1
|
Alex Thomas M, Cui X, Artinian LR, Cao Q, Jing J, Silva FC, Wang S, Zigman JM, Sun Y, Shi H, Xue B. Crosstalk between Gut Sensory Ghrelin Signaling and Adipose Tissue Sympathetic Outflow Regulates Metabolic Homeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.25.568689. [PMID: 38076894 PMCID: PMC10705268 DOI: 10.1101/2023.11.25.568689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
The stomach-derived orexigenic hormone ghrelin is a key regulator of energy homeostasis and metabolism in humans. The ghrelin receptor, growth hormone secretagogue receptor 1a (GHSR), is widely expressed in the brain and gastrointestinal vagal sensory neurons, and neuronal GHSR knockout results in a profoundly beneficial metabolic profile and protects against diet-induced obesity (DIO) and insulin resistance. Here we show that in addition to the well characterized vagal GHSR, GHSR is robustly expressed in gastrointestinal sensory neurons emanating from spinal dorsal root ganglia. Remarkably, sensory neuron GHSR deletion attenuates DIO through increased energy expenditure and sympathetic outflow to adipose tissue independent of food intake. In addition, neuronal viral tract tracing reveals prominent crosstalk between gut non-vagal sensory afferents and adipose sympathetic outflow. Hence, these findings demonstrate a novel gut sensory ghrelin signaling pathway critical for maintaining energy homeostasis.
Collapse
Affiliation(s)
- M. Alex Thomas
- Department of Biology, Georgia State University, Atlanta, GA
| | - Xin Cui
- Department of Biology, Georgia State University, Atlanta, GA
| | | | - Qiang Cao
- Department of Biology, Georgia State University, Atlanta, GA
| | - Jia Jing
- Department of Biology, Georgia State University, Atlanta, GA
| | - Felipe C. Silva
- Department of Biology, Georgia State University, Atlanta, GA
| | - Shirong Wang
- Department of Biology, Georgia State University, Atlanta, GA
| | - Jeffrey M. Zigman
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| | - Yuxiang Sun
- Department of Nutrition, Texas A & M University, College Station, TX
| | - Hang Shi
- Department of Biology, Georgia State University, Atlanta, GA
| | - Bingzhong Xue
- Department of Biology, Georgia State University, Atlanta, GA
| |
Collapse
|
2
|
Ladagu AD, Olopade FE, Chazot P, Oyagbemi AA, Ohiomokhare S, Folarin OR, Gilbert TT, Fuller M, Luong T, Adejare A, Olopade JO. Attenuation of Vanadium-Induced Neurotoxicity in Rat Hippocampal Slices (In Vitro) and Mice (In Vivo) by ZA-II-05, a Novel NMDA-Receptor Antagonist. Int J Mol Sci 2023; 24:16710. [PMID: 38069032 PMCID: PMC10706475 DOI: 10.3390/ijms242316710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/04/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Exposure to heavy metals, such as vanadium, poses an ongoing environmental and health threat, heightening the risk of neurodegenerative disorders. While several compounds have shown promise in mitigating vanadium toxicity, their efficacy is limited. Effective strategies involve targeting specific subunits of the NMDA receptor, a glutamate receptor linked to neurodegenerative conditions. The potential neuroprotective effects of ZA-II-05, an NMDA receptor antagonist, against vanadium-induced neurotoxicity were explored in this study. Organotypic rat hippocampal slices, and live mice, were used as models to comprehensively evaluate the compound's impact. Targeted in vivo fluorescence analyses of the hippocampal slices using propidium iodide as a marker for cell death was utilized. The in vivo study involved five dams, each with eight pups, which were randomly assigned to five experimental groups (n = 8 pups). After administering treatments intraperitoneally over six months, various brain regions were assessed for neuropathologies using different immunohistochemical markers. High fluorescence intensity was observed in the hippocampal slices treated with vanadium, signifying cell death. Vanadium-exposed mice exhibited demyelination, microgliosis, and neuronal cell loss. Significantly, treatment with ZA-II-05 resulted in reduced cellular death in the rat hippocampal slices and preserved cellular integrity and morphological architecture in different anatomical regions, suggesting its potential in countering vanadium-induced neurotoxicity.
Collapse
Affiliation(s)
- Amany Digal Ladagu
- Department of Veterinary Anatomy, University of Ibadan, Ibadan 200284, Nigeria; (A.D.L.); (O.R.F.); (T.T.G.); (J.O.O.)
| | | | - Paul Chazot
- Department of Biosciences, Durham University, County Durham DH1 3LE, UK;
| | - Ademola A. Oyagbemi
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan 200284, Nigeria;
| | - Samuel Ohiomokhare
- Department of Biosciences, Durham University, County Durham DH1 3LE, UK;
| | - Oluwabusayo Racheal Folarin
- Department of Veterinary Anatomy, University of Ibadan, Ibadan 200284, Nigeria; (A.D.L.); (O.R.F.); (T.T.G.); (J.O.O.)
| | - Taidinda Tashara Gilbert
- Department of Veterinary Anatomy, University of Ibadan, Ibadan 200284, Nigeria; (A.D.L.); (O.R.F.); (T.T.G.); (J.O.O.)
| | - Madison Fuller
- Department of Neuroscience, College of Arts and Sciences, Saint Joseph’s University, Philadelphia, PA 19131, USA; (M.F.); (T.L.)
| | - Toan Luong
- Department of Neuroscience, College of Arts and Sciences, Saint Joseph’s University, Philadelphia, PA 19131, USA; (M.F.); (T.L.)
| | - Adeboye Adejare
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, Saint Joseph’s University, Philadelphia, PA 19131, USA;
| | - James O. Olopade
- Department of Veterinary Anatomy, University of Ibadan, Ibadan 200284, Nigeria; (A.D.L.); (O.R.F.); (T.T.G.); (J.O.O.)
| |
Collapse
|
3
|
Ma X, Miraucourt LS, Qiu H, Sharif-Naeini R, Khadra A. Modulation of SK Channels via Calcium Buffering Tunes Intrinsic Excitability of Parvalbumin Interneurons in Neuropathic Pain: A Computational and Experimental Investigation. J Neurosci 2023; 43:5608-5622. [PMID: 37451982 PMCID: PMC10401647 DOI: 10.1523/jneurosci.0426-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/11/2023] [Accepted: 05/18/2023] [Indexed: 07/18/2023] Open
Abstract
Parvalbumin-expressing interneurons (PVINs) play a crucial role within the dorsal horn of the spinal cord by preventing touch inputs from activating pain circuits. In both male and female mice, nerve injury decreases PVINs' output via mechanisms that are not fully understood. In this study, we show that PVINs from nerve-injured male mice change their firing pattern from tonic to adaptive. To examine the ionic mechanisms responsible for this decreased output, we used a reparametrized Hodgkin-Huxley type model of PVINs, which predicted (1) the firing pattern transition is because of an increased contribution of small conductance calcium-activated potassium (SK) channels, enabled by (2) impairment in intracellular calcium buffering systems. Analyzing the dynamics of the Hodgkin-Huxley type model further demonstrated that a generalized Hopf bifurcation differentiates the two types of state transitions observed in the transient firing of PVINs. Importantly, this predicted mechanism holds true when we embed the PVIN model within the neuronal circuit model of the spinal dorsal horn. To experimentally validate this hypothesized mechanism, we used pharmacological modulators of SK channels and demonstrated that (1) tonic firing PVINs from naive male mice become adaptive when exposed to an SK channel activator, and (2) adapting PVINs from nerve-injured male mice return to tonic firing on SK channel blockade. Our work provides important insights into the cellular mechanism underlying the decreased output of PVINs in the spinal dorsal horn after nerve injury and highlights potential pharmacological targets for new and effective treatment approaches to neuropathic pain.SIGNIFICANCE STATEMENT Parvalbumin-expressing interneurons (PVINs) exert crucial inhibitory control over Aβ fiber-mediated nociceptive pathways at the spinal dorsal horn. The loss of their inhibitory tone leads to neuropathic symptoms, such as mechanical allodynia, via mechanisms that are not fully understood. This study identifies the reduced intrinsic excitability of PVINs as a potential cause for their decreased inhibitory output in nerve-injured condition. Combining computational and experimental approaches, we predict a calcium-dependent mechanism that modulates PVINs' electrical activity following nerve injury: a depletion of cytosolic calcium buffer allows for the rapid accumulation of intracellular calcium through the active membranes, which in turn potentiates SK channels and impedes spike generation. Our results therefore pinpoint SK channels as potential therapeutic targets for treating neuropathic symptoms.
