1
|
Yamamoto M, Shimizu T, Shimizu N, Fujieda M, Saito M. Brain carbon monoxide can suppress the rat micturition reflex through brain γ-aminobutyric acid receptors. Int J Urol 2024; 31:1052-1060. [PMID: 38884570 DOI: 10.1111/iju.15514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/02/2024] [Indexed: 06/18/2024]
Abstract
OBJECTIVES To investigate roles of brain carbon monoxide (CO), an endogenous gasotransmitter, in regulation of the rat micturition reflex. METHODS In urethane-anesthetized (0.8 g/kg, ip) male rats, evaluation of urodynamic parameters was started 1 h before intracerebroventricular administration of CORM-3 (CO donor) or ZnPP (non-selective inhibitor of heme oxygenase, a CO producing enzyme) and continued for 2 h after the administration. We also investigated effects of centrally pretreated SR95531 (GABAA receptor antagonist) or SCH50911 (GABAB receptor antagonist) on the CORM-3-induced response. RESULTS CORM-3 significantly prolonged intercontraction intervals (ICIs) without changing maximal voiding pressure (MVP), while ZnPP significantly shortened ICI and reduced single-voided volume and bladder capacity without affecting MVP, post-voided residual volume, or voiding efficiency. The ZnPP-induced ICI shortening was reversed by CORM-3. The CORM-3-induced ICI prolongation was significantly attenuated by centrally pretreated SR95531 or SCH50911, respectively. CONCLUSIONS Brain CO can suppress the rat micturition reflex through brain γ-aminobutyric acid (GABA) receptors.
Collapse
Affiliation(s)
- Masaki Yamamoto
- Department of Pediatrics, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Takahiro Shimizu
- Department of Pharmacology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Nobutaka Shimizu
- Pelvic Floor Center, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Mikiya Fujieda
- Department of Pediatrics, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Motoaki Saito
- Department of Pharmacology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| |
Collapse
|
2
|
Chen H, Sun H, Hua W, Chang H, Chen W, Ma S. Exogenous hydrogen sulfide ameliorates diabetes-associated cognitive dysfunction by regulating the nrf-2/HO-1 axis and the NLRP3 inflammasome pathway in diabetic rats. Eur J Pharmacol 2024; 966:176344. [PMID: 38280462 DOI: 10.1016/j.ejphar.2024.176344] [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: 09/17/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 01/29/2024]
Abstract
Diabetes-associated cognitive dysfunction (DACD) is a complication of diabetes mellitus that leads to an increased risk of cognitive impairment and dementia. However, the molecular mechanism underlying DACD has not been elucidated, and a promising therapy for this disease remains to be established. Hydrogen sulfide (H2S), a significant antioxidative and anti-inflammatory gasotransmitter, has emerged as a neuroprotective agent. In this study, we investigated the protective effects of H2S on DACD in a streptozotocin (STZ)-induced diabetic rat model. We applied the Morris water maze to evaluate spatial learning and memory abilities. We used Western blotting and immunohistochemical staining to investigate the expression of the Nrf-2/HO-1 axis and the NLRP3 inflammasome. After NaHS (H2S donor) administration, diabetic rats exhibited improved spatial learning and memory retrieval abilities in the Morris water maze. In STZ-induced diabetic rats, the protein expression levels of the Nrf-2/HO-1 axis, the NLRP3 inflammasome and subsequent inflammatory cytokines in the hippocampal region were elevated compared to those in control rats. Exogenous H2S triggered Nrf-2/HO-1 antioxidant activity and inhibited NLRP3 inflammasome activation and proinflammatory cytokine expression. These findings suggested that exogenous H2S has neuroprotective effects by modulating the Nrf-2/HO-1 axis and the NLRP3 inflammasome pathway, which were found to be associated with DACD. H2S treatment may be a promising therapeutic strategy for preventing the progression of tissue damage caused by DACD.
Collapse
Affiliation(s)
- Huinan Chen
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hongxue Sun
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Wei Hua
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hongye Chang
- Department of Neurology, Xi'an Central Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, China
| | - Wenjia Chen
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shuainan Ma
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| |
Collapse
|
3
|
Foster TC. Animal models for studies of alcohol effects on the trajectory of age-related cognitive decline. Alcohol 2023; 107:4-11. [PMID: 35504438 DOI: 10.1016/j.alcohol.2022.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 02/04/2023]
Abstract
There is growing interest in understanding how ethanol use interacts with advancing age to influence the brain and cognition. Animal models are employed to investigate the cellular and molecular mechanisms of brain aging and age-related neurodegenerative diseases that underlie cognitive decline. However, all too often research on problems and diseases of the elderly are conducted in healthy young animals, providing little clinical relevance. The validity of animal models is discussed, and confounds due to age-related differences in anxiety, sensory-motor function, and procedural learning are highlighted in order to enhance the successful translation of preclinical results into clinical settings. The mechanism of action of ethanol on brain aging will depend on the dose, acute or chronic treatment, or withdrawal from treatment and the age examined. Due to the fact that humans experience alcohol use throughout life, important questions concern the effects of the dose and duration of ethanol treatment on the trajectory of cognitive function. Central to this research will be questions of the specificity of alcohol effects on cognitive functions and related brain regions that decline with age, as well as the interaction of alcohol with mechanisms or biomarkers of brain aging. Alternatively, moderate alcohol use may provide a source of reserve and resilience against brain aging. Longitudinal studies have the advantage of being sensitive to detecting the effects of treatment on the emergence of cognitive impairment in middle age and can minimize effects of stress/anxiety associated with the novelty of alcohol exposure and behavioral testing, which disproportionately influence aged animals. Finally, the effect of alcohol on senescent neurophysiology and biomarkers of brain aging are discussed. In particular, the interaction of age and effects of alcohol on inflammation, oxidative stress, N-methyl-d-aspartate receptor function, and the balance of excitatory and inhibitory synaptic transmission are highlighted.
Collapse
Affiliation(s)
- Thomas C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States.
| |
Collapse
|
4
|
Pharmacological Modulations of Nrf2 and Therapeutic Implications in Aneurysmal Subarachnoid Hemorrhage. Molecules 2023; 28:molecules28041747. [PMID: 36838735 PMCID: PMC9963186 DOI: 10.3390/molecules28041747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
An aneurysmal subarachnoid hemorrhage (aSAH) is a subtype of stroke with high morbidity and mortality. The main causes of a poor prognosis include early brain injury (EBI) and delayed vasospasm, both of which play a significant role in the pathophysiological process. As an important mechanism of EBI and delayed vasospasm, oxidative stress plays an important role in the pathogenesis of aSAH by producing reactive oxygen species (ROS) through the mitochondria, hemoglobin, or enzymatic pathways in the early stages of aSAH. As a result, antioxidant therapy, which primarily targets the Nrf2-related pathway, can be employed as a potential strategy for treating aSAH. In the early stages of aSAH development, increasing the expression of antioxidant enzymes and detoxifying enzymes can relieve oxidative stress, reduce brain damage, and improve prognosis. Herein, the regulatory mechanisms of Nrf2 and related pharmacological compounds are reviewed, and Nrf2-targeted drugs are proposed as potential treatments for aSAH.
Collapse
|
5
|
Siracusa R, D’Amico R, Fusco R, Impellizzeri D, Peritore AF, Gugliandolo E, Crupi R, Interdonato L, Cordaro M, Cuzzocrea S, Di Paola R. Açai Berry Attenuates Cyclophosphamide-Induced Damage in Genitourinary Axis-Modulating Nrf-2/HO-1 Pathways. Antioxidants (Basel) 2022; 11:antiox11122355. [PMID: 36552563 PMCID: PMC9774754 DOI: 10.3390/antiox11122355] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Cyclophosphamide (CYP) is used to treat different malignancies and autoimmune disorders in men. This chemotherapy frequently reduces tumors, which is beneficial, but also causes infertility because of severe oxidative stress, inflammation, and apoptosis in the bladder and testes brought on by its metabolite, acrolein. The goal of this study was to assess the efficacy of a novel food, açai berry, in preventing CYP-induced damage in the bladder and testes. METHODS CYP was administered intraperitoneally once during the experiment at a dose of 200 mg/kg body weight diluted in 10 mL/kg b.w. of water. Açai berry was administered orally at a dose of 500 mg/kg. RESULTS The administration of açai berry was able to reduce inflammation, oxidative stress, lipid peroxidation, apoptosis, and histological changes in the bladder and testes after CYP injection. CONCLUSIONS Our findings show for the first time that açai berry modulates physiological antioxidant defenses to protect the bladder and testes against CYP-induced changes.
Collapse
Affiliation(s)
- Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy
| | - Ramona D’Amico
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy
| | - Alessio Filippo Peritore
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy
| | - Enrico Gugliandolo
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Rosalia Crupi
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Livia Interdonato
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy
| | - Marika Cordaro
- Department of Biomedical Dental and Morphological and Functional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
- Correspondence:
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, Saint Louis, MO 63104, USA
| | - Rosanna Di Paola
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| |
Collapse
|
6
|
Modification of Ischemia/Reperfusion-Induced Alterations in Subcellular Organelles by Ischemic Preconditioning. Int J Mol Sci 2022; 23:ijms23073425. [PMID: 35408783 PMCID: PMC8998910 DOI: 10.3390/ijms23073425] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 02/07/2023] Open
Abstract
It is now well established that ischemia/reperfusion (I/R) injury is associated with the compromised recovery of cardiac contractile function. Such an adverse effect of I/R injury in the heart is attributed to the development of oxidative stress and intracellular Ca2+-overload, which are known to induce remodeling of subcellular organelles such as sarcolemma, sarcoplasmic reticulum, mitochondria and myofibrils. However, repeated episodes of brief periods of ischemia followed by reperfusion or ischemic preconditioning (IP) have been shown to improve cardiac function and exert cardioprotective actions against the adverse effects of prolonged I/R injury. This protective action of IP in attenuating myocardial damage and subcellular remodeling is likely to be due to marked reductions in the occurrence of oxidative stress and intracellular Ca2+-overload in cardiomyocytes. In addition, the beneficial actions of IP have been attributed to the depression of proteolytic activities and inflammatory levels of cytokines as well as the activation of the nuclear factor erythroid factor 2-mediated signal transduction pathway. Accordingly, this review is intended to describe some of the changes in subcellular organelles, which are induced in cardiomyocytes by I/R for the occurrence of oxidative stress and intracellular Ca2+-overload and highlight some of the mechanisms for explaining the cardioprotective effects of IP.
