101
|
Moulis MF, Millet AM, Daloyau M, Miquel MC, Ronsin B, Wissinger B, Arnauné-Pelloquin L, Belenguer P. OPA1 haploinsufficiency induces a BNIP3-dependent decrease in mitophagy in neurons: relevance to Dominant Optic Atrophy. J Neurochem 2016; 140:485-494. [DOI: 10.1111/jnc.13894] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 09/28/2016] [Accepted: 10/24/2016] [Indexed: 12/31/2022]
Affiliation(s)
- Manon F Moulis
- Research Center on Animal Cognition (CRCA); Center for Integrative Biology (CBI); Toulouse University; CNRS UPS France
| | - Aurélie M Millet
- Research Center on Animal Cognition (CRCA); Center for Integrative Biology (CBI); Toulouse University; CNRS UPS France
| | - Marlène Daloyau
- Research Center on Animal Cognition (CRCA); Center for Integrative Biology (CBI); Toulouse University; CNRS UPS France
| | - Marie-Christine Miquel
- Research Center on Animal Cognition (CRCA); Center for Integrative Biology (CBI); Toulouse University; CNRS UPS France
| | - Brice Ronsin
- Center of Developmental Biology (CBD); Center for Integrative Biology (CBI); Toulouse University; CNRS UPS France
| | - Bernd Wissinger
- Center for Ophthalmology; University of Tübingen; Tübingen Germany
| | - Laetitia Arnauné-Pelloquin
- Research Center on Animal Cognition (CRCA); Center for Integrative Biology (CBI); Toulouse University; CNRS UPS France
| | - Pascale Belenguer
- Research Center on Animal Cognition (CRCA); Center for Integrative Biology (CBI); Toulouse University; CNRS UPS France
| |
Collapse
|
102
|
Dominant optic atrophy: updates on the pathophysiology and clinical manifestations of the optic atrophy 1 mutation. Curr Opin Ophthalmol 2016; 27:475-480. [PMID: 27585216 DOI: 10.1097/icu.0000000000000314] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
PURPOSE OF REVIEW Review recent advances in clinical and experimental studies of dominant optic atrophy (DOA) to better understand the complexities of pathophysiology caused by the optic atrophy 1 (OPA1) mutation. RECENT FINDINGS DOA is the most commonly diagnosed inherited optic atrophy, causing progressive bilateral visual loss that begins early in life. During the past 25 years, there has been substantial progress in the understanding of the clinical, genetic, and pathophysiological basis of this disease. The histopathological hallmark of DOA is the primary degeneration of retinal ganglion cells, preferentially in the papillomacular bundle, which results temporal optic disc pallor and cecocentral scotomata in patients with DOA. Loss of OPA1 protein function by OPA1 gene mutations causes mitochondrial dysfunction because of the loss of mitochondrial fusion, impaired mitochondrial oxidative phosphorylation, increases in reactive oxygen species, and altered calcium homeostasis. These factors lead to apoptosis of retinal ganglion cells by a haploinsufficiency mechanism. SUMMARY Improved understanding of the pathophysiology of DOA provides insights that can be used to develop therapeutic approaches to the DOA.
Collapse
|
103
|
Salim S. Oxidative Stress and the Central Nervous System. J Pharmacol Exp Ther 2016; 360:201-205. [PMID: 27754930 DOI: 10.1124/jpet.116.237503] [Citation(s) in RCA: 731] [Impact Index Per Article: 91.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 10/14/2016] [Indexed: 01/19/2023] Open
Abstract
Biochemical integrity of the brain is vital for normal functioning of the central nervous system (CNS). One of the factors contributing to cerebral biochemical impairment is a chemical process called oxidative stress. Oxidative stress occurs upon excessive free radical production resulting from an insufficiency of the counteracting antioxidant response system. The brain, with its high oxygen consumption and lipid-rich content, is highly susceptible to oxidative stress. Therefore, oxidative stress-induced damage to the brain has a strong potential to negatively impact normal CNS functions. Although oxidative stress has historically been considered to be involved mainly in neurodegenerative disorders such as Alzheimer disease, Huntington disease, and Parkinson disease, its involvement in neuropsychiatric disorders, including anxiety disorders and depression, is beginning to be recognized. This review is a discussion of the relevance of cerebral oxidative stress to impairment of emotional and mental well-being.
Collapse
Affiliation(s)
- Samina Salim
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| |
Collapse
|
104
|
Mitochondrial pathogenic mechanism and degradation in optineurin E50K mutation-mediated retinal ganglion cell degeneration. Sci Rep 2016; 6:33830. [PMID: 27654856 PMCID: PMC5031982 DOI: 10.1038/srep33830] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 09/02/2016] [Indexed: 01/15/2023] Open
Abstract
Mutations in optineurin (OPTN) are linked to the pathology of primary open angle glaucoma (POAG) and amyotrophic lateral sclerosis. Emerging evidence indicates that OPTN mutation is involved in accumulation of damaged mitochondria and defective mitophagy. Nevertheless, the role played by an OPTN E50K mutation in the pathogenic mitochondrial mechanism that underlies retinal ganglion cell (RGC) degeneration in POAG remains unknown. We show here that E50K expression induces mitochondrial fission-mediated mitochondrial degradation and mitophagy in the axons of the glial lamina of aged E50K−tg mice in vivo. While E50K activates the Bax pathway and oxidative stress, and triggers dynamics alteration-mediated mitochondrial degradation and mitophagy in RGC somas in vitro, it does not affect transport dynamics and fission of mitochondria in RGC axons in vitro. These results strongly suggest that E50K is associated with mitochondrial dysfunction in RGC degeneration in synergy with environmental factors such as aging and/or oxidative stress.
Collapse
|
105
|
Tiwari HS, Misra UK, Kalita J, Mishra A, Shukla S. Oxidative stress and glutamate excitotoxicity contribute to apoptosis in cerebral venous sinus thrombosis. Neurochem Int 2016; 100:91-96. [PMID: 27620812 DOI: 10.1016/j.neuint.2016.09.003] [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: 04/11/2016] [Revised: 08/30/2016] [Accepted: 09/06/2016] [Indexed: 02/06/2023]
Abstract
Cerebral venous sinus thrombosis (CVST) is an important cause of stroke in young especially in the developing countries. There is paucity of studies on the mechanism of cell damage in CVST. Aim of this study is to explore the role of glutamate excitotoxicity, oxidative stress; and apoptosis in an experimental model of CVST. Cerebral venous sinus thrombosis was induced by putting a strip of filter paper soaked in 40% ferric chloride on superior sagittal sinus. Brain was removed on day 1, 2 and 7 and oxidative stress markers (Mn-SOD, Catalase, GPx, LPO and GSH) were estimated spectrophotometrically. Glutamate level and its receptors NR1, NR2A and NR2B were estimated by real time polymerase chain reaction. The markers of apoptosis were evaluated by expression of cleaved Caspase-3 and specrtin break down product (SBDP) by western blot. In CVST, the evidence of oxidative stress (reduced activity of Mn-SOD, Catalase, GPx, GSH and increased level of LPO) was noted. Glutamate level was elevated and its receptors NR1, NR2A and NR2B were reduced. Increased expression of cleaved Caspase-3 on day 2 and 7 and SBDP on day 1 and 2 were noted. Oxidative stress, glutamate excitotoxicity and apoptosis seem to have important role in CVST induced cell damage.
Collapse
Affiliation(s)
- Hari Shanker Tiwari
- Department of Neurology, Sanjay Gandhi Post Graduate Medical Sciences, Lucknow, India
| | - Usha Kant Misra
- Department of Neurology, Sanjay Gandhi Post Graduate Medical Sciences, Lucknow, India.
| | - Jayantee Kalita
- Department of Neurology, Sanjay Gandhi Post Graduate Medical Sciences, Lucknow, India
| | - Akanksha Mishra
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Shubha Shukla
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, India
| |
Collapse
|
106
|
Role of NMDA Receptor-Mediated Glutamatergic Signaling in Chronic and Acute Neuropathologies. Neural Plast 2016; 2016:2701526. [PMID: 27630777 PMCID: PMC5007376 DOI: 10.1155/2016/2701526] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 06/13/2016] [Accepted: 06/29/2016] [Indexed: 12/11/2022] Open
Abstract
N-Methyl-D-aspartate receptors (NMDARs) have two opposing roles in the brain. On the one hand, NMDARs control critical events in the formation and development of synaptic organization and synaptic plasticity. On the other hand, the overactivation of NMDARs can promote neuronal death in neuropathological conditions. Ca(2+) influx acts as a primary modulator after NMDAR channel activation. An imbalance in Ca(2+) homeostasis is associated with several neurological diseases including schizophrenia, Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. These chronic conditions have a lengthy progression depending on internal and external factors. External factors such as acute episodes of brain damage are associated with an earlier onset of several of these chronic mental conditions. Here, we will review some of the current evidence of how traumatic brain injury can hasten the onset of several neurological conditions, focusing on the role of NMDAR distribution and the functional consequences in calcium homeostasis associated with synaptic dysfunction and neuronal death present in this group of chronic diseases.
Collapse
|
107
|
Kushnareva Y, Seong Y, Andreyev AY, Kuwana T, Kiosses WB, Votruba M, Newmeyer DD. Mitochondrial dysfunction in an Opa1(Q285STOP) mouse model of dominant optic atrophy results from Opa1 haploinsufficiency. Cell Death Dis 2016; 7:e2309. [PMID: 27468686 PMCID: PMC4973340 DOI: 10.1038/cddis.2016.160] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 04/29/2016] [Accepted: 05/02/2016] [Indexed: 12/22/2022]
Abstract
Mutations in the opa1 (optic atrophy 1) gene lead to autosomal dominant optic atrophy (ADOA), a hereditary eye disease. This gene encodes the Opa1 protein, a mitochondrial dynamin-related GTPase required for mitochondrial fusion and the maintenance of normal crista structure. The majority of opa1 mutations encode truncated forms of the protein, lacking a complete GTPase domain. It is unclear whether the phenotype results from haploinsufficiency or rather a deleterious effect of truncated Opa1 protein. We studied a heterozygous Opa1 mutant mouse carrying a defective allele with a stop codon in the beginning of the GTPase domain at residue 285, a mutation that mimics human pathological mutations. Using an antibody raised against an N-terminal portion of Opa1, we found that the level of wild-type protein was decreased in the mutant mice, as predicted. However, no truncated Opa1 protein was expressed. In embryonic fibroblasts isolated from the mutant mice, this partial loss of Opa1 caused mitochondrial respiratory deficiency and a selective loss of respiratory Complex IV subunits. Furthermore, partial Opa1 deficiency resulted in a substantial resistance to endoplasmic reticulum stress-induced death. On the other hand, the enforced expression of truncated Opa1 protein in cells containing normal levels of wild-type protein did not cause mitochondrial defects. Moreover, cells expressing the truncated Opa1 protein showed reduced Bax activation in response to apoptotic stimuli. Taken together, our results exclude deleterious dominant-negative or gain-of-function mechanisms for this type of Opa1 mutation and affirm haploinsufficiency as the mechanism underlying mitochondrial dysfunction in ADOA.