Collapse
Affiliation(s)
- Xinyue Ma
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec H3G 1Y6, Canada
- Alan Edwards Center for Research on Pain, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Loïs S Miraucourt
- Alan Edwards Center for Research on Pain, McGill University, Montreal, Quebec H3G 1Y6, Canada
- Department of Physiology, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Haoyi Qiu
- Alan Edwards Center for Research on Pain, McGill University, Montreal, Quebec H3G 1Y6, Canada
- Department of Physiology, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Reza Sharif-Naeini
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec H3G 1Y6, Canada
- Alan Edwards Center for Research on Pain, McGill University, Montreal, Quebec H3G 1Y6, Canada
- Department of Physiology, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Anmar Khadra
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec H3G 1Y6, Canada
- Department of Physiology, McGill University, Montreal, Quebec H3G 1Y6, Canada
- Department of Quantitative Life Sciences, McGill University, Montreal, Quebec H3G 1Y6, Canada
| |
Collapse
|
4
|
Role of Intracellular Na + in the Regulation of [Ca 2+] i in the Rat Suprachiasmatic Nucleus Neurons. Int J Mol Sci 2019; 20:ijms20194868. [PMID: 31575032 PMCID: PMC6801571 DOI: 10.3390/ijms20194868] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 09/28/2019] [Indexed: 12/17/2022] Open
Abstract
Transmembrane Ca2+ influx is essential to the proper functioning of the central clock in the suprachiasmatic nucleus (SCN). In the rat SCN neurons, the clearance of somatic Ca2+ following depolarization-induced Ca2+ transients involves Ca2+ extrusion via Na+/Ca2+ exchanger (NCX) and mitochondrial Ca2+ buffering. Here we show an important role of intracellular Na+ in the regulation of [Ca2+]i in these neurons. The effect of Na+ loading on [Ca2+]i was determined with the Na+ ionophore monensin and the cardiac glycoside ouabain to block Na+/K+-ATPase (NKA). Ratiometric Na+ and Ca2+ imaging was used to measure the change in [Na+]i and [Ca2+]i, and cell-attached recordings to investigate the effects of monensin and ouabain on spontaneous firing. Our results show that in spite of opposite effects on spontaneous firing and basal [Ca2+], both monensin and ouabain induced Na+ loading, and increased the peak amplitude, slowed the fast decay rate, and enhanced the slow decay phase of 20 mM K+-evoked Ca2+ transients. Furthermore, both ouabain and monensin preferentially enhanced nimodipine-insensitive Ca2+ transients. Together, our results indicate that in the SCN neurons the NKA plays an important role in regulating [Ca2+]i, in particular, associated with nimodipine-insensitive Ca2+ channels.
Collapse
|
5
|
Pioglitazone, a PPARγ agonist, reduces cisplatin-evoked neuropathic pain by protecting against oxidative stress. Pain 2019; 160:688-701. [PMID: 30507781 DOI: 10.1097/j.pain.0000000000001448] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Painful peripheral neuropathy is a dose-limiting side effect of cisplatin treatment. Using a murine model of cisplatin-induced hyperalgesia, we determined whether a PPARγ synthetic agonist, pioglitazone, attenuated the development of neuropathic pain and identified underlying mechanisms. Cisplatin produced mechanical and cold hyperalgesia and decreased electrical thresholds of Aδ and C fibers, which were attenuated by coadministration of pioglitazone (10 mg/kg, intraperitoneally [i.p.]) with cisplatin. Antihyperalgesic effects of pioglitazone were blocked by the PPARγ antagonist T0070907 (10 mg/kg, i.p.). We hypothesized that the ability of pioglitazone to reduce the accumulation of reactive oxygen species (ROS) in dorsal root ganglion (DRG) neurons contributed to its antihyperalgesic activity. Effects of cisplatin and pioglitazone on somatosensory neurons were studied on dissociated mouse DRG neurons after 24 hours in vitro. Incubation of DRG neurons with cisplatin (13 µM) for 24 hours increased the occurrence of depolarization-evoked calcium transients, and these were normalized by coincubation with pioglitazone (10 µM). Oxidative stress in DRG neurons was considered a significant contributor to cisplatin-evoked hyperalgesia because a ROS scavenger attenuated hyperalgesia and normalized the evoked calcium responses when cotreated with cisplatin. Pioglitazone increased the expression and activity of ROS-reducing enzymes in DRG and normalized cisplatin-evoked changes in oxidative stress and labeling of mitochondria with the dye MitoTracker Deep Red, indicating that the antihyperalgesic effects of pioglitazone were attributed to its antioxidant properties in DRG neurons. These data demonstrate clear benefits of broadening the use of the antidiabetic drug pioglitazone, or other PPARγ agonists, to minimize the development of cisplatin-induced painful neuropathy.
Collapse
|
6
|
Pan S, Conaway S, Deshpande DA. Mitochondrial regulation of airway smooth muscle functions in health and pulmonary diseases. Arch Biochem Biophys 2019; 663:109-119. [PMID: 30629957 DOI: 10.1016/j.abb.2019.01.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/28/2018] [Accepted: 01/04/2019] [Indexed: 12/24/2022]
Abstract
Mitochondria are important for airway smooth muscle physiology due to their diverse yet interconnected roles in calcium handling, redox regulation, and cellular bioenergetics. Increasing evidence indicates that mitochondria dysfunction is intimately associated with airway diseases such as asthma, IPF and COPD. In these pathological conditions, increased mitochondrial ROS, altered bioenergetics profiles, and calcium mishandling contribute collectively to changes in cellular signaling, gene expression, and ultimately changes in airway smooth muscle contractile/proliferative properties. Therefore, understanding the basic features of airway smooth muscle mitochondria and their functional contribution to airway biology and pathology are key to developing novel therapeutics for airway diseases. This review summarizes the recent findings of airway smooth muscle mitochondria focusing on calcium homeostasis and redox regulation, two key determinants of physiological and pathological functions of airway smooth muscle.
Collapse
Affiliation(s)
- Shi Pan
- Center for Translational Medicine, Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Stanley Conaway
- Center for Translational Medicine, Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Deepak A Deshpande
- Center for Translational Medicine, Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA.
| |
Collapse
|
7
|
Intrathecal Injection of miR-133b-3p or miR-143-3p Prevents the Development of Persistent Cold and Mechanical Allodynia Following a Peripheral Nerve Injury in Rats. Neuroscience 2018; 386:223-239. [PMID: 30018017 DOI: 10.1016/j.neuroscience.2018.06.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/01/2018] [Accepted: 06/24/2018] [Indexed: 12/24/2022]
Abstract
In DRG an increase in miR-133b-3p, miR-143-3p, and miR-1-3p correlates with the lack of development of neuropathic pain following a peripheral nerve injury. Using lentiviral (LV) vectors we found that a single injection of LV-miR-133b-3p or LV-miR-143-3p immediately after a peripheral nerve injury prevented the development of sustained mechanical and cold allodynia. Injection of LV-miR-133b-3p or LV-miR-143-3p by themselves or in combination, on day 3 post-injury produced a partial and transient reduction in mechanical allodynia and a sustained decrease in cold allodynia. Injection of LV-miR-1-3p has no effect. Co-injection of LV-miR-1a with miR-133b-3p or miR-143-3p on day 3 post-injury produced a sustained decrease in mechanical and cold allodynia. In DRG cultures, miR-133b-3p and miR-143-3p but not miR-1-3p, enhanced the depolarization-evoked cytoplasmic calcium increase. Using 3'UTR target clones containing a Gaussian luciferase reporter gene we found that with the 3'UTR-Scn2b, miR-133-3p and miR-143-3p reduced the expression while miR-1-3p enhanced the expression of the reporter gene. With the 3'UTR-TRPM8, miR-133-3p and miR-143-3p reduced the expression and miR-1-3p had no effect. With the 3'UTR-Piezo2, miR-133-3p increased the expression while miR-143-3p and miR-1-3p had no effect. LV-miR133b-3p, LV-miR-143-3p and LV-miR1a-3p reduced Scn2b-mRNA and Piezo2-mRNA. LV-miR133b-3p and LV-miR-143-3p reduced TRPM8-mRNA. LV-miR-133b-3p and LV-miR-143-3p prevent the development of chronic pain when injected immediately after the injury, but are only partially effective when injected at later times. LV-miR-1a-3p had no effect on pain, but complemented the actions of LV-miR-133b-3p or LV-miR-143-3p resulting in a sustained reversal of pain when co-injected 3 days following nerve injury.