Collapse
|
7
|
Amirshahrokhi K, Niapour A. Methylsulfonylmethane protects against ethanol-induced brain injury in mice through the inhibition of oxidative stress, proinflammatory mediators and apoptotic cell death. Int Immunopharmacol 2022; 106:108638. [PMID: 35203043 DOI: 10.1016/j.intimp.2022.108638] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 02/04/2022] [Accepted: 02/16/2022] [Indexed: 11/17/2022]
Abstract
Excessive ethanol consumption causes brain injury through oxidative stress, inflammation and apoptotic cell death. Methylsulfonylmethane (MSM) is a natural compound that has therapeutic effects on oxidative and inflammatory disorders. The aim of this study was to investigate the protective effect and underlying mechanisms of MSM on ethanol-induced brain injury in an experimental model. Male C57BL/6 mice were exposed to binge ethanol (5 g/kg/day, orally) and treated with MSM (200 and 400 mg/kg/day) concomitantly for 12 days. At the end of the experiment brain tissues were removed for histological and biochemical analysis. The results showed that MSM reduced ethanol-mediated oxidative stress by decreasing the levels of malondialdehyde (MDA) and carbonyl protein. The Nrf2/HO-1 pathway and the levels of cytoprotective antioxidants superoxide dismutase (SOD), catalase and glutathione (GSH) were increased by MSM in the brain tissue. MSM treatment reduced the ethanol-induced inflammatory factors including myeloperoxidase (MPO), iNOS/NO, cyclooxygenase (COX)-2, nuclear factor kappa B (NF-κB), NLRP3 inflammasome and proinflammatory cytokines including TNF-α, IL-1β, IL-6 and MCP-1. MSM also decreased the levels of pro-apoptotic caspase-3 and TUNEL positive cells while increased the level of anti-apoptotic Bcl-2 in the brain tissue. Our findings demonstrated that MSM protects against ethanol-induced brain injury by improving anti-oxidant defense mechanism and reducing ethanol-mediated inflammation and apoptosis. Therefore, MSM may be a potential protective approach for brain damage caused by high levels of alcohol.
Collapse
Affiliation(s)
- Keyvan Amirshahrokhi
- Department of Pharmacology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Ali Niapour
- Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
8
|
Habib CN, Mohamed MR, Tadros MG, Tolba MF, Menze ET, Masoud SI. The potential neuroprotective effect of diosmin in rotenone-induced model of Parkinson's disease in rats. Eur J Pharmacol 2022; 914:174573. [PMID: 34656609 DOI: 10.1016/j.ejphar.2021.174573] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/21/2022]
Abstract
Most treatments for Parkinson's disease (PD) focus on improving the symptoms and the dopaminergic effects; nevertheless, they cannot delay the disease progression. Diosmin (DM), a naturally occurring flavone that is obtained from citrus fruits, has demonstrated anti-apoptotic, anti-inflammatory and antioxidative properties in many diseases. This study aimed to assess the neuroprotective effects of diosmin in rotenone-induced rat model of PD and investigate its potential underlying mechanisms. A preliminary dose-response study was conducted where rats were treated with DM (50,100 and 200 mg/kg, p.o.) concomitantly with rotenone (2 mg/kg, s.c.) for 4 weeks. Catalepsy, motor impairment, spontaneous locomotion, body weight, histological examination and tyrosine hydroxylase (TH) immunoreactivity were evaluated in both the midbrains and striata of rats. Treatment with DM (200 mg/kg) showed the most promising outcome therefore, it was selected for further evaluation of α-synuclein, Bax, Bcl2, nuclear factor kappa B (NF-кB), nuclear factor erythroid 2- related factor 2 (Nrf2), and heme oxygenase-1 (HO-1), in addition to biochemical analysis of tumor necrosis factor-α (TNF-α). Results showed that DM (200 mg/kg, p.o.) prevented rotenone-induced motor impairment, weight reduction and histological damage. Furthermore, it significantly inhibited rotenone-induced decrease in TH expression. These results were correlated with reduction in α-synuclein immunoreactivity, together with improvement of Bax/Bcl2 ratio compared to rotenone group. DM also attenuated rotenone-induced increase in NF-кB expression as well as TNF- α levels. Moreover, DM inhibited rotenone-induced upregulation of Nrf2/HO-1 pathway. Thus, the current study suggests that DM might be a promising candidate for managing the neuropathological course of PD.
Collapse
Affiliation(s)
- Christine N Habib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt.
| | - Mohamed R Mohamed
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Mariane G Tadros
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Mai F Tolba
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt; School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, New Administrative Capital, Egypt
| | - Esther T Menze
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Somia I Masoud
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
9
|
Fibbi B, Marroncini G, Anceschi C, Naldi L, Peri A. Hyponatremia and Oxidative Stress. Antioxidants (Basel) 2021; 10:1768. [PMID: 34829639 PMCID: PMC8614907 DOI: 10.3390/antiox10111768] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/26/2021] [Accepted: 11/03/2021] [Indexed: 12/15/2022] Open
Abstract
Hyponatremia, i.e., the presence of a serum sodium concentration ([Na+]) < 136 mEq/L, is the most frequent electrolyte imbalance in the elderly and in hospitalized patients. Symptoms of acute hyponatremia, whose main target is the central nervous system, are explained by the "osmotic theory" and the neuronal swelling secondary to decreased extracellular osmolality, which determines cerebral oedema. Following the description of neurological and systemic manifestations even in mild and chronic hyponatremia, in the last decade reduced extracellular [Na+] was associated with detrimental effects on cellular homeostasis independently of hypoosmolality. Most of these alterations appeared to be elicited by oxidative stress. In this review, we focus on the role of oxidative stress on both osmolality-dependent and -independent impairment of cell and tissue functions observed in hyponatremic conditions. Furthermore, basic and clinical research suggested that oxidative stress appears to be a common denominator of the degenerative processes related to aging, cancer progression, and hyponatremia. Of note, low [Na+] is able to exacerbate multiple manifestations of senescence and to decrease progression-free and overall survival in oncologic patients.
Collapse
Affiliation(s)
- Benedetta Fibbi
- Pituitary Diseases and Sodium Alterations Unit, AOU Careggi, 50139 Florence, Italy; (B.F.); (G.M.)
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, AOU Careggi, 50139 Florence, Italy; (C.A.); (L.N.)
| | - Giada Marroncini
- Pituitary Diseases and Sodium Alterations Unit, AOU Careggi, 50139 Florence, Italy; (B.F.); (G.M.)
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, AOU Careggi, 50139 Florence, Italy; (C.A.); (L.N.)
| | - Cecilia Anceschi
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, AOU Careggi, 50139 Florence, Italy; (C.A.); (L.N.)
| | - Laura Naldi
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, AOU Careggi, 50139 Florence, Italy; (C.A.); (L.N.)
| | - Alessandro Peri
- Pituitary Diseases and Sodium Alterations Unit, AOU Careggi, 50139 Florence, Italy; (B.F.); (G.M.)
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, AOU Careggi, 50139 Florence, Italy; (C.A.); (L.N.)
| |
Collapse
|
10
|
Ahn YJ, Shin HJ, Jeong EA, An HS, Lee JY, Jang HM, Kim KE, Lee J, Shin MC, Roh GS. Exendin-4 Pretreatment Attenuates Kainic Acid-Induced Hippocampal Neuronal Death. Cells 2021; 10:cells10102527. [PMID: 34685508 PMCID: PMC8534217 DOI: 10.3390/cells10102527] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 12/15/2022] Open
Abstract
Exendin-4 (Ex-4) is a glucagon-like peptide-1 receptor (GLP-1R) agonist that protects against brain injury. However, little is known about the effect of Ex-4 on kainic acid (KA)-induced seizures and hippocampal cell death. Therefore, this study evaluated the neuroprotective effects of Ex-4 pretreatment in a mouse model of KA-induced seizures. Three days before KA treatment, mice were intraperitoneally injected with Ex-4. We found that Ex-4 pretreatment reversed KA-induced reduction of GLP-1R expression in the hippocampus and attenuated KA-induced seizure score, hippocampal neuronal death, and neuroinflammation. Ex-4 pretreatment also dramatically reduced hippocampal lipocalin-2 protein in KA-treated mice. Furthermore, immunohistochemical studies showed that Ex-4 pretreatment significantly alleviated blood–brain barrier leakage. Finally, Ex-4 pretreatment stimulated hippocampal expression of phosphorylated cyclic adenosine monophosphate (cAMP) response element-binding protein (p-CREB), a known target of GLP-1/GLP-1R signaling. These findings indicate that Ex-4 pretreatment may protect against KA-induced neuronal damage by regulating GLP-1R/CREB-mediated signaling pathways.
Collapse
Affiliation(s)
- Yu-Jeong Ahn
- Bio Anti-Aging Medical Research Center, Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (Y.-J.A.); (H.-J.S.); (E.-A.J.); (H.-S.A.); (J.-Y.L.); (H.-M.J.); (K.-E.K.); (J.L.)
| | - Hyun-Joo Shin
- Bio Anti-Aging Medical Research Center, Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (Y.-J.A.); (H.-J.S.); (E.-A.J.); (H.-S.A.); (J.-Y.L.); (H.-M.J.); (K.-E.K.); (J.L.)
| | - Eun-Ae Jeong
- Bio Anti-Aging Medical Research Center, Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (Y.-J.A.); (H.-J.S.); (E.-A.J.); (H.-S.A.); (J.-Y.L.); (H.-M.J.); (K.-E.K.); (J.L.)
| | - Hyeong-Seok An
- Bio Anti-Aging Medical Research Center, Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (Y.-J.A.); (H.-J.S.); (E.-A.J.); (H.-S.A.); (J.-Y.L.); (H.-M.J.); (K.-E.K.); (J.L.)
| | - Jong-Youl Lee
- Bio Anti-Aging Medical Research Center, Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (Y.-J.A.); (H.-J.S.); (E.-A.J.); (H.-S.A.); (J.-Y.L.); (H.-M.J.); (K.-E.K.); (J.L.)
| | - Hye-Min Jang
- Bio Anti-Aging Medical Research Center, Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (Y.-J.A.); (H.-J.S.); (E.-A.J.); (H.-S.A.); (J.-Y.L.); (H.-M.J.); (K.-E.K.); (J.L.)
| | - Kyung-Eun Kim
- Bio Anti-Aging Medical Research Center, Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (Y.-J.A.); (H.-J.S.); (E.-A.J.); (H.-S.A.); (J.-Y.L.); (H.-M.J.); (K.-E.K.); (J.L.)
| | - Jaewoong Lee
- Bio Anti-Aging Medical Research Center, Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (Y.-J.A.); (H.-J.S.); (E.-A.J.); (H.-S.A.); (J.-Y.L.); (H.-M.J.); (K.-E.K.); (J.L.)
| | - Meong-Cheol Shin
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Gyeongsang National University, Jinju 52828, Korea;
| | - Gu-Seob Roh
- Bio Anti-Aging Medical Research Center, Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (Y.-J.A.); (H.-J.S.); (E.-A.J.); (H.-S.A.); (J.-Y.L.); (H.-M.J.); (K.-E.K.); (J.L.)
