1
|
Liu C, Zhang W, Zhang H, Zhao C, Du X, Ren J, Qu X. Biomimetic engineering of a neuroinflammation-targeted MOF nanozyme scaffolded with photo-trigger released CO for the treatment of Alzheimer's disease. Chem Sci 2024; 15:13201-13208. [PMID: 39183930 PMCID: PMC11339965 DOI: 10.1039/d4sc02598a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/17/2024] [Indexed: 08/27/2024] Open
Abstract
Alzheimer's disease (AD) is one of the most fatal and irreversible neurodegenerative diseases, which causes a huge emotional and financial burden on families and society. Despite the progress made with recent clinical use of inhibitors of acetylcholinesterase and amyloid-β (Aβ) antibodies, the curative effects of AD treatment remain unsatisfactory, which is probably due to the complexity of pathogenesis and the multiplicity of therapeutic targets. Thus, modulating complex pathological networks could be an alternative approach to treat AD. Here, a neutrophil membrane-coated MOF nanozyme (denoted as Neu-MOF/Fla) is biomimetically engineered to disturb the malignant Aβ deposition-inflammation cycle and ameliorate the pathological network for effective AD treatment. Neu-MOF/Fla could recognize the pathological inflammatory signals of AD, and deliver the photo-triggered anti-inflammatory CO and MOF based hydrolytic nanozymes to the lesion area of the brain in a spontaneous manner. Based on the in vitro and in vivo studies, Neu-MOF/Fla significantly suppresses neuroinflammation, mitigates the Aβ burden, beneficially modulates the pro-inflammatory microglial phenotypes and improves the cognitive defects of AD mice models. Our work presents a good example for developing biomimetic multifunctional nanotherapeutics against AD by means of amelioration of multiple symptoms and improvement of cognitive defects.
Collapse
Affiliation(s)
- Chun Liu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230026 P. R. China
| | - Wenting Zhang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230026 P. R. China
| | - Haochen Zhang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230026 P. R. China
| | - Chuanqi Zhao
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230026 P. R. China
| | - Xiubo Du
- College of Life Sciences and Oceanography, Shenzhen Key Laboratory of Microbial Genetic Engineering, Shenzhen University Shenzhen 518060 China
| | - Jinsong Ren
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230026 P. R. China
| | - Xiaogang Qu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- University of Science and Technology of China Hefei Anhui 230026 P. R. China
| |
Collapse
|
2
|
Jin T, Lu H, Zhou Q, Chen D, Zeng Y, Shi J, Zhang Y, Wang X, Shen X, Cai X. H 2S-Releasing Versatile Montmorillonite Nanoformulation Trilogically Renovates the Gut Microenvironment for Inflammatory Bowel Disease Modulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308092. [PMID: 38308198 PMCID: PMC11005690 DOI: 10.1002/advs.202308092] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/20/2024] [Indexed: 02/04/2024]
Abstract
Abnormal activation of the intestinal mucosal immune system, resulting from damage to the intestinal mucosal barrier and extensive invasion by pathogens, contributes to the pathogenesis of inflammatory bowel disease (IBD). Current first-line treatments for IBD have limited efficacy and significant side effects. An innovative H2S-releasing montmorillonite nanoformulation (DPs@MMT) capable of remodeling intestinal mucosal immune homeostasis, repairing the mucosal barrier, and modulating gut microbiota is developed by electrostatically adsorbing diallyl trisulfide-loaded peptide dendrimer nanogels (DATS@PDNs, abbreviated as DPs) onto the montmorillonite (MMT) surface. Upon rectal administration, DPs@MMT specifically binds to and covers the damaged mucosa, promoting the accumulation and subsequent internalization of DPs by activated immune cells in the IBD site. DPs release H2S intracellularly in response to glutathione, initiating multiple therapeutic effects. In vitro and in vivo studies have shown that DPs@MMT effectively alleviates colitis by eliminating reactive oxygen species (ROS), inhibiting inflammation, repairing the mucosal barrier, and eradicating pathogens. RNA sequencing revealed that DPs@MMT exerts significant immunoregulatory and mucosal barrier repair effects, by activating pathways such as Nrf2/HO-1, PI3K-AKT, and RAS/MAPK/AP-1, and inhibiting the p38/ERK MAPK, p65 NF-κB, and JAK-STAT3 pathways, as well as glycolysis. 16S rRNA sequencing demonstrated that DPs@MMT remodels the gut microbiota by eliminating pathogens and increasing probiotics. This study develops a promising nanoformulation for IBD management.
Collapse
Affiliation(s)
- Ting Jin
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| | - Hongyang Lu
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| | - Qiang Zhou
- Department of OtolaryngologyRuian People's HospitalThe Third Affiliated Hospital of Wenzhou Medical UniversityWenzhou325016China
| | - Dongfan Chen
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| | - Youyun Zeng
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| | - Jiayi Shi
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| | - Yanmei Zhang
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| | - Xianwen Wang
- School of Biomedical EngineeringResearch and Engineering Center of Biomedical MaterialsAnhui Medical UniversityHefei230032China
| | - Xinkun Shen
- Department of OtolaryngologyRuian People's HospitalThe Third Affiliated Hospital of Wenzhou Medical UniversityWenzhou325016China
| | - Xiaojun Cai
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| |
Collapse
|
3
|
Buczyńska A, Sidorkiewicz I, Krętowski AJ, Zbucka-Krętowska M. The Role of Oxidative Stress in Trisomy 21 Phenotype. Cell Mol Neurobiol 2023; 43:3943-3963. [PMID: 37819608 PMCID: PMC10661812 DOI: 10.1007/s10571-023-01417-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 09/17/2023] [Indexed: 10/13/2023]
Abstract
Extensive research has been conducted to gain a deeper understanding of the deregulated metabolic pathways in the development of trisomy 21 (T21) or Down syndrome. This research has shed light on the hypothesis that oxidative stress plays a significant role in the manifestation of the T21 phenotype. Although in vivo studies have shown promising results in mitigating the detrimental effects of oxidative stress, there is currently a lack of introduced antioxidant treatment options targeting cognitive impairments associated with T21. To address this gap, a comprehensive literature review was conducted to provide an updated overview of the involvement of oxidative stress in T21. The review aimed to summarize the insights into the pathogenesis of the Down syndrome phenotype and present the findings of recent innovative research that focuses on improving cognitive function in T21 through various antioxidant interventions. By examining the existing literature, this research seeks to provide a holistic understanding of the role oxidative stress plays in the development of T21 and to explore novel approaches that target multiple aspects of antioxidant intervention to improve cognitive function in individuals with Down syndrome. The guides -base systematic review process (Hutton et al. 2015).
Collapse
Affiliation(s)
- Angelika Buczyńska
- Clinical Research Centre, Medical University of Białystok, ul. M. Skłodowskiej-Curie 24a, 15-276, Białystok, Poland.
| | - Iwona Sidorkiewicz
- Clinical Research Centre, Medical University of Białystok, ul. M. Skłodowskiej-Curie 24a, 15-276, Białystok, Poland
| | - Adam Jacek Krętowski
- Clinical Research Centre, Medical University of Białystok, ul. M. Skłodowskiej-Curie 24a, 15-276, Białystok, Poland
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Białystok, ul. Sklodowskiej-Curie 24a, 15-276, Białystok, Poland
| | - Monika Zbucka-Krętowska
- Department of Gynecological Endocrinology and Adolescent Gynecology, Medical University of Białystok, ul. M. Skłodowskiej-Curie 24a, 15-276, Białystok, Poland.
| |
Collapse
|
4
|
Sara Salahuddin H, Attaullah S, Ali Shah S, Khan S, Zahid M, Ullah M, Khayyam, Salahuddin S, Gul S, Alsugoor MH. Ranuncoside's attenuation of scopolamine-induced memory impairment in mice via Nrf2 and NF-ĸB signaling. Saudi Pharm J 2023; 31:101702. [PMID: 37533493 PMCID: PMC10391653 DOI: 10.1016/j.jsps.2023.101702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 07/07/2023] [Indexed: 08/04/2023] Open
Abstract
Scopolamine is a well-known pharmacological agent responsible for causing memory impairment in animals, as well as oxidative stress and neuroinflammation inducer which lead to the development of Alzheimer disease. Although a cure for Alzheimer's disease is unavailable. Ranuncoside, a metabolite obtained from a medicinal plant has demonstrated antioxidant and anti-inflammatory properties in vitro, making it a promising treatment with potential anti-Alzheimer disease properties. However, as ranuncoside has not been evaluated for its antioxidant and anti-neuroinflammatory properties in any in vivo model, our study aimed to evaluate its neurotherapeutic efficacy against scopolamine-induced memory impairment in adult male albino mice. Mice were randomly divided into four experimental groups. Mice of group I was injected with saline, group II was injected with scopolamine (1 mg/kg/day) for 3 weeks. After receiving a daily injection of scopolamine for 1 week, the mice of group III were injected with ranuncoside (10 mg/kg) every other day for 2 weeks along with scopolamine daily and group IV were injected with ranuncoside on 5th alternate days. Behavioral tests (i.e., Morris water maze and Y-maze) were performed to determine the memory-enhancing effect of ranuncoside against scopolamine's memory deleterious effect. Western blot analysis was also performed to further elucidate the anti-neuroinflammatory and antioxidant effects of ranuncoside against scopolamine-induced neuroinflammation and oxidative stress. Our results showed memory-enhancing, anti-neuroinflammatory effect, and antioxidant effects of ranuncoside against scopolamine by increasing the expression of the endogenous antioxidant system (i.e., Nrf2 and HO-1), followed by blocking neuroinflammatory markers such as NF-κB, COX-2, and TNF-α. The results also revealed that ranuncoside possesses hypoglycemic and hypolipidemic effects against scopolamine-induced hyperglycemia and hyperlipidemia in mice as well as scopolamine's hyperglycemic effect. In conclusion, our findings suggest that ranuncoside could be a potential agent for the management of Alzheimer's disease, hyperglycemia, and hyperlipidemia.