Collapse
|
8
|
Mandge D, Manchanda R. A biophysically detailed computational model of urinary bladder small DRG neuron soma. PLoS Comput Biol 2018; 14:e1006293. [PMID: 30020934 PMCID: PMC6066259 DOI: 10.1371/journal.pcbi.1006293] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 07/30/2018] [Accepted: 06/11/2018] [Indexed: 12/13/2022] Open
Abstract
Bladder small DRG neurons, which are putative nociceptors pivotal to urinary bladder function, express more than a dozen different ionic membrane mechanisms: ion channels, pumps and exchangers. Small-conductance Ca2+-activated K+ (SKCa) channels which were earlier thought to be gated solely by intracellular Ca2+ concentration ([Ca]i) have recently been shown to exhibit inward rectification with respect to membrane potential. The effect of SKCa inward rectification on the excitability of these neurons is unknown. Furthermore, studies on the role of KCa channels in repetitive firing and their contributions to different types of afterhyperpolarization (AHP) in these neurons are lacking. In order to study these phenomena, we first constructed and validated a biophysically detailed single compartment model of bladder small DRG neuron soma constrained by physiological data. The model includes twenty-two major known membrane mechanisms along with intracellular Ca2+ dynamics comprising Ca2+ diffusion, cytoplasmic buffering, and endoplasmic reticulum (ER) and mitochondrial mechanisms. Using modelling studies, we show that inward rectification of SKCa is an important parameter regulating neuronal repetitive firing and that its absence reduces action potential (AP) firing frequency. We also show that SKCa is more potent in reducing AP spiking than the large-conductance KCa channel (BKCa) in these neurons. Moreover, BKCa was found to contribute to the fast AHP (fAHP) and SKCa to the medium-duration (mAHP) and slow AHP (sAHP). We also report that the slow inactivating A-type K+ channel (slow KA) current in these neurons is composed of 2 components: an initial fast inactivating (time constant ∼ 25-100 ms) and a slow inactivating (time constant ∼ 200-800 ms) current. We discuss the implications of our findings, and how our detailed model can help further our understanding of the role of C-fibre afferents in the physiology of urinary bladder as well as in certain disorders.
Collapse
Affiliation(s)
- Darshan Mandge
- Computational Neurophysiology Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India 400076
| | - Rohit Manchanda
- Computational Neurophysiology Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India 400076
| |
Collapse
|
9
|
Hirabayashi Y, Kwon SK, Paek H, Pernice WM, Paul MA, Lee J, Erfani P, Raczkowski A, Petrey DS, Pon LA, Polleux F. ER-mitochondria tethering by PDZD8 regulates Ca 2+ dynamics in mammalian neurons. Science 2018; 358:623-630. [PMID: 29097544 DOI: 10.1126/science.aan6009] [Citation(s) in RCA: 317] [Impact Index Per Article: 52.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 08/21/2017] [Accepted: 09/20/2017] [Indexed: 01/06/2023]
Abstract
Interfaces between organelles are emerging as critical platforms for many biological responses in eukaryotic cells. In yeast, the ERMES complex is an endoplasmic reticulum (ER)-mitochondria tether composed of four proteins, three of which contain a SMP (synaptotagmin-like mitochondrial-lipid binding protein) domain. No functional ortholog for any ERMES protein has been identified in metazoans. Here, we identified PDZD8 as an ER protein present at ER-mitochondria contacts. The SMP domain of PDZD8 is functionally orthologous to the SMP domain found in yeast Mmm1. PDZD8 was necessary for the formation of ER-mitochondria contacts in mammalian cells. In neurons, PDZD8 was required for calcium ion (Ca2+) uptake by mitochondria after synaptically induced Ca2+-release from ER and thereby regulated cytoplasmic Ca2+ dynamics. Thus, PDZD8 represents a critical ER-mitochondria tethering protein in metazoans. We suggest that ER-mitochondria coupling is involved in the regulation of dendritic Ca2+ dynamics in mammalian neurons.
Collapse
Affiliation(s)
- Yusuke Hirabayashi
- Department of Neuroscience, Columbia University Medical Center, Columbia University, New York, NY 10027, USA.,Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA.,Kavli Institute for Brain Science, Columbia University, New York, NY 10027, USA.,Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan
| | - Seok-Kyu Kwon
- Department of Neuroscience, Columbia University Medical Center, Columbia University, New York, NY 10027, USA.,Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA.,Kavli Institute for Brain Science, Columbia University, New York, NY 10027, USA
| | - Hunki Paek
- Department of Neuroscience, Columbia University Medical Center, Columbia University, New York, NY 10027, USA.,Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA.,Kavli Institute for Brain Science, Columbia University, New York, NY 10027, USA
| | - Wolfgang M Pernice
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Maëla A Paul
- Department of Neuroscience, Columbia University Medical Center, Columbia University, New York, NY 10027, USA.,Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA.,Kavli Institute for Brain Science, Columbia University, New York, NY 10027, USA
| | - Jinoh Lee
- Department of Neuroscience, Columbia University Medical Center, Columbia University, New York, NY 10027, USA.,Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA.,Kavli Institute for Brain Science, Columbia University, New York, NY 10027, USA
| | - Parsa Erfani
- Department of Neuroscience, Columbia University Medical Center, Columbia University, New York, NY 10027, USA.,Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA.,Kavli Institute for Brain Science, Columbia University, New York, NY 10027, USA
| | - Ashleigh Raczkowski
- Simons Electron Microscopy Center, New York Structural Biology Center (NYSBC), New York, NY 10027, USA
| | - Donald S Petrey
- Center for Computational Biology and Bioinformatics, Department of Systems Biology, Columbia University, New York, NY 10032, USA.,Howard Hughes Medical Institute, Columbia University, New York, NY 10032, USA
| | - Liza A Pon
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA.,Institute of Human Nutrition, Columbia University, New York, NY 10032, USA
| | - Franck Polleux
- Department of Neuroscience, Columbia University Medical Center, Columbia University, New York, NY 10027, USA. .,Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA.,Kavli Institute for Brain Science, Columbia University, New York, NY 10027, USA
| |
Collapse
|
10
|
Tomer D, Chippalkatti R, Mitra K, Rikhy R. ERK regulates mitochondrial membrane potential in fission deficient Drosophila follicle cells during differentiation. Dev Biol 2018; 434:48-62. [DOI: 10.1016/j.ydbio.2017.11.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 11/04/2017] [Accepted: 11/15/2017] [Indexed: 01/10/2023]
|
11
|
Bustos G, Cruz P, Lovy A, Cárdenas C. Endoplasmic Reticulum-Mitochondria Calcium Communication and the Regulation of Mitochondrial Metabolism in Cancer: A Novel Potential Target. Front Oncol 2017; 7:199. [PMID: 28944215 PMCID: PMC5596064 DOI: 10.3389/fonc.2017.00199] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 08/18/2017] [Indexed: 01/15/2023] Open
Abstract
Cancer is characterized by an uncontrolled cell proliferation rate even under low nutrient availability, which is sustained by a metabolic reprograming now recognized as a hallmark of cancer. Warburg was the first to establish the relationship between cancer and mitochondria; however, he interpreted enhanced aerobic glycolysis as mitochondrial dysfunction. Today it is accepted that many cancer cell types need fully functional mitochondria to maintain their homeostasis. Calcium (Ca2+)—a key regulator of several cellular processes—has proven to be essential for mitochondrial metabolism. Inositol 1,4,5-trisphosphate receptor (IP3R)-mediated Ca2+ transfer from the endoplasmic reticulum to the mitochondria through the mitochondrial calcium uniporter (MCU) proves to be essential for the maintenance of mitochondrial function and cellular energy balance. Both IP3R and MCU are overexpressed in several cancer cell types, and the inhibition of the Ca2+ communication between these two organelles causes proliferation arrest, migration decrease, and cell death through mechanisms that are not fully understood. In this review, we summarize and analyze the current findings in this area, emphasizing the critical role of Ca2+ and mitochondrial metabolism in cancer and its potential as a novel therapeutic target.