- Correspondence: ; Tel.: +82-55-772-8035; Fax: +82-55-772-8039
| |
Collapse
|
11
|
Hidrox ® Roles in Neuroprotection: Biochemical Links between Traumatic Brain Injury and Alzheimer's Disease. Antioxidants (Basel) 2021; 10:antiox10050818. [PMID: 34065584 PMCID: PMC8161307 DOI: 10.3390/antiox10050818] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/07/2021] [Accepted: 05/19/2021] [Indexed: 12/22/2022] Open
Abstract
Traumatic brain injuries (TBI) are a serious public-health problem. Furthermore, subsequent TBI events can compromise TBI patients’ quality of life. TBI is linked to a number of long- and short-term complications such as cerebral atrophy and risk of developing dementia and Alzheimer’s Disease (AD). Following direct TBI damage, oxidative stress and the inflammatory response lead to tissue injury-associated neurodegenerative processes that are characteristic of TBI-induced secondary damage. Hidrox® showed positive effects in preclinical models of toxic oxidative stress and neuroinflammation; thus, the aim of this study was to evaluate the effect of Hidrox® administration on TBI-induced secondary injury and on the propagation of the AD-like neuropathology. Hidrox® treatment reduced histological damage after controlled cortical impact. Form a molecular point of view, hydroxytyrosol is able to preserve the cellular redox balance and protein homeostasis by activating the Nrf2 pathway and increasing the expression of phase II detoxifying enzymes such as HO-1, SOD, Catalase, and GSH, thus counteracting the neurodegenerative damage. Additionally, Hidrox® showed anti-inflammatory effects by reducing the activation of the NFkB pathway and related cytokines overexpression. From a behavioral point of view, Hidrox® treatment ameliorated the cognitive dysfunction and memory impairment induced by TBI. Additionally, Hidrox® was associated with a significant increased number of hippocampal neurons in the CA3 region, which were reduced post-TBI. In particular, Hidrox® decreased AD-like phenotypic markers such as ß-amyloid accumulation and APP and p-Tau overexpression. These findings indicate that Hidrox® could be a valuable treatment for TBI-induced secondary injury and AD-like pathological features.
Collapse
|
12
|
Fusco R, Salinaro AT, Siracusa R, D’Amico R, Impellizzeri D, Scuto M, Ontario ML, Crea R, Cordaro M, Cuzzocrea S, Di Paola R, Calabrese V. Hidrox ® Counteracts Cyclophosphamide-Induced Male Infertility through NRF2 Pathways in a Mouse Model. Antioxidants (Basel) 2021; 10:antiox10050778. [PMID: 34068924 PMCID: PMC8156985 DOI: 10.3390/antiox10050778] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/28/2021] [Accepted: 05/11/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Every year, men use cyclophosphamide to treat various cancers and autoimmune diseases. On the one hand, this chemotherapy often has the beneficial effect of regressing the tumor, but on the other hand, it leads to infertility due to excessive oxidative stress and apoptosis in the testes caused by its metabolite, acrolein. METHODS The objective of this study was to evaluate the beneficial power of a new compound called Hidrox®, containing 40-50% hydroxytyrosol, in counteracting the damage related to fertility induced by cyclophosphamide. The study was conducted using a single intraperitoneal injection of cyclophosphamide at a dose of 200 mg/kg b.w, in distilled water at 10 mL/kg b.w. The treatment was administered via the oral administration of Hidrox® at a dose of 50 mg/kg. RESULTS Our study confirms that the use of cyclophosphamide causes a series of sperm and histological alterations strongly connected with oxidative stress, lipid peroxidation, and apoptosis. CONCLUSION Our results demonstrate for the first time that Hidrox® protects testes from CYP-induced alterations by the modulation of physiological antioxidant defenses.
Collapse
Affiliation(s)
- Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (R.D.); (D.I.); (R.D.P.)
| | - Angela Trovato Salinaro
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95131 Catania, Italy; (A.T.S.); (M.S.); (M.L.O.); (V.C.)
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (R.D.); (D.I.); (R.D.P.)
| | - Ramona D’Amico
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (R.D.); (D.I.); (R.D.P.)
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (R.D.); (D.I.); (R.D.P.)
| | - Maria Scuto
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95131 Catania, Italy; (A.T.S.); (M.S.); (M.L.O.); (V.C.)
| | - Maria Laura Ontario
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95131 Catania, Italy; (A.T.S.); (M.S.); (M.L.O.); (V.C.)
| | - Roberto Crea
- Oliphenol LLC, 26225 Eden Landing Road, Unit C, Hayward, CA 94545, USA;
| | - Marika Cordaro
- Department of Biomedical, Dental and Morphological and Functional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
- Correspondence: (M.C.); (S.C.); Tel.: +39-090-676-5208 (M.C. & S.C.)
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (R.D.); (D.I.); (R.D.P.)
- Correspondence: (M.C.); (S.C.); Tel.: +39-090-676-5208 (M.C. & S.C.)
| | - Rosanna Di Paola
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (R.F.); (R.S.); (R.D.); (D.I.); (R.D.P.)
| | - Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95131 Catania, Italy; (A.T.S.); (M.S.); (M.L.O.); (V.C.)
| |
Collapse
|
13
|
Jayanti S, Vítek L, Tiribelli C, Gazzin S. The Role of Bilirubin and the Other "Yellow Players" in Neurodegenerative Diseases. Antioxidants (Basel) 2020; 9:E900. [PMID: 32971784 PMCID: PMC7555389 DOI: 10.3390/antiox9090900] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/16/2020] [Accepted: 09/20/2020] [Indexed: 02/07/2023] Open
Abstract
Bilirubin is a yellow endogenous derivate of the heme catabolism. Since the 1980s, it has been recognized as one of the most potent antioxidants in nature, able to counteract 10,000× higher intracellular concentrations of H2O2. In the recent years, not only bilirubin, but also its precursor biliverdin, and the enzymes involved in their productions (namely heme oxygenase and biliverdin reductase; altogether the "yellow players"-YPs) have been recognized playing a protective role in diseases characterized by a chronic prooxidant status. Based on that, there is an ongoing effort in inducing their activity as a therapeutic option. Nevertheless, the understanding of their specific contributions to pathological conditions of the central nervous system (CNS) and their role in these diseases are limited. In this review, we will focus on the most recent evidence linking the role of the YPs specifically to neurodegenerative and neurological conditions. Both the protective, as well as potentially worsening effects of the YP's activity will be discussed.
Collapse
Affiliation(s)
- Sri Jayanti
- Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163.5, Basovizza, 34149 Trieste, Italy; (S.J.); (C.T.)
- Faculty of Medicine, Universitas Hasanuddin, Makassar 90245, Indonesia
- Molecular Biomedicine Ph.D. Program, University of Trieste, 34127 Trieste, Italy
| | - Libor Vítek
- Institute of Medical Biochemistry and Laboratory Diagnostics, and 4th Department of Internal Medicine, Faculty General Hospital and 1st Faculty of Medicine, Charles University, 12000 Prague, Czech Republic;
| | - Claudio Tiribelli
- Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163.5, Basovizza, 34149 Trieste, Italy; (S.J.); (C.T.)
| | - Silvia Gazzin
- Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163.5, Basovizza, 34149 Trieste, Italy; (S.J.); (C.T.)
| |
Collapse
|
14
|
Severo L, Godinho D, Machado F, Hartmann D, Fighera MR, Soares FA, Furian AF, Oliveira MS, Royes LF. The role of mitochondrial bioenergetics and oxidative stress in depressive behavior in recurrent concussion model in mice. Life Sci 2020; 257:117991. [PMID: 32569782 DOI: 10.1016/j.lfs.2020.117991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 06/09/2020] [Accepted: 06/18/2020] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) is a public health problem in which even though 80 to 90% of cases are considered mild, usually starts a sequence of neurological disorders that can last a considerable time. Most of the research of this injury has been focused on oxidative stress and functional deficits; however, mechanisms that underlie the development of neuropsychiatric disorders remain little researched. Due to this, the present authors decided to investigate whether recurrent concussion protocols alter depressive-like phenotype behavior, and whether mitochondria play an indispensable role in this behavior or not. The experimental data revealed, for the first time, that the present protocol of recurrent concussions (4, 7, and 10 injuries) in mice did not alter immobility time during tail suspension tests (TSTs), but decreased hippocampal mitochondrial respiration and increased expression of proteins such as nuclear factor erythroid 2-related factor 2 (Nrf2) and superoxide (SOD2). This experimental data suggests that bioenergetic changes elicited by recurrent concussion did not induce depressive-like behavior, but activated the transcription factor of responsive antioxidant elements (ARE) that delay or prevent secondary cascades in this neurological disease.
Collapse
Affiliation(s)
- Leandro Severo
- Programa de Pós-Graduação em Ciências Biológicas, Bioquímica Toxicológica Universidade Federal de Santa Maria, 97105-900, Brazil; Laboratório de Bioquímica do Exercício Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil.
| | - Douglas Godinho
- Programa de Pós-Graduação em Ciências Biológicas, Bioquímica Toxicológica Universidade Federal de Santa Maria, 97105-900, Brazil; Laboratório de Bioquímica do Exercício Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | - Felipe Machado
- Laboratório de Bioquímica do Exercício Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | - Diane Hartmann
- Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria, 97105-900, Brazil
| | - Michele Rechia Fighera
- Programa de Pós-Graduação em Ciências Biológicas, Bioquímica Toxicológica Universidade Federal de Santa Maria, 97105-900, Brazil; Laboratório de Bioquímica do Exercício Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | - Félix Alexandre Soares
- Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria, 97105-900, Brazil
| | - Ana Flavia Furian
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia-Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | - Mauro Schneider Oliveira
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia-Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | - Luiz Fernando Royes
- Programa de Pós-Graduação em Ciências Biológicas, Bioquímica Toxicológica Universidade Federal de Santa Maria, 97105-900, Brazil; Laboratório de Bioquímica do Exercício Universidade Federal de Santa Maria, 97105-900 Santa Maria, RS, Brazil.
| |
Collapse
|
15
|
Zhou L, Zhou M, Tan H, Xiao M. Cypermethrin-induced cortical neurons apoptosis via the Nrf2/ARE signaling pathway. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2020; 165:104547. [PMID: 32359539 DOI: 10.1016/j.pestbp.2020.02.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 02/09/2020] [Accepted: 02/17/2020] [Indexed: 06/11/2023]
Abstract
Pesticide residue is a common problem worldwide. Cypermethrin is a type II pyrethroid pesticide that has been widely used in recent years. It has become a widespread residual pesticide in the environment and agricultural products. The neurotoxicity of cypermethrin remains a matter of concern. However, few studies have evaluated its toxicity on cerebral cortical neurons. As the center of the nervous system, the cerebral cortex is involved in a series of biological processes, such as learning, memory, emotions, and movement. The Nrf2/ARE signaling pathway has been considered to play a protective role in several central nervous system (CNS) diseases. We investigated whether this pathway plays a protective role in cypermethrin-induced apoptosis of the cortical neurons. We established a cypermethrin-induced apoptosis model in the cortical neurons using different cypermethrin doses and different incubation periods. The changes in Nrf2 protein and mRNA expression and its downstream genes HO-1 and NQO1 were detected by quantitative real-time PCR and Western blotting to study the role of the Nrf2/ARE pathway in cypermethrin-induced apoptosis of the cortical neurons. The results showed that the Nrf2/ARE signaling pathway has a protective effect in cypermethrin-induced apoptosis of the cortical neurons. However, this protective effect of the Nrf2/ARE pathway is very limited and is dependent on the exposure dose and exposure period of cypermethrin.
