including meta-learner, representation learning models, and tree-based models. We performed a comprehensive benchmark experiment based on nationwide healthcare claim data with application to Alzheimer's disease drug repurposing. We provided some challenges and opportunities in HTE estimation analysis in the healthcare domain to close the gap between innovative HTE models and deployment to real-world healthcare problems.
Collapse
Affiliation(s)
- Yaobin Ling
- Center for Secure Artificial Intelligence for Healthcare, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Fannin 7000, Houston, TX, United States.
| | - Pulakesh Upadhyaya
- Center for Secure Artificial Intelligence for Healthcare, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Fannin 7000, Houston, TX, United States.
| | - Luyao Chen
- Center for Secure Artificial Intelligence for Healthcare, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Fannin 7000, Houston, TX, United States.
| | - Xiaoqian Jiang
- Center for Secure Artificial Intelligence for Healthcare, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Fannin 7000, Houston, TX, United States.
| | - Yejin Kim
- Center for Secure Artificial Intelligence for Healthcare, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Fannin 7000, Houston, TX, United States.
| |
Collapse
|
25
|
The mechanisms underlying montelukast's neuropsychiatric effects - new insights from a combined metabolic and multiomics approach. Life Sci 2022; 310:121056. [DOI: 10.1016/j.lfs.2022.121056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 09/28/2022] [Accepted: 10/06/2022] [Indexed: 11/05/2022]
|
26
|
Montelukast and Acute Coronary Syndrome: The Endowed Drug. Pharmaceuticals (Basel) 2022; 15:ph15091147. [PMID: 36145367 PMCID: PMC9500901 DOI: 10.3390/ph15091147] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/06/2022] [Accepted: 09/09/2022] [Indexed: 12/16/2022] Open
Abstract
Acute coronary syndrome (ACS) is a set of signs and symptoms caused by a reduction of coronary blood flow with subsequent myocardial ischemia. ACS is associated with activation of the leukotriene (LT) pathway with subsequent releases of various LTs, including LTB4, LTC4, and LTD4, which cause inflammatory changes and induction of immunothrombosis. LTs through cysteine leukotriene (CysLT) induce activation of platelets and clotting factors with succeeding coronary thrombosis. CysLT receptor (CysLTR) antagonists such as montelukast (MK) may reduce the risk of the development of ACS and associated complications through suppression of the activation of platelet and clotting factors. Thus, this critical review aimed to elucidate the possible protective role of MK in the management of ACS. The LT pathway is implicated in the pathogenesis of atherosclerosis, cardiac hypertrophy, and heart failure. Inhibition of the LT pathway and CysL1TR by MK might be effective in preventing cardiovascular complications. MK could be an effective novel therapy in the management of ACS through inhibition of pro-inflammatory CysLT1R and modulation of inflammatory signaling pathways. MK can attenuate thrombotic events by inhibiting platelet activation and clotting factors that are activated during the development of ACS. In conclusion, MK could be an effective agent in reducing the severity of ACS and associated complications. Experimental, preclinical, and clinical studies are recommended to confirm the potential therapeutic of MK in the management of ACS.
Collapse
|
27
|
Marques CF, Marques MM, Justino GC. Leukotrienes vs. Montelukast—Activity, Metabolism, and Toxicity Hints for Repurposing. Pharmaceuticals (Basel) 2022; 15:ph15091039. [PMID: 36145259 PMCID: PMC9505853 DOI: 10.3390/ph15091039] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
Increasing environmental distress is associated with a growing asthma incidence; no treatments are available but montelukast (MTK)—an antagonist of the cysteinyl leukotrienes receptor 1—is widely used in the management of symptoms among adults and children. Recently, new molecular targets have been identified and MTK has been proposed for repurposing in other therapeutic applications, with several ongoing clinical trials. The proposed applications include neuroinflammation control, which could be explored in some neurodegenerative disorders, such as Alzheimer’s and Parkinson’s diseases (AD and PD). However, this drug has been associated with an increasing number of reported neuropsychiatric adverse drug reactions (ADRs). Besides, and despite being on the market since 1998, MTK metabolism is still poorly understood and the mechanisms underlying neuropsychiatric ADRs remain unknown. We review the role of MTK as a modulator of leukotriene pathways and systematize the current knowledge about MTK metabolism. Known toxic effects of MTK are discussed, and repurposing applications are presented comprehensively, with a focus on AD and PD.
Collapse
Affiliation(s)
- Cátia F. Marques
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
| | - Maria Matilde Marques
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
- Departamento de Engenharia Química, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
| | - Gonçalo C. Justino
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
- Correspondence:
| |
Collapse
|
28
|
Modulation of CREB and its associated upstream signaling pathways in pesticide-induced neurotoxicity. Mol Cell Biochem 2022; 477:2581-2593. [PMID: 35596844 PMCID: PMC9618525 DOI: 10.1007/s11010-022-04472-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 05/04/2022] [Indexed: 11/13/2022]
Abstract
Human beings are exposed to various environmental xenobiotics throughout their life consisting of a broad range of physical and chemical agents that impart bodily harm. Among these, pesticide exposure that destroys insects mainly by damaging their central nervous system also exerts neurotoxic effects on humans and is implicated in the etiology of several degenerative disorders. The connectivity between CREB (cAMP Response Element Binding Protein) signaling activation and neuronal activity is of broad interest and has been thoroughly studied in various diseased states. Several genes, as well as protein kinases, are involved in the phosphorylation of CREB, including BDNF (Brain-derived neurotrophic factor), Pi3K (phosphoinositide 3-kinase), AKT (Protein kinase B), RAS (Rat Sarcoma), MEK (Mitogen-activated protein kinase), PLC (Phospholipase C), and PKC (Protein kinase C) that play an essential role in neuronal plasticity, long-term potentiation, neuronal survival, learning, and memory formation, cognitive function, synaptic transmission, and suppressing apoptosis. These elements, either singularly or in a cascade, can result in the modulation of CREB, making it a vulnerable target for various neurotoxic agents, including pesticides. This review provides insight into how these various intracellular signaling pathways converge to bring about CREB activation and how the activated or deactivated CREB levels can affect the gene expression of the upstream molecules. We also discuss the various target genes within the cascade vulnerable to different types of pesticides. Thus, this review will facilitate future investigations associated with pesticide neurotoxicity and identify valuable therapeutic targets.