Collapse
Affiliation(s)
| | - Sobia Attaullah
- Department of Zoology, Islamia College, Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Shahid Ali Shah
- Neuro Molecular Medicine Research Centre (NMMRC), Ring Road, Peshawar, KPK, Pakistan
- The University of Haripur, KPK, Pakistan
| | - SanaUllah Khan
- Department of Zoology, University of Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Muhammad Zahid
- Department of Zoology, Islamia College, Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Mujeeb Ullah
- Department of Zoology, Islamia College, Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Khayyam
- Department of Zoology, Islamia College, Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Sidra Salahuddin
- Hayatabad Medical Complex, Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Seema Gul
- Department of Zoology, Islamia College, Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Mahdi H Alsugoor
- Department of Emergency Medical Services, College of Health Sciences-AlQunfudah, Umm Al- Qura University, Makkah 21912, Saudi Arabia
| |
Collapse
|
5
|
Wang L, Dan Q, Xu B, Chen Y, Zheng T. Research progress on gas signal molecular therapy for Parkinson's disease. Open Life Sci 2023; 18:20220658. [PMID: 37588999 PMCID: PMC10426759 DOI: 10.1515/biol-2022-0658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/22/2023] [Accepted: 06/14/2023] [Indexed: 08/18/2023] Open
Abstract
The pathogenesis of Parkinson's disease (PD) remains unclear. Among the pathological manifestations is the progressive degeneration of the nigrostriatal dopaminergic pathway, leading to massive loss of neurons in the substantia nigra pars compacta and dopamine (DA) depletion. Therefore, the current drug treatment is primarily based on DA supplementation and delaying the progression of the disease. However, as patients' symptoms continue to worsen, the drug effect will gradually decrease or even disappear, thereby further aggravating clinical symptoms. Gas signaling molecules, such as hydrogen sulfide (H2S), nitric oxide (NO), carbon monoxide (CO), and hydrogen (H2), exhibit pleiotropic biological functions and play crucial roles in physiological and pathological effects. In common neurodegenerative diseases including Alzheimer's disease and PD, gas signal molecules can prevent or delay disease occurrence via the primary mechanisms of antioxidation, anti-inflammatory response, and antiapoptosis. This article reviews the therapeutic progress of gas signaling molecules in PD models and discusses the possibility of their clinical applications.
Collapse
Affiliation(s)
- Linlin Wang
- Department of Hubei University of Medicine, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen518036, P. R. China
| | - Qing Dan
- Department of Hubei University of Medicine, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen518036, P. R. China
| | - Bingxuan Xu
- Department of Hubei University of Medicine, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen518036, P. R. China
| | - Yun Chen
- Department of Hubei University of Medicine, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen518036, P. R. China
| | - Tingting Zheng
- Department of Hubei University of Medicine, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen518036, P. R. China
| |
Collapse
|
6
|
da Silva A, Lepetre-Mouelhi S, Couvreur P. Micro- and nanocarriers for pain alleviation. Adv Drug Deliv Rev 2022; 187:114359. [PMID: 35654211 DOI: 10.1016/j.addr.2022.114359] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/10/2022] [Accepted: 05/20/2022] [Indexed: 12/28/2022]
Abstract
Acute or chronic pain is a major source of impairment in quality of life and affects a substantial part of the population. To date, pain is alleviated by a limited range of treatments with significant toxicity, increased risk of misuse and inconsistent efficacy, owing, in part, to lack of specificity and/or unfavorable pharmacokinetic properties. Thanks to the unique properties of nanoscaled drug carriers, nanomedicine may enhance drug biodistribution and targeting, thus contributing to improved bioavailability and lower off-target toxicity. After a brief overview of the current situation and the main critical issues regarding pain alleviation, this review will examine the most advanced approaches using nanomedicine of each drug class, from the preclinical stage to approved nanomedicines.
Collapse
|
7
|
Erekat NS. Apoptosis and its therapeutic implications in neurodegenerative diseases. Clin Anat 2021; 35:65-78. [PMID: 34558138 DOI: 10.1002/ca.23792] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/19/2021] [Accepted: 09/21/2021] [Indexed: 12/18/2022]
Abstract
Neurodegenerative disorders are characterized by progressive loss of particular populations of neurons. Apoptosis has been implicated in the pathogenesis of neurodegenerative diseases, including Parkinson disease, Alzheimer disease, Huntington disease, and amyotrophic lateral sclerosis. In this review, we focus on the existing notions relevant to comprehending the apoptotic death process, including the morphological features, mediators and regulators of cellular apoptosis. We also highlight the evidence of neuronal apoptotic death in Parkinson disease, Alzheimer disease, Huntington disease, and amyotrophic lateral sclerosis. Additionally, we present evidence of potential therapeutic agents that could modify the apoptotic pathway in the aforementioned neurodegenerative diseases and delay disease progression. Finally, we review the clinical trials that were conducted to evaluate the use of anti-apoptotic drugs in the treatment of the aforementioned neurodegenerative diseases, in order to highlight the essential need for early detection and intervention of neurodegenerative diseases in humans.
Collapse
Affiliation(s)
- Nour S Erekat
- Department of Anatomy, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
8
|
Park HR, Yang EJ. Oxidative Stress as a Therapeutic Target in Amyotrophic Lateral Sclerosis: Opportunities and Limitations. Diagnostics (Basel) 2021; 11:diagnostics11091546. [PMID: 34573888 PMCID: PMC8465946 DOI: 10.3390/diagnostics11091546] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/14/2021] [Accepted: 08/25/2021] [Indexed: 12/20/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS), also known as motor neuron disease (MND) and Lou Gehrig’s disease, is characterized by a loss of the lower motor neurons in the spinal cord and the upper motor neurons in the cerebral cortex. Due to the complex and multifactorial nature of the various risk factors and mechanisms that are related to motor neuronal degeneration, the pathological mechanisms of ALS are not fully understood. Oxidative stress is one of the known causes of ALS pathogenesis. This has been observed in patients as well as in cellular and animal models, and is known to induce mitochondrial dysfunction and the loss of motor neurons. Numerous therapeutic agents have been developed to inhibit oxidative stress and neuroinflammation. In this review, we describe the role of oxidative stress in ALS pathogenesis, and discuss several anti-inflammatory and anti-oxidative agents as potential therapeutics for ALS. Although oxidative stress and antioxidant fields are meaningful approaches to delay disease progression and prolong the survival in ALS, it is necessary to investigate various animal models or humans with different subtypes of sporadic and familial ALS.
Collapse
|
9
|
Pervin Z, Stephen JM. Effect of alcohol on the central nervous system to develop neurological disorder: pathophysiological and lifestyle modulation can be potential therapeutic options for alcohol-induced neurotoxication. AIMS Neurosci 2021; 8:390-413. [PMID: 34183988 PMCID: PMC8222771 DOI: 10.3934/neuroscience.2021021] [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: 01/20/2021] [Accepted: 03/01/2021] [Indexed: 12/06/2022] Open
Abstract
The central nervous system (CNS) is the major target for adverse effects of alcohol and extensively promotes the development of a significant number of neurological diseases such as stroke, brain tumor, multiple sclerosis (MS), Alzheimer's disease (AD), and amyotrophic lateral sclerosis (ALS). Excessive alcohol consumption causes severe neuro-immunological changes in the internal organs including irreversible brain injury and it also reacts with the defense mechanism of the blood-brain barrier (BBB) which in turn leads to changes in the configuration of the tight junction of endothelial cells and white matter thickness of the brain. Neuronal injury associated with malnutrition and oxidative stress-related BBB dysfunction may cause neuronal degeneration and demyelination in patients with alcohol use disorder (AUD); however, the underlying mechanism still remains unknown. To address this question, studies need to be performed on the contributing mechanisms of alcohol on pathological relationships of neurodegeneration that cause permanent neuronal damage. Moreover, alcohol-induced molecular changes of white matter with conduction disturbance in neurotransmission are a likely cause of myelin defect or axonal loss which correlates with cognitive dysfunctions in AUD. To extend our current knowledge in developing a neuroprotective environment, we need to explore the pathophysiology of ethanol (EtOH) metabolism and its effect on the CNS. Recent epidemiological studies and experimental animal research have revealed the association between excessive alcohol consumption and neurodegeneration. This review supports an interdisciplinary treatment protocol to protect the nervous system and to improve the cognitive outcomes of patients who suffer from alcohol-related neurodegeneration as well as clarify the pathological involvement of alcohol in causing other major neurological disorders.
Collapse
Affiliation(s)
- Zinia Pervin
- Department of Biomedical Engineering, University of New Mexico, Albuquerque, NM 87131, USA
| | - Julia M Stephen
- The Mind Research Network and Lovelace Biomedical and Environmental Research Institute, Albuquerque, NM 87106, USA
| |
Collapse
|
10
|
Ibi D, Hirashima K, Kojima Y, Sumiya K, Kondo S, Yamamoto M, Ando T, Hiramatsu M. Preventive Effects of Continuous Betaine Intake on Cognitive Impairment and Aberrant Gene Expression in Hippocampus of 3xTg Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2020; 79:639-652. [PMID: 33337369 DOI: 10.3233/jad-200972] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND The deposition of amyloid-β (Aβ) and hyperphosphorylation of tau are well-known as the pathophysiological features of Alzheimer's disease (AD), leading to oxidative stress and synaptic deficits followed by cognitive symptoms. We already demonstrated that betaine (glycine betaine) prevented cognitive impairment and hippocampal oxidative stress in mice intracerebroventricularly injected with an active fragment of Aβ, whereas the effect of betaine in chronic models of AD remains unknown. OBJECTIVE Our objective was to investigate the effects of chronic betaine intake on cognitive impairment and aberrant expression of genes involved in synapse and antioxidant activity in the hippocampus of a genetic AD model. METHODS We performed cognitive tests and RT-PCR in the hippocampus in 3xTg mice, a genetic AD model. RESULTS Cognitive impairment in the Y-maze and novel object recognition tests became evident in 3xTg mice at 9 months old, and not earlier, indicating that cognitive impairment in 3xTg mice developed age-dependently. To examine the preventive effect of betaine on such cognitive impairment, 3xTg mice were fed betaine-containing water for 3 months from 6 to 9 months old, and subsequently subjected to behavioral tests, in which betaine intake prevented the development of cognitive impairment in 3xTg mice. Additionally, the expression levels of genes involved in synapse and antioxidant activity were downregulated in hippocampus of 3xTg mice at 9 months old compared with age-matched wild-type mice, which were suppressed by betaine intake. CONCLUSION Betaine may be applicable as an agent preventing the progression of AD by improving the synaptic structure/function and/or antioxidant activity.
