1
|
Zhang Z, Zhang Y, Cao Y, Chen S, Zhu Y, Wang Y, Peng Y, Chen A, Wu Y. Integrated physiological and transcriptomic analysis reveals osmoregulatory collapse, oxidative stress, and apoptosis drive hypo-salinity-induced mortality in Solen grandis. Int J Biol Macromol 2025; 310:143495. [PMID: 40288715 DOI: 10.1016/j.ijbiomac.2025.143495] [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: 12/21/2024] [Revised: 03/31/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
This study systematically investigated the physiological and molecular responses of the razor clam Solen grandis to acute hypo-salinity stress through integrated survival, enzymatic, histological, and transcriptomic analyses. Exposure to 10 ‰ salinity for 96 h induced severe physiological disruption, reducing survival rates below 10 % and establishing a 13.216 ‰ 96-h LC50 threshold. Hypo-salinity suppressed significantly (p < 0.05) in feeding rate, ammonia excretion, and oxygen consumption compared to controls (28 ‰). Hepatopancreatic histopathology revealed acinar enlargement (3.45-fold area increase), luminal disorganization, and cytoplasmic vacuolization, indicating osmoregulatory failure. Enzymatic profiling demonstrated salinity-dependent dysregulation: Na+/K+-ATPase activity and MDA surged significantly (p < 0.05), while Ca2+/Mg2+-ATPase, antioxidant enzymes (T-SOD, CAT), digestive hydrolases (trypsin, α-AMS, LPS), and lysozyme activity declined significantly (p < 0.05). Transcriptomic analysis identified 2865 differentially expressed genes, highlighting activation of oxidative stress and apoptosis pathways alongside suppression of immune defense and nutrient metabolism networks. These findings delineate a coordinated stress response where energy reallocation to ion homeostasis compromises digestive efficiency and immune capacity, ultimately driving mortality. The study provides mechanistic insights into the ecological vulnerability of intertidal bivalves to salinity fluctuations, offering biomarkers for aquaculture health monitoring and foundational data for coastal ecosystem management under climate-driven hydrological changes.
Collapse
Affiliation(s)
- Zhidong Zhang
- Solen grandis Razor Clam Breeding and Propagation Center of Jiangsu Province, Marine Fisheries Research Institute of Jiangsu Province, Nantong 226007, China; Marine Economic Shellfish Research and Development Center of Jiangsu Province, Marine Fisheries Research Institute of Jiangsu Province, Nantong 226007, China
| | - Yu Zhang
- Solen grandis Razor Clam Breeding and Propagation Center of Jiangsu Province, Marine Fisheries Research Institute of Jiangsu Province, Nantong 226007, China; Marine Economic Shellfish Research and Development Center of Jiangsu Province, Marine Fisheries Research Institute of Jiangsu Province, Nantong 226007, China
| | - Yi Cao
- Solen grandis Razor Clam Breeding and Propagation Center of Jiangsu Province, Marine Fisheries Research Institute of Jiangsu Province, Nantong 226007, China; Marine Economic Shellfish Research and Development Center of Jiangsu Province, Marine Fisheries Research Institute of Jiangsu Province, Nantong 226007, China
| | - Suhua Chen
- Solen grandis Razor Clam Breeding and Propagation Center of Jiangsu Province, Marine Fisheries Research Institute of Jiangsu Province, Nantong 226007, China; Marine Economic Shellfish Research and Development Center of Jiangsu Province, Marine Fisheries Research Institute of Jiangsu Province, Nantong 226007, China
| | - Yanqing Zhu
- Marine Economic Shellfish Research and Development Center of Jiangsu Province, Marine Fisheries Research Institute of Jiangsu Province, Nantong 226007, China
| | - Yuchen Wang
- Solen grandis Razor Clam Breeding and Propagation Center of Jiangsu Province, Marine Fisheries Research Institute of Jiangsu Province, Nantong 226007, China; National Demonstration Centre for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China
| | - Yuheng Peng
- National Demonstration Centre for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China
| | - Aihua Chen
- Solen grandis Razor Clam Breeding and Propagation Center of Jiangsu Province, Marine Fisheries Research Institute of Jiangsu Province, Nantong 226007, China; Marine Economic Shellfish Research and Development Center of Jiangsu Province, Marine Fisheries Research Institute of Jiangsu Province, Nantong 226007, China
| | - Yangping Wu
- Solen grandis Razor Clam Breeding and Propagation Center of Jiangsu Province, Marine Fisheries Research Institute of Jiangsu Province, Nantong 226007, China; Marine Economic Shellfish Research and Development Center of Jiangsu Province, Marine Fisheries Research Institute of Jiangsu Province, Nantong 226007, China.
| |
Collapse
|
2
|
Murray KE, Ravula AR, Stiritz VA, Cominski TP, Delic V, Marín de Evsikova C, Rama Rao KV, Chandra N, Beck KD, Pfister BJ, Citron BA. Sex and Genotype Affect Mouse Hippocampal Gene Expression in Response to Blast-Induced Traumatic Brain Injury. Mol Neurobiol 2025:10.1007/s12035-025-04879-5. [PMID: 40178780 DOI: 10.1007/s12035-025-04879-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 03/21/2025] [Indexed: 04/05/2025]
Abstract
Blast-induced traumatic brain injury (bTBI) has been identified as an increasingly prevalent cause of morbidity and mortality in both military and civilian populations over the past few decades. Functional outcomes following bTBI vary widely among individuals, and chronic neurodegenerative effects including cognitive impairments can develop without effective diagnosis and treatment. Genetic predispositions and sex differences may affect gene expression changes in response to bTBI and influence an individual's probability of sustaining long-term damage or exhibiting resilience and tissue repair. Male and female mice from eight genetically diverse and distinct strains (129S1/SvImJ, A/J, C57BL/6J, CAST/EiJ, NOD/ShiLtJ, NZO/HlLtJ, PWK/PhJ, WSB/EiJ) which encompassed 90% of the genetic variability in commercially available laboratory mice were exposed to a single bTBI (180 kPa) using a well-established shock tube system. Subacute changes in hippocampal gene expression due to blast exposure were assessed using RNA-seq at 1-month post-injury. We identified patterns of dysregulation in gene ontology terms and canonical pathways related to mitochondrial function, ribosomal structure, synaptic plasticity, protein degradation, and intracellular signaling that varied by sex and/or strain, including significant changes in genes encoding respiratory complex I of the electron transport chain in male WSB/EiJ mice and the glutamatergic synapse across more than half of our groups. This study represents a multi-level examination of how genetic variability may influence response to bTBI and provides a foundation for the identification of potential therapeutic targets that could be modulated to improve the health of Veterans and others with histories of blast exposures.
Collapse
Affiliation(s)
- Kathleen E Murray
- Laboratory of Molecular Biology, Research & Development, U.S. Department of Veterans Affairs, VA New Jersey Health Care System, East Orange, NJ, USA
- School of Graduate Studies, Rutgers Health, Newark, NJ, USA
| | - Arun Reddy Ravula
- Molecular Neurotherapeutics Laboratory, Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Victoria A Stiritz
- Neurobehavioral Research Laboratory, Research & Development, U.S. Department of Veterans Affairs, VA New Jersey Health Care System, East Orange, NJ, USA
- School of Graduate Studies, Rutgers Health, Newark, NJ, USA
| | - Tara P Cominski
- Neurobehavioral Research Laboratory, Research & Development, U.S. Department of Veterans Affairs, VA New Jersey Health Care System, East Orange, NJ, USA
- Division of Life Sciences, School of Arts and Sciences, Rutgers University, New Brunswick, NJ, USA
| | - Vedad Delic
- Laboratory of Molecular Biology, Research & Development, U.S. Department of Veterans Affairs, VA New Jersey Health Care System, East Orange, NJ, USA
- School of Graduate Studies, Rutgers Health, Newark, NJ, USA
- Department of Pharmacology, Physiology & Neuroscience, Rutgers-New Jersey Medical School, Newark, NJ, 07101, USA
| | - Caralina Marín de Evsikova
- Epigenetics and Functional Genomics Laboratory, Research & Development, U.S. Department of Veterans Affairs, Bay Pines VA Healthcare System, Bay Pines, FL, USA
- Department of Molecular Medicine, University of South Florida, Tampa, FL, USA
| | - Kakulavarapu V Rama Rao
- Center for Injury Biomechanics, Materials, and Medicine, Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Namas Chandra
- Center for Injury Biomechanics, Materials, and Medicine, Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Kevin D Beck
- Neurobehavioral Research Laboratory, Research & Development, U.S. Department of Veterans Affairs, VA New Jersey Health Care System, East Orange, NJ, USA
- School of Graduate Studies, Rutgers Health, Newark, NJ, USA
- Department of Pharmacology, Physiology & Neuroscience, Rutgers-New Jersey Medical School, Newark, NJ, 07101, USA
| | - Bryan J Pfister
- Center for Injury Biomechanics, Materials, and Medicine, Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Bruce A Citron
- Laboratory of Molecular Biology, Research & Development, U.S. Department of Veterans Affairs, VA New Jersey Health Care System, East Orange, NJ, USA.