Collapse
|
29
|
Josiah SS, Famusiwa CD, Crown OO, Lawal AO, Olaleye MT, Akindahunsi AA, Akinmoladun AC. Neuroprotective effects of catechin and quercetin in experimental Parkinsonism through modulation of dopamine metabolism and expression of IL-1β, TNF-α, NF-κB, IκKB, and p53 genes in male Wistar rats. Neurotoxicology 2022; 90:158-171. [PMID: 35337893 DOI: 10.1016/j.neuro.2022.03.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 10/18/2022]
Abstract
The neurobehavioral, brain redox-stabilizing and neurochemical modulatory properties of catechin and quercetin in rotenone-induced Parkinsonism, and the involvement of NF-κB-mediated inflammation, were investigated. Male Wistar rats subcutaneously administered with multiple doses of 1.5mg/kg rotenone were post-treated with 5-20mg/kg catechin or quercetin. This was followed by neurobehavioral evaluation, biochemical estimations, and assessment of neurotransmitter metabolism in the striatum. Expression of genes involved in the canonical pathway for the activation of NF-κB mediated inflammation (IL-1β, TNF-α, NF-κB, and IκKB) and the pro-apoptotic gene, p53, in the striatum was determined by RT-qPCR. Catechin and quercetin mitigated neurobehavioral deficits caused by rotenone. Both flavonoids attenuated striatal redox stress and neurochemical dysfunction, optimized disturbed dopamine metabolism, and improved depletion of neuron density caused by rotenone toxicity. While administration of catechin produced a more pronounced attenuating effect on IL-1β, TNF-α, and p53 genes, the attenuating effect of quercetin (20mg/kg) was more pronounced on NF-κB and IκKB gene expressions when compared to the group administered with rotenone only. Comparatively, quercetin demonstrated superior protection against rotenone neurotoxicity. It is concluded that catechin and quercetin have potential relevance in Parkinson's disease therapy through amelioration of redox stress, optimization of dopamine metabolism, and modulation of anti-inflammatory and anti-apoptotic pathways.
Collapse
Affiliation(s)
- Sunday Solomon Josiah
- Department of Biochemistry, School of Life Sciences, The Federal University of Technology, P.M.B. 704, Akure 340001, Nigeria; Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Hatherly Laboratories, Exeter EX4 4PS, UK
| | - Courage Dele Famusiwa
- Department of Biochemistry, School of Life Sciences, The Federal University of Technology, P.M.B. 704, Akure 340001, Nigeria; Department of Chemical sciences, Skyline University Nigeria, Kano, Nigeria
| | - Olamide Olajusi Crown
- Department of Biochemistry, School of Life Sciences, The Federal University of Technology, P.M.B. 704, Akure 340001, Nigeria; Department of Chemistry, Physics and Atmospheric Science, Jackson State University, Jackson, MS 39204, USA
| | - Akeem O Lawal
- Department of Biochemistry, School of Life Sciences, The Federal University of Technology, P.M.B. 704, Akure 340001, Nigeria
| | - Mary Tolulope Olaleye
- Department of Biochemistry, School of Life Sciences, The Federal University of Technology, P.M.B. 704, Akure 340001, Nigeria
| | - Afolabi Akintunde Akindahunsi
- Department of Biochemistry, School of Life Sciences, The Federal University of Technology, P.M.B. 704, Akure 340001, Nigeria
| | - Afolabi Clement Akinmoladun
- Department of Biochemistry, School of Life Sciences, The Federal University of Technology, P.M.B. 704, Akure 340001, Nigeria.
| |
Collapse
|
30
|
Ahmed S, El-Sayed MM, Kandeil MA, Khalaf MM. Empagliflozin attenuates Neurodegeneration through Antioxidant, Anti-inflammatory, and Modulation of α-synuclein and Parkin Levels in Rotenone-Induced Parkinson’s Disease in Rats. Saudi Pharm J 2022; 30:863-873. [PMID: 35812142 PMCID: PMC9257853 DOI: 10.1016/j.jsps.2022.03.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 03/10/2022] [Indexed: 10/26/2022] Open
|
31
|
Ma J, Shi X, Li M, Chen S, Gu Q, Zheng J, Li D, Wu S, Yang H, Li X. MicroRNA-181a-2-3p shuttled by mesenchymal stem cell-secreted extracellular vesicles inhibits oxidative stress in Parkinson's disease by inhibiting EGR1 and NOX4. Cell Death Discov 2022; 8:33. [PMID: 35075150 PMCID: PMC8786891 DOI: 10.1038/s41420-022-00823-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 12/10/2021] [Accepted: 01/06/2022] [Indexed: 12/18/2022] Open
Abstract
The current study investigated the physiological mechanisms by which extracellular vesicle (EV)-encapsulated miR-181a-2-3p derived from mesenchymal stem cells (MSCs) might mediate oxidative stress (OS) in Parkinson's disease (PD). First, 6-hydroxydopamine (6-OHDA)-induced PD cell and mouse models were established, after which miR-181a-2-3p, EGR1, and NOX4 expression patterns were determined in SH-SY5Y cells and substantia nigra (SN) of PD mice. Next, the binding affinity among miR-181a-2-3p, EGR1, and NOX4 was identified using multiple assays. Gain- or loss-of-function experiments were further adopted to detect SH-SY5Y cell proliferation and apoptosis and to measure the levels of SOD, MDA, and ROS. Finally, the effects of miR-181a-2-3p from MSC-derived EVs in PD mouse models were also explored. It was found that miR-181a-2-3p was poorly expressed in 6-OHDA-induced SH-SY5Y cells, whereas miR-181a-2-3p from MSCs could be transferred into SH-SY5Y cells via EVs. In addition, miR-181a-2-3p could target and inhibit EGR1, which promoted the expression of NOX4. The aforementioned miR-181a-2-3p shuttled by MSC-derived EVs facilitated SH-SY5Y proliferation and SOD levels, but suppressed apoptosis and MDA and ROS levels by regulating EGR1 via inhibition of NOX4/p38 MAPK, so as to repress OS of PD. Furthermore, in PD mice, miR-181a-2-3p was carried by EVs from MSCs to alleviate apoptosis of dopamine neurons and OS, accompanied by increased expressions of α-syn and decreased 4-HNE in SN tissues. Collectively, our findings revealed that MSC-derived EV-loaded miR-181a-2-3p downregulated EGR1 to inhibit OS via the NOX4/p38 MAPK axis in PD.
Collapse
Affiliation(s)
- Jianjun Ma
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, 450003, P. R. China.
- Department of Neurology, Zhengzhou University People's Hospital, Zhengzhou, 450003, P. R. China.
- Department of Neurology, Henan University People's Hospital, Zhengzhou, 450003, P. R. China.
