101
|
Mandal A, Pinter K, Drerup CM. Analyzing Neuronal Mitochondria in vivo Using Fluorescent Reporters in Zebrafish. Front Cell Dev Biol 2018; 6:144. [PMID: 30410881 PMCID: PMC6209631 DOI: 10.3389/fcell.2018.00144] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 10/08/2018] [Indexed: 01/08/2023] Open
Abstract
Despite their importance for cellular viability, the actual life history and properties of mitochondria in neurons are still unclear. These organelles are distributed throughout the entirety of the neuron and serve many functions, including: energy production (ATP), iron homeostasis and processing, calcium buffering, and metabolite production, as well as many other lesser known activities. Given their importance, understanding how these organelles are positioned and how their health and function is maintained is critical for many aspects of cell biology. This is best illustrated by the diverse disease literature which demonstrates that abnormal mitochondrial movement, localization, size, or function often correlates with neural pathology. In the following methods article, we will describe the techniques and tools we have optimized to directly visualize mitochondria and analyze mitochondrial lifetime, health, and function in neurons in vivo using fluorescent reporters in the zebrafish. The zebrafish system is ideal for in vivo studies of mitochondrial biology as: (1) neuronal circuits develop rapidly, within days; (2) it is genetically accessible; and (3) embryos and larvae are translucent allowing imaging in a completely intact vertebrate nervous system. Using these tools and techniques, the field is poised to answer questions of mitochondrial biology in the context of neuronal health and function in normal and disease states.
Collapse
Affiliation(s)
- Amrita Mandal
- Unit on Neuronal Cell Biology, NICHD, National Institutes of Health, Bethesda, MD, United States
| | - Katherine Pinter
- Unit on Neuronal Cell Biology, NICHD, National Institutes of Health, Bethesda, MD, United States
| | - Catherine M Drerup
- Unit on Neuronal Cell Biology, NICHD, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
102
|
Metabolic regulation and glucose sensitivity of cortical radial glial cells. Proc Natl Acad Sci U S A 2018; 115:10142-10147. [PMID: 30224493 DOI: 10.1073/pnas.1808066115] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The primary stem cells of the cerebral cortex are the radial glial cells (RGCs), and disturbances in their operation lead to myriad brain disorders in all mammals from mice to humans. Here, we found in mice that maternal gestational obesity and hyperglycemia can impair the maturation of RGC fibers and delay cortical neurogenesis. To investigate potential mechanisms, we used optogenetic live-imaging approaches in embryonic cortical slices. We found that Ca2+ signaling regulates mitochondrial transport and is crucial for metabolic support in RGC fibers. Cyclic intracellular Ca2+ discharge from localized RGC fiber segments detains passing mitochondria and ensures their proper distribution and enrichment at specific sites such as endfeet. Impairment of mitochondrial function caused an acute loss of Ca2+ signaling, while hyperglycemia decreased Ca2+ activity and impaired mitochondrial transport, leading to degradation of the RGC scaffold. Our findings uncover a physiological mechanism indicating pathways by which gestational metabolic disturbances can interfere with brain development.
Collapse
|
103
|
Toglia P, Demuro A, Mak DOD, Ullah G. Data-driven modeling of mitochondrial dysfunction in Alzheimer's disease. Cell Calcium 2018; 76:23-35. [PMID: 30248575 DOI: 10.1016/j.ceca.2018.09.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 09/02/2018] [Accepted: 09/03/2018] [Indexed: 02/06/2023]
Abstract
Intracellular accumulation of oligomeric forms of β amyloid (Aβ) are now believed to play a key role in the earliest phase of Alzheimer's disease (AD) as their rise correlates well with the early symptoms of the disease. Extensive evidence points to impaired neuronal Ca2+ homeostasis as a direct consequence of the intracellular Aβ oligomers. However, little is known about the downstream effects of the resulting Ca2+ rise on the many intracellular Ca2+-dependent pathways. Here we use multiscale modeling in conjunction with patch-clamp electrophysiology of single inositol 1,4,5-trisphosphate (IP3) receptor (IP3R) and fluorescence imaging of whole-cell Ca2+ response, induced by exogenously applied intracellular Aβ42 oligomers to show that Aβ42 inflicts cytotoxicity by impairing mitochondrial function. Driven by patch-clamp experiments, we first model the kinetics of IP3R, which is then extended to build a model for the whole-cell Ca2+ signals. The whole-cell model is then fitted to fluorescence signals to quantify the overall Ca2+ release from the endoplasmic reticulum by intracellular Aβ42 oligomers through G-protein-mediated stimulation of IP3 production. The estimated IP3 concentration as a function of intracellular Aβ42 content together with the whole-cell model allows us to show that Aβ42 oligomers impair mitochondrial function through pathological Ca2+ uptake and the resulting reduced mitochondrial inner membrane potential, leading to an overall lower ATP and increased production of reactive oxygen species and H2O2. We further show that mitochondrial function can be restored by the addition of Ca2+ buffer EGTA, in accordance with the observed abrogation of Aβ42 cytotoxicity by EGTA in our live cells experiments.
Collapse
Affiliation(s)
- Patrick Toglia
- Department of Physics, University of South Florida, Tampa, FL 33620, USA
| | - Angelo Demuro
- Department of Neurobiology and Behavior and Physiology and Biophysics, University of California, Irvine, CA 92697, USA
| | - Don-On Daniel Mak
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ghanim Ullah
- Department of Physics, University of South Florida, Tampa, FL 33620, USA.
| |
Collapse
|
104
|
Parrado-Fernández C, Schneider B, Ankarcrona M, Conti MM, Cookson MR, Kivipelto M, Cedazo-Mínguez Á, Sandebring-Matton A. Reduction of PINK1 or DJ-1 impair mitochondrial motility in neurites and alter ER-mitochondria contacts. J Cell Mol Med 2018; 22:5439-5449. [PMID: 30133157 PMCID: PMC6201361 DOI: 10.1111/jcmm.13815] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 07/02/2018] [Indexed: 12/31/2022] Open
Abstract
Subcellular distribution of mitochondria in neurons is crucial for meeting the energetic demands, as well as the necessity to buffer Ca2+ within the axon, dendrites and synapses. Mitochondrial impairment is an important feature of Parkinson disease (PD), in which both familial parkinsonism genes DJ-1 and PINK1 have a great impact on mitochondrial function. We used differentiated human dopaminergic neuroblastoma cell lines with stable PINK1 or DJ-1 knockdown to study live motility of mitochondria in neurites. The frequency of anterograde and retrograde mitochondrial motility was decreased in PINK1 knockdown cells and the frequency of total mitochondrial motility events was reduced in both cell lines. However, neither the distribution nor the size of mitochondria in the neurites differed from the control cells even after downregulation of the mitochondrial fission protein, Drp1. Furthermore, mitochondria from PINK1 knockdown cells, in which motility was most impaired, had increased levels of GSK3βSer9 and higher release of mitochondrial Ca2+ when exposed to CCCP-induced mitochondrial uncoupling. Further analysis of the ER-mitochondria contacts involved in Ca2+ shuttling showed that PINK1 knockdown cells had reduced contacts between the two organelles. Our results give new insight on how PINK1 and DJ-1 influence mitochondria, thus providing clues to novel PD therapies.
Collapse
Affiliation(s)
- Cristina Parrado-Fernández
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Bernadette Schneider
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Maria Ankarcrona
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Melissa M Conti
- Laboratory of Neurogenetics, National Institute on Aging/NIH, Bethesda, Maryland
| | - Mark R Cookson
- Laboratory of Neurogenetics, National Institute on Aging/NIH, Bethesda, Maryland
| | - Miia Kivipelto
- Division of Clinical Geriatrics, Center for Alzheimer Research, NVS, Karolinska Institutet, Stockholm, Sweden.,Aging Research Center, Karolinska Institutet-Stockholm University, Stockholm, Sweden.,Research & Development Unit, Stockholms Sjukhem, Stockholm, Sweden
| | - Ángel Cedazo-Mínguez
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Anna Sandebring-Matton
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
105
|
Abstract
This review systematically examines the evidence for shifts in flux through energy generating biochemical pathways in Huntington’s disease (HD) brains from humans and model systems. Compromise of the electron transport chain (ETC) appears not to be the primary or earliest metabolic change in HD pathogenesis. Rather, compromise of glucose uptake facilitates glucose flux through glycolysis and may possibly decrease flux through the pentose phosphate pathway (PPP), limiting subsequent NADPH and GSH production needed for antioxidant protection. As a result, oxidative damage to key glycolytic and tricarboxylic acid (TCA) cycle enzymes further restricts energy production so that while basal needs may be met through oxidative phosphorylation, those of excessive stimulation cannot. Energy production may also be compromised by deficits in mitochondrial biogenesis, dynamics or trafficking. Restrictions on energy production may be compensated for by glutamate oxidation and/or stimulation of fatty acid oxidation. Transcriptional dysregulation generated by mutant huntingtin also contributes to energetic disruption at specific enzymatic steps. Many of the alterations in metabolic substrates and enzymes may derive from normal regulatory feedback mechanisms and appear oscillatory. Fine temporal sequencing of the shifts in metabolic flux and transcriptional and expression changes associated with mutant huntingtin expression remain largely unexplored and may be model dependent. Differences in disease progression among HD model systems at the time of experimentation and their varying states of metabolic compensation may explain conflicting reports in the literature. Progressive shifts in metabolic flux represent homeostatic compensatory mechanisms that maintain the model organism through presymptomatic and symptomatic stages.
Collapse
Affiliation(s)
- Janet M Dubinsky
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
106
|
Hill RL, Kulbe JR, Singh IN, Wang JA, Hall ED. Synaptic Mitochondria are More Susceptible to Traumatic Brain Injury-induced Oxidative Damage and Respiratory Dysfunction than Non-synaptic Mitochondria. Neuroscience 2018; 386:265-283. [PMID: 29960045 DOI: 10.1016/j.neuroscience.2018.06.028] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 06/12/2018] [Accepted: 06/18/2018] [Indexed: 12/17/2022]
Abstract
Traumatic brain injury (TBI) results in mitochondrial dysfunction and induction of lipid peroxidation (LP). Lipid peroxidation-derived neurotoxic aldehydes such as 4-HNE and acrolein bind to mitochondrial proteins, inducing additional oxidative damage and further exacerbating mitochondrial dysfunction and LP. Mitochondria are heterogeneous, consisting of both synaptic and non-synaptic populations. Synaptic mitochondria are reported to be more vulnerable to injury; however, this is the first study to characterize the temporal profile of synaptic and non-synaptic mitochondria following TBI, including investigation of respiratory dysfunction and oxidative damage to mitochondrial proteins between 3 and 120 h following injury. These results indicate that synaptic mitochondria are indeed the more vulnerable population, showing both more rapid and severe impairments than non-synaptic mitochondria. By 24 h, synaptic respiration is significantly impaired compared to synaptic sham, whereas non-synaptic respiration does not decline significantly until 48 h. Decreases in respiration are associated with increases in oxidative damage to synaptic and non-synaptic mitochondrial proteins at 48 h and 72 h, respectively. These results indicate that the therapeutic window for mitochondria-targeted pharmacological neuroprotectants to prevent respiratory dysfunction is shorter for the more vulnerable synaptic mitochondria than for the non-synaptic population.
Collapse
Affiliation(s)
- Rachel L Hill
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States
| | - Jacqueline R Kulbe
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States; Department of Neuroscience, University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States
| | - Indrapal N Singh
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States; Department of Neuroscience, University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States
| | - Juan A Wang
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States
| | - Edward D Hall
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States; Department of Neuroscience, University of Kentucky College of Medicine, 741 S. Limestone St, Lexington, KY 40536-0509, United States.
| |
Collapse
|
107
|
ROS Control Mitochondrial Motility through p38 and the Motor Adaptor Miro/Trak. Cell Rep 2018; 21:1667-1680. [PMID: 29117569 DOI: 10.1016/j.celrep.2017.10.060] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 09/18/2017] [Accepted: 10/15/2017] [Indexed: 01/20/2023] Open
Abstract
Mitochondrial distribution and motility are recognized as central to many cellular functions, but their regulation by signaling mechanisms remains to be elucidated. Here, we report that reactive oxygen species (ROS), either derived from an extracellular source or intracellularly generated, control mitochondrial distribution and function by dose-dependently, specifically, and reversibly decreasing mitochondrial motility in both rat hippocampal primary cultured neurons and cell lines. ROS decrease motility independently of cytoplasmic [Ca2+], mitochondrial membrane potential, or permeability transition pore opening, known effectors of oxidative stress. However, multiple lines of genetic and pharmacological evidence support that a ROS-activated mitogen-activated protein kinase (MAPK), p38α, is required for the motility inhibition. Furthermore, anchoring mitochondria directly to kinesins without involvement of the physiological adaptors between the organelles and the motor protein prevents the H2O2-induced decrease in mitochondrial motility. Thus, ROS engage p38α and the motor adaptor complex to exert changes in mitochondrial motility, which likely has both physiological and pathophysiological relevance.
