1
|
Chakrabarti A, Verma S. Identifying potential genes driving ferroptosis in the substantia nigra and dopaminergic neurons in Parkinson's disease. Mol Cell Neurosci 2025; 132:103993. [PMID: 39848553 DOI: 10.1016/j.mcn.2025.103993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/07/2025] [Accepted: 01/16/2025] [Indexed: 01/25/2025] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder marked by dopaminergic (DA) neuron degeneration in the substantia nigra (SN). Conventional dopamine replacement therapies provide limited long-term efficacy and significant side effects. Emerging evidence suggests ferroptosis-a form of cell death driven by iron-dependent lipid peroxidation-contributes to PD pathology, though direct evidence linking dysregulation of ferroptosis-related genes in DA neuron loss in PD remains limited. This study explores the expression of ferroptosis-associated genes in the SN and DA neurons of PD patients, identifying potential therapeutic targets. We analyzed two independent RNA-seq datasets, GSE7621 and GSE8397 (GPL-96), from the GEO database to identify common differentially expressed ferroptosis-related genes in the SN of PD patients. We also conducted Gene Ontology and pathway enrichment analyses of these genes to explore the underlying mechanisms and constructed a protein-protein interaction network. The findings were further validated using an additional dataset, GSE49036. We further explored the dysregulation of these ferroptosis-related genes in DA neurons using RNA-seq data GSE169755, derived from DA neurons isolated from the SN of PD patients and controls. Lastly, the proposed hypothesis was experimentally validated in an in vitro PD model. This comprehensive multi-dataset analysis uncovers novel insights into the expression of ferroptosis-related genes in PD, suggesting potential biomarkers and therapeutic targets for mitigating DA neuron loss and PD progression.
Collapse
Affiliation(s)
- Ardra Chakrabarti
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sonia Verma
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
2
|
Buneeva O, Medvedev A. Monoamine Oxidase Inhibitors in Toxic Models of Parkinsonism. Int J Mol Sci 2025; 26:1248. [PMID: 39941014 PMCID: PMC11818313 DOI: 10.3390/ijms26031248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/10/2025] [Accepted: 01/12/2025] [Indexed: 02/16/2025] Open
Abstract
Monoamine oxidase inhibitors are widely used for the symptomatic treatment of Parkinson's disease (PD). They demonstrate antiparkinsonian activity in different toxin-based models induced by 6-hydroxydopamine, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), and pesticides (rotenone and paraquat). In some models, such as MPTP-induced PD, MAO inhibitors prevent the formation of the neurotoxin MPP+ from the protoxin MPTP. Regardless of the toxin's nature, potent MAO inhibitors prevent dopamine loss reduction, the formation of hydrogen peroxide, hydrogen peroxide signaling, and the accumulation of hydrogen peroxide-derived reactive oxygen species responsible for the development of oxidative stress. It becomes increasingly clear that some metabolites of MAO inhibitors (e.g., the rasagiline metabolite 1-R-aminoindan) possess their own bio-pharmacological activities unrelated to the parent compound. In addition, various MAO inhibitors exhibit multitarget action, in which MAO-independent effects prevail. This opens new prospects in the development of novel therapeutics based on simultaneous actions on several prospective targets for the therapy of PD.
Collapse
Affiliation(s)
| | - Alexei Medvedev
- Institute of Biomedical Chemistry, 10 Pogodinskaya Street, 119121 Moscow, Russia;
| |
Collapse
|
3
|
Goudarzi S, Mohammad Jafari R, Farsiu N, Amini B, Manavi MA, Fahanik-Babaei J, Ejtemaei-Mehr S, Dehpour AR. Protective effects of licofelone on scopolamine-induced spatial learning and memory impairment by enhancing parkin-dependent mitophagy and promotion of neural regeneration and in adult mice. Eur J Pharmacol 2024; 984:177025. [PMID: 39395583 DOI: 10.1016/j.ejphar.2024.177025] [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: 05/06/2024] [Revised: 10/01/2024] [Accepted: 10/01/2024] [Indexed: 10/14/2024]
Abstract
Inhibition of COX and LOX could contribute to memory formation and prevention of neurodegeneration, by alleviation of neuroinflammation and improvement of mitochondrial homeostasis. We aimed to assess the effect of licofelone, a dual COX and 5-LOX inhibitor on memory formation, neural apoptosis, neural regeneration, and mitophagy in acute and chronic dosages, given that licofelone could regulate nitric oxide levels. Y-maze and Passive Avoidance tests were used to evaluate memory function in NMRI mice using the EthoVision setting, following scopolamine administration (1 mg/kg, i.p.) as an acute amnestic drug. Hippocampi were used to evaluate the levels of apoptosis via TUNEL assay, neural regeneration via immunohistochemistry method detecting doublecortin and nestin, and mitophagy via Western blot of mitophagy proteins Parkin and ATG5. While acute high-dose licofelone (20 mg/kg) could reverse amnestic effects of scopolamine in passive avoidance test (p = 0.0001), Chronic licofelone (10 mg/kg for 10 consecutive days) could improve performance in Y-maze (p = 0.0007). Molecular analysis revealed that the chronic form of the drug could enhance neural regeneration in CA1 and SGZ regions, reset mitophagy levels as much as the healthy state, and reduce apoptosis rate. Licofelone appears to show a desirable anti-amnestic profile in a low dose chronically; it is hence recommended for future clinical studies on the prevention of neuroinflammation and memory deficit.
Collapse
Affiliation(s)
- Sepideh Goudarzi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Razieh Mohammad Jafari
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Nikou Farsiu
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behnam Amini
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Manavi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Javad Fahanik-Babaei
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahram Ejtemaei-Mehr
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Liu J, Chen L, Sun Z, Tao Z, Pavel V, Li Y, Wang F, Cui W, Liu S. Unidirectional gene delivery electrospun fibrous membrane via charge repulsion for tendon repair. Bioact Mater 2024; 37:191-205. [PMID: 38549775 PMCID: PMC10972767 DOI: 10.1016/j.bioactmat.2024.03.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 03/01/2024] [Accepted: 03/07/2024] [Indexed: 11/12/2024] Open
Abstract
Gene therapy is capable of efficiently regulating the expression of abnormal genes in diseased tissues and expected to be a therapeutic option for refractory diseases. However, unidirectional targeting gene therapy is always desired at the tissue interface. In this study, inspired by the principle that like charges repulse each other, a positively charged micro-nano electrospun fibrous membrane with dual-layer structure was developed by electrospinning technology to achieve unidirectional delivery of siRNA-loaded cationic nanocarriers, thus realizing unidirectional gene therapy at the tendon-paratenon interface. Under the charge repulsion of positively charged layer, more cationic COX-2 siRNA nanocarriers were enriched in peritendinous tissue, which not only improved the bioavailability of the gene drug to prevent the peritendinous adhesion formation, but also avoided adverse effects on the fragile endogenous healing of tendon itself. In summary, this study provides an innovative strategy for unidirectional targeting gene therapy of tissue interface diseases by utilizing charge repulsion to facilitate unidirectional delivery of gene drugs.
Collapse
Affiliation(s)
- Jingwen Liu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China
| | - Liang Chen
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Zhenyu Sun
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China
| | - Zaijin Tao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China
| | - Volotovski Pavel
- Republican Scientific and Practical Center of Traumatology and Orthopedics, Belarusian State Medical University, Minsk 220024, Belarus
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Fei Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Shen Liu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China
| |
Collapse
|
5
|
Xu Y, Jia B, Li J, Li Q, Luo C. The Interplay between Ferroptosis and Neuroinflammation in Central Neurological Disorders. Antioxidants (Basel) 2024; 13:395. [PMID: 38671843 PMCID: PMC11047682 DOI: 10.3390/antiox13040395] [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: 03/11/2024] [Revised: 03/23/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Central neurological disorders are significant contributors to morbidity, mortality, and long-term disability globally in modern society. These encompass neurodegenerative diseases, ischemic brain diseases, traumatic brain injury, epilepsy, depression, and more. The involved pathogenesis is notably intricate and diverse. Ferroptosis and neuroinflammation play pivotal roles in elucidating the causes of cognitive impairment stemming from these diseases. Given the concurrent occurrence of ferroptosis and neuroinflammation due to metabolic shifts such as iron and ROS, as well as their critical roles in central nervous disorders, the investigation into the co-regulatory mechanism of ferroptosis and neuroinflammation has emerged as a prominent area of research. This paper delves into the mechanisms of ferroptosis and neuroinflammation in central nervous disorders, along with their interrelationship. It specifically emphasizes the core molecules within the shared pathways governing ferroptosis and neuroinflammation, including SIRT1, Nrf2, NF-κB, Cox-2, iNOS/NO·, and how different immune cells and structures contribute to cognitive dysfunction through these mechanisms. Researchers' findings suggest that ferroptosis and neuroinflammation mutually promote each other and may represent key factors in the progression of central neurological disorders. A deeper comprehension of the common pathway between cellular ferroptosis and neuroinflammation holds promise for improving symptoms and prognosis related to central neurological disorders.