| | - Xiaoxue Shi
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, 450003, P. R. China
- Department of Neurology, Zhengzhou University People's Hospital, Zhengzhou, 450003, P. R. China
| | - Mingjian Li
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, 450003, P. R. China
- Department of Neurology, Henan University People's Hospital, Zhengzhou, 450003, P. R. China
| | - Siyuan Chen
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, 450003, P. R. China
- Department of Neurology, Zhengzhou University People's Hospital, Zhengzhou, 450003, P. R. China
- Department of Neurology, Henan University People's Hospital, Zhengzhou, 450003, P. R. China
| | - Qi Gu
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, 450003, P. R. China
- Department of Neurology, Zhengzhou University People's Hospital, Zhengzhou, 450003, P. R. China
- Department of Neurology, Henan University People's Hospital, Zhengzhou, 450003, P. R. China
| | - Jinhua Zheng
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, 450003, P. R. China
- Department of Neurology, Zhengzhou University People's Hospital, Zhengzhou, 450003, P. R. China
- Department of Neurology, Henan University People's Hospital, Zhengzhou, 450003, P. R. China
| | - Dongsheng Li
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, 450003, P. R. China
- Department of Neurology, Zhengzhou University People's Hospital, Zhengzhou, 450003, P. R. China
- Department of Neurology, Henan University People's Hospital, Zhengzhou, 450003, P. R. China
| | - Shaopu Wu
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, 450003, P. R. China
- Department of Neurology, Zhengzhou University People's Hospital, Zhengzhou, 450003, P. R. China
- Department of Neurology, Henan University People's Hospital, Zhengzhou, 450003, P. R. China
| | - Hongqi Yang
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, 450003, P. R. China
- Department of Neurology, Zhengzhou University People's Hospital, Zhengzhou, 450003, P. R. China
- Department of Neurology, Henan University People's Hospital, Zhengzhou, 450003, P. R. China
| | - Xue Li
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, 450003, P. R. China
- Department of Neurology, Zhengzhou University People's Hospital, Zhengzhou, 450003, P. R. China
- Department of Neurology, Henan University People's Hospital, Zhengzhou, 450003, P. R. China
| |
Collapse
|
32
|
L-Theanine Ameliorated Rotenone-Induced Parkinsonism-like Symptoms in Rats. Neurotox Res 2022; 40:241-258. [PMID: 34988886 DOI: 10.1007/s12640-021-00451-w] [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: 09/09/2021] [Revised: 11/17/2021] [Accepted: 11/23/2021] [Indexed: 10/19/2022]
Abstract
Rotenone (RO)-induced neurotoxicity exhibits pathophysiological features similar to those reported in patients with Parkinson's disease (PD), such as nitrosative and oxidative stress, mitochondrial dysfunction, and neural cytoarchitecture alterations in the substantia nigra pars compacta (SNpc)/striatum (ST), which has been used for decades as an animal model of PD in humans. L-Theanine (LT), a major amino acid component of green tea, exhibits potent antioxidant and anti-inflammatory activities and protects against various neural injuries. We investigated the potential therapeutic effects of LT on RO-induced behavioral and neurochemical dysfunction in rats and the neuroprotective mechanisms underlying these effects. Unilateral stereotaxic intranigral infusion of RO into the SNpc to induce PD-like manifestations induced significant behavioral impairment as evaluated using an open field test, rotarod test, grip strength measurement, and beam-crossing task in rats. LT treatment (300 mg/kg i.p., 21 days) ameliorated most RO-induced behavioral impairments. In addition, LT treatment reduced nitric oxide level and lipid peroxidation production, increased mitochondrial function and integrity, as well as the activities of mitochondrial complexes I, II, IV, and V, and reduced the levels of neuroinflammatory and apoptotic markers in the SNpc and ameliorated the levels of catecholamines, GABA and glutamate in the ST induced by RO. These results demonstrate the possible therapeutic effects of LT against RO-induced behavioral impairments, including antioxidative effects, prevention of mitochondrial dysfunction, prevention of neurochemical deficiency, anti-neuroinflammatory effects, and anti-apoptotic effects. This is the first report on the neuroprotective effect of LT against RO-induced behavioral impairments, and the above evidence provides a potential clinically relevant role for LT in the management of human PD.
Collapse
|
33
|
El-Shamarka ME, Eliwa HA, Ahmed MAE. Inhibition of boldenone-induced aggression in rats by curcumin: Targeting TLR4/MyD88/TRAF-6/NF-κB pathway. J Biochem Mol Toxicol 2021; 36:e22936. [PMID: 34719837 DOI: 10.1002/jbt.22936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 09/22/2021] [Accepted: 10/18/2021] [Indexed: 11/08/2022]
Abstract
The illicit abuse of anabolic steroids is associated with brutal aggression, which represents a serious health hazard and social threat. Boldenone is commonly used for doping by athletes and adolescents for esthetic purposes and to enhance performance and endurance during competitions. However, the mechanistic pathways underlying boldenone-induced behavioral deviations and neuronal toxicity have not yet been elucidated. On the other hand, the natural polyphenol curcumin is appreciated for its relative safety, potent antioxidant activity, and anti-inflammatory properties. Therefore, the present study was initiated to explore the signaling pathways underlying boldenone-induced anxiety and aggression in rats, and the protective effects of curcumin. To achieve this aim, male Wistar albino rats were randomly distributed into control, curcumin (100 mg/kg in sesame oil, p.o., once daily), boldenone (5 mg/kg, intramuscular, once weekly), and combination groups. Rats were challenged across the open field, irritability, defensive aggression, and resident-intruder tests. The prefrontal cortex was used to assess serotonin level, oxidative stress markers, and mRNA expression of myeloid differentiation primary response gene (MyD88), TNFR-associated factor 6 (TRAF-6), tumor necrosis factor α (TNF-α), interleukin-1β (IL-1β), protein expression of toll-like receptor 4 (TLR4), and phosphorylated nuclear factor-κB transcription factor (NF-κB p65). Unprecedented, the current results showed that boldenone elicited aggression in rats accompanied by depleted serotonin, enhanced oxidative stress, and exaggerated inflammatory response via upregulation of TLR4/MyD88/TRAF-6/NF-κB pathway. Interestingly, curcumin mitigated boldenone-induced neurobehavioral disturbances in rats, normalized the oxidant/antioxidant balance, and suppressed TLR4/MyD88/TRAF-6/NF-κB pathway and its downstream proinflammatory signaling molecules TNF-α and IL-1β.
Collapse
Affiliation(s)
- Marwa E El-Shamarka
- Department of Narcotics, Ergogenic Aids and Poisons, National Research Center, Dokki, Egypt
| | - Hesham A Eliwa
- Department of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST), Giza, Egypt
| | - Maha A E Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST), Giza, Egypt
| |
Collapse
|
34
|
Behl T, Madaan P, Sehgal A, Singh S, Sharma N, Bhatia S, Al-Harrasi A, Chigurupati S, Alrashdi I, Bungau SG. Elucidating the Neuroprotective Role of PPARs in Parkinson's Disease: A Neoteric and Prospective Target. Int J Mol Sci 2021; 22:10161. [PMID: 34576325 PMCID: PMC8467926 DOI: 10.3390/ijms221810161] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 09/17/2021] [Accepted: 09/19/2021] [Indexed: 12/13/2022] Open
Abstract
One of the utmost frequently emerging neurodegenerative diseases, Parkinson's disease (PD) must be comprehended through the forfeit of dopamine (DA)-generating nerve cells in the substantia nigra pars compacta (SN-PC). The etiology and pathogenesis underlying the emergence of PD is still obscure. However, expanding corroboration encourages the involvement of genetic and environmental factors in the etiology of PD. The destruction of numerous cellular components, namely oxidative stress, ubiquitin-proteasome system (UPS) dysfunction, autophagy-lysosome system dysfunction, neuroinflammation and programmed cell death, and mitochondrial dysfunction partake in the pathogenesis of PD. Present-day pharmacotherapy can alleviate the manifestations, but no therapy has been demonstrated to cease disease progression. Peroxisome proliferator-activated receptors (PPARs) are ligand-directed transcription factors pertaining to the class of nuclear hormone receptors (NHR), and are implicated in the modulation of mitochondrial operation, inflammation, wound healing, redox equilibrium, and metabolism of blood sugar and lipids. Numerous PPAR agonists have been recognized to safeguard nerve cells from oxidative destruction, inflammation, and programmed cell death in PD and other neurodegenerative diseases. Additionally, various investigations suggest that regular administration of PPAR-activating non-steroidal anti-inflammatory drugs (NSAIDs) (ibuprofen, indomethacin), and leukotriene receptor antagonists (montelukast) were related to the de-escalated evolution of neurodegenerative diseases. The present review elucidates the emerging evidence enlightening the neuroprotective outcomes of PPAR agonists in in vivo and in vitro models experiencing PD. Existing articles up to the present were procured through PubMed, MEDLINE, etc., utilizing specific keywords spotlighted in this review. Furthermore, the authors aim to provide insight into the neuroprotective actions of PPAR agonists by outlining the pharmacological mechanism. As a conclusion, PPAR agonists exhibit neuroprotection through modulating the expression of a group of genes implicated in cellular survival pathways, and may be a propitious target in the therapy of incapacitating neurodegenerative diseases like PD.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (P.M.); (A.S.); (S.S.); (N.S.)
