51
|
Liu Y, Chu JMT, Yan T, Zhang Y, Chen Y, Chang RCC, Wong GTC. Short-term resistance exercise inhibits neuroinflammation and attenuates neuropathological changes in 3xTg Alzheimer's disease mice. J Neuroinflammation 2020; 17:4. [PMID: 31900170 PMCID: PMC6942350 DOI: 10.1186/s12974-019-1653-7] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 11/20/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Both human and animal studies have shown beneficial effects of physical exercise on brain health but most tend to be based on aerobic rather than resistance type regimes. Resistance exercise has the advantage of improving both muscular and cardiovascular function, both of which can benefit the frail and the elderly. However, the neuroprotective effects of resistance training in cognitive impairment are not well characterized. METHODS We evaluated whether short-term resistant training could improve cognitive function and pathological changes in mice with pre-existing cognitive impairment. Nine-month-old 3xTg mouse underwent a resistance training protocol of climbing up a 1-m ladder with a progressively heavier weight loading. RESULTS Compared with sedentary counterparts, resistance training improved cognitive performance and reduced neuropathological and neuroinflammatory changes in the frontal cortex and hippocampus of mice. In line with these results, inhibition of pro-inflammatory intracellular pathways was also demonstrated. CONCLUSIONS Short-term resistance training improved cognitive function in 3xTg mice, and conferred beneficial effects on neuroinflammation, amyloid and tau pathology, as well as synaptic plasticity. Resistance training may represent an alternative exercise strategy for delaying disease progression in Alzheimer's disease.
Collapse
Affiliation(s)
- Yan Liu
- Department of Anaesthesiology, LKS Faculty of Medicine, The University of Hong Kong, Room K424, Queen Mary Hospital, Pokfulam, Hong Kong, SAR, China.,Laboratory of Neurodegenerative Diseases, LKS Faculty of MedicineSchool of Biomedical Sciences, The University of Hong Kong, Hong Kong, SAR, China
| | - John Man Tak Chu
- Department of Anaesthesiology, LKS Faculty of Medicine, The University of Hong Kong, Room K424, Queen Mary Hospital, Pokfulam, Hong Kong, SAR, China.,Laboratory of Neurodegenerative Diseases, LKS Faculty of MedicineSchool of Biomedical Sciences, The University of Hong Kong, Hong Kong, SAR, China
| | - Tim Yan
- Laboratory of Neurodegenerative Diseases, LKS Faculty of MedicineSchool of Biomedical Sciences, The University of Hong Kong, Hong Kong, SAR, China
| | - Yan Zhang
- Department of Anaesthesiology, LKS Faculty of Medicine, The University of Hong Kong, Room K424, Queen Mary Hospital, Pokfulam, Hong Kong, SAR, China.,Laboratory of Neurodegenerative Diseases, LKS Faculty of MedicineSchool of Biomedical Sciences, The University of Hong Kong, Hong Kong, SAR, China
| | - Ying Chen
- Department of Anaesthesiology, LKS Faculty of Medicine, The University of Hong Kong, Room K424, Queen Mary Hospital, Pokfulam, Hong Kong, SAR, China.,Laboratory of Neurodegenerative Diseases, LKS Faculty of MedicineSchool of Biomedical Sciences, The University of Hong Kong, Hong Kong, SAR, China
| | - Raymond Chuen Chung Chang
- Laboratory of Neurodegenerative Diseases, LKS Faculty of MedicineSchool of Biomedical Sciences, The University of Hong Kong, Hong Kong, SAR, China. .,State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, L4-49, Laboratory Block, Pokfulam, Hong Kong, SAR, China.
| | - Gordon Tin Chun Wong
- Department of Anaesthesiology, LKS Faculty of Medicine, The University of Hong Kong, Room K424, Queen Mary Hospital, Pokfulam, Hong Kong, SAR, China.
| |
Collapse
|
52
|
Zhang Y, Anoopkumar-Dukie S, Arora D, Davey AK. Review of the anti-inflammatory effect of SIRT1 and SIRT2 modulators on neurodegenerative diseases. Eur J Pharmacol 2020; 867:172847. [DOI: 10.1016/j.ejphar.2019.172847] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 12/03/2019] [Indexed: 12/22/2022]
|
53
|
Dang C, Han B, Li Q, Han R, Hao J. Up-regulation of PGC-1α in neurons protects against experimental autoimmune encephalomyelitis. FASEB J 2019; 33:14811-14824. [PMID: 31718280 DOI: 10.1096/fj.201901149rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Reactive oxygen species (ROS) generation and mitochondrial dysfunction are related to neuron loss in multiple sclerosis (MS). Although peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α) appears to play a key role in modulating levels of mitochondrial ROS, antioxidants, and uncoupling proteins (UCPs), and PGC-1α expression is reduced in the neocortex of patients with MS, it is unclear what its role is in neurons and in the manifestation of clinical symptoms of MS. Here, we show in wild-type (WT) experimental autoimmune encephalomyelitis (EAE) mice that PGC-1α is decreased 13 d after EAE induction followed by a steady decline up to 20 d. These changes were accompanied by parallel alterations in levels of superoxide dismutase 2, peroxiredoxin 3, thioredoxin 2, UCP4, and UCP5. In transgenic (TG) mice with neuron-specific overexpression of PGC-1α (PGC-1αf/fEno2-Cre), clinical symptoms after EAE induction were delayed and less severe than in WT mice. The degrees of apoptotic neuron loss and demyelination were also less severe in PGC-1α-TG mice. Overexpression of PGC-1α in neuronal neuroblastoma spinal cord 34 cells subjected to EAE inflammatory conditions showed similar results to those obtained in vivo. RNA sequencing analysis showed that apoptotic processes were significantly enriched in the top 10 significant gene ontology (GO) terms of differentially expressed genes, and the apoptotic pathway was significantly enriched in Kyoto Encyclopedia of Genes and Genomes pathway analysis. Our findings indicate that up-regulation of neuronal PGC-1α protected neurons from apoptosis in EAE. Manipulating PGC-1α levels in MS may help stave off this devastating disease.-Dang, C., Han, B., Li, Q., Han, R., Hao, J. Up-regulation of PGC-1α in neurons protects against experimental autoimmune encephalomyelitis.
