1
|
Du R, Xiao N, Han L, Guo K, Li K, Chen Z, Zhang H, Zhou Z, Huang Y, Zhao X, Bian H. Dexrazoxane inhibits the growth of esophageal squamous cell carcinoma by attenuating SDCBP/MDA-9/syntenin-mediated EGFR-PI3K-Akt pathway activation. Sci Rep 2024; 14:9167. [PMID: 38649770 PMCID: PMC11035576 DOI: 10.1038/s41598-024-59665-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 04/12/2024] [Indexed: 04/25/2024] Open
Abstract
Syndecan-binding protein (SDCBP) was reported to stimulate the advancement of esophageal squamous cell carcinoma (ESCC) and could potentially be a target for ESCC treatment. There is a growing corpus of research on the anti-tumor effects of iron chelators; however, very few studies have addressed the involvement of dexrazoxane in cancer. In this study, structure-based virtual screening was employed to select drugs targeting SDCBP from the Food and Drug Administration (FDA)-approved drug databases. The sepharose 4B beads pull-down assay revealed that dexrazoxane targeted SDCBP by interacting with its PDZ1 domain. Additionally, dexrazoxane inhibited ESCC cell proliferation and anchorage-independent colony formation via SDCBP. ESCC cell apoptosis and G2 phase arrest were induced as measured by the flow cytometry assay. Subsequent research revealed that dexrazoxane attenuated the binding ability between SDCBP and EGFR in an immunoprecipitation assay. Furthermore, dexrazoxane impaired EGFR membrane localization and inactivated the EGFR/PI3K/Akt pathway. In vivo, xenograft mouse experiments indicated that dexrazoxane suppressed ESCC tumor growth. These data indicate that dexrazoxane might be established as a potential anti-cancer agent in ESCC by targeting SDCBP.
Collapse
Affiliation(s)
- Ruijuan Du
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang, 473004, Henan, People's Republic of China.
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, No. 80, Changjiang Road, Nanyang, 473004, Henan, People's Republic of China.
| | - Nan Xiao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
| | - Li Han
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang, 473004, Henan, People's Republic of China
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, No. 80, Changjiang Road, Nanyang, 473004, Henan, People's Republic of China
| | - KeLei Guo
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang, 473004, Henan, People's Republic of China
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, No. 80, Changjiang Road, Nanyang, 473004, Henan, People's Republic of China
| | - Kai Li
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang, 473004, Henan, People's Republic of China
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, No. 80, Changjiang Road, Nanyang, 473004, Henan, People's Republic of China
| | - Zhiguo Chen
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang, 473004, Henan, People's Republic of China
| | - Hui Zhang
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang, 473004, Henan, People's Republic of China
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, No. 80, Changjiang Road, Nanyang, 473004, Henan, People's Republic of China
| | - Zijun Zhou
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang, 473004, Henan, People's Republic of China
| | - Yunlong Huang
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang, 473004, Henan, People's Republic of China
| | - Xulin Zhao
- Oncology Department, Nanyang First People's Hospital, Nan Yang, 473004, Henan, People's Republic of China
| | - Hua Bian
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang, 473004, Henan, People's Republic of China.
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, No. 80, Changjiang Road, Nanyang, 473004, Henan, People's Republic of China.
| |
Collapse
|
2
|
Ding XS, Gao L, Han Z, Eleuteri S, Shi W, Shen Y, Song ZY, Su M, Yang Q, Qu Y, Simon DK, Wang XL, Wang B. Ferroptosis in Parkinson's disease: Molecular mechanisms and therapeutic potential. Ageing Res Rev 2023; 91:102077. [PMID: 37742785 DOI: 10.1016/j.arr.2023.102077] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/26/2023] [Accepted: 09/21/2023] [Indexed: 09/26/2023]
Abstract
Parkinson's Disease (PD) is characterized by the progressive loss of dopaminergic neurons in the substantia nigra (SN), leading to motor and non-motor symptoms. While the exact mechanisms remain complex and multifaceted, several molecular pathways have been implicated in PD pathology, including accumulation of misfolded proteins, impaired mitochondrial function, oxidative stress, inflammation, elevated iron levels, etc. Overall, PD's molecular mechanisms involve a complex interplay between genetic, environmental, and cellular factors that disrupt cellular homeostasis, and ultimately lead to the degeneration of dopaminergic neurons. Recently, emerging evidence highlights ferroptosis, an iron-dependent non-apoptotic cell death process, as a pivotal player in the advancement of PD. Notably, oligomeric α-synuclein (α-syn) generates reactive oxygen species (ROS) and lipid peroxides within cellular membranes, potentially triggering ferroptosis. The loss of dopamine, a hallmark of PD, could predispose neurons to ferroptotic vulnerability. This unique form of cell demise unveils fresh insights into PD pathogenesis, necessitating an exploration of the molecular intricacies connecting ferroptosis and PD progression. In this review, the molecular and regulatory mechanisms of ferroptosis and their connection with the pathological processes of PD have been systematically summarized. Furthermore, the features of ferroptosis in PD animal models and clinical trials targeting ferroptosis as a therapeutic approach in PD patients' management are scrutinized.