Collapse
Affiliation(s)
- Y Kushnareva
- Immune Regulation, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Y Seong
- Immune Regulation, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - A Y Andreyev
- Department of Pharmacology, University of California San Diego, La Jolla, CA 92093, USA
| | - T Kuwana
- Immune Regulation, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - W B Kiosses
- Immune Regulation, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - M Votruba
- School of Optometry and Vision Sciences, Cardiff University, Cardiff CF24 4LU, UK.,Cardiff Eye Unit, University Hospital Wales, Cardiff CF14 4XW, UK
| | - D D Newmeyer
- Immune Regulation, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| |
Collapse
|
108
|
Abstract
Cerebellar disorders trigger the symptoms of movement problems, imbalance, incoordination, and frequent fall. Cerebellar disorders are shown in various CNS illnesses including a drinking disorder called alcoholism. Alcoholism is manifested as an inability to control drinking in spite of adverse consequences. Human and animal studies have shown that cerebellar symptoms persist even after complete abstinence from drinking. In particular, the abrupt termination (ethanol withdrawal) of long-term excessive ethanol consumption has shown to provoke a variety of neuronal and mitochondrial damage to the cerebellum. Upon ethanol withdrawal, excitatory neurotransmitter molecules such as glutamate are overly released in brain areas including cerebellum. This is particularly relevant to the cerebellar neuronal network as glutamate signals are projected to Purkinje neurons through granular cells that are the most populated neuronal type in CNS. This excitatory neuronal signal may be elevated by ethanol withdrawal stress, which promotes an increase in intracellular Ca(2+) level and a decrease in a Ca(2+)-binding protein, both of which result in the excessive entry of Ca(2+) to the mitochondria. Subsequently, mitochondria undergo a prolonged opening of mitochondrial permeability transition pore and the overproduction of harmful free radicals, impeding adenosine triphosphate (ATP)-generating function. This in turn provokes the leakage of mitochondrial molecule cytochrome c to the cytosol, which triggers a cascade of adverse cytosol reactions. Upstream to this pathway, cerebellum under the condition of ethanol withdrawal has shown aberrant gene modifications through altered DNA methylation, histone acetylation, or microRNA expression. Interplay between these events and molecules may result in functional damage to cerebellar mitochondria and consequent neuronal degeneration, thereby contributing to motoric deficit. Mitochondria-targeting research may help develop a powerful new therapy to manage cerebellar disorders associated with hyperexcitatory CNS disorders like ethanol withdrawal.
Collapse
Affiliation(s)
- Marianna E Jung
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie Blvd., Fort Worth, TX, 76107-2699, USA,
| |
Collapse
|
109
|
Bolaños JP. Bioenergetics and redox adaptations of astrocytes to neuronal activity. J Neurochem 2016; 139 Suppl 2:115-125. [PMID: 26968531 PMCID: PMC5018236 DOI: 10.1111/jnc.13486] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 11/19/2015] [Accepted: 11/23/2015] [Indexed: 12/14/2022]
Abstract
Neuronal activity is a high‐energy demanding process recruiting all neural cells that adapt their metabolism to sustain the energy and redox balance of neurons. During neurotransmission, synaptic cleft glutamate activates its receptors in neurons and in astrocytes, before being taken up by astrocytes through energy costly transporters. In astrocytes, the energy requirement for glutamate influx is likely to be met by glycolysis. To enable this, astrocytes are constitutively glycolytic, robustly expressing 6‐phosphofructo‐2‐kinase/fructose‐2,6‐bisphosphatase‐3 (PFKFB3), an enzyme that is negligibly present in neurons by continuous degradation because of the ubiquitin‐proteasome pathway via anaphase‐promoting complex/cyclosome (APC)‐Cdh1. Additional factors contributing to the glycolytic frame of astrocytes may include 5′‐AMP‐activated protein kinase (AMPK), hypoxia‐inducible factor‐1 (HIF‐1), pyruvate kinase muscle isoform‐2 (PKM2), pyruvate dehydrogenase kinase‐4 (PDK4), lactate dehydrogenase‐B, or monocarboxylate transporter‐4 (MCT4). Neurotransmission‐associated messengers, such as nitric oxide or ammonium, stimulate lactate release from astrocytes. Astrocyte‐derived glycolytic lactate thus sustains the energy needs of neurons, which in contrast to astrocytes mainly rely on oxidative phosphorylation. Neuronal activity unavoidably triggers reactive oxygen species, but the antioxidant defense of neurons is weak; hence, they use glucose for oxidation through the pentose‐phosphate pathway to preserve the redox status. Furthermore, neural activity is coupled with erythroid‐derived erythroid‐derived 2‐like 2 (Nrf2) mediated transcriptional activation of antioxidant genes in astrocytes, which boost the de novo glutathione biosynthesis in neighbor neurons. Thus, the bioenergetics and redox programs of astrocytes are adapted to sustain neuronal activity and survival. Developing therapeutic strategies to interfere with these pathways may be useful to combat neurological diseases.
Our current knowledge on brain's management of bioenergetics and redox requirements associated with neural activity is herein revisited. The astrocyte‐neuronal lactate shuttle (ANLS) explains the energy needs of neurotransmission. Furthermore, neurotransmission unavoidably triggers increased mitochondrial reactive oxygen species in neurons. By coupling glutamatergic activity with transcriptional activation of antioxidant genes, astrocytes provide neurons with neuroprotective glutathione through an astrocyte‐neuronal glutathione shuttle (ANGS).
This article is part of the60th Anniversary special issue.
Collapse
Affiliation(s)
- Juan P Bolaños
- Institute of Functional Biology and Genomics (IBFG), University of Salamanca-CSIC-IBSAL, Salamanca, Spain.
| |
Collapse
|
110
|
|
111
|
The Response to Oxidative DNA Damage in Neurons: Mechanisms and Disease. Neural Plast 2016; 2016:3619274. [PMID: 26942017 PMCID: PMC4752990 DOI: 10.1155/2016/3619274] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 12/24/2015] [Indexed: 11/26/2022] Open
Abstract
There is a growing body of evidence indicating that the mechanisms that control genome stability are of key importance in the development and function of the nervous system. The major threat for neurons is oxidative DNA damage, which is repaired by the base excision repair (BER) pathway. Functional mutations of enzymes that are involved in the processing of single-strand breaks (SSB) that are generated during BER have been causally associated with syndromes that present important neurological alterations and cognitive decline. In this review, the plasticity of BER during neurogenesis and the importance of an efficient BER for correct brain function will be specifically addressed paying particular attention to the brain region and neuron-selectivity in SSB repair-associated neurological syndromes and age-related neurodegenerative diseases.
Collapse
|
112
|
Luchtman D, Gollan R, Ellwardt E, Birkenstock J, Robohm K, Siffrin V, Zipp F. In vivo and in vitro effects of multiple sclerosis immunomodulatory therapeutics on glutamatergic excitotoxicity. J Neurochem 2016; 136:971-80. [PMID: 26662167 DOI: 10.1111/jnc.13456] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 11/23/2015] [Accepted: 11/24/2015] [Indexed: 11/27/2022]
Abstract
In multiple sclerosis (MS), a candidate downstream mechanism for neuronal injury is glutamate (Glu)-induced excitotoxicity, leading to toxic increases in intraneuronal Ca(2+) . Here, we used in vivo two-photon imaging in the brain of TN-XXL transgenic Ca(2+) reporter mice to test whether promising oral MS therapeutics, namely fingolimod, dimethyl fumarate, and their respective metabolites fingolimod-phosphate and monomethyl fumarate, can protect neurons against acute glutamatergic excitotoxic damage. We also assessed whether these drugs can protect against excitotoxicity in vitro using primary cortical neurons, and whether they can directly inhibit Glu release from pathogenic T-helper 17 lymphocytes. In vivo, direct and acute (1 h) administration of 100 mM Glu to the brainstem resulted in a rapid and significant up-regulation in neuronal Ca(2+) signaling as well as morphological excitotoxic changes that were attenuated by the NMDA-receptor antagonist MK801. Direct CNS administration of MS drugs prior to Glu significantly delayed or reduced, but did not prevent the neuronal Ca(2+) increase or morphological changes. In vitro, prolonged (24 h) treatment of primary neurons with the fumarates significantly protected against neurotoxicity induced by Glu as well as NMDA, similar to MK801. Furthermore, monomethyl fumerate significantly reduced Glu release from pathogenic T-helper 17 lymphocytes. Overall, these data suggest that MS drugs may mediate neuroprotection via excitotoxicity modulating effects. Evidence suggests MS pathogenesis may involve neuronal excitotoxicity, induced by local release of glutamate. However, current MS drugs, including dimethyl fumerate (DMF) and fingolimod (FTY720) are largely anti-inflammatory and not yet fully tested for their neuroprotective potential. Here, we show that the drugs, in particular DMF metabolite monomethyl fumerate (MMF), protect neurons by excitotoxicity modulating effects. Th17, T-helper 17.
Collapse
Affiliation(s)
- Dirk Luchtman
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn²), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - René Gollan
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn²), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Erik Ellwardt
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn²), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Jérôme Birkenstock
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn²), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Kerstin Robohm
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn²), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Volker Siffrin
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn²), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn²), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
113
|
Muneer A. The Neurobiology of Bipolar Disorder: An Integrated Approach. Chonnam Med J 2016; 52:18-37. [PMID: 26865997 PMCID: PMC4742607 DOI: 10.4068/cmj.2016.52.1.18] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 11/26/2015] [Accepted: 11/30/2015] [Indexed: 12/27/2022] Open
Abstract
Bipolar disorder is a heterogeneous condition with myriad clinical manifestations and many comorbidities leading to severe disabilities in the biopsychosocial realm. The objective of this review article was to underline recent advances in knowledge regarding the neurobiology of bipolar disorder. A further aim was to draw attention to new therapeutic targets in the treatment of bipolar disorder. To accomplish these goals, an electronic search was undertaken of the PubMed database in August 2015 of literature published during the last 10 years on the pathophysiology of bipolar disorder. A wide-ranging evaluation of the existing work was done with search terms such as "mood disorders and biology," "bipolar disorder and HPA axis," "bipolar disorder and cytokines," "mood disorders and circadian rhythm," "bipolar disorder and oxidative stress," etc. This endeavor showed that bipolar disorder is a diverse condition sharing neurobiological mechanisms with major depressive disorder and psychotic spectrum disorders. There is convincing evidence of crosstalk between different biological systems that act in a deleterious manner causing expression of the disease in genetically predisposed individuals. Inflammatory mediators act in concert with oxidative stress to dysregulate hormonal, metabolic, and circadian homeostasis in precipitating and perpetuating the illness. Stress, whether biologically or psychologically mediated, is responsible for the initiation and progression of the diathesis. Bipolar spectrum disorders have a strong genetic component; severe life stresses acting through various paths cause the illness phenotype.
Collapse
Affiliation(s)
- Ather Muneer
- Department of Psychiatry, Islamic International Medical College, Riphah International University, Rawalpindi, Pakistan
| |
Collapse
|
114
|
Green Tea Polyphenols Attenuated Glutamate Excitotoxicity via Antioxidative and Antiapoptotic Pathway in the Primary Cultured Cortical Neurons. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:2050435. [PMID: 26788243 PMCID: PMC4692997 DOI: 10.1155/2016/2050435] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/13/2015] [Indexed: 12/01/2022]
Abstract
Green tea polyphenols are a natural product which has antioxidative and antiapoptotic effects. It has been shown that glutamate excitotoxicity induced oxidative stress is linked to neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. In this study we explored the neuroprotective effect of green teen polyphenols against glutamate excitotoxicity in the primary cultured cortical neurons. We found that green tea polyphenols protected against glutamate induced neurotoxicity in the cortical neurons as measured by MTT and TUNEL assays. Green tea polyphenols were then showed to inhibit the glutamate induced ROS release and SOD activity reduction in the neurons. Furthermore, our results demonstrated that green tea polyphenols restored the dysfunction of mitochondrial pro- or antiapoptotic proteins Bax, Bcl-2, and caspase-3 caused by glutamate. Interestingly, the neuroprotective effect of green tea polyphenols was abrogated when the neurons were incubated with siBcl-2. Taken together, these results demonstrated that green tea polyphenols protected against glutamate excitotoxicity through antioxidative and antiapoptotic pathways.
