1
|
Szilágyi A, Takács B, Szekeres R, Tarjányi V, Nagy D, Priksz D, Bombicz M, Kiss R, Szabó AM, Lehoczki A, Gesztelyi R, Juhász B, Szilvássy Z, Varga B. Effects of voluntary and forced physical exercise on the retinal health of aging Wistar rats. GeroScience 2024; 46:4707-4728. [PMID: 38795184 PMCID: PMC11336036 DOI: 10.1007/s11357-024-01208-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 05/13/2024] [Indexed: 05/27/2024] Open
Abstract
Aging is accompanied by an increased prevalence of degenerative conditions, including those affecting ocular health, which significantly impact quality of life and increase the burden on healthcare systems. Among these, retinal aging is of particular concern due to its direct link to vision impairment, a leading cause of disability in the elderly. Vision loss in the aging population is associated with heightened risks of cognitive decline, social isolation, and morbidity. This study addresses the critical gap in our understanding of modifiable lifestyle factors, such as physical exercise, that may mitigate retinal aging and its related pathologies. We investigated the effects of different exercise regimens-voluntary (recreational-type) and forced (high-intensity)-on the retinal health of aging Wistar rats (18-month-old), serving as a model for studying the translational potential of exercise interventions in humans. Male Wistar rats were divided into four groups: a young control (3-month-old) for baseline comparison, an aged sedentary control, an aged group engaging in voluntary exercise via a running wheel in their cage, and an aged group subjected to forced exercise on a treadmill for six sessions of 20 min each per week. After a 6-month experimental period, we assessed retinal function via electroretinography (ERG), measured retinal thickness histologically, and analyzed protein expression changes relevant to oxidative stress, inflammation, and anti-aging mechanisms. Our findings reveal that voluntary exercise positively impacts retinal function and morphology, reducing oxidative stress and inflammation markers while enhancing anti-aging protein expression. In contrast, forced exercise showed diminished benefits. These insights underscore the importance of exercise intensity and preference in preserving retinal health during aging. The study highlights the potential of recreational physical activity as a non-invasive strategy to counteract retinal aging, advocating for further research into exercise regimens as preventative therapies for age-related ocular degenerations.
Collapse
Affiliation(s)
- Anna Szilágyi
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Barbara Takács
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Réka Szekeres
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Vera Tarjányi
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Dávid Nagy
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Dániel Priksz
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Mariann Bombicz
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Rita Kiss
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Adrienn Mónika Szabó
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Andrea Lehoczki
- Departments of Hematology and Stem Cell Transplantation, South Pest Central Hospital, National Institute of Hematology and Infectious Diseases, Saint Ladislaus Campus, Budapest, Hungary
- Department of Public Health, Semmelweis University, Budapest, Hungary
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary
| | - Rudolf Gesztelyi
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Béla Juhász
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Zoltán Szilvássy
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary
| | - Balázs Varga
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98., 4032, Debrecen, Hungary.
| |
Collapse
|
2
|
Datta C, Das P, Dutta S, Prasad T, Banerjee A, Gehlot S, Ghosal A, Dhabal S, Biswas P, De D, Chaudhuri S, Bhattacharjee A. AMPK activation reduces cancer cell aggressiveness via inhibition of monoamine oxidase A (MAO-A) expression/activity. Life Sci 2024; 352:122857. [PMID: 38914305 DOI: 10.1016/j.lfs.2024.122857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/14/2024] [Accepted: 06/16/2024] [Indexed: 06/26/2024]
Abstract
AIM AMPK can be considered as an important target molecule for cancer for its unique ability to directly recognize cellular energy status. The main aim of this study is to explore the role of different AMPK activators in managing cancer cell aggressiveness and to understand the mechanistic details behind the process. MAIN METHODS First, we explored the AMPK expression pattern and its significance in different subtypes of lung cancer by accessing the TCGA data sets for LUNG, LUAD and LUSC patients and then established the correlation between AMPK expression pattern and overall survival of lung cancer patients using Kaplan-Meire plot. We further carried out several cell-based assays by employing different wet lab techniques including RT-PCR, Western Blot, proliferation, migration and invasion assays to fulfil the aim of the study. KEY FINDINGS SIGNIFICANCE: This study identifies the importance of AMPK activators as a repurposing agent for combating lung and colon cancer cell aggressiveness. It also suggests SRT-1720 as a potent repurposing agent for cancer treatment especially in NSCLC patients where a point mutation is present in LKB1.
Collapse
Affiliation(s)
- Chandreyee Datta
- Department of Biotechnology, National Institute of Technology, Durgapur, Mahatma Gandhi Avenue, 713209 Burdwan, West Bengal, India
| | - Payel Das
- Department of Biotechnology, National Institute of Technology, Durgapur, Mahatma Gandhi Avenue, 713209 Burdwan, West Bengal, India
| | - Subhajit Dutta
- Department of Biotechnology, National Institute of Technology, Durgapur, Mahatma Gandhi Avenue, 713209 Burdwan, West Bengal, India
| | - Tuhina Prasad
- Department of Biotechnology, National Institute of Technology, Durgapur, Mahatma Gandhi Avenue, 713209 Burdwan, West Bengal, India
| | - Abhineet Banerjee
- Department of Biotechnology, National Institute of Technology, Durgapur, Mahatma Gandhi Avenue, 713209 Burdwan, West Bengal, India
| | - Sameep Gehlot
- Department of Biotechnology, National Institute of Technology, Durgapur, Mahatma Gandhi Avenue, 713209 Burdwan, West Bengal, India
| | - Arpa Ghosal
- Department of Biotechnology, National Institute of Technology, Durgapur, Mahatma Gandhi Avenue, 713209 Burdwan, West Bengal, India
| | - Sukhamoy Dhabal
- Department of Biotechnology, National Institute of Technology, Durgapur, Mahatma Gandhi Avenue, 713209 Burdwan, West Bengal, India
| | - Pritam Biswas
- Department of Biotechnology, National Institute of Technology, Durgapur, Mahatma Gandhi Avenue, 713209 Burdwan, West Bengal, India
| | - Debojyoti De
- Department of Biotechnology, National Institute of Technology, Durgapur, Mahatma Gandhi Avenue, 713209 Burdwan, West Bengal, India
| | - Surabhi Chaudhuri
- Department of Biotechnology, National Institute of Technology, Durgapur, Mahatma Gandhi Avenue, 713209 Burdwan, West Bengal, India
| | - Ashish Bhattacharjee
- Department of Biotechnology, National Institute of Technology, Durgapur, Mahatma Gandhi Avenue, 713209 Burdwan, West Bengal, India.
| |
Collapse
|
3
|
Li Q, Jiang L, Feng J, Wang X, Wang X, Xu X, Chu W. Aged polystyrene microplastics exacerbate alopecia associated with tight junction injuries and apoptosis via oxidative stress pathway in skin. ENVIRONMENT INTERNATIONAL 2024; 186:108638. [PMID: 38593689 DOI: 10.1016/j.envint.2024.108638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/13/2024] [Accepted: 04/06/2024] [Indexed: 04/11/2024]
Abstract
Microplastics (MPs) are pervasive pollutants in the natural environment and contribute to increased levels of illness in both animals and humans. However, thespecific impacts of MPs on skin damage and alopeciaare not yet well understood. In this study, we have examined the effects of two types of polystyrene MPs (pristine and aged) on skin and hair follicle damage in mice. UV irradiation changed the chemical and physical properties of the aged MPs, including functional groups, surface roughness, and contact angles. In both in vivo and in vitro experiments, skin and cell injuries related to oxidative stress, apoptosis, tight junctions (TJs), alopecia, mitochondrial dysfunction, and other damages were observed. Mechanistically, MPs and aged MPs can induce TJs damage via the oxidative stress pathway and inhibition of antioxidant-related proteins, and this can lead to alopecia. The regulation of cell apoptosis was also observed, and this is involved in the ROS-mediated mitochondrial signaling pathway. Importantly, aged MPs showed exacerbated toxicity, which may be due to their elevated surface irregularities and altered chemical compositions. Collectively, this study suggests a potential therapeutic approach for alopecia and hair follicle damage caused by MPs pollution.
Collapse
Affiliation(s)
- Qian Li
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Lehua Jiang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Jianhai Feng
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Xinhui Wang
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Xusheng Wang
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Xuejuan Xu
- Department of Endocrinology, The First People's Hospital of Foshan, Foshan, Guangdong, China
| | - Weiwei Chu
- Department of Plastic Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China.
| |
Collapse
|
4
|
Lu X, Hong J, Zhang J, Liu Q, Liao G, Shi Y, Tang H, Liu X. Triphenyl phosphate disrupts placental tryptophan metabolism by activating MAOA/ROS/NFκB. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 904:166688. [PMID: 37659542 DOI: 10.1016/j.scitotenv.2023.166688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/05/2023] [Accepted: 08/28/2023] [Indexed: 09/04/2023]
Abstract
Triphenyl phosphate (TPhP) is an organophosphate flame retardant widely distributed in the environment. The neurodevelopmental toxicity of TPhP has been observed in animals and humans. Previously, we found that prenatal TPhP exposure disturbed placental tryptophan metabolism, impaired neurodevelopment in male offspring, and induced abnormal neurobehavior; however, the underlying mechanisms are unknown. In this study, using the trophoblast cell line JEG-3, we found that TPhP altered gene and protein expression in the tryptophan metabolism pathway, inhibited the tryptophan-serotonin pathway, and activated the tryptophan-kynurenine pathway. Meanwhile, TPhP induced oxidative stress by activating monoamine oxidase A (MAOA), promoting inflammatory factors including nuclear factor kappa-B (NFκB), interleukin-6, and tumor necrosis factor α. The NFκB inhibitor sulfasalazine could alleviate the effects of TPhP on tryptophan metabolism disturbance. The MAOA inhibitor clorgyline or the antioxidant N-acetylcysteine can mitigate oxidative stress and eliminate TPhP-induced inflammatory factors and tryptophan metabolism disturbances. The data above suggest that TPhP disturbed tryptophan metabolism by activating NFκB through MAOA-mediated oxidative stress. Finally, using the mouse intrauterine exposure model, the results confirmed that TPhP induced oxidative stress, activated inflammatory factors, disturbed tryptophan metabolism, and increased the levels of the tryptophan metabolites serotonin, kynurenine, 3-hydroxykynurenine, and 3-hydroxyanthranilic acid in the placenta during the second trimester of pregnancy. Overall, TPhP can disturb placental tryptophan metabolism by activating the inflammatory factor NFκB, which was induced by MAOA-induced oxidative stress. The results of this study confirm that indirect exposure to xenobiotic compounds at an early life stage can impair offspring development and provide a novel perspective on the neurodevelopmental toxicity of TPhP.
Collapse
Affiliation(s)
- Xiaoxun Lu
- The First Dongguan Affiliated Hospital, Dongguan Key Laboratory of Environmental Medicine, Guangdong Medical University, Guangdong 523-808, China
| | - Jiabin Hong
- The Third People's Hospital of Zhuhai, Zhuhai 519000, Guangdong, China
| | - Jing Zhang
- The First Dongguan Affiliated Hospital, Dongguan Key Laboratory of Environmental Medicine, Guangdong Medical University, Guangdong 523-808, China
| | - Qian Liu
- The First Dongguan Affiliated Hospital, Dongguan Key Laboratory of Environmental Medicine, Guangdong Medical University, Guangdong 523-808, China
| | - Ganzhong Liao
- The First Dongguan Affiliated Hospital, Dongguan Key Laboratory of Environmental Medicine, Guangdong Medical University, Guangdong 523-808, China
| | - Yanwei Shi
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Huanwen Tang
- The First Dongguan Affiliated Hospital, Dongguan Key Laboratory of Environmental Medicine, Guangdong Medical University, Guangdong 523-808, China.
| | - Xiaoshan Liu
- The First Dongguan Affiliated Hospital, Dongguan Key Laboratory of Environmental Medicine, Guangdong Medical University, Guangdong 523-808, China.
| |
Collapse
|
5
|
Long X, Wu H, Zhou Y, Wan Y, Kan X, Gong J, Zhao X. Preventive Effect of Limosilactobacillus fermentum SCHY34 on Lead Acetate-Induced Neurological Damage in SD Rats. Front Nutr 2022; 9:852012. [PMID: 35571929 PMCID: PMC9094495 DOI: 10.3389/fnut.2022.852012] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 04/04/2022] [Indexed: 02/03/2023] Open
Abstract
Lead poisoning caused by lead pollution seriously affects people's health. Lactic acid bacteria has been shown to be useful for biological scavenging of lead. In this experiment, Sprague-Dawley (SD) rats were treated with 200 mg/L of lead acetate solution daily to induce chronic lead poisoning, and oral Limosilactobacillus fermentum (L. fermentum) SCHY34 to study its mitigation effects and mechanisms on rat neurotoxicity. The L. fermentum SCHY34 showed competent results on in vitro survival rate and the lead ion adsorption rate. Animal experiments showed that L. fermentum SCHY34 maintained the morphology of rat liver, kidney, and hippocampi, reduced the accumulation of lead in the blood, liver, kidney, and brain tissue. Further, L. fermentum SCHY34 alleviated the lead-induced decline in spatial memory and response capacity of SD rats, and also regulated the secretion of neurotransmitters and related enzyme activities in the brain tissue of rats, such as glutamate (Glu), monoamine oxidase (MAO), acetylcholinesterase (AchE), cyclic adenosine monophosphate (cAMP), and adenylate cyclase (AC). In addition, the expression of genes related to cognitive capacity, antioxidation, and anti-apoptotic in rat brain tissues were increased L. fermentum SCHY34 treatment, such as brain-derived neurotrophic factor (BDNF), c-fos, c-jun, superoxide dismutase (SOD)1/2, Nuclear factor erythroid 2-related factor 2 (Nrf2), and B-cell lymphoma 2 (Bcl-2), and so on. L. fermentum SCHY34 showed a great biological scavenging and potential effect on alleviating the toxicity of lead ions.