Collapse
Affiliation(s)
- Xv-Shen Ding
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China; Basic Medicine School, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China
| | - Li Gao
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China
| | - Zheng Han
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China
| | - Simona Eleuteri
- Department of Neurology, Beth Israel Deaconess Medical Center, 3 Blackfan Circle 628H, Boston, MA 02215, USA
| | - Wei Shi
- Department of Neurosurgery, PLA 960th hospital, JiNan, Shandong Province, 250031, China
| | - Yun Shen
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China
| | - Zi-Yao Song
- Basic Medicine School, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China
| | - Mingming Su
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China
| | - Qian Yang
- Department of Experimental Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China
| | - Yan Qu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China.
| | - David K Simon
- Department of Neurology, Beth Israel Deaconess Medical Center, 3 Blackfan Circle 628H, Boston, MA 02215, USA.
| | - Xue-Lian Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China.
| | - Bao Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China.
| |
Collapse
|
3
|
Miller SJ, Darji RY, Walaieh S, Lewis JA, Logan R. Senolytic and senomorphic secondary metabolites as therapeutic agents in Drosophila melanogaster models of Parkinson's disease. Front Neurol 2023; 14:1271941. [PMID: 37840914 PMCID: PMC10568035 DOI: 10.3389/fneur.2023.1271941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/04/2023] [Indexed: 10/17/2023] Open
Abstract
Drosophila melanogaster is a valuable model organism for a wide range of biological exploration. The well-known advantages of D. melanogaster include its relatively simple biology, the ease with which it is genetically modified, the relatively low financial and time costs associated with their short gestation and life cycles, and the large number of offspring they produce per generation. D. melanogaster has facilitated the discovery of many significant insights into the pathology of Parkinson's disease (PD) and has served as an excellent preclinical model of PD-related therapeutic discovery. In this review, we provide an overview of the major D. melanogaster models of PD, each of which provide unique insights into PD-relevant pathology and therapeutic targets. These models are discussed in the context of their past, current, and future potential use for studying the utility of secondary metabolites as therapeutic agents in PD. Over the last decade, senolytics have garnered an exponential interest in their ability to mitigate a broad spectrum of diseases, including PD. Therefore, an emphasis is placed on the senolytic and senomorphic properties of secondary metabolites. It is expected that D. melanogaster will continue to be critical in the effort to understand and improve treatment of PD, including their involvement in translational studies focused on secondary metabolites.
Collapse
Affiliation(s)
- Sean J. Miller
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT, United States
| | - Rayyan Y. Darji
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT, United States
| | - Sami Walaieh
- Department of Biology, Eastern Nazarene College, Quincy, MA, United States
| | - Jhemerial A. Lewis
- Department of Biology, Eastern Nazarene College, Quincy, MA, United States
| | - Robert Logan
- Department of Biology, Eastern Nazarene College, Quincy, MA, United States
| |
Collapse
|
4
|
Grosu L, Grosu AI, Crisan D, Zlibut A, Perju-Dumbrava L. Parkinson's disease and cardiovascular involvement: Edifying insights (Review). Biomed Rep 2023; 18:25. [PMID: 36846617 PMCID: PMC9944619 DOI: 10.3892/br.2023.1607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/02/2023] [Indexed: 02/16/2023] Open
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative illnesses, and is a major healthcare burden with prodigious consequences on life-quality, morbidity, and survival. Cardiovascular diseases are the leading cause of mortality worldwide and growing evidence frequently reports their co-existence with PD. Cardiac dysautonomia due to autonomic nervous system malfunction is the most prevalent type of cardiovascular manifestation in these patients, comprising orthostatic and postprandial hypotension, along with supine and postural hypertension. Moreover, many studies have endorsed the risk of patients with PD to develop ischemic heart disease, heart failure and even arrhythmias, but the underlying mechanisms are not entirely clear. As importantly, the medication used in treating PD, such as levodopa, dopamine agonists or anticholinergic agents, is also responsible for cardiovascular adverse reactions, but further studies are required to elucidate the underlying mechanisms. The purpose of this review was to provide a comprehensive overview of current available data regarding the overlapping cardiovascular disease in patients with PD.
