1
|
Ghaith WZ, Wadie W, El-Yamany MF. Crosstalk between SIRT1/Nrf2 signaling and NLRP3 inflammasome/pyroptosis as a mechanistic approach for the neuroprotective effect of linagliptin in Parkinson's disease. Int Immunopharmacol 2025; 145:113716. [PMID: 39642562 DOI: 10.1016/j.intimp.2024.113716] [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: 08/31/2024] [Revised: 11/24/2024] [Accepted: 11/24/2024] [Indexed: 12/09/2024]
Abstract
In recent years, special attention has been paid to highlighting the antiparkinsonian effect of linagliptin. However, the mechanism of its action has not yet been well investigated. The present study aimed to verify the neuroprotective effect of linagliptin in the rotenone model of Parkinson's disease (PD) and further explore its potential molecular mechanisms. Rats were intoxicated with rotenone (2 mg/kg/day; sc) and treated with linagliptin (10 mg/kg/day; po) for 14 consecutive days. The present finding showed that linagliptin ameliorated the histopathological changes of rotenone on substantia nigra and striata. Linagliptin decreased α-synuclein immunoreactivity along with an increase in tyrosine hydroxylase immunoreactivity and striatal dopamine content. This was reflected in the marked improvement of the behavior and motor deficits in rotenone-intoxicated rats. On the molecular level, linagliptin upregulated sirtuin 1 (SIRT1)/ nuclear factor erythroid 2-related factor 2 (Nrf2) signaling, reduced ionized calcium-binding adaptor molecule 1 (Iba1) protein expression, restored glutathione (GSH) content, and elevated heme oxygenase-1 (HO-1) level in rats with rotenone intoxication. Moreover, linagliptin inhibited NOD-like receptor protein 3 (NLRP3)/caspase-1/interleukin-1β (IL-1β) cascade with subsequent reduction in gasdermin D (GSDMD) expression. Therefore, the present study reveals the ability of linagliptin, through the activation of SIRT1/Nrf2 signaling, to suppress NLRP3 inflammasome-mediated pyroptosis and protect against rotenone-induced parkinsonism.
Collapse
Affiliation(s)
| | - Walaa Wadie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Egypt
| | - Mohammed F El-Yamany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Egypt.
| |
Collapse
|
2
|
Sood R, Sanjay, Kang SU, Yoon NY, Lee HJ. Malvidin-3-O-Glucoside Mitigates α-Syn and MPTP Co-Induced Oxidative Stress and Apoptosis in Human Microglial HMC3 Cells. Int J Mol Sci 2024; 25:12733. [PMID: 39684444 DOI: 10.3390/ijms252312733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/08/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Parkinson's disease (PD) is a widespread age-related neurodegenerative disorder characterized by the presence of an aggregated protein, α-synuclein (α-syn), which is encoded by the SNCA gene and localized to presynaptic terminals in a normal human brain. The α-syn aggregation is induced by the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mitochondrial neurotoxin and is therefore used to mimic PD-like pathology in various in vitro and in vivo models. However, in vitro PD-like pathology using α-syn and MPTP in human microglial cells has not yet been reported. Malvidin-3-O-glucoside (M3G) is a major anthocyanin primarily responsible for pigmentation in various fruits and beverages and has been reported to possess various bioactivities. However, the neuroprotective effects of M3G in humanized in vitro PD-like pathologies have not been reported. Therefore, individual and co-treatments of α-syn and MPTP in a human microglial (HMC3) cell line were used to establish a humanized PD-like pathology model in vitro. The individual treatments were significantly less cytotoxic when compared to the α-syn and MPTP co-treatment. This study examined the neuroprotective effects of M3G by treating HMC3 cells with α-syn (8 μg/mL) and MPTP (2 mM) individually or in a co-treatment in the presence or absence of M3G (50 μM). M3G demonstrated anti-apoptotic, anti-inflammatory, and antioxidative properties against the α-syn- and MPTP-generated humanized in vitro PD-like pathology. This study determined that the cytoprotective effects of M3G are mediated by nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase (HO)-1 signaling.
Collapse
Affiliation(s)
- Rachit Sood
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam 13120, Republic of Korea
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam 13120, Republic of Korea
| | - Sanjay
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam 13120, Republic of Korea
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam 13120, Republic of Korea
| | - Sung-Ung Kang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Na Young Yoon
- Food Safety and Processing Research Division, National Institute of Fisheries Science, Busan 46083, Republic of Korea
| | - Hae-Jeung Lee
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam 13120, Republic of Korea
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam 13120, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Science and Technology (GAIHST), Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
3
|
He S, Chen Y, Wang H, Li S, Wei Y, Zhang H, Gao Q, Wang F, Zhang R. Neuroprotective effects of chlorogenic acid: Modulation of Akt/Erk1/2 signaling to prevent neuronal apoptosis in Parkinson's disease. Free Radic Biol Med 2024; 222:275-287. [PMID: 38925315 DOI: 10.1016/j.freeradbiomed.2024.06.018] [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: 05/13/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 06/28/2024]
Abstract
As a prevalent neurodegenerative disorder, Parkinson's disease is associated with oxidative stress. Our recent investigations revealed that reactive oxygen species (ROS) and PD-toxins like 6-hydroxydopamine (6-OHDA) can induce neuronal apoptosis through over-activation of Akt signaling. Chlorogenic acid (CGA), a natural acid phenol abundant in the human diet, is well-documented for its ability to mitigate intracellular ROS. In this study, we utilized CGA to treat experimental models of PD both in vitro and in vivo. Our study results demonstrated that SH-SY5Y and primary neurons exhibited cell apoptosis in response to 6-OHDA. Pretreatment with CGA significantly attenuated PD toxins-induced large amount of ROS, inhibiting Erk1/2 activation, preventing Akt inhibition, and hindering neuronal cell death. Combining the Erk1/2 inhibitor U0126 with CGA could reverse 6-OHDA-induced Akt inhibition, ROS, and apoptosis in the cells. Crucially, the Akt activator SC79 and ROS scavenger NAC both could eliminate excessive ROS via Akt and Erk1/2 signaling pathways, and CGA further potentiated these effects in PD models. Behavioral experiments revealed that CGA could alleviate gait abnormalities in PD model mice. The neuroprotective effects have been demonstrated in several endocrine regions and in the substantia nigra tissue, which shows the positive tyrosine hydroxylase (TH). Overall, our results suggest that CGA prevents the activation of Erk1/2 and inactivation of Akt by removing excess ROS in PD models. These findings propose a potential strategy for mitigating neuronal degeneration in Parkinson's disease by modulating the Akt/Erk1/2 signaling pathway through the administration of CGA and/or the use of antioxidants to alleviate oxidative stress.
Collapse
Affiliation(s)
- Shuai He
- College of Life Sciences, Anhui Medical University, Anhui, 230032, PR China
| | - Yuxiang Chen
- College of Life Sciences, Anhui Medical University, Anhui, 230032, PR China
| | - Hui Wang
- College of Life Sciences, Anhui Medical University, Anhui, 230032, PR China
| | - Shupei Li
- College of Life Sciences, Anhui Medical University, Anhui, 230032, PR China
| | - Yu Wei
- College of Life Sciences, Anhui Medical University, Anhui, 230032, PR China
| | - Hui Zhang
- College of Life Sciences, Anhui Medical University, Anhui, 230032, PR China
| | - Qian Gao
- College of Life Sciences, Anhui Medical University, Anhui, 230032, PR China
| | - Fengsong Wang
- College of Life Sciences, Anhui Medical University, Anhui, 230032, PR China.
| | - Ruijie Zhang
- College of Life Sciences, Anhui Medical University, Anhui, 230032, PR China.
| |
Collapse
|
4
|
Yin X, Wang M, Li F, Wang Z, Gao Z. Sjögren's syndrome and Parkinson's disease: a bidirectional Mendelian randomization study. Front Genet 2024; 15:1370245. [PMID: 39104742 PMCID: PMC11298492 DOI: 10.3389/fgene.2024.1370245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024] Open
Abstract
Background Previous epidemiological studies have reported an association between Sjögren's syndrome (SS) and Parkinson's disease (PD); however, the causality and direction of this relationship remain unclear. In this study, we aimed to investigate the causal relationship between genetically determined SS and the risk of PD using bidirectional Mendelian randomization (MR). Methods Summary statistics for Sjögren's syndrome used as exposure were obtained from the FinnGen database, comprising 1,290 cases and 213,145 controls. The outcome dataset for PD was derived from the United Kingdom Biobank database, including 6,998 cases and 415,466 controls. Various MR methods, such as inverse variance weighted (IVW), Mendelian randomization Egger regression (MR-Egger), weighted median (WM), simple mode, weighted mode, MR-pleiotropy residual sum and outlier (MR-PRESSO), and robust adjusted profile score (RAPS), were employed to investigate the causal effects of SS on PD. Instrumental variable strength evaluation and sensitivity analyses were conducted to ensure the reliability of the results. In addition, reverse MR analysis was performed to examine the causal effects of PD on SS. Results The WM, IVW, RAPS and MR-PRESSO methods demonstrated a significant association between genetically predicted SS and reduced risk of PD (odds ratio ORWM = 0.9988, ORIVW = 0.9987, ORRAPS = 0.9987, ORMR-PRESSO = 0.9987, respectively, P < 0.05). None of the MR analyses showed evidence of horizontal pleiotropy (P > 0.05) based on the MR-Egger and MR-PRESSO tests, and there was no statistical heterogeneity in the test results of the MR-Egger and IVW methods. The leave-one-out sensitivity analysis confirmed the robustness of the causal relationship between SS and PD. Furthermore, reverse MR analysis did not support any causal effects of PD on SS. Conclusion Our MR study supports a potential causal association between SS and a reduced risk of PD. Further extensive clinical investigations and comprehensive fundamental research are warranted to elucidate the underlying mechanisms linking SS and PD.
Collapse
Affiliation(s)
| | | | | | - Zhenfu Wang
- Department of Neurology, The Second Medical Center and National Clinical Research Center for Geriatric Disease, Chinese PLA General Hospital, Beijing, China
| | - Zhongbao Gao
- Department of Neurology, The Second Medical Center and National Clinical Research Center for Geriatric Disease, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
5
|
Bandiwadekar A, Jose J, Gopan G, Augustin V, Ashtekar H, Khot KB. Transdermal delivery of resveratrol loaded solid lipid nanoparticle as a microneedle patch: a novel approach for the treatment of Parkinson's disease. Drug Deliv Transl Res 2024:10.1007/s13346-024-01656-0. [PMID: 38949746 DOI: 10.1007/s13346-024-01656-0] [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] [Accepted: 06/15/2024] [Indexed: 07/02/2024]
Abstract
Parkinson's disease (PD), affecting millions of people worldwide and expected to impact 10 million by 2030, manifests a spectrum of motor and non-motor symptoms linked to the decline of dopaminergic neurons. Current therapies manage PD symptoms but lack efficacy in slowing disease progression, emphasizing the urgency for more effective treatments. Resveratrol (RSV), recognized for its neuroprotective and antioxidative properties, encounters challenges in clinical use for PD due to limited bioavailability. Researchers have investigated lipid-based nanoformulations, specifically solid lipid nanoparticles (SLNs), to enhance RSV stability. Oral drug delivery via SLNs faces obstacles, prompting exploration into transdermal delivery using SLNs integrated with microneedles (MNs) for improved patient compliance. In this study, an RSV-loaded SLNs (RSV -SLNs) incorporated into the MN patch was developed for transdermal RSV delivery to improve its stability and patient compliance. Characterization studies demonstrated favorable physical properties of SLNs with a sustained drug release profile of 78.36 ± 0.74%. The developed MNs exhibited mechanical robustness and skin penetration capabilities. Ex vivo permeation studies displayed substantial drug permeation of 68.39 ± 1.4% through the skin. In an in vivo pharmacokinetic study, the RSV-SLNs delivered through MNs exhibited a significant increase in Cmax, Tmax, and AUC0 - t values, alongside a reduced elimination rate in blood plasma in contrast to the administration of pure RSV via MNs. Moreover, an in vivo study showcased enhanced behavioral functioning and increased brain antioxidant levels in the treated animals. In-vivo skin irritation study revealed no signs of irritation till 24 h which permits long-term MNs application. Histopathological analysis showed notable changes in the brain regions of the rat, specifically the striatum and substantia nigra, after the completion of the treatment. Based on these findings, the development of an RSV-SLN loaded MNs (RSVSNLMP) patch presents a novel approach, with the potential to enhance the drug's efficiency, patient compliance, and therapeutic outcomes for PD, offering a promising avenue for advanced PD therapy.
Collapse
Affiliation(s)
- Akshay Bandiwadekar
- NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics,, NITTE Deemed-to-be University, Mangalore, 575018, India
| | - Jobin Jose
- NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics,, NITTE Deemed-to-be University, Mangalore, 575018, India.
| | - Gopika Gopan
- NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics,, NITTE Deemed-to-be University, Mangalore, 575018, India
| | - Varsha Augustin
- NGSM Institute of Pharmaceutical Sciences, NITTE Deemed-to-be University, Department of NITTE University Center for Animal Research & Experimentation (NUCARE), Mangalore, 575018, India
| | - Harsha Ashtekar
- NGSM Institute of Pharmaceutical Sciences, Department of Pharmacology, NITTE Deemed-to-be University, Mangalore, 575018, India
| | - Kartik Bhairu Khot
- NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics,, NITTE Deemed-to-be University, Mangalore, 575018, India
| |
Collapse
|
6
|
Azzini E, Peña-Corona SI, Hernández-Parra H, Chandran D, Saleena LAK, Sawikr Y, Peluso I, Dhumal S, Kumar M, Leyva-Gómez G, Martorell M, Sharifi-Rad J, Calina D. Neuroprotective and anti-inflammatory effects of curcumin in Alzheimer's disease: Targeting neuroinflammation strategies. Phytother Res 2024; 38:3169-3189. [PMID: 38616356 DOI: 10.1002/ptr.8200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 03/17/2024] [Accepted: 03/18/2024] [Indexed: 04/16/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by the accumulation of amyloid-beta plaques and neurofibrillary tangles, leading to neuronal loss. Curcumin, a polyphenolic compound derived from Curcuma longa, has shown potential neuroprotective effects due to its anti-inflammatory and antioxidant properties. This review aims to synthesize current preclinical data on the anti-neuroinflammatory mechanisms of curcumin in the context of AD, addressing its pharmacokinetics, bioavailability, and potential as a therapeutic adjunct. An exhaustive literature search was conducted, focusing on recent studies within the last 10 years related to curcumin's impact on neuroinflammation and its neuroprotective role in AD. The review methodology included sourcing articles from specialized databases using specific medical subject headings terms to ensure precision and relevance. Curcumin demonstrates significant neuroprotective properties by modulating neuroinflammatory pathways, scavenging reactive oxygen species, and inhibiting the production of pro-inflammatory cytokines. Despite its potential, challenges remain regarding its limited bioavailability and the scarcity of comprehensive human clinical trials. Curcumin emerges as a promising therapeutic adjunct in AD due to its multimodal neuroprotective benefits. However, further research is required to overcome challenges related to bioavailability and to establish effective dosing regimens in human subjects. Developing novel delivery systems and formulations may enhance curcumin's therapeutic potential in AD treatment.
