1
|
Murray KE, Ratliff WA, Delic V, Citron BA. Gulf War toxicant-induced reductions in dendritic arbors and spine densities of dentate granule cells are improved by treatment with a Nrf2 activator. Brain Res 2024; 1823:148682. [PMID: 37989436 DOI: 10.1016/j.brainres.2023.148682] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/08/2023] [Accepted: 11/15/2023] [Indexed: 11/23/2023]
Abstract
Gulf War Illness (GWI) is a chronic multi-symptom disorder affecting approximately 30 % of Veterans deployed to the Persian Gulf from 1990 to 91. GWI encompasses a wide spectrum of symptoms which frequently include neurological problems such as learning and memory impairments, mood disorders, and an increased incidence of neurodegenerative disorders. Combined exposure to both reversible and irreversible acetylcholinesterase (AChE) inhibitors has been identified as a likely risk factor for GWI. It is possible that the exposures affected connectivity in the brain, and it was also unknown whether this could benefit from treatment. We assessed chronic changes in dendritic architecture in granule cells of the dentate gyrus following exposure to pyridostigmine bromide (PB, 0.7 mg/kg), chlorpyrifos (CPF, 12.5 mg/kg), and N,N-diethyl-m-toluamide (DEET, 7.5 mg/kg) in male C57Bl/6J mice. We also evaluated the therapeutic effects of dietary administration for eight weeks of 1 % tert-butylhydroquinone (tBHQ), a Nrf2 activator, on long-term neuronal morphology. We found that Gulf War toxicant exposure resulted in reduced dendritic length and branching as well as overall spine density in dentate granule cells at 14 weeks post-exposure and that these effects were ameliorated by treatment with tBHQ. These findings indicate that Gulf War toxicant exposure results in chronic changes to dentate granule cell morphology and that modulation of neuroprotective transcription factors such as Nrf2 may improve long-term neuronal health in the hippocampus.
Collapse
Affiliation(s)
- Kathleen E Murray
- Laboratory of Molecular Biology, Research & Development, Department of Veterans Affairs, VA New Jersey Health Care System, East Orange, NJ 07018, USA; School of Graduate Studies, Rutgers University, Newark, NJ 07103, USA
| | - Whitney A Ratliff
- Research & Development, Department of Veterans Affairs, Bay Pines VA Healthcare System, Bay Pines, FL 33744, USA
| | - Vedad Delic
- Laboratory of Molecular Biology, Research & Development, Department of Veterans Affairs, VA New Jersey Health Care System, East Orange, NJ 07018, USA; School of Graduate Studies, Rutgers University, Newark, NJ 07103, USA; Department of Pharmacology, Physiology, and Neuroscience, Rutgers-New Jersey Medical School, Newark, NJ 07103, USA
| | - Bruce A Citron
- Laboratory of Molecular Biology, Research & Development, Department of Veterans Affairs, VA New Jersey Health Care System, East Orange, NJ 07018, USA; School of Graduate Studies, Rutgers University, Newark, NJ 07103, USA; Department of Pharmacology, Physiology, and Neuroscience, Rutgers-New Jersey Medical School, Newark, NJ 07103, USA.
| |
Collapse
|
2
|
Zhang E, Wu T, Zhuo Y, Cui J, Sun S, Wu G, Zhang G. Effect of Nrf2 on brain injury induced by hydraulic shock via regulation of mitophagy and apoptosis. Aging (Albany NY) 2023; 15:13422-13433. [PMID: 38019476 DOI: 10.18632/aging.205250] [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: 03/30/2023] [Accepted: 10/11/2023] [Indexed: 11/30/2023]
Abstract
The specific protective mechanism of mitophagy and Nrf2 in brain injury has not been fully clarified. This study aimed to reveal the effect of Nrf2 on hydraulic shock brain injury in mice, and explore its possible mechanism. Twenty-four Nrf2 knockout (Nrf2-/-) and wild-type mice (WT) of C57BL/6J were randomly divided into two groups: control group (C) and brain injury group (TBI). Hematoxylin-eosin staining (HE) assay was used for the histomorphological observation. The apoptotic state of brain tissue was detected by TUNEL. Mechanical damage in vitro models of glial cells were prepared. The wild-type (WT) and Nrf2 knockout (KO) mice were constructed to investigate the changes of mitophagy and apoptosis-related indicators by Western blotting. The experimental results showed that 24 h after TBI, the tissue structure was highly porous, the cells were highly edema, the neuronal space increased significantly, the neuron degeneration, and the cell vacuolation was obvious. Meanwhile, the number of apoptotic cells and the apoptosis rate of glial cells increased significantly. After injury, the relative expression of Parkin, Pink, Beclin and LC-3II proteins were significantly decreased in all mice. The protein expressions of Caspase3 and Caspase12 were significantly increased. However, in the TBI group, KO mice were more impaired than WT mice. In conclusion, Nrf2 plays a protective role by promoting mitophagy to inhibit apoptosis in the process of brain injury caused by hydraulic shock in mice, which provides a new idea for the effective treatment of brain injury.
Collapse
Affiliation(s)
- Erwei Zhang
- The Second Hospital of Hebei Medical University Department of Neurosurgery, Shijiazhuang, China
| | - Tongmao Wu
- The Second Hospital of Hebei Medical University Department of Neurosurgery, Shijiazhuang, China
| | - Yayu Zhuo
- The Second Hospital of Hebei Medical University Department of Neurosurgery, Shijiazhuang, China
| | - Junling Cui
- The Second Hospital of Hebei Medical University Department of Neurosurgery, Shijiazhuang, China
| | - Si Sun
- The Second Hospital of Hebei Medical University Department of Neurosurgery, Shijiazhuang, China
| | - Guobiao Wu
- The Second Hospital of Hebei Medical University Department of Neurosurgery, Shijiazhuang, China
| | - Gengshen Zhang
- The Second Hospital of Hebei Medical University Department of Neurosurgery, Shijiazhuang, China
| |
Collapse
|
3
|
Wei T, Li JD, Wang YJ, Zhao W, Duan F, Wang Y, Xia LL, Jiang ZB, Song X, Zhu YQ, Shao WY, Wang Z, Bi KS, Li H, Zhang XC, Jiao DL. p-Nrf2/HO-1 Pathway Involved in Methamphetamine-induced Executive Dysfunction through Endoplasmic Reticulum Stress and Apoptosis in the Dorsal Striatum. Neurotox Res 2023; 41:446-458. [PMID: 37199892 DOI: 10.1007/s12640-023-00650-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/25/2023] [Accepted: 05/04/2023] [Indexed: 05/19/2023]
Abstract
Methamphetamine (METH) abuse is known to cause executive dysfunction. However, the molecular mechanism underlying METH induced executive dysfunction remains unclear. Go/NoGo experiment was performed in mice to evaluate METH-induced executive dysfunction. Immunoblot analysis of Nuclear factor-E2-related factor 2 (Nrf2), phosphorylated Nrf2 (p-Nrf2), heme-oxygenase-1 (HO-1), Glucose Regulated Protein 78(GRP78), C/EBP homologous protein (CHOP), Bcl-2, Bax and Caspase3 was performed to evaluate the levels of oxidative stress, endoplasmic reticulum (ER) stress and apoptosis in the dorsal striatum (Dstr). Malondialdehyde (MDA) levels and glutathione peroxidase (GSH-Px) activity was conducted to evaluate the level of oxidative stress. TUNEL staining was conducted to detect apoptotic neurons. The animal Go/NoGo testing confirmed that METH abuse impaired the inhibitory control ability of executive function. Meanwhile, METH down-regulated the expression of p-Nrf2, HO-1 and GSH-Px and activated ER stress and apoptosis in the Dstr. Microinjection of Tert-butylhydroxyquinone (TBHQ), an Nrf2 agonist, into the Dstr increased the expression of p-Nrf2, HO-1, and GSH-Px, ameliorated ER stress, apoptosis and executive dysfunction caused by METH. Our results indicated that the p-Nrf2/HO-1 pathway was potentially involved in mediating methamphetamine-induced executive dysfunction by inducing endoplasmic reticulum stress and apoptosis in the dorsal striatum.
Collapse
Affiliation(s)
- Tao Wei
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
- Huainan First People's Hospital, Huainan, 232007, Anhui, China
| | - Jun-Da Li
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Yu-Jing Wang
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Wei Zhao
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Fan Duan
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Yan Wang
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Ling-Ling Xia
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Zhao-Bin Jiang
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Xun Song
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Yu-Qiong Zhu
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Wen-Yi Shao
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Ze Wang
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Kang-Sheng Bi
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Hui Li
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Xiao-Chu Zhang
- CAS Key Laboratory of Brain Function and Disease and School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China.
| | - Dong-Liang Jiao
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China.
| |
Collapse
|
4
|
Lynch DG, Narayan RK, Li C. Multi-Mechanistic Approaches to the Treatment of Traumatic Brain Injury: A Review. J Clin Med 2023; 12:jcm12062179. [PMID: 36983181 PMCID: PMC10052098 DOI: 10.3390/jcm12062179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability worldwide. Despite extensive research efforts, the majority of trialed monotherapies to date have failed to demonstrate significant benefit. It has been suggested that this is due to the complex pathophysiology of TBI, which may possibly be addressed by a combination of therapeutic interventions. In this article, we have reviewed combinations of different pharmacologic treatments, combinations of non-pharmacologic interventions, and combined pharmacologic and non-pharmacologic interventions for TBI. Both preclinical and clinical studies have been included. While promising results have been found in animal models, clinical trials of combination therapies have not yet shown clear benefit. This may possibly be due to their application without consideration of the evolving pathophysiology of TBI. Improvements of this paradigm may come from novel interventions guided by multimodal neuromonitoring and multimodal imaging techniques, as well as the application of multi-targeted non-pharmacologic and endogenous therapies. There also needs to be a greater representation of female subjects in preclinical and clinical studies.
Collapse
Affiliation(s)
- Daniel G. Lynch
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Zucker School of Medicine at Hofstra/Northwell Health, Hempstead, NY 11549, USA
| | - Raj K. Narayan
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Department of Neurosurgery, St. Francis Hospital, Roslyn, NY 11576, USA
| | - Chunyan Li
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Zucker School of Medicine at Hofstra/Northwell Health, Hempstead, NY 11549, USA
- Department of Neurosurgery, Northwell Health, Manhasset, NY 11030, USA
- Correspondence:
| |
Collapse
|
5
|
An Y, Li H, Wang M, Xia Z, Ding L, Xia X. Nuclear factor erythroid 2-related factor 2 agonist protects retinal ganglion cells in glutamate excitotoxicity retinas. Biomed Pharmacother 2022; 153:113378. [DOI: 10.1016/j.biopha.2022.113378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 06/27/2022] [Accepted: 07/06/2022] [Indexed: 11/16/2022] Open
|
6
|
The Role of the NRF2 Pathway in Maintaining and Improving Cognitive Function. Biomedicines 2022; 10:biomedicines10082043. [PMID: 36009590 PMCID: PMC9405981 DOI: 10.3390/biomedicines10082043] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/10/2022] [Accepted: 08/17/2022] [Indexed: 11/24/2022] Open
Abstract
Nuclear factor (erythroid-derived 2)-like 2 (NRF2) is a redox-sensitive transcription factor that binds to the antioxidant response element consensus sequence, decreasing reactive oxygen species and regulating the transcription of a wide array of genes, including antioxidant and detoxifying enzymes, regulating genes involved in mitochondrial function and biogenesis. Moreover, NRF2 has been shown to directly regulate the expression of anti-inflammatory mediators reducing the expression of pro-inflammatory cytokines. In recent years, attention has turned to the role NRF2 plays in the brain in different diseases such Alzheimer’s disease, Parkinson’s disease, Huntington’s disease and others. This review focused on the evidence, derived in vitro, in vivo and from clinical trials, supporting a role for NRF2 activation in maintaining and improving cognitive function and how its activation can be used to elicit neuroprotection and lead to cognitive enhancement. The review also brings a critical discussion concerning the possible prophylactic and/or therapeutic use of NRF2 activators in treating cognitive impairment-related conditions.
Collapse
|
7
|
Ratliff WA, Saykally JN, Keeley KL, Driscoll DC, Murray KE, Okuka M, Mervis RF, Delic V, Citron BA. Sidestream Smoke Affects Dendritic Complexity and Astrocytes After Model Mild Closed Head Traumatic Brain Injury. Cell Mol Neurobiol 2022; 42:1453-1463. [PMID: 33417143 PMCID: PMC8263792 DOI: 10.1007/s10571-020-01036-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 12/28/2020] [Indexed: 11/26/2022]
Abstract
Mild traumatic brain injuries can have long-term consequences that interfere with the life of the patient and impose a burden on our health care system. Oxidative stress has been identified as a contributing factor for the progression of neurodegeneration following TBI. A major source of oxidative stress for many veterans is cigarette smoking and second-hand smoke, which has been shown to have an effect on TBI recovery. To examine the potential influences of second-hand smoke during recovery from TBI, we utilized a mouse model of closed head injury, followed by repeated exposure to cigarette smoke and treatment with a neuroprotective antioxidant. We found that neither the mild injuries nor the smoke exposure produced axonal damage detectable with amino cupric silver staining. However, complexity in the dendritic arbors was significantly reduced after mild TBI plus smoke exposure. In the hippocampus, there were astrocytic responses, including Cyp2e1 upregulation, after the injury and tobacco smoke insult. This study provides useful context for the importance of lifestyle changes, such as reducing or eliminating cigarette smoking, during recovery from TBI.