Collapse
|
108
|
Mitochondrial therapy promotes regeneration of injured hippocampal neurons. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3001-3012. [PMID: 29913215 DOI: 10.1016/j.bbadis.2018.06.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 05/12/2018] [Accepted: 06/13/2018] [Indexed: 11/20/2022]
Abstract
Due to the inhibitory microenvironment and reduced intrinsic growth capacity of neurons, neuronal regeneration of central nervous system remains challenging. Neurons are highly energy demanding and require sufficient mitochondria to support cellular activities. In response to stimuli, mitochondria undergo fusion/fission cycles to adapt to environment. It is thus logical to hypothesize that the plasticity of mitochondrial dynamics is required for neuronal regeneration. In this study, we examined the role of mitochondrial dynamics during regeneration of rat hippocampal neurons. Quantitative analysis showed that injury induced mitochondrial fission. As mitochondrial dysfunction has been implicated in neurodegenerative diseases, we tested the possibility that the mitochondrial therapy may promote neuronal regeneration. Supplying freshly isolated mitochondria to the injured hippocampal neurons not only significantly increased neurite re-growth but also restored membrane potential of injured hippocampal neurons. Together, our findings support the importance of mitochondrial dynamics during regeneration of injured hippocampal neurons and highlight the therapeutic prospect of mitochondria to the injured central nervous system.
Collapse
|
109
|
Brain Mitochondria, Aging, and Parkinson's Disease. Genes (Basel) 2018; 9:genes9050250. [PMID: 29751692 PMCID: PMC5977190 DOI: 10.3390/genes9050250] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 04/27/2018] [Accepted: 05/07/2018] [Indexed: 02/08/2023] Open
Abstract
This paper reconsiders the role of mitochondria in aging and in Parkinson’s Disease (PD). The most important risk factor for PD is aging. Alterations in mitochondrial activity are typical of aging. Mitochondrial aging is characterized by decreased oxidative phosphorylation, proteasome activity decrease, altered autophagy, and mitochondrial dysfunction. Beyond declined oxidative phosphorylation, mitochondrial dysfunction consists of a decline of beta-oxidation as well as of the Krebs cycle. Not inherited mitochondrial DNA (mtDNA) mutations are acquired over time and parallel the decrease in oxidative phosphorylation. Many of these mitochondrial alterations are also found in the PD brain specifically in the substantia nigra (SN). mtDNA deletions and development of respiratory chain deficiency in SN neurons of aged individuals as well as of individuals with PD converge towards a shared pathway, which leads to neuronal dysfunction and death. Finally, several nuclear genes that are mutated in hereditary PD are usually implicated in mitochondrial functioning to a various extent and their mutation may cause mitochondrial impairment. In conclusion, a tight link exists between mitochondria, aging, and PD.
Collapse
|
110
|
Kraus K, Kleene R, Braren I, Loers G, Lutz D, Schachner M. A fragment of adhesion molecule L1 is imported into mitochondria, and regulates mitochondrial metabolism and trafficking. J Cell Sci 2018; 131:jcs.210500. [PMID: 29632241 DOI: 10.1242/jcs.210500] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 03/13/2018] [Indexed: 02/05/2023] Open
Abstract
The cell adhesion molecule L1 (also known as L1CAM) plays important roles in the mammalian nervous system under physiological and pathological conditions. We have previously reported that proteolytic cleavage of L1 by myelin basic protein leads to the generation of a 70 kDa transmembrane L1 fragment (L1-70) that promotes neuronal migration and neuritogenesis. Here, we provide evidence that L1-70 is imported from the cytoplasm into mitochondria. Genetic ablation of L1, inhibition of mitochondrial import of L1-70 or prevention of myelin basic protein-mediated generation of L1-70 all lead to reduced mitochondrial complex I activity, and impaired mitochondrial membrane potential, fusion, fission and motility, as well as increased retrograde transport. We identified NADH dehydrogenase ubiquinone flavoprotein 2 as a binding partner for L1, suggesting that L1-70 interacts with this complex I subunit to regulate complex I activity. The results of our study provide insights into novel functions of L1 in mitochondrial metabolism and cellular dynamics. These functions are likely to ameliorate the consequences of acute nervous system injuries and chronic neurodegenerative diseases.
Collapse
Affiliation(s)
- Kristina Kraus
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Ralf Kleene
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Ingke Braren
- Vector Core Unit, Institut für Experimentelle Pharmakologie und Toxikologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Gabriele Loers
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - David Lutz
- Institut für Strukturelle Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong 515041, China
| |
Collapse
|
111
|
Mitochondrial cAMP-PKA signaling: What do we really know? BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2018; 1859:868-877. [PMID: 29694829 DOI: 10.1016/j.bbabio.2018.04.005] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/06/2018] [Accepted: 04/18/2018] [Indexed: 12/22/2022]
Abstract
Mitochondria are key organelles for cellular homeostasis. They generate the most part of ATP that is used by cells through oxidative phosphorylation. They also produce reactive oxygen species, neurotransmitters and other signaling molecules. They are important for calcium homeostasis and apoptosis. Considering the role of this organelle, it is not surprising that most mitochondrial dysfunctions are linked to the development of pathologies. Various mechanisms adjust mitochondrial activity according to physiological needs. The cAMP-PKA signaling emerged in recent years as a direct and powerful mean to regulate mitochondrial functions. Multiple evidence demonstrates that such pathway can be triggered from cytosol or directly within mitochondria. Notably, specific anchor proteins target PKA to mitochondria whereas enzymes necessary for generation and degradation of cAMP are found directly in these organelles. Mitochondrial PKA targets proteins localized in different compartments of mitochondria, and related to various functions. Alterations of mitochondrial cAMP-PKA signaling affect the development of several physiopathological conditions, including neurodegenerative diseases. It is however difficult to discriminate between the effects of cAMP-PKA signaling triggered from cytosol or directly in mitochondria. The specific roles of PKA localized in different mitochondrial compartments are also not completely understood. The aim of this work is to review the role of cAMP-PKA signaling in mitochondrial (patho)physiology.
Collapse
|
112
|
Kimball EC, Jefferys JL, Pease ME, Oglesby EN, Nguyen C, Schaub J, Pitha I, Quigley HA. The effects of age on mitochondria, axonal transport, and axonal degeneration after chronic IOP elevation using a murine ocular explant model. Exp Eye Res 2018; 172:78-85. [PMID: 29625080 PMCID: PMC5994189 DOI: 10.1016/j.exer.2018.04.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 03/07/2018] [Accepted: 04/02/2018] [Indexed: 01/03/2023]
Abstract
The purpose of this study was to compare younger and older mice after chronic intraocular pressure (IOP) elevation lasting up to 4 days with respect to mitochondrial density, structure, and movement, as well as axonal integrity, in an ex vivo explant model. We studied 2 transgenic mouse strains, both on a C57BL/6J background, one expressing yellow fluorescent protein (YFP) in selected axons and one expressing cyan fluorescent protein (CFP) in all mitochondria. Mice of 4 months or 14 months of age were exposed to chronic IOP by anterior chamber microbead injection for 14 h, 1, 3, or 4 days. The optic nerve head of globe--optic nerve explants were examined by laser scanning microscopy. Mitochondrial density, structure, and movement were quantified in the CFP explants, and axonal integrity was quantified in YFP explants. In control mice, there was a trend towards decreased mitochondrial density (# per mm2) with age when comparing younger to older, control mice, but this was not significant (1947 ± 653 vs 1412 ± 356; p = 0.19). Mitochondrial density decreased after IOP elevation, significantly, by 31%, in younger mice (p = 0.04) but trending towards a decrease, by 22%, in older mice (p = 0.82) compared to age matched controls. Mitochondrial mean size was not altered after chronic IOP elevation for 14 h or more (p ≥ 0.16). When assessing mitochondrial movement, in younger mice, 5% were mobile at any given time; 4% in the anterograde direction and 1% retrograde. In younger untreated tissue, only 75% of explants had moving mitochondria (mean = 15.8 moving/explant), while after glaucoma induction only 24% of explants had moving mitochondria (mean = 4.2 moving/explant; difference from control, p = 0.03). The distance mitochondria traveled in younger mice was unchanged after glaucoma exposure, but in older glaucoma explants the distance traveled was less than half of older controls (p < 0.0003). In younger mice, mitochondrial speed increased after 14 h of elevated IOP (p = 0.006); however, in older glaucoma explants, movement was actually slower than controls (p = 0.02). In RGC-YFP explants, axonal integrity declined significantly after 4 days of IOP elevation to a similar degree in both younger and older mice. Older mice underwent greater loss of mitochondrial movement with chronic IOP elevation than younger mice, but suffered similar short-term axonal fragmentation in C57BL/6J mice. These transgenic strains, studied in explants, permit observations of alterations in intracellular structure and organelle activity in experimental glaucoma.
Collapse
Affiliation(s)
- Elizabeth C Kimball
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA.
| | - Joan L Jefferys
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Mary E Pease
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Ericka N Oglesby
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Cathy Nguyen
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Julie Schaub
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Ian Pitha
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA; The Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Harry A Quigley
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
113
|
Transitional correlation between inner-membrane potential and ATP levels of neuronal mitochondria. Sci Rep 2018; 8:2993. [PMID: 29445117 PMCID: PMC5813116 DOI: 10.1038/s41598-018-21109-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 01/30/2018] [Indexed: 12/11/2022] Open
Abstract
The importance of highly active mitochondria and their contribution to neuronal function has been of recent interest. In most cases, however, mitochondrial activity is estimated using measurements of mitochondrial inner membrane potential (IMPmito), and little is known about the dynamics of native mitochondrial ATP (ATPmito). This study conducted simultaneous imaging of IMPmito and ATPmito in neurons to explore their behaviour and their correlation during physiological mitochondrial/neuronal activity. We found that mitochondrial size, transport velocity and transport direction are not dependent on ATPmito or IMPmito. However, changes in ATPmito and IMPmito during mitochondrial fission/fusion were found; IMPmito depolarized via mitochondrial fission and hyperpolarized via fusion, and ATPmito levels increased after fusion. Because the density of mitochondria is higher in growth cones (GCs) than in axonal processes, integrated ATPmito signals (density × ATPmito) were higher in GCs. This integrated signal in GCs correlated with axonal elongation. However, while the averaged IMPmito was relatively hyperpolarized in GCs, there was no correlation between IMPmito in GCs and axonal elongation. A detailed time-course analysis performed to clarify the reason for these discrepancies showed that IMPmito and ATPmito levels did not always correlate accurately; rather, there were several correlation patterns that changed over time.
Collapse
|
114
|
Shiba-Fukushima K, Ishikawa KI, Inoshita T, Izawa N, Takanashi M, Sato S, Onodera O, Akamatsu W, Okano H, Imai Y, Hattori N. Evidence that phosphorylated ubiquitin signaling is involved in the etiology of Parkinson's disease. Hum Mol Genet 2018; 26:3172-3185. [PMID: 28541509 DOI: 10.1093/hmg/ddx201] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 05/23/2017] [Indexed: 01/07/2023] Open
Abstract
The ubiquitin (Ub) kinase PINK1 and the E3 Ub ligase Parkin, two gene products associated with young-onset Parkinson's disease (PD), participate in mitochondrial quality control. The phosphorylation of mitochondrial polyUb by PINK1, which is activated in a mitochondrial membrane potential (ΔΨm)-dependent manner, facilitates the mitochondrial translocation and concomitant enzymatic activation of Parkin, leading to the clearance of phospho-polyUb-tagged mitochondria via mitophagy. Thus, Ub phosphorylation is a key event in PINK1-Parkin-mediated mitophagy. Here, we examined the role of phospho-Ub signaling in the pathogenesis of PD using fly PD models, human brain tissue and dopaminergic neurons derived from induced pluripotent stem cells (iPSCs) containing Parkin or PINK1 mutations, as well as normal controls. We report that phospho-Ub signaling is highly conserved between humans and Drosophila, and that phospho-Ub signaling and the relocation of axonal mitochondria upon ΔΨm reduction are indeed compromised in human dopaminergic neurons containing Parkin or PINK1 mutations. Moreover, phospho-Ub signaling is prominent in tyrosine hydroxylase-positive neurons compared with tyrosine hydroxylase-negative neurons, suggesting that PINK1-Parkin signaling is more required for dopaminergic neurons. These results shed light on the particular vulnerability of dopaminergic neurons to mitochondrial stress.
