1
|
Scibetta S, Miceli M, Iuliano M, Stefanuto L, Carbone E, Piscopo P, Petrozza V, Romeo G, Mangino G, Calogero A, Gasperi T, Rosa P. In Vitro Evaluation of the Antioxidant Capacity of 3,3-Disubstituted-3H-benzofuran-2-one Derivatives in a Cellular Model of Neurodegeneration. Life (Basel) 2024; 14:422. [PMID: 38672695 PMCID: PMC11051253 DOI: 10.3390/life14040422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024] Open
Abstract
Oxidative stress represents a hallmark for many degenerative pathologies of the Central Nervous System. Throughout life, the constant pressure of noxious stimuli and/or episodes of traumatic events may expose the brain to a microenvironment where the non-balanced reactive oxygen species inevitably lead to neuronal loss and cognitive decline. HO-1, a 32 kDa heat-shock protein catalyzing the degradation of heme into carbon monoxide (CO), iron and biliverdin/bilirubin is considered one of the main antioxidant defense mechanisms playing pivotal roles in neuroprotection. Restoring the redox homeostasis is the goal of many natural or synthetic antioxidant molecules pursuing beneficial effects on brain functions. Here, we investigated the antioxidant capacity of four selected benzofuran-2-one derivatives in a cellular model of neurodegeneration represented by differentiated SH-SY5Y cells exposed to catechol-induced oxidative stress. Our main results highlight how all the molecules have antioxidant properties, especially compound 9, showing great abilities in reducing intracellular ROS levels and protecting differentiated SH-SY5Y cells from catechol-induced death. This compound above all seems to boost HO-1 mRNA and perinuclear HO-1 protein isoform expression when cells are exposed to the oxidative insult. Our findings open the way to consider benzofuran-2-ones as a novel and promising adjuvant antioxidant strategy for many neurodegenerative disorders.
Collapse
Affiliation(s)
- Sofia Scibetta
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome Sapienza, Polo Pontino, 04100 Latina, Italy; (S.S.); (M.I.); (V.P.); (G.R.); (G.M.); (A.C.)
| | - Martina Miceli
- Department of Science, University of Roma Tre, 00146 Rome, Italy; (M.M.); (L.S.)
| | - Marco Iuliano
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome Sapienza, Polo Pontino, 04100 Latina, Italy; (S.S.); (M.I.); (V.P.); (G.R.); (G.M.); (A.C.)
| | - Luca Stefanuto
- Department of Science, University of Roma Tre, 00146 Rome, Italy; (M.M.); (L.S.)
| | - Elena Carbone
- Department of Neuroscience, Italian National Institute of Health, 00161 Rome, Italy; (E.C.); (P.P.)
| | - Paola Piscopo
- Department of Neuroscience, Italian National Institute of Health, 00161 Rome, Italy; (E.C.); (P.P.)
| | - Vincenzo Petrozza
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome Sapienza, Polo Pontino, 04100 Latina, Italy; (S.S.); (M.I.); (V.P.); (G.R.); (G.M.); (A.C.)
- Istituto Chirurgico Ortopedico Traumatologico (ICOT), 04100 Latina, Italy
| | - Giovanna Romeo
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome Sapienza, Polo Pontino, 04100 Latina, Italy; (S.S.); (M.I.); (V.P.); (G.R.); (G.M.); (A.C.)
| | - Giorgio Mangino
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome Sapienza, Polo Pontino, 04100 Latina, Italy; (S.S.); (M.I.); (V.P.); (G.R.); (G.M.); (A.C.)
| | - Antonella Calogero
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome Sapienza, Polo Pontino, 04100 Latina, Italy; (S.S.); (M.I.); (V.P.); (G.R.); (G.M.); (A.C.)
- Istituto Chirurgico Ortopedico Traumatologico (ICOT), 04100 Latina, Italy
| | - Tecla Gasperi
- Department of Science, University of Roma Tre, 00146 Rome, Italy; (M.M.); (L.S.)
- National Institute of Biostructures and Biosystems (INBB), 00136 Rome, Italy
| | - Paolo Rosa
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome Sapienza, Polo Pontino, 04100 Latina, Italy; (S.S.); (M.I.); (V.P.); (G.R.); (G.M.); (A.C.)
- Istituto Chirurgico Ortopedico Traumatologico (ICOT), 04100 Latina, Italy
| |
Collapse
|
2
|
Cho W, Oh H, Abd El-Aty AM, Mobarak EH, Jeong JH, Jung TW. IL-38 alleviates atherogenic responses via SIRT6/HO-1 signaling: A promising strategy against obesity-related atherosclerosis. Biochem Biophys Res Commun 2024; 694:149407. [PMID: 38154209 DOI: 10.1016/j.bbrc.2023.149407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 12/30/2023]
Abstract
Interleukin-38 (IL-38), a member of the IL-1 family, is known for its anti-inflammatory properties mediated through ligand signaling in various disease models. It plays a significant role in atherosclerosis development, forming a theoretical basis for therapeutic strategies. However, the direct effects of IL-38 on atherogenic responses in the vascular endothelium and monocytes remain unclear. In this investigation, IL-38 treatment reduced THP-1 monocyte adhesion to HUVECs, decreased the expression of vascular adhesion molecules, and mitigated inflammation in the presence of palmitate. IL-38 treatment upregulated SIRT6 expression and enhanced autophagy markers such as LC3 conversion and p62 degradation. The effects of IL-38 were nullified by siRNA-mediated suppression of SIRT6 or heme oxygenase-1 (HO-1) in HUVECs and palmitate-treated THP-1 cells. These findings reveal that IL-38 mitigates inflammation through the SIRT6/HO-1 pathway, offering a potential therapeutic approach for addressing obesity-related atherosclerosis.
Collapse
Affiliation(s)
- Wonjun Cho
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Heeseung Oh
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - A M Abd El-Aty
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, 12211, Giza, Egypt; Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum, 25240, Turkey.
| | - Enas H Mobarak
- Department of Restorative Dentistry, Faculty of Dentistry, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - Tae Woo Jung
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Pan S, Hale AT, Lemieux ME, Raval DK, Garton TP, Sadler B, Mahaney KB, Strahle JM. Iron homeostasis and post-hemorrhagic hydrocephalus: a review. Front Neurol 2024; 14:1287559. [PMID: 38283681 PMCID: PMC10811254 DOI: 10.3389/fneur.2023.1287559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 11/21/2023] [Indexed: 01/30/2024] Open
Abstract
Iron physiology is regulated by a complex interplay of extracellular transport systems, coordinated transcriptional responses, and iron efflux mechanisms. Dysregulation of iron metabolism can result in defects in myelination, neurotransmitter synthesis, and neuronal maturation. In neonates, germinal matrix-intraventricular hemorrhage (GMH-IVH) causes iron overload as a result of blood breakdown in the ventricles and brain parenchyma which can lead to post-hemorrhagic hydrocephalus (PHH). However, the precise mechanisms by which GMH-IVH results in PHH remain elusive. Understanding the molecular determinants of iron homeostasis in the developing brain may lead to improved therapies. This manuscript reviews the various roles iron has in brain development, characterizes our understanding of iron transport in the developing brain, and describes potential mechanisms by which iron overload may cause PHH and brain injury. We also review novel preclinical treatments for IVH that specifically target iron. Understanding iron handling within the brain and central nervous system may provide a basis for preventative, targeted treatments for iron-mediated pathogenesis of GMH-IVH and PHH.
Collapse
Affiliation(s)
- Shelei Pan
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Andrew T. Hale
- Department of Neurosurgery, University of Alabama at Birmingham School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mackenzie E. Lemieux
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Dhvanii K. Raval
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Thomas P. Garton
- Department of Neurology, Johns Hopkins University School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Brooke Sadler
- Department of Pediatrics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Hematology and Oncology, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Kelly B. Mahaney
- Department of Neurosurgery, Stanford University School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Jennifer M. Strahle
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Pediatrics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Orthopedic Surgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
4
|
Jung TW, Kim H, Park SY, Cho W, Oh H, Lee HJ, Abd El-Aty AM, Hacimuftuoglu A, Jeong JH. Stachydrine alleviates lipid-induced skeletal muscle insulin resistance via AMPK/HO-1-mediated suppression of inflammation and endoplasmic reticulum stress. J Endocrinol Invest 2022; 45:2181-2191. [PMID: 35834165 DOI: 10.1007/s40618-022-01866-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 07/06/2022] [Indexed: 10/17/2022]
Abstract
OBJECTIVE Insulin resistance develops due to skeletal muscle inflammation and endoplasmic reticulum (ER) stress. Stachydrine (STA), extracted from Leonurus heterophyllus, has been shown to suppress proliferation and induce apoptosis in breast cancer cells and exert anti-inflammatory properties in the brain, heart, and liver. However, the roles of STA in insulin signaling in skeletal muscle remain unclear. Herein, we examined the impacts of STA on insulin signaling in skeletal muscle under hyperlipidemic conditions and its related molecular mechanisms. METHODS Various protein expression levels were determined by Western blotting. Levels of mouse serum cytokines were measured by ELISA. RESULTS We found that STA-ameliorated inflammation and ER stress, leading to attenuation of insulin resistance in palmitate-treated C2C12 myocytes. STA dose-dependently enhanced AMPK phosphorylation and HO-1 expression. Administration of STA attenuated not only insulin resistance but also inflammation and ER stress in the skeletal muscle of high-fat diet (HFD)-fed mice. Additionally, STA-ameliorated glucose tolerance and insulin sensitivity, as well as serum TNFα and MCP-1, in mice fed a HFD. Small interfering (si) RNA-associated suppression of AMPK or HO-1 expression abolished the effects of STA in C2C12 myocytes. CONCLUSIONS These results suggest that STA activates AMPK/HO-1 signaling, resulting in reduced inflammation and ER stress, thereby improving skeletal muscle insulin resistance. Using STA as a natural ingredient, this research successfully treated insulin resistance and type 2 diabetes.
Collapse
Affiliation(s)
- T W Jung
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - H Kim
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - S Y Park
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - W Cho
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - H Oh
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - H J Lee
- Department of Anatomy and Cell Biology, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Korea
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - A M Abd El-Aty
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
- Department of Medical Pharmacology, Medical Faculty, Ataturk University, 25240, Erzurum, Türkiye
| | - A Hacimuftuoglu
- Department of Medical Pharmacology, Medical Faculty, Ataturk University, 25240, Erzurum, Türkiye
| | - J H Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea.
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea.
| |
Collapse
|
5
|
Lim JS, Oh J, Yun HS, Lee JS, Hahn D, Kim JS. Anti-neuroinflammatory activity of 6,7-dihydroxy-2,4-dimethoxy phenanthrene isolated from Dioscorea batatas Decne partly through suppressing the p38 MAPK/NF-κB pathway in BV2 microglial cells. JOURNAL OF ETHNOPHARMACOLOGY 2022; 282:114633. [PMID: 34520827 DOI: 10.1016/j.jep.2021.114633] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/02/2021] [Accepted: 09/09/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The rhizome of Dioscorea batatas Decne (called Chinses yam) widely distributed in East Asian countries including China, Japan, Korea and Taiwan has long been used in oriental folk medicine owing to its tonic, antitussive, expectorant and anti-ulcerative effects. It has been reported to have anti-inflammatory, antioxidative, cholesterol-lowering, anticholinesterase, growth hormone-releasing, antifungal and immune cell-stimulating activities. AIM OF THE STUDY Neuroinflammation caused by activated microglia contributes to neuronal dysfunction and neurodegeneration. In the present study, the anti-neuroinflammatory activity of 6,7-dihydroxy-2,4-dimethoxy phenanthrene (DHDMP), a phenanthrene compound isolated from Dioscorea batatas Decne, was examined in microglial and neuronal cells. MATERIALS AND METHODS A natural phenanthrene compound, DHDMP, was isolated from the peel of Dioscorea batatas Decne. The anti-neuroinflammatory capability of the compound was examined using the co-culture system of BV2 murine microglial and HT22 murine neuronal cell lines. The expression levels of inflammatory mediators and cytoprotective proteins in the cells were quantified by enzyme-linked immunosorbent assay and Western blot analysis. RESULTS DHDMP at the concentrations of ≤1 μg/mL did not exhibit a cytotoxic effect for BV2 and HT22 cells. Rather DHDMP effectively restored the growth rate of HT22 cells, which was reduced by co-culture with lipopolysaccharide (LPS)-treated BV2 cells. DHDMP significantly decreased the production of proinflammatory mediators, such as nitric oxide, tumor necrosis factor-α, interleukin-6, inducible nitric oxide synthase, and cyclooxygenase-2 in BV2 cells. Moreover, DHDMP strongly inhibited the nuclear translocation of nuclear factor κB (NF-κB) and phosphorylation of p38 mitogen-activated protein kinase (MAPK) in BV2 cells. The compound did not affect the levels and phosphorylation of ERK and JNK. Concurrently, DHDMP increased the expression of heme oxygenase-1 (HO-1), an inducible cytoprotective enzyme, in HT22 cells. CONCLUSIONS Our findings indicate that DHDMP effectively dampened LPS-mediated inflammatory responses in BV2 microglial cells by suppressing transcriptional activity of NF-κB and its downstream mediators and contributed to HT22 neuronal cell survival. This study provides insight into the therapeutic potential of DHDMP for inflammation-related neurological diseases.