Collapse
Affiliation(s)
- Whitney A Ratliff
- Laboratory of Molecular Biology, Research and Development 151, Bay Pines VA Healthcare System, Bay Pines, FL, 33744, USA
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
| | - Jessica N Saykally
- Laboratory of Molecular Biology, Research and Development 151, Bay Pines VA Healthcare System, Bay Pines, FL, 33744, USA
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
| | - Kristen L Keeley
- Laboratory of Molecular Biology, Research and Development 151, Bay Pines VA Healthcare System, Bay Pines, FL, 33744, USA
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
| | - David C Driscoll
- Laboratory of Molecular Biology, Research and Development 151, Bay Pines VA Healthcare System, Bay Pines, FL, 33744, USA
| | - Kathleen E Murray
- VA New Jersey Health Care System, Research & Development, East Orange, NJ, 07018, USA
- Department of Pharmacology, Physiology, & Neuroscience, Rutgers - New Jersey Medical School, Newark, NJ, 07103, USA
| | - Maja Okuka
- Department of Obstetrics and Gynecology, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
| | | | - Vedad Delic
- VA New Jersey Health Care System, Research & Development, East Orange, NJ, 07018, USA
- Department of Pharmacology, Physiology, & Neuroscience, Rutgers - New Jersey Medical School, Newark, NJ, 07103, USA
| | - Bruce A Citron
- Laboratory of Molecular Biology, Research and Development 151, Bay Pines VA Healthcare System, Bay Pines, FL, 33744, USA.
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA.
- VA New Jersey Health Care System, Research & Development, East Orange, NJ, 07018, USA.
- Department of Pharmacology, Physiology, & Neuroscience, Rutgers - New Jersey Medical School, Newark, NJ, 07103, USA.
| |
Collapse
|
8
|
Farina M, Vieira LE, Buttari B, Profumo E, Saso L. The Nrf2 Pathway in Ischemic Stroke: A Review. Molecules 2021; 26:5001. [PMID: 34443584 PMCID: PMC8399750 DOI: 10.3390/molecules26165001] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/13/2021] [Accepted: 08/14/2021] [Indexed: 02/07/2023] Open
Abstract
Ischemic stroke, characterized by the sudden loss of blood flow in specific area(s) of the brain, is the leading cause of permanent disability and is among the leading causes of death worldwide. The only approved pharmacological treatment for acute ischemic stroke (intravenous thrombolysis with recombinant tissue plasminogen activator) has significant clinical limitations and does not consider the complex set of events taking place after the onset of ischemic stroke (ischemic cascade), which is characterized by significant pro-oxidative events. The transcription factor Nuclear factor erythroid 2-related factor 2 (Nrf2), which regulates the expression of a great number of antioxidant and/or defense proteins, has been pointed as a potential pharmacological target involved in the mitigation of deleterious oxidative events taking place at the ischemic cascade. This review summarizes studies concerning the protective role of Nrf2 in experimental models of ischemic stroke, emphasizing molecular events resulting from ischemic stroke that are, in parallel, modulated by Nrf2. Considering the acute nature of ischemic stroke, we discuss the challenges in using a putative pharmacological strategy (Nrf2 activator) that relies upon transcription, translation and metabolically active cells in treating ischemic stroke patients.
Collapse
Affiliation(s)
- Marcelo Farina
- Department of Biochemistry, Federal University of Santa Catarina, 88040-900 Florianópolis, Brazil;
| | - Leonardo Eugênio Vieira
- Department of Biochemistry, Federal University of Santa Catarina, 88040-900 Florianópolis, Brazil;
| | - Brigitta Buttari
- Department of Cardiovascular, Endocrine-Metabolic Diseases, and Aging, Italian National Institute of Health, 00161 Rome, Italy; (B.B.); (E.P.)
| | - Elisabetta Profumo
- Department of Cardiovascular, Endocrine-Metabolic Diseases, and Aging, Italian National Institute of Health, 00161 Rome, Italy; (B.B.); (E.P.)
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
9
|
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
|
10
|
Ratliff WA, Delic V, Pick CG, Citron BA. Dendritic arbor complexity and spine density changes after repetitive mild traumatic brain injury and neuroprotective treatments. Brain Res 2020; 1746:147019. [PMID: 32681835 DOI: 10.1016/j.brainres.2020.147019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/25/2020] [Accepted: 07/10/2020] [Indexed: 12/17/2022]
Abstract
Traumatic brain injury has been described as the signature affliction of recent military conflicts and repetitive TBIs, particularly associated with military and athletic activities, typically result in more severe clinical effects. The majority of TBIs are mild, but they can result in long term cognitive deficits for which there is no effective treatment. One of the most significant deficits observed in TBI patients is memory loss, which suggests that TBI can induce pathological changes within the hippocampus. tert-butylhydroquinone (tBHQ) and pioglitazone activate the Nrf2 and PPAR-γ transcription factors, respectively, and both have been shown to be neuroprotective in model systems. We examined the morphological changes within the hippocampus following repetitive mild TBI and simultaneous treatment with both factors. We utilized a closed head injury mouse model with five injuries over 5 weeks. Our results showed marked morphological changes among the dendrites and dendritic spines of the neurons of the dentate gyrus of the hippocampus. We observed decreases in overall dendritic length, as well as in the quantity and density of dendritic spines. Our treatment partially ameliorated these effects, suggesting that the Nrf2 and PPAR-γ transcription factors may be important targets for future drug development in the treatment of TBI in humans.
Collapse
Affiliation(s)
- Whitney A Ratliff
- Laboratory of Molecular Biology, Bay Pines VA Healthcare System, Research and Development 151, Bldg. 22 Rm. 123, 10000 Bay Pines Blvd, Bay Pines, FL 33744, United States
| | - Vedad Delic
- Laboratory of Molecular Biology, VA New Jersey Health Care System, Research & Development (15), Bldg. 16, Rm 16-176 385 Tremont Ave, East Orange, NJ 07018, United States
| | - Chaim G Pick
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel; Dr. Miriam and Sheldon G. Adelson Chair and Center for the Biology of Addictive Diseases, Tel Aviv University, Tel Aviv, Israel
| | - Bruce A Citron
- Laboratory of Molecular Biology, VA New Jersey Health Care System, Research & Development (15), Bldg. 16, Rm 16-176 385 Tremont Ave, East Orange, NJ 07018, United States; Laboratory of Molecular Biology, Bay Pines VA Healthcare System, Research and Development 151, Bldg. 22 Rm. 123, 10000 Bay Pines Blvd, Bay Pines, FL 33744, United States; Department of Pharmacology, Physiology & Neuroscience, Rutgers-New Jersey Medical School, Newark, NJ 07103, United States.
| |
Collapse
|
11
|
Nna VU, Ujah GA, Suleiman JB, Mohamed M, Nwokocha C, Akpan TJ, Ekuma HC, Fubara VV, Kekung-Asu CB, Osim EE. Tert-butylhydroquinone preserve testicular steroidogenesis and spermatogenesis in cisplatin-intoxicated rats by targeting oxidative stress, inflammation and apoptosis. Toxicology 2020; 441:152528. [PMID: 32565124 DOI: 10.1016/j.tox.2020.152528] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/08/2020] [Accepted: 06/16/2020] [Indexed: 12/14/2022]
Abstract
Cisplatin (Cis) is an effective chemotherapeutic intervention against many cancer types. However, the oxidative stress-related toxicities associated with cancer cell resistance-induced dose scaling has limited its long-term use. In the present study, we explored the benefits of the antioxidant, tert-butylhydroquinone (tBHQ; 50 mg/kg b.w./day, for 14 days) against Cis single dose injection (7 mg/kg b.w., i.p on Day 8), on testicular toxicity of male Wistar rats. Cis triggered testicular and epididymal oxidative stress, testicular inflammation (upregulated NF-κB, TNF-α and IL-1β mRNA levels, and downregulated IL-10 mRNA level), increased testicular apoptosis (increased Bax/Bcl2 and caspase-3 mRNA levels) and decreased testicular germ cells proliferation. Further, Cis decreased testicular steroidogenesis (decreased expression of StAR, CYP11A1, 3β-HSD and 17β-HSD mRNA and proteins) and decreased follicle stimulating hormone, luteinizing hormone and testosterone levels. Cis also decreased sperm count, motility, viability, normal morphology and Johnsen score. However, intervention with tBHQ significantly decreased oxidative stress by upregulating Nrf2 gene, suppressed inflammation, apoptosis and increased testicular germ cells proliferation. tBHQ also increased steroidogenesis and improved sperm parameters. Taken together, tBHQ improves steroidogenesis and spermatogenesis in Cis-intoxicated rats by improving antioxidant status, dampening inflammation and apoptosis, thus improving the proliferative capacity of spermatogenic cells.
Collapse
Affiliation(s)
- Victor Udo Nna
- Department of Physiology, College of Medical Sciences, University of Calabar, P.M.B. 1115 Calabar, Cross River State, Nigeria.
| | - Godwin Adakole Ujah
- Department of Physiology, College of Medical Sciences, University of Calabar, P.M.B. 1115 Calabar, Cross River State, Nigeria
| | - Joseph Bagi Suleiman
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, 16150, Kelantan, Malaysia; Department of Science Laboratory, Technology, Akanu Ibiam Federal, Polytechnic, Unwana, Nigeria
| | - Mahaneem Mohamed
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, 16150, Kelantan, Malaysia; Unit of Integrative Medicine, Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, 16150, Kelantan, Malaysia
| | - Chukwuemeka Nwokocha
- Department of Basic Medical Sciences (Physiology Section). The University of the West Indies, Mona, Kingston 7, Jamaica
| | - Timothy Joe Akpan
- Department of Physiology, College of Medical Sciences, University of Calabar, P.M.B. 1115 Calabar, Cross River State, Nigeria
| | - Hope Chinaza Ekuma
- Department of Physiology, College of Medical Sciences, University of Calabar, P.M.B. 1115 Calabar, Cross River State, Nigeria
| | - Victoria Victor Fubara
- Department of Physiology, College of Medical Sciences, University of Calabar, P.M.B. 1115 Calabar, Cross River State, Nigeria
| | - Catherine Barong Kekung-Asu
- Department of Physiology, College of Medical Sciences, University of Calabar, P.M.B. 1115 Calabar, Cross River State, Nigeria
| | - Eme Efiom Osim
- Department of Physiology, College of Medical Sciences, University of Calabar, P.M.B. 1115 Calabar, Cross River State, Nigeria
| |
Collapse
|
12
|
Ratliff WA, Qubty D, Delic V, Pick CG, Citron BA. Repetitive Mild Traumatic Brain Injury and Transcription Factor Modulation. J Neurotrauma 2020; 37:1910-1917. [PMID: 32292111 DOI: 10.1089/neu.2020.7005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The worldwide incidence of traumatic brain injury (TBI) is ∼0.5% per year and the frequency is significantly higher among military personnel and athletes. Repetitive TBIs are associated with military and athletic activities, and typically involve more severe consequences. The majority of TBIs are mild; however, these still can result in long-term cognitive deficits, and there is currently no effective treatment. tert-Butylhydroquinone (tBHQ) and pioglitazone can activate the nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and peroxisome proliferator-activated receptor-gamma (PPAR-γ) transcription factors, respectively, and each has been shown to be neuroprotective in various model systems. We examined behavioral and gene expression changes after repetitive mild TBI followed by simultaneous treatment with both factors. We used a repetitive closed head injury of mice involving five injuries with a 1-week interval between each TBI. We found that memory performance was significantly reduced by the injuries, unless the TBIs were followed by the tBHQ and pioglitazone administrations. Certain genes; for example, growth hormone and osteopontin, were downregulated by the injury, and this was reversed by the treatment, whereas other genes; for example, a tumor necrosis factor receptor, were upregulated by the injury and restored if the post-injury treatment was administered. Analysis of gene expression levels affected by the injury and/or the treatment point to potential mechanisms that could be exploited therapeutically.
Collapse
Affiliation(s)
- Whitney A Ratliff
- Laboratory of Molecular Biology, Bay Pines VA Healthcare System, Research and Development, Bay Pines, Florida, USA
| | - Doaa Qubty
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Vedad Delic
- Laboratory of Molecular Biology, VA New Jersey Health Care System, Research and Development, East Orange, New Jersey, USA
| | - Chaim G Pick
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.,Dr. Miriam and Sheldon G. Adelson Chair and Center for the Biology of Addictive Diseases, Tel Aviv University, Tel Aviv, Israel
| | - Bruce A Citron
- Laboratory of Molecular Biology, Bay Pines VA Healthcare System, Research and Development, Bay Pines, Florida, USA.,Laboratory of Molecular Biology, VA New Jersey Health Care System, Research and Development, East Orange, New Jersey, USA.,Department of Pharmacology, Physiology, and Neuroscience, Rutgers-New Jersey Medical School, Newark, New Jersey, USA
| |
Collapse
|
13
|
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
|
14
|
Lorente L, Martín MM, Pérez-Cejas A, González-Rivero AF, Argueso M, Ramos L, Solé-Violán J, Cáceres JJ, Jiménez A, García-Marín V. Serum caspase-3 levels during the first week of traumatic brain injury. Med Intensiva 2019; 45:131-137. [PMID: 31677852 DOI: 10.1016/j.medin.2019.09.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/10/2019] [Accepted: 09/01/2019] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Confluence between the intrinsic and extrinsic apoptosis pathways is reached at the point of caspase-3 activation, which induces death cell. Higher serum caspase-3 levels have been recorded on day 1 of traumatic brain injury (TBI) in 30-day non-survivors compared to survivors. The objectives of this study therefore were to determine whether serum caspase-3 levels are persistently higher in non-survivors than in survivors, and whether these levels may be used to predict 30-day mortality. DESIGN A prospective observational study was carried out. SETTING Six Spanish Intensive Care Units. PATIENTS Patients with severe isolated TBI (defined as Glasgow Coma Scale <9 points and non-cranial Injury Severity Score <10 points). INTERVENTIONS Serum caspase-3 concentrations were measured on days 1, 4 and 8 of TBI. MAIN VARIABLES OF INTEREST Thirty-day mortality was considered as the study endpoint. RESULTS In comparison with non-survivors (n=34), 30-day survivors (n=90) showed lower serum caspase-3 levels on days 1 (p=0.001), 4 (p<0.001) and 8 (p<0.001) of TBI. Analysis of the ROC curves showed serum caspase-3 concentrations on days 1, 4 and 8 of TBI to have an AUC (95% CI) in predicting 30-day mortality of 0.70 (0.61-0.78; p=0.001), 0.83 (0.74-0.89; p<0.001) and 0.87 (0.79-0.93; p<0.001), respectively. CONCLUSIONS The novel findings of our study were that serum caspase-3 levels during the first week of TBI were lower in survivors and could predict 30-day mortality.
