1
|
Prasad K, Raghu KS, Maruthiyodan S, Wadhwa R, Kaul SC, Satyamoorthy K, Guruprasad KP. Promotion of cellular differentiation and DNA repair potential in brain cancer cells by Clitoria ternatea L. with rasayana properties in vitro. J Ayurveda Integr Med 2025; 16:101050. [PMID: 39798267 PMCID: PMC11773022 DOI: 10.1016/j.jaim.2024.101050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 06/03/2024] [Accepted: 08/08/2024] [Indexed: 01/15/2025] Open
Abstract
BACKGROUND Brain ageing is accompanied by the diminution of neuronal plasticity, which is correlated with the inability to respond to loss of memory, various stress-induced stimuli, and increased risk of neurodegenerative disorders. In the recent past, plant based herbal medicines are of interest over synthetic drugs for therapeutic purposes due to lower side effects. The Indian traditional medicine Ayurveda describes several herbal remedies, such as rasayana (elixirs for rejuvenation), to treat many age-related diseases. Medhya rasayana (brain elixir) is a class of rasayana used for its nootropic functions, such as enhancement of memory and intellect, in addition to promoting normal health. Clitoria ternatea L. is one such plant used in the preparation of medhya rasayana. OBJECTIVE To investigate the neuronal differentiation and DNA repair potential of Shankhpushpi (Clitoria ternatea L.) in neuroblastoma cells. MATERIALS & METHODS The effect of Clitoria ternatea L. on neuronal cell differentiation, DNA repair (base excision repair, comet, γH2AX immunostaining assays), autophagy by cadaverine uptake and mitochondrial functions by fluorescent dye staining through flow cytometry were evaluated. RESULTS The results revealed that Clitoria ternatea L. enhanced DNA repair and mitochondrial membrane potential and reduced autophagy and reactive oxygen species (ROS) in IMR-32 neuroblastoma cells. Treatment of IMR-32 neuroblastoma and C6 glioblastoma cells with shankhpushpi induced neuronal differentiation and exhibited markers such as MAP2, Mortalin and GFAP. CONCLUSION Neurobiological pathways and molecular mechanisms influenced by rasayana herb shankhpushpi suggests its therapeutic potential for neurodegenerative diseases.
Collapse
Affiliation(s)
- Keshava Prasad
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Planetarium Complex, Manipal, 576 104, Karnataka, India
| | - Kothanahalli S Raghu
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Planetarium Complex, Manipal, 576 104, Karnataka, India
| | - Swathi Maruthiyodan
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Planetarium Complex, Manipal, 576 104, Karnataka, India
| | - Renu Wadhwa
- Drug Discovery and Assets Innovation DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Biomedical Research Institute, National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba, 305 8565, Japan
| | - Sunil C Kaul
- Drug Discovery and Assets Innovation DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Biomedical Research Institute, National Institute of Advanced Industrial Science & Technology (AIST), Tsukuba, 305 8565, Japan
| | - Kapaettu Satyamoorthy
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Planetarium Complex, Manipal, 576 104, Karnataka, India; Shri Dharmasthala Manjunatheshwara (SDM) University, SDM College of Medical Sciences and Hospital, Manjushree Nagar, Sattur, Dharwad, 580009, Karnataka, India
| | - Kanive Parashiva Guruprasad
- Centre for Ayurvedic Biology, Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| |
Collapse
|
2
|
Zhang Y, Zhang L, Zhao Y, He J, Zhang Y, Zhang X. PGC-1α inhibits M2 macrophage polarization and alleviates liver fibrosis following hepatic ischemia reperfusion injury. Cell Death Discov 2023; 9:337. [PMID: 37679346 PMCID: PMC10484946 DOI: 10.1038/s41420-023-01636-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 08/17/2023] [Accepted: 08/29/2023] [Indexed: 09/09/2023] Open
Abstract
Oxidative stress can induce inflammation, promoting macrophage polarization and liver fibrosis following hepatic ischemia-reperfusion (I/R). Peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) has anti-oxidant and anti-inflammatory activity. However, how PGC-1α regulates macrophage polarization following hepatic I/R remains largely unknown. Male C57BL/6 wild-type mice were pre-treated with vehicle or trichostatin A (TSA) for 2 days and subjected to surgical induction of I/R. Liver injury and fibrosis in individual mice were examined longitudinally and the expression levels of IL-6, STAT3, M2-type macrophage markers, Collagen I and α-SMA in the liver of mice were analyzed by immunohistochemistry, RT-qPCR and Western blot. The potential interaction of PGC-1α with phosphorylated NF-kBp65 was determined by immunoprecipitation. The impacts of PGC-1α deficiency in hepatocytes on their IL-6 production and macrophage polarization were tested in a Transwell co-culture system. Moreover, the M2-type macrophage polarization and liver fibrosis were examined in hepatocyte-specific PGC-1α knockout mice and AAV8-mediated PGC-1α over-expressing mice following liver I/R. The down-regulated PGC-1α expression by I/R was negatively correlated with IL-6 levels in the liver of I/R mice and PGC-1α deficiency enhanced IL-6 expression, STAT3 activation and M2-type macrophage polarization in the I/R mice, which were abrogated by TSA treatment. In addition, PGC-1α directly interacted with phosphorylated NF-kBp65 in I/R livers. Hepatocyte-specific PGC-1α deficiency increased IL-6 production and promoted macrophage polarization toward M2 type when co-culture. More importantly, administration with AAV8-PGC-1α rescued the I/R-induced liver fibrosis by inhibiting the IL-6/JAK2/STAT3 signaling and M2-type macrophage polarization in the liver. These results suggest that PGC-1α may alleviate the I/R-induced liver fibrosis by attenuating the IL-6/JAK2/STAT3 signaling to limit M2-type macrophage polarization. PGC-1α may be a therapeutic target for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Yanting Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Linzhong Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Department of Gastroenterology, Air Force Medical Center, Beijing, China
| | - Yanmian Zhao
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jing He
- Department of Internal Medicine, School Hospital, Communication University of China, Beijing, China
| | - Yanghao Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiuying Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.
| |
Collapse
|
3
|
Esteras N, Abramov AY. Nrf2 as a regulator of mitochondrial function: Energy metabolism and beyond. Free Radic Biol Med 2022; 189:136-153. [PMID: 35918014 DOI: 10.1016/j.freeradbiomed.2022.07.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/20/2022] [Accepted: 07/19/2022] [Indexed: 12/14/2022]
Abstract
Mitochondria are unique and essential organelles that mediate many vital cellular processes including energy metabolism and cell death. The transcription factor Nrf2 (NF-E2 p45-related factor 2) has emerged in the last few years as an important modulator of multiple aspects of mitochondrial function. Well-known for controlling cellular redox homeostasis, the cytoprotective effects of Nrf2 extend beyond its ability to regulate a diverse network of antioxidant and detoxification enzymes. Here, we review the role of Nrf2 in the regulation of mitochondrial function and structure. We focus on Nrf2 involvement in promoting mitochondrial quality control and regulation of basic aspects of mitochondrial function, including energy production, reactive oxygen species generation, calcium signalling, and cell death induction. Given the importance of mitochondria in the development of multiple diseases, these findings reinforce the pharmacological activation of Nrf2 as an attractive strategy to counteract mitochondrial dysfunction.
Collapse
Affiliation(s)
- Noemí Esteras
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London, UK.
| | - Andrey Y Abramov
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London, UK.
| |
Collapse
|
4
|
Soni D, Kumar P. GSK-3β-mediated regulation of Nrf2/HO-1 signaling as a new therapeutic approach in the treatment of movement disorders. Pharmacol Rep 2022; 74:557-569. [PMID: 35882765 DOI: 10.1007/s43440-022-00390-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/09/2022] [Accepted: 07/12/2022] [Indexed: 11/30/2022]
Abstract
Movement disorders are neurological conditions characterized by involuntary motor movements, such as dystonia, ataxia, chorea myoclonus, tremors, Huntington's disease (HD), and Parkinson's disease (PD). It is classified into two categories: hypokinetic and hyperkinetic movements. Globally, movement disorders are a major cause of death. The pathophysiological process is initiated by excessive ROS generation, mitochondrial dysfunction, neuroinflammation, and neurotransmitters imbalance that lead to motor dysfunction in PD and HD patients. Several endogenous targets including Nrf2 maintain oxidative balance in the body. Activation of Nrf2 signaling is regulated by the enzyme glycogen synthase kinase (GSK-3β). In the cytoplasm, inhibition of GSK-3β regulates cellular proliferation, homeostasis, and apoptotic process by stimulating the nuclear factor erythroid 2 (Nrf2) pathway which is involved in the elevation of the cellular antioxidant enzymes which controls the ROS generation. The activation of Nrf2 increases the expression of antioxidant response elements (ARE), such as (Hemeoxygenase-1) HO-1, which decreases excessive cellular stress, mitochondrial dysfunction, apoptosis, and neuronal degeneration, which is the major cause of motor dysfunction. The present review explores the GSK-3β-mediated neuroprotection in various movement disorders through the Nrf2/HO-1 antioxidant pathway. This review provides a link between GSK-3β and the Nrf2/HO-1 signaling pathway in the treatment of PD and HD. In addition to that it highlights various GSK-3β inhibitors and the Nrf2/HO-1 activators, which exert robust neuroprotection against motor disorders. Therefore, the present review will help in the discovery of new therapy for PD and HD patients.
Collapse
Affiliation(s)
- Divya Soni
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India.
| |
Collapse
|
5
|
Functional Deficits of 5×FAD Neural Stem Cells Are Ameliorated by Glutathione Peroxidase 4. Cells 2022; 11:cells11111770. [PMID: 35681465 PMCID: PMC9179411 DOI: 10.3390/cells11111770] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/23/2022] [Accepted: 05/26/2022] [Indexed: 12/31/2022] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia affecting millions of people around the globe. Impaired neurogenesis is reported in AD as well as in AD animal models, although the underlying mechanism remains unclear. Elevated lipid peroxidation products are well-documented in AD. In current study, the role of lipid peroxidation on neural stem cell (NSCs) function is tested. Neural stem cells (NSCs) from 5×FAD mice, a widely used AD model with impaired neurogenesis, were observed to have increased levels of lipid reactive oxygen species compared to NSCs from control WT mice. 5×FAD NSCs exhibited altered differentiation potential as revealed by their propensity to differentiate into astrocytic lineage instead of neuronal lineage compared to WT NSCs. In addition, 5×FAD NSCs showed a reduced level of Gpx4, a key enzyme in reducing hydroperoxides in membrane lipids, and this reduction appeared to be caused by enhanced autophagy-lysosomal degradation of Gpx4 protein. To test if increasing Gpx4 could restore differentiation potential, NSCs from 5×FAD and Gpx4 double transgenic mice, i.e., 5×FAD/GPX4 mice were studied. Remarkably, upon differentiation, neuronal linage cells increased significantly in 5×FAD/GPX4 cultures compared to 5×FAD cultures. Taken together, the findings suggest that deficiency of lipid peroxidation defense contributes to functional decline of NSCs in AD.
Collapse
|
6
|
Reichmann H. [Caffeine, Chocolate and Adenosine A2A Receptor Antagonists in the Treatment of Parkinson's Disease]. FORTSCHRITTE DER NEUROLOGIE-PSYCHIATRIE 2022. [PMID: 35584767 DOI: 10.1055/a-1785-3632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND Patients with Parkinson's disease can apparently benefit from caffeine consumption, as a number of experimental and clinical studies have already shown. METHODS The review examined the available literature on caffeine and Parkinson's disease. RESULTS Caffeine can penetrate the blood-brain barrier and exerts its biological effects mainly by antagonizing adenosine receptors. Numerous studies indicate that caffeine and its derivatives theobromine and theophylline are associated with a reduced risk of Parkinson's disease. Caffeine and adenosine antagonists reduce the excitotoxicity caused by glutamate. Evidence from animal models supports the potential of A2A receptor antagonism as an innovative disease-modifying target in Parkinson's disease CONCLUSION: The present review shows that the investigation and synthesis of xanthine derivatives as well as their analysis in clinical studies could be a promising approach in the therapy of neurodegenerative diseases.