Collapse
Affiliation(s)
- Yejia Xu
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China
- Hebei Key Laboratory of Forensic Medicine, College of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Bowen Jia
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China
| | - Jing Li
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China
| | - Qianqian Li
- NHC Key Laboratory of Drug Addiction Medicine, Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming 650500, China
- School of Forensic Medicine, Wannan Medical College, Wuhu 241002, China
| | - Chengliang Luo
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China
- Hebei Key Laboratory of Forensic Medicine, College of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, China
- NHC Key Laboratory of Drug Addiction Medicine, Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming 650500, China
| |
Collapse
|
6
|
Shen Y, Li X, Wang H, Wang Y, Tao L, Wang P, Zhang H. Bisphenol A induced neuronal apoptosis and enhanced autophagy in vitro through Nrf2/HO-1 and Akt/mTOR pathways. Toxicology 2023; 500:153678. [PMID: 38006930 DOI: 10.1016/j.tox.2023.153678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/10/2023] [Accepted: 11/18/2023] [Indexed: 11/27/2023]
Abstract
Bisphenol A (BPA) was traditionally used in epoxy resins and polycarbonate plastics, but it was found to be harmful to human health due to its endocrine-disrupting effects. It can affect various biological functions of human beings and interfere with brain development. However, the neurotoxic mechanisms of BPA on brain development and associated neurodegeneration remain poorly understood. Here, we reported that BPA (100, 250, 500 μM) inhibited cell viability of neural cells PC12, SH-SY5Y and caused dose-dependent cell death. In addition, BPA exposure increased intracellular reactive oxygen species (ROS) and mitochondrial ROS (mtROS) levels, decreased mitochondrial membrane potential, reduced the expression of cytochrome c oxidase IV (COX4), downregulated Bcl-2, and initiated apoptosis. Moreover, BPA treatment resulted in the accumulation of intracellular acidic vacuoles and increased the autophagy marker LC3 II to LC3 I ratio. Furthermore, BPA exposure inhibited Nrf2/ HO-1 and AKT/mTOR pathways and mediated cellular oxidative stress, apoptosis, and excessive autophagy, leading to neuronal degeneration. The interactions between oxidative stress, autophagy, and apoptosis during BPA-induced neurotoxicity remain unclear and require further in vivo confirmation.
Collapse
Affiliation(s)
- Yue Shen
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Xinying Li
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Hongyan Wang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Yicheng Wang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Liqing Tao
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China; School of Life Sciences, Shaoxing University, Shaoxing, Zhejiang, China
| | - Pingping Wang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Heng Zhang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China; School of Life Sciences, Shaoxing University, Shaoxing, Zhejiang, China.
| |
Collapse
|
7
|
Vell MS, Krishnan A, Wangensteen K, Serper M, Seeling KS, Hehl L, Rendel MD, Zandvakili I, Vujkovic M, Scorletti E, Creasy KT, Trautwein C, Rader DJ, Alqahtani S, Schneider KM, Schneider CV. Aspirin is associated with a reduced incidence of liver disease in men. Hepatol Commun 2023; 7:e0268. [PMID: 37708453 PMCID: PMC10503677 DOI: 10.1097/hc9.0000000000000268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 07/31/2023] [Indexed: 09/16/2023] Open
Abstract
BACKGROUND The hepatoprotective effects of aspirin have been observed in individuals with viral hepatitis; however, its impact on the general population remains uncertain. Understanding the association between aspirin use and the development of liver diseases is crucial for optimizing preventive strategies. METHODS We identified individuals with aspirin use in the UK Biobank and the Penn Medicine Biobank, as well as propensity-score-matched controls. Outcome measures included new liver disease development, diagnosed by MRI or "International Classification of Diseases and Related Health Problems" coding, and incidences of gastrointestinal bleeding and ulcers. RESULTS In the UK Biobank cohort, regular aspirin use was associated with an 11.2% reduction in the risk of developing new liver diseases during the average 11.84 ± 2.01-year follow-up period (HR=0.888, 95% CI = 0.819-0.963; p = 4.1 × 10-3). Notably, the risk of metabolic dysfunction-associated steatotic liver disease (ICD-10 K76.0) and MRI-diagnosed steatosis was significantly lower among aspirin users (HR = 0.882-0.911), whereas no increased risk of gastrointestinal bleeding or ulcers was observed. These findings were replicated in the Penn Medicine Biobank cohort, in which the protective effect of aspirin appeared to be dependent on the duration of intake. The greatest risk reduction for new liver disease development was observed after at least 1 year of aspirin use (HR = 0.569, 95% CI = 0.425-0.762; p = 1.6 × 10-4). Intriguingly, when considering general risk factors, only men exhibited a lower risk of MRI-confirmed or ICD-coded steatosis with aspirin use (HRs = 0.806-0.906), while no significant protective effect of aspirin was observed in females. CONCLUSION This cohort study demonstrated that regular aspirin use was associated with a reduced risk of liver disease in men without an elevated risk of gastrointestinal bleeding or ulcers. Further investigation is warranted to elucidate potential sex-related differences in the effects of aspirin and to inform tailored preventive strategies for liver diseases.
Collapse
Affiliation(s)
- Mara Sophie Vell
- Department of Medicine III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Arunkumar Krishnan
- Section of Gastroenterology and Hepatology, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| | - Kirk Wangensteen
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Marina Serper
- Department of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Katharina Sophie Seeling
- Department of Medicine III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Leonida Hehl
- Department of Medicine III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Miriam Daphne Rendel
- Department of Medicine III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Inuk Zandvakili
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Marijana Vujkovic
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Eleonora Scorletti
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kate Townsend Creasy
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christian Trautwein
- Department of Medicine III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Daniel James Rader
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Saleh Alqahtani
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Liver Transplant Center, King Faisal Specialist Hospital & Research Center, Riyadh
| | - Kai Markus Schneider
- Department of Medicine III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Carolin Victoria Schneider
- Department of Medicine III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- The Institute for Translational Medicine and Therapeutics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
8
|
Liu X, Yang L, Zhang G, Ling J. Neuroprotective Effects of Phenolic Antioxidant Tert-butylhydroquinone (tBHQ) in Brain Diseases. Mol Neurobiol 2023; 60:4909-4923. [PMID: 37191855 DOI: 10.1007/s12035-023-03370-3] [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: 10/18/2022] [Accepted: 04/25/2023] [Indexed: 05/17/2023]
Abstract
Human life and health are gravely threatened by brain diseases. The onset and progression of the illnesses are influenced by a variety of factors, including pathogenic causes, environmental factors, mental issues, etc. According to scientific studies, neuroinflammation and oxidative stress play a significant role in the development and incidence of brain diseases by producing pro-inflammatory cytokines and oxidative tissue damage to induce inflammation and apoptosis. Neuroinflammation, oxidative stress, and oxidative stress-related changes are inseparable factors in the etiology of several brain diseases. Numerous neurodegenerative diseases have undergone substantial research into the therapeutic alternatives that target oxidative stress, the function of oxidative stress, and the possible therapeutic use of antioxidants. Formerly, tBHQ is a synthetic phenolic antioxidant, which has been widely used as a food additive. According to recent researches, tBHQ can suppress the processes that lead to neuroinflammation and oxidative stress, which offers a fresh approach to treating brain diseases. In order to achieve the goal of decreasing inflammation and apoptosis, tBHQ is a specialized nuclear factor erythroid 2-related factor (Nrf2) activator that decreases oxidative stress and enhances antioxidant status by upregulating the Nrf2 gene and reducing nuclear factor kappa-B (NF-κB) activity. This article reviews the effects of tBHQ on neuroinflammation and oxidative stress in recent years and looks into how tBHQ inhibits neuroinflammation and oxidative stress through human, animal, and cell experiments to play a neuroprotective role in Alzheimer's disease (AD), stroke, depression, and Parkinson's disease (PD). It is anticipated that this article will be useful as a reference for upcoming research and the creation of drugs to treat brain diseases.