Collapse
Affiliation(s)
- Chun Dang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Bin Han
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qian Li
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ranran Han
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Junwei Hao
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
54
|
Nashine S, Subramaniam SR, Chwa M, Nesburn A, Kuppermann BD, Federoff H, Kenney MC. PU-91 drug rescues human age-related macular degeneration RPE cells; implications for AMD therapeutics. Aging (Albany NY) 2019; 11:6691-6713. [PMID: 31477635 PMCID: PMC6756897 DOI: 10.18632/aging.102179] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 08/09/2019] [Indexed: 12/24/2022]
Abstract
Since mitochondrial dysfunction is implicated in the pathogenesis of AMD, this study is based on the premise that repurposing of mitochondria-stabilizing FDA-approved drugs such as PU-91, might rescue AMD RPE cells from AMD mitochondria-induced damage. The PU-91 drug upregulates PGC-1α which is a critical regulator of mitochondrial biogenesis. Herein, we tested the therapeutic potential of PU-91 drug and examined the additive effects of treatment with PU-91 and esterase inhibitors i.e., EI-12 and EI-78, using the in vitro transmitochondrial AMD cell model. This model was created by fusing platelets obtained from AMD patients with Rho0 i.e., mitochondria-deficient, ARPE-19 cell lines. The resulting AMD RPE cell lines have identical nuclei but differ in their mitochondrial DNA content, which is derived from individual AMD patients. Briefly, we report significant improvement in cell survival, mitochondrial health, and antioxidant potential in PU-91-treated AMD RPE cells compared to their untreated counterparts. In conclusion, this study identifies PU 91 as a therapeutic candidate drug for AMD and repurposing of PU-91 will be a smoother transition from lab bench to clinic since the pharmacological profiles of PU-91 have been examined already.
Collapse
Affiliation(s)
- Sonali Nashine
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA 92697, USA
| | | | - Marilyn Chwa
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA 92697, USA
| | - Anthony Nesburn
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA 92697, USA.,Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Baruch D Kuppermann
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA 92697, USA
| | - Howard Federoff
- Department of Neurology, University of California Irvine, Irvine, CA 92697, USA
| | - M Cristina Kenney
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA 92697, USA.,Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|
55
|
Xu WN, Yang RZ, Zheng HL, Yu W, Zheng XF, Li B, Jiang SD, Jiang LS. PGC-1α acts as an mediator of Sirtuin2 to protect annulus fibrosus from apoptosis induced by oxidative stress through restraining mitophagy. Int J Biol Macromol 2019; 136:1007-1017. [PMID: 31238070 DOI: 10.1016/j.ijbiomac.2019.06.163] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 06/21/2019] [Accepted: 06/21/2019] [Indexed: 12/22/2022]
Abstract
Apoptosis of annulus fibrosus (AF) is observed widely in intervertebral disc degeneration (IVDD) which causes weaken of tension in the annulus of intervertebral disc. Previous studies reported that apoptosis of AF is induced mainly by oxidative stress. SIRT2 is a major regulator of mitochondria to mediate ROS production. However, the mechanism of SIRT2 in IVDD remains unclear. Here, the expression of SIRT2 was detected in AF cells exposed to tert-Butyl hydroperoxide (TBHP) by western blotting. Autophagic flux and apoptosis were assessed by western blotting, flow cytometry and immunofluorescence respectively. Safranin O staining, HE, and immunohistochemical were used to assess the IVDD after 3, 6 and 9 months of surgical procedure in vivo. The expression of SIRT2 was decreased in AF cells treated with TBHP. Repression of mitophagy alleviated the apoptosis of AF cells caused by TBHP. Overexpression of PGC-1α prevented AF cells from apoptosis and mitophagy after applying Lenti-PGC-1α to transfect AF cells. These protections of PGC-1α were reduced by FCCP. Furthermore, the expression of PGC-1α was reduced and the level of mitophagy was increased in IVDD models. In conclusion, this study indicates that the regulation of PGC-1α expression provide a new theoretical basis for the mechanism of IVDD.
Collapse
Affiliation(s)
- Wen-Ning Xu
- Department of Clinic of Spine Center, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200082, China
| | - Run-Ze Yang
- Department of Clinic of Spine Center, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200082, China
| | - Huo-Liang Zheng
- Department of Clinic of Spine Center, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200082, China
| | - Wei Yu
- Department of Orthopaedics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Xin-Feng Zheng
- Department of Clinic of Spine Center, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200082, China
| | - Bo Li
- Department of Clinic of Spine Center, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200082, China
| | - Sheng-Dan Jiang
- Department of Clinic of Spine Center, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200082, China.
| | - Lei-Sheng Jiang
- Department of Clinic of Spine Center, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200082, China.