Collapse
Affiliation(s)
- Galdo Bustos
- Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile.,Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Pablo Cruz
- Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile.,Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Alenka Lovy
- Department of Neuroscience, Center for Neuroscience Research, Tufts University School of Medicine, Boston, MA, United States
| | - César Cárdenas
- Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile.,Geroscience Center for Brain Health and Metabolism, Santiago, Chile.,Buck Institute for Research on Aging, Novato, CA, United States.,Department of Chemistry and Biochemistry, University of California, Santa Barbara, Santa Barbara, CA, United States
| |
Collapse
|
12
|
Chen J, Wang J, Zhang J, Pu C. 3- n-Butylphthalide reduces the oxidative damage of muscles in an experimental autoimmune myositis animal model. Exp Ther Med 2017; 14:2085-2093. [PMID: 28962128 PMCID: PMC5609169 DOI: 10.3892/etm.2017.4766] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 03/23/2017] [Indexed: 01/18/2023] Open
Abstract
3-n-Butylphthalide (NBP) protects the mitochondria and reduces apoptosis in multiple disease models. However, it remains to be determined whether NBP can protect muscle cells from oxidative stress, lipid peroxidation and apoptosis in myositis. In the present study, a myosin immunization protocol was applied to induce experimental autoimmune myositis (EAM) in guinea pigs. After 4 weeks, a low- or high-dose NBP solution was injected intraperitoneally into the guinea pigs, with saline solution serving as the negative control. After 10 days, the guinea pigs were sacrificed and muscle cells were isolated for analysis. The results revealed that NBP increased the superoxide dismutase and catalase activity, and reduced malondialdehyde activity in the EAM model. Furthermore, NBP enhanced ATPase activity in muscle mitochondrial membranes and muscle fiber membranes, reduced the number of apoptotic cells, and differentially regulated the Bcl-2, Bax and BAD mRNA and protein expression levels in muscle tissues and sera. NBP directly protects muscle mitochondria and muscle cells from oxidative damage. Notably, NBP reduced muscle cell apoptosis. Thus, it is speculated that, as an antioxidant treatment, NBP may benefit individuals with myopathy.
Collapse
Affiliation(s)
- Juan Chen
- Department of Neurology, Chinese PLA Medical School, Beijing 100853, P.R. China.,Department of Neurology, The 309th Hospital of PLA, Beijing 100091, P.R. China
| | - Jingyang Wang
- Department of Neurology, Chinese PLA Medical School, Beijing 100853, P.R. China
| | - Jiyan Zhang
- Laboratory of Immunology, Academy of Military Medical Sciences, Beijing 100850, P.R. China
| | - Chuanqiang Pu
- Department of Neurology, Chinese PLA Medical School, Beijing 100853, P.R. China
| |
Collapse
|
13
|
Todorova V, Blokland A. Mitochondria and Synaptic Plasticity in the Mature and Aging Nervous System. Curr Neuropharmacol 2017; 15:166-173. [PMID: 27075203 PMCID: PMC5327446 DOI: 10.2174/1570159x14666160414111821] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 03/23/2016] [Accepted: 04/11/2016] [Indexed: 01/11/2023] Open
Abstract
Synaptic plasticity in the adult brain is believed to represent the cellular mechanisms of learning and memory. Mitochondria are involved in the regulation of the complex processes of synaptic plasticity. This paper reviews the current knowledge on the regulatory roles of mitochondria in the function and plasticity of synapses and the implications of mitochondrial dysfunctions in synaptic transmission. First, the importance of mitochondrial distribution and motility for maintenance and strengthening of dendritic spines, but also for new spines/synapses formation is presented. Secondly, the major mitochondrial functions as energy supplier and calcium buffer organelles are considered as possible explanation for their essential and regulatory roles in neuronal plasticity processes. Thirdly, the effects of synaptic potentiation on mitochondrial gene expression are discussed. And finally, the relation between age-related alterations in synaptic plasticity and mitochondrial dysfunctions is considered. It appears that memory loss and neurodegeneration during aging are related to mitochondrial (dys)function. Although, it is clear that mitochondria are essential for synaptic plasticity, further studies are indicated to scrutinize the intracellular and molecular processes that regulate the functions of mitochondria in synaptic plasticity.
Collapse
Affiliation(s)
- Vyara Todorova
- Institute II for Anatomy, Medical Faculty, University of Cologne, Cologne, Germany
| | | |
Collapse
|
14
|
Kannurpatti SS. Mitochondrial calcium homeostasis: Implications for neurovascular and neurometabolic coupling. J Cereb Blood Flow Metab 2017; 37:381-395. [PMID: 27879386 PMCID: PMC5381466 DOI: 10.1177/0271678x16680637] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Mitochondrial function is critical to maintain high rates of oxidative metabolism supporting energy demands of both spontaneous and evoked neuronal activity in the brain. Mitochondria not only regulate energy metabolism, but also influence neuronal signaling. Regulation of "energy metabolism" and "neuronal signaling" (i.e. neurometabolic coupling), which are coupled rather than independent can be understood through mitochondria's integrative functions of calcium ion (Ca2+) uptake and cycling. While mitochondrial Ca2+ do not affect hemodynamics directly, neuronal activity changes are mechanistically linked to functional hyperemic responses (i.e. neurovascular coupling). Early in vitro studies lay the foundation of mitochondrial Ca2+ homeostasis and its functional roles within cells. However, recent in vivo approaches indicate mitochondrial Ca2+ homeostasis as maintained by the role of mitochondrial Ca2+ uniporter (mCU) influences system-level brain activity as measured by a variety of techniques. Based on earlier evidence of subcellular cytoplasmic Ca2+ microdomains and cellular bioenergetic states, a mechanistic model of Ca2+ mobilization is presented to understand systems-level neurovascular and neurometabolic coupling. This integrated view from molecular and cellular to the systems level, where mCU plays a major role in mitochondrial and cellular Ca2+ homeostasis, may explain the wide range of activation-induced coupling across neuronal activity, hemodynamic, and metabolic responses.
Collapse
|
15
|
Mishra J, Jhun BS, Hurst S, O-Uchi J, Csordás G, Sheu SS. The Mitochondrial Ca 2+ Uniporter: Structure, Function, and Pharmacology. Handb Exp Pharmacol 2017; 240:129-156. [PMID: 28194521 PMCID: PMC5554456 DOI: 10.1007/164_2017_1] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Mitochondrial Ca2+ uptake is crucial for an array of cellular functions while an imbalance can elicit cell death. In this chapter, we briefly reviewed the various modes of mitochondrial Ca2+ uptake and our current understanding of mitochondrial Ca2+ homeostasis in regards to cell physiology and pathophysiology. Further, this chapter focuses on the molecular identities, intracellular regulators as well as the pharmacology of mitochondrial Ca2+ uniporter complex.