Collapse
Affiliation(s)
- Elena Azzini
- Research Centre for Food and Nutrition, Council for Agricultural Research and Economics (CREA-AN), Rome, Italy
| | - Sheila I Peña-Corona
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Héctor Hernández-Parra
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Deepak Chandran
- Department of Veterinary Sciences and Animal Husbandry, Amrita School of Agricultural Sciences, Amrita Vishwa Vidyapeetham University, Coimbatore, India
| | | | - Yousef Sawikr
- Department of Pharmacology and Toxicology, Faculty of Medicine University of Ajdabiya, Ajdabiya, Libya
| | - Ilaria Peluso
- Research Centre for Food and Nutrition, Council for Agricultural Research and Economics (CREA-AN), Rome, Italy
| | - Sangram Dhumal
- Division of Horticulture, RCSM College of Agriculture, Kolhapur, India
| | - Manoj Kumar
- Chemical and Biochemical Processing Division, ICAR-Central Institute for Research on Cotton Technology, Mumbai, India
| | - Gerardo Leyva-Gómez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Miquel Martorell
- Department of Nutrition and Dietetics, Faculty of Pharmacy, and Centre for Healthy Living, University of Concepción, Concepción, Chile
| | | | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| |
Collapse
|
7
|
Al‐Qahtani Z, Al‐kuraishy HM, Al‐Gareeb AI, Albuhadily AK, Ali NH, Alexiou A, Papadakis M, Saad HM, Batiha GE. The potential role of brain renin-angiotensin system in the neuropathology of Parkinson disease: Friend, foe or turncoat? J Cell Mol Med 2024; 28:e18495. [PMID: 38899551 PMCID: PMC11187740 DOI: 10.1111/jcmm.18495] [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: 11/04/2023] [Revised: 01/15/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024] Open
Abstract
Parkinson disease (PD) is one of the most common neurodegenerative diseases of the brain. Of note, brain renin-angiotensin system (RAS) is intricate in the PD neuropathology through modulation of oxidative stress, mitochondrial dysfunction and neuroinflammation. Therefore, modulation of brain RAS by angiotensin receptor blockers (ARBs) and angiotensin-converting enzyme inhibitors (ACEIs) may be effective in reducing the risk and PD neuropathology. It has been shown that all components including the peptides and enzymes of the RAS are present in the different brain areas. Brain RAS plays a critical role in the regulation of memory and cognitive function, and in the controlling of central blood pressure. However, exaggerated brain RAS is implicated in the pathogenesis of different neurodegenerative diseases including PD. Two well-known pathways of brain RAS are recognized including; the classical pathway which is mainly mediated by AngII/AT1R has detrimental effects. Conversely, the non-classical pathway which is mostly mediated by ACE2/Ang1-7/MASR and AngII/AT2R has beneficial effects against PD neuropathology. Exaggerated brain RAS affects the viability of dopaminergic neurons. However, the fundamental mechanism of brain RAS in PD neuropathology was not fully elucidated. Consequently, the purpose of this review is to disclose the mechanistic role of RAS in in the pathogenesis of PD. In addition, we try to revise how the ACEIs and ARBs can be developed for therapeutics in PD.
Collapse
Affiliation(s)
- Zainah Al‐Qahtani
- Neurology Section, Internal Medicine Department, College of MedicineKing khaled universityAbhaSaudi Arabia
| | - Hayder M. Al‐kuraishy
- Clinical pharmacology and medicine, college of medicineMustansiriyah UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Clinical pharmacology and medicine, college of medicineMustansiriyah UniversityBaghdadIraq
| | - Ali K. Albuhadily
- Clinical pharmacology and medicine, college of medicineMustansiriyah UniversityBaghdadIraq
| | - Naif H. Ali
- Department of Internal Medicine, Medical CollegeNajran UniversityNajranSaudi Arabia
| | - Athanasios Alexiou
- University Centre for Research & DevelopmentChandigarh UniversityMohaliIndia
- Department of Science and EngineeringNovel Global Community Educational FoundationHebershamNew South WalesAustralia
- Department of Research & Development, FunogenAthensGreece
- Department of Research & DevelopmentAFNP MedWienAustria
| | - Marios Papadakis
- Department of Surgery IIUniversity Hospital Witten‐HerdeckeWuppertalGermany
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary MedicineMatrouh UniversityMatrouhEgypt
| | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhour UniversityDamanhourAlBeheiraEgypt
| |
Collapse
|
8
|
Abdolmaleki A, Karimian A, Khoshnazar SM, Asadi A, Samarein ZA, Smail SW, Bhattacharya D. The role of Nrf2 signaling pathways in nerve damage repair. Toxicol Res (Camb) 2024; 13:tfae080. [PMID: 38799411 PMCID: PMC11116835 DOI: 10.1093/toxres/tfae080] [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: 10/25/2023] [Revised: 03/05/2024] [Accepted: 05/17/2024] [Indexed: 05/29/2024] Open
Abstract
The protein, Nuclear factor-E2-related factor 2 (Nrf2), is a transitory protein that acts as a transcription factor and is involved in the regulation of many cytoprotective genes linked to xenobiotic metabolism and antioxidant responses. Based on the existing clinical and experimental data, it can be inferred that neurodegenerative diseases are characterized by an excessive presence of markers of oxidative stress (OS) and a reduced presence of antioxidant defense systems in both the brain and peripheral tissues. The presence of imbalances in the homeostasis between oxidants and antioxidants has been recognized as a substantial factor in the pathogenesis of neurodegenerative disorders. The dysregulations include several cellular processes such as mitochondrial failure, protein misfolding, and neuroinflammation. These dysregulations all contribute to the disruption of proteostasis in neuronal cells, leading to their eventual mortality. A noteworthy component of Nrf2, as shown by recent research undertaken over the last decade, is to its role in the development of resistance to OS. Nrf2 plays a pivotal role in regulating systems that defend against OS. Extant research offers substantiation for the protective and defensive roles of Nrf2 in the context of neurodegenerative diseases. The purpose of this study is to provide a comprehensive analysis of the influence of Nrf2 on OS and its function in regulating antioxidant defense systems within the realm of neurodegenerative diseases. Furthermore, we evaluate the most recent academic inquiries and empirical evidence about the beneficial and potential role of certain Nrf2 activator compounds within the realm of therapeutic interventions.
Collapse
Affiliation(s)
- Arash Abdolmaleki
- Department of Biophysics, Faculty of Advanced Technologies, University of Mohaghegh Ardabili, PO Box: 179, Ardabil, 11367-56199, Iran
| | - Aida Karimian
- Department of Biology, Faculty of Science, University of Mohaghegh Ardabili, PO Box: 179, Ardabil, 11367-56199, Iran
| | - Seyedeh Mahdieh Khoshnazar
- Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Imam Khomeini Highway, Mustafa Khomeini Boulevard, Ibn Sina, Kerman, 9986598, Iran
| | - Asadollah Asadi
- Department of Biology, Faculty of Science, University of Mohaghegh Ardabili, PO Box: 179, Ardabil, 11367-56199, Iran
| | - Zahra Akhavi Samarein
- Department of Counseling, Faculty of Education and Psychology, University of Mohaghegh Ardabili, PO Box: 179, Ardabil, 11367-56199, Iran
| | - Shukur Wasman Smail
- Department of Medical Microbiology, College of Science, Cihan University-Erbil, Kurdistan Region, 1235897, Iraq
| | - Deepak Bhattacharya
- Ph.D., Policy, Nursing, At Fight-Cancer at Home, Medicinal Toxicology & QC, Sri Radha Krishna Raas Mandir, KedarGouri Road, Bhubaneswar, Odisa 751002, India
| |
Collapse
|
9
|
Kryl'skii ED, Razuvaev GA, Popova TN, Oleinik SA, Medvedeva SM, Shikhaliev KS. 6-Hydroxy-2,2,4-trimethyl-1,2,3,4-tetrahydroquinoline Demonstrates Neuroprotective Properties in Experimental Parkinson's Disease by Enhancing the Antioxidant System, Normalising Chaperone Activity and Suppressing Apoptosis. Neurochem Res 2024; 49:1387-1405. [PMID: 38502411 DOI: 10.1007/s11064-024-04125-9] [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: 11/14/2023] [Revised: 02/06/2024] [Accepted: 02/10/2024] [Indexed: 03/21/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease, whereby disturbances within the antioxidant defence system, increased aggregation of proteins, and activation of neuronal apoptosis all have a crucial role in the pathogenesis. In this context, exploring the neuroprotective capabilities of compounds that sustain the effectiveness of cellular defence systems in neurodegenerative disorders is worthwhile. During this study, we assessed how 6-hydroxy-2,2,4-trimethyl-1,2,3,4-tetrahydroquinoline (HTHQ), which has antioxidant properties, affects the functioning of the antioxidant system, the activity of NADPH-generating enzymes and chaperones, and the level of apoptotic processes in rats with rotenone-induced PD. Six groups of animals were formed for our experiment, each with 12 animals. These were: a control group, animals with rotenone-induced PD, rats with PD given HTHQ at a dose of 50 mg/kg, rats with PD given HTHQ at a dose of 25 mg/kg, animals with pathology who were administered a comparison drug rasagiline, and control animals who were administered HTHQ at a dose of 50 mg/kg. The study results indicate that administering HTHQ led to a significant decrease in oxidative stress in PD rats. The enhanced redox status in animal tissues was linked with the recovery of antioxidant enzyme activities and NADPH-generating enzyme function, as well as an upsurge in the mRNA expression levels of antioxidant genes and factors Nrf2 and Foxo1. Administering HTHQ to rats with PD normalized the chaperone-like activity and mRNA levels of heat shock protein 70. Rats treated with the compound displayed lower apoptosis intensity when compared to animals with pathology. Therefore, owing to its antioxidant properties, HTHQ demonstrated a beneficial impact on the antioxidant system, resulting in decreased requirements for chaperone activation and the inhibition of apoptosis processes triggered in PD. HTHQ at a dose of 50 mg/kg had a greater impact on the majority of the examined variables compared to rasagiline.
Collapse
Affiliation(s)
- Evgenii D Kryl'skii
- Department of Medical Biochemistry, Molecular and Cell Biology, Voronezh State University, Universitetskaya Sq. 1, Voronezh, Russia, 394018.
| | - Grigorii A Razuvaev
- Department of Medical Biochemistry, Molecular and Cell Biology, Voronezh State University, Universitetskaya Sq. 1, Voronezh, Russia, 394018
| | - Tatyana N Popova
- Department of Medical Biochemistry, Molecular and Cell Biology, Voronezh State University, Universitetskaya Sq. 1, Voronezh, Russia, 394018
| | - Sergei A Oleinik
- Department of Medical Biochemistry, Molecular and Cell Biology, Voronezh State University, Universitetskaya Sq. 1, Voronezh, Russia, 394018
| | - Svetlana M Medvedeva
- Department of Organic Chemistry, Voronezh State University, Universitetskaya Sq. 1, Voronezh, Russia, 394018
| | - Khidmet S Shikhaliev
- Department of Organic Chemistry, Voronezh State University, Universitetskaya Sq. 1, Voronezh, Russia, 394018
| |
Collapse
|
10
|
Kim Y, Cho M, Jang CH, Lee JS, Kim JS, Oh J, Lim J. Oral Administration of Euonymus alatus Leaf Extract Ameliorates Alzheimer's Disease Phenotypes in 5xFAD Transgenic Mice. Foods 2024; 13:682. [PMID: 38472795 DOI: 10.3390/foods13050682] [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: 12/26/2023] [Revised: 02/17/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease and is frequently characterized by progressive and irreversible impairment of cognitive functions. However, its etiology remains poorly understood, limiting therapeutic interventions. Our previous study showed that the ethanol extract of Euonymus alatus leaves (EA) positively affected scopolamine-induced hypomnesia in the normal mouse model by promoting nuclear factor E2-related factor 2 (Nrf2) activation. Herein, we examined whether EA administration could ameliorate major AD phenotypes that are manifested in 5xFAD transgenic mice. Two-month-old mice were orally administered with EA at a dose of 50, 100, or 150 mg/kg body weight/day thrice a week for 14 weeks. We observed that EA administration improved behavioral deficits as assessed by the passive avoidance, Morris water maze, and Y-maze tasks; decreased the plasma levels of pro-inflammatory cytokines, including TNFα and IL-1β; decreased the protein expression levels of inflammatory mediators in the hippocampus; and attenuated histological damage and amyloid beta plaques in the hippocampal region of 5xFAD mouse brain. Interestingly, our data demonstrated that the effectiveness was partially attributed to quercetin, which was noted to be a component of EA. Hence, these findings suggest that a long-term administration of EA could alleviate AD symptoms and delay its progression.