Collapse
|
115
|
Clemens LE, Weber JJ, Wlodkowski TT, Yu-Taeger L, Michaud M, Calaminus C, Eckert SH, Gaca J, Weiss A, Magg JCD, Jansson EKH, Eckert GP, Pichler BJ, Bordet T, Pruss RM, Riess O, Nguyen HP. Olesoxime suppresses calpain activation and mutant huntingtin fragmentation in the BACHD rat. Brain 2015; 138:3632-53. [PMID: 26490331 DOI: 10.1093/brain/awv290] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 08/11/2015] [Indexed: 12/14/2022] Open
Abstract
Huntington's disease is a fatal human neurodegenerative disorder caused by a CAG repeat expansion in the HTT gene, which translates into a mutant huntingtin protein. A key event in the molecular pathogenesis of Huntington's disease is the proteolytic cleavage of mutant huntingtin, leading to the accumulation of toxic protein fragments. Mutant huntingtin cleavage has been linked to the overactivation of proteases due to mitochondrial dysfunction and calcium derangements. Here, we investigated the therapeutic potential of olesoxime, a mitochondria-targeting, neuroprotective compound, in the BACHD rat model of Huntington's disease. BACHD rats were treated with olesoxime via the food for 12 months. In vivo analysis covered motor impairments, cognitive deficits, mood disturbances and brain atrophy. Ex vivo analyses addressed olesoxime's effect on mutant huntingtin aggregation and cleavage, as well as brain mitochondria function. Olesoxime improved cognitive and psychiatric phenotypes, and ameliorated cortical thinning in the BACHD rat. The treatment reduced cerebral mutant huntingtin aggregates and nuclear accumulation. Further analysis revealed a cortex-specific overactivation of calpain in untreated BACHD rats. Treated BACHD rats instead showed significantly reduced levels of mutant huntingtin fragments due to the suppression of calpain-mediated cleavage. In addition, olesoxime reduced the amount of mutant huntingtin fragments associated with mitochondria, restored a respiration deficit, and enhanced the expression of fusion and outer-membrane transport proteins. In conclusion, we discovered the calpain proteolytic system, a key player in Huntington's disease and other neurodegenerative disorders, as a target of olesoxime. Our findings suggest that olesoxime exerts its beneficial effects by improving mitochondrial function, which results in reduced calpain activation. The observed alleviation of behavioural and neuropathological phenotypes encourages further investigations on the use of olesoxime as a therapeutic for Huntington's disease.
Collapse
Affiliation(s)
- Laura E Clemens
- 1 Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Calwerstrasse 7, 72076 Tuebingen, Germany 2 Centre for Rare Diseases, University of Tuebingen, Calwerstrasse 7, 72076 Tuebingen, Germany
| | - Jonasz J Weber
- 1 Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Calwerstrasse 7, 72076 Tuebingen, Germany 2 Centre for Rare Diseases, University of Tuebingen, Calwerstrasse 7, 72076 Tuebingen, Germany
| | - Tanja T Wlodkowski
- 1 Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Calwerstrasse 7, 72076 Tuebingen, Germany 2 Centre for Rare Diseases, University of Tuebingen, Calwerstrasse 7, 72076 Tuebingen, Germany
| | - Libo Yu-Taeger
- 1 Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Calwerstrasse 7, 72076 Tuebingen, Germany 2 Centre for Rare Diseases, University of Tuebingen, Calwerstrasse 7, 72076 Tuebingen, Germany
| | - Magali Michaud
- 3 Trophos SA., Parc Scientifique de Luminy Case 931, 13288 Marseille Cedex 9, France
| | - Carsten Calaminus
- 4 Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tuebingen, Roentgenweg 13, 72076 Tuebingen, Germany
| | - Schamim H Eckert
- 5 Department of Pharmacology, Goethe University Frankfurt am Main, Max-von-Laue Str. 9, 60438 Frankfurt, Germany
| | - Janett Gaca
- 5 Department of Pharmacology, Goethe University Frankfurt am Main, Max-von-Laue Str. 9, 60438 Frankfurt, Germany
| | - Andreas Weiss
- 6 Novartis Institutes for BioMedical Research, Klybeckstrasse 141, 4057 Basel, Switzerland
| | - Janine C D Magg
- 1 Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Calwerstrasse 7, 72076 Tuebingen, Germany 2 Centre for Rare Diseases, University of Tuebingen, Calwerstrasse 7, 72076 Tuebingen, Germany
| | - Erik K H Jansson
- 1 Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Calwerstrasse 7, 72076 Tuebingen, Germany 2 Centre for Rare Diseases, University of Tuebingen, Calwerstrasse 7, 72076 Tuebingen, Germany
| | - Gunter P Eckert
- 5 Department of Pharmacology, Goethe University Frankfurt am Main, Max-von-Laue Str. 9, 60438 Frankfurt, Germany
| | - Bernd J Pichler
- 4 Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tuebingen, Roentgenweg 13, 72076 Tuebingen, Germany
| | - Thierry Bordet
- 3 Trophos SA., Parc Scientifique de Luminy Case 931, 13288 Marseille Cedex 9, France
| | - Rebecca M Pruss
- 3 Trophos SA., Parc Scientifique de Luminy Case 931, 13288 Marseille Cedex 9, France
| | - Olaf Riess
- 1 Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Calwerstrasse 7, 72076 Tuebingen, Germany 2 Centre for Rare Diseases, University of Tuebingen, Calwerstrasse 7, 72076 Tuebingen, Germany
| | - Huu P Nguyen
- 1 Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Calwerstrasse 7, 72076 Tuebingen, Germany 2 Centre for Rare Diseases, University of Tuebingen, Calwerstrasse 7, 72076 Tuebingen, Germany
| |
Collapse
|
116
|
Bertholet AM, Delerue T, Millet AM, Moulis MF, David C, Daloyau M, Arnauné-Pelloquin L, Davezac N, Mils V, Miquel MC, Rojo M, Belenguer P. Mitochondrial fusion/fission dynamics in neurodegeneration and neuronal plasticity. Neurobiol Dis 2015; 90:3-19. [PMID: 26494254 DOI: 10.1016/j.nbd.2015.10.011] [Citation(s) in RCA: 255] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/16/2015] [Accepted: 10/13/2015] [Indexed: 12/17/2022] Open
Abstract
Mitochondria are dynamic organelles that continually move, fuse and divide. The dynamic balance of fusion and fission of mitochondria determines their morphology and allows their immediate adaptation to energetic needs, keeps mitochondria in good health by restoring or removing damaged organelles or precipitates cells in apoptosis in cases of severe defects. Mitochondrial fusion and fission are essential in mammals and their disturbances are associated with several diseases. However, while mitochondrial fusion/fission dynamics, and the proteins that control these processes, are ubiquitous, associated diseases are primarily neurological disorders. Accordingly, inactivation of the main actors of mitochondrial fusion/fission dynamics is associated with defects in neuronal development, plasticity and functioning, both ex vivo and in vivo. Here, we present the central actors of mitochondrial fusion and fission and review the role of mitochondrial dynamics in neuronal physiology and pathophysiology. Particular emphasis is placed on the three main actors of these processes i.e. DRP1,MFN1-2, and OPA1 as well as on GDAP1, a protein of the mitochondrial outer membrane preferentially expressed in neurons. This article is part of a Special Issue entitled: Mitochondria & Brain.
Collapse
Affiliation(s)
- A M Bertholet
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France
| | - T Delerue
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France
| | - A M Millet
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France
| | - M F Moulis
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France
| | - C David
- CNRS, Institut de Biochimie et Génétique Cellulaires (IBGC), UMR5095, Bordeaux, France; Université de Bordeaux, Institut de Biochimie et Génétique Cellulaires (IBGC), UMR5095, Bordeaux, France
| | - M Daloyau
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France
| | - L Arnauné-Pelloquin
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France
| | - N Davezac
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France
| | - V Mils
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France
| | - M C Miquel
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France
| | - M Rojo
- CNRS, Institut de Biochimie et Génétique Cellulaires (IBGC), UMR5095, Bordeaux, France; Université de Bordeaux, Institut de Biochimie et Génétique Cellulaires (IBGC), UMR5095, Bordeaux, France.
| | - P Belenguer
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France.
| |
Collapse
|
117
|
Ahmed N, Ronchi D, Comi GP. Genes and Pathways Involved in Adult Onset Disorders Featuring Muscle Mitochondrial DNA Instability. Int J Mol Sci 2015; 16:18054-76. [PMID: 26251896 PMCID: PMC4581235 DOI: 10.3390/ijms160818054] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 07/27/2015] [Accepted: 07/28/2015] [Indexed: 12/31/2022] Open
Abstract
Replication and maintenance of mtDNA entirely relies on a set of proteins encoded by the nuclear genome, which include members of the core replicative machinery, proteins involved in the homeostasis of mitochondrial dNTPs pools or deputed to the control of mitochondrial dynamics and morphology. Mutations in their coding genes have been observed in familial and sporadic forms of pediatric and adult-onset clinical phenotypes featuring mtDNA instability. The list of defects involved in these disorders has recently expanded, including mutations in the exo-/endo-nuclease flap-processing proteins MGME1 and DNA2, supporting the notion that an enzymatic DNA repair system actively takes place in mitochondria. The results obtained in the last few years acknowledge the contribution of next-generation sequencing methods in the identification of new disease loci in small groups of patients and even single probands. Although heterogeneous, these genes can be conveniently classified according to the pathway to which they belong. The definition of the molecular and biochemical features of these pathways might be helpful for fundamental knowledge of these disorders, to accelerate genetic diagnosis of patients and the development of rational therapies. In this review, we discuss the molecular findings disclosed in adult patients with muscle pathology hallmarked by mtDNA instability.
Collapse
Affiliation(s)
- Naghia Ahmed
- Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Dino Ferrari Centre, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, via Francesco Sforza 35, Milan 20122, Italy.
| | - Dario Ronchi
- Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Dino Ferrari Centre, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, via Francesco Sforza 35, Milan 20122, Italy.
| | - Giacomo Pietro Comi
- Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Dino Ferrari Centre, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, via Francesco Sforza 35, Milan 20122, Italy.
| |
Collapse
|
118
|
Marshall J, Wong KY, Rupasinghe CN, Tiwari R, Zhao X, Berberoglu ED, Sinkler C, Liu J, Lee I, Parang K, Spaller MR, Hüttemann M, Goebel DJ. Inhibition of N-Methyl-D-aspartate-induced Retinal Neuronal Death by Polyarginine Peptides Is Linked to the Attenuation of Stress-induced Hyperpolarization of the Inner Mitochondrial Membrane Potential. J Biol Chem 2015; 290:22030-48. [PMID: 26100636 DOI: 10.1074/jbc.m115.662791] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Indexed: 02/02/2023] Open
Abstract
It is widely accepted that overactivation of NMDA receptors, resulting in calcium overload and consequent mitochondrial dysfunction in retinal ganglion neurons, plays a significant role in promoting neurodegenerative disorders such as glaucoma. Calcium has been shown to initiate a transient hyperpolarization of the mitochondrial membrane potential triggering a burst of reactive oxygen species leading to apoptosis. Strategies that enhance cell survival signaling pathways aimed at preventing this adverse hyperpolarization of the mitochondrial membrane potential may provide a novel therapeutic intervention in retinal disease. In the retina, brain-derived neurotrophic factor has been shown to be neuroprotective, and our group previously reported a PSD-95/PDZ-binding cyclic peptide (CN2097) that augments brain-derived neurotrophic factor-induced pro-survival signaling. Here, we examined the neuroprotective properties of CN2097 using an established retinal in vivo NMDA toxicity model. CN2097 completely attenuated NMDA-induced caspase 3-dependent and -independent cell death and PARP-1 activation pathways, blocked necrosis, and fully prevented the loss of long term ganglion cell viability. Although neuroprotection was partially dependent upon CN2097 binding to the PDZ domain of PSD-95, our results show that the polyarginine-rich transport moiety C-R(7), linked to the PDZ-PSD-95-binding cyclic peptide, was sufficient to mediate short and long term protection via a mitochondrial targeting mechanism. C-R(7) localized to mitochondria and was found to reduce mitochondrial respiration, mitochondrial membrane hyperpolarization, and the generation of reactive oxygen species, promoting survival of retinal neurons.