Collapse
Affiliation(s)
- Jyotsna Mishra
- Center for Translational Medicine, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Suite 543D, Philadelphia, PA, 19107, USA
| | - Bong Sook Jhun
- Cardiovascular Research Center, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, 02903, USA
| | - Stephen Hurst
- Center for Translational Medicine, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Suite 543D, Philadelphia, PA, 19107, USA
| | - Jin O-Uchi
- Cardiovascular Research Center, Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, 02903, USA.
| | - György Csordás
- MitoCare Center for Mitochondrial Imaging Research and Diagnostics, Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA.
| | - Shey-Shing Sheu
- Center for Translational Medicine, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Suite 543D, Philadelphia, PA, 19107, USA.
| |
Collapse
|
16
|
Guleria A, Singh V, Chandna S. An attenuated calcium signaling and pre-emptive activation of UPR pathway together contribute to ER and calcium stress resilience of Lepidopteran insect cells. Biochim Biophys Acta Gen Subj 2016; 1861:504-521. [PMID: 27908702 DOI: 10.1016/j.bbagen.2016.11.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Revised: 11/22/2016] [Accepted: 11/25/2016] [Indexed: 11/26/2022]
Affiliation(s)
- Ayushi Guleria
- Division of Natural Radiation Response Mechanisms, Institute of Nuclear Medicine and Allied Sciences, Delhi 110054, India
| | - Vijaypal Singh
- Division of Natural Radiation Response Mechanisms, Institute of Nuclear Medicine and Allied Sciences, Delhi 110054, India
| | - Sudhir Chandna
- Division of Natural Radiation Response Mechanisms, Institute of Nuclear Medicine and Allied Sciences, Delhi 110054, India.
| |
Collapse
|
17
|
Essential protective role of tumor necrosis factor receptor 2 in neurodegeneration. Proc Natl Acad Sci U S A 2016; 113:12304-12309. [PMID: 27791020 DOI: 10.1073/pnas.1605195113] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Despite the recognized role of tumor necrosis factor (TNF) in inflammation and neuronal degeneration, anti-TNF therapeutics failed to treat neurodegenerative diseases. Animal disease models had revealed the antithetic effects of the two TNF receptors (TNFR) in the central nervous system, whereby TNFR1 has been associated with inflammatory degeneration and TNFR2 with neuroprotection. We here show the therapeutic potential of selective inhibition of TNFR1 and activation of TNFR2 by ATROSAB, a TNFR1-selective antagonistic antibody, and EHD2-scTNFR2, an agonistic TNFR2-selective TNF, respectively, in a mouse model of NMDA-induced acute neurodegeneration. Coadministration of either ATROSAB or EHD2-scTNFR2 into the magnocellular nucleus basalis significantly protected cholinergic neurons and their cortical projections against cell death, and reverted the neurodegeneration-associated memory impairment in a passive avoidance paradigm. Simultaneous blocking of TNFR1 and TNFR2 signaling, however, abrogated the therapeutic effect. Our results uncover an essential role of TNFR2 in neuroprotection. Accordingly, the therapeutic activity of ATROSAB is mediated by shifting the balance of the antithetic activity of endogenous TNF toward TNFR2, which appears essential for neuroprotection. Our data also explain earlier results showing that complete blocking of TNF activity by anti-TNF drugs was detrimental rather than protective and argue for the use of next-generation TNFR-selective TNF therapeutics as an effective approach in treating neurodegenerative diseases.
Collapse
|
18
|
Kwon SK, Hirabayashi Y, Polleux F. Organelle-Specific Sensors for Monitoring Ca 2+ Dynamics in Neurons. Front Synaptic Neurosci 2016; 8:29. [PMID: 27695411 PMCID: PMC5025517 DOI: 10.3389/fnsyn.2016.00029] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 08/30/2016] [Indexed: 11/16/2022] Open
Abstract
Calcium (Ca2+) plays innumerable critical functions in neurons ranging from regulation of neurotransmitter release and synaptic plasticity to activity-dependent transcription. Therefore, more than any other cell types, neurons are critically dependent on spatially and temporally controlled Ca2+ dynamics. This is achieved through an exquisite level of compartmentalization of Ca2+ storage and release from various organelles. The function of these organelles in the regulation of Ca2+ dynamics has been studied for decades using electrophysiological and optical methods combined with pharmacological and genetic alterations. Mitochondria and the endoplasmic reticulum (ER) are among the organelles playing the most critical roles in Ca2+ dynamics in neurons. At presynaptic boutons, Ca2+ triggers neurotransmitter release and synaptic plasticity, and postsynaptically, Ca2+ mobilization mediates long-term synaptic plasticity. To explore Ca2+ dynamics in live cells and intact animals, various synthetic and genetically encoded fluorescent Ca2+ sensors were developed, and recently, many groups actively increased the sensitivity and diversity of genetically encoded Ca2+ indicators (GECIs). Following conjugation with various signal peptides, these improved GECIs can be targeted to specific subcellular compartments, allowing monitoring of organelle-specific Ca2+ dynamics. Here, we review recent findings unraveling novel roles for mitochondria- and ER-dependent Ca2+ dynamics in neurons and at synapses.
Collapse
Affiliation(s)
- Seok-Kyu Kwon
- Department of Neuroscience, Mortimer B. Zuckerman Mind Brain Behavior Institute, Kavli Institute for Brain Science, Columbia University Medical Center New York, NY, USA
| | - Yusuke Hirabayashi
- Department of Neuroscience, Mortimer B. Zuckerman Mind Brain Behavior Institute, Kavli Institute for Brain Science, Columbia University Medical Center New York, NY, USA
| | - Franck Polleux
- Department of Neuroscience, Mortimer B. Zuckerman Mind Brain Behavior Institute, Kavli Institute for Brain Science, Columbia University Medical Center New York, NY, USA
| |
Collapse
|
19
|
Wagner S, De Bortoli S, Schwarzländer M, Szabò I. Regulation of mitochondrial calcium in plants versus animals. JOURNAL OF EXPERIMENTAL BOTANY 2016; 67:3809-29. [PMID: 27001920 DOI: 10.1093/jxb/erw100] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Ca(2+) acts as an important cellular second messenger in eukaryotes. In both plants and animals, a wide variety of environmental and developmental stimuli trigger Ca(2+) transients of a specific signature that can modulate gene expression and metabolism. In animals, mitochondrial energy metabolism has long been considered a hotspot of Ca(2+) regulation, with a range of pathophysiology linked to altered Ca(2+) control. Recently, several molecular players involved in mitochondrial Ca(2+) signalling have been identified, including those of the mitochondrial Ca(2+) uniporter. Despite strong evidence for sophisticated Ca(2+) regulation in plant mitochondria, the picture has remained much less clear. This is currently changing aided by live imaging and genetic approaches which allow dissection of subcellular Ca(2+) dynamics and identification of the proteins involved. We provide an update on our current understanding in the regulation of mitochondrial Ca(2+) and signalling by comparing work in plants and animals. The significance of mitochondrial Ca(2+) control is discussed in the light of the specific metabolic and energetic needs of plant and animal cells.
Collapse
Affiliation(s)
- Stephan Wagner
- Plant Energy Biology Lab, Institute of Crop Science and Resource Conservation (INRES), University of Bonn, Friedrich-Ebert-Allee 144, D-53113 Bonn, Germany
| | - Sara De Bortoli
- Department of Biology and CNR Institute of Neurosciences, University of Padova, Viale G. Colombo 3, 35121 Padova, Italy
| | - Markus Schwarzländer
- Plant Energy Biology Lab, Institute of Crop Science and Resource Conservation (INRES), University of Bonn, Friedrich-Ebert-Allee 144, D-53113 Bonn, Germany
| | - Ildikò Szabò
- Department of Biology and CNR Institute of Neurosciences, University of Padova, Viale G. Colombo 3, 35121 Padova, Italy
| |
Collapse
|
20
|
Reilly JM, Dharmalingam B, Marsh SJ, Thompson V, Goebel A, Brown DA. Effects of serum immunoglobulins from patients with complex regional pain syndrome (CRPS) on depolarisation-induced calcium transients in isolated dorsal root ganglion (DRG) neurons. Exp Neurol 2015; 277:96-102. [PMID: 26708558 DOI: 10.1016/j.expneurol.2015.12.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 12/10/2015] [Accepted: 12/15/2015] [Indexed: 12/28/2022]
Abstract
Complex regional pain syndrome (CRPS) is thought to have an auto-immune component. One such target recently proposed from the effects of auto-immune IgGs on Ca(2+) transients in cardiac myocytes and cell lines is the α1-adrenoceptor. We have tested whether such IgGs exerted comparable effects on nociceptive sensory neurons isolated from rat dorsal root ganglia. Depolarisation-induced [Ca(2+)]i transients were generated by applying 30 mM KCl for 2 min and monitored by Fura-2 fluorescence imaging. No IgGs tested (including 3 from CRPS patients) had any significant effect on these [Ca(2+)]i transients. However, IgG from one CRPS patient consistently and significantly reduced the K(+)-induced response of cells that had been pre-incubated for 24h with a mixture of inflammatory mediators (1 μM histamine, 5-hydroxytryptamine, bradykinin and PGE2). Since this pre-incubation also appeared to induce a comparable inhibitory response to the α1-agonist phenylephrine, this is compatible with the α1-adrenoceptor as a target for CRPS auto-immunity. A mechanism whereby this might enhance pain is suggested.