| |
Collapse
|
56
|
Dal Yontem F, Kim SH, Ding Z, Grimm E, Ekmekcioglu S, Akcakaya H. Mitochondrial dynamic alterations regulate melanoma cell progression. J Cell Biochem 2019; 120:2098-2108. [PMID: 30256441 DOI: 10.1002/jcb.27518] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 07/27/2018] [Indexed: 01/24/2023]
Abstract
Research on mitochondrial fusion and fission (mitochondrial dynamics) has gained much attention in recent years, as it is important for understanding many biological processes, including the maintenance of mitochondrial functions, apoptosis, and cancer. The rate of mitochondrial biosynthesis and degradation can affect various aspects of tumor progression. However, the role of mitochondrial dynamics in melanoma progression remains controversial and requires a mechanistic understanding to target the altered metabolism of cancer cells. Therefore, in our study, we disrupted mitochondrial fission with mdivi-1, the reported inhibitor of dynamin related protein 1 (Drp1), and knocked down Drp1 and Mfn2 to evaluate the effects of mitochondrial dynamic alterations on melanoma cell progression. Our confocal study results showed that mitochondrial fission was inhibited both in mdivi-1 and in Drp1 knockdown cells and, in parallel, mitochondrial fusion was induced. We also found that mitochondrial fission inhibition by mdivi-1 induced cell death in melanoma cells. However, silencing Drp1 and Mfn2 did not affect cell viability, but enhanced melanoma cell migration. We further show that dysregulated mitochondrial fusion by Mfn2 knockdowns suppressed the oxygen consumption rate of melanoma cells. Together, our findings suggest that mitochondrial dynamic alterations regulate melanoma cell migration and progression.
Collapse
Affiliation(s)
- Fulya Dal Yontem
- Department of Biophysics, Istanbul University, Istanbul Medical Faculty, Istanbul, Turkey.,Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sun-Hee Kim
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zhen Ding
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elizabeth Grimm
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Suhendan Ekmekcioglu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Handan Akcakaya
- Department of Biophysics, Istanbul University, Istanbul Medical Faculty, Istanbul, Turkey
| |
Collapse
|
57
|
Elfawy HA, Das B. Crosstalk between mitochondrial dysfunction, oxidative stress, and age related neurodegenerative disease: Etiologies and therapeutic strategies. Life Sci 2018; 218:165-184. [PMID: 30578866 DOI: 10.1016/j.lfs.2018.12.029] [Citation(s) in RCA: 229] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 12/10/2018] [Accepted: 12/15/2018] [Indexed: 12/21/2022]
Abstract
Mitochondrial function is vital for normal cellular processes. Mitochondrial damage and oxidative stress have been greatly implicated in the progression of aging, along with the pathogenesis of age-related neurodegenerative diseases (NDs), such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. Although antioxidant therapy has been proposed for the prevention and treatment of age-related NDs, unraveling the molecular mechanisms of mitochondrial dysfunction can lead to significant progress in the development of effective treatments against such diseases. Aging is associated with the generation and accumulation of reactive oxygen species (ROS) that are the major contributors to oxidative stress. Oxidative stress is caused because of the imbalance between the production of ROS and their oxidation, which can affect the mitochondrial respiratory chain function, thereby altering the membrane permeability and calcium homeostasis, along with increasing the heteroplasmic mtDNA and weakening the mitochondrial defense systems. Mitochondrial dysfunction mainly affects mitochondrial biogenesis and dynamics that are prominent in several age-related NDs. Mitochondrial dysfunction has a crucial role in the pathophysiology of age-related NDs. Several mitochondria targeted strategies, such as enhancing the antioxidant bioavailability via novel delivery systems, identifying unique mitochondrial proteins as specific drug targets, investigating the signaling pathways of mitochondrial biogenesis and dynamics, and identifying effective natural products are potentially effective to counteract mitochondrial dysfunction-related NDs.
Collapse
Affiliation(s)
- Hasnaa A Elfawy
- School of Biotechnology, KIIT deemed to be University, Campus XI, Bhubaneswar 751024, Odisha, India
| | - Biswadeep Das
- School of Biotechnology, KIIT deemed to be University, Campus XI, Bhubaneswar 751024, Odisha, India.
| |
Collapse
|
58
|
Neuroprotective Mechanisms of Resveratrol in Alzheimer's Disease: Role of SIRT1. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:8152373. [PMID: 30510627 PMCID: PMC6232815 DOI: 10.1155/2018/8152373] [Citation(s) in RCA: 234] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 07/21/2018] [Accepted: 08/15/2018] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a progressive and neurodegenerative disorder of the cortex and hippocampus, which eventually leads to cognitive impairment. Although the etiology of AD remains unclear, the presence of β-amyloid (Aβ) peptides in these learning and memory regions is a hallmark of AD. Therefore, the inhibition of Aβ peptide aggregation has been considered the primary therapeutic strategy for AD treatment. Many studies have shown that resveratrol has antioxidant, anti-inflammatory, and neuroprotective properties and can decrease the toxicity and aggregation of Aβ peptides in the hippocampus of AD patients, promote neurogenesis, and prevent hippocampal damage. In addition, the antioxidant activity of resveratrol plays an important role in neuronal differentiation through the activation of silent information regulator-1 (SIRT1). SIRT1 plays a vital role in the growth and differentiation of neurons and prevents the apoptotic death of these neurons by deacetylating and repressing p53 activity; however, the exact mechanisms remain unclear. Resveratrol also has anti-inflammatory effects as it suppresses M1 microglia activation, which is involved in the initiation of neurodegeneration, and promotes Th2 responses by increasing anti-inflammatory cytokines and SIRT1 expression. This review will focus on the antioxidant and anti-inflammatory neuroprotective effects of resveratrol, specifically on its role in SIRT1 and the association with AD pathophysiology.