Collapse
Affiliation(s)
- Ji Sun Lim
- Institute of Agricultural Science and Technology, Kyungpook National University, Daegu, 41566, South Korea.
| | - Jisun Oh
- Institute of Agricultural Science and Technology, Kyungpook National University, Daegu, 41566, South Korea.
| | - Hyun Seok Yun
- School of Food Science and Biotechnology, Kyungpook National University, Daegu, 41566, South Korea.
| | - Jeong Soon Lee
- Forest Resources Development Institute of Gyeongsangbuk-do, Andong, 36605, South Korea.
| | - Dongyup Hahn
- School of Food Science and Biotechnology, Kyungpook National University, Daegu, 41566, South Korea.
| | - Jong-Sang Kim
- Institute of Agricultural Science and Technology, Kyungpook National University, Daegu, 41566, South Korea; School of Food Science and Biotechnology, Kyungpook National University, Daegu, 41566, South Korea.
| |
Collapse
|
6
|
Moeinabadi-Bidgoli K, Babajani A, Yazdanpanah G, Farhadihosseinabadi B, Jamshidi E, Bahrami S, Niknejad H. Translational insights into stem cell preconditioning: From molecular mechanisms to preclinical applications. Biomed Pharmacother 2021; 142:112026. [PMID: 34411911 DOI: 10.1016/j.biopha.2021.112026] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 08/05/2021] [Accepted: 08/07/2021] [Indexed: 02/06/2023] Open
Abstract
Cell-based therapy (CBT) is a revolutionary approach for curing a variety of degenerative diseases. Stem cell-based regenerative medicine is a novel strategy for treating tissue damages regarding stem cells unique properties such as differentiation potential, paracrine impacts, and self-renewal ability. However, the current cell-based treatments encounter considerable challenges to be translated into clinical practice, including low cell survival, migration, and differentiation rate of transplanted stem cells. The poor stem cell therapy outcomes mainly originate from the unfavorable condition of damaged tissues for transplanted stem cells. The promising method of preconditioning improves cell resistance against the host environment's stress by imposing certain conditions similar to the harsh microenvironment of the damaged tissues on the transplanted stem cells. Various pharmacological, biological, and physical inducers are able to establish preconditioning. In addition to their known pharmacological effects on tissues and cells, these preconditioning agents improve cell biological aspects such as cell survival, proliferation, differentiation, migration, immunomodulation, paracrine impacts, and angiogenesis. This review focuses on different protocols and inducers of preconditioning along with underlying molecular mechanisms of their effects on stem cell behavior. Moreover, preclinical applications of preconditioned stem cells in various damaged organs such as heart, lung, brain, bone, cartilage, liver, and kidney are discussed with prospects of their translation into the clinic.
Collapse
Affiliation(s)
- Kasra Moeinabadi-Bidgoli
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirhesam Babajani
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ghasem Yazdanpanah
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA
| | | | - Elham Jamshidi
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soheyl Bahrami
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in AUVA Research Center, Vienna, Austria
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Zhang C, Yuan Y, Ou M. Mangiferin attenuates cigarette smoke-induced chronic obstructive pulmonary disease in male albino rats. Microvasc Res 2021; 138:104208. [PMID: 34139206 DOI: 10.1016/j.mvr.2021.104208] [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: 02/16/2021] [Revised: 05/18/2021] [Accepted: 06/07/2021] [Indexed: 11/30/2022]
Abstract
We analyzed the ability of mangiferin to suppress cigarette smoke-induced chronic obstructive pulmonary disease. Control rats showed a marked decrease in the ratio of the forced expiratory volume at 0.1 s to forced vital capacity. The decreases in the peak expiratory flow and maximal mid-expiratory flow indicated airway remodeling and enlargement. The expression levels of superoxide dismutase (SOD), heme oxygenase-1 (HO-1), γ-glutamylcysteine synthetase, nuclear factor erythroid 2-related factor 2, and activating transcription factor 4 were increased in the control rats. The levels of oxidative stress, malondialdehyde, and reactive oxygen species peaked after 24 weeks, whereas the SOD and HO-1 levels and the total antioxidant capacity were reduced in control rats. Mangiferin restored the levels of reactive oxygen species, malondialdehyde, SOD, HO-1, and T-AOC to near normal. Increased numbers of infiltrating inflammatory cells were observed in control rats but were significantly reduced by mangiferin. In addition, edema and airway inflammation were reduced by mangiferin.
Collapse
Affiliation(s)
- Chao Zhang
- The Sixth Department of Health Care, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100048, China
| | - Yi Yuan
- Beijing Academy of Food Sciences, Beijing 100068, China
| | - Min Ou
- The Sixth Department of Health Care, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100048, China.
| |
Collapse
|
8
|
Heme oxygenase-1 in blood and saliva during acute psychosis: A pilot study. Psychiatry Res 2021; 299:113857. [PMID: 33756209 DOI: 10.1016/j.psychres.2021.113857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 03/04/2021] [Indexed: 11/22/2022]
Abstract
Despite the extensive prevalence of psychosis and schizophrenia spectrum disorders, their biological underpinnings remain largely unexplained. Recently, the overproduction of heme oxygenase-1 (HO-1), an enzyme that catalyzes the degradation of heme, was associated with oxidative stress and a neurologic phenotype similar to schizophrenia in transgenic mice. We sought to evaluate, by comparing patients experiencing an acute psychotic episode, and age/sex-matched healthy control participants, whether there was an association between HO-1 overexpression and psychosis. This cross-sectional pilot study included 16 patients and 17 control participants. Enzyme-linked immunosorbent assay and quantitative real-time polymerase chain reaction were used to quantify HO-1 expression in blood and saliva. Four psychiatric questionnaires were used to measure psychiatric symptoms in participants. Higher levels of salivary HO-1 expression were detected in patients experiencing an acute psychotic episode when compared to control participants (84.01 vs. 61.26 ng/ml, p = 0.026), but plasma and lymphocyte HO-1 expression did not significantly differ between groups. Overexpression of HO-1 in saliva specimens was also positively associated with psychiatric symptom severity and disability. The overexpression of HO-1 in the saliva of patients with psychosis suggests that it may serve as a potential biomarker for this symptom which should be explored in larger clinical trials.
Collapse
|
9
|
Chen J, Lan C, An H, Jin Y, Li Q, Ge S, Yu Y, Shen G, Pan B, Xu Y, Ye R, Li Z, Wang B. Potential interference on the lipid metabolisms by serum copper in a women population: A repeated measurement study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 760:143375. [PMID: 33189376 DOI: 10.1016/j.scitotenv.2020.143375] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 06/11/2023]
Abstract
The relationship between excess copper (Cu) intake and lipid metabolic disorders is not well-studied, and most studies on this topic have a cross-sectional design. Here, we investigated the relationship between Cu exposure and blood lipid metabolism in women population, as well as potential mediation effects of oxidative stress and inflammation, using a repeated-measurement study. A total of 35 women in northern China were included, and each individual was visited for five times. Blood samples were collected, and the following serum biomarkers were analyzed: heme oxygenase-1 (HO-1), monocyte chemotactic protein-1 (MCP-1), interleukin (IL)-6, IL-8, and lipids [triglycerides (TG), total cholesterol (TC), low-density lipoprotein cholesterol (LDL), high-density lipoprotein cholesterol (HDL), and lipoprotein(a) (Lp(a))]. A linear mixed-effect model was used to analyze the associations between Cu and the individual biomarkers in serum. The results showed that Cu was positively associated with TG (β = 0.0007, P = 0.01), TC (β = 0.0006, P = 0.002), LDL (β = 0.0004, P ≤ 0.001), and Lp(a) (β = 0.0004, P = 0.01), but not associated with HDL (β = 0.0001, P = 0.19). Likewise, serum Cu was positively associated with HO-1 (β = 0.0004, P = 0.03) and negatively associated with MCP-1 (β = -0.0006, P = 0.003) and IL-8 (β = -0.002, P = 0.03). Among the biomarkers of oxidative stress, inflammation, and lipids in serum, only IL-8 was negatively associated with HDL (β = -0.0004, P = 0.009). No other associations were observed. We conclude that high Cu exposure may elevate blood lipid levels as well as disturb processes related to oxidative stress and inflammation responses.
Collapse
Affiliation(s)
- Junxi Chen
- Institute of Reproductive and Child Health, Peking University, Beijing 100191, PR China; Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, PR China; Key Laboratory of Reproductive Health, National Health Commission of the People's Republic of China, Beijing 100191, PR China
| | - Changxin Lan
- Institute of Reproductive and Child Health, Peking University, Beijing 100191, PR China; Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, PR China; Key Laboratory of Reproductive Health, National Health Commission of the People's Republic of China, Beijing 100191, PR China
| | - Hang An
- Institute of Reproductive and Child Health, Peking University, Beijing 100191, PR China; Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, PR China; Key Laboratory of Reproductive Health, National Health Commission of the People's Republic of China, Beijing 100191, PR China
| | - Yu Jin
- Institute of Reproductive and Child Health, Peking University, Beijing 100191, PR China; Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, PR China; Key Laboratory of Reproductive Health, National Health Commission of the People's Republic of China, Beijing 100191, PR China
| | - Qi Li
- School of Resources and Environmental Engineering, Jiangxi University of Science and Technology, Ganzhou 341000, PR China
| | - Shufang Ge
- School of Environment, Beijing Normal University, Beijing 100875, PR China
| | - Yanxin Yu
- School of Environment, Beijing Normal University, Beijing 100875, PR China
| | - Guofeng Shen
- College of Urban and Environmental Sciences, Peking University, Beijing 100871, PR China
| | - Bo Pan
- Department of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, 650051, PR China
| | - Ying Xu
- Department of Building Science, Tsinghua University, Beijing, 100084, PR China
| | - Rongwei Ye
- Institute of Reproductive and Child Health, Peking University, Beijing 100191, PR China; Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, PR China; Key Laboratory of Reproductive Health, National Health Commission of the People's Republic of China, Beijing 100191, PR China
| | - Zhiwen Li
- Institute of Reproductive and Child Health, Peking University, Beijing 100191, PR China; Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, PR China; Key Laboratory of Reproductive Health, National Health Commission of the People's Republic of China, Beijing 100191, PR China
| | - Bin Wang
- Institute of Reproductive and Child Health, Peking University, Beijing 100191, PR China; Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, PR China; Key Laboratory of Reproductive Health, National Health Commission of the People's Republic of China, Beijing 100191, PR China.
| |
Collapse
|
10
|
A Review on Potential Footprints of Ferulic Acid for Treatment of Neurological Disorders. Neurochem Res 2021; 46:1043-1057. [PMID: 33547615 DOI: 10.1007/s11064-021-03257-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 01/16/2021] [Accepted: 01/22/2021] [Indexed: 02/06/2023]
Abstract
Ferulic acid is being screened in preclinical settings to combat various neurological disorders. It is a naturally occurring dietary flavonoid commonly found in grains, fruits, and vegetables such as rice, wheat, oats, tomatoes, sweet corn etc., which exhibits protective effects against a number of neurological diseases such as epilepsy, depression, ischemia-reperfusion injury, Alzheimer's disease, and Parkinson's disease. Ferulic acid prevents and treats different neurological diseases pertaining to its potent anti-oxidative and anti-inflammatory effects, beside modulating unique neuro-signaling pathways. It stays in the bloodstream for longer periods than other dietary polyphenols and antioxidants and easily crosses blood brain barrier. The use of novel drug delivery systems such as solid-lipid nanoparticles (SLNs) or its salt forms (sodium ferulate, ethyl ferulate, and isopentyl ferulate) further enhance its bioavailability and cerebral penetration. Based on reported studies, ferulic acid appears to be a promising molecule for treatment of neurological disorders; however, more preclinical (in vitro and in vivo) mechanism-based studies should be planned and conceived followed by its testing in clinical settings.