Collapse
Affiliation(s)
- Yoonsu Kim
- Department of Integrative Biology, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Minjung Cho
- Department of Integrative Biology, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Chan Ho Jang
- Institute of Agricultural Science and Technology, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jeong Soon Lee
- Forest Environment Research Institute of Gyeongsangbuk-do, Gyeongju 38174, Republic of Korea
| | - Jong-Sang Kim
- Department of Integrative Biology, Kyungpook National University, Daegu 41566, Republic of Korea
- Institute of Agricultural Science and Technology, Kyungpook National University, Daegu 41566, Republic of Korea
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jisun Oh
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Jinkyu Lim
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea
| |
Collapse
|
11
|
Qin Z, Xie L, Li W, Wang C, Li Y. New Insights into Mechanisms Traditional Chinese Medicine for Allergic Rhinitis by Regulating Inflammatory and Oxidative Stress Pathways. J Asthma Allergy 2024; 17:97-112. [PMID: 38405022 PMCID: PMC10888064 DOI: 10.2147/jaa.s444923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/23/2024] [Indexed: 02/27/2024] Open
Abstract
Allergy rhinitis (AR) is becoming more common and has serious medical and societal consequences. Sneezing, paroxysmal nasal blockage, nasal itching, mucosal edema, coughing, and rhinorrhea are symptoms of this type I allergic immunological illness. Immunoglobulin E-mediated inflammation is the cause of it. Because AR is prone to recurrent attacks, extended medication therapy may impair its effectiveness. In addition to negatively affecting the patients' physical health, this can also negatively impact their mental health. During AR development, there are inflammatory and oxidative stress responses that are linked to problems in a number of signal transduction pathways. By using the terms "allergic rhinitis", "traditional Chinese medicine", "inflammation", and "oxidative stress", we screened for pertinent research published over the previous five years in databases like PubMed. We saw that NF-KB, TLR, IL-33/ST2, PI3K/AKT, MAPK, and Nrf2 are some of the most important inflammatory and oxidative stress pathways in AR. Studies have revealed that antioxidant and anti-inflammatory therapy reduced the risk of AR and was therapeutic; however, the impact of the therapy varies widely. The Chinese medical system places a high value on traditional Chinese medicine (TCM), which has been there for virtually all of China's 5000-year history. By influencing signaling pathways related to inflammation and oxidative stress, Chinese herbal medicine and its constituent compounds have been shown to prevent allergic rhinitis. This review will focus on this evidence and provide references for clinical treatment and scientific research applications.
Collapse
Affiliation(s)
- Zhu Qin
- Department of Otolaryngology, Graduate School of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| | - Liangzhen Xie
- Department of Otolaryngology, Graduate School of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
- Department of Otolaryngology, First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| | - Wentao Li
- Department of Otolaryngology, First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| | - Chao Wang
- Department of Otolaryngology, Graduate School of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| | - Yan Li
- Department of Otolaryngology, First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| |
Collapse
|
12
|
Tao L, Yu W, Liu Z, Zhao D, Lin S, Szalóki D, Kicsák M, Kurtán T, Zhang H. JE-133 Suppresses LPS-Induced Neuroinflammation Associated with the Regulation of JAK/STAT and Nrf2 Signaling Pathways. ACS Chem Neurosci 2024; 15:258-267. [PMID: 38181172 DOI: 10.1021/acschemneuro.3c00454] [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] [Indexed: 01/07/2024] Open
Abstract
Neuroinflammation plays an important role in the pathogenesis of neurodegenerative diseases, and interrupting the microglial-mediated neuroinflammation has been suggested as a promising strategy to delay or prevent the progression of neurodegeneration. In this study, we investigated the effects of JE-133, an optically active isochroman-2H-chromene conjugate containing a 1,3-disubstituted isochroman unit, on lipopolysaccharide (LPS)-induced microglial neuroinflammation and underlying mechanisms both in vitro and in vivo. First, JE-133 treatment decreased LPS-induced overproduction of interleukin-1 beta (IL-1β), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), nitrite, and nitric oxide synthase (iNOS) in BV2 microglial cells. Further study revealed that JE-133 downregulated the phosphorylation level of JAK/STAT and upregulated the protein level of Nrf2/HO-1 in LPS-stimulated BV2 microglial cells and verified that JE-133 directly bound to Keap1 by a pull-down assay. Next, JE-133 administration also inhibited neuroinflammation in vivo, as indicated by a reduced CD11b protein level and an overexpressed mRNA level of the pro-inflammatory cytokine TNF-α in the hippocampus of LPS-injected mice. Moreover, the regulative effects of JE-133 on the JAK/STAT and Nrf2/HO-1 pathways were also verified in the hippocampus of LPS-injected mice. Taken together, our study for the first time reports that JE-133 exhibits inhibitory effects against LPS-stimulated neuroinflammation both in vitro and in vivo, which might be associated with the simultaneous regulation of the JAK/STAT and Nrf2 pathways. Our findings may provide important clues for the discovery of effective drug leads/candidates against neuroinflammation-associated neurodegeneration.
Collapse
Affiliation(s)
- Lingxue Tao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Lingang Laboratory, Shanghai 200031, China
| | - Weichen Yu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ziyi Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Nanchang University, Jiangxi 330031, China
| | - Danfeng Zhao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Sijin Lin
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dóra Szalóki
- Department of Organic Chemistry, University of Debrecen, Debrecen, P.O. Box 400, Debrecen H-4002, Hungary
| | - Máté Kicsák
- Department of Organic Chemistry, University of Debrecen, Debrecen, P.O. Box 400, Debrecen H-4002, Hungary
| | - Tibor Kurtán
- Department of Organic Chemistry, University of Debrecen, Debrecen, P.O. Box 400, Debrecen H-4002, Hungary
| | - Haiyan Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
13
|
Wang J, Cao Y, Lu Y, Zhu H, Zhang J, Che J, Zhuang R, Shao J. Recent progress and applications of small molecule inhibitors of Keap1-Nrf2 axis for neurodegenerative diseases. Eur J Med Chem 2024; 264:115998. [PMID: 38043492 DOI: 10.1016/j.ejmech.2023.115998] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/18/2023] [Accepted: 11/23/2023] [Indexed: 12/05/2023]
Abstract
The Kelch-like ECH-associated protein 1 (Keap1)-nuclear factor erythroid 2-related factor 2 (Nrf2) pathway serves as a crucial regulator against oxidative stress (OS) damage in various cells and organs. It has garnered significant attention as a potential therapeutic target for neurodegenerative diseases (NDD). Although progress has been achieved in strategies to regulate the Keap1-Nrf2 pathway, the availability of Nrf2 activators applicable to NDD is currently limited. Currently, the FDA has approved the Nrf2 activators dimethyl fumarate (DMF) and Omaveloxolone (Omav) as novel first-line oral drugs for the treatment of patients with relapsing forms of multiple sclerosis and Friedreich's ataxia. A promising alternative approach involves the direct inhibition of Keap1-Nrf2 protein-protein interactions (PPI), which offers numerous advantages over the use of electrophilic Nrf2 activators, primarily in avoiding off-target effects. This review examines the compelling evidence supporting the beneficial role of Nrf2 in NDD and explores the potential of Keap1 inhibitors and Keap1-Nrf2 PPI inhibitors as therapeutic agents, with the aim to provide further insights into the development of inhibitors targeting this pathway for the treatment of NDD.
Collapse
Affiliation(s)
- Jing Wang
- School of Medicine, Hangzhou City University, Hangzhou, 310015, Zhejiang Province, China; Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, China; College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang Province, China
| | - Yu Cao
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, 310023, China
| | - Yang Lu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang Province, China
| | - Huajian Zhu
- School of Medicine, Hangzhou City University, Hangzhou, 310015, Zhejiang Province, China; Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, China; College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang Province, China
| | - Jiankang Zhang
- School of Medicine, Hangzhou City University, Hangzhou, 310015, Zhejiang Province, China; Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, China; College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang Province, China
| | - Jinxin Che
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang Province, China.
| | - Rangxiao Zhuang
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, 310023, China.
| | - Jiaan Shao
- School of Medicine, Hangzhou City University, Hangzhou, 310015, Zhejiang Province, China; Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, China; College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang Province, China.
| |
Collapse
|
14
|
Costantini E, Carrarini C, Calisi D, De Rosa M, Simone M, Di Crosta A, Palumbo R, Cipollone A, Aielli L, De Laurentis M, Colarusso L, Pilotto A, Padovani A, Konstantinidou F, Gatta V, Stuppia L, Cipollone F, Di Nicola M, Reale M, Bonanni L. Search in the Periphery for Potential Inflammatory Biomarkers of Dementia with Lewy Bodies and Alzheimer's Disease. J Alzheimers Dis 2024; 99:1147-1158. [PMID: 38759010 PMCID: PMC11191525 DOI: 10.3233/jad-231471] [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] [Accepted: 04/04/2024] [Indexed: 05/19/2024]
Abstract
Background Neuroinflammation, with altered peripheral proinflammatory cytokine production, plays a major role in the pathogenesis of neurodegenerative diseases, such as Alzheimer's disease (AD), while the role of inflammation in dementia with Lewy bodies (DLB) is less known and the results of different studies are often in disagreement. Objective The present study aimed to investigate the levels of TNFα and IL-6 in serum and supernatants, and the related DNA methylation in patients affected by DLB and AD compared to healthy controls (HCs), to clarify the role of epigenetic mechanisms of DNA promoter methylation on of pro-inflammatory cytokines overproduction. Methods Twenty-one patients with DLB and fourteen with AD were frequency-matched for age and sex with eleven HCs. Clinical evaluation, TNFα and IL-6 gene methylation status, cytokine gene expression levels and production in serum and peripheral blood mononuclear cell (PBMC) supernatants were performed. Results In AD and DLB patients, higher serum levels of IL-6 and TNFα were detected than in HCs. Differences in LPS-stimulated versus spontaneous PBMCs were observed between DLB, AD, and HC in the levels of TNFα (p = 0.027) and IL-6 (p < 0.001). Higher levels were also revealed for sIL-6R in DLB (p < 0.001) and AD (p < 0.001) in comparison with HC.DNA hypomethylation in IL-6 and TNFα CpG promoter sites was detected for DLB and AD patients compared to the corresponding site in HCs. Conclusions Our preliminary study documented increased levels of IL-6 and TNFα in DLB and AD patients to HCs. This overproduction can be due to epigenetic mechanisms regarding the hypomethylation of DNA promoters.
Collapse
Affiliation(s)
- Erica Costantini
- Department of Medicine and Aging Sciences, University “G. d’Annunzio”, Chieti, Italy
| | - Claudia Carrarini
- Department of Neuroscience, Catholic University of Sacred Heart, Rome, Italy
- IRCCS San Raffaele, Rome, Italy
| | - Dario Calisi
- Department of Neurosciences, Imaging and Clinical Sciences, University “G. d’Annunzio”, Chieti, Italy
| | - Matteo De Rosa
- Department of Neurosciences, Imaging and Clinical Sciences, University “G. d’Annunzio”, Chieti, Italy
| | - Marianna Simone
- Clinics of Neurology SS. Annunziata Hospital of Chieti, Chieti, Italy
| | - Adolfo Di Crosta
- Department of Psychological Health and Territorial Sciences, University “G. d’Annunzio”, Chieti, Italy
| | - Rocco Palumbo
- Department of Psychological Health and Territorial Sciences, University “G. d’Annunzio”, Chieti, Italy
| | - Alessia Cipollone
- Department of Medicine and Aging Sciences, University “G. d’Annunzio”, Chieti, Italy
| | - Lisa Aielli
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio”, Chieti, Italy
| | | | | | - Andrea Pilotto
- Department of Clinical and Experimental Sciences, Neurology Unit, University of Brescia, Brescia, Italy
- Parkinson’s Disease Rehabilitation Centre, FERB ONLUS-S, Isidoro Hospital, Trescore Balneario, Italy
| | - Alessandro Padovani
- Department of Clinical and Experimental Sciences, Neurology Unit, University of Brescia, Brescia, Italy
| | - Fani Konstantinidou
- Department of Psychological Health and Territorial Sciences, University “G. d’Annunzio”, Chieti, Italy
| | - Valentina Gatta
- Department of Psychological Health and Territorial Sciences, University “G. d’Annunzio”, Chieti, Italy
| | - Liborio Stuppia
- Department of Psychological Health and Territorial Sciences, University “G. d’Annunzio”, Chieti, Italy
| | - Francesco Cipollone
- Department of Medicine and Aging Sciences, University “G. d’Annunzio”, Chieti, Italy
| | - Marta Di Nicola
- Department of Medical and Oral Sciences and Biotechnologies, University “G. d’Annunzio”, Chieti, Italy
| | - Marcella Reale
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio”, Chieti, Italy
| | - Laura Bonanni
- Department of Medicine and Aging Sciences, University “G. d’Annunzio”, Chieti, Italy
| |
Collapse
|
15
|
Nies YH, Yahaya MF, Lim WL, Teoh SL. Microarray-based Analysis of Differential Gene Expression Profile in Rotenone-induced Parkinson's Disease Zebrafish Model. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:761-772. [PMID: 37291778 DOI: 10.2174/1871527322666230608122552] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 05/04/2023] [Accepted: 05/04/2023] [Indexed: 06/10/2023]
Abstract
BACKGROUND & OBJECTIVES Despite much clinical and laboratory research that has been performed to explore the mechanisms of Parkinson's disease (PD), its pathogenesis remains elusive to date. Therefore, this study aimed to identify possible regulators of neurodegeneration by performing microarray analysis of the zebrafish PD model's brain following rotenone exposure. METHODS A total of 36 adult zebrafish were divided into two groups: control (n = 17) and rotenonetreated (n = 19). Fish were treated with rotenone water (5 μg/L water) for 28 days and subjected to locomotor behavior analysis. Total RNA was extracted from the brain tissue after rotenone treatment. The cDNA synthesized was subjected to microarray analysis and subsequently validated by qPCR. RESULTS Administration of rotenone has significantly reduced locomotor activity in zebrafish (p < 0.05), dysregulated dopamine-related gene expression (dat, th1, and th2, p < 0.001), and reduced dopamine level in the brain (p < 0.001). In the rotenone-treated group, genes involved in cytotoxic T lymphocytes (gzm3, cd8a, p < 0.001) and T cell receptor signaling (themis, lck, p < 0.001) were upregulated significantly. Additionally, gene expression involved in microgliosis regulation (tyrobp, p < 0.001), cellular response to IL-1 (ccl34b4, il2rb, p < 0.05), and regulation of apoptotic process (dedd1, p < 0.001) were also upregulated significantly. CONCLUSION The mechanisms of T cell receptor signaling, microgliosis regulation, cellular response to IL-1, and apoptotic signaling pathways have potentially contributed to PD development in rotenonetreated zebrafish.