| | - Piyush Madaan
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (P.M.); (A.S.); (S.S.); (N.S.)
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (P.M.); (A.S.); (S.S.); (N.S.)
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (P.M.); (A.S.); (S.S.); (N.S.)
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (P.M.); (A.S.); (S.S.); (N.S.)
| | - Saurabh Bhatia
- Natural & Medical Sciences Research Centre, University of Nizwa, Birkat Al Mauz 616, Nizwa P.O. Box 33, Oman; (S.B.); (A.A.-H.)
- School of Health Science, University of Petroleum and Energy Studies, Dehradun 248007, India
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Centre, University of Nizwa, Birkat Al Mauz 616, Nizwa P.O. Box 33, Oman; (S.B.); (A.A.-H.)
| | - Sridevi Chigurupati
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraydah 52571, Saudi Arabia;
| | - Ibrahim Alrashdi
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne NE1 7RU, UK;
| | - Simona Gabriela Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410073 Oradea, Romania
| |
Collapse
|
35
|
Potential Effects of Leukotriene Receptor Antagonist Montelukast in Treatment of Neuroinflammation in Parkinson's Disease. Int J Mol Sci 2021; 22:ijms22115606. [PMID: 34070609 PMCID: PMC8198163 DOI: 10.3390/ijms22115606] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/16/2021] [Accepted: 05/21/2021] [Indexed: 12/11/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder where misfolded alpha-synuclein-enriched aggregates called Lewy bodies are central in pathogenesis. No neuroprotective or disease-modifying treatments are currently available. Parkinson’s disease is considered a multifactorial disease and evidence from multiple patient studies and animal models has shown a significant immune component during the course of the disease, highlighting immunomodulation as a potential treatment strategy. The immune changes occur centrally, involving microglia and astrocytes but also peripherally with changes to the innate and adaptive immune system. Here, we review current understanding of different components of the PD immune response with a particular emphasis on the leukotriene pathway. We will also describe evidence of montelukast, a leukotriene receptor antagonist, as a possible anti-inflammatory treatment for PD.
Collapse
|
36
|
Al-Kuraishy HM, Al-Gareeb AI, Almulaiky YQ, Cruz-Martins N, El-Saber Batiha G. Role of leukotriene pathway and montelukast in pulmonary and extrapulmonary manifestations of Covid-19: The enigmatic entity. Eur J Pharmacol 2021; 904:174196. [PMID: 34004207 PMCID: PMC8123523 DOI: 10.1016/j.ejphar.2021.174196] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/28/2021] [Accepted: 05/12/2021] [Indexed: 02/06/2023]
Abstract
Severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2), the responsible agent for the coronavirus disease 2019 (Covid-19), has its entry point through interaction with angiotensin converting enzyme 2 (ACE2) receptors, highly expressed in lung type II alveolar cells and other tissues, like heart, pancreas, brain, and vascular endothelium. This review aimed to elucidate the potential role of leukotrienes (LTs) in the pathogenesis and clinical presentation of SARS-CoV-2 infection, and to reveal the critical role of LT pathway receptor antagonists and inhibitors in Covid-19 management. A literature search was done in PubMed, Scopus, Web of Science and Google Scholar databases to find the potential role of montelukast and other LT inhibitors in the management of pulmonary and extra-pulmonary manifestations triggered by SARS-CoV-2. Data obtained so far underline that pulmonary and extra-pulmonary manifestations in Covid-19 are attributed to a direct effect of SARS-CoV-2 in expressed ACE2 receptors or indirectly through NF-κB dependent induction of a cytokine storm. Montelukast can ameliorate extra-pulmonary manifestations in Covid-19 either directly through blocking of Cys-LTRs in different organs or indirectly through inhibition of the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, Baghdad, Iraq.
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, Baghdad, Iraq.
| | - Yaaser Q Almulaiky
- University of Jeddah, College of Sciences and Arts at Khulis, Department of Chemistry, Jeddah, Saudi Arabia.
| | - Natália Cruz-Martins
- Faculty of Medicine, University of Porto, Alameda Prof. Hernani Monteiro, 4200-319, Porto, Portugal; Institute for Research and Innovation in Health (i3S), University of Porto, Rua Alfredo Allen, 4200-135, Porto, Portugal; Laboratory of Neuropsychophysiology, Faculty of Psychology and Education Sciences, University of Porto, Portugal.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt.
| |
Collapse
|
37
|
Han B, Zhang YY, Ye ZQ, Xiao Y, Rasouli J, Wu WC, Ye SM, Guo XY, Zhu L, Rostami A, Wang LB, Zhang Y, Li X. Montelukast alleviates inflammation in experimental autoimmune encephalomyelitis by altering Th17 differentiation in a mouse model. Immunology 2021; 163:185-200. [PMID: 33480040 DOI: 10.1111/imm.13308] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 12/23/2020] [Accepted: 01/04/2021] [Indexed: 12/25/2022] Open
Abstract
Montelukast is a leukotriene receptor antagonist that is known to prevent allergic rhinitis and asthma. Blocking the Cysteinyl leukotriene receptor (CysLTR1), one of the primary receptors of leukotrienes, has been demonstrated to be efficacious in ameliorating experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS), through disrupting chemotaxis of infiltrating T cells. However, the role of CysLTR1 in the pathogenesis of MS is not well understood. Here, we show that MS patients had higher expression of CysLTR1 in the circulation and central nervous system (CNS). The majority of CD4+ T cells expressed CysLTR1 in MS lesions. Among T-cell subsets, Th17 cells had the highest expression of CysLTR1, and blocking CysLTR1 signalling abrogated their development in vitro. Inhibition of CysLTR1 by montelukast suppressed EAE development in both a prophylactic and therapeutic manner and inhibited myelin loss in EAE mice. Similarly, the in vivo results showed that montelukast inhibited Th17 response in EAE mice and that Th17 cells treated with montelukast had reduced encephalitogenic in adoptive EAE. Our findings strongly suggest that targeting Th17 response by inhibiting CysLTR1 signalling could be a promising therapeutic strategy for the treatment of MS and CNS inflammatory diseases.