Collapse
|
11
|
Solár P, Brázda V, Levin S, Zamani A, Jančálek R, Dubový P, Joukal M. Subarachnoid Hemorrhage Increases Level of Heme Oxygenase-1 and Biliverdin Reductase in the Choroid Plexus. Front Cell Neurosci 2020; 14:593305. [PMID: 33328892 PMCID: PMC7732689 DOI: 10.3389/fncel.2020.593305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/04/2020] [Indexed: 11/18/2022] Open
Abstract
Subarachnoid hemorrhage is a specific, life-threatening form of hemorrhagic stroke linked to high morbidity and mortality. It has been found that the choroid plexus of the brain ventricles forming the blood-cerebrospinal fluid barrier plays an important role in subarachnoid hemorrhage pathophysiology. Heme oxygenase-1 and biliverdin reductase are two of the key enzymes of the hemoglobin degradation cascade. Therefore, the aim of present study was to investigate changes in protein levels of heme oxygenase-1 and biliverdin reductase in the rat choroid plexus after experimental subarachnoid hemorrhage induced by injection of non-heparinized autologous blood to the cisterna magna. Artificial cerebrospinal fluid of the same volume as autologous blood was injected to mimic increased intracranial pressure in control rats. Immunohistochemical and Western blot analyses were used to monitor changes in the of heme oxygenase-1 and biliverdin reductase levels in the rat choroid plexus after induction of subarachnoid hemorrhage or artificial cerebrospinal fluid application for 1, 3, and 7 days. We found increased levels of heme oxygenase-1 and biliverdin reductase protein in the choroid plexus over the entire period following subarachnoid hemorrhage induction. The level of heme oxygenase-1 was the highest early (1 and 3 days) after subarachnoid hemorrhage indicating its importance in hemoglobin degradation. Increased levels of heme oxygenase-1 were also observed in the choroid plexus epithelial cells at all time points after application of artificial cerebrospinal fluid. Biliverdin reductase protein was detected mainly in the choroid plexus epithelial cells, with levels gradually increasing during subarachnoid hemorrhage. Our results suggest that heme oxygenase-1 and biliverdin reductase are involved not only in hemoglobin degradation but probably also in protecting choroid plexus epithelial cells and the blood-cerebrospinal fluid barrier from the negative effects of subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Peter Solár
- Department of Anatomy, Faculty of Medicine, Cellular and Molecular Neurobiology Research Group, Masaryk University, Brno, Czechia.,Department of Neurosurgery - St. Anne's University Hospital Brno, Faculty of Medicine, Masaryk University, Brno, Czechia.,Department of Neurosurgery, St. Anne's University Hospital Brno, Brno, Czechia
| | - Václav Brázda
- Department of Anatomy, Faculty of Medicine, Cellular and Molecular Neurobiology Research Group, Masaryk University, Brno, Czechia.,Institute of Biophysics of the Czech Academy of Sciences, Brno, Czechia
| | - Shahaf Levin
- Department of Anatomy, Faculty of Medicine, Cellular and Molecular Neurobiology Research Group, Masaryk University, Brno, Czechia
| | - Alemeh Zamani
- Department of Anatomy, Faculty of Medicine, Cellular and Molecular Neurobiology Research Group, Masaryk University, Brno, Czechia
| | - Radim Jančálek
- Department of Neurosurgery - St. Anne's University Hospital Brno, Faculty of Medicine, Masaryk University, Brno, Czechia.,Department of Neurosurgery, St. Anne's University Hospital Brno, Brno, Czechia
| | - Petr Dubový
- Department of Anatomy, Faculty of Medicine, Cellular and Molecular Neurobiology Research Group, Masaryk University, Brno, Czechia
| | - Marek Joukal
- Department of Anatomy, Faculty of Medicine, Cellular and Molecular Neurobiology Research Group, Masaryk University, Brno, Czechia
| |
Collapse
|
12
|
Ahmad R, Khan A, Lee HJ, Ur Rehman I, Khan I, Alam SI, Kim MO. Lupeol, a Plant-Derived Triterpenoid, Protects Mice Brains against Aβ-Induced Oxidative Stress and Neurodegeneration. Biomedicines 2020; 8:biomedicines8100380. [PMID: 32993092 PMCID: PMC7601269 DOI: 10.3390/biomedicines8100380] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/11/2020] [Accepted: 09/25/2020] [Indexed: 12/15/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder that represents 60–70% of all dementia cases. AD is characterized by the formation and accumulation of amyloid-beta (Aβ) plaques, neurofibrillary tangles, and neuronal cell loss. Further accumulation of Aβ in the brain induces oxidative stress, neuroinflammation, and synaptic and memory dysfunction. In this study, we investigated the antioxidant and neuroprotective effects of the natural triterpenoid lupeol in the Aβ1-42 mouse model of AD. An Intracerebroventricular injection (i.c.v.) of Aβ (3 µL/5 min/mouse) into the brain of a mouse increased the reactive oxygen species (ROS) levels, neuroinflammation, and memory and cognitive dysfunction. The oral administration of lupeol at a dose of 50 mg/kg for two weeks significantly decreased the oxidative stress, neuroinflammation, and memory impairments. Lupeol decreased the oxidative stress via the activation of nuclear factor erythroid 2-related factor-2 (Nrf-2) and heme oxygenase-1 (HO-1) in the brain of adult mice. Moreover, lupeol treatment prevented neuroinflammation by suppressing activated glial cells and inflammatory mediators. Additionally, lupeol treatment significantly decreased the accumulation of Aβ and beta-secretase-1 (BACE-1) expression and enhanced the memory and cognitive function in the Aβ-mouse model of AD. To the best of our knowledge, this is the first study to investigate the anti-oxidative and neuroprotective effects of lupeol against Aβ1-42-induced neurotoxicity. Our findings suggest that lupeol could serve as a novel, promising, and accessible neuroprotective agent against progressive neurodegenerative diseases such as AD.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Myeong Ok Kim
- Correspondence: ; Tel.: +82-55-772-1345; Fax: +82-55-772-2656
| |
Collapse
|
13
|
Li X, Liu W, Li R, Guo S, Fan H, Wei B, Zhang X, He X, Duan C. TSG-6 Attenuates Oxidative Stress-Induced Early Brain Injury in Subarachnoid Hemorrhage Partly by the HO-1 and Nox2 Pathways. J Stroke Cerebrovasc Dis 2020; 29:104986. [PMID: 32992175 DOI: 10.1016/j.jstrokecerebrovasdis.2020.104986] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 04/11/2020] [Accepted: 05/18/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Early brain injury (EBI) refers to acute brain injury during the first 72 h after subarachnoid hemorrhage (SAH), which is one of the major causes of poor prognosis after SAH. Here, we investigated the effect and the related mechanism of TSG-6 on EBI after SAH. MATERIALS AND METHODS The Sprague-Dawley rat model of SAH was developed by the endovascular perforation method. TSG-6 (5μg) was administered by an intraventricular injection within 1.5 h after SAH. The effects of TSG-6 on EBI were assessed by neurological score, brain water content (BWC) and TUNEL staining. Immunofluorescence staining was used to assay NF-κB/p-NF-κB expression in microglia. Protein expression levels of heme oxygenase-1 (HO-1), NADPH oxidase 2 (Nox2), Bcl-2, Bax, and cleaved-caspase-3 were measured to investigate the potential mechanism. The enzyme activity of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) and the level of reactive oxygen species (ROS) were analyzed using commercially available kits. RESULTS The results showed that TSG-6 treatment alleviated the neurobehavioral dysfunction and reduced BWC and the number of TUNEL-positive neurons in EBI after SAH. TSG-6 decreased the ROS level and enhanced the enzyme activity of SOD and GSH-Px after SAH. Furthermore TSG-6 inhibited the NF-κB activation, increased the protein expression levels of HO-1 and Bcl-2 and decreased the expression levels of Nox2, Bax, and cleaved-caspase-3. The administration of TSG-6 siRNA abolished the protective effects of TSG-6 on EBI after SAH. CONCLUSION We found that TSG-6 attenuated oxidative stress and apoptosis in EBI after SAH partly by inhibiting NF-κB and activating HO-1 pathway in brain tissue.
Collapse
Affiliation(s)
- Xifeng Li
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 253# Industry Road, Guangdong 510282, China
| | - Wenchao Liu
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 253# Industry Road, Guangdong 510282, China
| | - Ran Li
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 253# Industry Road, Guangdong 510282, China
| | - Shenquan Guo
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 253# Industry Road, Guangdong 510282, China
| | - Haiyan Fan
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 253# Industry Road, Guangdong 510282, China
| | - Boyang Wei
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 253# Industry Road, Guangdong 510282, China
| | - Xin Zhang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 253# Industry Road, Guangdong 510282, China
| | - Xuying He
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 253# Industry Road, Guangdong 510282, China
| | - Chuanzhi Duan
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 253# Industry Road, Guangdong 510282, China.
| |
Collapse
|
14
|
Zhou J, Terluk MR, Basso L, Mishra UR, Orchard PJ, Cloyd JC, Schröder H, Kartha RV. N-acetylcysteine Provides Cytoprotection in Murine Oligodendrocytes through Heme Oxygenase-1 Activity. Biomedicines 2020; 8:biomedicines8080240. [PMID: 32717964 PMCID: PMC7460204 DOI: 10.3390/biomedicines8080240] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/21/2020] [Accepted: 07/21/2020] [Indexed: 02/06/2023] Open
Abstract
Oligodendrocytic injury by oxidative stress can lead to demyelination, contributing to neurodegeneration. We investigated the mechanisms by which an antioxidant, N-acetylcysteine (NAC), reduces oxidative stress in murine oligodendrocytes. We used normal 158N and mutant 158JP cells with endogenously high reactive oxygen species (ROS) levels. Oxidative stress was induced in 158N cells using hydrogen peroxide (H2O2, 500 μM), and both cells were treated with NAC (50 µM to 500 µM). ROS production, total glutathione (GSH) and cell survival were measured 24 h after treatment. In normal cells, H2O2 treatment resulted in a ~5.5-fold increase in ROS and ~50% cell death. These deleterious effects of oxidative stress were attenuated by NAC, resulting in improved cell survival. Similarly, NAC treatment resulted in decreased ROS levels in 158JP cells. Characterization of mechanisms underlying cytoprotection in both cell lines revealed an increase in GSH levels by NAC, which was partially blocked by an inhibitor of GSH synthesis. Interestingly, we observed heme oxygenase-1 (HO-1), a cytoprotective enzyme, play a critical role in cytoprotection. Inhibition of HO-1 activity abolished the cytoprotective effect of NAC with a corresponding decrease in total antioxidant capacity. Our results indicate that NAC promotes oligodendrocyte survival in oxidative stress-related conditions through multiple pathways.
Collapse
Affiliation(s)
- Jie Zhou
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, 2001 6th Street SE, Minneapolis, MN 55455, USA; (J.Z.); (M.R.T.); (L.B.); (U.R.M.); (J.C.C.)
| | - Marcia R. Terluk
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, 2001 6th Street SE, Minneapolis, MN 55455, USA; (J.Z.); (M.R.T.); (L.B.); (U.R.M.); (J.C.C.)
| | - Lisa Basso
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, 2001 6th Street SE, Minneapolis, MN 55455, USA; (J.Z.); (M.R.T.); (L.B.); (U.R.M.); (J.C.C.)
| | - Usha R. Mishra
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, 2001 6th Street SE, Minneapolis, MN 55455, USA; (J.Z.); (M.R.T.); (L.B.); (U.R.M.); (J.C.C.)
| | - Paul J. Orchard
- Division of Pediatric Blood and Marrow Transplantation, Department of Pediatrics, Medical School, University of Minnesota, 425 East River Parkway, Minneapolis, MN 55455, USA;
| | - James C. Cloyd
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, 2001 6th Street SE, Minneapolis, MN 55455, USA; (J.Z.); (M.R.T.); (L.B.); (U.R.M.); (J.C.C.)
| | - Henning Schröder
- Department of Pharmaceutics, University of Minnesota, 308 Harvard Street SE, Minneapolis, MN 55455, USA;
| | - Reena V. Kartha
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, 2001 6th Street SE, Minneapolis, MN 55455, USA; (J.Z.); (M.R.T.); (L.B.); (U.R.M.); (J.C.C.)
- Correspondence: ; Tel.: +1-612-626-2436
| |
Collapse
|
15
|
Feltham BA, Louis XL, Eskin MNA, Suh M. Docosahexaenoic Acid: Outlining the Therapeutic Nutrient Potential to Combat the Prenatal Alcohol-Induced Insults on Brain Development. Adv Nutr 2020; 11:724-735. [PMID: 31989167 PMCID: PMC7231602 DOI: 10.1093/advances/nmz135] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 12/05/2019] [Accepted: 12/22/2019] [Indexed: 01/20/2023] Open
Abstract
Brain development is markedly affected by prenatal alcohol exposure, leading to cognitive and behavioral problems in the children. Protecting neuronal damage from prenatal alcohol could improve neural connections and functioning of the brain. DHA, a n-3 (ω-3) long-chain PUFA, is involved in the development of neurons. Insufficient concentrations of DHA impair neuronal development and plasticity of synaptic junctions and affect neurotransmitter concentrations in the brain. Alcohol consumption during pregnancy decreases the maternal DHA status and reduces the placental transfer of DHA to the fetus, resulting in less DHA being available for brain development. It is important to know whether DHA could induce beneficial effects on various physiological functions that promote neuronal development. This review will discuss the current evidence for the beneficial role of DHA in protecting against neuronal damage and its potential in mitigating the teratogenic effects of alcohol.
