251
|
Bartolome F, Esteras N, Martin-Requero A, Boutoleau-Bretonniere C, Vercelletto M, Gabelle A, Le Ber I, Honda T, Dinkova-Kostova AT, Hardy J, Carro E, Abramov AY. Pathogenic p62/SQSTM1 mutations impair energy metabolism through limitation of mitochondrial substrates. Sci Rep 2017; 7:1666. [PMID: 28490746 PMCID: PMC5431917 DOI: 10.1038/s41598-017-01678-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 03/30/2017] [Indexed: 12/21/2022] Open
Abstract
Abnormal mitochondrial function has been found in patients with frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). Mutations in the p62 gene (also known as SQSTM1) which encodes the p62 protein have been reported in both disorders supporting the idea of an ALS/FTD continuum. In this work the role of p62 in energy metabolism was studied in fibroblasts from FTD patients carrying two independent pathogenic mutations in the p62 gene, and in a p62-knock-down (p62 KD) human dopaminergic neuroblastoma cell line (SH-SY5Y). We found that p62 deficiency is associated with inhibited complex I mitochondrial respiration due to lack of NADH for the electron transport chain. This deficiency was also associated with increased levels of NADPH reflecting a higher activation of pentose phosphate pathway as this is accompanied with higher cytosolic reduced glutathione (GSH) levels. Complex I inhibition resulted in lower mitochondrial membrane potential and higher cytosolic ROS production. Pharmacological activation of transcription factor Nrf2 increased mitochondrial NADH levels and restored mitochondrial membrane potential in p62-deficient cells. Our results suggest that the phenotype is caused by a loss-of-function effect, because similar alterations were found both in the mutant fibroblasts and the p62 KD model. These findings highlight the implication of energy metabolism in pathophysiological events associated with p62 deficiency.
Collapse
Affiliation(s)
- Fernando Bartolome
- Neurodegenerative Disorders group, Instituto de Investigacion Hospital 12 de Octubre (i+12), Av Cordoba, Madrid, 28041, Spain. .,Biomedical Research Networking Centre on Neurodegenerative Diseases (CIBERNED), Madrid, Spain. .,Department of Molecular Neuroscience, UCL Institute of Neurology Queen Square, London, WC1N 3BG, UK.
| | - Noemi Esteras
- Department of Molecular Neuroscience, UCL Institute of Neurology Queen Square, London, WC1N 3BG, UK
| | - Angeles Martin-Requero
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, Madrid, 28040, Spain.,Biomedical Research Networking Centre on Rare Diseases (CIBERER), Madrid, Spain
| | - Claire Boutoleau-Bretonniere
- Laboratoire d'études des mécanismes cognitifs, EA 3082, Université Lyon 2, Bron, F-69500, France.,CHU Nantes, Centre de Mémoire et de Ressource et Recherche (CM2R), Nantes, France.,Inserm, CIC 04, Nantes, France
| | - Martine Vercelletto
- CHU Nantes, Centre de Mémoire et de Ressource et Recherche (CM2R), Nantes, France.,Inserm, CIC 04, Nantes, France
| | - Audrey Gabelle
- Memory Research and Resources Center, Department of Neurology, Montpellier University Hospital, Montpellier, France
| | - Isabelle Le Ber
- CNR-MAJ, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France.,ICM, Inserm U1127, CNRS UMR 7225, Sorbonne Universités, UPMC-P6 UMR S 1127 - Hôpital Pitié-Salpêtrière, Paris, France
| | - Tadashi Honda
- Department of Chemistry and Institute of Chemical Biology & Drug Discovery Stony Brook University Stony Brook, New York, 11794, USA
| | | | - John Hardy
- Department of Molecular Neuroscience, UCL Institute of Neurology Queen Square, London, WC1N 3BG, UK.,Reta Lilla Weston Laboratories, London, WC1N 3BG, UK
| | - Eva Carro
- Neurodegenerative Disorders group, Instituto de Investigacion Hospital 12 de Octubre (i+12), Av Cordoba, Madrid, 28041, Spain.,Biomedical Research Networking Centre on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Andrey Y Abramov
- Department of Molecular Neuroscience, UCL Institute of Neurology Queen Square, London, WC1N 3BG, UK.
| |
Collapse
|
252
|
Abstract
Across all kingdoms in the tree of life, calcium (Ca2+) is an essential element used by cells to respond and adapt to constantly changing environments. In multicellular organisms, it plays fundamental roles during fertilization, development and adulthood. The inability of cells to regulate Ca2+ can lead to pathological conditions that ultimately culminate in cell death. One such pathological condition is manifested in Parkinson's disease, the second most common neurological disorder in humans, which is characterized by the aggregation of the protein, α-synuclein. This Review discusses current evidence that implicates Ca2+ in the pathogenesis of Parkinson's disease. Understanding the mechanisms by which Ca2+ signaling contributes to the progression of this disease will be crucial for the development of effective therapies to combat this devastating neurological condition.
Collapse
Affiliation(s)
- Sofia V Zaichick
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Kaitlyn M McGrath
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Gabriela Caraveo
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
253
|
Zhang Y, Nguyen DT, Olzomer EM, Poon GP, Cole NJ, Puvanendran A, Phillips BR, Hesselson D. Rescue of Pink1 Deficiency by Stress-Dependent Activation of Autophagy. Cell Chem Biol 2017; 24:471-480.e4. [PMID: 28366621 DOI: 10.1016/j.chembiol.2017.03.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 01/02/2017] [Accepted: 03/02/2017] [Indexed: 10/19/2022]
Abstract
Stimulating autophagy is a promising therapeutic strategy for slowing the progression of neurodegenerative disease. Neurons are insensitive to current approaches based on mTOR inhibition for activating autophagy, and instead may rely on the Parkinson's disease-associated proteins PINK1 and PARKIN to activate the autophagy-lysosomal pathway in response to mitochondrial damage. We developed a multifactorial zebrafish drug-screening platform combining Pink1 deficiency with an environmental toxin to compromise mitochondrial function and trigger dopaminergic neuron loss. Using a phenotypic screening strategy, we identified a series of piperazine phenothiazines, including trifluoperazine, which rescued Pink1 deficiency by activating autophagy selectively in stressed zebrafish and human cells. We show that trifluoperazine acts downstream of, or parallel to, PINK1/PARKIN to stimulate transcription factor EB nuclear translocation and the expression of autophagy-lysosomal target genes. These data suggest that stress-dependent pharmacological reactivation of autophagy could prevent the loss of vulnerable neurons to slow neurodegeneration.
Collapse
Affiliation(s)
- Yuxi Zhang
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - David T Nguyen
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Ellen M Olzomer
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Gin P Poon
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Nicholas J Cole
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Anita Puvanendran
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Brigitte R Phillips
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Daniel Hesselson
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; St. Vincent's Clinical School, UNSW Australia, Sydney, NSW 2010, Australia.
| |
Collapse
|
254
|
Gelmetti V, De Rosa P, Torosantucci L, Marini ES, Romagnoli A, Di Rienzo M, Arena G, Vignone D, Fimia GM, Valente EM. PINK1 and BECN1 relocalize at mitochondria-associated membranes during mitophagy and promote ER-mitochondria tethering and autophagosome formation. Autophagy 2017; 13:654-669. [PMID: 28368777 DOI: 10.1080/15548627.2016.1277309] [Citation(s) in RCA: 260] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Mitophagy is a highly specialized process to remove dysfunctional or superfluous mitochondria through the macroautophagy/autophagy pathway, aimed at protecting cells from the damage of disordered mitochondrial metabolism and apoptosis induction. PINK1, a neuroprotective protein mutated in autosomal recessive Parkinson disease, has been implicated in the activation of mitophagy by selectively accumulating on depolarized mitochondria, and promoting PARK2/Parkin translocation to them. While these steps have been characterized in depth, less is known about the process and site of autophagosome formation upon mitophagic stimuli. A previous study reported that, in starvation-induced autophagy, the proautophagic protein BECN1/Beclin1 (which we previously showed to interact with PINK1) relocalizes at specific regions of contact between the endoplasmic reticulum (ER) and mitochondria called mitochondria-associated membranes (MAM), from which the autophagosome originates. Here we show that, following mitophagic stimuli, autophagosomes also form at MAM; moreover, endogenous PINK1 and BECN1 were both found to relocalize at MAM, where they promoted the enhancement of ER-mitochondria contact sites and the formation of omegasomes, that represent autophagosome precursors. PARK2 was also enhanced at MAM following mitophagy induction. However, PINK1 silencing impaired BECN1 enrichment at MAM independently of PARK2, suggesting a novel role for PINK1 in regulating mitophagy. MAM have been recently implicated in many key cellular events. In this light, the observed prevalent localization of PINK1 at MAM may well explain other neuroprotective activities of this protein, such as modulation of mitochondrial calcium levels, mitochondrial dynamics, and apoptosis.
Collapse
Affiliation(s)
- Vania Gelmetti
- a Neurogenetics Unit, IRCCS Santa Lucia Foundation , Rome , Italy
| | - Priscilla De Rosa
- b IRCCS Casa Sollievo della Sofferenza, CSS-Mendel Institute , Rome , Italy
| | | | | | - Alessandra Romagnoli
- d National Institute for Infectious Diseases "LazzaroSpallanzani" IRCCS , Rome , Italy
| | - Martina Di Rienzo
- d National Institute for Infectious Diseases "LazzaroSpallanzani" IRCCS , Rome , Italy.,e Department of Biology , "Tor Vergata" University , Rome , Italy
| | - Giuseppe Arena
- f IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université Montpellier, Institutrégional du Cancer Montpellier , Montpellier , France
| | | | - Gian Maria Fimia
- d National Institute for Infectious Diseases "LazzaroSpallanzani" IRCCS , Rome , Italy.,h Department of Biological and Environmental Sciences and Technologies (DiSTeBA) , University of Salento , Lecce , Italy
| | - Enza Maria Valente
- a Neurogenetics Unit, IRCCS Santa Lucia Foundation , Rome , Italy.,i Department of Molecular Medicine , University of Pavia , Pavia , Italy
| |
Collapse
|
255
|
Lehmann S, Loh SHY, Martins LM. Enhancing NAD + salvage metabolism is neuroprotective in a PINK1 model of Parkinson's disease. Biol Open 2017; 6:141-147. [PMID: 28011627 PMCID: PMC5312101 DOI: 10.1242/bio.022186] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Familial forms of Parkinson's disease (PD) caused by mutations in PINK1 are linked to mitochondrial impairment. Defective mitochondria are also found in Drosophila models of PD with pink1 mutations. The co-enzyme nicotinamide adenine dinucleotide (NAD+) is essential for both generating energy in mitochondria and nuclear DNA repair through NAD+-consuming poly(ADP-ribose) polymerases (PARPs). We found alterations in NAD+ salvage metabolism in Drosophila pink1 mutants and showed that a diet supplemented with the NAD+ precursor nicotinamide rescued mitochondrial defects and protected neurons from degeneration. Additionally, a mutation of Parp improved mitochondrial function and was neuroprotective in the pink1 mutants. We conclude that enhancing the availability of NAD+ by either the use of a diet supplemented with NAD+ precursors or the inhibition of NAD+-dependent enzymes, such as PARPs, which compete with mitochondria for NAD+, is a viable approach to preventing neurotoxicity associated with mitochondrial defects.
Collapse
Affiliation(s)
- Susann Lehmann
- MRC Toxicology Unit, Lancaster Road, Leicester LE1 9HN, UK
| | | | | |
Collapse
|
256
|
Wang Y, He H, Wang S, Liu Y, Hu M, Cao Y, Kong S, Wei X, Wang C. Photostimulation by femtosecond laser triggers restorable fragmentation in single mitochondrion. JOURNAL OF BIOPHOTONICS 2017; 10:286-293. [PMID: 26847091 DOI: 10.1002/jbio.201500281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 01/14/2016] [Accepted: 01/15/2016] [Indexed: 05/08/2023]
Abstract
Mitochondrial research is important to the study of ageing, apoptosis, and metabolic diseases. Over the years, mitochondria have been studied with stimulation by chemical agents in a global manner for basic and applied research. This approach lacks of precision and accuracy in terms of spatial and temporal resolution. Here we demonstrate a direct and well-defined photostimulation targeting on single mitochondrial tubular structure using a tightly-focused femtosecond (fs) laser that could precisely activate mitochondria at single tubule level to show restorable fragmentation and subsequent recovery after tens of seconds. In these two processes, a series of mitochondrial reactive oxygen species (mROS) flashes was observed and found critical to the mitochondrial fragmentation. Meanwhile, transient openings of mitochondrial permeability transition pores (mPTP) were seen with oscillations of mitochondrial membrane potential. These activities were crucial for the recovery through scavenging the mROS. Without the feedback mechanisms, the fragmented mitochondria could not return back to their original tubular structure. These interesting observations show that photostimulation by fs laser is an active, precise, clean and well-defined approach to dissect the role of mitochondria in normal physiology and different kinds of diseases.