Collapse
|
7
|
Babaei P, Azari HB. Exercise Training Improves Memory Performance in Older Adults: A Narrative Review of Evidence and Possible Mechanisms. Front Hum Neurosci 2022; 15:771553. [PMID: 35153701 PMCID: PMC8829997 DOI: 10.3389/fnhum.2021.771553] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/16/2021] [Indexed: 12/15/2022] Open
Abstract
As human life expectancy increases, cognitive decline and memory impairment threaten independence and quality of life. Therefore, finding prevention and treatment strategies for memory impairment is an important health concern. Moreover, a better understanding of the mechanisms involved underlying memory preservation will enable the development of appropriate pharmaceuticals drugs for those who are activity limited. Exercise training as a non-pharmacological tool, has been known to increase the mean lifespan by maintaining general body health and improving the cardiovascular and nervous systems function. Among different exercise training protocols, aerobic exercise has been reported to prevent the progression of memory decline, provided adequate exertion level, duration, and frequency. Mechanisms underlying exercise training effects on memory performance have not been understood yet. Convergent evidence suggest several direct and indirect mechanisms at molecular and supramolecular levels. The supramolecular level includes improvement in blood circulation, synaptic plasticity and neurogenesis which are under controls of complex molecular signaling of neurotransmitters, neurotrophic factors, exerkines, and epigenetics factors. Among these various factors, irisin/BDNF signaling seems to be one of the important mediators of crosstalk between contracted skeletal muscles and the brain during exercise training. This review provides an affordable and effective method to improve cognitive function in old ages, particularly those who are most vulnerable to neurodegenerative disorders.
Collapse
Affiliation(s)
- Parvin Babaei
- Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Helya Bolouki Azari
- Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Physiology, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Bisdemethoxycurcumin Protects Small Intestine from Lipopolysaccharide-Induced Mitochondrial Dysfunction via Activating Mitochondrial Antioxidant Systems and Mitochondrial Biogenesis in Broiler Chickens. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9927864. [PMID: 34795844 PMCID: PMC8595021 DOI: 10.1155/2021/9927864] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 10/04/2021] [Accepted: 10/25/2021] [Indexed: 12/11/2022]
Abstract
Bisdemethoxycurcumin is one of the three curcuminoids of turmeric and exhibits good antioxidant activity in animal models. This study is aimed at investigating the effect of bisdemethoxycurcumin on small intestinal mitochondrial dysfunction in lipopolysaccharide- (LPS-) treated broilers, especially on the mitochondrial thioredoxin 2 system and mitochondrial biogenesis. A total of 320 broiler chickens were randomly assigned into four experimental diets using a 2 × 2 factorial arrangement with diet (0 and 150 mg/kg bisdemethoxycurcumin supplementation) and stress (saline or LPS challenge) for 20 days. Broilers received a dose of LPS (1 mg/kg body weight) or sterile saline intraperitoneally on days 16, 18, and 20 of the trial. Bisdemethoxycurcumin mitigated the mitochondrial dysfunction of jejunum and ileum induced by LPS, as evident by the reduced reactive oxygen species levels and the increased mitochondrial membrane potential. Bisdemethoxycurcumin partially reversed the decrease in the mitochondrial DNA copy number and the depletion of ATP levels. Bisdemethoxycurcumin activated the mitochondrial antioxidant response, including the prevention of lipid peroxidation, enhancement of manganese superoxide dismutase activity, and the upregulation of the mitochondrial glutaredoxin 5 and thioredoxin 2 system. The enhanced mitochondrial respiratory complex activities in jejunum and ileum were also attributed to bisdemethoxycurcumin treatment. In addition, bisdemethoxycurcumin induced mitochondrial biogenesis via transcriptional regulation of proliferator-activated receptor-gamma coactivator-1alpha pathway. In conclusion, our results demonstrated the potential of bisdemethoxycurcumin to attenuate small intestinal mitochondrial dysfunction, which might be mediated via activating the mitochondrial antioxidant system and mitochondrial biogenesis in LPS-treated broilers.
Collapse
|
9
|
Burtscher J, Millet GP, Place N, Kayser B, Zanou N. The Muscle-Brain Axis and Neurodegenerative Diseases: The Key Role of Mitochondria in Exercise-Induced Neuroprotection. Int J Mol Sci 2021; 22:6479. [PMID: 34204228 PMCID: PMC8235687 DOI: 10.3390/ijms22126479] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/13/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022] Open
Abstract
Regular exercise is associated with pronounced health benefits. The molecular processes involved in physiological adaptations to exercise are best understood in skeletal muscle. Enhanced mitochondrial functions in muscle are central to exercise-induced adaptations. However, regular exercise also benefits the brain and is a major protective factor against neurodegenerative diseases, such as the most common age-related form of dementia, Alzheimer's disease, or the most common neurodegenerative motor disorder, Parkinson's disease. While there is evidence that exercise induces signalling from skeletal muscle to the brain, the mechanistic understanding of the crosstalk along the muscle-brain axis is incompletely understood. Mitochondria in both organs, however, seem to be central players. Here, we provide an overview on the central role of mitochondria in exercise-induced communication routes from muscle to the brain. These routes include circulating factors, such as myokines, the release of which often depends on mitochondria, and possibly direct mitochondrial transfer. On this basis, we examine the reported effects of different modes of exercise on mitochondrial features and highlight their expected benefits with regard to neurodegeneration prevention or mitigation. In addition, knowledge gaps in our current understanding related to the muscle-brain axis in neurodegenerative diseases are outlined.
Collapse
Affiliation(s)
- Johannes Burtscher
- Institute of Sport Sciences, University of Lausanne, CH-1015 Lausanne, Switzerland; (G.P.M.); (N.P.); (B.K.); (N.Z.)
- Department of Biomedical Sciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| | - Grégoire P. Millet
- Institute of Sport Sciences, University of Lausanne, CH-1015 Lausanne, Switzerland; (G.P.M.); (N.P.); (B.K.); (N.Z.)
- Department of Biomedical Sciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| | - Nicolas Place
- Institute of Sport Sciences, University of Lausanne, CH-1015 Lausanne, Switzerland; (G.P.M.); (N.P.); (B.K.); (N.Z.)
- Department of Biomedical Sciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| | - Bengt Kayser
- Institute of Sport Sciences, University of Lausanne, CH-1015 Lausanne, Switzerland; (G.P.M.); (N.P.); (B.K.); (N.Z.)
- Department of Biomedical Sciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| | - Nadège Zanou
- Institute of Sport Sciences, University of Lausanne, CH-1015 Lausanne, Switzerland; (G.P.M.); (N.P.); (B.K.); (N.Z.)
- Department of Biomedical Sciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| |
Collapse
|
10
|
Dodson M, Anandhan A, Zhang DD, Madhavan L. An NRF2 Perspective on Stem Cells and Ageing. FRONTIERS IN AGING 2021; 2:690686. [PMID: 36213179 PMCID: PMC9536878 DOI: 10.3389/fragi.2021.690686] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 06/03/2021] [Indexed: 04/24/2023]
Abstract
Redox and metabolic mechanisms lie at the heart of stem cell survival and regenerative activity. NRF2 is a major transcriptional controller of cellular redox and metabolic homeostasis, which has also been implicated in ageing and lifespan regulation. However, NRF2's role in stem cells and their functioning with age is only just emerging. Here, focusing mainly on neural stem cells, which are core to adult brain plasticity and function, we review recent findings that identify NRF2 as a fundamental player in stem cell biology and ageing. We also discuss NRF2-based molecular programs that may govern stem cell state and function with age, and implications of this for age-related pathologies.
Collapse
Affiliation(s)
- Matthew Dodson
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, United States
| | - Annadurai Anandhan
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, United States
- Department of Neurology, University of Arizona, Tucson, AZ, United States
| | - Donna D. Zhang
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, United States
| | - Lalitha Madhavan
- Department of Neurology, University of Arizona, Tucson, AZ, United States
- Evelyn F. McKnight Brain Institute and Bio5 Institute, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
11
|
Chaudhary R, Agarwal V, Kaushik AS, Rehman M. Involvement of myocyte enhancer factor 2c in the pathogenesis of autism spectrum disorder. Heliyon 2021; 7:e06854. [PMID: 33981903 PMCID: PMC8082549 DOI: 10.1016/j.heliyon.2021.e06854] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/09/2020] [Accepted: 04/15/2021] [Indexed: 12/29/2022] Open
Abstract
Myocyte enhancer factor 2 (MEF2), a family of transcription factor of MADS (minichromosome maintenance 1, agamous, deficiens and serum response factor)-box family needed in the growth and differentiation of a variety of human cells, such as neural, immune, endothelial, and muscles. As per existing literature, MEF2 transcription factors have also been associated with synaptic plasticity, the developmental mechanisms governing memory and learning, and several neurologic conditions, like autism spectrum disorders (ASDs). Recent genomic findings have ascertained a link between MEF2 defects, particularly in the MEF2C isoform and the ASD. In this review, we summarized a concise overview of the general regulation, structure and functional roles of the MEF2C transcription factor. We further outlined the potential role of MEF2C as a risk factor for various neurodevelopmental disorders, such as ASD, MEF2C Haploinsufficiency Syndrome and Fragile X syndrome.
Collapse
Affiliation(s)
- Rishabh Chaudhary
- Department of Pharmaceutical Sciences, School of Biosciences and Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Vipul Agarwal
- Department of Pharmaceutical Sciences, School of Biosciences and Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Arjun Singh Kaushik
- Department of Pharmaceutical Sciences, School of Biosciences and Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Mujeeba Rehman
- Department of Pharmaceutical Sciences, School of Biosciences and Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| |
Collapse
|
12
|
Luo M, Gan RY, Li BY, Mao QQ, Shang A, Xu XY, Li HY, Li HB. Effects and Mechanisms of Tea on Parkinson’s Disease, Alzheimer’s Disease and Depression. FOOD REVIEWS INTERNATIONAL 2021. [DOI: 10.1080/87559129.2021.1904413] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Min Luo
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou
| | - Ren-You Gan
- Research Center for Plants and Human Health, Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences (CAAS), Chengdu, China
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), Sichuan Engineering & Technology Research Center of Coarse Cereal Industralization, Chengdu University, Chengdu, China
| | - Bang-Yan Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou
| | - Qian-Qian Mao
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou
| | - Ao Shang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou
| | - Xiao-Yu Xu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou
| | - Hang-Yu Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou
| | - Hua-Bin Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou
| |
Collapse
|
13
|
Chen X, Zhang Y, Wang Q, Qin Y, Yang X, Xing Z, Shen Y, Wu H, Qi Y. The function of SUMOylation and its crucial roles in the development of neurological diseases. FASEB J 2021; 35:e21510. [PMID: 33710677 DOI: 10.1096/fj.202002702r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 02/02/2021] [Accepted: 02/22/2021] [Indexed: 11/11/2022]
Abstract
Neurological diseases are relatively complex diseases of a large system; however, the detailed mechanism of their pathogenesis has not been completely elucidated, and effective treatment methods are still lacking for some of the diseases. The SUMO (small ubiquitin-like modifier) modification is a dynamic and reversible process that is catalyzed by SUMO-specific E1, E2, and E3 ligases and reversed by a family of SENPs (SUMO/Sentrin-specific proteases). SUMOylation covalently conjugates numerous cellular proteins, and affects their cellular localization and biological activity in numerous cellular processes. A wide range of neuronal proteins have been identified as SUMO substrates, and the disruption of SUMOylation results in defects in synaptic plasticity, neuronal excitability, and neuronal stress responses. SUMOylation disorders cause many neurodegenerative diseases, such as Parkinson's disease, Alzheimer's disease, and Huntington's disease. By modulating the ion channel subunit, SUMOylation imbalance is responsible for the development of various channelopathies. The regulation of protein SUMOylation in neurons may provide a new strategy for the development of targeted therapeutic drugs for neurodegenerative diseases and channelopathies.
Collapse
Affiliation(s)
- Xu Chen
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yuhong Zhang
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Qiqi Wang
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yuanyuan Qin
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Xinyi Yang
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Zhengcao Xing
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yajie Shen
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Hongmei Wu
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yitao Qi
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
14
|
Sheng X, Yang S, Wen X, Zhang X, Ye Y, Zhao P, Zang L, Peng K, Du E, Li S. Neuroprotective effects of Shende'an tablet in the Parkinson's disease model. Chin Med 2021; 16:18. [PMID: 33549148 PMCID: PMC7866695 DOI: 10.1186/s13020-021-00429-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 12/20/2020] [Accepted: 01/28/2021] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Shende'an tablet (SDA) is a newly capsuled Chinese herbal formula derived from the Chinese traditional medicine Zhengan Xifeng Decoction which is approved for the treatment of neurasthenia and insomnia in China. This study aimed to investigate the neuroprotective effects of SDA against Parkinson's disease (PD) in vitro and in vivo. METHODS In the present work, the neuroprotective effects and mechanism of SDA were evaluated in the cellular PD model. Male C57BL/6J mice were subject to a partial MPTP lesion alongside treatment with SDA. Behavioural test and tyrosine-hydroxylase immunohistochemistry were used to evaluate nigrostriatal tract integrity. HPLC analysis and Western blotting were used to assess the effect of SDA on dopamine metabolism and the expression of HO-1, PGC-1α and Nrf2, respectively. RESULTS Our results demonstrated that SDA had neuroprotective effect in dopaminergic PC12 cells with 6-OHDA lesion. It had also displayed efficient dopaminergic neuronal protection and motor behavior alleviation properties in MPTP-induced PD mice. In the PC12 cells and MPTP-induced Parkinson's disease animal models, SDA was highly efficacious in α-synuclein clearance associated with the activation of PGC-1α/Nrf2 signal pathway. CONCLUSIONS SDA demonstrated potential as a future therapeutic modality in PD through protecting dopamine neurons and alleviating the motor symptoms, mediated by the activation of PGC-1α/Nrf2 signal pathway.