Collapse
Affiliation(s)
- Daisuke Ibi
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Tenpaku-ku, Nagoya, Japan
| | - Kazuki Hirashima
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Tenpaku-ku, Nagoya, Japan
| | - Yuya Kojima
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Tenpaku-ku, Nagoya, Japan
| | - Kahori Sumiya
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Tenpaku-ku, Nagoya, Japan
| | - Sari Kondo
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Tenpaku-ku, Nagoya, Japan
| | - Mirai Yamamoto
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Tenpaku-ku, Nagoya, Japan
| | - Toshihiro Ando
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Tenpaku-ku, Nagoya, Japan
| | - Masayuki Hiramatsu
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Tenpaku-ku, Nagoya, Japan
| |
Collapse
|
11
|
Gao X, Jiang L, Gong Y, Chen X, Ying M, Zhu H, He Q, Yang B, Cao J. Stress granule: A promising target for cancer treatment. Br J Pharmacol 2019; 176:4421-4433. [PMID: 31301065 DOI: 10.1111/bph.14790] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/26/2019] [Accepted: 07/02/2019] [Indexed: 12/14/2022] Open
Abstract
Stress granules (SGs) are primarily composed of mRNAs that stall at translation initiation and usually appear in the cytoplasm under unusual physiological or pathological conditions such as hypoxia, oxidative stress, and viral infection. Recent studies have indicated that several components of SGs participate in tumourigenesis and cancer metastasis through tumour-associated signalling pathways as well as other mechanisms. Furthermore, some chemotherapy drugs have been reported to induce SGs. Thus, the roles of SGs in cancer treatment have attracted considerable interest. Importantly, disturbing the recruitment of SGs components or microtubule polymerization, as well as other strategies that can abolish SGs formation, is reported to inhibit tumour progression, suggesting that targeting SGs could be a promising strategy for cancer treatment. In this review, we summarize the relationship between SGs and cancer, as well as recent advances in targeting SGs, in the interest of providing new opportunities for cancer treatment.
Collapse
Affiliation(s)
- Xiaomeng Gao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Li Jiang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yanling Gong
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xiaobing Chen
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Meidan Ying
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Hong Zhu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ji Cao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
12
|
Yu H, Yuan B, Chu Q, Wang C, Bi H. Protective roles of isoastilbin against Alzheimer's disease via Nrf2‑mediated antioxidation and anti‑apoptosis. Int J Mol Med 2019; 43:1406-1416. [PMID: 30664148 PMCID: PMC6365075 DOI: 10.3892/ijmm.2019.4058] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 01/07/2019] [Indexed: 12/18/2022] Open
Abstract
By analyzing the L‑glutamic acid (L‑Glu)‑induced apoptosis of PC12 cells and an AlCl3 combined with D‑galactose (D‑gal)‑developed Alzheimer's disease (AD) mouse model, the protective effects of isoastilbin (IAB) against AD were systematically investigated in the present study. Pre‑incubation with IAB for 3 h prior to treatment with 25 mM L‑Glu decreased cell viability and inhibited apoptosis, suppressed the accumulation of intracellular reactive oxygen species, and restored mitochondrial membrane potential in PC12 cells induced by L‑Glu. In mice with AD, the reduced escape latency time in the water maze test, suppressed chronic movement in the center area of an open field test and enhanced ability to seek hidden food in a Y maze test indicated that abnormal behaviors had improved after 28 days of treatment with IAB. Furthermore, IAB reduced the deposition of amyloid β (Aβ) and the expression of phosphorylated‑Tau in the mouse brain and enhanced the serum levels of Aβ. IAB ameliorated the oxidative stress via modulating the levels of associated enzymes and improved the functioning of the central cholinergic system, as indicated by an increase in acetylcholine and choline acetyltransferase concentrations. The expression levels of acetylcholine esterase were reduced in the mouse brain in response to IAB pre‑treatment. In cells and brain tissue, IAB regulated the expression levels of pro‑ and anti‑apoptotic proteins and enhanced the nuclear levels of NF‑E2p45‑related factor 2 (Nrf2); subsequently, IAB further enhanced the expression of superoxide dismutase 1, catalase, and heme oxygenase‑1 and ‑2. The findings of the present study indicated that the protection of IAB against AD is at least partially associated with its antioxidation and anti‑apoptotic properties.
Collapse
Affiliation(s)
- Hong Yu
- Departments of Otolaryngology Head and Neck Surgery, Jilin 130021, P.R. China
| | - Bo Yuan
- Urology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Qiubo Chu
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Chunyue Wang
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Hui Bi
- Department of Anesthesiology, Hospital of Stomatology, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
13
|
Protective roles of Amanita caesarea polysaccharides against Alzheimer's disease via Nrf2 pathway. Int J Biol Macromol 2019; 121:29-37. [DOI: 10.1016/j.ijbiomac.2018.09.216] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/12/2018] [Accepted: 09/29/2018] [Indexed: 02/07/2023]
|
14
|
Lv J, Jiang S, Yang Z, Hu W, Wang Z, Li T, Yang Y. PGC-1α sparks the fire of neuroprotection against neurodegenerative disorders. Ageing Res Rev 2018; 44:8-21. [PMID: 29580918 DOI: 10.1016/j.arr.2018.03.004] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 03/12/2018] [Accepted: 03/20/2018] [Indexed: 12/30/2022]
Abstract
Recently, growing evidence has demonstrated that peroxisome proliferator activated receptor γ (PPARγ) coactivator-1α (PGC-1α) is a superior transcriptional regulator that acts via controlling the expression of anti-oxidant enzymes and uncoupling proteins and inducing mitochondrial biogenesis, which plays a beneficial part in the central nervous system (CNS). Given the significance of PGC-1α, we summarize the current literature on the molecular mechanisms and roles of PGC-1α in the CNS. Thus, in this review, we first briefly introduce the basic characteristics regarding PGC-1α. We then depict some of its important cerebral functions and discuss upstream modulators, partners, and downstream effectors of the PGC-1α signaling pathway. Finally, we highlight recent progress in research on the involvement of PGC-1α in certain major neurodegenerative disorders (NDDs), including Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. Collectively, the data presented here may be useful for supporting the future potential of PGC-1α as a therapeutic target.
Collapse
Affiliation(s)
- Jianjun Lv
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China; Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Shuai Jiang
- Department of Aerospace Medicine, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Zhi Yang
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Wei Hu
- Department of Immunology, The Fourth Military Medical University,169 Changle West Road, Xi'an 710032, China
| | - Zheng Wang
- Department of Cadio-Thoracic Surgery, Wuhan General Hospital of The People's Liberation Army, 627 Wuluo Road, Wuhan 430070, China
| | - Tian Li
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China; Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China.
| |
Collapse
|
15
|
Ropivacaine regulates the expression and function of heme oxygenase-1. Biomed Pharmacother 2018; 103:284-289. [PMID: 29656184 DOI: 10.1016/j.biopha.2018.04.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 03/29/2018] [Accepted: 04/02/2018] [Indexed: 01/03/2023] Open
Abstract
As a new generation of amide-type local anesthetics (LAs), ropivacaine has been widely used for pain management in clinical settings. Increasing evidence has shown that administration of ropivacaine causes cytotoxic effects and apoptosis. However, the underlying molecular mechanisms still need to be elucidated. In the current study, our results indicated that ropivacaine treatment caused a significant induction of heme oxygenase-1 (HO-1) at both the mRNA and protein levels in human SHSY5Y cells. Levels of HO-1 mRNA and protein peaked at 1 h and 18 h, respectively, in response to ropivacaine treatment. Additionally, ropivacaine treatment enhanced HO-1 activity in a dose-dependent manner. Interestingly, we found that ropivacaine treatment induced phosphorylation of p38. Blockage of p38 phosphorylation with its specific inhibitor SB203580 or by transfection with p38 siRNA restrained ropivacaine-stimulated HO-1 expression. Additionally, we found that ropivacaine treatment promoted nuclear translocation of Nrf2 and amplified ARE promoter activity. Silencing of Nrf2 abolished ropivacaine-induced HO-1 expression. Notably, we found that inhibition of HO-1 activity promoted ropivacaine-induced production of reactive oxygen species (ROS), deletion of reduced glutathione (GSH), and release of lactate dehydrogenase (LDH), suggesting that induction of HO-1 by ropivacaine acted as a compensatory survival response against ropivacaine.
Collapse
|
16
|
Cui Y, Ma S, Zhang C, Li D, Yang B, Lv P, Xing Q, Huang T, Yang GL, Cao W, Guan F. Pharmacological activation of the Nrf2 pathway by 3H-1, 2-dithiole-3-thione is neuroprotective in a mouse model of Alzheimer disease. Behav Brain Res 2018; 336:219-226. [DOI: 10.1016/j.bbr.2017.09.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 08/19/2017] [Accepted: 09/05/2017] [Indexed: 12/20/2022]
|
17
|
Onodera Y, Teramura T, Takehara T, Obora K, Mori T, Fukuda K. miR-155 induces ROS generation through downregulation of antioxidation-related genes in mesenchymal stem cells. Aging Cell 2017; 16:1369-1380. [PMID: 28967703 PMCID: PMC5676067 DOI: 10.1111/acel.12680] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2017] [Indexed: 12/21/2022] Open
Abstract
Inflammation‐induced reactive oxygen species (ROS) are implicated in cellular dysfunction and an important trigger for aging‐ or disease‐related tissue degeneration. Inflammation‐induced ROS in stem cells lead to deterioration of their properties, altering tissue renewal or regeneration. Pathological ROS generation can be induced by multiple steps, and dysfunction of antioxidant systems is a major cause. The identification of the central molecule mediating the above‐mentioned processes would pave the way for the development of novel therapeutics for aging, aging‐related diseases, or stem cell therapies. In recent years, microRNAs (miRNAs) have been shown to play important roles in many biological reactions, including inflammation and stem cell functions. In inflammatory conditions, certain miRNAs are highly expressed and mediate some cytotoxic actions. Here, we focused on miR‐155, which is one of the most prominent miRNAs in inflammation and hypothesized that miR‐155 participates to inflammation‐induced ROS generation in stem cells. We observed mesenchymal stem cells (MSCs) from 1.5‐year‐old aged mice and determined that antioxidants, Nfe2l2, Sod1, and Hmox1, were suppressed, while miR‐155‐5p was highly expressed. Subsequent in vitro studies demonstrated that miR‐155‐5p induces ROS generation by suppression of the antioxidant genes by targeting the common transcription factor C/ebpβ. Moreover, this mechanism occurred during the cell transplantation process, in which ROS generation is triggering loss of transplanted stem cells. Finally, attenuation of antioxidants and ROS accumulation were partially prevented in miR‐155 knockout MSCs. In conclusion, our study suggests that miR‐155 is an important mediator connecting aging, inflammation, and ROS generation in stem cells.