Collapse
Affiliation(s)
- Lihua Zhou
- School of Public Health, Bengbu Medical College, Bengbu, Anhui 233030, China.
| | - Mengqing Zhou
- School of Public Health, Bengbu Medical College, Bengbu, Anhui 233030, China
| | - Handan Tan
- School of Public Health, Bengbu Medical College, Bengbu, Anhui 233030, China
| | - Mengxi Xiao
- School of Public Health, Bengbu Medical College, Bengbu, Anhui 233030, China
| |
Collapse
|
16
|
Song S, Chen Y, Han F, Dong M, Xiang X, Sui J, Li Y, Yang H, Liu J. Aloperine activates the Nrf2-ARE pathway when ameliorating early brain injury in a subarachnoid hemorrhage model. Exp Ther Med 2018; 15:3847-3855. [PMID: 29563984 PMCID: PMC5858125 DOI: 10.3892/etm.2018.5896] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Accepted: 10/13/2017] [Indexed: 12/14/2022] Open
Abstract
Aloperine (ALO) exhibits neuroprotective effects against oxidative stress in vitro; however, its protective effect in early brain injury (EBI) following experimental subarachnoid hemorrhage (SAH) remains to be elucidated. The aim of the current study was to evaluate the antioxidant activity of ALO in EBI, and its association with nuclear factor erythroid-related factor 2 and the antioxidant responsive element (Nrf2-ARE) survival pathway. In the present study, an experimental SAH model was induced in rats following a prechiasmatic cistern injection. All rats were randomly divided into five groups: Sham, SAH, SAH+ vehicle, and an SAH+ ALO group (including low and high doses). ALO was administrated intraperitoneally at 2 and 24 h following induction of the SAH model. Brain samples were collected from each group at 48 h after SAH induction. Subsequently, western blotting, immunohistochemistry and cell apoptosis assays were performed, along with assessments for brain edema, neurological deficit, and the activity of oxidant/antioxidant factors. It was observed that the expression of Nrf2-ARE pathway-associated agents, including Nrf2, and heme oxygenase-1, were markedly increased in the high concentration ALO group compared with that of the SAH group. In addition, the level of oxidative damage was reduced. Furthermore, early brain damage, including brain edema, neurological deficit and cellular apoptosis were significantly ameliorated. In conclusion, the results of the present study indicate that ALO can ameliorate oxidative damage against EBI following SAH, most likely via the Nrf2-ARE survival pathway.
Collapse
Affiliation(s)
- Shibin Song
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Yimin Chen
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Feng Han
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Minghao Dong
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Xin Xiang
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Jianmei Sui
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Yuming Li
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Hua Yang
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Jian Liu
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| |
Collapse
|
17
|
Fang Y, Liu X, Zhao L, Wei Z, Jiang D, Shao H, Zang Y, Xu J, Wang Q, Liu Y, Peng Y, Yin X. RhGLP-1 (7-36) protects diabetic rats against cerebral ischemia-reperfusion injury via up-regulating expression of Nrf2/HO-1 and increasing the activities of SOD. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2017; 21:475-485. [PMID: 28883752 PMCID: PMC5587598 DOI: 10.4196/kjpp.2017.21.5.475] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 11/14/2016] [Accepted: 11/14/2016] [Indexed: 12/13/2022]
Abstract
The present study aimed to explore the neuroprotective effect and possible mechanisms of rhGLP-1 (7–36) against transient ischemia/reperfusion injuries induced by middle cerebral artery occlusion (MCAO) in type 2 diabetic rats. First, diabetic rats were established by a combination of a high-fat diet and low-dose streptozotocin (STZ) (30 mg/kg, intraperitoneally). Second, they were subjected to MCAO for 2 h, then treated with rhGLP-1 (7–36) (10, 20, 40 µg/kg i.p.) at the same time of reperfusion. In the following 3 days, they were injected with rhGLP-1 (7–36) at the same dose and route for three times each day. After 72 h, hypoglycemic effects were assessed by blood glucose changes, and neuroprotective effects were evaluated by neurological deficits, infarct volume and histomorphology. Mechanisms were investigated by detecting the distribution and expression of the nuclear factor erythroid-derived factor 2 related factor 2 (Nrf2) in ischemic brain tissue, the levels of phospho-PI3 kinase (PI3K)/PI3K ratio and heme-oxygenase-1 (HO-l), as well as the activities of superoxide dismutase (SOD) and the contents of malondialdehyde (MDA). Our results showed that rhGLP-1 (7–36) significantly reduced blood glucose and infarction volume, alleviated neurological deficits, enhanced the density of surviving neurons and vascular proliferation. The nuclear positive cells ratio and expression of Nrf2, the levels of P-PI3K/PI3K ratio and HO-l increased, the activities of SOD increased and the contents of MDA decreased. The current results indicated the protective effect of rhGLP-1 (7–36) in diabetic rats following MCAO/R that may be concerned with reducing blood glucose, up-regulating expression of Nrf2/HO-1 and increasing the activities of SOD.
Collapse
Affiliation(s)
- Yi Fang
- Department of Pharmacy, Peking University People's Hospital, Beijing 100044, China
| | - Xiaofang Liu
- Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Libo Zhao
- Department of Pharmacy, Peking University People's Hospital, Beijing 100044, China
| | - Zhongna Wei
- Department of Pharmacy, Guizhou Orthopedics Hospital, Guizhou 550002, China
| | - Daoli Jiang
- Department of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Hua Shao
- Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yannan Zang
- Department of Pharmacy, Peking University People's Hospital, Beijing 100044, China
| | - Jia Xu
- Department of Pharmacy, Mawangdui Hospital, Changsha 410016, China
| | - Qian Wang
- Department of Pharmacy, Peking University People's Hospital, Beijing 100044, China
| | - Yang Liu
- Department of Pharmacy, Peking University People's Hospital, Beijing 100044, China
| | - Ye Peng
- Department of oncology, Harrison International Peace Hospital, Hengshui 053000, China
| | - Xiaoxing Yin
- Department of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| |
Collapse
|
18
|
Duncan JW, Johnson S, Zhang X, Zheng B, Luo J, Ou XM, Stockmeier CA, Wang JM. Up-Regulation of PKR Signaling Pathway by Ethanol Displays an Age of Onset-Dependent Relationship. Alcohol Clin Exp Res 2016; 40:2320-2328. [PMID: 27647657 DOI: 10.1111/acer.13209] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 08/01/2016] [Indexed: 12/16/2022]
Abstract
BACKGROUND Ethanol (EtOH) neurotoxicity can result in devastating effects on brain and behavior by disrupting homeostatic signaling cascades and inducing cell death. One such mechanism involves double-stranded RNA activated protein kinase (PKR), a primary regulator of protein translation and cell viability in the presence of a virus or other external stimuli. EtOH-mediated up-regulation of interferon-gamma (IFN-γ; the oxidative stress-inducible regulator of PKR), PKR, and its target, p53, are still being fully elucidated. METHODS Using Western blot analysis, immunofluorescence, and linear regression analyses, changes in the IFN-γ-PKR-p53 pathway following chronic EtOH treatment in the frontal cortex of rodents were examined. The role of PKR on cell viability was also assessed in EtOH-treated cells using PKR overexpression vector and PKR inhibitor (PKRI). RESULTS In rats chronically fed EtOH, PKR, phosphorylated PKR (p-PKR), IFN-γ, and p53 were significantly increased following chronic EtOH exposure. Linear regression revealed a significant correlation between IFN-γ and p-PKR protein levels, as well as p-PKR expression and age of EtOH exposure. Overexpression of PKR resulted in greater cell death, while use of PKRI enhanced cell viability in EtOH-treated cells. CONCLUSIONS Chronic EtOH exposure activates the IFN-γ-PKR-p53 pathway in the frontal cortex of rodents. p-PKR expression is greater in brains of rodents exposed to EtOH at earlier ages compared to later life, suggesting a mechanism by which young brains could be more susceptible to EtOH-related brain injury. PKR and p-PKR were also colocalized in neurons and astrocytes of rats. This study provides additional insight into biochemical mechanisms underlying alcohol use disorder related neuropathology and warrants further investigation of PKR as a potential pharmacotherapeutic target to combat EtOH-related neurotoxicity, loss of protein translation and brain injury.
Collapse
Affiliation(s)
- Jeremy W Duncan
- Program in Neuroscience , University of Mississippi Medical Center, Jackson, Mississippi.,Department of Psychiatry and Human Behavior , University of Mississippi Medical Center, Jackson, Mississippi
| | - Shakevia Johnson
- Department of Psychiatry and Human Behavior , University of Mississippi Medical Center, Jackson, Mississippi
| | - Xiao Zhang
- Program in Neuroscience , University of Mississippi Medical Center, Jackson, Mississippi.,Department of Psychiatry and Human Behavior , University of Mississippi Medical Center, Jackson, Mississippi
| | - Baoying Zheng
- Department of Pathology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jia Luo
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky.,Department of Internal Medicine, University of Kentucky, Lexington, Kentucky
| | - Xiao-Ming Ou
- Department of Psychiatry and Human Behavior , University of Mississippi Medical Center, Jackson, Mississippi
| | - Craig A Stockmeier
- Department of Psychiatry and Human Behavior , University of Mississippi Medical Center, Jackson, Mississippi
| | - Jun Ming Wang
- Program in Neuroscience , University of Mississippi Medical Center, Jackson, Mississippi. .,Department of Psychiatry and Human Behavior , University of Mississippi Medical Center, Jackson, Mississippi. .,Department of Pathology, University of Mississippi Medical Center, Jackson, Mississippi.
| |
Collapse
|
19
|
Choi DJ, Cho S, Seo JY, Lee HB, Park YI. Neuroprotective effects of the Phellinus linteus ethyl acetate extract against H2O2-induced apoptotic cell death of SK-N-MC cells. Nutr Res 2015; 36:31-43. [PMID: 26773779 DOI: 10.1016/j.nutres.2015.11.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 10/16/2015] [Accepted: 11/09/2015] [Indexed: 01/09/2023]
Abstract
Numerous studies have suggested that neuronal cells are protected against oxidative stress-induced cell damage by antioxidants, such as polyphenolic compounds. Phellinus linteus (PL) has traditionally been used to treat various symptoms in East Asian countries. In the present study, we prepared an ethyl acetate extract from the fruiting bodies of PL (PLEA) using hot water extraction, ethanol precipitation, and ethyl acetate extraction. The PLEA contained polyphenols as its major chemical component, and thus, we predicted that it may exhibit antioxidant and neuroprotective effects against oxidative stress. The results showed that the pretreatment of human brain neuroblastoma SK-N-MC cells with the PLEA (0.1-5 μg/mL) significantly and dose-dependently reduced the cytotoxicity of H2O2 and the intracellular ROS levels and enhanced the expression of HO-1 (heme oxygenase-1) and antioxidant enzymes, such as CAT (catalase), GPx-1 (glutathione peroxidase-1), and SOD-1 and -2 (superoxide dismutase-1 and -2). The PLEA also directly scavenged free radicals. PLEA pretreatment also significantly attenuated DNA fragmentation and suppressed the mRNA expression and activation of mitogen-activated protein kinases extracellular signal-regulated kinase, c-Jun N-terminal kinase, and p38 kinase, which are induced by oxidative stress and lead to cell death. PLEA pretreatment inhibited the activation of the apoptosis-related proteins caspase-3 and poly (ADP-ribose) polymerase. These results demonstrate that the PLEA has neuroprotective effects against oxidative stress (H2O2)-induced neuronal cell death via its antioxidant and anti-apoptotic properties. PLEA should be investigated in an in vivo model on its potential to prevent or ameliorate neurodegenerative disease.