Collapse
Affiliation(s)
- L Lorente
- Intensive Care Unit, Hospital Universitario de Canarias, La Laguna, Santa Cruz de Tenerife, Spain.
| | - M M Martín
- Intensive Care Unit, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - A Pérez-Cejas
- Laboratory Department, Hospital Universitario de Canarias, La Laguna, Santa Cruz de Tenerife, Spain
| | - A F González-Rivero
- Laboratory Department, Hospital Universitario de Canarias, La Laguna, Santa Cruz de Tenerife, Spain
| | - M Argueso
- Intensive Care Unit, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - L Ramos
- Intensive Care Unit, Hospital General de La Palma, Breña Alta, La Palma, Spain
| | - J Solé-Violán
- Intensive Care Unit, Hospital Universitario Dr. Negrín, CIBERES, Las Palmas de Gran Canaria, Spain
| | - J J Cáceres
- Intensive Care Unit, Hospital Insular, Las Palmas de Gran Canaria, Spain
| | - A Jiménez
- Research Unit, Hospital Universitario de Canarias, La Laguna, Santa Cruz de Tenerife, Spain
| | - V García-Marín
- Department of Neurosurgery, Hospital Universitario de Canarias, La Laguna, Santa Cruz de Tenerife, Spain
| |
Collapse
|
15
|
High Serum Caspase-Cleaved Cytokeratin-18 Levels and Mortality of Traumatic Brain Injury Patients. Brain Sci 2019; 9:brainsci9100269. [PMID: 31658711 PMCID: PMC6826452 DOI: 10.3390/brainsci9100269] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/09/2019] [Accepted: 10/09/2019] [Indexed: 01/28/2023] Open
Abstract
Objective: Apoptosis increases in traumatic brain injury (TBI). Caspase-cleaved cytokeratin (CCCK)-18 in blood during apoptosis could appear. At the time of admission due to TBI, higher blood CCCK-18 levels were found in non-surviving than in surviving patients. Therefore, the objective of our study was to analyze whether serum CCCK-18 levels determined during the first week after TBI could predict early mortality (at 30 days). Methods: Severe TBI patients were included (considering severe when Glasgow Coma Scale < 9) in this observational and multicentre study. Serum CCCK-18 levels were determined at day 1 of TBI, and at days 4 and 8 after TBI. Results: Serum CCCK-18 levels at day 1 of TBI, and in the days 4 and 8 after TBI were higher (p < 0.001) in non-surviving than in surviving patients (34 and 90 patients, respectively) and could predict early mortality (p < 0.001 in the area under the curve). Conclusions: The new findings from our study were that serum CCCK-18 levels at any moment of the first week of TBI were higher in non-surviving patients and were able to predict early mortality.
Collapse
|
16
|
Analgesic and Antidepressant Effects of Oltipraz on Neuropathic Pain in Mice by Modulating Microglial Activation. J Clin Med 2019; 8:jcm8060890. [PMID: 31234342 PMCID: PMC6616658 DOI: 10.3390/jcm8060890] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 06/12/2019] [Accepted: 06/15/2019] [Indexed: 12/12/2022] Open
Abstract
Nerve injury provokes microglial activation, contributing to the sensory and emotional disorders associated with neuropathic pain that do not completely resolve with treatment. In C57BL/6J mice with neuropathic pain induced by chronic constriction of the sciatic nerve (CCI), we evaluated the effects of oltipraz, an antioxidant and anticancer compound, on (1) allodynia and hyperalgesia, (2) microglial activation and pain signaling pathways, (3) oxidative stress, and (4) depressive-like behaviors. Twenty-eight days after surgery, we assessed the effects of oltipraz on the expression of CD11b/c (a microglial marker), phosphoinositide 3-kinase (PI3K)/ phosphorylated protein kinase B (p-Akt), nuclear factor-κB (NF-κB) transcription factor, and mitogen activated protein kinases (MAPK) in the spinal cord, hippocampus, and prefrontal cortex. Our results show that oltipraz alleviates neuropathic pain by inhibiting microglial activation and PI3K/p-Akt, phosphorylated inhibitor of κBα (p-IκBα), and MAPK overexpression, and by normalizing and/or enhancing the expression of antioxidant proteins, nuclear factor erythroid derived-2-related factor 2 (Nrf2), heme oxygenase 1 (HO-1), and NAD(P)H:quinone oxidoreductase-1 (NQO1) in the spinal cord. The inhibition of microglial activation and induction of the Nrf2/HO-1/NQO1 signaling pathway in the hippocampus and/or prefrontal cortex may explain the antidepressant effects of oltipraz during neuropathic pain. These data demonstrate the analgesic and antidepressant effects of oltipraz and reveal its protective and antioxidant properties during chronic pain.
Collapse
|
17
|
Song H, Xu Y, Yang X, Rong X, Wang Y, Wei N. Tertiary butylhydroquinone alleviates gestational diabetes mellitus in C57BL/KsJ-Lep db/+ mice by suppression of oxidative stress. J Cell Biochem 2019; 120:15310-15319. [PMID: 31050362 DOI: 10.1002/jcb.28798] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/30/2019] [Accepted: 02/04/2019] [Indexed: 12/14/2022]
Abstract
Gestational diabetes mellitus (GDM) is a common disorder characterized by abnormal glucose metabolism during pregnancy, affecting 2% to 5% of pregnant women. Currently, clinical treatment for GDM is very limited. The present study was designed to investigate the effect and underlying molecular mechanism of tertiary butylhydroquinone (TBHQ) in a pregnant C57BL/KsJ-Lep db/+ (referred to as db+) GDM mouse model. The results showed that nonpregnant db/+ mice did not show a diabetic phenotype, and TBHQ had no effect on glucose and insulin tolerance in these mice. Moreover, in db/+ pregnant mice exhibiting typical diabetes symptoms, such as hyperglycemia and hypoinsulinemia, TBHQ could remarkably decrease the blood glucose level, increase insulin level, and improve glucose and insulin intolerance. The results also revealed that TBHQ could inhibit oxidative stress in pregnant db/+ mice. Furthermore, TBHQ greatly improved offspring survival rate, glucose metabolism, and insulin tolerance. In addition, TBHQ inhibited oxidative stress by reducing malondialdehyde (MDA) and reactive oxygen species (ROS) levels and increased superoxide dismutase (SOD), and glutathione peroxidase (GPx) activities. Moreover, we found that TBHQ activated the nuclear factor erythroid 2-related factor 2 (Nrf2), thereby increasing the levels of Nrf2, and ultimately upregulating the expression of heme oxygenase 1 (NO-1) and superoxide dismutase 2 (SOD2). In conclusion, our findings demonstrated that TBHQ alleviated GDM via Nrf2 activation.
Collapse
Affiliation(s)
- Hongbi Song
- Department of Obstetrics, Guizhou Provincial People's Hospital, Guiyang, People's Republic of China
| | - Yin Xu
- Department of Obstetrics, Guizhou Provincial People's Hospital, Guiyang, People's Republic of China
| | - Xiaowu Yang
- Department of Obstetrics, Guizhou Provincial People's Hospital, Guiyang, People's Republic of China
| | - Xiaoting Rong
- Department of Obstetrics, Guizhou Provincial People's Hospital, Guiyang, People's Republic of China
| | - Ying Wang
- Department of Obstetrics, Guizhou Provincial People's Hospital, Guiyang, People's Republic of China
| | - Na Wei
- Department of Obstetrics, Guizhou Provincial People's Hospital, Guiyang, People's Republic of China
| |
Collapse
|
18
|
Neuroprotective Role of the Nrf2 Pathway in Subarachnoid Haemorrhage and Its Therapeutic Potential. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6218239. [PMID: 31191800 PMCID: PMC6525854 DOI: 10.1155/2019/6218239] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/17/2019] [Accepted: 03/20/2019] [Indexed: 12/11/2022]
Abstract
The mechanisms underlying poor outcome following subarachnoid haemorrhage (SAH) are complex and multifactorial. They include early brain injury, spreading depolarisation, inflammation, oxidative stress, macroscopic cerebral vasospasm, and microcirculatory disturbances. Nrf2 is a global promoter of the antioxidant and anti-inflammatory response and has potential protective effects against all of these mechanisms. It has been shown to be upregulated after SAH, and Nrf2 knockout animals have poorer functional and behavioural outcomes after SAH. There are many agents known to activate the Nrf2 pathway. Of these, the actions of sulforaphane, curcumin, astaxanthin, lycopene, tert-butylhydroquinone, dimethyl fumarate, melatonin, and erythropoietin have been studied in SAH models. This review details the different mechanisms of injury after SAH including the contribution of haemoglobin (Hb) and its breakdown products. It then summarises the evidence that the Nrf2 pathway is active and protective after SAH and finally examines the evidence supporting Nrf2 upregulation as a therapy after SAH.
Collapse
|
19
|
Maggio N, Rubovitch V, Hoffer BJ, Citron BA, Greig NH, Pick CG. Neuronal Hyperexcitability Following mTBI. NEUROSENSORY DISORDERS IN MILD TRAUMATIC BRAIN INJURY 2019:67-81. [DOI: 10.1016/b978-0-12-812344-7.00006-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
20
|
Lazaro I, Lopez-Sanz L, Bernal S, Oguiza A, Recio C, Melgar A, Jimenez-Castilla L, Egido J, Madrigal-Matute J, Gomez-Guerrero C. Nrf2 Activation Provides Atheroprotection in Diabetic Mice Through Concerted Upregulation of Antioxidant, Anti-inflammatory, and Autophagy Mechanisms. Front Pharmacol 2018; 9:819. [PMID: 30108504 PMCID: PMC6080546 DOI: 10.3389/fphar.2018.00819] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/09/2018] [Indexed: 12/19/2022] Open
Abstract
Interactive relationships between metabolism, inflammation, oxidative stress, and autophagy in the vascular system play a key role in the pathogenesis of diabetic cardiovascular disease. Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is a stress-sensitive guarantor of cellular homeostasis, which cytoprotective contributions extend beyond the antioxidant defense. We investigated the beneficial effects and underlying mechanisms of the Nrf2 inducer tert-butyl hydroquinone (tBHQ) on diabetes-driven atherosclerosis. In the experimental model of streptozotocin-induced diabetes in apolipoprotein E-deficient mice, treatment with tBHQ increased Nrf2 activity in macrophages and vascular smooth muscle cells within atherosclerotic lesions. Moreover, tBHQ significantly decreased the size, extension and lipid content of atheroma plaques, and attenuated inflammation by reducing lesional macrophages (total number and M1/M2 phenotype balance), foam cell size and chemokine expression. Atheroprotection was accompanied by both systemic and local antioxidant effects, characterized by lower levels of superoxide anion and oxidative DNA marker 8-hydroxy-2'-deoxyguanosine, reduced expression of NADPH oxidase subunits, and increased antioxidant capacity. Interestingly, tBHQ treatment upregulated the gene and protein expression of autophagy-related molecules and also enhanced autophagic flux in diabetic mouse aorta. In vitro, Nrf2 activation by tBHQ suppressed cytokine-induced expression of pro-inflammatory and oxidative stress genes, altered macrophage phenotypes, and promoted autophagic activity. Our results reinforce pharmacological Nrf2 activation as a promising atheroprotective approach in diabetes, according to the plethora of cytoprotective mechanisms involved in the resolution of inflammation and oxidative stress, and restoring autophagy.