Collapse
Affiliation(s)
- Xingyao Long
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing Engineering Research Center of Functional Food, Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing, China
| | - Haibo Wu
- Department of Neurosurgery, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| | - Yujing Zhou
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing Engineering Research Center of Functional Food, Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing, China
| | - Yunxiao Wan
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing Engineering Research Center of Functional Food, Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing, China
| | - Xuemei Kan
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing Engineering Research Center of Functional Food, Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing, China
| | - Jianjun Gong
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing Engineering Research Center of Functional Food, Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing, China
| | - Xin Zhao
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing Engineering Research Center of Functional Food, Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing, China
| |
Collapse
|
6
|
Bathini M, Raghushaker CR, Mahato KK. The Molecular Mechanisms of Action of Photobiomodulation Against Neurodegenerative Diseases: A Systematic Review. Cell Mol Neurobiol 2022. [PMID: 33301129 DOI: 10.1007/s10571-020-01016-9,33301129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
Neurodegenerative diseases might be slow but relentless, as we continue to fail in treating or delaying their progression. Given the complexity in the pathogenesis of these diseases, a broad-acting approach like photobiomodulation can prove promising. Photobiomodulation (PBM) uses red and infrared light for therapeutic benefits, working by stimulating growth and proliferation. The implications of photobiomodulation have been studied in several neurodegenerative disease models. It has been shown to improve cell survival, decrease apoptosis, alleviate oxidative stress, suppress inflammation, and rescue mitochondrial function. In in vivo models, it has reportedly preserved motor and cognitive skills. Beyond mitochondrial stimulation, the molecular mechanisms by which photobiomodulation protects against neurodegeneration have not been very well studied. This review has systematically been undertaken to study the effects of photobiomodulation at a molecular level and identify the different biochemical pathways and molecular changes in the process. The data showed the involvement of pathways like extracellular signal-regulated kinase (ERK), mitogen-activated protein kinase (MAPK), and protein kinase B (Akt). In addition, the expression of several genes and proteins playing different roles in the disease mechanisms was found to be influenced by PBM, such as neurotrophic factors and secretases. Studying the literature indicated that PBM can be translated to a potential therapeutic tool, acting through a spectrum of mechanisms that work together to decelerate disease progression in the organism, which is difficult to achieve through pharmacological interventions.
Collapse
Affiliation(s)
- Mayukha Bathini
- Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Chandavalli Ramappa Raghushaker
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Krishna Kishore Mahato
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
7
|
Szilágyi A, Takács B, Szekeres R, Tarjányi V, Bombicz M, Priksz D, Kovács A, Juhász B, Frecska E, Szilvássy Z, Varga B. Therapeutic Properties of Ayahuasca Components in Ischemia/Reperfusion Injury of the Eye. Biomedicines 2022; 10:997. [PMID: 35625734 PMCID: PMC9138933 DOI: 10.3390/biomedicines10050997] [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: 02/21/2022] [Revised: 04/13/2022] [Accepted: 04/23/2022] [Indexed: 02/06/2023] Open
Abstract
Ischemic eye diseases are major causes of vision impairment. Thus, potential retinoprotective effects of N'N-dimethyltryptamine (DMT) were investigated. To inhibit its rapid breakdown by monoamine-oxidase A (MAO-A) enzyme, DMT was co-administered with harmaline, a β-carboline in the Amazonian Ayahuasca brew. Using ligation, 60 min of ischemia was provoked in eyes of rats, followed by 7 days of reperfusion whilst animals received harmaline alone, DMT + harmaline, or vehicle treatment. After 1 week of reperfusion, electroretinographical (ERG) measurements, histological analysis, and Western blot were performed. Harmaline alone exhibited retinoprotection in ischemia-reperfusion (I/R) which was, surprisingly, counterbalanced by DMT in case of co-administration. As both MAO-A inhibition and DMT increase serotoninergic tone synergistically, communicated to be anti-ischemic, thus, involvement of other pathways was investigated. Based on our experiments, DMT and harmaline exert opposite effects on important ocular proteins such as PARP1, NFκB, MMP9, or HSP70, each having a critical role in a different mechanism of eye-ischemia-related pathologies, e.g., cell death, inflammation, tissue destruction, and oxidative stress. Since DMT is proclaimed to be a promising drug candidate, its potentially undesirable effect on eye-ischemia should be further investigated. Meanwhile, this experiment revealed the potential therapeutic effect of MAO-A inhibitor harmaline in I/R-related eye diseases.
Collapse
Affiliation(s)
- Anna Szilágyi
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Nagyerdei Krt 98, H-4032 Debrecen, Hungary; (A.S.); (B.T.); (R.S.); (V.T.); (M.B.); (D.P.); (B.J.); (Z.S.)
| | - Barbara Takács
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Nagyerdei Krt 98, H-4032 Debrecen, Hungary; (A.S.); (B.T.); (R.S.); (V.T.); (M.B.); (D.P.); (B.J.); (Z.S.)
| | - Réka Szekeres
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Nagyerdei Krt 98, H-4032 Debrecen, Hungary; (A.S.); (B.T.); (R.S.); (V.T.); (M.B.); (D.P.); (B.J.); (Z.S.)
| | - Vera Tarjányi
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Nagyerdei Krt 98, H-4032 Debrecen, Hungary; (A.S.); (B.T.); (R.S.); (V.T.); (M.B.); (D.P.); (B.J.); (Z.S.)
| | - Mariann Bombicz
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Nagyerdei Krt 98, H-4032 Debrecen, Hungary; (A.S.); (B.T.); (R.S.); (V.T.); (M.B.); (D.P.); (B.J.); (Z.S.)
| | - Dániel Priksz
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Nagyerdei Krt 98, H-4032 Debrecen, Hungary; (A.S.); (B.T.); (R.S.); (V.T.); (M.B.); (D.P.); (B.J.); (Z.S.)
| | - Attila Kovács
- Department of Psychiatry, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98, H-4032 Debrecen, Hungary; (A.K.); (E.F.)
| | - Béla Juhász
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Nagyerdei Krt 98, H-4032 Debrecen, Hungary; (A.S.); (B.T.); (R.S.); (V.T.); (M.B.); (D.P.); (B.J.); (Z.S.)
| | - Ede Frecska
- Department of Psychiatry, Faculty of Medicine, University of Debrecen, Nagyerdei Krt 98, H-4032 Debrecen, Hungary; (A.K.); (E.F.)
| | - Zoltán Szilvássy
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Nagyerdei Krt 98, H-4032 Debrecen, Hungary; (A.S.); (B.T.); (R.S.); (V.T.); (M.B.); (D.P.); (B.J.); (Z.S.)
| | - Balázs Varga
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Nagyerdei Krt 98, H-4032 Debrecen, Hungary; (A.S.); (B.T.); (R.S.); (V.T.); (M.B.); (D.P.); (B.J.); (Z.S.)
| |
Collapse
|
8
|
Chen W, Liang R, Yi Y, Zhu J, Zhang J. P38α deficiency in macrophages ameliorates murine experimental colitis by regulating inflammation and immune process. Pathol Res Pract 2022; 233:153881. [DOI: 10.1016/j.prp.2022.153881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 03/23/2022] [Accepted: 04/01/2022] [Indexed: 11/28/2022]
|
9
|
Wu KC, Lee CY, Chern Y, Lin CJ. Amelioration of lipopolysaccharide-induced memory impairment in equilibrative nucleoside transporter-2 knockout mice is accompanied by the changes in glutamatergic pathways. Brain Behav Immun 2021; 96:187-199. [PMID: 34058310 DOI: 10.1016/j.bbi.2021.05.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 05/18/2021] [Accepted: 05/26/2021] [Indexed: 11/17/2022] Open
Abstract
Neuroinflammation has been implicated in cognitive deficits in neurological and neurodegenerative diseases. Lipopolysaccharide (LPS)-induced neuroinflammation and the breakdown of the blood-brain barrier can be attenuated in mice with equilibrative nucleoside transporter-2 (ENT2/Ent2) deletion. The present study was aimed to investigate the role of ENT2 in cognitive and neuronal functions under physiological and inflammatory conditions, in terms of behavioral performance and synaptic plasticity in saline- and LPS-treated Ent2 knockout (KO) mice and their wild-type (WT) littermate controls. Repeated administrations of LPS significantly impaired spatial memory formation in Morris water maze and hippocampal-dependent long-term potentiation (LTP) in WT mice. The LPS-treated WT mice exhibited significant synaptic and neuronal damage in the hippocampus. Notably, the LPS-induced impairment in spatial memory and LTP performance were attenuated in Ent2 KO mice, along with the preservation of neuronal survival. The beneficial effects were accompanied by the normalization of excessive extracellular glutamate and aberrant downstream signaling of glutamate receptor activation, including the upregulation of phosphorylated p38 mitogen-activated protein kinase and the downregulation of phosphorylated cyclic adenosine monophosphate-response element-binding protein. There was no significant difference in behavioral outcome and all tested parameters between these two genotypes under physiological condition. These results suggest that ENT2 plays an important role in regulating inflammation-associated cognitive decline and neuronal damage.
Collapse
Affiliation(s)
- Kuo-Chen Wu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chih-Yu Lee
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yijuang Chern
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chun-Jung Lin
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
10
|
Jones DN, Raghanti MA. The role of monoamine oxidase enzymes in the pathophysiology of neurological disorders. J Chem Neuroanat 2021; 114:101957. [PMID: 33836221 DOI: 10.1016/j.jchemneu.2021.101957] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 04/03/2021] [Accepted: 04/04/2021] [Indexed: 12/12/2022]
Abstract
Monoamine oxidase enzymes are responsible for the degredation of serotonin, dopamine, and norepinephrine in the central neurvous system. Although it has been nearly 100 years since they were first described, we are still learning about their role in the healthy brain and how they are altered in various disease states. The present review provides a survey of our current understanding of monoamine oxidases, with a focus on their contributions to neuropsychiatric, neurodevelopmental, and neurodegenerative disease. Important species differences in monoamine oxidase function and development in the brain are highlighted. Sex-specific monoamine oxidase regulatory mechanisms and their implications for various neurological disorders are also discussed. While our understanding of these critical enzymes has expanded over the last century, gaps exist in our understanding of sex and species differences and the roles monoamine oxidases may play in conditions often comorbid with neurological disorders.