Collapse
Affiliation(s)
- Yintao Wang
- Ultrafast Laser Laboratory, Key Laboratory of Optoelectronic Information Technology (Ministry of Education), College of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin, 300072, P.R. China
| | - Hao He
- Ultrafast Laser Laboratory, Key Laboratory of Optoelectronic Information Technology (Ministry of Education), College of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin, 300072, P.R. China
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Shaoyang Wang
- Ultrafast Laser Laboratory, Key Laboratory of Optoelectronic Information Technology (Ministry of Education), College of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin, 300072, P.R. China
| | - Yaohui Liu
- Key Laboratory of microbial functional genomics of Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300072, P.R. China
| | - Minglie Hu
- Ultrafast Laser Laboratory, Key Laboratory of Optoelectronic Information Technology (Ministry of Education), College of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin, 300072, P.R. China
| | - Youjia Cao
- Key Laboratory of microbial functional genomics of Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300072, P.R. China
| | - Siukai Kong
- School of Life Sciences, the Chinese University of Hong Kong, Hong Kong
| | - Xunbin Wei
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Chingyue Wang
- Ultrafast Laser Laboratory, Key Laboratory of Optoelectronic Information Technology (Ministry of Education), College of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin, 300072, P.R. China
| |
Collapse
|
257
|
Li Y, Qiu L, Liu X, Hou Z, Yu B. PINK1 alleviates myocardial hypoxia-reoxygenation injury by ameliorating mitochondrial dysfunction. Biochem Biophys Res Commun 2017; 484:118-124. [DOI: 10.1016/j.bbrc.2017.01.061] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 01/13/2017] [Indexed: 10/20/2022]
|
258
|
Human iPSC-Derived Neural Progenitors Are an Effective Drug Discovery Model for Neurological mtDNA Disorders. Cell Stem Cell 2017; 20:659-674.e9. [PMID: 28132834 DOI: 10.1016/j.stem.2016.12.013] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 11/04/2016] [Accepted: 12/19/2016] [Indexed: 01/19/2023]
Abstract
Mitochondrial DNA (mtDNA) mutations frequently cause neurological diseases. Modeling of these defects has been difficult because of the challenges associated with engineering mtDNA. We show here that neural progenitor cells (NPCs) derived from human induced pluripotent stem cells (iPSCs) retain the parental mtDNA profile and exhibit a metabolic switch toward oxidative phosphorylation. NPCs derived in this way from patients carrying a deleterious homoplasmic mutation in the mitochondrial gene MT-ATP6 (m.9185T>C) showed defective ATP production and abnormally high mitochondrial membrane potential (MMP), plus altered calcium homeostasis, which represents a potential cause of neural impairment. High-content screening of FDA-approved drugs using the MMP phenotype highlighted avanafil, which we found was able to partially rescue the calcium defect in patient NPCs and differentiated neurons. Overall, our results show that iPSC-derived NPCs provide an effective model for drug screening to target mtDNA disorders that affect the nervous system.
Collapse
|
259
|
Ma MW, Wang J, Zhang Q, Wang R, Dhandapani KM, Vadlamudi RK, Brann DW. NADPH oxidase in brain injury and neurodegenerative disorders. Mol Neurodegener 2017; 12:7. [PMID: 28095923 PMCID: PMC5240251 DOI: 10.1186/s13024-017-0150-7] [Citation(s) in RCA: 309] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 01/05/2017] [Indexed: 12/11/2022] Open
Abstract
Oxidative stress is a common denominator in the pathology of neurodegenerative disorders such as Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis, and multiple sclerosis, as well as in ischemic and traumatic brain injury. The brain is highly vulnerable to oxidative damage due to its high metabolic demand. However, therapies attempting to scavenge free radicals have shown little success. By shifting the focus to inhibit the generation of damaging free radicals, recent studies have identified NADPH oxidase as a major contributor to disease pathology. NADPH oxidase has the primary function to generate free radicals. In particular, there is growing evidence that the isoforms NOX1, NOX2, and NOX4 can be upregulated by a variety of neurodegenerative factors. The majority of recent studies have shown that genetic and pharmacological inhibition of NADPH oxidase enzymes are neuroprotective and able to reduce detrimental aspects of pathology following ischemic and traumatic brain injury, as well as in chronic neurodegenerative disorders. This review aims to summarize evidence supporting the role of NADPH oxidase in the pathology of these neurological disorders, explores pharmacological strategies of targeting this major oxidative stress pathway, and outlines obstacles that need to be overcome for successful translation of these therapies to the clinic.
Collapse
Affiliation(s)
- Merry W Ma
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Jing Wang
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Quanguang Zhang
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Ruimin Wang
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Krishnan M Dhandapani
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neurosurgery, Medical College of Georgia, Augusta University, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Ratna K Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health Science Center, 7703 Medical Drive, San Antonio, TX, 78229, USA
| | - Darrell W Brann
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA. .,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA.
| |
Collapse
|
260
|
Rastogi R, Geng X, Li F, Ding Y. NOX Activation by Subunit Interaction and Underlying Mechanisms in Disease. Front Cell Neurosci 2017; 10:301. [PMID: 28119569 PMCID: PMC5222855 DOI: 10.3389/fncel.2016.00301] [Citation(s) in RCA: 153] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 12/20/2016] [Indexed: 12/19/2022] Open
Abstract
Nicotinamide adenine dinucleotide phosphate (NAPDH) oxidase (NOX) is an enzyme complex with the sole function of producing superoxide anion and reactive oxygen species (ROS) at the expense of NADPH. Vital to the immune system as well as cellular signaling, NOX is also involved in the pathologies of a wide variety of disease states. Particularly, it is an integral player in many neurological diseases, including stroke, TBI, and neurodegenerative diseases. Pathologically, NOX produces an excessive amount of ROS that exceed the body’s antioxidant ability to neutralize them, leading to oxidative stress and aberrant signaling. This prevalence makes it an attractive therapeutic target and as such, NOX inhibitors have been studied and developed to counter NOX’s deleterious effects. However, recent studies of NOX have created a better understanding of the NOX complex. Comprised of independent cytosolic subunits, p47-phox, p67-phox, p40-phox and Rac, and membrane subunits, gp91-phox and p22-phox, the NOX complex requires a unique activation process through subunit interaction. Of these subunits, p47-phox plays the most important role in activation, binding and translocating the cytosolic subunits to the membrane and anchoring to p22-phox to organize the complex for NOX activation and function. Moreover, these interactions, particularly that between p47-phox and p22-phox, are dependent on phosphorylation initiated by upstream processes involving protein kinase C (PKC). This review will look at these interactions between subunits and with PKC. It will focus on the interaction involving p47-phox with p22-phox, key in bringing the cytosolic subunits to the membrane. Furthermore, the implication of these interactions as a target for NOX inhibitors such as apocynin will be discussed as a potential avenue for further investigation, in order to develop more specific NOX inhibitors based on the inhibition of NOX assembly and activation.
Collapse
Affiliation(s)
- Radhika Rastogi
- Department of Neurosurgery, Wayne State University School of Medicine Detroit, MI, USA
| | - Xiaokun Geng
- Department of Neurosurgery, Wayne State University School of MedicineDetroit, MI, USA; China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical UniversityBeijing, China; Department of Neurology, Beijing Luhe Hospital, Capital Medical UniversityBeijing, China
| | - Fengwu Li
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University Beijing, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of MedicineDetroit, MI, USA; China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical UniversityBeijing, China
| |
Collapse
|
261
|
Soman S, Keatinge M, Moein M, Da Costa M, Mortiboys H, Skupin A, Sugunan S, Bazala M, Kuznicki J, Bandmann O. Inhibition of the mitochondrial calcium uniporter rescues dopaminergic neurons in pink1 -/- zebrafish. Eur J Neurosci 2016; 45:528-535. [PMID: 27859782 PMCID: PMC5324670 DOI: 10.1111/ejn.13473] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 11/03/2016] [Accepted: 11/04/2016] [Indexed: 11/26/2022]
Abstract
Mutations in PTEN-induced putative kinase 1 (PINK1) are a cause of early onset Parkinson's disease (PD). Loss of PINK1 function causes dysregulation of mitochondrial calcium homeostasis, resulting in mitochondrial dysfunction and neuronal cell death. We report that both genetic and pharmacological inactivation of the mitochondrial calcium uniporter (MCU), located in the inner mitochondrial membrane, prevents dopaminergic neuronal cell loss in pink1Y431 * mutant zebrafish (Danio rerio) via rescue of mitochondrial respiratory chain function. In contrast, genetic inactivation of the voltage dependent anion channel 1 (VDAC1), located in the outer mitochondrial membrane, did not rescue dopaminergic neurons in PINK1 deficient D. rerio. Subsequent gene expression studies revealed specific upregulation of the mcu regulator micu1 in pink1Y431 * mutant zebrafish larvae and inactivation of micu1 also results in rescue of dopaminergic neurons. The functional consequences of PINK1 deficiency and modified MCU activity were confirmed using a dynamic in silico model of Ca2+ triggered mitochondrial activity. Our data suggest modulation of MCU-mediated mitochondrial calcium homeostasis as a possible neuroprotective strategy in PINK1 mutant PD.
Collapse
Affiliation(s)
- Smijin Soman
- International Institute of Molecular and Cell Biology, Warsaw, Poland.,Bateson Centre, University of Sheffield, Sheffield, UK.,Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
| | - Marcus Keatinge
- Bateson Centre, University of Sheffield, Sheffield, UK.,Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
| | - Mahsa Moein
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
| | - Marc Da Costa
- Bateson Centre, University of Sheffield, Sheffield, UK.,Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
| | - Heather Mortiboys
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
| | - Alexander Skupin
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg.,National Centre for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA, USA
| | - Sreedevi Sugunan
- International Institute of Molecular and Cell Biology, Warsaw, Poland.,Bateson Centre, University of Sheffield, Sheffield, UK.,Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
| | - Michal Bazala
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Jacek Kuznicki
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Oliver Bandmann
- Bateson Centre, University of Sheffield, Sheffield, UK.,Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
| |
Collapse
|
262
|
Nardin A, Schrepfer E, Ziviani E. Counteracting PINK/Parkin Deficiency in the Activation of Mitophagy: A Potential Therapeutic Intervention for Parkinson's Disease. Curr Neuropharmacol 2016; 14:250-9. [PMID: 26517048 PMCID: PMC4857628 DOI: 10.2174/1570159x13666151030104414] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 05/20/2015] [Accepted: 05/20/2015] [Indexed: 12/21/2022] Open
Abstract
Parkinson’s Disease (PD) related genes PINK1, a protein kinase [1], and Parkin, an E3 ubiquitin ligase [2], operate within the same pathway [3-5], which controls, via specific elimination of dysfunctional mitochondria, the quality of the organelle network [6]. Parkin translocates to impaired mitochondria and drives their elimination via autophagy, a process known as mitophagy [6]. PINK1 regulates Parkin translocation through a not yet completely understood mechanism [7, 8]. Mitochondrial outer membrane proteins Mitofusin (MFN), VDAC, Fis1 and TOM20 were found to be targets for Parkin mediated ubiquitination [9-11]. By adding ubiquitin molecules to its targets expressed on mitochondria, Parkin tags and selects dysfunctional mitochondria for clearance, contributing to maintain a functional and healthy mitochondrial network. Abnormal accumulation of misfolded proteins and unfunctional mitochondria is a characteristic hallmark of PD pathology. Therefore a therapeutic approach to enhance clearance of misfolded proteins and potentiate the ubiquitin-proteosome system (UPS) could be instrumental to ameliorate the progression of the disease. Recently, much effort has been put to identify specific de-ubiquitinating enzymes (DUBs) that oppose Parkin in the ubiquitination of its targets. Similar to other post-translational modifications, such as phosphorylation and acetylation, ubiquitination is also a reversible modification, mediated by a large family of DUBs [12]. DUBs inhibitors or activators can affect cellular response to stimuli that induce mitophagy via ubiquitination of mitochondrial outer membrane proteins MFN, VDAC, Fis1 and TOM20. In this respect, the identification of a Parkin-opposing DUB in the regulation of mitophagy, might be instrumental to develop specific isopeptidase inhibitors or activators that can modulate the fundamental biological process of mitochondria clearance and impact on cell survival.
Collapse
Affiliation(s)
| | | | - Elena Ziviani
- Department of Biology, University of Padova, via Ugo Bassi 58b, 35121, Padova, Italy.
| |
Collapse
|
263
|
Arena G, Valente EM. PINK1 in the limelight: multiple functions of an eclectic protein in human health and disease. J Pathol 2016; 241:251-263. [PMID: 27701735 DOI: 10.1002/path.4815] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Revised: 09/04/2016] [Accepted: 09/23/2016] [Indexed: 01/02/2023]
Abstract
The gene PINK1 [phosphatase and tensin homologue (PTEN)-induced putative kinase 1] encodes a serine/threonine kinase which was initially linked to the pathogenesis of a familial form of Parkinson's disease. Research on PINK1 has recently unravelled that its multiple functions extend well beyond neuroprotection, implicating this eclectic protein in a growing number of human pathologies, including cancer, diabetes, cardiopulmonary dysfunctions, and inflammation. Extensive studies have identified PINK1 as a crucial player in the mitochondrial quality control pathway, required to label damaged mitochondria and promote their elimination through an autophagic process (mitophagy). Mounting evidence now indicates that PINK1 activities are not restricted solely to mitophagy, and that different subcellular and even sub-mitochondrial pools of PINK1 are involved in distinct signalling cascades to regulate cell metabolism and survival. In this review, we provide a concise overview on the different functions of PINK1 and their potential role in human diseases. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Giuseppe Arena
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Montpellier, France.,INSERM, U1194, Montpellier, France.,Université Montpellier, Montpellier, France.,Institut Régional du Cancer Montpellier, Montpellier, France
| | - Enza Maria Valente
- Section of Neurosciences, Department of Medicine and Surgery, University of Salerno, Salerno, Italy.,Neurogenetics Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| |
Collapse
|
264
|
Abstract
Reactive species play an important role in physiological functions. Overproduction of reactive species, notably reactive oxygen (ROS) and nitrogen (RNS) species along with the failure of balance by the body's antioxidant enzyme systems results in destruction of cellular structures, lipids, proteins, and genetic materials such as DNA and RNA. Moreover, the effects of reactive species on mitochondria and their metabolic processes eventually cause a rise in ROS/RNS levels, leading to oxidation of mitochondrial proteins, lipids, and DNA. Oxidative stress has been considered to be linked to the etiology of many diseases, including neurodegenerative diseases (NDDs) such as Alzheimer diseases, Amyotrophic lateral sclerosis, Friedreich's ataxia, Huntington's disease, Multiple sclerosis, and Parkinson's diseases. In addition, oxidative stress causing protein misfold may turn to other NDDs include Creutzfeldt-Jakob disease, Bovine Spongiform Encephalopathy, Kuru, Gerstmann-Straussler-Scheinker syndrome, and Fatal Familial Insomnia. An overview of the oxidative stress and mitochondrial dysfunction-linked NDDs has been summarized in this review.