Collapse
Affiliation(s)
- Laura Grosu
- Department of Neurology, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Neurology, Municipal Clinical Hospital, 400139 Cluj-Napoca, Romania
| | - Alin Ionut Grosu
- Department of Internal Medicine, 5th Medical Clinic, Iuliu Hatieganu University of Medicine and Pharmacy, 400139 Cluj-Napoca, Romania
- Department of Cardiology, Municipal Clinical Hospital, 400139 Cluj-Napoca, Romania
| | - Dana Crisan
- Department of Internal Medicine, 5th Medical Clinic, Iuliu Hatieganu University of Medicine and Pharmacy, 400139 Cluj-Napoca, Romania
- Department of Internal Medicine, Municipal Clinical Hospital, 400139 Cluj-Napoca, Romania
| | - Alexandru Zlibut
- Department of Internal Medicine, 5th Medical Clinic, Iuliu Hatieganu University of Medicine and Pharmacy, 400139 Cluj-Napoca, Romania
- Department of Cardiology, Emergency Clinical Hospital of Bucharest, 014461 Bucharest, Romania
| | - Lacramioara Perju-Dumbrava
- Department of Neurology, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| |
Collapse
|
5
|
Sethi B, Kumar V, Mahato K, Coulter DW, Mahato RI. Recent advances in drug delivery and targeting to the brain. J Control Release 2022; 350:668-687. [PMID: 36057395 PMCID: PMC9884093 DOI: 10.1016/j.jconrel.2022.08.051] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/19/2022] [Accepted: 08/26/2022] [Indexed: 02/01/2023]
Abstract
Our body keeps separating the toxic chemicals in the blood from the brain. A significant number of drugs do not enter the central nervous system (CNS) due to the blood-brain barrier (BBB). Certain diseases, such as tumor growth and stroke, are known to increase the permeability of the BBB. However, the heterogeneity of this permeation makes it difficult and unpredictable to transport drugs to the brain. In recent years, research has been directed toward increasing drug penetration inside the brain, and nanomedicine has emerged as a promising approach. Active targeting requires one or more specific ligands on the surface of nanoparticles (NPs), which brain endothelial cells (ECs) recognize, allowing controlled drug delivery compared to conventional targeting strategies. This review highlights the mechanistic insights about different cell types contributing to the development and maintenance of the BBB and summarizes the recent advancement in brain-specific NPs for different pathological conditions. Furthermore, fundamental properties of brain-targeted NPs will be discussed, and the standard lesion features classified by neurological pathology are summarized.
Collapse
Affiliation(s)
- Bharti Sethi
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha NE 68198, USA
| | - Virender Kumar
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha NE 68198, USA
| | - Kalika Mahato
- College of Medicine, University of Nebraska Medical Center, Omaha NE 68198, USA
| | - Donald W Coulter
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha NE 68198, USA.