Collapse
Affiliation(s)
- Iolanda Lazaro
- Renal, Vascular and Diabetes Research Lab, IIS-Fundacion Jimenez Diaz, Autonoma University of Madrid, Madrid, Spain
| | - Laura Lopez-Sanz
- Renal, Vascular and Diabetes Research Lab, IIS-Fundacion Jimenez Diaz, Autonoma University of Madrid, Madrid, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, Madrid, Spain
| | - Susana Bernal
- Renal, Vascular and Diabetes Research Lab, IIS-Fundacion Jimenez Diaz, Autonoma University of Madrid, Madrid, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, Madrid, Spain
| | - Ainhoa Oguiza
- Renal, Vascular and Diabetes Research Lab, IIS-Fundacion Jimenez Diaz, Autonoma University of Madrid, Madrid, Spain
| | - Carlota Recio
- Renal, Vascular and Diabetes Research Lab, IIS-Fundacion Jimenez Diaz, Autonoma University of Madrid, Madrid, Spain
| | - Ana Melgar
- Renal, Vascular and Diabetes Research Lab, IIS-Fundacion Jimenez Diaz, Autonoma University of Madrid, Madrid, Spain
| | - Luna Jimenez-Castilla
- Renal, Vascular and Diabetes Research Lab, IIS-Fundacion Jimenez Diaz, Autonoma University of Madrid, Madrid, Spain
| | - Jesus Egido
- Renal, Vascular and Diabetes Research Lab, IIS-Fundacion Jimenez Diaz, Autonoma University of Madrid, Madrid, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, Madrid, Spain
| | - Julio Madrigal-Matute
- Department of Developmental and Molecular Biology, Institute for Aging Studies, Albert Einstein College of Medicine, New York City, NY, United States
| | - Carmen Gomez-Guerrero
- Renal, Vascular and Diabetes Research Lab, IIS-Fundacion Jimenez Diaz, Autonoma University of Madrid, Madrid, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, Madrid, Spain
| |
Collapse
|
21
|
Zhang J, Tucker LD, DongYan, Lu Y, Yang L, Wu C, Li Y, Zhang Q. Tert-butylhydroquinone post-treatment attenuates neonatal hypoxic-ischemic brain damage in rats. Neurochem Int 2018; 116:1-12. [PMID: 29530758 PMCID: PMC5895521 DOI: 10.1016/j.neuint.2018.03.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 01/16/2018] [Accepted: 03/08/2018] [Indexed: 11/23/2022]
Abstract
Hypoxic-ischemic (HI) encephalopathy is a leading cause of dire mortality and morbidity in neonates. Unfortunately, no effective therapies have been developed as of yet. Oxidative stress plays a critical role in pathogenesis and progression of neonatal HI. Previously, as a Nrf2 activator, tert-butylhydroquinone (TBHQ) has been demonstrated to exert neuroprotection on brain trauma and ischemic stroke models, as well as oxidative stress-induced cytotoxicity in neurons. It is, however, still unknown whether TBHQ administration can protect against oxidative stress in neonatal HI brain injury. This study was undertaken to determine the neuroprotective effects and mechanisms of TBHQ post-treatment on neonatal HI brain damage. Using a neonatal HI rat model, we demonstrated that TBHQ markedly abated oxidative stress compared to the HI group, as evidenced by decreased oxidative stress indexes, enhanced Nrf2 nuclear accumulation and DNA binding activity, and up-regulated expression of Nrf2 downstream antioxidative genes. Administration of TBHQ likewise significantly suppressed reactive gliosis and release of inflammatory cytokines, and inhibited apoptosis and neuronal degeneration in the neonatal rat cerebral cortex. In addition, infarct size and neuronal damage were attenuated distinctly. These beneficial effects were accompanied by improved neurological reflex and motor coordination as well as amelioration of spatial learning and memory deficits. Overall, our results provide the first documentation of the beneficial effects of TBHQ in neonatal HI model, in part conferred by activation of Nrf2 mediated antioxidative signaling pathways.
Collapse
Affiliation(s)
- Juan Zhang
- Department of Pharmacology, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712046, PR China; Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912, USA
| | - Lorelei Donovan Tucker
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912, USA
| | - DongYan
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912, USA
| | - Yujiao Lu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912, USA
| | - Luodan Yang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912, USA
| | - Chongyun Wu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912, USA
| | - Yong Li
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912, USA
| | - Quanguang Zhang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912, USA.
| |
Collapse
|
22
|
Saykally JN, Hatic H, Keeley KL, Jain SC, Ravindranath V, Citron BA. Withania somnifera Extract Protects Model Neurons from In Vitro Traumatic Injury. Cell Transplant 2018; 26:1193-1201. [PMID: 28933215 PMCID: PMC5657733 DOI: 10.1177/0963689717714320] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Withania somnifera has been used in traditional medicine for a variety of neural disorders. Recently, chronic neurodegenerative conditions have been shown to benefit from treatment with this extract. To evaluate the action of this extract on traumatically injured neurons, the efficacy of W. somnifera root extract as a neuroprotective agent was examined in cultured model neurons exposed to an in vitro injury system designed to mimic mild traumatic brain injury (TBI). Neuronal health was evaluated by staining with annexin V (an early, apoptotic feature) and monitoring released lactate dehydrogenase activity (a terminal cell loss parameter). Potential mechanisms underlying the observed neuroprotection were examined. Additionally, morphological changes were monitored following injury and treatment. Although no differences were found in the expression of the antioxidant transcription factor nuclear factor erythroid 2-like 2 (Nrf2) or other Nrf2-related downstream components, significant changes were seen in apoptotic signaling. Treatment with the extract resulted in an increased length of neurites projecting from the neuronal cell body after injury. W. somnifera extract treatment also resulted in reduced cell death in the model neuron TBI system. The cell death factor Bax was involved (its expression was reduced 2-fold by the treatment) and injury-induced reduction in neurite lengths and numbers was reversed by the treatment. This all indicates that W. somnifera root extract was neuroprotective and could have therapeutic potential to target factors involved in secondary injury and long-term sequelae of mild TBI.
Collapse
Affiliation(s)
- Jessica N Saykally
- 1 Laboratory of Molecular Biology, Research and Development, Bay Pines VA Healthcare System, Bay Pines, FL, USA.,2 Department of Molecular Medicine, USF Morsani College of Medicine, Tampa, FL, USA
| | - Haris Hatic
- 1 Laboratory of Molecular Biology, Research and Development, Bay Pines VA Healthcare System, Bay Pines, FL, USA
| | - Kristen L Keeley
- 1 Laboratory of Molecular Biology, Research and Development, Bay Pines VA Healthcare System, Bay Pines, FL, USA
| | - Subhash C Jain
- 3 Department of Chemistry, University of Delhi, Delhi, India
| | | | - Bruce A Citron
- 1 Laboratory of Molecular Biology, Research and Development, Bay Pines VA Healthcare System, Bay Pines, FL, USA.,2 Department of Molecular Medicine, USF Morsani College of Medicine, Tampa, FL, USA
| |
Collapse
|
23
|
Zagorski JW, Turley AE, Freeborn RA, VanDenBerg KR, Dover HE, Kardell BR, Liby KT, Rockwell CE. Differential effects of the Nrf2 activators tBHQ and CDDO-Im on the early events of T cell activation. Biochem Pharmacol 2018; 147:67-76. [PMID: 29155145 PMCID: PMC5905342 DOI: 10.1016/j.bcp.2017.11.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 11/13/2017] [Indexed: 12/30/2022]
Abstract
We previously demonstrated that activation of the transcription factor, nuclear factor erythroid 2-related factor 2 (Nrf2) promotes CD4+ Th2 differentiation. In the current study, we assessed the role of Nrf2 in early events following T cell activation. The Nrf2 activators, tBHQ (tert-butylhydroquinone) and CDDO-Im (the imidazolide derivative of the triterpenoid CDDO), were used in conjunction with splenocytes derived from wild-type and Nrf2-null mice to distinguish between Nrf2-specific and off-target effects. CDDO-Im inhibited early IFNγ production in a largely Nrf2-dependent manner. In contrast, tBHQ and CDDO-Im had little effect on expression of CD25 or CD69. Furthermore, tBHQ inhibited GM-CSF and IL-2 production in both wild-type and Nrf2-null T cells, suggesting this effect is Nrf2-independent. Conversely, CDDO-Im caused a concentration-dependent increase in IL-2 secretion in wild-type, but not Nrf2-null, splenocytes, suggesting that Nrf2 promotes IL-2 production. Interestingly, both compounds inhibit NFκB DNA binding, where the suppression by tBHQ is Nrf2-independent and CDDO-Im is Nrf2-dependent. Surprisingly, as compared to wild-type splenocytes, Nrf2-null splenocytes showed lower nuclear accumulation of c-Jun, a member of the AP-1 family of transcription factors, which have been shown to drive multiple immune genes, including IL-2. Both Nrf2 activators caused a Nrf2-dependent trend toward increased nuclear accumulation of c-Jun. These data suggest that modulation of cytokine secretion by tBHQ likely involves multiple pathways, including AP-1, NFκB, and Nrf2. Overall, the data suggest that Nrf2 activation inhibits secretion of the Th1 cytokine IFNγ, and increases early production of IL-2, which has been shown to promote Th2 differentiation, and may support the later occurrence of Th2 polarization.
Collapse
Affiliation(s)
- Joseph W Zagorski
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, United States; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, United States; Cell and Molecular Biology Program, Michigan State University, East Lansing, MI 48824, United States
| | - Alexandra E Turley
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, United States; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, United States
| | - Robert A Freeborn
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, United States; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, United States
| | - Kelly R VanDenBerg
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, United States; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, United States
| | - Heather E Dover
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, United States; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, United States
| | - Brian R Kardell
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, United States; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, United States
| | - Karen T Liby
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, United States
| | - Cheryl E Rockwell
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, United States; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, United States; Cell and Molecular Biology Program, Michigan State University, East Lansing, MI 48824, United States.
| |
Collapse
|
24
|
Gleixner AM, Hutchison DF, Sannino S, Bhatia TN, Leak LC, Flaherty PT, Wipf P, Brodsky JL, Leak RK. N-Acetyl-l-Cysteine Protects Astrocytes against Proteotoxicity without Recourse to Glutathione. Mol Pharmacol 2017; 92:564-575. [PMID: 28830914 PMCID: PMC5635514 DOI: 10.1124/mol.117.109926] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 08/17/2017] [Indexed: 02/06/2023] Open
Abstract
N-acetyl-l-cysteine (NAC) exhibits protective properties in brain injury models and has undergone a number of clinical trials. Most studies of NAC have focused on neurons. However, neuroprotection may be complemented by the protection of astrocytes because healthier astrocytes can better support the viability of neurons. Here, we show that NAC can protect astrocytes against protein misfolding stress (proteotoxicity), the hallmark of neurodegenerative disorders. Although NAC is thought to be a glutathione precursor, NAC protected primary astrocytes from the toxicity of the proteasome inhibitor MG132 without eliciting any increase in glutathione. Furthermore, glutathione depletion failed to attenuate the protective effects of NAC. MG132 elicited a robust increase in the folding chaperone heat shock protein 70 (Hsp70), and NAC mitigated this effect. Nevertheless, three independent inhibitors of Hsp70 function ablated the protective effects of NAC, suggesting that NAC may help preserve Hsp70 chaperone activity and improve protein quality control without need for Hsp70 induction. Consistent with this view, NAC abolished an increase in ubiquitinated proteins in MG132-treated astrocytes. However, NAC did not affect the loss of proteasome activity in response to MG132, demonstrating that it boosted protein homeostasis and cell viability without directly interfering with the efficacy of this proteasome inhibitor. The thiol-containing molecules l-cysteine and d-cysteine both mimicked the protective effects of NAC, whereas the thiol-lacking molecule N-acetyl-S-methyl-l-cysteine failed to exert protection or blunt the rise in ubiquitinated proteins. Collectively, these findings suggest that the thiol group in NAC is required for its effects on glial viability and protein quality control.
Collapse
Affiliation(s)
- Amanda M Gleixner
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Daniel F Hutchison
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Sara Sannino
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Tarun N Bhatia
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Lillian C Leak
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Patrick T Flaherty
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Peter Wipf
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Jeffrey L Brodsky
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| |
Collapse
|
25
|
Lorente L. Biomarkers Associated with the Outcome of Traumatic Brain Injury Patients. Brain Sci 2017; 7:brainsci7110142. [PMID: 29076989 PMCID: PMC5704149 DOI: 10.3390/brainsci7110142] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 09/24/2017] [Accepted: 10/20/2017] [Indexed: 12/16/2022] Open
Abstract
This review focuses on biomarkers associated with the outcome of traumatic brain injury (TBI) patients, such as caspase-3; total antioxidant capacity; melatonin; S100B protein; glial fibrillary acidic protein (GFAP); glutamate; lactate; brain-derived neurotrophic factor (BDNF); substance P; neuron-specific enolase (NSE); ubiquitin carboxy-terminal hydrolase L-1 (UCH-L1); tau; decanoic acid; and octanoic acid.
Collapse
Affiliation(s)
- Leonardo Lorente
- Intensive Care Unit, Hospital Universitario de Canarias, La Laguna, Santa Cruz de Tenerife 38320, Spain.
| |
Collapse
|
26
|
Bai J, Yu XJ, Liu KL, Wang FF, Li HB, Shi XL, Zhang Y, Huo CJ, Li X, Gao HL, Qi J, Liu JJ, Zhu GQ, Chen WS, Cui W, Kang YM. Tert-butylhydroquinone attenuates oxidative stress and inflammation in hypothalamic paraventricular nucleus in high salt-induced hypertension. Toxicol Lett 2017; 281:1-9. [PMID: 28844481 DOI: 10.1016/j.toxlet.2017.08.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 08/18/2017] [Accepted: 08/20/2017] [Indexed: 11/25/2022]
Abstract
Excessive oxidative stress and inflammation in hypothalamic paraventricular nucleus (PVN) are implicated in the pathogenesis of hypertension. It is reported that tert-butylhydroquinone (tBHQ), a nuclear factor erythroid 2-related factor 2(Nrf2)-inducer, has a variety of pharmacological activities such as anti-oxidation and anti-inflammatory effect. The objective of this study was to investigate the effects of tBHQ in high salt induced hypertension and to identify whether the beneficial effects were induced by inhibiting PVN oxidative stress and inflammation. Male Sprague-Dawley rats were fed with high salt diet (HS, 8% NaCl) or normal salt diet (NS, 0.3% NaCl). These rats were administration of tBHQ (150mg/kg/d) by oral gavage for 16 weeks. Our results showed that high salt intake resulted in higher mean arterial pressure, cardiac hypertrophy as well as increased plasma level of norepinephrine and interleukin (IL)-1β, IL-6 compared with NS rats. It increased PVN level of reactive oxygen species, gp91phox, IL-1β, IL-6, p-IKKβ and nuclear factor-kappa B (NF-κB) activity, decreased PVN level of Nrf2 and Cu/Zn-SOD. Chronic administration of tBHQ significantly attenuated these changes in HS rats. These data suggest that the protective effects of tBHQ in salt induced hypertension are partly due to inhibiting oxidative stress and inflammation in PVN.