Collapse
Affiliation(s)
- Danielle N Jones
- Department of Anthropology and School of Biomedical Sciences, Kent State University, Kent, OH, USA; Brain Health Research Institute, Kent State University, Kent, OH, USA.
| | - Mary Ann Raghanti
- Department of Anthropology and School of Biomedical Sciences, Kent State University, Kent, OH, USA; Brain Health Research Institute, Kent State University, Kent, OH, USA
| |
Collapse
|
11
|
Bathini M, Raghushaker CR, Mahato KK. The Molecular Mechanisms of Action of Photobiomodulation Against Neurodegenerative Diseases: A Systematic Review. Cell Mol Neurobiol 2020; 42:955-971. [PMID: 33301129 PMCID: PMC8942959 DOI: 10.1007/s10571-020-01016-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 11/18/2020] [Indexed: 01/08/2023]
Abstract
Neurodegenerative diseases might be slow but relentless, as we continue to fail in treating or delaying their progression. Given the complexity in the pathogenesis of these diseases, a broad-acting approach like photobiomodulation can prove promising. Photobiomodulation (PBM) uses red and infrared light for therapeutic benefits, working by stimulating growth and proliferation. The implications of photobiomodulation have been studied in several neurodegenerative disease models. It has been shown to improve cell survival, decrease apoptosis, alleviate oxidative stress, suppress inflammation, and rescue mitochondrial function. In in vivo models, it has reportedly preserved motor and cognitive skills. Beyond mitochondrial stimulation, the molecular mechanisms by which photobiomodulation protects against neurodegeneration have not been very well studied. This review has systematically been undertaken to study the effects of photobiomodulation at a molecular level and identify the different biochemical pathways and molecular changes in the process. The data showed the involvement of pathways like extracellular signal-regulated kinase (ERK), mitogen-activated protein kinase (MAPK), and protein kinase B (Akt). In addition, the expression of several genes and proteins playing different roles in the disease mechanisms was found to be influenced by PBM, such as neurotrophic factors and secretases. Studying the literature indicated that PBM can be translated to a potential therapeutic tool, acting through a spectrum of mechanisms that work together to decelerate disease progression in the organism, which is difficult to achieve through pharmacological interventions.
Collapse
Affiliation(s)
- Mayukha Bathini
- Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Chandavalli Ramappa Raghushaker
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Krishna Kishore Mahato
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
12
|
Liu J, Liu J, Bai M, Wang H. Protective effect of puerarin against burn-induced heart injury in rats. Exp Ther Med 2020; 20:275-282. [PMID: 32536996 PMCID: PMC7282049 DOI: 10.3892/etm.2020.8696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 02/07/2020] [Indexed: 12/13/2022] Open
Abstract
The present study evaluated the potential protective effects of puerarin and its associated mechanism on burn-induced myocardial damage. A total of 40 healthy adult Wistar rats were randomly divided into four groups: i) Sham; ii) burn; iii) burn + puerarin; and iv) puerarin. Serum levels of interleukin (IL)-1β, tumor necrosis factor-α (TNF-α) and IL-6 were measured using ELISA. Myeloperoxidase (MPO) activity and malondialdehyde (MDA) levels were determined in myocardial homogenates using a commercial assay kit. TUNEL staining and western blot analysis of cleaved and pro-caspase-3 were also performed to assess apoptosis. Activation of p38-MAPK, ERK, JNK and AKT were measured using western blot analysis. Left ventricular systolic pressure, maximum rates of increase/decrease in left ventricular pressure, creatine kinase MB activity and cardiac troponin T levels were found to be altered in the burn group 12 h after burn, which were reversed by puerarin treatment. Injection of puerarin following burn injury also reduced heart water content. Serum levels of IL-1β, TNF-α and IL-6 were significantly higher in the burn group compared with those in the sham group. Puerarin treatment reduced serum levels of IL-1β, TNF-α and IL-6, in addition to reducing MPO activity and MDA levels in myocardial tissues. Puerarin inhibited the activation of caspase-3, p38, ERK and JNK following severe burn, but elevated Akt activation following severe burn. In conclusion, puerarin improved cardiac function in rats following severe burn injury, which may be due to reduced myocardial injury, inhibition of cardiomyocyte apoptosis and reduced oxidative inflammatory stress; the MAPK and AKT signaling pathways are proposed to the underlying mechanism of these findings.
Collapse
Affiliation(s)
- Junling Liu
- Department of Burn, Gansu Provincial Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Jianyun Liu
- Department of Burn, Gansu Provincial Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Mingming Bai
- Department of Burn, Gansu Provincial Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Hui Wang
- Department of Burn, Gansu Provincial Hospital, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
13
|
Godisela KK, Reddy SS, Reddy PY, Kumar CU, Reddy VS, Ayyagari R, Reddy GB. Role of sorbitol-mediated cellular stress response in obesity-associated retinal degeneration. Arch Biochem Biophys 2020; 679:108207. [PMID: 31760123 DOI: 10.1016/j.abb.2019.108207] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 11/16/2019] [Accepted: 11/20/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE Obesity is a global health problem associated with several diseases including ocular complications. Earlier we reported progressive retinal degeneration because of obesity in a spontaneous obese rat (WNIN/Ob) model. In the current study, we examined the molecular mechanisms leading to retinal degeneration in WNIN/Ob rat. METHODS Sorbitol was estimated by the fluorometric method in the retina of WNIN/Ob rats at different age (3-, 6- and 12- months), along with their respective lean rats. Immunoblotting was performed in the retina to assess the status of the insulin signaling pathway, ER stress and cellular stress (p38MAPK and ERK1/2). Human SK-N-SH cells were treated with 0.5 and 1.0 M sorbitol for 30 min to study insulin signaling, ER stress, and cellular stress. TUNEL assay was done to measure apoptosis. The retinal function in the rats was determined by electroretinogram. RESULTS A gradual but significantly higher intracellular sorbitol accumulation was observed in the retina of obese rats from 3- to 12-months. The cellular osmotic stress has activated the insulin signaling mechanism without activating AKT and also triggered ER stress. Both the stresses activated the ERK and p38MAPK signaling causing apoptosis in the retina leading to retinal degeneration. Retinal dysfunction was confirmed by altered scotopic and photopic electroretinogram responses. These in vivo results were mimicked in SK-N-SH cells when exposed to sorbitol in vitro. CONCLUSIONS These results suggest cellular stress due to sorbitol accumulation impairing the ER function, thereby leading to progressive retinal degeneration under obese conditions.
Collapse
Affiliation(s)
- Kishore K Godisela
- Biochemistry Division, National Institute of Nutrition, Hyderabad, India
| | | | - P Yadagiri Reddy
- Biochemistry Division, National Institute of Nutrition, Hyderabad, India
| | - Ch Uday Kumar
- Biochemistry Division, National Institute of Nutrition, Hyderabad, India
| | - V Sudhakar Reddy
- Biochemistry Division, National Institute of Nutrition, Hyderabad, India
| | - Radha Ayyagari
- Shiley Eye Institute, University of California San Diego, La Jolla, CA, USA
| | | |
Collapse
|
14
|
Babbar M, Huang Y, Curtiss CM, Sheikh MS. CHTM1 regulates cancer cell sensitivity to metabolic stress via p38-AIF1 pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:271. [PMID: 31221176 PMCID: PMC6587271 DOI: 10.1186/s13046-019-1253-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 05/28/2019] [Indexed: 11/10/2022]
Abstract
BACKGROUND Recently, we have reported the characterization of a novel protein named Coiled-coil Helix Tumor and Metabolism 1 (CHTM1). CHTM1 localizes to both cytosol and mitochondria. Sequence corresponding to CHTM1 is also annotated in the database as CHCHD5. CHTM1 is deregulated in human breast and colon cancers and its deficiency in human cancer cells leads to defective lipid metabolism and poor growth under glucose/glutamine starvation. METHODS Human cancer cell lines and tissue specimens were used. CHTM1 knockdown was done via lentiviral approach. CHTM1-expresssion constructs were developed and mutants were generated via site-directed mutagenesis approach. Western blotting, immunostaining, immunohistochemistry, cell fractionation and luciferase assays were performed. Reactive oxygen species and reactive nitrogen species were also measured. RESULTS Here we report that CHTM1 deficiency sensitizes human lung cancer cells to metabolic stress-induced cell death mediated by glucose/glutamine deprivation and metformin treatment. CHTM1 interacts with Apoptosis Inducing Factor 1 (AIF1) that is one of the important death inducing molecules. CHTM1 appears to negatively regulate AIF1 by preventing AIF1 translocation to cytosol/nucleus and thereby inhibit AIF1-mediated caspase-independent cell death. Our results also indicate that p38, a stress kinase, plays a critical role in metabolic stress-induced cell death in CHTM1-deficient cells. Furthermore, p38 appears to enhance AIF1 translocation from mitochondria to cytosol particularly in metabolically stressed CHTM1-deficient cells and CHTM1 negatively regulates p38 kinase activity. The expression status of CHTM1 in lung cancer patient samples is also investigated and our results indicate that CHTM1 levels are increased in the majority of lung tumors when compared to their matching normal tissues. CONCLUSION Thus, CHTM1 appears to be an important metabolic marker that regulates cancer cell survival under metabolic stress conditions, and has the potential to be developed as a predictive tumor marker.
Collapse
Affiliation(s)
- Mansi Babbar
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA.,Present address: Mansi Babbar, Laboratory of Molecular Gerontology, National Institute on Aging, NIH, Baltimore, MD, 21224, USA
| | - Ying Huang
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Christopher M Curtiss
- Department of Pathology, State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA
| | - M Saeed Sheikh
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA.
| |
Collapse
|
15
|
Choi JY, Yun J, Hwang CJ, Lee HP, Kim HD, Chun H, Park PH, Choi DY, Han SB, Hong JT. (E)-2-methoxy-4-(3-(4-methoxyphenyl) prop-1-en-1-yl) Phenol Ameliorates MPTP-Induced Dopaminergic Neurodegeneration by Inhibiting the STAT3 Pathway. Int J Mol Sci 2019; 20:ijms20112632. [PMID: 31146332 PMCID: PMC6600543 DOI: 10.3390/ijms20112632] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 01/08/2023] Open
Abstract
Neuroinflammation is implicated in dopaminergic neurodegeneration. We have previously demonstrated that (E)-2-methoxy-4-(3-(4-methoxyphenyl) prop-1-en-1-yl) phenol (MMPP), a selective signal transducer and activator of transcription 3 (STAT3) inhibitor, has anti-inflammatory properties in several inflammatory disease models. We investigated whether MMPP could protect against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced dopaminergic cell loss and behavioral impairment. Imprinting control region (ICR) mice (8 weeks old, n = 10 per group) were administered MMPP (5 mg/kg) in drinking water for 1 month, and injected with MPTP (15 mg/kg, four times with 2 h intervals) during the last 7 days of treatment. MMPP decreased MPTP-induced behavioral impairments in rotarod, pole, and gait tests. We also showed that MMPP ameliorated dopamine depletion in the striatum and inflammatory marker elevation in primary cultured neurons by high-performance liquid chromatography and immunohistochemical analysis. Increased activation of STAT3, p38, and monoamine oxidase B (MAO-B) were observed in the substantia nigra and striatum after MPTP injection, effects that were attenuated by MMPP treatment. Furthermore, MMPP inhibited STAT3 activity and expression of neuroinflammatory proteins, including ionized calcium binding adaptor molecule 1 (Iba1), inducible nitric oxide synthase (iNOS), and glial fibrillary acidic protein (GFAP) in 1-methyl-4-phenylpyridinium (MPP+; 0.5 mM)-treated primary cultured cells. However, mitogen-activated protein kinase (MAPK) inhibitors augmented the activity of MMPP. Collectively, our results suggest that MMPP may be an anti-inflammatory agent that attenuates dopaminergic neurodegeneration and neuroinflammation through MAO-B and MAPK pathway-dependent inhibition of STAT3 activation.
Collapse
Affiliation(s)
- Ji Yeon Choi
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Cheongju 28160, Korea.
| | - Jaesuk Yun
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Cheongju 28160, Korea.
| | - Chul Ju Hwang
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Cheongju 28160, Korea.
| | - Hee Pom Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Cheongju 28160, Korea.
| | - Hae Deun Kim
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Cheongju 28160, Korea.
| | - Hyungok Chun
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Cheongju 28160, Korea.
| | - Pil-Hoon Park
- College of Pharmacy, Yeungnam University, 280, Daehak-ro, Gyeongsan, Gyeongbuk 38541, Korea.
| | - Dong Young Choi
- College of Pharmacy, Yeungnam University, 280, Daehak-ro, Gyeongsan, Gyeongbuk 38541, Korea.
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Cheongju 28160, Korea.