| |
Collapse
|
6
|
Nian Y, Hu X, Zhang R, Feng J, Du J, Li F, Bu L, Zhang Y, Chen Y, Tao C. Mining on Alzheimer's diseases related knowledge graph to identity potential AD-related semantic triples for drug repurposing. BMC Bioinformatics 2022; 23:407. [PMID: 36180861 PMCID: PMC9523633 DOI: 10.1186/s12859-022-04934-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To date, there are no effective treatments for most neurodegenerative diseases. Knowledge graphs can provide comprehensive and semantic representation for heterogeneous data, and have been successfully leveraged in many biomedical applications including drug repurposing. Our objective is to construct a knowledge graph from literature to study the relations between Alzheimer's disease (AD) and chemicals, drugs and dietary supplements in order to identify opportunities to prevent or delay neurodegenerative progression. We collected biomedical annotations and extracted their relations using SemRep via SemMedDB. We used both a BERT-based classifier and rule-based methods during data preprocessing to exclude noise while preserving most AD-related semantic triples. The 1,672,110 filtered triples were used to train with knowledge graph completion algorithms (i.e., TransE, DistMult, and ComplEx) to predict candidates that might be helpful for AD treatment or prevention. RESULTS Among three knowledge graph completion models, TransE outperformed the other two (MR = 10.53, Hits@1 = 0.28). We leveraged the time-slicing technique to further evaluate the prediction results. We found supporting evidence for most highly ranked candidates predicted by our model which indicates that our approach can inform reliable new knowledge. CONCLUSION This paper shows that our graph mining model can predict reliable new relationships between AD and other entities (i.e., dietary supplements, chemicals, and drugs). The knowledge graph constructed can facilitate data-driven knowledge discoveries and the generation of novel hypotheses.
Collapse
Affiliation(s)
- Yi Nian
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, 7000 Fannin St, Houston, TX 77030 USA
| | - Xinyue Hu
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, 7000 Fannin St, Houston, TX 77030 USA
| | - Rui Zhang
- Department of Pharmaceutical Care & Health System (PCHS) and the Institute for Health Informatics (IHI), University of Minnesota, 7-115A Weaver-Densford Hall, Minneapolis, MN 55455 USA
| | - Jingna Feng
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, 7000 Fannin St, Houston, TX 77030 USA
| | - Jingcheng Du
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, 7000 Fannin St, Houston, TX 77030 USA
| | - Fang Li
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, 7000 Fannin St, Houston, TX 77030 USA
| | - Larry Bu
- University of Maryland School of Medicine, 655 W Baltimore St S, Baltimore, MD 21201 USA
| | - Yuji Zhang
- University of Maryland School of Medicine, 655 W Baltimore St S, Baltimore, MD 21201 USA
| | - Yong Chen
- Department of Biostatistics, Epidemiology and Informatics (DBEI), the Perelman School of Medicine, University of Pennsylvania, 602 Blockley Hall, 423 Guardian Drive, Philadelphia, PA 19104 USA
| | - Cui Tao
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, 7000 Fannin St, Houston, TX 77030 USA
| |
Collapse
|
7
|
Sun Y, He L, Wang W, Xie Z, Zhang X, Wang P, Wang L, Yan C, Liu Z, Zhao J, Cui Z, Wang Y, Tang L, Zhang Z. Activation of Atg7-dependent autophagy by a novel inhibitor of the Keap1-Nrf2 protein-protein interaction from Penthorum chinense Pursh. attenuates 6-hydroxydopamine-induced ferroptosis in zebrafish and dopaminergic neurons. Food Funct 2022; 13:7885-7900. [PMID: 35776077 DOI: 10.1039/d2fo00357k] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The death of dopaminergic neurons is a dominant factor during the occurrence and development of Parkinson's disease (PD). Previous studies demonstrated that ferroptosis is implicated in the death of dopaminergic neurons. Besides, polyphenols have been proven to be effective in preventing the death of dopaminergic neurons. This work aims to explore the neuroprotective effect and mechanism of thonningianin A (Th A), a polyphenolic compound in natural plant foods, against 6-hydroxydopamine (6-OHDA)-induced ferroptosis in dopaminergic cells. The results of molecular docking and other binding assays collectively demonstrated that Th A can strongly target the Kelch domain of Keap1. Th A treatment significantly facilitated the nuclear factor erythroid 2-like 2 (Nrf2) nuclear translocation and subsequently increased the heme oxygenase-1 (HO-1) protein level through inhibiting the protein-protein interaction (PPI) of Keap1 and Nrf2. Compared with the nomifensine (Nomi) treatment, Th A had a more potent protective effect on 6-OHDA-induced ferroptosis during PD pathology in zebrafish, which was associated with assuaging the reduction of the total swimming distance, glutathione (GSH) depletion, iron accumulation, lipid peroxidation, and aggregation of α-synuclein (α-syn). Furthermore, Th A also exhibited a strong protective effect against 6-OHDA-induced ferroptosis in vitro in the human neuroblastoma cell line SH-SY5Y. Th A degraded Keap1 protein through activating Atg7-dependent autophagy. Additionally, Th A treatment facilitated the degradation of Keap1 protein by promoting the interaction between p62/SQSTM1 (sequestosome 1, hereafter referred to as p62) and Keap1. Taken together, our findings indicated that Th A protects dopaminergic cells against 6-OHDA-induced ferroptosis through activating the Nrf2-based cytoprotective system, thus enabling a potential application of Keap1-Nrf2 PPI inhibitors in the restraint of ferroptosis and treatment of PD.