Collapse
Affiliation(s)
- Juan Bai
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an 710061, China
| | - Xiao-Jing Yu
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an 710061, China.
| | - Kai-Li Liu
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an 710061, China
| | - Fang-Fang Wang
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an 710061, China
| | - Hong-Bao Li
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an 710061, China
| | - Xiao-Lian Shi
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an 710061, China; Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Yan Zhang
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an 710061, China
| | - Chan-Juan Huo
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an 710061, China
| | - Xiang Li
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an 710061, China
| | - Hong-Li Gao
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an 710061, China
| | - Jie Qi
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an 710061, China
| | - Jin-Jun Liu
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an 710061, China
| | - Guo-Qing Zhu
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, 210029, China
| | - Wen-Sheng Chen
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Wei Cui
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Yu-Ming Kang
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an 710061, China.
| |
Collapse
|
27
|
Saykally JN, Ratliff WA, Keeley KL, Pick CG, Mervis RF, Citron BA. Repetitive Mild Closed Head Injury Alters Protein Expression and Dendritic Complexity in a Mouse Model. J Neurotrauma 2017; 35:139-148. [PMID: 28701108 DOI: 10.1089/neu.2017.5070] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Worldwide head injuries are a growing problem. In the United States alone, 1.7 million people suffer a head injury each year. While most of these injuries are mild, head injury sufferers still sustain symptoms that can have major medical and economical impacts. Moreover, repetitive mild head injuries, like those observed in active military personnel and athletes, have demonstrated a more severe and long-term set of consequences. In an effort to better understand the delayed pathological changes following multiple mild head injuries, we used a mouse model of mild closed head injury (with no motor deficits observed by rotarod testing) and measured dendritic complexity at 30 days after injury and potentially related factors up to 60 days post-injury. We found an increase in TDP-43 protein at 60 days post-injury in the hippocampus and a decrease in autophagy factors three days post-injury. Alterations in dendritic complexity were neuronal subtype and location specific. Measurements of neurotropic factors suggest that an increase in complexity in the cortex may be a consequence of neuronal loss of the less connected neurons.
Collapse
Affiliation(s)
- Jessica N Saykally
- 1 Laboratory of Molecular Biology, Research and Development 151, Bay Pines VA Healthcare System , Bay Pines, Florida.,2 Department of Molecular Medicine, University of South Florida College of Medicine , Tampa, Florida
| | - Whitney A Ratliff
- 1 Laboratory of Molecular Biology, Research and Development 151, Bay Pines VA Healthcare System , Bay Pines, Florida.,2 Department of Molecular Medicine, University of South Florida College of Medicine , Tampa, Florida
| | - Kristen L Keeley
- 1 Laboratory of Molecular Biology, Research and Development 151, Bay Pines VA Healthcare System , Bay Pines, Florida.,2 Department of Molecular Medicine, University of South Florida College of Medicine , Tampa, Florida
| | - Chaim G Pick
- 3 Department of Anatomy and Anthropology, Sackler School of Medicine, Tel Aviv University , Tel Aviv, Israel
| | - Ronald F Mervis
- 4 NeuroStructural Research Laboratories, Inc. , Tampa, Florida.,5 Center for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine , Tampa, Florida
| | - Bruce A Citron
- 1 Laboratory of Molecular Biology, Research and Development 151, Bay Pines VA Healthcare System , Bay Pines, Florida.,2 Department of Molecular Medicine, University of South Florida College of Medicine , Tampa, Florida
| |
Collapse
|
28
|
Xu W, Li F, Xu Z, Sun B, Cao J, Liu Y. Tert-butylhydroquinone protects PC12 cells against ferrous sulfate-induced oxidative and inflammatory injury via the Nrf2/ARE pathway. Chem Biol Interact 2017; 273:28-36. [DOI: 10.1016/j.cbi.2017.05.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/22/2017] [Accepted: 05/30/2017] [Indexed: 12/17/2022]
|
29
|
Zagorski JW, Maser TP, Liby KT, Rockwell CE. Nrf2-Dependent and -Independent Effects of tert-Butylhydroquinone, CDDO-Im, and H 2O 2 in Human Jurkat T Cells as Determined by CRISPR/Cas9 Gene Editing. J Pharmacol Exp Ther 2017; 361:259-267. [PMID: 28280124 PMCID: PMC5399638 DOI: 10.1124/jpet.116.238899] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 03/03/2017] [Indexed: 12/13/2022] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a stress-activated transcription factor activated by stimuli such as electrophilic compounds and other reactive xenobiotics. Previously, we have shown that the commonly used food additive and Nrf2 activator tert-butylhydroquinone (tBHQ) suppresses interleukin-2 (IL-2) production, CD25 expression, and NFκB activity in human Jurkat T cells. The purpose of the current studies was to determine whether these effects were dependent upon Nrf2 by developing a human Nrf2-null T cell model using clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 technology. The current studies show that suppression of CD25 expression by tBHQ is partially dependent on Nrf2, whereas inhibition of IL-2 secretion is largely Nrf2-independent. Interestingly, tBHQ inhibited NFκB activation in an Nrf2-independent manner. This was an unexpected finding since Nrf2 inhibits NFκB activation in other models. These results led us to investigate another more potent Nrf2 activator, the synthetic triterpenoid 1[2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oyl]imidazole (CDDO-Im). Treatment of wild-type and Nrf2-null Jurkat T cells with CDDO-Im resulted in an Nrf2-dependent suppression of IL-2. Furthermore, susceptibility to reactive oxygen species was significantly enhanced in the Nrf2-null clones as determined by decreased mitochondrial membrane potential and cell viability. Importantly, this study is the first to describe the generation of a human Nrf2-null model, which is likely to have multiple applications in immunology and cancer biology. Collectively, this study demonstrates a role for Nrf2 in the effects of CDDO-Im on CD25 and IL-2 expression, whereas the effect of tBHQ on these parameters is complex and likely involves modulation of multiple stress-activated transcription factors, including NFκB and Nrf2.
Collapse
Affiliation(s)
- Joseph W Zagorski
- Department of Pharmacology and Toxicology (J.W.Z., T.P.M., K.T.L., C.E.R.), Institute for Integrated Toxicology (J.W.Z., C.E.R.), and Cell and Molecular Biology Program (J.W.Z. C.E.R.), Michigan State University, East Lansing, Michigan
| | - Tyler P Maser
- Department of Pharmacology and Toxicology (J.W.Z., T.P.M., K.T.L., C.E.R.), Institute for Integrated Toxicology (J.W.Z., C.E.R.), and Cell and Molecular Biology Program (J.W.Z. C.E.R.), Michigan State University, East Lansing, Michigan
| | - Karen T Liby
- Department of Pharmacology and Toxicology (J.W.Z., T.P.M., K.T.L., C.E.R.), Institute for Integrated Toxicology (J.W.Z., C.E.R.), and Cell and Molecular Biology Program (J.W.Z. C.E.R.), Michigan State University, East Lansing, Michigan
| | - Cheryl E Rockwell
- Department of Pharmacology and Toxicology (J.W.Z., T.P.M., K.T.L., C.E.R.), Institute for Integrated Toxicology (J.W.Z., C.E.R.), and Cell and Molecular Biology Program (J.W.Z. C.E.R.), Michigan State University, East Lansing, Michigan
| |
Collapse
|
30
|
Nrf2-ARE signaling provides neuroprotection in traumatic brain injury via modulation of the ubiquitin proteasome system. Neurochem Int 2017; 111:32-44. [PMID: 28465088 DOI: 10.1016/j.neuint.2017.04.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 04/26/2017] [Indexed: 12/29/2022]
Abstract
The nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant response element (ARE) pathway exhibits protective effects in a variety of neurological diseases. However, the role of this pathway in traumatic brain injury (TBI) is not fully understood. This study investigates whether the Nrf2-ARE pathway provides neuroprotection following TBI via regulation of the ubiquitin proteasome system (UPS), and examines the involvement of this pathway in redox homeostasis. We found that activation the Nrf2-ARE pathway can mitigate secondary brain injury induced by TBI. Furthermore, we found that inhibiting the Nrf2-ARE pathway weakened the UPS following TBI. Treatment of TBI with the proteasome inhibitor, MG132, increased neuronal apoptosis, and evidence of brain water content was found. These data suggest that the Nrf2-ARE pathway provides neuroprotection following TBI via modulation of the UPS. In addition, the results indicated that the content of glutathione (GSH) was significantly increased after activation of Nrf2, and the level of ROS decreased; however, this effect contradictory in the Nrf2 knockout mice. Further studies found that treatment with the ROS agonist, ferric ammonium citrate (FAC), resulted in additional damage exerted by the ubiquitin proteasome pathways, and a significant increase in the amount of ubiquitinated proteins. In contrast, the activity of the ubiquitin proteasome pathways was vastly enhanced, and the level of ubiquitination proteins was significantly decreased following treatment with the inhibitor, N-acetylcysteine (NAC). The above mentioned results were also verified in in vitro experiments. In conclusion, the activation the Nrf2-ARE pathway improves neurological impairment caused by TBI via modulation of the UPS, and the redox homeostasis is one of the vital regulatory mechanisms.
Collapse
|
31
|
Zeng XP, Li XJ, Zhang QY, Liu QW, Li L, Xiong Y, He CX, Wang YF, Ye QF. Tert-Butylhydroquinone Protects Liver Against Ischemia/Reperfusion Injury in Rats Through Nrf2-Activating Anti-Oxidative Activity. Transplant Proc 2017; 49:366-372. [DOI: 10.1016/j.transproceed.2016.12.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 12/13/2016] [Indexed: 01/07/2023]
|
32
|
Zhang L, Yang J, Wu S, Jin C, Lu X, Hu X, Sun Y, Gao X, Cai Y. Activation of Nrf2/ARE signaling pathway attenuates lanthanum chloride induced injuries in primary rat astrocytes. Metallomics 2017; 9:1120-1131. [DOI: 10.1039/c7mt00182g] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Lanthanum (La) exposure can lead to learning and memory disorder in animals; however, the underlying mechanism of La induced neurotoxicity is still unknown.
Collapse
Affiliation(s)
- Lijin Zhang
- Department of Toxicology
- School of Public Health
- China Medical University
- Shenyang 110122
- People's Republic of China
| | - Jinghua Yang
- Department of Toxicology
- School of Public Health
- China Medical University
- Shenyang 110122
- People's Republic of China
| | - Shengwen Wu
- Department of Toxicology
- School of Public Health
- China Medical University
- Shenyang 110122
- People's Republic of China
| | - Cuihong Jin
- Department of Toxicology
- School of Public Health
- China Medical University
- Shenyang 110122
- People's Republic of China
| | - Xiaobo Lu
- Department of Toxicology
- School of Public Health
- China Medical University
- Shenyang 110122
- People's Republic of China
| | - Xiaoyu Hu
- Department of Toxicology
- School of Public Health
- China Medical University
- Shenyang 110122
- People's Republic of China
| | - Yaling Sun
- Department of Toxicology
- School of Public Health
- China Medical University
- Shenyang 110122
- People's Republic of China
| | - Xiang Gao
- Department of Toxicology
- School of Public Health
- China Medical University
- Shenyang 110122
- People's Republic of China
| | - Yuan Cai
- Department of Toxicology
- School of Public Health
- China Medical University
- Shenyang 110122
- People's Republic of China
| |
Collapse
|
33
|
Tweedie D, Fukui K, Li Y, Yu QS, Barak S, Tamargo IA, Rubovitch V, Holloway HW, Lehrmann E, Wood WH, Zhang Y, Becker KG, Perez E, Van Praag H, Luo Y, Hoffer BJ, Becker RE, Pick CG, Greig NH. Cognitive Impairments Induced by Concussive Mild Traumatic Brain Injury in Mouse Are Ameliorated by Treatment with Phenserine via Multiple Non-Cholinergic and Cholinergic Mechanisms. PLoS One 2016; 11:e0156493. [PMID: 27254111 PMCID: PMC4890804 DOI: 10.1371/journal.pone.0156493] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 05/16/2016] [Indexed: 11/18/2022] Open
Abstract
Traumatic brain injury (TBI), often caused by a concussive impact to the head, affects an estimated 1.7 million Americans annually. With no approved drugs, its pharmacological treatment represents a significant and currently unmet medical need. In our prior development of the anti-cholinesterase compound phenserine for the treatment of neurodegenerative disorders, we recognized that it also possesses non-cholinergic actions with clinical potential. Here, we demonstrate neuroprotective actions of phenserine in neuronal cultures challenged with oxidative stress and glutamate excitotoxicity, two insults of relevance to TBI. These actions translated into amelioration of spatial and visual memory impairments in a mouse model of closed head mild TBI (mTBI) two days following cessation of clinically translatable dosing with phenserine (2.5 and 5.0 mg/kg BID x 5 days initiated post mTBI) in the absence of anti-cholinesterase activity. mTBI elevated levels of thiobarbituric acid reactive substances (TBARS), a marker of oxidative stress. Phenserine counteracted this by augmenting homeostatic mechanisms to mitigate oxidative stress, including superoxide dismutase [SOD] 1 and 2, and glutathione peroxidase [GPx], the activity and protein levels of which were measured by specific assays. Microarray analysis of hippocampal gene expression established that large numbers of genes were exclusively regulated by each individual treatment with a substantial number of them co-regulated between groups. Molecular pathways associated with lipid peroxidation were found to be regulated by mTBI, and treatment of mTBI animals with phenserine effectively reversed injury-induced regulations in the ‘Blalock Alzheimer’s Disease Up’ pathway. Together these data suggest that multiple phenserine-associated actions underpin this compound’s ability to ameliorate cognitive deficits caused by mTBI, and support the further evaluation of the compound as a therapeutic for TBI.