Collapse
Affiliation(s)
- Bradley A Feltham
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
| | - Xavier L Louis
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
| | - Michael N A Eskin
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Miyoung Suh
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
| |
Collapse
|
16
|
Karampoor S, Zahednasab H, Amini R, Esghaei M, Sholeh M, Keyvani H. Maraviroc attenuates the pathogenesis of experimental autoimmune encephalitis. Int Immunopharmacol 2020; 80:106138. [PMID: 32007705 DOI: 10.1016/j.intimp.2019.106138] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/06/2019] [Accepted: 12/18/2019] [Indexed: 12/26/2022]
Abstract
It has been shown that the blockade of chemokine receptor type 5 can dampen inflammatory reaction within the central nervous system (CNS). In the present study, we utilized maraviroc, a potent antagonist o CCR5, to examine whether this drug can mitigate neuroinflammation in the spinal cord of mice induced by experimental autoimmune encephalitis (EAE), considered a murine model of multiple sclerosis (MS). For this aim, mice were immunized with myelin oligodendrocyte glycoprotein 35-55 (MOG35-55), followed by pertussis toxin to induce paralysis in EAE mice. The animals intraperitoneally received various doses of maraviroc (5, 25, and 50 mg/kg body weight) when the early clinical signs of EAE appeared. The results demonstrated that the administration of maraviroc led to a marked decrease in the clinical score and improvement in behavioral motor functions. Moreover, our finding indicated that the administration of maraviroc significantly attenuates the infiltration of inflammatory cells to the spinal cord, microgliosis, astrogliosis, pro-inflammatory cytokines, and cell death in EAE mice. The flow cytometry data indicated that a decreased number of CD4+ and CD8+ T cells in the peripheral blood of mice with EAE without affecting the number of T regulatory cells (CD4 + CD25+ forkhead box protein 3+). Finally, it seems that maraviroc is well-tolerated, and targeting CCR5 could open up a new horizon in the treatment of MS.
Collapse
Affiliation(s)
- Sajad Karampoor
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamid Zahednasab
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Razieh Amini
- Department of Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Maryam Esghaei
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sholeh
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Keyvani
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
Kumar A, Ghosh DK, Ranjan A. Mefloquine binding to human acyl-CoA binding protein leads to redox stress-mediated apoptotic death of human neuroblastoma cells. Neurotoxicology 2020; 77:169-180. [PMID: 31987860 DOI: 10.1016/j.neuro.2020.01.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 01/12/2020] [Accepted: 01/16/2020] [Indexed: 12/19/2022]
Abstract
Malaria is an infectious disease that is caused by different species of Plasmodium. Several antimalarial drugs are used to counter the spread and infectivity of Plasmodium species. However, humans are also vulnerable to many of the antimalarial drugs, including the quinoline-based drugs. In particular, the antimalarial mefloquine has been reported to show adverse neuropsychiatric effects in humans. Though mefloquine is known to be neurotoxic, the molecular mechanisms associated with this phenomenon are still obscure. In this study, we show that mefloquine binds to and inactivates the human acyl-CoA binding protein (hACBP), potentially inducing redox stress in human neuroblastoma cells (IMR-32). Mefloquine occupies the acyl-CoA binding pocket of hACBP by interacting with several of the critical acyl-CoA binding amino acids. This leads to the competitive inhibition of acyl-CoA(s) binding to hACBP and to the accumulation of lipid droplets inside the IMR-32 cells. The accumulation of cytosolic lipid globules and oxidative stress finally correlates with the apoptotic death of cells. Taken together, our study deciphers a mechanistic detail of how mefloquine leads to the death of human cells by perturbing the activity of hACBP and lipid homeostasis.
Collapse
Affiliation(s)
- Abhishek Kumar
- Computational and Functional Genomics Group, Centre for DNA Fingerprinting and Diagnostics, Uppal, Hyderabad, 500039, Telangana, India; Graduate studies, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Debasish Kumar Ghosh
- Computational and Functional Genomics Group, Centre for DNA Fingerprinting and Diagnostics, Uppal, Hyderabad, 500039, Telangana, India
| | - Akash Ranjan
- Computational and Functional Genomics Group, Centre for DNA Fingerprinting and Diagnostics, Uppal, Hyderabad, 500039, Telangana, India.
| |
Collapse
|
18
|
Lee H, Ko W, Chowdhury A, Li B, Kim SC, Oh H, Kim YC, Woo ER, Baek NI, Lee DS. Brassicaphenanthrene A from Brassica rapa protects HT22 neuronal cells through the regulation of Nrf2‑mediated heme oxygenase‑1 expression. Mol Med Rep 2019; 21:493-500. [PMID: 31746357 DOI: 10.3892/mmr.2019.10824] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/09/2019] [Indexed: 11/05/2022] Open
Abstract
Brain cell damage that results from oxidative toxicity contributes to neuronal degeneration. The transcription factor nuclear factor‑E2‑related factor 2 (Nrf2) regulates the expression of heme oxygenase (HO)‑1 and glutathione (GSH), and serves a key role in the pathogenesis of neurological diseases. Brassica rapa is a turnip that is unique to Ganghwa County, and is used mainly for making kimchi, a traditional Korean food. In the current study, brassicaphenanthrene A (BrPA) from B. rapa was demonstrated to exhibit protective effects against neurotoxicity induced by glutamate via Nrf2‑mediated HO‑1 expression. Similarly, BrPA increased the expression of cellular glutathione and glutamine‑cysteine ligase genes. Furthermore, BrPA caused the nuclear translocation of Nrf2 and increased antioxidant response element (ARE) promoter activity. Nrf2 also mediated HO‑1 induction by BrPA through the PI3K/Akt and JNK regulatory pathways. The results of the present study indicated the neuroprotective effect of BrPA, a natural food component from B. rapa.
Collapse
Affiliation(s)
- Hwan Lee
- College of Pharmacy, Chosun University, Dong‑gu, Gwangju 61452, Republic of Korea
| | - Wonmin Ko
- College of Pharmacy, Chosun University, Dong‑gu, Gwangju 61452, Republic of Korea
| | | | - Bin Li
- Department of Pharmacy, Qingdao University of Science and Technology, Qingdao, Shandong 266042, P.R. China
| | - Sam Cheol Kim
- Department of Family Practice and Community Medicine, Chosun University College of Medicine, Dong‑gu, Gwangju 61452, Republic of Korea
| | - Hyuncheol Oh
- College of Pharmacy, Wonkwang University, Iksan, Jeollabuk-do 54538, Republic of Korea
| | - Youn-Chul Kim
- College of Pharmacy, Wonkwang University, Iksan, Jeollabuk-do 54538, Republic of Korea
| | - Eun-Rhan Woo
- College of Pharmacy, Chosun University, Dong‑gu, Gwangju 61452, Republic of Korea
| | - Nam-In Baek
- Graduate School of Biotechnology and Department of Oriental Medicinal Biotechnology, Kyung‑Hee University, Yongin, Gyeonggi-do 17104, Republic of Korea
| | - Dong-Sung Lee
- College of Pharmacy, Chosun University, Dong‑gu, Gwangju 61452, Republic of Korea
| |
Collapse
|
19
|
Gong X, Liu C, Wang H, Fan J, Jiang C, Zou Y. Effect of heme oxygenase 1 and renin/prorenin receptor on oxidized low-density lipoprotein-induced human umbilical vein endothelial cells. Exp Ther Med 2019; 18:1752-1760. [PMID: 31410134 PMCID: PMC6676210 DOI: 10.3892/etm.2019.7769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 04/12/2019] [Indexed: 12/12/2022] Open
Abstract
The incidence of depression has previously been correlated to hypertension. The aim of the present study was to explore the mechanisms of depression and hypertension by examining the expression and interaction of renin/prorenin receptor (PRR) and heme oxygenase 1 (HO-1) in vascular endothelial cells. A case-control study was conducted, and general data and serum factors were compared between hypertension patients complicated with depression and patients with hypertension alone. Logistic regression analysis was used to detect risk factors associated with hypertension complicated with depression. In addition, human umbilical vein endothelial cells (HUVECs) were treated with oxidized low-density lipoprotein (ox-LDL) and/or PRR gene silencing, and a Cell Counting Kit-8 (CCK-8) assay was performed to test their proliferation. The concentrations of inflammatory factors and oxidative stress factor were also detected using enzyme-linked immunosorbent assay and chemical colorimetry. Western blot analysis and reverse transcription-quantitative polymerase chain reaction were applied to detect protein and mRNA expression levels, respectively. The results revealed that HO-1 and renin precursor (Rep) were independent factors that affected hypertension complicated with depression. Serum HO-1 levels in patients with hypertension complicated with depression were significantly lower than that in hypertensive patients without depression, while Rep levels in patients with hypertension complicated with depression were significantly higher than that in hypertensive patients without depression. In HUVECs, ox-LDL reduced the cell proliferation in a dose-dependent manner, upregulated the expression of PRR gene and downregulated the expression of HO-1 gene. It was also observed that silencing of the PRR gene promoted the expression of the HO-1 gene. Furthermore, ox-LDL upregulated the inflammatory response and oxidative stress levels, while PRR gene silencing inhibited the ox-LDL-induced inflammatory factor and oxidative stress levels in HUVECs. Thus, regulating the expression levels of HO-1 and PRR to inhibit the oxidative stress and pro-inflammatory effect of ox-LDL may provide new insight for the treatment of hypertension patients with depression.
Collapse
Affiliation(s)
- Xin Gong
- Department of Integrated Chinese and Western Medicine, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Congyang Liu
- Department of Integrated Chinese and Western Medicine, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Hongling Wang
- Central Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Jinying Fan
- Department of Cardiology, Yantaishan Hospital, Yantai, Shandong 264000, P.R. China
| | - Cuihong Jiang
- Department of Integrated Chinese and Western Medicine, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Yong Zou
- Department of Integrated Chinese and Western Medicine, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| |
Collapse
|
20
|
Jin Y, Wang H. Naringenin Inhibit the Hydrogen Peroxide-Induced SH-SY5Y Cells Injury Through Nrf2/HO-1 Pathway. Neurotox Res 2019; 36:796-805. [DOI: 10.1007/s12640-019-00046-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 04/08/2019] [Accepted: 04/10/2019] [Indexed: 01/06/2023]
|
21
|
Abstract
Mild environmental stress might have beneficial effects in aging by activating maintenance and repair processes in cells and organs. These beneficial stress effects fit to the concept of hormesis. Prominent stressors acting in a hormetic way are physical exercises, fasting, cold and heat. This review will introduce some toxins, which have been found to induce hormetic responses in animal models of aging research. To highlight the molecular signature of these hormetic effects we will depict signaling pathways affected by low doses of toxins on cellular and organismic level. As prominent examples for signaling pathways involved in both aging processes as well as toxin responses, PI3K/Akt/mTOR- and AMPK-signal transduction will be described in more detail. Due to the striking overlap of signaling pathways mediating toxin induced responses and aging processes we propose considering the ability of low doses of toxins to slow down the rate of aging.
Collapse
|
22
|
Cen J, Zhao N, Huang WW, Liu L, Xie YY, Gan Y, Wang CJ, Ji BS. Polyamine analogue QMA attenuated ischemic injury in MCAO rats via ERK and Akt activated Nrf2/HO-1 signaling pathway. Eur J Pharmacol 2019; 844:165-174. [DOI: 10.1016/j.ejphar.2018.12.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 12/03/2018] [Accepted: 12/10/2018] [Indexed: 02/06/2023]
|
23
|
Cui B, Zhang S, Wang Y, Guo Y. Farrerol attenuates β-amyloid-induced oxidative stress and inflammation through Nrf2/Keap1 pathway in a microglia cell line. Biomed Pharmacother 2019; 109:112-119. [DOI: 10.1016/j.biopha.2018.10.053] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 10/10/2018] [Accepted: 10/10/2018] [Indexed: 12/19/2022] Open
|
24
|
Zahednasab H, Firouzi M, Kaboudanian-Ardestani S, Mojallal-Tabatabaei Z, Karampour S, Keyvani H. The protective effect of rifampicin on behavioral deficits, biochemical, and neuropathological changes in a cuprizone model of demyelination. Cytokine 2019; 113:417-426. [DOI: 10.1016/j.cyto.2018.10.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 09/16/2018] [Accepted: 10/18/2018] [Indexed: 12/19/2022]
|
25
|
The sinister face of heme oxygenase-1 in brain aging and disease. Prog Neurobiol 2019; 172:40-70. [DOI: 10.1016/j.pneurobio.2018.06.008] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 06/19/2018] [Accepted: 06/30/2018] [Indexed: 11/23/2022]
|
26
|
Luu BE, Storey KB. Solving Donor Organ Shortage with Insights from Freeze Tolerance in Nature. Bioessays 2018; 40:e1800092. [DOI: 10.1002/bies.201800092] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 08/02/2018] [Indexed: 01/06/2023]
Affiliation(s)
- Bryan E. Luu
- Institute of Biochemistry and Department of BiologyCarleton University1125 Colonel By DriveOttawaON, K1S 5B6
| | - Kenneth B. Storey
- Institute of Biochemistry and Department of BiologyCarleton University1125 Colonel By DriveOttawaON, K1S 5B6
| |
Collapse
|
27
|
Heme Oxygenase 1 in the Nervous System: Does It Favor Neuronal Cell Survival or Induce Neurodegeneration? Int J Mol Sci 2018; 19:ijms19082260. [PMID: 30071692 PMCID: PMC6121636 DOI: 10.3390/ijms19082260] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 07/25/2018] [Accepted: 07/30/2018] [Indexed: 02/08/2023] Open
Abstract
Heme oxygenase 1 (HO-1) up-regulation is recognized as a pivotal mechanism of cell adaptation to stress. Under control of different transcription factors but with a prominent role played by Nrf2, HO-1 induction is crucial also in nervous system response to damage. However, several lines of evidence have highlighted that HO-1 expression is associated to neuronal damage and neurodegeneration especially in Alzheimer’s and Parkinson’s diseases. In this review, we summarize the current literature regarding the role of HO-1 in nervous system pointing out different molecular mechanisms possibly responsible for HO-1 up-regulation in nervous system homeostasis and neurodegeneration.