Collapse
Affiliation(s)
- Yong Hui Nies
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Mohamad Fairuz Yahaya
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Wei Ling Lim
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Selangor, Malaysia
| | - Seong Lin Teoh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
16
|
Wu K, Wang X, Gong L, Zhai X, Wang K, Qiu X, Zhang H, Tang Z, Jiang H, Wang X. Screening of H 2S donors with a red emission mitochondria-targetable fluorescent probe: Toward discovering a new therapeutic strategy for Parkinson's disease. Biosens Bioelectron 2023; 237:115521. [PMID: 37429146 DOI: 10.1016/j.bios.2023.115521] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/28/2023] [Accepted: 07/05/2023] [Indexed: 07/12/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder caused by various factors such as neuroinflammation, oxidative stress, mitochondrial dysfunction, and neuronal apoptosis. Recent studies have shown that H2S supplementation reverses neuronal loss and mitigates motor deficits in PD patients through anti-inflammatory, antioxidant, improved mitochondrial function and proautophagic. Therefore, the discovery and use of H2S donors may be an exciting and intriguing strategy for the treatment of PD. Herein, we report a red emission mitochondria-targetable fluorescent probe, Rho-H2S, which can specifically and sensitively detect H2S with a limit of detection of 62.5 nM. Bioimaging experiments have shown that the probe has excellent mitochondrial targeting and good imaging capabilities for the detection of exogenous and endogenous H2S in cells. More importantly, based on the Rho-H2S probe, we first confirmed the sulforaphane (SFN) among 15 glucosinolate and isothiocyanate compounds from cruciferous vegetables with an outstanding ability to release H2S and we further proved that SFN could alleviate the symptoms of PD in vivo. All results demonstrate that Rho-H2S could be an effective tool for screening H2S donors and can contribute to the development of new therapeutic strategies for PD.
Collapse
Affiliation(s)
- Ke Wu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China
| | - Xumei Wang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China
| | - Lili Gong
- Experimental Center, Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China
| | - Xinyuan Zhai
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China
| | - Kai Wang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China
| | - Xiao Qiu
- Experimental Center, Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China
| | - Hao Zhang
- Experimental Center, Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China
| | - Zhixin Tang
- Experimental Center, Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China.
| | - Haiqiang Jiang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China.
| | - Xiaoming Wang
- Experimental Center, Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China.
| |
Collapse
|
17
|
Darwish SF, Elbadry AMM, Elbokhomy AS, Salama GA, Salama RM. The dual face of microglia (M1/M2) as a potential target in the protective effect of nutraceuticals against neurodegenerative diseases. FRONTIERS IN AGING 2023; 4:1231706. [PMID: 37744008 PMCID: PMC10513083 DOI: 10.3389/fragi.2023.1231706] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/23/2023] [Indexed: 09/26/2023]
Abstract
The pathophysiology of different neurodegenerative illnesses is significantly influenced by the polarization regulation of microglia and macrophages. Traditional classifications of macrophage phenotypes include the pro-inflammatory M1 and the anti-inflammatory M2 phenotypes. Numerous studies demonstrated dynamic non-coding RNA modifications, which are catalyzed by microglia-induced neuroinflammation. Different nutraceuticals focus on the polarization of M1/M2 phenotypes of microglia and macrophages, offering a potent defense against neurodegeneration. Caeminaxin A, curcumin, aromatic-turmerone, myricetin, aurantiamide, 3,6'-disinapoylsucrose, and resveratrol reduced M1 microglial inflammatory markers while increased M2 indicators in Alzheimer's disease. Amyloid beta-induced microglial M1 activation was suppressed by andrographolide, sulforaphane, triptolide, xanthoceraside, piperlongumine, and novel plant extracts which also prevented microglia-mediated necroptosis and apoptosis. Asarone, galangin, baicalein, and a-mangostin reduced oxidative stress and pro-inflammatory cytokines, such as interleukin (IL)-1, IL-6, and tumor necrosis factor-alpha in M1-activated microglia in Parkinson's disease. Additionally, myrcene, icariin, and tenuigenin prevented the nod-like receptor family pyrin domain-containing 3 inflammasome and microglial neurotoxicity, while a-cyperone, citronellol, nobiletin, and taurine prevented NADPH oxidase 2 and nuclear factor kappa B activation. Furthermore, other nutraceuticals like plantamajoside, swertiamarin, urolithin A, kurarinone, Daphne genkwa flower, and Boswellia serrata extracts showed promising neuroprotection in treating Parkinson's disease. In Huntington's disease, elderberry, curcumin, iresine celosia, Schisandra chinensis, gintonin, and pomiferin showed promising results against microglial activation and improved patient symptoms. Meanwhile, linolenic acid, resveratrol, Huperzia serrata, icariin, and baicalein protected against activated macrophages and microglia in experimental autoimmune encephalomyelitis and multiple sclerosis. Additionally, emodin, esters of gallic and rosmarinic acids, Agathisflavone, and sinomenine offered promising multiple sclerosis treatments. This review highlights the therapeutic potential of using nutraceuticals to treat neurodegenerative diseases involving microglial-related pathways.
Collapse
Affiliation(s)
- Samar F. Darwish
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Cairo, Egypt
| | - Abdullah M. M. Elbadry
- Faculty of Pharmacy, Badr University in Cairo (BUC), Cairo, Egypt
- Nanotechnology Research Center (NTRC), The British University in Egypt (BUE), El-Sherouk City, Egypt
| | | | - Ghidaa A. Salama
- Faculty of Pharmacy, Badr University in Cairo (BUC), Cairo, Egypt
| | - Rania M. Salama
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Misr International University, Cairo, Egypt
| |
Collapse
|
18
|
Lanza M, Cucinotta L, Casili G, Filippone A, Basilotta R, Capra AP, Campolo M, Paterniti I, Cuzzocrea S, Esposito E. The Transcription Factor Nrf2 Mediates the Effects of Antrodia camphorata Extract on Neuropathological Changes in a Mouse Model of Parkinson's Disease. Int J Mol Sci 2023; 24:ijms24119250. [PMID: 37298200 DOI: 10.3390/ijms24119250] [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: 04/20/2023] [Revised: 05/11/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Parkinson's disease (PD) is a disorder that is characterized by progressive and selective neuronal injury and cell death. Recent studies have provided accumulating evidence for a significant role of the immune system and neuroinflammation in PD pathogenesis. On this basis, many scientific articles have highlighted the anti-inflammatory and neuroprotective properties of Antrodia camphorata (AC), an edible fungus containing various bioactive compounds. This study aimed to evaluate the inhibitory effect of AC administration on neuroinflammation and oxidative stress in a murine model of MPTP-induced dopaminergic degeneration. AC (10, 30, 100 mg/kg) was administered daily by oral gavage starting 24 h after the first administration of MPTP, and mice were sacrificed 7 days after MPTP induction. In this study, treatment with AC significantly reduced the alteration of PD hallmarks, increasing tyrosine hydroxylase expression and reducing the number of alpha-synuclein-positive neurons. In addition, AC treatment restored the myelination process of neurons associated with PD and attenuated the neuroinflammatory state. Furthermore, our study demonstrated that AC was able to reduce the oxidative stress induced by MPTP injection. In conclusion, this study highlighted that AC could be a potential therapeutic agent for the treatment of neurodegenerative disorders such as PD.
Collapse
Affiliation(s)
- Marika Lanza
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 7 Viale Ferdinando Stagno D'Alcontres, 31, 98166 Messina, Italy
| | - Laura Cucinotta
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 7 Viale Ferdinando Stagno D'Alcontres, 31, 98166 Messina, Italy
| | - Giovanna Casili
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 7 Viale Ferdinando Stagno D'Alcontres, 31, 98166 Messina, Italy
| | - Alessia Filippone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 7 Viale Ferdinando Stagno D'Alcontres, 31, 98166 Messina, Italy
| | - Rossella Basilotta
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 7 Viale Ferdinando Stagno D'Alcontres, 31, 98166 Messina, Italy
| | - Anna Paola Capra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 7 Viale Ferdinando Stagno D'Alcontres, 31, 98166 Messina, Italy
| | - Michela Campolo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 7 Viale Ferdinando Stagno D'Alcontres, 31, 98166 Messina, Italy
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 7 Viale Ferdinando Stagno D'Alcontres, 31, 98166 Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 7 Viale Ferdinando Stagno D'Alcontres, 31, 98166 Messina, Italy
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 7 Viale Ferdinando Stagno D'Alcontres, 31, 98166 Messina, Italy
| |
Collapse
|
19
|
Ravenhill SM, Evans AH, Crewther SG. Escalating Bi-Directional Feedback Loops between Proinflammatory Microglia and Mitochondria in Ageing and Post-Diagnosis of Parkinson's Disease. Antioxidants (Basel) 2023; 12:antiox12051117. [PMID: 37237983 DOI: 10.3390/antiox12051117] [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: 03/22/2023] [Revised: 05/12/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Parkinson's disease (PD) is a chronic and progressive age-related neurodegenerative disease affecting up to 3% of the global population over 65 years of age. Currently, the underlying physiological aetiology of PD is unknown. However, the diagnosed disorder shares many common non-motor symptoms associated with ageing-related neurodegenerative disease progression, such as neuroinflammation, microglial activation, neuronal mitochondrial impairment, and chronic autonomic nervous system dysfunction. Clinical PD has been linked to many interrelated biological and molecular processes, such as escalating proinflammatory immune responses, mitochondrial impairment, lower adenosine triphosphate (ATP) availability, increasing release of neurotoxic reactive oxygen species (ROS), impaired blood brain barrier integrity, chronic activation of microglia, and damage to dopaminergic neurons consistently associated with motor and cognitive decline. Prodromal PD has also been associated with orthostatic hypotension and many other age-related impairments, such as sleep disruption, impaired gut microbiome, and constipation. Thus, this review aimed to present evidence linking mitochondrial dysfunction, including elevated oxidative stress, ROS, and impaired cellular energy production, with the overactivation and escalation of a microglial-mediated proinflammatory immune response as naturally occurring and damaging interlinked bidirectional and self-perpetuating cycles that share common pathological processes in ageing and PD. We propose that both chronic inflammation, microglial activation, and neuronal mitochondrial impairment should be considered as concurrently influencing each other along a continuum rather than as separate and isolated linear metabolic events that affect specific aspects of neural processing and brain function.
Collapse
Affiliation(s)
| | - Andrew Howard Evans
- Department of Medicine, The Walter and Eliza Hall Institute of Medical Research, Melbourne 3052, Australia
- Epworth Hospital, Richmond 3121, Australia
- Department of Neurology, Royal Melbourne Hospital, Melbourne 3050, Australia
| | | |
Collapse
|
20
|
Zhang J, Li L, Liu Q, Zhao Z, Su D, Xiao C, Jin T, Chen L, Xu C, You Z, Zhou T. Gastrodin programs an Arg-1 + microglial phenotype in hippocampus to ameliorate depression- and anxiety-like behaviors via the Nrf2 pathway in mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 113:154725. [PMID: 36867963 DOI: 10.1016/j.phymed.2023.154725] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/05/2023] [Accepted: 02/20/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Regulating the microglial phenotype is an attractive strategy for treating diseases of the central nervous system such as depression and anxiety. Gastrodin can quickly cross the blood-brain barrier and mitigate microglia-mediated inflammation, which widely used to treat a variety of central nervous system diseases associated with microglial dysfunction. However, the molecular mechanism by which gastrodin regulates the functional phenotype of microglia remains unclear. PURPOSE Since the transcription factor "nuclear factor erythroid 2-related factor 2″ (Nrf2) is associated with the anti-inflammatory effects of gastrodin, we hypothesized that gastrodin induces Nrf2 expression in microglia and thereby programs an anti-inflammatory phenotype. STUDY DESIGN Male C57BL/6 mice, treated or not with gastrodin, were given lipopolysaccharide (LPS) at 0.25 mg/kg/d for 10 days to induce chronic neuroinflammation. The effects of gastrodin on microglial phenotypes, neuroinflammation and depression- and anxiety-like behaviors were evaluated. In another experiment, animals were treated with Nrf2 inhibitor ML385 throughout the 13-day gastrodin intervention period. METHODS The effects of gastrodin on depression- and anxiety-like behaviors were evaluated through the sucrose preference test, forced swimming test, open field test and elevated plus-maze test; as well as its effects on morphology and molecular and functional phenotypes of hippocampal microglia through immunohistochemistry, real-time PCR and enzyme-linked immunosorbent assays. RESULTS Chronic exposure to LPS caused hippocampal microglia to secrete inflammatory cytokines, their somata to enlarge, and their dendrites to lose branches. These changes were associated with depression- and anxiety-like behaviors. Gastrodin blocked these LPS-induced alterations and promoted an Arg-1+ microglial phenotype that protected neurons from injury. The effects of gastrodin were associated with Nrf2 activation, whereas blockade of Nrf2 antagonized gastrodin. CONCLUSION These results suggest that gastrodin acts via Nrf2 to promote an Arg-1+ microglial phenotype, which buffers the harmful effects of LPS-induced neuroinflammation. Gastrodin may be a promising drug against central nervous system diseases that involve microglial dysfunction.
Collapse
Affiliation(s)
- Jinqiang Zhang
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Liangyuan Li
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Qin Liu
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Zhihuang Zhao
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Dapeng Su
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Chenghong Xiao
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Ting Jin
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Li Chen
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Chunyun Xu
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Zili You
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China.
| | - Tao Zhou
- Resource Institute for Chinese & Ethnic Materia Medica, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| |
Collapse
|
21
|
Tryphena KP, Singh G, Jain N, Famta P, Srivastava S, Singh SB, Khatri DK. Integration of miRNA's Theranostic Potential with Nanotechnology: Promises and Challenges for Parkinson's Disease Therapeutics. Mech Ageing Dev 2023; 211:111800. [PMID: 36958539 DOI: 10.1016/j.mad.2023.111800] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/03/2023] [Accepted: 03/17/2023] [Indexed: 03/25/2023]
Abstract
Despite the wide research going on in Parkinson's disease (PD), the burden of PD still remains high and continues to increase. The current drugs available for the treatment of PD are only aimed at symptomatic control. Hence, research is mainly focused on identifying the novel therapeutic targets that can be effectively targeted in order to slow down or culminate the disease progression. Recently the role of microRNAs (miRNAs) in the regulation of various pathological mechanisms of PD has been thoroughly explored and many of them were found to be dysregulated in the biological samples of PD patients. These miRNAs can be used as diagnostic markers and novel therapeutic options to manage PD. The delivery of miRNAs to the target site in brain is a challenging job owing to their nature of degradability by endonucleases as well as poor blood brain barrier (BBB) permeability. Nanoparticles appear to be the best solution to effectively encase the miRNA in their core as well as cross the BBB to deliver them into brain. Functionalisation of these nanoparticles further enhances the site-specific delivery.