- School of Graduate Studies, Rutgers Health, Newark, NJ, USA.
- Department of Pharmacology, Physiology & Neuroscience, Rutgers-New Jersey Medical School, Newark, NJ, 07101, USA.
| |
Collapse
|
3
|
Bhat AA, Moglad E, Goyal A, Afzal M, Thapa R, Almalki WH, Kazmi I, Alzarea SI, Ali H, Gaur A, Singh TG, Singh SK, Dua K, Gupta G. Nrf2 pathways in neuroprotection: Alleviating mitochondrial dysfunction and cognitive impairment in aging. Life Sci 2024; 357:123056. [PMID: 39277133 DOI: 10.1016/j.lfs.2024.123056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/27/2024] [Accepted: 09/11/2024] [Indexed: 09/17/2024]
Abstract
Mitochondrial dysfunction and cognitive impairment are widespread phenomena among the elderly, being crucial factors that contribute to neurodegenerative diseases. Nuclear factor erythroid 2-related factor 2 (Nrf2) is an important regulator of cellular defense systems, including that against oxidative stress. As such, increased Nrf2 activity may serve as a strategy to avert mitochondrial dysfunction and cognitive decline. Scientific data on Nrf2-mediated neuroprotection was collected from PubMed, Google Scholar, and Science Direct, specifically addressing mitochondrial dysfunction and cognitive impairment in older people. Search terms included "Nrf2", "mitochondrial dysfunction," "cognitive impairment," and "neuroprotection." Studies focusing on in vitro and in vivo models and clinical investigations were included to review Nrf2's therapeutic potential comprehensively. The relative studies have demonstrated that increased Nrf2 activity could improve mitochondrial performance, decrease oxidative pressure, and mitigate cognitive impairment. To a large extent, this is achieved through the modulation of critical cellular signalling pathways such as the Keap1/Nrf2 pathway, mitochondrial biogenesis, and neuroinflammatory responses. The present review summarizes the recent progress in comprehending the molecular mechanisms regarding the neuroprotective benefits mediated by Nrf2 through its substantial role against mitochondrial dysfunction and cognitive impairment. This review also emphasizes Nrf2-target pathways and their contribution to cognitive function improvement and rescue from mitochondria-related abnormalities as treatment strategies for neurodegenerative diseases that often affect elderly individuals.
Collapse
Affiliation(s)
- Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, U.P., India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589 Jeddah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, 72341 Sakaka, Al-Jouf, Saudi Arabia
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Ashish Gaur
- Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India; Graphic Era Hill University, Clement Town, Dehradun 248002, India
| | | | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates.
| |
Collapse
|
4
|
Guo B, Zheng C, Cao J, Qiu X, Luo F, Li H, Lee SM, Yang X, Zhang G, Sun Y, Zhang Z, Wang Y. Tetramethylpyrazine Nitrone Promotes the Clearance of Alpha-Synuclein via Nrf2-Mediated Ubiquitin-Proteasome System Activation. Neuromolecular Med 2024; 26:9. [PMID: 38568291 DOI: 10.1007/s12017-024-08775-4] [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: 10/29/2023] [Accepted: 02/21/2024] [Indexed: 04/05/2024]
Abstract
Aggregation of α-synuclein (α-syn) and α-syn cytotoxicity are hallmarks of sporadic and familial Parkinson's disease (PD). Nuclear factor (erythroid-derived 2)-like 2 (Nrf2)-dependent enhancement of the expression of the 20S proteasome core particles (20S CPs) and regulatory particles (RPs) increases proteasome activity, which can promote α-syn clearance in PD. Activation of peroxisome proliferator-activated receptor γ co-activator 1α (PGC-1α) may reduce oxidative stress by strongly inducing Nrf2 gene expression. In the present study, tetramethylpyrazine nitrone (TBN), a potent-free radical scavenger, promoted α-syn clearance by the ubiquitin-proteasome system (UPS) in cell models overexpressing the human A53T mutant α-syn. In the α-syn transgenic mice model, TBN improved motor impairment, decreased the products of oxidative damage, and down-regulated the α-syn level in the serum. TBN consistently up-regulated PGC-1α and Nrf2 expression in tested models of PD. Additionally, TBN similarly enhanced the proteasome 20S subunit beta 8 (Psmb8) expression, which is linked to chymotrypsin-like proteasome activity. Furthermore, TBN increased the mRNA levels of both the 11S RPs subunits Pa28αβ and a proteasome chaperone, known as the proteasome maturation protein (Pomp). Interestingly, specific siRNA targeting of Nrf2 blocked TBN's effects on Psmb8, Pa28αβ, Pomp expression, and α-syn clearance. In conclusion, TBN promotes the clearance of α-syn via Nrf2-mediated UPS activation, and it may serve as a potentially disease-modifying therapeutic agent for PD.
Collapse
Affiliation(s)
- Baojian Guo
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-Cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, 601# Huangpu Road, Guangzhou, 510632, China
| | - Chengyou Zheng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-Cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, 601# Huangpu Road, Guangzhou, 510632, China
- School of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen, 518055, China
| | - Jie Cao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-Cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, 601# Huangpu Road, Guangzhou, 510632, China
| | - Xiaoling Qiu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-Cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, 601# Huangpu Road, Guangzhou, 510632, China
| | - Fangcheng Luo
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-Cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, 601# Huangpu Road, Guangzhou, 510632, China
| | - Haitao Li
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China
| | - Simon Mingyuan Lee
- Institute of Chinese Medical Sciences and State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Avenida da Universidade, Taipa, 999078, Macao SAR, China
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Center for Disease Control and Prevention, No. 8, Longyuan Road, Nanshan District, Shenzhen, 518055, China
| | - Gaoxiao Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-Cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, 601# Huangpu Road, Guangzhou, 510632, China
| | - Yewei Sun
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-Cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, 601# Huangpu Road, Guangzhou, 510632, China.
| | - Zaijun Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-Cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, 601# Huangpu Road, Guangzhou, 510632, China.
| | - Yuqiang Wang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-Cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, 601# Huangpu Road, Guangzhou, 510632, China
| |
Collapse
|
5
|
Wang P, Fan S, Hu X, Luo L, Ying J, Li J. MG132, Attenuates the Retinal Vascular Injury Through the Upregulation of Nrf2 Expression. J Ocul Pharmacol Ther 2023; 39:661-671. [PMID: 37729070 DOI: 10.1089/jop.2023.0053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2023] Open
Abstract
Purpose: This study clarifies the beneficial effects of MG132, a proteasomal inhibitor, on retinal vascular injury mediated by diabetes-induced oxidative stress through nuclear factor erythroid 2-related factor 2 (Nrf2). Methods: Diabetic rats and control animals were randomly assigned to receive MG132 or vehicle for 24 weeks, and human retinal endothelial cells (HRECs) were incubated with normal or high glucose with or without MG132. 26S proteasome activity in the rat retinas or cultured HRECs was measured using Suc-LLVY-7-amido-4-methylcoumarin. NADPH-quinone oxidoreduc-tase (NQO1), heme oxygenase (HO)-1, kelch-like ECH-associated protein 1 (Keap1) and Nrf2 were examined by Western blotting and real-time reverse transcription polymerase chain reaction. Cell apoptosis is measured through flow cytometry assay, mitochondrial reactive oxygen species (ROS) production, and retinal vascular leakage were assayed using CM-H2DCFDA fluorescent probes and Evans blue, respectively. Results: MG132 significantly inhibited the activation of 26S proteasome induced by diabetes or elevated glucose, and subsequently increased the expression of Nrf2, NQO1, and HO-1, and further reduced ROS accumulation. These changes were associated with a decrease of diabetes-induced retinal vascular leakage and retinal capillary cell apoptosis. Conclusions: MG132 decreases diabetes-induced 26S proteasome activation and exerts protective effects against retinal microvascular dysfunction in diabetic rats in association with the alleviation of retinal oxidative stress mediated by Nrf2.