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31 Osongsaemgmyeong 1-ro, Cheongju 28160, Korea.
| |
Collapse
|
16
|
Agi E, Namvar A, Moradi S, Bolhassani A. Expression of the Neuron-Related Genes in hNGF-β-Treated PC12 Cells. NEUROPHYSIOLOGY+ 2019. [DOI: 10.1007/s11062-019-09770-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
17
|
Ugun-Klusek A, Theodosi TS, Fitzgerald JC, Burté F, Ufer C, Boocock DJ, Yu-Wai-Man P, Bedford L, Billett EE. Monoamine oxidase-A promotes protective autophagy in human SH-SY5Y neuroblastoma cells through Bcl-2 phosphorylation. Redox Biol 2019; 20:167-181. [PMID: 30336354 PMCID: PMC6197572 DOI: 10.1016/j.redox.2018.10.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/05/2018] [Accepted: 10/06/2018] [Indexed: 02/07/2023] Open
Abstract
Monoamine oxidases (MAOs) are located on the outer mitochondrial membrane and are drug targets for the treatment of neurological disorders. MAOs control the levels of neurotransmitters in the brain via oxidative deamination and contribute to reactive oxygen species (ROS) generation through their catalytic by-product H2O2. Increased ROS levels may modulate mitochondrial function and mitochondrial dysfunction is implicated in a vast array of disorders. However, the downstream effects of MAO-A mediated ROS production in a neuronal model has not been previously investigated. In this study, using MAO-A overexpressing neuroblastoma cells, we demonstrate that higher levels of MAO-A protein/activity results in increased basal ROS levels with associated increase in protein oxidation. Increased MAO-A levels result in increased Lysine-63 linked ubiquitination of mitochondrial proteins and promotes autophagy through Bcl-2 phosphorylation. Furthermore, ROS generated locally on the mitochondrial outer membrane by MAO-A promotes phosphorylation of dynamin-1-like protein, leading to mitochondrial fragmentation and clearance without complete loss of mitochondrial membrane potential. Cellular ATP levels are maintained following MAO-A overexpression and complex IV activity/protein levels increased, revealing a close relationship between MAO-A levels and mitochondrial function. Finally, the downstream effects of increased MAO-A levels are dependent on the availability of amine substrates and in the presence of exogenous substrate, cell viability is dramatically reduced. This study shows for the first time that MAO-A generated ROS is involved in quality control signalling, and increase in MAO-A protein levels leads to a protective cellular response in order to mediate removal of damaged macromolecules/organelles, but substrate availability may ultimately determine cell fate. The latter is particularly important in conditions such as Parkinson's disease, where a dopamine precursor is used to treat disease symptoms and highlights that the fate of MAO-A containing dopaminergic neurons may depend on both MAO-A levels and catecholamine substrate availability.
Collapse
Affiliation(s)
- Aslihan Ugun-Klusek
- School of Science and Technology, Nottingham Trent University, Nottingham, UK.
| | | | - Julia C Fitzgerald
- Hertie-Institute for Clinical Brain Research, University of Tübingen and German Centre for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Florence Burté
- Wellcome Trust Centre for Mitochondrial Research, Institute of Genetic Medicine, Newcastle University, Newcastle Upon Tyne, UK
| | - Christoph Ufer
- Institute of Biochemistry, University Medicine Berlin-Charité, Berlin, Germany
| | - David J Boocock
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, UK
| | - Patrick Yu-Wai-Man
- NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology, London, UK; Cambridge Eye Unit, Addenbrooke's Hospital, Cambridge University Hospitals, Cambridge, UK; MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK; Cambridge Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Lynn Bedford
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - E Ellen Billett
- School of Science and Technology, Nottingham Trent University, Nottingham, UK
| |
Collapse
|
18
|
Yusufzai SK, Khan MS, Sulaiman O, Osman H, Lamjin DN. Molecular docking studies of coumarin hybrids as potential acetylcholinesterase, butyrylcholinesterase, monoamine oxidase A/B and β-amyloid inhibitors for Alzheimer's disease. Chem Cent J 2018; 12:128. [PMID: 30515636 PMCID: PMC6768047 DOI: 10.1186/s13065-018-0497-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 11/21/2018] [Indexed: 01/11/2023] Open
Abstract
Coumarins are the phytochemicals, which belong to the family of benzopyrone, that display interesting pharmacological properties. Several natural, synthetic and semisynthetic coumarin derivatives have been discovered in decades for their applicability as lead structures as drugs. Coumarin based conjugates have been described as potential AChE, BuChE, MAO and β-amyloid inhibitors. Therefore, the objective of this review is to focus on the construction of these pharmacologically important coumarin analogues with anti-Alzheimer’s activities, highlight their docking studies and structure–activity relationships based on their substitution pattern with respect to the selected positions on the chromen ring by emphasising on the research reports conducted in between year 1968 to 2017.![]()
Collapse
Affiliation(s)
- Samina Khan Yusufzai
- School of Industrial Technology, Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - Mohammad Shaheen Khan
- Industrial Chemistry Programme, Faculty of Science and Natural Resources, Universiti Malaysia Sabah, 88400, Kota Kinabalu, Sabah, Malaysia.
| | - Othman Sulaiman
- School of Industrial Technology, Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - Hasnah Osman
- School of Chemical Sciences, Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - Dalily Nabilah Lamjin
- Industrial Chemistry Programme, Faculty of Science and Natural Resources, Universiti Malaysia Sabah, 88400, Kota Kinabalu, Sabah, Malaysia
| |
Collapse
|
19
|
Hwang CJ, Lee HP, Choi DY, Jeong HS, Kim TH, Lee TH, Kim YM, Moon DB, Park SS, Kim SY, Oh KW, Hwang DY, Han SB, Lee HJ, Hong JT. Inhibitory effect of thiacremonone on MPTP-induced dopaminergic neurodegeneration through inhibition of p38 activation. Oncotarget 2018; 7:46943-46958. [PMID: 27409674 PMCID: PMC5216915 DOI: 10.18632/oncotarget.10504] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 06/30/2016] [Indexed: 12/11/2022] Open
Abstract
Neuroinflammation is implicated for dopaminergic neurodegeneration. Sulfur compounds extracted from garlic have been shown to have anti-inflammatory properties. Previously, we have investigated that thiacremonone, a sulfur compound isolated from garlic has anti-inflammatory effects on several inflammatory disease models. To investigate the protective effect of thiacremonone against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced behavioral impairment and dopaminergic neurodegeneration, 8 week old ICR mice were given thiacremonone (10 mg/kg) in drinking water for 1 month and received intraperitoneal injection of MPTP (15 mg/kg, four times with 2 h interval) during the last 7 days of treatment. Our data showed that thiacremonone decreased MPTP-induced behavioral impairments (Rotarod test, Pole test, and Gait test), dopamine depletion and microglia and astrocytes activations as well as neuroinflammation. Higher activation of p38 was found in the substantia nigra and striatum after MPTP injection, but p38 activation was reduced in thiacremonone treated group. In an in vitro study, thiacremonone (1, 2, and 5 μg/ml) effectively decreased MPP+ (0.5 mM)-induced glial activation, inflammatory mediators generation and dopaminergic neurodegeneration in cultured astrocytes and microglial BV-2 cells. Moreover, treatment of p38 MAPK inhibitor SB203580 (10 μM) further inhibited thiacremonone induced reduction of neurodegeneration and neuroinflammation. These results indicated that the anti-inflammatory compound, thiacremonone, inhibited neuroinflammation and dopaminergic neurodegeneration through inhibition of p38 activation.
Collapse
Affiliation(s)
- Chul Ju Hwang
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Hee Pom Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Dong-Young Choi
- College of Pharmacy, Yeungnam University, Daehak-Ro, Gyeongsan, Gyeongbuk, Republic of Korea
| | - Heon Sang Jeong
- College of Agriculture, Life and Environments Sciences, Chungbuk National University, Osongsaengmyeong, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Tae Hoon Kim
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Tae Hyung Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Young Min Kim
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Dae Bong Moon
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Sung Sik Park
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Sun Young Kim
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Ki-Wan Oh
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Dae Yeon Hwang
- College of Natural Resources & Life Science, Pusan National University, Pusan, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Hwa-Jeong Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong, Osong-eup, Heungduk-gu, Cheongju, Chungbuk, Republic of Korea
| |
Collapse
|
20
|
Naoi M, Maruyama W, Shamoto-Nagai M. Type A and B monoamine oxidases distinctly modulate signal transduction pathway and gene expression to regulate brain function and survival of neurons. J Neural Transm (Vienna) 2017; 125:1635-1650. [DOI: 10.1007/s00702-017-1832-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 12/18/2017] [Indexed: 02/01/2023]
|
21
|
Apoptosis signal-regulating kinase 1 mediates the inhibitory effect of hepatocyte nuclear factor-4α on hepatocellular carcinoma. Oncotarget 2017; 7:27408-21. [PMID: 27050273 PMCID: PMC5053659 DOI: 10.18632/oncotarget.8478] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 03/16/2016] [Indexed: 12/24/2022] Open
Abstract
Previous studies provided substantial evidence of a striking suppressive effect of hepatocyte nuclear factor 4α (HNF4α) on hepatocellular carcinoma (HCC). Apoptosis signal-regulating kinase 1 (ASK1) is involved in death receptor-mediated apoptosis and may acts as a tumor suppressor in hepatocarcinogenesis. However, the status and function of ASK1 during HCC progression are unclear. In this study, we found that HNF4α increased ASK1 expression by directly binding to its promoter. ASK1 expression was dramatically suppressed and correlated with HNF4α levels in HCC tissues. Reduced ASK1 expression was associated with aggressive tumors and poor prognosis for human HCC. Moreover, ASK1 inhibited the malignant phenotype of HCC cells in vitro. Intratumoral ASK1 injection significantly suppressed the growth of subcutaneous HCC xenografts in nude mice. More interestingly, systemic ASK1 delivery strikingly inhibited the growth of orthotopic HCC nodules in NOD/SCID mice. In addition, inhibition of endogenous ASK1 partially reversed the suppressive effects of HNF4α on HCC. Collectively, this study highlights the suppressive effect of ASK1 on HCC and its biological significance in HCC development. These outcomes broaden the knowledge of ASK1 function in HCC progression, and provide a novel potential prognostic biomarker and therapeutic target for advanced HCC.
Collapse
|
22
|
Hussain ASM, Renno WM, Sadek HL, Kayali NM, Al-Salem A, Rao MS, Khan KM. Monoamine oxidase-B inhibitor protects degenerating spinal neurons, enhances nerve regeneration and functional recovery in sciatic nerve crush injury model. Neuropharmacology 2017; 128:231-243. [PMID: 29054367 DOI: 10.1016/j.neuropharm.2017.10.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/28/2017] [Accepted: 10/15/2017] [Indexed: 12/29/2022]
Abstract
Monoamine oxidase-B (MAOB), a flavin adenine dinucleotide (FAD), is an enzyme which catalyzes the oxidation of amines. MAOB is proposed to play a major role in the pathogenesis of neurodegeneration through the production of reactive oxygen species (ROS) and neurotoxins. The present study was designed to outline the effects of the MAOB inhibitor (MAOB-I) on neuroprotection of spinal neurons, regeneration of sciatic nerve fibers, and recovery of sensory-motor functions in the sciatic nerve crush injury model. Male Wistar rats (4-months-old) were assigned to i) Naïve (N), ii) Sham (S), iii) Sciatic nerve crush and treated with saline (CRUSH + SALINE) and iv) Sciatic nerve crush and treated with MAOB inhibitor (CRUSH + MAOB-I) groups (n = 10/group). In groups iii and iv, the crush injury was produced by crushing the sciatic nerve followed by treatment with saline or MAOB-I (Selegiline® 2.5 mg/kg) intraperitoneally for 10 days. Behavioral tests were conducted from week 1 to week 6. At the end of the study, sciatic nerve and lumbar spinal cord were examined by immunohistochemistry, light and electron microscopy. MAOB-I treatment showed significant improvement in sensory and motor functions compared to saline treatment (p < 0.05-0.001) in injured nerves. The morphological study showed a significantly increased number of nerve fibers in sciatic nerve distal to the site of injury (p < 0.05), with better myelination pattern in CRUSH + MAOB-I treated group compared to CRUSH + SALINE group. Spinal cord ventral horns showed a significant increase in the number of NeuN-immunoreactive neurons in the MAOB-I treated group compared to Saline treated group (p < 0.01). MAOB-I has a significant potential for protecting the degenerating spinal cord neurons and enhancing the regeneration of injured sciatic nerve fibers following crush injury.