Collapse
Affiliation(s)
- Joanne M Reilly
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Backialakshmi Dharmalingam
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK; Pain Research Institute, Department of Translational Medicine, University of Liverpool, Liverpool, L9 7AL, UK
| | - Stephen J Marsh
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Victoria Thompson
- Pain Research Institute, Department of Translational Medicine, University of Liverpool, Liverpool, L9 7AL, UK
| | - Andreas Goebel
- Pain Research Institute, Department of Translational Medicine, University of Liverpool, Liverpool, L9 7AL, UK.
| | - David A Brown
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| |
Collapse
|
21
|
Félix-Martínez GJ, Godínez-Fernández JR. Modeling Ca(2+) currents and buffered diffusion of Ca(2+) in human β-cells during voltage clamp experiments. Math Biosci 2015; 270:66-80. [PMID: 26476144 DOI: 10.1016/j.mbs.2015.09.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 09/03/2015] [Accepted: 09/28/2015] [Indexed: 11/27/2022]
Abstract
Macroscopic Ca(2+) currents of the human β-cells were characterized using the Hodgkin-Huxley formalism. Expressions describing the Ca(2+)-dependent inactivation process of the L-type Ca(2+) channels in terms of the concentration of Ca(2+) were obtained. By coupling the modeled Ca(2+) currents to a three-dimensional model of buffered diffusion of Ca(2+), we simulated the Ca(2+) transients formed in the immediate vicinity of the cell membrane during voltage clamp experiments performed in high buffering conditions. Our modeling approach allowed us to consider the distribution of the Ca(2+) sources over the cell membrane. The effect of exogenous (EGTA) and endogenous Ca(2+) buffers on the temporal course of the Ca(2+) transients was evaluated. We show that despite the high Ca(2+) buffering capacity, nanodomains are formed in the submembrane space, where a peak Ca(2+) concentration between ∼76 and 143 µM was estimated from our simulations. In addition, the contribution of each Ca(2+) current to the formation of the Ca(2+) nanodomains was also addressed. Here we provide a general framework to incorporate the spatial aspects to the models of the pancreatic β-cell, such as a more detailed and realistic description of Ca(2+) dynamics in response to electrical activity in physiological conditions can be provided by future models.
Collapse
Affiliation(s)
- Gerardo J Félix-Martínez
- Department of Electrical Engineering, Universidad Autónoma Metropolitana Iztapalapa, México, D.F., Mexico .
| | - J Rafael Godínez-Fernández
- Department of Electrical Engineering, Universidad Autónoma Metropolitana Iztapalapa, México, D.F., Mexico
| |
Collapse
|
22
|
Matthews EA, Dietrich D. Buffer mobility and the regulation of neuronal calcium domains. Front Cell Neurosci 2015; 9:48. [PMID: 25750615 PMCID: PMC4335178 DOI: 10.3389/fncel.2015.00048] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 01/31/2015] [Indexed: 11/13/2022] Open
Abstract
The diffusion of calcium inside neurons is determined in part by the intracellular calcium binding species that rapidly bind to free calcium ions upon entry. It has long been known that some portion of a neuron's intracellular calcium binding capacity must be fixed or poorly mobile, as calcium diffusion is strongly slowed in the intracellular environment relative to diffusion in cytosolic extract. The working assumption was that these immobile calcium binding sites are provided by structural proteins bound to the cytoskeleton or intracellular membranes and may thereby be relatively similar in composition and capacity across different cell types. However, recent evidence suggests that the immobile buffering capacity can vary greatly between cell types and that some mobile calcium binding proteins may alter their mobility upon binding calcium, thus blurring the line between mobile and immobile. The ways in which immobile buffering capacity might be relevant to different calcium domains within neurons has been explored primarily through modeling. In certain regimes, the presence of immobile buffers and the interaction between mobile and immobile buffers have been shown to result in complex spatiotemporal patterns of free calcium. In total, these experimental and modeling findings call for a more nuanced consideration of the local intracellular calcium microenvironment. In this review we focus on the different amounts, affinities, and mobilities of immobile calcium binding species; propose a new conceptual category of physically diffusible but functionally immobile buffers; and discuss how these buffers might interact with mobile calcium binding partners to generate characteristic calcium domains.
Collapse
Affiliation(s)
- Elizabeth A. Matthews
- Experimental Neurophysiology, Department of Neurosurgery, University Clinic BonnBonn, Germany
| | | |
Collapse
|
23
|
Abstract
The neuromuscular junction (NMJ) is a synapse between motor neurons and skeletal muscle fibers, and is critical for control of muscle contraction. Its formation requires neuronal agrin that acts by binding to LRP4 to stimulate MuSK. Mutations have been identified in agrin, MuSK, and LRP4 in patients with congenital myasthenic syndrome, and patients with myasthenia gravis develop antibodies against agrin, LRP4, and MuSK. However, it remains unclear whether the agrin signaling pathway is critical for NMJ maintenance because null mutation of any of the three genes is perinatal lethal. In this study, we generated imKO mice, a mutant strain whose LRP4 gene can be deleted in muscles by doxycycline (Dox) treatment. Ablation of the LRP4 gene in adult muscle enabled studies of its role in NMJ maintenance. We demonstrate that Dox treatment of P30 mice reduced muscle strength and compound muscle action potentials. AChR clusters became fragmented with diminished junctional folds and synaptic vesicles. The amplitude and frequency of miniature endplate potentials were reduced, indicating impaired neuromuscular transmission and providing cellular mechanisms of adult LRP4 deficiency. We showed that LRP4 ablation led to the loss of synaptic agrin and the 90 kDa fragments, which occurred ahead of other prejunctional and postjunctional components, suggesting that LRP4 may regulate the stability of synaptic agrin. These observations demonstrate that LRP4 is essential for maintaining the structural and functional integrity of the NMJ and that loss of muscle LRP4 in adulthood alone is sufficient to cause myasthenic symptoms.