Collapse
Affiliation(s)
- Peipei Wang
- Department of Ophthalmology, Lishui Municipal Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, P.R. China
- Department of Stomatology, Lishui Hospital, Zhejiang University School of Medicine, Lishui, P.R. China
| | - Shipei Fan
- Department of Ophthalmology, Lishui Municipal Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, P.R. China
- Department of Ophthalmology, Lishui Hospital, Zhejiang University School of Medicine, Lishui, P.R. China
| | - Xin Hu
- Department of Ophthalmology, Lishui Municipal Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, P.R. China
- Department of Ophthalmology, Lishui Hospital, Zhejiang University School of Medicine, Lishui, P.R. China
| | - Li Luo
- Department of Ophthalmology, Lishui Municipal Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, P.R. China
- Department of Ophthalmology, Lishui Hospital, Zhejiang University School of Medicine, Lishui, P.R. China
| | - Jia Ying
- Department of Ophthalmology, Lishui Municipal Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, P.R. China
- Department of Ophthalmology, Lishui Hospital, Zhejiang University School of Medicine, Lishui, P.R. China
| | - Jun Li
- Department of Ophthalmology, Lishui Municipal Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, P.R. China
- Department of Ophthalmology, Lishui Hospital, Zhejiang University School of Medicine, Lishui, P.R. China
| |
Collapse
|
6
|
Muneer PMA. Nrf2 as a Potential Therapeutic Target for Traumatic Brain Injury. J Integr Neurosci 2023; 22:81. [PMID: 37519172 DOI: 10.31083/j.jin2204081] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/07/2023] [Accepted: 03/20/2023] [Indexed: 08/01/2023] Open
Abstract
In this review, we discuss the possibility and feasibility of nuclear factor erythroid 2-related factor 2 (Nrf2) as a therapeutic target to minimize the devastating effects of a brain injury. To complete this review, comprehensive literature searches were conducted in MEDLINE, PubMed, Embase, and PsycINFO databases for English scientific peer-reviewed articles through December 2022. This short review addressed the different sources of oxidative stress and its effects on blood-brain barrier (BBB) dysfunction, mitochondrial damage, and changes in a variety of inflammatory molecules associated with central nervous system (CNS) injury. At last, we explained the potential efficacy of the Nrf2 transcription factor in reducing oxidative stress-mediated secondary damages after a CNS injury. The role of CPUY192018, an inhibitor of Nrf2-Keap1 protein-protein interaction in protecting the injured brain cells is given as evidence of Nrf2's role in activating antioxidant genes. Overall, the scope of Nrf2 in developing therapeutic interventions for a variety of pathophysiological conditions associated with CNS injury-induced free radical/inflammatory signaling is acknowledged. Nrf2 has a widespread application in basic and clinical neuroscience for understanding and treating free radical/inflammatory signaling disorders, including neurological diseases. The development of innovative therapeutic strategies using Nrf2-inducing agents can be applied to reduce the complications of TBI before advancing it to posttraumatic stress disorder (PTSD).
Collapse
Affiliation(s)
- P M Abdul Muneer
- Laboratory of CNS Injury and Molecular Therapy, JFK Neuroscience Institute, Hackensack Meridian JFK University Medical Center, Edison, NJ 08820, USA
| |
Collapse
|
7
|
Cheng H, Wang P, Wang N, Dong W, Chen Z, Wu M, Wang Z, Yu Z, Guan D, Wang L, Zhao R. Neuroprotection of NRF2 against Ferroptosis after Traumatic Brain Injury in Mice. Antioxidants (Basel) 2023; 12:731. [PMID: 36978979 PMCID: PMC10044792 DOI: 10.3390/antiox12030731] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/11/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023] Open
Abstract
Ferroptosis and iron-related redox imbalance aggravate traumatic brain injury (TBI) outcomes. NRF2 is the predominant transcription factor regulating oxidative stress and neuroinflammation in TBI, but its role in iron-induced post-TBI damage is unclear. We investigated ferroptotic neuronal damage in the injured cortex and observed neurological deficits post-TBI. These were ameliorated by the iron chelator deferoxamine (DFO) in wild-type mice. In Nrf2-knockout (Nrf2-/-) mice, more sever ferroptosis and neurological deficits were detected. Dimethyl fumarate (DMF)-mediated NRF2 activation alleviated neural dysfunction in TBI mice, partly due to TBI-induced ferroptosis mitigation. Additionally, FTH-FTL and FSP1 protein levels, associated with iron metabolism and the ferroptotic redox balance, were highly NRF2-dependent post-TBI. Thus, NRF2 is neuroprotective against TBI-induced ferroptosis through both the xCT-GPX4- and FTH-FTL-determined free iron level and the FSP1-regulated redox status. This yields insights into the neuroprotective role of NRF2 in TBI-induced neuronal damage and its potential use in TBI treatment.
Collapse
Affiliation(s)
- Hao Cheng
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
- School of Forensic Medicine, Kunming Medical University, Kunming 650500, China
| | - Pengfei Wang
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
- Collaborative Laboratory of Intelligentized Forensic Science, Shenyang 110034, China
| | - Ning Wang
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
- Collaborative Laboratory of Intelligentized Forensic Science, Shenyang 110034, China
| | - Wenwen Dong
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
- Collaborative Laboratory of Intelligentized Forensic Science, Shenyang 110034, China
| | - Ziyuan Chen
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
- Collaborative Laboratory of Intelligentized Forensic Science, Shenyang 110034, China
| | - Mingzhe Wu
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
- Collaborative Laboratory of Intelligentized Forensic Science, Shenyang 110034, China
| | - Ziwei Wang
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
- Collaborative Laboratory of Intelligentized Forensic Science, Shenyang 110034, China
| | - Ziqi Yu
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
- Collaborative Laboratory of Intelligentized Forensic Science, Shenyang 110034, China
| | - Dawei Guan
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang 110122, China
- Collaborative Laboratory of Intelligentized Forensic Science, Shenyang 110034, China
| | - Linlin Wang
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang 110122, China
- Collaborative Laboratory of Intelligentized Forensic Science, Shenyang 110034, China
| | - Rui Zhao
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang 110122, China
- Collaborative Laboratory of Intelligentized Forensic Science, Shenyang 110034, China
| |
Collapse
|
8
|
Bai X, Bian Z, Zhang M. Targeting the Nrf2 signaling pathway using phytochemical ingredients: A novel therapeutic road map to combat neurodegenerative diseases. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 109:154582. [PMID: 36610130 DOI: 10.1016/j.phymed.2022.154582] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/11/2022] [Accepted: 11/29/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Nuclear factor erythroid 2-related factor 2 (Nrf2) is a classical nuclear transcription factor that regulates the system's anti-oxidative stress response. The activation of Nrf2 induces the expression of antioxidant proteins and improves the system's anti-oxidative stress ability. Accumulating evidence suggests that Nrf2-centered signaling pathways may be a key pharmacological target for the treatment of neurodegenerative diseases (NDDs). However, phytochemicals as new therapeutic agents against NDDs have not been clearly delineated. PURPOSE To review the therapeutic effects of phytochemical ingredients on NDDs by activating Nrf2 and reducing oxidative stress injury. METHODS A comprehensive search of published articles was performed using various literature databases including PubMed, Google Scholar, and China National Knowledge Infrastructure. The search terms included "Nrf2", "phytochemical ingredients", "natural bioactive agents", "neurodegenerative diseases", "Antioxidant", "Alzheimer's disease", "Parkinson's disease", "Huntington's disease", "amyotrophic lateral sclerosis" "multiple sclerosis", "toxicity", and combinations of these keywords. A total of 769 preclinical studies were retrieved until August 2022, and we included 39 of these articless on phytochemistry, pharmacology, toxicology and other fields. RESULTS Numerous in vivo and in vitro studies showed that phytochemical ingredients could act as an Nrf2 activator in the treatment of NDDs through the antioxidant defense mechanism. These phytochemical ingredients, such as salidroside, naringenin, resveratrol, sesaminol, ellagic acid, ginsenoside Re, tanshinone I, sulforaphane, curcumin, naringin, tetramethylpyrazine, withametelin, magnolol, piperine, and myricetin, had the potential to improve Nrf2 signaling, thereby combatting NDDs. CONCLUSION As Nrf2 activators, phytochemical ingredients may provide a novel potential strategy for the treatment of NDDs. Here, we reviewed the interaction between phytochemical ingredients, Nrf2, and its antioxidant damaging pathway in NDDs and explored the advantages of phytochemical ingredients in anti-oxidative stress, which provides a reliable basis for improving the treatment of NDDs. However, further clinical trials are needed to determine the safety and efficacy of Nrf2 activators for NDDs.
Collapse
Affiliation(s)
- Xue Bai
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, 110004, Shenyang, Liaoning, PR China
| | - Zhigang Bian
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, 110004, Shenyang, Liaoning, PR China
| | - Meng Zhang
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, 110004, Shenyang, Liaoning, PR China.
| |
Collapse
|
9
|
Wang Q, Botchway BOA, Zhang Y, Liu X. Ellagic acid activates the Keap1-Nrf2-ARE signaling pathway in improving Parkinson's disease: A review. Biomed Pharmacother 2022; 156:113848. [PMID: 36242848 DOI: 10.1016/j.biopha.2022.113848] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/26/2022] [Accepted: 10/06/2022] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is a familiar neurodegenerative disease, accompanied by motor retardation, static tremor, memory decline and dementia. Heredity, environment, age and oxidative stress have been suggested as key factors in the instigation of PD. The Keap1-Nrf2-ARE signaling is one of the most significant anti- oxidative stress (OS) pathways. The Keap1 is a negative regulator of the Nrf2. The Keap1-Nrf2-ARE pathway can induce cell oxidation resistance and reduce nerve injury to treat neurodegenerative diseases. Ellagic acid (EA) can inhibit the Keap1 to accumulate the Nrf2 in the nucleus, and act on the ARE to produce target proteins, which in turn may alleviate the impact of OS on neuronal cells of PD. This review analyzes the structure and physiological role of EA, along with the structure, composition and functions of the Keap1-Nrf2-ARE signaling pathway. We further expound on the mechanism of ellagic acid in its activation of the Keap1-Nrf2-ARE signaling pathway, as well as the relationship between EA in impairing the TLR4/Myd88/NF-κB and Nrf2 pathways. Ellagic acid has the potentiality of improving PD by activating the Keap1-Nrf2-ARE signaling pathway and scavenging free radicals.