Collapse
Affiliation(s)
| | - Waleed M Renno
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait.
| | - Hanaa L Sadek
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait
| | - Noura M Kayali
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait
| | - Aseel Al-Salem
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait
| | - Muddanna S Rao
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait
| | - Khalid M Khan
- Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait
| |
Collapse
|
23
|
Individual Amino Acid Supplementation Can Improve Energy Metabolism and Decrease ROS Production in Neuronal Cells Overexpressing Alpha-Synuclein. Neuromolecular Med 2017. [PMID: 28620826 DOI: 10.1007/s12017-017-8448-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by alpha-synuclein accumulation and loss of dopaminergic neurons in the substantia nigra (SN) region of the brain. Increased levels of alpha-synuclein have been shown to result in loss of mitochondrial electron transport chain complex I activity leading to increased reactive oxygen species (ROS) production. WT alpha-synuclein was stably overexpressed in human BE(2)-M17 neuroblastoma cells resulting in increased levels of an alpha-synuclein multimer, but no increase in alpha-synuclein monomer levels. Oxygen consumption was decreased by alpha-synuclein overexpression, but ATP levels did not decrease and ROS levels did not increase. Treatment with ferrous sulfate, a ROS generator, resulted in decreased oxygen consumption in both control and alpha-synuclein overexpressing cells. However, this treatment only decreased ATP levels and increased ROS production in the cells overexpressing alpha-synuclein. Similarly, paraquat, another ROS generator, decreased ATP levels in the alpha-synuclein overexpressing cells, but not in the control cells, further demonstrating how alpha-synuclein sensitized the cells to oxidative insult. Proteomic analysis yielded molecular insights into the cellular adaptations to alpha-synuclein overexpression, such as the increased abundance of many mitochondrial proteins. Many amino acids and citric acid cycle intermediates and their ester forms were individually supplemented to the cells with L-serine, L-proline, L-aspartate, or L-glutamine decreasing ROS production in oxidatively stressed alpha-synuclein overexpressing cells, while diethyl oxaloacetate or L-valine supplementation increased ATP levels. These results suggest that dietary supplementation with individual metabolites could yield bioenergetic improvements in PD patients to delay loss of dopaminergic neurons.
Collapse
|
24
|
Inaba-Hasegawa K, Shamoto-Nagai M, Maruyama W, Naoi M. Type B and A monoamine oxidase and their inhibitors regulate the gene expression of Bcl-2 and neurotrophic factors in human glioblastoma U118MG cells: different signal pathways for neuroprotection by selegiline and rasagiline. J Neural Transm (Vienna) 2017; 124:1055-1066. [PMID: 28577058 DOI: 10.1007/s00702-017-1740-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 05/30/2017] [Indexed: 12/01/2022]
Abstract
Type B monoamine oxidase (MAO-B) in glial cells has been considered to be associated with neuronal death in Parkinson's disease. MAO-B inhibitors, rasagiline and selegiline [(-)deprenyl], protect neurons in animal and cellular models of neurodegeneration. However, the role of MAO-B itself in the regulation of cell death processing remains elusive, whereas type A MAO (MAO-A) mediates the induction of anti-apoptotic Bcl-2 genes by rasagiline and selegiline. In this paper, the involvement of MAOs in the induction of neuroprotective genes by MAO inhibitors was investigated in human glioblastoma U118MG cells expressing mainly MAO-B. Selegiline significantly increased Mao-B, which was suppressed by Mao-A knockdown with short interfering (si)RNA, whereas rasagiline less markedly increased Mao-B, which was not affected by Mao-A knockdown. Mao-A mRNA was also markedly increased by rasagiline and selegiline, and Mao-B knockdown significantly enhanced the induction by selegiline, but not by rasagiline. Mao-B knockdown also significantly increased mRNA levels of Bcl-2, brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF). Selegiline synergistically enhanced the expression of these genes in Mao-B knockdown cells, but Mao-A knockdown suppressed the increase. Rasagiline increased BDNF and GDNF, which Mao-B and Mao-A knockdown inhibited. These results show that MAO-B might function as a repressor and MAO-A as a mediator in the constitutional expression of pro-survival genes, and that MAO-B and MAO-A might regulate different signal pathways for rasagiline and selegiline to induce neuroprotective genes. The novel role of glial MAOs in the regulation of gene expression is discussed.
Collapse
Affiliation(s)
- Keiko Inaba-Hasegawa
- Department of Health and Nutrition, Faculty of Psychological and Physical Science, Aichi Gakuin University, 12 Araike, Iwasaki-cho, Nisshin, Aichi, 470-0195, Japan
| | - Masayo Shamoto-Nagai
- Department of Health and Nutrition, Faculty of Psychological and Physical Science, Aichi Gakuin University, 12 Araike, Iwasaki-cho, Nisshin, Aichi, 470-0195, Japan
| | - Wakako Maruyama
- Department of Health and Nutrition, Faculty of Psychological and Physical Science, Aichi Gakuin University, 12 Araike, Iwasaki-cho, Nisshin, Aichi, 470-0195, Japan
| | - Makoto Naoi
- Department of Health and Nutrition, Faculty of Psychological and Physical Science, Aichi Gakuin University, 12 Araike, Iwasaki-cho, Nisshin, Aichi, 470-0195, Japan.
| |
Collapse
|
25
|
MAOA-a novel decision maker of apoptosis and autophagy in hormone refractory neuroendocrine prostate cancer cells. Sci Rep 2017; 7:46338. [PMID: 28402333 PMCID: PMC5389346 DOI: 10.1038/srep46338] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 03/15/2017] [Indexed: 12/13/2022] Open
Abstract
Autophagy and apoptosis are two well-controlled mechanisms regulating cell fate. An understanding of decision-making between these two pathways is in its infancy. Monoamine oxidase A (MAOA) is a mitochondrial enzyme that is well-known in psychiatric research. Emerging reports showed that overexpression MAOA is associated with prostate cancer (PCa). Here, we show that MAOA is involved in mediating neuroendocrine differentiation of PCa cells, a feature associated with hormone-refractory PCa (HRPC), a lethal type of disease. Following recent reports showing that NED of PCa requires down-regulation of repressor element-1 silencing transcription factor (REST) and activation of autophagy; we observe that MAOA is a novel direct target gene of REST. Reactive oxygen species (ROS) produced by overexpressed MAOA plays an essential role in inhibiting apoptosis and activating autophagy in NED PCa cells. MAOA inhibitors significantly reduced NED and autophagy activation of PCa cells. Our results here show MAOA as a new decision-maker for activating autophagy and MAOA inhibitors may be useful as a potential therapy for neuroendocrine tumors.
Collapse
|
26
|
Yang Z, Wang X, Yang J, Sun M, Wang Y, Wang X. Aberrant CpG Methylation Mediates Abnormal Transcription of MAO-A Induced by Acute and Chronic l-3,4-Dihydroxyphenylalanine Administration in SH-SY5Y Neuronal Cells. Neurotox Res 2016; 31:334-347. [DOI: 10.1007/s12640-016-9686-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 11/27/2016] [Accepted: 12/01/2016] [Indexed: 01/07/2023]
|
27
|
Xie H, Li C, Dang Q, Chang LS, Li L. Infiltrating mast cells increase prostate cancer chemotherapy and radiotherapy resistances via modulation of p38/p53/p21 and ATM signals. Oncotarget 2016; 7:1341-53. [PMID: 26625310 PMCID: PMC4811464 DOI: 10.18632/oncotarget.6372] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 11/06/2015] [Indexed: 12/11/2022] Open
Abstract
Early studies indicated that mast cells in prostate tumor microenvironment might influence prostate cancer (PCa) progression. Their impacts to PCa therapy, however, remained unclear. Here we found PCa could recruit more mast cells than normal prostate epithelial cells then alter PCa chemotherapy and radiotherapy sensitivity, leading to PCa more resistant to these therapies. Mechanism dissection revealed that infiltrated mast cells could increase p21 expression via modulation of p38/p53 signals, and interrupting p38-p53 signals via siRNAs of p53 or p21 could reverse mast cell-induced docetaxel chemotherapy resistance of PCa. Furthermore, recruited mast cells could also increase the phosphorylation of ATM at ser-1981 site, and inhibition of ATM activity could reverse mast cell-induced radiotherapy resistance. The in vivo mouse model with xenografted PCa C4-2 cells co-cultured with mast cells also confirmed that mast cells could increase PCa chemotherapy resistance via activating p38/p53/p21 signaling. Together, our results provide a new mechanism showing infiltrated mast cells could alter PCa chemotherapy and radiotherapy sensitivity via modulating the p38/p53/p21 signaling and phosphorylation of ATM. Targeting this newly identified signaling may help us better suppress PCa chemotherapy and radiotherapy resistance.
Collapse
Affiliation(s)
- Hongjun Xie
- Chawnshang Chang Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| | - Chong Li
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Science, Beijing 100101, China
| | - Qiang Dang
- Chawnshang Chang Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| | - Luke S Chang
- Chawnshang Chang Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| | - Lei Li
- Chawnshang Chang Sex Hormone Research Center, Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
28
|
Hwang CJ, Choi DY, Jung YY, Lee YJ, Yun JS, Oh KW, Han SB, Oh S, Park MH, Hong JT. Inhibition of p38 pathway-dependent MPTP-induced dopaminergic neurodegeneration in estrogen receptor alpha knockout mice. Horm Behav 2016; 80:19-29. [PMID: 26836768 DOI: 10.1016/j.yhbeh.2016.01.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 01/24/2016] [Accepted: 01/29/2016] [Indexed: 01/20/2023]
Abstract
Approximately, 7-10 million people in the world suffer from Parkinson's disease (PD). Recently, increasing evidence has suggested the protective effect of estrogens against nigrostriatal dopaminergic damage in PD. In this study, we investigated whether estrogen affects 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced behavioral impairment in estrogen receptor alpha (ERα)-deficient mice. MPTP (15mg/kg, four times with 1.5-h interval)-induced dopaminergic neurodegeneration was evaluated in ERα wild-type (WT) and knockout (KO) mice. Larger dopamine depletion, behavioral impairments (Rotarod test, Pole test, and Gait test), activation of microglia and astrocytes, and neuroinflammation after MPTP injection were observed in ERα KO mice compared to those in WT mice. Immunostaining for tyrosine hydroxylase (TH) after MPTP injection showed fewer TH-positive neurons in ERα KO mice than WT mice. Levels of dopamine and 3,4-dihydroxyphenylacetic acid (DOPAC, metabolite of dopamine) were also lowered in ERα KO mice after MPTP injection. Interestingly, a higher immunoreactivity for monoamine oxidase (MAO) B was found in the substantia nigra and striatum of ERα KO mice after MPTP injection. We also found an increased activation of p38 kinase (which positively regulates MAO B expression) in ERα KO mice. In vitro estrogen treatment inhibited neuroinflammation in 1-methyl-4-phenyl pyridium (MPP+)-treated cultured astrocyte cells; however, these inhibitory effects were removed by p38 inhibitor. These results indicate that ERα might be important for dopaminergic neuronal survival through inhibition of p38 pathway.
Collapse
Affiliation(s)
- Chul Ju Hwang
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-31, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 361-951, South Korea
| | - Dong-Young Choi
- College of Pharmacy, Yeungnam University, 280, Daehak-ro, Gyeongsan, Gyeongbuk 712-749, South Korea
| | - Yu Yeon Jung
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-31, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 361-951, South Korea
| | - Young-Jung Lee
- School of Equine Science, Cheju Halla University, 38 Halladaehak-ro, Jeju-si, Jeju Special Self-Governing Province 690-708, South Korea
| | - Jae Suk Yun
- Osong Health Technology Administration Complex, 187 Osongsaengmyeong2(i)-ro, Osong-eup, Cheongju, Chungbuk 363-700, South Korea
| | - Ki-Wan Oh
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-31, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 361-951, South Korea
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-31, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 361-951, South Korea
| | - Seikwan Oh
- Department of Neuroscience and Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul 158-710, South Korea
| | - Mi Hee Park
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-31, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 361-951, South Korea.
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro 194-31, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 361-951, South Korea.
| |
Collapse
|
29
|
Duncan JW, Zhang X, Wang N, Johnson S, Harris S, Udemgba C, Ou XM, Youdim MB, Stockmeier CA, Wang JM. Binge ethanol exposure increases the Krüppel-like factor 11-monoamine oxidase (MAO) pathway in rats: Examining the use of MAO inhibitors to prevent ethanol-induced brain injury. Neuropharmacology 2016; 105:329-340. [PMID: 26805422 DOI: 10.1016/j.neuropharm.2016.01.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 01/14/2016] [Accepted: 01/20/2016] [Indexed: 12/25/2022]
Abstract
Binge drinking induces several neurotoxic consequences including oxidative stress and neurodegeneration. Because of these effects, drugs which prevent ethanol-induced damage to the brain may be clinically beneficial. In this study, we investigated the ethanol-mediated KLF11-MAO cell death cascade in the frontal cortex of Sprague-Dawley rats exposed to a modified Majchowicz 4-day binge ethanol model and control rats. Moreover, MAO inhibitors (MAOIs) were investigated for neuroprotective activity against binge ethanol. Binge ethanol-treated rats demonstrated a significant increase in KLF11, both MAO isoforms, protein oxidation and caspase-3, as well as a reduction in BDNF expression in the frontal cortex compared to control rats. MAOIs prevented these binge ethanol-induced changes, suggesting a neuroprotective benefit. Neither binge ethanol nor MAOI treatment significantly affected protein expression levels of the oxidative stress enzymes, SOD2 or catalase. Furthermore, ethanol-induced antinociception was enhanced following exposure to the 4-day ethanol binge. These results demonstrate that the KLF11-MAO pathway is activated by binge ethanol exposure and MAOIs are neuroprotective by preventing the binge ethanol-induced changes associated with this cell death cascade. This study supports KLF11-MAO as a mechanism of ethanol-induced neurotoxicity and cell death that could be targeted with MAOI drug therapy to alleviate alcohol-related brain injury. Further examination of MAOIs to reduce alcohol use disorder-related brain injury could provide pivotal insight to future pharmacotherapeutic opportunities.