Collapse
|
24
|
Scheff NN, Yilmaz E, Gold MS. The properties, distribution and function of Na(+)-Ca(2+) exchanger isoforms in rat cutaneous sensory neurons. J Physiol 2014; 592:4969-93. [PMID: 25239455 PMCID: PMC4259538 DOI: 10.1113/jphysiol.2014.278036] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 09/10/2014] [Indexed: 11/08/2022] Open
Abstract
The Na(+)-Ca(2+) exchanger (NCX) appears to play an important role in the regulation of the high K(+)-evoked Ca(2+) transient in putative nociceptive dorsal root ganglion (DRG) neurons. The purpose of the present study was to (1) characterize the properties of NCX activity in subpopulations of DRG neurons, (2) identify the isoform(s) underlying NCX activity, and (3) begin to assess the function of the isoform(s) in vivo. In retrogradely labelled neurons from the glabrous skin of adult male Sprague-Dawley rats, NCX activity, as assessed with fura-2-based microfluorimetry, was only detected in putative nociceptive IB4+ neurons. There were two modes of NCX activity: one was evoked in response to relatively large and long lasting (∼325 nm for >12 s) increases in the concentration of intracellular Ca(2+) ([Ca(2+)]i), and a second was active at resting [Ca(2+)]i > ∼150 nm. There also were two modes of evoked activity: one that decayed relatively rapidly (<5 min) and a second that persisted (>10 min). Whereas mRNA encoding all three NCX isoforms (NCX1-3) was detected in putative nociceptive cutaneous neurons with single cell PCR, pharmacological analysis and small interfering RNA (siRNA) knockdown of each isoform in vivo suggested that NCX2 and 3 were responsible for NCX activity. Western blot analyses suggested that NCX isoforms were differentially distributed within sensory neurons. Functional assays of excitability, action potential propagation, and nociceptive behaviour suggest NCX activity has little influence on excitability per se, but instead influences axonal conduction velocity, resting membrane potential, and nociceptive threshold. Together these results indicate that the function of NCX in the regulation of [Ca(2+)]i in putative nociceptive neurons may be unique relative to other cells in which these exchanger isoforms have been characterized and it has the potential to influence sensory neuron properties at multiple levels.
Collapse
Affiliation(s)
- N N Scheff
- The Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA Department of Anesthesiology, University of Pittsburgh, Pittsburgh, PA, USA Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - E Yilmaz
- The Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA Department of Anesthesiology, University of Pittsburgh, Pittsburgh, PA, USA Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - M S Gold
- The Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA Department of Anesthesiology, University of Pittsburgh, Pittsburgh, PA, USA Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
25
|
Hogan QH, Sprick C, Guo Y, Mueller S, Bienengraeber M, Pan B, Wu HE. Divergent effects of painful nerve injury on mitochondrial Ca(2+) buffering in axotomized and adjacent sensory neurons. Brain Res 2014; 1589:112-25. [PMID: 25251590 DOI: 10.1016/j.brainres.2014.09.040] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Revised: 09/09/2014] [Accepted: 09/15/2014] [Indexed: 02/05/2023]
Abstract
Mitochondria critically regulate cytoplasmic Ca(2+) concentration ([Ca(2+)]c), but the effects of sensory neuron injury have not been examined. Using FCCP (1µM) to eliminate mitochondrial Ca(2+) uptake combined with oligomycin (10µM) to prevent ATP depletion, we first identified features of depolarization-induced neuronal [Ca(2+)]c transients that are sensitive to blockade of mitochondrial Ca(2+) buffering in order to assess mitochondrial contributions to [Ca(2+)]c regulation. This established the loss of a shoulder during the recovery of the depolarization (K(+))-induced transient, increased transient peak and area, and elevated shoulder level as evidence of diminished mitochondrial Ca(2+) buffering. We then examined transients in Control neurons and neurons from the 4th lumbar (L4) and 5th lumbar (L5) dorsal root ganglia after L5 spinal nerve ligation (SNL). The SNL L4 neurons showed decreased transient peak and area compared to control neurons, while the SNL L5 neurons showed increased shoulder level. Additionally, SNL L4 neurons developed shoulders following transients with lower peaks than Control neurons. Application of FCCP plus oligomycin elevated resting [Ca(2+)]c in SNL L4 neurons more than in Control neurons. Whereas application of FCCP plus oligomycin 2s after neuronal depolarization initiated mitochondrial Ca(2+) release in most Control and SNL L4 neurons, this usually failed to release mitochondrial Ca(2+) from SNL L5 neurons. For comparable cytoplasmic Ca(2+) loads, the releasable mitochondrial Ca(2+) in SNL L5 neurons was less than Control while it was increased in SNL L4 neurons. These findings show diminished mitochondrial Ca(2+) buffering in axotomized SNL L5 neurons but enhanced Ca(2+) buffering by neurons in adjacent SNL L4 neurons.
Collapse
Affiliation(s)
- Quinn H Hogan
- Department of Anesthesiology, Medical College of Wisconsin, 8701 W Watertown Plank, Milwaukee, WI 53226, USA; Zablocki VA Medical Center, 5000 W National Avenue, Milwaukee, WI 53295, USA
| | - Chelsea Sprick
- Department of Anesthesiology, Medical College of Wisconsin, 8701 W Watertown Plank, Milwaukee, WI 53226, USA
| | - Yuan Guo
- Department of Anesthesiology, Medical College of Wisconsin, 8701 W Watertown Plank, Milwaukee, WI 53226, USA
| | - Samantha Mueller
- Department of Anesthesiology, Medical College of Wisconsin, 8701 W Watertown Plank, Milwaukee, WI 53226, USA
| | - Martin Bienengraeber
- Department of Anesthesiology, Medical College of Wisconsin, 8701 W Watertown Plank, Milwaukee, WI 53226, USA; Department of Pharmacology and Toxicology, Medical College of Wisconsin, USA
| | - Bin Pan
- Department of Anesthesiology, Medical College of Wisconsin, 8701 W Watertown Plank, Milwaukee, WI 53226, USA
| | - Hsiang-En Wu
- Department of Anesthesiology, Medical College of Wisconsin, 8701 W Watertown Plank, Milwaukee, WI 53226, USA.
| |
Collapse
|
26
|
Kim MS, Shutov LP, Gnanasekaran A, Lin Z, Rysted JE, Ulrich JD, Usachev YM. Nerve growth factor (NGF) regulates activity of nuclear factor of activated T-cells (NFAT) in neurons via the phosphatidylinositol 3-kinase (PI3K)-Akt-glycogen synthase kinase 3β (GSK3β) pathway. J Biol Chem 2014; 289:31349-60. [PMID: 25231981 DOI: 10.1074/jbc.m114.587188] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The Ca(2+)/calcineurin-dependent transcription factor nuclear factor of activated T-cells (NFAT) plays an important role in regulating many neuronal functions, including excitability, axonal growth, synaptogenesis, and neuronal survival. NFAT can be activated by action potential firing or depolarization that leads to Ca(2+)/calcineurin-dependent dephosphorylation of NFAT and its translocation to the nucleus. Recent data suggest that NFAT and NFAT-dependent functions in neurons can also be potently regulated by NGF and other neurotrophins. However, the mechanisms of NFAT regulation by neurotrophins are not well understood. Here, we show that in dorsal root ganglion sensory neurons, NGF markedly facilitates NFAT-mediated gene expression induced by mild depolarization. The effects of NGF were not associated with changes in [Ca(2+)]i and were independent of phospholipase C activity. Instead, the facilitatory effect of NGF depended on activation of the PI3K/Akt pathway downstream of the TrkA receptor and on inhibition of glycogen synthase kinase 3β (GSK3β), a protein kinase known to phosphorylate NFAT and promote its nuclear export. Knockdown or knockout of NFATc3 eliminated this facilitatory effect. Simultaneous monitoring of EGFP-NFATc3 nuclear translocation and [Ca(2+)]i changes in dorsal root ganglion neurons indicated that NGF slowed the rate of NFATc3 nuclear export but did not affect its nuclear import rate. Collectively, our data suggest that NGF facilitates depolarization-induced NFAT activation by stimulating PI3K/Akt signaling, inactivating GSK3β, and thereby slowing NFATc3 export from the nucleus. We propose that NFAT serves as an integrator of neurotrophin action and depolarization-driven calcium signaling to regulate neuronal gene expression.