Collapse
|
59
|
Govindarajulu M, Pinky PD, Bloemer J, Ghanei N, Suppiramaniam V, Amin R. Signaling Mechanisms of Selective PPAR γ Modulators in Alzheimer's Disease. PPAR Res 2018; 2018:2010675. [PMID: 30420872 PMCID: PMC6215547 DOI: 10.1155/2018/2010675] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/31/2018] [Accepted: 09/13/2018] [Indexed: 01/22/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease characterized by abnormal protein accumulation, synaptic dysfunction, and cognitive impairment. The continuous increase in the incidence of AD with the aged population and mortality rate indicates the urgent need for establishing novel molecular targets for therapeutic potential. Peroxisome proliferator-activated receptor gamma (PPARγ) agonists such as rosiglitazone and pioglitazone reduce amyloid and tau pathologies, inhibit neuroinflammation, and improve memory impairments in several rodent models and in humans with mild-to-moderate AD. However, these agonists display poor blood brain barrier permeability resulting in inadequate bioavailability in the brain and thus requiring high dosing with chronic time frames. Furthermore, these dosing levels are associated with several adverse effects including increased incidence of weight gain, liver abnormalities, and heart failure. Therefore, there is a need for identifying novel compounds which target PPARγ more selectively in the brain and could provide therapeutic benefits without a high incidence of adverse effects. This review focuses on how PPARγ agonists influence various pathologies in AD with emphasis on development of novel selective PPARγ modulators.
Collapse
Affiliation(s)
- Manoj Govindarajulu
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, USA
| | - Priyanka D. Pinky
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, USA
| | - Jenna Bloemer
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, USA
| | - Nila Ghanei
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, USA
| | - Vishnu Suppiramaniam
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, USA
- Center for Neuroscience, Auburn University, Auburn, AL, USA
| | - Rajesh Amin
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, USA
- Center for Neuroscience, Auburn University, Auburn, AL, USA
| |
Collapse
|
60
|
Corpas R, Griñán-Ferré C, Rodríguez-Farré E, Pallàs M, Sanfeliu C. Resveratrol Induces Brain Resilience Against Alzheimer Neurodegeneration Through Proteostasis Enhancement. Mol Neurobiol 2018; 56:1502-1516. [DOI: 10.1007/s12035-018-1157-y] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 05/28/2018] [Indexed: 12/27/2022]
|
61
|
Azimi M, Gharakhanlou R, Naghdi N, Khodadadi D, Heysieattalab S. Moderate treadmill exercise ameliorates amyloid-β-induced learning and memory impairment, possibly via increasing AMPK activity and up-regulation of the PGC-1α/FNDC5/BDNF pathway. Peptides 2018; 102:78-88. [PMID: 29309801 DOI: 10.1016/j.peptides.2017.12.027] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 12/27/2017] [Accepted: 12/29/2017] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder associated with loss of memory and cognitive abilities. Previous evidence suggested that exercise ameliorates learning and memory deficits by increasing brain derived neurotrophic factor (BDNF) and activating downstream pathways in AD animal models. However, upstream pathways related to increase BDNF induced by exercise in AD animal models are not well known. We investigated the effects of moderate treadmill exercise on Aβ-induced learning and memory impairment as well as the upstream pathway responsible for increasing hippocampal BDNF in an animal model of AD. Animals were divided into five groups: Intact, Sham, Aβ1-42, Sham-exercise (Sham-exe) and Aβ1-42-exercise (Aβ-exe). Aβ was microinjected into the CA1 area of the hippocampus and then animals in the exercise groups were subjected to moderate treadmill exercise (for 4 weeks with 5 sessions per week) 7 days after microinjection. In the present study the Morris water maze (MWM) test was used to assess spatial learning and memory. Hippocampal mRNA levels of BDNF, peroxisome proliferator-activated receptor gamma co-activator 1 alpha (PGC-1α), fibronectin type III domain-containing 5 (FNDC5) as well as protein levels of AMPK-activated protein kinase (AMPK), PGC-1α, BDNF, phosphorylation of AMPK were measured. Our results showed that intra-hippocampal injection of Aβ1-42 impaired spatial learning and memory which was accompanied by reduced AMPK activity (p-AMPK/total-AMPK ratio) and suppression of the PGC-1α/FNDC5/BDNF pathway in the hippocampus of rats. In contrast, moderate treadmill exercise ameliorated the Aβ1-42-induced spatial learning and memory deficit, which was accompanied by restored AMPK activity and PGC-1α/FNDC5/BDNF levels. Our results suggest that the increased AMPK activity and up-regulation of the PGC-1α/FNDC5/BDNF pathway by exercise are likely involved in mediating the beneficial effects of exercise on Aβ-induced learning and memory impairment.
Collapse
Affiliation(s)
- Mohammad Azimi
- Department of Physical Education and Sport Sciences, Faculty of Humanities, Tarbiat Modares University, Tehran, Iran
| | - Reza Gharakhanlou
- Department of Physical Education and Sport Sciences, Faculty of Humanities, Tarbiat Modares University, Tehran, Iran.
| | - Nasser Naghdi
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, 13164, Tehran, Iran
| | - Davar Khodadadi
- Department of Physical Education and Sport Sciences, Faculty of Humanities, Tarbiat Modares University, Tehran, Iran
| | - Soomaayeh Heysieattalab
- Cognitive Neuroscience Division, Faculty of Education and Psychology, University of Tabriz, Tabriz, Iran
| |
Collapse
|
62
|
Peroxisomal Acyl-CoA Oxidase Type 1: Anti-Inflammatory and Anti-Aging Properties with a Special Emphasis on Studies with LPS and Argan Oil as a Model Transposable to Aging. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6986984. [PMID: 29765501 PMCID: PMC5889864 DOI: 10.1155/2018/6986984] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 01/23/2018] [Indexed: 12/11/2022]
Abstract
To clarify appropriateness of current claims for health and wellness virtues of argan oil, studies were conducted in inflammatory states. LPS induces inflammation with reduction of PGC1-α signaling and energy metabolism. Argan oil protected the liver against LPS toxicity and interestingly enough preservation of peroxisomal acyl-CoA oxidase type 1 (ACOX1) activity against depression by LPS. This model of LPS-driven toxicity circumvented by argan oil along with a key anti-inflammatory role attributed to ACOX1 has been here transposed to model aging. This view is consistent with known physiological role of ACOX1 in yielding precursors of specialized proresolving mediators (SPM) and with characteristics of aging and related disorders including reduced PGC1-α function and improvement by strategies rising ACOX1 (via hormonal gut FGF19 and nordihydroguaiaretic acid in metabolic syndrome and diabetes conditions) and SPM (neurodegenerative disorders, atherosclerosis, and stroke). Delay of aging to resolve inflammation results from altered production of SPM, SPM improving most aging disorders. The strategic metabolic place of ACOX1, upstream of SPM biosynthesis, along with ability of ACOX1 preservation/induction and SPM to improve aging-related disorders and known association of aging with drop in ACOX1 and SPM, all converge to conclude that ACOX1 represents a previously unsuspected and currently emerging antiaging protein.