Collapse
Affiliation(s)
- Bing Han
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yan-Yan Zhang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Ze-Qing Ye
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yun Xiao
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Javad Rasouli
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Wen-Cheng Wu
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Su-Min Ye
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Xin-Yue Guo
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Lin Zhu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | | | - Li-Bin Wang
- The General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yuan Zhang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Xing Li
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
38
|
Tesfaye BA, Hailu HG, Zewdie KA, Ayza MA, Berhe DF. Montelukast: The New Therapeutic Option for the Treatment of Epilepsy. J Exp Pharmacol 2021; 13:23-31. [PMID: 33505173 PMCID: PMC7829127 DOI: 10.2147/jep.s277720] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 12/29/2020] [Indexed: 12/16/2022] Open
Abstract
Currently, there is no definitive cure for epilepsy. The available medications relieve symptoms and reduce seizure attacks. The major challenge with the available antiepileptic medication is safety and affordability. The repurposing of montelukast for epilepsy can be an alternative medication with a better safety profile. Montelukast is a leukotriene receptor antagonist that binds to the cysteinyl leukotrienes (CysLT) receptors used in the treatment of bronchial asthma and seasonal allergies. Emerging evidence suggests that montelukast's anti-inflammatory effect can help to maintain BBB integrity. The drug has also neuroprotective and anti-oxidative activities to reduce seizure incidence and epilepsy. The present review summarizes the neuropharmacological actions of montelukast in epilepsy with an emphasis on the recent findings associated with CysLT and cell-specific effects.
Collapse
Affiliation(s)
- Bekalu Amare Tesfaye
- Department of Pharmacology and Toxicology, School of Pharmacy, Mekelle University, Mekelle, Ethiopia
| | - Haftom Gebregergs Hailu
- Department of Pharmacology and Toxicology, School of Pharmacy, Mekelle University, Mekelle, Ethiopia
| | - Kaleab Alemayehu Zewdie
- Department of Pharmacology and Toxicology, School of Pharmacy, Mekelle University, Mekelle, Ethiopia
| | - Muluken Altaye Ayza
- Department of Pharmacology and Toxicology, School of Pharmacy, Mekelle University, Mekelle, Ethiopia
| | - Derbew Fikadu Berhe
- Department of Pharmacology and Toxicology, School of Pharmacy, Mekelle University, Mekelle, Ethiopia
| |
Collapse
|
39
|
Oxidative Stress in Parkinson's Disease: Potential Benefits of Antioxidant Supplementation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2360872. [PMID: 33101584 PMCID: PMC7576349 DOI: 10.1155/2020/2360872] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 09/06/2020] [Accepted: 09/21/2020] [Indexed: 12/11/2022]
Abstract
Parkinson's disease (PD) occurs in approximately 1% of the population over 65 years of age and has become increasingly more common with advances in age. The number of individuals older than 60 years has been increasing in modern societies, as well as life expectancy in developing countries; therefore, PD may pose an impact on the economic, social, and health structures of these countries. Oxidative stress is highlighted as an important factor in the genesis of PD, involving several enzymes and signaling molecules in the underlying mechanisms of the disease. This review presents updated data on the involvement of oxidative stress in the disease, as well as the use of antioxidant supplements in its therapy.
Collapse
|
40
|
Augusto RL, Mendonça IP, de Albuquerque Rego GN, Pereira DD, da Penha Gonçalves LV, Dos Santos ML, de Souza RF, Moreno GMM, Cardoso PRG, de Souza Andrade D, da Silva-Júnior JC, Pereira MC, Peixoto CA, Medeiros-Linard CFB, de Souza IA, Andrade-da-Costa BLDS. Purified anacardic acids exert multiple neuroprotective effects in pesticide model of Parkinson's disease: in vivo and in silico analysis. IUBMB Life 2020; 72:1765-1779. [PMID: 32449271 DOI: 10.1002/iub.2304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 11/08/2022]
Abstract
Parkinson's disease (PD) induced by environmental toxins involves a multifactorial cascade of harmful factors, thus motivating the search for therapeutic agents able to act on the greatest number of molecular targets. This study evaluated the efficacy of 50 mg/kg purified anacardic acids (AAs), isolated from cashew nut shell liquid, on multiple steps of oxidative stress and inflammation induced by rotenone in the substantia nigra (SN) and striatum. Adult mice were divided into four groups: Control, rotenone, AAs + rotenone, and AAs alone. Lipoperoxidation, nitric oxide (NO) levels, and reduced glutathione (GSH)/oxidized gluthatione (GSSG) ratio were evaluated. NF-kB-p65, pro-IL-1β, cleaved IL-1β, metalloproteinase-9, Tissue Inhibitory Factor-1 (TIMP-1), tyrosine hydroxylase (TH), and glial fibrillary acidic protein (GFAP) levels were assessed by Western blot. In silico studies were also made using the SwissADME web tool. Rotenone increased lipoperoxidation and NO production and reduced TH levels and GSH/GSSG ratio in both SN and striatum. It also enhanced NF-kB-p65, pro, and cleaved IL-1β, MMP-9, GFAP levels compared to control and AAs groups. The AAs alone reduced pro-IL-1β in the striatum while they augmented TIMP1 and reduced MMP-9 amounts in both regions. AAs reversed rotenone-induced effects on lipoperoxidation, NO production, and GSH/GSSG ratio, as well as increased TH and attenuated pro-IL-1β and MMP-9 levels in both regions, NF-kB-p65 in the SN and GFAP in the striatum. Altogether, the in vivo and in silico analysis reinforced multiple and defined molecular targets of AAs, identifying that they are promising neuroprotective drug candidates for PD, acting against oxidative and inflammatory conditions induced by rotenone.