Collapse
Affiliation(s)
- Yuta Onodera
- Division of Cell Biology for Regenerative Medicine Institute of Advanced Clinical Medicine Kindai University Faculty of Medicine Osaka Japan
| | - Takeshi Teramura
- Division of Cell Biology for Regenerative Medicine Institute of Advanced Clinical Medicine Kindai University Faculty of Medicine Osaka Japan
| | - Toshiyuki Takehara
- Division of Cell Biology for Regenerative Medicine Institute of Advanced Clinical Medicine Kindai University Faculty of Medicine Osaka Japan
| | - Kayoko Obora
- Department of Rehabilitation Medicine Kindai University Faculty of Medicine Osaka Japan
| | - Tatsufumi Mori
- Kindai University Life Science Research Institute Osaka Japan
| | - Kanji Fukuda
- Division of Cell Biology for Regenerative Medicine Institute of Advanced Clinical Medicine Kindai University Faculty of Medicine Osaka Japan
- Department of Rehabilitation Medicine Kindai University Faculty of Medicine Osaka Japan
| |
Collapse
|
18
|
Li H, Qin T, Li M, Ma S. Thymol improves high-fat diet-induced cognitive deficits in mice via ameliorating brain insulin resistance and upregulating NRF2/HO-1 pathway. Metab Brain Dis 2017; 32:385-393. [PMID: 27761760 DOI: 10.1007/s11011-016-9921-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 10/06/2016] [Indexed: 01/08/2023]
Abstract
The impaired insulin signaling has been recognized as a common pathogenetic mechanism between diabetes and Alzheimer's disease (AD). In the progression of AD, brain is characterized by defective insulin receptor substrate-1 (IRS-1) and increased oxidative stress. Thymol, a monoterpene phenol isolated from medicinal herbs, has exhibited robust neuroprotective effects. The present study was designed to investigate the protective effect of thymol on HFD-induced cognitive deficits, and explore the possible mechanisms. C57BL/6 J mice were fed for 12 weeks with either HFD or normal diet. The mice fed with HFD were dosed with metformin (200 mg/kg) or thymol (20, 40 mg/kg) daily. It was observed that thymol treatment significantly reversed the gain of body weight and peripheral insulin resistance induced by HFD. Meanwhile, thymol improved the cognitive impairments in the Morris Water Maze (MWM) test and decreased HFD-induced Aβ deposition and tau hyperphosphorylation in the hippocampus, which may be correlated with the inhibition of hippocampal oxidative stress and inflammation. In addition, thymol down-regulated the level of P-Ser307 IRS-1, and hence enhancing the expression of P-Ser473 AKT and P-Ser9 GSK3β. We further found that the protective effects of thymol on cognitive impairments were associated with the up-regulation of nuclear respiratory factor (Nrf2)/heme oxygenase-1(HO-1) pathway. In conclusion, thymol exhibited beneficial effects on HFD-induced cognitive deficits through improving hippocampal insulin resistance, and activating Nrf2/HO-1 signaling.
Collapse
Affiliation(s)
- Hongyan Li
- Department of Pharmacology of Chinese Materia Medica, China PharmaceuticalUniversity, Nanjing, 210009, People's Republic of China
| | - Tingting Qin
- Department of Pharmacology of Chinese Materia Medica, China PharmaceuticalUniversity, Nanjing, 210009, People's Republic of China
| | - Min Li
- Department of Pharmacology of Chinese Materia Medica, China PharmaceuticalUniversity, Nanjing, 210009, People's Republic of China
| | - Shiping Ma
- Department of Pharmacology of Chinese Materia Medica, China PharmaceuticalUniversity, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
19
|
Neuroprotective Properties of Panax notoginseng Saponins via Preventing Oxidative Stress Injury in SAMP8 Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:8713561. [PMID: 28250796 PMCID: PMC5303860 DOI: 10.1155/2017/8713561] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 01/11/2017] [Indexed: 12/14/2022]
Abstract
Inhibiting oxidative damage in early stage of Alzheimer's disease (AD) is considered as a strategy for AD treatment. Our previous study has shown that Panax notoginseng saponins (PNS) have an antiaging action by increasing the levels of superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GSH-PX) in the serum of aged rats. In this study, we aimed to investigate the effects of PNS on antioxidant enzymes and uncoupling proteins (UCPs) involved in oxidative stress in AD mice. The results showed that PNS prevented neuronal loss in hippocampal CA1 region and alleviated pathomorphological change of neurons in CA1 region. Moreover, PNS inhibited the production of 8-hydroxydeoxyguanosine (8-OHdG), enhanced the expressions and activities of SOD, CAT, and GSH-PX, and improved the mRNA and protein levels of UCP4 and UCP5 in the brains of SAMP8 mice. Together, our study shows that PNS has the ability to protect neurons in AD brain from oxidative stress damage through attenuating the production of 8-OHdG, enhancing the activities of antioxidant enzymes and the expressions levels of UCP4 and UCP5. Accordingly, PNS may be a promising agent for AD treatment.
Collapse
|
20
|
Relationships between Stress Granules, Oxidative Stress, and Neurodegenerative Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:1809592. [PMID: 28194255 PMCID: PMC5286466 DOI: 10.1155/2017/1809592] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 12/28/2016] [Indexed: 12/13/2022]
Abstract
Cytoplasmic stress granules (SGs) are critical for facilitating stress responses and for preventing the accumulation of misfolded proteins. SGs, however, have been linked to the pathogenesis of neurodegenerative diseases, in part because SGs share many components with neuronal granules. Oxidative stress is one of the conditions that induce SG formation. SGs regulate redox levels, and SG formation in turn is differently regulated by various types of oxidative stress. These associations and other evidences suggest that SG formation contributes to the development of neurodegenerative diseases. In this paper, we review the regulation of SG formation/assembly and discuss the interactions between oxidative stress and SG formation. We then discuss the links between SGs and neurodegenerative diseases and the current therapeutic approaches for neurodegenerative diseases that target SGs.
Collapse
|
21
|
Wang L, Wang M, Hu J, Shen W, Hu J, Yao Y, Wang X, Afzal CM, Ma R, Li G. Protective effect of 3H-1, 2-dithiole-3-thione on cellular model of Alzheimer's disease involves Nrf2/ARE signaling pathway. Eur J Pharmacol 2016; 795:115-123. [PMID: 27939991 DOI: 10.1016/j.ejphar.2016.12.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 12/05/2016] [Accepted: 12/07/2016] [Indexed: 10/20/2022]
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a major regulator for a battery of genes encoding detoxifying and antioxidative enzymes. 3H-1, 2-dithiole-3-thione (D3T), a potent free radical scavenger, is able to activate Nrf2 signaling pathway. In the present study, N2a/APPswe cells were used as the Alzheimer's disease (AD) cellular model and we investigated the protective effect of D3T on N2a/APPswe cells and the potential mechanisms. Our assays demonstrated that D3T was able to attenuate reactive oxygen species generation, increase MMP level as well as decrease MDA content. Furthermore, treatment of the cells with 40μM D3T for 24h, showed significant suppression of Aβ level in N2a/APPswe cells. The current study also found that D3T significantly upregulated the Nrf2 mRNA level and protein expression, and subsequently enhanced mRNA expression of HO-1 and NQO1 in N2a/APPswe cells. Meanwhile, down-regulation of Nrf2 by small interference RNA abolished cytoprotection of D3T. Taken together, these results demonstrate that D3T provides neuroprotection in vitro model and therefore may be a potential complement for AD therapy.
Collapse
Affiliation(s)
- Lan Wang
- Department of Neurology, Puai Hospital, Tongji Medical College of Huazhong University of Science & Technology, Wuhan 430033, China
| | - Min Wang
- Department of Neurology, Puai Hospital, Tongji Medical College of Huazhong University of Science & Technology, Wuhan 430033, China; Department of Neurology, Union Hospital, Tongji Medical College of Huazhong University of Science & Technology, Wuhan 430022, China
| | - Jing Hu
- Department of Neurology, Huaihe Hospital, Henan University, Kaifeng 47500, China
| | - Wei Shen
- Department of Neurology, Puai Hospital, Tongji Medical College of Huazhong University of Science & Technology, Wuhan 430033, China
| | - Junjie Hu
- Department of Neurology, Union Hospital, Tongji Medical College of Huazhong University of Science & Technology, Wuhan 430022, China
| | - Yi Yao
- Department of Neurology, Union Hospital, Tongji Medical College of Huazhong University of Science & Technology, Wuhan 430022, China
| | - Xifeng Wang
- Department of Neurology, Puai Hospital, Tongji Medical College of Huazhong University of Science & Technology, Wuhan 430033, China
| | - Curimbacus M Afzal
- Department of Neurology, Union Hospital, Tongji Medical College of Huazhong University of Science & Technology, Wuhan 430022, China
| | - Rong Ma
- Department of Pharmacology, Tongji Medical College of Huazhong University of Science & Technology, Wuhan 430030, China
| | - Gang Li
- Department of Neurology, Union Hospital, Tongji Medical College of Huazhong University of Science & Technology, Wuhan 430022, China.