Collapse
Affiliation(s)
- Kahori Shiba-Fukushima
- Department of Treatment and Research in Multiple Sclerosis and Neuro-Intractable Disease
| | - Kei-Ichi Ishikawa
- Center for Genomic and Regenerative Medicine.,Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Tsuyoshi Inoshita
- Department of Treatment and Research in Multiple Sclerosis and Neuro-Intractable Disease
| | - Nana Izawa
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan.,Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Masashi Takanashi
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Shigeto Sato
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Osamu Onodera
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | | | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Yuzuru Imai
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan.,Department of Research for Parkinson's Disease, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Treatment and Research in Multiple Sclerosis and Neuro-Intractable Disease.,Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan.,Department of Research for Parkinson's Disease, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
115
|
Melkov A, Abdu U. Regulation of long-distance transport of mitochondria along microtubules. Cell Mol Life Sci 2018; 75:163-176. [PMID: 28702760 PMCID: PMC11105322 DOI: 10.1007/s00018-017-2590-1] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 07/05/2017] [Accepted: 07/06/2017] [Indexed: 11/29/2022]
Abstract
Mitochondria are cellular organelles of crucial importance, playing roles in cellular life and death. In certain cell types, such as neurons, mitochondria must travel long distances so as to meet metabolic demands of the cell. Mitochondrial movement is essentially microtubule (MT) based and is executed by two main motor proteins, Dynein and Kinesin. The organization of the cellular MT network and the identity of motors dictate mitochondrial transport. Tight coupling between MTs, motors, and the mitochondria is needed for the organelle precise localization. Two adaptor proteins are involved directly in mitochondria-motor coupling, namely Milton known also as TRAK, which is the motor adaptor, and Miro, which is the mitochondrial protein. Here, we discuss the active mitochondria transport process, as well as motor-mitochondria coupling in the context of MT organization in different cell types. We focus on mitochondrial trafficking in different cell types, specifically neurons, migrating cells, and polarized epithelial cells.
Collapse
Affiliation(s)
- Anna Melkov
- Department of Life Sciences, Ben-Gurion University, 8410500, Beersheba, Israel
| | - Uri Abdu
- Department of Life Sciences, Ben-Gurion University, 8410500, Beersheba, Israel.
| |
Collapse
|
116
|
Rumora AE, Lentz SI, Hinder LM, Jackson SW, Valesano A, Levinson GE, Feldman EL. Dyslipidemia impairs mitochondrial trafficking and function in sensory neurons. FASEB J 2018; 32:195-207. [PMID: 28904018 PMCID: PMC6191072 DOI: 10.1096/fj.201700206r] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 08/21/2017] [Indexed: 01/11/2023]
Abstract
Mitochondrial trafficking plays a central role in dorsal root ganglion (DRG) neuronal cell survival and neurotransmission by transporting mitochondria from the neuronal cell body throughout the bundles of DRG axons. In type 2 diabetes (T2DM), dyslipidemia and hyperglycemia damage DRG neurons and induce mitochondrial dysfunction; however, the impact of free fatty acids and glucose on mitochondrial trafficking in DRG neurons remains unknown. To evaluate the impact of free fatty acids compared to hyperglycemia on mitochondrial transport, primary adult mouse DRG neuron cultures were treated with physiologic concentrations of palmitate and glucose and assessed for alterations in mitochondrial trafficking, mitochondrial membrane potential, and mitochondrial bioenergetics. Palmitate treatment significantly reduced the number of motile mitochondria in DRG axons, but physiologic concentrations of glucose did not impair mitochondrial trafficking dynamics. Palmitate-treated DRG neurons also exhibited a reduction in mitochondrial velocity, and impaired mitochondrial trafficking correlated with mitochondrial depolarization in palmitate-treated DRG neurons. Finally, we found differential bioenergetic effects of palmitate and glucose on resting and energetically challenged mitochondria in DRG neurons. Together, these results suggest that palmitate induces DRG neuron mitochondrial depolarization, inhibiting axonal mitochondrial trafficking and altering mitochondrial bioenergetic capacity.-Rumora, A. E., Lentz, S. I., Hinder, L. M., Jackson, S. W., Valesano, A., Levinson, G. E., Feldman, E. L. Dyslipidemia impairs mitochondrial trafficking and function in sensory neurons.
Collapse
Affiliation(s)
- Amy E Rumora
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Stephen I Lentz
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Lucy M Hinder
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Samuel W Jackson
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Andrew Valesano
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Gideon E Levinson
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA;
| |
Collapse
|
117
|
Jang DH, Seeger SC, Grady ME, Shofer FS, Eckmann DM. Mitochondrial dynamics and respiration within cells with increased open pore cytoskeletal meshes. Biol Open 2017; 6:1831-1839. [PMID: 29109116 PMCID: PMC5769657 DOI: 10.1242/bio.029009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The cytoskeletal architecture directly affects the morphology, motility, and tensional homeostasis of the cell. In addition, the cytoskeleton is important for mitosis, intracellular traffic, organelle motility, and even cellular respiration. The organelle responsible for a majority of the energy conversion for the cell, the mitochondrion, has a dependence on the cytoskeleton for mobility and function. In previous studies, we established that cytoskeletal inhibitors altered the movement of the mitochondria, their morphology, and their respiration in human dermal fibroblasts. Here, we use this protocol to investigate applicability of power law diffusion to describe mitochondrial locomotion, assessment of rates of fission and fusion in healthy and diseased cells, and differences in mitochondria locomotion in more open networks either in response to cytoskeletal destabilizers or by cell line. We found that mitochondria within fibrosarcoma cells and within fibroblast cells treated with an actin-destabilizing toxin resulted in increased net travel, increased average velocity, and increased diffusion of mitochondria when compared to control fibroblasts. Although the mitochondria within the fibrosarcoma travel further than mitochondria within their healthy counterparts, fibroblasts, the dependence on mitochondria for respiration is much lower with higher rates ofhydrogen peroxide production and was confirmed using the OROBOROS O2K. We also found that rates of fission and fusion of the mitochondria equilibrate despite significant alteration of the cytoskeleton. Rates ranged from 15% to 25%, where the highest rates were observed within the fibrosarcoma cell line. This result is interesting because the fibrosarcoma cell line does not have increased respiration metrics including when compared to fibroblast. Mitochondria travel further, faster, and have an increase in percent mitochondria splitting or joining while not dependent on the mitochondria for a majority of its energy production. This study illustrates the complex interaction between mitochondrial movement and respiration through the disruption of the cytoskeleton. Summary: We assessed the effect of cytoskeletal inhibitors and the use of a calcium ionophore as an additional stressor on mitochondrial motility and bioenergetic function in fibroblasts and fibrosarcoma cells.
Collapse
Affiliation(s)
- David H Jang
- Department of Emergency Medicine, Division of Medical Toxicology and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, 3620 Hamilton Walk, John Morgan Building Room 12, Philadelphia, PA 19104, USA
| | - Sarah C Seeger
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Martha E Grady
- Department of Mechanical Engineering, University of Kentucky, 151 RGAN Building, Lexington, KY 40506, USA
| | - Frances S Shofer
- Department of Emergency Medicine, Division of Medical Toxicology and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, 3620 Hamilton Walk, John Morgan Building Room 12, Philadelphia, PA 19104, USA
| | - David M Eckmann
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, 3620 Hamilton Walk, John Morgan Building Room 27B, Philadelphia, PA 19104, USA .,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
118
|
Misgeld T, Schwarz TL. Mitostasis in Neurons: Maintaining Mitochondria in an Extended Cellular Architecture. Neuron 2017; 96:651-666. [PMID: 29096078 DOI: 10.1016/j.neuron.2017.09.055] [Citation(s) in RCA: 351] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Revised: 09/25/2017] [Accepted: 09/28/2017] [Indexed: 02/06/2023]
Abstract
Neurons have more extended and complex shapes than other cells and consequently face a greater challenge in distributing and maintaining mitochondria throughout their arbors. Neurons can last a lifetime, but proteins turn over rapidly. Mitochondria, therefore, need constant rejuvenation no matter how far they are from the soma. Axonal transport of mitochondria and mitochondrial fission and fusion contribute to this rejuvenation, but local protein synthesis is also likely. Maintenance of a healthy mitochondrial population also requires the clearance of damaged proteins and organelles. This involves degradation of individual proteins, sequestration in mitochondria-derived vesicles, organelle degradation by mitophagy and macroautophagy, and in some cases transfer to glial cells. Both long-range transport and local processing are thus at work in achieving neuronal mitostasis-the maintenance of an appropriately distributed pool of healthy mitochondria for the duration of a neuron's life. Accordingly, defects in the processes that support mitostasis are significant contributors to neurodegenerative disorders.
Collapse
Affiliation(s)
- Thomas Misgeld
- Technical University of Munich, Institute of Neuronal Cell Biology, Munich, Germany; German Center for Neurodegenerative Diseases, Munich, Germany; Munich Cluster for Systems Neurology, Munich, Germany; Center of Integrated Protein Science, Munich, Germany.
| | - Thomas L Schwarz
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA; F.M. Kirby Neurobiology Center, Children's Hospital, Boston, MA, USA.
| |
Collapse
|
119
|
Jackson JG, Robinson MB. Regulation of mitochondrial dynamics in astrocytes: Mechanisms, consequences, and unknowns. Glia 2017; 66:1213-1234. [PMID: 29098734 DOI: 10.1002/glia.23252] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 09/20/2017] [Accepted: 10/09/2017] [Indexed: 12/15/2022]
Abstract
Astrocytes are the major glial cell in the central nervous system. These polarized cells possess numerous processes that ensheath the vasculature and contact synapses. Astrocytes play important roles in synaptic signaling, neurotransmitter synthesis and recycling, control of nutrient uptake, and control of local blood flow. Many of these processes depend on local metabolism and/or energy utilization. While astrocytes respond to increases in neuronal activity and metabolic demand by upregulating glycolysis and glycogenolysis, astrocytes also possess significant capacity for oxidative (mitochondrial) metabolism. Mitochondria mediate energy supply and metabolism, cellular survival, ionic homeostasis, and proliferation. These organelles are dynamic structures undergoing extensive fission and fusion, directed movement along cytoskeletal tracts, and degradation. While many of the mechanisms underlying the dynamics of these organelles and their physiologic roles have been characterized in neurons and other cells, the roles that mitochondrial dynamics play in glial physiology is less well understood. Recent work from several laboratories has demonstrated that mitochondria are present within the fine processes of astrocytes, that their movement is regulated, and that they contribute to local Ca2+ signaling within the astrocyte. They likely play a role in local ATP production and metabolism, particularly that of glutamate. Here we will review these and other findings describing the mechanism by which mitochondrial dynamics are regulated in astrocytes, how mitochondrial dynamics might influence astrocyte and brain metabolism, and draw parallels to mitochondrial dynamics in neurons. Additionally, we present new analyses of the size, distribution, and dynamics of mitochondria in astrocytes performed using in vivo using 2-photon microscopy.