Collapse
Affiliation(s)
- Doo Jin Choi
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Gyeonggi-do, 420-743, Republic of Korea
| | - Sarang Cho
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Gyeonggi-do, 420-743, Republic of Korea
| | - Jeong Yeon Seo
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Gyeonggi-do, 420-743, Republic of Korea
| | - Hyang Burm Lee
- Division of Food Technology, Biotechnology & Agrochemistry, Chonnam National University, Buk-Gu, Gwangju, 500-757, Republic of Korea
| | - Yong Il Park
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Gyeonggi-do, 420-743, Republic of Korea.
| |
Collapse
|
20
|
Han Y, Yi W, Qin J, Zhao Y, Zhang J, Chang X. Carbon monoxide offers neuroprotection from hippocampal cell damage induced by recurrent febrile seizures through the PERK-activated ER stress pathway. Neurosci Lett 2015; 585:126-31. [DOI: 10.1016/j.neulet.2014.11.040] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 11/05/2014] [Accepted: 11/25/2014] [Indexed: 12/11/2022]
|
21
|
Astaxanthin activates nuclear factor erythroid-related factor 2 and the antioxidant responsive element (Nrf2-ARE) pathway in the brain after subarachnoid hemorrhage in rats and attenuates early brain injury. Mar Drugs 2014; 12:6125-41. [PMID: 25528957 PMCID: PMC4278222 DOI: 10.3390/md12126125] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 11/19/2014] [Accepted: 12/08/2014] [Indexed: 01/25/2023] Open
Abstract
Astaxanthin (ATX) has been proven to ameliorate early brain injury (EBI) after experimental subarachnoid hemorrhage (SAH) by modulating cerebral oxidative stress. This study was performed to assess the effect of ATX on the Nrf2-ARE pathway and to explore the underlying molecular mechanisms of antioxidant properties of ATX in EBI after SAH. A total of 96 male SD rats were randomly divided into four groups. Autologous blood was injected into the prechiasmatic cistern of the rat to induce an experimental SAH model. Rats in each group were sacrificed at 24 h after SAH. Expressions of Nrf2 and heme oxygenase-1 (HO-1) were measured by Western blot and immunohistochemistry analysis. The mRNA levels of HO-1, NAD (P) H: quinone oxidoreductase 1 (NQO-1), and glutathione S-transferase-α1 (GST-α1) were determined by real-time polymerase chain reaction (PCR). It was observed that administration of ATX post-SAH could up-regulate the cortical expression of these agents, mediated in the Nrf2-ARE pathway at both pretranscriptional and posttranscriptional levels. Meanwhile, oxidative damage was reduced. Furthermore, ATX treatment significantly attenuated brain edema, blood–brain barrier (BBB) disruption, cellular apoptosis, and neurological dysfunction in SAH models. This study demonstrated that ATX treatment alleviated EBI in SAH model, possibly through activating the Nrf2-ARE pathway by inducing antioxidant and detoxifying enzymes.
Collapse
|
22
|
Giuliani C, Peri A. Effects of Hyponatremia on the Brain. J Clin Med 2014; 3:1163-77. [PMID: 26237597 PMCID: PMC4470176 DOI: 10.3390/jcm3041163] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/18/2014] [Accepted: 10/10/2014] [Indexed: 12/31/2022] Open
Abstract
Hyponatremia is a very common electrolyte disorder, especially in the elderly, and is associated with significant morbidity, mortality and disability. In particular, the consequences of acute hyponatremia on the brain may be severe, including permanent disability and death. Also chronic hyponatremia can affect the health status, causing attention deficit, gait instability, increased risk of falls and fractures, and osteoporosis. Furthermore, an overly rapid correction of hyponatremia can be associated with irreversible brain damage, which may be the result of the osmotic demyelination syndrome. This review analyzes the detrimental consequences of acute and chronic hyponatremia and its inappropriate correction on the brain and the underlying physiopathological mechanisms, with a particular attention to the less known in vivo and in vitro effects of chronic hyponatremia.
Collapse
Affiliation(s)
- Corinna Giuliani
- Endocrine Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence 50139, Italy.
| | - Alessandro Peri
- Endocrine Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence 50139, Italy.
| |
Collapse
|
23
|
Choi DJ, Kim SL, Choi JW, Park YI. Neuroprotective effects of corn silk maysin via inhibition of H2O2-induced apoptotic cell death in SK-N-MC cells. Life Sci 2014; 109:57-64. [PMID: 24928367 DOI: 10.1016/j.lfs.2014.05.020] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 04/21/2014] [Accepted: 05/30/2014] [Indexed: 02/04/2023]
Abstract
AIMS Neuroprotective effects of maysin, which is a flavone glycoside that was isolated from the corn silk (CS, Zea mays L.) of a Korean hybrid corn Kwangpyeongok, against oxidative stress (H2O2)-induced apoptotic cell death of human neuroblastoma SK-N-MC cells were investigated. MAIN METHODS Maysin cytotoxicity was determined by measuring cell viability using MTT and lactate dehydrogenase (LDH) assays. Intracellular reactive oxygen species (ROS) were measured using a 2,7-dichlorofluorescein diacetate (DCF-DA) assay. Apoptotic cell death was monitored by annexin V-FITC/PI double staining and by a TUNEL assay. Antioxidant enzyme mRNA levels were determined by real-time PCR. The cleavage of poly (ADP-ribose) polymerase (PARP) was measured by western blotting. KEY FINDINGS Maysin pretreatment reduced the cytotoxic effect of H2O2 on SK-N-MC cells, as shown by the increase in cell viability and by reduced LDH release. Maysin pretreatment also dose-dependently reduced the intracellular ROS level and inhibited PARP cleavage. In addition, DNA damage and H2O2-induced apoptotic cell death were significantly attenuated by maysin pretreatment. Moreover, maysin pretreatment (5-50 μg/ml) for 2h significantly and dose-dependently increased the mRNA levels of antioxidant enzymes (CAT, GPx-1, SOD-1, SOD-2 and HO-1) in H2O2 (200 μM)-insulted cells. SIGNIFICANCE These results suggest that CS maysin has neuroprotective effects against oxidative stress (H2O2)-induced apoptotic death of human brain SK-N-MC cells through its antioxidative action. This report is the first regarding neuroprotective health benefits of corn silk maysin by its anti-apoptotic action and by triggering the expression of intracellular antioxidant enzyme systems in SK-N-MC cells.
Collapse
Affiliation(s)
- Doo Jin Choi
- Department of Biotechnology, The CUK Agromedical Research Center, The Catholic University of Korea, Bucheon, Gyeonggi-do 420-743, Republic of Korea
| | - Sun-Lim Kim
- National Institute of Crop Science, RDA, Suwon, Gyeonggi-do 441-857, Republic of Korea
| | - Ji Won Choi
- Department of Biotechnology, The CUK Agromedical Research Center, The Catholic University of Korea, Bucheon, Gyeonggi-do 420-743, Republic of Korea
| | - Yong Il Park
- Department of Biotechnology, The CUK Agromedical Research Center, The Catholic University of Korea, Bucheon, Gyeonggi-do 420-743, Republic of Korea.
| |
Collapse
|
24
|
Abstract
Traumatic brain injury (TBI) is the most important cause of disability in individuals under the age of 45 years and thus represents a significant social and economic burden. Evidence strongly suggests that oxidative stress is a cornerstone event leading to and propagating secondary injury mechanisms such as excitotoxicity, mitochondrial dysfunction, apoptosis, autophagy, brain edema, and inflammation. TBI has defied conventional approaches to diagnosis and therapy development because of its heterogeneity and complexity. Therefore, it is necessary to explore alternative approaches to therapy development for TBI. The aim of this review is to present a therapeutic approach for TBI, taking into account the evidence supporting the role for oxidative stress in the pathophysiological processes of secondary brain injury. The role of agents such as mitochondria-targeted antioxidants (melatonin and new mitochondria-targeted antioxidants), nicotinamide adenine dinucleotide phosphate (NADPH) inhibitors (antioxidant vitamins and apocynin), and other compounds having mainly antioxidant properties (hydrogen-rich saline, sulforaphane, U-83836E, omega-3, and polyphenols) is covered. The rationale for innovative antioxidant therapies based on current knowledge and particularly the most recent studies regarding this field is discussed. Particular considerations and translational potential of new TBI treatments are examined and a novel therapeutic proposal for TBI is presented.
Collapse
|
25
|
Moretti M, Budni J, Freitas AE, Rosa PB, Rodrigues ALS. Antidepressant-like effect of ascorbic acid is associated with the modulation of mammalian target of rapamycin pathway. J Psychiatr Res 2014; 48:16-24. [PMID: 24209999 DOI: 10.1016/j.jpsychires.2013.10.014] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 09/17/2013] [Accepted: 10/17/2013] [Indexed: 01/02/2023]
Abstract
The present study investigated the involvement of the PI3K, GSK-3β, heme oxygenase-1 (HO-1) and mTOR in the antidepressant-like effect of ascorbic acid in the tail suspension test (TST). Male Swiss mice were pretreated with ascorbic acid (1 mg/kg, p.o.) or vehicle and 45 min after, LY294002 (10 μg/site, i.c.v., reversible PI3K inhibitor), rapamycin (0.2 nmol/site, i.c.v., selective mTOR inhibitor), zinc protoporphyrin (ZnPP - 10 ng/site, i.c.v., HO-1 inhibitor) or vehicle was administered. We also investigated the synergistic effect of ascorbic acid (0.1 mg/kg, p.o., sub-effective dose in the TST) with lithium chloride (10 mg/kg, p.o., non-selective GSK-3β inhibitor), AR-A014418 (0.01 μg/site, i.c.v., selective GSK-3β inhibitor) or cobalt protoporphyrin (CoPP - 0.01 μg/site, i.c.v., HO-1 inducer) in the TST. The antidepressant-like effect of ascorbic acid (1 mg/kg, p.o.) was prevented by the treatment of mice with LY294002, rapamycin or ZnPP. In addition, sub-effective doses of lithium chloride, AR-A014418 or CoPP, combined with a sub-effective dose of ascorbic acid produced a synergistic antidepressant-like effect. We also demonstrated that 1 h after its administration, ascorbic acid increased the phosphorylation of p70S6K and the immunocontent of PSD-95 in the hippocampus of mice. These results indicate that the antidepressant-like effect of ascorbic acid in the TST might be dependent on the activation of PI3K and mTOR, inhibition of GSK-3β as well as induction of HO-1, reinforcing the notion that these are important targets for antidepressant activity and contributing to better elucidate the mechanisms underlying the antidepressant-like effect of ascorbic acid.