Collapse
Affiliation(s)
- Xiaoyan Sheng
- Nursing Department, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, Guangdong, China
| | - Shuiyuan Yang
- Department of Pharmacy, Guangdong Second Provincial General Hospital, Guangzhou, 510317, Guangdong, China
| | - Xiaomin Wen
- The Centre of Preventive, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, Guangdong, China
| | - Xin Zhang
- Department of Pharmacy, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, Guangdong, China
| | - Yongfeng Ye
- Department of Pharmacy, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, Guangdong, China
| | - Peng Zhao
- The Centre of Preventive, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, Guangdong, China
| | - Limin Zang
- Zhengzhou Yihe Hospital of Henan University, Zhengzhou, 450047, Henan, China
| | - Kang Peng
- The Centre of Preventive, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, Guangdong, China.
| | - Enming Du
- Henan Eye Institute, Henan Eye Hospital, Henan Key Laboratory of Ophthalmology and Visual Science, People's Hospital of Zhengzhou University, Henan University, School of Medicine, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China.
| | - Sai Li
- Department of Pharmacy, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, Guangdong, China.
| |
Collapse
|
15
|
Fracassi A, Marcatti M, Zolochevska O, Tabor N, Woltjer R, Moreno S, Taglialatela G. Oxidative Damage and Antioxidant Response in Frontal Cortex of Demented and Nondemented Individuals with Alzheimer's Neuropathology. J Neurosci 2021; 41:538-554. [PMID: 33239403 PMCID: PMC7821866 DOI: 10.1523/jneurosci.0295-20.2020] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 11/21/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by progressive neurodegeneration in the cerebral cortex, histopathologically hallmarked by amyloid β (Aβ) extracellular plaques and intracellular neurofibrillary tangles, constituted by hyperphosphorylated tau protein. Correlation between these pathologic features and dementia has been challenged by the emergence of "nondemented with Alzheimer's neuropathology" (NDAN) individuals, cognitively intact despite displaying pathologic features of AD. The existence of these subjects suggests that some unknown mechanisms are triggered to resist Aβ-mediated detrimental events. Aβ accumulation affects mitochondrial redox balance, increasing oxidative stress status, which in turn is proposed as a primary culprit in AD pathogenesis. To clarify the relationship linking Aβ, oxidative stress, and cognitive impairment, we performed a comparative study on AD, NDAN, and aged-matched human postmortem frontal cortices of either sex. We quantitatively analyzed immunofluorescence distribution of oxidative damage markers, and of SOD2 (superoxide dismutase 2), PGC1α [peroxisome proliferator-activated receptor (PPAR) γ-coactivator 1α], PPARα, and catalase as key factors in antioxidant response, as well as the expression of miRNA-485, as a PGC1α upstream regulator. Our results confirm dramatic redox imbalance, associated with impaired antioxidant defenses in AD brain. By contrast, NDAN individuals display low oxidative damage, which is associated with high levels of scavenging systems, possibly resulting from a lack of PGC1α miRNA-485-related inhibition. Comparative analyses in neurons and astrocytes further highlighted cell-specific mechanisms to counteract redox imbalance. Overall, our data emphasize the importance of transcriptional and post-transcriptional regulation of antioxidant response in AD. This suggests that an efficient PGC1α-dependent "safety mechanism" may prevent Aβ-mediated oxidative stress, supporting neuroprotective therapies aimed at ameliorating defects in antioxidant response pathways in AD patients.
Collapse
Affiliation(s)
- Anna Fracassi
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch (UTMB), Galveston, Texas 77550
| | - Michela Marcatti
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch (UTMB), Galveston, Texas 77550
| | - Olga Zolochevska
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch (UTMB), Galveston, Texas 77550
| | - Natalie Tabor
- Neuroscience Summer Undergraduate Program, University of Texas Medical Branch, Galveston, Texas 77555
| | - Randall Woltjer
- Department of Pathology, Oregon Health and Science University, Portland, Oregon 97239-3098
| | - Sandra Moreno
- Department of Science, LIME, University Roma Tre, 00146 Rome, Italy
| | - Giulio Taglialatela
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch (UTMB), Galveston, Texas 77550
| |
Collapse
|
16
|
The Nrf2/PGC1 α Pathway Regulates Antioxidant and Proteasomal Activity to Alter Cisplatin Sensitivity in Ovarian Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4830418. [PMID: 33294122 PMCID: PMC7714579 DOI: 10.1155/2020/4830418] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/13/2020] [Accepted: 10/21/2020] [Indexed: 01/07/2023]
Abstract
Drug resistance remains a barrier in the clinical treatment of ovarian cancer. Proteasomal and antioxidant activities play important roles in tumor drug resistance, and increasing evidence suggests the existence of an interaction between antioxidant and proteasomal activities. However, the mechanism of the synergistic effects of proteasomal activity and antioxidation on tumor drug resistance is not completely clear. In this study, we compared two ovarian cancer cells, A2780 and SKOV3 cells. Among them, SKOV3 cell is a human clear cell carcinoma cell line that is resistant to platinum. We found that compared with the findings in A2780 cells, SKOV3 cells were less sensitive to both proteasomal inhibitor and cisplatin. Proteasomal inhibition enhanced the sensitivity of A2780 cells, but not SKOV3 cells, to cisplatin. Notably, the Nrf2-mediated antioxidant pathway was identified as a resistance mechanism in proteasome inhibitor-resistant cells, but this was not the only factor identified in our research. In SKOV3 cells, PGC1α regulated the antioxidant activity of Nrf2 by increasing the phosphorylation of GSK3β, and in turn, Nrf2 regulated the transcriptional activity of PGC1α. Thus, Nrf2 and PGC1α synergistically participate in the regulation of proteasomal activity. Furthermore, the Nrf2/PGC1α pathway participated in the regulation of mitochondrial function and homeostasis, further regulating proteasomal activity in SKOV3 cells. Therefore, exploring the roles of PGC1α and Nrf2 in the regulation of proteasomal activity by antioxidant and mitochondrial functions may provide new avenues for reversing drug resistance in ovarian cancer.
Collapse
|
17
|
Schepici G, Silvestro S, Bramanti P, Mazzon E. Caffeine: An Overview of Its Beneficial Effects in Experimental Models and Clinical Trials of Parkinson's Disease. Int J Mol Sci 2020; 21:ijms21134766. [PMID: 32635541 PMCID: PMC7369844 DOI: 10.3390/ijms21134766] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/30/2020] [Accepted: 07/03/2020] [Indexed: 12/20/2022] Open
Abstract
Parkinson’s Disease (PD) is a neurological disease characterized by the progressive degeneration of the nigrostriatal dopaminergic pathway with consequent loss of neurons in the substantia nigra pars compacta and dopamine depletion. The cytoplasmic inclusions of α-synuclein (α-Syn), known as Lewy bodies, are the cytologic hallmark of PD. The presence of α-Syn aggregates causes mitochondrial degeneration, responsible for the increase in oxidative stress and consequent neurodegeneration. PD is a progressive disease that shows a complicated pathogenesis. The current therapies are used to alleviate the symptoms of the disease without changing its clinical course. Recently, phytocompounds with neuroprotective effects and antioxidant properties such as caffeine have aroused the interest of researchers. The purpose of this review is to summarize the preclinical studies present in the literature and clinical trials recorded in ClinicalTrial.gov, aimed at illustrating the effects of caffeine used as a nutraceutical compound combined with the current PD therapies. Therefore, the preventive effects of caffeine in the neurodegeneration of dopaminergic neurons encourage the use of this alkaloid as a supplement to reduce the progress of the PD.
Collapse
|
18
|
Zhou ZD, Xie SP, Saw WT, Ho PGH, Wang H, Lei Z, Yi Z, Tan EK. The Therapeutic Implications of Tea Polyphenols Against Dopamine (DA) Neuron Degeneration in Parkinson's Disease (PD). Cells 2019; 8:cells8080911. [PMID: 31426448 PMCID: PMC6721683 DOI: 10.3390/cells8080911] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 08/14/2019] [Indexed: 12/15/2022] Open
Abstract
: Accumulative evidence indicated that the pathologically accumulated metal ions (iron species and Mn3+) and abnormally up-regulated monoamine oxidase B (MAOB) activity induced oxidation of endogenous dopamine (DA) can lead to mitochondria impairment, lysosome dysfunction, proteasome inhibition, and selective DA neuron vulnerability, which is implicated in the pathogenesis of Parkinson's disease (PD). The DA oxidation can generate deleterious reactive oxygen species (ROS) and highly reactive DA quinones (DAQ) to induce DA-related toxicity, which can be alleviated by DA oxidation suppressors, ROS scavengers, DAQ quenchers, and MAOB inhibitors. On the other hand, the nuclear factor erythroid 2-related factor 2 (Nrf2)-Keap1 and Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) anti-oxidative and proliferative signaling pathways play roles in anti-oxidative cell defense and mitochondria biogenesis, which is implicated in DA neuron protections. Therefore, agents with capabilities to suppress DA-related toxicity including inhibition of DA oxidation, scavenge of ROS, detoxification of DAQ, inhibition of MAOB, and modulations of anti-oxidative signaling pathways can be protective to DA neurons. Accumulative evidence shows that tea or coffee consumptions and smoking are related to deceased PD prevalence with unknown mechanisms. In this study, we investigate the protective capabilities of tea polyphenols and other PD relevant agents to inhibit DA-related toxicity and protect against environmental or genetic factors induced DA neuron degeneration in vitro and in vivo. We find that tea polyphenols can significantly suppress DA-related toxicity to protect DA neurons. The tea polyphenols can protect DA neurons via inhibition of DA oxidation, conjugation with DAQ, scavenge of ROS, inhibition of MAOB, and modulations of Nrf2-Keap1 and PGC-1α anti-oxidative signaling pathways. The tea polyphenols with more phenolic hydroxyl groups and ring structures have stronger protective functions. The protective capabilities of tea polyphenols is further strengthened by evidence that phenolic hydroxyl groups can directly conjugate with DAQ. However, GSH and other sulfhydyl groups containing agents have weaker capabilities to abrogate DA oxidation, detoxify ROS and DAQ and inhibit MAOB; whereas nicotine (NICO) and caffeine (CAF) can only modulate Nrf2-Keap1 and PGC-1α pathways to protect DA neurons weakly. The tea polyphenols are identified to protect against overexpression of mutant A30P α-synuclein (α-syn) induced DA neuron degeneration and PD-like symptoms in transgenic Drosophila. Based on achievements from current studies, the excellent and versatile protective capabilities of tea polyphenols are highlighted, which will contribute and benefit to future anti-PD therapy.
Collapse
Affiliation(s)
- Zhi Dong Zhou
- Department of Research, National Neuroscience Institute, Singapore 308433, Singapore.
- Signature Research Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore 169857, Singapore.
| | - Shao Ping Xie
- Department of Research, National Neuroscience Institute, Singapore 308433, Singapore
| | - Wuan Ting Saw
- Department of Research, National Neuroscience Institute, Singapore 308433, Singapore
| | - Patrick Ghim Hoe Ho
- Department of Research, National Neuroscience Institute, Singapore 308433, Singapore
| | - Hongyan Wang
- Signature Research Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Zhou Lei
- Ocular Proteomics Laboratory, Singapore Eye Research Institute, Singapore 169856, Singapore
- Singapore Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Research Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Zhao Yi
- Department of Neurology, Singapore General Hospital, Singapore 169608, Singapore
| | - Eng King Tan
- Department of Research, National Neuroscience Institute, Singapore 308433, Singapore.
- Signature Research Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore 169857, Singapore.