Collapse
Affiliation(s)
- Yiran Sun
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, P.R. China.
| | - Libo He
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, Sichuan, China.
| | - Wang Wang
- School of Basic Medicine, Nanchang Medical College, Nanchang 330052, Jiangxi, China
| | - Zhishen Xie
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, P.R. China.
| | - Xiaowei Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, P.R. China.
| | - Pan Wang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, P.R. China.
| | - Lan Wang
- College of Chemical and Food Engineering, Zhengzhou Institute of Technology, Zhengzhou 450044, China
| | - Chenchen Yan
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, P.R. China.
| | - Zhiwen Liu
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, P.R. China.
| | - Jie Zhao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, P.R. China.
| | - Zhenghao Cui
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, P.R. China.
| | - Yida Wang
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, Sichuan, China.
| | - Lin Tang
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, Sichuan, China.
| | - Zhenqiang Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, P.R. China.
| |
Collapse
|
8
|
Fu XX, Wang J, Cai HY, Jiang H, Jiang JZ, Chen HH, Han S. Co-Application of C16 and Ang-1 Improves the Effects of Levodopa in Parkinson Disease Treatment. J Inflamm Res 2022; 15:3797-3814. [PMID: 35836722 PMCID: PMC9273834 DOI: 10.2147/jir.s368291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/29/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Levodopa is regarded as a standard medication in Parkinson disease (PD) treatment. However, long-term administration of levodopa leads to levodopa-induced dyskinesia (LID), which can markedly affect patient quality of life. Previous studies have shown that neuroinflammation in the brain plays a role in LID and increases potential neuroinflammatory mediators associated with the side effects of levodopa. OBJECTIVE The treatment effect of C16 (a peptide that competitively binds integrin αvβ3 and inhibits inflammatory cell infiltration) and angiopoietin-1 (Ang-1; a vascular endothelial growth factor vital for blood vessel protection), along with levodopa, was evaluated in a rodent model of PD. METHODS We administered a combination of C16 and Ang-1 in a rodent model of PD induced by MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine). Seventy-five mice were randomly divided into five treatment groups: control, vehicle, levodopa, C16+Ang-1, and levodopa+C16+Ang-1. Behavioral, histological, and electrophysiological experiments were used to determine neuron function and recovery. RESULTS The results showed that C16+Ang-1 treatment alleviated neuroinflammation in the CNS and promoted the recovery effects of levodopa on neural function. CONCLUSION Our study suggests that C16+Ang-1 can compensate for the shortcomings of levodopa, improve the CNS microenvironment, and ameliorate the effects of levodopa. This treatment strategy could be developed as a combinatorial therapeutic in the future.