Collapse
Affiliation(s)
- Joshua G Jackson
- Children's Hospital of Philadelphia Research Institute, University of Pennsylvania, Philadelphia, PA, 19104.,Departments of Pediatrics, University of Pennsylvania, Philadelphia, PA, 19104
| | - Michael B Robinson
- Children's Hospital of Philadelphia Research Institute, University of Pennsylvania, Philadelphia, PA, 19104.,Departments of Pediatrics, University of Pennsylvania, Philadelphia, PA, 19104.,Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, 19104
| |
Collapse
|
120
|
Kulbe JR, Hall ED. Chronic traumatic encephalopathy-integration of canonical traumatic brain injury secondary injury mechanisms with tau pathology. Prog Neurobiol 2017; 158:15-44. [PMID: 28851546 PMCID: PMC5671903 DOI: 10.1016/j.pneurobio.2017.08.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 08/09/2017] [Accepted: 08/17/2017] [Indexed: 12/14/2022]
Abstract
In recent years, a new neurodegenerative tauopathy labeled Chronic Traumatic Encephalopathy (CTE), has been identified that is believed to be primarily a sequela of repeated mild traumatic brain injury (TBI), often referred to as concussion, that occurs in athletes participating in contact sports (e.g. boxing, American football, Australian football, rugby, soccer, ice hockey) or in military combatants, especially after blast-induced injuries. Since the identification of CTE, and its neuropathological finding of deposits of hyperphosphorylated tau protein, mechanistic attention has been on lumping the disorder together with various other non-traumatic neurodegenerative tauopathies. Indeed, brains from suspected CTE cases that have come to autopsy have been confirmed to have deposits of hyperphosphorylated tau in locations that make its anatomical distribution distinct for other tauopathies. The fact that these individuals experienced repetitive TBI episodes during their athletic or military careers suggests that the secondary injury mechanisms that have been extensively characterized in acute TBI preclinical models, and in TBI patients, including glutamate excitotoxicity, intracellular calcium overload, mitochondrial dysfunction, free radical-induced oxidative damage and neuroinflammation, may contribute to the brain damage associated with CTE. Thus, the current review begins with an in depth analysis of what is known about the tau protein and its functions and dysfunctions followed by a discussion of the major TBI secondary injury mechanisms, and how the latter have been shown to contribute to tau pathology. The value of this review is that it might lead to improved neuroprotective strategies for either prophylactically attenuating the development of CTE or slowing its progression.
Collapse
Affiliation(s)
- Jacqueline R Kulbe
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, United States; Department of Neuroscience, University of Kentucky College of Medicine, United States
| | - Edward D Hall
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, United States; Department of Neuroscience, University of Kentucky College of Medicine, United States.
| |
Collapse
|
121
|
Graham LC, Eaton SL, Brunton PJ, Atrih A, Smith C, Lamont DJ, Gillingwater TH, Pennetta G, Skehel P, Wishart TM. Proteomic profiling of neuronal mitochondria reveals modulators of synaptic architecture. Mol Neurodegener 2017; 12:77. [PMID: 29078798 PMCID: PMC5659037 DOI: 10.1186/s13024-017-0221-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 10/19/2017] [Indexed: 02/16/2023] Open
Abstract
Background Neurons are highly polarized cells consisting of three distinct functional domains: the cell body (and associated dendrites), the axon and the synapse. Previously, it was believed that the clinical phenotypes of neurodegenerative diseases were caused by the loss of entire neurons, however it has recently become apparent that these neuronal sub-compartments can degenerate independently, with synapses being particularly vulnerable to a broad range of stimuli. Whilst the properties governing the differential degenerative mechanisms remain unknown, mitochondria consistently appear in the literature, suggesting these somewhat promiscuous organelles may play a role in affecting synaptic stability. Synaptic and non-synaptic mitochondrial subpools are known to have different enzymatic properties (first demonstrated by Lai et al., 1977). However, the molecular basis underpinning these alterations, and their effects on morphology, has not been well documented. Methods The current study has employed electron microscopy, label-free proteomics and in silico analyses to characterize the morphological and biochemical properties of discrete sub-populations of mitochondria. The physiological relevance of these findings was confirmed in-vivo using a molecular genetic approach at the Drosophila neuromuscular junction. Results Here, we demonstrate that mitochondria at the synaptic terminal are indeed morphologically different to non-synaptic mitochondria, in both rodents and human patients. Furthermore, generation of proteomic profiles reveals distinct molecular fingerprints – highlighting that the properties of complex I may represent an important specialisation of synaptic mitochondria. Evidence also suggests that at least 30% of the mitochondrial enzymatic activity differences previously reported can be accounted for by protein abundance. Finally, we demonstrate that the molecular differences between discrete mitochondrial sub-populations are capable of selectively influencing synaptic morphology in-vivo. We offer several novel mitochondrial candidates that have the propensity to significantly alter the synaptic architecture in-vivo. Conclusions Our study demonstrates discrete proteomic profiles exist dependent upon mitochondrial subcellular localization and selective alteration of intrinsic mitochondrial proteins alters synaptic morphology in-vivo. Electronic supplementary material The online version of this article (10.1186/s13024-017-0221-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Laura C Graham
- Division of Neurobiology, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK.,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
| | - Samantha L Eaton
- Division of Neurobiology, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK.,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
| | - Paula J Brunton
- Division of Neurobiology, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Abdelmadjid Atrih
- FingerPrints Proteomics Facility, College of Life Sciences, University of Dundee, Dundee, UK
| | - Colin Smith
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.,Department of Academic Neuropathology, University of Edinburgh, CCBS, Chancellor's Building, Little France, Edinburgh, UK
| | - Douglas J Lamont
- FingerPrints Proteomics Facility, College of Life Sciences, University of Dundee, Dundee, UK
| | - Thomas H Gillingwater
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.,Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, Edinburgh, UK
| | - Giuseppa Pennetta
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.,Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, Edinburgh, UK
| | - Paul Skehel
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.,Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, Edinburgh, UK
| | - Thomas M Wishart
- Division of Neurobiology, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK. .,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
122
|
Hamid HS, Hayes JM, Feldman EL, Lentz SI. Three-dimensional Imaging and Analysis of Mitochondria within Human Intraepidermal Nerve Fibers. J Vis Exp 2017. [PMID: 28994751 DOI: 10.3791/53369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The goal of this protocol is to study mitochondria within intraepidermal nerve fibers. Therefore, 3D imaging and analysis techniques were developed to isolate nerve-specific mitochondria and evaluate disease-induced alterations of mitochondria in the distal tip of sensory nerves. The protocol combines fluorescence immunohistochemistry, confocal microscopy and 3D image analysis techniques to visualize and quantify nerve-specific mitochondria. Detailed parameters are defined throughout the procedures in order to provide a concrete example of how to use these techniques to isolate nerve-specific mitochondria. Antibodies were used to label nerve and mitochondrial signals within tissue sections of skin punch biopsies, which was followed by indirect immunofluorescence to visualize nerves and mitochondria with a green and red fluorescent signal respectively. Z-series images were acquired with confocal microscopy and 3D analysis software was used to process and analyze the signals. It is not necessary to follow the exact parameters described within, but it is important to be consistent with the ones chosen throughout the staining, acquisition and analysis steps. The strength of this protocol is that it is applicable to a wide variety of circumstances where one fluorescent signal is used to isolate other signals that would otherwise be impossible to study alone.
Collapse
|
123
|
Murray JW, Yin D, Wolkoff AW. Reduction of organelle motility by removal of potassium and other solutes. PLoS One 2017; 12:e0184898. [PMID: 28922372 PMCID: PMC5602639 DOI: 10.1371/journal.pone.0184898] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 09/01/2017] [Indexed: 12/02/2022] Open
Abstract
There are surprisingly few studies that describe how the composition of cell culture medium may affect the trafficking of organelles. Here we utilize time lapse multi-channel fluorescent imaging to show that short term exposure of Huh-7 cells to medium lacking potassium, sodium, or chloride strongly reduces but does not eliminate the characteristic back and forth and cell-traversing movement of fluorescent EGF (FL-EGF) containing organelles. We focused on potassium because of its relatively low abundance in media and serum and its energy requiring accumulation into cells. Upon exposure to potassium free medium, organelle motility declined steadily through 90 min and then persisted at a low level. Reduced motility was confirmed in 5 independent cell lines and for organelles of the endocytic pathway (FL-EGF and Lysotracker), autophagosomes (LC3-GFP), and mitochondria (TMRE). As has been previously established, potassium free medium also inhibited endocytosis. We expected that diminished cellular metabolism would precede loss of organelle motility. However, extracellular flux analysis showed near normal mitochondrial oxygen consumption and only a small decrease in extracellular acidification, the latter suggesting decreased glycolysis or proton efflux. Other energy dependent activities such as the accumulation of Lysotracker, TMRE, DiBAC4(3), and the exclusion of propidium iodide remained intact, as did the microtubule cytoskeleton. We took advantage of cell free in vitro motility assays and found that removal of potassium or sodium from the reconstituted cytosolic medium decreased the movement of endosomes on purified microtubules. The results indicate that although changes in proton homeostasis and cell energetics under solute depletion are not fully understood, potassium as well as sodium appear to be directly required by the motile machinery of organelles for optimal trafficking.
Collapse
Affiliation(s)
- John W. Murray
- Marion Bessin Liver Research Center, Division of Gastroenterology and Liver Diseases, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, United States of America
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, United States of America
- * E-mail:
| | - David Yin
- Marion Bessin Liver Research Center, Division of Gastroenterology and Liver Diseases, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, United States of America
| | - Allan W. Wolkoff
- Marion Bessin Liver Research Center, Division of Gastroenterology and Liver Diseases, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, United States of America
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, United States of America
| |
Collapse
|
124
|
Norkett R, Modi S, Kittler JT. Mitochondrial roles of the psychiatric disease risk factor DISC1. Schizophr Res 2017; 187:47-54. [PMID: 28087269 DOI: 10.1016/j.schres.2016.12.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 12/17/2016] [Accepted: 12/22/2016] [Indexed: 12/31/2022]
Abstract
Ion transport during neuronal signalling utilizes the majority of the brain's energy supply. Mitochondria are key sites for energy provision through ATP synthesis and play other important roles including calcium buffering. Thus, tightly regulated distribution and function of these organelles throughout the intricate architecture of the neuron is essential for normal synaptic communication. Therefore, delineating mechanisms coordinating mitochondrial transport and function is essential for understanding nervous system physiology and pathology. While aberrant mitochondrial transport and dynamics have long been associated with neurodegenerative disease, they have also more recently been linked to major mental illness including schizophrenia, autism and depression. However, the underlying mechanisms have yet to be elucidated, due to an incomplete understanding of the combinations of genetic and environmental factors contributing to these conditions. Consequently, the DISC1 gene has undergone intense study since its discovery at the site of a balanced chromosomal translocation, segregating with mental illness in a Scottish pedigree. The precise molecular functions of DISC1 remain elusive. Reported functions of DISC1 include regulation of intracellular signalling pathways, neuronal migration and dendritic development. Intriguingly, a role for DISC1 in mitochondrial homeostasis and transport is fast emerging. Therefore, a major function of DISC1 in regulating mitochondrial distribution, ATP synthesis and calcium buffering may be disrupted in psychiatric disease. In this review, we discuss the links between DISC1 and mitochondria, considering both trafficking of these organelles and their function, and how, via these processes, DISC1 may contribute to the regulation of neuronal behavior in normal and psychiatric disease states.
Collapse
Affiliation(s)
- R Norkett
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK
| | - S Modi
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK
| | - J T Kittler
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, UK.
| |
Collapse
|
125
|
Roberts RC. Postmortem studies on mitochondria in schizophrenia. Schizophr Res 2017; 187:17-25. [PMID: 28189530 PMCID: PMC5550365 DOI: 10.1016/j.schres.2017.01.056] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 01/27/2017] [Accepted: 01/30/2017] [Indexed: 01/02/2023]
Abstract
The aim of this paper is to provide a brief review of mitochondrial structure as it relates to function and then present abnormalities in mitochondria in postmortem schizophrenia with a focus on ultrastructure. Function, morphology, fusion, fission, motility, ΔΨmem, ATP production, mitochondrial derived vesicles, and mitochondria-associated ER membranes will be briefly covered. Pathology in mitochondria has long been implicated in schizophrenia, as shown by genetic, proteomic, enzymatic and anatomical abnormalities. The cortex and basal ganglia will be reviewed. In the anterior cingulate cortex, the number of mitochondria per neuronal somata in layers 5/6 in schizophrenia is decreased by 43%. There are also fewer mitochondria in terminals forming axospinous synapses. In the caudate and putamen the number of mitochondria is abnormal in both glial cells and neurons in schizophrenia subjects, the extent of which depends on treatment, response and predominant lifetime symptoms. Treatment-responsive schizophrenia subjects had about a 40% decrease in the number of mitochondria per synapse in the caudate nucleus and putamen, while treatment resistant cases had normal values. A decrease in mitochondrial density in the neuropil distinguishes paranoid from undifferentiated schizophrenia. The appearance, size and density of mitochondria were normal in the nucleus accumbens. In the substantia nigra, COX subunits were affected in rostral regions. Mitochondrial hyperplasia occurs within axon terminals that synapse onto dopamine neurons, but mitochondria in dopamine neuronal somata are similar in size and number. In schizophrenia, mitochondria are differentially affected depending on the brain region, cell type, subcellular location, treatment status, treatment response and symptoms.