Collapse
Affiliation(s)
- Xiaojin Liu
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Department of Pharmacy, Shandong Medical College, Linyi, 276000, China
| | - Luodan Yang
- College of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Guoying Zhang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Jianya Ling
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China.
| |
Collapse
|
9
|
Zhang R, Teng L, Zhong Y, Ma P, Xu L, Xiao P. Neuroprotection of isookanin against MPTP-induced cell death of SH-SY5Y cells via BCL2/BAX and PI3K/AKT pathways. Psychopharmacology (Berl) 2023:10.1007/s00213-023-06383-x. [PMID: 37256377 DOI: 10.1007/s00213-023-06383-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 05/07/2023] [Indexed: 06/01/2023]
Abstract
BACKGROUND AND PURPOSE Isookanin, an important antioxidant component in Coreopsis tinctoria Nutt., has shown remarkable hypolipidemic, hypoglycemic, and hypotensive effects. However, the neuroprotective effect of isookanin has not been reported yet. Here, the neuroprotective effects and relevant molecular mechanisms of isookanin are explored for the first time. METHODS The SH-SY5Y cells were exposed to neurotoxic H2O2, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), and Aβ25-35, respectively. Cell viability and apoptosis were evaluated by MTT, lactate dehydrogenase (LDH), and TUNEL assays. Intercellular ROS and mitochondrial membrane potential were assessed by DCFH-DA and JC-1 assay. Western blot and qRT-PCR were used to explore the perturbed signaling at the gene and protein levels. Molecular docking analysis and in vitro assay were further applied to confirm potential target. RESULTS Among the three in vitro models, isookanin showed the best neuroprotection against MPTP-induced damage. Isookanin attenuated the levels of LDH, intracellular ROS, and mitochondrial membrane potential. Isookanin upregulated phosphorylation of AKT and PI3K, and increased BCL2/BAX ratio. Isookanin possessed a powerful affinity toward AKT. Besides, the protective effects of isookanin disappeared when cells were co-treated with an AKT inhibitor (AZD5363). CONCLUSION Isookanin regulated BCL2/BAX and PI3K/AKT pathways to reduce mitochondrial damage and cellular apoptosis. Isookanin may be a new protector for neurodegenerative diseases.
Collapse
Affiliation(s)
- Rong Zhang
- Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151 Malianwa North Road, Haidian District, 100193, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, 100193, Beijing, China
| | - Lili Teng
- Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151 Malianwa North Road, Haidian District, 100193, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, 100193, Beijing, China
| | - Yi Zhong
- Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151 Malianwa North Road, Haidian District, 100193, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, 100193, Beijing, China
| | - Pei Ma
- Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151 Malianwa North Road, Haidian District, 100193, Beijing, China.
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, 100193, Beijing, China.
| | - Lijia Xu
- Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151 Malianwa North Road, Haidian District, 100193, Beijing, China.
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, 100193, Beijing, China.
| | - Peigen Xiao
- Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151 Malianwa North Road, Haidian District, 100193, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, 100193, Beijing, China
| |
Collapse
|
10
|
Montero-Cosme TG, Pascual-Mathey LI, Hernández-Aguilar ME, Herrera-Covarrubias D, Rojas-Durán F, Aranda-Abreu GE. Potential drugs for the treatment of Alzheimer's disease. Pharmacol Rep 2023; 75:544-559. [PMID: 37005970 DOI: 10.1007/s43440-023-00481-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/24/2023] [Accepted: 03/26/2023] [Indexed: 04/04/2023]
Abstract
It is well known that amyloid precursor protein (APP), the enzyme β-secretase 1 (BACE1), cyclooxygenase 2 (COX-2), nicastrin (NCT), and hyperphosphorylated tau protein (p-tau) are closely related to the development of Alzheimer's disease (AD). In addition, recent evidence shows that neuroinflammation also contributes to the pathogenesis of AD. Although the mechanism is not clearly known, such inflammation could alter the activity of the aforementioned molecules. Therefore, the use of anti-inflammatory agents could slow the progression of the disease. Nimesulide, resveratrol, and citalopram are three anti-inflammatory agents that could contribute to a decrease in neuroinflammation and consequently to a decrease in the overexpression of APP, BACE1, COX-2, NCT, and p-Tau, as they possess anti-inflammatory effects that could regulate the expression of APP, BACE1, COX-2, NCT, and p-Tau of potent pro-inflammatory markers indirectly involved in the expression of APP, BACE1, NCT, COX-2, and p-Tau; therefore, their use could be beneficial as preventive treatment as well as in the early stages of AD.
Collapse
Affiliation(s)
| | | | | | | | - Fausto Rojas-Durán
- Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa, Veracruz, México
| | | |
Collapse
|
11
|
Wang J, Meng X, Chen K, Feng J. An autophagy-related four-lncRNA signature helps to predict progression-free survival of neuroblastoma patients. Front Oncol 2022; 12:1014845. [PMID: 36530992 PMCID: PMC9753905 DOI: 10.3389/fonc.2022.1014845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/14/2022] [Indexed: 09/14/2023] Open
Abstract
BACKGROUND This study aimed to identify autophagy-related long non-coding RNAs (lncRNAs) associated with progression of neuroblastoma (NB), and to build an autophagy-related lncRNA signature that helps to predict progression-free survival (PFS) of NB. METHODS Three independent gene expression datasets were utilized in this study. Autophagy-related genes (ARG) associated with PFS of NB patients were firstly identified by univariate Cox survival analysis. lncRNAs correlated with those PFS-related ARGs were then identified. The least absolute shrinkage and selection operator (LASSO) regression and multivariate Cox regression analyses were performed to select out those lncRNAs with the best prognostic value for PFS. The Receiver Operating Characteristic (ROC) and Area Under Curve (AUC) analyses were performed to assess the prediction accuracy. RESULTS Four autophagy-related lncRNAs (AL356599.1, AC022075.1, AC020928.1 and LINC02076) were found to be with the best prognostic value and integrated into a four-lncRNA risk signature for predicting PFS of NB patients. The four-lncRNA signature significantly stratify NB patients into two risk groups, with high-risk group has significantly poorer PFS than the low-risk group. The prognostic role of the lncRNA signature was independent with other clinical risk factors. The ROC curves revealed that the lncRNA signature has a good performance in predicting PFS (AUC > 0.70). A nomogram based on COG (Children's Oncology Group) risk and the lncRNA risk score was constructed, showing good prediction accuracy (C-index = 0.700). The prognostic ability of the nomogram was better than that of COG risk alone (AUC = 0.790 versus AUC = 0.748). GSEA analyses revealed that multiple autophagy-related gene sets are significantly enriched in the low-risk group. CONCLUSIONS We identified an autophagy-related four-lncRNA signature that could help to predict the PFS of NB patients. Autophagy-related gene sets are significantly enriched in low-risk group, suggesting tumor suppressive roles of autophagy in NB.
Collapse
Affiliation(s)
| | | | | | - Jiexiong Feng
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
12
|
Naoi M, Maruyama W, Shamoto-Nagai M. Neuroprotective Function of Rasagiline and Selegiline, Inhibitors of Type B Monoamine Oxidase, and Role of Monoamine Oxidases in Synucleinopathies. Int J Mol Sci 2022; 23:ijms231911059. [PMID: 36232361 PMCID: PMC9570229 DOI: 10.3390/ijms231911059] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/09/2022] [Accepted: 09/14/2022] [Indexed: 11/27/2022] Open
Abstract
Synucleinopathies are a group of neurodegenerative disorders caused by the accumulation of toxic species of α-synuclein. The common clinical features are chronic progressive decline of motor, cognitive, behavioral, and autonomic functions. They include Parkinson’s disease, dementia with Lewy body, and multiple system atrophy. Their etiology has not been clarified and multiple pathogenic factors include oxidative stress, mitochondrial dysfunction, impaired protein degradation systems, and neuroinflammation. Current available therapy cannot prevent progressive neurodegeneration and “disease-modifying or neuroprotective” therapy has been proposed. This paper presents the molecular mechanisms of neuroprotection by the inhibitors of type B monoamine oxidase, rasagiline and selegiline. They prevent mitochondrial apoptosis, induce anti-apoptotic Bcl-2 protein family, and pro-survival brain- and glial cell line-derived neurotrophic factors. They also prevent toxic oligomerization and aggregation of α-synuclein. Monoamine oxidase is involved in neurodegeneration and neuroprotection, independently of the catalytic activity. Type A monoamine oxidases mediates rasagiline-activated signaling pathways to induce neuroprotective genes in neuronal cells. Multi-targeting propargylamine derivatives have been developed for therapy in various neurodegenerative diseases. Preclinical studies have presented neuroprotection of rasagiline and selegiline, but beneficial effects have been scarcely presented. Strategy to improve clinical trials is discussed to achieve disease-modification in synucleinopathies.