Collapse
Affiliation(s)
- Md Torequl Islam
- a Northeast Biotechnology Network (RENORBIO), Postgraduate Program in Biotechnology , Federal University of Piauí (UFPI) , Teresina , Brazil.,b Department of Pharmacy, Faculty of Science and Engineering , Southern University Bangladesh (SUB) , Chittagong , Bangladesh
| |
Collapse
|
265
|
Requejo-Aguilar R, Bolaños JP. Mitochondrial control of cell bioenergetics in Parkinson's disease. Free Radic Biol Med 2016; 100:123-137. [PMID: 27091692 PMCID: PMC5065935 DOI: 10.1016/j.freeradbiomed.2016.04.012] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/13/2016] [Accepted: 04/14/2016] [Indexed: 12/15/2022]
Abstract
Parkinson disease (PD) is a neurodegenerative disorder characterized by a selective loss of dopaminergic neurons in the substantia nigra. The earliest biochemical signs of the disease involve failure in mitochondrial-endoplasmic reticulum cross talk and lysosomal function, mitochondrial electron chain impairment, mitochondrial dynamics alterations, and calcium and iron homeostasis abnormalities. These changes are associated with increased mitochondrial reactive oxygen species (mROS) and energy deficiency. Recently, it has been reported that, as an attempt to compensate for the mitochondrial dysfunction, neurons invoke glycolysis as a low-efficient mode of energy production in models of PD. Here, we review how mitochondria orchestrate the maintenance of cellular energetic status in PD, with special focus on the switch from oxidative phosphorylation to glycolysis, as well as the implication of endoplasmic reticulum and lysosomes in the control of bioenergetics.
Collapse
Affiliation(s)
- Raquel Requejo-Aguilar
- Department of Biochemistry and Molecular Biology, University of Cordoba, Institute Maimonides of Biomedical Investigation of Cordoba (IMIBIC), Cordoba, Spain
| | - Juan P Bolaños
- Institute of Functional Biology and Genomics (IBFG), University of Salamanca-CSIC, Zacarias Gonzalez, 2, 37007 Salamanca, Spain.
| |
Collapse
|
266
|
Angelova PR, Abramov AY. Functional role of mitochondrial reactive oxygen species in physiology. Free Radic Biol Med 2016; 100:81-85. [PMID: 27296839 DOI: 10.1016/j.freeradbiomed.2016.06.005] [Citation(s) in RCA: 185] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 06/06/2016] [Accepted: 06/07/2016] [Indexed: 02/07/2023]
Abstract
The major energy generator in the cell - mitochondria produce reactive oxygen species as a by-product of a number of enzymatic reactions and the production of ATP. Emerging evidence suggests that mitochondrial ROS regulate diverse physiological parameters and that dysregulated ROS signalling may contribute to a development of processes which lead to human diseases. ROS produced in mitochondrial enzymes are triggers of monoamine-induced calcium signal in astrocytes, playing important role in physiological and pathophysiological response to dopamine. Generation of ROS in mitochondria leads to peroxidation of lipids, which is considered to be one of the most important mechanisms of cell injury under condition of oxidative stress. However, it also can induce activation of mitochondrial and cellular phospholipases that can trigger a variety of the signals - from activation of ion channels to stimulation of calcium signal. Mitochondria are shown to be the oxygen sensor in astrocytes, therefore inhibition of respiration by hypoxia induces ROS production which leads to lipid peroxidation, activation of phospholipase C and induction of IP3-mediated calcium signal. Propagation of astrocytic calcium signal stimulates breathing activity in response to hypoxia. Thus, ROS produced by mitochondrial enzymes or electron transport chain can be used as a trigger for signalling cascades in central nervous system and deregulation of this process leads to pathology.
Collapse
Affiliation(s)
- Plamena R Angelova
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK.
| | - Andrey Y Abramov
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK.
| |
Collapse
|
267
|
Blacker TS, Duchen MR. Investigating mitochondrial redox state using NADH and NADPH autofluorescence. Free Radic Biol Med 2016; 100:53-65. [PMID: 27519271 PMCID: PMC5145803 DOI: 10.1016/j.freeradbiomed.2016.08.010] [Citation(s) in RCA: 254] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 08/02/2016] [Accepted: 08/08/2016] [Indexed: 11/27/2022]
Abstract
The redox states of the NAD and NADP pyridine nucleotide pools play critical roles in defining the activity of energy producing pathways, in driving oxidative stress and in maintaining antioxidant defences. Broadly speaking, NAD is primarily engaged in regulating energy-producing catabolic processes, whilst NADP may be involved in both antioxidant defence and free radical generation. Defects in the balance of these pathways are associated with numerous diseases, from diabetes and neurodegenerative disease to heart disease and cancer. As such, a method to assess the abundance and redox state of these separate pools in living tissues would provide invaluable insight into the underlying pathophysiology. Experimentally, the intrinsic fluorescence of the reduced forms of both redox cofactors, NADH and NADPH, has been used for this purpose since the mid-twentieth century. In this review, we outline the modern implementation of these techniques for studying mitochondrial redox state in complex tissue preparations. As the fluorescence spectra of NADH and NADPH are indistinguishable, interpreting the signals resulting from their combined fluorescence, often labelled NAD(P)H, can be complex. We therefore discuss recent studies using fluorescence lifetime imaging microscopy (FLIM) which offer the potential to discriminate between the two separate pools. This technique provides increased metabolic information from cellular autofluorescence in biomedical investigations, offering biochemical insights into the changes in time-resolved NAD(P)H fluorescence signals observed in diseased tissues.
Collapse
Affiliation(s)
- Thomas S Blacker
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK; Department of Physics and Astronomy, University College London, London WC1E 6BT, UK
| | - Michael R Duchen
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| |
Collapse
|
268
|
Kang R, Zeng L, Xie Y, Yan Z, Zhou B, Cao L, Klionsky DJ, Tracey KJ, Li J, Wang H, Billiar TR, Jiang J, Tang D. A novel PINK1- and PARK2-dependent protective neuroimmune pathway in lethal sepsis. Autophagy 2016; 12:2374-2385. [PMID: 27754761 DOI: 10.1080/15548627.2016.1239678] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Although the PINK1-PARK2 pathway contributes to the pathogenesis of Parkinson disease, its roles in sepsis (a major challenge for critical care) were previously unknown. Here, we show that pink1-/- and park2-/- mice are more sensitive to polymicrobial sepsis-induced multiple organ failure and death. The decrease in the circulating level of the neurotransmitter dopamine in pink1-/- and park2-/- mice accelerates the release of a late sepsis mediator, HMGB1, via HIF1A-dependent anaerobic glycolysis and subsequent NLRP3-dependent inflammasome activation. Genetic depletion of Nlrp3 or Hif1a in pink1-/- and park2-/- mice confers protection against lethal polymicrobial sepsis. Moreover, pharmacological administration of dopamine agonist (e.g., pramipexole), HMGB1-inhibitor (e.g., neutralizing antibody or glycyrrhizin), or NLRP3-inhibitor (e.g., MCC950) reduces septic death in pink1-/- and park2-/- mice. The mRNA expression of HIF1A and NLRP3 is upregulated, whereas the mRNA expression of PINK1 and PARK2 is downregulated in peripheral blood mononuclear cells of patients with sepsis. Thus, an impaired PINK1-PARK2-mediated neuroimmunology pathway contributes to septic death and may represent a novel therapeutic target in critical care medicine.
Collapse
Affiliation(s)
- Rui Kang
- a Department of Surgery , University of Pittsburgh , Pittsburgh , PA , USA
| | - Ling Zeng
- a Department of Surgery , University of Pittsburgh , Pittsburgh , PA , USA.,b State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Research institute for Traffic Medicine of People's Liberation Army, Daping Hospital, Third Military Medical University , Chongqing , China
| | - Yangchun Xie
- a Department of Surgery , University of Pittsburgh , Pittsburgh , PA , USA
| | - Zhengwen Yan
- a Department of Surgery , University of Pittsburgh , Pittsburgh , PA , USA.,c Department of Neurology , Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University , Guangzhou, Guangdong , China
| | - Borong Zhou
- d Department of Neurology.,e Centre of DAMP Biology, Third Affiliated Hospital of Guangzhou Medical University , Guangzhou, Guangdong , China
| | - Lizhi Cao
- f Department of Pediatrics , Xiangya Hospital, Central South University , Changsha, Hunan , China
| | - Daniel J Klionsky
- g Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology , University of Michigan , Ann Arbor , MI , USA
| | - Kevin J Tracey
- h Laboratory of Emergency Medicine, The Feinstein Institute for Medical Research , Manhasset , NY , USA
| | - Jianhua Li
- h Laboratory of Emergency Medicine, The Feinstein Institute for Medical Research , Manhasset , NY , USA
| | - Haichao Wang
- h Laboratory of Emergency Medicine, The Feinstein Institute for Medical Research , Manhasset , NY , USA
| | - Timothy R Billiar
- a Department of Surgery , University of Pittsburgh , Pittsburgh , PA , USA
| | - Jianxin Jiang
- b State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Research institute for Traffic Medicine of People's Liberation Army, Daping Hospital, Third Military Medical University , Chongqing , China
| | - Daolin Tang
- a Department of Surgery , University of Pittsburgh , Pittsburgh , PA , USA.,e Centre of DAMP Biology, Third Affiliated Hospital of Guangzhou Medical University , Guangzhou, Guangdong , China
| |
Collapse
|
269
|
Metallomic Biomarkers in Cerebrospinal fluid and Serum in patients with Parkinson's disease in Indian population. Sci Rep 2016; 6:35097. [PMID: 27752066 PMCID: PMC5067653 DOI: 10.1038/srep35097] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 09/22/2016] [Indexed: 12/28/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease with the absence of markers for diagnosis. Several studies on PD reported the elements imbalance in biofluids as biomarkers. However, their results remained inconclusive. This study integrates metallomics, multivariate and artificial neural network (ANN) to understand element variations in CSF and serum of PD patients from the largest cohort of Indian population to solve the inconsistent results of previous studies. Also, this study is aimed to (1) ascertain a common element signature between CSF and serum. (2) Assess cross sectional element variation with clinical symptoms. (3) Develop ANN models for rapid diagnosis. A metallomic profile of 110 CSF and 530 serum samples showed significant variations in 10 elements of CSF and six in serum of patients compared to controls. Consistent variations in elements pattern were noticed for Calcium, Magnesium and Iron in both the fluids of PD, which provides feasible diagnosis from serum. Furthermore, implementing multivariate analyses showed clear classification between normal and PD in both the fluids. Also, ANN provides 99% accuracy in detection of disease from CSF and serum. Overall, our analyses demonstrate that elements profile in biofluids of PD will be useful in development of diagnostic markers for PD.