Collapse
Affiliation(s)
- Qianhui Wang
- Department of Histology and Embryology, Medical College, Shaoxing University, Zhejiang, China
| | - Benson O A Botchway
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
| | - Yong Zhang
- Department of Histology and Embryology, Medical College, Shaoxing University, Zhejiang, China
| | - Xuehong Liu
- Department of Histology and Embryology, Medical College, Shaoxing University, Zhejiang, China.
| |
Collapse
|
10
|
Han Y, Wang C, Li X, Liang G. Oleanolic acid reduces oxidative stress and neuronal apoptosis after experimental subarachnoid hemorrhage by regulating Nrf2/HO-1 pathway. Drug Dev Res 2022; 83:680-687. [PMID: 34820872 DOI: 10.1002/ddr.21899] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/10/2021] [Accepted: 11/15/2021] [Indexed: 11/11/2022]
Abstract
Oxidative stress is an early major pathological feature after subarachnoid hemorrhage (SAH) and involves in the development of acute brain injury, neuronal apoptosis and cerebral vasospasm following SAH. Antioxidant stress is an effective way to improve the prognosis of SAH. Oleanolic acid is a widely used triterpenoid from plants, which has strong antioxidant activities, hepatoprotective, anti-inflammatory and anti-cancer activities. However, whether oleanolic acid exerts its anti-oxidant effect after SAH and the underlying mechanisms involved in it is unclear. In current study, the SAH model was established on Sprague Dawley rats using a standard intravascular puncture model. We found OA treatment significantly reduced malondialdehyde levels, and increased the levels of superoxide dismutase, catalase and GSH-Px after SAH, and reduced many EBI-related indicators, including brain edema, BBB disruption, SAH grades, and neurological score. In addition, the activation of Nrf2/HO-1 pathway after SAH was also detected. And by using Nrf2 siRNA intracerebroventricular injections, apoptosis related factors downstream of Nrf2/HO-1 pathway were detected. By TUNEL staining, OA treatment obviously reduced neuronal apoptosis. Therefore, we suggest that OA could alleviate oxidative stress and reduce neuronal apoptosis through activating Nrf 2/HO-1 pathway.
Collapse
Affiliation(s)
- Yuwei Han
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Chenchen Wang
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Xiaoming Li
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Guobiao Liang
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| |
Collapse
|
11
|
Mi Z, Liu H, Rose ME, Ma J, Reay DP, Ma X, Henchir JJ, Dixon CE, Graham SH. Mutation of a Ubiquitin Carboxy Terminal Hydrolase L1 Lipid Binding Site Alleviates Cell Death, Axonal Injury, and Behavioral Deficits After Traumatic Brain Injury in Mice. Neuroscience 2021; 475:127-136. [PMID: 34508847 DOI: 10.1016/j.neuroscience.2021.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/19/2021] [Accepted: 09/01/2021] [Indexed: 11/17/2022]
Abstract
Ubiquitin carboxy terminal hydrolase L1 (UCHL1) is a protein highly expressed in neurons that may play important roles in the ubiquitin proteasome pathway (UPP) in neurons, axonal integrity, and motor function after traumatic brain injury (TBI). Binding of reactive lipid species to cysteine 152 of UCHL1 results in unfolding, aggregation, and inactivation of the enzyme. To test the role of this mechanism in TBI, mice bearing a cysteine to alanine mutation at site 152 (C152A mice) that renders UCHL1 resistant to inactivation by reactive lipids were subjected to the controlled cortical impact model (CCI) of TBI and compared to wild type (WT) controls. Alterations in protein ubiquitination and activation of autophagy pathway markers in traumatized brain were detected by immunoblotting. Cell death and axonal injury were determined by histological assessment and anti-amyloid precursor protein (APP) immunohistochemistry. Behavioral outcomes were determined using the beam balance and Morris water maze tests. C152A mice had reduced accumulation of ubiquitinated proteins, decreased activation of the autophagy markers Beclin-1 and LC3B, a decreased number of abnormal axons, decreased CA1 cell death, and improved motor and cognitive function compared to WT controls after CCI; no significant change in spared tissue volume was observed. These results suggest that binding of lipid substrates to cysteine 152 of UCHL1 is important in the pathogenesis of injury and recovery after TBI and may be a novel target for future therapeutic approaches.
Collapse
Affiliation(s)
- Zhiping Mi
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA.
| | - Hao Liu
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA
| | - Marie E Rose
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA.
| | - Jie Ma
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA.
| | - Daniel P Reay
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA.
| | - Xiecheng Ma
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurosurgery, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Department of Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Jeremy J Henchir
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurosurgery, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Department of Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA.
| | - C Edward Dixon
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurosurgery, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Department of Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA.
| | - Steven H Graham
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA; Department of Neurology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
12
|
Buhlman LM, Krishna G, Jones TB, Thomas TC. Drosophila as a model to explore secondary injury cascades after traumatic brain injury. Biomed Pharmacother 2021; 142:112079. [PMID: 34463269 PMCID: PMC8458259 DOI: 10.1016/j.biopha.2021.112079] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/11/2021] [Accepted: 08/17/2021] [Indexed: 12/14/2022] Open
Abstract
Drosophilae are emerging as a valuable model to study traumatic brain injury (TBI)-induced secondary injury cascades that drive persisting neuroinflammation and neurodegenerative pathology that imposes significant risk for long-term neurological deficits. As in mammals, TBI in Drosophila triggers axonal injury, metabolic crisis, oxidative stress, and a robust innate immune response. Subsequent neurodegeneration stresses quality control systems and perpetuates an environment for neuroprotection, regeneration, and delayed cell death via highly conserved cell signaling pathways. Fly injury models continue to be developed and validated for both whole-body and head-specific injury to isolate, evaluate, and modulate these parallel pathways. In conjunction with powerful genetic tools, the ability for longitudinal evaluation, and associated neurological deficits that can be tested with established behavioral tasks, Drosophilae are an attractive model to explore secondary injury cascades and therapeutic intervention after TBI. Here, we review similarities and differences between mammalian and fly pathophysiology and highlight strategies for their use in translational neurotrauma research.
Collapse
Affiliation(s)
- Lori M Buhlman
- Biomedical Sciences Program, Midwestern University, Glendale, AZ, USA.
| | - Gokul Krishna
- Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA; Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA
| | - T Bucky Jones
- Department of Anatomy, Midwestern University, Glendale, AZ, USA
| | - Theresa Currier Thomas
- Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA; Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Phoenix VA Health Care System, Phoenix, AZ, USA.
| |
Collapse
|
13
|
Saikumar J, Bonini NM. Synergistic effects of brain injury and aging: common mechanisms of proteostatic dysfunction. Trends Neurosci 2021; 44:728-740. [PMID: 34301397 DOI: 10.1016/j.tins.2021.06.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/26/2021] [Accepted: 06/08/2021] [Indexed: 01/09/2023]
Abstract
The aftermath of TBI is associated with an acute stress response and the accumulation of insoluble protein aggregates. Even after the symptoms of TBI are resolved, insidious molecular processes continue to develop, which often ultimately result in the development of age-associated neurodegenerative disorders. The precise molecular cascades that drive unhealthy brain aging are still largely unknown. In this review, we discuss proteostatic dysfunction as a converging mechanism contributing to accelerated brain aging after TBI. We examine evidence from human tissue and in vivo animal models, spanning both the aging and injury contexts. We conclude that TBI has a sustained debilitating effect on the proteostatic machinery, which may contribute to the accelerated pathological and cognitive hallmarks of aging that are observed following injury.
Collapse
Affiliation(s)
- Janani Saikumar
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nancy M Bonini
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
14
|
Ghiam MK, Patel SD, Hoffer A, Selman WR, Hoffer BJ, Hoffer ME. Drug Repurposing in the Treatment of Traumatic Brain Injury. Front Neurosci 2021; 15:635483. [PMID: 33833663 PMCID: PMC8021906 DOI: 10.3389/fnins.2021.635483] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/19/2021] [Indexed: 01/02/2023] Open
Abstract
Traumatic brain injury (TBI) is the most common cause of morbidity among trauma patients; however, an effective pharmacological treatment has not yet been approved. Individuals with TBI are at greater risk of developing neurological illnesses such as Alzheimer's disease (AD) and Parkinson's disease (PD). The approval process for treatments can be accelerated by repurposing known drugs to treat the growing number of patients with TBI. This review focuses on the repurposing of N-acetyl cysteine (NAC), a drug currently approved to treat hepatotoxic overdose of acetaminophen. NAC also has antioxidant and anti-inflammatory properties that may be suitable for use in therapeutic treatments for TBI. Minocycline (MINO), a tetracycline antibiotic, has been shown to be effective in combination with NAC in preventing oligodendrocyte damage. (-)-phenserine (PHEN), an anti-acetylcholinesterase agent with additional non-cholinergic neuroprotective/neurotrophic properties initially developed to treat AD, has demonstrated efficacy in treating TBI. Recent literature indicates that NAC, MINO, and PHEN may serve as worthwhile repositioned therapeutics in treating TBI.