Collapse
Affiliation(s)
- Ricielle L Augusto
- Departamento de Fisiologia e Farmacologia, Universidade Federal de Pernambuco, UFPE, Recife, Brazil
| | - Ingrid P Mendonça
- Departamento de Fisiologia e Farmacologia, Universidade Federal de Pernambuco, UFPE, Recife, Brazil.,Departamento de Entomologia, Laboratório de Ultraestrutura, Instituto Aggeu Magalhães-FIOCRUZ, Recife, Brazil.,Instituto Nacional de Ciência e Tecnologia de Neuroimunomodulação (NIM), Rio de Janeiro, Brazil
| | | | - Danielle D Pereira
- Departamento de Fisiologia e Farmacologia, Universidade Federal de Pernambuco, UFPE, Recife, Brazil
| | | | - Maria L Dos Santos
- Instituto de Química, Divisão de Química orgânica, Universidade de Brasília, UnB, Brasilia, Brazil
| | - Raphael F de Souza
- Departamento de Fisiologia e Farmacologia, Universidade Federal de Pernambuco, UFPE, Recife, Brazil.,Departamento de Educação Física, Universidade Federal de Sergipe, UFS, São Cristóvam, Brazil
| | - Giselle M M Moreno
- Departamento de Fisiologia e Farmacologia, Universidade Federal de Pernambuco, UFPE, Recife, Brazil
| | - Pablo R G Cardoso
- Departamento de Fisiologia e Farmacologia, Universidade Federal de Pernambuco, UFPE, Recife, Brazil
| | - Daniele de Souza Andrade
- Departamento de Fisiologia e Farmacologia, Universidade Federal de Pernambuco, UFPE, Recife, Brazil
| | - José C da Silva-Júnior
- Departamento de Fisiologia e Farmacologia, Universidade Federal de Pernambuco, UFPE, Recife, Brazil
| | - Michelly C Pereira
- Departamento de Fisiologia e Farmacologia, Universidade Federal de Pernambuco, UFPE, Recife, Brazil
| | - Christina A Peixoto
- Departamento de Entomologia, Laboratório de Ultraestrutura, Instituto Aggeu Magalhães-FIOCRUZ, Recife, Brazil.,Instituto Nacional de Ciência e Tecnologia de Neuroimunomodulação (NIM), Rio de Janeiro, Brazil
| | | | - Ivone A de Souza
- Departamento de Antibióticos, Universidade Federal de Pernambuco, Recife, Brazil
| | | |
Collapse
|
41
|
Xu W, Zhang L, Geng Y, Liu Y, Zhang N. Long noncoding RNA GAS5 promotes microglial inflammatory response in Parkinson's disease by regulating NLRP3 pathway through sponging miR-223-3p. Int Immunopharmacol 2020; 85:106614. [PMID: 32470877 DOI: 10.1016/j.intimp.2020.106614] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 04/24/2020] [Accepted: 05/14/2020] [Indexed: 01/01/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder. Neuroinflammation induced by microglia plays an important role in the pathogenesis of PD. Long noncoding RNA GAS5 was showed to have significant effects on regulating inflammatory response. Here, we aim to investigate the effects of GAS5 on the inflammatory response of PD, and the underlying mechanism. An in vivo model of PD was established in C57BL/6 mice by rotenone and an in vitro cell model was conducted on microglia by lipopolysaccharide (LPS). Our results indicated that GAS5 was upregulated in tissues in a mice model of PD and microglia activated by LPS. Gain- and loss- of functional experiments demonstrated that GAS5 promoted the inflammation of microglia in vitro. Besides, the knockdown of GAS5 repressed the PD progression in vivo. Mechanistically, GAS5 positively regulated the NLRP3 expression via competitively sponging miR-223-3p. Overall, our finding illuminates that GAS5 accelerates PD progression through targeting miR-223-3p/NLRP3 axis.
Collapse
Affiliation(s)
- Wei Xu
- Department of Neurology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China.
| | - Ling Zhang
- College of Health Management, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yu Geng
- College of Health Management, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Ye Liu
- Electrocardial Center, the First Affiliated Hospital of Jinzhou Medical University. Jinzhou, Liaoning, China
| | - Ning Zhang
- Department of Hematology, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| |
Collapse
|
42
|
Hormati A, Ahmadpour S, Afkhami Ardekani M, Khodadust F, Refahi S. Radioprotective effects of montelukast, a selective leukotriene CysLT1 receptor antagonist, against nephrotoxicity induced by gamma radiation in mice. J Biochem Mol Toxicol 2020; 34:e22479. [PMID: 32125029 DOI: 10.1002/jbt.22479] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/24/2019] [Accepted: 02/14/2020] [Indexed: 12/30/2022]
Abstract
PURPOSE In this study, we evaluated the renal protective effects of montelukast (MLK) against ionizing radiation (IR) induced nephrotoxicity in mice. MATERIALS AND METHODS Radioprotective effects of MLK were assessed by biochemical analysis including measurements of kidney malondialdehyde (MDA), reduced glutathione (GSH), and serum creatinine and urea levels. Besides, for further evaluation of protective effects of MLK on renal system, 99m Tc-dimercaptosuccinic acid (DMSA) has been applied. The total antioxidant capacity of MLK was measured by using 1,1-diphenyl-2-picryl hydrazyl radical reagents and compared with butylated hydroxyl toluene standard antioxidant. RESULTS The biochemical evaluation revealed that better results have been achieved for the groups administered with MLK than the only radiation group. Besides only IR-treated mice group, those treated with MLK demonstrated a significant decrease in urea and creatinine levels. Statistically, significant differences of MDA and SHG levels (P < .05) were found between the radiation group and MLK plus IR-treated group. Also, 99m Tc-DMSA kidney uptake value (%ID/g) was observed lower for MLK plus IR-treated mice group than only radiation-treated mice group. CONCLUSIONS According to our findings, MLK has a potential role to be used as a renal protective agent against gamma radiation in radiotherapy.
Collapse
Affiliation(s)
- Ahmad Hormati
- Gastroenterology and Hepatology Disease Research Center, Qom University of Medical Sciences, Qom, Iran.,Gastrointestinal and Liver Disease Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Sajjad Ahmadpour
- Gastroenterology and Hepatology Disease Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Mahdieh Afkhami Ardekani
- Department of Radiology, Faculty of Para-Medicine, Hormozgan University of Medical Sciences, Bandare Abbas, Iran
| | - Fatemeh Khodadust
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Soheila Refahi
- Department of Medical Physics, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
43
|
Hafez HM, Hassanein H. Montelukast ameliorates doxorubicin-induced cardiotoxicity via modulation of p-glycoprotein and inhibition of ROS-mediated TNF-α/NF-κB pathways. Drug Chem Toxicol 2020; 45:548-559. [DOI: 10.1080/01480545.2020.1730885] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Heba M. Hafez
- Department of Pharmacology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Hanaa Hassanein
- Department of Histology, Faculty of Medicine, Minia University, Minia, Egypt
| |
Collapse
|
44
|
Duarte Azevedo M, Sander S, Tenenbaum L. GDNF, A Neuron-Derived Factor Upregulated in Glial Cells during Disease. J Clin Med 2020; 9:E456. [PMID: 32046031 PMCID: PMC7073520 DOI: 10.3390/jcm9020456] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/31/2020] [Accepted: 02/03/2020] [Indexed: 12/20/2022] Open
Abstract
In a healthy adult brain, glial cell line-derived neurotrophic factor (GDNF) is exclusively expressed by neurons, and, in some instances, it has also been shown to derive from a single neuronal subpopulation. Secreted GDNF acts in a paracrine fashion by forming a complex with the GDNF family receptor α1 (GFRα1), which is mainly expressed by neurons and can act in cis as a membrane-bound factor or in trans as a soluble factor. The GDNF/GFRα1 complex signals through interactions with the "rearranged during transfection" (RET) receptor or via the neural cell adhesion molecule (NCAM) with a lower affinity. GDNF can also signal independently from GFRα1 by interacting with syndecan-3. RET, which is expressed by neurons involved in several pathways (nigro-striatal dopaminergic neurons, motor neurons, enteric neurons, sensory neurons, etc.), could be the main determinant of the specificity of GDNF's pro-survival effect. In an injured brain, de novo expression of GDNF occurs in glial cells. Neuroinflammation has been reported to induce GDNF expression in activated astrocytes and microglia, infiltrating macrophages, nestin-positive reactive astrocytes, and neuron/glia (NG2) positive microglia-like cells. This disease-related GDNF overexpression can be either beneficial or detrimental depending on the localization in the brain and the level and duration of glial cell activation. Some reports also describe the upregulation of RET and GFRα1 in glial cells, suggesting that GDNF could modulate neuroinflammation.