Collapse
Affiliation(s)
- Makoto Naoi
- Correspondence: ; Tel.: +81-05-6173-1111 (ext. 3494); Fax: +81-561-731-142
| | | | | |
Collapse
|
13
|
COX-2/sEH Dual Inhibitor Alleviates Hepatocyte Senescence in NAFLD Mice by Restoring Autophagy through Sirt1/PI3K/AKT/mTOR. Int J Mol Sci 2022; 23:ijms23158267. [PMID: 35897843 PMCID: PMC9332821 DOI: 10.3390/ijms23158267] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/20/2022] [Accepted: 07/25/2022] [Indexed: 02/07/2023] Open
Abstract
We previously found that the disorder of soluble epoxide hydrolase (sEH)/cyclooxygenase-2 (COX-2)-mediated arachidonic acid (ARA) metabolism contributes to the pathogenesis of the non-alcoholic fatty liver disease (NAFLD) in mice. However, the exact mechanism has not been elucidated. Accumulating evidence points to the essential role of cellular senescence in NAFLD. Herein, we investigated whether restoring the balance of sEH/COX-2-mediated ARA metabolism attenuated NAFLD via hepatocyte senescence. A promised dual inhibitor of sEH and COX-2, PTUPB, was used in our study to restore the balance of sEH/COX-2-mediated ARA metabolism. In vivo, NAFLD was induced by a high-fat diet (HFD) using C57BL/6J mice. In vitro, mouse hepatocytes (AML12) and mouse hepatic astrocytes (JS1) were used to investigate the effects of PTUPB on palmitic acid (PA)-induced hepatocyte senescence and its mechanism. PTUPB alleviated liver injury, decreased collagen and lipid accumulation, restored glucose tolerance, and reduced hepatic triglyceride levels in HFD-induced NAFLD mice. Importantly, PTUPB significantly reduced the expression of liver senescence-related molecules p16, p53, and p21 in HFD mice. In vitro, the protein levels of γH2AX, p53, p21, COX-2, and sEH were increased in AML12 hepatocytes treated with PA, while Ki67 and PCNA were significantly decreased. PTUPB decreased the lipid content, the number of β-gal positive cells, and the expression of p53, p21, and γH2AX proteins in AML12 cells. Meanwhile, PTUPB reduced the activation of hepatic astrocytes JS1 by slowing the senescence of AML12 cells in a co-culture system. It was further observed that PTUPB enhanced the ratio of autophagy-related protein LC3II/I in AML12 cells, up-regulated the expression of Fundc1 protein, reduced p62 protein, and suppressed hepatocyte senescence. In addition, PTUPB enhanced hepatocyte autophagy by inhibiting the PI3K/AKT/mTOR pathway through Sirt1, contributing to the suppression of senescence. PTUPB inhibits the PI3K/AKT/mTOR pathway through Sirt1, improves autophagy, slows down the senescence of hepatocytes, and alleviates NAFLD.
Collapse
|
14
|
Zhang L, Li M, Wang W, Yu W, Liu H, Wang K, Chang M, Deng C, Ji Y, Shen Y, Qi L, Sun H. Celecoxib alleviates denervation-induced muscle atrophy by suppressing inflammation and oxidative stress and improving microcirculation. Biochem Pharmacol 2022; 203:115186. [PMID: 35882305 DOI: 10.1016/j.bcp.2022.115186] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/16/2022] [Accepted: 07/19/2022] [Indexed: 11/25/2022]
Abstract
The molecular mechanism underlying denervation-induced muscle atrophy is complex and incompletely understood. Our previous results suggested that inflammation may play an important role in the early stages of muscle atrophy. Celecoxib is reported to exert anti-inflammatory effects. Here, we explored the effect of celecoxib on denervation-induced muscle atrophy and sought to identify the mechanism involved. We found that celecoxib treatment significantly increased the wet weight ratio and CSA of the tibialisanteriormuscle. Additionally, celecoxib downregulated the levels of COX-2, inflammatory factors and reduced inflammatory cell infiltration. GO and KEGG pathway enrichment analysis indicated that after 3 days of celecoxib treatment in vivo, the differentially expressed genes (DEGs) were mainly associated with the regulation of immune responses related to complement activation; after 14 days, the DEGs were mainly involved in the regulation of oxidative stress and inflammation-related responses. Celecoxib administration reduced the levels of ROS and oxidative stress-related proteins. Furthermore, we found that celecoxib treatment inhibited the denervation-induced up-regulation of the ubiquitin-proteasome and autophagy-lysosomal systems related proteins; decreased mitophagy in target muscles; and increased levels of MHC. Finally, celecoxib also attenuated microvascular damage in denervated skeletal muscle. Combined, our findings demonstrated that celecoxib inhibits inflammation and oxidative stress in denervated skeletal muscle, thereby suppressing mitophagy and proteolysis, improving blood flow in target muscles, and, ultimately, alleviating denervation-induced muscle atrophy. Our results confirmed that inflammatory responses play a key role in denervation-induced muscle atrophy and highlight a novel strategy for the prevention and treatment of this condition.
Collapse
Affiliation(s)
- Lilei Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu Province 226001, P. R. China
| | - Ming Li
- Department of Laboratory Medicine, Department of Endocrinology, Binhai County People's Hospital affiliated to Kangda College of Nanjing Medical University, Yancheng, Jiangsu Province 224500, P. R. China
| | - Wei Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu Province 226001, P. R. China; Department of Pathology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, P. R. China
| | - Weiran Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu Province 226001, P. R. China
| | - Hua Liu
- Department of Orthopedics, Haian Hospital of Traditional Chinese Medicine, 55 Ninghai Middle Road, Haian, Nantong, Jiangsu Province 226600, P. R. China
| | - Kexin Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu Province 226001, P. R. China
| | - Mengyuan Chang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu Province 226001, P. R. China
| | - Chunyan Deng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu Province 226001, P. R. China
| | - Yanan Ji
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu Province 226001, P. R. China
| | - Yuntian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu Province 226001, P. R. China.
| | - Lei Qi
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province 226001, P. R. China.
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu Province 226001, P. R. China.
| |
Collapse
|
15
|
Subkorn P, Norkaew C, Deesrisak K, Tanyong D. Punicalagin, a pomegranate compound, induces apoptosis and autophagy in acute leukemia. PeerJ 2021; 9:e12303. [PMID: 34760363 PMCID: PMC8570173 DOI: 10.7717/peerj.12303] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 09/22/2021] [Indexed: 12/14/2022] Open
Abstract
Background Punicalagin is the major phenolic compound found in pomegranate peels. It has several reported medical benefits, including antioxidant, anti-inflammatory, and anticancer properties. The present study investigated the anti-leukemic effects and the molecular mechanism of punicalagin on NB4 and MOLT-4 leukemic cell lines. Methods Leukemic cells were treated with punicalagin and cell viability was determined using MTS assay. Apoptosis and autophagy were analyzed by flow cytometry using Annexin V-FITC/PI and anti-LC3/FITC antibodies staining, respectively. Apoptotic and autophagic mRNA expression were determined using reverse transcription-quantitative PCR. STITCH bioinformatics tools were used to predict the interaction between punicalagin and its proposed target proteins. Results Results indicated that punicalagin decreased NB4 and MOLT-4 cell viability in a dose-dependent manner. Punicalagin, in combination with daunorubicin, exhibited synergistic cytotoxic effects. Punicalagin induced apoptosis through the upregulation of caspase-3/-8/-9, Bax and the downregulation of Bcl-2 expression. Punicalagin also promoted autophagy via the downregulation of mTOR and the upregulation of ULK1 expression. Cyclooxygenase-2 and toll-like receptor 4 were found to be involved in punicalagin-induced cell death in punicalagin-targeted protein interactions. Conclusions These results suggest that punicalagin exerts cytotoxic activities by suppressing proliferation and promoting apoptosis and autophagy by activating the caspase cascade, altering Bax and Bcl-2, and regulating autophagy via mTOR/ULK1 signaling.