Collapse
Affiliation(s)
- Morgana Moretti
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, 88040-900 SC, Brazil.
| | | | | | | | | |
Collapse
|
26
|
Li Y, Gao C, Shi Y, Tang Y, Liu L, Xiong T, Du M, Xing M, Liu L, Yao P. Carbon monoxide alleviates ethanol-induced oxidative damage and inflammatory stress through activating p38 MAPK pathway. Toxicol Appl Pharmacol 2013; 273:53-8. [PMID: 23994557 DOI: 10.1016/j.taap.2013.08.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Revised: 08/06/2013] [Accepted: 08/13/2013] [Indexed: 12/15/2022]
Abstract
Stress-inducible protein heme oxygenase-1(HO-1) is well-appreciative to counteract oxidative damage and inflammatory stress involving the pathogenesis of alcoholic liver diseases (ALD). The potential role and signaling pathways of HO-1 metabolite carbon monoxide (CO), however, still remained unclear. To explore the precise mechanisms, ethanol-dosed adult male Balb/c mice (5.0g/kg.bw.) or ethanol-incubated primary rat hepatocytes (100mmol/L) were pretreated by tricarbonyldichlororuthenium (II) dimmer (CORM-2, 8mg/kg for mice or 20μmol/L for hepatocytes), as well as other pharmacological reagents. Our data showed that CO released from HO-1 induction by quercetin prevented ethanol-derived oxidative injury, which was abolished by CO scavenger hemoglobin. The protection was mimicked by CORM-2 with the attenuation of GSH depletion, SOD inactivation, MDA overproduction, and the leakage of AST, ALT or LDH in serum and culture medium induced by ethanol. Moreover, CORM-2 injection or incubation stimulated p38 phosphorylation and suppressed abnormal Tnfa and IL-6, accompanying the alleviation of redox imbalance induced by ethanol and aggravated by inflammatory factors. The protective role of CORM-2 was abolished by SB203580 (p38 inhibitor) but not by PD98059 (ERK inhibitor) or SP600125 (JNK inhibitor). Thus, HO-1 released CO prevented ethanol-elicited hepatic oxidative damage and inflammatory stress through activating p38 MAPK pathway, suggesting a potential therapeutic role of gaseous signal molecule on ALD induced by naturally occurring phytochemicals.
Collapse
Affiliation(s)
- Yanyan Li
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, 13 Hangkong Road, Wuhan 430030, PR China; Ministry of Education Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, 13 Hangkong Road, Wuhan 430030, PR China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, 13 Hangkong Road, Wuhan 430030, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Benvenuti S, Deledda C, Luciani P, Modi G, Bossio A, Giuliani C, Fibbi B, Peri A. Low Extracellular Sodium Causes Neuronal Distress Independently of Reduced Osmolality in an Experimental Model of Chronic Hyponatremia. Neuromolecular Med 2013; 15:493-503. [DOI: 10.1007/s12017-013-8235-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 05/09/2013] [Indexed: 10/26/2022]
|
28
|
Hu JL, Xiao L, Li ZY, Wang Q, Chang Y, Jin Y. Upregulation of HO-1 is accompanied by activation of p38MAPK and mTOR in human oesophageal squamous carcinoma cells. Cell Biol Int 2013; 37:584-92. [PMID: 23412940 DOI: 10.1002/cbin.10075] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 02/06/2013] [Indexed: 12/31/2022]
Affiliation(s)
- Jian-Li Hu
- Cancer Centre, Union Hospital, Huazhong University of Science and Technology; 1277 Jiefangdadao Jianghan District, Wuhan; Hubei; 430022; PR China
| | - Lan Xiao
- Department of Obstetrics and Gynecology; First Affiliated Hospital, An Hui Medical College; 218 Jixi Road, Hefei; AnHui; 230022; PR China
| | - Zhen-Yun Li
- Cancer Centre, Union Hospital, Huazhong University of Science and Technology; 1277 Jiefangdadao Jianghan District, Wuhan; Hubei; 430022; PR China
| | - Qiong Wang
- Cancer Centre, Union Hospital, Huazhong University of Science and Technology; 1277 Jiefangdadao Jianghan District, Wuhan; Hubei; 430022; PR China
| | - Yu Chang
- Cancer Centre, Union Hospital, Huazhong University of Science and Technology; 1277 Jiefangdadao Jianghan District, Wuhan; Hubei; 430022; PR China
| | - Yi Jin
- Laboratory Department; Union Hospital, Huazhong University of Science and Technology; 1277 Jiefangdadao Jianghan District, Wuhan; Hubei; 430022; PR China
| |
Collapse
|
29
|
Jie Q, Tang Y, Deng Y, Li Y, Shi Y, Gao C, Xing M, Wang D, Liu L, Yao P. Bilirubin participates in protecting of heme oxygenase-1 induction by quercetin against ethanol hepatotoxicity in cultured rat hepatocytes. Alcohol 2013; 47:141-8. [PMID: 23265624 DOI: 10.1016/j.alcohol.2012.10.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 10/12/2012] [Accepted: 10/19/2012] [Indexed: 12/17/2022]
Abstract
To attenuate alcohol liver disease (ALD) is extremely urgent since ALD has been emerged as a major liver disease. The aim of the present study is to investigate the hepatoprotective effect against ethanol-induced injury of bilirubin, a product of heme metabolism degradation via HO and biliverdin reductase catalysis. Ethanol-incubated primary rat hepatocytes (100 mmol/L) were treated by quercetin, bilirubin, inflammatory factors, and/or HO-1 inducer/inhibitor for 24 h, and the cellular damage was assayed. Quercetin lowered ethanol-induced glutathione depletion and superoxide dismutase inactivation, inhibited the overproduction of malondialdehyde and reactive oxygen species, and decreased the leakage of cellular aspartate aminotransferase and lactate dehydrogenase, accompanying the normalization of bilirubin level. The effect of quercetin was mimicked by exogenous bilirubin in a dose-dependent manner to some extent (within 25 μmol/L) and pharmacological HO-1 inducer hemin, but abolished by HO-1 inhibitor zinc protoporphyrin-IX. Inflammatory challenge of TNF-α plus IL-6 further aggravated ethanol-induced oxidative damage, which was also attenuated by bilirubin in part. These findings shed a light on the anti-oxidative and anti-inflammatory role of bilirubin released from quercetin/HO-1 and biliverdin reductase pathway against ethanol hepatotoxicity and highlight a prospective strategy of nutritional intervention for ALD by naturally occurring quercetin to induce HO-1 with the release of bioactive end-products.
Collapse
|
30
|
Akt/Nrf2 Activated Upregulation of Heme Oxygenase-1 Involves in the Role of Rg1 Against Ferrous Iron-Induced Neurotoxicity in SK-N-SH Cells. Neurotox Res 2012. [DOI: 10.1007/s12640-012-9362-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
31
|
Huang YN, Wang JY, Lee CT, Lin CH, Lai CC, Wang JY. L-ascorbate attenuates methamphetamine neurotoxicity through enhancing the induction of endogenous heme oxygenase-1. Toxicol Appl Pharmacol 2012; 265:241-52. [PMID: 23022510 DOI: 10.1016/j.taap.2012.08.036] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2012] [Revised: 08/13/2012] [Accepted: 08/27/2012] [Indexed: 12/20/2022]
Abstract
Methamphetamine (METH) is a drug of abuse which causes neurotoxicity and increased risk of developing neurodegenerative diseases. We previously found that METH induces heme oxygenase (HO)-1 expression in neurons and glial cells, and this offers partial protection against METH toxicity. In this study, we investigated the effects of l-ascorbate (vitamin C, Vit. C) on METH toxicity and HO-1 expression in neuronal/glial cocultures. Cell viability and damage were evaluated by 3-(4,5-dimethylthianol-2-yl)-2,5 diphenyl tetrazolium bromide (MTT) reduction and lactate dehydrogenase (LDH) release, respectively. Neuronal and glial localization of HO-1 were identified by double immunofluorescence staining. Reactive oxygen species (ROS) production was measured using the fluorochrome 2',7'-dichlorofluorescin diacetate. HO-1 mRNA and protein expression were examined by RT-qPCR and Western blotting, respectively. Results show that Vit. C induced HO-1 mRNA and protein expressions in time- and concentration-dependent manners. Inhibition of p38 mitogen-activated protein kinase (MAPK) but not extracellular signal-regulated kinase (ERK) significantly blocked induction of HO-1 by Vit. C. HO-1 mRNA and protein expressions were significantly elevated by a combination of Vit. C and METH, compared to either Vit. C or METH alone. Pretreatment with Vit. C enhanced METH-induced HO-1 expression and attenuated METH-induced ROS production and neurotoxicity. Pharmacological inhibition of HO activity abolished suppressive effects of Vit. C on METH-induced ROS production and attenuated neurotoxicity. We conclude that induction of HO-1 expression contributes to the attenuation of METH-induced ROS production and neurotoxicity by Vit. C. We suggest that HO-1 induction by Vit. C may serve as a strategy to alleviate METH neurotoxicity.
Collapse
Affiliation(s)
- Ya-Ni Huang
- Department of Nursing, Hsin Sheng College of Medical Care and Management, Taoyuan, Taiwan
| | | | | | | | | | | |
Collapse
|
32
|
Dal-Cim T, Molz S, Egea J, Parada E, Romero A, Budni J, Martín de Saavedra MD, Barrio LD, Tasca CI, López MG. Guanosine protects human neuroblastoma SH-SY5Y cells against mitochondrial oxidative stress by inducing heme oxigenase-1 via PI3K/Akt/GSK-3β pathway. Neurochem Int 2012; 61:397-404. [DOI: 10.1016/j.neuint.2012.05.021] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 05/17/2012] [Accepted: 05/23/2012] [Indexed: 12/20/2022]
|
33
|
Yao C, Wei G, Lu XCM, Yang W, Tortella FC, Dave JR. Selective brain cooling in rats ameliorates intracerebral hemorrhage and edema caused by penetrating brain injury: possible involvement of heme oxygenase-1 expression. J Neurotrauma 2012; 28:1237-45. [PMID: 21463155 DOI: 10.1089/neu.2010.1678] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Brain edema formation associated with trauma-induced intracerebral hemorrhage (ICH) is a clinical complication with high mortality. Studies have shown that heme oxygenase-1 (HO-1) plays an important role in ICH-induced brain edema. In order to understand the role of HO-1 in the protective effect of selective brain cooling (SBC), we investigated the time course of HO-1 changes following penetrating ballistic-like brain injury (PBBI) in rats. Samples were collected from injured and control animals at 6, 24, 48, and 72 h, and 7 days post-injury to evaluate HO-1 expression, heme concentration, brain water content, and immunohistochemistry (IHC). Following a 10% frontal PBBI, HO-1 mRNA and protein was increased at all time points studied, reaching maximum expression levels at 24-48 h post-injury. An increase in the heme concentration and the development of brain edema coincided with the upregulation of HO-1 mRNA and protein during the 7-day post-injury period. SBC significantly decreased PBBI-induced heme concentration, attenuated HO-1 upregulation, and concomitantly reduced brain water content. These results suggest that the neuroprotective effects of SBC may be partially mediated by reducing the heme accumulation, which reduced injury-mediated upregulation of HO-1, and in turn ameliorated edema formation. Collectively, these results suggest a potential value of HO-1 as a diagnostic and/or therapeutic biomarker in hemorrhagic brain injury.