Collapse
Affiliation(s)
- Jeremy W Duncan
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS 39216, USA; Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Xiao Zhang
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS 39216, USA; Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Niping Wang
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Shakevia Johnson
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Sharonda Harris
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Chinelo Udemgba
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Xiao-Ming Ou
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Moussa B Youdim
- Technion-Rappaport Family Faculty of Medicine, Eve Topf Center of Excellence, For Neurodegenerative Diseases Research, Haifa, 31096, Israel
| | - Craig A Stockmeier
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Jun Ming Wang
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS 39216, USA; Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA; Department of Pathology, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| |
Collapse
|
30
|
Curcumol Inhibits Growth and Induces Apoptosis of Colorectal Cancer LoVo Cell Line via IGF-1R and p38 MAPK Pathway. Int J Mol Sci 2015; 16:19851-67. [PMID: 26307972 PMCID: PMC4581329 DOI: 10.3390/ijms160819851] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 07/27/2015] [Accepted: 07/30/2015] [Indexed: 01/07/2023] Open
Abstract
Curcumol, isolated from the traditional medical plant Rhizoma Curcumae, is the bioactive component of Zedoary oil, whose potential anti-tumor effect has attracted considerable attention in recent years. Though many researchers have reported curcumol and its bioactivity, the potential molecular mechanism for its anti-cancer effect in colorectal cancer LoVo cells still remains unclear. In the present study, we found that curcumol showed growth inhibition and induced apoptosis of LoVo cells in a dose- and time-dependent manner. The occurrence of its proliferation inhibition and apoptosis came with suppression of IGF-1R expression, and then increased the phosphorylation of p38 mitogen activated protein kinase (MAPK), which might result in a cascade response by inhibiting the CREB survival pathway and finally triggered Bax/Bcl-2 and poly(ADP-ribose) polymerase 1 (PARP-1) apoptosis signals. Moreover, curcumol inhibited colorectal cancer in xenograft models of nude mice. Immunohistochemical and Western blot analysis revealed that curcumol could decrease the expression of ki-67, Bcl-2 as well as CREB1, and increase the expression of Bax and the phosphorylation of p38, which were consistent with our in vitro study. Overall, our in vitro and in vivo data confirmed the anti-cancer activity of curcumol, which was related to a significant inhibition of IGF-1R and activation of p38 MAPKs, indicating that curcumol may be a potential anti-tumor agent for colorectal carcinoma therapy.
Collapse
|
31
|
Naoi M, Riederer P, Maruyama W. Modulation of monoamine oxidase (MAO) expression in neuropsychiatric disorders: genetic and environmental factors involved in type A MAO expression. J Neural Transm (Vienna) 2015; 123:91-106. [PMID: 25604428 DOI: 10.1007/s00702-014-1362-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 12/27/2014] [Indexed: 12/18/2022]
Abstract
Monoamine oxidase types A and B (MAO-A, MAO-B) regulate the levels of monoamine neurotransmitters in the brain, and their dysfunction may be involved in the pathogenesis and influence the clinical phenotypes of neuropsychiatric disorders. Reversible MAO-A inhibitors, such as moclobemide and befloxatone, are currently employed in the treatment of emotional disorders by inhibiting the enzymatic degradation of dopamine, serotonin and norepinephrine in the central nervous system (CNS). It has been suggested that the irreversible MAO-B inhibitors selegiline and rasagiline exert a neuroprotective effect in Parkinson's and Alzheimer's diseases. This effect, however, is not related to their inhibition of MAO activity; in animal and cellular models, selegiline and rasagiline protect neuronal cells through their anti-apoptotic activity and induction of pro-survival genes. There is increasing evidence that MAO-A activity, but not that of MAO-B, is implicated in the pathophysiology of neurodegenerative disorders, but also in gene induction by MAO-B inhibitors; on the other hand, selegiline and rasagiline increase MAO-A mRNA, protein, and enzyme activity levels. Taken together, these results suggest that each MAO subtype exerts effects that modulate the expression and activity of the other isoenzyme. The roles of MAO-A and -B in the CNS should therefore be re-evaluated with respect to the "type-specificity" of their inhibitors, which may not be unconditional during chronic treatment. Mao-a expression, in particular, may be implicated in pathogenesis and phenotypes in neuropsychiatric disorders. MAO-A expression is modified by mao polymorphisms affecting its transcriptional efficiency, as well as by mutations and polymorphism of parkin, Sirt1, FOXO, microRNA, presenilin-1, and other regulatory proteins. In addition, childhood maltreatment has been shown to have an impact upon adolescent social behavior in children with mao-a polymorphisms of low transcriptional activity. Low MAO-A activity may increase the levels of serotonin and norepinephrine, resulting in disturbed neurotransmitter system development and behavior. This review discusses genetic and environmental factors involved in the regulation of MAO-A expression, in the contexts of neuropsychiatric function and of the regulation of neuronal survival and death.
Collapse
Affiliation(s)
- Makoto Naoi
- Department of Health and Nutrition, Faculty of Psychological and Physical Science, Aichi Gakuin University, 12 Araike, Iwasaki-cho, Nisshin, Aichi, 470-0195, Japan.
| | - Peter Riederer
- Clinical Neurochemistry, National Parkinson's Foundation Centre of Excellence Laboratories, Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital Würzburg, Würzburg, Germany
| | - Wakako Maruyama
- Department of Cognitive Brain Science, National Research Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| |
Collapse
|
32
|
Shan Z, Hou Q, Zhang N, Guo L, Zhang X, Ma Y, Zhou Y. Overexpression of oxidored-nitro domain containing protein 1 induces growth inhibition and apoptosis in human prostate cancer PC3 cells. Oncol Rep 2014; 32:1939-46. [PMID: 25118646 DOI: 10.3892/or.2014.3407] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 07/29/2014] [Indexed: 11/06/2022] Open
Abstract
Previous studies have reported that oxidored-nitro domain containing protein 1 (NOR1) is a novel tumor suppressor gene identified in various types of cancer, such as nasopharyngeal carcinoma and cervical cancer. Recent studies have shown that NOR1 expression is lower in prostate cancer compared with normal prostate tissue. However, the specific function and exact mechanism of NOR1 in prostate cancer remains to be clarified. The present study aimed to investigate the function and mechanism of NOR1 in prostate cancer PC3 cells. DU145 and PC3 cells were transduced with a vector and cell viability, proliferation and apoptosis were determined. As predicted, NOR1 overexpression significantly inhibited growth and apoptosis in PC3 cells. NOR1 overexpression decreased the expression of the anti-apoptotic genes Bcl-2 and Bcl-xl and increased the level of the pro-apoptotic genes Bax and Bak in PC3 cells. Further investigation demonstrated that NOR1 overexpression activates caspase-3. Silencing of NOR1 did not inhibit growth or induce apoptosis in PC3 cells. Moreover, NOR1 inhibited proliferation and induced apoptosis via the activation of MAPK. The overexpression of NOR1 significantly inhibited tumor growth in PC3 tumor-bearing nude mice. The results suggest that the upregulated NOR1 expression was able to inhibit the progression of prostate cancer. Thus, NOR1 may be an ideal target for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Zhongjie Shan
- Department of Urology, The People's Hospital of Zhengzhou, Zhengzhou, Henan 450003, P.R. China
| | - Qinglan Hou
- Department of Urology, The People's Hospital of Zhengzhou, Zhengzhou, Henan 450003, P.R. China
| | - Nan Zhang
- Department of Urology, The People's Hospital of Zhengzhou, Zhengzhou, Henan 450003, P.R. China
| | - Liang Guo
- Department of Urology, The People's Hospital of Zhengzhou, Zhengzhou, Henan 450003, P.R. China
| | - Xinheng Zhang
- Department of Urology, The Central Hospital of Sanmenxia, Sanmenxia, Henan 472000, P.R. China
| | - Yaohui Ma
- Department of Urology, The People's Hospital of Zhengzhou, Zhengzhou, Henan 450003, P.R. China
| | - Yudong Zhou
- Department of Urology, The People's Hospital of Zhengzhou, Zhengzhou, Henan 450003, P.R. China
| |
Collapse
|
33
|
Naoi M, Maruyama W, Inaba-Hasegawa K. Revelation in the neuroprotective functions of rasagiline and selegiline: the induction of distinct genes by different mechanisms. Expert Rev Neurother 2014; 13:671-84. [PMID: 23739004 DOI: 10.1586/ern.13.60] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
In Parkinson's disease, cell death of dopamine neurons in the substantia nigra progresses and neuroprotective therapy is required to halt neuronal loss. In cellular and animal models, selegiline [(-)deprenyl] and rasagiline, inhibitors of type B monoamine oxidase (MAO)-B, protect neuronal cells from programmed cell death. In this paper, the authors review their recent results on the molecular mechanisms by which MAO inhibitors prevent the cell death through the induction of antiapoptotic, prosurvival genes. MAO-A mediates the induction of antiapoptotic bcl-2 and mao-a itself by rasagiline, whereas a different mechanism is associated with selegiline. Rasagiline and selegiline preferentially increase GDNF and BDNF in nonhuman primates and Parkinsonian patients, respectively. Enhanced neurotrophic factors might be applicable to monitor the neurorescuing activity of neuroprotection.
Collapse
Affiliation(s)
- Makoto Naoi
- Department of Health and Nutrition, Faculty of Psychological and Physical Science, Aichi Gakuin University, Nisshin, Aichi, Japan.
| | | | | |
Collapse
|
34
|
β-Arrestin prevents cell apoptosis through pro-apoptotic ERK1/2 and p38 MAPKs and anti-apoptotic Akt pathways. Apoptosis 2013; 17:1019-26. [PMID: 22699970 DOI: 10.1007/s10495-012-0741-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Our previous studies have shown that β-arrestin 2 plays an anti-apoptotic effect. However, the mechanisms by which β-arrestin contribute to anti-apoptotic role remain unclear. In this study, we show that a deficiency of either β-arrestin 1 or β-arrestin 2 significantly increases serum deprivation (SD)-induced percentage of apoptotic cells. β-arrestin 2 deficient-induced apoptosis was inhibited by transfection with β-arrestin 2 full-length plasmid, revealing that SD-induced apoptosis is dependent on β-arrestin 2. Furthermore, in the absence of either β-arrestin 1 or β-arrestin 2 significantly enhances SD-induced the level of pro-apoptotic proteins, including cleaved caspase-3, extracellular-signal regulated kinase 1/2 (ERK1/2) and p38, members of mitogen-activated protein kinases (MAPKs). In addition, a deficiency of either β-arrestin 1 or β-arrestin 2 inhibits phosphorylation of Akt. The SD-induced changes in cleaved caspase-3, ERK1/2 and p38 MAPKs, Akt, and apoptotic cell numbers could be blocked by double knockout of β-arrestin 1/2. Our study thus demonstrates that β-arrestin inhibits cell apoptosis through pro-apoptotic ERK1/2 and p38 MAPKs and anti-apoptotic Akt signaling pathways.
Collapse
|
35
|
Fitzgerald JC, Ugun-Klusek A, Allen G, De Girolamo LA, Hargreaves I, Ufer C, Abramov AY, Billett EE. Monoamine oxidase-A knockdown in human neuroblastoma cells reveals protection against mitochondrial toxins. FASEB J 2013; 28:218-29. [PMID: 24051032 DOI: 10.1096/fj.13-235481] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The study examined how the mitochondrial enzyme monoamine oxidase-A (MAO-A), which produces hydrogen peroxide as a catalytic by-product, influences death and survival mechanisms. Targeted microRNA (miRNA) was used to stably knock down MAO-A mRNA, protein, and catalytic activity by 60-70% in SH-SY5Y human neuroblastoma cells. The effects of MAO-A knockdown (KD) on ATP, oxidative stress, electron transport chain, and survival following exposure to mitochondrial toxins were assessed. In control cells, complex I inhibition resulted in caspase-mediated cell death linked with ROS production and reduced ATP, followed by up-regulation of MAO-A mRNA, protein, and enzyme activity levels. Inhibition of complex III and IV resulted in a similar increase in MAO-A expression, while up-regulation of MAO-A was lower following complex II inhibition. MAO-A KD decreased basal reactive oxygen species levels by 50% and increased levels of ATP and reduced glutathione and Bcl-2. MAO-A KD specifically increased the activity of complex I but had no effect on complex II-IV activities. Furthermore, MAO-A KD protected against inhibitors of complex I, III, and IV. In summary, endogenous MAO-A levels influence mitochondrial function, notably complex I activity, and MAO-A may be a target for protection against neurodegenerative conditions that involve oxidative stress and mitochondrial dysfunction as underlying pathogenic factors.