Collapse
Affiliation(s)
- Paweena Subkorn
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| | - Chosita Norkaew
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| | - Kamolchanok Deesrisak
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| | - Dalina Tanyong
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| |
Collapse
|
16
|
Zhang J, Liu L, Zhang L, Chen S, Chen Y, Cai C. Targeted fatty acid metabolomics to discover Parkinson's disease associated metabolic alteration. JOURNAL OF MASS SPECTROMETRY : JMS 2021; 56:e4781. [PMID: 34523199 DOI: 10.1002/jms.4781] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/31/2021] [Accepted: 08/07/2021] [Indexed: 06/13/2023]
Abstract
The pathogenesis of Parkinson's disease (PD) remains to be elucidated, and the metabolomics analysis has the potential to identify metabolic profiles that are involved in PD pathogenesis. Here we applied a target metabolomics approach to measure the plasma levels of 158 fatty acid metabolites in a discovery cohort including 42 PD patients and 54 health volunteers, and found two upregulated (arachidonic acid and 13-hydroxy-octadecatrienoic acid) and eleven down-regulated (docosahexaenoic acid, lyso-platelet-activating factor, 12-hydroxy-eicosatetraenoic acid, dihydroxy-eicosatrienoic acids, dihidroxy-octadecenoic acids, 17,18-dihydroxy-eicosatetraenoic acid, and hydroperoxy-octadecadienoic acids) metabolites as primary candidate marker of PD. A support vector machine algorithm with primary candidate marker was used in an independent validation cohort to identify PD. Arachidonic acid and 13-hydroxy-octadecatrienoic acid were evaluated as an effective tool in that area under the receiver operating characteristic curve reached 0.995 and 0.912 in the validation set for diagnosing PD from healthy volunteers. Besides, the sensitivity and specificity of arachidonic acid as diagnostic factor of PD in validation set were 100% and 94.10%. Similarly, the sensitivity and specificity of 13-hydroxy-octadecatrienoic acid were 100% and 82.40% for identifying PD. This target fatty acid metabolomics demonstrated a series of plasma fatty acid metabolite as PD candidate marker with high efficiency and provided insights into the understanding of PD metabolic regulation.
Collapse
Affiliation(s)
- Junjie Zhang
- Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, China
- School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Lulu Liu
- Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, China
- School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Lijiang Zhang
- Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, China
- School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Simei Chen
- Neurology Department, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Yusen Chen
- Neurology Department, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Chun Cai
- Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, China
- School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
17
|
Wang S, Ma J, Zeng Y, Zhou G, Wang Y, Zhou W, Sun X, Wu M. Icariin, an Up-and-Coming Bioactive Compound Against Neurological Diseases: Network Pharmacology-Based Study and Literature Review. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:3619-3641. [PMID: 34447243 PMCID: PMC8384151 DOI: 10.2147/dddt.s310686] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 06/25/2021] [Indexed: 12/12/2022]
Abstract
Icariin is a biologically active substance in Epimedii herba that is used for the treatment of neurologic disorders. However, a comprehensive analysis of the molecular mechanisms of icariin is lacking. In this review, we present a brief history of the use of icariin for medicinal purposes; describe the active chemical components of Epimedii herba; and examine the evidence from experimental studies that have uncovered molecular targets of icariin in different diseases. We also constructed a protein–protein interaction network and carried out Gene Ontology and Kyoto Encyclopedia of Genes and Genomes functional enrichment analyses to predict the therapeutic actions of icariin in nervous system diseases including Alzheimer disease, Parkinson disease, ischemic stroke, depressive disorder, multiple sclerosis, glioblastoma, and hereditary spastic paraplegias. The results of our analyses can guide future studies on the application of icariin to the treatment of neurologic disorders.
Collapse
Affiliation(s)
- Shuangqiu Wang
- Department of Neurology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, People's Republic of China.,Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210046, People's Republic of China.,State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, People's Republic of China
| | - Jiarui Ma
- Provincial Key Laboratory of Drug Target and Drug for Degenerative Disease, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, People's Republic of China
| | - Yanqi Zeng
- First Clinical Medical School, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, People's Republic of China
| | - Guowei Zhou
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Yuxuan Wang
- Department of Neurology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, People's Republic of China.,Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210046, People's Republic of China.,State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, People's Republic of China
| | - Wenjuan Zhou
- First Clinical Medical School, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, People's Republic of China
| | - Xiaohe Sun
- First Clinical Medical School, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, People's Republic of China
| | - Minghua Wu
- Department of Neurology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, People's Republic of China.,First Clinical Medical School, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, People's Republic of China
| |
Collapse
|
18
|
Niranjan R, Kishor S, Kumar A. Matrix metalloproteinases in the pathogenesis of dengue viral disease: Involvement of immune system and newer therapeutic strategies. J Med Virol 2021; 93:4629-4637. [PMID: 33634515 DOI: 10.1002/jmv.26903] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/17/2021] [Accepted: 02/22/2021] [Indexed: 12/17/2022]
Abstract
Globally, the burden due to dengue infection is increasing with a recent estimate of 96 million progressing to the disease every year. Dengue pathogenesis and the factors influencing it are not completely known. It is now widely speculated that there is an important role of matrix metalloproteinases (MMPs) in the initiation and progression of dengue pathogenesis; however, their exact roles are not fully understood. Overactivation of matrix metalloproteinases may contribute to the severity of dengue pathogenesis. Cytokines and various other mediators of inflammation interact with the vascular endothelium and matrix metalloproteinases may be one of the components among them. Extensive plasma leakage into tissue spaces may result in a shock. It is evident in the literature that MMP2 and MMP9 increase in dengue patients is correlated with the severity of the disease; however, the underlying mechanism is still unknown. Activation of innate cells and adaptive immune cells which include, B and T cells, macrophages or monocytes and dendritic cells also contribute to the dengue pathology. Newer therapeutic strategies include microRNAs, such as miR-134 (targets MMP3 and MMP1) and MicroRNA-320d, (targets MMP/TIMP proteolytic system). The use of antibodies-based therapeutics like (Andecaliximab; anti-matrix metalloproteinase-9 antibody) is also suggested against MMPs in dengue. In this review, we summarize some recent developments associated with the involvement of immune cells and their mediators associated with the matrix metalloproteinases mediated dengue pathogenesis. We highlight that, there is still very little knowledge about the MMPs in dengue pathogenesis which needs attention and extensive investigations.
Collapse
Affiliation(s)
- Rituraj Niranjan
- Immunology Laboratory, ICMR-Vector Control Research Center, Puducherry, India
| | - Sumitha Kishor
- Immunology Laboratory, ICMR-Vector Control Research Center, Puducherry, India
| | - Ashwani Kumar
- Immunology Laboratory, ICMR-Vector Control Research Center, Puducherry, India
| |
Collapse
|
19
|
Ou Z, Zhou Y, Wang L, Xue L, Zheng J, Chen L, Tong Q. NLRP3 Inflammasome Inhibition Prevents α-Synuclein Pathology by Relieving Autophagy Dysfunction in Chronic MPTP-Treated NLRP3 Knockout Mice. Mol Neurobiol 2021; 58:1303-1311. [PMID: 33169332 DOI: 10.1007/s12035-020-02198-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 11/03/2020] [Indexed: 12/21/2022]
Abstract
Recent researches showed that nucleotide-binding domain and leucine-rich repeat protein 3 (NLRP3) inflammasome inhibition exerted dopaminergic neuroprotection in cellular or animal models of Parkinson's disease (PD). NLRP3 inflammasome has been proposed as a drug target for treatment of PD. However, the interplay between chronic NLRP3 inflammasome and progressive α-synuclein pathology keeps poorly understood. Moreover, the potential mechanism keeps unknown. In the present study, we investigate whether NLRP3 inflammasome inhibition prevents α-synuclein pathology by relieving autophagy dysfunction in the chronic 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) mouse model of PD. NLRP3 knockout mice and their wild-type counterparts were treated with continuous MPTP administration via osmotic mini-pumps. Dopaminergic neuronal degeneration was assessed by western blotting and immunohistochemistry (IHC). The levels of dopamine and its metabolites were determined using high-performance liquid chromatography. NLRP3 inflammasome activation and autophagy biomarkers were assessed by western blot. The expressions of pro-inflammatory cytokines were measured by ELISA. The glial reaction and α-synuclein pathology were assessed by IHC and immunofluorescence. Our results show that NLRP3 inflammasome inhibition via NLRP3 knockout not only protects against nigral dopaminergic degeneration and striatal dopamine deletion but also prevents nigral pathological α-synuclein formation in PD mice. Furthermore, it significantly suppresses MPTP-induced glial reaction accompanied by the secretion of pro-inflammatory cytokines in the midbrain of mice. Most importantly, it relieves autophagy dysfunction in the midbrain of PD mice. Collectively, we demonstrate for the first time that improving autophagy function is involved in the preventive effect of NLRP3 inflammasome inhibition on α-synuclein pathology in PD.