| |
Collapse
|
22
|
Cai M, Wang H, Li JJ, Zhang YL, Xin L, Li F, Lou SJ. The signaling mechanisms of hippocampal endoplasmic reticulum stress affecting neuronal plasticity-related protein levels in high fat diet-induced obese rats and the regulation of aerobic exercise. Brain Behav Immun 2016; 57:347-359. [PMID: 27189035 DOI: 10.1016/j.bbi.2016.05.010] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 05/12/2016] [Accepted: 05/13/2016] [Indexed: 12/24/2022] Open
Abstract
High fat diet (HFD)-induced obesity has been shown to reduce the levels of neuronal plasticity-related proteins, specifically brain-derived neurotrophic factor (BDNF) and synaptophysin (SYN), in the hippocampus. However, the underlying mechanisms are not fully clear. Endoplasmic reticulum stress (ERS) has been reported to play a key role in regulating gene expression and protein production by affecting stress signaling pathways and ER functions of protein folding and post-translational modification in peripheral tissues of obese rodent models. Additionally, HFD that is associated with hyperglycemia could induce hippocampal ERS, thus impairing insulin signaling and cognitive health in HFD mice. One goal of this study was to determine whether hyperglycemia and hyperlipidemia could cause hippocampal ERS in HFD-induced obese SD rats, and explore the potential mechanisms of ERS regulating hippocampal BDNF and SYN proteins production. Additionally, although regular aerobic exercise could reduce central inflammation and elevate hippocampal BDNF and SYN levels in obese rats, the regulated mechanisms are poorly understood. Nrf2-HO-1 pathways play roles in anti-ERS, anti-inflammation and anti-apoptosis in peripheral tissues. Therefore, the other goal of this study was to determine whether aerobic exercise could activate Nrf2-HO-1 in hippocampus to alleviate obesity-induced hippocampal ERS, which would lead to increased BDNF and SYN levels. Male SD rats were fed on HFD for 8weeks to establish the obese model. Then, 8weeks of aerobic exercise treadmill intervention was arranged for the obese rats. Results showed that HFD-induced obesity caused hyperglycemia and hyperlipidemia, and significantly promoted hippocampal glucose transporter 3 (GLUT3) and fatty acid transport protein 1 (FATP1) protein expression. These results were associated with the activation of hippocampal ERS and ERS-mediated apoptosis. At the same time, we found that excessive hippocampal ERS not only significantly decreased proBDNF-the precursor of mature BDNF, but also attenuated p38/ERK-CREB signaling pathways and activated NLRP3-IL-1β pathways in obese rats. These results were associated with reduced BDNF and SYN protein production. However, these adverse changes were obviously reversed by aerobic exercise intervention through activating the Nrf2-HO-1 pathways. These results suggest that dietary obesity could induce hippocampal ERS in male SD rats, and excessive hippocampal ERS plays a critical role in decreasing the levels of BDNF and SYN. Moreover, aerobic exercise could activate hippocampal Nrf2 and HO-1 to relieve ERS and heighten BDNF and SYN production in obese rats.
Collapse
Affiliation(s)
- Ming Cai
- Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Hong Wang
- Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, China; College of Rehabilitation Sciences, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Jing-Jing Li
- Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Yun-Li Zhang
- Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Lei Xin
- Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Feng Li
- Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Shu-Jie Lou
- Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, China.
| |
Collapse
|
23
|
Hsu YM, Yin MC. EPA or DHA enhanced oxidative stress and aging protein expression in brain of d-galactose treated mice. Biomedicine (Taipei) 2016; 6:17. [PMID: 27514534 PMCID: PMC4980825 DOI: 10.7603/s40681-016-0017-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 06/20/2016] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Effects of eicosapentaenoic acid (EPA, 20:5) and docosahexaenoic acid (DHA, 22:6) upon fatty acid composition, oxidative and inflammatory factors and aging proteins in brain of d-galactose (DG) treated aging mice were examined. METHODS Each fatty acid at 7 mg/kg BW/week was supplied for 8 weeks. Brain aging was induced by DG treatment (100 mg/kg body weight) via daily subcutaneous injection for 8 weeks. RESULTS DG, EPA and DHA treatments changed brain fatty acid composition. DG down-regulated brain Bcl-2 expression and up-regulated Bax expression. Compared with DG groups, EPA and DHA further enhanced Bax expression. DG decreased glutathione content, increased reactive oxygen species (ROS) and oxidized glutathione (GSSG) production, the intake of EPA or DHA caused greater ROS and GSSG formation. DG treatments up-regulated the protein expression of p47(phox) and gp91(phox), and the intake of EPA or DHA led to greater p47(phox) and gp91(phox) expression. DG increased brain prostaglandin E2 (PGE2) levels, and cyclooxygenase (COX)-2 expression and activity, the intake of EPA or DHA reduced brain COX-2 activity and PGE2 formation. DG enhanced brain p53, p16 and p21 expression. EPA and DHA intake led to greater p21 expression, and EPA only caused greater p53 and p16 expression. CONCLUSION These findings suggest that these two PUFAs have toxic effects toward aging brain.
Collapse
Affiliation(s)
- Yuan-Man Hsu
- Department of Biological Science and Technology, China Medical University, 404, Taichung, Taiwan
| | - Mei-Chin Yin
- Department of Nutrition, China Medical University, 91, Hsueh-shih Rd., 404, Taichung, Taiwan.
| |
Collapse
|
24
|
de Oliveira MR. Phloretin-induced cytoprotective effects on mammalian cells: A mechanistic view and future directions. Biofactors 2016; 42:13-40. [PMID: 26826024 DOI: 10.1002/biof.1256] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 11/19/2015] [Indexed: 11/11/2022]
Abstract
Phloretin (C15 H14 O5 ), a dihydrochalcone flavonoid, is mainly found in fruit, leaves, and roots of apple tree. Phloretin exerts antioxidant, anti-inflammatory, and anti-tumor activities in mammalian cells through mechanisms that have been partially elucidated throughout the years. Phloretin bioavailability is well known in humans, but still remains to be better studied in experimental animals, such as mouse and rat. The focus of the present review is to gather information regarding the mechanisms involved in the phloretin-elicited effects in different in vitro and in vivo experimental models. Several manuscripts were analyzed and data raised by authors were described and discussed here in a mechanistic manner. Comparisons between the effects elicited by phloretin and phloridzin were made whenever possible, as well as with other polyphenols, clarifying questions about the use of phloretin as a potential therapeutic agent. Toxicological aspects associated to phloretin exposure were also discussed here. Furthermore, a special section containing future directions was created as a suggestive guide towards the elucidation of phloretin-related actions in mammalian cells and tissues.
Collapse
Affiliation(s)
- Marcos Roberto de Oliveira
- Department of Chemistry/ICET, Postgraduate Program in Chemistry (PPGQ), Federal University of Mato Grosso (UFMT), CEP, Cuiaba, MT, Brazil
| |
Collapse
|
25
|
BQ123 Stimulates Skeletal Muscle Antioxidant Defense via Nrf2 Activation in LPS-Treated Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:2356853. [PMID: 26823945 PMCID: PMC4707360 DOI: 10.1155/2016/2356853] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 09/24/2015] [Accepted: 10/11/2015] [Indexed: 01/02/2023]
Abstract
Little is understood of skeletal muscle tissue in terms of oxidative stress and inflammation. Endothelin-1 is an endogenous, vasoconstrictive peptide which can induce overproduction of reactive oxygen species and proinflammatory cytokines. The aim of this study was to evaluate whether BQ123, an endothelin-A receptor antagonist, influences the level of TNF-α, IL-6, SOD-1, HO-1, Nrf2 mRNA, and NF-κB subunit RelA/p65 mRNA in the femoral muscle obtained from endotoxemic rats. Male Wistar rats were divided into 4 groups (n = 6) and received iv (1) saline (control), (2) LPS (15 mg/kg), (3) BQ123 (1 mg/kg), (4) BQ123 (1 mg/kg), and LPS (15 mg/kg, resp.) 30 min later. Injection of LPS led to significant increase in levels of RelA/p65 mRNA, TNF-α, and IL-6, while content of SOD-1, HO-1, and Nrf2 mRNA was unchanged. Administration of BQ123 prior to LPS challenge resulted in a significant reduction in RelA/p65 mRNA, TNF-α, and IL-6 levels, as well as markedly elevated concentrations of SOD-1, HO-1, and Nrf2 mRNA. BQ123 appears to enhance antioxidant defense and prevent production of TNF-α and IL-6 in skeletal muscle of LPS-treated rat. In conclusion, endothelin-A receptor antagonism exerts significant impact on the skeletal muscle favouring anti-inflammatory effects and protection against oxidative stress.
Collapse
|
26
|
Wang B, Zhang Q, Yao R, Liu X, Qu Z. 7, 8-Dihydroxyflavone Protects an Endothelial Cell Line from H2O2 Damage. PLoS One 2015; 10:e0135345. [PMID: 26266800 PMCID: PMC4534426 DOI: 10.1371/journal.pone.0135345] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 07/22/2015] [Indexed: 12/31/2022] Open
Abstract
7, 8-dihydroxyflavone (7, 8-DHF), a selective agonist for TrkB receptors, has been well studied for its neurotrophic functions. However, its roles outside the neural tissues have scarcely been studied as yet. In this study, we investigated the protecting roles of 7, 8-DHF in EA.hy926 cells, a human umbilic vein endothelial cell line which was exposed to hydrogen peroxide (H2O2). We found that 7, 8-DHF significantly protected the cells from being damaged by H2O2 through suppression of apoptosis, attenuation of inflammatory factor releasing and inhibition of reactive oxygen species generation. The potent biological effects of 7, 8-DHF were probably executed via its binding to TrkB receptors because the receptor specific antagonist ANA-12 significantly blocked its protecting effects. The protecting roles of 7, 8-DHF in EA.hy926 cells suggest that it will be a promising compound to be developed into a health product that definitely benefits endothelial functions and prevents cardiovascular diseases.