Collapse
Affiliation(s)
- Michael K. Ghiam
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Shrey D. Patel
- University of Miami Miller School of Medicine, Miami, FL, United States
| | - Alan Hoffer
- Department of Neurological Surgery, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Warren R. Selman
- Department of Neurological Surgery, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Barry J. Hoffer
- Department of Neurological Surgery, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Michael E. Hoffer
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
15
|
Zhang Z, Yu J, Wang P, Lin L, Liu R, Zeng R, Ma H, Zhao Y. iTRAQ-based proteomic profiling reveals protein alterations after traumatic brain injury and supports thyroxine as a potential treatment. Mol Brain 2021; 14:25. [PMID: 33504361 PMCID: PMC7839205 DOI: 10.1186/s13041-021-00739-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 01/16/2021] [Indexed: 12/25/2022] Open
Abstract
Traumatic brain injury (TBI) is a primary cause of disability and death across the world. Previously, RNA analysis was widely used to study the pathophysiological mechanisms underlying TBI; however, the relatively low correlation between the transcriptome and proteome revealed that RNA transcription abundance does not reliably predict protein abundance, which led to the emergence of proteomic research. In this study, an iTRAQ proteomics approach was applied to detect protein alterations after TBI on a large scale. A total of 3937 proteins were identified, and 146 proteins were significantly changed after TBI. Moreover, 23 upregulated proteins were verified by parallel reaction monitoring (PRM), and fold changes in 16 proteins were consistent with iTRAQ outcomes. Transthyretin (Ttr) upregulation has been demonstrated at the transcriptional level, and this study further confirmed this at the protein level. After treatment with thyroxine (T4), which is transported by Ttr, the effects of T4 on neuronal histopathology and behavioral performance were determined in vivo (TBI + T4 group). Brain edema was alleviated, and the integrity of the blood brain barrier (BBB) improved. Escape latency in the Morris water maze (MWM) declined significantly compared with the group without T4 treatment. Modified neurological severity scores (mNSS) of the TBI + T4 group decreased from day 1 to day 7 post-TBI compared with the TBI + saline group. These results indicate that T4 treatment has potential to alleviate pathologic and behavioral abnormalities post-TBI. Protein alterations after T4 treatment were also detected by iTRAQ proteomics. Upregulation of proteins like Lgals3, Gfap and Apoe after TBI were reversed by T4 treatment. GO enrichment showed T4 mainly affected intermediate filament organization, cholesterol transportation and axonal regeneration. In summary, iTRAQ proteomics provides information about the impact of TBI on protein alterations and yields insight into underlying mechanisms and pathways involved in TBI and T4 treatment. Finally, Ttr and other proteins identified by iTRAQ may become potential novel treatment targets post-TBI.
Collapse
Affiliation(s)
- Zhongxiang Zhang
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
- Hubei Clinical Research Center for Emergency and Resuscitation, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
| | - Jiangtao Yu
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
- Hubei Clinical Research Center for Emergency and Resuscitation, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
| | - Pengcheng Wang
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
- Hubei Clinical Research Center for Emergency and Resuscitation, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
| | - Lian Lin
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
- Hubei Clinical Research Center for Emergency and Resuscitation, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
| | - Ruining Liu
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
- Hubei Clinical Research Center for Emergency and Resuscitation, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
| | - Rong Zeng
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
- Hubei Clinical Research Center for Emergency and Resuscitation, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
| | - Haoli Ma
- Hubei Clinical Research Center for Emergency and Resuscitation, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
| | - Yan Zhao
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
- Hubei Clinical Research Center for Emergency and Resuscitation, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China
| |
Collapse
|
16
|
Cheng J, Zhang M, Cheng S, Li F, Zhang B, Sun X, Hu H, Chen L, Zhao Z, Hu H, Zhang Z. Low-dose alcohol ameliorated high fat diet-induced anxiety-related behavior via enhancing adiponectin expression and activating the Nrf2 pathway. Food Funct 2021; 12:241-251. [PMID: 33295905 DOI: 10.1039/d0fo02704a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Long-term high-fat-diet (HFD)-induced obesity is associated with many comorbidities, such as cognitive impairment and anxiety, which are increasing public health burdens that have gained prevalence in adolescents. Although low-dose alcohol could attenuate the risk of cardiovascular disease, its mechanism on HFD-induced anxiety-related behavior remains not clear. The mice were divided into 4 groups, Control (Con), Alcohol (Alc), HFD and HFD + Alc groups. To verify the effects of low-dose alcohol on HFD-induced anxiety-related behavior, the mice were fed with HFD for 16 weeks. At the beginning of week 13, the HFD-fed mice were administered intragastrically with low-dose alcohol (0.8 g kg-1) for 4 weeks. After 4 weeks of oral administration, low-dose alcohol decreased body weight and Lee's index in HFD-induced obese mice. Moreover, low-dose alcohol alleviated the anxiety-related behaviors of obese mice in the open field test and the elevated plus maze test. The HFD-induced damage to the hippocampus was improved in hematoxylin-eosin staining assay in mice. In addition, low-dose alcohol also suppressed HFD-induced oxidative stress and increased HFD-suppressed adiponectin (APN) expression and nuclear factor erythroid 2-related factor 2 (Nrf2) activation in the hippocampus. Taken together, low-dose alcohol significantly ameliorates HFD-induced obesity, oxidative stress and anxiety-related behavior in mice, which might be related to APN upregulation, Nrf2 activation and related antioxidase expression including SOD1, HO-1, and catalase.
Collapse
Affiliation(s)
- Jie Cheng
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| | - Meng Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| | - Shaoli Cheng
- Basic Medical Experiment Teaching Center, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Fan Li
- Basic Medical Experiment Teaching Center, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Bingyi Zhang
- Basic Medical Experiment Teaching Center, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xiaoming Sun
- Basic Medical Experiment Teaching Center, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Huijuan Hu
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| | - Lina Chen
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China. and Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China, Xi'an, Shaanxi 710061, China
| | - Zhenghang Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China. and Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China, Xi'an, Shaanxi 710061, China
| | - Hao Hu
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China. and Basic Medical Experiment Teaching Center, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China and Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China, Xi'an, Shaanxi 710061, China
| | - Zhanqin Zhang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
17
|
Xu C, Zhang M, Zhang G, Yan S, Yan W. Hydrogen Sulfide Improves Functional Recovery in Rat Traumatic Spinal Cord Injury Model by Inducing Nuclear Translocation of NF-E2-Related Factor 2. Biol Pharm Bull 2021; 44:1093-1100. [PMID: 34334495 DOI: 10.1248/bpb.b21-00259] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hydrogen sulfide (H2S), an important gaseous messenger, is known to have neuroprotective effects in many neurological disorders. This study examined the neuroprotective effects and the associated mechanisms of H2S in the model Sprague-Dawley (SD) rats with spinal cord injury (SCI). We found that H2S showed neuroprotective effects in SCI model rats, improved the symptoms of neurological impairment, reduced the secretion of inflammatory factors, nerve cell apoptosis, and endoplasmic reticulum (ER), and oxidative stresses. Moreover, these effects were produced by activation of nuclear factor-erythroid 2-related factor 2 (Nrf2) protein. Our results suggest that H2S supplementation could be a potential therapeutic strategy to promote SCI recovery.
Collapse
Affiliation(s)
- Chang Xu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine
| | - Man Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine
| | - Guangping Zhang
- Department of Orthopedic Surgery, The Second Traditional Chinese Medicine Hospital of Yibin City
| | - Sheng Yan
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine
| | - Weiqi Yan
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine
| |
Collapse
|
18
|
Mi Z, Liu H, Rose ME, Ma X, Reay DP, Ma J, Henchir J, Dixon CE, Graham SH. Abolishing UCHL1's hydrolase activity exacerbates TBI-induced axonal injury and neuronal death in mice. Exp Neurol 2020; 336:113524. [PMID: 33159930 DOI: 10.1016/j.expneurol.2020.113524] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 10/14/2020] [Accepted: 10/30/2020] [Indexed: 01/13/2023]
Abstract
Ubiquitin (Ub) C-terminal hydrolase L1 (UCHL1) is a multifunctional protein that is expressed in neurons throughout brain at high levels. UCHL1 deletion is associated with axonal degeneration, progressive sensory motor ataxia, and premature death in mice. UCHL1 has been hypothesized to play a role in the pathogenesis of neurodegenerative diseases and recovery after neuronal injury. UCHL1 hydrolyzes Ub from polyubiquitinated (poly-Ub) proteins, but also may ligate Ub to select neuronal proteins, and interact with cytoskeletal proteins. These and other mechanisms have been hypothesized to underlie UCHL1's role in neurodegeneration and response to brain injury. A UCHL1 knockin mouse containing a C90A mutation (C90A) devoid of hydrolase activity was constructed. The C90A mouse did not develop the sensory and motor deficits, degeneration of the gracile nucleus and tract, or premature death as seen in UCHL1 deficient mice. C90A and wild type (WT) mice were subjected to the controlled cortical impact (CCI) model of traumatic brain injury (TBI), and cell death, axonal injury and behavioral outcome were assessed. C90A mice exhibited decreased spared tissue volume, greater loss of CA1 hippocampal neurons and greater axonal injury as detected using anti-amyloid precursor protein (APP) antibody and anti- non-phosphorylated neurofilament H (SMI-32) antibody immunohistochemistry after CCI compared to WT controls. Poly-Ub proteins and Beclin-1 were elevated after CCI in C90A mice compared to WT controls. Vestibular motor deficits assessed using the beam balance test resolved by day 5 after CCI in WT mice but not in C90A mice. These results suggest that the hydrolase activity of UCHL1 does not account for the progressive neurodegeneration and premature death seen in mice that do not express full length UCHL1. The hydrolase activity of UCHL1 contributes to the function of the ubiquitin proteasome pathway (UPP), ameliorates activation of autophagy, and improves motor recovery after CCI. Thus, UCHL1 hydrolase activity plays an important role in acute injury response after TBI.