Collapse
Affiliation(s)
- Rosalinda C. Roberts
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama, Birmingham, AL 35294
| |
Collapse
|
126
|
Murphy LC, Millar JK. Regulation of mitochondrial dynamics by DISC1, a putative risk factor for major mental illness. Schizophr Res 2017; 187:55-61. [PMID: 28082141 DOI: 10.1016/j.schres.2016.12.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 12/22/2016] [Accepted: 12/23/2016] [Indexed: 12/22/2022]
Abstract
Mitochondria are dynamic organelles that are essential to power the process of neurotransmission. Neurons must therefore ensure that mitochondria maintain their functional integrity and are efficiently transported along the full extent of the axons and dendrites, from soma to synapses. Mitochondrial dynamics (trafficking, fission and fusion) co-ordinately regulate mitochondrial quality control and function. DISC1 is a component of the mitochondrial transport machinery and regulates mitochondrial dynamics. DISC1's role in this is adversely affected by sequence variants connected to brain structure/function and disease risk, and by mutant truncation. The DISC1 interactors NDE1 and GSK3β are also involved, indicating a convergence of putative risk factors for psychiatric illness upon mitochondrial dynamics.
Collapse
Affiliation(s)
- Laura C Murphy
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetic and Molecular Medicine at the University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| | - J Kirsty Millar
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetic and Molecular Medicine at the University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK.
| |
Collapse
|
127
|
Afreen S, Riherd Methner DN, Ferreira A. Tau 45-230 association with the cytoskeleton and membrane-bound organelles: Functional implications in neurodegeneration. Neuroscience 2017; 362:104-117. [PMID: 28844006 DOI: 10.1016/j.neuroscience.2017.08.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 08/10/2017] [Accepted: 08/14/2017] [Indexed: 12/15/2022]
Abstract
The dysregulation of posttranslational modifications of the microtubule-associated protein (MAP) tau plays a key role in Alzheimer's disease (AD) and related disorders. Thus, we have previously shown that beta amyloid (Aβ)-induced neurotoxicity was mediated, at least in part, by tau cleavage into the tau45-230 fragment. However, the mechanisms underlying the toxicity of tau45-230 remain unknown. To get insights into such mechanisms, we first determined the subcellular localization of this tau fragment in hippocampal neurons. Tau45-230 was easily detectable in cell bodies and processes extended by these neurons. In addition, cell extraction experiments performed using Triton X-100 and saponin showed that a pool of tau45-230 was associated with the cytoskeleton and the cytoskeleton plus membrane-bound organelles, respectively, in cultured hippocampal neurons. Furthermore, they suggested that these associations were independent of the presence of full-length tau. We also assessed whether this tau fragment could alter axonal transport. Our results indicated that tau45-230 significantly reduced the number of organelles transported along hippocampal axons. This altered axonal transport did not correlate with changes in the total number of organelles present in these cells or in motor protein levels. Together these results suggested that tau45-230 could exert its toxic effects by partially blocking axonal transport along microtubules thus contributing to the early pathology of AD.
Collapse
Affiliation(s)
- Sana Afreen
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - D Nicole Riherd Methner
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Adriana Ferreira
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
128
|
Akabane S, Uno M, Tani N, Shimazaki S, Ebara N, Kato H, Kosako H, Oka T. PKA Regulates PINK1 Stability and Parkin Recruitment to Damaged Mitochondria through Phosphorylation of MIC60. Mol Cell 2017; 62:371-384. [PMID: 27153535 DOI: 10.1016/j.molcel.2016.03.037] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 02/25/2016] [Accepted: 03/31/2016] [Indexed: 01/09/2023]
Abstract
A mitochondrial kinase, PTEN-induced putative kinase 1 (PINK1), selectively recruits the ubiquitin ligase Parkin to damaged mitochondria, which modifies mitochondria by polyubiquitination, leading to mitochondrial autophagy. Here, we report that treatment with an adenylate cyclase agonist or expression of protein kinase A (PKA) impairs Parkin recruitment to damaged mitochondria and decreases PINK1 protein levels. We identified a mitochondrial membrane protein, MIC60 (also known as mitofilin), as a PKA substrate. Mutational and mass spectrometric analyses revealed that the Ser528 residue of MIC60 undergoes PKA-dependent phosphorylation. MIC60 transiently interacts with PINK1, and MIC60 downregulation leads to a reduction in PINK1 and mislocalization of Parkin. Phosphorylation-mimic mutants of MIC60 fail to restore the defect in Parkin recruitment in MIC60-knocked down cells, whereas a phosphorylation-deficient MIC60 mutant facilitates the mitochondrial localization of Parkin. Our findings indicate that PKA-mediated phosphorylation of MIC60 negatively regulates mitochondrial clearance that is initiated by PINK1 and Parkin.
Collapse
Affiliation(s)
- Shiori Akabane
- Department of Life Science, Rikkyo University, Toshima-ku, Tokyo 171-8501, Japan
| | - Midori Uno
- Department of Life Science, Rikkyo University, Toshima-ku, Tokyo 171-8501, Japan
| | - Naoki Tani
- Liaison Laboratory Research Promotion Center, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Shunta Shimazaki
- Department of Life Science, Rikkyo University, Toshima-ku, Tokyo 171-8501, Japan
| | - Natsumi Ebara
- Department of Life Science, Rikkyo University, Toshima-ku, Tokyo 171-8501, Japan
| | - Hiroki Kato
- Division of Oral Health, Growth, and Development, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Hidetaka Kosako
- Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Toshihiko Oka
- Department of Life Science, Rikkyo University, Toshima-ku, Tokyo 171-8501, Japan.
| |
Collapse
|
129
|
Han SM, Baig HS, Hammarlund M. Mitochondria Localize to Injured Axons to Support Regeneration. Neuron 2017; 92:1308-1323. [PMID: 28009276 DOI: 10.1016/j.neuron.2016.11.025] [Citation(s) in RCA: 174] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 08/31/2016] [Accepted: 11/08/2016] [Indexed: 12/19/2022]
Abstract
Axon regeneration is essential to restore the nervous system after axon injury. However, the neuronal cell biology that underlies axon regeneration is incompletely understood. Here we use in vivo, single-neuron analysis to investigate the relationship between nerve injury, mitochondrial localization, and axon regeneration. Mitochondria translocate into injured axons so that average mitochondria density increases after injury. Moreover, single-neuron analysis reveals that axons that fail to increase mitochondria have poor regeneration. Experimental alterations to axonal mitochondrial distribution or mitochondrial respiratory chain function result in corresponding changes to regeneration outcomes. Axonal mitochondria are specifically required for growth-cone migration, identifying a key energy challenge for injured neurons. Finally, mitochondrial localization to the axon after injury is regulated in part by dual-leucine zipper kinase 1 (DLK-1), a conserved regulator of axon regeneration. These data identify regulation of axonal mitochondria as a new cell-biological mechanism that helps determine the regenerative response of injured neurons.
Collapse
Affiliation(s)
- Sung Min Han
- Departments of Genetics and Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| | - Huma S Baig
- Departments of Genetics and Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| | - Marc Hammarlund
- Departments of Genetics and Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA.
| |
Collapse
|
130
|
Chen Y, Deng J, Wang P, Yang M, Chen X, Zhu L, Liu J, Lu B, Shen Y, Fushimi K, Xu Q, Wu JY. PINK1 and Parkin are genetic modifiers for FUS-induced neurodegeneration. Hum Mol Genet 2017; 25:5059-5068. [PMID: 27794540 DOI: 10.1093/hmg/ddw310] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 09/02/2016] [Indexed: 12/13/2022] Open
Abstract
Dysregulation of Fused in Sarcoma (FUS) gene expression is associated with fronto-temporal lobar degeneration (FTLD), and missense mutations in the FUS gene have been identified in patients affected by amyotrophic lateral sclerosis (ALS). However, molecular and cellular defects underlying FUS proteinopathy remain to be elucidated. Here, we examined whether genes important for mitochondrial quality control play a role in FUS proteinopathy. In our genetic screening, Pink1 and Park genes were identified as modifiers of neurodegeneration phenotypes induced by wild type (Wt) or ALS-associated P525L-mutant human FUS. Down-regulating expression of either Pink1 or Parkin genes ameliorated FUS-induced neurodegeneration phenotypes. The protein levels of PINK1 and Parkin were elevated in cells overexpressing FUS. Remarkably, ubiquitinylation of Miro1 protein, a downstream target of the E3 ligase activity of Parkin, was also increased in cells overexpressing FUS protein. In fly motor neurons expressing FUS, both motility and processivity of mitochondrial axonal transport were reduced by expression of either Wt- or P525L-mutant FUS. Finally, down-regulating PINK1 or Parkin partially rescued the locomotive defects and enhanced the survival rate in transgenic flies expressing FUS. Our data indicate that PINK1 and Parkin play an important role in FUS-induced neurodegeneration. This study has uncovered a previously unknown link between FUS proteinopathy and PINK1/Parkin genes, providing new insights into the pathogenesis of FUS proteinopathy.
Collapse
Affiliation(s)
- Yanbo Chen
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Science, Beijing, China.,Department of Neurology, Center for Genetic Medicine, Lurie Cancer Center, Northwestern University Feinberg School of Medicine, 303 E. Superior, Chicago, IL, USA.,National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences & Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, China
| | - Jianwen Deng
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Science, Beijing, China
| | - Peng Wang
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Science, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Mengxue Yang
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Science, Beijing, China.,Department of Neurology, Center for Genetic Medicine, Lurie Cancer Center, Northwestern University Feinberg School of Medicine, 303 E. Superior, Chicago, IL, USA
| | - Xiaoping Chen
- Department of Neurology, Center for Genetic Medicine, Lurie Cancer Center, Northwestern University Feinberg School of Medicine, 303 E. Superior, Chicago, IL, USA
| | - Li Zhu
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Science, Beijing, China
| | - Jianghong Liu
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Science, Beijing, China
| | - Bingwei Lu
- Department of Pathology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Yan Shen
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences & Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, China
| | - Kazuo Fushimi
- Department of Neurology, Center for Genetic Medicine, Lurie Cancer Center, Northwestern University Feinberg School of Medicine, 303 E. Superior, Chicago, IL, USA
| | - Qi Xu
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences & Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, China
| | - Jane Y Wu
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Science, Beijing, China.,Department of Neurology, Center for Genetic Medicine, Lurie Cancer Center, Northwestern University Feinberg School of Medicine, 303 E. Superior, Chicago, IL, USA
| |
Collapse
|
131
|
Compartmentalized Regulation of Parkin-Mediated Mitochondrial Quality Control in the Drosophila Nervous System In Vivo. J Neurosci 2017; 36:7375-91. [PMID: 27413149 DOI: 10.1523/jneurosci.0633-16.2016] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/18/2016] [Indexed: 12/14/2022] Open
Abstract
UNLABELLED In neurons, the normal distribution and selective removal of mitochondria are considered essential for maintaining the functions of the large asymmetric cell and its diverse compartments. Parkin, a E3 ubiquitin ligase associated with familial Parkinson's disease, has been implicated in mitochondrial dynamics and removal in cells including neurons. However, it is not clear how Parkin functions in mitochondrial turnover in vivo, or whether Parkin-dependent events of the mitochondrial life cycle occur in all neuronal compartments. Here, using the live Drosophila nervous system, we investigated the involvement of Parkin in mitochondrial dynamics, distribution, morphology, and removal. Contrary to our expectations, we found that Parkin-deficient animals do not accumulate senescent mitochondria in their motor axons or neuromuscular junctions; instead, they contain far fewer axonal mitochondria, and these displayed normal motility behavior, morphology, and metabolic state. However, the loss of Parkin did produce abnormal tubular and reticular mitochondria restricted to the motor cell bodies. In addition, in contrast to drug-treated, immortalized cells in vitro, mature motor neurons rarely displayed Parkin-dependent mitophagy. These data indicate that the cell body is the focus of Parkin-dependent mitochondrial quality control in neurons, and argue that a selection process allows only healthy mitochondria to pass from cell bodies to axons, perhaps to limit the impact of mitochondrial dysfunction. SIGNIFICANCE STATEMENT Parkin has been proposed to police mitochondrial fidelity by binding to dysfunctional mitochondria via PTEN (phosphatase and tensin homolog)-induced putative kinase 1 (PINK1) and targeting them for autophagic degradation. However, it is unknown whether and how the PINK1/Parkin pathway regulates the mitochondrial life cycle in neurons in vivo Using Drosophila motor neurons, we show that parkin disruption generates an abnormal mitochondrial network in cell bodies in vivo and reduces the number of axonal mitochondria without producing any defects in their axonal transport, morphology, or metabolic state. Furthermore, while cultured neurons display Parkin-dependent axonal mitophagy, we find this is vanishingly rare in vivo under normal physiological conditions. Thus, both the spatial distribution and mechanism of mitochondrial quality control in vivo differ substantially from those observed in vitro.