Collapse
Affiliation(s)
- Bingxiang Wang
- Center for Medical Research, the First Affiliated Hospital, Qingdao University, Qingdao, 266555, China; Department of Physiology, Taishan Medical College, Taian, 271000, China
| | - Qian Zhang
- Center for Medical Research, the First Affiliated Hospital, Qingdao University, Qingdao, 266555, China
| | - Ruyong Yao
- Center for Medical Research, the First Affiliated Hospital, Qingdao University, Qingdao, 266555, China
| | - Xiangping Liu
- Center for Medical Research, the First Affiliated Hospital, Qingdao University, Qingdao, 266555, China
| | - Zhiqiang Qu
- Center for Medical Research, the First Affiliated Hospital, Qingdao University, Qingdao, 266555, China
| |
Collapse
|
27
|
Peng T, Wang J, Lu J, Lu H, Teng J, Jia Y. Neuroprotective effects of α-melanocyte-stimulating hormone against the neurotoxicity of 1-methyl-4-phenylpyridinium. IUBMB Life 2015; 69:315-320. [PMID: 26136377 DOI: 10.1002/iub.1385] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 03/02/2015] [Accepted: 04/20/2015] [Indexed: 11/07/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease in humans. The hormone α-melanocyte-stimulating hormone (α-MSH) has been reported to be neuroprotective in previous studies. The aim of this study is to investigate the neuroprotective effects of α-MSH against the neurotoxicity of 1-methyl-4-phenylpyridinium (MPP+). Our results indicated that treatment with α-MSH in M17 cells attenuated MPP+-induced oxidative stress, embodied by exacerbated reactive oxygen species and protein carbonyls. In addition, we found that α-MSH could improve mitochondrial function in M17 cells through increasing the level of adenosine triphosphate and mitochondrial membrane potential. Furthermore, treatment with α-MSH restored the reduction of cell viability and the induction of lactate dehydrogenase release induced by α-MSH. Importantly, Hoechst staining results indicated that α-MSH treatment significantly reduces the number of apoptotic cells after treatment with MPP+. Mechanically, we found that α-MSH prevented apoptosis signals through reducing the level of cleaved caspase-3 and attenuating cytochrome c release. All these data imply that α-MSH produces a protective effect in PD. © 2015 IUBMB Life, 69(5):315-320, 2017.
Collapse
Affiliation(s)
- Tao Peng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jingtao Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jingjing Lu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Hong Lu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Junfang Teng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yanjie Jia
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
28
|
Peers C, Boyle JP. Oxidative modulation of K+ channels in the central nervous system in neurodegenerative diseases and aging. Antioxid Redox Signal 2015; 22:505-21. [PMID: 25333910 DOI: 10.1089/ars.2014.6007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
SIGNIFICANCE Oxidative stress and damage are well-established components of neurodegenerative diseases, contributing to neuronal death during disease progression. Here, we consider key K(+) channels as target proteins that can undergo oxidative modulation, describe what is understood about how this influences disease progression, and consider regulation of these channels by gasotransmitters as a means of cellular protection. RECENT ADVANCES Oxidative regulation of the delayed rectifier Kv2.1 and the Ca(2+)- and voltage-sensitive BK channel are established, but recent studies contest how their redox sensitivity contributes to altered excitability, progression of neurodegenerative diseases, and healthy aging. CRITICAL ISSUES Both Kv2.1 and BK channels have recently been established as target proteins for regulation by the gasotransmitters carbon monoxide and hydrogen sulfide. Establishing the molecular basis of such regulation, and exactly how this influences excitability and vulnerability to apoptotic cell death will determine whether such regulation can be exploited for therapeutic benefit. FUTURE DIRECTIONS Developing a more comprehensive picture of the oxidative modulation of K(+) channels (and, indeed, other ion channels) within the central nervous system in health and disease will enable us to better understand processes associated with healthy aging as well as distinct processes underlying progression of neurodegenerative diseases. Advances in the growing understanding of how gasotransmitters can regulate ion channels, including redox-sensitive K(+) channels, are a research priority for this field, and will establish their usefulness in design of future approaches for the treatment of such diseases.
Collapse
Affiliation(s)
- Chris Peers
- Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), Faculty of Medicine and Health, University of Leeds , Leeds, United Kingdom
| | | |
Collapse
|
29
|
Zhou Q, Chen J, Yi S, Lou Y, Tang W, Liu Y, Zhang P. Zhichan powder regulates nigrostriatal dopamine synthesis and metabolism in Parkinson's disease rats. Neural Regen Res 2015; 7:2107-14. [PMID: 25558223 PMCID: PMC4281411 DOI: 10.3969/j.issn.1673-5374.2012.27.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 08/06/2012] [Indexed: 12/20/2022] Open
Abstract
In this study, rat models of Parkinson's disease induced by substantia nigra injection of 6-hydroxy-dopamine were intragastrically administered Zhichan powder daily for 50 days. Reverse transcription PCR results showed that tyrosine hydroxylase mRNA expression in the rat substantia nigra was significantly increased, while monoamine oxidase B mRNA expression was significantly decreased in the Zhichan powder group, compared with the model group. In addition, the levels of striatal dopamine and homovanillic acid, the ratio of dopamine to homovanillic acid, and the activity of blood superoxide dismutase were all higher in the Zhichan powder group than in the model group, but the content of malondialdehyde in blood was lower. Our experimental findings indicate that Zhichan powder has an antioxidant effect, it can regulate the expression of monoamine oxidase B and tyrosine hydroxylase in the substantia nigra of Parkinson's disease rats, and it can facilitate the secretion of striatal dopamine and its metabolite homovanillic acid.
Collapse
Affiliation(s)
- Qingwei Zhou
- Department of Biochemistry and Molecular Biology, Institute of Regenerative Medical Sciences, Jilin University, Changchun 130021, Jilin Province, China
| | - Jiajun Chen
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun 130013, Jilin Province, China
| | - Shihong Yi
- Department of Immunology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, Jilin Province, China
| | - Yongwei Lou
- Department of Clinical Laboratory, Changchun Municipal People's Hospital, Changchun 130041, Jilin Province, China
| | - Weimin Tang
- Department of Immunology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, Jilin Province, China
| | - Yongmao Liu
- Department of Immunology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, Jilin Province, China
| | - Pengguo Zhang
- Department of Radiology, Second Clinical Hospital of Jilin University, Changchun 130021, Jilin Province, China
| |
Collapse
|
30
|
Noda M. Protective effect of molecular hydrogen against white matter ischemic injury. ACTA ACUST UNITED AC 2015. [DOI: 10.16977/cbfm.26.2_77] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Mami Noda
- Laboratory of Pathophysiology, Graduate School of Pharmaceutical Sciences, Kyushu University
- Division of Neurofunctional Genomics, Medical Institute of Bioregulation, Kyushu University
| |
Collapse
|
31
|
Zhai X, Chen X, Ohta S, Sun X. Review and prospect of the biomedical effects of hydrogen. Med Gas Res 2014; 4:19. [PMID: 25485090 PMCID: PMC4256831 DOI: 10.1186/s13618-014-0019-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 11/17/2014] [Indexed: 01/25/2023] Open
Abstract
Hydrogen, the simplest gas in nature, was recently reported as a therapeutic antioxidant through selectively reducing cytotoxic oxygen radicals. Though hundreds of studies on curative effects of hydrogen were published and justified, the mechanism remains unclear. We proposed several promising directions in this area by relatively in-depth analysis. Firstly, the physiological function of hydrogen was regarded neutralizing free radicals at a low dose; however, physiological effects of an excessive dose of hydrogen were necessary for the comprehensive understanding. Secondly, the therapeutic effects and mechanisms were explained by anti-oxidative, anti-inflammatory and apoptosis ways, while the limitation was obvious and needed update. Thirdly, further studies might be focused on the possible networks including effecters and receptors of hydrogen, and the evolutionary perspective was a good point of view. In conclusion, this review might be a reference and guidance for relative scholars.
Collapse
Affiliation(s)
- Xiao Zhai
- Graduate Management Unit, Changhai hospital affiliated to the Second Military Medical University, Shanghai, PR China
| | - Xiao Chen
- Department of Orthopedics, Changhai hospital affiliated to the Second Military Medical University, Shanghai, PR China
| | - Shigeo Ohta
- Department of Biochemistry and Cell Biology, Institute of Development and Aging Sciences, Graduate School of Medicine, Nippon Medical School, 1-396 Kosugi-machi, Nakahara-ku, Kawasaki-city, Kanagawa-ken 211-8533 Japan
| | - Xuejun Sun
- Department of Navy Aeromedicine, Second Military Medical University, 168 Changhai, Shanghai, China
| |
Collapse
|
32
|
Xing S, Shen D, Chen C, Wang J, Yu Z. Early induction of oxidative stress in a mouse model of Alzheimer's disease with heme oxygenase activity. Mol Med Rep 2014; 10:599-604. [PMID: 24858858 PMCID: PMC4094768 DOI: 10.3892/mmr.2014.2252] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 04/03/2014] [Indexed: 11/06/2022] Open
Abstract
Evidence suggests that brain tissues of patients with Alzheimer's disease (AD) are easily attacked by oxidative stress, and numerous studies indicate that heme oxygenase (HO) is a major cell adaptive responder to stress. However, whether HO‑1 and HO‑2 play different roles in this process has not yet been studied. In the present study, it was shown in an AD model that HO‑1 and HO‑2 have different roles in the early stages of AD. Learning and memory ability was tested in APPswe/PS1ΔE9 (APP/PS1) transgenic and wild‑type mice using the Morris water maze. β‑amyloid plaques were measured using immunofluorescence staining. Changes in reactive oxygen species (ROS) levels in the hippocampi were measured using a fluorescence technique. The results indicated that the escape latency, amyloid plaque deposition and ROS production increased in the hippocampi of APP/PS1 transgenic mice compared with wild‑type mice. Furthermore, using double‑immunofluorescence staining and western blot analysis, it was found that the expression of HO‑1 and HO‑2 increased in the hippocampi of APP/PS1 mice and, notably, HO‑2 was also found to be overexpressed in astrocytes. Little difference was observed in the plasma HO‑1 concentrations between the two groups, while the plasma HO‑2 concentration of the APP/PS1 mice was lower than that of the wild‑type mice, shown by ELISA. In conclusion, HO‑2 overexpression is an early event and plays a more critical role in the progression of AD.