Collapse
Affiliation(s)
- Zhiping Mi
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, PA, USA; Department of Neurology, University of Pittsburgh School of Medicine, PA, USA
| | - Hao Liu
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, PA, USA; Department of Neurology, University of Pittsburgh School of Medicine, PA, USA
| | - Marie E Rose
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, PA, USA; Department of Neurology, University of Pittsburgh School of Medicine, PA, USA
| | - Xiecheng Ma
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, PA, USA; Department of Neurosurgery, University of Pittsburgh, PA 15216, USA; Department of Critical Care Medicine, University of Pittsburgh, PA 15216, USA
| | - Daniel P Reay
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, PA, USA; Department of Neurology, University of Pittsburgh School of Medicine, PA, USA
| | - Jie Ma
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, PA, USA; Department of Neurology, University of Pittsburgh School of Medicine, PA, USA
| | - Jeremy Henchir
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, PA, USA; Department of Neurosurgery, University of Pittsburgh, PA 15216, USA; Department of Critical Care Medicine, University of Pittsburgh, PA 15216, USA
| | - C Edward Dixon
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, PA, USA; Department of Neurosurgery, University of Pittsburgh, PA 15216, USA; Department of Critical Care Medicine, University of Pittsburgh, PA 15216, USA
| | - Steven H Graham
- Geriatric Research Educational and Clinical Center, V.A. Pittsburgh Healthcare System, PA, USA; Department of Neurology, University of Pittsburgh School of Medicine, PA, USA.
| |
Collapse
|
19
|
Luo M, Luo Y. Imperatorin Relieved Ulcerative Colitis by Regulating the Nrf-2/ARE/HO-1 Pathway in Rats. Inflammation 2020; 44:558-569. [PMID: 33098052 DOI: 10.1007/s10753-020-01353-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/01/2020] [Accepted: 09/28/2020] [Indexed: 01/23/2023]
Abstract
Ulcerative colitis is a common intestinal inflammatory disease. Imperatorin (IMP) has been reported to alleviate mast cell-mediated allergic responses by suppressing the expression of Nrf-2, ARE, and HO-1. However, whether IMP can relieve ulcerative colitis by regulating Nrf-2/ARE/HO-1 pathway is unclear. Thus, this study aims to investigate the effect of IMP on ulcerative colitis in rats and elucidate the potential mechanism. In our study, rats were treated with 2,4,6-trinitro-benzenesulfonic acid (TNBS) to induce the animal model of ulcerative colitis. Next, these rats were treated with diverse doses of IMP (15 mg/kg, 30 mg/kg, and 60 mg/kg) and sacrificed at different time points (3 days, 7 days, and 14 days). The levels of inflammatory factors (TNF-α and IL-6) in colon tissues were detected with ELISA kits. H&E staining was performed to observe the pathologic changes of the colon tissues. The expression of Nrf-2, ARE, and HO-1 in colon tissues was determined with the immunofluorescence and Western blotting. The results showed that application of IMP alleviated the symptoms of ulcerative colitis and inhibited the secretion of TNF-α and IL-6. Besides, treatment of IMP promoted the expression of Nrf-2, ARE, and HO-1 in the early stage of this disease (the third day), but suppressed the expression of Nrf-2, ARE, and HO-1 in the advanced stage of the ulcerative colitis (the fourteenth day). Collectively, IMP relieved the symptoms of ulcerative colitis by regulating the Nrf-2/ARE/HO-1 pathway, which might provide a new therapeutic drug to support the clinical treatment of ulcerative colitis.
Collapse
Affiliation(s)
- Min Luo
- Department of Gastroenterology, the Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, China. .,Research Center of Digestive Disease, Central South University, Changsha City, Hunan Province, China.
| | - Yin Luo
- Department of Paediatrics, Suining People's Hospital, Suining City, Hunan Province, China
| |
Collapse
|
20
|
Saikumar J, Byrns CN, Hemphill M, Meaney DF, Bonini NM. Dynamic neural and glial responses of a head-specific model for traumatic brain injury in Drosophila. Proc Natl Acad Sci U S A 2020; 117:17269-17277. [PMID: 32611818 PMCID: PMC7382229 DOI: 10.1073/pnas.2003909117] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is the strongest environmental risk factor for the accelerated development of neurodegenerative diseases. There are currently no therapeutics to address this due to lack of insight into mechanisms of injury progression, which are challenging to study in mammalian models. Here, we have developed and extensively characterized a head-specific approach to TBI in Drosophila, a powerful genetic system that shares many conserved genes and pathways with humans. The Drosophila TBI (dTBI) device inflicts mild, moderate, or severe brain trauma by precise compression of the head using a piezoelectric actuator. Head-injured animals display features characteristic of mammalian TBI, including severity-dependent ataxia, life span reduction, and brain degeneration. Severe dTBI is associated with cognitive decline and transient glial dysfunction, and stimulates antioxidant, proteasome, and chaperone activity. Moreover, genetic or environmental augmentation of the stress response protects from severe dTBI-induced brain degeneration and life span deficits. Together, these findings present a tunable, head-specific approach for TBI in Drosophila that recapitulates mammalian injury phenotypes and underscores the ability of the stress response to mitigate TBI-induced brain degeneration.
Collapse
Affiliation(s)
- Janani Saikumar
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104
| | - China N Byrns
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Matthew Hemphill
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
| | - David F Meaney
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA 19104
| | - Nancy M Bonini
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104;
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
21
|
Li X, Zhang J, Rong H, Zhang X, Dong M. Ferulic Acid Ameliorates MPP +/MPTP-Induced Oxidative Stress via ERK1/2-Dependent Nrf2 Activation: Translational Implications for Parkinson Disease Treatment. Mol Neurobiol 2020; 57:2981-2995. [PMID: 32445087 DOI: 10.1007/s12035-020-01934-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 05/13/2020] [Indexed: 12/27/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder closely associated with oxidative stress. The biochemical and cellular alterations that occur after cell and mouse treatment with the parkinsonism-inducing neurotoxin MPP+/MPTP are remarkably similar to those observed in idiopathic PD. Previously, we showed that ferulic acid (FA) has antioxidant properties and the ability to activate nuclear factor E2-related factor 2 (Nrf2). The present study tested the hypothesis that FA attenuates MPP+/MPTP-induced oxidative stress by regulating crosstalk between sirtuin 2 (SIRT2) and Nrf2 pathways. To test this hypothesis, we performed in vitro and in vivo studies using MPP+/MPTP-challenged SH-SY5Y cells or mice treated with or not with FA. FA marginally inhibited SIRT2 in parallel with α-synuclein at levels of transcription and translation in SH-SY5Y cells challenged with MPP+. Moreover, FA attenuated MPP+-induced oxidative stress, as indicated by reactive oxygen species, lipid hydroperoxides, GSH/GSSG ratio, and NAD+/NADH ratio. Mechanistically, FA strongly upregulated the glutamate cysteine ligase catalytic subunit and heme oxygenase-1 expression at the levels of transcription and translation. Interestingly, FA-mediated extracellular signal-regulated kinases 1 and 2 (ERK1/2) activation contributed to nuclear accumulation of Nrf2 via de novo synthesis, which was validated by the use of dominant negative ERK2. Surprisingly, activation of the ERK1/2 and inhibition of SIRT2 by FA are mediated by independent mechanisms. Furthermore, FA ameliorated motor deficits and oxidative stress in the ventral midbrain in MPTP-treated (25 mg/kg, i.p., daily for 5 days) wild-type mice and α-synuclein knockout mice, but not in Nrf2 knockout mice. Collectively, FA exerts antioxidant effects through ERK1/2-mediated activation of the Nrf2 pathway, and these results may have important translational value for the treatment of PD.