Collapse
Affiliation(s)
- Kamatham Pushpa Tryphena
- Molecular and cellular neuroscience lab, Department of pharmacology and toxicology, National Institute of Pharmaceutical Education and Research (NIPER)- Hyderabad
| | - Gurpreet Singh
- Molecular and cellular neuroscience lab, Department of pharmacology and toxicology, National Institute of Pharmaceutical Education and Research (NIPER)- Hyderabad
| | - Naitik Jain
- Department of pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)- Hyderabad
| | - Paras Famta
- Department of pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)- Hyderabad
| | - Saurabh Srivastava
- Department of pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)- Hyderabad.
| | - Shashi Bala Singh
- Molecular and cellular neuroscience lab, Department of pharmacology and toxicology, National Institute of Pharmaceutical Education and Research (NIPER)- Hyderabad
| | - Dharmendra Kumar Khatri
- Molecular and cellular neuroscience lab, Department of pharmacology and toxicology, National Institute of Pharmaceutical Education and Research (NIPER)- Hyderabad.
| |
Collapse
|
22
|
Muhammad F, Liu Y, Wang N, Zhao L, Zhou Y, Yang H, Li H. Rose essential oil diminishes dopaminergic neuron degenerations and reduces α-synuclein aggregation in Caenorhabditis elegans models of Parkinson's disease. Phytother Res 2023. [PMID: 36920348 DOI: 10.1002/ptr.7783] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 10/10/2022] [Accepted: 01/29/2023] [Indexed: 03/16/2023]
Abstract
Parkinson's disease (P.D.) is the second most progressive neurodegenerative disorder in the elderly. Degeneration of dopaminergic (DA) neurons and α-synuclein (α-Syn) accumulated toxicity is the major contributor to this disease. At present, the disease has no effective treatment. Many recent studies focus on identifying novel therapeutics that provide benefits to stop the disease progression in P.D. patients. Screening novel and effective drugs in P.D. animal models is time- and cost-consuming. Rose Essential Oil (REO) extracted from Rosa Rugosa species (R. Setate × R. Rugosa). REO contains Citronellol, Geraniol, and Octadiene that possess anti-Aβ, anti-oxidative, and anti-depression-like properties, but no reports have defined the REO effect on P.D. yet. The present study examines the REO neuroprotective potential in transgenic Caenorhabditis elegans P.D. models. We observed that REO reduced α-Syn aggregations and diminished DA neuron degenerations induced by 6-OHDA, reduced food-sensing behavioural disabilities, and prolonged the lifespan of the nematode. Moreover, REO augmented the chymotrypsin-like proteasome and SOD-3 activities. Further, we observed the anti-oxidative role of REO by reducing internal cells ROS. Together, these findings supported REO as an anti-PD drug and may exert its effects by lowering oxidative stress via the anti-oxidative pathway.
Collapse
Affiliation(s)
- Fahim Muhammad
- College of Life Sciences, Lanzhou University, Lanzhou, China
| | - Yan Liu
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Ningbo Wang
- College of Life Sciences, Lanzhou University, Lanzhou, China.,School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Longhe Zhao
- College of Life Sciences, Lanzhou University, Lanzhou, China
| | - Yangtao Zhou
- Department of Neurology, Clinical Center for Parkinson's Disease, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Hui Yang
- Institute of Biology Gansu Academy of Sciences, Lanzhou, China
| | - Hongyu Li
- College of Life Sciences, Lanzhou University, Lanzhou, China.,School of Pharmacy, Lanzhou University, Lanzhou, China
| |
Collapse
|
23
|
Huang B, Hu G, Zong X, Yang S, He D, Gao X, Liu D. α-Cyperone protects dopaminergic neurons and inhibits neuroinflammation in LPS-induced Parkinson's disease rat model via activating Nrf2/HO-1 and suppressing NF-κB signaling pathway. Int Immunopharmacol 2023; 115:109698. [PMID: 36634417 DOI: 10.1016/j.intimp.2023.109698] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 12/21/2022] [Accepted: 01/03/2023] [Indexed: 01/12/2023]
Abstract
Our previous study showed that α-Cyperone inhibited the inflammatory response triggered by activated microglia and protected dopaminergic neuron in in vitro cell model of Parkinson's disease (PD). It is unclear the effect of α-Cyperone in animal models of PD. In this study, our results indicated that α-Cyperone ameliorated motor dysfunction, protected dopaminergic neurons, and inhibited the reduction of dopamine and its metabolites in lipopolysaccharide (LPS)-induced PD rat model. Moreover, α-Cyperone suppressed the activation of microglia and the expression of neuroinflammatory factor (TNF-α, IL-6, IL-1β, iNOS, COX-2 and ROS). Furthermore, the molecular mechanism research revealed that α-Cyperone inhibited neuroinflammation and oxidative stress to exert protective effect in microglia by activating Nrf2/HO-1 and suppressing NF-κB signaling pathway. Moreover, α-Cyperone upregulated the expression of antioxidant enzymes (GCLC, GCLM and NQO1) in microglia. In conclusion, our study demonstrates α-Cyperone alleviates dopaminergic neurodegeneration by inhibiting neuroinflammation and oxidative stress in LPS-induced PD rat model via activating Nrf2/HO-1 and suppressing NF-κB signaling pathway.
Collapse
Affiliation(s)
- Bingxu Huang
- Department of Laboratory Animals, College of Animal Science, Jilin University, Changchun, China
| | - Guiqiu Hu
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiaofeng Zong
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Shuo Yang
- Department of Laboratory Animals, College of Animal Science, Jilin University, Changchun, China
| | - Dewei He
- Department of Laboratory Animals, College of Animal Science, Jilin University, Changchun, China
| | - Xiyu Gao
- Department of Laboratory Animals, College of Animal Science, Jilin University, Changchun, China
| | - Dianfeng Liu
- Department of Laboratory Animals, College of Animal Science, Jilin University, Changchun, China.
| |
Collapse
|
24
|
Narrative Review Concerning the Clinical Spectrum of Ophthalmological Impairments in Parkinson's Disease. Neurol Int 2023; 15:140-161. [PMID: 36810467 PMCID: PMC9944508 DOI: 10.3390/neurolint15010012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 01/30/2023] Open
Abstract
Ophthalmic non-motor impairments are common in Parkinson's disease patients, from the onset of the neurodegenerative disease and even prior to the development of motor symptoms. This is a very crucial component of the potential for early detection of this disease, even in its earliest stages. Since the ophthalmological disease is extensive and impacts all extraocular and intraocular components of the optical analyzer, a competent assessment of it would be beneficial for the patients. Because the retina is an extension of the nervous system and has the same embryonic genesis as the central nervous system, it is helpful to investigate the retinal changes in Parkinson's disease in order to hypothesize insights that may also be applicable to the brain. As a consequence, the detection of these symptoms and signs may improve the medical evaluation of PD and predict the illness' prognosis. Another valuable aspect of this pathology is the fact that the ophthalmological damage contributes significantly to the decrease in the quality of life of patients with Parkinson's disease. We provide an overview of the most significant ophthalmologic impairments associated with Parkinson's disease. These results certainly constitute a large number of the prevalent visual impairments experienced by PD patients.
Collapse
|
25
|
Anwar MM, Özkan E, Shomalizadeh N, Sapancı S, Özler C, Kesibi J, Gürsoy-Özdemir Y. Assessing the role of primary healthy microglia and gap junction blocker in hindering Alzheimer's disease neuroinflammatory type: Early approaches for therapeutic intervention. Front Neurosci 2023; 16:1041461. [PMID: 36704003 PMCID: PMC9871931 DOI: 10.3389/fnins.2022.1041461] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
Alzheimer's disease (AD) is a predominantly heterogeneous disease with a highly complex pathobiology. The presence of amyloid-beta (Aβ) depositions and the accumulation of hyperphosphorylated tau protein remain the characteristic hallmarks of AD. These hallmarks can be detected throughout the brain and other regions, including cerebrospinal fluid (CSF) and the spinal cord. Microglia cells, the brain-resident macrophage type of the brain, are implicated in maintaining healthy brain homeostasis. The localized administration of primary healthy microglia (PHM) is suggested to play a role in mitigating AD hallmark depositions and associated cognitive dysfunction. Carbenoxolone (CBX) is the most common gap junction blocker. It cannot effectively cross the blood-brain barrier (BBB) under systemic administration. Therefore, localized administration of CBX may be a recommended intervention against AD by acting as an antioxidant and anti-inflammatory agent. This study aims to determine whether the localized intracerebroventricular (ICV) administration of PHM and CBX may act as an effective therapeutic intervention for AD neuroinflammatory type. In addition, this study also aims to reveal whether detecting AD hallmarks in the spinal cord and CSF can be considered functional and effective during AD early diagnosis. Male albino rats were divided into four groups: control (group 1), lipopolysaccharide (LPS)-induced AD neuroinflammatory type (group 2), ICV injection of LPS + isolated PHM (group 3), and ICV injection of LPS + CBX (group 4). Morris water maze (MWM) was conducted to evaluate spatial working memory. The brain and spinal cord were isolated from each rat with the collection of CSF. Our findings demonstrate that the localized administration of PHM and CBX can act as promising therapeutic approaches against AD. Additionally, Aβ and tau toxic aggregates were detected in the spinal cord and the CSF of the induced AD model concomitant with the brain tissues. Overall, it is suggested that the ICV administration of PHM and CBX can restore normal brain functions and alleviate AD hallmark depositions. Detecting these depositions in the spinal cord and CSF may be considered in AD early diagnosis. As such, conducting clinical research is recommended to reveal the benefits of related therapeutic approaches compared with preclinical findings.
Collapse
Affiliation(s)
- Mai M. Anwar
- Department of Biochemistry, National Organization for Drug Control and Research/Egyptian Drug Authority, Cairo, Egypt
| | - Esra Özkan
- Koç University Research Center for Translational Medicine, KUTTAM, Koç University, Istanbul, Turkey
| | - Narges Shomalizadeh
- Koç University Research Center for Translational Medicine, KUTTAM, Koç University, Istanbul, Turkey
| | - Selin Sapancı
- Koç University Research Center for Translational Medicine, KUTTAM, Koç University, Istanbul, Turkey
| | - Ceyda Özler
- Koç University Research Center for Translational Medicine, KUTTAM, Koç University, Istanbul, Turkey
| | - Judy Kesibi
- Koç University Research Center for Translational Medicine, KUTTAM, Koç University, Istanbul, Turkey
| | - Yasemin Gürsoy-Özdemir
- Koç University Research Center for Translational Medicine, KUTTAM, Koç University, Istanbul, Turkey
- Department of Neurology, School of Medicine, Koç University, Istanbul, Turkey
| |
Collapse
|
26
|
Martinez Ramirez CE, Ruiz-Pérez G, Stollenwerk TM, Behlke C, Doherty A, Hillard CJ. Endocannabinoid signaling in the central nervous system. Glia 2023; 71:5-35. [PMID: 36308424 PMCID: PMC10167744 DOI: 10.1002/glia.24280] [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: 02/01/2022] [Revised: 09/02/2022] [Accepted: 09/29/2022] [Indexed: 11/07/2022]
Abstract
It is hard to overestimate the influence of the endocannabinoid signaling (ECS) system on central nervous system (CNS) function. In the 40 years since cannabinoids were found to trigger specific cell signaling cascades, studies of the ECS system continue to cause amazement, surprise, and confusion! CB1 cannabinoid receptors are expressed widely in the CNS and regulate cell-cell communication via effects on the release of both neurotransmitters and gliotransmitters. CB2 cannabinoid receptors are difficult to detect in the CNS but seem to "punch above their weight" as compounds targeting these receptors have significant effects on inflammatory state and behavior. Positive and negative allosteric modulators for both receptors have been identified and examined in preclinical studies. Concentrations of the endocannabinoid ligands, N-arachidonoylethanolamine and 2-arachidonoylglycerol (2-AG), are regulated by a combination of enzymatic synthesis and degradation and inhibitors of these processes are available and making their way into clinical trials. Importantly, ECS regulates many essential brain functions, including regulation of reward, anxiety, inflammation, motor control, and cellular development. While the field is on the cusp of preclinical discoveries providing impactful clinical and therapeutic insights into many CNS disorders, there is still much to be learned about this remarkable and versatile modulatory system.
Collapse
Affiliation(s)
- César E Martinez Ramirez
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Gonzalo Ruiz-Pérez
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Todd M Stollenwerk
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Christina Behlke
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Ashley Doherty
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Cecilia J Hillard
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
27
|
Actaea racemosa L. Rhizome Protect against MPTP-Induced Neurotoxicity in Mice by Modulating Oxidative Stress and Neuroinflammation. Antioxidants (Basel) 2022; 12:antiox12010040. [PMID: 36670902 PMCID: PMC9854773 DOI: 10.3390/antiox12010040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/09/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Parkinson's disease (PD) is a dopaminergic neuron-related neurodegenerative illness. Treatments exist that alleviate symptoms but have a variety of negative effects. Recent research has revealed that oxidative stress, along with neuroinflammation, is a major factor in the course of this disease. Therefore, the aim of our study was to observe for the first time the effects of a natural compound such as Actaea racemosa L. rhizome in an in vivo model of PD induced by neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). For the study, mice received four injections of MPTP (20 mg/kg) for the induction of PD. Starting 24 h after the first administration of MPTP we treated mice with Actaea racemosa L. rhizome (100 mg/kg) daily for seven days. Our findings clearly demonstrated that Actaea racemosa L. rhizome treatment decreases oxidative stress by activating redox balance enzymes such as Nrf2/HO-1. We also demonstrated that Actaea racemosa L. rhizome is capable of modulating inflammatory indicators involved in PD, such as IκB-α, NF-κB, GFAP and Iba1, thus reducing the degeneration of dopaminergic neurons and motor and non-motor alterations. To summarize, Actaea racemosa L. rhizome, which is subject to fewer regulations than traditional medications, could be used as a dietary supplement to improve patients' brain health and could be a promising nutraceutical choice to slow the course and symptoms of PD.