- Department of Neurology, Singapore General Hospital, Singapore 169608, Singapore.
| |
Collapse
|
19
|
SC79, a novel Akt activator, protects dopaminergic neuronal cells from MPP + and rotenone. Mol Cell Biochem 2019; 461:81-89. [PMID: 31342299 DOI: 10.1007/s11010-019-03592-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/15/2019] [Indexed: 12/31/2022]
Abstract
In pathogenesis of Parkinson's disease (PD), mitochondrial dysfunction causes substantial reactive oxygen species (ROS) production and oxidative stress, leading to dopaminergic (DA) neuronal cell death. Mitochondrial toxins, including MPP+ (1-methyl-4-phenylpyridinium ion) and rotenone, induce oxidative injury in cultured DA neuronal cells. The current study tested the potential effect of SC79, a first-in-class small-molecule Akt activator, against the process. In SH-SY5Y cells and primary murine DA neurons, SC79 significantly attenuated MPP+- and rotenone-induced viability reduction, cell death, and apoptosis. SC79 activated Akt signaling in DA neuronal cells. Akt inhibition (by LY294002 and MK-2206) or CRISPR-Cas9-mediated Akt1 knockout completely abolished SC79-induced DA neuroprotection against MPP+. Further studies demonstrated that SC79 attenuated MPP+- and rotenone-induced ROS production, mitochondrial depolarization, and lipid peroxidation in SH-SY5Y cells and primary DA neurons. Moreover, upregulation of Nrf2-dependent genes (HO1 and NQO1) and Nrf2 protein stabilization were detected in SC79-treated SH-SY5Y cells and primary DA neurons. Together we show that SC79 protects DA neuronal cells from mitochondrial toxins possibly via activation of Akt-Nrf2 signaling.
Collapse
|
20
|
MitoQ ameliorates testis injury from oxidative attack by repairing mitochondria and promoting the Keap1-Nrf2 pathway. Toxicol Appl Pharmacol 2019; 370:78-92. [DOI: 10.1016/j.taap.2019.03.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/21/2019] [Accepted: 03/01/2019] [Indexed: 11/21/2022]
|
21
|
Sarni AR, Baroni L. Milk and Parkinson disease: Could galactose be the missing link. MEDITERRANEAN JOURNAL OF NUTRITION AND METABOLISM 2019. [DOI: 10.3233/mnm-180234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
| | - Luciana Baroni
- Primary Care Unit, Northern District, Local Health Unit 2 Marca Trevigiana, Treviso, Italy
| |
Collapse
|
22
|
Lv J, Jiang S, Yang Z, Hu W, Wang Z, Li T, Yang Y. PGC-1α sparks the fire of neuroprotection against neurodegenerative disorders. Ageing Res Rev 2018; 44:8-21. [PMID: 29580918 DOI: 10.1016/j.arr.2018.03.004] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 03/12/2018] [Accepted: 03/20/2018] [Indexed: 12/30/2022]
Abstract
Recently, growing evidence has demonstrated that peroxisome proliferator activated receptor γ (PPARγ) coactivator-1α (PGC-1α) is a superior transcriptional regulator that acts via controlling the expression of anti-oxidant enzymes and uncoupling proteins and inducing mitochondrial biogenesis, which plays a beneficial part in the central nervous system (CNS). Given the significance of PGC-1α, we summarize the current literature on the molecular mechanisms and roles of PGC-1α in the CNS. Thus, in this review, we first briefly introduce the basic characteristics regarding PGC-1α. We then depict some of its important cerebral functions and discuss upstream modulators, partners, and downstream effectors of the PGC-1α signaling pathway. Finally, we highlight recent progress in research on the involvement of PGC-1α in certain major neurodegenerative disorders (NDDs), including Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. Collectively, the data presented here may be useful for supporting the future potential of PGC-1α as a therapeutic target.
Collapse
Affiliation(s)
- Jianjun Lv
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China; Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Shuai Jiang
- Department of Aerospace Medicine, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Zhi Yang
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Wei Hu
- Department of Immunology, The Fourth Military Medical University,169 Changle West Road, Xi'an 710032, China
| | - Zheng Wang
- Department of Cadio-Thoracic Surgery, Wuhan General Hospital of The People's Liberation Army, 627 Wuluo Road, Wuhan 430070, China
| | - Tian Li
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China; Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China.
| |
Collapse
|
23
|
Xi Y, Feng D, Tao K, Wang R, Shi Y, Qin H, Murphy MP, Yang Q, Zhao G. MitoQ protects dopaminergic neurons in a 6-OHDA induced PD model by enhancing Mfn2-dependent mitochondrial fusion via activation of PGC-1α. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2859-2870. [PMID: 29842922 DOI: 10.1016/j.bbadis.2018.05.018] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 05/22/2018] [Accepted: 05/24/2018] [Indexed: 11/16/2022]
Abstract
Parkinson's disease (PD) is characterized by the degeneration of dopaminergic neurons in the substantia nigra compacta (SNc). Although mitochondrial dysfunction is the critical factor in the pathogenesis of PD, the underlying molecular mechanisms are not well understood, and as a result, effective medical interventions are lacking. Mitochondrial fission and fusion play important roles in the maintenance of mitochondrial function and cell viability. Here, we investigated the effects of MitoQ, a mitochondria-targeted antioxidant, in 6-hydroxydopamine (6-OHDA)-induced in vitro and in vivo PD models. We observed that 6-OHDA enhanced mitochondrial fission by decreasing the expression of Mfn1, Mfn2 and OPA1 as well as by increasing the expression of Drp1 in the dopaminergic (DA) cell line SN4741. Notably, MitoQ treatment particularly upregulated the Mfn2 protein and mRNA levels and promoted mitochondrial fusion in the presence of 6-OHDA in a Mfn2-dependent manner. In addition, MitoQ also stabilized mitochondrial morphology and function in the presence of 6-OHDA, which further suppressed the formation of reactive oxygen species (ROS), as well as ameliorated mitochondrial fragmentation and cellular apoptosis. Moreover, the activation of peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) was attributed to the upregulation of Mfn2 induced by MitoQ. Consistent with these findings, administration of MitoQ in 6-OHDA-treated mice significantly rescued the decrease of Mfn2 expression and the loss of DA neurons in the SNc. Taken together, our findings suggest that MitoQ protects DA neurons in a 6-OHDA induced PD model by activating PGC-1α to enhance Mfn2-dependent mitochondrial fusion.
Collapse
Affiliation(s)
- Ye Xi
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Dayun Feng
- Department of Neurosurgery and Institute for Functional Brain Disorders, Tangdu Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Kai Tao
- Department of Neurosurgery and Institute for Functional Brain Disorders, Tangdu Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Ronglin Wang
- Department of Neurosurgery and Institute for Functional Brain Disorders, Tangdu Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Yajun Shi
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Huaizhou Qin
- Department of Neurosurgery and Institute for Functional Brain Disorders, Tangdu Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Qian Yang
- Department of Neurosurgery and Institute for Functional Brain Disorders, Tangdu Hospital, Fourth Military Medical University, Xi'an, PR China.
| | - Gang Zhao
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China.
| |
Collapse
|
24
|
Paul KC, Sinsheimer JS, Cockburn M, Bronstein JM, Bordelon Y, Ritz B. NFE2L2, PPARGC1α, and pesticides and Parkinson's disease risk and progression. Mech Ageing Dev 2018; 173:1-8. [PMID: 29630901 DOI: 10.1016/j.mad.2018.04.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 02/27/2018] [Accepted: 04/04/2018] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To investigate three expression-altering NFE2L2 SNPs and four PPARGC1α previously implicated SNPs and pesticides on Parkinson's disease (PD) risk and symptom progression. METHODS In 472 PD patients and 532 population-based controls, we examined variants and their interactions with maneb and paraquat (MB/PQ) pesticide exposure on PD onset (logistic regression) and progression of motor symptoms and cognitive decline (n = 192; linear repeated measures). RESULTS NFE2L2 rs6721961 T allele was associated with a reduced risk of PD (OR = 0.70, 95% CI = 0.53, 0.94) and slower cognitive decline (β = 0.095; p = 0.0004). None of the PPARGC1α SNPs were marginally associated with PD risk. We estimate statistical interactions between MB/PQ and PPARGC1α rs6821591 (interaction p = 0.009) and rs8192678 (interaction p = 0.05), such that those with high exposure and the variant allele were at an increased risk of PD (OR ≥ 1.30, p ≤ 0.05). PPARGC1α rs6821591 was also associated with faster motor symptom progression as measured with the UPDRS-III (β = 0.234; p = 0.001). CONCLUSION Our study provides support for the involvement of both NFE2L2 and PPARGC1α in PD susceptibility and progression, marginally and through pathways involving MB/PQ exposure.
Collapse
Affiliation(s)
- Kimberly C Paul
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Janet S Sinsheimer
- Department of Biostatistics, UCLA Fielding School of Public Health, Los Angeles, CA, USA; Departments of Human Genetics and Biomathematics, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Myles Cockburn
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, CA, USA
| | - Jeff M Bronstein
- Department of Neurology, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Yvette Bordelon
- Department of Neurology, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Beate Ritz
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA, USA; Department of Neurology, David Geffen School of Medicine, Los Angeles, CA, USA.
| |
Collapse
|
25
|
Zhang X, Ji R, Sun H, Peng J, Ma X, Wang C, Fu Y, Bao L, Jin Y. Scutellarin ameliorates nonalcoholic fatty liver disease through the PPARγ/PGC-1α-Nrf2 pathway. Free Radic Res 2018; 52:198-211. [PMID: 29400110 DOI: 10.1080/10715762.2017.1422602] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/26/2017] [Accepted: 12/26/2017] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterised by excessive accumulation of hepatic lipids and oxidative injury of hepatocytes. Scutellarin is a flavonoid glycoside having antioxidative stress activity. Our current study aims to investigate the molecular mechanism of scutellarin ameliorating NAFLD. Scutellarin treatment was applied to male C57BL/6 mice maintained on a high-fat diet (HFD) and HepG2 cells challenged with oleic acid. The antioxidation biochemical indicators and lipid levels in the liver and cells were detected by kits. Liver pathology was observed by light microscope, Oil Red O staining, and transmission electron microscope (TEM). In addition, quantitative real-time polymerase chain reactions (qRT-PCR) and western blot assays were employed to detect the mRNA and protein levels of various antioxidative-related genes in the presence or absence of peroxisome proliferator-activated receptor gamma (PPARγ); inhibitor GW9662. Our results showed that scutellarin could significantly reduce blood lipid levels and enhance antioxidative capacities in both the models. In addition, scutellarin treatment conspicuously activated PPARγ, peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α), nuclear factor erythroid-2-related factor (Nrf2), haem oxygenase-1 (HO-1), glutathione S-transferase (GST), and NAD(P)H quinone dehydrogenase one (NQO1), while it significantly inhibited nuclear factor kappa B (NF-κB), Kelch-like ECH-associated protein 1 (Keap1) at both the mRNA and protein levels. However, after interfered by GW9662, scutellarin effect was significantly decreased. The experimental data demonstrated that scutellarin showed strong hypolipidaemic, antioxidative, and liver protective activity which could be attributed to its regulating activity in the PPARγ/PGC-1α-Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Xiaoxue Zhang
- a College of Pharmacy , Dalian Medical University , Dalian , China
| | - Renpeng Ji
- a College of Pharmacy , Dalian Medical University , Dalian , China
| | - Huijun Sun
- a College of Pharmacy , Dalian Medical University , Dalian , China
| | - Jinyong Peng
- a College of Pharmacy , Dalian Medical University , Dalian , China
| | - Xiaodong Ma
- a College of Pharmacy , Dalian Medical University , Dalian , China
| | - ChangYuan Wang
- a College of Pharmacy , Dalian Medical University , Dalian , China
| | - Yufeng Fu
- a College of Pharmacy , Dalian Medical University , Dalian , China
| | - Liuchi Bao
- a College of Pharmacy , Dalian Medical University , Dalian , China
| | - Yue Jin
- a College of Pharmacy , Dalian Medical University , Dalian , China
| |
Collapse
|
26
|
Mandel M, de Uña-Álvarez J, Simon DK, Betensky RA. Inverse probability weighted Cox regression for doubly truncated data. Biometrics 2017; 74:481-487. [PMID: 28886206 DOI: 10.1111/biom.12771] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 07/01/2017] [Accepted: 08/01/2017] [Indexed: 11/28/2022]
Abstract
Doubly truncated data arise when event times are observed only if they fall within subject-specific, possibly random, intervals. While non-parametric methods for survivor function estimation using doubly truncated data have been intensively studied, only a few methods for fitting regression models have been suggested, and only for a limited number of covariates. In this article, we present a method to fit the Cox regression model to doubly truncated data with multiple discrete and continuous covariates, and describe how to implement it using existing software. The approach is used to study the association between candidate single nucleotide polymorphisms and age of onset of Parkinson's disease.