Collapse
|
270
|
O'Hare Doig RL, Bartlett CA, Smith NM, Hodgetts SI, Dunlop SA, Hool L, Fitzgerald M. Specific combinations of ion channel inhibitors reduce excessive Ca 2+ influx as a consequence of oxidative stress and increase neuronal and glial cell viability in vitro. Neuroscience 2016; 339:450-462. [PMID: 27725216 DOI: 10.1016/j.neuroscience.2016.10.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 09/15/2016] [Accepted: 10/02/2016] [Indexed: 01/02/2023]
Abstract
Combinations of Ca2+ channel inhibitors have been proposed as an effective means to prevent excess Ca2+ flux and death of neurons and glia following neurotrauma in vivo. However, it is not yet known if beneficial outcomes such as improved viability have been due to direct effects on intracellular Ca2+ concentrations. Here, the effects of combinations of Lomerizine (Lom), 2,3-dioxo-7-(1H-imidazol-1-yl)6-nitro-1,2,3,4-tetrahydro-1-quinoxalinyl]acetic acid monohydrate (YM872), 3,5-dimethyl-1-adamantanamine (memantine (Mem)) and/or adenosine 5'-triphosphate periodate oxidized sodium salt (oxATP) to block voltage-gated Ca2+ channels, Ca2+ permeable α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, NMDA receptors and purinergic P2X7 receptors (P2X7R) respectively, on Ca2+ concentration and viability of rat primary mixed cortical (MC) cultures exposed to hydrogen peroxide (H2O2) insult, were assessed. The contribution of ryanodine-sensitive intracellular stores to intracellular Ca2+ concentration was also assessed. Live cell calcium imaging revealed that a 30min H2O2 insult induced a slow increase in intracellular Ca2+, in part from intracellular sources, associated with loss of cell viability by 6h. Most combinations of inhibitors that included oxATP significantly decreased Ca2+ influx and increased cell viability when administered simultaneously with H2O2. However, reductions in intracellular Ca2+ concentration were not always linked to improved cell viability. Examination of the density of specific cell subpopulations demonstrated that most combinations of inhibitors that included oxATP preserved NG2+ non-oligodendroglial cells, but preservation of astrocytes and neurons required additional inhibitors. Olig2+ oligodendroglia and ED-1+ activated microglia/macrophages were not preserved by any of the inhibitor combinations. These data indicate that following H2O2 insult, limiting intracellular Ca2+ entry via P2X7R is generally associated with increased cell viability. Protection of NG2+ non-oligodendroglial cells by Ca2+ channel inhibitor combinations may contribute to observed beneficial outcomes in vivo.
Collapse
Affiliation(s)
- Ryan L O'Hare Doig
- Experimental and Regenerative Neurosciences, The University of Western Australia, Crawley, Western Australia, Australia; School of Animal Biology, The University of Western Australia, Crawley, Western Australia, Australia; School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Carole A Bartlett
- Experimental and Regenerative Neurosciences, The University of Western Australia, Crawley, Western Australia, Australia; School of Animal Biology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Nicole M Smith
- Experimental and Regenerative Neurosciences, The University of Western Australia, Crawley, Western Australia, Australia; School of Animal Biology, The University of Western Australia, Crawley, Western Australia, Australia; School of Chemistry and Biochemistry, The University of Western Australia, Crawley, Western Australia, Australia
| | - Stuart I Hodgetts
- Experimental and Regenerative Neurosciences, The University of Western Australia, Crawley, Western Australia, Australia; School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Sarah A Dunlop
- Experimental and Regenerative Neurosciences, The University of Western Australia, Crawley, Western Australia, Australia; School of Animal Biology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Livia Hool
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Western Australia, Australia; Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
| | - Melinda Fitzgerald
- Experimental and Regenerative Neurosciences, The University of Western Australia, Crawley, Western Australia, Australia; School of Animal Biology, The University of Western Australia, Crawley, Western Australia, Australia.
| |
Collapse
|
271
|
Delgado-Camprubi M, Esteras N, Soutar MP, Plun-Favreau H, Abramov AY. Deficiency of Parkinson's disease-related gene Fbxo7 is associated with impaired mitochondrial metabolism by PARP activation. Cell Death Differ 2016; 24:120-131. [PMID: 27689878 PMCID: PMC5260490 DOI: 10.1038/cdd.2016.104] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 07/28/2016] [Accepted: 08/31/2016] [Indexed: 12/20/2022] Open
Abstract
The Parkinson's disease (PD)-related protein F-box only protein 7 (Fbxo7) is the substrate-recognition component of the Skp1-Cullin-F-box protein E3 ubiquitin ligase complex. We have recently shown that PD-associated mutations in Fbxo7 disrupt mitochondrial autophagy (mitophagy), suggesting a role for Fbxo7 in modulating mitochondrial homeostasis. Here we report that Fbxo7 deficiency is associated with reduced cellular NAD+ levels, which results in increased mitochondrial NADH redox index and impaired activity of complex I in the electron transport chain. Under these conditions of compromised respiration, mitochondrial membrane potential and ATP contents are reduced, and cytosolic reactive oxygen species (ROS) production is increased. ROS activates poly (ADP-ribose) polymerase (PARP) activity in Fbxo7-deficient cells. PARP inhibitor restores cellular NAD+ content and redox index and ATP pool, suggesting that PARP overactivation is cause of decreased complex I-driven respiration. These findings bring new insight into the mechanism of Fbxo7 deficiency, emphasising the importance of mitochondrial dysfunction in PD.
Collapse
Affiliation(s)
- Marta Delgado-Camprubi
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
| | - Noemi Esteras
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
| | - Marc Pm Soutar
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
| | - Helene Plun-Favreau
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
| | - Andrey Y Abramov
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
| |
Collapse
|
272
|
Farfán-García ED, Pérez-Rodríguez M, Espinosa-García C, Castillo-Mendieta NT, Maldonado-Castro M, Querejeta E, Trujillo-Ferrara JG, Soriano-Ursúa MA. Disruption of motor behavior and injury to the CNS induced by 3-thienylboronic acid in mice. Toxicol Appl Pharmacol 2016; 307:130-137. [PMID: 27495897 DOI: 10.1016/j.taap.2016.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 07/30/2016] [Accepted: 08/01/2016] [Indexed: 02/07/2023]
Abstract
The scarcity of studies on boron containing compounds (BCC) in the medicinal field is gradually being remedied. Efforts have been made to explore the effects of BCCs due to the properties that boron confers to molecules. Research has shown that the safety of some BCCs is similar to that found for boron-free compounds (judging from the acute toxicological evaluation). However, it has been observed that the administration of 3-thienylboronic acid (3TB) induced motor disruption in CD1 mice. In the current contribution we studied in deeper form the disruption of motor performance produced by the intraperitoneal administration of 3TB in mice from two strains (CD1 and C57BL6). Disruption of motor activity was dependent not only on the dose of 3TB administered, but also on the DMSO concentration in the vehicle. The ability of 3TB to enter the Central Nervous System (CNS) was evidenced by Raman spectroscopy as well as morphological effects on the CNS, such as loss of neurons yielding biased injury to the substantia nigra and striatum at doses ≥200mg/kg, and involving granular cell damage at doses of 400mg/kg but less injury in the motor cortex. Our work acquaints about the use of this compound in drug design, but the interesting profile as neurotoxic agent invite us to study it regarding the damage on the motor system.
Collapse
Affiliation(s)
- E D Farfán-García
- Academias de Fisiología Humana, Bioquímica y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, 11340 Ciudad de México, Mexico
| | - M Pérez-Rodríguez
- Academias de Fisiología Humana, Bioquímica y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, 11340 Ciudad de México, Mexico
| | - C Espinosa-García
- Departamento de Biología de la Reproducción, Universidad Autónoma Metropolitana (UAM), 09310 Ciudad de México, Mexico
| | - N T Castillo-Mendieta
- Academias de Fisiología Humana, Bioquímica y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, 11340 Ciudad de México, Mexico
| | - M Maldonado-Castro
- Academias de Fisiología Humana, Bioquímica y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, 11340 Ciudad de México, Mexico
| | - E Querejeta
- Academias de Fisiología Humana, Bioquímica y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, 11340 Ciudad de México, Mexico
| | - J G Trujillo-Ferrara
- Academias de Fisiología Humana, Bioquímica y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, 11340 Ciudad de México, Mexico
| | - M A Soriano-Ursúa
- Academias de Fisiología Humana, Bioquímica y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, 11340 Ciudad de México, Mexico.
| |
Collapse
|
273
|
Filippi-Chiela EC, Viegas MS, Thomé MP, Buffon A, Wink MR, Lenz G. Modulation of Autophagy by Calcium Signalosome in Human Disease. Mol Pharmacol 2016; 90:371-84. [PMID: 27436127 DOI: 10.1124/mol.116.105171] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 07/18/2016] [Indexed: 02/14/2025] Open
Abstract
Autophagy is a catabolic process that is largely regulated by extracellular and intracellular signaling pathways that are central to cellular metabolism and growth. Mounting evidence has shown that ion channels and transporters are important for basal autophagy functioning and influence autophagy to handle stressful situations. Besides its role in cell proliferation and apoptosis, intracellular Ca(2+) is widely recognized as a key regulator of autophagy, acting through the modulation of pathways such as the mechanistic target of rapamycin complex 1, calcium/calmodulin-dependent protein kinase kinase 2, and protein kinase C. Proper spatiotemporal Ca(2+) availability, coupled with a controlled ionic flow among the extracellular milieu, storage compartments, and the cytosol, is critical in determining the role played by Ca(2+) on autophagy and on cell fate. The crosstalk between Ca(2+) and autophagy has a central role in cellular homeostasis and survival during several physiologic and pathologic conditions. Here we review the main findings concerning the mechanisms and roles of Ca(2+)-modulated autophagy, focusing on human disorders ranging from cancer to neurologic diseases and immunity. By identifying mechanisms, players, and pathways that either induce or suppress autophagy, new promising approaches for preventing and treating human disorders emerge, including those based on the modulation of Ca(2+)-mediated autophagy.
Collapse
Affiliation(s)
- Eduardo Cremonese Filippi-Chiela
- Graduate Program in Hepatology and Gastroenterology, Faculty of Medicine (E.C.F.-C.), and Gene Therapy Center (M.S.V.), Hospital de Clínicas de Porto Alegre; Department of Biophysics and Center of Biotechnology (M.P.T., G.L.) and Laboratory of Biochemical and Cytological Analysis, Faculty of Pharmacy (M.R.W.), Federal University of Rio Grande do Sul (UFRGS); and Department of Health Sciences and Cell Biology Laboratory, Federal University of Health Sciences of Porto Alegre (A.B.), Porto Allegre, Brazil
| | - Michelle S Viegas
- Graduate Program in Hepatology and Gastroenterology, Faculty of Medicine (E.C.F.-C.), and Gene Therapy Center (M.S.V.), Hospital de Clínicas de Porto Alegre; Department of Biophysics and Center of Biotechnology (M.P.T., G.L.) and Laboratory of Biochemical and Cytological Analysis, Faculty of Pharmacy (M.R.W.), Federal University of Rio Grande do Sul (UFRGS); and Department of Health Sciences and Cell Biology Laboratory, Federal University of Health Sciences of Porto Alegre (A.B.), Porto Allegre, Brazil
| | - Marcos Paulo Thomé
- Graduate Program in Hepatology and Gastroenterology, Faculty of Medicine (E.C.F.-C.), and Gene Therapy Center (M.S.V.), Hospital de Clínicas de Porto Alegre; Department of Biophysics and Center of Biotechnology (M.P.T., G.L.) and Laboratory of Biochemical and Cytological Analysis, Faculty of Pharmacy (M.R.W.), Federal University of Rio Grande do Sul (UFRGS); and Department of Health Sciences and Cell Biology Laboratory, Federal University of Health Sciences of Porto Alegre (A.B.), Porto Allegre, Brazil
| | - Andreia Buffon
- Graduate Program in Hepatology and Gastroenterology, Faculty of Medicine (E.C.F.-C.), and Gene Therapy Center (M.S.V.), Hospital de Clínicas de Porto Alegre; Department of Biophysics and Center of Biotechnology (M.P.T., G.L.) and Laboratory of Biochemical and Cytological Analysis, Faculty of Pharmacy (M.R.W.), Federal University of Rio Grande do Sul (UFRGS); and Department of Health Sciences and Cell Biology Laboratory, Federal University of Health Sciences of Porto Alegre (A.B.), Porto Allegre, Brazil
| | - Marcia R Wink
- Graduate Program in Hepatology and Gastroenterology, Faculty of Medicine (E.C.F.-C.), and Gene Therapy Center (M.S.V.), Hospital de Clínicas de Porto Alegre; Department of Biophysics and Center of Biotechnology (M.P.T., G.L.) and Laboratory of Biochemical and Cytological Analysis, Faculty of Pharmacy (M.R.W.), Federal University of Rio Grande do Sul (UFRGS); and Department of Health Sciences and Cell Biology Laboratory, Federal University of Health Sciences of Porto Alegre (A.B.), Porto Allegre, Brazil
| | - Guido Lenz
- Graduate Program in Hepatology and Gastroenterology, Faculty of Medicine (E.C.F.-C.), and Gene Therapy Center (M.S.V.), Hospital de Clínicas de Porto Alegre; Department of Biophysics and Center of Biotechnology (M.P.T., G.L.) and Laboratory of Biochemical and Cytological Analysis, Faculty of Pharmacy (M.R.W.), Federal University of Rio Grande do Sul (UFRGS); and Department of Health Sciences and Cell Biology Laboratory, Federal University of Health Sciences of Porto Alegre (A.B.), Porto Allegre, Brazil
| |
Collapse
|
274
|
Emerging (and converging) pathways in Parkinson's disease: keeping mitochondrial wellness. Biochem Biophys Res Commun 2016; 483:1020-1030. [PMID: 27581196 DOI: 10.1016/j.bbrc.2016.08.153] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 08/25/2016] [Accepted: 08/27/2016] [Indexed: 12/31/2022]
Abstract
The selective cell loss in the ventral component of the substantia nigra pars compacta and the presence of alpha-synuclein (α-syn)-rich intraneuronal inclusions called Lewy bodies are the pathological hallmarks of Parkinson's disease (PD), the most common motor system disorder whose aetiology remains largely elusive. Although most cases of PD are idiopathic, there are rare familial forms of the disease that can be traced to single gene mutations that follow Mendelian inheritance pattern. The study of several nuclear encoded proteins whose mutations are linked to the development of autosomal recessive and dominant forms of familial PD enhanced our understanding of biochemical and cellular mechanisms contributing to the disease and suggested that many signs of neurodegeneration result from compromised mitochondrial function. Here we present an overview of the current understanding of PD-related mitochondrial dysfunction including defects in bioenergetics and Ca2+ homeostasis, mitochondrial DNA mutations, altered mitochondrial dynamics and autophagy. We emphasize, in particular, the convergence of many "apparently" different pathways towards a common route involving mitochondria. Understanding whether mitochondrial dysfunction in PD represents the cause or the consequence of the disease is challenging and will help to define the pathogenic processes at the basis of the PD onset and progression.