Collapse
|
28
|
Inhibition of the TLR/NF- κB Signaling Pathway and Improvement of Autophagy Mediates Neuroprotective Effects of Plumbagin in Parkinson's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1837278. [PMID: 36589679 PMCID: PMC9800084 DOI: 10.1155/2022/1837278] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 12/05/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
A naphthoquinone molecule known as plumbagin (PL), which has a wide range of pharmacological properties including antitumor, antioxidation, anti-inflammation, and neuroprotective effects, is extracted from the roots of the medicinal herb Plumbago zeylanica L. Plumbagin has been studied for its potential to treat Parkinson's disease (PD). However, its effectiveness and mechanism are still unknown. This study intends to evaluate plumbagin's effectiveness against PD in vitro and in vivo. Plumbagin partially repaired the loss of dopaminergic neurons in the nigral substantia nigra and the resulting behavioural impairment caused by MPTP or MPTP/probenecid in mice. Furthermore, plumbagin treatment significantly inhibited the TLR/NF-κB pathways. It reduced the TNF-α, IL-6, and IL-1β mRNA expression in PD mice induced by MPTP or MPTP/probenecid, which was consistent with the findings in the inflammatory model of BV2 cells induced by MPP+ or LPS. In addition, plumbagin treatment enhanced the microtubule-associated protein 1 light chain 3 beta (LC3) LC3-II/LC3-I levels while decreasing the p-mTOR and p62 protein accumulation in PD mice induced by MPTP or MPTP/probenecid, which was similar to the results obtained from the experiments in SH-SY5Y and PC12 cells induced by MPP+. Consequently, our results support the hypothesis that plumbagin, by promoting autophagy and inhibiting the activation of the TLR/NF-κB signaling pathway, is a promising treatment agent for treating Parkinson's disease (PD). However, to confirm plumbagin's anti-PD action more thoroughly, other animal and cell PD models must be used in future studies.
Collapse
|
29
|
Shao J, Liu X, Lian M, Mao Y. Citronellol Prevents 6-OHDA-Induced Oxidative Stress, Mitochondrial Dysfunction, and Apoptosis in Parkinson Disease Model of SH-SY5Y Cells via Modulating ROS-NO, MAPK/ERK, and PI3K/Akt Signaling Pathways. Neurotox Res 2022; 40:2221-2237. [PMID: 36097250 DOI: 10.1007/s12640-022-00558-8] [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: 04/26/2022] [Revised: 07/13/2022] [Accepted: 08/09/2022] [Indexed: 12/31/2022]
Abstract
Parkinson disease is a neurodegenerative disorder distinguished by dopaminergic shortage in the striatum and the accumulation of α-synuclein neuronal aggregates in the brains of patients. Since, there is no accurate treatment available for Parkinson disease, researches are designed to alleviate the pathognomonic symptoms such as neuroinflammation, oxidative stress, mitochondrial dysfunction, and apoptosis. Accordingly, a number of compounds have been reported to inhibit these pathognomonic symptoms. In this study, we have assessed the neuroprotective potential of citronellol against 6-OHDA-induced neurotoxicity in SH-SY5Y cells. The results found that citronellol treatment effectively hindered the cell death caused by 6-OHDA and thereby maintaining the cell viability in SH-SY5Y cells at 50 µg/mL concentration. As expected, the citronellol treatment significantly reduced the 6-OHDA-induced secretion of inflammatory factors (IL-1β, IL-6, and TNF-α), which was obtained through ELISA technique. Similarly, citronellol hindered the 6-OHDA-induced oxidative stress by lowering the intracellular ROS and NO level and MDA leakage along with increased expression of SOD level in SH-SY5Y cells. The JC-1 staining showed that 6-OHDA increased the number of green fluorescent dots with ruptured mitochondrial membrane potential, while citronellol increased the amount of red fluorescent, showing the rescue potential against the 6-OHDA-induced mitochondrial dysfunction. Furthermore, citronellol hampered the 6-OHDA-induced apoptosis via the suppression of Bcl-2/Bax pathway. The western blotting results hypothesized that citronellol rescued SH-SY5Y cells from 6-OHDA-induced neurotoxicity via modulating ROS-NO, MAPK/ERK, and PI3K/Akt signaling pathways. However, further clinical trials are required to verify the anti-Parkinson efficacy.
Collapse
Affiliation(s)
- Jiahui Shao
- Department of Neurology, The First People's Hospital of Wenling, Zhejiang Province, Wenling, 317500, China
| | - Xuan Liu
- Department of Neurology, The First People's Hospital of Wenling, Zhejiang Province, Wenling, 317500, China
| | - Mengjia Lian
- Department of Neurology, The First People's Hospital of Wenling, Zhejiang Province, Wenling, 317500, China
| | - Youbing Mao
- Department of Special Inspection Section, The First People's Hospital of Wenling, No. 333, Chuanan South Road, Chengxi StreetZhejiang Province, Wenling, 317500, China.
| |
Collapse
|
30
|
Davis J, Kolaski E, Babcock DT. Vexed mutations promote degeneration of dopaminergic neurons through excessive activation of the innate immune response. NPJ Parkinsons Dis 2022; 8:147. [PMID: 36323700 PMCID: PMC9630459 DOI: 10.1038/s41531-022-00417-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 10/14/2022] [Indexed: 11/11/2022] Open
Abstract
The hallmark of Parkinson's disease (PD) is the loss of dopaminergic (DA) neurons in the brain. However, little is known about why DA neurons are selectively vulnerable to PD. We previously completed a screen identifying genes associated with the progressive degeneration of DA neurons. Here we describe the role of a previously uncharacterized gene, CG42339, in the loss of DA neurons using Drosophila Melanogaster. CG42339 mutants display a progressive loss of DA neurons and locomotor dysfunction, along with an accumulation of advanced glycation end products (AGEs) in the brain. Based on this phenotype, we refer to CG42339 as vexed. We demonstrate that vexed is specifically required within cortex glia to maintain neuronal viability. Loss of vexed function results in excessive activation of the innate immune response in the brain, leading to loss of DA neurons. We show that activation of the innate immune response leads to increased nitric oxide signaling and accumulation of AGEs, which ultimately result in neurodegeneration. These results provide further insight into the relationship between the role of the immune response in the central nervous system and how this impacts neuronal viability.
Collapse
Affiliation(s)
- Jacinta Davis
- grid.259029.50000 0004 1936 746XDepartment of Biological Sciences, Lehigh University, Bethlehem, PA USA
| | - Elizabeth Kolaski
- grid.259029.50000 0004 1936 746XDepartment of Biological Sciences, Lehigh University, Bethlehem, PA USA
| | - Daniel T. Babcock
- grid.259029.50000 0004 1936 746XDepartment of Biological Sciences, Lehigh University, Bethlehem, PA USA
| |
Collapse
|
31
|
Qin Z, Wang H, Dou Q, Xu L, Xu Z, Jia R. Protective effect of fluoxetine against oxidative stress induced by renal ischemia-reperfusion injury via the regulation of miR-450b-5p/Nrf2 axis. Aging (Albany NY) 2022; 15:15640-15656. [PMID: 36126189 PMCID: PMC10781502 DOI: 10.18632/aging.204289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 09/01/2022] [Indexed: 11/25/2022]
Abstract
The present study was performed to assess the protective effect of fluoxetine (FLX) on renal ischemia-reperfusion injury (IRI) via the regulation of miR-450b-5p/Nrf2 axis in male rats. In vivo, these male rats were randomly divided into different treatment groups. The rats were administered with FLX (20 mg/kg, intraperitoneally) once daily for 3 days before operation. The pathomorphological changes of renal tissues were assessed by histological examination and Masson staining. In vitro, HK-2 cells were used to detect the activity by CCK-8 assay in Hypoxia/Reoxygenation (H/R) group and Hypoxia/Reoxygenation+Fluoxetine (H/R+FLX) group. In addition, the oxidative stress biomarkers were evaluated. Subsequently, Nrf2, NF-κB, and Nrf2-dependent antioxidant enzymes, were detected by Western blot assay. In vivo, the pathological changes and serological renal function were significantly relieved in the rats with the pre-treatment of FLX, compared to IRI group. After FLX stimulation, the expression levels of oxidative stress indices significantly decreased, while tissue antioxidant indices significantly increased, compared to IRI group. The differently expressed miRNAs on renal IRI in male rats were screened out by miRNA microarray, especially showing that miR-450b-5p was selected as the target miRNA. Following miR-450b-5p agomir injection, the pathological changes and oxidative stress biomarkers significantly aggravated, whether in IRI group or IRI+FLX group. Bioinformatics analysis and double-luciferase reporter assay demonstrated that miR-450b-5p directly targeted Nrf2. The expression level of NF-κB significantly increased, while the expression levels of Nrf2 and Nrf2-dependent antioxidant enzymes significantly decreased after miR-450b-5p agomir injection. Furthermore, the expression levels of Nrf2 and it-dependent antioxidant enzymes were apparently increased in ischemic kidney after the transfection of miR-450b-5p mimic+recombination protein Nrf2, as well as the decreased expression levels of intracellular ROS and iNOS. In vitro, FLX significantly increased HK-2 cell viability, and relieved H/R HK-2 cell oxidative injury via down-regulating ROS and iNOS. In addition, H/R-induced oxidative damage was recovered with miR-450b-5p mimic and recombination protein Nrf2. Consequently, FLX played an important protective role in renal IRI-induced oxidative damage by promoting antioxidation via targeting miR-450b-5p/Nrf2 axis.
Collapse
Affiliation(s)
- Zhiqiang Qin
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Hao Wang
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Quanliang Dou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Luwei Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Zheng Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Ruipeng Jia
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| |
Collapse
|
32
|
Ding MR, Qu YJ, Hu B, An HM. Signal pathways in the treatment of Alzheimer's disease with traditional Chinese medicine. Biomed Pharmacother 2022; 152:113208. [PMID: 35660246 DOI: 10.1016/j.biopha.2022.113208] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 11/28/2022] Open
Abstract
AIM OF THE REVIEW This study aimed to reveal the classical signal pathways and important potential targets of traditional Chinese medicine (TCM) for treating Alzheimer's disease (AD), and provide support for further investigation on TCM and its active ingredients. MATERIALS AND METHODS Literature survey was conducted using PubMed, Web of Science, Google Scholar, CNKI, and other databases, with "Alzheimer's disease," "traditional Chinese medicine," "medicinal herb," "Chinese herb," and "natural plant" as the primary keywords. RESULTS TCM could modulate signal pathways related to AD pathological progression, including NF-κB, Nrf2, JAK/STAT, ubiquitin-proteasome pathway, autophagy-lysosome pathway-related AMPK/mTOR, GSK-3/mTOR, and PI3K/Akt/mTOR, as well as SIRT1 and PPARα pathway. It could regulate crosstalk between pathways through a multitarget, thus maintaining chronic inflammatory interaction balance, inhibiting oxidative stress damage, regulating ubiquitin-proteasome system function, modulating autophagy, and eventually improving cognitive impairment in patients with AD. CONCLUSION TCM could be multilevel, multitargeted, and multifaceted to prevent and treat AD. In-depth research on the prevention and treatment of AD with TCM could provide new ideas for exploring the pathogenesis of AD and developing new anti-AD drugs.
Collapse
Affiliation(s)
- Min-Rui Ding
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yan-Jie Qu
- Department of Neurology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Bing Hu
- Institute of Traditional Chinese Medicine in Oncology, Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Hong-Mei An
- Department of Science & Technology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| |
Collapse
|
33
|
Soni D, Kumar P. GSK-3β-mediated regulation of Nrf2/HO-1 signaling as a new therapeutic approach in the treatment of movement disorders. Pharmacol Rep 2022; 74:557-569. [PMID: 35882765 DOI: 10.1007/s43440-022-00390-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/09/2022] [Accepted: 07/12/2022] [Indexed: 11/30/2022]
Abstract
Movement disorders are neurological conditions characterized by involuntary motor movements, such as dystonia, ataxia, chorea myoclonus, tremors, Huntington's disease (HD), and Parkinson's disease (PD). It is classified into two categories: hypokinetic and hyperkinetic movements. Globally, movement disorders are a major cause of death. The pathophysiological process is initiated by excessive ROS generation, mitochondrial dysfunction, neuroinflammation, and neurotransmitters imbalance that lead to motor dysfunction in PD and HD patients. Several endogenous targets including Nrf2 maintain oxidative balance in the body. Activation of Nrf2 signaling is regulated by the enzyme glycogen synthase kinase (GSK-3β). In the cytoplasm, inhibition of GSK-3β regulates cellular proliferation, homeostasis, and apoptotic process by stimulating the nuclear factor erythroid 2 (Nrf2) pathway which is involved in the elevation of the cellular antioxidant enzymes which controls the ROS generation. The activation of Nrf2 increases the expression of antioxidant response elements (ARE), such as (Hemeoxygenase-1) HO-1, which decreases excessive cellular stress, mitochondrial dysfunction, apoptosis, and neuronal degeneration, which is the major cause of motor dysfunction. The present review explores the GSK-3β-mediated neuroprotection in various movement disorders through the Nrf2/HO-1 antioxidant pathway. This review provides a link between GSK-3β and the Nrf2/HO-1 signaling pathway in the treatment of PD and HD. In addition to that it highlights various GSK-3β inhibitors and the Nrf2/HO-1 activators, which exert robust neuroprotection against motor disorders. Therefore, the present review will help in the discovery of new therapy for PD and HD patients.
Collapse
Affiliation(s)
- Divya Soni
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India.
| |
Collapse
|
34
|
Yang XX, Yang R, Zhang F. Role of Nrf2 in Parkinson's Disease: Toward New Perspectives. Front Pharmacol 2022; 13:919233. [PMID: 35814229 PMCID: PMC9263373 DOI: 10.3389/fphar.2022.919233] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/12/2022] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease (PD) is one of the most common and chronic degenerative diseases in the central nervous system. The main pathology of PD formation is the progressive loss of dopaminergic neurons in substantia nigra and the formation of α-synuclein-rich Lewy bodies. The pathogenesis of PD is not caused by any single independent factor. The diversity of these independent factors of PD, such as iron accumulation, oxidative stress, neuroinflammation, mitochondrial dysfunction, age, environment, and heredity, makes the research progress of PD slow. Nrf2 has been well-known to be closely associated with the pathogenesis of PD and could regulate these induced factors development. Nrf2 activation could protect dopaminergic neurons and slow down the progression of PD. This review summarized the role of Nrf2 pathway on the pathogenesis of PD. Regulation of Nrf2 pathway might be one of the promising strategies to prevent and treat PD.