Collapse
Affiliation(s)
- Micha Mandel
- Department of Statistics, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jacobo de Uña-Álvarez
- Department of Statistics and OR and Center for Biomedical Research (CINBIO), University of Vigo, Vigo, Spain
| | - David K Simon
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, U.S.A
| | - Rebecca A Betensky
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, U.S.A
| |
Collapse
|
27
|
Ay M, Luo J, Langley M, Jin H, Anantharam V, Kanthasamy A, Kanthasamy AG. Molecular mechanisms underlying protective effects of quercetin against mitochondrial dysfunction and progressive dopaminergic neurodegeneration in cell culture and MitoPark transgenic mouse models of Parkinson's Disease. J Neurochem 2017; 141:766-782. [PMID: 28376279 PMCID: PMC5643047 DOI: 10.1111/jnc.14033] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 03/24/2017] [Accepted: 03/28/2017] [Indexed: 12/22/2022]
Abstract
Quercetin, one of the major flavonoids in plants, has been recently reported to have neuroprotective effects against neurodegenerative processes. However, since the molecular signaling mechanisms governing these effects are not well clarified, we evaluated quercetin's effect on the neuroprotective signaling events in dopaminergic neuronal models and further tested its efficacy in the MitoPark transgenic mouse model of Parkinson's disease (PD). Western blot analysis revealed that quercetin significantly induced the activation of two major cell survival kinases, protein kinase D1 (PKD1) and Akt in MN9D dopaminergic neuronal cells. Furthermore, pharmacological inhibition or siRNA knockdown of PKD1 blocked the activation of Akt, suggesting that PKD1 acts as an upstream regulator of Akt in quercetin-mediated neuroprotective signaling. Quercetin also enhanced cAMP response-element binding protein phosphorylation and expression of the cAMP response-element binding protein target gene brain-derived neurotrophic factor. Results from qRT-PCR, Western blot analysis, mtDNA content analysis, and MitoTracker assay experiments revealed that quercetin augmented mitochondrial biogenesis. Quercetin also increased mitochondrial bioenergetics capacity and protected MN9D cells against 6-hydroxydopamine-induced neurotoxicity. To further evaluate the neuroprotective efficacy of quercetin against the mitochondrial dysfunction underlying PD, we used the progressive dopaminergic neurodegenerative MitoPark transgenic mouse model of PD. Oral administration of quercetin significantly reversed behavioral deficits, striatal dopamine depletion, and TH neuronal cell loss in MitoPark mice. Together, our findings demonstrate that quercetin activates the PKD1-Akt cell survival signaling axis and suggest that further exploration of quercetin as a promising neuroprotective agent for treating PD may offer clinical benefits.
Collapse
Affiliation(s)
- Muhammet Ay
- Parkinson’s Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, Iowa 50011, USA
| | - Jie Luo
- Parkinson’s Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, Iowa 50011, USA
| | - Monica Langley
- Parkinson’s Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, Iowa 50011, USA
| | - Huajun Jin
- Parkinson’s Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, Iowa 50011, USA
| | - Vellareddy Anantharam
- Parkinson’s Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, Iowa 50011, USA
| | - Arthi Kanthasamy
- Parkinson’s Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, Iowa 50011, USA
| | - Anumantha G. Kanthasamy
- Parkinson’s Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, Iowa 50011, USA
| |
Collapse
|
28
|
Haslam IS, Jadkauskaite L, Szabó IL, Staege S, Hesebeck-Brinckmann J, Jenkins G, Bhogal RK, Lim FL, Farjo N, Farjo B, Bíró T, Schäfer M, Paus R. Oxidative Damage Control in a Human (Mini-) Organ: Nrf2 Activation Protects against Oxidative Stress-Induced Hair Growth Inhibition. J Invest Dermatol 2017; 137:295-304. [DOI: 10.1016/j.jid.2016.08.035] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 08/22/2016] [Accepted: 08/23/2016] [Indexed: 01/08/2023]
|
29
|
Simon DK, Simuni T, Elm J, Clark-Matott J, Graebner AK, Baker L, Dunlop SR, Emborg M, Kamp C, Morgan JC, Ross GW, Sharma S, Ravina B. Peripheral Biomarkers of Parkinson's Disease Progression and Pioglitazone Effects. JOURNAL OF PARKINSONS DISEASE 2016; 5:731-6. [PMID: 26444095 PMCID: PMC5061495 DOI: 10.3233/jpd-150666] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Pioglitazone, an oral hypoglycemic agent, recently failed to show promise as a disease-modifying agent in a 44-week phase 2 placebo-controlled study in 210 Parkinson's disease (PD) subjects. We analyzed peripheral biomarkers, including leukocyte PGC-1α and target gene expression, plasma interleukin 6 (IL-6) as a marker of inflammation, and urine 8-hydroxydeoxyguanosine (8OHdG) as a marker of oxidative DNA damage. Baseline or changes from baseline in biomarker levels were not associated with the rate of progression of PD. Pioglitazone did not significantly alter biomarker levels. Other agents that more effectively target these mechanisms remain of potential interest as disease modifying therapies in PD.
Collapse
Affiliation(s)
- David K Simon
- Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Tanya Simuni
- Neurology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Jordan Elm
- Biostatistics, Medical University of South Carolina, Charleston, SC, USA
| | - Joanne Clark-Matott
- Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Allison K Graebner
- Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Liana Baker
- Clinical Trials Coordination Center, University of Rochester, Medical Center, Rochester, NY, USA
| | | | - Marina Emborg
- Wisconsin National Primate Research Center and Department of Medical Physics, University of Wisconsin, Madison, WI, USA
| | - Cornelia Kamp
- Clinical Materials Services Unit, University of Rochester, Medical Center, Rochester, NY, USA
| | - John C Morgan
- Neurology, Medical College of Georgia, Augusta, GA, USA
| | - G Webster Ross
- Neurology, Veterans Affairs Pacific Islands Health Care System, Honolulu, HI, USA
| | - Saloni Sharma
- Clinical Trials Coordination Center, University of Rochester, Medical Center, Rochester, NY, USA
| | | | | |
Collapse
|
30
|
Choong CJ, Mochizuki H. Gene therapy targeting mitochondrial pathway in Parkinson's disease. J Neural Transm (Vienna) 2016; 124:193-207. [PMID: 27638713 DOI: 10.1007/s00702-016-1616-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 08/31/2016] [Indexed: 01/11/2023]
Abstract
Parkinson's disease (PD) presents a relative selective localization of pathology to substantia nigra and well-defined motor symptoms caused by dopaminergic degeneration that makes it an ideal target for gene therapy. Parallel progress in viral vector systems enables the delivery of therapeutic genes directly into brain with reasonable safety along with sustained transgene expression. To date, gene therapy for PD that has reached clinical trial evaluation is mainly based on symptomatic approach that involves enzyme replacement strategy and restorative approach that depends on the addition of neurotrophic factors. Mitochondrial dysregulation, such as reduced complex I activity, increased mitochondria-derived reactive oxygen species (ROS) production, ROS-mediated mitochondrial DNA damage, bioenergetic failure, and perturbation of mitochondrial dynamics and mitophagy, has long been implicated in the pathogenesis of PD. Many of mutated genes linked to familial forms of PD affect these mitochondrial features. In this review, we discuss the recent progress that has been made in preclinical development of gene therapy targeting the mitochondrial pathway as disease modifying approach for PD. This review focuses on the potential therapeutic efficacy of candidate genes, including Parkin, PINK1, alpha synuclein, PGC-1 alpha, and anti-apoptotic molecules.
Collapse
Affiliation(s)
- Chi-Jing Choong
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan
| | - Hideki Mochizuki
- Department of Neurology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
31
|
Zhang T, Zhang Q, Guo J, Yuan H, Peng H, Cui L, Yin J, Zhang L, Zhao J, Li J, White A, Carmichael PL, Westmoreland C, Peng S. Non-cytotoxic concentrations of acetaminophen induced mitochondrial biogenesis and antioxidant response in HepG2 cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 46:71-79. [PMID: 27438896 DOI: 10.1016/j.etap.2016.06.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 06/27/2016] [Accepted: 06/30/2016] [Indexed: 06/06/2023]
Abstract
Mitochondrial dysfunction has been implicated in acute, severe liver injury caused by overdose of acetaminophen (APAP). However, whether mitochondrial biogenesis is involved is unclear. Here we demonstrated that mitochondrial biogenesis, as indicated by the amounts of mitochondrial DNA and proteins, increased significantly in HepG2 cells exposed to low, non-cytotoxic concentrations of APAP. This heightened response was accompanied by upregulated expression of PGC-1α, NRF-1 and TFAM, which are key transcriptional regulators of mitochondrial biogenesis. Additionally, antioxidants including glutathione, MnSOD, HO-1, NQO1, and Nrf2 were also significantly upregulated. In contrast, for HepG2 cells exposed to high, cytotoxic concentration of APAP, mitochondrial biogenesis was inhibited and the expression of its regulatory proteins and antioxidants were concentration-dependently downregulated. In summary, our study indicated that mitochondrial biogenesis, along with antioxidant induction, may be an important cellular adaptive mechanism counteracting APAP-induced toxicity and overwhelming this cytoprotective capacity could result in liver injury.
Collapse
Affiliation(s)
- Tingfen Zhang
- Evaluation and Research Centre for Toxicology, Institute of Disease Control and Prevention, The Academy of Military Medical Sciences, Beijing, PR China
| | - Qiang Zhang
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Jiabin Guo
- Evaluation and Research Centre for Toxicology, Institute of Disease Control and Prevention, The Academy of Military Medical Sciences, Beijing, PR China
| | - Haitao Yuan
- Evaluation and Research Centre for Toxicology, Institute of Disease Control and Prevention, The Academy of Military Medical Sciences, Beijing, PR China
| | - Hui Peng
- Evaluation and Research Centre for Toxicology, Institute of Disease Control and Prevention, The Academy of Military Medical Sciences, Beijing, PR China
| | - Lan Cui
- Evaluation and Research Centre for Toxicology, Institute of Disease Control and Prevention, The Academy of Military Medical Sciences, Beijing, PR China
| | - Jian Yin
- Evaluation and Research Centre for Toxicology, Institute of Disease Control and Prevention, The Academy of Military Medical Sciences, Beijing, PR China
| | - Li Zhang
- Evaluation and Research Centre for Toxicology, Institute of Disease Control and Prevention, The Academy of Military Medical Sciences, Beijing, PR China
| | - Jun Zhao
- Evaluation and Research Centre for Toxicology, Institute of Disease Control and Prevention, The Academy of Military Medical Sciences, Beijing, PR China
| | - Jin Li
- Unilever Safety and Environmental Assurance Center, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Andrew White
- Unilever Safety and Environmental Assurance Center, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Paul L Carmichael
- Unilever Safety and Environmental Assurance Center, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Carl Westmoreland
- Unilever Safety and Environmental Assurance Center, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, UK
| | - Shuangqing Peng
- Evaluation and Research Centre for Toxicology, Institute of Disease Control and Prevention, The Academy of Military Medical Sciences, Beijing, PR China.
| |
Collapse
|
32
|
Erlich AT, Tryon LD, Crilly MJ, Memme JM, Moosavi ZSM, Oliveira AN, Beyfuss K, Hood DA. Function of specialized regulatory proteins and signaling pathways in exercise-induced muscle mitochondrial biogenesis. Integr Med Res 2016; 5:187-197. [PMID: 28462117 PMCID: PMC5390460 DOI: 10.1016/j.imr.2016.05.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 05/04/2016] [Indexed: 12/19/2022] Open
Abstract
Skeletal muscle mitochondrial content and function are regulated by a number of specialized molecular pathways that remain to be fully defined. Although a number of proteins have been identified to be important for the maintenance of mitochondria in quiescent muscle, the requirement for these appears to decrease with the activation of multiple overlapping signaling events that are triggered by exercise. This makes exercise a valuable therapeutic tool for the treatment of mitochondrially based metabolic disorders. In this review, we summarize some of the traditional and more recently appreciated pathways that are involved in mitochondrial biogenesis in muscle, particularly during exercise.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - David A. Hood
- Corresponding author. Muscle Health Research Centre, School of Kinesiology and Health Science York University, Toronto, Ontario M3J1P3, Canada.
| |
Collapse
|
33
|
Osborne NN, Núñez-Álvarez C, Joglar B, Del Olmo-Aguado S. Glaucoma: Focus on mitochondria in relation to pathogenesis and neuroprotection. Eur J Pharmacol 2016; 787:127-33. [PMID: 27090928 DOI: 10.1016/j.ejphar.2016.04.032] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 03/25/2016] [Accepted: 04/14/2016] [Indexed: 12/23/2022]
Abstract
Primary open-angle glaucoma (POAG) is a common form of glaucoma in which retinal ganglion cells (RGCs) die at varying intervals. Primary open-angle glaucoma is often associated with an increased intraocular pressure (IOP), which when reduced, can slow down the progression of the disease. However, it is essential to develop better modes of treatments for glaucoma patients. In this overview, we discuss the hypothesis that RGC mitochondria are affected during the initiation of POAG, by becoming gradually weakened, but at different rates because of their specific receptor profiles. With this in mind, we argue that neuroprotection in the context of glaucoma should focus on preserving RGC mitochondrial function and suggest a number of ways by which this can theoretically be achieved. Since POAG is a chronic disease, any neuroprotective treatment strategy must be tolerated over many years. Theoretically, topically applied substances should have the fewest side effects, but it is questionable whether sufficient compounds can reach RGC mitochondria to be effective. Therefore, other delivery procedures that might result in greater concentrations of neuroprotectants reaching RGC mitochondria are being developed. Red-light therapy represents another therapeutic alternative for enhancing RGC mitochondrial functions and has the advantage of being both non-toxic and non-invasive.