Collapse
Affiliation(s)
- John Marshall
- From the Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, Rhode Island, 02912
| | - Kwoon Y Wong
- the Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan 48105
| | - Chamila N Rupasinghe
- the Geisel School of Medicine at Dartmouth, Department of Pharmacology and Toxicology, and Norris Cotton Cancer Center, Lebanon, New Hampshire 03756
| | - Rakesh Tiwari
- the Chapman University School of Pharmacy, Irvine, California 92618
| | - Xiwu Zhao
- the Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan 48105
| | | | - Christopher Sinkler
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan 48201, and
| | - Jenney Liu
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan 48201, and
| | - Icksoo Lee
- the College of Medicine, Dankook University, Cheonan-si, Chungcheongnam-do, 330-714, Republic of Korea
| | - Keykavous Parang
- the Chapman University School of Pharmacy, Irvine, California 92618
| | - Mark R Spaller
- the Geisel School of Medicine at Dartmouth, Department of Pharmacology and Toxicology, and Norris Cotton Cancer Center, Lebanon, New Hampshire 03756
| | - Maik Hüttemann
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan 48201, and
| | | |
Collapse
|
119
|
Sanderson TH, Raghunayakula S, Kumar R. Neuronal hypoxia disrupts mitochondrial fusion. Neuroscience 2015; 301:71-8. [PMID: 26049142 PMCID: PMC4504771 DOI: 10.1016/j.neuroscience.2015.05.078] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 05/07/2015] [Accepted: 05/29/2015] [Indexed: 01/04/2023]
Abstract
Brain ischemia/reperfusion injury results in death of vulnerable neurons and extensive brain damage. It is well known that mitochondrial release of cytochrome c (cyto c) is a hallmark of neuronal death, however the molecular events underlying this release are largely unknown. We tested the hypothesis that cyto c release is regulated by breakdown of the cristae architecture maintenance protein, optic atrophy 1 (OPA1), located in the inner mitochondrial membrane. We simulated ischemia/reperfusion in isolated primary rat neurons and interrogated OPA1 release from the mitochondria, OPA1 oligomeric breakdown, and concomitant dysfunction of mitochondrial dynamic state. We found that ischemia/reperfusion induces cyto c release and cell death that corresponds to multiple changes in OPA1, including: (i) translocation of the mitochondrial fusion protein OPA1 from the mitochondria to the cytosol, (ii) increase in the short isoform of OPA1, suggestive of proteolytic processing, (iii) breakdown of OPA1 oligomers in the mitochondria, and (iv) increased mitochondrial fission. Thus, we present novel evidence of a connection between release of cyto c from mitochondria and disruption of the mitochondrial fusion.
Collapse
Affiliation(s)
- T H Sanderson
- Department of Emergency Medicine, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI, USA; Cardiovascular Research Institute, Wayne State University School of Medicine, 421E. Canfield, Detroit, MI, USA
| | - S Raghunayakula
- Department of Emergency Medicine, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI, USA
| | - R Kumar
- Department of Emergency Medicine, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI, USA; Cardiovascular Research Institute, Wayne State University School of Medicine, 421E. Canfield, Detroit, MI, USA; Department of Physiology, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI, USA.
| |
Collapse
|
120
|
Rodriguez M, Rodriguez-Sabate C, Morales I, Sanchez A, Sabate M. Parkinson's disease as a result of aging. Aging Cell 2015; 14:293-308. [PMID: 25677794 PMCID: PMC4406659 DOI: 10.1111/acel.12312] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2014] [Indexed: 12/15/2022] Open
Abstract
It is generally considered that Parkinson's disease is induced by specific agents that degenerate a clearly defined population of dopaminergic neurons. Data commented in this review suggest that this assumption is not as clear as is often thought and that aging may be critical for Parkinson's disease. Neurons degenerating in Parkinson's disease also degenerate in normal aging, and the different agents involved in the etiology of this illness are also involved in aging. Senescence is a wider phenomenon affecting cells all over the body, whereas Parkinson's disease seems to be restricted to certain brain centers and cell populations. However, reviewed data suggest that Parkinson's disease may be a local expression of aging on cell populations which, by their characteristics (high number of synaptic terminals and mitochondria, unmyelinated axons, etc.), are highly vulnerable to the agents promoting aging. The development of new knowledge about Parkinson's disease could be accelerated if the research on aging and Parkinson's disease were planned together, and the perspective provided by gerontology gains relevance in this field.
Collapse
Affiliation(s)
- Manuel Rodriguez
- Laboratory of Neurobiology and Experimental Neurology, Department of Physiology, Faculty of Medicine, University of La LagunaLa Laguna, Spain
- Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED)La Laguna, Spain
| | - Clara Rodriguez-Sabate
- Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED)La Laguna, Spain
| | - Ingrid Morales
- Laboratory of Neurobiology and Experimental Neurology, Department of Physiology, Faculty of Medicine, University of La LagunaLa Laguna, Spain
- Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED)La Laguna, Spain
| | - Alberto Sanchez
- Laboratory of Neurobiology and Experimental Neurology, Department of Physiology, Faculty of Medicine, University of La LagunaLa Laguna, Spain
| | - Magdalena Sabate
- Rehabilitation Service, Department of Pharmacology and Physical Medicine, Faculty of Medicine, University of La LagunaLa Laguna, Spain
| |
Collapse
|
121
|
Chintala S, Cheng M, Zhang X. Decreased Expression of DREAM Promotes the Degeneration of Retinal Neurons. PLoS One 2015; 10:e0127776. [PMID: 26020793 PMCID: PMC4447264 DOI: 10.1371/journal.pone.0127776] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 04/18/2015] [Indexed: 12/27/2022] Open
Abstract
The intrinsic mechanisms that promote the degeneration of retinal ganglion cells (RGCs) following the activation of N-Methyl-D-aspartic acid-type glutamate receptors (NMDARs) are unclear. In this study, we have investigated the role of downstream regulatory element antagonist modulator (DREAM) in NMDA-mediated degeneration of the retina. NMDA, phosphate-buffered saline (PBS), and MK801 were injected into the vitreous humor of C57BL/6 mice. At 12, 24, and 48 hours after injection, expression of DREAM in the retina was determined by immunohistochemistry, western blot analysis, and electrophoretic mobility-shift assay (EMSA). Apoptotic death of cells in the retina was determined by terminal deoxynucleotidyl transferace dUTP nick end labeling (TUNEL) assays. Degeneration of RGCs in cross sections and in whole mount retinas was determined by using antibodies against Tuj1 and Brn3a respectively. Degeneration of amacrine cells and bipolar cells was determined by using antibodies against calretinin and protein kinase C (PKC)-alpha respectively. DREAM was expressed constitutively in RGCs, amacrine cells, bipolar cells, as well as in the inner plexiform layer (IPL). NMDA promoted a progressive decrease in DREAM levels in all three cell types over time, and at 48 h after NMDA-treatment very low DREAM levels were evident in the IPL only. DREAM expression in retinal nuclear proteins was decreased progressively after NMDA-treatment, and correlated with its decreased binding to the c-fos-DRE oligonucleotides. A decrease in DREAM expression correlated significantly with apoptotic death of RGCs, amacrine cells and bipolar cells. Treatment of eyes with NMDA antagonist MK801, restored DREAM expression to almost normal levels in the retina, and significantly decreased NMDA-mediated apoptotic death of RGCs, amacrine cells, and bipolar cells. Results presented in this study show for the first time that down-regulation of DREAM promotes the degeneration of RGCs, amacrine cells, and bipolar cells.
Collapse
Affiliation(s)
- Shravan Chintala
- Laboratory of Ophthalmic Neurobiology, and Eye Research Institute of Oakland University, Rochester, Michigan, United States of America
- * E-mail:
| | - Mei Cheng
- Laboratory of Ophthalmic Neurobiology, and Eye Research Institute of Oakland University, Rochester, Michigan, United States of America
| | - Xiao Zhang
- Laboratory of Ophthalmic Neurobiology, and Eye Research Institute of Oakland University, Rochester, Michigan, United States of America
| |
Collapse
|
122
|
Moghimi E, Solomon JA, Gianforcaro A, Hamadeh MJ. Dietary Vitamin D3 Restriction Exacerbates Disease Pathophysiology in the Spinal Cord of the G93A Mouse Model of Amyotrophic Lateral Sclerosis. PLoS One 2015; 10:e0126355. [PMID: 26020962 PMCID: PMC4447353 DOI: 10.1371/journal.pone.0126355] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 03/22/2015] [Indexed: 12/11/2022] Open
Abstract
Background Dietary vitamin D3 (D3) restriction reduces paw grip endurance and motor performance in G93A mice, and increases inflammation and apoptosis in the quadríceps of females. ALS, a neuromuscular disease, causes progressive degeneration of motor neurons in the brain and spinal cord. Objective We analyzed the spinal cords of G93A mice following dietary D3 restriction at 2.5% the adequate intake (AI) for oxidative damage (4-HNE, 3-NY), antioxidant enzymes (SOD2, catalase, GPx1), inflammation (TNF-α, IL-6, IL-10), apoptosis (bax/bcl-2 ratio, cleaved/pro-caspase 3 ratio), neurotrophic factor (GDNF) and neuron count (ChAT, SMI-36/SMI-32 ratio). Methods Beginning at age 25 d, 42 G93A mice were provided food ad libitum with either adequate (AI;1 IU D3/g feed; 12 M, 11 F) or deficient (DEF; 0.025 IU D3/g feed; 10 M, 9 F) D3. At age 113 d, the spinal cords were analyzed for protein content. Differences were considered significant at P ≤ 0.10, since this was a pilot study. Results DEF mice had 16% higher 4-HNE (P = 0.056), 12% higher GPx1 (P = 0.057) and 23% higher Bax/Bcl2 ratio (P = 0.076) vs. AI. DEF females had 29% higher GPx1 (P = 0.001) and 22% higher IL-6 (P = 0.077) vs. AI females. DEF males had 23% higher 4-HNE (P = 0.066) and 18% lower SOD2 (P = 0.034) vs. AI males. DEF males had 27% lower SOD2 (P = 0.004), 17% lower GPx1 (P = 0.070), 29% lower IL-6 (P = 0.023) and 22% lower ChAT (P = 0.082) vs. DEF females. Conclusion D3 deficiency exacerbates disease pathophysiology in the spinal cord of G93A mice, the exact mechanisms are sex-specific. This is in accord with our previous results in the quadriceps, as well as functional and disease outcomes.