Collapse
Affiliation(s)
- Xiao-Xiao Fu
- Institute of Anatomy and Sir Run Run Shaw Hospital, Medical College, Zhejiang University, Hangzhou, People’s Republic of China
| | - Jin Wang
- Institute of Anatomy and Sir Run Run Shaw Hospital, Medical College, Zhejiang University, Hangzhou, People’s Republic of China
| | - Hua-Ying Cai
- Institute of Anatomy and Sir Run Run Shaw Hospital, Medical College, Zhejiang University, Hangzhou, People’s Republic of China
| | - Hong Jiang
- Institute of Anatomy and Sir Run Run Shaw Hospital, Medical College, Zhejiang University, Hangzhou, People’s Republic of China
| | - Jin-Zhan Jiang
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, Jinhua, People’s Republic of China
| | - Hao-Hao Chen
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, Jinhua, People’s Republic of China
- Hao-Hao Chen, Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, 1188 Wuzhou Steet, Jinhua, People’s Republic of China, Tel +86-579-82265128, Fax +86-579-82265110, Email
| | - Shu Han
- Institute of Anatomy and Sir Run Run Shaw Hospital, Medical College, Zhejiang University, Hangzhou, People’s Republic of China
- Correspondence: Shu Han, Institute of Anatomy, Sir Run Run Shaw Hospital, Medical College, Zhejiang University, 866 Yuhangtang Road, Hangzhou, People’s Republic of China, Tel +86-571-88208160, Fax +86-571-88208094, Email
| |
Collapse
|
9
|
Mishra A, Bandopadhyay R, Singh PK, Mishra PS, Sharma N, Khurana N. Neuroinflammation in neurological disorders: pharmacotherapeutic targets from bench to bedside. Metab Brain Dis 2021; 36:1591-1626. [PMID: 34387831 DOI: 10.1007/s11011-021-00806-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 07/22/2021] [Indexed: 02/07/2023]
Abstract
Neuroinflammation is one of the host defensive mechanisms through which the nervous system protects itself from pathogenic and or infectious insults. Moreover, neuroinflammation occurs as one of the most common pathological outcomes in various neurological disorders, makes it the promising target. The present review focuses on elaborating the recent advancement in understanding molecular mechanisms of neuroinflammation and its role in the etiopathogenesis of various neurological disorders, especially Alzheimer's disease (AD), Parkinson's disease (PD), and Epilepsy. Furthermore, the current status of anti-inflammatory agents in neurological diseases has been summarized in light of different preclinical and clinical studies. Finally, possible limitations and future directions for the effective use of anti-inflammatory agents in neurological disorders have been discussed.
Collapse
Affiliation(s)
- Awanish Mishra
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, India.
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, 781101, India.
| | - Ritam Bandopadhyay
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, India
| | - Prabhakar Kumar Singh
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, India
| | - Pragya Shakti Mishra
- Department of Nuclear Medicine, Sanjay Gandhi Post Graduate Institute of Medical Sciences (SGPGIMS), Raebareli Road, Lucknow, 226014, India
| | - Neha Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, India
| | - Navneet Khurana
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, India
| |
Collapse
|
10
|
Mustapha M, Taib CNM. MPTP-induced mouse model of Parkinson's disease: A promising direction of therapeutic strategies. Bosn J Basic Med Sci 2021; 21:422-433. [PMID: 33357211 PMCID: PMC8292858 DOI: 10.17305/bjbms.2020.5181] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/10/2020] [Indexed: 12/23/2022] Open
Abstract
Among the popular animal models of Parkinson's disease (PD) commonly used in research are those that employ neurotoxins, especially 1-methyl- 4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP). This neurotoxin exerts it neurotoxicity by causing a barrage of insults, such as oxidative stress, mitochondrial apoptosis, inflammation, excitotoxicity, and formation of inclusion bodies acting singly and in concert, ultimately leading to dopaminergic neuronal damage in the substantia nigra pars compacta and striatum. The selective neurotoxicity induced by MPTP in the nigrostriatal dopaminergic neurons of the mouse brain has led to new perspectives on PD. For decades, the MPTP-induced mouse model of PD has been the gold standard in PD research even though it does not fully recapitulate PD symptomatology, but it does have the advantages of simplicity, practicability, affordability, and fewer ethical considerations and greater clinical correlation than those of other toxin models of PD. The model has rejuvenated PD research and opened new frontiers in the quest for more novel therapeutic and adjuvant agents for PD. Hence, this review summarizes the role of MPTP in producing Parkinson-like symptoms in mice and the experimental role of the MPTP-induced mouse model. We discussed recent developments of more promising PD therapeutics to enrich our existing knowledge about this neurotoxin using this model.