Collapse
Affiliation(s)
- Neville N Osborne
- Fundación de Investigación Oftalmológica, Avda. Doctores Fernández-Vega 34, E-33012 Oviedo, Asturias, Spain.
| | - Claudia Núñez-Álvarez
- Fundación de Investigación Oftalmológica, Avda. Doctores Fernández-Vega 34, E-33012 Oviedo, Asturias, Spain
| | - Belen Joglar
- Fundación de Investigación Oftalmológica, Avda. Doctores Fernández-Vega 34, E-33012 Oviedo, Asturias, Spain
| | - Susana Del Olmo-Aguado
- Fundación de Investigación Oftalmológica, Avda. Doctores Fernández-Vega 34, E-33012 Oviedo, Asturias, Spain
| |
Collapse
|
34
|
Patil J, Matte A, Nissbrandt H, Mallard C, Sandberg M. Sustained Effects of Neonatal Systemic Lipopolysaccharide on IL-1β and Nrf2 in Adult Rat Substantia Nigra Are Partly Normalized by a Spirulina-Enriched Diet. Neuroimmunomodulation 2016; 23:250-259. [PMID: 27931028 PMCID: PMC5266664 DOI: 10.1159/000452714] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 10/19/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND/AIM Neonatal infection can sensitize the adult substantia nigra (SN) to secondary insults, causing a decrease in antioxidant capacity which may lead to Parkinson's disease in adults. We studied the prolonged effect of systemic infection by (i.p.) administration of lipopolysaccharide (LPS) on interleukin (IL)-1β, the antioxidant regulator nuclear factor-erythroid 2-related factor 2 (Nrf2), and the peroxisome proliferator-activated receptor γ coactivator (PGC)-1α in rat SN. METHOD AND RESULTS Five-day-old rat pups were treated with LPS (i.p. 2 mg/kg). After 65 days, the mRNA level of IL-1β was significantly increased, in parallel with a decrease in that of the rate-limiting enzyme in glutathione synthesis, the γ-glutamylcysteine ligase catalytic subunit (γGCLc), Nrf2, and brain-derived neurotrophic factor (BDNF). Protein levels of γGCLc and Nrf2 were decreased while IL-1β protein was significantly increased. These LPS-induced long-term changes correlated with a decrease in phosphorylated (active) AKT (pAKT) and phosphorylated (inactive) GSK-3β (pGSK-3β). In another set of experiments, a 0.1% Spirulina-containing diet was given to lactating mothers 24 h before the LPS treatment of the pups. The Spirulina-supplemented diet decreased IL-1β protein expression in SN and elevated the mRNA level of γGCLc, Nrf2 protein, PGC-1α protein, and pAKT. CONCLUSION Early-life infection can negatively affect Nrf2, pAKT, and pGSK-3β for a long time in SN. A diet enriched with antioxidant and anti-inflammatory phytochemicals can partly restore some, but not all, of the effects on the antioxidant defense, possibly via normalizing effects on pAKT.
Collapse
Affiliation(s)
- Jaspal Patil
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Sweden
| | - Ashok Matte
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Sweden
| | - Hans Nissbrandt
- Department of Pharmacology, Institute of Neuroscience and Physiology, University of Gothenburg, Sweden
| | - Carina Mallard
- Department of Physiology, Institute of Neuroscience and Physiology, University of Gothenburg, Sweden
| | - Mats Sandberg
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Sweden
| |
Collapse
|
35
|
Xu P, Zhang Y, Wang W, Yuan Q, Liu Z, Rasoul LM, Wu Q, Liu M, Ye X, Li D, Ren G. Long-Term Administration of Fibroblast Growth Factor 21 Prevents Chemically-Induced Hepatocarcinogenesis in Mice. Dig Dis Sci 2015; 60:3032-43. [PMID: 26003555 DOI: 10.1007/s10620-015-3711-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 05/07/2015] [Indexed: 12/31/2022]
Abstract
PURPOSE In this study, we explored whether treatment with FGF-21 could prevent diethylnitrosamine (DEN) induced hepatocarcinogenesis in mice. METHODS & RESULTS Hepatoma was induced by injection of DEN every three days for 18 weeks. For the prophylactic experiment, mice were firstly injected with FGF-21 for 2 weeks, then FGF-21 was administered to the mice once daily in association with DEN injection till the end of the experiment. The hepatoma incidence of mice treated with FGF-21 was 13.3%, while the incidence of mice treated with saline was 61.5%. To understand the mechanisms, we compared the expression of βklotho (KLB) and oxidative stress level in the livers between the mice treated with FGF-21 and saline. We found that FGF-21 could suppress DEN-induced oxidative stress and up-regulate the expression of KLB in the livers. To confirm these results, we compared the expression of KLB in L02 cells stimulated with or without FGF-21. Besides, we established DEN-induced oxidative stress cell model to affirm the relationship between FGF-21 and DEN-induced oxidative stress in vitro. Results showed that FGF-21 increased the expression of KLB and diminished the DEN-induced oxidative stress in vitro in a dose dependent manner. CONCLUSION Systemic administration of FGF-21 can prevent DEN-induced hepatocarcinogenesis via suppressing oxidative stress and increasing the expression of KLB.
Collapse
Affiliation(s)
- Pengfei Xu
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, 59 Mucai Street, Harbin, 150030, People's Republic of China
| | - Yingjie Zhang
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, 59 Mucai Street, Harbin, 150030, People's Republic of China
| | - Wenfei Wang
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, 59 Mucai Street, Harbin, 150030, People's Republic of China
| | - Qingyan Yuan
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, 59 Mucai Street, Harbin, 150030, People's Republic of China
| | - Zhihang Liu
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, 59 Mucai Street, Harbin, 150030, People's Republic of China
| | - Lubna Muhi Rasoul
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, 59 Mucai Street, Harbin, 150030, People's Republic of China.,College of Science, University of Baghdad, Baghdad, Iraq
| | - Qiang Wu
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, 59 Mucai Street, Harbin, 150030, People's Republic of China
| | - Mingyao Liu
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, 59 Mucai Street, Harbin, 150030, People's Republic of China
| | - Xianlong Ye
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, 59 Mucai Street, Harbin, 150030, People's Republic of China
| | - Deshan Li
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, 59 Mucai Street, Harbin, 150030, People's Republic of China. .,Key Laboratory of Agricultural Biological Functional Gene, Northeast Agricultural University, Harbin, People's Republic of China.
| | - Guiping Ren
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, 59 Mucai Street, Harbin, 150030, People's Republic of China.
| |
Collapse
|
36
|
Huang Y, Li W, Su ZY, Kong ANT. The complexity of the Nrf2 pathway: beyond the antioxidant response. J Nutr Biochem 2015; 26:1401-13. [PMID: 26419687 DOI: 10.1016/j.jnutbio.2015.08.001] [Citation(s) in RCA: 317] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 08/03/2015] [Indexed: 12/11/2022]
Abstract
The NF-E2-related factor 2 (Nrf2)-mediated signalling pathway provides living organisms an efficient and pivotal line of defensive to counteract environmental insults and endogenous stressors. Nrf2 coordinates the basal and inducible expression of antioxidant and Phase II detoxification enzymes to adapt to different stress conditions. The stability and cellular distribution of Nrf2 is tightly controlled by its inhibitory binding protein Kelch-like ECH-associated protein 1. Nrf2 signalling is also regulated by posttranslational, transcriptional, translational and epigenetic mechanisms, as well as by other protein partners, including p62, p21 and IQ motif-containing GTPase activating protein 1. Many studies have demonstrated that Nrf2 is a promising target for preventing carcinogenesis and other chronic diseases, including cardiovascular diseases, neurodegenerative diseases and pulmonary injury. However, constitutive activation of Nrf2 in advanced cancer cells may confer drug resistance. Here, we review the molecular mechanisms of Nrf2 signalling, the diverse classes of Nrf2 activators, including bioactive nutrients and other chemicals, and the cellular functions and disease relevance of Nrf2 and discuss the dual role of Nrf2 in different contexts.
Collapse
Affiliation(s)
- Ying Huang
- Department of Pharmaceutics, Earnest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Wenji Li
- Department of Pharmaceutics, Earnest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Zheng-yuan Su
- Department of Pharmaceutics, Earnest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Ah-Ng Tony Kong
- Department of Pharmaceutics, Earnest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA.
| |
Collapse
|
37
|
Ek CJ, D'Angelo B, Lehner C, Nathanielsz P, Li C, Mallard C. Expression of tight junction proteins and transporters for xenobiotic metabolism at the blood-CSF barrier during development in the nonhuman primate (P. hamadryas). Reprod Toxicol 2015; 56:32-44. [PMID: 26092209 DOI: 10.1016/j.reprotox.2015.06.047] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Revised: 06/04/2015] [Accepted: 06/05/2015] [Indexed: 10/23/2022]
Abstract
The choroid plexus (CP) is rich in barrier mechanisms including transporters and enzymes which can influence drug disposition between blood and brain. We have limited knowledge of their state in fetus. We have studied barrier mechanisms along with metabolism and transporters influencing xenobiotics, using RNAseq and protein analysis, in the CP during the second-half of gestation in a nonhuman primate (Papio hamadryas). There were no differences in the expression of the tight-junctions at the CP suggesting a well-formed fetal blood-CSF barrier during this period of gestation. Further, the fetal CP express many enzymes for phase I-III metabolisms as well as transporters suggesting that it can greatly influence drug disposition and has a significant machinery to deactivate reactive molecules with only minor gestational changes. In summary, the study suggests that from, at least, midgestation, the CP in the nonhuman primate is restrictive and express most known genes associated with barrier function and transport.
Collapse
Affiliation(s)
- C Joakim Ek
- Institute for Neuroscience and Physiology, Department of Physiology, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden.
| | - Barbara D'Angelo
- Institute for Neuroscience and Physiology, Department of Physiology, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Christine Lehner
- Institute of Tendon and Bone Regeneration, Paracelsus Medical University, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Salzburg, Austria; Department of Traumatology and Sports Injuries, Paracelsus Medical University Salzburg, Austria; Austrian Cluster for Tissue Regeneration Vienna, Austria
| | - Peter Nathanielsz
- Wyoming Pregnancy and Life Course Health Center, University of Wyoming, Laramie, WY 82071, United States
| | - Cun Li
- Department of Traumatology and Sports Injuries, Paracelsus Medical University Salzburg, Austria
| | - Carina Mallard
- Institute for Neuroscience and Physiology, Department of Physiology, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| |
Collapse
|
38
|
Oxidative Stress during the Progression of β-Amyloid Pathology in the Neocortex of the Tg2576 Mouse Model of Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:967203. [PMID: 25973140 PMCID: PMC4418010 DOI: 10.1155/2015/967203] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 03/26/2015] [Accepted: 03/27/2015] [Indexed: 12/05/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, characterized by progressive neurodegeneration. Pathogenetic mechanisms, triggered by β-amyloid (Aβ) accumulation, include oxidative stress, derived from energy homeostasis deregulation and involving mitochondria and peroxisomes. We here addressed the oxidative stress status and the elicited cellular response at the onset and during the progression of Aβ pathology, studying the neocortex of Tg2576 model of AD. Age-dependent changes of oxidative damage markers, antioxidant enzymes, and related transcription factors were analysed in relation to the distribution of Aβ peptide and oligomers, by a combined molecular/morphological approach. Nucleic acid oxidative damage, accompanied by defective antioxidant defences, and decreased PGC1α expression are already detected in 3-month-old Tg2576 neurons. Conversely, PPARα is increased in these cells, with its cytoplasmic localization suggesting nongenomic, anti-inflammatory actions. At 6 months, when intracellular Aβ accumulates, PMP70 is downregulated, indicating impairment of fatty acids peroxisomal translocation and their consequent harmful accumulation. In 9-month-old Tg2576 neocortex, Aβ oligomers and acrolein deposition correlate with GFAP, GPX1, and PMP70 increases, supporting a compensatory response, involving astroglial peroxisomes. At severe pathological stages, when senile plaques disrupt cortical cytoarchitecture, antioxidant capacity is gradually lost. Overall, our data suggest early therapeutic intervention in AD, also targeting peroxisomes.