Collapse
|
275
|
The Cytomegalovirus protein pUL37×1 targets mitochondria to mediate neuroprotection. Sci Rep 2016; 6:31373. [PMID: 27562039 PMCID: PMC4999870 DOI: 10.1038/srep31373] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 07/19/2016] [Indexed: 12/22/2022] Open
Abstract
There is substantial evidence that mitochondrial dysfunction plays a significant role in the pathogenesis of Parkinson disease (PD). This contribution probably encompasses defects of oxidative phosphorylation, mitochondrial turnover (mitophagy), mitochondrial derived oxidative stress, and apoptotic signalling. Human cytomegalovirus immediate-early protein pUL37 × 1 induces Bax mitochondrial translocation and inactivation to prevent apoptosis. Over-expressing pUL37 × 1 in neuronal cells protects against staurosporin and 6-hydroxydopamine induced apoptosis and cell death. Protection is not enhanced by bax silencing in pUL37 × 1 over-expressing cells, suggesting a bax-dependent mechanism of action. pUL37 × 1 increases glycolysis and induces mitochondrial hyperpolarization, a bax independent anti-apoptotic action. pUL37 × 1 increases glycolysis through activation of phosphofructokinase by a calcium-dependent pathway. The dual anti-apoptotic mechanism of pUL37 × 1 may be considered a novel neuroprotective strategy in diseases where mitochondrial dysfunction and apoptotic pathways are involved.
Collapse
|
276
|
Coherent and Contradictory Facts, Feats and Fictions Associated with Metal Accumulation in Parkinson's Disease: Epicenter or Outcome, Yet a Demigod Question. Mol Neurobiol 2016; 54:4738-4755. [PMID: 27480264 DOI: 10.1007/s12035-016-0016-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 07/12/2016] [Indexed: 01/30/2023]
Abstract
Unwarranted exposure due to liberal use of metals for maintaining the lavish life and to achieve the food demand for escalating population along with an incredible boost in the average human life span owing to orchestrated progress in rejuvenation therapy have gradually increased the occurrence of Parkinson's disease (PD). Etiology is albeit elusive; association of PD with metal accumulation has never been overlooked due to noteworthy similitude between metal-exposure symptoms and a few cardinal features of disease. Even though metals are entailed in the vital functions, a hysterical shift, primarily augmentation, escorts the stern nigrostriatal dopaminergic neurodegeneration. An increase in the passage of metals through the blood brain barrier and impaired metabolic activity and elimination system could lead to metal accumulation in the brain, which eventually makes dopaminergic neurons quite susceptible. In the present article, an update on implication of metal accumulation in PD/Parkinsonism has been provided. Moreover, encouraging and paradoxical facts and fictions associated with metal accumulation in PD/Parkinsonism have also been compiled. Systematic literature survey of PD is performed to describe updated information if metal accumulation is an epicenter or merely an outcome. Finally, a perspective on the association of metal accumulation with pesticide-induced Parkinsonism has been explained to unveil the likely impact of the former in the latter.
Collapse
|
277
|
Abstract
The power of Drosophila genetics has attracted attention in tackling important biomedical challenges such as the understanding and prevention of neurodegenerative diseases. Parkinson's disease (PD) is the most common neurodegenerative movement disorder which results from the relentless degeneration of midbrain dopaminergic neurons. Over the past two decades tremendous advances have been made in identifying genes responsible for inherited forms of PD. The ease of genetic manipulation in Drosophila has spurred the development of numerous models of PD, including expression of human genes carrying pathogenic mutations or the targeted mutation of conserved orthologs. The genetic and cellular analysis of these models is beginning to reveal fundamental insights into the pathogenic mechanisms. Numerous pathways and processes are disrupted in these models but some common themes are emerging. These often implicate aberrant synaptic function, protein aggregation, autophagy, oxidative stress, and mitochondrial dysfunction. Moreover, an impressive list of small molecule compounds have been identified as effective in reversing pathogenic phenotypes, paving the way to explore these for therapeutic interventions.
Collapse
Affiliation(s)
- V L Hewitt
- Medical Research Council Mitochondrial Biology Unit, Cambridge, United Kingdom
| | - A J Whitworth
- Medical Research Council Mitochondrial Biology Unit, Cambridge, United Kingdom.
| |
Collapse
|
278
|
Lee S, Yang M, Kim J, Kang S, Kim J, Kim JC, Jung C, Shin T, Kim SH, Moon C. Trimethyltin-induced hippocampal neurodegeneration: A mechanism-based review. Brain Res Bull 2016; 125:187-99. [PMID: 27450702 DOI: 10.1016/j.brainresbull.2016.07.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 07/19/2016] [Indexed: 12/22/2022]
Abstract
Trimethyltin (TMT), a toxic organotin compound, induces neurodegeneration selectively involving the limbic system and especially prominent in the hippocampus. Neurodegeneration-associated behavioral abnormalities, such as hyperactivity, aggression, cognitive deficits, and epileptic seizures, occur in both exposed humans and experimental animal models. Previously, TMT had been used generally in industry and agriculture, but the use of TMT has been limited because of its dangers to people. TMT has also been used to make a promising in vivo rodent model of neurodegeneration because of its region-specific characteristics. Several studies have demonstrated that TMT-treated animal models of epileptic seizures can be used as tools for researching hippocampus-specific neurotoxicity as well as the molecular mechanisms leading to hippocampal neurodegeneration. This review summarizes the in vivo and in vitro underlying mechanisms of TMT-induced hippocampal neurodegeneration (oxidative stress, inflammatory responses, and neuronal death/survival). Thus, the present review may be helpful to provide general insights into TMT-induced neurodegeneration and approaches to therapeutic interventions for neurodegenerative diseases, including temporal lobe epilepsy.
Collapse
Affiliation(s)
- Sueun Lee
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186, South Korea
| | - Miyoung Yang
- Department of Anatomy, School of Medicine and Institute for Environmental Science, Wonkwang University, Jeonbuk 54538, South Korea
| | - Jinwook Kim
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186, South Korea
| | - Sohi Kang
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186, South Korea
| | - Juhwan Kim
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186, South Korea
| | - Jong-Choon Kim
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186, South Korea
| | - Chaeyong Jung
- Department of Anatomy, Chonnam National University Medical School, Gwangju 61469, South Korea
| | - Taekyun Shin
- Department of Veterinary Anatomy, College of Veterinary Medicine, Jeju National University, Jeju 63243, South Korea
| | - Sung-Ho Kim
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186, South Korea
| | - Changjong Moon
- Departments of Veterinary Anatomy and Veterinary Toxicology, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju 61186, South Korea.
| |
Collapse
|
279
|
Agnihotri S, Golbourn B, Huang X, Remke M, Younger S, Cairns RA, Chalil A, Smith CA, Krumholtz SL, Mackenzie D, Rakopoulos P, Ramaswamy V, Taccone MS, Mischel PS, Fuller GN, Hawkins C, Stanford WL, Taylor MD, Zadeh G, Rutka JT. PINK1 Is a Negative Regulator of Growth and the Warburg Effect in Glioblastoma. Cancer Res 2016; 76:4708-19. [PMID: 27325644 DOI: 10.1158/0008-5472.can-15-3079] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 06/02/2016] [Indexed: 11/16/2022]
Abstract
Proliferating cancer cells are characterized by high rates of glycolysis, lactate production, and altered mitochondrial metabolism. This metabolic reprogramming provides important metabolites for proliferation of tumor cells, including glioblastoma. These biological processes, however, generate oxidative stress that must be balanced through detoxification of reactive oxygen species (ROS). Using an unbiased retroviral loss-of-function screen in nontransformed human astrocytes, we demonstrate that mitochondrial PTEN-induced kinase 1 (PINK1) is a regulator of the Warburg effect and negative regulator of glioblastoma growth. We report that loss of PINK1 contributes to the Warburg effect through ROS-dependent stabilization of hypoxia-inducible factor-1A and reduced pyruvate kinase muscle isozyme 2 activity, both key regulators of aerobic glycolysis. Mechanistically, PINK1 suppresses ROS and tumor growth through FOXO3a, a master regulator of oxidative stress and superoxide dismutase 2. These findings highlight the importance of PINK1 and ROS balance in normal and tumor cells. PINK1 loss was observed in a significant number of human brain tumors including glioblastoma (n > 900) and correlated with poor patient survival. PINK1 overexpression attenuates in vivo glioblastoma growth in orthotopic mouse xenograft models and a transgenic glioblastoma model in Drosophila Cancer Res; 76(16); 4708-19. ©2016 AACR.
Collapse
Affiliation(s)
- Sameer Agnihotri
- The Arthur and Sonia Labatt Brain Tumor Research Centre, Hospital for Sick Children, Toronto, Ontario, Canada. The Arthur and Sonia Labatt Brain Tumor Research Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Brian Golbourn
- The Arthur and Sonia Labatt Brain Tumor Research Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Xi Huang
- The Arthur and Sonia Labatt Brain Tumor Research Centre, Hospital for Sick Children, Toronto, Ontario, Canada. Department of Physiology, Howard Hughes Medical Institute, University of California, San Francisco, California. Department of Biophysics, Howard Hughes Medical Institute, University of California, San Francisco, California. Department of Biochemistry, Howard Hughes Medical Institute, University of California, San Francisco, California
| | - Marc Remke
- The Arthur and Sonia Labatt Brain Tumor Research Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Susan Younger
- Department of Physiology, Howard Hughes Medical Institute, University of California, San Francisco, California. Department of Biophysics, Howard Hughes Medical Institute, University of California, San Francisco, California. Department of Biochemistry, Howard Hughes Medical Institute, University of California, San Francisco, California
| | - Rob A Cairns
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Alan Chalil
- The Arthur and Sonia Labatt Brain Tumor Research Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Christian A Smith
- The Arthur and Sonia Labatt Brain Tumor Research Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Stacey-Lynn Krumholtz
- The Arthur and Sonia Labatt Brain Tumor Research Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Danielle Mackenzie
- The Arthur and Sonia Labatt Brain Tumor Research Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Patricia Rakopoulos
- The Arthur and Sonia Labatt Brain Tumor Research Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Vijay Ramaswamy
- The Arthur and Sonia Labatt Brain Tumor Research Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Michael S Taccone
- The Arthur and Sonia Labatt Brain Tumor Research Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Paul S Mischel
- Ludwig Institute for Cancer Research, University of California, San Diego, California
| | - Gregory N Fuller
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cynthia Hawkins
- The Arthur and Sonia Labatt Brain Tumor Research Centre, Hospital for Sick Children, Toronto, Ontario, Canada. Department of Pathology, Hospital for Sick Children, Ontario, Canada
| | - William L Stanford
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research, Ontario, Canada
| | - Michael D Taylor
- The Arthur and Sonia Labatt Brain Tumor Research Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Gelareh Zadeh
- The Arthur and Sonia Labatt Brain Tumor Research Centre, Hospital for Sick Children, Toronto, Ontario, Canada. Department of Biophysics, Howard Hughes Medical Institute, University of California, San Francisco, California. Department of Neurosurgery, Toronto Western Hospital, Ontario, Canada. Gelareh Zadeh, University Health Network, Toronto, Ontario, Canada
| | - James T Rutka
- The Arthur and Sonia Labatt Brain Tumor Research Centre, Hospital for Sick Children, Toronto, Ontario, Canada. RS McLaughlin, Professor and Chairman, University of Toronto, Toronto, Ontario, Canada. Department of Surgery, Hospital for Sick Children, Ontario, Canada.
| |
Collapse
|
280
|
Glucose Metabolism and AMPK Signaling Regulate Dopaminergic Cell Death Induced by Gene (α-Synuclein)-Environment (Paraquat) Interactions. Mol Neurobiol 2016; 54:3825-3842. [PMID: 27324791 DOI: 10.1007/s12035-016-9906-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 05/03/2016] [Indexed: 10/21/2022]
Abstract
While environmental exposures are not the single cause of Parkinson's disease (PD), their interaction with genetic alterations is thought to contribute to neuronal dopaminergic degeneration. However, the mechanisms involved in dopaminergic cell death induced by gene-environment interactions remain unclear. In this work, we have revealed for the first time the role of central carbon metabolism and metabolic dysfunction in dopaminergic cell death induced by the paraquat (PQ)-α-synuclein interaction. The toxicity of PQ in dopaminergic N27 cells was significantly reduced by glucose deprivation, inhibition of hexokinase with 2-deoxy-D-glucose (2-DG), or equimolar substitution of glucose with galactose, which evidenced the contribution of glucose metabolism to PQ-induced cell death. PQ also stimulated an increase in glucose uptake, and in the levels of glucose transporter type 4 (GLUT4) and Na+-glucose transporters isoform 1 (SGLT1) proteins, but only inhibition of GLUT-like transport with STF-31 or ascorbic acid reduced PQ-induced cell death. Importantly, while autophagy protein 5 (ATG5)/unc-51 like autophagy activating kinase 1 (ULK1)-dependent autophagy protected against PQ toxicity, the inhibitory effect of glucose deprivation on cell death progression was largely independent of autophagy or mammalian target of rapamycin (mTOR) signaling. PQ selectively induced metabolomic alterations and adenosine monophosphate-activated protein kinase (AMPK) activation in the midbrain and striatum of mice chronically treated with PQ. Inhibition of AMPK signaling led to metabolic dysfunction and an enhanced sensitivity of dopaminergic cells to PQ. In addition, activation of AMPK by PQ was prevented by inhibition of the inducible nitric oxide syntase (iNOS) with 1400W, but PQ had no effect on iNOS levels. Overexpression of wild type or A53T mutant α-synuclein stimulated glucose accumulation and PQ toxicity, and this toxic synergism was reduced by inhibition of glucose metabolism/transport and the pentose phosphate pathway (6-aminonicotinamide). These results demonstrate that glucose metabolism and AMPK regulate dopaminergic cell death induced by gene (α-synuclein)-environment (PQ) interactions.