Collapse
|
63
|
Pohland M, Pellowska M, Asseburg H, Hagl S, Reutzel M, Joppe A, Berressem D, Eckert SH, Wurglics M, Schubert‐Zsilavecz M, Eckert GP. MH84 improves mitochondrial dysfunction in a mouse model of early Alzheimer's disease. Alzheimers Res Ther 2018; 10:18. [PMID: 29433569 PMCID: PMC5809956 DOI: 10.1186/s13195-018-0342-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 01/12/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND Current approved drugs for Alzheimer's disease (AD) only attenuate symptoms, but do not cure the disease. The pirinixic acid derivate MH84 has been characterized as a dual gamma-secretase/proliferator activated receptor gamma (PPARγ) modulator in vitro. Pharmacokinetic studies in mice showed that MH84 is bioavailable after oral administration and reaches the brain. We recently demonstrated that MH84 improved mitochondrial dysfunction in a cellular model of AD. In the present study, we extended the pharmacological characterization of MH84 to 3-month-old Thy-1 AβPPSL mice (harboring the Swedish and London mutation in human amyloid precursor protein (APP)) which are characterized by enhanced AβPP processing and cerebral mitochondrial dysfunction, representing a mouse model of early AD. METHODS Three-month-old Thy-1 AβPPSL mice received 12 mg/kg b.w. MH84 by oral gavage once a day for 21 days. Mitochondrial respiration was analyzed in isolated brain mitochondria, and mitochondrial membrane potential and ATP levels were determined in dissociated brain cells. Citrate synthase (CS) activity was determined in brain tissues and MitoTracker Green fluorescence was measured in HEK293-AβPPwt and HEK293-AβPPsw cells. Soluble Aβ1-40 and Aβ1-42 levels were determined using ELISA. Western blot analysis and qRT-PCR were used to measure protein and mRNA levels, respectively. RESULTS MH84 reduced cerebral levels of the β-secretase-related C99 peptide and of Aβ40 levels. Mitochondrial dysfunction was ameliorated by restoring complex IV (cytochrome-c oxidase) respiration, mitochondrial membrane potential, and levels of ATP. Induction of PPARγ coactivator-1α (PGC-1α) mRNA and protein expression was identified as a possible mode of action that leads to increased mitochondrial mass as indicated by enhanced CS activity, OXPHOS levels, and MitoTracker Green fluorescence. CONCLUSIONS MH84 modulates β-secretase processing of APP and improves mitochondrial dysfunction by a PGC-1α-dependent mechanism. Thus, MH84 seems to be a new promising therapeutic agent with approved in-vivo activity for the treatment of AD.
Collapse
Affiliation(s)
| | - Maren Pellowska
- Institute of Pharmaceutical Chemistry, Goethe University, Frankfurt, Germany
| | - Heike Asseburg
- Institute of Pharmacology, Goethe University, Frankfurt, Germany
- Institute of Nutritional Sciences, Justus-Liebig-University, Giessen, Germany
| | - Stephanie Hagl
- Institute of Pharmacology, Goethe University, Frankfurt, Germany
| | - Martina Reutzel
- Institute of Nutritional Sciences, Justus-Liebig-University, Giessen, Germany
| | - Aljoscha Joppe
- Institute of Pharmacology, Goethe University, Frankfurt, Germany
| | - Dirk Berressem
- Institute of Pharmacology, Goethe University, Frankfurt, Germany
| | | | - Mario Wurglics
- Institute of Pharmaceutical Chemistry, Goethe University, Frankfurt, Germany
| | | | - Gunter P. Eckert
- Institute of Nutritional Sciences, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
64
|
Sun W, Nguyen KD, Fitch WL, Banister SD, Tang H, Zhang X, Yu L, Engleman EG, Rajadas J. In vitro and in vivo metabolite identification of a novel benzimidazole compound ZLN005 by LC-MS/MS. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2018; 32:480-488. [PMID: 29334584 DOI: 10.1002/rcm.8060] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 12/05/2017] [Accepted: 01/04/2018] [Indexed: 06/07/2023]
Abstract
RATIONALE A novel benzimidazole compound ZLN005 was previously identified as a transcriptional activator of peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) in certain metabolic tissues. Upregulation of PGC-1α by ZLN005 has been shown to have beneficial effect in a diabetic mouse model and in a coronary artery disease model in vitro. ZLN005 could also have therapeutic potential in neurodegenerative diseases involving down-regulation of PGC-1α. Given the phenotypic efficacy of ZLN005 in several animal models of human disease, its metabolic profile was investigated to guide the development of novel therapeutics using ZLN005 as the lead compound. METHODS ZLN005 was incubated with both rat and human liver microsomes and S9 fractions to identify in vitro metabolites. Urine from rats dosed with ZLN005 was used to identify in vivo metabolites. Extracted metabolites were analyzed by LC-MS/MS using a hybrid linear ion trap triple quadrupole mass spectrometer under full scan, enhanced product ion scan, neutral loss scan and precursor scan modes. Metabolites in plasma and brain of ZLN005-treated rats were also profiled using multiple reaction monitoring. RESULTS Identified in vitro transformations of ZLN005 include mono- and dihydroxylation, further oxidation to carboxylic acids, and mono-O-glucuronide and sulfate conjugation to hydroxy ZLN005 as well as glutathione conjugation. Identified in vivo metabolites are mainly glucuronide and sulfate conjugates of dihydroxyl, carboxyl, and hydroxy acid of the parent compound. The parent compound as well as several major phase I metabolites were found in rat plasma and brain. CONCLUSIONS Using both in vitro and in vivo methods, we elucidated the metabolic pathway of ZLN005. Phase I metabolites with hydroxylation and carboxylation, as well as phase II metabolites with glucuronide, sulfate and glutathione conjugation, were identified.