Collapse
Affiliation(s)
- Elnaz Moghimi
- School of Kinesiology and Health Science, Faculty of Health, York University, Toronto, Ontario, Canada
- Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Jesse A. Solomon
- School of Kinesiology and Health Science, Faculty of Health, York University, Toronto, Ontario, Canada
- Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Alexandro Gianforcaro
- School of Kinesiology and Health Science, Faculty of Health, York University, Toronto, Ontario, Canada
- Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Mazen J. Hamadeh
- School of Kinesiology and Health Science, Faculty of Health, York University, Toronto, Ontario, Canada
- Muscle Health Research Centre, York University, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
123
|
Ju WK, Kim KY, Noh YH, Hoshijima M, Lukas TJ, Ellisman MH, Weinreb RN, Perkins GA. Increased mitochondrial fission and volume density by blocking glutamate excitotoxicity protect glaucomatous optic nerve head astrocytes. Glia 2015; 63:736-53. [PMID: 25557093 PMCID: PMC4373968 DOI: 10.1002/glia.22781] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 12/03/2014] [Indexed: 12/31/2022]
Abstract
Abnormal structure and function of astrocytes have been observed within the lamina cribrosa region of the optic nerve head (ONH) in glaucomatous neurodegeneration. Glutamate excitotoxicity-mediated mitochondrial alteration has been implicated in experimental glaucoma. However, the relationships among glutamate excitotoxicity, mitochondrial alteration and ONH astrocytes in the pathogenesis of glaucoma remain unknown. We found that functional N-methyl-d-aspartate (NMDA) receptors (NRs) are present in human ONH astrocytes and that glaucomatous human ONH astrocytes have increased expression levels of NRs and the glutamate aspartate transporter. Glaucomatous human ONH astrocytes exhibit mitochondrial fission that is linked to increased expression of dynamin-related protein 1 and its phosphorylation at Serine 616. In BAC ALDH1L1 eGFP or Thy1-CFP transgenic mice, NMDA treatment induced axon loss as well as hypertrophic morphology and mitochondrial fission in astrocytes of the glial lamina. In human ONH astrocytes, NMDA treatment in vitro triggered mitochondrial fission by decreasing mitochondrial length and number, thereby reducing mitochondrial volume density. However, blocking excitotoxicity by memantine (MEM) prevented these alterations by increasing mitochondrial length, number and volume density. In glaucomatous DBA/2J (D2) mice, blocking excitotoxicity by MEM inhibited the morphological alteration as well as increased mitochondrial number and volume density in astrocytes of the glial lamina. However, blocking excitotoxicity decreased autophagosome/autolysosome volume density in both astrocytes and axons in the glial lamina of glaucomatous D2 mice. These findings provide evidence that blocking excitotoxicity prevents ONH astrocyte dysfunction in glaucomatous neurodegeneration by increasing mitochondrial fission, increasing mitochondrial volume density and length, and decreasing autophagosome/autolysosome formation. GLIA 2015;63:736-753.
Collapse
Affiliation(s)
- Won-Kyu Ju
- Department of Ophthalmology, Laboratory for Optic Nerve Biology, Hamilton Glaucoma Center, University of California San DiegoLa Jolla, California
| | - Keun-Young Kim
- Center for Research on Biological Systems, National Center for Microscopy and Imaging Research and Department of Neuroscience, University of California San DiegoLa Jolla, California
| | - You Hyun Noh
- Department of Ophthalmology, Laboratory for Optic Nerve Biology, Hamilton Glaucoma Center, University of California San DiegoLa Jolla, California
| | - Masahiko Hoshijima
- Center for Research on Biological Systems, National Center for Microscopy and Imaging Research and Department of Neuroscience, University of California San DiegoLa Jolla, California
- Department of Medicine, University of California San DiegoLa Jolla, California
| | - Thomas J Lukas
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern UniversityChicago, Illinois
| | - Mark H Ellisman
- Center for Research on Biological Systems, National Center for Microscopy and Imaging Research and Department of Neuroscience, University of California San DiegoLa Jolla, California
| | - Robert N Weinreb
- Department of Ophthalmology, Laboratory for Optic Nerve Biology, Hamilton Glaucoma Center, University of California San DiegoLa Jolla, California
| | - Guy A Perkins
- Center for Research on Biological Systems, National Center for Microscopy and Imaging Research and Department of Neuroscience, University of California San DiegoLa Jolla, California
| |
Collapse
|
124
|
Iommarini L, Peralta S, Torraco A, Diaz F. Mitochondrial Diseases Part II: Mouse models of OXPHOS deficiencies caused by defects in regulatory factors and other components required for mitochondrial function. Mitochondrion 2015; 22:96-118. [PMID: 25640959 DOI: 10.1016/j.mito.2015.01.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 01/22/2015] [Indexed: 01/21/2023]
Abstract
Mitochondrial disorders are defined as defects that affect the oxidative phosphorylation system (OXPHOS). They are characterized by a heterogeneous array of clinical presentations due in part to a wide variety of factors required for proper function of the components of the OXPHOS system. There is no cure for these disorders owing to our poor knowledge of the pathogenic mechanisms of disease. To understand the mechanisms of human disease numerous mouse models have been developed in recent years. Here we summarize the features of several mouse models of mitochondrial diseases directly related to those factors affecting mtDNA maintenance, replication, transcription, translation as well as other proteins that are involved in mitochondrial dynamics and quality control which affect mitochondrial OXPHOS function without being intrinsic components of the system. We discuss how these models have contributed to our understanding of mitochondrial diseases and their pathogenic mechanisms.
Collapse
Affiliation(s)
- Luisa Iommarini
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, Via Irnerio 42, 40128 Bologna, Italy.
| | - Susana Peralta
- Department of Neurology, University of Miami, Miller School of Medicine, Miami, FL 33136, USA.
| | - Alessandra Torraco
- Unit for Neuromuscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesù Children's Hospital, IRCCS, Viale di San Paolo, 15 - 00146, Rome, Italy.
| | - Francisca Diaz
- Department of Neurology, University of Miami, Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
125
|
Cooke Bailey JN, Yaspan BL, Pasquale LR, Hauser MA, Kang JH, Loomis SJ, Brilliant M, Budenz DL, Christen WG, Fingert J, Gaasterland D, Gaasterland T, Kraft P, Lee RK, Lichter PR, Liu Y, McCarty CA, Moroi SE, Richards JE, Realini T, Schuman JS, Scott WK, Singh K, Sit AJ, Vollrath D, Wollstein G, Zack DJ, Zhang K, Pericak-Vance MA, Allingham RR, Weinreb RN, Haines JL, Wiggs JL. Hypothesis-independent pathway analysis implicates GABA and acetyl-CoA metabolism in primary open-angle glaucoma and normal-pressure glaucoma. Hum Genet 2014; 133:1319-30. [PMID: 25037249 PMCID: PMC4273559 DOI: 10.1007/s00439-014-1468-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 07/08/2014] [Indexed: 12/15/2022]
Abstract
Primary open-angle glaucoma (POAG) is a leading cause of blindness worldwide. Using genome-wide association single-nucleotide polymorphism data from the Glaucoma Genes and Environment study and National Eye Institute Glaucoma Human Genetics Collaboration comprising 3,108 cases and 3,430 controls, we assessed biologic pathways as annotated in the KEGG database for association with risk of POAG. After correction for genic overlap among pathways, we found 4 pathways, butanoate metabolism (hsa00650), hematopoietic cell lineage (hsa04640), lysine degradation (hsa00310) and basal transcription factors (hsa03022) related to POAG with permuted p < 0.001. In addition, the human leukocyte antigen (HLA) gene family was significantly associated with POAG (p < 0.001). In the POAG subset with normal-pressure glaucoma (NPG), the butanoate metabolism pathway was also significantly associated (p < 0.001) as well as the MAPK and Hedgehog signaling pathways (hsa04010 and hsa04340), glycosaminoglycan biosynthesis-heparan sulfate pathway (hsa00534) and the phenylalanine, tyrosine and tryptophan biosynthesis pathway (hsa0400). The butanoate metabolism pathway overall, and specifically the aspects of the pathway that contribute to GABA and acetyl-CoA metabolism, was the only pathway significantly associated with both POAG and NPG. Collectively these results implicate GABA and acetyl-CoA metabolism in glaucoma pathogenesis, and suggest new potential therapeutic targets.
Collapse
Affiliation(s)
| | - Brian L. Yaspan
- Center for Human Genetics Research, Vanderbilt University, Nashville, TN, USA
| | - Louis R. Pasquale
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael A. Hauser
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, USA
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Jae H. Kang
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Stephanie J. Loomis
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Murray Brilliant
- Center for Human Genetics, Marshfield Clinic Research Foundation, Marshfield, WI, USA
| | - Donald L. Budenz
- Department of Ophthalmology, University of North Carolina, Chapel Hill, NC, USA
| | | | - John Fingert
- Department of Ophthalmology, College of Medicine, University of Iowa, Iowa City, IO, USA
- Department of A natomy/Cell Biology, College of Medicine, University of Iowa, Iowa City, IO, USA
| | | | - Terry Gaasterland
- Scripps Genome Center, University of California at San Diego, San Diego, CA, USA
| | - Peter Kraft
- Department of Biostatistics, Harvard School of Public Health, Boston, MA, USA
| | - Richard K. Lee
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Paul R. Lichter
- Department of Ophthalmology and V isual Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Yutao Liu
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, USA
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | | | - Sayoko E. Moroi
- Department of Ophthalmology and V isual Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Julia E. Richards
- Department of Ophthalmology and V isual Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Tony Realini
- Department of Ophthalmology, WVU Eye Institute, Morgantown, WV, USA
| | - Joel S. Schuman
- Department of Ophthalmology, UPMC Eye Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - William K. Scott
- Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kuldev Singh
- Department of Ophthalmology, Stanford University, Palo Alto, CA, USA
| | - Arthur J. Sit
- Department of Ophthalmology, Mayo Clinic, Rochester, MN, USA
| | | | - Gadi Wollstein
- Department of Ophthalmology, UPMC Eye Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Donald J. Zack
- Wilmer Eye Institute, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Kang Zhang
- Department of Ophthalmology, Hamilton Eye Center, University of California, San Diego, CA, USA
| | | | - R. Rand Allingham
- Department of Ophthalmology, Duke University Medical Center, Durham, NC, USA
| | - Robert N. Weinreb
- Department of Ophthalmology, Hamilton Eye Center, University of California, San Diego, CA, USA
| | - Jonathan L. Haines
- Center for Human Genetics Research, Vanderbilt University, Nashville, TN, USA
| | - Janey L. Wiggs
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA, USA
| |
Collapse
|
126
|
Golomb BA, Allison M, Koperski S, Koslik HJ, Devaraj S, Ritchie JB. Coenzyme Q10 benefits symptoms in Gulf War veterans: results of a randomized double-blind study. Neural Comput 2014; 26:2594-651. [PMID: 25149705 DOI: 10.1162/neco_a_00659] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
We sought to assess whether coenzyme Q10 (CoQ10) benefits the chronic multisymptom problems that affect one-quarter to one-third of 1990-1 Gulf War veterans, using a randomized, double-blind, placebo-controlled study. Participants were 46 veterans meeting Kansas and Centers for Disease Control criteria for Gulf War illness. Intervention was PharmaNord (Denmark) CoQ10 100 mg per day (Q100), 300 mg per day (Q300), or an identical-appearing placebo for 3.5 ± 0.5 months. General self-rated health (GSRH), the primary outcome, differed across randomization arms at baseline, and sex significantly predicted GSRH change, compelling adjustment for baseline GSRH and prompting sex-stratified analysis. GSRH showed no significant benefit in the combined-sex sample. Among males (85% of participants), Q100 significantly benefited GSRH versus placebo and versus Q300, providing emphasis on Q100. Physical function (summary performance score, SPS) improved on Q100 versus placebo. A rise in CoQ10 approached significance as a predictor of improvement in GSRH and significantly predicted SPS improvement. Among 20 symptoms each present in half or more of the enrolled veterans, direction-of-difference on Q100 versus placebo was favorable for all except sleep problems; sign test 19:1, p=0.00004) with several symptoms individually significant. Significance for these symptoms despite the small sample underscores large effect sizes, and an apparent relation of key outcomes to CoQ10 change increases prospects for causality. In conclusion, Q100 conferred benefit to physical function and symptoms in veterans with Gulf War illness. Examination in a larger sample is warranted, and findings from this study can inform the conduct of a larger trial.