Collapse
|
281
|
Genetic variations associated with six-white-point coat pigmentation in Diannan small-ear pigs. Sci Rep 2016; 6:27534. [PMID: 27270507 PMCID: PMC4897638 DOI: 10.1038/srep27534] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 05/18/2016] [Indexed: 11/08/2022] Open
Abstract
A common phenotypic difference among domestic animals is variation in coat color. Six-white-point is a pigmentation pattern observed in varying pig breeds, which seems to have evolved through several different mechanistic pathways. Herein, we re-sequenced whole genomes of 31 Diannan small-ear pigs from China and found that the six-white-point coat color in Diannan small-ear pigs is likely regulated by polygenic loci, rather than by the MC1R locus. Strong associations were observed at three loci (EDNRB, CNTLN, and PINK1), which explain about 20 percent of the total coat color variance in the Diannan small-ear pigs. We found a mutation that is highly differentiated between six-white-point and black Diannan small-ear pigs, which is located in a conserved noncoding sequence upstream of the EDNRB gene and is a putative binding site of the CEBPB protein. This study advances our understanding of coat color evolution in Diannan small-ear pigs and expands our traditional knowledge of coat color being a monogenic trait.
Collapse
|
282
|
Voigt A, Berlemann LA, Winklhofer KF. The mitochondrial kinase PINK1: functions beyond mitophagy. J Neurochem 2016; 139 Suppl 1:232-239. [PMID: 27251035 DOI: 10.1111/jnc.13655] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 04/05/2016] [Accepted: 04/22/2016] [Indexed: 11/29/2022]
Abstract
Mutations in the genes encoding the mitochondrial kinase PINK1 and the E3 ubiquitin ligase Parkin cause autosomal recessive Parkinson's disease (PD). Pioneering work in Drosophila melanogaster revealed that the loss of PINK1 or Parkin function causes similar phenotypes including dysfunctional mitochondria. Further research showed that PINK1 can act upstream of Parkin in a mitochondrial quality control pathway to induce removal of damaged mitochondria in a process called mitophagy. Albeit the PINK1/Parkin-induced mitophagy pathway is well established and has recently been elucidated in great detail, its pathophysiological relevance is being debated. Mounting evidence indicates that PINK1 has additional functions, for example, in regulating complex I activity and maintaining neuronal viability in response to stress. Here, we discuss mitophagy-dependent and -independent functions of PINK1 and their possible role in PD pathogenesis. Mutations in the PINK1 gene, encoding a mitochondrial kinase, are associated with autosomal recessive Parkinson's disease. In this review, we summarize and discuss the functional roles of PINK1 in maintaining mitochondrial integrity, eliminating damaged mitochondria, and promoting cell survival. This article is part of a special issue on Parkinson disease.
Collapse
Affiliation(s)
- Aaron Voigt
- Department of Neurology, University Hospital, RWTH Aachen University, Aachen, Germany.
| | - Lena A Berlemann
- Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Germany
| | - Konstanze F Winklhofer
- Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Germany.
| |
Collapse
|
283
|
Gautier CA, Erpapazoglou Z, Mouton-Liger F, Muriel MP, Cormier F, Bigou S, Duffaure S, Girard M, Foret B, Iannielli A, Broccoli V, Dalle C, Bohl D, Michel PP, Corvol JC, Brice A, Corti O. The endoplasmic reticulum-mitochondria interface is perturbed in PARK2 knockout mice and patients with PARK2 mutations. Hum Mol Genet 2016; 25:2972-2984. [PMID: 27206984 DOI: 10.1093/hmg/ddw148] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 05/10/2016] [Accepted: 05/10/2016] [Indexed: 12/13/2022] Open
Abstract
Mutations in PARK2, encoding the E3 ubiquitin protein ligase Parkin, are a common cause of autosomal recessive Parkinson's disease (PD). Loss of PARK2 function compromises mitochondrial quality by affecting mitochondrial biogenesis, bioenergetics, dynamics, transport and turnover. We investigated the impact of PARK2 dysfunction on the endoplasmic reticulum (ER)-mitochondria interface, which mediates calcium (Ca2+) exchange between the two compartments and is essential for Parkin-dependent mitophagy. Confocal and electron microscopy analyses showed the ER and mitochondria to be in closer proximity in primary fibroblasts from PARK2 knockout (KO) mice and PD patients with PARK2 mutations than in controls. Ca2+ flux to the cytosol was also modified, due to enhanced ER-to-mitochondria Ca2+ transfers, a change that was also observed in neurons derived from induced pluripotent stem cells of a patient with PARK2 mutations. Subcellular fractionation showed the abundance of the Parkin substrate mitofusin 2 (Mfn2), which is known to modulate the ER-mitochondria interface, to be specifically higher in the mitochondrion-associated ER membrane compartment in PARK2 KO tissue. Mfn2 downregulation or the exogenous expression of normal Parkin restored cytosolic Ca2+ transients in fibroblasts from patients with PARK2 mutations. In contrast, a catalytically inactive PD-related Parkin variant had no effect. Overall, our data suggest that Parkin is directly involved in regulating ER-mitochondria contacts and provide new insight into the role of the loss of Parkin function in PD development.
Collapse
Affiliation(s)
- Clément A Gautier
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Inserm, U1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, CNRS, UMR 7225, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
| | - Zoi Erpapazoglou
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Inserm, U1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, CNRS, UMR 7225, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
| | - François Mouton-Liger
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Inserm, U1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, CNRS, UMR 7225, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
| | - Marie Paule Muriel
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Inserm, U1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, CNRS, UMR 7225, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
| | - Florence Cormier
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Inserm, U1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, CNRS, UMR 7225, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
- National Research Council (CNR), Institute of Neuroscience, Milan, Italy
| | - Stéphanie Bigou
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Inserm, U1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, CNRS, UMR 7225, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
| | - Sophie Duffaure
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Inserm, U1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, CNRS, UMR 7225, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
| | - Mathilde Girard
- CECS, I-Stem, AFM, Institute of Stem Cell Therapy and Exploration of Monogenic Diseases, 91030 Evry cedex, France
| | - Benjamin Foret
- CECS, I-Stem, AFM, Institute of Stem Cell Therapy and Exploration of Monogenic Diseases, 91030 Evry cedex, France
| | - Angelo Iannielli
- National Research Council (CNR), Institute of Neuroscience, Milan, Italy
- Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Vania Broccoli
- National Research Council (CNR), Institute of Neuroscience, Milan, Italy
- Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Carine Dalle
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Inserm, U1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, CNRS, UMR 7225, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
| | - Delphine Bohl
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Inserm, U1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, CNRS, UMR 7225, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
| | - Patrick P Michel
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Inserm, U1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, CNRS, UMR 7225, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
| | - Jean-Christophe Corvol
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Inserm, U1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, CNRS, UMR 7225, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
- Assistance-publique Hôpitaux de Paris, Inserm, CIC-1422, Department of Neurology, Hôpital Pitié-Salpêtrière, F-75013 Paris, France
| | - Alexis Brice
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Inserm, U1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, CNRS, UMR 7225, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
| | - Olga Corti
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Inserm, U1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, CNRS, UMR 7225, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France
- Bases moléculaires, physiopathologie et traitement des maladies neurodégénératives, Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
| |
Collapse
|
284
|
Chung KM, Jeong EJ, Park H, An HK, Yu SW. Mediation of Autophagic Cell Death by Type 3 Ryanodine Receptor (RyR3) in Adult Hippocampal Neural Stem Cells. Front Cell Neurosci 2016; 10:116. [PMID: 27199668 PMCID: PMC4858590 DOI: 10.3389/fncel.2016.00116] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 04/21/2016] [Indexed: 11/15/2022] Open
Abstract
Cytoplasmic Ca2+ actively engages in diverse intracellular processes from protein synthesis, folding and trafficking to cell survival and death. Dysregulation of intracellular Ca2+ levels is observed in various neuropathological states including Alzheimer’s and Parkinson’s diseases. Ryanodine receptors (RyRs) and inositol 1,4,5-triphosphate receptors (IP3Rs), the main Ca2+ release channels located in endoplasmic reticulum (ER) membranes, are known to direct various cellular events such as autophagy and apoptosis. Here we investigated the intracellular Ca2+-mediated regulation of survival and death of adult hippocampal neural stem (HCN) cells utilizing an insulin withdrawal model of autophagic cell death (ACD). Despite comparable expression levels of RyR and IP3R transcripts in HCN cells at normal state, the expression levels of RyRs—especially RyR3—were markedly upregulated upon insulin withdrawal. While treatment with the RyR agonist caffeine significantly promoted the autophagic death of insulin-deficient HCN cells, treatment with its inhibitor dantrolene prevented the induction of autophagy following insulin withdrawal. Furthermore, CRISPR/Cas9-mediated knockout of the RyR3 gene abolished ACD of HCN cells. This study delineates a distinct, RyR3-mediated ER Ca2+ regulation of autophagy and programmed cell death in neural stem cells. Our findings provide novel insights into the critical, yet understudied mechanisms underlying the regulatory function of ER Ca2+ in neural stem cell biology.
Collapse
Affiliation(s)
- Kyung Min Chung
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST) Daegu, South Korea
| | - Eun-Ji Jeong
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST) Daegu, South Korea
| | - Hyunhee Park
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST) Daegu, South Korea
| | - Hyun-Kyu An
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST) Daegu, South Korea
| | - Seong-Woon Yu
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST) Daegu, South Korea
| |
Collapse
|
285
|
Meclizine-induced enhanced glycolysis is neuroprotective in Parkinson disease cell models. Sci Rep 2016; 6:25344. [PMID: 27145922 PMCID: PMC4857109 DOI: 10.1038/srep25344] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 03/21/2016] [Indexed: 11/08/2022] Open
Abstract
Meclizine is a well-tolerated drug routinely used as an anti-histamine agent in the management of disequilibrium. Recently, meclizine has been assessed for its neuroprotective properties in ischemic stroke and Huntington disease models. We found that meclizine protected against 6-hydroxydopamine-induced apoptosis and cell death in both SH-SY5Y cells and rat primary cortical cultures. Meclizine increases the level of 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3), which activates phosphofructokinase, a rate-determining enzyme of glycolysis. This protection is therefore mediated by meclizine’s ability to enhance glycolysis and increase mitochondrial hyperpolarization. Meclizine represents an interesting candidate for further investigation to re-purpose for its potential to be neuroprotective in Parkinson disease.
Collapse
|
286
|
Parpura V, Sekler I, Fern R. Plasmalemmal and mitochondrial Na+-Ca2+exchange in neuroglia. Glia 2016; 64:1646-54. [DOI: 10.1002/glia.22975] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 01/07/2016] [Accepted: 01/25/2016] [Indexed: 12/20/2022]
Affiliation(s)
- Vladimir Parpura
- Department of Neurobiology; Center for Glial Biology in Medicine, Civitan International Research Center, Atomic Force Microscopy & Nanotechnology Laboratories, and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham; Birmingham Alabama
| | - Israel Sekler
- Department of Physiology, Faculty of Health Science; Ben-Gurion University; Ben-Guion Av 84105 POB 653
| | - Robert Fern
- Peninsular School of Medicine and Dentistry; University of Plymouth; Plymouth PL6 8BU United Kingdom
| |
Collapse
|
287
|
Esteras N, Dinkova-Kostova AT, Abramov AY. Nrf2 activation in the treatment of neurodegenerative diseases: a focus on its role in mitochondrial bioenergetics and function. Biol Chem 2016; 397:383-400. [PMID: 26812787 DOI: 10.1515/hsz-2015-0295] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 01/07/2016] [Indexed: 12/16/2022]
Abstract
The nuclear factor erythroid-derived 2 (NF-E2)-related factor 2 (Nrf2) is a transcription factor well-known for its function in controlling the basal and inducible expression of a variety of antioxidant and detoxifying enzymes. As part of its cytoprotective activity, increasing evidence supports its role in metabolism and mitochondrial bioenergetics and function. Neurodegenerative diseases are excellent candidates for Nrf2-targeted treatments. Most neurodegenerative conditions such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, frontotemporal dementia and Friedreich's ataxia are characterized by oxidative stress, misfolded protein aggregates, and chronic inflammation, the common targets of Nrf2 therapeutic strategies. Together with them, mitochondrial dysfunction is implicated in the pathogenesis of most neurodegenerative disorders. The recently recognized ability of Nrf2 to regulate intermediary metabolism and mitochondrial function makes Nrf2 activation an attractive and comprehensive strategy for the treatment of neurodegenerative disorders. This review aims to focus on the potential therapeutic role of Nrf2 activation in neurodegeneration, with special emphasis on mitochondrial bioenergetics and function, metabolism and the role of transporters, all of which collectively contribute to the cytoprotective activity of this transcription factor.