Collapse
|
132
|
Analysis of mitochondrial organization and function in the Drosophila blastoderm embryo. Sci Rep 2017; 7:5502. [PMID: 28710464 PMCID: PMC5511145 DOI: 10.1038/s41598-017-05679-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 06/01/2017] [Indexed: 11/25/2022] Open
Abstract
Mitochondria are inherited maternally as globular and immature organelles in metazoan embryos. We have used the Drosophila blastoderm embryo to characterize their morphology, distribution and functions in embryogenesis. We find that mitochondria are relatively small, dispersed and distinctly distributed along the apico-basal axis in proximity to microtubules by motor protein transport. Live imaging, photobleaching and photoactivation analyses of mitochondrially targeted GFP show that they are mobile in the apico-basal axis along microtubules and are immobile in the lateral plane thereby associating with one syncytial cell. Photoactivated mitochondria distribute equally to daughter cells across the division cycles. ATP depletion by pharmacological and genetic inhibition of the mitochondrial electron transport chain (ETC) activates AMPK and decreases syncytial metaphase furrow extension. In summary, we show that small and dispersed mitochondria of the Drosophila blastoderm embryo localize by microtubule transport and provide ATP locally for the fast syncytial division cycles. Our study opens the possibility of use of Drosophila embryogenesis as a model system to study the impact of maternal mutations in mitochondrial morphology and metabolism on embryo patterning and differentiation.
Collapse
|
133
|
Cobalt inhibits motility of axonal mitochondria and induces axonal degeneration in cultured dorsal root ganglion cells of rat. Cell Biol Toxicol 2017; 34:93-107. [PMID: 28656345 DOI: 10.1007/s10565-017-9402-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Accepted: 06/15/2017] [Indexed: 10/19/2022]
Abstract
Cobalt is a trace element that localizes in the human body as cobalamin, also known as vitamin B12. Excessive cobalt exposure induces a peripheral neuropathy, the mechanisms of which are yet to be elucidated. We investigated how cobalt may affect mitochondrial motility in primary cultures of rat dorsal root ganglion (DRG). We observed mitochondrial motility by time-lapse imaging after DsRed2 tagging via lentivirus, mitochondrial structure using transmission electron microscopy (TEM), and axonal swelling using immunocytochemical staining. The concentration of cobaltous ion (Co2+) required to significantly suppress mitochondrial motility is lower than that required to induce axonal swelling following a 24-h treatment. Exposure to relatively low concentrations of Co2+ for 48 h suppressed mitochondrial motility without leading to axonal swelling. TEM images indicated that Co2+ induces mitochondrial destruction. Our results show that destruction of the axonal mitochondria precedes the axonal degeneration induced by Co2+ exposure.
Collapse
|
134
|
Robinson BJ, Stanisavljevic B, Silverman MA, Scalettar BA. Stochastic Subcellular Organization of Dense-Core Vesicles Revealed by Point Pattern Analysis. Biophys J 2017; 111:852-863. [PMID: 27558728 DOI: 10.1016/j.bpj.2016.07.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 06/20/2016] [Accepted: 07/05/2016] [Indexed: 12/20/2022] Open
Abstract
Dense-core vesicles (DCVs) are regulated secretory organelles found in many types of neurons. In neurons of the hippocampus, their cargo includes proteins that mediate several pivotal processes, including differentiation and synaptic plasticity. Motivated by interest in DCV distribution and its impact on cargo action, we have used fluorescence microscopy and statistical analysis to develop a quantitative model of the subcellular organization of DCVs in hippocampal neurons that are spontaneously active (their most prevalent state). We also have tested the functionally motivated hypothesis that these organelles are synaptically enriched. Variance-to-mean ratio, frequency distribution, and Moran's autocorrelation analyses reveal that DCV distribution along shafts, and within synapses, follows Poisson statistics, establishing that stochastically dictated organization sustains cargo function. Occupancy in boutons exceeds that at nearby extrasynaptic axonal sites by approximately threefold, revealing significant local presynaptic enrichment. Widespread stochastic organization is consistent with the emerging functional importance of synaptically and extrasynaptically localized DCVs. Presynaptic enrichment is consistent with the established importance of protecting presynaptic sites from depletion of DCV cargo. These results enhance understanding of the link between DCV organization and mechanisms of cargo action, and they reinforce the emerging theme that randomness is a prevalent aspect of synaptic organization and composition.
Collapse
Affiliation(s)
- Benjamin J Robinson
- Department of Physics, Lewis & Clark College, Portland, Oregon; Department of Mathematics, Lewis & Clark College, Portland, Oregon
| | - Bogdan Stanisavljevic
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Michael A Silverman
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Bethe A Scalettar
- Department of Physics, Lewis & Clark College, Portland, Oregon; Program in Biochemistry and Molecular Biology, Lewis & Clark College, Portland, Oregon.
| |
Collapse
|
135
|
Sheng ZH. The Interplay of Axonal Energy Homeostasis and Mitochondrial Trafficking and Anchoring. Trends Cell Biol 2017; 27:403-416. [PMID: 28228333 PMCID: PMC5440189 DOI: 10.1016/j.tcb.2017.01.005] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 01/15/2017] [Accepted: 01/20/2017] [Indexed: 01/02/2023]
Abstract
Mitochondria are key cellular power plants essential for neuronal growth, survival, function, and regeneration after injury. Given their unique morphological features, neurons face exceptional challenges in maintaining energy homeostasis at distal synapses and growth cones where energy is in high demand. Efficient regulation of mitochondrial trafficking and anchoring is critical for neurons to meet altered energy requirements. Mitochondrial dysfunction and impaired transport have been implicated in several major neurological disorders. Thus, research into energy-mediated regulation of mitochondrial recruitment and redistribution is an important emerging frontier. In this review, I discuss new insights into the mechanisms regulating mitochondrial trafficking and anchoring, and provide an updated overview of how mitochondrial motility maintains energy homeostasis in axons, thus contributing to neuronal growth, regeneration, and synaptic function.
Collapse
Affiliation(s)
- Zu-Hang Sheng
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706, USA.
| |
Collapse
|
136
|
Yin X, Kidd GJ, Ohno N, Perkins GA, Ellisman MH, Bastian C, Brunet S, Baltan S, Trapp BD. Proteolipid protein-deficient myelin promotes axonal mitochondrial dysfunction via altered metabolic coupling. J Cell Biol 2017; 215:531-542. [PMID: 27872255 PMCID: PMC5119941 DOI: 10.1083/jcb.201607099] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/01/2016] [Accepted: 10/25/2016] [Indexed: 12/12/2022] Open
Abstract
The authors show that central nervous system myelin lacking proteolipid protein (PLP) induces mitochondrial dysfunction, including altered motility, degeneration, and ectopic smooth endoplasmic reticulum interactions, leading to axonal structural defects and degeneration. Mutated PLP occurs in hereditary spastic paraplegia, and these cellular effects provide potential insight into the pathology of the disease. Hereditary spastic paraplegia (HSP) is a neurological syndrome characterized by degeneration of central nervous system (CNS) axons. Mutated HSP proteins include myelin proteolipid protein (PLP) and axon-enriched proteins involved in mitochondrial function, smooth endoplasmic reticulum (SER) structure, and microtubule (MT) stability/function. We characterized axonal mitochondria, SER, and MTs in rodent optic nerves where PLP is replaced by the peripheral nerve myelin protein, P0 (P0-CNS mice). Mitochondrial pathology and degeneration were prominent in juxtaparanodal axoplasm at 1 mo of age. In wild-type (WT) optic nerve axons, 25% of mitochondria–SER associations occurred on extensions of the mitochondrial outer membrane. Mitochondria–SER associations were reduced by 86% in 1-mo-old P0-CNS juxtaparanodal axoplasm. 1-mo-old P0-CNS optic nerves were more sensitive to oxygen-glucose deprivation and contained less adenosine triphosphate (ATP) than WT nerves. MT pathology and paranodal axonal ovoids were prominent at 6 mo. These data support juxtaparanodal mitochondrial degeneration, reduced mitochondria–SER associations, and reduced ATP production as causes of axonal ovoid formation and axonal degeneration.
Collapse
Affiliation(s)
- Xinghua Yin
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Grahame J Kidd
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Nobuhiko Ohno
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Guy A Perkins
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA 92093
| | - Mark H Ellisman
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA 92093
| | - Chinthasagar Bastian
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Sylvain Brunet
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Selva Baltan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Bruce D Trapp
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| |
Collapse
|
137
|
Fedorovich SV, Waseem TV, Puchkova LV. Biogenetic and morphofunctional heterogeneity of mitochondria: the case of synaptic mitochondria. Rev Neurosci 2017; 28:363-373. [DOI: 10.1515/revneuro-2016-0077] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 12/29/2016] [Indexed: 12/26/2022]
Abstract
AbstractThe mitochondria of different cells are different in their morphological and biochemical properties. These organelles generate free radicals during activity, leading inevitably to mitochondrial DNA damage. It is not clear how this problem is addressed in long-lived cells, such as neurons. We propose the hypothesis that mitochondria within the same cell also differ in lifespan and ability to divide. According to our suggestion, cells have a pool of ‘stem’ mitochondria with low metabolic activity and a pool of ‘differentiated’ mitochondria with significantly shorter lifespans and high metabolic activity. We consider synaptic mitochondria as a possible example of ‘differentiated’ mitochondria. They are significantly smaller than mitochondria from the cell body, and they are different in key enzyme activity levels, proteome, and lipidome. Synaptic mitochondria are more sensitive to different damaging factors. It has been established that neurons have a sorting mechanism that sends mitochondria with high membrane potential to presynaptic endings. This review describes the properties of synaptic mitochondria and their role in the regulation of synaptic transmission.
Collapse
Affiliation(s)
- Sergei V. Fedorovich
- Institute of Biophysics and Cell Engineering, Akademicheskaya St., 27, Minsk 220072, Belarus
| | - Tatyana V. Waseem
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Ludmila V. Puchkova
- Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya str., 29, St. Petersburg 195251, Russia
- ITMO University, Kronverksky av., 49, St.Petersburg 197101, Russia
- Institute of Experimental Medicine, Pavlova str., 12, St.Petersburg 197376, Russia
| |
Collapse
|
138
|
Improvement of mitochondrial function mediated the neuroprotective effect of 5-(4-hydroxy-3-dimethoxybenzylidene)-2-thioxo-4-thiazolidinone in rats with cerebral ischemia-reperfusion injuries. Oncotarget 2017; 8:61193-61202. [PMID: 28977856 PMCID: PMC5617416 DOI: 10.18632/oncotarget.18048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 04/25/2017] [Indexed: 11/25/2022] Open
Abstract
Deficits in mitochondrial function is a critical inducement in the major pathways that drive neuronal cell death in ischemic process particularly. Drugs target to improve the mitochondrial function may be a feasible therapeutic choice in treatment with ischemic diseases. In the present study, we investigated whether 5-(4-hydroxy-3-dimethoxybenzylidene)-2-thioxo-4-thiazolidinone (RD-1), a compound derived from rhodanine, could protect against ischemic neuronal damage via improving mitochondrial function. We tested the neuroprotective effect of RD-1 both in rats modeled by middle cerebral artery occlusion reperfusion in vivo and in primary cortical neurons subjected to hypoxia/reperfusion injury in vitro. Results showed that treatment with RD-1 for 14 days remarkably reduced infarct size, decreased neurological deficit score and accelerated the recovery of somatosensory function in vivo. Meanwhile, RD-1 also increased the cellular viability after 48 h treatment in vitro. In addition, RD-1 protected the primary cortical neurons against mitochondrial damage as evidenced by stabilizing the mitochondrial membrane potential and reducing the overproduction of reactive oxygen species. Furthermore, hypoxia/reperfusion injury induced damaged mitochondrial axonal transport and consequently neurotransmitter release disorder, which were ameliorated by RD-1 treatment. Besides, RD-1 inhibited the downregulation of proteins related with mitochondrial transport and neurotransmitter release induced by ischemic injury both in vivo and in vitro. The obtained data demonstrated the neuroprotective effect of RD-1 and the involved mechanisms were partially attributed to the improvement in mitochondrial function and the synaptic activity. Our study indicated that RD-1 may be a potential therapeutic drug for the ischemic stroke therapy.