Collapse
|
28
|
Ye F, Li X, Liu Y, Chang W, Liu W, Yuan J, Chen J. Hemin provides protection against lead neurotoxicity through heme oxygenase 1/carbon monoxide activation. J Appl Toxicol 2018; 38:1353-1364. [PMID: 29797346 DOI: 10.1002/jat.3646] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/23/2018] [Accepted: 04/17/2018] [Indexed: 12/19/2022]
Abstract
The neurotoxicity of lead (Pb) is well established, and oxidative stress is strongly associated with Pb-induced neurotoxicity. Heme oxygenase 1 (HO-1) is an important antioxidative enzyme for protection against oxidative stress in many disease models. In this study, we applied hemin, the substrate and a well-known inducer of HO-1, to investigate the possible role of HO-1 in protecting against Pb neurotoxicity. Hemin can significantly attenuate Pb acetate-induced cell death and oxidative stress in the hippocampus and frontal cortex of developmental rats. Consistent with in vivo results, the protective effects of hemin were also observed in SH-SY5Y cells after inducing cell survival and maintaining redox balance. However, knocking down HO-1 could significantly abolish the cytoprotective action of hemin against Pb toxicity, confirming HO-1 contributed to the protection. Finally, the HO-1-derived production of carbon monoxide, but not of bilirubin or Fe2+ , mediated the protective effects of HO-1 activation induced by hemin treatment against Pb-induced cell death and oxidative stress in SHSY5Y cells. Overall, this study showed that hemin provided protection against Pb neurotoxicity by HO-1/carbon monoxide activation.
Collapse
Affiliation(s)
- Fang Ye
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.,Ministry of Education Key Lab for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xiaoyi Li
- Center for Translational Medicine, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yawen Liu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.,Ministry of Education Key Lab for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Wei Chang
- Department of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, People's Republic of China
| | - Wenqi Liu
- Department of Parasitology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Jing Yuan
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.,Ministry of Education Key Lab for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Jun Chen
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.,Ministry of Education Key Lab for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
29
|
Candida albicans β-Glucan-Containing Particles Increase HO-1 Expression in Oral Keratinocytes via a Reactive Oxygen Species/p38 Mitogen-Activated Protein Kinase/Nrf2 Pathway. Infect Immun 2018; 86:IAI.00575-17. [PMID: 29311246 DOI: 10.1128/iai.00575-17] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 01/02/2018] [Indexed: 12/20/2022] Open
Abstract
Oral keratinocytes provide the first line of host defense against oral candidiasis. We speculated that interactions of fungal cell wall components with oral keratinocytes regulate the stress response against Candida infection and examined the expression of genes induced by heat-killed Candida albicans in oral immortalized keratinocytes using a cDNA microarray technique. Of 24,000 genes revealed by that analysis, we focused on HO-1, a stress-inducible gene, as its expression was increased by both heat-killed and live C. albicans In histological findings, HO-1 expression in the superficial layers of the oral epithelium following Candida infection was elevated compared to that in healthy epithelium. We then investigated fungal cell wall components involved in induction of HO-1 expression and found that β-glucan-containing particles (β-GPs) increased its expression. Furthermore, β-glucan was observed on the surface of both heat-killed C. albicans and Candida cells that had invaded the oral epithelium. Fungal β-GPs also promoted induction of intracellular reactive oxygen species (ROS), NADPH oxidase activation, and p38 mitogen-activated protein kinase (MAPK) phosphorylation, while those specific inhibitors inhibited the HO-1 expression induced by fungal β-GPs. Moreover, fungal β-GPs induced Nrf2 translocation into nuclei via p38 MAPK signaling, while the HO-1 expression induced by fungal β-GPs was inhibited by Nrf2-specific small interfering RNA (siRNA). Finally, knockdown of cells by HO-1- and Nrf2-specific siRNAs resulted in increased β-GP-mediated ROS production compared to that in the control cells. Our results show that the HO-1 induced by fungal β-GPs via ROS/p38 MAPK/Nrf2 from oral keratinocytes may have important roles in host defense against the stress caused by Candida infection in the oral epithelium.
Collapse
|
30
|
Brum AM, van de Peppel J, Nguyen L, Aliev A, Schreuders-Koedam M, Gajadien T, van der Leije CS, van Kerkwijk A, Eijken M, van Leeuwen JPTM, van der Eerden BCJ. Using the Connectivity Map to discover compounds influencing human osteoblast differentiation. J Cell Physiol 2018; 233:4895-4906. [PMID: 29194609 DOI: 10.1002/jcp.26298] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 11/28/2017] [Indexed: 12/24/2022]
Abstract
Osteoporosis is a common skeletal disorder characterized by low bone mass leading to increased bone fragility and fracture susceptibility. Identification of factors influencing osteoblast differentiation and bone formation is very important. Previously, we identified parbendazole to be a novel compound that stimulates osteogenic differentiation of human mesenchymal stromal cells (hMSCs), using gene expression profiling and bioinformatic analyzes, including the Connectivity Map (CMap), as an in-silico approach. The aim for this paper is to identify additional compounds affecting osteoblast differentiation using the CMap. Gene expression profiling was performed on hMSCs differentiated to osteoblasts using Illumina microarrays. Our osteoblast gene signature, the top regulated genes 6 hr after induction by dexamethasone, was uploaded into CMap (www.broadinstitute.org/cmap/). Through this approach we identified compounds with gene signatures positively correlating (withaferin-A, calcium folinate, amylocaine) or negatively correlating (salbutamol, metaraminol, diprophylline) to our osteoblast gene signature. All positively correlating compounds stimulated osteogenic differentiation, as indicated by increased mineralization compared to control treated cells. One of three negatively correlating compounds, salbutamol, inhibited dexamethasone-induced osteoblastic differentiation, while the other two had no effect. Based on gene expression data of withaferin-A and salbutamol, we identified HMOX1 and STC1 as being strongly differentially expressed . shRNA knockdown of HMOX1 or STC1 in hMSCs inhibited osteoblast differentiation. These results confirm that the CMap is a powerful approach to identify positively compounds that stimulate osteogenesis of hMSCs, and through this approach we can identify genes that play an important role in osteoblast differentiation and could be targets for novel bone anabolic therapies.
Collapse
Affiliation(s)
- Andrea M Brum
- Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Jeroen van de Peppel
- Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Linh Nguyen
- Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Abidin Aliev
- Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | | | - Tarini Gajadien
- Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | | | | | | | | | - B C J van der Eerden
- Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| |
Collapse
|
31
|
Nway NC, Fujitani Y, Hirano S, Mar O, Win-Shwe TT. Role of TLR4 in olfactory-based spatial learning activity of neonatal mice after developmental exposure to diesel exhaust origin secondary organic aerosol. Neurotoxicology 2017; 63:155-165. [DOI: 10.1016/j.neuro.2017.10.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 09/27/2017] [Accepted: 10/03/2017] [Indexed: 01/24/2023]
|
32
|
Sheng XJ, Tu HJ, Chien WL, Kang KH, Lu DH, Liou HH, Lee MJ, Fu WM. Antagonism of proteasome inhibitor-induced heme oxygenase-1 expression by PINK1 mutation. PLoS One 2017; 12:e0183076. [PMID: 28806787 PMCID: PMC5555616 DOI: 10.1371/journal.pone.0183076] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 07/29/2017] [Indexed: 11/25/2022] Open
Abstract
PTEN-induced putative kinase 1 (PINK1) is an integral protein in the mitochondrial membrane and maintains mitochondrial fidelity. Pathogenic mutations in PINK1 have been identified as a cause of early-onset autosomal recessive familial Parkinson’s disease (PD). The ubiquitin proteasome pathway is associated with neurodegenerative diseases. In this study, we investigated whether mutations of PINK1 affects the cellular stress response following proteasome inhibition. Administration of MG132, a peptide aldehyde proteasome inhibitor, significantly increased the expression of heme oxygenase-1 (HO-1) in rat dopaminergic neurons in the substantia nigra and in the SH-SY5Y neuronal cell line. The induction of HO-1 expression by proteasome inhibition was reduced in PINK1 G309D mutant cells. MG132 increased the levels of HO-1 through the Akt, p38, and Nrf2 signaling pathways. Compared with the cells expressing WT-PINK1, the phosphorylation of Akt and p38 was lower in those cells expressing the PINK1 G309D mutant, which resulted in the inhibition of the nuclear translocation of Nrf2. Furthermore, MG132-induced neuronal death was enhanced by the PINK1 G309D mutation. In this study, we demonstrated that the G309D mutation impairs the neuroprotective function of PINK1 following proteasome inhibition, which may be related to the pathogenesis of PD.
Collapse
Affiliation(s)
- Xiang-Jun Sheng
- Pharmacological Institute, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hunag-Ju Tu
- Pharmacological Institute, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wei-Lin Chien
- Pharmacological Institute, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kai-Hsiang Kang
- Pharmacological Institute, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Dai-Hua Lu
- Pharmacological Institute, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Horng-Huei Liou
- Pharmacological Institute, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ming-Jen Lee
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
- * E-mail: (WF); (ML)
| | - Wen-Mei Fu
- Pharmacological Institute, College of Medicine, National Taiwan University, Taipei, Taiwan
- * E-mail: (WF); (ML)
| |
Collapse
|
33
|
Belhaj A, Dewachter L, Rorive S, Remmelink M, Weynand B, Melot C, Hupkens E, Dewachter C, Creteur J, Mc Entee K, Naeije R, Rondelet B. Mechanical versus humoral determinants of brain death-induced lung injury. PLoS One 2017; 12:e0181899. [PMID: 28753621 PMCID: PMC5533440 DOI: 10.1371/journal.pone.0181899] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 07/10/2017] [Indexed: 12/29/2022] Open
Abstract
Background The mechanisms of brain death (BD)-induced lung injury remain incompletely understood, as uncertainties persist about time-course and relative importance of mechanical and humoral perturbations. Methods Brain death was induced by slow intracranial blood infusion in anesthetized pigs after randomization to placebo (n = 11) or to methylprednisolone (n = 8) to inhibit the expression of pro-inflammatory mediators. Pulmonary artery pressure (PAP), wedged PAP (PAWP), pulmonary vascular resistance (PVR) and effective pulmonary capillary pressure (PCP) were measured 1 and 5 hours after Cushing reflex. Lung tissue was sampled to determine gene expressions of cytokines and oxidative stress molecules, and pathologically score lung injury. Results Intracranial hypertension caused a transient increase in blood pressure followed, after brain death was diagnosed, by persistent increases in PAP, PCP and the venous component of PVR, while PAWP did not change. Arterial PO2/fraction of inspired O2 (PaO2/FiO2) decreased. Brain death was associated with an accumulation of neutrophils and an increased apoptotic rate in lung tissue together with increased pro-inflammatory interleukin (IL)-6/IL-10 ratio and increased heme oxygenase(HO)-1 and hypoxia inducible factor(HIF)-1 alpha expression. Blood expressions of IL-6 and IL-1β were also increased. Methylprednisolone pre-treatment was associated with a blunting of increased PCP and PVR venous component, which returned to baseline 5 hours after BD, and partially corrected lung tissue biological perturbations. PaO2/FiO2 was inversely correlated to PCP and lung injury score. Conclusions Brain death-induced lung injury may be best explained by an initial excessive increase in pulmonary capillary pressure with increased pulmonary venous resistance, and was associated with lung activation of inflammatory apoptotic processes which were partially prevented by methylprednisolone.