Collapse
Affiliation(s)
- Wenchao Sun
- Biomaterials and Advance Drug Delivery Lab, School of Medicine, Stanford University, USA
| | - Khoa Dinh Nguyen
- Department of Pathology, School of Medicine, Stanford University, USA
| | - William L Fitch
- Biomaterials and Advance Drug Delivery Lab, School of Medicine, Stanford University, USA
| | - Samuel D Banister
- Medicinal Chemistry Knowledge Center, ChEM-H, Stanford University, USA
| | - Hongxiang Tang
- Biomaterials and Advance Drug Delivery Lab, School of Medicine, Stanford University, USA
| | - Xiaolan Zhang
- Biomaterials and Advance Drug Delivery Lab, School of Medicine, Stanford University, USA
| | - Lewis Yu
- Department of Pathology, School of Medicine, Stanford University, USA
| | - Edgar G Engleman
- Department of Pathology, School of Medicine, Stanford University, USA
| | - Jayakumar Rajadas
- Biomaterials and Advance Drug Delivery Lab, School of Medicine, Stanford University, USA
- Department of Bioengineering and Therapeutic Sciences, School of Pharmacy, University of California, San Francisco
| |
Collapse
|
65
|
Sajan MP, Hansen BC, Higgs MG, Kahn CR, Braun U, Leitges M, Park CR, Diamond DM, Farese RV. Atypical PKC, PKCλ/ι, activates β-secretase and increases Aβ 1-40/42 and phospho-tau in mouse brain and isolated neuronal cells, and may link hyperinsulinemia and other aPKC activators to development of pathological and memory abnormalities in Alzheimer's disease. Neurobiol Aging 2018; 61:225-237. [PMID: 29032894 PMCID: PMC5705272 DOI: 10.1016/j.neurobiolaging.2017.09.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 08/25/2017] [Accepted: 09/03/2017] [Indexed: 12/31/2022]
Abstract
Hyperinsulinemia activates brain Akt and PKC-λ/ι and increases Aβ1-40/42 and phospho-tau in insulin-resistant animals. Here, we examined underlying mechanisms in mice, neuronal cells, and mouse hippocampal slices. Like Aβ1-40/42, β-secretase activity was increased in insulin-resistant mice and monkeys. In insulin-resistant mice, inhibition of hepatic PKC-λ/ι sufficient to correct hepatic abnormalities and hyperinsulinemia simultaneously reversed increases in Akt, atypical protein kinase C (aPKC), β-secretase, and Aβ1-40/42, and restored acute Akt activation. However, 2 aPKC inhibitors additionally blocked insulin's ability to activate brain PKC-λ/ι and thereby increase β-secretase and Aβ1-40/42. Furthermore, direct blockade of brain aPKC simultaneously corrected an impairment in novel object recognition in high-fat-fed insulin-resistant mice. In neuronal cells and/or mouse hippocampal slices, PKC-ι/λ activation by insulin, metformin, or expression of constitutive PKC-ι provoked increases in β-secretase, Aβ1-40/42, and phospho-thr-231-tau that were blocked by various PKC-λ/ι inhibitors, but not by an Akt inhibitor. PKC-λ/ι provokes increases in brain β-secretase, Aβ1-40/42, and phospho-thr-231-tau. Excessive signaling via PKC-λ/ι may link hyperinsulinemia and other PKC-λ/ι activators to pathological and functional abnormalities in Alzheimer's disease.
Collapse
Affiliation(s)
- Mini P Sajan
- Internal Medicine and Research Services, James A. Haley Veterans Medical Center, Tampa, Florida, USA; Department of Internal Medicine, University of South Florida College of Medicine, Tampa, Florida, USA
| | - Barbara C Hansen
- Internal Medicine and Research Services, James A. Haley Veterans Medical Center, Tampa, Florida, USA
| | - Margaret G Higgs
- Internal Medicine and Research Services, James A. Haley Veterans Medical Center, Tampa, Florida, USA
| | - C Ron Kahn
- Joslin Research Foundation, Harvard Medical School, Boston, MA, USA
| | | | | | - Collin R Park
- Departments of Psychology, Molecular Pharmacology and Physiology, Center for Preclinical and Clinical Research on PTSD, University of South Florida, Tampa, Florida, USA
| | - David M Diamond
- Departments of Psychology, Molecular Pharmacology and Physiology, Center for Preclinical and Clinical Research on PTSD, University of South Florida, Tampa, Florida, USA
| | - Robert V Farese
- Internal Medicine and Research Services, James A. Haley Veterans Medical Center, Tampa, Florida, USA; Department of Internal Medicine, University of South Florida College of Medicine, Tampa, Florida, USA.