Collapse
Affiliation(s)
- David Tweedie
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, United States of America
| | - Koji Fukui
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, United States of America
- Division of Bioscience and Engineering, Shibaura Institute of Technology, Saitama 3378570, Japan
| | - Yazhou Li
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, United States of America
| | - Qian-sheng Yu
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, United States of America
| | - Shani Barak
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, 69978 Israel
| | - Ian A. Tamargo
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, United States of America
| | - Vardit Rubovitch
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, 69978 Israel
| | - Harold W. Holloway
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, United States of America
| | - Elin Lehrmann
- Laboratory of Genetics and Genomics, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, United States of America
| | - William H. Wood
- Laboratory of Genetics and Genomics, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, United States of America
| | - Yongqing Zhang
- Laboratory of Genetics and Genomics, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, United States of America
| | - Kevin G. Becker
- Laboratory of Genetics and Genomics, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, United States of America
| | - Evelyn Perez
- Laboratory of Behavioral Neuroscience, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, United States of America
| | - Henriette Van Praag
- Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, United States of America
| | - Yu Luo
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, OH, United States of America
| | - Barry J. Hoffer
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, OH, United States of America
| | - Robert E. Becker
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, United States of America
- Independent Researcher, 7123 Pinebrook Road, Park City, UT 94098, United States of America
| | - Chaim G. Pick
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, 69978 Israel
- Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, 69978 Israel
| | - Nigel H. Greig
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, United States of America
- * E-mail:
| |
Collapse
|
34
|
Duan X, Li J, Li W, Xing X, Zhang Y, Li W, Zhao L, Sun G, Gao XH, Li B. Antioxidant tert-butylhydroquinone ameliorates arsenic-induced intracellular damages and apoptosis through induction of Nrf2-dependent antioxidant responses as well as stabilization of anti-apoptotic factor Bcl-2 in human keratinocytes. Free Radic Biol Med 2016; 94:74-87. [PMID: 26878773 DOI: 10.1016/j.freeradbiomed.2016.02.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 01/28/2016] [Accepted: 02/10/2016] [Indexed: 01/27/2023]
Abstract
UNLABELLED Human skin is a known target site of inorganic arsenic with effects ranging from hyperkeratosis to dermal malignancies. Tert-butylhydroquinone (tBHQ), approved food-grade phenolic antioxidant, is demonstrated to induce remarkable antioxidant activity in a variety of cells and tissues. The present study aimed at the protective effects of tBHQ on arsenic-induced cytotoxicity and apoptosis in human keratinocytes. Our results demonstrated that tBHQ antagonized arsenic-induced decrease of cell viability, generation of reactive oxygen species (ROS) and lipid peroxidation, as well as reduction of antioxidative enzymes superoxide dismutase (SOD) and catalase (CAT) activities. We also found that tBHQ relieved the G2/M phase arrest by arsenic exposure, which was associated with altering the expression of cell cycle regulators cyclin D1 and CDK4. tBHQ treatment further reduced the numbers of arsenic-induced mitochondrial-mediated apoptotic cells, which occurred concomitantly with the effective recovery of mitochondrial membrane potential (ΔΨm) depolarization, the release of cytochrome c releasing from the mitochondrial as well as the survival signal related factor caspase 3 activation. Our experiments then confirmed that tBHQ activated nuclear factor E2-related factor 2 (NRF2) pathway by increasing NRF2 protein in both nucleus and cytoplasm and upregulating NRF2 downstream targets NAD(P)H quinine oxidoreductase 1 (NQO1) and heme oxygenase-1 (HO-1). More interestingly, arsenic-induced decrease of anti-apoptotic factor B-cell lymphoma-2 (Bcl-2) and increase of pro-apoptotic factor Bcl-2-associated X protein (Bax) could all be reversed by tBHQ pretreatment. These results suggested together that tBHQ could ameliorate arsenic-induced cytotoxicity and apoptosis, which might be linked with the induction of Nrf2-dependent antioxidant responses as well as stabilization of anti-apoptotic factor Bcl-2 in human keratinocytes.
Collapse
Affiliation(s)
- Xiaoxu Duan
- Department of Occupational and Environmental Health, Key Laboratory of Arsenic-related Biological Effects and Prevention and Treatment in Liaoning Province, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110013, China
| | - Jinlong Li
- Department of Occupational and Environmental Health, Key Laboratory of Arsenic-related Biological Effects and Prevention and Treatment in Liaoning Province, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110013, China
| | - Wei Li
- Health Care Department, Maternal and Child Health Care Center of Cangzhou, Cangzhou 061000, China
| | - Xiaoyue Xing
- Student Office, China Medical University, Shenyang 110013, China
| | - Yang Zhang
- Department of Occupational and Environmental Health, Key Laboratory of Arsenic-related Biological Effects and Prevention and Treatment in Liaoning Province, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110013, China
| | - Wei Li
- Department of Occupational and Environmental Health, Key Laboratory of Arsenic-related Biological Effects and Prevention and Treatment in Liaoning Province, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110013, China
| | - Lu Zhao
- Department of Occupational and Environmental Health, Key Laboratory of Arsenic-related Biological Effects and Prevention and Treatment in Liaoning Province, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110013, China
| | - Guifan Sun
- Environment and Non-Communicable Diseases Research Center, School of Public Health, China Medical University, Shenyang 110013, China
| | - Xing-Hua Gao
- Department of Dermatology, No. 1 Hospital of China Medical University, Shenyang 110001, China.
| | - Bing Li
- Department of Occupational and Environmental Health, Key Laboratory of Arsenic-related Biological Effects and Prevention and Treatment in Liaoning Province, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110013, China.
| |
Collapse
|
35
|
Novel pharmaceutical treatments for minimal traumatic brain injury and evaluation of animal models and methodologies supporting their development. J Neurosci Methods 2016; 272:69-76. [PMID: 26868733 DOI: 10.1016/j.jneumeth.2016.02.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 02/01/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND The need for effective pharmaceuticals within animal models of traumatic brain injury (TBI) continues to be paramount, as TBI remains the major cause of brain damage for children and young adults. While preventative measures may act to reduce the incidence of initial blunt trauma, well-tolerated drugs are needed to target the neurologically damaging internal cascade of molecular mechanisms that follow. Such processes, known collectively as the secondary injury phase, include inflammation, excitotoxicity, and apoptosis among other changes still subject to research. In this article positive treatment findings to mitigate this secondary injury in rodent TBI models will be overviewed, and include recent studies on Exendin-4, N-Acetyl-l-cycteine, Salubrinal and Thrombin. CONCLUSIONS These studies provide representative examples of methodologies that can be combined with widely available in vivo rodent models to evaluate therapeutic approaches of translational relevance, as well as drug targets and biochemical cascades that may slow or accelerate the degenerative processes induced by TBI. They employ well-characterized tests such as the novel object recognition task for assessing cognitive deficits. The application of such methodologies provides both decision points and a gateway for implementation of further translational studies to establish the feasibility of clinical efficacy of potential therapeutic interventions.
Collapse
|
36
|
Jiang S, Deng C, Lv J, Fan C, Hu W, Di S, Yan X, Ma Z, Liang Z, Yang Y. Nrf2 Weaves an Elaborate Network of Neuroprotection Against Stroke. Mol Neurobiol 2016; 54:1440-1455. [PMID: 26846360 DOI: 10.1007/s12035-016-9707-7] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Accepted: 01/05/2016] [Indexed: 12/24/2022]
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a neuroprotective transcription factor that has recently attracted increased attention. Stroke, a common and serious neurological disease, is currently a leading cause of death in the USA so far. It is therefore of vital importance to explore how Nrf2 behaves in stroke. In this review, we first introduce the structural features of Nrf2 and Kelch-like ECH-associated protein 1 (Keap1) and briefly depict the activation, inactivation, and regulation processes of the Nrf2 pathway. Next, we discuss the physiopathological mechanisms, upstream modulators, and downstream targets of the Nrf2 pathway. Following this background, we expand our discussion to the roles of Nrf2 in ischemic and hemorrhagic stroke and provide several potential future directions. The information presented here may be useful in the design of future experimental research and increase the likelihood of using Nrf2 as a therapeutic target for stroke in the future.
Collapse
Affiliation(s)
- Shuai Jiang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East, Zhengzhou, 450052, China.,Department of Aerospace Medicine, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710032, China
| | - Chao Deng
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Jianjun Lv
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, China
| | - Chongxi Fan
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an, 710038, China
| | - Wei Hu
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, China
| | - Shouyin Di
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, China
| | - Xiaolong Yan
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, China
| | - Zhiqiang Ma
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, China
| | - Zhenxing Liang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East, Zhengzhou, 450052, China.
| | - Yang Yang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East, Zhengzhou, 450052, China. .,Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, China.
| |
Collapse
|
37
|
Gazaryan IG, Thomas B. The status of Nrf2-based therapeutics: current perspectives and future prospects. Neural Regen Res 2016; 11:1708-1711. [PMID: 28123399 PMCID: PMC5204211 DOI: 10.4103/1673-5374.194706] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
This mini-review presents the authors' vision on the current status and future trends in the development of neuroprotective agents working via activation of nuclear factor erythroid 2-related factor 2 (Nrf2), and in particular, via disruption of Nrf2-Keap1 interaction. There are two opposite "chemical" mechanisms underlying such activation: the first one is a non-specific covalent modification of Keap1 thiols, resulting in side effects of varied severity, and the second one is the shift of the Nrf2-Kelch-like ECH associated protein-1 (Keap1) binding equilibrium in the presence of a competitive and chemically benign displacement agent. At this point, no displacement activators exhibit sufficient biological activity in comparison with common Nrf2 activators working via Keap1 thiol modification. Hence, the hope in therapeutics is now linked to the FDA approved dimethylfumarate, whose derivative, monomethylfumarate, as we demonstrated recently, is much less toxic but equally biologically potent and an ideal candidate for clinical trials right now. A newly emerging player is a nuclear inhibitor of Nrf2, BTB domain and CNC homolog 1 (Bach1). The commercially developed Bach1 inhibitors are currently under investigation in our laboratory showing promising results. In our viewpoint, the perfect future drug will present the combination of a displacement activator and Bach1 inhibitor to insure safety and efficiency of Nrf2 activation.
Collapse
Affiliation(s)
- Irina G Gazaryan
- Department of Cell Biology and Anatomy, School of Medicine, New York Medical College, Valhalla, NY, USA; and Department of Chemistry and Physical Sciences, Dyson College, Pace University, Pleasantville, NY, USA
| | - Bobby Thomas
- Departments of Pharmacology, Toxicology and Neurology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| |
Collapse
|
38
|
t-BHQ Provides Protection against Lead Neurotoxicity via Nrf2/HO-1 Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:2075915. [PMID: 26798413 PMCID: PMC4698940 DOI: 10.1155/2016/2075915] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 10/11/2015] [Indexed: 12/30/2022]
Abstract
The neurotoxicity of lead has been well established, and oxidative stress is strongly associated with lead-induced neurotoxicity. Nrf2 is important for protection against oxidative stress in many disease models. We applied t-BHQ, which is an Nrf2 activator, to investigate the possible role of Nrf2 in the protection against lead neurotoxicity. t-BHQ significantly attenuated the oxidative stress in developmental rats by decreasing MDA level, as well as by increasing SOD activity and GSH content, in the hippocampus and frontal cortex. Furthermore, neuronal apoptosis was detected by Nissl staining, and Bax expression was inhibited in the t-BHQ-treated group. Results showed that t-BHQ suppressed ROS production and caspase 3/7 activity but increased intracellular GSH content, in SH-SY5Y cells under lead exposure. Moreover, in vivo and in vitro, t-BHQ enhanced the nuclear translocation of Nrf2 and binding to ARE areas but did not induce Nrf2 transcription. These phenomena were confirmed using RT-PCR, EMSA, Western blot, and immunofluorescence analyses. Subsequent upregulation of the expression of HO-1, NQO1, and GCLC was observed. However, knockdown of Nrf2 or HO-1 adversely affected the protective effects of t-BHQ against lead toxicity in SH-SY5Y cells. Thus, t-BHQ can protect against lead neurotoxicity, depending on the Nrf2/HO-1 pathway.