Collapse
Affiliation(s)
- Asmae Belhaj
- Department of Cardio-Vascular, Thoracic Surgery and Lung Transplantation, CHU UcL Namur, Université Catholique de Louvain, Yvoir, Belgium
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
- * E-mail: ,
| | - Laurence Dewachter
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Sandrine Rorive
- Department of Anatomopathology, Erasmus Academic Hospital, Brussels, Belgium
- DIAPATH—Center for Microscopy and Molecular Imaging (CMMI), Gosselies, Belgium
| | - Myriam Remmelink
- Department of Anatomopathology, Erasmus Academic Hospital, Brussels, Belgium
| | - Birgit Weynand
- Department of Anatomopathology, UZ Leuven, Katholiek Universiteit Leuven, Brussels, Belgium
| | - Christian Melot
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
- Department of Emergency, Erasmus Academic Hospital, Brussels, Belgium
| | - Emeline Hupkens
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Céline Dewachter
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Jacques Creteur
- Department of Intensive Care, Erasmus Academic Hospital, Brussels, Belgium
| | - Kathleen Mc Entee
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Robert Naeije
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Benoît Rondelet
- Department of Cardio-Vascular, Thoracic Surgery and Lung Transplantation, CHU UcL Namur, Université Catholique de Louvain, Yvoir, Belgium
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
34
|
Wang S, Zhang T, Yang Z, Lin J, Cai B, Ke Q, Lan W, Shi J, Wu S, Lin W. Heme oxygenase-1 protects spinal cord neurons from hydrogen peroxide-induced apoptosis via suppression of Cdc42/MLK3/MKK7/JNK3 signaling. Apoptosis 2017; 22:449-462. [PMID: 27864650 DOI: 10.1007/s10495-016-1329-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The mechanisms by which oxidative stress induces spinal cord neuron death has not been completely understood. Investigation on the molecular signal pathways involved in oxidative stress-mediated neuronal death is important for development of new therapeutics for oxidative stress-associated spinal cord disorders. In current study we examined the role of heme oxygenase-1 (HO-1) in the modulation of MLK3/MKK7/JNK3 signaling, which is a pro-apoptotic pathway, after treating primary spinal cord neurons with H2O2. We found that MLK3/MKK7/JNK3 signaling was substantially activated by H2O2 in a time-dependent manner, demonstrated by increase of activating phosphorylation of MLK3, MKK7 and JNK3. H2O2 also induced expression of HO-1. Transduction of neurons with HO-1-expressing adeno-associated virus before H2O2 treatment introduced expression of exogenous HO-1 in neurons. Exogenous HO-1 reduced phosphorylation of MLK3, MKK7 and JNK3. Consistent with its inhibitory effect on MLK3/MKK7/JNK3 signaling, exogenous HO-1 decreased H2O2-induced neuronal apoptosis and necrosis. Furthermore, we found that exogenous HO-1 inhibited expression of Cdc42, which is crucial for MLK3 activation. In addition, HO-1-induced down-regulation of MLK3/MKK7/JNK3 signaling might be related to up-regulation of microRNA-137 (mir-137). A mir-137 inhibitor alleviated the inhibitory effect of HO-1 on JNK3 activation. This inhibitor also increased neuronal death even when exogenous HO-1 was expressed. Therefore, our study suggests a novel mechanism by which HO-1 exerted its neuroprotective efficacy on oxidative stress.
Collapse
Affiliation(s)
- Siyuan Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Fujian Medical University, 34 North Zhongshan Road, Quanzhou, 362000, China
| | - Tao Zhang
- Department of Orthopedic Surgery, The Second Hospital of Fuzhou Affiliated to Xiamen University, Fuzhou, 350007, China
| | - Zhen Yang
- Department of Orthopedic Surgery, The People's Hospital of Guizhou Province, Guiyang, 550002, China
| | - Jianhua Lin
- Department of Orthopedic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China
| | - Bin Cai
- Department of Neurology and Institute of Neurology, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China
| | - Qingfeng Ke
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Fujian Medical University, 34 North Zhongshan Road, Quanzhou, 362000, China
| | - Wenbin Lan
- Department of Orthopedic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350004, China
| | - Jinxing Shi
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Fujian Medical University, 34 North Zhongshan Road, Quanzhou, 362000, China
| | - Shiqiang Wu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Fujian Medical University, 34 North Zhongshan Road, Quanzhou, 362000, China
| | - Wenping Lin
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Fujian Medical University, 34 North Zhongshan Road, Quanzhou, 362000, China.
| |
Collapse
|
35
|
Liu L, Huang W, Wang J, Song H, Cen J, Ji B. Anthraquinone derivative exerted hormetic effect on the apoptosis in oxygen-glucose deprivation-induced PC12 cells via ERK and Akt activated Nrf2/HO-1 signaling pathway. Chem Biol Interact 2016; 262:1-11. [PMID: 27923643 DOI: 10.1016/j.cbi.2016.12.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 11/24/2016] [Accepted: 12/02/2016] [Indexed: 12/13/2022]
Abstract
There were accumulated evidences that agents may attenuate neurological disorders through a hormetic effect. This study was designed to investigate hormetic effect of BME on the oxygen-glucose deprivation (OGD)-induced mitochondrial apoptosis in NGF-differentiated PC12 cells. The effect of BME on the intracellular reactive oxygen species (iROS) formation and pro-survival signals mediated by ERK and Akt as well as transcription factor nuclear factor-erythroid 2 p45-related factor 2 (Nrf2) pathways was also determined. The present results showed that, at low concentrations, pretreatment with BME triggered stress response by causing ROS production, then, activated survival-promoting signals via ERK and Akt activated Nrf2/HO-1 signaling pathway, resulting in decrease in cytotoxicity induced by the OGD. It may be accepted that mild pretreatment with BME stimulated transient and moderate ROS production, but activated hormetic signals and induced stress responsive genes. In contrast, high concentrations of BME displayed toxic action due to massive ROS production. These results suggested that the effect of BME on the OGD-induced PC12 cells may be hormetic mechanism including induction of oxidative stress and subsequent activation of stress response gene expression.
Collapse
Affiliation(s)
- Lu Liu
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng 475001, People's Republic of China
| | - Weiwei Huang
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng 475001, People's Republic of China
| | - Jianhong Wang
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng 475001, People's Republic of China
| | - Huiling Song
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng 475001, People's Republic of China
| | - Juan Cen
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng 475001, People's Republic of China.
| | - Biansheng Ji
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng 475001, People's Republic of China.
| |
Collapse
|
36
|
Shah SA, Amin FU, Khan M, Abid MN, Rehman SU, Kim TH, Kim MW, Kim MO. Anthocyanins abrogate glutamate-induced AMPK activation, oxidative stress, neuroinflammation, and neurodegeneration in postnatal rat brain. J Neuroinflammation 2016; 13:286. [PMID: 27821173 PMCID: PMC5100309 DOI: 10.1186/s12974-016-0752-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 10/26/2016] [Indexed: 12/20/2022] Open
Abstract
Background Glutamate-induced excitotoxicity, oxidative damage, and neuroinflammation are believed to play an important role in the development of a number of CNS disorders. We recently reported that a high dose of glutamate could induce AMPK-mediated neurodegeneration in the postnatal day 7 (PND7) rat brain. Yet, the mechanism of glutamate-induced oxidative stress and neuroinflammation in the postnatal brain is not well understood. Here, we report for the first time the mechanism of glutamate-induced oxidative damage, neuroinflammation, and neuroprotection by polyphenolic anthocyanins in PND7. Methods PND7 rat brains, SH-SY5Y, and BV2 cells treated either alone with glutamate or in combination with anthocyanins and compound C were examined with Western blot and immunofluorescence techniques. Additionally, reactive oxygen species (ROS) assay and other ELISA kit assays were employed to know the therapeutic efficacy of anthocyanins against glutamate. Results A single injection of glutamate to developing rats significantly increased brain glutamate levels, activated and phosphorylated AMPK induction, and inhibited nuclear factor-E2-related factor 2 (Nrf2) after 2, 3, and 4 h in a time-dependent manner. In contrast, anthocyanin co-treatment significantly reduced glutamate-induced AMPK induction, ROS production, neuroinflammation, and neurodegeneration in the developing rat brain. Most importantly, anthocyanins increased glutathione (GSH and GSSG) levels and stimulated the endogenous antioxidant system, including Nrf2 and heme oxygenase-1 (HO-1), against glutamate-induced oxidative stress. Interestingly, blocking AMPK with compound C in young rats abolished glutamate-induced neurotoxicity. Similarly, all these experiments were replicated in SH-SY5Y cells by silencing AMPK with siRNA, which suggests that AMPK is the key mediator in glutamate-induced neurotoxicity. Conclusions Here, we report for the first time that anthocyanins can potentially decrease glutamate-induced neurotoxicity in young rats. Our work demonstrates that glutamate is toxic to the developing rat brain and that anthocyanins can minimize the severity of glutamate-induced neurotoxicity in an AMPK-dependent manner. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0752-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shahid Ali Shah
- Neuroscience Pioneer Research Center, Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Faiz Ul Amin
- Neuroscience Pioneer Research Center, Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Mehtab Khan
- Neuroscience Pioneer Research Center, Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Muhammad Noman Abid
- Neuroscience Pioneer Research Center, Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Shafiq Ur Rehman
- Neuroscience Pioneer Research Center, Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Tae Hyun Kim
- Neuroscience Pioneer Research Center, Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Min Woo Kim
- Neuroscience Pioneer Research Center, Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea
| | - Myeong Ok Kim
- Neuroscience Pioneer Research Center, Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, 660-701, Republic of Korea.
| |
Collapse
|
37
|
Ozkan A, Parlak H, Tanriover G, Dilmac S, Ulker SN, Birsen I, Agar A. The protective mechanism of docosahexaenoic acid in mouse model of Parkinson: The role of hemeoxygenase. Neurochem Int 2016; 101:S0197-0186(16)30159-0. [PMID: 27984168 DOI: 10.1016/j.neuint.2016.10.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 10/18/2016] [Accepted: 10/26/2016] [Indexed: 12/17/2022]
Abstract
Parkinson's disease (PD) is characterized by degeneration of the dopaminergic neurons in substantia nigra (SN). Its major clinical symptoms are tremor, rigidity, bradykinesia and postural instability. Docosahexaenoic acid (DHA) is an essential fatty acid for neural functions that resides within the neural membrane. A decline in fatty acid concentration is observed in case of neurodegenerative diseases such as PD. The present study aimed to explore the role of the heme oxygenase (HO) enzyme in protective effects of DHA administration in an experimental model of PD by using the neurotoxin 1-Methly-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Three-month old male C57BL/6 mice were randomly divided into 4 groups as Control, DHA-treated (DHA), MPTP-injected (MPTP) and DHA-treated + MPTP injected (DHA + MPTP). DHA was administered daily (36 mg kg-1 day-1) by gavage to DHA and DHA + MPTP groups for 30 days. On the 23rd day of DHA administration, MPTP was intraperitoneally injected at a dose of 4 × 20 mg kg-1 with 2-hr. intervals. Motor activities of mice were evaluated by pole test, locomotor activity and rotarod tests on the 7th day of the utilization of experimental Parkinson's model. Total brain tissues were used in immunohistochemical analysis of the tyrosine hydroxylase (TH) and Nuclear factor E2 related factor2 (Nrf2). SN tissues were extracted for biochemical analysis. HO-1 and HO-2 protein levels were detected by western blotting. Further, HO activity was measured by spectrophotometric assay. As an indicator of motor coordination and balance, the rotarod test at 40 rpm showed that MPTP-treated animals exhibited shorter time on the rotating rod mill, which was significantly increased by DHA treatment in DHA + MPTP group. The total locomotor activity, ambulatory movement and total distance were decreased in MPTP group, whereas they were improved upon DHA treatment. The results of the pole test indicating the intensity of the bradykinesia showed that the T-turn and T-total were increased in MPTP group, while DHA treatment significantly shortened both parameters. The number of TH-positive cells in SN was significantly reduced in MPTP group compared to the Control and DHA + MPTP groups. Also, immunoreactive Nrf2 levels were clearly increased in MPTP group compared to DHA + MPTP group. HO-1 expression level decreased in the DHA + MPTP group compared to MPTP group. The results of the present study indicated that DHA has protective effects on dopaminergic neurons in MPTP-induced experimental model of PD. In addition, the pathways of HO-1 and HO-2 might participate in this protective mechanism.