Collapse
Affiliation(s)
| | | | - Liliane Tenenbaum
- Laboratory of Molecular Neurotherapies and NeuroModulation, Center for Neuroscience Research, Lausanne University Hospital, CHUV-Pavillon 3, av de Beaumont, CH-1010 Lausanne, Switzerland; (M.D.A.); (S.S.)
| |
Collapse
|
45
|
Modulation of neuroinflammation by cysteinyl leukotriene 1 and 2 receptors: implications for cerebral ischemia and neurodegenerative diseases. Neurobiol Aging 2019; 87:1-10. [PMID: 31986345 DOI: 10.1016/j.neurobiolaging.2019.12.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 12/04/2019] [Accepted: 12/14/2019] [Indexed: 12/21/2022]
Abstract
Neuroinflammation is a complex biological process and has been known to play an important role in age-related cerebrovascular and neurodegenerative disorders, such as cerebral ischemia, Alzheimer's disease, and Parkinson's disease. Cysteinyl leukotrienes (CysLTs) are potent inflammatory lipid mediators that exhibit actions mainly through activating type 1 and type 2 CysLT receptors (CysLT1 and CysLT2). Accumulating evidence shows that CysLT1 and CysLT2 are activated at different stages of pathological process in various cell types in the brain such as vascular endothelial cells, astrocytes, microglia, and neurons in response to insults. However, the precise roles and mechanisms of CysLT1 and CysLT2 in regulating the pathogenesis of cerebral ischemia, Alzheimer's disease, and Parkinson's disease are not fully understood. In this article, we focus on current advances that link activation of CysLT1 and CysLT2 to the pathological process during brain ischemia and neurodegeneration and discuss mechanisms by which CysLT1 and CysLT2 mediate inflammatory process and brain injury. Multitarget anti-inflammatory potentials of CysLT1 and CysLT2 antagonism for neuroinflammation and brain injury will also be reviewed.
Collapse
|
46
|
Tseng HC, Wang MH, Chang KC, Soung HS, Fang CH, Lin YW, Li KY, Yang CC, Tsai CC. Protective Effect of (-)Epigallocatechin-3-gallate on Rotenone-Induced Parkinsonism-like Symptoms in Rats. Neurotox Res 2019; 37:669-682. [PMID: 31811588 DOI: 10.1007/s12640-019-00143-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 11/13/2019] [Accepted: 11/21/2019] [Indexed: 01/29/2023]
Abstract
Rotenone (ROT)-induced neurotoxicity has been used for decades as an animal model of Parkinson's disease (PD) in humans. This model exhibits pathophysiological features similar to those reported in patients with PD, namely, striatal nitrosative and oxidative stress, mitochondrial dysfunction, and neural cytoarchitecture alteration. (-)Epigallocatechin-3-gallate (EGCG), the most abundant and potent green tea catechin, has notable anti-oxidative, anti-inflammatory, and neuroprotective effects. The objective of the present study was to investigate the potential protective effects of EGCG on ROT-induced motor and neurochemical dysfunctions in rats. Furthermore, we also aimed to study the neuroprotective mechanisms underlying these effects. ROT treatment (0.5 mg/kg s.c., 21 days) reduced body weight and induced significant motor impairments as assessed using an open-field test, rotarod test, grip strength measurement, and beam-crossing task. EGCG treatment (100 or 300 mg/kg i.p., 60 min prior to ROT administration, 21 days) prevented most of the ROT-induced motor impairments. Moreover, EGCG treatment reduced ROT-induced nitric oxide (NO) level and lipid peroxidation (LPO) production; increased the activity of succinate dehydrogenase (SDH), ATPase, and ETC enzymes and the levels of catecholamines in the striatum; and reduced the levels of neuroinflammatory and apoptotic markers. These results demonstrate the possible neuroprotective effects of EGCG against ROT-induced motor impairments, including anti-oxidatory effect, prevention of mitochondrial dysfunction, prevention of neurochemical deficiency, anti-neuroinflammatory effect, and anti-apoptotic effect. This is the first report about the neuroprotective effect of EGCG against ROT-induced motor impairments, and the above evidence provides a potential clinically relevant role for EGCG in delaying or treating human PD.
Collapse
Affiliation(s)
- Hsiang-Chien Tseng
- Department of Anesthesiology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, 11101, Taiwan, Republic of China.,School of Medicine, Fu Jen Catholic University, New Taipei City, 24205, Taiwan, Republic of China
| | - Mao-Hsien Wang
- Department of Anesthesia, En Chu Kon Hospital, Sanshia District, New Taipei City, 23702, Taiwan, Republic of China
| | - Kuo-Chi Chang
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei, 10608, Taiwan, Republic of China
| | - Hung-Sheng Soung
- Department of Psychiatry, Yuan-Shan Br. of Taipei Veteran General Hospital, Yilan County, 26604, Taiwan, Republic of China.,Department of Biomedical Engineering, National Defense Medical Center, Taipei, 11490, Taiwan, Republic of China
| | - Chih-Hsiang Fang
- Institute of Biomedical Engineering, National Taiwan University, Taipei, 10051, Taiwan, Republic of China
| | - Yi-Wen Lin
- Institute of Biomedical Engineering, National Taiwan University, Taipei, 10051, Taiwan, Republic of China
| | - Keng-Yuan Li
- Institute of Biomedical Engineering, National Taiwan University, Taipei, 10051, Taiwan, Republic of China
| | - Chih-Chuan Yang
- Department of Neurosurgery, Mackay Memorial Hospital, Taipei, 10449, Taiwan, Republic of China.,Department of Medicine, Mackay Medical College, New Taipei City, 252, Taiwan, Republic of China
| | - Cheng-Chia Tsai
- Department of Neurosurgery, Mackay Memorial Hospital, Taipei, 10449, Taiwan, Republic of China. .,Department of Medicine, Mackay Medical College, New Taipei City, 252, Taiwan, Republic of China.