Collapse
|
39
|
Shabbir A, Bianchetti E, Cargonja R, Petrovic A, Mladinic M, Pilipović K, Nistri A. Role of HSP70 in motoneuron survival after excitotoxic stress in a rat spinal cord injury modelin vitro. Eur J Neurosci 2015; 42:3054-65. [DOI: 10.1111/ejn.13108] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 10/15/2015] [Accepted: 10/15/2015] [Indexed: 01/06/2023]
Affiliation(s)
- Ayisha Shabbir
- Neuroscience Department; Scuola Internazionale Superiore di Studi Avanzati (SISSA); Via Bonomea 265 34136 Trieste Italy
| | - Elena Bianchetti
- Neuroscience Department; Scuola Internazionale Superiore di Studi Avanzati (SISSA); Via Bonomea 265 34136 Trieste Italy
| | - Renato Cargonja
- Neuroscience Department; Scuola Internazionale Superiore di Studi Avanzati (SISSA); Via Bonomea 265 34136 Trieste Italy
- Department of Biotechnology; University of Rijeka; Rijeka Croatia
| | - Antonela Petrovic
- Neuroscience Department; Scuola Internazionale Superiore di Studi Avanzati (SISSA); Via Bonomea 265 34136 Trieste Italy
- Department of Biotechnology; University of Rijeka; Rijeka Croatia
| | - Miranda Mladinic
- Neuroscience Department; Scuola Internazionale Superiore di Studi Avanzati (SISSA); Via Bonomea 265 34136 Trieste Italy
- Department of Biotechnology; University of Rijeka; Rijeka Croatia
| | - Kristina Pilipović
- Neuroscience Department; Scuola Internazionale Superiore di Studi Avanzati (SISSA); Via Bonomea 265 34136 Trieste Italy
| | - Andrea Nistri
- Neuroscience Department; Scuola Internazionale Superiore di Studi Avanzati (SISSA); Via Bonomea 265 34136 Trieste Italy
| |
Collapse
|
40
|
Shu L, Wang C, Wang J, Zhang Y, Zhang X, Yang Y, Zhuo J, Liu J. The neuroprotection of hypoxic preconditioning on rat brain against traumatic brain injury by up-regulated transcription factor Nrf2 and HO-1 expression. Neurosci Lett 2015; 611:74-80. [PMID: 26590328 DOI: 10.1016/j.neulet.2015.11.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 10/23/2015] [Accepted: 11/09/2015] [Indexed: 01/30/2023]
Abstract
Hypoxic preconditioning (HPC) increases the inherent tolerance of brain tissue suffering from severe hypoxia or ischemia insult by stimulating the protective ability of the brain. However, little is known concerning the effect of HPC on traumatic brain injury (TBI). We designed this study to investigate the effect of HPC on TBI and explore its underlying mechanisms. We found that HPC significantly alleviates neurological dysfunction, lessens brain edema, reduces cell apoptosis, increases neuronal survival, up-regulates the expressions of Nrf2 and HO-1, and decreases the inducer of protein carbonyls, 4-hydroxy-2-nonenal, and 8-hydroxy-2-deoxyguanosine in the brain tissue of rats 24h after brain injury. However, no influence was observed in normal rats after only 3d of hypoxic training. Results further indicated that HPC protects the brain against traumatic damage. This protective effect may be achieved by up-regulating Nrf2 and HO-1 expression and alleviating oxidative stress damage.
Collapse
Affiliation(s)
- Longfei Shu
- Department of Neurosurgery, 105 Hospital of PLA, Clinical College of PLA, Anhui Medical University, Hefei, Anhui 230031, China
| | - Chunlin Wang
- Department of Neurosurgery, 105 Hospital of PLA, Clinical College of PLA, Anhui Medical University, Hefei, Anhui 230031, China
| | - Jinbiao Wang
- Department of Neurosurgery, 105 Hospital of PLA, Clinical College of PLA, Anhui Medical University, Hefei, Anhui 230031, China
| | - Yongming Zhang
- Department of Neurosurgery, 105 Hospital of PLA, Clinical College of PLA, Anhui Medical University, Hefei, Anhui 230031, China
| | - Xing Zhang
- Department of Neurosurgery, 105 Hospital of PLA, Clinical College of PLA, Anhui Medical University, Hefei, Anhui 230031, China
| | - Yanyan Yang
- Department of Neurosurgery, 105 Hospital of PLA, Clinical College of PLA, Anhui Medical University, Hefei, Anhui 230031, China
| | - Jianwei Zhuo
- Department of Neurosurgery, 105 Hospital of PLA, Clinical College of PLA, Anhui Medical University, Hefei, Anhui 230031, China
| | - Jiachuan Liu
- Department of Neurosurgery, 105 Hospital of PLA, Clinical College of PLA, Anhui Medical University, Hefei, Anhui 230031, China.
| |
Collapse
|
41
|
Lorente L, Martín MM, Argueso M, Ramos L, Solé-Violán J, Riaño-Ruiz M, Jiménez A, Borreguero-León JM. Serum caspase-3 levels and mortality are associated in patients with severe traumatic brain injury. BMC Neurol 2015; 15:228. [PMID: 26545730 PMCID: PMC4636758 DOI: 10.1186/s12883-015-0485-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 10/29/2015] [Indexed: 01/10/2023] Open
Abstract
Background Different apoptosis pathways activate caspase-3. In a study involving 27 patients with traumatic brain injury (TBI), higher caspase-3 levels were found in contusion brain tissue resected from non-survivors than from survivors. The objective of this study was to determine whether there is an association in TBI patients between serum caspase-3 levels (thus using an easier, quicker, less expensive and less invasive procedure) and mortality, in a larger series of patients. Methods We carried out a prospective, observational and multicenter study in six Spanish Hospital Intensive Care Units including 112 patients with severe TBI. All had Glasgow Coma Scale (GCS) scores lower than 9. Patients with an Injury Severity Score (ISS) in non-cranial aspects higher than 9 were excluded. Blood samples were collected on day 1 of TBI to measure serum caspas-3 levels. The endpoint was 30-day mortality. Results We found that non-surviving patients (n = 31) showed higher (p = 0.003) serum caspase-3 levels compared to survivors (n = 81). Kaplan-Meier survival analysis showed a higher risk of death in TBI patients with serum caspase-3 levels >0.20 ng/mL than in patients with lower concentrations (Hazard Ratio = 3.15; 95 % CI = 1.40 to 7.08; P < 0.001). Multiple logistic regression analysis showed that serum caspase-3 levels > 0.20 ng/mL were associated with mortality at 30 days in TBI patients controlling for Marshall CT classification, age and GCS (Odds ratio = 7.99; 95 % CI = 2.116 to 36.744; P = 0.001). Conclusions The association between serum caspase-3 levels and mortality in TBI patients was the major novel finding of our study.
Collapse
Affiliation(s)
- Leonardo Lorente
- Intensive Care Unit, Hospital Universitario de Canarias, Ofra, s/n. La Laguna, 38320, Santa Cruz de Tenerife, Spain.
| | - María M Martín
- Intensive Care Unit, Hospital Universitario Nuestra Señora de Candelaria, Crta del Rosario s/n, Santa Cruz de Tenerife, 38010, Spain.
| | - Mónica Argueso
- Intensive Care Unit, Hospital Clínico Universitario de Valencia, Avda. Blasco Ibáñez n°17-19, Valencia, 46004, Spain.
| | - Luis Ramos
- Intensive Care Unit, Hospital General de La Palma, Buenavista de Arriba s/n, Breña Alta, La Palma, 38713, Spain.
| | - Jordi Solé-Violán
- Intensive Care Unit, Hospital Universitario Dr. Negrín, CIBERES, Barranco de la Ballena s/n, Las Palmas de Gran Canaria, 35010, Spain.
| | - Marta Riaño-Ruiz
- Servicio de Bioquímica Clínica, Complejo Hospitalario Universitario Insular Materno-Infantil, Plaza Dr. Pasteur s/n, Las Palmas de Gran Canaria, 35016, Spain.
| | - Alejandro Jiménez
- Research Unit, Hospital Universitario de Canarias, Ofra, s/n. La Laguna, 38320, Santa Cruz de Tenerife, Spain.
| | - Juan M Borreguero-León
- Laboratory Deparment, Hospital Universitario de Canarias, Ofra, s/n. La Laguna, 38320, Santa Cruz de Tenerife, Spain.
| |
Collapse
|
42
|
Wang LF, Su SW, Wang L, Zhang GQ, Zhang R, Niu YJ, Guo YS, Li CY, Jiang WB, Liu Y, Guo HC. Tert-butylhydroquinone ameliorates doxorubicin-induced cardiotoxicity by activating Nrf2 and inducing the expression of its target genes. Am J Transl Res 2015; 7:1724-1735. [PMID: 26692920 PMCID: PMC4656753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Accepted: 10/11/2015] [Indexed: 06/05/2023]
Abstract
UNLABELLED Oxidative stress plays an important role in doxorubicin (DOX)-induced cardiotoxicity. Nuclear factor E2-related factor-2 (Nrf2) is a transcription factor that orchestrates the antioxidant and cytoprotective responses to oxidative stress. In the present study, we tested whether tert-butylhydroquinone (tBHQ) could protect against DOX-induced cardiotoxicity in vivo and, if so, whether the protection was associated with the up-regulation of the Nrf2 pathway. The results showed that treatment with tBHQ significantly decreased the DOX-induced cardiac injury in wild-type mice. Moreover, tBHQ ameliorated the DOX-induced oxidative stress and apoptosis. Further studies suggested that tBHQ increased the nuclear accumulation of Nrf2 and the Nrf2-regulated gene expression, including heme oxygenase-1 (HO-1) and NAD(P)H quinone oxido-reductase-1 (NQO-1) expression. Knocking out Nrf2 in mice abolished the protective effect of tBHQ on the DOX-induced cardiotoxicity. These results indicate that tBHQ has a beneficial effect on DOX-induced cardiotoxicity, and this effect was associated with the enhanced expression of Nrf2 and its downstream antioxidant genes, HO-1 and NQO-1.
Collapse
Affiliation(s)
- Lin-Feng Wang
- Department of Spine Surgery, The Third Hospital of Hebei Medical UniversityShijiazhuang, Hebei Province, 050017, China
| | - Su-Wen Su
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Department of Pharmacology, Hebei Medical UniversityShijiazhuang, Hebei Province, 050017, China
| | - Lei Wang
- Department of Toxicology, Hebei Medical UniversityShijiazhuang, Hebei Province, 050017, China
| | - Guo-Qiang Zhang
- Department of Toxicology, Hebei Medical UniversityShijiazhuang, Hebei Province, 050017, China
| | - Rong Zhang
- Department of Toxicology, Hebei Medical UniversityShijiazhuang, Hebei Province, 050017, China
| | - Yu-Jie Niu
- Department of Toxicology, Hebei Medical UniversityShijiazhuang, Hebei Province, 050017, China
| | - Yan-Su Guo
- Department of Neurology, The Second Hospital of Hebei Medical UniversityShijiazhuang, Hebei Province, 050017, China
| | - Chun-Yan Li
- Department of Neurology, The Second Hospital of Hebei Medical UniversityShijiazhuang, Hebei Province, 050017, China
| | - Wen-Bo Jiang
- Department of Toxicology, Hebei Medical UniversityShijiazhuang, Hebei Province, 050017, China
| | - Yi Liu
- Department of Toxicology, Hebei Medical UniversityShijiazhuang, Hebei Province, 050017, China
| | - Hui-Cai Guo
- Department of Toxicology, Hebei Medical UniversityShijiazhuang, Hebei Province, 050017, China
| |
Collapse
|
43
|
Tert-butylhydroquinone Ameliorates Early Brain Injury After Experimental Subarachnoid Hemorrhage in Mice by Enhancing Nrf2-Independent Autophagy. Neurochem Res 2015; 40:1829-38. [DOI: 10.1007/s11064-015-1672-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 07/10/2015] [Accepted: 07/17/2015] [Indexed: 01/06/2023]
|
44
|
The antioxidant compound tert-butylhydroquinone activates Akt in myocardium, suppresses apoptosis and ameliorates pressure overload-induced cardiac dysfunction. Sci Rep 2015; 5:13005. [PMID: 26260024 PMCID: PMC4531315 DOI: 10.1038/srep13005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 07/10/2015] [Indexed: 02/06/2023] Open
Abstract
Tert-butylhydroquinone (TBHQ) is an antioxidant compound which shows multiple cytoprotective actions. We evaluated the effects of TBHQ on pathological cardiac remodeling and dysfunction induced by chronic overload. Pressure overload was created by transverse aortic constriction (TAC) in male C57BL/6 mice. TBHQ was incorporated in the diet and administered for 4 weeks. TBHQ treatment prevented left ventricular dilatation and cardiac dysfunction induced by TAC, and decreased the prevalence of myocardial apoptosis. The beneficial effects of TBHQ were associated with an increase in Akt activation, but not related to activations of Nrf2 or AMP-activated protein kinase. TBHQ-induced Akt activation was accompanied by increased phosphorylation of Bad, glycogen synthase kinase-3β (GSK-3β) and mammalian target of rapamycin (mTOR). Mechanistically, we showed that in cultured H9c2 cells and primary cardiac myocytes, TBHQ stimulated Akt phosphorylation and suppressed oxidant-induced apoptosis; this effect was abolished by wortmannin or an Akt inhibitor. Blockade of the Akt pathway in vivo accelerated cardiac dysfunction, and abrogated the protective effects of TBHQ. TBHQ also reduced the reactive aldehyde production and protein carbonylation in stressed myocardium. We suggest that TBHQ treatment may represent a novel strategy for timely activation of the cytoprotective Akt pathway in stressed myocardium.