Collapse
Affiliation(s)
- Ayse Ozkan
- Akdeniz University, Faculty of Medicine, Department of Physiology, Antalya, Turkey.
| | - Hande Parlak
- Akdeniz University, Faculty of Medicine, Department of Physiology, Antalya, Turkey
| | - Gamze Tanriover
- Akdeniz University, Faculty of Medicine, Department of Histology and Embryology, Antalya, Turkey
| | - Sayra Dilmac
- Akdeniz University, Faculty of Medicine, Department of Histology and Embryology, Antalya, Turkey
| | | | - Ilknur Birsen
- Akdeniz University, Faculty of Medicine, Department of Physiology, Antalya, Turkey
| | - Aysel Agar
- Akdeniz University, Faculty of Medicine, Department of Physiology, Antalya, Turkey
| |
Collapse
|
38
|
Lee DS, Jeong GS. Butein provides neuroprotective and anti-neuroinflammatory effects through Nrf2/ARE-dependent haem oxygenase 1 expression by activating the PI3K/Akt pathway. Br J Pharmacol 2016; 173:2894-909. [PMID: 27465039 PMCID: PMC5055139 DOI: 10.1111/bph.13569] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 07/12/2016] [Accepted: 07/16/2016] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Butein, 3,4,2',4'-tetrahydroxychalcone, has various pharmacological effects. However, no study has demonstrated the specific neurobiological mechanisms of the effects of butein in neuronal cells. The present study examined the role of butein as an antioxidative and anti-inflammatory inducer of haem oxygenase 1 (HO1) in mouse hippocampal HT22, BV2 microglial and primary mouse hippocampus neurons. EXPERIMENTAL APPROACH We investigated the neuroprotective effects of butein on glutamate-induced HT22 cell and primary mouse hippocampal neuron death and its anti-neuroinflammatory effects on LPS-induced activation of BV2 cells. We elucidated the underlying mechanisms by assessing the involvement of NF-κB, HO1, nuclear factor-E2-related factor 2 (Nrf2) and Akt signalling. KEY RESULTS Butein decreased cellular oxidative injury and the production of ROS in glutamate-treated HT22 cells and primary mouse hippocampal neurons. Furthermore, butein suppressed LPS-induced pro-inflammatory enzymes and mediators in BV2 microglia. Butein inhibited IL-6, IL-1β and TNF-α production and mRNA expression. In addition, butein decreased NO and PGE2 production and inducible NOS and COX-2 expression through the NF-κB signalling pathway. Butein up-regulated Nrf2/ARE-mediated HO1 expression through the PI3K/Akt pathway and this was positively associated with its cytoprotective effects and anti-neuroinflammatory actions. CONCLUSION AND IMPLICATIONS Our results indicate that butein effectively prevents glutamate-induced oxidative damage and LPS-induced activation and that the induction of HO1 by butein through the PI3K/Akt pathway and Nrf2 activation appears to play a pivotal role in its effects on neuronal cells. Our results provide evidence for the neuroprotective properties of butein.
Collapse
Affiliation(s)
- Dong-Sung Lee
- College of Pharmacy, Chosun University, Gwangju, Korea
| | | |
Collapse
|
39
|
Sung HY, Choi BO, Jeong JH, Kong KA, Hwang J, Ahn JH. Amyloid Beta-Mediated Hypomethylation of Heme Oxygenase 1 Correlates with Cognitive Impairment in Alzheimer's Disease. PLoS One 2016; 11:e0153156. [PMID: 27058954 PMCID: PMC4825942 DOI: 10.1371/journal.pone.0153156] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 03/24/2016] [Indexed: 01/02/2023] Open
Abstract
To identify epigenetically regulated genes involved in the pathogenesis of Alzheimer’s disease (AD) we analyzed global mRNA expression and methylation profiles in amyloid precursor protein (APP)-Swedish mutant-expressing AD model cells, H4-sw and selected heme oxygenase-1 (HMOX1), which is associated with pathological features of AD such as neurofibrillary tangles and senile plaques. We examined the epigenetic regulatory mechanism of HMOX1 and its application as a diagnostic and prognostic biomarker for AD. Our results show that HMOX1 mRNA and protein expression was approximately 12.2-fold and 7.9-fold increased in H4-sw cells, respectively. Increased HMOX1 expression was also detected in the brain, particularly the hippocampus, of AD model transgenic mice. However, the methylation of specific CpG sites within its promoter, particularly at CpG located −374 was significantly decreased in H4-sw cells. Treatment of neuroglioma cells with the demethylating agent 5-aza-2′-deoxycytidine resulted in reduced methylation of HMOX1 promoter accompanied by enhanced HMOX1 expression strongly supporting DNA methylation-dependent transcriptional regulation of HMOX1. Toxic Aβ-induced aberrant hypomethylation of HMOX1 at −374 promoter CpG site was correlated with increased HMOX1expression. In addition to neuroglioma cells, we also found Aβ-induced epigenetic regulation of HMOX1 in human T lymphocyte Jurkat cells. We evaluated DNA methylation status of HMOX1 at −374 promoter CpG site in blood samples from AD patients, patients with mild cognitive impairment (MCI), and control individuals using quantitative methylation-specific polymerase chain reaction. We observed lower methylation of HMOX1 at the −374 promoter CpG site in AD patients compared to MCI and control individuals, and a correlation between Mini-Mental State Examination score and demethylation level. Receiver operating characteristics analysis revealed good discrimination of AD patients from MCI patients and control individuals. Our findings suggest that the methylation status of HMOX1 at a specific promoter CpG site is related to AD progression.
Collapse
Affiliation(s)
- Hye Youn Sung
- Department of Biochemistry, School of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Byung-Ok Choi
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jee Hyang Jeong
- Department of Neurology, Ewha Womans University Mokdong Hospital, Seoul, Republic of Korea
| | - Kyoung Ae Kong
- Clinical Trial Center, Ewha Womans University Medical Center, Seoul, Republic of Korea
| | - Jinha Hwang
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea
| | - Jung-Hyuck Ahn
- Department of Biochemistry, School of Medicine, Ewha Womans University, Seoul, Republic of Korea
- Department of Biochemistry, Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
40
|
Han LT, Fang Y, Cao Y, Wu FH, Liu E, Mo GY, Huang F. Triterpenoid saponin flaccidoside II from Anemone flaccida triggers apoptosis of NF1-associated malignant peripheral nerve sheath tumors via the MAPK-HO-1 pathway. Onco Targets Ther 2016; 9:1969-79. [PMID: 27103823 PMCID: PMC4827896 DOI: 10.2147/ott.s95597] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are highly aggressive soft tissue neoplasms that are extremely rare and are frequently associated with neurofibromatosis type 1 patients. MPNSTs are typically fatal, and there is no effective treatment so far. In our previous study, we showed that flaccidoside II, one of the triterpenoid saponins isolated from Anemone flaccida Fr. Schmidt, has antitumor potential by inducing apoptosis. In the present study, we found that flaccidoside II inhibits proliferation and facilitates apoptosis in MPNST cell lines ST88-14 and S462. Furthermore, this study provides a mechanism by which the downregulation of heme oxygenase-1 via extracellular signal-regulated kinase-1/2 and p38 mitogen-activated protein kinase pathways is involved in the apoptotic role of flaccidoside II. This study suggested the potential of flaccidoside II as a novel pharmacotherapeutic approach for MPNSTs.
Collapse
Affiliation(s)
- Lin-Tao Han
- China Key Laboratory of TCM Resource and Prescription, Ministry of Education, Wuhan, Hubei, People's Republic of China
| | - Yin Fang
- China Key Laboratory of TCM Resource and Prescription, Ministry of Education, Wuhan, Hubei, People's Republic of China
| | - Yan Cao
- Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, People's Republic of China
| | - Feng-Hua Wu
- China Key Laboratory of TCM Resource and Prescription, Ministry of Education, Wuhan, Hubei, People's Republic of China
| | - E Liu
- Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, People's Republic of China
| | - Guo-Yan Mo
- Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, People's Republic of China
| | - Fang Huang
- China Key Laboratory of TCM Resource and Prescription, Ministry of Education, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
41
|
Inhibition of Ectodermal-Neural Cortex 1 Protects Neural Cells from Apoptosis Induced by Hypoxia and Hypoglycemia. J Mol Neurosci 2016; 59:126-34. [DOI: 10.1007/s12031-016-0742-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 03/22/2016] [Indexed: 10/22/2022]
|
42
|
Jao HY, Hsu JD, Lee YR, Lo CS, Lee HJ. Mulberry water extract regulates the osteoblast/osteoclast balance in an ovariectomic rat model. Food Funct 2016; 7:4753-4763. [DOI: 10.1039/c6fo00852f] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mulberry has favorable antioxidant ability.
Collapse
Affiliation(s)
- Hsing-Yu Jao
- Institute of Biochemistry
- Microbiology and Immunology
- Medical College
- Chung Shan Medical University
- Taichung
| | - Jeng-Dong Hsu
- Department of Pathology
- Chung Shan Medical University Hospital
- Taichung
- Taiwan
| | - Yi-Ru Lee
- Institute of Biochemistry
- Microbiology and Immunology
- Medical College
- Chung Shan Medical University
- Taichung
| | - Chien-Sheng Lo
- Department of Orthopaediology
- Show Chwuan Memory Hospital
- Changhua
- Taiwan
| | - Huei-Jane Lee
- Institute of Biochemistry
- Microbiology and Immunology
- Medical College
- Chung Shan Medical University
- Taichung
| |
Collapse
|
43
|
Li L, Peng Y, Hui Y, Zhang S, Zhou Y, Li D, Li J, Si Z, Li J, Wang D, Li Y, Dong M, Gao X. Overexpression of Heme Oxygenase 1 Impairs Cognitive Ability and Changes the Plasticity of the Synapse. J Alzheimers Dis 2015; 47:595-608. [DOI: 10.3233/jad-150027] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Lisha Li
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China
- State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Harbin, China
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, China
| | - Yahui Peng
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China
- State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Harbin, China
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, China
| | - Yang Hui
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China
- State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Harbin, China
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, China
| | - Shuai Zhang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China
| | - You Zhou
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China
| | - Dan Li
- People’s Hospital of Yuxi City, Yuki, China
| | - Jihong Li
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China
- State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Harbin, China
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, China
| | - Zizhen Si
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China
| | - Jing Li
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China
| | - Dayong Wang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China
- State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Harbin, China
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, China
| | - Yanze Li
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China
- State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Harbin, China
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, China
| | - Min Dong
- GE Healthcare Life Sciences, Shanghai, China
| | - Xu Gao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China
- State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Harbin, China
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, China
| |
Collapse
|
44
|
Li BZ, Guo B, Zhang HY, Liu J, Tao SS, Pan HF, Ye DQ. Therapeutic potential of HO-1 in autoimmune diseases. Inflammation 2015; 37:1779-88. [PMID: 24818708 DOI: 10.1007/s10753-014-9908-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Heme oxygenase-1 (HO-1), the inducible isoform of heme oxygenase (HO), has raised a lot of concerns in recent years due to its multiple functions. HO-1 was found to be a pivotal cytoprotective, antioxidant, anti-apoptotic, immunosuppressive, as well as anti-inflammatory molecule. Recent studies have clarified its significant functions in many diseases with substantial findings. In autoimmune diseases, HO-1 may have promising therapeutic potential. Here, we briefly reviewed recent advances in this field, aiming at hopefully exploring the potential therapeutic roles of HO-1, and design HO-1-based strategies for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Bao-Zhu Li
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | | | | | | | | | | | | |
Collapse
|
45
|
Bobermin LD, Wartchow KM, Flores MP, Leite MC, Quincozes-Santos A, Gonçalves CA. Ammonia-induced oxidative damage in neurons is prevented by resveratrol and lipoic acid with participation of heme oxygenase 1. Neurotoxicology 2015; 49:28-35. [PMID: 26003724 DOI: 10.1016/j.neuro.2015.05.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 05/14/2015] [Accepted: 05/15/2015] [Indexed: 12/29/2022]
Abstract
Ammonia is a metabolite that, at high concentrations, is implicated in neurological disorders, such as hepatic encephalopathy (HE), which is associated with acute or chronic liver failure. Astrocytes are considered the primary target of ammonia toxicity in the central nervous system (CNS) because glutamine synthetase (GS), responsible for ammonia metabolism in CNS, is an astrocytic enzyme. Thus, neuronal dysfunction has been associated as secondary to astrocytic impairment. However, we demonstrated that ammonia can induce direct effects on neuronal cells. The cell viability was decreased by ammonia in SH-SY5Y cells and cerebellar granule neurons. In addition, ammonia induced increased reactive oxygen species (ROS) production and decreased GSH intracellular content, the main antioxidant in CNS. As ammonia neurotoxicity is strongly associated with oxidative stress, we also investigated the potential neuroprotective roles of the antioxidants, resveratrol (RSV) and lipoic acid (LA), against ammonia toxicity in cerebellar granule neurons. RSV and LA were able to prevent the oxidative damage induced by ammonia, maintaining the levels of ROS production and GSH close to basal values. Both antioxidants also decreased ROS production and increased GSH content under basal conditions (in the absence of ammonia). Moreover, we showed that heme oxygenase 1 (HO1), a protein associated with protection against stress conditions, is involved in the beneficial effects of RSV and LA in cerebellar granule neurons. Thus, this study reinforces the neuroprotective effects of RSV and LA. Although more studies in vivo are required, RSV and LA could represent interesting therapeutic strategies for the management of HE.