| |
Collapse
|
47
|
Köse E, Oğuz F, Vardi N, Ediz Sarihan M, Beytur A, Yücel A, Polat A, Eki Nci N. Therapeutic and protective effects of montelukast against doxorubicin-induced acute kidney damage in rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2019; 22:407-411. [PMID: 31168345 PMCID: PMC6535195 DOI: 10.22038/ijbms.2019.33493.7996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Objective(s): The current study was designed to investigate the therapeutic and protective effects of montelukast (ML) against doxorubicin (DOX)-induced acute kidney damage in rats. Materials and Methods: Thirty-five Wistar albino female rats were randomly divided into 5 groups as follows: Group I: Control; Group II: Control+ML; Group III: DOX; Group IV: DOX+ML; Group V: ML+DOX. At the end of the experiment, the kidney tissues of rats were collected. Thiobarbituric acid reactive substance (TBARS), reduced glutathione, superoxide dismutase (SOD), and catalase levels were determined from the kidney tissues. In addition, the kidney tissues were examined histologically. Results: DOX induced a significant increase in the kidney TBARS levels, whereas SOD contents significantly decreased when compared with the control group. On the other hand, ML administration before and after DOX injection caused significant decreases in TBARS production and also increases in SOD levels. Histologically, the most remarkable damage was glomerulosclerosis and tubular changes in the DOX group. Moreover, marked tubular necrosis and swelling in tubular epithelial cells were observed in this group. Contrarily, although glomerulosclerosis was recognized as alleviated also in both DOX+ML and ML+DOX groups, the lesions did not completely ameliorate. However, treatment with ML after DOX injection was more effective than treatment with ML before DOX injection with respect to the protection of tubular structures. Conclusion: It was determined that ML treatment after DOX injection caused therapeutic effects against DOX-induced kidney damage. Thence, ML treatment is of some clinical properties for oxidative stress damage in kidney tissues.
Collapse
Affiliation(s)
- Evren Köse
- Department of Anatomy, School of Medicine, Inonu University, Malatya, Turkey
| | - Fatih Oğuz
- Department of Urology, School of Medicine, Inonu University, Malatya, Turkey
| | - Nigar Vardi
- Department of Histology-Embryology, School of Medicine, Inonu University, Malatya, Turkey
| | - Mehmet Ediz Sarihan
- Department of Emergency, School of Medicine, Inonu University, Malatya, Turkey
| | - Ali Beytur
- Department of Urology, School of Medicine, Inonu University, Malatya, Turkey
| | - Aytaç Yücel
- Department of Anesthesiology and Reanimation, School of Medicine, Inonu University, Malatya, Turkey
| | - Alaadin Polat
- Department of Physiology, School of Medicine, Inonu University, Malatya, Turkey
| | - Nihat Eki Nci
- Department of Anatomy, School of Medicine, Erciyes University, Kayseri, Turkey
| |
Collapse
|
48
|
Jiao Z, Zhang W, Chen C, Zhu X, Chen X, Zhou M, Peng G, Liu H, Qiu J, Lin Y, Huang S, Mo M, Yang X, Qu S, Xu P. Gene Dysfunction Mediates Immune Response to Dopaminergic Degeneration in Parkinson's Disease. ACS Chem Neurosci 2019; 10:803-811. [PMID: 30289236 DOI: 10.1021/acschemneuro.8b00373] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Many publications reported that genetic dysfunction mediates abnormal immune responses in the brain, which is important for the development of neurodegenerative diseases, especially for Parkinson's disease (PD). This immune disorder results in subsequent inflammatory reaction, which stimulates microglia or other immune cells to secrete cytokines and chemokines and disturbs the proportion of peripheral blood lymphocyte subsets contributing to dopaminergic (DA) neuron apoptosis. Furthermore, the abnormal immune related signal pathways caused by genetic variants promote chronic inflammation destroying the blood-brain barrier, which allows infiltration of different molecules and blood cells into the central nervous system (CNS) exerting toxicity on DA neurons. As a result, the inflammatory reaction in the CNS accelerates the progression of Parkinson's disease and promotes α-synuclein aggregation and diffusion among DA neurons in the procession of Parkinson's disease. Thus, for disease evaluation, the genetic mediated abnormal immune response in PD may be assessed based on the multiple immune molecules and inflammatory factors, as well as the ratio of lymphocyte subsets from PD patient's peripheral blood as potential biomarkers.
Collapse
Affiliation(s)
- Zhigang Jiao
- Central Laboratory, Shunde Hospital, Southern Medical University, Foshan 528300, China
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Foshan 528300, China
| | - Wenlong Zhang
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Chaojun Chen
- Department of Neurology, Guangzhou Chinese Medical Integrated Hospital (Huadu), Guangdong 510800, China
| | - Xiaoqin Zhu
- Guangzhou Medical University, Guangzhou 511436, China
| | - Xiang Chen
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Miaomiao Zhou
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Guoyou Peng
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Hanqun Liu
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Jiewen Qiu
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Yuwan Lin
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Shuxuan Huang
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Mingshu Mo
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Xinling Yang
- Department of Neurology, the Third Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Shaogang Qu
- Central Laboratory, Shunde Hospital, Southern Medical University, Foshan 528300, China
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Foshan 528300, China
| | - Pingyi Xu
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| |
Collapse
|
49
|
Abstract
Parkinson’s disease (PD) is a neurodegenerative disease characterized by a progressive loss of dopaminergic neurons from the nigrostriatal pathway, formation of Lewy bodies, and microgliosis. During the past decades multiple cellular pathways have been associated with PD pathology (i.e., oxidative stress, endosomal-lysosomal dysfunction, endoplasmic reticulum stress, and immune response), yet disease-modifying treatments are not available. We have recently used genetic data from familial and sporadic cases in an unbiased approach to build a molecular landscape for PD, revealing lipids as central players in this disease. Here we extensively review the current knowledge concerning the involvement of various subclasses of fatty acyls, glycerolipids, glycerophospholipids, sphingolipids, sterols, and lipoproteins in PD pathogenesis. Our review corroborates a central role for most lipid classes, but the available information is fragmented, not always reproducible, and sometimes differs by sex, age or PD etiology of the patients. This hinders drawing firm conclusions about causal or associative effects of dietary lipids or defects in specific steps of lipid metabolism in PD. Future technological advances in lipidomics and additional systematic studies on lipid species from PD patient material may improve this situation and lead to a better appreciation of the significance of lipids for this devastating disease.
Collapse
|
50
|
Bohush A, Niewiadomska G, Filipek A. Role of Mitogen Activated Protein Kinase Signaling in Parkinson's Disease. Int J Mol Sci 2018; 19:ijms19102973. [PMID: 30274251 PMCID: PMC6213537 DOI: 10.3390/ijms19102973] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 09/25/2018] [Accepted: 09/26/2018] [Indexed: 12/31/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder caused by insufficient dopamine production due to the loss of 50% to 70% of dopaminergic neurons. A shortage of dopamine, which is predominantly produced by the dopaminergic neurons within the substantia nigra, causes clinical symptoms such as reduction of muscle mass, impaired body balance, akinesia, bradykinesia, tremors, postural instability, etc. Lastly, this can lead to a total loss of physical movement and death. Since no cure for PD has been developed up to now, researchers using cell cultures and animal models focus their work on searching for potential therapeutic targets in order to develop effective treatments. In recent years, genetic studies have prominently advocated for the role of improper protein phosphorylation caused by a dysfunction in kinases and/or phosphatases as an important player in progression and pathogenesis of PD. Thus, in this review, we focus on the role of selected MAP kinases such as JNKs, ERK1/2, and p38 MAP kinases in PD pathology.
Collapse
Affiliation(s)
- Anastasiia Bohush
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland.
| | - Grazyna Niewiadomska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland.
| | - Anna Filipek
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland.
| |
Collapse
|