Collapse
Affiliation(s)
- Sanli Xing
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, P.R. China
| | - Dingzhu Shen
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, P.R. China
| | - Chuan Chen
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, P.R. China
| | - Jian Wang
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, P.R. China
| | - Zhihua Yu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, P.R. China
| |
Collapse
|
33
|
Szczesny B, Módis K, Yanagi K, Coletta C, Le Trionnaire S, Perry A, Wood ME, Whiteman M, Szabo C. AP39, a novel mitochondria-targeted hydrogen sulfide donor, stimulates cellular bioenergetics, exerts cytoprotective effects and protects against the loss of mitochondrial DNA integrity in oxidatively stressed endothelial cells in vitro. Nitric Oxide 2014; 41:120-30. [PMID: 24755204 DOI: 10.1016/j.niox.2014.04.008] [Citation(s) in RCA: 218] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 04/08/2014] [Accepted: 04/14/2014] [Indexed: 12/12/2022]
Abstract
The purpose of the current study was to investigate the effect of the recently synthesized mitochondrially-targeted H2S donor, AP39 [(10-oxo-10-(4-(3-thioxo-3H-1,2-dithiol-5yl)phenoxy)decyl) triphenylphosphonium bromide], on bioenergetics, viability, and mitochondrial DNA integrity in bEnd.3 murine microvascular endothelial cells in vitro, under normal conditions, and during oxidative stress. Intracellular H2S was assessed by the fluorescent dye 7-azido-4-methylcoumarin. For the measurement of bioenergetic function, the XF24 Extracellular Flux Analyzer was used. Cell viability was estimated by the combination of the MTT and LDH methods. Oxidative protein modifications were measured by the Oxyblot method. Reactive oxygen species production was monitored by the MitoSOX method. Mitochondrial and nuclear DNA integrity were assayed by the Long Amplicon PCR method. Oxidative stress was induced by addition of glucose oxidase. Addition of AP39 (30-300 nM) to bEnd.3 cells increased intracellular H2S levels, with a preferential response in the mitochondrial regions. AP39 exerted a concentration-dependent effect on mitochondrial activity, which consisted of a stimulation of mitochondrial electron transport and cellular bioenergetic function at lower concentrations (30-100 nM) and an inhibitory effect at the higher concentration of 300 nM. Under oxidative stress conditions induced by glucose oxidase, an increase in oxidative protein modification and an enhancement in MitoSOX oxidation was noted, coupled with an inhibition of cellular bioenergetic function and a reduction in cell viability. AP39 pretreatment attenuated these responses. Glucose oxidase induced a preferential damage to the mitochondrial DNA; AP39 (100 nM) pretreatment protected against it. In conclusion, the current paper documents antioxidant and cytoprotective effects of AP39 under oxidative stress conditions, including a protection against oxidative mitochondrial DNA damage.
Collapse
Affiliation(s)
- Bartosz Szczesny
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Katalin Módis
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Kazunori Yanagi
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Ciro Coletta
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Sophie Le Trionnaire
- University of Exeter Medical School, St. Luke's Campus, Exeter, England, United Kingdom
| | - Alexis Perry
- Biosciences, College of Life and Environmental Science, University of Exeter, England, United Kingdom
| | - Mark E Wood
- Biosciences, College of Life and Environmental Science, University of Exeter, England, United Kingdom
| | - Matthew Whiteman
- University of Exeter Medical School, St. Luke's Campus, Exeter, England, United Kingdom.
| | - Csaba Szabo
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
34
|
Carrillo-Salinas FJ, Navarrete C, Mecha M, Feliú A, Collado JA, Cantarero I, Bellido ML, Muñoz E, Guaza C. A cannabigerol derivative suppresses immune responses and protects mice from experimental autoimmune encephalomyelitis. PLoS One 2014; 9:e94733. [PMID: 24727978 PMCID: PMC3984273 DOI: 10.1371/journal.pone.0094733] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 03/19/2014] [Indexed: 01/25/2023] Open
Abstract
Phytocannabinoids that do not produce psychotropic effects are considered of special interest as novel therapeutic agents in CNS diseases. A cannabigerol quinone, the compound VCE-003, has been shown to alleviate symptoms in a viral model of multiple sclerosis (MS). Hence, we studied T cells and macrophages as targets for VCE-003 and its efficacy in an autoimmune model of MS. Proliferation, cell cycle, expression of activation markers was assessed by FACs in human primary T cells, and cytokine and chemokine production was evaluated. Transcription was studied in Jurkat cells and RAW264.7 cells were used to study the effects of VCE-003 on IL-17-induced macrophage polarization to a M1 phenotype. Experimental autoimmune encephalomyelitis (EAE) was induced by myelin oligodendrocyte glycoprotein (MOG35–55) immunization and spinal cord pathology was assessed by immunohistochemistry. Neurological impairment was evaluated using disease scores. We show here that VCE-003 inhibits CD3/CD28-induced proliferation, cell cycle progression and the expression of the IL-2Rα and ICAM-1 activation markers in human primary T cells. VCE-003 inhibits the secretion of Th1/Th17 cytokines and chemokines in primary murine T cells, and it reduces the transcriptional activity of the IL-2, IL-17 and TNFα promoters induced by CD3/CD28. In addition, VCE-003 and JWH-133, a selective CB2 agonist, dampened the IL-17-induced polarization of macrophages to a pro-inflammatory M1 profile. VCE-003 also prevented LPS-induced iNOS expression in microglia. VCE-003 ameliorates the neurological defects and the severity of MOG-induced EAE in mice through CB2 and PPARγ receptor activation. A reduction in cell infiltrates, mainly CD4+ T cells, was observed, and Th1 and Th17 responses were inhibited in the spinal cord of VCE-003-treated mice, accompanied by weaker microglial activation, structural preservation of myelin sheets and reduced axonal damage. This study highlights the therapeutic potential of VCE-003 as an agent for the treatment of human immune diseases with both inflammatory and autoimmune components.
Collapse
Affiliation(s)
| | - Carmen Navarrete
- VivaCell Biotechnology España, Parque Científico Tecnológico de Córdoba, Córdoba, Spain
| | - Miriam Mecha
- Neuroimmunology Group, Functional and System Neurobiology Department, Instituto Cajal, Madrid, Spain
| | - Ana Feliú
- Neuroimmunology Group, Functional and System Neurobiology Department, Instituto Cajal, Madrid, Spain
| | - Juan A. Collado
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, Universidad de Córdoba, Córdoba, Spain
| | - Irene Cantarero
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, Universidad de Córdoba, Córdoba, Spain
| | - María L. Bellido
- VivaCell Biotechnology España, Parque Científico Tecnológico de Córdoba, Córdoba, Spain
| | - Eduardo Muñoz
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, Universidad de Córdoba, Córdoba, Spain
- * E-mail: (EM); (CG)
| | - Carmen Guaza
- Neuroimmunology Group, Functional and System Neurobiology Department, Instituto Cajal, Madrid, Spain
- * E-mail: (EM); (CG)
| |
Collapse
|
35
|
Zhang L, Dong L, Liu X, Jiang Y, Zhang L, Zhang X, Li X, Zhang Y. α-Melanocyte-stimulating hormone protects retinal vascular endothelial cells from oxidative stress and apoptosis in a rat model of diabetes. PLoS One 2014; 9:e93433. [PMID: 24695675 PMCID: PMC3973693 DOI: 10.1371/journal.pone.0093433] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 03/04/2014] [Indexed: 12/14/2022] Open
Abstract
AIMS Oxidative stress and apoptosis are among the earliest lesions of diabetic retinopathy. This study sought to examine the anti-oxidative and anti-apoptotic effects of α-melanocyte-stimulating hormone (α-MSH) in early diabetic retinas and to explore the underlying mechanisms in retinal vascular endothelial cells. METHODS Sprague-Dawley rats were injected intravenously with streptozocin to induce diabetes. The diabetic rats were injected intravitreally with α-MSH or saline. At week 5 after diabetes, the retinas were analyzed for reactive oxygen species (ROS) and gene expression. One week later, the retinas were processed for terminal deoxynucleotidyl transferase dUTP nick-end labeling staining and transmission electron microscopy. Retinal vascular endothelial cells were stimulated by high glucose (HG) with or without α-MSH. The expression of Forkhead box O genes (Foxos) was examined through real-time PCR. The Foxo4 gene was overexpressed in endothelial cells by transient transfection prior to α-MSH or HG treatment, and oxidative stress and apoptosis were analyzed through CM-H2DCFDA and annexin-V assays, respectively. RESULTS In diabetic retinas, the levels of H2O2 and ROS and the total anti-oxidant capacity were normalized, the apoptotic cell number was reduced, and the ultrastructural injuries were ameliorated by α-MSH. Treatment with α-MSH also corrected the aberrant changes in eNOS, iNOS, ICAM-1, and TNF-α expression levels in diabetic retinas. Furthermore, α-MSH inhibited Foxo4 up-regulation in diabetic retinas and in endothelial cells exposed to HG, whereas Foxo4 overexpression abrogated the anti-oxidative and anti-apoptotic effects of α-MSH in HG-stimulated retinal vascular endothelial cells. CONCLUSIONS α-MSH normalized oxidative stress, reduced apoptosis and ultrastructural injuries, and corrected gene expression levels in early diabetic retinas. The protective effects of α-MSH in retinal vascular endothelial cells may be mediated through the inhibition of Foxo4 up-regulation induced by HG. This study suggests an α-MSH-mediated potential intervention approach to early diabetic retinopathy and a novel regulatory mechanism involving Foxo4.
Collapse
Affiliation(s)
- Lijuan Zhang
- Tianjin Medical University Eye Hospital/Eye Institute, Tianjin, China
| | - Lijie Dong
- Tianjin Medical University Eye Hospital/Eye Institute, Tianjin, China
| | - Xun Liu
- Tianjin Medical University Eye Hospital/Eye Institute, Tianjin, China
| | - Yuanfeng Jiang
- Tianjin Medical University Eye Hospital/Eye Institute, Tianjin, China
| | - Lingjun Zhang
- Tianjin Medical University Eye Hospital/Eye Institute, Tianjin, China
| | - Xiaomin Zhang
- Tianjin Medical University Eye Hospital/Eye Institute, Tianjin, China
| | - Xiaorong Li
- Tianjin Medical University Eye Hospital/Eye Institute, Tianjin, China
| | - Yan Zhang
- Tianjin Medical University Eye Hospital/Eye Institute, Tianjin, China
| |
Collapse
|
36
|
Sakurai T, Ito T, Wakame K, Kitadate K, Arai T, Ogasawara J, Kizaki T, Sato S, Ishibashi Y, Fujiwara T, Akagawa K, Ishida H, Ohno H. Enzyme-treated Asparagus officinalis Extract Shows Neuroprotective Effects and Attenuates Cognitive Impairment in Senescence-accelerated Mice. Nat Prod Commun 2014. [DOI: 10.1177/1934578x1400900130] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Increases in the number of patients with dementia involving Alzheimer's disease (AD) are seen as a grave public health problem. In neurodegenerative disorders involving AD, biological stresses, such as oxidative and inflammatory stress, induce neural cell damage. Asparagus ( Asparagus officinalis) is a popular vegetable, and an extract prepared from this reportedly possesses various beneficial biological activities. In the present study, we investigated the effects of enzyme-treated asparagus extract (ETAS) on neuronal cells and early cognitive impairment of senescence-accelerated mouse prone 8 (SAMP8) mice. The expression of mRNAs for factors that exert cytoprotective and anti-apoptotic functions, such as heat-shock protein 70 and heme oxygenase-1, was upregulated in NG108–15 neuronal cells by treatment with ETAS. Moreover, when release of lactate dehydrogenase from damaged NG108–15 cells was increased for cells cultured in medium containing either the nitric oxide donor sodium nitroprusside or the hypoxia mimic reagent cobalt chloride, ETAS significantly attenuated this cell damage. Also, when contextual fear memory, which is considered to be a hippocampus-dependent memory, was significantly impaired in SAMP8 mice, ETAS attenuated the cognitive impairment. These results suggest that ETAS produces cytoprotective effects in neuronal cells and attenuates the effects on the cognitive impairment of SAMP8 mice.