Collapse
|
139
|
Liao PC, Tandarich LC, Hollenbeck PJ. ROS regulation of axonal mitochondrial transport is mediated by Ca2+ and JNK in Drosophila. PLoS One 2017; 12:e0178105. [PMID: 28542430 PMCID: PMC5436889 DOI: 10.1371/journal.pone.0178105] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/06/2017] [Indexed: 12/31/2022] Open
Abstract
Mitochondria perform critical functions including aerobic ATP production and calcium (Ca2+) homeostasis, but are also a major source of reactive oxygen species (ROS) production. To maintain cellular function and survival in neurons, mitochondria are transported along axons, and accumulate in regions with high demand for their functions. Oxidative stress and abnormal mitochondrial axonal transport are associated with neurodegenerative disorders. However, we know little about the connection between these two. Using the Drosophila third instar larval nervous system as the in vivo model, we found that ROS inhibited mitochondrial axonal transport more specifically, primarily due to reduced flux and velocity, but did not affect transport of other organelles. To understand the mechanisms underlying these effects, we examined Ca2+ levels and the JNK (c-Jun N-terminal Kinase) pathway, which have been shown to regulate mitochondrial transport and general fast axonal transport, respectively. We found that elevated ROS increased Ca2+ levels, and that experimental reduction of Ca2+ to physiological levels rescued ROS-induced defects in mitochondrial transport in primary neuron cell cultures. In addition, in vivo activation of the JNK pathway reduced mitochondrial flux and velocities, while JNK knockdown partially rescued ROS-induced defects in the anterograde direction. We conclude that ROS have the capacity to regulate mitochondrial traffic, and that Ca2+ and JNK signaling play roles in mediating these effects. In addition to transport defects, ROS produces imbalances in mitochondrial fission-fusion and metabolic state, indicating that mitochondrial transport, fission-fusion steady state, and metabolic state are closely interrelated in the response to ROS.
Collapse
Affiliation(s)
- Pin-Chao Liao
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, United States of America
| | - Lauren C. Tandarich
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, United States of America
| | - Peter J. Hollenbeck
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, United States of America
- * E-mail:
| |
Collapse
|
140
|
|
141
|
Di Benedetto G, Gerbino A, Lefkimmiatis K. Shaping mitochondrial dynamics: The role of cAMP signalling. Biochem Biophys Res Commun 2017; 500:65-74. [PMID: 28501614 DOI: 10.1016/j.bbrc.2017.05.041] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 05/07/2017] [Indexed: 12/25/2022]
Abstract
In recent years, our idea of mitochondria evolved from "mere" energy and metabolite producers to key regulators of many cellular functions. In order to preserve and protect their functional status, these organelles engage a number of dynamic processes that allow them to decrease accumulated burden and maintain their homeostasis. Indeed, mitochondria can unite (fusion), divide (fission), position themselves strategically in the cell (motility/trafficking) and if irreversibly damaged or dysfunctional eliminated (mitophagy). These dynamic processes can be controlled both by mitochondrial and cellular signalling pathways, hence allowing mitochondria to tune their function to the cellular needs. Among the regulatory mechanisms, reversible phosphorylation downstream the cyclic AMP (cAMP) signalling cascade was shown to deeply influence mitochondrial dynamics. This review explores the emerging evidence suggesting that cAMP is a key player in the orchestration of mitochondrial fusion/fission, motility and mitophagy, extending the repertoire of this second messenger, which is now recognised as a major regulator of mitochondrial homeostasis.
Collapse
Affiliation(s)
- Giulietta Di Benedetto
- Neuroscience Institute, Italian National Research Council (CNR), Venetian Institute of Molecular Medicine, 35131, Padova, Italy
| | - Andrea Gerbino
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Bari, Italy
| | - Konstantinos Lefkimmiatis
- Neuroscience Institute, Italian National Research Council (CNR), Venetian Institute of Molecular Medicine, 35131, Padova, Italy.
| |
Collapse
|
142
|
Hawley ZCE, Campos-Melo D, Droppelmann CA, Strong MJ. MotomiRs: miRNAs in Motor Neuron Function and Disease. Front Mol Neurosci 2017; 10:127. [PMID: 28522960 PMCID: PMC5415563 DOI: 10.3389/fnmol.2017.00127] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 04/18/2017] [Indexed: 12/12/2022] Open
Abstract
MiRNAs are key regulators of the mammalian transcriptome that have been increasingly linked to degenerative diseases of the motor neurons. Although many of the miRNAs currently incriminated as participants in the pathogenesis of these diseases are also important to the normal development and function of motor neurons, at present there is no knowledge of the complete miRNA profile of motor neurons. In this review, we examine the current understanding with respect to miRNAs that are specifically required for motor neuron development, function and viability, and provide evidence that these should be considered as a functional network of miRNAs which we have collectively termed MotomiRs. We will also summarize those MotomiRs currently known to be associated with both amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA), and discuss their potential use as biomarkers.
Collapse
Affiliation(s)
- Zachary C E Hawley
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western UniversityLondon, ON, Canada
| | - Danae Campos-Melo
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western UniversityLondon, ON, Canada
| | - Cristian A Droppelmann
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western UniversityLondon, ON, Canada
| | - Michael J Strong
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western UniversityLondon, ON, Canada.,Department of Pathology, Schulich School of Medicine and Dentistry, Western UniversityLondon, ON, Canada.,Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western UniversityLondon, ON, Canada
| |
Collapse
|
143
|
Lin MY, Cheng XT, Tammineni P, Xie Y, Zhou B, Cai Q, Sheng ZH. Releasing Syntaphilin Removes Stressed Mitochondria from Axons Independent of Mitophagy under Pathophysiological Conditions. Neuron 2017; 94:595-610.e6. [PMID: 28472658 PMCID: PMC5484086 DOI: 10.1016/j.neuron.2017.04.004] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 02/14/2017] [Accepted: 04/03/2017] [Indexed: 01/12/2023]
Abstract
Chronic mitochondrial stress is a central problem associated with neurodegenerative diseases. Early removal of defective mitochondria from axons constitutes a critical step of mitochondrial quality control. Here we investigate axonal mitochondrial response to mild stress in wild-type neurons and chronic mitochondrial defects in Amytrophic Lateral Sclerosis (ALS)- and Alzheimer's disease (AD)-linked neurons. We show that stressed mitochondria are removed from axons triggered by the bulk release of mitochondrial anchoring protein syntaphilin via a new class of mitochondria-derived cargos independent of Parkin, Drp1, and autophagy. Immuno-electron microscopy and super-resolution imaging show the budding of syntaphilin cargos, which then share a ride on late endosomes for transport toward the soma. Releasing syntaphilin is also activated in the early pathological stages of ALS- and AD-linked mutant neurons. Our study provides new mechanistic insights into the maintenance of axonal mitochondrial quality through SNPH-mediated coordination of mitochondrial stress and motility before activation of Parkin-mediated mitophagy. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Mei-Yao Lin
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706, USA
| | - Xiu-Tang Cheng
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706, USA
| | - Prasad Tammineni
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Yuxiang Xie
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706, USA
| | - Bing Zhou
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706, USA
| | - Qian Cai
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Zu-Hang Sheng
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706, USA.
| |
Collapse
|
144
|
Mazel T. Crosstalk of cell polarity signaling pathways. PROTOPLASMA 2017; 254:1241-1258. [PMID: 28293820 DOI: 10.1007/s00709-017-1075-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/02/2017] [Indexed: 06/06/2023]
Abstract
Cell polarity, the asymmetric organization of cellular components along one or multiple axes, is present in most cells. From budding yeast cell polarization induced by pheromone signaling, oocyte polarization at fertilization to polarized epithelia and neuronal cells in multicellular organisms, similar mechanisms are used to determine cell polarity. Crucial role in this process is played by signaling lipid molecules, small Rho family GTPases and Par proteins. All these signaling circuits finally govern the cytoskeleton, which is responsible for oriented cell migration, cell shape changes, and polarized membrane and organelle trafficking. Thus, typically in the process of cell polarization, most cellular constituents become polarized, including plasma membrane lipid composition, ion concentrations, membrane receptors, and proteins in general, mRNA, vesicle trafficking, or intracellular organelles. This review gives a brief overview how these systems talk to each other both during initial symmetry breaking and within the signaling feedback loop mechanisms used to preserve the polarized state.
Collapse
Affiliation(s)
- Tomáš Mazel
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Albertov 4, 128 00, Prague 2, Czech Republic.
- State Institute for Drug Control, Šrobárova 48, 100 41, Prague 10, Czech Republic.
| |
Collapse
|
145
|
Wang Y, Santerre M, Tempera I, Martin K, Mukerjee R, Sawaya BE. HIV-1 Vpr disrupts mitochondria axonal transport and accelerates neuronal aging. Neuropharmacology 2017; 117:364-375. [PMID: 28212984 PMCID: PMC5397298 DOI: 10.1016/j.neuropharm.2017.02.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 02/08/2017] [Accepted: 02/09/2017] [Indexed: 12/24/2022]
Abstract
Disruption of mitochondria axonal transport, essential for the maintenance of synaptic and neuronal integrity and function, has been identified in neurodegenerative diseases. Whether HIV-1 viral proteins affect mitochondria axonal transport is unknown, albeit HIV-associated neurocognitive disorders occur in around half of the patients living with HIV. Therefore, we sought to examine the effect of HIV-1 viral protein R (Vpr) on mitochondria axonal transport. Using mice primary neuronal cultures, we demonstrated that 4-day Vpr treatment reduced the ratio of moving mitochondria associated with (i) less energy (ATP) supply, (ii) reduction in Miro-1 and (iii) increase of α-synuclein which led to loss of microtubule stability as demonstrated by inconsecutive distribution of acetylated α-tubulin along the axons. Interestingly, the effect of Vpr on mitochondria axonal transport was partially restored in the presence of bongkrekic acid, a compound that negatively affected the Vpr-adenine nucleotide translocator (ANT) interaction and totally restored the ATP level in neurons. This indicated Vpr impaired mitochondria axonal transport partially related to its interaction with ANT. The above effect of Vpr was similar to the data obtained from hippocampal tissues isolated from 18-month-old aging mice compared to 5-month-old mice. In accord with previous clinical findings that HIV infection prematurely ages the brain and increases the susceptibility to HAND, we found that Vpr induced aging markers in neurons. Thus, we concluded that instead of causing cell death, low concentration of HIV-1 Vpr altered neuronal function related with inhibition of mitochondria axonal transport which might contribute to the accelerated neuronal aging.