| |
Collapse
|
66
|
Lin H, Magrane J, Rattelle A, Stepanova A, Galkin A, Clark EM, Dong YN, Halawani SM, Lynch DR. Early cerebellar deficits in mitochondrial biogenesis and respiratory chain complexes in the KIKO mouse model of Friedreich ataxia. Dis Model Mech 2017; 10:1343-1352. [PMID: 29125827 PMCID: PMC5719255 DOI: 10.1242/dmm.030502] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 08/11/2017] [Indexed: 12/14/2022] Open
Abstract
Friedreich ataxia (FRDA), the most common recessive inherited ataxia, results from deficiency of frataxin, a small mitochondrial protein crucial for iron-sulphur cluster formation and ATP production. Frataxin deficiency is associated with mitochondrial dysfunction in FRDA patients and animal models; however, early mitochondrial pathology in FRDA cerebellum remains elusive. Using frataxin knock-in/knockout (KIKO) mice and KIKO mice carrying the mitoDendra transgene, we show early cerebellar deficits in mitochondrial biogenesis and respiratory chain complexes in this FRDA model. At asymptomatic stages, the levels of PGC-1α (PPARGC1A), the mitochondrial biogenesis master regulator, are significantly decreased in cerebellar homogenates of KIKO mice compared with age-matched controls. Similarly, the levels of the PGC-1α downstream effectors, NRF1 and Tfam, are significantly decreased, suggesting early impaired cerebellar mitochondrial biogenesis pathways. Early mitochondrial deficiency is further supported by significant reduction of the mitochondrial markers GRP75 (HSPA9) and mitofusin-1 in the cerebellar cortex. Moreover, the numbers of Dendra-labeled mitochondria are significantly decreased in cerebellar cortex, confirming asymptomatic cerebellar mitochondrial biogenesis deficits. Functionally, complex I and II enzyme activities are significantly reduced in isolated mitochondria and tissue homogenates from asymptomatic KIKO cerebella. Structurally, levels of the complex I core subunit NUDFB8 and complex II subunits SDHA and SDHB are significantly lower than those in age-matched controls. These results demonstrate complex I and II deficiency in KIKO cerebellum, consistent with defects identified in FRDA patient tissues. Thus, our findings identify early cerebellar mitochondrial biogenesis deficits as a potential mediator of cerebellar dysfunction and ataxia, thereby providing a potential therapeutic target for early intervention of FRDA.
Collapse
Affiliation(s)
- Hong Lin
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jordi Magrane
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY 10065, USA
| | - Amy Rattelle
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Anna Stepanova
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY 10065, USA
- Queen's University Belfast, School of Biological Sciences, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Alexander Galkin
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY 10065, USA
- Queen's University Belfast, School of Biological Sciences, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Elisia M Clark
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yi Na Dong
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Sarah M Halawani
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - David R Lynch
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
67
|
Grings M, Moura AP, Parmeggiani B, Pletsch JT, Cardoso GMF, August PM, Matté C, Wyse ATS, Wajner M, Leipnitz G. Bezafibrate prevents mitochondrial dysfunction, antioxidant system disturbance, glial reactivity and neuronal damage induced by sulfite administration in striatum of rats: Implications for a possible therapeutic strategy for sulfite oxidase deficiency. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2135-2148. [PMID: 28529047 DOI: 10.1016/j.bbadis.2017.05.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 04/30/2017] [Accepted: 05/17/2017] [Indexed: 12/13/2022]
Abstract
Sulfite accumulates in tissues of patients affected by sulfite oxidase (SO) deficiency, a neurometabolic disease characterized by seizures and progressive encephalopathy, often resulting in early death. We investigated the effects of sulfite on mitochondrial function, antioxidant system, glial reactivity and neuronal damage in rat striatum, as well as the potential protective effects of bezafibrate on sulfite-induced toxicity. Thirty-day-old rats were intrastriatally administered with sulfite (2μmol) or NaCl (2μmol; control) and euthanized 30min after injection for evaluation of biochemical parameters and western blotting, or 7days after injection for analysis of glial reactivity and neuronal damage. Treatment with bezafibrate (30 or 100mg/kg/day) was performed by gavage during 7days before (pre-treatment) or after sulfite administration. Sulfite decreased creatine kinase and citrate synthase activities, mitochondrial mass, and PGC-1α nuclear content whereas bezafibrate pre-treatment prevented these alterations. Sulfite also diminished cytochrome c oxidase (COX) IV-1 content, glutathione levels and the activities of glutathione peroxidase (GPx), glutathione reductase (GR), glutathione S-transferase (GST) and glucose-6-phosphate dehydrogenase (G6PDH). On the other hand, catalase activity was increased by sulfite. Bezafibrate pre-treatment prevented the reduction of GPx, GR, GST and G6PDH activities. Finally, sulfite induced glial reactivity and neuronal damage, which were prevented by bezafibrate when administered before or after sulfite administration. Our findings provide strong evidence that sulfite induces neurotoxicity that leads to glial reactivity and neuronal damage. Since bezafibrate exerts neuroprotective effects against sulfite toxicity, it may be an attractive agent for the development of novel therapeutic strategies for SO-deficient patients.
Collapse
Affiliation(s)
- Mateus Grings
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Alana Pimentel Moura
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Belisa Parmeggiani
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Julia Tauana Pletsch
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Gabriela Miranda Fernandez Cardoso
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Pauline Maciel August
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Cristiane Matté
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil; Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Angela T S Wyse
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil; Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Moacir Wajner
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil; Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil; Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, CEP 90035-903 Porto Alegre, RS, Brazil
| | - Guilhian Leipnitz
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil; Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil.
| |
Collapse
|
68
|
Chandran JS, Scarrott JM, Shaw PJ, Azzouz M. Gene Therapy in the Nervous System: Failures and Successes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1007:241-257. [PMID: 28840561 DOI: 10.1007/978-3-319-60733-7_13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Genetic disorders, caused by deleterious changes in the DNA sequence away from the normal genomic sequence, affect millions of people worldwide. Gene therapy as a treatment option for patients is an attractive proposition due to its conceptual simplicity. In principle, gene therapy involves correcting the genetic disorder by either restoring a normal functioning copy of a gene or reducing the toxicity arising from a mutated gene. In this way specific genetic function can be restored without altering the expression of other genes and the proteins they encode. The reality however is much more complex, and as a result the vector systems used to deliver gene therapies have by necessity continued to evolve and improve over time with respect to safety profile, efficiency, and long-term expression. In this chapter we examine the current approaches to gene therapy, assess the different gene delivery systems utilized, and highlight the failures and successes of relevant clinical trials. We do not intend for this chapter to be a comprehensive and exhaustive assessment of all clinical trials that have been conducted in the CNS, but instead will focus on specific diseases that have seen successes and failures with different gene therapy vehicles to gauge how preclinical models have informed the design of clinical trials.