Collapse
|
39
|
Torrent R, De Angelis Rigotti F, Dell'Era P, Memo M, Raya A, Consiglio A. Using iPS Cells toward the Understanding of Parkinson's Disease. J Clin Med 2015; 4:548-66. [PMID: 26239346 PMCID: PMC4470155 DOI: 10.3390/jcm4040548] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 02/10/2015] [Accepted: 02/10/2015] [Indexed: 12/24/2022] Open
Abstract
Cellular reprogramming of somatic cells to human pluripotent stem cells (iPSC) represents an efficient tool for in vitro modeling of human brain diseases and provides an innovative opportunity in the identification of new therapeutic drugs. Patient-specific iPSC can be differentiated into disease-relevant cell types, including neurons, carrying the genetic background of the donor and enabling de novo generation of human models of genetically complex disorders. Parkinson’s disease (PD) is the second most common age-related progressive neurodegenerative disease, which is mainly characterized by nigrostriatal dopaminergic (DA) neuron degeneration and synaptic dysfunction. Recently, the generation of disease-specific iPSC from patients suffering from PD has unveiled a recapitulation of disease-related cell phenotypes, such as abnormal α-synuclein accumulation and alterations in autophagy machinery. The use of patient-specific iPSC has a remarkable potential to uncover novel insights of the disease pathogenesis, which in turn will open new avenues for clinical intervention. This review explores the current Parkinson’s disease iPSC-based models highlighting their role in the discovery of new drugs, as well as discussing the most challenging limitations iPSC-models face today.
Collapse
Affiliation(s)
- Roger Torrent
- Institute for Biomedicine of the University of Barcelona (IBUB), Barcelona Science Park, Barcelona 08028, Spain.
| | - Francesca De Angelis Rigotti
- Institute for Biomedicine of the University of Barcelona (IBUB), Barcelona Science Park, Barcelona 08028, Spain.
| | - Patrizia Dell'Era
- Department of Molecular and Translational Medicine, Fibroblast Reprogramming Unit, University of Brescia, Brescia 25123, Italy.
| | - Maurizio Memo
- Department of Molecular and Translational Medicine, Fibroblast Reprogramming Unit, University of Brescia, Brescia 25123, Italy.
| | - Angel Raya
- Control of Stem Cell Potency Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona 08028, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona 08010, Spain.
- Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain.
- Center of Regenerative Medicine in Barcelona, Dr. Aiguader 88, Barcelona 08003, Spain.
| | - Antonella Consiglio
- Institute for Biomedicine of the University of Barcelona (IBUB), Barcelona Science Park, Barcelona 08028, Spain.
- Department of Molecular and Translational Medicine, Fibroblast Reprogramming Unit, University of Brescia, Brescia 25123, Italy.
| |
Collapse
|
40
|
Xiong W, MacColl Garfinkel AE, Li Y, Benowitz LI, Cepko CL. NRF2 promotes neuronal survival in neurodegeneration and acute nerve damage. J Clin Invest 2015; 125:1433-45. [PMID: 25798616 DOI: 10.1172/jci79735] [Citation(s) in RCA: 208] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 02/05/2015] [Indexed: 12/30/2022] Open
Abstract
Oxidative stress contributes to the loss of neurons in many disease conditions as well as during normal aging; however, small-molecule agents that reduce oxidation have not been successful in preventing neurodegeneration. Moreover, even if an efficacious systemic reduction of reactive oxygen and/or nitrogen species (ROS/NOS) could be achieved, detrimental side effects are likely, as these molecules regulate normal physiological processes. A more effective and targeted approach might be to augment the endogenous antioxidant defense mechanism only in the cells that suffer from oxidation. Here, we created several adeno-associated virus (AAV) vectors to deliver genes that combat oxidation. These vectors encode the transcription factors NRF2 and/or PGC1a, which regulate hundreds of genes that combat oxidation and other forms of stress, or enzymes such as superoxide dismutase 2 (SOD2) and catalase, which directly detoxify ROS. We tested the effectiveness of this approach in 3 models of photoreceptor degeneration and in a nerve crush model. AAV-mediated delivery of NRF2 was more effective than SOD2 and catalase, while expression of PGC1a accelerated photoreceptor death. Since the NRF2-mediated neuroprotective effects extended to photoreceptors and retinal ganglion cells, which are 2 very different types of neurons, these results suggest that this targeted approach may be broadly applicable to many diseases in which cells suffer from oxidative damage.
Collapse
|
41
|
Zhao XR, Gonzales N, Aronowski J. Pleiotropic role of PPARγ in intracerebral hemorrhage: an intricate system involving Nrf2, RXR, and NF-κB. CNS Neurosci Ther 2014; 21:357-66. [PMID: 25430543 DOI: 10.1111/cns.12350] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 10/10/2014] [Accepted: 10/11/2014] [Indexed: 12/13/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a subtype of stroke involving formation of hematoma within brain parenchyma, which accounts for 8-15% of all strokes in Western societies and 20-30% among Asian populations, and has a 1-year mortality rate >50%. The high mortality and severe morbidity make ICH a major public health problem. Only a few evidence-based targeted treatments are used for ICH management, and interventions focus primarily on supportive care and comorbidity prevention. Even in patients who survive the ictus, extravasated blood (including plasma components) and subsequent intrahematoma hemolytic products trigger a series of adverse events within the brain parenchyma, leading to secondary brain injury, edema and severe neurological deficits or death. Although the hematoma in humans gradually resolves within months, full restoration of neurological function can be slow and often incomplete, leaving survivors with devastating neurological deficits. During past years, peroxisome proliferator-activated receptor gamma (PPARγ) transcription factor and its agonists received recognition as important players in regulating not only glucose and lipid metabolism (which underlies its therapeutic effect in type 2 diabetes mellitus), and more recently, as an instrumental pleiotropic regulator of antiinflammation, antioxidative regulation, and phagocyte-mediated cleanup processes. PPARγ agonists have emerged as potential therapeutic target for stroke. The use of PPARγ as a therapeutic target appears to have particularly strong compatibility toward pathogenic components of ICH. In addition to its direct genomic effect, PPARγ may interact with transcription factor, NF-κB, which may underlie many aspects of the antiinflammatory effect of PPARγ. Furthermore, PPARγ appears to regulate expression of Nrf2, another transcription factor and master regulator of detoxification and antioxidative regulation. Finally, the synergistic costimulation of PPARγ and retinoid X receptor, RXR, may play an additional role in the therapeutic modulation of PPARγ function. In this article, we outline the main components of the role of PPARγ in ICH pathogenesis.
Collapse
Affiliation(s)
- Xiu-Rong Zhao
- Department of Neurology, Stroke Research Center, University of Texas Medical School - Houston, Houston, TX, USA
| | | | | |
Collapse
|
42
|
Osborne NN, Álvarez CN, del Olmo Aguado S. Targeting mitochondrial dysfunction as in aging and glaucoma. Drug Discov Today 2014; 19:1613-22. [DOI: 10.1016/j.drudis.2014.05.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Revised: 04/16/2014] [Accepted: 05/20/2014] [Indexed: 12/21/2022]
|
43
|
Ye D, Wang Y, Li H, Jia W, Man K, Lo CM, Wang Y, Lam KSL, Xu A. Fibroblast growth factor 21 protects against acetaminophen-induced hepatotoxicity by potentiating peroxisome proliferator-activated receptor coactivator protein-1α-mediated antioxidant capacity in mice. Hepatology 2014; 60:977-89. [PMID: 24590984 DOI: 10.1002/hep.27060] [Citation(s) in RCA: 145] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 02/02/2014] [Indexed: 12/12/2022]
Abstract
UNLABELLED Acetaminophen (APAP) overdose is a leading cause of drug-induced hepatotoxicity and acute liver failure worldwide, but its pathophysiology remains incompletely understood. Fibroblast growth factor 21 (FGF21) is a hepatocyte-secreted hormone with pleiotropic effects on glucose and lipid metabolism. This study aimed to investigate the pathophysiological role of FGF21 in APAP-induced hepatotoxicity in mice. In response to APAP overdose, both hepatic expression and circulating levels of FGF21 in mice were dramatically increased as early as 3 hours, prior to elevations of the liver injury markers alanine aminotransferase (ALT) and aspartate aminotransferase (AST). APAP overdose-induced liver damage and mortality in FGF21 knockout (KO) mice were markedly aggravated, which was accompanied by increased oxidative stress and impaired antioxidant capacities as compared to wild-type (WT) littermates. By contrast, replenishment of recombinant FGF21 largely reversed APAP-induced hepatic oxidative stress and liver injury in FGF21 KO mice. Mechanistically, FGF21 induced hepatic expression of peroxisome proliferator-activated receptor coactivator protein-1α (PGC-1α), thereby increasing the nuclear abundance of nuclear factor erythroid 2-related factor 2 (Nrf2) and subsequent up-regulation of several antioxidant genes. The beneficial effects of recombinant FGF21 on up-regulation of Nrf2 and antioxidant genes and alleviation of APAP-induced oxidative stress and liver injury were largely abolished by adenovirus-mediated knockdown of hepatic PGC-1α expression, whereas overexpression of PGC-1α was sufficient to counteract the increased susceptibility of FGF21 KO mice to APAP-induced hepatotoxicity. CONCLUSION The marked elevation of FGF21 by APAP overdose may represent a compensatory mechanism to protect against the drug-induced hepatotoxicity, by enhancing PGC-1α/Nrf2-mediated antioxidant capacity in the liver.
Collapse
Affiliation(s)
- Dewei Ye
- State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong, China; Department of Medicine, University of Hong Kong, Hong Kong, China; Department of Pharmacology & Pharmacy, University of Hong Kong, Hong Kong, China
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Zhou J, Qu XD, Li ZY, Wei-Ji, Liu Q, Ma YH, He JJ. Salvianolic acid B attenuates toxin-induced neuronal damage via Nrf2-dependent glial cells-mediated protective activity in Parkinson's disease models. PLoS One 2014; 9:e101668. [PMID: 24991814 PMCID: PMC4081637 DOI: 10.1371/journal.pone.0101668] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 06/09/2014] [Indexed: 12/30/2022] Open
Abstract
Salvianolic acid B (SalB), a bioactive compound isolated from the plant-derived medicinal herb Danshen, has been shown to exert various anti-oxidative and anti-inflammatory activities in several neurological disorders. In this study, we sought to investigate the potential protective effects and associated molecular mechanisms of SalB in Parkinson’s disease (PD) models. To determine the neuroprotective effects of SalB in vitro, MPP+- or lipopolysaccharide (LPS)-induced neuronal injury was achieved using primary cultures with different compositions of neurons, microglia and astrocytes. Our results showed that SalB reduced both LPS- and MPP+-induced toxicity of dopamine neurons in a dose-dependent manner. Additionally, SalB treatment inhibited the release of microglial pro-inflammatory cytokines and resulted in an increase in the expression and release of glial cell line-derived neurotrophic factor (GDNF) from astrocytes. Western blot analysis illustrated that SalB increased the expression and nuclear translocation of nuclear factor (erythroid-derived 2)-like 2 (Nrf2). The knockdown of Nrf2 using specific small interfering RNA (siRNA) partially reversed the SalB-induced GDNF expression and anti-inflammatory activity. Moreover, SalB treatment significantly attenuated dopaminergic (DA) neuronal loss, inhibited neuroinflammation, increased GDNF expression and improved the neurological function in MPTP-treated mice. Collectively, these findings demonstrated that SalB protects DA neurons by an Nrf-2 -mediated dual action: reducing microglia activation-mediated neuroinflammation and inducing astrocyte activation-dependent GDNF expression. Importantly the present study also highlights critical roles of glial cells as targets for developing new strategies to alter the progression of neurodegenerative disorders.