Collapse
Affiliation(s)
- Xu Li
- The Institute of Medicine, Qiqihar Medical University, 333 BuKui Street, JianHua District, Qiqihar, 161006, China
| | - Jing Zhang
- Department of Hematology, the First Affiliated Hospital, Harbin Medical University, Harbin, 150001, China
| | - Hua Rong
- The Institute of Medicine, Qiqihar Medical University, 333 BuKui Street, JianHua District, Qiqihar, 161006, China
| | - Xiaojie Zhang
- The Institute of Medicine, Qiqihar Medical University, 333 BuKui Street, JianHua District, Qiqihar, 161006, China
| | - Miaoxian Dong
- The Institute of Medicine, Qiqihar Medical University, 333 BuKui Street, JianHua District, Qiqihar, 161006, China.
| |
Collapse
|
22
|
Song JY, Wang XG, Zhang ZY, Che L, Fan B, Li GY. Endoplasmic reticulum stress and the protein degradation system in ophthalmic diseases. PeerJ 2020; 8:e8638. [PMID: 32117642 PMCID: PMC7036270 DOI: 10.7717/peerj.8638] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 01/26/2020] [Indexed: 12/16/2022] Open
Abstract
Objective Endoplasmic reticulum (ER) stress is involved in the pathogenesis of various ophthalmic diseases, and ER stress-mediated degradation systems play an important role in maintaining ER homeostasis during ER stress. The purpose of this review is to explore the potential relationship between them and to find their equilibrium sites. Design This review illustrates the important role of reasonable regulation of the protein degradation system in ER stress-mediated ophthalmic diseases. There were 128 articles chosen for review in this study, and the keywords used for article research are ER stress, autophagy, UPS, ophthalmic disease, and ocular. Data sources The data are from Web of Science, PubMed, with no language restrictions from inception until 2019 Jul. Results The ubiquitin proteasome system (UPS) and autophagy are important degradation systems in ER stress. They can restore ER homeostasis, but if ER stress cannot be relieved in time, cell death may occur. However, they are not independent of each other, and the relationship between them is complementary. Therefore, we propose that ER stability can be achieved by adjusting the balance between them. Conclusion The degradation system of ER stress, UPS and autophagy are interrelated. Because an imbalance between the UPS and autophagy can cause cell death, regulating that balance may suppress ER stress and protect cells against pathological stress damage.
Collapse
Affiliation(s)
- Jing-Yao Song
- Department of Ophthalmology, Second Hospital of Jilin University, ChangChun, China
| | - Xue-Guang Wang
- Department of Traumatic Orthopedics, Third People's Hospital of Jinan, Jinan, China
| | - Zi-Yuan Zhang
- Department of Ophthalmology, Second Hospital of Jilin University, ChangChun, China
| | - Lin Che
- Department of Ophthalmology, Second Hospital of Jilin University, ChangChun, China
| | - Bin Fan
- Department of Ophthalmology, Second Hospital of Jilin University, ChangChun, China
| | - Guang-Yu Li
- Department of Ophthalmology, Second Hospital of Jilin University, ChangChun, China
| |
Collapse
|
23
|
Transcriptional activation of antioxidant gene expression by Nrf2 protects against mitochondrial dysfunction and neuronal death associated with acute and chronic neurodegeneration. Exp Neurol 2020; 328:113247. [PMID: 32061629 DOI: 10.1016/j.expneurol.2020.113247] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/04/2020] [Accepted: 02/11/2020] [Indexed: 02/07/2023]
Abstract
Mitochondria are both a primary source of reactive oxygen species (ROS) and a sensitive target of oxidative stress; damage to mitochondria can result in bioenergetic dysfunction and both necrotic and apoptotic cell death. These relationships between mitochondria and cell death are particularly strong in both acute and chronic neurodegenerative disorders. ROS levels are affected by both the production of superoxide and its toxic metabolites and by antioxidant defense mechanisms. Mitochondrial antioxidant activities include superoxide dismutase 2, glutathione peroxidase and reductase, and intramitochondrial glutathione. When intracellular conditions disrupt the homeostatic balance between ROS production and detoxification, a net increase in ROS and an oxidized shift in cellular redox state ensues. Cells respond to this imbalance by increasing the expression of genes that code for proteins that protect against oxidative stress and inhibit cytotoxic oxidation of proteins, DNA, and lipids. If, however, the genomic response to mitochondrial oxidative stress is insufficient to maintain homeostasis, mitochondrial bioenergetic dysfunction and release of pro-apoptotic mitochondrial proteins into the cytosol initiate a variety of cell death pathways, ultimately resulting in potentially lethal damage to vital organs, including the brain. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a translational activating protein that enters the nucleus in response to oxidative stress, resulting in increased expression of numerous cytoprotective genes, including genes coding for mitochondrial and non-mitochondrial antioxidant proteins. Many experimental and some FDA-approved drugs promote this process. Since mitochondria are targets of ROS, it follows that protection against mitochondrial oxidative stress by the Nrf2 pathway of gene expression contributes to neuroprotection by these drugs. This document reviews the evidence that Nrf2 activation increases mitochondrial antioxidants, thereby protecting mitochondria from dysfunction and protecting neural cells from damage and death. New experimental results are provided demonstrating that post-ischemic administration of the Nrf2 activator sulforaphane protects against hippocampal neuronal death and neurologic injury in a clinically-relevant animal model of cardiac arrest and resuscitation.
Collapse
|
24
|
Parastan RH, Christopher M, Torrys YS, Mahadewa TGB. Combined Therapy Potential of Apocynin and Tert-butylhydroquinone as a Therapeutic Agent to Prevent Secondary Progression to Traumatic Brain Injury. Asian J Neurosurg 2020; 15:10-15. [PMID: 32181166 PMCID: PMC7057894 DOI: 10.4103/ajns.ajns_231_19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 01/23/2020] [Indexed: 11/06/2022] Open
Abstract
Traumatic brain injury is caused by physical collision (primary injury). It changes the brain's biochemistry and disturbs the normal brain function such as memory loss and consciousness disturbance (secondary injury). The severity can be measured with the Glasgow Coma Scale. The secondary injury will cause oxidative stress that leads to the nervous cells death, so treatment is needed before it gets worse. Primary injury results in excess of reactive oxidative stress (ROS) which is known from NADPH oxidase 2 (Nox2). Excessive ROS is deadly to the nerve cells. Excessive ROS will activate nuclear factor erythroid 2-like 2 (Nrf2). Nrf2 will bind to antioxidant response elements, to protect multi organs against ROS, including this brain injury. However, this does not last long, so it requires handling excess ROS. Apocynin can inhibit the activation of Nox2, and reduce the neuron injuries in the hippocampus. It also protects the tissues from oxidative stress. While Nrf2 can be activated by tert-butylhydroquinone, to protect cells. The combination may reduce the secondary brain injury, improve the neurologic recovery, cognitive function, and reduce the secondary cortical lesion.
Collapse
Affiliation(s)
| | - Michael Christopher
- Department of Medicine, Faculty of Medicine, Udayana University, Bali, Indonesia
| | | | | |
Collapse
|
25
|
Bhowmick S, D'Mello V, Caruso D, Abdul-Muneer PM. Traumatic brain injury-induced downregulation of Nrf2 activates inflammatory response and apoptotic cell death. J Mol Med (Berl) 2019; 97:1627-1641. [PMID: 31758217 DOI: 10.1007/s00109-019-01851-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/11/2019] [Accepted: 11/05/2019] [Indexed: 12/17/2022]
Abstract
Recent studies from our group and others have demonstrated that oxidative stress, Ca2+ signaling, and neuroinflammation are major mechanisms contributing to post-traumatic neurodegeneration. The present study investigated the mechanisms of regulation of nuclear factor E2-related factor 2 (Nrf2) and its role in regulating antioxidant genes and oxidative stress-induced neuroinflammation and neurodegeneration following TBI. Nrf2 transcriptional system is the major regulator of endogenous defense mechanisms operating within the cells. Wild-type (Nrf2+/+) and Nrf2-deficient mice (Nrf2-/-) were subjected to 15 psi fluid percussion injury and demonstrated the regulatory role of Nrf2 in the expression antioxidant genes and oxidative stress, neuroinflammation, and cell death. Immunohistochemistry, q-RT-PCR, and western blotting techniques detected downregulation of Nrf2 and antioxidant proteins such as HO-1, GPx1, GSTm1, and NQO1 in mouse brain samples. Further, our study demonstrated that the downregulation of Nrf2 and antioxidant genes in TBI correlated with the induction of free radical-generating enzyme NADPH oxidase 1 and inducible nitric oxide synthase and their corresponding oxidative/nitrosative stress markers 4-hydroxynonenal and 3-nitrotyrosine. The decrease in Nrf2 with subsequent increase in oxidative stress markers led to the activation of MMP3/9, TGF-β1, and NF-kB that further led to neuroinflammation and apoptosis. The absence of Nrf2 function in mice resulted in exacerbated brain injury as shown by the increased oxidative stress markers, pro-inflammatory cytokines, and apoptosis markers at 24 h after TBI. In conclusion, this study could establish the significance of Nrf2 in transforming into a novel preventive approach against the pathophysiology of TBI. KEY MESSAGES: • Traumatic brain injury impairs Nrf2 signaling in mouse. • Nrf2-mediated activation of antioxidant genes are altered after TBI. • Impairment of Nrf2 signaling leads to oxidative stress. • TBI-induced downregulation of Nrf2 activates MMPs, TGF-β1, and NF-kB. • Nrf2 regulates neuroinflammation and apoptotic cell death in TB.