Collapse
Affiliation(s)
- Changping Yao
- Department of Applied Neurobiology, Division of Psychiatry and Neuroscience, Walter Reed Army Institute of Research, 503 Robert Grant AvenueSilver Spring, MD 20910, USA
| | | | | | | | | | | |
Collapse
|
34
|
Gerjevic LN, Lu S, Chaky JP, Harrison-Findik DD. Regulation of heme oxygenase expression by alcohol, hypoxia and oxidative stress. World J Biol Chem 2011; 2:252-60. [PMID: 22216371 PMCID: PMC3247681 DOI: 10.4331/wjbc.v2.i12.252] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Revised: 10/11/2011] [Accepted: 10/17/2011] [Indexed: 02/05/2023] Open
Abstract
AIM: To study the effect of both acute and chronic alcohol exposure on heme oxygenases (HOs) in the brain, liver and duodenum.
METHODS: Wild-type C57BL/6 mice, heterozygous Sod2 knockout mice, which exhibit attenuated manganese superoxide dismutase activity, and liver-specific ARNT knockout mice were used to investigate the role of alcohol-induced oxidative stress and hypoxia. For acute alcohol exposure, ethanol was administered in the drinking water for 1 wk. Mice were pair-fed with regular or ethanol-containing Lieber De Carli liquid diets for 4 wk for chronic alcohol studies. HO expression was analyzed by real-time quantitative polymerase chain reaction and Western blotting.
RESULTS: Chronic alcohol exposure downregulated HO-1 expression in the brain but upregulated it in the duodenum of wild-type mice. It did not alter liver HO-1 expression, nor HO-2 expression in the brain, liver or duodenum. In contrast, acute alcohol exposure decreased both liver HO-1 and HO-2 expression, and HO-2 expression in the duodenum of wild-type mice. The decrease in liver HO-1 expression was abolished in ARNT+/- mice. Sod2+/- mice with acute alcohol exposure did not exhibit any changes in liver HO-1 and HO-2 expression or in brain HO-2 expression. However, alcohol inhibited brain HO-1 and duodenal HO-2 but increased duodenal HO-1 expression in Sod2+/- mice. Collectively, these findings indicate that acute and chronic alcohol exposure regulates HO expression in a tissue-specific manner. Chronic alcohol exposure alters brain and duodenal, but not liver HO expression. However, acute alcohol exposure inhibits liver HO-1 and HO-2, and also duodenal HO-2 expression.
CONCLUSION: The inhibition of liver HO expression by acute alcohol-induced hypoxia may play a role in the early phases of alcoholic liver disease progression.
Collapse
Affiliation(s)
- Lisa Nicole Gerjevic
- Lisa Nicole Gerjevic, Sizhao Lu, Jonathan Pascal Chaky, Duygu Dee Harrison-Findik, Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5820, United States
| | | | | | | |
Collapse
|
35
|
Guan L, Zhang YL, Wen T, Wang XF, Zhu MX, Zhao JY. Dynamic changes of heme oxygenase-1 in the hippocampus of rats after acute carbon monoxide poisoning. ARCHIVES OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2011; 60:165-172. [PMID: 20422170 DOI: 10.1007/s00244-010-9524-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2009] [Accepted: 04/05/2010] [Indexed: 05/29/2023]
Abstract
Heme oxygenase-1 (HO-1), an inducible enzyme, degrades heme to carbon monoxide (CO), iron, and bilirubin. We have investigated the relationship among HO-1 protein expression, HO activity, and CO concentrations in the hippocampus of CO-exposed rats. Western blotting and immunohistochemistry revealed that the enzyme is predominantly localized in hippocampal CA1 and CA3 pyramidal cells and in granule cells of the dentate gyrus. HO enzyme activity was reduced immediately following CO exposure, while expression of HO-1 protein was consistently upregulated in a time-dependent manner. Local CO concentrations in hippocampus rose immediately following exposure, but the elevation was maintained for ~20 h despite the decline in blood carboxyhemoglobin levels toward baseline. We suggest that CO initially inhibits HO enzyme activity, whereas at later time points the inhibition is released and local CO generation is maintained by the activity of the endogenous HO enzyme. And the noninducible form of heme oxygenase, HO-2, was not altered following CO administration. Together these results indicate that the HO/CO pathway in the rat hippocampus is induced by acute CO exposure; local CO production may play a regulatory role in brain injury following CO poisoning.
Collapse
Affiliation(s)
- Li Guan
- Research Center of Occupational Medicine, Peking University Third Hospital, Beijing, People's Republic of China
| | | | | | | | | | | |
Collapse
|
36
|
Hong Y, Yan W, Chen S, Sun CR, Zhang JM. The role of Nrf2 signaling in the regulation of antioxidants and detoxifying enzymes after traumatic brain injury in rats and mice. Acta Pharmacol Sin 2010; 31:1421-30. [PMID: 20953205 DOI: 10.1038/aps.2010.101] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
AIM To determine whether Nrf2 signaling pathway activation could attenuate oxidative stress and neuronal damage following traumatic brain injury (TBI). METHODS Controlled cortical impact (CCI) injury was performed in Sprague-Dawley rats and Nrf2-knockout or control mice. Sulforaphane (SFN), a potent Nrf2 activator, was used to activate Nrf2. Oxidative stress, lesion volume, neuron degeneration, and neurologic dysfunction were determined using biochemical, histopathological and neuroethologic approaches. Protein and mRNA levels of Nrf2 and the antioxidant enzymes heme oxygenase 1 (HO-1) and NAD(P)H:quinine oxidoreductase 1 (NQO1) were assessed using Western blot analysis and RT-PCR. RESULTS Activation of Nrf2 by SFN( 5 mg/kg, ip) induced the nuclear translocation and activation of Nrf2, which resulted in an up-regulation of Nrf2-dependent antioxidant enzymes and a reduction of oxidative damage after TBI. In accordance with these biochemical changes, SFN also significantly reduced neuronal death, contusion volume, and neurological dysfunction after TBI. Furthermore, Nrf2-knockout mice showed more severe oxidative stress and neurologic deficits after TBI and did not benefit from the effects of SFN. CONCLUSION Nrf2 plays a pivotal role in cell defenses against the oxidative stress of TBI. In addition, pharmacological activation of the Nrf2 signaling pathway by small molecule inducers such as SFN attenuated oxidative stress and neuronal damage following TBI.
Collapse
|
37
|
Synthetic Triterpenoid CDDO Derivatives Modulate Cytoprotective or Immunological Properties in Astrocytes, Neurons, and Microglia. J Neuroimmune Pharmacol 2010; 6:107-20. [DOI: 10.1007/s11481-010-9240-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Accepted: 08/11/2010] [Indexed: 12/28/2022]
|
38
|
Menk M, von Haefen C, Funke-Kaiser H, Sifringer M, Schefe JH, Kirsch S, Seidel K, Reinemund J, Steckelings UM, Unger T, Spies CD. Ethanol-induced downregulation of the angiotensin AT2 receptor in murine fibroblasts is mediated by PARP-1. Alcohol 2010; 44:495-506. [PMID: 20693103 DOI: 10.1016/j.alcohol.2010.05.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2009] [Revised: 05/04/2010] [Accepted: 05/12/2010] [Indexed: 01/13/2023]
Abstract
Molecular mechanisms accompanying ethanol-induced cytotoxicity remain to be defined. The renin-angiotensin system with its respective receptors, the angiotensin AT1 and AT2 receptor (AT1R and AT2R), has been implicated in these processes. The AT2R seems to counteract the pro-inflammatory, pro-hypertrophic, and pro-fibrotic actions of the AT1R and is involved in cellular differentiation and tissue repair. Recently, we identified poly(ADP-ribose) polymerase-1 (PARP-1) as a novel negative transcriptional regulator of the AT2R. However, the complex interactions between ethanol, PARP-1, and the AT2R are largely unknown. In this in vitro study, we aimed to clarify whether acute ethanol treatment modifies AT2R promoter activity or AT2R mRNA and protein levels and whether PARP-1 is involved in ethanol-mediated regulation of the AT2R. Murine fibroblasts of the R3T3 and MEF line (murine embryonic fibroblasts) were exposed to ethanol for 24h. AT2R promoter activity, mRNA and protein levels were analyzed with and without PARP-1 inhibition and in PARP-1 knockout MEF cells. Expression of PARP-1 was analyzed over course of time, and cell viability and DNA fragmentation were measured on single-cell level by flow cytometry. Ethanol exposition induced substantial downregulation of the AT2R on promoter, mRNA and protein levels in a dose-dependent manner. Pharmacological inhibition or ablation of PARP-1 completely abolished this effect. Ethanol treatment did not have any effect on AT1R mRNA and protein levels in MEF cells. Further, acute ethanol treatment promoted DNA fragmentation and caused transcriptional induction of PARP-1. Our findings reveal that PARP-1 is an upstream transcriptional regulator of the AT2 receptor in the context of ethanol exposure and represses the AT2R gene in fibroblasts in vitro. Variations in expression of the potentially tissue-protective AT2R might contribute to ethanol-mediated pathology.
Collapse
Affiliation(s)
- Mario Menk
- Department of Anesthesiology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum/Campus Charité Mitte, 13353 Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Leasure JL, Nixon K. Exercise neuroprotection in a rat model of binge alcohol consumption. Alcohol Clin Exp Res 2009; 34:404-14. [PMID: 20028365 DOI: 10.1111/j.1530-0277.2009.01105.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Excessive alcohol intake produces structural and functional deficits in corticolimbic pathways that are thought to underlie cognitive deficits in the alcohol use disorders (AUDs). Animal models of binge alcohol administration support the direct link of high levels of alcohol consumption and neurotoxicity in the hippocampus and surrounding cortex. In contrast, voluntary wheel running enhances hippocampal neurogenesis and generally promotes the health of neurons. METHODS We investigated whether voluntary exercise prior to binge alcohol exposure could protect against alcohol-induced cell loss. Female Long-Evans rats exercised voluntarily for 14 days before undergoing 4 days of binge alcohol consumption. Brains were harvested immediately after the last dose of alcohol and examined for various histological markers of neurodegeneration, including both cell death (FluoroJade B) and cell birth (Ki67) markers. RESULTS Rats that exercised prior to binge exposure were significantly less behaviorally intoxicated, which was not a result of enhanced hepatic metabolism. Rats that exercised prior to binge alcohol consumption had reduced loss of dentate gyrus granule cells and fewer FluoroJade B positive cells in the dentate gyrus and associated entorhinal-perirhinal cortex compared to nonexercisers. However, exercise did not protect against cell death in the piriform cortex nor protect against alcohol-induced decreases in cell proliferation, evidenced by a similar alcohol-induced reduction in Ki67 labeled cells between exercise and sedentary rats. CONCLUSIONS We conclude that exercise can reduce behavioral sensitivity to ethanol intoxication and protect vulnerable brain areas from alcohol-induced cell death. Exercise neuroprotection of alcohol-induced brain damage has important implications in understanding the neurobiology of the AUDs as well as in developing novel treatment strategies.