Collapse
Affiliation(s)
- Julia C Fitzgerald
- 2School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, UK.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Na+/H+ exchanger 1 inhibition contributes to K562 leukaemic cell differentiation. Cell Biol Int 2012; 36:739-45. [PMID: 22494070 DOI: 10.1042/cbi20100919] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The effect of hypoxia on the differentiation of chronic myeloid leukaemic K562 cells were studied, as was the role of the NHE1 (Na+/H+ exchanger 1). Hypoxia induced differentiation of K562 cells as seen by modifications in their morphological features, up-regulation of C/EBPα (CCAAT/enhancer-binding protein α), and marked IL-8 (interleukin-8) release. Inhibition of NHE1 under hypoxia additionally enhanced the level of C/EBPα and further promoted leukaemic cells differentiation. Pharmacological inhibition of p38 MAPK (mitogen-activated protein kinase) also significantly suppressed C/EBPα expression under hypoxia conditions after NHE1 inhibition. These results indicate the enhancement of hypoxia-induced K562 differentiation by NHE1 inhibition, which may be due to up-regulation of C/EBPα via p38 MAPK signalling pathway, which suggests a possible therapeutic target of NHE1 under hypoxia microenvironment in the treatment of leukaemic diseases.
Collapse
|
37
|
Inaba-Hasegawa K, Akao Y, Maruyama W, Naoi M. Rasagiline and selegiline, inhibitors of type B monoamine oxidase, induce type A monoamine oxidase in human SH-SY5Y cells. J Neural Transm (Vienna) 2012; 120:435-44. [PMID: 22968599 DOI: 10.1007/s00702-012-0899-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2012] [Accepted: 09/03/2012] [Indexed: 02/03/2023]
Abstract
Type B monoamine oxidase (MAO-B) is proposed to be involved in the pathogenesis of neurodegenerative disorders, such as Parkinson's disease, through oxidative stress and synthesis of neurotoxins. MAO-B inhibitors, rasagiline and selegiline [(-)deprenyl], protect neuronal cells by direct intervention in mitochondrial death signaling and induction of pro-survival Bcl-2 and neurotrophic factors. Recently, type A MAO (MAO-A) was found to mediate the induction of anti-apoptotic Bcl-2 by rasagiline, whereas MAO-A increases in neuronal death and also serves as a target of neurotoxins. These controversial results suggest that MAO-A may play a decisive role in neuronal survival and death. This paper reports that rasagiline and selegiline increased the mRNA, protein and catalytic activity of MAO-A in SH-SY5Y cells. Silencing MAO-A expression with small interfering (si)RNA suppressed rasagiline-dependent MAO-A expression, but MAO-B overexpression in SH-SY5Y cells did not affect, suggesting that MAO-A, not MAO-B, might be associated with MAO-A upregulation. Rasagiline reduced R1, a MAO-A specific repressor, but selegiline did not. Mithramycin-A, an inhibitor of Sp1 binding, and actinomycin-D, a transcriptional inhibitor, reduced the rasagiline-dependent upregulation of MAO-A mRNA, indicating that rasagiline induced MAO-A transcriptionally through R1-Sp1 pathway, whereas selegiline by another non-defined pathway. These results are discussed in relation to the role of MAO-A and these MAO-B inhibitors in neuronal death and neuroprotection.
Collapse
Affiliation(s)
- Keiko Inaba-Hasegawa
- Department of Neurosciences, Gifu International Institute of Biotechnology, Kakamigahara, Gifu, Japan
| | | | | | | |
Collapse
|
38
|
Choi DY, Lee MK, Hong JT. Lack of CCR5 modifies glial phenotypes and population of the nigral dopaminergic neurons, but not MPTP-induced dopaminergic neurodegeneration. Neurobiol Dis 2012; 49:159-68. [PMID: 22922220 DOI: 10.1016/j.nbd.2012.08.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 07/12/2012] [Accepted: 08/02/2012] [Indexed: 11/17/2022] Open
Abstract
Constitutive expression of C-C chemokine receptor (CCR) 5 has been detected in astrocytes, microglia and neurons, but its physiological roles in the central nervous system are obscure. The bidirectional interactions between neuron and glial cells through CCR5 and its ligands were thought to be crucial for maintaining normal neuronal activities. No study has described function of CCR5 in the dopaminergic neurodegeneration in Parkinson's disease. In order to examine effects of CCR5 on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced dopaminergic neurodegeneration, we employed CCR5 wild type (WT) and knockout (KO) mice. Immunostainings for tyrosine hydroxylase (TH) exhibited that CCR5 KO mice had lower number of TH-positive neurons even in the absence of MPTP. Difference in MPTP (15mg/kg×4 times, 2hr interval)-mediated loss of TH-positive neurons was subtle between CCR5 WT and KO mice, but there was larger dopamine depletion, behavioral impairments and microglial activation in CCR5 deficient mice. Intriguingly, CCR5 KO brains contained higher immunoreactivity for monoamine oxidase (MAO) B which was mainly localized within astrocytes. In agreement with upregulation of MAO B, concentration of MPP+ was higher in the substantia nigra and striatum of CCR5 KO mice after MPTP injection. We found remarkable activation of p38 MAPK in CCR5 deficient mice, which positively regulates MAO B expression. These results indicate that CCR5 deficiency modifies the nigrostriatal dopaminergic neuronal system and bidirectional interaction between neurons and glial cells via CCR5 might be important for dopaminergic neuronal survival.
Collapse
Affiliation(s)
- Dong-Young Choi
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 712-749, Republic of Korea; College of Pharmacy, Chungbuk National University, 12 Gaesin-dong, Heungduk-gu, Cheongju, Chungbuk, 361-763, Republic of Korea; Medical Research Center, Chungbuk National University, 12 Gaesin-dong, Heungduk-gu, Cheongju, Chungbuk, 361-763, Republic of Korea
| | - Myung Koo Lee
- College of Pharmacy, Chungbuk National University, 12 Gaesin-dong, Heungduk-gu, Cheongju, Chungbuk, 361-763, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy, Chungbuk National University, 12 Gaesin-dong, Heungduk-gu, Cheongju, Chungbuk, 361-763, Republic of Korea; Medical Research Center, Chungbuk National University, 12 Gaesin-dong, Heungduk-gu, Cheongju, Chungbuk, 361-763, Republic of Korea.
| |
Collapse
|
39
|
Aspartic acid substitutions in monoamine oxidase-A reveal both catalytic-dependent and -independent influences on cell viability and proliferation. J Neural Transm (Vienna) 2012; 119:1285-94. [DOI: 10.1007/s00702-012-0779-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Accepted: 02/16/2012] [Indexed: 12/17/2022]
|
40
|
Type A monoamine oxidase is associated with induction of neuroprotective Bcl-2 by rasagiline, an inhibitor of type B monoamine oxidase. J Neural Transm (Vienna) 2011; 119:405-14. [PMID: 22065207 DOI: 10.1007/s00702-011-0730-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Accepted: 10/24/2011] [Indexed: 12/15/2022]
Abstract
Rasagiline and (-)deprenyl (selegiline), irreversible type B monoamine oxidase (MAO-B) inhibitors, protect neuronal cells through gene induction of pro-survival Bcl-2 and neurotrophic factors in the cellular models of neurodegenerative disorders. In this paper, the role of MAO in the up-regulation of neuroprotective Bcl-2 gene by these inhibitors was studied using type A MAO (MAO-A) expressing wild SH-SY5Y cells and the transfection-enforced MAO-B overexpressed cells. Rasagiline and (-)deprenyl, and also befloxatone, a reversible MAO-A inhibitor, increased Bcl-2 mRNA and protein in SH-SY5Y cells. Silencing MAO-A expression with short interfering (si) RNA suppressed Bcl-2 induction by rasagiline, but not by (-)deprenyl. MAO-B overexpression inhibited Bcl-2 induction by rasagiline and befloxatone, but did not affect that by (-)deprenyl, suggesting the different mechanisms behind Bcl-2 gene induction by these MAO-B inhibitors. The novel role of MAO-A in Bcl-2 induction by rasagiline is discussed with regard to the molecular mechanism underlying neuroprotection by the MAO inhibitors.
Collapse
|
41
|
Johnson S, Stockmeier CA, Meyer JH, Austin MC, Albert PR, Wang J, May WL, Rajkowska G, Overholser JC, Jurjus G, Dieter L, Johnson C, Sittman DB, Ou XM. The reduction of R1, a novel repressor protein for monoamine oxidase A, in major depressive disorder. Neuropsychopharmacology 2011; 36:2139-48. [PMID: 21654740 PMCID: PMC3158311 DOI: 10.1038/npp.2011.105] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The novel transcriptional repressor protein, R1 (JPO2/CDCA7L/RAM2), inhibits monoamine oxidase A (MAO A) gene expression and influences cell proliferation and survival. MAO A is implicated in several neuropsychiatric illnesses and highly elevated in major depressive disorder (MDD); however, whether R1 is involved in these disorders is unknown. This study evaluates the role of R1 in depressed subjects either untreated or treated with antidepressant drugs. R1 protein levels were determined in the postmortem prefrontal cortex of 18 untreated MDD subjects and 12 medicated MDD subjects compared with 18 matched psychiatrically normal control subjects. Western blot analysis showed that R1 was significantly decreased by 37.5% (p<0.005) in untreated MDD subjects. The R1 level in medicated MDD subjects was also significantly lower (by 30%; p<0.05) compared with control subjects, but was not significantly different compared with untreated MDD subjects. Interestingly, the reduction in R1 was significantly correlated with an increase (approximately 40%; p<0.05) in MAO A protein levels within the MDD groups compared with controls. Consistent with the change in MAO A protein expression, the MAO A catalytic activity was significantly greater in both MDD groups compared with controls. These results suggest that reduced R1 may lead to elevated MAO A levels in untreated and treated MDD subjects; moreover, the reduction of R1 has been implicated in apoptotic cell death and apoptosis has also been observed in the brains of MDD subjects. Therefore, modulation of R1 levels may provide a new therapeutic target in the development of more effective strategies to treat MDD.
Collapse
Affiliation(s)
- Shakevia Johnson
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
| | - Craig A Stockmeier
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA,Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA
| | - Jeffrey H Meyer
- Department of Psychiatry and Centre for Addiction and Mental Health, University of Toronto, Toronto, ON, Canada
| | - Mark C Austin
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
| | - Paul R Albert
- Department of Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Junming Wang
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Warren L May
- Biostatistics Center, University of Mississippi Medical Center, Jackson, MS, USA
| | - Grazyna Rajkowska
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
| | - James C Overholser
- Department of Psychology, Case Western Reserve University, Cleveland, OH, USA
| | - George Jurjus
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA
| | - Lesa Dieter
- Department of Psychology, Case Western Reserve University, Cleveland, OH, USA
| | - Chandra Johnson
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
| | - Donald B Sittman
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, MS, USA
| | - Xiao-Ming Ou
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA,Department of Psychiatry and Human Behavior (G-109), University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS 39216, USA, Tel: +1 601 984 5893, Fax: +1 601 984 5899, E-mail:
| |
Collapse
|
42
|
Shih JC, Wu JB, Chen K. Transcriptional regulation and multiple functions of MAO genes. J Neural Transm (Vienna) 2011; 118:979-86. [PMID: 21359973 PMCID: PMC3125068 DOI: 10.1007/s00702-010-0562-9] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Accepted: 12/08/2010] [Indexed: 11/29/2022]
Abstract
Monoamine oxidase (MAO) A and MAO B are a crucial pair of isoenzymes, which oxidatively deaminate monoamine neurotransmitters and dietary amines with a production of hydrogen peroxide. These two isoenzymes have different but overlapping substrate and inhibitor specificities. MAO A and MAO B share 70% amino acid sequence identity and show different temporal and spatial expressions in both humans and mice. Abnormal MAO A or MAO B activity has been implicated in numerous neurological and psychiatric disorders. A better understanding of the transcriptional regulation of MAO A and MAO B genes may help explain the differential tissue-specific expression of these two isoenzymes and provide insights into the molecular basis of the disorders associated with MAO dysfunction. This review discusses the recent progress in the transcriptional regulation and multiple functions of MAO A and MAO B genes.