Collapse
Affiliation(s)
- Saurav Bhowmick
- Laboratory of CNS Injury and Molecular Therapy, JFK Neuroscience Institute, Hackensack Meridian Health JFK Medical Center, 65 James St, Edison, NJ, 08820, USA
| | - Veera D'Mello
- Laboratory of CNS Injury and Molecular Therapy, JFK Neuroscience Institute, Hackensack Meridian Health JFK Medical Center, 65 James St, Edison, NJ, 08820, USA
| | - Danielle Caruso
- Laboratory of CNS Injury and Molecular Therapy, JFK Neuroscience Institute, Hackensack Meridian Health JFK Medical Center, 65 James St, Edison, NJ, 08820, USA
| | - P M Abdul-Muneer
- Laboratory of CNS Injury and Molecular Therapy, JFK Neuroscience Institute, Hackensack Meridian Health JFK Medical Center, 65 James St, Edison, NJ, 08820, USA.
| |
Collapse
|
26
|
Wang M, Zhu Z. Nrf2 is involved in osmoregulation, antioxidation and immunopotentiation in Coilia nasus under salinity stress. BIOTECHNOL BIOTEC EQ 2019. [DOI: 10.1080/13102818.2019.1673671] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Affiliation(s)
- Meiyao Wang
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, P.R. China
- Department of Biotechnology, Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, P.R. China
- Aquatic Animal Genome Center, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, P.R. China
| | - Zhixiang Zhu
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, P.R. China
| |
Collapse
|
27
|
Sosunov A, Olabarria M, Goldman JE. Alexander disease: an astrocytopathy that produces a leukodystrophy. Brain Pathol 2019; 28:388-398. [PMID: 29740945 DOI: 10.1111/bpa.12601] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 03/02/2018] [Indexed: 02/02/2023] Open
Abstract
Alexander Disease (AxD) is a degenerative disorder caused by mutations in the GFAP gene, which encodes the major intermediate filament of astrocytes. As other cells in the CNS do not express GFAP, AxD is a primary astrocyte disease. Astrocytes acquire a large number of pathological features, including changes in morphology, the loss or diminution of a number of critical astrocyte functions and the activation of cell stress and inflammatory pathways. AxD is also characterized by white matter degeneration, a pathology that has led it to be included in the "leukodystrophies." Furthermore, variable degrees of neuronal loss take place. Thus, the astrocyte pathology triggers alterations in other cell types. Here, we will review the neuropathology of AxD and discuss how a disease of astrocytes can lead to severe pathologies in non-astrocytic cells. Our knowledge of the pathophysiology of AxD will also lead to a better understanding of how astrocytes interact with other CNS cells and how astrocytes in the gliosis that accompanies many neurological disorders can damage the function and survival of other cells.
Collapse
Affiliation(s)
| | - Markel Olabarria
- Departments of Pathology and Cell Biology, Columbia University, New York, NY
| | - James E Goldman
- Departments of Pathology and Cell Biology, Columbia University, New York, NY
| |
Collapse
|
28
|
Zhou Y, Wang HD, Zhou XM, Fang J, Zhu L, Ding K. N-acetylcysteine amide provides neuroprotection via Nrf2-ARE pathway in a mouse model of traumatic brain injury. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:4117-4127. [PMID: 30584276 PMCID: PMC6284532 DOI: 10.2147/dddt.s179227] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background Increasing evidence demonstrate N-acetylcysteine amide (NACA) provides neuroprotection and attenuated oxidative stress in rats following traumatic brain injury (TBI). The nuclear factor erythroid 2-related factor 2 (Nrf2)–antioxidant response element (ARE) signal pathway is activated after TBI and provides a protective effect against TBI. However, the function and mechanism of NACA in mice after TBI remain unknown. This study was to evaluate the neuroprotection of NACA and the potential action of the Nrf2-ARE pathway in a weight-drop mouse model of TBI. Materials and methods Four groups of animals were randomly divided into sham, TBI, TBI+vehicle, and TBI+NACA (100 mg/kg, administered intraperitoneally). The protein levels of Nrf2, heme oxygenase-1 (HO-1), NAD(P)H: quinine oxidoreductase-1 (NQO1), cleaved caspase-3 and the mRNA levels of HO-1 and NQO1 were detected. The neurobehavior, neuronal degeneration, apoptosis and oxidative stress were also assessed. Results Treatment with NACA significantly improved neurologic status at days 1 and 3 following TBI. Moreover, NACA promoted Nrf2 activation a day after TBI. The protein and mRNA levels of HO-1 and NQO1 were upregulated by NACA. Meanwhile, NACA treatment significantly reduced the level of malondialdehyde (MDA) and enhanced the activity of superoxide dismutase (SOD) and glutathione peroxidase (GPx), which indicated NACA attenuated oxidative stress following TBI. NACA prominently reduced the protein level of cleaved caspase-3 and TUNEL-positive cells, indicating its antiapoptotic effect. Additionally, Fluoro-Jade C staining showed NACA alleviated neuronal degeneration a day after TBI. Conclusions Our study reveals that NACA potentially provides neuroprotection via the activation of the Nrf2-ARE signaling pathway after TBI in mice.
Collapse
Affiliation(s)
- Yuan Zhou
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Jiangsu, China,
| | - Han-Dong Wang
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Jiangsu, China,
| | - Xiao-Ming Zhou
- Department of Neurosurgery, Jinling Hospital, Jiangsu, China
| | - Jiang Fang
- Department of Neurosurgery, Jinling Hospital, Jiangsu, China
| | - Lin Zhu
- Department of Neurosurgery, Jinling Hospital, Jiangsu, China
| | - Ke Ding
- Department of Neurosurgery, Jinling Hospital, Jiangsu, China
| |
Collapse
|
29
|
Dong W, Yang B, Wang L, Li B, Guo X, Zhang M, Jiang Z, Fu J, Pi J, Guan D, Zhao R. Curcumin plays neuroprotective roles against traumatic brain injury partly via Nrf2 signaling. Toxicol Appl Pharmacol 2018; 346:28-36. [PMID: 29571711 DOI: 10.1016/j.taap.2018.03.020] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 03/15/2018] [Accepted: 03/19/2018] [Indexed: 12/28/2022]
Abstract
Traumatic brain injury (TBI), which leads to high mortality and morbidity, is a prominent public health problem worldwide with no effective treatment. Curcumin has been shown to be beneficial for neuroprotection in vivo and in vitro, but the underlying mechanism remains unclear. This study determined whether the neuroprotective role of curcumin in mouse TBI is dependent on the NF-E2-related factor (Nrf2) pathway. The Feeney weight-drop contusion model was used to mimic TBI. Curcumin was administered intraperitoneally 15 min after TBI induction, and brains were collected at 24 h after TBI. The levels of Nrf2 and its downstream genes (Hmox-1, Nqo1, Gclm, and Gclc) were detected by Western blot and qRT-PCR at 24 h after TBI. In addition, edema, oxidative damage, cell apoptosis and inflammatory reactions were evaluated in wild type (WT) and Nrf2-knockout (Nrf2-KO) mice to explore the role of Nrf2 signaling after curcumin treatment. In wild type mice, curcumin treatment resulted in reduced ipsilateral cortex injury, neutrophil infiltration, and microglia activation, improving neuron survival against TBI-induced apoptosis and degeneration. These effects were accompanied by increased expression and nuclear translocation of Nrf2, and enhanced expression of antioxidant enzymes. However, Nrf2 deletion attenuated the neuroprotective effects of curcumin in Nrf2-KO mice after TBI. These findings demonstrated that curcumin effects on TBI are associated with the activation the Nrf2 pathway, providing novel insights into the neuroprotective role of Nrf2 and the potential therapeutic use of curcumin for TBI.
Collapse
Affiliation(s)
- Wenwen Dong
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
| | - Bei Yang
- Department of Histology and Embryology, College of Basic Medical Sciences, China Medical University, Shenyang 110122, China
| | - Linlin Wang
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
| | - Bingxuan Li
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
| | - Xiangshen Guo
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
| | - Miao Zhang
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
| | - Zhenfei Jiang
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
| | - Jingqi Fu
- Program of Environmental Toxicology, School of Public Health, China Medical Univeristy, Shenyang 110122, China
| | - Jingbo Pi
- Program of Environmental Toxicology, School of Public Health, China Medical Univeristy, Shenyang 110122, China
| | - Dawei Guan
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China.
| | - Rui Zhao
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China.
| |
Collapse
|