Collapse
Affiliation(s)
- J Leigh Leasure
- Department of Psychology, University of Houston, Houston, Texas, USA
| | | |
Collapse
|
40
|
Yao P, Hao L, Nussler N, Lehmann A, Song F, Zhao J, Neuhaus P, Liu L, Nussler A. The protective role of HO-1 and its generated products (CO, bilirubin, and Fe) in ethanol-induced human hepatocyte damage. Am J Physiol Gastrointest Liver Physiol 2009; 296:G1318-23. [PMID: 19325051 DOI: 10.1152/ajpgi.00555.2007] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
It has been reported that naturally occurring quercetin exerts hepatoprotective effects through heme oxygenase-1 (HO-1) induction. However, the precise mechanism of how ethanol-associated liver damage is counteracted by quercetin-enhanced HO-1 metabolism still remains unclear. To further decipher the protective role of quercetin on ethanol-induced liver damage, we treated human hepatocytes with quercetin and various (end) products of the HO-1 pathway. Our data clearly showed that quercetin treatment attenuated ethanol-induced damage, whereas hemoglobin and zinc protoporphyrin 9 (ZnPP) abolished such effects. Iron-II aggravated ethanol toxicity and was only partially reduced by quercetin. In contrast, carbon monoxide (CO) dose dependently inhibited ethanol-induced cytochrome P450 2E1 (CYP 2E1) activity and hepatotoxicity but had no influence on CYP 2E1 protein expression. Similarly, hemoglobin dramatically stimulated CYP 2E1 activity but not the protein expression in quercetin- and ethanol-cotreated hepatocytes. ZnPP significantly promoted CYP 2E1 protein expression in the presence and absence of CO treatment but inhibited ethanol-induced CYP 2E1 activation following CO incubation in quercetin- and ethanol-cotreated hepatocytes. These results suggested that quercetin virtually attenuated ethanol-derived oxidative damage via HO-1 induction. Heme degradation and CO release may mediate the protective effects through inhibiting ethanol-induced CYP 2E1 synthesis and enzymatic activity, respectively.
Collapse
Affiliation(s)
- Ping Yao
- Universitätsmedizin Berlin, Charité, Campus Virchow, Department of General, Visceral, and Transplantation Surgery, Berlin, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
González-Reyes S, Orozco-Ibarra M, Guzmán-Beltrán S, Molina-Jijón E, Massieu L, Pedraza-Chaverri J. Neuroprotective role of heme-oxygenase 1 against iodoacetate-induced toxicity in rat cerebellar granule neurons: Role of bilirubin. Free Radic Res 2009; 43:214-23. [PMID: 19177228 DOI: 10.1080/10715760802676670] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Heme oxygenase (HO) catalyses the breakdown of heme to iron, carbon monoxide and biliverdin, the latter being further reduced to bilirubin. A protective role of the inducible isoform, HO-1, has been described in pathological conditions associated with the production of reactive oxygen species (ROS). The aim of this study was to investigate the role of HO-1 in the neurotoxicity induced by iodoacetate (IAA) in primary cultures of cerebellar granule neurons (CGNs). IAA, an inhibitor of the glycolysis pathway, reduces cell survival, increases ROS production and enhances HO-1 expression in CGNs. Furthermore, the induction of HO-1 expression by cobalt protoporphyrin (CoPP) prevented cell death and ROS production induced by IAA, whereas the inhibition of HO activity with tin mesoporphyrin exacerbated the IAA-induced neurotoxicity. The protective effect elicited by CoPP was reproduced by bilirubin addition, suggesting that this molecule may be involved in the protective effect of HO-1 induction in this experimental model.
Collapse
|
42
|
Orozco-Ibarra M, Estrada-Sánchez AM, Massieu L, Pedraza-Chaverrí J. Heme oxygenase-1 induction prevents neuronal damage triggered during mitochondrial inhibition: role of CO and bilirubin. Int J Biochem Cell Biol 2008; 41:1304-14. [PMID: 19063990 DOI: 10.1016/j.biocel.2008.11.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2008] [Revised: 11/10/2008] [Accepted: 11/11/2008] [Indexed: 12/11/2022]
Abstract
Heme oxygenase (HO) catalyzes the breakdown of heme to iron, carbon monoxide (CO), and biliverdin, the latter being further reduced to bilirubin (BR). A protective role of the inducible isoform, HO-1, has been described in pathological conditions associated with reactive oxygen species (ROS) and oxidative damage. The aim of this study was to investigate the role of HO-1 in the neurotoxicity induced by the mitochondrial toxin 3-nitropropionic acid (3-NP) in primary cultures of cerebellar granule neurons (CGNs). Toxicity of 3-NP is associated with ROS production, and this metabolic toxin has been used to mimic pathological conditions such as Huntington's disease. We found that cell death caused by 3-NP exposure was exacerbated by inhibition of HO with tin mesoporphyrin (SnMP). In addition, HO-1 up-regulation induced by the exposure to cobalt protoporphyrin (CoPP) before the incubation with 3-NP, prevented the cell death and the increase in ROS induced by 3-NP. Interestingly, addition of SnMP to CoPP-pretreated CGNs exposed to 3-NP, abolished the protective effect of CoPP suggesting that HO activity was responsible for this protective effect. This was additionally supported by the fact that CORM-2, a CO-releasing molecule, and BR, were able to protect against cell death and the increase in ROS induced by 3-NP. Our data clearly show that HO-1 elicits in CGNs a neuroprotective action against the neurotoxicity of 3-NP and that CO and BR may be involved, at least in part, in this protective effect. The present results increase our knowledge about the role of HO-1 in neuropathological conditions.
Collapse
Affiliation(s)
- Marisol Orozco-Ibarra
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510 México DF, Mexico
| | | | | | | |
Collapse
|
43
|
Yan W, Wang HD, Hu ZG, Wang QF, Yin HX. Activation of Nrf2-ARE pathway in brain after traumatic brain injury. Neurosci Lett 2007; 431:150-4. [PMID: 18162315 DOI: 10.1016/j.neulet.2007.11.060] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2007] [Revised: 11/14/2007] [Accepted: 11/21/2007] [Indexed: 10/22/2022]
Abstract
Secondary brain injury plays a pivotal role in the outcome of patients suffering from traumatic brain injury (TBI). The mechanisms underlying secondary brain injury are complex and interrelated. Previous studies focused on one of these mechanisms have been proved to be ineffective in clinical practice. Therefore, a target, which can interrupt multi-mechanisms underlying TBI, is desirable. Nrf2-ARE pathway has been proved to be the key regulator in reducing oxidative stress, inflammatory damage and accumulation of toxic metabolites, which are all involved in TBI. However, whether Nrf2-ARE pathway is activated after TBI has not been studied. In the present study, the nuclear Nrf2 protein level was detected by Western blot, and the mRNA levels of heme oxygenase-1 (HO-1) and NAD(P)H: quinone oxidoreductase-1 (NQO1), two Nrf2-regulated gene products, were determined using reverse-transcriptase polymerase chain reaction (RT-PCR) 24h after TBI. Furthermore, we also localized the expression of Nrf2 and HO-1 using immunohistochemical study. After TBI, the nuclear Nrf2 protein level was significantly increased, and the mRNA levels of both HO-1 and NQO1 were also up regulated. Moreover, both Nrf2 and HO-1 were localized in the same types of cells. According to these results, it could be postulated that Nrf2-ARE pathway was activated in brain after TBI.
Collapse
Affiliation(s)
- Wei Yan
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, 305 East Zhongshan Road, Nanjing 210002, Jiangsu Province, PR China
| | | | | | | | | |
Collapse
|
44
|
Ku BM, Lee YK, Jeong JY, Mun J, Han JY, Roh GS, Kim HJ, Cho GJ, Choi WS, Yi GS, Kang SS. Ethanol-induced oxidative stress is mediated by p38 MAPK pathway in mouse hippocampal cells. Neurosci Lett 2007; 419:64-7. [PMID: 17420100 DOI: 10.1016/j.neulet.2007.03.049] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2007] [Revised: 03/23/2007] [Accepted: 03/24/2007] [Indexed: 10/23/2022]
Abstract
It has been known that ethanol causes neuronal cell death through oxidative stress. Ethanol itself and reactive oxygen species (ROS) produced by ethanol modulate intracellular signaling pathways including mitogen-activated protein kinase (MAPK) cascades. This study was conducted to examine the impact of ethanol on MAPK signaling in HT22 cells. Ethanol (100 and 400mM) caused activation of ERK, p38 MAPK, and JNK. ERK activation occurred in early time and p38 MAPK activation was evident when ERK activation was diminished. Specific inhibitor of p38 MAPK (SB203580) protected HT22 cells against ethanol, which was accompanied by an inhibition of ROS accumulation. However, inhibitors of ERK (U0126) and JNK (SP600125) had no effects on ethanol-induced neuronal cell death when they are treated with ethanol for 24h. These results suggest that p38 MAPK may have important roles in ROS accumulation during ethanol-induced oxidative stress in HT22 cells.
Collapse
Affiliation(s)
- Bo Mi Ku
- Department of Anatomy and Neurobiology, Institute of Health Sciences, School of Medicine, Gyeongsang National University, 92 Chilam-dong, Jinju, Gyeongnam 660-751, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Pascal T, Debacq-Chainiaux F, Boilan E, Ninane N, Raes M, Toussaint O. Heme oxygenase-1 and interleukin-11 are overexpressed in stress-induced premature senescence of human WI-38 fibroblasts induced by tert-butylhydroperoxide and ethanol. Biogerontology 2007; 8:409-22. [PMID: 17295091 DOI: 10.1007/s10522-007-9084-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2007] [Accepted: 01/12/2007] [Indexed: 01/25/2023]
Abstract
Acute repeated exposures to subcytotoxic concentrations of tert-butylhydroperoxide and ethanol trigger premature senescence of human diploid fibroblasts. In the present work we found an increased mRNA and protein level of interleukin-11 and heme oxygenase-1 in premature senescence of WI-38 human diploid foetal lung fibroblasts induced by both tert-butylhydroperoxide and ethanol. We tested whether interleukin-11 and heme oxygenase-1 could protect against tert-butylhydroperoxide- or ethanol-induced premature senescence when stable overexpression was established using a retroviral vector-based transduction. No protective effect was found against the decrease of the proliferative potential, the increase of the proportion of senescence-associated ss-galactosidase positive cells and the increase of the mRNA levels of six senescence-associated genes.
Collapse
Affiliation(s)
- Thierry Pascal
- Unit of Research on Cellular Biology (URBC), Department of Biology, University of Namur (FUNDP), Rue de Bruxelles, 5000, Namur, Belgium
| | | | | | | | | | | |
Collapse
|