Collapse
Affiliation(s)
- Xin-xing Yang
- Laboratory Animal Center and Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Rong Yang
- Laboratory Animal Center and Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Feng Zhang
- Laboratory Animal Center and Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| |
Collapse
|
35
|
Amin R, Quispe C, Docea AO, Alibek Y, Kulbayeva M, Durna Daştan S, Calina D, Sharifi-Rad J. The role of Tumour Necrosis Factor in neuroinflammation associated with Parkinson's disease and targeted therapies. Neurochem Int 2022; 158:105376. [PMID: 35667491 DOI: 10.1016/j.neuint.2022.105376] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 05/29/2022] [Accepted: 05/30/2022] [Indexed: 12/21/2022]
Abstract
Neurodegenerative disorders Parkinson's disease is a progressive neurodegenerative disorder associated with neuroinflammatory responses that lead to the neurodegeneration of the dopaminergic neurons. These neuroinflammatory mechanisms involve various cytokines produced by the activated glial cells. Tumour Necrosis factor α (TNF α) is one of the major mediators of the neuroinflammation associated with neurodegeneration. TNF α has a dual role of neuroprotection and neurotoxicity in the brain. The effective pathways of TNF involve various signalling pathways transduced by the receptors TNFR1 and TNFR2. Effective therapeutic strategies have been produced targeting the neurotoxic behaviour of the Tumour Necrosis Factor and the associated neurodegeneration which includes the use of Dominant Negative Tumour Necrosis Factor (DN-TNF) inhibitors like XENP 345 and XPro®1595 and peroxisome proliferator receptor gamma (PPAR-γ) agonists.
Collapse
Affiliation(s)
- Ruhul Amin
- Faculty of Pharmaceutical Science, Assam Down Town University, Panikhaiti, Guwahati, Assam, India.
| | - Cristina Quispe
- Facultad de Ciencias de la Salud, Universidad Arturo Prat, Avda. Arturo Prat 2120, Iquique, 1110939, Chile.
| | - Anca Oana Docea
- Department of Toxicology, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania
| | - Ydyrys Alibek
- Biomedical Research Centre, Al-Farabi Kazakh National University, Al-Farabi av. 71, 050040, Almaty, Kazakhstan.
| | - Marzhan Kulbayeva
- Department of Biophysics, Biomedicine and Neuroscience, Al-Farabi Kazakh National University, Al-Farabi av. 71, 050040, Almaty, Kazakhstan.
| | - Sevgi Durna Daştan
- Department of Biology, Faculty of Science, Sivas Cumhuriyet University, 58140, Sivas, Turkey; Beekeeping Development Application and Research Center, Sivas Cumhuriyet University, 58140, Sivas, Turkey.
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| | | |
Collapse
|
36
|
Polarization of Microglia and Its Therapeutic Potential in Sepsis. Int J Mol Sci 2022; 23:ijms23094925. [PMID: 35563317 PMCID: PMC9101892 DOI: 10.3390/ijms23094925] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/17/2022] [Accepted: 01/17/2022] [Indexed: 02/04/2023] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection, leaving the inflammation process without a proper resolution, leading to tissue damage and possibly sequelae. The central nervous system (CNS) is one of the first regions affected by the peripheral inflammation caused by sepsis, exposing the neurons to an environment of oxidative stress, triggering neuronal dysfunction and apoptosis. Sepsis-associated encephalopathy (SAE) is the most frequent sepsis-associated organ dysfunction, with symptoms such as deliriums, seizures, and coma, linked to increased mortality, morbidity, and cognitive disability. However, the current therapy does not avoid those patients’ symptoms, evidencing the search for a more optimal approach. Herein we focus on microglia as a prominent therapeutic target due to its multiple functions maintaining CNS homeostasis and its polarizing capabilities, stimulating and resolving neuroinflammation depending on the stimuli. Microglia polarization is a target of multiple studies involving nerve cell preservation in diseases caused or aggravated by neuroinflammation, but in sepsis, its therapeutic potential is overlooked. We highlight the peroxisome proliferator-activated receptor gamma (PPARγ) neuroprotective properties, its role in microglia polarization and inflammation resolution, and the interaction with nuclear factor-κB (NF-κB) and mitogen-activated kinases (MAPK), making PPARγ a molecular target for sepsis-related studies to come.
Collapse
|
37
|
Li K, Ning P, Liu B, Yang H, Zhu Y, Yin W, Zhou C, Ren H, Yang X. Downregulation of CHCHD2 may Contribute to Parkinson's Disease by Reducing Expression of NFE2L2 and RQCD1. Curr Neurovasc Res 2022; 19:19-29. [PMID: 35388756 DOI: 10.2174/1567202619666220406082221] [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: 12/10/2021] [Revised: 12/23/2021] [Accepted: 12/28/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Parkinson's disease (PD) is associated with coiled-coil-helix-coiled-coil-helix domain containing 2 (CHCHD2) downregulation, which has been linked to reduced cyclocytase activity and increased levels of oxygen free radicals, leading to mitochondrial fragmentation and apoptosis. Little is known about how CHCHD2 normally functions in the cell and, therefore, how its downregulation may contribute to PD. OBJECTIVE This study aimed to identify such target genes using chromatin immunoprecipitation sequencing from SH-SY5Y human neuroblastoma cells treated with neurotoxin 1-methyl-4-phenylpyridinium (MPP+) as a PD model. METHODS In this study, we established a MPP+ -reated SH-SY5Y cell model and evaluated the effects of CHCHD2 overexpression on cell proliferation and apoptosis. At the same time, we used high-throughput chromatin immunoprecipitation sequencing to identify its downstream target gene in SH-SY5Y cells. In addition, we verified the possible downstream target genes and discussed their mechanisms. RESULTS The expression level of α-synuclein increased in SH-SY5Y cells treated with MPP+, while the protein expression level of CHCHD2 decreased significantly, especially after 24 h of treatment. Chip-IP results showed that CHCHD2 may regulate potential target genes such as HDX, ACP1, RAVER2, C1orf229, RN7SL130, GNPTG, erythroid 2 Like 2 (NFE2L2), required for cell differentiation 1 homologue (RQCD1), solute carrier family 5 member 7 (SLA5A7), and N-Acetyltransferase 8 Like (NAT8L). NFE2L2 and RQCD1 were validated as targets using PCR and western blotting of immunoprecipitates, and these two genes together with SLA5A7 and NAT8L were upregulated in SH-SY5Y cells overexpressing CHCHD2. Downregulation of CHCHD2 may contribute to PD by leading to inadequate expression of NFE2L2 and RQCD1 as well as, potentially, SLA5A7 and NAT8L. CONCLUSION Our results suggest that CHCHD2 plays a protective role by maintaining mitochondrial homeostasis and promoting proliferation in neurons. In this study, the changes of CHCHD2 and downstream target genes such as NFE2L2/RQCD1 may have potential application prospects in the future. These findings provide leads to explore PD pathogenesis and potential treatments.
Collapse
Affiliation(s)
- Kelu Li
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, PR China
| | - Pingping Ning
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Bin Liu
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, PR China.,Yunnan Province Clinical Research Center for Gerontology, Kunming, Yunnan Province, PR China
| | - Hongju Yang
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, PR China.,Yunnan Province Clinical Research Center for Gerontology, Kunming, Yunnan Province, PR China
| | - Yongyun Zhu
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, PR China
| | - WeiFang Yin
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, PR China
| | - Chuanbin Zhou
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, PR China
| | - Hui Ren
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, PR China.,Yunnan Province Clinical Research Center for Gerontology, Kunming, Yunnan Province, PR China
| | - Xinglong Yang
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, PR China.,Yunnan Province Clinical Research Center for Gerontology, Kunming, Yunnan Province, PR China
| |
Collapse
|
38
|
Chakkittukandiyil A, Sajini DV, Karuppaiah A, Selvaraj D. The principal molecular mechanisms behind the activation of Keap1/Nrf2/ARE pathway leading to neuroprotective action in Parkinson's disease. Neurochem Int 2022; 156:105325. [PMID: 35278519 DOI: 10.1016/j.neuint.2022.105325] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 03/03/2022] [Accepted: 03/06/2022] [Indexed: 02/07/2023]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder. PD is associated with the loss of dopaminergic neurons in the substantia nigra pars compacta region of the midbrain. Present therapies for PD provide only symptomatic relief by restoring the dopamine (DA) level. However, they are not disease modifying agents and so they do not delay the disease progression. Alpha-synuclein aggregation, oxidative stress, mitochondrial dysfunction and chronic inflammation are considered to be the major pathological mechanisms mediating neurodegeneration in PD. To resist oxidative stress, the human body has an antioxidant defence mechanism consisting of many antioxidants and cytoprotective genes. The expression of those genes are largely controlled by the Kelch-like ECH-associated protein 1/Nuclear factor - erythroid - 2 - related factor 2/Antioxidant response element (Keap1/Nrf2/ARE) signalling pathway. The transcription factor Nrf2 is activated in response to oxidative or electrophilic stress and protects the cells from oxidative stress and inflammation. Nrf2 has been widely considered as a therapeutic target for neurodegeneration and several drugs are now being tested in clinical trials. Regulation of the Keap1/Nrf2/ARE pathway by small molecules which can act as Nrf2 activators could be effective for treating oxidative stress and neuroinflammation in PD. In this review, we had discussed the principal molecular mechanisms behind the neuroprotective effects of Keap1/Nrf2/ARE pathway in PD. Additionally, we also discussed the small molecules and phytochemicals that could activate the Nrf2 mediated anti-oxidant pathway for neuroprotection in PD.
Collapse
Affiliation(s)
- Amritha Chakkittukandiyil
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Deepak Vasudevan Sajini
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Arjunan Karuppaiah
- Department of Pharmaceutics, PSG College of Pharmacy, Peelamedu, Coimbatore, Tamil Nadu, India
| | - Divakar Selvaraj
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India.
| |
Collapse
|
39
|
Hatton SL, Pandey MK. Fat and Protein Combat Triggers Immunological Weapons of Innate and Adaptive Immune Systems to Launch Neuroinflammation in Parkinson's Disease. Int J Mol Sci 2022; 23:1089. [PMID: 35163013 PMCID: PMC8835271 DOI: 10.3390/ijms23031089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 01/27/2023] Open
Abstract
Parkinson's disease (PD) is the second-most common neurodegenerative disease in the world, affecting up to 10 million people. This disease mainly happens due to the loss of dopaminergic neurons accountable for memory and motor function. Partial glucocerebrosidase enzyme deficiency and the resultant excess accumulation of glycosphingolipids and alpha-synuclein (α-syn) aggregation have been linked to predominant risk factors that lead to neurodegeneration and memory and motor defects in PD, with known and unknown causes. An increasing body of evidence uncovers the role of several other lipids and their association with α-syn aggregation, which activates the innate and adaptive immune system and sparks brain inflammation in PD. Here, we review the emerging role of a number of lipids, i.e., triglyceride (TG), diglycerides (DG), glycerophosphoethanolamines (GPE), polyunsaturated fatty acids (PUFA), sphingolipids, gangliosides, glycerophospholipids (GPL), and cholesterols, and their connection with α-syn aggregation as well as the induction of innate and adaptive immune reactions that trigger neuroinflammation in PD.
Collapse
Affiliation(s)
- Shelby Loraine Hatton
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Cincinnati, OH 45229, USA;
| | - Manoj Kumar Pandey
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Cincinnati, OH 45229, USA;
- Department of Pediatrics, Division of Human Genetics, College of Medicine, University of Cincinnati, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| |
Collapse
|
40
|
Lang Y, Zhang H, Yu H, Li Y, Liu X, Li M. Long non-coding RNA myocardial infarction-associated transcript promotes 1-Methyl-4-phenylpyridinium ion-induced neuronal inflammation and oxidative stress in Parkinson's disease through regulating microRNA-221-3p/ transforming growth factor /nuclear factor E2-related factor 2 axis. Bioengineered 2021; 13:930-940. [PMID: 34967706 PMCID: PMC8805986 DOI: 10.1080/21655979.2021.2015527] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
This study attempted to evaluate the role of long non-coding RNA myocardial infarction-associated transcript (LncRNA MIAT) in Parkinson’s disease (PD). The mouse model was established through intraperitoneal injection with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), and in vitro model was induced by administrating cell with 1-Methyl-4-phenylpyridinium ion (MPP+). Rotarod test was conducted to evaluate the motor coordination of PD mice. In order to investigate the roles of LncRNA MIAT in neuronal inflammation and oxidative stress, MIAT shRNA (shMIAT) was transfected into MPP+-treated cells, and cell viability, cell apoptosis and oxidative stress response were evaluated. To evaluate the interactions between LncRNA MIAT and microRNA-221-3p (miR-221-3p)/TGF-β1/Nrf2, miR-221-3p mimic, miR-221-3p inhibitor, NC-inhibitor and transforming growth factor-β1 shRNA (shTGF-β1) were subsequently transfected into MPP+-treated cells. Dual-luciferase reporter gene assays were performed to determine the interaction of miR-221-3p with MIAT or TGFB receptor 1 (TGFBR1). The expressions of LncRNA MIAT, miR-221-3p, TGFBR1, transforming growth factor (TGF-β1) and nuclear factor E2-related factor 2 (Nrf2) were measured by quantitative reverse-transcription polymerase chain reaction (RT-qPCR) and immunoblotting. As a result, LncRNA MIAT was abundantly expressed in PD mice and cells, while downregulation of LncRNA MIAT promoted the survival of neurons, inhibited apoptosis and oxidative stress in neurons. LncRNA MIAT bound to miR-221-3p, and there was a negative correlation between miR-221-3p and LncRNA MIAT expression. In addition, miR-221-3p targeted TGFBR1 and suppressed TGF-β1 expression but increased Nrf2 expression. LncRNA MIAT promoted MPP+-induced neuronal injury in PD via regulating TGF-β1/Nrf2 axis through binding with miR-221-3p.
Collapse
Affiliation(s)
- Yue Lang
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian City, Liaoning Province, China
| | - Hui Zhang
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian City, Liaoning Province, China
| | - Haojia Yu
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian City, Liaoning Province, China
| | - Yu Li
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian City, Liaoning Province, China
| | - Xiao Liu
- Graduate School, Dalian Medical University, Dalian City, Liaoning Province, China
| | - Minjie Li
- Graduate School, Dalian Medical University, Dalian City, Liaoning Province, China
| |
Collapse
|
41
|
Emodin ameliorates antioxidant capacity and exerts neuroprotective effect via PKM2-mediated Nrf2 transactivation. Food Chem Toxicol 2021; 160:112790. [PMID: 34971761 DOI: 10.1016/j.fct.2021.112790] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 12/21/2021] [Accepted: 12/24/2021] [Indexed: 11/20/2022]
Abstract
Pyruvate kinase M2 (PKM2) is overexpressed in neuronal cells. However, there are few studies on the involvement of PKM2 modulators in neurodegenerative diseases. Emodin, a dominating anthraquinone derivative extracting from the rhizome of rhubarb, has received expanding consideration due to its pharmacological properties. Our data reveal that emodin could resist hydrogen peroxide- or 6-hydroxydopamine-mediated mitochondrial fission and apoptosis in PC12 cells (a neuron-like rat pheochromocytoma cell line). Notably, emodin at nontoxic concentrations significantly inhibits PKM2 activity and promotes dissociation of tetrameric PKM2 into dimers in cells. The PKM2 dimerization enhances the interaction of PKM2 and NFE2-related factor 2 (Nrf2), which further triggers the activation of the Nrf2/ARE pathway to upregulate a panel of cytoprotective genes. Modulating the PKM2/Nrf2/ARE axis by emodin unveils a novel mechanism for understanding the pharmacological functions of emodin. Our findings indicate that emodin is a potential candidate for the treatment of oxidative stress-related neurodegenerative disorders.