Collapse
Affiliation(s)
- Takuya Sakurai
- Department of Molecular Predictive Medicine and Sport Science, Kyorin University, School of Medicine, Tokyo 181–8611, Japan
| | - Tomohiro Ito
- Amino Up Chemical Co., Ltd., Sapporo 004-0839, Japan
| | - Koji Wakame
- Amino Up Chemical Co., Ltd., Sapporo 004-0839, Japan
| | | | - Takashi Arai
- Department of Clinical Research, Saitama Children's Medical Center, Saitama 339–8551, Japan
| | - Junetsu Ogasawara
- Department of Molecular Predictive Medicine and Sport Science, Kyorin University, School of Medicine, Tokyo 181–8611, Japan
| | - Takako Kizaki
- Department of Molecular Predictive Medicine and Sport Science, Kyorin University, School of Medicine, Tokyo 181–8611, Japan
| | - Shogo Sato
- Department of Molecular Predictive Medicine and Sport Science, Kyorin University, School of Medicine, Tokyo 181–8611, Japan
| | - Yoshinaga Ishibashi
- Department of Molecular Predictive Medicine and Sport Science, Kyorin University, School of Medicine, Tokyo 181–8611, Japan
| | - Tomonori Fujiwara
- Department of Cell Physiology, Kyorin University, School of Medicine, Tokyo 181–8611, Japan
| | - Kimio Akagawa
- Department of Cell Physiology, Kyorin University, School of Medicine, Tokyo 181–8611, Japan
| | - Hitoshi Ishida
- Third Department of Internal Medicine, Kyorin University, School of Medicine, Tokyo 181–8611, Japan
| | - Hideki Ohno
- Department of Molecular Predictive Medicine and Sport Science, Kyorin University, School of Medicine, Tokyo 181–8611, Japan
| |
Collapse
|
37
|
Kim H, Britton GL, Peng T, Holland CK, McPherson DD, Huang SL. Nitric oxide-loaded echogenic liposomes for treatment of vasospasm following subarachnoid hemorrhage. Int J Nanomedicine 2013; 9:155-65. [PMID: 24379666 PMCID: PMC3873237 DOI: 10.2147/ijn.s48856] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Delayed cerebral vasospasm following subarachnoid hemorrhage causes severe ischemic neurologic deficits leading to permanent neurologic dysfunction or death. Reduced intravascular and perivascular nitric oxide (NO) availability is a primary pathophysiology of cerebral vasospasm. In this study, we evaluated NO-loaded echogenic liposomes (NO-ELIP) for ultrasound-facilitated NO delivery to produce vasodilation for treatment of vasospasm following subarachnoid hemorrhage. We investigated the vasodilative effects of NO released from NO-ELIP both ex vivo and in vivo. Liposomes containing phospholipids and cholesterol were prepared, and NO was encapsulated. The encapsulation and release of NO from NO-ELIP were determined by the syringe/vacuum method and ultrasound imaging. The ex vivo vasodilative effect of NO-ELIP was investigated using rabbit carotid arteries. Arterial vasodilation was clearly observed with NO-ELIP exposed to Doppler ultrasound whereas there was little vasodilative effect without exposure to Doppler ultrasound in the presence of red blood cells. Penetration of NO into the arterial wall was determined by fluorescent microscopy. The vasodilative effects of intravenously administered NO-ELIP in vivo were determined in a rat subarachnoid hemorrhage model. NO-ELIP with ultrasound activation over the carotid artery demonstrated effective arterial vasodilation in vivo resulting in improved neurologic function. This novel methodology for ultrasound-controlled delivery of NO has the potential for therapeutic treatment of vasospasm following subarachnoid hemorrhage. This ultrasound-controlled release strategy provides a new avenue for targeted bioactive gas and therapeutic delivery for improved stroke treatment.
Collapse
Affiliation(s)
- Hyunggun Kim
- Division of Cardiovascular Medicine, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - George L Britton
- Division of Cardiovascular Medicine, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Tao Peng
- Division of Cardiovascular Medicine, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Christy K Holland
- Division of Cardiovascular Diseases, Department of Internal Medicine, and Department of Biomedical Engineering and Radiology, University of Cincinnati, Cincinnati, OH, USA
| | - David D McPherson
- Division of Cardiovascular Medicine, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Shao-Ling Huang
- Division of Cardiovascular Medicine, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
38
|
Xue H, Yuan P, Ni J, Li C, Shao D, Liu J, Shen Y, Wang Z, Zhou L, Zhang W, Huang Y, Yu C, Wang R, Lu L. H(2)S inhibits hyperglycemia-induced intrarenal renin-angiotensin system activation via attenuation of reactive oxygen species generation. PLoS One 2013; 8:e74366. [PMID: 24058553 PMCID: PMC3772925 DOI: 10.1371/journal.pone.0074366] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 07/31/2013] [Indexed: 01/09/2023] Open
Abstract
Decrease in endogenous hydrogen sulfide (H2S) was reported to participate in the pathogenesis of diabetic nephropathy (DN). This study is aimed at exploring the relationship between the abnormalities in H2S metabolism, hyperglycemia-induced oxidative stress and the activation of intrarenal renin-angiotensin system (RAS). Cultured renal mesangial cells (MCs) and streptozotocin (STZ) induced diabetic rats were used for the studies. The expressions of angiotensinogen (AGT), angiotensin converting enzyme (ACE), angiotensin II (Ang II) type I receptor (AT1), transforming growth factor-β1 (TGF-β1) and collagen IV were measured by real time PCR and Western blot. Reactive oxygen species (ROS) production was assessed by fluorescent probe assays. Cell proliferation was analyzed by 5'-bromo-2'-deoxyuridine incorporation assay. Ang II concentration was measured by an enzyme immunoassay. AGT, ACE and AT1 receptor mRNA levels and Ang II concentration were increased in high glucose (HG) -treated MCs, the cell proliferation rate and the production of TGF-β1 and of collagen IV productions were also increased. The NADPH oxidase inhibitor diphenylenechloride iodonium (DPI) was able to reverse the HG-induced RAS activation and the changes in cell proliferation and collagen synthesis. Supplementation of H2S attenuated HG-induced elevations in ROS and RAS activation. Blockade on H2S biosynthesis from cystathione-γ-lyase (CSE) by DL-propargylglycine (PPG) resulted in effects similar to that of HG treatment. In STZ-induced diabetic rats, the changes in RAS were also reversed by H2S supplementation without affecting blood glucose concentration. These data suggested that the decrease in H2S under hyperglycemic condition leads to an imbalance between oxidative and reductive species. The increased oxidative species results in intrarenal RAS activation, which, in turn, contributes to the pathogenesis of renal dysfunction.
Collapse
MESH Headings
- Acetophenones/pharmacology
- Angiotensin II Type 1 Receptor Blockers/pharmacology
- Angiotensinogen/genetics
- Angiotensinogen/metabolism
- Animals
- Blood Glucose/metabolism
- Cell Proliferation/drug effects
- Cells, Cultured
- Collagen Type IV/metabolism
- Cystathionine beta-Synthase/genetics
- Cystathionine beta-Synthase/metabolism
- Cystathionine gamma-Lyase/genetics
- Cystathionine gamma-Lyase/metabolism
- Diabetes Mellitus, Experimental/enzymology
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/pathology
- Glucose/pharmacology
- Hydrogen Sulfide/pharmacology
- Hyperglycemia/enzymology
- Hyperglycemia/genetics
- Hyperglycemia/metabolism
- Hyperglycemia/pathology
- Kidney/drug effects
- Kidney/pathology
- Losartan/pharmacology
- Mesangial Cells/drug effects
- Mesangial Cells/metabolism
- Mesangial Cells/pathology
- NADPH Oxidases/metabolism
- Onium Compounds/pharmacology
- Peptidyl-Dipeptidase A/genetics
- Peptidyl-Dipeptidase A/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Reactive Oxygen Species/metabolism
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Renin-Angiotensin System/drug effects
- Transforming Growth Factor beta1/metabolism
Collapse
Affiliation(s)
- Hong Xue
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ping Yuan
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pulmonary Circulation Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jun Ni
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chen Li
- Department of Nephrology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Decui Shao
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jia Liu
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yang Shen
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhen Wang
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Li Zhou
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Zhang
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu Huang
- School of Biomedical Sciences and Institute of Vascular Medicine, Chinese University of Hong Kong, Hong Kong, China
| | - Chen Yu
- Department of Nephrology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Rui Wang
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Biology, Lakehead University, Thunder Bay, Canada
| | - Limin Lu
- Department of Physiology and Pathophysiology, Shanghai Medical College, Fudan University, Shanghai, China
- * E-mail:
| |
Collapse
|
39
|
Deng YN, Shi J, Liu J, Qu QM. Celastrol protects human neuroblastoma SH-SY5Y cells from rotenone-induced injury through induction of autophagy. Neurochem Int 2013; 63:1-9. [DOI: 10.1016/j.neuint.2013.04.005] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 04/02/2013] [Accepted: 04/08/2013] [Indexed: 01/30/2023]
|