Collapse
Affiliation(s)
- Ying Wang
- Molecular Studies of Neurodegenerative Diseases Lab, United States; Department of Neurology, The Fels Institute for Cancer Research & Molecular Biology, United States.
| | - Maryline Santerre
- Molecular Studies of Neurodegenerative Diseases Lab, United States; Department of Neurology, The Fels Institute for Cancer Research & Molecular Biology, United States
| | - Italo Tempera
- Department of Neurology, The Fels Institute for Cancer Research & Molecular Biology, United States
| | - Kayla Martin
- Department of Neurology, The Fels Institute for Cancer Research & Molecular Biology, United States
| | - Ruma Mukerjee
- Molecular Studies of Neurodegenerative Diseases Lab, United States; Department of Neurology, The Fels Institute for Cancer Research & Molecular Biology, United States
| | - Bassel E Sawaya
- Molecular Studies of Neurodegenerative Diseases Lab, United States; Department of Neurology, The Fels Institute for Cancer Research & Molecular Biology, United States; Temple University School of Medicine, Philadelphia, PA 19140, United States.
| |
Collapse
|
146
|
Kimball EC, Pease ME, Steinhart MR, Oglesby EN, Pitha I, Nguyen C, Quigley HA. A mouse ocular explant model that enables the study of living optic nerve head events after acute and chronic intraocular pressure elevation: Focusing on retinal ganglion cell axons and mitochondria. Exp Eye Res 2017; 160:106-115. [PMID: 28414059 DOI: 10.1016/j.exer.2017.04.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 01/03/2017] [Accepted: 04/09/2017] [Indexed: 11/16/2022]
Abstract
We developed an explant model of the mouse eye and optic nerve that facilitates the study of retinal ganglion cell axons and mitochondria in the living optic nerve head (ONH) in an ex vivo environment. Two transgenic mouse strains were used, one expressing yellow fluorescent protein in selected axons and a second strain expressing cyan fluorescent protein in all mitochondria. We viewed an explanted mouse eye and optic nerve by laser scanning microscopy at and behind the ONH, the site of glaucoma injury. Explants from previously untreated mice were studied with the intraocular pressure (IOP) set artificially at normal or elevated levels for several hours. Explants were also studied from eyes that had undergone chronic IOP elevation from 14 h to 6 weeks prior to ex vivo study. Image analysis in static images and video of individual mitochondria or axonal structure determined effects of acute and chronic IOP elevation. At normal IOP, fluorescent axonal structure was stable for up to 3 h under ex vivo conditions. After chronic IOP elevation, axonal integrity index values indicated fragmentation of axon structure in the ONH. In mice with fluorescent mitochondria, the normal density decreased with distance behind the ONH by 45% (p = 0.002, t-test). Density increased with prior chronic IOP elevation to 21,300 ± 4176 mitochondria/mm2 compared to control 16,110 ± 3159 mitochondria/mm2 (p = 0.025, t-test), but did not increase significantly after 4 h, acute IOP elevation (1.5% decrease in density, p = 0.83, t-test). Mean normal mitochondrial length of 2.3 ± 1.4 μm became 13% smaller after 4 h of IOP elevation ex vivo compared to baseline (p = 0.015, t-test, N-10). Normal mitochondrial speed of movement was significantly slower in the anterograde direction (towards the brain) than retrograde, but there were more mitochondria in motion and traveling longer lengths in anterograde direction. The percent of mitochondria in motion decreased by >50% with acute IOP increase to 30 mm Hg after 60 min. A new ocular explant model implemented with eyes from transgenic mice with fluorescent cellular components provided real time measurement of the early events in experimental glaucoma and quantitative outcomes for neuroprotection therapy experiments.
Collapse
Affiliation(s)
- Elizabeth C Kimball
- From the Glaucoma Center of Excellence, Wilmer Ophthalmological Institute, Johns Hopkins University, Baltimore, MD, USA.
| | - Mary E Pease
- From the Glaucoma Center of Excellence, Wilmer Ophthalmological Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Matthew R Steinhart
- From the Glaucoma Center of Excellence, Wilmer Ophthalmological Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Ericka N Oglesby
- From the Glaucoma Center of Excellence, Wilmer Ophthalmological Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Ian Pitha
- From the Glaucoma Center of Excellence, Wilmer Ophthalmological Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Cathy Nguyen
- From the Glaucoma Center of Excellence, Wilmer Ophthalmological Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Harry A Quigley
- From the Glaucoma Center of Excellence, Wilmer Ophthalmological Institute, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
147
|
Regulation of Synaptic Amyloid-β Generation through BACE1 Retrograde Transport in a Mouse Model of Alzheimer's Disease. J Neurosci 2017; 37:2639-2655. [PMID: 28159908 DOI: 10.1523/jneurosci.2851-16.2017] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Revised: 01/02/2017] [Accepted: 01/09/2017] [Indexed: 12/20/2022] Open
Abstract
Amyloid-β (Aβ) peptides play a key role in synaptic damage and memory deficits in the early pathogenesis of Alzheimer's disease (AD). Abnormal accumulation of Aβ at nerve terminals leads to synaptic pathology and ultimately to neurodegeneration. β-site amyloid precursor protein (APP) cleaving enzyme 1 (BACE1) is the major neuronal β-secretase for Aβ generation. However, the mechanisms regulating BACE1 distribution in axons and β cleavage of APP at synapses remain largely unknown. Here, we reveal that dynein-Snapin-mediated retrograde transport regulates BACE1 trafficking in axons and APP processing at presynaptic terminals. BACE1 is predominantly accumulated within late endosomes at the synapses of AD-related mutant human APP (hAPP) transgenic (Tg) mice and patient brains. Defective retrograde transport by genetic ablation of snapin in mice recapitulates late endocytic retention of BACE1 and increased APP processing at presynaptic sites. Conversely, overexpressing Snapin facilitates BACE1 trafficking and reduces synaptic BACE1 accumulation by enhancing the removal of BACE1 from distal AD axons and presynaptic terminals. Moreover, elevated Snapin expression via stereotactic hippocampal injections of adeno-associated virus particles in mutant hAPP Tg mouse brains decreases synaptic Aβ levels and ameliorates synapse loss, thus rescuing cognitive impairments associated with hAPP mice. Altogether, our study provides new mechanistic insights into the complex regulation of BACE1 trafficking and presynaptic localization through Snapin-mediated dynein-driven retrograde axonal transport, thereby suggesting a potential approach of modulating Aβ levels and attenuating synaptic deficits in AD.SIGNIFICANCE STATEMENT β-Site amyloid precursor protein (APP) cleaving enzyme 1 (BACE1) trafficking and synaptic localization significantly influence its β secretase activity and amyloid-β (Aβ) production. In AD brains, BACE1 is accumulated within dystrophic neurites, which is thought to augment Aβ-induced synaptotoxicity by Aβ overproduction. However, it remains largely unknown whether axonal transport regulates synaptic APP processing. Here, we demonstrate that Snapin-mediated retrograde transport plays a critical role in removing BACE1 from presynaptic terminals toward the soma, thus reducing synaptic Aβ production. Adeno-associated virus-mediated Snapin overexpression in the hippocampus of mutant hAPP mice significantly decreases synaptic Aβ levels, attenuates synapse loss, and thus rescues cognitive deficits. Our study uncovers a new pathway that controls synaptic APP processing by enhancing axonal BACE1 trafficking, thereby advancing our fundamental knowledge critical for ameliorating Aβ-linked synaptic pathology.
Collapse
|
148
|
Xu Y, Chen M, Hu B, Huang R, Hu B. In vivo Imaging of Mitochondrial Transport in Single-Axon Regeneration of Zebrafish Mauthner Cells. Front Cell Neurosci 2017; 11:4. [PMID: 28174522 PMCID: PMC5258718 DOI: 10.3389/fncel.2017.00004] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 01/09/2017] [Indexed: 12/17/2022] Open
Abstract
Mitochondrial transport is essential for neuronal function, but the evidence of connections between mitochondrial transport and axon regeneration in the central nervous system (CNS) of living vertebrates remains limited. Here, we developed a novel model to explore mitochondrial transport in a single Mauthner axon (M axon) of zebrafish with non-invasive in vivo imaging. To confirm the feasibility of using this model, we treated labeled zebrafish with nocodazole and demonstrated that it could disrupt mitochondrial transport. We also used two-photon laser axotomy to precisely axotomize M axons and simultaneously recorded their regeneration and the process of mitochondrial transport in living zebrafish larvae. The findings showed that the injured axons with stronger regenerative capability maintain greater mitochondrial motility. Furthermore, to stimulate axon regeneration, treatment with dibutyryl cyclic adenosine monophosphate (db-cAMP) could also augment mitochondrial motility. Taken together, our results provide new evidence that mitochondrial motility is positively correlated with axon regeneration in the living vertebrate CNS. This promising model will be useful for further studies on the interaction between axon regeneration and mitochondrial dynamics, using various genetic and pharmacological techniques.
Collapse
Affiliation(s)
- Yang Xu
- Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, and School of Life Sciences, University of Science and Technology of China Hefei, China
| | - Min Chen
- Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, and School of Life Sciences, University of Science and Technology of China Hefei, China
| | - Bingbing Hu
- Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, and School of Life Sciences, University of Science and Technology of China Hefei, China
| | - Rongchen Huang
- Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, and School of Life Sciences, University of Science and Technology of China Hefei, China
| | - Bing Hu
- Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, and School of Life Sciences, University of Science and Technology of China Hefei, China
| |
Collapse
|
149
|
Tammineni P, Ye X, Feng T, Aikal D, Cai Q. Impaired retrograde transport of axonal autophagosomes contributes to autophagic stress in Alzheimer's disease neurons. eLife 2017; 6. [PMID: 28085665 PMCID: PMC5235353 DOI: 10.7554/elife.21776] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 12/22/2016] [Indexed: 12/20/2022] Open
Abstract
Neurons face unique challenges of transporting nascent autophagic vacuoles (AVs) from distal axons toward the soma, where mature lysosomes are mainly located. Autophagy defects have been linked to Alzheimer’s disease (AD). However, the mechanisms underlying altered autophagy remain unknown. Here, we demonstrate that defective retrograde transport contributes to autophagic stress in AD axons. Amphisomes predominantly accumulate at axonal terminals of mutant hAPP mice and AD patient brains. Amyloid-β (Aβ) oligomers associate with AVs in AD axons and interact with dynein motors. This interaction impairs dynein recruitment to amphisomes through competitive interruption of dynein-Snapin motor-adaptor coupling, thus immobilizing them in distal axons. Consistently, deletion of Snapin in mice causes AD-like axonal autophagic stress, whereas overexpressing Snapin in hAPP neurons reduces autophagic accumulation at presynaptic terminals by enhancing AV retrograde transport. Altogether, our study provides new mechanistic insight into AD-associated autophagic stress, thus establishing a foundation for ameliorating axonal pathology in AD. DOI:http://dx.doi.org/10.7554/eLife.21776.001 Alzheimer’s disease is the result of protein fragments called amyloid-β peptides accumulating in the brain and forming clumps. These protein “aggregates” disrupt cellular activities and cause serious problems. To combat this process, healthy cells use a process called autophagy to destroy aggregated proteins. The aggregates are first loaded into structures called autophagosomes that then fuse with cell compartments called lysosomes, which contain enzymes that can break down the proteins. Brain cells called neurons have an unusual shape with branch-like structures and a long projection called an axon that all form off the main cell body. Autophagosomes predominantly form in the axons and need to move toward the cell body where the lysosomes are found. A motor protein called dynein drives the movement of autophagosomes by interacting with an adaptor protein known as Snapin on the surface of these structures. Autophagosomes tend to accumulate within the neurons of individuals with Alzheimer’s disease, but it is not known why. Cai et al. examined the ability of autophagosomes to move to the cell body of neurons from a mouse model of Alzheimer’s disease in which human amyloid-β peptides accumulate in the brain, and in the brains of human patients with Alzheimer’s disease. The experiments show that autophagosomes predominantly accumulate in the axons and at the ends of axons during Alzheimer’s disease. Amyloid-β aggregates associate with autophagosomes in the axons and interact with dynein motors. This disrupts the interaction between dynein and Snapin and impairs dynein binding to the autophagosomes, trapping the autophagosomes in the axons. Increasing the production of Snapin proteins inside the mouse neurons enhances dynein binding to autophagosomes and thus helps these structures move to the cell body. The next step is to investigate whether increasing the ability of autophagosomes to move to the cell body reduces the symptoms of Alzheimer’s disease in the mutant mice. This will help to build a foundation for the future development of new strategies to treat Alzheimer’s disease and other neurodegenerative disorders that are caused by protein aggregates. DOI:http://dx.doi.org/10.7554/eLife.21776.002
Collapse
Affiliation(s)
- Prasad Tammineni
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, United States
| | - Xuan Ye
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, United States
| | - Tuancheng Feng
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, United States
| | - Daniyal Aikal
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, United States
| | - Qian Cai
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, United States
| |
Collapse
|
150
|
Abstract
Although tctp expression in many areas of the human brain was reported more than 15 years ago, little was known about how it functions in neurons. The early notion that Tctp is primarily expressed in mitotic cells, together with reports suggesting a relative low abundance in the brain, has perhaps potentiated this almost complete disregard for the study of Tctp in the context of neuron biology. However, recent evidence has challenged this view, as a number of independent genome-wide profiling studies identified tctp mRNA among the most enriched in the axonal compartment across diverse neuronal populations, including embryonic retinal ganglion cells. Considering the emerging parallels between axon guidance and cancer cell invasion, the axonal expression of cancer-associated tctp was suggestive of it holding an unexplored role in the wiring of neuronal circuits. Our study revealed that Tctp is necessary for the accurate and timely development of axon projections during the formation of vertebrate retinal circuits via its association with the survival machinery of the axon. Globally, the findings indicate that compromised pro-survival signaling in Tctp-deficient axons results in mitochondrial dysfunction and a subsequent decrease in axonal mitochondrial density. These effects likely translate into a metabolic state inadequate to support the normal guidance and extension processes of a developing axon.
Collapse
Affiliation(s)
- Cláudio Gouveia Roque
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA.
| | - Christine E Holt
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, England, UK
| |
Collapse
|