Collapse
Affiliation(s)
- Zhou Ou
- Department of Neurology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, 1 Huanghe Road West, Huaian, 223300, Jiangsu, China
| | - Yuanzhang Zhou
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, Jiangsu, China
| | - Lijun Wang
- Department of Neurology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, 1 Huanghe Road West, Huaian, 223300, Jiangsu, China
| | - Liujun Xue
- Department of Neurology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, 1 Huanghe Road West, Huaian, 223300, Jiangsu, China
| | - Jinlong Zheng
- Department of Neurology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, 1 Huanghe Road West, Huaian, 223300, Jiangsu, China
| | - Liam Chen
- Division of Neuropathology, Department of Pathology, Johns Hopkins University School of Medicine, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Qiang Tong
- Department of Neurology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, 1 Huanghe Road West, Huaian, 223300, Jiangsu, China.
- Division of Neuropathology, Department of Pathology, Johns Hopkins University School of Medicine, 720 Rutland Ave, Baltimore, MD, 21205, USA.
| |
Collapse
|
20
|
Chen H, Qian Z, Zhang S, Tang J, Fang L, Jiang F, Ge D, Chang J, Cao J, Yang L, Cao X. Silencing COX-2 blocks PDK1/TRAF4-induced AKT activation to inhibit fibrogenesis during skeletal muscle atrophy. Redox Biol 2021; 38:101774. [PMID: 33152664 PMCID: PMC7645269 DOI: 10.1016/j.redox.2020.101774] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/26/2020] [Accepted: 10/27/2020] [Indexed: 02/08/2023] Open
Abstract
Skeletal muscle atrophy with high prevalence can induce weakness and fatigability and place huge burden on both health and quality of life. During skeletal muscle degeneration, excessive fibroblasts and extracellular matrix (ECM) accumulated to replace and impair the resident muscle fiber and led to loss of muscle mass. Cyclooxygenase-2 (COX-2), the rate-limiting enzyme in synthesis of prostaglandin, has been identified as a positive regulator in pathophysiological process like inflammation and oxidative stress. In our study, we found injured muscles of human subjects and mouse model overexpressed COX-2 compared to the non-damaged region and COX-2 was also upregulated in fibroblasts following TGF-β stimulation. Then we detected the effect of selective COX-2 inhibitor celecoxib on fibrogenesis. Celecoxib mediated anti-fibrotic effect by inhibiting fibroblast differentiation, proliferation and migration as well as inactivating TGF-β-dependent signaling pathway, non-canonical TGF-β pathways and suppressing generation of reactive oxygen species (ROS) and oxidative stress. In vivo pharmacological inhibition of COX-2 by celecoxib decreased tissue fibrosis and increased skeletal muscle fiber preservation reflected by less ECM formation and myofibroblast accumulation with decreased p-ERK1/2, p-Smad2/3, TGF-βR1, VEGF, NOX2 and NOX4 expression. Expression profiling further found that celecoxib could suppress PDK1 expression. The interaction between COX-2 and PDK1/AKT signaling remained unclear, here we found that COX-2 could bind to PDK1/AKT to form compound. Knockdown of COX-2 in fibroblasts by pharmacological inactivation or by siRNA restrained PDK1 expression and AKT phosphorylation induced by TGF-β treatment. Besides, si-COX-2 prevented TGF-β-induced K63-ubiquitination of AKT by blocking the interaction between AKT and E3 ubiquitin ligase TRAF4. In summary, we found blocking COX-2 inhibited fibrogenesis after muscle atrophy induced by injury and suppressed AKT signaling pathway by inhibiting upstream PDK1 expression and preventing the recruitment of TRAF4 to AKT, indicating that COX-2/PDK1/AKT signaling pathway promised to be target for treating muscle atrophy in the future.
Collapse
Affiliation(s)
- Hongtao Chen
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhanyang Qian
- Department of Orthopedics, Zhongda Hospital of Southeast University, Nanjing, Jiangsu, China
| | - Sheng Zhang
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jian Tang
- Department of Plastic and Burn Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Le Fang
- Department of Critical Care Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fan Jiang
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dawei Ge
- Department of Orthopedics, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jie Chang
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiang Cao
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Yang
- Department of Orthopedics, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Xiaojian Cao
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
21
|
Meng X, Li H, Fang E, Feng J, Zhao X. Comparison of Stage 4 and Stage 4s Neuroblastoma Identifies Autophagy-Related Gene and LncRNA Signatures Associated With Prognosis. Front Oncol 2020; 10:1411. [PMID: 32974147 PMCID: PMC7466450 DOI: 10.3389/fonc.2020.01411] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/03/2020] [Indexed: 01/01/2023] Open
Abstract
Background: The spontaneous regression of neuroblastoma (NB) is most prevalent and well-documented in stage 4s NB patients. However, whether autophagy plays roles in the spontaneous regression of NB is unknown. Objective: This study aimed to identify autophagy-related genes (ARGs) and autophagy-related long non-coding RNAs (lncRNAs) differentially expressed in stage 4 and stage 4s NB and to build prognostic risk signatures on the basis of the ARGs and autophagy-related lncRNAs. Methods: One RNA-sequence (RNA-Seq) dataset (TARGET NBL, n = 153) was utilized as discovery cohort, and two microarray datasets (n = 498 and n = 223) were used as validation cohorts. Differentially expressed ARGs were identified by comparing stage 4s and stage 4 NB samples. An ARG signature risk score and an autophagy-related lncRNA signature risk score were constructed. The receiver operating characteristic (ROC) curve analyses were used to evaluate the survival prediction ability of the two signatures. Gene function annotation and Gene Set Enrichment Analysis (GSEA) were performed to clarify the autophagic biological processes enriched in different risk groups. Results: Nine ARGs were integrated into the ARG signature. Patients in the high-risk group of the ARG signature had significantly poorer overall survival (OS) than patients in the low-risk group. The ROC curves analyses revealed that the ARG signature performed very well in predicting OS [5-year area under the curve (AUC) = 0.81]. Seven autophagy-related lncRNAs were integrated into the autophagy-related lncRNA signature. Patients in the high-risk group of the lncRNA signature had significantly poorer OS than patients in the low-risk group. The ROC curve analyses also revealed that the lncRNA signature performed well in predicting OS (5-year AUC = 0.77). Both the ARG signature and lncRNA signature are independent with other clinical risk factors in the multivariate Cox regression survival analyses. GSEAs revealed that autophagy-related biological processes are enriched in low-risk groups. Conclusions: Autophagy-related genes and lncRNAs are differentially expressed between stage 4 and stage 4s NB. The ARG signature and autophagy-related lncRNA signature successfully stratified NB patients into two risk groups. Autophagy-related biological processes are highly enriched in low-risk NB groups.
Collapse
Affiliation(s)
| | | | | | | | - Xiang Zhao
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
22
|
Maiese K. Dysregulation of metabolic flexibility: The impact of mTOR on autophagy in neurodegenerative disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 155:1-35. [PMID: 32854851 DOI: 10.1016/bs.irn.2020.01.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Non-communicable diseases (NCDs) that involve neurodegenerative disorders and metabolic disease impact over 400 million individuals globally. Interestingly, metabolic disorders, such as diabetes mellitus, are significant risk factors for the development of neurodegenerative diseases. Given that current therapies for these NCDs address symptomatic care, new avenues of discovery are required to offer treatments that affect disease progression. Innovative strategies that fill this void involve the mechanistic target of rapamycin (mTOR) and its associated pathways of mTOR complex 1 (mTORC1), mTOR complex 2 (mTORC2), AMP activated protein kinase (AMPK), trophic factors that include erythropoietin (EPO), and the programmed cell death pathways of autophagy and apoptosis. These pathways are intriguing in their potential to provide effective care for metabolic and neurodegenerative disorders. Yet, future work is necessary to fully comprehend the entire breadth of the mTOR pathways that can effectively and safely translate treatments to clinical medicine without the development of unexpected clinical disabilities.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, NY, United States.
| |
Collapse
|
23
|
Fu X, Tan T, Liu P. Regulation of Autophagy by Non-Steroidal Anti-Inflammatory Drugs in Cancer. Cancer Manag Res 2020; 12:4595-4604. [PMID: 32606952 PMCID: PMC7305821 DOI: 10.2147/cmar.s253345] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 05/12/2020] [Indexed: 12/23/2022] Open
Abstract
Cancer is the leading cause of death, placing a substantial global health burden. The development of the most effective treatment regimen is the unmet clinical need for cancer. Inflammation plays a role in tumorigenesis and progression, and anti-inflammation may be a promising option for cancer management and prevention. Emerging studies have shown that non-steroidal anti-inflammatory drugs (NSAIDs) display anticarcinogenic and chemopreventive properties through the regulation of autophagy in certain types of cancer. In this review, we summarize the pharmacological functions and side effects of NSAIDs as chemotherapeutic agents, and focus on its mode of action on autophagy regulation, which increases our knowledge of NSAIDs and cancer-related inflammation, and contributes to a putative addition of NSAIDs in the chemoprevention and treatment of cancer.