Collapse
Affiliation(s)
- Musa Mustapha
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor (Darul Ehsan), Malaysia
- Department of Human Anatomy, Faculty of Basic Sciences, College of Medical Sciences, Ahmadu Bello University, Zaria, Nigeria
| | - Che Norma Mat Taib
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor (Darul Ehsan), Malaysia
| |
Collapse
|
11
|
Huang X, Hussain B, Chang J. Peripheral inflammation and blood-brain barrier disruption: effects and mechanisms. CNS Neurosci Ther 2021; 27:36-47. [PMID: 33381913 PMCID: PMC7804893 DOI: 10.1111/cns.13569] [Citation(s) in RCA: 247] [Impact Index Per Article: 82.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 01/08/2023] Open
Abstract
The blood-brain barrier (BBB) is an important physiological barrier that separates the central nervous system (CNS) from the peripheral circulation, which contains inflammatory mediators and immune cells. The BBB regulates cellular and molecular exchange between the blood vessels and brain parenchyma. Normal functioning of the BBB is crucial for the homeostasis and proper function of the brain. It has been demonstrated that peripheral inflammation can disrupt the BBB by various pathways, resulting in different CNS diseases. Recently, clinical research also showed CNS complications following SARS-CoV-2 infection and chimeric antigen receptor (CAR)-T cell therapy, which both lead to a cytokine storm in the circulation. Therefore, elucidation of the mechanisms underlying the BBB disruption induced by peripheral inflammation will provide an important basis for protecting the CNS in the context of exacerbated peripheral inflammatory diseases. In the present review, we first summarize the physiological properties of the BBB that makes the CNS an immune-privileged organ. We then discuss the relevance of peripheral inflammation-induced BBB disruption to various CNS diseases. Finally, we elaborate various factors and mechanisms of peripheral inflammation that disrupt the BBB.
Collapse
Affiliation(s)
- Xiaowen Huang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular ImmunomodulationInstitute of Biomedicine and BiotechnologyShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- University of Chinese Academy of SciencesBeijingChina
| | - Basharat Hussain
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular ImmunomodulationInstitute of Biomedicine and BiotechnologyShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- University of Chinese Academy of SciencesBeijingChina
| | - Junlei Chang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular ImmunomodulationInstitute of Biomedicine and BiotechnologyShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
| |
Collapse
|
12
|
Mendonça IP, Duarte-Silva E, Chaves-Filho AJM, Andrade da Costa BLDS, Peixoto CA. Neurobiological findings underlying depressive behavior in Parkinson's disease: A review. Int Immunopharmacol 2020; 83:106434. [PMID: 32224442 DOI: 10.1016/j.intimp.2020.106434] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 03/19/2020] [Indexed: 12/19/2022]
Abstract
Parkinson's disease (PD) is one of the most prevalent neurodegenerative diseases in the world with a harmful impact on the quality of life. Although its clinical diagnosis is based on motor symptoms such as resting tremor, postural instability, slow gait, and muscle stiffness, this disorder is also characterized by the presence of early emotional impairment, including features such as depression, anxiety, fatigue, and apathy. Depression is the main emotional manifestation associated with PD and the mechanisms involved in its pathophysiology have been extensively investigated however, it is not yet completely elucidated. In addition to monoaminergic imbalance, immunological and gut microbiota changes have been associated with depression in PD. Besides, a patient group appears be refractory to the treatment available currently. This review emphasizes the mainly neuromolecular findings of the PD-associated depression as well as discuss novel and potential pharmacological and non-pharmacological therapeutic strategies.
Collapse
Affiliation(s)
- Ingrid Prata Mendonça
- Laboratory of Ultrastructure, AggeuMagalhães Institute (IAM), Oswaldo Cruz Foundation (FIOCRUZ), PE, Brazil; Postgraduate Program in Biological Sciences (PPGCB), Federal University of Pernambuco (UFPE), Brazil.
| | - Eduardo Duarte-Silva
- Laboratory of Ultrastructure, AggeuMagalhães Institute (IAM), Oswaldo Cruz Foundation (FIOCRUZ), PE, Brazil; Postgraduate Program in Biosciences and Biotechnology for Health (PPGBBS), Oswaldo Cruz Foundation (FIOCRUZ-PE)/AggeuMagalhães Institute (IAM), Recife, PE, Brazil
| | - Adriano José Maia Chaves-Filho
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | | | - Christina Alves Peixoto
- Laboratory of Ultrastructure, AggeuMagalhães Institute (IAM), Oswaldo Cruz Foundation (FIOCRUZ), PE, Brazil; National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.