Collapse
Affiliation(s)
- Larissa Daniele Bobermin
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Krista Minéia Wartchow
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Marianne Pires Flores
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Marina Concli Leite
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - André Quincozes-Santos
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Carlos-Alberto Gonçalves
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
46
|
Kalaiselvan I, Samuthirapandi M, Govindaraju A, Sheeja Malar D, Kasi PD. Olive oil and its phenolic compounds (hydroxytyrosol and tyrosol) ameliorated TCDD-induced heptotoxicity in rats via inhibition of oxidative stress and apoptosis. PHARMACEUTICAL BIOLOGY 2015; 54:338-346. [PMID: 25955957 DOI: 10.3109/13880209.2015.1042980] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
CONTEXT Naturally occurring polyphenols including olive oil (OO) and its constituents hydroxytyrosol (HT) and tyrosol (TY), consumed in the Mediterranean diet, have shown to treat various ailments due to their remarkable antioxidant properties. OBJECTIVE The present study investigates the hepatoprotective effects of OO and its phenolic compounds HT and TY against TCDD-induced hepatotoxicity in male Wistar rats. MATERIALS AND METHODS TCDD was administered at a dose of 100 ng/kg p.o. for 20 d. Administration of OO (10 ml/kg; oral), HT (0.5 mg/kg; oral), and TY (30 mg/kg; i.p) was started 5 d prior to TCDD administration, and continued for 25 d with or without TCDD administration. At the end of the experiment (25 d), blood was taken for biochemical analyses and liver for the measurement of macromolecular damages, antioxidant status, expressions of CYP1A1, and apoptotic factors. RESULTS TCDD administration resulted in significant (p < 0.05) increase in the level of hepatic stress markers ALT (101.6 ± 3.07 IU/l), AST (295.0 ± 3.0 IU/l), and ALP (266.66 ± 3.7 IU/l). Also, biochemical analyses of liver reported elevation in nitrite and protein carbonyl content and depletion of NQO1 and HO. However, OO, HT, and TY restored the antioxidant status. Protein expressions by Western Blot technique showed an increase in the level of CYP1A1 and Bax and a decreased level of Bcl-2 on TCDD treatment, and vice versa on OO, HT, and TY treatment. DISCUSSION AND CONCLUSION Our work concludes that dietary supplementation of OO, HT, and TY could serve as a potential preventive drug for TCDD-induced hepatotoxicity.
Collapse
Affiliation(s)
- Ilavarasi Kalaiselvan
- a Department of Biotechnology , Alagappa University , Karaikudi , Tamil Nadu , India and
| | - Muniasamy Samuthirapandi
- b Department of Animal Science , Bharathidasan University , Tiruchirappalli , Tamil Nadu , India
| | - Archunan Govindaraju
- b Department of Animal Science , Bharathidasan University , Tiruchirappalli , Tamil Nadu , India
| | - Dicson Sheeja Malar
- a Department of Biotechnology , Alagappa University , Karaikudi , Tamil Nadu , India and
| | - Pandima Devi Kasi
- a Department of Biotechnology , Alagappa University , Karaikudi , Tamil Nadu , India and
| |
Collapse
|
47
|
A heme oxygenase-1 transducer model of degenerative and developmental brain disorders. Int J Mol Sci 2015; 16:5400-19. [PMID: 25761244 PMCID: PMC4394483 DOI: 10.3390/ijms16035400] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 01/28/2015] [Accepted: 02/22/2015] [Indexed: 12/17/2022] Open
Abstract
Heme oxygenase-1 (HO-1) is a 32 kDa protein which catalyzes the breakdown of heme to free iron, carbon monoxide and biliverdin. The Hmox1 promoter contains numerous consensus sequences that render the gene exquisitely sensitive to induction by diverse pro-oxidant and inflammatory stimuli. In “stressed” astroglia, HO-1 hyperactivity promotes mitochondrial iron sequestration and macroautophagy and may thereby contribute to the pathological iron deposition and bioenergetic failure documented in Alzheimer disease, Parkinson disease and certain neurodevelopmental conditions. Glial HO-1 expression may also impact neuroplasticity and cell survival by modulating brain sterol metabolism and the proteasomal degradation of neurotoxic proteins. The glial HO-1 response may represent a pivotal transducer of noxious environmental and endogenous stressors into patterns of neural damage and repair characteristic of many human degenerative and developmental CNS disorders.
Collapse
|
48
|
Han H, Cui W, Wang L, Xiong Y, Liu L, Sun X, Hao L. Lutein prevents high fat diet-induced atherosclerosis in ApoE-deficient mice by inhibiting NADPH oxidase and increasing PPAR expression. Lipids 2015; 50:261-73. [PMID: 25663235 DOI: 10.1007/s11745-015-3992-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 01/16/2015] [Indexed: 01/06/2023]
Abstract
Epidemiological and experimental studies provide supportive evidence that lutein, a major carotenoid, may act as a chemopreventive agent against atherosclerosis, although the underlying molecular mechanisms are not well understood. The main aim of this study was to investigate the effects of lutein on the alleviation of atherosclerosis and its molecular mechanisms involved in oxidative stress and lipid metabolism. Male apolipoprotein E knockout mice (n = 55) were fed either a normal chow diet or a high fat diet (HFD) supplemented with or without lutein for 24 weeks. The results showed that a HFD induced atherosclerosis formation, lipid metabolism disorders and oxidative stress, but noticeable improvements were observed in the lutein treated group. Additionally, lutein supplementation reversed the decreased protein expression of aortic heme oxygenase-1 and increased the mRNA and protein expressions of aortic nicotinamide-adenine dinucleotide phosphate oxidase stimulated by a HFD. Furthermore, the decreased mRNA and protein expression levels of hepatic peroxisome proliferator-activated receptor-α, carnitine palmitoyltransferase 1A, acyl CoA oxidase 1, low density lipoprotein receptors and scavenger receptor class B type I observed in mice with atherosclerosis were markedly enhanced after treatment with lutein. Taken together, these data add new evidence supporting the anti-atherogenic properties of lutein and describing its mechanisms of action in atherosclerosis prevention, including oxidative stress and lipid metabolism improvements.
Collapse
Affiliation(s)
- Hao Han
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
49
|
Suk Lee D, Kim BN, Lim S, Lee J, Kim J, Jeong JG, Kim S. Effective suppression of nitric oxide production by HX106N through transcriptional control of heme oxygenase-1. Exp Biol Med (Maywood) 2015; 240:1136-46. [PMID: 25605059 DOI: 10.1177/1535370214567612] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 11/25/2014] [Indexed: 11/16/2022] Open
Abstract
Heme oxygenase-1 (HO-1) has been suggested to be a key neuroprotective enzyme because of its widespread distribution in the brain as well as its strong antioxidative effects. HX106N, a water-soluble botanical formulation, has previously been demonstrated to prevent amyloid β-induced memory impairment and oxidative stress in mice by upregulating HO-1 levels. In this study, the underlying molecular mechanisms of HX106N-induced HO-1 expression were investigated using BV-2 cells, a murine microglial cell line, and primary microglia. Treatment with HX106N induced the expression of HO-1 at the transcriptional level through the stress-responsive element-containing enhancer present in the ho-1 promoter. Nuclear factor E2-related factor 2 (Nrf2) was activated in cells treated with HX106N. The results from knockdown assay showed that small interfering RNA of Nrf2 attenuated HX106N-mediated HO-1 expression. Pharmacological inhibitors of p38 and JNK mitogen-activated protein kinases suppressed the HX106N-mediated induction of HO-1. The NF-κB signaling pathway was activated by HX106N and played a role in HX106N-induced HO-1 expression. Furthermore, HO-1 and one of its by-products during the enzymatic degradation of heme, CO, were found to be involved in HX106N-mediated suppression of NO production. Taken together, these data indicate that HX106N exerts potent antioxidative effects by increasing the expression of HO-1 through multiple signaling pathways, leading to the suppression of NO production.
Collapse
Affiliation(s)
- Doo Suk Lee
- School of Biological Sciences, Seoul National University, Seoul 151-742, Korea ViroMed Co., Ltd., Seoul 151-747, Korea
| | - Binna N Kim
- Interdisciplinary Program in Neuroscience, College of Natural Sciences, Seoul National University, Seoul 151-742, Korea
| | - Seonung Lim
- School of Biological Sciences, Seoul National University, Seoul 151-742, Korea
| | - Junsub Lee
- School of Biological Sciences, Seoul National University, Seoul 151-742, Korea
| | - Jiyoung Kim
- School of Biological Sciences, Seoul National University, Seoul 151-742, Korea Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 151-742, Korea
| | | | - Sunyoung Kim
- School of Biological Sciences, Seoul National University, Seoul 151-742, Korea ViroMed Co., Ltd., Seoul 151-747, Korea
| |
Collapse
|
50
|
Hascalovici JR, Song W, Liberman A, Vaya J, Khatib S, Holcroft C, Laferla F, Schipper HM. Neural HO-1/sterol interactions in vivo: implications for Alzheimer's disease. Neuroscience 2014; 280:40-9. [PMID: 25218961 DOI: 10.1016/j.neuroscience.2014.09.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 08/29/2014] [Accepted: 09/02/2014] [Indexed: 10/24/2022]
Abstract
BACKGROUND Up-regulation of heme oxygenase-1 (HO-1) and altered cholesterol (CH) metabolism are characteristic of Alzheimer-diseased (AD) neural tissues. We previously provided evidence of significant HO-1/sterol interactions in vitro (cultured rat astroglia) and in post-mortem human AD brain (Religious Orders Study). METHODS The current experiments were designed to further delineate these interactions in vivo by comparing the behavior of HO-1/sterol interactions in two mouse models; (1) a novel HO-1 transgenic mouse (GFAP.HMOX1) engineered to selectively express human HO-1 in the astrocytic compartment and (2) the previously described triple transgenic AD mouse (3xTg-AD). In samples of frontal cortex, total CH, CH precursors and relevant oxysterols were quantified by gas chromatography-mass spectrometry (GC-MS) and HO-1 protein expression was assessed by ELISA. The relationships of HO-1 expression to total CH, CH precursors and total oxysterols were determined for both mouse models using linear regression analysis. RESULTS HO-1 expression is increased in GFAP.HMOX1 mice relative to wild type and in 11-12-month-old 3xTg-AD mice (with AD-like phenotype) relative to control mice and 5-6-month-old 3xTg-AD mice (no AD-like phenotype). Total oxysterols significantly decreased as HO-1 expression increased in GFAP.HMOX1 mice expressing high levels of HO-1, whereas total oxysterols increased as HO-1 expression increased in aged 3xTg-AD mice. Total CH and total CH precursors increased as HO-1 protein expression increased in 11-12-month-old 3xTg-AD mice relative to 5-6-month old 3xTg-AD mice. CONCLUSIONS Our findings indicate a differential impact of HO-1 on patterns of brain sterol and redox homeostasis that is contingent on the presence or absence of AD-like neuropathology. These data provide fresh insight concerning the regulation of sterol homeostasis within the aging and degenerating CNS which may inform the development of novel therapeutic and preventive strategies for the management of AD and related conditions.
Collapse
Affiliation(s)
- J R Hascalovici
- Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Cote Sainte Catherine Road, Montreal, Quebec H3T 1E2, Canada; Department of Neurology and Neurosurgery, McGill University, Jewish General Hospital, 3755 Cote Sainte Catherine Road, Montreal, Quebec H3T 1E2, Canada
| | - W Song
- Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Cote Sainte Catherine Road, Montreal, Quebec H3T 1E2, Canada
| | - A Liberman
- Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Cote Sainte Catherine Road, Montreal, Quebec H3T 1E2, Canada
| | - J Vaya
- Laboratory of Natural Medicinal Compounds, Migal-Galilee Technological Center and Tel Hai College, Kiryat-Shmona 11016, Israel
| | - S Khatib
- Laboratory of Natural Medicinal Compounds, Migal-Galilee Technological Center and Tel Hai College, Kiryat-Shmona 11016, Israel
| | - C Holcroft
- Centre for Clinical Epidemiology and Community Studies, Jewish General Hospital, 3755 Cote Sainte Catherine Road, Montreal, Quebec H3T 1E2, Canada
| | - F Laferla
- Department of Neurobiology and Behavior and Institute for Brain Aging and Dementia, University of California, Irvine, Irvine, CA 92697-4545, USA
| | - H M Schipper
- Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Cote Sainte Catherine Road, Montreal, Quebec H3T 1E2, Canada; Department of Neurology and Neurosurgery, McGill University, Jewish General Hospital, 3755 Cote Sainte Catherine Road, Montreal, Quebec H3T 1E2, Canada.
| |
Collapse
|