Collapse
|
288
|
|
289
|
Regulation of mitochondrial functions by protein phosphorylation and dephosphorylation. Cell Biosci 2016; 6:25. [PMID: 27087918 PMCID: PMC4832502 DOI: 10.1186/s13578-016-0089-3] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 04/01/2016] [Indexed: 12/02/2022] Open
Abstract
The mitochondria are double membrane-bound organelles found in most eukaryotic cells. They generate most of the cell’s energy supply of adenosine triphosphate (ATP). Protein phosphorylation and dephosphorylation are critical mechanisms in the regulation of cell signaling networks and are essential for almost all the cellular functions. For many decades, mitochondria were considered autonomous organelles merely functioning to generate energy for cells to survive and proliferate, and were thought to be independent of the cellular signaling networks. Consequently, phosphorylation and dephosphorylation processes of mitochondrial kinases and phosphatases were largely neglected. However, evidence accumulated in recent years on mitochondria-localized kinases/phosphatases has changed this longstanding view. Mitochondria are increasingly recognized as a hub for cell signaling, and many kinases and phosphatases have been reported to localize in mitochondria and play important functions. However, the strength of the evidence on mitochondrial localization and the activities of the reported kinases and phosphatases vary greatly, and the detailed mechanisms on how these kinases/phosphatases translocate to mitochondria, their subsequent function, and the physiological and pathological implications of their localization are still poorly understood. Here, we provide an updated perspective on the recent advancement in this area, with an emphasis on the implications of mitochondrial kinases/phosphatases in cancer and several other diseases.
Collapse
|
290
|
Hsu P, Liu X, Zhang J, Wang HG, Ye JM, Shi Y. Cardiolipin remodeling by TAZ/tafazzin is selectively required for the initiation of mitophagy. Autophagy 2016; 11:643-52. [PMID: 25919711 DOI: 10.1080/15548627.2015.1023984] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Tafazzin (TAZ) is a phospholipid transacylase that catalyzes the remodeling of cardiolipin, a mitochondrial phospholipid required for oxidative phosphorylation. Mutations of TAZ cause Barth syndrome, which is characterized by mitochondrial dysfunction and dilated cardiomyopathy, leading to premature death. However, the molecular mechanisms underlying the cause of mitochondrial dysfunction in Barth syndrome remain poorly understood. Here we investigated the role of TAZ in regulating mitochondrial function and mitophagy. Using primary mouse embryonic fibroblasts (MEFs) with doxycycline-inducible knockdown of Taz, we showed that TAZ deficiency in MEFs caused defective mitophagosome biogenesis, but not other autophagic processes. Consistent with a key role of mitophagy in mitochondria quality control, TAZ deficiency in MEFs also led to impaired oxidative phosphorylation and severe oxidative stress. Together, these findings provide key insights on mitochondrial dysfunction in Barth syndrome, suggesting that pharmacological restoration of mitophagy may provide a novel treatment for this lethal condition.
Collapse
Key Words
- AdGFP-LC3, recombinant adenovirus expressing GFP tagged MAP1LC3B
- AdTAZ, recombinant adenovirus expressing Myc-tagged TAZ
- BTHS, Barth syndrome
- BafA1, bafilomycin A1
- Barth syndrome
- CCCP, carbonyl cyanide m-chlorophenylhydrazone
- CL, cardiolipin
- Dox, doxycycline
- FCCP, carbonyl cyanide p-triflouromethoxyphenylhydrazone
- LTG, LysoTracker Green
- MAP1LC3B/LC3B, microtubule-associated protein 1 light chain 3 beta
- MEF, mouse embryonic fibroblast
- MLCL, monolysocardiolipin
- MTR, MitoTracker Red
- PARK2, parkin RBR E3 ubiquitin protein ligase
- PINK1, PTEN-induced putative kinase 1
- SOD2, superoxide dismutase 2 mitochondrial
- TAZ, tafazzin
- TLCL, tetralinoleoyl-cardiolipin
- autophagy
- cardiolipin
- mitochondrial dysfunction
- mitophagosome
- mitophagy
- tafazzin
Collapse
Affiliation(s)
- Paul Hsu
- a Department of Cellular and Molecular Physiology ; Hershey , PA USA
| | | | | | | | | | | |
Collapse
|
291
|
The Dual Function of Reactive Oxygen/Nitrogen Species in Bioenergetics and Cell Death: The Role of ATP Synthase. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:3869610. [PMID: 27034734 PMCID: PMC4806282 DOI: 10.1155/2016/3869610] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 02/15/2016] [Indexed: 01/11/2023]
Abstract
Reactive oxygen species (ROS) and reactive nitrogen species (RNS) targeting mitochondria are major causative factors in disease pathogenesis. The mitochondrial permeability transition pore (PTP) is a mega-channel modulated by calcium and ROS/RNS modifications and it has been described to play a crucial role in many pathophysiological events since prolonged channel opening causes cell death. The recent identification that dimers of ATP synthase form the PTP and the fact that posttranslational modifications caused by ROS/RNS also affect cellular bioenergetics through the modulation of ATP synthase catalysis reveal a dual function of these modifications in the cells. Here, we describe mitochondria as a major site of production and as a target of ROS/RNS and discuss the pathophysiological conditions in which oxidative and nitrosative modifications modulate the catalytic and pore-forming activities of ATP synthase.
Collapse
|
292
|
Aroso M, Ferreira R, Freitas A, Vitorino R, Gomez-Lazaro M. New insights on the mitochondrial proteome plasticity in Parkinson's disease. Proteomics Clin Appl 2016; 10:416-29. [PMID: 26749507 DOI: 10.1002/prca.201500092] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 12/09/2015] [Accepted: 01/04/2016] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative diseases whose relentless progression results in severe disability. Although PD aetiology is unknown, growing evidences point to the mitochondrial involvement in the pathobiology of this disorder. So, it seems imperative to understand the means by which the molecular pathways harboured in this organelle are regulated. With the advances in MS-based proteomics, there is a substantial expectation in the increased knowledge of mitochondrial protein dynamics. Still, few studies have been performed on mitochondrial protein profiling in the context of PD. In order to integrate data from these studies, network analyses were performed taking into consideration variables such as model of PD, cell line, or tissue origin. Overall, data retrieved from these analyses highlighted the modulation of the biological processes related with "generation of energy," "cellular metabolism," and "mitochondrial transport" in PD. However, it was noted that the impact of sample type and/or PD model on the biological processes was modulated by the disease. Moreover, technical considerations related to protein characterization using gel-based or gel-free MS approaches should be considered in data comparison among different studies. Data from the present review will help to envisage future studies targeting these mechanisms.
Collapse
Affiliation(s)
- Miguel Aroso
- Department of Medical Sciences, iBiMED, University of Aveiro, Aveiro, Portugal
| | - Rita Ferreira
- Department of Chemistry, QOPNA, Mass Spectrometry Center, University of Aveiro, Aveiro, Portugal
| | - Ana Freitas
- Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, University of Porto, Porto, Portugal.,Faculty of Medicine, University of Porto, Porto, Portugal
| | - Rui Vitorino
- Department of Medical Sciences, iBiMED, University of Aveiro, Aveiro, Portugal.,Department of Chemistry, QOPNA, Mass Spectrometry Center, University of Aveiro, Aveiro, Portugal.,Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Maria Gomez-Lazaro
- Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, University of Porto, Porto, Portugal
| |
Collapse
|
293
|
Choi I, Woo JH, Jou I, Joe EH. PINK1 Deficiency Decreases Expression Levels of mir-326, mir-330, and mir-3099 during Brain Development and Neural Stem Cell Differentiation. Exp Neurobiol 2016; 25:14-23. [PMID: 26924929 PMCID: PMC4766110 DOI: 10.5607/en.2016.25.1.14] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 01/27/2016] [Accepted: 01/27/2016] [Indexed: 12/17/2022] Open
Abstract
PTEN-induced putative kinase 1 (PINK1) is a Parkinson's disease (PD) gene. We examined miRNAs regulated by PINK1 during brain development and neural stem cell (NSC) differentiation, and found that lvels of miRNAs related to tumors and inflammation were different between 1-day-old-wild type (WT) and PINK1-knockout (KO) mouse brains. Notably, levels of miR-326, miR-330 and miR-3099, which are related to astroglioma, increased during brain development and NSC differentiation, and were significantly reduced in the absence of PINK1. Interestingly, in the presence of ciliary neurotrophic factor (CNTF), which pushes differentiation of NSCs into astrocytes, miR-326, miR-330, and miR-3099 levels in KO NSCs were also lower than those in WT NSCs. Furthermore, mimics of all three miRNAs increased expression of the astrocytic marker glial fibrillary acidic protein (GFAP) during differentiation of KO NSCs, but inhibitors of these miRNAs decreased GFAP expression in WT NSCs. Moreover, these miRNAs increased the translational efficacy of GFAP through the 3'-UTR of GFAP mRNA. Taken together, these results suggest that PINK1 deficiency reduce expression levels of miR-326, miR-330 and miR-3099, which may regulate GFAP expression during NSC differentiation and brain development.
Collapse
Affiliation(s)
- Insup Choi
- Neuroscience Graduate Program Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, Korea.; Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon 16499, Korea
| | - Joo Hong Woo
- Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon 16499, Korea
| | - Ilo Jou
- Neuroscience Graduate Program Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, Korea.; Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon 16499, Korea.; Department of Pharmacology, Ajou University School of Medicine, Suwon 16499, Korea
| | - Eun-Hye Joe
- Neuroscience Graduate Program Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, Korea.; Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon 16499, Korea.; Department of Pharmacology, Ajou University School of Medicine, Suwon 16499, Korea.; Department of Brain Science, Ajou University School of Medicine, Suwon 16499, Korea.; Brain Disease Research Center, Ajou University School of Medicine, Suwon 16499, Korea
| |
Collapse
|
294
|
Saric A, Andreau K, Armand AS, Møller IM, Petit PX. Barth Syndrome: From Mitochondrial Dysfunctions Associated with Aberrant Production of Reactive Oxygen Species to Pluripotent Stem Cell Studies. Front Genet 2016; 6:359. [PMID: 26834781 PMCID: PMC4719219 DOI: 10.3389/fgene.2015.00359] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 12/15/2015] [Indexed: 12/22/2022] Open
Abstract
Mutations in the gene encoding the enzyme tafazzin, TAZ, cause Barth syndrome (BTHS). Individuals with this X-linked multisystem disorder present cardiomyopathy (CM) (often dilated), skeletal muscle weakness, neutropenia, growth retardation, and 3-methylglutaconic aciduria. Biopsies of the heart, liver and skeletal muscle of patients have revealed mitochondrial malformations and dysfunctions. It is the purpose of this review to summarize recent results of studies on various animal or cell models of Barth syndrome, which have characterized biochemically the strong cellular defects associated with TAZ mutations. Tafazzin is a mitochondrial phospholipidlysophospholipid transacylase that shuttles acyl groups between phospholipids and regulates the remodeling of cardiolipin (CL), a unique inner mitochondrial membrane phospholipid dimer consisting of two phosphatidyl residues linked by a glycerol bridge. After their biosynthesis, the acyl chains of CLs may be modified in remodeling processes involving up to three different enzymes. Their characteristic acyl chain composition depends on the function of tafazzin, although the enzyme itself surprisingly lacks acyl specificity. CLs are crucial for correct mitochondrial structure and function. In addition to their function in the basic mitochondrial function of ATP production, CLs play essential roles in cardiac function, apoptosis, autophagy, cell cycle regulation and Fe-S cluster biosynthesis. Recent developments in tafazzin research have provided strong insights into the link between mitochondrial dysfunction and the production of reactive oxygen species (ROS). An important tool has been the generation of BTHS-specific induced pluripotent stem cells (iPSCs) from BTHS patients. In a complementary approach, disease-specific mutations have been introduced into wild-type iPSC lines enabling direct comparison with isogenic controls. iPSC-derived cardiomyocytes were then characterized using biochemical and classical bioenergetic approaches. The cells are tested in a "heart-on-chip" assay to model the pathophysiology in vitro, to characterize the underlying mechanism of BTHS deriving from TAZ mutations, mitochondrial deficiencies and ROS production and leading to tissue defects, and to evaluate potential therapies with the use of mitochondrially targeted antioxidants.