Collapse
Affiliation(s)
- Beatrice A Golomb
- Departments of Medicine and of Family and Preventive Medicine, University of California, San Diego, La Jolla, CA 92093, U.S.A.
| | | | | | | | | | | |
Collapse
|
127
|
Kiang AS, Humphries MM, Campbell M, Humphries P. Antioxidant therapy for retinal disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 801:783-9. [PMID: 24664771 DOI: 10.1007/978-1-4614-3209-8_98] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Disease mechanisms associated with retinal disease are of immense complexity, mutations within 45 genes having been implicated, for example, in retinitis pigmentosa, while interplay between genetic, environmental, and demographic factors can lead to diabetic retinopathy, age-related macular degeneration, and glaucoma. In light of such diversity, any therapeutic modality that can be targeted to an early molecular process instrumental in multiple forms of disease, such as oxidative stress, holds much attraction. Here, we provide a brief overview of a selection of compounds displaying antioxidant activity, which have been shown to slow down degeneration of retinal tissues and highlight suggested modes of action.
Collapse
Affiliation(s)
- Anna-Sophia Kiang
- Ocular Genetics Unit, School of Genetics and Microbiology, Trinity College Dublin, Dublin 2, Ireland,
| | | | | | | |
Collapse
|
128
|
Yu W, Bonnet M, Farso M, Ma K, Chabot JG, Martin E, Torriglia A, Guan Z, McLaurin J, Quirion R, Krantic S. The expression of apoptosis inducing factor (AIF) is associated with aging-related cell death in the cortex but not in the hippocampus in the TgCRND8 mouse model of Alzheimer's disease. BMC Neurosci 2014; 15:73. [PMID: 24915960 PMCID: PMC4070095 DOI: 10.1186/1471-2202-15-73] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 05/30/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Recent evidence has suggested that Alzheimer's disease (AD)-associated neuronal loss may occur via the caspase-independent route of programmed cell death (PCD) in addition to caspase-dependent mechanisms. However, the brain region specificity of caspase-independent PCD in AD-associated neurodegeneration is unknown. We therefore used the transgenic CRND8 (TgCRND8) AD mouse model to explore whether the apoptosis inducing factor (AIF), a key mediator of caspase-independent PCD, contributes to cell loss in selected brain regions in the course of aging. RESULTS Increased expression of truncated AIF (tAIF), which is directly responsible for cell death induction, was observed at both 4- and 6-months of age in the cortex. Concomitant with the up-regulation of tAIF was an increase in the nuclear translocation of this protein. Heightened tAIF expression or translocation was not observed in the hippocampus or cerebellum, which were used as AD-vulnerable and relatively AD-spared regions, respectively. The cortical alterations in tAIF levels were accompanied by increased Bax expression and mitochondrial translocation. This effect was preceded by a significant reduction in ATP content and an increase in reactive oxygen species (ROS) production, detectable at 2 months of age despite negligible amounts of amyloid-beta peptides (Aβ). CONCLUSIONS Taken together, these data suggest that AIF is likely to play a region-specific role in AD-related caspase-independent PCD, which is consistent with aging-associated mitochondrial impairment and oxidative stress.
Collapse
Affiliation(s)
- Wenfeng Yu
- Key laboratory of Molecular Biology, Guiyang Medical University, Guiyang 550004, China
- Department of Psychiatry, Douglas Mental Health University Institute (DMHUI), McGill University, Verdun Montréal, Québec H4H 1R3, Canada
| | - Mathilde Bonnet
- Department of Psychiatry, Douglas Mental Health University Institute (DMHUI), McGill University, Verdun Montréal, Québec H4H 1R3, Canada
| | - Mark Farso
- Department of Psychiatry, Douglas Mental Health University Institute (DMHUI), McGill University, Verdun Montréal, Québec H4H 1R3, Canada
| | - Keran Ma
- Department Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Jean-Guy Chabot
- Department of Psychiatry, Douglas Mental Health University Institute (DMHUI), McGill University, Verdun Montréal, Québec H4H 1R3, Canada
| | | | | | - Zhizhong Guan
- Key laboratory of Molecular Biology, Guiyang Medical University, Guiyang 550004, China
- Department of Pathology in the Affiliated Hospital of Guiyang Medical University, Guiyang 550004, China
| | - JoAnne McLaurin
- Department Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Rémi Quirion
- Department of Psychiatry, Douglas Mental Health University Institute (DMHUI), McGill University, Verdun Montréal, Québec H4H 1R3, Canada
| | - Slavica Krantic
- Department of Psychiatry, Douglas Mental Health University Institute (DMHUI), McGill University, Verdun Montréal, Québec H4H 1R3, Canada
- Centre de Recherche des Cordeliers, UMRS872, Paris, France
| |
Collapse
|
129
|
The db/db mouse: a useful model for the study of diabetic retinal neurodegeneration. PLoS One 2014; 9:e97302. [PMID: 24837086 PMCID: PMC4023966 DOI: 10.1371/journal.pone.0097302] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 04/17/2014] [Indexed: 01/20/2023] Open
Abstract
Background To characterize the sequential events that are taking place in retinal neurodegeneration in a murine model of spontaneous type 2 diabetes (db/db mouse). Methods C57BLKsJ-db/db mice were used as spontaneous type 2 diabetic animal model, and C57BLKsJ-db/+ mice served as the control group. To assess the chronological sequence of the abnormalities the analysis was performed at different ages (8, 16 and 24 weeks). The retinas were evaluated in terms of morphological and functional abnormalities [electroretinography (ERG)]. Histological markers of neurodegeneration (glial activation and apoptosis) were evaluated by immunohistochemistry. In addition glutamate levels and glutamate/aspartate transporter (GLAST) expression were assessed. Furthermore, to define gene expression changes associated with early diabetic retinopathy a transcriptome analyses was performed at 8 week. Furthermore, an additional interventional study to lower blood glucose levels was performed. Results Glial activation was higher in diabetic than in non diabetic mice in all the stages (p<0.01). In addition, a progressive loss of ganglion cells and a significant reduction of neuroretinal thickness were also observed in diabetic mice. All these histological hallmarks of neurodegeneration were less pronounced at week 8 than at week 16 and 24. Significant ERG abnormalities were present in diabetic mice at weeks 16 and 24 but not at week 8. Moreover, we observed a progressive accumulation of glutamate in diabetic mice associated with an early downregulation of GLAST. Morphological and ERG abnormalities were abrogated by lowering blood glucose levels. Finally, a dysregulation of several genes related to neurotransmission and oxidative stress such as UCP2 were found at week 8. Conclusions Our results suggest that db/db mouse reproduce the features of the neurodegenerative process that occurs in the human diabetic eye. Therefore, it seems an appropriate model for investigating the underlying mechanisms of diabetes-induced retinal neurodegeneration and for testing neuroprotective drugs.
Collapse
|
130
|
Givvimani S, Pushpakumar S, Veeranki S, Tyagi SC. Dysregulation of Mfn2 and Drp-1 proteins in heart failure. Can J Physiol Pharmacol 2014; 92:583-91. [PMID: 24905188 DOI: 10.1139/cjpp-2014-0060] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Therapeutic approaches for cardiac regenerative mechanisms have been explored over the past decade to target various cardiovascular diseases (CVD). Structural and functional aberrations of mitochondria have been observed in CVD. The significance of mitochondrial maturation and function in cardiomyocytes is distinguished by their attribution to embryonic stem cell differentiation into adult cardiomyocytes. An abnormal fission process has been implicated in heart failure, and treatment with mitochondrial division inhibitor 1 (Mdivi-1), a specific inhibitor of dynamin related protein-1 (Drp-1), has been shown to improve cardiac function. We recently observed that the ratio of mitofusin 2 (Mfn2; a fusion protein) and Drp-1 (a fission protein) was decreased during heart failure, suggesting increased mitophagy. Treatment with Mdivi-1 improved cardiac function by normalizing this ratio. Aberrant mitophagy and enhanced oxidative stress in the mitochondria contribute to abnormal activation of MMP-9, leading to degradation of the important gap junction protein connexin-43 (Cx-43) in the ventricular myocardium. Reduced Cx-43 levels were associated with increased fibrosis and ventricular dysfunction in heart failure. Treatment with Mdivi-1 restored MMP-9 and Cx-43 expression towards normal. In this review, we discuss mitochondrial dynamics, its relation to MMP-9 and Cx-43, and the therapeutic role of fission inhibition in heart failure.
Collapse
Affiliation(s)
- Srikanth Givvimani
- Department of Physiology & Biophysics, School of Medicine, University of Louisville, KY 40202, USA
| | | | | | | |
Collapse
|
131
|
Lee D, Shim MS, Kim KY, Noh YH, Kim H, Kim SY, Weinreb RN, Ju WK. Coenzyme Q10 inhibits glutamate excitotoxicity and oxidative stress-mediated mitochondrial alteration in a mouse model of glaucoma. Invest Ophthalmol Vis Sci 2014; 55:993-1005. [PMID: 24458150 DOI: 10.1167/iovs.13-12564] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE To test whether a diet supplemented with coenzyme Q10 (CoQ10) ameliorates glutamate excitotoxicity and oxidative stress-mediated retinal ganglion cell (RGC) degeneration by preventing mitochondrial alterations in the retina of glaucomatous DBA/2J mice. METHODS Preglaucomatous DBA/2J and age-matched control DBA/2J-Gpnmb(+) mice were fed with CoQ10 (1%) or a control diet daily for 6 months. The RGC survival and axon preservation were measured by Brn3a and neurofilament immunohistochemistry and by conventional transmission electron microscopy. Glial fibrillary acidic protein (GFAP), superoxide dismutase-2 (SOD2), heme oxygenase-1 (HO1), N-methyl-d-aspartate receptor (NR) 1 and 2A, and Bax and phosphorylated Bad (pBad) protein expression was measured by Western blot analysis. Apoptotic cell death was assessed by TUNEL staining. Mitochondrial DNA (mtDNA) content and mitochondrial transcription factor A (Tfam)/oxidative phosphorylation (OXPHOS) complex IV protein expression were measured by real-time PCR and Western blot analysis. RESULTS Coenzyme Q10 promoted RGC survival by approximately 29% and preserved the axons in the optic nerve head (ONH), as well as inhibited astroglial activation by decreasing GFAP expression in the retina and ONH of glaucomatous DBA/2J mice. Intriguingly, CoQ10 significantly blocked the upregulation of NR1 and NR2A, as well as of SOD2 and HO1 protein expression in the retina of glaucomatous DBA/2J mice. In addition, CoQ10 significantly prevented apoptotic cell death by decreasing Bax protein expression or by increasing pBad protein expression. More importantly, CoQ10 preserved mtDNA content and Tfam/OXPHOS complex IV protein expression in the retina of glaucomatous DBA/2J mice. CONCLUSIONS Our findings suggest that CoQ10 may be a promising therapeutic strategy for ameliorating glutamate excitotoxicity and oxidative stress in glaucomatous neurodegeneration.