Collapse
|
42
|
Insulin-like Growth Factor II Prevents MPP+ and Glucocorticoid Mitochondrial-Oxidative and Neuronal Damage in Dopaminergic Neurons. Antioxidants (Basel) 2021; 11:antiox11010041. [PMID: 35052545 PMCID: PMC8773450 DOI: 10.3390/antiox11010041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/19/2021] [Accepted: 12/22/2021] [Indexed: 11/17/2022] Open
Abstract
Stress seems to contribute to Parkinson’s disease (PD) neuropathology, probably by dysregulation of the hypothalamic–pituitary–adrenal axis. Key factors in this pathophysiology are oxidative stress and mitochondrial dysfunction and neuronal glucocorticoid-induced toxicity. The insulin-like growth factor II (IGF-II), a pleiotropic hormone, has shown antioxidant and neuroprotective effects in some neurodegenerative disorders. Our aim was to examine the protective effect of IGF-II on a dopaminergic cellular combined model of PD and mild to moderate stress measuring oxidative stress parameters, mitochondrial and neuronal markers, and signalling pathways. IGF-II counteracts the mitochondrial-oxidative damage produced by the toxic synergistic effect of corticosterone and 1-methyl-4-phenylpyridinium, protecting dopaminergic neurons from death and neurodegeneration. IGF-II promotes PKC activation and nuclear factor (erythroid-derived 2)-like 2 antioxidant response in a glucocorticoid receptor-dependent pathway, preventing oxidative cell damage and maintaining mitochondrial function. Thus, IGF-II is a potential therapeutic tool for treatment and prevention of disease progression in PD patients suffering mild to moderate emotional stress.
Collapse
|
43
|
Boas SM, Joyce KL, Cowell RM. The NRF2-Dependent Transcriptional Regulation of Antioxidant Defense Pathways: Relevance for Cell Type-Specific Vulnerability to Neurodegeneration and Therapeutic Intervention. Antioxidants (Basel) 2021; 11:antiox11010008. [PMID: 35052512 PMCID: PMC8772787 DOI: 10.3390/antiox11010008] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/09/2021] [Accepted: 12/14/2021] [Indexed: 12/11/2022] Open
Abstract
Oxidative stress has been implicated in the etiology and pathobiology of various neurodegenerative diseases. At baseline, the cells of the nervous system have the capability to regulate the genes for antioxidant defenses by engaging nuclear factor erythroid 2 (NFE2/NRF)-dependent transcriptional mechanisms, and a number of strategies have been proposed to activate these pathways to promote neuroprotection. Here, we briefly review the biology of the transcription factors of the NFE2/NRF family in the brain and provide evidence for the differential cellular localization of NFE2/NRF family members in the cells of the nervous system. We then discuss these findings in the context of the oxidative stress observed in two neurodegenerative diseases, Parkinson's disease (PD) and amyotrophic lateral sclerosis (ALS), and present current strategies for activating NFE2/NRF-dependent transcription. Based on the expression of the NFE2/NRF family members in restricted populations of neurons and glia, we propose that, when designing strategies to engage these pathways for neuroprotection, the relative contributions of neuronal and non-neuronal cell types to the overall oxidative state of tissue should be considered, as well as the cell types which have the greatest intrinsic capacity for producing antioxidant enzymes.
Collapse
Affiliation(s)
- Stephanie M. Boas
- Department of Neuroscience, Southern Research, 2000 9th Avenue South, Birmingham, AL 35205, USA; (S.M.B.); (K.L.J.)
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, 1720 2nd Avenue South, Birmingham, AL 35294, USA
| | - Kathlene L. Joyce
- Department of Neuroscience, Southern Research, 2000 9th Avenue South, Birmingham, AL 35205, USA; (S.M.B.); (K.L.J.)
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, 1720 2nd Avenue South, Birmingham, AL 35294, USA
| | - Rita M. Cowell
- Department of Neuroscience, Southern Research, 2000 9th Avenue South, Birmingham, AL 35205, USA; (S.M.B.); (K.L.J.)
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, 1720 2nd Avenue South, Birmingham, AL 35294, USA
- Correspondence:
| |
Collapse
|
44
|
Parga JA, Rodriguez-Perez AI, Garcia-Garrote M, Rodriguez-Pallares J, Labandeira-Garcia JL. NRF2 Activation and Downstream Effects: Focus on Parkinson's Disease and Brain Angiotensin. Antioxidants (Basel) 2021; 10:antiox10111649. [PMID: 34829520 PMCID: PMC8614768 DOI: 10.3390/antiox10111649] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/17/2021] [Accepted: 10/18/2021] [Indexed: 12/18/2022] Open
Abstract
Reactive oxygen species (ROS) are signalling molecules used to regulate cellular metabolism and homeostasis. However, excessive ROS production causes oxidative stress, one of the main mechanisms associated with the origin and progression of neurodegenerative disorders such as Parkinson's disease. NRF2 (Nuclear Factor-Erythroid 2 Like 2) is a transcription factor that orchestrates the cellular response to oxidative stress. The regulation of NRF2 signalling has been shown to be a promising strategy to modulate the progression of the neurodegeneration associated to Parkinson's disease. The NRF2 pathway has been shown to be affected in patients with this disease, and activation of NRF2 has neuroprotective effects in preclinical models, demonstrating the therapeutic potential of this pathway. In this review, we highlight recent advances regarding the regulation of NRF2, including the effect of Angiotensin II as an endogenous signalling molecule able to regulate ROS production and oxidative stress in dopaminergic neurons. The genes regulated and the downstream effects of activation, with special focus on Kruppel Like Factor 9 (KLF9) transcription factor, provide clues about the mechanisms involved in the neurodegenerative process as well as future therapeutic approaches.
Collapse
Affiliation(s)
- Juan A. Parga
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.I.R.-P.); (M.G.-G.); (J.R.-P.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, CIMUS, Department of Morphological Sciences, University of Santiago de Compostela, R/ San Francisco s/n, 15782 Santiago de Compostela, Spain
- Correspondence: (J.A.P.); (J.L.L.-G.)
| | - Ana I. Rodriguez-Perez
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.I.R.-P.); (M.G.-G.); (J.R.-P.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, CIMUS, Department of Morphological Sciences, University of Santiago de Compostela, R/ San Francisco s/n, 15782 Santiago de Compostela, Spain
| | - Maria Garcia-Garrote
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.I.R.-P.); (M.G.-G.); (J.R.-P.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, CIMUS, Department of Morphological Sciences, University of Santiago de Compostela, R/ San Francisco s/n, 15782 Santiago de Compostela, Spain
| | - Jannette Rodriguez-Pallares
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.I.R.-P.); (M.G.-G.); (J.R.-P.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, CIMUS, Department of Morphological Sciences, University of Santiago de Compostela, R/ San Francisco s/n, 15782 Santiago de Compostela, Spain
| | - Jose L. Labandeira-Garcia
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.I.R.-P.); (M.G.-G.); (J.R.-P.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, CIMUS, Department of Morphological Sciences, University of Santiago de Compostela, R/ San Francisco s/n, 15782 Santiago de Compostela, Spain
- Correspondence: (J.A.P.); (J.L.L.-G.)
| |
Collapse
|
45
|
Singh A, Yadawa AK, Chaturvedi S, Wahajuddin M, Mishra A, Singh S. Mechanism for antiParkinsonian effect of resveratrol: Involvement of transporters, synaptic proteins, dendrite arborization, biochemical alterations, ER stress and apoptosis. Food Chem Toxicol 2021; 155:112433. [PMID: 34302886 DOI: 10.1016/j.fct.2021.112433] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 07/15/2021] [Accepted: 07/17/2021] [Indexed: 11/21/2022]
Abstract
The present study was undertaken to evaluate the mechanism for antiParkinsonian effect of resveratrol employing 6-hydroxydopamine (6-OHDA) induced experimental model of Parkinson's disease (PD). Resveratrol treatment significantly protects the PD related pathological markers like level of tyrosine hydroxylase, dopamine and apoptotic proteins (Bax and cleaved caspase-3). Disease pathology involves significantly decreased level of dopamine transporter, synaptophysin and postsynaptic density protein 95 (PSD-95) along with augmented level of vesicular monoamine transporter and considerably affected the dendrite arborization. Such affected neuronal communication was significantly restored with resveratrol treatment. Biochemical alterations include the depleted level of glutathione (GSH), mitochondrial complex-I activity with concomitant increased level of lipid peroxidation, nitrite level and calcium levels, which were also significantly inhibited with resveratrol treatment. Altered calcium level induces the endoplasmic reticulum (ER) stress related signalling and phosphorylated Nuclear factor erythroid 2-related factor 2 (Nrf2), and with resveratrol treatment the level of phosphorylated Nrf2 was further increased. The concurrent depleted level of proteasome activity was observed which was attenuated with resveratrol treatment. Proinflammatory cytokines and activated astrocytes were observed which was inhibited with resveratrol treatment. In conclusion, findings suggested that resveratrol exhibits the interference in neuronal communication, oxidative stress, mitochondrial pathophysiology, ER stress, protein degradation mechanism and inflammatory responses and could be utilize in clinics to treat the PD patients.
Collapse
Affiliation(s)
- Ashish Singh
- Division of Toxicology and Experimental Medicine, Central Drug Research Institute, Lucknow, 226031, India
| | - Arun Kumar Yadawa
- Division of Toxicology and Experimental Medicine, Central Drug Research Institute, Lucknow, 226031, India
| | - Swati Chaturvedi
- Division of Pharmacokinetics and Pharmaceutics, Central Drug Research Institute, Lucknow, 226031, India
| | - M Wahajuddin
- Division of Pharmacokinetics and Pharmaceutics, Central Drug Research Institute, Lucknow, 226031, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology, Jodhpur, Rajasthan, 342011, India
| | - Sarika Singh
- Division of Toxicology and Experimental Medicine, Central Drug Research Institute, Lucknow, 226031, India.
| |
Collapse
|
46
|
Mavroeidi P, Xilouri M. Neurons and Glia Interplay in α-Synucleinopathies. Int J Mol Sci 2021; 22:4994. [PMID: 34066733 PMCID: PMC8125822 DOI: 10.3390/ijms22094994] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/29/2021] [Accepted: 05/04/2021] [Indexed: 11/16/2022] Open
Abstract
Accumulation of the neuronal presynaptic protein alpha-synuclein within proteinaceous inclusions represents the key histophathological hallmark of a spectrum of neurodegenerative disorders, referred to by the umbrella term a-synucleinopathies. Even though alpha-synuclein is expressed predominantly in neurons, pathological aggregates of the protein are also found in the glial cells of the brain. In Parkinson's disease and dementia with Lewy bodies, alpha-synuclein accumulates mainly in neurons forming the Lewy bodies and Lewy neurites, whereas in multiple system atrophy, the protein aggregates mostly in the glial cytoplasmic inclusions within oligodendrocytes. In addition, astrogliosis and microgliosis are found in the synucleinopathy brains, whereas both astrocytes and microglia internalize alpha-synuclein and contribute to the spread of pathology. The mechanisms underlying the pathological accumulation of alpha-synuclein in glial cells that under physiological conditions express low to non-detectable levels of the protein are an area of intense research. Undoubtedly, the presence of aggregated alpha-synuclein can disrupt glial function in general and can contribute to neurodegeneration through numerous pathways. Herein, we summarize the current knowledge on the role of alpha-synuclein in both neurons and glia, highlighting the contribution of the neuron-glia connectome in the disease initiation and progression, which may represent potential therapeutic target for a-synucleinopathies.
Collapse
Affiliation(s)
| | - Maria Xilouri
- Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece;
| |
Collapse
|
47
|
Mogroside V Alleviates Lipopolysaccharide-Induced Neuroinflammation via Inhibition of TLR4-MyD88 and Activation of AKT/AMPK-Nrf2 Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:5521519. [PMID: 34012471 PMCID: PMC8105091 DOI: 10.1155/2021/5521519] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/19/2021] [Accepted: 03/30/2021] [Indexed: 02/08/2023]
Abstract
As innate immune effector cells in the central nervous system (CNS), microglia not only are essential for the normal development of nervous system but also act on different neurological diseases, including Alzheimer's disease (AD), Huntington's disease (HD), and other neuroinflammatory diseases. Mogroside V (Mog), a natural plant active ingredient and isolated form of Momordica grosvenori, has been shown to possess anti-inflammatory action, but few studies were carried out to investigate the effects of Mog on neuroinflammation. This study aimed to investigate the role of Mog in lipopolysaccharide- (LPS-) induced neuroinflammation and neuronal damage, revealing the underlying mechanisms. Our data indicated that Mog significantly inhibited the LPS-induced production of proinflammatory factors, such as tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), IL-18, IL-6, cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), and high mobility group box 1 (HMGB1) in BV-2 cells. We found that Mog also suppressed toll-like receptor 4 (TLR4), myeloid differentiation factor 88 (MyD88), the phosphorylation of mitogen-activated protein kinases (MAPKs), adenosine 5'-monophosphate- (AMP-) activated protein kinase (AMPK), nuclear factor kappa-B (NF-κB), and protein kinase B (AKT). Moreover, Mog also enhanced the expression of γ-glutamyl cysteine synthetase catalytic subunit (GCLC), modifier subunit (GCLM), heme oxygenase-1 (HO-1), and quinine oxidoreductase 1 (NQO1) proteins, mostly depending on the nuclear translation of nuclear factor erythroid-2 related factor 2 (Nrf2). In contrast, pretreatment with inhibitors of AKT can suppress the phosphorylation of AMPK, Nrf2, and its downstream proteins expression. In summary, Mog might play a protective role against LPS-induced neurotoxicity by inhibiting the TLR4-MyD88 and activation of AMPK/AKT-Nrf2 signaling pathway.
Collapse
|