Collapse
Affiliation(s)
- Xiangjie Fu
- Cholestatic Liver Diseases Center and Department of Gastroenterology, Southwest Hospital, Third Military Medical University, Chongqing, People’s Republic of China
| | - Tan Tan
- Translational Medicine Institute, The First Affiliated Hospital of Chenzhou, University of South China, Hunan, People’s Republic of China
| | - Peijun Liu
- Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Shanxi, People’s Republic of China
| |
Collapse
|
24
|
Wang C, Wang T, Hu R, Dai J, Liu H, Li N, Schneider U, Yang Z, Wang J. Cyclooxygenase-2 Facilitates Newcastle Disease Virus Proliferation and Is as a Target for Canthin-6-One Antiviral Activity. Front Microbiol 2020; 11:987. [PMID: 32508794 PMCID: PMC7251056 DOI: 10.3389/fmicb.2020.00987] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 04/23/2020] [Indexed: 11/18/2022] Open
Abstract
Cyclooxygenase-2 (COX-2), one of the mediators of inflammation in response to viral infection, plays an important role in host antiviral defense system. But its role in Newcastle disease virus (NDV) proliferation process remains unclear. This study revealed that inhibition of COX-2 could benefit NDV proliferation and overexpression of COX-2 dose-dependently suppressed NDV proliferation. Overexpression of COX-2 also showed inhibitory effect on NDV-induced endoplasmic reticulum (ER)-stress and autophagy, also promoted the expression of antiviral genes. However, prostaglandin E2 (PGE2), the major product of COX-2, had indistinctive effects on NDV proliferation. At variant time point post viral infection, a tight regulation pattern of COX-2 by NDV was observed. Using inhibitors and siRNA against signaling molecules, the nuclear factor-κB (NF-κB) and melanoma differentiation-associated gene 5 (MDA5) were identified as critical factors for NDV induced COX-2 expression. Nonetheless, at late stage of NDV proliferation, substantial suppression of COX-2 protein synthesis could be detected, accompanied by a decrease in mRNA half-life. Furthermore, three C ring-truncated canthin-6-one analogs were used to activate COX-2 expression and showed inhibitory effect on NDV proliferation with the effective concentrations on μM level. Taken together, these results illustrated a novel NDV-regulated cellular mechanism and indicated that COX-2 is an important regulator of NDV proliferation which can serve as a potential target for anti-NDV agents.
Collapse
Affiliation(s)
- Chongyang Wang
- College of Chemistry and Pharmacy, Northwest A&F University, Yangling, China
| | - Ting Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Ruochen Hu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Jiangkun Dai
- College of Chemistry and Pharmacy, Northwest A&F University, Yangling, China
| | - Haijin Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Na Li
- College of Food Science and Technology, Northwest University, Xi'an, China
| | - Uwe Schneider
- School of Chemistry, The University of Edinburgh, Edinburgh, United Kingdom
| | - Zengqi Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Junru Wang
- College of Chemistry and Pharmacy, Northwest A&F University, Yangling, China
| |
Collapse
|
25
|
COX-2 in liver fibrosis. Clin Chim Acta 2020; 506:196-203. [PMID: 32184095 DOI: 10.1016/j.cca.2020.03.024] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/13/2020] [Accepted: 03/13/2020] [Indexed: 02/07/2023]
Abstract
As a vital inducible sensor, cyclooxygenase-2 (COX-2) plays an important role in the progress of hepatic fibrogenesis. Activation of hepatic stellate cells (HSCs) in the liver can significantly accelerate the onset and development of liver fibrosis. COX-2 overexpression triggers inflammation that is an important inducer in hepatic fibrosis. Increasing evidence indicates that COX-2 is involved in the main pathogenesis of liver fibrosis, such as inflammation, apoptosis, and cell senescence. Moreover, COX-2 expression is altered in patients and animal models with non-alcoholic fatty liver disease or cirrhosis. These findings suggest that COX-2 has a broad and critical role in the development of liver fibrosis. In this review, we summarize the latest advances in the regulation and signal transduction of COX-2 and its impact on liver fibrosis.
Collapse
|
26
|
Alieva AK, Rudenok MM, Novosadova EV, Vlasov IN, Arsenyeva EL, Rosinskaya AV, Grivennikov IA, Slominsky PA, Shadrina MI. Whole-Transcriptome Analysis of Dermal Fibroblasts, Derived from Three Pairs of Monozygotic Twins, Discordant for Parkinson's Disease. J Mol Neurosci 2020; 70:284-293. [PMID: 31823283 PMCID: PMC7222158 DOI: 10.1007/s12031-019-01452-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/17/2019] [Indexed: 01/24/2023]
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative diseases. In most cases, the development of the disease is sporadic and is not associated with any currently known mutations associated with PD. It is believed that changes associated with the epigenetic regulation of gene expression may play an important role in the pathogenesis of this disease. The study of individuals with an almost identical genetic background, such as monozygotic twins, is one of the best approaches to the analysis of such changes. A whole-transcriptome analysis of dermal fibroblasts obtained from three pairs of monozygotic twins discordant for PD was carried out in this work. Twenty-nine differentially expressed genes were identified in the three pairs of twins. These genes were included in seven processes within two clusters, according to the results of an enrichment analysis. The cluster with the greatest statistical significance included processes associated with the regulation of the differentiation of fat cells, the action potential, and the regulation of glutamatergic synaptic transmission. The most significant genes, which occupied a central position in this cluster, were PTGS2, SCN9A, and GRIK2. These genes can be considered as potential candidate genes for PD.
Collapse
Affiliation(s)
- Anelya Kh. Alieva
- Institute of Molecular Genetics of Russian Academy of Sciences, 2 Kurchatova sq, Moscow, 123182 Russia
| | - Margarita M. Rudenok
- Institute of Molecular Genetics of Russian Academy of Sciences, 2 Kurchatova sq, Moscow, 123182 Russia
| | - Ekaterina V. Novosadova
- Institute of Molecular Genetics of Russian Academy of Sciences, 2 Kurchatova sq, Moscow, 123182 Russia
| | - Ivan N. Vlasov
- Institute of Molecular Genetics of Russian Academy of Sciences, 2 Kurchatova sq, Moscow, 123182 Russia
| | - Elena L. Arsenyeva
- Institute of Molecular Genetics of Russian Academy of Sciences, 2 Kurchatova sq, Moscow, 123182 Russia
| | - Anna V. Rosinskaya
- State Public Health Institution Primorsk Regional Clinical Hospital No. 1, 57 Aleutskaya St, Vladivostok, 690091 Russia
| | - Igor A. Grivennikov
- Institute of Molecular Genetics of Russian Academy of Sciences, 2 Kurchatova sq, Moscow, 123182 Russia
| | - Petr A. Slominsky
- Institute of Molecular Genetics of Russian Academy of Sciences, 2 Kurchatova sq, Moscow, 123182 Russia
| | - Maria I. Shadrina
- Institute of Molecular Genetics of Russian Academy of Sciences, 2 Kurchatova sq, Moscow, 123182 Russia
| |
Collapse
|
27
|
Neuroprotective Effect of Optimized Yinxieling Formula in 6-OHDA-Induced Chronic Model of Parkinson's Disease through the Inflammation Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:2529641. [PMID: 31929812 PMCID: PMC6942822 DOI: 10.1155/2019/2529641] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/24/2019] [Accepted: 11/21/2019] [Indexed: 12/14/2022]
Abstract
Parkinson's disease (PD) is characterized by progressive degeneration of dopaminergic neurons in the substantia nigra (SN)-striatum circuit, which is associated with glial activation and consequent chronic neuroinflammation. Optimized Yinxieling Formula (OYF) is a Chinese medicine that exerts therapeutical effect and antiinflammation property on psoriasis. Our previous study has proven that pretreatment with OYF could regulate glia-mediated inflammation in an acute mouse model of PD induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Given that PD is a chronic degeneration disorder, this study applied another PD animal model induced by striatal injection of 6-hydroxydopamine (6-OHDA) to mimic the progressive damage of the SN-striatum dopamine system in rats. The OYF was administrated in the manner of pretreatment plus treatment. The effects of the OYF on motor behaviors were assessed with the apomorphine-induced rotation test and adjusting steps test. To confirm the effect of OYF on dopaminergic neurons and glia activation in this model, we analyzed the expression of tyrosine hydroxylase (TH) and glia markers, ionized calcium-binding adapter molecule 1 (Iba-1), and glial fibrillary acidic protein (GFAP) in the SN region of the rat PD model. Inflammation-associated factors, including tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), IL-6, inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2), were further evaluated in this model and in interferon-γ- (INF-γ-) induced murine macrophages RAW264.7 cells. The results from the in vivo study showed that OYF reversed the motor behavioral dysfunction in 6-OHDA-induced PD rats, upregulated the TH expression, decreased the immunoreactivity of Iba-1 and GFAP, and downregulated the mRNA levels of TNF-α and COX-2. The OYF also trended to decrease the mRNA levels of IL-1β and iNOS in vivo. The results from the in vitro study showed that OYF significantly decreased the mRNA levels of TNF-α, IL-1β, IL-6, iNOS, and COX-2. Therefore, this study suggests that OYF exerts antiinflammatory effects, which might be related to the protection of dopaminergic neurons in 6-OHDA-induced chronic neurotoxicity.