Collapse
|
45
|
Lu XY, Wang HD, Xu JG, Ding K, Li T. Deletion of Nrf2 Exacerbates Oxidative Stress After Traumatic Brain Injury in Mice. Cell Mol Neurobiol 2015; 35:713-21. [PMID: 25732597 PMCID: PMC11486234 DOI: 10.1007/s10571-015-0167-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 02/23/2015] [Indexed: 12/21/2022]
Abstract
Traumatic brain injury (TBI) is a worldwide public health and medical problem. Oxidative stress is recognized as an important contributing factor in the pathogenesis of TBI. The present study was designed to explore the anti-oxidative effect of Nuclear factor erythroid 2-related factor 2 (Nrf2) on brain damage induced by traumatic injury in a mouse model. Moderate weight-drop impact head injury was induced in adult male mice. The mice were randomly divided into four groups: Nrf2(+/+) sham-operation, Nrf2(-/-) sham-operation, Nrf2(+/+) TBI, and Nrf2(-/-) TBI group. Neurological scores were evaluated 24 h after TBI, followed by collection of the brain specimens. Brain edema was detected by the wet-dry ratio method. The expression of NOX2 protein in the brain specimen was investigated using Western Blot analysis and immunohistochemical staining. In addition, malondialdehyde (MDA) level and superoxide dismutase (SOD) activity were evaluated in the brain tissues. Twenty-four hours after TBI, our results showed Nrf2(+/+) TBI mice have more severe neurological deficits and brain edema than Nrf2(+/+) sham group. On the other hand, the Nrf2(-/-) TBI mice were found to have significantly increased neurological deficits and brain edema, compared to Nrf2(+/+) TBI mice (P < 0.05). At the same time, we found that the expression of NOX2 protein, MDA level were significantly increased in Nrf2(-/-) mice, while SOD activity was considerably decreased after TBI compared to Nrf2(+/+) mice (P < 0.05). We demonstrated that deletion of Nrf2 exacerbates brain injury after TBI in mice, suggesting that Nrf2 may play an important role in protecting brain injury after TBI, possibly by modulating oxidative stress.
Collapse
Affiliation(s)
- Xin-Yu Lu
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002 Jiangsu China
| | - Han-Dong Wang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002 Jiangsu China
| | - Jian-Guo Xu
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002 Jiangsu China
| | - Ke Ding
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002 Jiangsu China
| | - Tao Li
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing, 210002 Jiangsu China
| |
Collapse
|
46
|
Crum TS, Gleixner AM, Posimo JM, Mason DM, Broeren MT, Heinemann SD, Wipf P, Brodsky JL, Leak RK. Heat shock protein responses to aging and proteotoxicity in the olfactory bulb. J Neurochem 2015; 133:780-794. [PMID: 25640060 DOI: 10.1111/jnc.13041] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 12/22/2014] [Accepted: 01/12/2015] [Indexed: 12/13/2022]
Abstract
The olfactory bulb is one of the most vulnerable brain regions in age-related proteinopathies. Proteinopathic stress is mitigated by the heat shock protein (Hsp) family of chaperones. Here, we describe age-related decreases in Hsc70 in the olfactory bulb of the female rat and higher levels of Hsp70 and Hsp25 in middle and old age than at 2-4 months. To model proteotoxic and oxidative stress in the olfactory bulb, primary olfactory bulb cultures were treated with the proteasome inhibitors lactacystin and MG132 or the pro-oxidant paraquat. Toxin-induced increases were observed in Hsp70, Hsp25, and Hsp32. To determine the functional consequences of the increase in Hsp70, we attenuated Hsp70 activity with two mechanistically distinct inhibitors. The Hsp70 inhibitors greatly potentiated the toxicity of sublethal lactacystin or MG132 but not of paraquat. Although ubiquitinated protein levels were unchanged with aging in vivo or with sublethal MG132 in vitro, there was a large, synergistic increase in ubiquitinated proteins when proteasome and Hsp70 functions were simultaneously inhibited. Our study suggests that olfactory bulb cells rely heavily on Hsp70 chaperones to maintain homeostasis during mild proteotoxic, but not oxidative insults, and that Hsp70 prevents the accrual of ubiquitinated proteins in these cells. The olfactory bulb is affected in the early phases of many age-related neurodegenerative disorders. Here, we described the impact of aging on multiple heat shock proteins (Hsps), such as Hsp70, in the female rat olfactory bulb in vivo. Using multiple proteasome and Hsp70 inhibitors (see schematic), we found that proteotoxicity elicited a compensatory increase in Hsp70 in primary olfactory bulb cells in vitro. Hsp70 then reduced the proteotoxic buildup of ubiquitinated proteins and robustly protected against cell death according to three independent viability assays. Thus, olfactory bulb neurons can mount impressive natural adaptations to proteotoxic injury, perhaps explaining why neurodegenerative disorders are so delayed in onset and so slow to progress.
Collapse
Affiliation(s)
- Tyler S Crum
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh PA
| | - Amanda M Gleixner
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh PA
| | - Jessica M Posimo
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh PA
| | - Daniel M Mason
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh PA
| | - Matthew T Broeren
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh PA
| | - Scott D Heinemann
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh PA
| | - Peter Wipf
- Departments of Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh PA
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh PA
| | - Rehana K Leak
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh PA
| |
Collapse
|
47
|
Posimo JM, Weilnau JN, Gleixner AM, Broeren MT, Weiland NL, Brodsky JL, Wipf P, Leak RK. Heat shock protein defenses in the neocortex and allocortex of the telencephalon. Neurobiol Aging 2015; 36:1924-37. [PMID: 25771395 DOI: 10.1016/j.neurobiolaging.2015.02.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 01/30/2015] [Accepted: 02/11/2015] [Indexed: 11/18/2022]
Abstract
The telencephalic allocortex develops protein inclusions before the neocortex in many age-related proteinopathies. One major defense mechanism against proteinopathic stress is the heat shock protein (Hsp) network. We therefore contrasted Hsp defenses in stressed primary neocortical and allocortical cells. Neocortical neurons were more resistant to the proteasome inhibitor MG132 than neurons from 3 allocortical subregions: entorhinal cortex, piriform cortex, and hippocampus. However, allocortical neurons exhibited higher MG132-induced increases in Hsp70 and heat shock cognate 70 (Hsc70). MG132-treated allocortical neurons also exhibited greater levels of protein ubiquitination. Inhibition of Hsp70/Hsc70 activity synergistically exacerbated MG132 toxicity in allocortical neurons more than neocortical neurons, suggesting that the allocortex is more reliant on these Hsp defenses. In contrast, astrocytes harvested from the neocortex or allocortex did not differ in their response to Hsp70/Hsc70 inhibition. Consistent with the idea that chaperones are maximally engaged in allocortical neurons, an increase in Hsp70/Hsc70 activity was protective only in neocortical neurons. Finally, the levels of select Hsps were altered in the neocortex and allocortex in vivo with aging.
Collapse
Affiliation(s)
- Jessica M Posimo
- Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | - Justin N Weilnau
- Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | - Amanda M Gleixner
- Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | - Matthew T Broeren
- Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | - Nicole L Weiland
- Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA.
| |
Collapse
|
48
|
Li Y, Cao Y, Wang F, Pu S, Zhang Y, Li C. Tert-butylhydroquinone attenuates scrotal heat-induced damage by regulating Nrf2-antioxidant system in the mouse testis. Gen Comp Endocrinol 2014; 208:12-20. [PMID: 25260249 DOI: 10.1016/j.ygcen.2014.09.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 08/01/2014] [Accepted: 09/16/2014] [Indexed: 01/07/2023]
Abstract
Tert-butylhydroquinone (tBHQ), a widely used nuclear factor erythroid 2-related factor 2 (Nrf2) activator, was always employed to investigate the potential protective role of Nrf2 activation. In this study, to elucidate the effect of tBHQ on scrotal heat-induced damage and Nrf2-antioxidant system in mouse testes, eight-week-old mice were administrated with or without dietary tBHQ (1%w/w) for 1week and afterward subjected to a single scrotal heat treatment (42°C for 25min). Trunk blood and testes were collected 3h or 1, 2, or 7days later. Mice displayed less germ cell loss in testes, higher relative testis weight and lower testosterone concentration on day 2 in tBHQ treatment group. Before heat treatment, there were significant increases in malondialdehyde (MDA) concentration in tBHQ treatment group. After heat treatment, mice in tBHQ treatment group showed lower MDA concentration than those in non-tBHQ treatment group. In addition, both tBHQ pretreatment and scrotal heat treatment induced markedly increased Nrf2 protein expression in cytoplasm and nuclei of interstitial cells, accompanying with elevated mRNA expression of Nrf2 and Nrf2-regulated genes in mice testes. Our data indicated that pretreatment to tBHQ induced a mild oxidative stress, and further enhanced the cellular antioxidative ability to protect testicular cells against scrotal heat-induced damage via a mechanism that might involve the Nrf2-antioxidant system in mice testes.
Collapse
Affiliation(s)
- Yansen Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yun Cao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Fei Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Shaoxia Pu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yonghui Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China
| | - ChunMei Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
49
|
Xu J, Wang H, Ding K, Zhang L, Wang C, Li T, Wei W, Lu X. Luteolin provides neuroprotection in models of traumatic brain injury via the Nrf2-ARE pathway. Free Radic Biol Med 2014; 71:186-195. [PMID: 24642087 DOI: 10.1016/j.freeradbiomed.2014.03.009] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 02/16/2014] [Accepted: 03/07/2014] [Indexed: 01/09/2023]
Abstract
UNLABELLED Luteolin has recently been proven to exert neuroprotection in a variety of neurological diseases; however, its roles and the underlying mechanisms in traumatic brain injury are not fully understood. The present study was aimed to investigate the neuroprotective effects of luteolin in models of traumatic brain injury (TBI) and the possible role of the Nrf2-ARE pathway in the putative neuroprotection. A modified Marmarou׳s weight-drop model in mice and the scratch model in mice primary cultured neurons were used to induce TBI. We determined that luteolin significantly ameliorated secondary brain injury induced by TBI, including neurological deficits, brain water content, and neuronal apoptosis. Furthermore, the level of malondialdehyde (MDA) and the activity of glutathione peroxidase (GPx) were restored in the group with luteolin treatment. in vitro studies showed that luteolin administration lowered the intracellular reactive oxygen species (ROS) level and increased the neuron survival. Moreover, luteolin enhanced the translocation of Nrf2 to the nucleus both in vivo and in vitro, which was proved by the results of Western blot, immunohistochemistry, and electrophoretic mobility shift assay (EMSA). Subsequently upregulation of the expression of the downstream factors such as heme oxygenase 1 (HO1) and NAD(P)H quinone oxidoreductase 1 (NQO1) was also examined. However, luteolin treatment failed to provide neuroprotection after TBI in Nrf2(-/-) mice. Taken together, these in vivo and in vitro data demonstrated that luteolin provided neuroprotective effects in the models of TBI, possibly through the activation of the Nrf2-ARE pathway.
Collapse
Affiliation(s)
- Jianguo Xu
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, People׳s Republic of China
| | - Handong Wang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, People׳s Republic of China.
| | - Ke Ding
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, People׳s Republic of China
| | - Li Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, People׳s Republic of China
| | - Chunxi Wang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, People׳s Republic of China
| | - Tao Li
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, People׳s Republic of China
| | - Wuting Wei
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Southern Medical University (Guangzhou), Nanjing, Jiangsu Province, People׳s Republic of China
| | - Xinyu Lu
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, People׳s Republic of China
| |
Collapse
|
50
|
Wang Z, Ji C, Wu L, Qiu J, Li Q, Shao Z, Chen G. Tert-butylhydroquinone alleviates early brain injury and cognitive dysfunction after experimental subarachnoid hemorrhage: role of Keap1/Nrf2/ARE pathway. PLoS One 2014; 9:e97685. [PMID: 24848277 PMCID: PMC4029824 DOI: 10.1371/journal.pone.0097685] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 04/23/2014] [Indexed: 11/18/2022] Open
Abstract
Tert-butylhydroquinone (tBHQ), an Nrf2 activator, has demonstrated neuroprotection against brain trauma and ischemic stroke in vivo. However, little work has been done with respect to its effect on early brain injury (EBI) after subarachnoid hemorrhage (SAH). At the same time, as an oral medication, it may have extensive clinical applications for the treatment of SAH-induced cognitive dysfunction. This study was undertaken to evaluate the influence of tBHQ on EBI, secondary deficits of learning and memory, and the Keap1/Nrf2/ARE pathway in a rat SAH model. SD rats were divided into four groups: (1) Control group (n=40); (2) SAH group (n=40); (3) SAH+vehicle group (n=40); and (4) SAH+tBHQ group (n=40). All SAH animals were subjected to injection of autologous blood into the prechiasmatic cistern once in 20 s. In SAH+tBHQ group, tBHQ was administered via oral gavage at a dose of 12.5 mg/kg at 2 h, 12 h, 24 h, and 36 h after SAH. In the first set of experiments, brain samples were extracted and evaluated 48 h after SAH. In the second set of experiments, changes in cognition and memory were investigated in a Morris water maze. Results shows that administration of tBHQ after SAH significantly ameliorated EBI-related problems, such as brain edema, blood-brain barrier (BBB) impairment, clinical behavior deficits, cortical apoptosis, and neurodegeneration. Learning deficits induced by SAH was markedly alleviated after tBHQ therapy. Treatment with tBHQ markedly up-regulated the expression of Keap1, Nrf2, HO-1, NQO1, and GSTα1 after SAH. In conclusion, the administration of tBHQ abated the development of EBI and cognitive dysfunction in this SAH model. Its action was probably mediated by activation of the Keap1/Nrf2/ARE pathway.
Collapse
Affiliation(s)
- Zhong Wang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Chengyuan Ji
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Lingyun Wu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Jiaoxue Qiu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Qi Li
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhong Shao
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Gang Chen
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|