Collapse
Affiliation(s)
- Dongwook Lee
- Laboratory for Optic Nerve Biology, Hamilton Glaucoma Center and Department of Ophthalmology, University of California, San Diego, La Jolla, California
| | | | | | | | | | | | | | | |
Collapse
|
132
|
Early onset Alzheimer's disease and oxidative stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:375968. [PMID: 24669286 PMCID: PMC3942075 DOI: 10.1155/2014/375968] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 11/18/2013] [Indexed: 01/30/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in elderly adults. It is estimated that 10% of the world's population aged more than 60-65 years could currently be affected by AD, and that in the next 20 years, there could be more than 30 million people affected by this pathology. One of the great challenges in this regard is that AD is not just a scientific problem; it is associated with major psychosocial and ethical dilemmas and has a negative impact on national economies. The neurodegenerative process that occurs in AD involves a specific nervous cell dysfunction, which leads to neuronal death. Mutations in APP, PS1, and PS2 genes are causes for early onset AD. Several animal models have demonstrated that alterations in these proteins are able to induce oxidative damage, which in turn favors the development of AD. This paper provides a review of many, although not all, of the mutations present in patients with familial Alzheimer's disease and the association between some of these mutations with both oxidative damage and the development of the pathology.
Collapse
|
133
|
Ambrosi G, Cerri S, Blandini F. A further update on the role of excitotoxicity in the pathogenesis of Parkinson’s disease. J Neural Transm (Vienna) 2014; 121:849-59. [DOI: 10.1007/s00702-013-1149-z] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 12/19/2013] [Indexed: 11/30/2022]
|
134
|
Ramachandran A, McGill MR, Xie Y, Ni HM, Ding WX, Jaeschke H. Receptor interacting protein kinase 3 is a critical early mediator of acetaminophen-induced hepatocyte necrosis in mice. Hepatology 2013; 58:2099-108. [PMID: 23744808 PMCID: PMC3791212 DOI: 10.1002/hep.26547] [Citation(s) in RCA: 204] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2012] [Accepted: 05/20/2013] [Indexed: 12/13/2022]
Abstract
UNLABELLED Acetaminophen (APAP) overdose is a major cause of hepatotoxicity and acute liver failure in the U.S., but the pathophysiology is incompletely understood. Despite evidence for apoptotic signaling, hepatic cell death after APAP is generally considered necrotic in mice and in humans. Recent findings suggest that the receptor interacting protein kinase 3 (RIP3) acts as a switch from apoptosis to necrosis (programmed necrosis). Thus, the aim of the current investigation was to determine if RIP3 is involved in APAP-induced liver cell death. APAP (200-300 mg/kg) caused glutathione depletion and protein adduct formation, oxidant stress, mitochondrial release of apoptosis inducing factor, and nuclear DNA fragmentation resulting in centrilobular necrosis in C57Bl/6J mice. Inhibiting RIP3 protein induction with antisense morpholinos in wild-type animals or using RIP3-deficient mice had no effect on protein adduct formation but attenuated all other parameters, including necrotic cell death, at 6 hours after APAP. In addition, cultured hepatocytes from RIP3-deficient mice showed reduced injury compared to wild-type cells after 24 hours. Interestingly, APAP-induced mitochondrial translocation of dynamin-related protein 1 (Drp1), the initiator of mitochondrial fission, was inhibited by reduced RIP3 protein expression and the Drp1 inhibitor MDIVI reduced APAP-induced cell death at 24 hours. All of these protective effects were lost after 24 hours in vivo or 48 hours in vitro. CONCLUSION RIP3 is an early mediator of APAP hepatotoxicity, involving modulation of mitochondrial dysfunction and oxidant stress. Controlling RIP3 expression could be a promising new approach to reduce APAP-induced liver injury, but requires complementary strategies to control mitochondrial dysfunction for long-term protection.
Collapse
Affiliation(s)
- Anup Ramachandran
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS
| | | | | | | | | | | |
Collapse
|
135
|
Alavi MV, Fuhrmann N. Dominant optic atrophy, OPA1, and mitochondrial quality control: understanding mitochondrial network dynamics. Mol Neurodegener 2013; 8:32. [PMID: 24067127 PMCID: PMC3856479 DOI: 10.1186/1750-1326-8-32] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 09/16/2013] [Indexed: 12/13/2022] Open
Abstract
Mitochondrial quality control is fundamental to all neurodegenerative diseases, including the most prominent ones, Alzheimer’s Disease and Parkinsonism. It is accomplished by mitochondrial network dynamics – continuous fission and fusion of mitochondria. Mitochondrial fission is facilitated by DRP1, while MFN1 and MFN2 on the mitochondrial outer membrane and OPA1 on the mitochondrial inner membrane are essential for mitochondrial fusion. Mitochondrial network dynamics are regulated in highly sophisticated ways by various different posttranslational modifications, such as phosphorylation, ubiquitination, and proteolytic processing of their key-proteins. By this, mitochondria process a wide range of different intracellular and extracellular parameters in order to adapt mitochondrial function to actual energetic and metabolic demands of the host cell, attenuate mitochondrial damage, recycle dysfunctional mitochondria via the mitochondrial autophagy pathway, or arrange for the recycling of the complete host cell by apoptosis. Most of the genes coding for proteins involved in this process have been associated with neurodegenerative diseases. Mutations in one of these genes are associated with a neurodegenerative disease that originally was described to affect retinal ganglion cells only. Since more and more evidence shows that other cell types are affected as well, we would like to discuss the pathology of dominant optic atrophy, which is caused by heterozygous sequence variants in OPA1, in the light of the current view on OPA1 protein function in mitochondrial quality control, in particular on its function in mitochondrial fusion and cytochrome C release. We think OPA1 is a good example to understand the molecular basis for mitochondrial network dynamics.
Collapse
Affiliation(s)
- Marcel V Alavi
- Department of Ophthalmology, University of California, San Francisco, 10 Koret Way, 94143-0730 San Francisco, CA, USA.
| | | |
Collapse
|
136
|
Dapper JD, Crish SD, Pang IH, Calkins DJ. Proximal inhibition of p38 MAPK stress signaling prevents distal axonopathy. Neurobiol Dis 2013; 59:26-37. [PMID: 23859799 DOI: 10.1016/j.nbd.2013.07.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 07/04/2013] [Indexed: 12/21/2022] Open
Abstract
The p38 mitogen-activated protein kinase (MAPK) isoforms are phosphorylated by a variety of stress stimuli in neurodegenerative disease and act as upstream activators of myriad pathogenic processes. Thus, p38 MAPK inhibitors are of growing interest as possible therapeutic interventions. Axonal dysfunction is an early component of most neurodegenerative disorders, including the most prevalent optic neuropathy, glaucoma. Sensitivity to intraocular pressure at an early stage disrupts anterograde transport along retinal ganglion cell (RGC) axons to projection targets in the brain with subsequent degeneration of the axons themselves; RGC body loss is much later. Here we show that elevated ocular pressure in rats increases p38 MAPK activation in retina, especially in RGC bodies. Topical eye-drop application of a potent and selective inhibitor of the p38 MAPK catalytic domain (Ro3206145) prevented both the degradation of anterograde transport to the brain and degeneration of axons in the optic nerve. Ro3206145 reduced in the retina phosphorylation of tau and heat-shock protein 27, both down-stream targets of p38 MAPK activation implicated in glaucoma, as well as expression of two inflammatory responses. We also observed increased p38 MAPK activation in mouse models. Thus, inhibition of p38 MAPK signaling in the retina may represent a therapeutic target for preventing early pathogenesis in optic neuropathies.
Collapse
Affiliation(s)
- Jason D Dapper
- The Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | | | | | | |
Collapse
|
137
|
Kwan P. Sarcopenia: the gliogenic perspective. Mech Ageing Dev 2013; 134:349-55. [PMID: 23831860 DOI: 10.1016/j.mad.2013.06.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Revised: 06/20/2013] [Accepted: 06/22/2013] [Indexed: 12/15/2022]
Abstract
It has been approximately 25 years since Dr. Rosenberg first brought attention to sarcopenia. To date, this aging-associated condition is recognized as a chronic loss of muscle mass and is usually accompanied by dynapenia. Despite its poly-etiological factors, sarcopenia has a strong neurogenic component underlying this chrono-degeneration of muscle mass, as shown in recent studies. As it seems plausible to explain the origin of sarcopenia through a motor neuron degeneration model, the focus of sarcopenia research should combine neuroscience with the study of the original myocyte and satellite cells. Although a complete mechanism underlying the development of sarcopenia has yet to be elucidated, we propose that the primary trigger of sarcopenia could be gliogenic in origin based on the close relationship between the glia, neurons and non-neural cells, for example, the motor unit and its associated glia in both the central nervous system (CNS) and the peripheral nervous system (PNS). In addition to muscle cells, both of the neural cells are affected by aging.
Collapse
Affiliation(s)
- Ping Kwan
- ST013a, Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hong Kong.
| |
Collapse
|
138
|
Guo BL, Sui BD, Wang XY, Wei YY, Huang J, Chen J, Wu SX, Li YQ, Wang YY, Yang YL. Significant changes in mitochondrial distribution in different pain models of mice. Mitochondrion 2013; 13:292-7. [DOI: 10.1016/j.mito.2013.03.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 02/23/2013] [Accepted: 03/19/2013] [Indexed: 11/25/2022]
|
139
|
Mitochondrial fusion proteins and human diseases. Neurol Res Int 2013; 2013:293893. [PMID: 23781337 PMCID: PMC3678461 DOI: 10.1155/2013/293893] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Revised: 04/28/2013] [Accepted: 05/01/2013] [Indexed: 01/19/2023] Open
Abstract
Mitochondria are highly dynamic, complex organelles that continuously alter their shape, ranging between two opposite processes, fission and fusion, in response to several stimuli and the metabolic demands of the cell. Alterations in mitochondrial dynamics due to mutations in proteins involved in the fusion-fission machinery represent an important pathogenic mechanism of human diseases. The most relevant proteins involved in the mitochondrial fusion process are three GTPase dynamin-like proteins: mitofusin 1 (MFN1) and 2 (MFN2), located in the outer mitochondrial membrane, and optic atrophy protein 1 (OPA1), in the inner membrane. An expanding number of degenerative disorders are associated with mutations in the genes encoding MFN2 and OPA1, including Charcot-Marie-Tooth disease type 2A and autosomal dominant optic atrophy. While these disorders can still be considered rare, defective mitochondrial dynamics seem to play a significant role in the molecular and cellular pathogenesis of more common neurodegenerative diseases, for example, Alzheimer's and Parkinson's diseases. This review provides an overview of the basic molecular mechanisms involved in mitochondrial fusion and focuses on the alteration in mitochondrial DNA amount resulting from impairment of mitochondrial dynamics. We also review the literature describing the main disorders associated with the disruption of mitochondrial fusion.
Collapse
|
140
|
Celojevic D, Nilsson S, Kalaboukhova L, Tasa G, Juronen E, Sjölander A, Zetterberg H, Zetterberg M. Genetic Variation of Superoxide Dismutases in Patients with Primary Open-angle Glaucoma. Ophthalmic Genet 2013; 35:79-84. [DOI: 10.3109/13816810.2013.793364] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
141
|
Abstract
Dexras1, a small G-protein localized predominantly to the brain, is transcriptionally upregulated by the synthetic glucocorticoid dexamethasone. It has close homology to the Ras subfamily but differs in that Dexras1 contains an extended 7 kDa C-terminal tail. Previous studies in our laboratory showed that NMDA receptor activation, via NO and Dexras1, physiologically stimulates DMT1, the major iron importer. A membrane-permeable iron chelator substantially reduces NMDA excitotoxicity, suggesting that Dexras1-mediated iron influx plays a crucial role in NMDA/NO-mediated cell death. We here report that iron influx is elicited by nitric oxide but not by other proapoptotic stimuli, such as H₂O₂ or staurosporine. Deletion of Dexras1 in mice attenuates NO-mediated cell death in dissociated primary cortical neurons and retinal ganglion cells in vivo. Thus, Dexras1 appears to mediate NMDA-elicited neurotoxicity via NO and iron influx.