Collapse
Affiliation(s)
- Ana Saric
- INSERM U 1124 "Toxicologie, Pharmacologie et Signalisation Cellulaire" and "FR 3567" CNRS Chimie, Toxicologie, Signalisation Cellulaire et Cibles Thérapeutiques, Université Paris Descartes - Centre Universitaire des Saints-PèresParis, France; Division of Molecular Medicine, Ruđer Bošković InstituteZagreb, Croatia
| | - Karine Andreau
- INSERM U 1124 "Toxicologie, Pharmacologie et Signalisation Cellulaire" and "FR 3567" CNRS Chimie, Toxicologie, Signalisation Cellulaire et Cibles Thérapeutiques, Université Paris Descartes - Centre Universitaire des Saints-Pères Paris, France
| | - Anne-Sophie Armand
- INSERM U 1124 "Toxicologie, Pharmacologie et Signalisation Cellulaire" and "FR 3567" CNRS Chimie, Toxicologie, Signalisation Cellulaire et Cibles Thérapeutiques, Université Paris Descartes - Centre Universitaire des Saints-Pères Paris, France
| | - Ian M Møller
- Department of Molecular Biology and Genetics, Aarhus University Slagelse, Denmark
| | - Patrice X Petit
- INSERM U 1124 "Toxicologie, Pharmacologie et Signalisation Cellulaire" and "FR 3567" CNRS Chimie, Toxicologie, Signalisation Cellulaire et Cibles Thérapeutiques, Université Paris Descartes - Centre Universitaire des Saints-Pères Paris, France
| |
Collapse
|
295
|
Choi I, Choi DJ, Yang H, Woo JH, Chang MY, Kim JY, Sun W, Park SM, Jou I, Lee SH, Lee SH, Joe EH. PINK1 expression increases during brain development and stem cell differentiation, and affects the development of GFAP-positive astrocytes. Mol Brain 2016; 9:5. [PMID: 26746235 PMCID: PMC4706723 DOI: 10.1186/s13041-016-0186-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 01/04/2016] [Indexed: 01/19/2023] Open
Abstract
Background Mutation of PTEN-induced putative kinase 1 (PINK1) causes autosomal recessive early-onset Parkinson’s disease (PD). Despite of its ubiquitous expression in brain, its roles in non-neuronal cells such as neural stem cells (NSCs) and astrocytes were poorly unknown. Results We show that PINK1 expression increases from embryonic day 12 to postnatal day 1 in mice, which represents the main period of brain development. PINK1 expression also increases during neural stem cell (NSC) differentiation. Interestingly, expression of GFAP (a marker of astrocytes) was lower in PINK1 knockout (KO) mouse brain lysates compared to wild-type (WT) lysates at postnatal days 1-8, whereas there was little difference in the expression of markers for other brain cell types (e.g., neurons and oligodendrocytes). Further experiments showed that PINK1-KO NSCs were defective in their differentiation to astrocytes, producing fewer GFAP-positive cells compared to WT NSCs. However, the KO and WT NSCs did not differ in their self-renewal capabilities or ability to differentiate to neurons and oligodendrocytes. Interestingly, during differentiation of KO NSCs there were no defects in mitochondrial function, and there were not changes in signaling molecules such as SMAD1/5/8, STAT3, and HES1 involved in differentiation of NSCs into astrocytes. In brain sections, GFAP-positive astrocytes were more sparsely distributed in the corpus callosum and substantia nigra of KO animals compared with WT. Conclusion Our study suggests that PINK1 deficiency causes defects in GFAP-positive astrogliogenesis during brain development and NSC differentiation, which may be a factor to increase risk for PD. Electronic supplementary material The online version of this article (doi:10.1186/s13041-016-0186-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Insup Choi
- Neuroscience Graduate Program Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea. .,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Korea.
| | - Dong-Joo Choi
- Neuroscience Graduate Program Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea.
| | - Haijie Yang
- Department of Pharmacology, Ajou University School of Medicine san-5, Woncheon-dong, Youngtong-gu, Suwon, Kyunggi-do, 442-721, Korea.
| | - Joo Hong Woo
- Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Korea.
| | - Mi-Yoon Chang
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Korea.
| | - Joo Yeon Kim
- Department of Anatomy and Division of Brain Korea 21 Plus Biomedical Science, Korea University College of Medicine, Seoul, 136-705, Korea.
| | - Woong Sun
- Department of Anatomy and Division of Brain Korea 21 Plus Biomedical Science, Korea University College of Medicine, Seoul, 136-705, Korea.
| | - Sang-Myun Park
- Neuroscience Graduate Program Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea. .,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Korea. .,Department of Pharmacology, Ajou University School of Medicine san-5, Woncheon-dong, Youngtong-gu, Suwon, Kyunggi-do, 442-721, Korea.
| | - Ilo Jou
- Neuroscience Graduate Program Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea. .,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Korea. .,Department of Pharmacology, Ajou University School of Medicine san-5, Woncheon-dong, Youngtong-gu, Suwon, Kyunggi-do, 442-721, Korea.
| | - Sang-Hun Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Korea.
| | - Sang Hoon Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Korea.
| | - Eun-Hye Joe
- Neuroscience Graduate Program Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea. .,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Korea. .,Department of Pharmacology, Ajou University School of Medicine san-5, Woncheon-dong, Youngtong-gu, Suwon, Kyunggi-do, 442-721, Korea. .,Department of Brain Science, Ajou University School of Medicine, Suwon, Korea. .,Brain Disease Research Center, Ajou University School of Medicine, Suwon, Korea.
| |
Collapse
|
296
|
Choi J, Polcher A, Joas A. Systematic literature review on Parkinson's disease and Childhood Leukaemia and mode of actions for pesticides. ACTA ACUST UNITED AC 2016. [DOI: 10.2903/sp.efsa.2016.en-955] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
297
|
de Oliveira MR. Phloretin-induced cytoprotective effects on mammalian cells: A mechanistic view and future directions. Biofactors 2016; 42:13-40. [PMID: 26826024 DOI: 10.1002/biof.1256] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 11/19/2015] [Indexed: 11/11/2022]
Abstract
Phloretin (C15 H14 O5 ), a dihydrochalcone flavonoid, is mainly found in fruit, leaves, and roots of apple tree. Phloretin exerts antioxidant, anti-inflammatory, and anti-tumor activities in mammalian cells through mechanisms that have been partially elucidated throughout the years. Phloretin bioavailability is well known in humans, but still remains to be better studied in experimental animals, such as mouse and rat. The focus of the present review is to gather information regarding the mechanisms involved in the phloretin-elicited effects in different in vitro and in vivo experimental models. Several manuscripts were analyzed and data raised by authors were described and discussed here in a mechanistic manner. Comparisons between the effects elicited by phloretin and phloridzin were made whenever possible, as well as with other polyphenols, clarifying questions about the use of phloretin as a potential therapeutic agent. Toxicological aspects associated to phloretin exposure were also discussed here. Furthermore, a special section containing future directions was created as a suggestive guide towards the elucidation of phloretin-related actions in mammalian cells and tissues.
Collapse
Affiliation(s)
- Marcos Roberto de Oliveira
- Department of Chemistry/ICET, Postgraduate Program in Chemistry (PPGQ), Federal University of Mato Grosso (UFMT), CEP, Cuiaba, MT, Brazil
| |
Collapse
|
298
|
Santo-Domingo J, Wiederkehr A, De Marchi U. Modulation of the matrix redox signaling by mitochondrial Ca 2+. World J Biol Chem 2015; 6:310-323. [PMID: 26629314 PMCID: PMC4657127 DOI: 10.4331/wjbc.v6.i4.310] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 09/04/2015] [Accepted: 10/13/2015] [Indexed: 02/05/2023] Open
Abstract
Mitochondria sense, shape and integrate signals, and thus function as central players in cellular signal transduction. Ca2+ waves and redox reactions are two such intracellular signals modulated by mitochondria. Mitochondrial Ca2+ transport is of utmost physio-pathological relevance with a strong impact on metabolism and cell fate. Despite its importance, the molecular nature of the proteins involved in mitochondrial Ca2+ transport has been revealed only recently. Mitochondrial Ca2+ promotes energy metabolism through the activation of matrix dehydrogenases and down-stream stimulation of the respiratory chain. These changes also alter the mitochondrial NAD(P)H/NAD(P)+ ratio, but at the same time will increase reactive oxygen species (ROS) production. Reducing equivalents and ROS are having opposite effects on the mitochondrial redox state, which are hard to dissect. With the recent development of genetically encoded mitochondrial-targeted redox-sensitive sensors, real-time monitoring of matrix thiol redox dynamics has become possible. The discoveries of the molecular nature of mitochondrial transporters of Ca2+ combined with the utilization of the novel redox sensors is shedding light on the complex relation between mitochondrial Ca2+ and redox signals and their impact on cell function. In this review, we describe mitochondrial Ca2+ handling, focusing on a number of newly identified proteins involved in mitochondrial Ca2+ uptake and release. We further discuss our recent findings, revealing how mitochondrial Ca2+ influences the matrix redox state. As a result, mitochondrial Ca2+ is able to modulate the many mitochondrial redox-regulated processes linked to normal physiology and disease.
Collapse
|
299
|
Von Stockum S, Nardin A, Schrepfer E, Ziviani E. Mitochondrial dynamics and mitophagy in Parkinson's disease: A fly point of view. Neurobiol Dis 2015; 90:58-67. [PMID: 26550693 DOI: 10.1016/j.nbd.2015.11.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 11/02/2015] [Accepted: 11/05/2015] [Indexed: 12/27/2022] Open
Abstract
Mitochondria are double membrane-bounded organelles residing in the cytoplasm of almost all eukaryotic cells, which convert energy from the disposal of organic substrates into an electrochemical gradient that is in turn converted into ATP. However, the ion gradient that is generated through the oxidation of nutrients, may lead to the production of reactive oxygen species (ROS), which can generate free radicals, damaging cells and contributing to disease. Originally described as static structures, to date they are considered extremely plastic and dynamic organelles. In this respect, mitochondrial dynamics is crucial to prevent potential damage that is generated by ROS. For instance, mitochondria elongate to dilute oxidized proteins into the mitochondrial network, and they fragment to allow selective elimination of dysfunctional mitochondria via mitophagy. Accordingly, mitochondrial dynamics perturbation may compromise the selective elimination of damaged proteins and dysfunctional organelles and lead to the development of different diseases including neurodegenerative diseases. In recent years the fruit fly Drosophila melanogaster has proved to be a valuable model system to evaluate the consequences of mitochondria quality control dysfunction in vivo, particularly with respect to PINK1/Parkin dependent dysregulation of mitophagy in the onset of Parkinson's Disease (PD). The current challenge is to be able to use fly based genetic strategies to gain further insights into molecular mechanisms underlying disease in order to develop new therapeutic strategies. This article is part of a Special Issue entitled: Role of mitochondria in physiological and pathophysiological functions in the central nervous system.
Collapse
Affiliation(s)
| | - Alice Nardin
- Department of Biochemistry, University of Padova, via Ugo Bassi 56, Padova, Italy
| | - Emilie Schrepfer
- Department of Biochemistry, University of Padova, via Ugo Bassi 56, Padova, Italy; VIMM, Venetian Institute of Molecular Medicine, Via Giuseppe Orus 2, Padova, Italy
| | - Elena Ziviani
- Fondazione Ospedale San Camillo, IRCCS, Lido di Venezia, Venezia, Italy; Department of Biochemistry, University of Padova, via Ugo Bassi 56, Padova, Italy.
| |
Collapse
|
300
|
Bernardi P, Rasola A, Forte M, Lippe G. The Mitochondrial Permeability Transition Pore: Channel Formation by F-ATP Synthase, Integration in Signal Transduction, and Role in Pathophysiology. Physiol Rev 2015; 95:1111-55. [PMID: 26269524 DOI: 10.1152/physrev.00001.2015] [Citation(s) in RCA: 439] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The mitochondrial permeability transition (PT) is a permeability increase of the inner mitochondrial membrane mediated by a channel, the permeability transition pore (PTP). After a brief historical introduction, we cover the key regulatory features of the PTP and provide a critical assessment of putative protein components that have been tested by genetic analysis. The discovery that under conditions of oxidative stress the F-ATP synthases of mammals, yeast, and Drosophila can be turned into Ca(2+)-dependent channels, whose electrophysiological properties match those of the corresponding PTPs, opens new perspectives to the field. We discuss structural and functional features of F-ATP synthases that may provide clues to its transition from an energy-conserving into an energy-dissipating device as well as recent advances on signal transduction to the PTP and on its role in cellular pathophysiology.
Collapse
Affiliation(s)
- Paolo Bernardi
- Department of Biomedical Sciences and Consiglio Nazionale delle Ricerche Neuroscience Institute, University of Padova, Padova, Italy; Vollum Institute, Oregon Health and Sciences University, Portland, Oregon; and Department of Food Science, University of Udine, Udine, Italy
| | - Andrea Rasola
- Department of Biomedical Sciences and Consiglio Nazionale delle Ricerche Neuroscience Institute, University of Padova, Padova, Italy; Vollum Institute, Oregon Health and Sciences University, Portland, Oregon; and Department of Food Science, University of Udine, Udine, Italy
| | - Michael Forte
- Department of Biomedical Sciences and Consiglio Nazionale delle Ricerche Neuroscience Institute, University of Padova, Padova, Italy; Vollum Institute, Oregon Health and Sciences University, Portland, Oregon; and Department of Food Science, University of Udine, Udine, Italy
| | - Giovanna Lippe
- Department of Biomedical Sciences and Consiglio Nazionale delle Ricerche Neuroscience Institute, University of Padova, Padova, Italy; Vollum Institute, Oregon Health and Sciences University, Portland, Oregon; and Department of Food Science, University of Udine, Udine, Italy
| |
Collapse
|