Collapse
Affiliation(s)
- Jayanth S Chandran
- Sheffield Institute for Translational Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
| | - Joseph M Scarrott
- Sheffield Institute for Translational Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
| | - Mimoun Azzouz
- Sheffield Institute for Translational Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK.
| |
Collapse
|
69
|
Jęśko H, Wencel P, Strosznajder RP, Strosznajder JB. Sirtuins and Their Roles in Brain Aging and Neurodegenerative Disorders. Neurochem Res 2016; 42:876-890. [PMID: 27882448 PMCID: PMC5357501 DOI: 10.1007/s11064-016-2110-y] [Citation(s) in RCA: 186] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 10/21/2016] [Accepted: 11/14/2016] [Indexed: 02/07/2023]
Abstract
Sirtuins (SIRT1-SIRT7) are unique histone deacetylases (HDACs) whose activity depends on NAD+ levels and thus on the cellular metabolic status. SIRTs regulate energy metabolism and mitochondrial function. They orchestrate the stress response and damage repair. Through these functions sirtuins modulate the course of aging and affect neurodegenerative diseases. SIRTSs interact with multiple signaling proteins, transcription factors (TFs) and poly(ADP-ribose) polymerases (PARPs) another class of NAD+-dependent post-translational protein modifiers. The cross-talk between SIRTs TFs and PARPs is a highly promising research target in a number of brain pathologies. This review describes updated results on sirtuins in brain aging/neurodegeneration. It focuses on SIRT1 but also on the roles of mitochondrial SIRTs (SIRT3, 4, 5) and on SIRT6 and SIRT2 localized in the nucleus and in cytosol, respectively. The involvement of SIRTs in regulation of insulin-like growth factor signaling in the brain during aging and in Alzheimer's disease was also focused. Moreover, we analyze the mechanism(s) and potential significance of interactions between SIRTs and several TFs in the regulation of cell survival and death. A critical view is given on the application of SIRT activators/modulators in therapy of neurodegenerative diseases.
Collapse
Affiliation(s)
- Henryk Jęśko
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego st., 02106, Warsaw, Poland
| | - Przemysław Wencel
- Laboratory of Preclinical Research and Environmental Agents, Department of Neurosurgery, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego st., 02106, Warsaw, Poland
| | - Robert P Strosznajder
- Laboratory of Preclinical Research and Environmental Agents, Department of Neurosurgery, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego st., 02106, Warsaw, Poland.
| | - Joanna B Strosznajder
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego st., 02106, Warsaw, Poland
| |
Collapse
|
70
|
Jha SK, Jha NK, Kumar D, Ambasta RK, Kumar P. Linking mitochondrial dysfunction, metabolic syndrome and stress signaling in Neurodegeneration. Biochim Biophys Acta Mol Basis Dis 2016; 1863:1132-1146. [PMID: 27345267 DOI: 10.1016/j.bbadis.2016.06.015] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 06/16/2016] [Accepted: 06/17/2016] [Indexed: 12/13/2022]
Abstract
Mounting evidence suggests a link between metabolic syndrome (MetS) such as diabetes, obesity, non-alcoholic fatty liver disease in the progression of Alzheimer's disease (AD), Parkinson's disease (PD) and other neurodegenerative diseases (NDDs). For instance, accumulated Aβ oligomer is enhancing neuronal Ca2+ release and neural NO where increased NO level in the brain through post translational modification is modulating the level of insulin production. It has been further confirmed that irrespective of origin; brain insulin resistance triggers a cascade of the neurodegeneration phenomenon which can be aggravated by free reactive oxygen species burden, ER stress, metabolic dysfunction, neuorinflammation, reduced cell survival and altered lipid metabolism. Moreover, several studies confirmed that MetS and diabetic sharing common mechanisms in the progression of AD and NDDs where mitochondrial dynamics playing a critical role. Any mutation in mitochondrial DNA, exposure of environmental toxin, high-calorie intake, homeostasis imbalance, glucolipotoxicity is causative factors for mitochondrial dysfunction. These cumulative pleiotropic burdens in mitochondria leads to insulin resistance, increased ROS production; enhanced stress-related enzymes that is directly linked MetS and diabetes in neurodegeneration. Since, the linkup mechanism between mitochondrial dysfunction and disease phenomenon of both MetS and NDDs is quite intriguing, therefore, it is pertinent for the researchers to identify and implement the therapeutic interventions for targeting MetS and NDDs. Herein, we elucidated the pertinent role of MetS induced mitochondrial dysfunction in neurons and their consequences in NDDs. Further, therapeutic potential of well-known biomolecules and chaperones to target altered mitochondria has been comprehensively documented. This article is part of a Special Issue entitled: Oxidative Stress and Mitochondrial Quality in Diabetes/Obesity and Critical Illness Spectrum of Diseases - edited by P. Hemachandra Reddy.
Collapse
Affiliation(s)
- Saurabh Kumar Jha
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India
| | - Niraj Kumar Jha
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India
| | - Dhiraj Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India.
| |
Collapse
|