Collapse
|
142
|
Schiavone S, Jaquet V, Trabace L, Krause KH. Severe life stress and oxidative stress in the brain: from animal models to human pathology. Antioxid Redox Signal 2013; 18:1475-90. [PMID: 22746161 PMCID: PMC3603496 DOI: 10.1089/ars.2012.4720] [Citation(s) in RCA: 220] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Revised: 06/25/2012] [Accepted: 07/01/2012] [Indexed: 12/27/2022]
Abstract
SIGNIFICANCE Severe life stress (SLS), as opposed to trivial everyday stress, is defined as a serious psychosocial event with the potential of causing an impacting psychological traumatism. RECENT ADVANCES Numerous studies have attempted to understand how the central nervous system (CNS) responds to SLS. This response includes a variety of morphological and neurochemical modifications; among them, oxidative stress is almost invariably observed. Oxidative stress is defined as disequilibrium between oxidant generation and the antioxidant response. CRITICAL ISSUES In this review, we discuss how SLS leads to oxidative stress in the CNS, and how the latter impacts pathophysiological outcomes. We also critically discuss experimental methods that measure oxidative stress in the CNS. The review covers animal models and human observations. Animal models of SLS include sleep deprivation, maternal separation, and social isolation in rodents, and the establishment of hierarchy in non-human primates. In humans, SLS, which is caused by traumatic events such as child abuse, war, and divorce, is also accompanied by oxidative stress in the CNS. FUTURE DIRECTIONS The outcome of SLS in humans ranges from resilience, over post-traumatic stress disorder, to development of chronic mental disorders. Defining the sources of oxidative stress in SLS might in the long run provide new therapeutic avenues.
Collapse
Affiliation(s)
- Stefania Schiavone
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland.
| | | | | | | |
Collapse
|
143
|
Leonelli M, Martins DO, Britto LRG. Retinal cell death induced by TRPV1 activation involves NMDA signaling and upregulation of nitric oxide synthases. Cell Mol Neurobiol 2013; 33:379-92. [PMID: 23324998 DOI: 10.1007/s10571-012-9904-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Accepted: 12/29/2012] [Indexed: 01/23/2023]
Abstract
The activation of the transient receptor potential vanilloid type 1 channel (TRPV1) has been correlated with oxidative and nitrosative stress and cell death in the nervous system. Our previous results indicate that TRPV1 activation in the adult retina can lead to constitutive and inducible nitric oxide synthase-dependent protein nitration and apoptosis. In this report, we have investigated the potential effects of TRPV1 channel activation on nitric oxide synthase (NOS) expression and function, and the putative participation of ionotropic glutamate receptors in retinal TRPV1-induced protein nitration, lipid peroxidation, and DNA fragmentation. Intravitreal injections of the classical TRPV1 agonist capsaicin up-regulated the protein expression of the inducible and endothelial NOS isoforms. Using 4,5-diaminofluorescein diacetate for nitric oxide (NO) imaging, we found that capsaicin also increased the production of NO in retinal blood vessels. Processes and perikarya of TRPV1-expressing neurons in the inner nuclear layer of the retina were found in the vicinity of nNOS-positive neurons, but those two proteins did not colocalize. Retinal explants exposed to capsaicin presented high protein nitration, lipid peroxidation, and cell death, which were observed in the inner nuclear and plexiform layers and in ganglion cells. This effect was partially blocked by AP-5, a NMDA glutamate receptor antagonist, but not by CNQX, an AMPA/kainate receptor antagonist. These data support a potential role for TRPV1 channels in physiopathological retinal processes mediated by NO, which at least in part involve glutamate release.
Collapse
Affiliation(s)
- Mauro Leonelli
- Laboratory of Cellular Neurobiology, Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1524, São Paulo, SP, 05508-000, Brazil.
| | | | | |
Collapse
|
144
|
Metabolic effects of TiO2 nanoparticles, a common component of sunscreens and cosmetics, on human keratinocytes. Cell Death Dis 2013; 4:e549. [PMID: 23519118 PMCID: PMC3615742 DOI: 10.1038/cddis.2013.76] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The long-term health risks of nanoparticles remain poorly understood, which is a serious concern given their prevalence in the environment from increased industrial and domestic use. The extent to which such compounds contribute to cellular toxicity is unclear, and although it is known that induction of oxidative stress pathways is associated with this process, the proteins and the metabolic pathways involved with nanoparticle-mediated oxidative stress and toxicity are largely unknown. To investigate this problem further, the effect of TiO2 on the HaCaT human keratinocyte cell line was examined. The data show that although TiO2 does not affect cell cycle phase distribution, nor cell death, these nanoparticles have a considerable and rapid effect on mitochondrial function. Metabolic analysis was performed to identify 268 metabolites of the specific pathways involved and 85 biochemical metabolites were found to be significantly altered, many of which are known to be associated with the cellular stress response. Importantly, the uptake of nanoparticles into the cultured cells was restricted to phagosomes, TiO2 nanoparticles did not enter into the nucleus or any other cytoplasmic organelle. No other morphological changes were detected after 24-h exposure consistent with a specific role of mitochondria in this response.
Collapse
|
145
|
Abstract
Sarcopenia is an aging-associated condition, which is currently characterized by the loss of muscle mass and muscle strength. However, there is no consensus regarding its characterization hitherto. As the world older adult population is on the rise, the impact of sarcopenia becomes greater. Due to the lack of effective treatments, sarcopenia is still a persisting problem among the global older adults and should not be overlooked. As a result, it is vital to investigate deeper into the mechanism underlying the pathogenesis of sarcopenia in order to develop more effective therapeutic interventions and to inscribe a more uniform characterization. The etiology of sarcopenia is currently found to be multifactorial, and most of the pharmacological researches are focused on the muscular factors in aging. Although the complete mechanism underlying the development of sarcopenia is still waiting to be elucidated, we propose in this article that the primary trigger of sarcopenia may be neurogenic in origin based on the intimate relationship between the nervous and muscular system, namely, the motor neuron and its underlying muscle fibers. Both of them are affected by the cellular environment and their physiological activity.
Collapse
|
146
|
Rufini A, Tucci P, Celardo I, Melino G. Senescence and aging: the critical roles of p53. Oncogene 2013; 32:5129-43. [PMID: 23416979 DOI: 10.1038/onc.2012.640] [Citation(s) in RCA: 753] [Impact Index Per Article: 68.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 11/30/2012] [Accepted: 12/07/2012] [Indexed: 11/09/2022]
Abstract
p53 functions as a transcription factor involved in cell-cycle control, DNA repair, apoptosis and cellular stress responses. However, besides inducing cell growth arrest and apoptosis, p53 activation also modulates cellular senescence and organismal aging. Senescence is an irreversible cell-cycle arrest that has a crucial role both in aging and as a robust physiological antitumor response, which counteracts oncogenic insults. Therefore, via the regulation of senescence, p53 contributes to tumor growth suppression, in a manner strictly dependent by its expression and cellular context. In this review, we focus on the recent advances on the contribution of p53 to cellular senescence and its implication for cancer therapy, and we will discuss p53's impact on animal lifespan. Moreover, we describe p53-mediated regulation of several physiological pathways that could mediate its role in both senescence and aging.
Collapse
Affiliation(s)
- A Rufini
- Medical Research Council, Toxicology Unit, Leicester University, Leicester, UK
| | | | | | | |
Collapse
|
147
|
Brain energy metabolism in glutamate-receptor activation and excitotoxicity: role for APC/C-Cdh1 in the balance glycolysis/pentose phosphate pathway. Neurochem Int 2013; 62:750-6. [PMID: 23416042 DOI: 10.1016/j.neuint.2013.02.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 01/25/2013] [Accepted: 02/03/2013] [Indexed: 02/07/2023]
Abstract
Recent advances in the field of brain energy metabolism strongly suggest that glutamate receptor-mediated neurotransmission is coupled with molecular signals that switch-on glucose utilization pathways to meet the high energetic requirements of neurons. Failure to adequately coordinate energy supply for neurotransmission ultimately results in a positive amplifying loop of receptor over-activation leading to neuronal death, a process known as excitotoxicity. In this review, we revisited current concepts in excitotoxic mechanisms, their involvement in energy substrate utilization, and the signaling pathways that coordinate both processes. In particular, we have focused on the novel role played by the E3 ubiquitin ligase, anaphase-promoting complex/cyclosome (APC/C)-Cdh1, in cell metabolism. Our laboratory identified 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 (PFKFB3) -a key glycolytic-promoting enzyme- as an APC/C-Cdh1 substrate. Interestingly, APC/C-Cdh1 activity is inhibited by over-activation of glutamate receptors through a Ca(2+)-mediated mechanism. Furthermore, by inhibiting APC/C-Cdh1 activity, glutamate-receptors activation promotes PFKFB3 stabilization, leading to increased glycolysis and decreased pentose-phosphate pathway activity. This causes a loss in neuronal ability to regenerate glutathione, triggering oxidative stress and delayed excitotoxicity. Further investigation is critical to identify novel molecules responsible for the coupling of energy metabolism with glutamatergic neurotransmission and excitotoxicity, as well as to help developing new therapeutic strategies against neurodegeneration.
Collapse
|
148
|
Heimfarth L, Loureiro SO, Dutra MF, Petenuzzo L, de Lima BO, Fernandes CG, da Rocha JBT, Pessoa-Pureur R. Disrupted cytoskeletal homeostasis, astrogliosis and apoptotic cell death in the cerebellum of preweaning rats injected with diphenyl ditelluride. Neurotoxicology 2013. [DOI: 10.1016/j.neuro.2012.10.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
149
|
D'Onofrio PM, Koeberle PD. What can we learn about stroke from retinal ischemia models? Acta Pharmacol Sin 2013. [PMID: 23202803 DOI: 10.1038/aps.2012.165] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Retinal ischemia is a very useful model to study the impact of various cell death pathways, such as apoptosis and necrosis, in the ischemic retina. However, it is important to note that the retina is formed as an outpouching of the diencephalon and is part of the central nervous system. As such, the cell death pathways initiated in response to ischemic damage in the retina reflect those found in other areas of the central nervous system undergoing similar trauma. The retina is also more accessible than other areas of the central nervous system, thus making it a simpler model to work with and study. By utilizing the retinal model, we can greatly increase our knowledge of the cell death processes initiated by ischemia which lead to degeneration in the central nervous system. This paper examines work that has been done so far to characterize various aspects of cell death in the retinal ischemia model, such as various pathways which are activated, and the role neurotrophic factors, and discusses how these are relevant to the treatment of ischemic damage in both the retina and the greater central nervous system.
Collapse
|
150
|
Abstract
Mitochondria play a central role in cell fate after stressors such as ischemic brain injury. The convergence of intracellular signaling pathways on mitochondria and their release of critical factors are now recognized as a default conduit to cell death or survival. Besides the individual processes that converge on or emanate from mitochondria, a mitochondrial organellar response to changes in the cellular environment has recently been described. Whereas mitochondria have previously been perceived as a major center for cellular signaling, one can postulate that the organelle's dynamics themselves affect cell survival. This brief perspective review puts forward the concept that disruptions in mitochondrial dynamics--biogenesis, clearance, and fission/fusion events--may underlie neural diseases and thus could be targeted as neuroprotective strategies in the context of ischemic injury. To do so, we present a general overview of the current understanding of mitochondrial dynamics and regulation. We then review emerging studies that correlate mitochondrial biogenesis, mitophagy, and fission/fusion events with neurologic disease and recovery. An overview of the system as it is currently understood is presented, and current assessment strategies and their limitations are discussed.
Collapse
|