Collapse
Affiliation(s)
- Jean C Shih
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Room 518, Los Angeles, CA 90089, USA.
| | | | | |
Collapse
|
43
|
Prabhakaran K, Chapman GD, Gunasekar PG. α-Synuclein overexpression enhances manganese-induced neurotoxicity through the NF-κB-mediated pathway. Toxicol Mech Methods 2011; 21:435-43. [DOI: 10.3109/15376516.2011.560210] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
44
|
Naoi M, Maruyama W, Inaba-Hasegawa K, Akao Y. Type A monoamine oxidase regulates life and death of neurons in neurodegeneration and neuroprotection. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2011; 100:85-106. [PMID: 21971004 DOI: 10.1016/b978-0-12-386467-3.00005-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In Parkinson's disease, type B monoamine oxidase (MAO-B) is proposed to play an important role in the pathogenesis through production of reactive oxygen species and neurotoxins from protoxicants, such as 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. In addition, inhibitors of MAO-B protect neurons in the cellular and animal models of Parkinson's and Alzheimer's diseases. However, the role of type A MAO (MAO-A) in neuronal death and neuroprotection by MAO-B inhibitors has been scarcely elucidated. This chapter presents our recent results on the involvement of MAO-A in the activation of mitochondrial death signal pathway and in the induction of prosurvival genes to prevent cell death with MAO-B inhibitors. The roles of MAO-A in the regulation of neuronal survival and death are discussed in concern to find a novel strategy to protect neurons in age-associated neurodegenerative disorders and depression.
Collapse
Affiliation(s)
- Makoto Naoi
- Department of Neurosciences, Gifu International Institute of Biotechnology, Kakamigahara, Gifu, Japan
| | | | | | | |
Collapse
|
45
|
Nigam M, Singh N, Ranjan V, Zaidi D, Sharma R, Nigam D, Gupta DK, Sundaram S, Balapure AK. Centchroman mediated apoptosis involves cross-talk between extrinsic/intrinsic pathways and oxidative regulation. Life Sci 2010; 87:750-8. [DOI: 10.1016/j.lfs.2010.10.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2010] [Revised: 09/29/2010] [Accepted: 10/15/2010] [Indexed: 01/02/2023]
|
46
|
Pan YC, Li CF, Ko CY, Pan MH, Chen PJ, Tseng JT, Wu WC, Chang WC, Huang AM, Sterneck E, Wang JM. CEBPD reverses RB/E2F1-mediated gene repression and participates in HMDB-induced apoptosis of cancer cells. Clin Cancer Res 2010; 16:5770-80. [PMID: 20971808 DOI: 10.1158/1078-0432.ccr-10-1025] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE Recent evidence indicates that a tumor suppressor gene CEBPD (CCAAT/enhancer-binding protein delta) is downregulated in many cancers including cervical cancer, which provides a therapeutic potential associated with its reactivation. However, little is known for CEBPD activators and the effect of reactivation of CEBPD transcription upon anticancer drug treatment. In this study, we identified a novel CEBPD activator, 1-(2-hydroxy-5-methylphenyl)-3-phenyl-1,3-propanedione (HMDB). The purpose of this study is to characterize the mechanism of HMDB-induced CEBPD activation and its potential effect in cancer therapy. EXPERIMENTAL DESIGN Methylation-specific PCR assay, reporter assay, and chromatin immunoprecipitation (ChIP) assay were performed to dissect the signaling pathway of HMDB-induced CEBPD transcription. Furthermore, a consequence of HMDB-induced CEBPD expression was linked with E2F1 and retinoblastoma (RB), which discloses the scenario of CEBPD, E2F1, and RB bindings and transcriptional regulation on the promoters of proapoptotic genes, PPARG2 and GADD153. Finally, the anticancer effect of HMDB was examined in xenograft mice. RESULTS We demonstrate that CEBPD plays an essential role in HMDB-mediated apoptosis of cancer cells. HMDB up-regulates CEBPD transcription through the p38/CREB pathway, thus leading to transcriptional activation of PPARG2 and GADD153. Furthermore, increased level of CEBPD attenuates E2F1-induced cancer cell proliferation and partially rescues RB/E2F1-mediated repression of PPARG2 and GADD153 transcription. Moreover, HMDB treatment attenuates the growth of A431 xenografts in severe combined immunodeficient mice mice. CONCLUSIONS These results clearly demonstrate that HMDB kills cancer cells through activation of CEBPD pathways and suggest that HMDB can serve as a superior chemotherapeutic agent with limited potential for adverse side effects.
Collapse
Affiliation(s)
- Yen-Chun Pan
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Chaturvedi R, Bansal K, Narayana Y, Kapoor N, Sukumar N, Togarsimalemath SK, Chandra N, Mishra S, Ajitkumar P, Joshi B, Katoch VM, Patil SA, Balaji KN. The multifunctional PE_PGRS11 protein from Mycobacterium tuberculosis plays a role in regulating resistance to oxidative stress. J Biol Chem 2010; 285:30389-403. [PMID: 20558725 DOI: 10.1074/jbc.m110.135251] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Mycobacterium tuberculosis utilizes unique strategies to survive amid the hostile environment of infected host cells. Infection-specific expression of a unique mycobacterial cell surface antigen that could modulate key signaling cascades can act as a key survival strategy in curtailing host effector responses like oxidative stress. We demonstrate here that hypothetical PE_PGRS11 ORF encodes a functional phosphoglycerate mutase. The transcriptional analysis revealed that PE_PGRS11 is a hypoxia-responsive gene, and enforced expression of PE_PGRS11 by recombinant adenovirus or Mycobacterium smegmatis imparted resistance to alveolar epithelial cells against oxidative stress. PE_PGRS11-induced resistance to oxidative stress necessitated the modulation of genetic signatures like induced expression of Bcl2 or COX-2. This modulation of specific antiapoptotic molecular signatures involved recognition of PE_PGRS11 by TLR2 and subsequent activation of the PI3K-ERK1/2-NF-κB signaling axis. Furthermore, PE_PGRS11 markedly diminished H(2)O(2)-induced p38 MAPK activation. Interestingly, PE_PGRS11 protein was exposed at the mycobacterial cell surface and was involved in survival of mycobacteria under oxidative stress. Furthermore, PE_PGRS11 displayed differential B cell responses during tuberculosis infection. Taken together, our investigation identified PE_PGRS11 as an in vivo expressed immunodominant antigen that plays a crucial role in modulating cellular life span restrictions imposed during oxidative stress by triggering TLR2-dependent expression of COX-2 and Bcl2. These observations clearly provide a mechanistic basis for the rescue of pathogenic Mycobacterium-infected lung epithelial cells from oxidative stress.
Collapse
Affiliation(s)
- Rashmi Chaturvedi
- Department of Microbiology and Cell Biology, Supercomputer Education and Research Centre, Indian Institute of Science, Bangalore 560012, India
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Chang CW, Tsai WH, Chuang WJ, Lin YS, Wu JJ, Liu CC, Tsai PJ, Lin MT. Procaspase 8 and Bax are up-regulated by distinct pathways in Streptococcal pyrogenic exotoxin B-induced apoptosis. J Biol Chem 2009; 284:33195-205. [PMID: 19801665 DOI: 10.1074/jbc.m109.020586] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have previously identified integrin alpha(v)beta(3) and Fas as receptors for the streptococcal pyrogenic exotoxin B (SPE B), and G308S, a mutant of SPE B that binds to Fas only. In the current study we found that after binding to alpha(v)beta(3), SPE B stimulated the tyrosine phosphorylation of JAK2 and STAT1. STAT1 tyrosine phosphorylation was inhibited by a JAK2 inhibitor, AG490, short interfering RNA (siRNA) silencing of JAK2, and anti-alpha(V)beta(3) antibody. AG490 also decreased the binding of tyrosine-phosphorylated STAT1 to the procaspase 8 promoter, decreasing procaspase 8 expression, suggesting that SPE B up-regulates procaspase 8 expression via the JAK2/STAT1 pathway. Alternatively, both SPE B and G308S increased STAT1 phosphorylation at serine 727, which was inhibited by anti-Fas antibody, a p38 inhibitor, SB203580, and siRNA silencing of p38. In addition, SPE B and G308S increased binding of serine-phosphorylated STAT1 to the Bax promoter and Bax expression, which was decreased by SB203580. SPE B and G308S-stimulated Bax expression was also inhibited by anti-Fas antibody. These findings suggest that Fas mediate SPE B-induced Bax expression through p38. Silencing of JAK2 or p38 by siRNA blocked procaspase 8 expression, whereas only p38 siRNA decreased Bax expression. Furthermore, JAK2 inhibition and p38 inhibition reduced SPE B-induced apoptosis, but only p38 inhibition blocked G308S-induced apoptosis.
Collapse
Affiliation(s)
- Chia-Wen Chang
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan, 701 Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Tikoo K, Ali IY, Gupta J, Gupta C. 5-Azacytidine prevents cisplatin induced nephrotoxicity and potentiates anticancer activity of cisplatin by involving inhibition of metallothionein, pAKT and DNMT1 expression in chemical induced cancer rats. Toxicol Lett 2009; 191:158-66. [PMID: 19723570 DOI: 10.1016/j.toxlet.2009.08.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2009] [Revised: 08/22/2009] [Accepted: 08/24/2009] [Indexed: 11/26/2022]
Abstract
5-Azactydine inhibits cell growth by direct cytotoxic action as well as by inhibition of DNA methyl transferase enzyme. Inhibitors of DNMT have been reported to potentiate the therapeutic activity of cisplatin in vitro. Dose dependent bone marrow toxicity, neurotoxicity and nephrotoxicity are the major side effects of cisplatin, limiting its use as an effective chemotherapeutic agent. The present study was aimed to reduce the nephrotoxic potential of cisplatin without compensating its potency. To best of our knowledge, this is the first report which shows that the combination of 5-azacytidine with cisplatin leads to remarkable reduction in nephrotoxicity, by involving inhibition of cisplatin induced metallothionein expression. 5-Azacytidine treatment with cisplatin leads to maximum reduction in tumor size in DMH induced colon cancer and tumor volume in DMBA induced breast cancer bearing SD rats. This combination regimen prevents phosphorylation and acetylation of histone H3 which may be involved in inhibition of aberrant gene expression in colon tumors. Further, 5-azacytidine potentiated cisplatin induced antitumor activity by involving decreased expression of pAKT, DNMT1 and an increased expression of p38 in colon tumors. Thus, combination of 5-azactydine with cisplatin attenuates the cisplatin induced nephrotoxicity and potentiates the anti-cancer activity which can have profound clinical implications.
Collapse
Affiliation(s)
- Kulbhushan Tikoo
- Laboratory of Chromatin Biology, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Mohali, Punjab 160 062, India.
| | | | | | | |
Collapse
|
50
|
Cao X, Rui L, Pennington PR, Chlan-Fourney J, Jiang Z, Wei Z, Li XM, Edmondson DE, Mousseau DD. Serine 209 resides within a putative p38(MAPK) consensus motif and regulates monoamine oxidase-A activity. J Neurochem 2009; 111:101-10. [PMID: 19650872 DOI: 10.1111/j.1471-4159.2009.06300.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The p38 mitogen-activated protein kinase (MAPK) cascade as well as the enzyme monoamine oxidase-A (MAO-A) have both been associated with oxidative stress. We observed that the specific inhibition of the p38(MAPK) protein [using either a chemical inhibitor or a dominant-negative p38(MAPK) clone] selectively induces MAO-A activity and MAO-A-sensitive toxicity in several neuronal cell lines, including primary cortical neurons. Over-expression of a constitutively active p38(MAPK) results in the phosphorylation of the MAO-A protein and inhibition of MAO-A activity. The MAO-A(Ser209Glu) phosphomimic - bearing a targeted substitution within a putative p38(MAPK) consensus motif - is neither active nor neurotoxic. In contrast, the MAO-A(Ser209Ala) variant (mimics dephosphorylation) does not associate with p38(MAPK), and is both very active and very toxic. Substitution of the homologous serine in the MAO-B isoform, i.e. Ser200, with either Glu or Ala does not affect the catalytic activity of the corresponding over-expressed proteins. These combined in vitro data strongly suggest a direct p38(MAPK)-dependent inhibition of MAO-A function. Based on published observations, this endogenous means of selectively regulating MAO-A function could provide for an adaptive response to oxidative stress associated with disorders as diverse as depression, reperfusion/ischemia, and the early stages of Alzheimer's disease.
Collapse
Affiliation(s)
- Xia Cao
- Cell Signalling Laboratory, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|