Collapse
Affiliation(s)
- Jie Zhou
- Department of Neurosurgery, Lanzhou General Hospital, Lanzhou Command of PLA, Lanzhou, Gansu, China
| | - Xiao-Dong Qu
- Department of Neurosurgery, Lanzhou General Hospital, Lanzhou Command of PLA, Lanzhou, Gansu, China
| | - Zhi-Yun Li
- Department of Neurosurgery, Lanzhou General Hospital, Lanzhou Command of PLA, Lanzhou, Gansu, China
| | - Wei-Ji
- Department of Neurosurgery, Lanzhou General Hospital, Lanzhou Command of PLA, Lanzhou, Gansu, China
| | - Qi Liu
- Department of Neurosurgery, Lanzhou General Hospital, Lanzhou Command of PLA, Lanzhou, Gansu, China
| | - Yi-Hui Ma
- Department of Neurosurgery, Lanzhou General Hospital, Lanzhou Command of PLA, Lanzhou, Gansu, China
| | - Jiao-Jiang He
- Department of Neurosurgery, Lanzhou General Hospital, Lanzhou Command of PLA, Lanzhou, Gansu, China
| |
Collapse
|
45
|
Narasimhan M, Hong J, Atieno N, Muthusamy VR, Davidson CJ, Abu-Rmaileh N, Richardson RS, Gomes AV, Hoidal JR, Rajasekaran NS. Nrf2 deficiency promotes apoptosis and impairs PAX7/MyoD expression in aging skeletal muscle cells. Free Radic Biol Med 2014; 71:402-414. [PMID: 24613379 PMCID: PMC4493911 DOI: 10.1016/j.freeradbiomed.2014.02.023] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Revised: 02/17/2014] [Accepted: 02/25/2014] [Indexed: 12/20/2022]
Abstract
Skeletal muscle redox homeostasis is transcriptionally regulated by nuclear erythroid-2-p45-related factor-2 (Nrf2). We recently demonstrated that age-associated stress impairs Nrf2-ARE (antioxidant-response element) transcriptional signaling. Here, we hypothesize that age-dependent decline or genetic ablation of Nrf2 leads to accelerated apoptosis and skeletal muscle degeneration. Under basal-physiological conditions, disruption of Nrf2 significantly downregulates antioxidants and causes oxidative stress. Surprisingly, Nrf2-null mice had enhanced antioxidant capacity identical to wild-type (WT) upon acute endurance exercise stress (AEES), suggesting activation of Nrf2-independent mechanisms (i.e., PGC1α) against oxidative stress. Analysis of prosurvival pathways in the basal state reveals decreased AKT levels, whereas p-p53, a repressor of AKT, was increased in Nrf2-null vs WT mice. Upon AEES, AKT and p-AKT levels were significantly (p < 0.001) increased (>10-fold) along with profound downregulation of p-p53 (p < 0.01) in Nrf2-null vs WT skeletal muscle, indicating the onset of prosurvival mechanisms to compensate for the loss of Nrf2 signaling. However, we found a decreased stem cell population (PAX7) and MyoD expression (differentiation) along with profound activation of ubiquitin and apoptotic pathways in Nrf2-null vs WT mice upon AEES, suggesting that compensatory prosurvival mechanisms failed to overcome the programmed cell death and degeneration in skeletal muscle. Further, the impaired regeneration was sustained in Nrf2-null vs WT mice after 1 week of post-AEES recovery. In an age-associated oxidative stress condition, ablation of Nrf2 results in induction of apoptosis and impaired muscle regeneration.
Collapse
Affiliation(s)
- Madhusudhanan Narasimhan
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Cardiac Aging and Redox Signaling Laboratory, Division of Cardiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Jennifer Hong
- Cardiac Aging and Redox Signaling Laboratory, Division of Cardiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Nancy Atieno
- Cardiac Aging and Redox Signaling Laboratory, Division of Cardiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Vasanthi R Muthusamy
- Cardiac Aging and Redox Signaling Laboratory, Division of Cardiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Christopher J Davidson
- Cardiac Aging and Redox Signaling Laboratory, Division of Cardiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Naser Abu-Rmaileh
- Cardiac Aging and Redox Signaling Laboratory, Division of Cardiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Russell S Richardson
- Division of Geriatrics, and Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA; Department of Exercise & Sports Sciences, College of Health, University of Utah, Salt Lake City, UT 84112, USA; Geriatric Research, Education, and Clinical Center, Salt Lake City Veteran's Medical Center
| | | | - John R Hoidal
- Division of Pulmonary Medicine, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Namakkal S Rajasekaran
- Cardiac Aging and Redox Signaling Laboratory, Division of Cardiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA; Department of Exercise & Sports Sciences, College of Health, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
46
|
Isogenic human iPSC Parkinson's model shows nitrosative stress-induced dysfunction in MEF2-PGC1α transcription. Cell 2013; 155:1351-64. [PMID: 24290359 DOI: 10.1016/j.cell.2013.11.009] [Citation(s) in RCA: 350] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 08/11/2013] [Accepted: 11/04/2013] [Indexed: 01/16/2023]
Abstract
Parkinson's disease (PD) is characterized by loss of A9 dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc). An association has been reported between PD and exposure to mitochondrial toxins, including environmental pesticides paraquat, maneb, and rotenone. Here, using a robust, patient-derived stem cell model of PD allowing comparison of A53T α-synuclein (α-syn) mutant cells and isogenic mutation-corrected controls, we identify mitochondrial toxin-induced perturbations in A53T α-syn A9 DA neurons (hNs). We report a pathway whereby basal and toxin-induced nitrosative/oxidative stress results in S-nitrosylation of transcription factor MEF2C in A53T hNs compared to corrected controls. This redox reaction inhibits the MEF2C-PGC1α transcriptional network, contributing to mitochondrial dysfunction and apoptotic cell death. Our data provide mechanistic insight into gene-environmental interaction (GxE) in the pathogenesis of PD. Furthermore, using small-molecule high-throughput screening, we identify the MEF2C-PGC1α pathway as a therapeutic target to combat PD.
Collapse
|
47
|
Neuroprotective Properties of a Novel Non-Thiazoledinedione Partial PPAR- γ Agonist against MPTP. PPAR Res 2013; 2013:582809. [PMID: 24223584 PMCID: PMC3808726 DOI: 10.1155/2013/582809] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Revised: 07/24/2013] [Accepted: 07/25/2013] [Indexed: 12/25/2022] Open
Abstract
Activation of the peroxisome proliferator activated receptor-gamma (PPAR)-γ is proposed as a neuroprotective strategy to treat neurodegenerative disorders. In this study, we examined if LSN862 (LSN), a novel non-thiazoledinedione partial PPAR-γ agonist, was neuroprotective in a mouse model of Parkinson's disease (PD) and assessed possible mechanisms of action. LSN (3, 10, or 30 mg/kg) or vehicle was orally administered daily to C57BL/6 and antioxidant response element-human placental alkaline phosphatase (ARE-hPAP) reporter mice 3 days prior to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP; 30 mg/kg, i.p. × 5 days) or PBS administration. LSN elicited a dose-dependent preservation of dopaminergic nigrostriatal innervation that was not associated with inhibition of MPTP metabolism or activation of Nrf2-ARE, although changes in NQO1 and SOD2 mRNA were observed. A significant dose-dependent downregulation in MAC-1 and GFAP positive cells was observed in MPTP + LSN-treated mice as well as significant downregulation of mRNA expression levels of these inflammatory markers. MPTP-induced increases in PPAR-γ and PGC1α expression were ameliorated by LSN dosing. Our results demonstrate that oral administration of LSN is neuroprotective against MPTP-induced neurodegeneration, and this effect is associated with downregulation of neuroinflammation, decreased oxidative stress, and modulation of PPAR-γ and PGC1α expression. These results suggest that LSN can be a candidate alternative non-thiazoledinedione partial PPAR-γ agonist for neuroprotective treatment of PD.
Collapse
|
48
|
Baldelli S, Aquilano K, Ciriolo MR. Punctum on two different transcription factors regulated by PGC-1α: Nuclear factor erythroid-derived 2-like 2 and nuclear respiratory factor 2. Biochim Biophys Acta Gen Subj 2013; 1830:4137-46. [DOI: 10.1016/j.bbagen.2013.04.006] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 03/21/2013] [Accepted: 04/02/2013] [Indexed: 12/30/2022]
|
49
|
Zolotukhin P, Kozlova Y, Dovzhik A, Kovalenko K, Kutsyn K, Aleksandrova A, Shkurat T. Oxidative status interactome map: towards novel approaches in experiment planning, data analysis, diagnostics and therapy. MOLECULAR BIOSYSTEMS 2013; 9:2085-96. [PMID: 23698602 DOI: 10.1039/c3mb70096h] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Experimental evidence suggests an immense variety of processes associated with and aimed at producing reactive oxygen and/or nitrogen species. Clinical studies implicate an enormous range of pathologies associated with reactive oxygen/nitrogen species metabolism deregulation, particularly oxidative stress. Recent advances in biochemistry, proteomics and molecular biology/biophysics of cells suggest oxidative stress to be an endpoint of complex dysregulation events of conjugated pathways consolidated under the term, proposed here, "oxidative status". The oxidative status concept, in order to allow for novel diagnostic and therapeutic approaches, requires elaboration of a new logic system comprehending all the features, versatility and complexity of cellular pro- and antioxidative components of different nature. We have developed a curated and regularly updated interactive interactome map of human cellular-level oxidative status allowing for systematization of the related most up-to-date experimental data. A total of more than 600 papers were selected for the initial creation of the map. The map comprises more than 300 individual factors with respective interactions, all subdivided hierarchically for logical analysis purposes. The pilot application of the interactome map suggested several points for further development of oxidative status-based technologies.
Collapse
Affiliation(s)
- Peter Zolotukhin
- Southern Federal University, Stachki av., 194/1, Rostov-on-Don, Russia.
| | | | | | | | | | | | | |
Collapse
|
50
|
Fanelli F, Sepe S, D’Amelio M, Bernardi C, Cristiano L, Cimini A, Cecconi F, Ceru' MP, Moreno S. Age-dependent roles of peroxisomes in the hippocampus of a transgenic mouse model of Alzheimer's disease. Mol Neurodegener 2013; 8:8. [PMID: 23374228 PMCID: PMC3599312 DOI: 10.1186/1750-1326-8-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 01/29/2013] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Alzheimer's Disease (AD) is a progressive neurodegenerative disease, especially affecting the hippocampus. Impairment of cognitive and memory functions is associated with amyloid β-peptide-induced oxidative stress and alterations in lipid metabolism. In this scenario, the dual role of peroxisomes in producing and removing ROS, and their function in fatty acids β-oxidation, may be critical. This work aims to investigating the possible involvement of peroxisomes in AD onset and progression, as studied in a transgenic mouse model, harboring the human Swedish familial AD mutation. We therefore characterized the peroxisomal population in the hippocampus, focusing on early, advanced, and late stages of the disease (3, 6, 9, 12, 18 months of age). Several peroxisome-related markers in transgenic and wild-type hippocampal formation were comparatively studied, by a combined molecular/immunohistochemical/ultrastructural approach. RESULTS Our results demonstrate early and significant peroxisomal modifications in AD mice, compared to wild-type. Indeed, the peroxisomal membrane protein of 70 kDa and acyl-CoA oxidase 1 are induced at 3 months, possibly reflecting the need for efficient fatty acid β-oxidation, as a compensatory response to mitochondrial dysfunction. The concomitant presence of oxidative damage markers and the altered expression of antioxidant enzymes argue for early oxidative stress in AD. During physiological and pathological brain aging, important changes in the expression of peroxisome-related proteins, also correlating with ongoing gliosis, occur in the hippocampus. These age- and genotype-based alterations, strongly dependent on the specific marker considered, indicate metabolic and/or numerical remodeling of peroxisomal population. CONCLUSIONS Overall, our data support functional and biogenetic relationships linking peroxisomes to mitochondria and suggest peroxisomal proteins as biomarkers/therapeutic targets in pre-symptomatic AD.
Collapse
Affiliation(s)
- Francesca Fanelli
- Department of Biology-LIME, University “Roma Tre”, viale Marconi 446, 00146, Rome, Italy
- University Campus Bio-Medico, via Alvaro del Portillo 21, 00128, Rome, Italy
| | - Sara Sepe
- Department of Biology-LIME, University “Roma Tre”, viale Marconi 446, 00146, Rome, Italy
| | - Marcello D’Amelio
- IRCCS S. Lucia Foundation, via del Fosso di Fiorano 65, 00143, Rome, Italy
- University Campus Bio-Medico, via Alvaro del Portillo 21, 00128, Rome, Italy
| | - Cinzia Bernardi
- Department of Radiological Sciences and Laboratory Medicine, UOC Pathological Anatomy, San Filippo Neri Hospital, via Martinotti 20, 00135, Rome, Italy
| | - Loredana Cristiano
- Department of Life, Health and Environmental Sciences, University of L’Aquila, piazzale Salvatore Tommasi 1, 67100, Coppito, (AQ), Italy
| | - AnnaMaria Cimini
- Department of Life, Health and Environmental Sciences, University of L’Aquila, piazzale Salvatore Tommasi 1, 67100, Coppito, (AQ), Italy
| | - Francesco Cecconi
- IRCCS S. Lucia Foundation, via del Fosso di Fiorano 65, 00143, Rome, Italy
- Department of Biology, University of Rome ‘Tor Vergata’, via della Ricerca Scientifica, 00133, Rome, Italy
| | - Maria Paola Ceru'
- Department of Life, Health and Environmental Sciences, University of L’Aquila, piazzale Salvatore Tommasi 1, 67100, Coppito, (AQ), Italy
| | - Sandra Moreno
- Department of Biology-LIME, University “Roma Tre”, viale Marconi 446, 00146, Rome, Italy
| |
Collapse
|