| |
Collapse
|
13
|
Rai SN, Singh P. Advancement in the modelling and therapeutics of Parkinson's disease. J Chem Neuroanat 2020; 104:101752. [PMID: 31996329 DOI: 10.1016/j.jchemneu.2020.101752] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 01/19/2020] [Accepted: 01/20/2020] [Indexed: 02/08/2023]
Abstract
Since the discovery of L-dopa in the middle of the 20th century (1960s), there is not any neuroprotective therapy available although significant development has been made in the treatment of symptomatic Parkinson's disease (PD). Neurological disorders like PD can be modelled in animals so as to recapitulates most of the symptoms seen in PD patients. In aging population, PD is the second most common neurodegenerative disease after Alzheimer's disease, even though significant outcomes have been achieved in PD research yet it still is a mystery to solve the treatments for PD. In the last two decades, PD models have provided enhanced precision into the understanding of the process of PD disease, its etiology, pathology, and molecular mechanisms behind it. Furthermore, at the same time as cellular models have helped to recognize specific events, animal models, both toxic and genetic, have replicated almost all of the hallmarks of PD and are very helpful for testing and finding new strategies for neuroprotection. Recently, in both classical and newer models, major advances have been done in the modelling of supplementary PD features have come into the light. In this review, we have try to provide an updated summary of the characteristics of these models related to in vitro and in vivo models, animal models for PD, stem cell model for PD, newer 3D model as well as the strengths and limitations of these most popular PD models.
Collapse
Affiliation(s)
- Sachchida Nand Rai
- Department of Zoology, Mahila Maha Vidhyalaya, Institute of Science, Banaras Hindu University, Varanasi, India.
| | - Payal Singh
- Department of Zoology, Mahila Maha Vidhyalaya, Institute of Science, Banaras Hindu University, Varanasi, India.
| |
Collapse
|
14
|
Si L, Wang H, Wang L. Suppression of miR-193a alleviates neuroinflammation and improves neurological function recovery after traumatic brain injury (TBI) in mice. Biochem Biophys Res Commun 2020; 523:527-534. [PMID: 31924304 DOI: 10.1016/j.bbrc.2019.11.095] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 11/15/2019] [Indexed: 02/06/2023]
Abstract
Traumatic brain injury (TBI) is a leading cause of morbidity and mortality in the world, and is tightly associated with microglia-regulated neuroinflammation. However, the activation profile of microglia during the pathophysiological responses is still not fully understood. Micro-RNAs (miRs), as noncoding RNAs, are involved in the progression of TBI. In this study, we attempted to explore the effects of miR-193a on TBI using the in vivo and in vitro studies. Following experimental TBI in mice, we found that miR-193a expression was significantly up-regulated in ipsilateral cortical tissues and in the microglia/macrophages isolated from the ipsilateral cortical tissues, which was accompanied with markedly enhanced expression of pro-inflammatory factors. We then found that miR-193a hairpin inhibitor (antagomir) markedly reduced lesion volume, brain water contents and neuron death in TBI mice induced by the controlled cortical impact (CCI). In addition, cognitive dysfunction in TBI mice was markedly improved after miR-193a antagomir injection. Of note, CCI-induced activation of microglia was repressed by miR-193a inhibition, along with significantly reduced expression of neuroinflammatory markers, which were associated with the blockage of nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome. The anti-neuroinflammation effects of miR-193a suppression were verified in lipopolysaccharide (LPS)-incubated microglial cells transfected with miR-193a inhibitor. In contrast, LPS-induced activation of microglial cells and the expression of pro-inflammatory factors was markedly further accelerated by the transfection of miR-193a mimic. Taken together, TBI resulted in a robust neuroinflammatory response that was closely associated with the up-regulated miR-193a expression mainly in microglia/macrophages; however, miR-193a suppression significantly alleviated post-traumatic neuroinflammation and cognitive dysfunction. Therefore, miR-193a might be a promising therapeutic target for the treatment of TBI-associated neuroinflammation.
Collapse
Affiliation(s)
- Lili Si
- Department of Neurology, Guangrao County People's Hospital, Dongying City, Shandong Province, 257300, China
| | - Haifeng Wang
- Department of Geriatrics, Tongji Hospital, School of Medicine, Tongji University, Putuo District, Shanghai, 200065, China
| | - Leyuan Wang
- Department of Neurology, Changle People's Hospital, Shandong, 262400, China.
| |
Collapse
|