Collapse
|
28
|
Jha NK, Kar R, Niranjan R. ABC Transporters in Neurological Disorders: An Important Gateway for Botanical Compounds Mediated Neuro-Therapeutics. Curr Top Med Chem 2019; 19:795-811. [PMID: 30977450 DOI: 10.2174/1568026619666190412121811] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 02/27/2019] [Accepted: 03/07/2019] [Indexed: 12/13/2022]
Abstract
Neurodegeneration is a distinguishing feature of many age related disorders and other vector borne neuroinflammatory diseases. There are a number of factors that can modulate the pathology of these disorders. ATP-binding cassette (ABC) transporters are primarily involved in the maintenance of normal brain homeostasis by eliminating toxic peptides and compounds from the brain. Also, ABC transporters protect the brain from the unwanted effects of endogenous and exogenous toxins that can enter the brain parenchyma. Therefore, these transporters have the ability to determine the pathological outcomes of several neurological disorders. For instance, ABC transporters like P-glycoprotein (ABCB1), and BCRP (ABCG2) have been reported to facilitate the clearance of peptides such as amyloid-β (Aβ) that accumulate in the brain during Alzheimer's disease (AD) progression. Other members such as ABCA1, ABCA2, ABCC8, ABCC9, ABCG1 and ABCG4 also have been reported to be involved in the progression of various brain disorders such as HIV-associated dementia, Multiple sclerosis (MS), Ischemic stroke, Japanese encephalitis (JE) and Epilepsy. However, these defective transporters can be targeted by numerous botanical compounds such as Verapamil, Berberine and Fascalpsyn as a therapeutic target to treat these neurological outcomes. These compounds are already reported to modulate ABC transporter activity in the CNS. Nonetheless, the exact mechanisms involving the ABC transporters role in normal brain functioning, their role in neuronal dysfunction and how these botanical compounds ensure and facilitate their therapeutic action in association with defective transporters still remain elusive. This review therefore, summarizes the role of ABC transporters in neurological disorders, with a special emphasis on its role in AD brains. The prospect of using botanical/natural compounds as modulators of ABC transporters in neurological disorders is discussed in the latter half of the article.
Collapse
Affiliation(s)
- Niraj Kumar Jha
- Department of Biotechnology, Noida Institute of Engineering & Technology (NIET), Greater Noida, India
| | - Rohan Kar
- Department of Biotechnology, Noida Institute of Engineering & Technology (NIET), Greater Noida, India
| | - Rituraj Niranjan
- Unit of Microbiology and Molecular Biology, ICMR-Vector Control Research Center, Puducherry-605006, India
| |
Collapse
|
29
|
Lee SB, Youn J, Jang W, Yang HO. Neuroprotective effect of anodal transcranial direct current stimulation on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced neurotoxicity in mice through modulating mitochondrial dynamics. Neurochem Int 2019; 129:104491. [PMID: 31229553 DOI: 10.1016/j.neuint.2019.104491] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 06/07/2019] [Accepted: 06/16/2019] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the accumulation of protein inclusions and the loss of dopaminergic neurons. Abnormal mitochondrial homeostasis is thought to be important for the pathogenesis of PD. Transcranial direct current stimulation (tDCS), a noninvasive brain stimulation technique, constitutes a promising approach for promoting recovery of various neurological conditions. However, little is known about its mechanism of action. The present study elucidated the neuroprotective effects of tDCS on the mitochondrial quality control pathway in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse model. We used the MPTP-induced neurotoxicity in vivo model. Mice were stimulated for 5 consecutive days with MPTP treatment. After observation of behavioral alteration using the rotarod test, mice were sacrificed for the measurement of the PD- and mitochondrial quality control-related protein levels in the substantia nigra. tDCS improved the behavioral alterations and changes in tyrosine hydroxylase levels in MPTP-treated mice. Furthermore, tDCS attenuated mitochondrial damage, as indicated by diminished mitochondrial swelling and mitochondrial glutamate dehydrogenase activity in the MPTP-induced PD mouse model. MPTP significantly increased mitophagy and decreased mitochondrial biogenesis-related proteins. These changes were attenuated by tDCS. Furthermore, MPTP significantly increased fission-related protein dynamin-related protein 1 with no effect on fusion-related protein mitofusin-2, and tDCS attenuated these changes. Our findings demonstrated the neuroprotective effect of anodal tDCS on the MPTP-induced neurotoxic mouse model through suppressing excessive mitophagy and balancing mitochondrial dynamics. The neuroprotective effect of anodal tDCS with modulation of mitochondrial dynamics provides a new therapeutic strategy for the treatment of PD.
Collapse
Affiliation(s)
- Sang-Bin Lee
- Natural Medicine Center, Korea Institute of Science and Technology, Gangneung, 25457, Republic of Korea; School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jinyoung Youn
- Department of Neurology, Samsung Medical Center, School of Medicine, Sungkyunkwan University, Seoul, Republic of Korea
| | - Wooyoung Jang
- Department of Neurology, Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung, Republic of Korea.
| | - Hyun Ok Yang
- Natural Medicine Center, Korea Institute of Science and Technology, Gangneung, 25457, Republic of Korea; Division of Bio-Medical Science &Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea.
| |
Collapse
|
30
|
Recent advances in the mechanisms of neuroinflammation and their roles in neurodegeneration. Neurochem Int 2018; 120:13-20. [PMID: 30016687 DOI: 10.1016/j.neuint.2018.07.003] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/07/2018] [Accepted: 07/13/2018] [Indexed: 12/11/2022]
Abstract
Neuroinflammation is associated with the pathogenesis of many neurological disorders including Parkinson's disease, Alzheimer's disease, Amyotrophic lateral sclerosis and Huntington disease. Current studies in this area have advanced the mechanism of neuroinflammation and its role in neurodegeneration. Studies from epidemiologic, clinical and animal models also contributed in the various new mechanisms of neuroinflammation. In this line, activation of monocytes is an important emerging mechanism that has a, profound role in neuroinflammation and neurodegeneration. Ion channels, matrix metalloproteases and microRNAs are also found to be the key players in the pathogenesis of neuroinflammation. In particular, microRNA-32 regulates microglia-mediated neuroinflammation and thus neurodegeneration. Notably, some important studies describe the role of Th17 cells in neuroinflammation, but, very little knowledge is available about their mechanism of action. Particularly, the role of autophagy gets emphasized, which plays a very critical role in protein aggregation and neurodegeneration. In this review, we highlight and discuss the mechanisms of these mediators of inflammation by which they contribute to the disease progression. In conclusion, we focus on the various newer molecular mechanisms that are associated with the basic understanding of neuroinflammation in neurodegeneration.
Collapse
|