Collapse
Affiliation(s)
- Man-Su Kim
- From the Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242 and the College of Pharmacy, Inje University, Gimhae 621-749, Korea
| | - Leonid P Shutov
- From the Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242 and
| | - Aswini Gnanasekaran
- From the Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242 and
| | - Zhihong Lin
- From the Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242 and
| | - Jacob E Rysted
- From the Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242 and
| | - Jason D Ulrich
- From the Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242 and
| | - Yuriy M Usachev
- From the Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242 and
| |
Collapse
|
27
|
Gao J, Yao H, Pan XD, Xie AM, Zhang L, Song JH, Ma AJ, Liu ZC. Alteration of mitochondrial function and ultrastructure in the hippocampus of pilocarpine-treated rat. Epilepsy Res 2014; 108:162-70. [DOI: 10.1016/j.eplepsyres.2013.11.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 09/17/2013] [Accepted: 11/21/2013] [Indexed: 11/26/2022]
|
28
|
Brustovetsky T, Pellman JJ, Yang XF, Khanna R, Brustovetsky N. Collapsin response mediator protein 2 (CRMP2) interacts with N-methyl-D-aspartate (NMDA) receptor and Na+/Ca2+ exchanger and regulates their functional activity. J Biol Chem 2014; 289:7470-82. [PMID: 24474686 DOI: 10.1074/jbc.m113.518472] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Collapsin response mediator protein 2 (CRMP2) is traditionally viewed as an axonal growth protein involved in axon/dendrite specification. Here, we describe novel functions of CRMP2. A 15-amino acid peptide from CRMP2, fused to the TAT cell-penetrating motif of the HIV-1 protein, TAT-CBD3, but not CBD3 without TAT, attenuated N-methyl-d-aspartate receptor (NMDAR) activity and protected neurons against glutamate-induced Ca(2+) dysregulation, suggesting the key contribution of CRMP2 in these processes. In addition, TAT-CBD3, but not CBD3 without TAT or TAT-scramble peptide, inhibited increases in cytosolic Ca(2+) mediated by the plasmalemmal Na(+)/Ca(2+) exchanger (NCX) operating in the reverse mode. Co-immunoprecipitation experiments revealed an interaction between CRMP2 and NMDAR as well as NCX3 but not NCX1. TAT-CBD3 disrupted CRMP2-NMDAR interaction without change in NMDAR localization. In contrast, TAT-CBD3 augmented the CRMP2-NCX3 co-immunoprecipitation, indicating increased interaction or stabilization of a complex between these proteins. Immunostaining with an anti-NCX3 antibody revealed that TAT-CBD3 induced NCX3 internalization, suggesting that both reverse and forward modes of NCX might be affected. Indeed, the forward mode of NCX, evaluated in experiments with ionomycin-induced Ca(2+) influx into neurons, was strongly suppressed by TAT-CBD3. Knockdown of CRMP2 with short interfering RNA (siRNA) prevented NCX3 internalization in response to TAT-CBD3 exposure. Moreover, CRMP2 down-regulation strongly attenuated TAT-CBD3-induced inhibition of reverse NCX. Overall, our results demonstrate that CRMP2 interacts with NCX and NMDAR and that TAT-CBD3 protects against glutamate-induced Ca(2+) dysregulation most likely via suppression of both NMDAR and NCX activities. Our results further clarify the mechanism of action of TAT-CBD3 and identify a novel regulatory checkpoint for NMDAR and NCX function based on CRMP2 interaction with these proteins.
Collapse
|
29
|
Evidence for A1 and A 3 receptors mediating adenosine-induced intracellular calcium release in the dorsal root ganglion neurons by using confocal microscopy imaging. Lasers Med Sci 2013; 29:1209-15. [PMID: 24352876 DOI: 10.1007/s10103-013-1511-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 12/05/2013] [Indexed: 10/25/2022]
Abstract
Adenosine exerts a key role in analgesia. In the present study, adenosine-induced Ca(2+) responses were revealed by using confocal microscopy imaging in the rat dorsal root ganglia (DRG) neurons in vitro. Our results showed that adenosine could evoke increases in the intracellular Ca(2+) concentration in the DRG neurons. In addition, by application of selective receptor antagonists, two types of receptors, A1R and A3R, were identified to be involved in the adenosine-induced Ca(2+) release from intracellular stores in neurons. Altogether, these results suggest that confocal microscopy imaging combined with fluorescent dyes could help to detect the analgesic-induced ion signaling in single cell.
Collapse
|
30
|
Cytosolic calcium regulation in rat afferent vagal neurons during anoxia. Cell Calcium 2013; 54:416-27. [PMID: 24189167 DOI: 10.1016/j.ceca.2013.10.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2013] [Revised: 09/30/2013] [Accepted: 10/06/2013] [Indexed: 12/12/2022]
Abstract
Sensory neurons are able to detect tissue ischaemia and both transmit information to the brainstem as well as release local vasoactive mediators. Their ability to sense tissue ischaemia is assumed to be primarily mediated through proton sensing ion channels, lack of oxygen however may also affect sensory neuron function. In this study we investigated the effects of anoxia on isolated capsaicin sensitive neurons from rat nodose ganglion. Acute anoxia triggered a reversible increase in [Ca2+]i that was mainly due to Ca2+-efflux from FCCP sensitive stores and from caffeine and CPA sensitive ER stores. Prolonged anoxia resulted in complete depletion of ER Ca2+-stores. Mitochondria were partially depolarised by acute anoxia but mitochondrial Ca2+-uptake/buffering during voltage-gated Ca2+-influx was unaffected. The process of Ca2+-release from mitochondria and cytosolic Ca2+-clearance following Ca2+ influx was however significantly slowed. Anoxia was also found to inhibit SERCA activity and, to a lesser extent, PMCA activity. Hence, anoxia has multiple influences on [Ca2+]i homeostasis in vagal afferent neurons, including depression of ATP-driven Ca2+-pumps, modulation of the kinetics of mitochondrial Ca2+ buffering/release and Ca2+-release from, and depletion of, internal Ca2+-stores. These effects are likely to influence sensory neuronal function during ischaemia.
Collapse
|
31
|
Tsutsui S, Stys PK. Metabolic injury to axons and myelin. Exp Neurol 2013; 246:26-34. [DOI: 10.1016/j.expneurol.2012.04.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Revised: 03/20/2012] [Accepted: 04/23/2012] [Indexed: 12/31/2022]
|
32
|
Abstract
Calcium (Ca(2+)) uptake into the mitochondrial matrix is critically important to cellular function. As a regulator of matrix Ca(2+) levels, this flux influences energy production and can initiate cell death. If large, this flux could potentially alter intracellular Ca(2+) ([Ca(2+)]i) signals. Despite years of study, fundamental disagreements on the extent and speed of mitochondrial Ca(2+) uptake still exist. Here, we review and quantitatively analyze mitochondrial Ca(2+) uptake fluxes from different tissues and interpret the results with respect to the recently proposed mitochondrial Ca(2+) uniporter (MCU) candidate. This quantitative analysis yields four clear results: (i) under physiological conditions, Ca(2+) influx into the mitochondria via the MCU is small relative to other cytosolic Ca(2+) extrusion pathways; (ii) single MCU conductance is ∼6-7 pS (105 mM [Ca(2+)]), and MCU flux appears to be modulated by [Ca(2+)]i, suggesting Ca(2+) regulation of MCU open probability (P(O)); (iii) in the heart, two features are clear: the number of MCU channels per mitochondrion can be calculated, and MCU probability is low under normal conditions; and (iv) in skeletal muscle and liver cells, uptake per mitochondrion varies in magnitude but total uptake per cell still appears to be modest. Based on our analysis of available quantitative data, we conclude that although Ca(2+) critically regulates mitochondrial function, the mitochondria do not act as a significant dynamic buffer of cytosolic Ca(2+) under physiological conditions. Nevertheless, with prolonged (superphysiological) elevations of [Ca(2+)]i, mitochondrial Ca(2+) uptake can increase 10- to 1,000-fold and begin to shape [Ca(2+)]i dynamics.
Collapse
|
33
|
Separate Ca2+ sources are buffered by distinct Ca2+ handling systems in aplysia neuroendocrine cells. J Neurosci 2013; 33:6476-91. [PMID: 23575846 DOI: 10.1523/jneurosci.6384-11.2013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Although the contribution of Ca(2+) buffering systems can vary between neuronal types and cellular compartments, it is unknown whether distinct Ca(2+) sources within a neuron have different buffers. As individual Ca(2+) sources can have separate functions, we propose that each is handled by unique systems. Using Aplysia californica bag cell neurons, which initiate reproduction through an afterdischarge involving multiple Ca(2+)-dependent processes, we investigated the role of endoplasmic reticulum (ER) and mitochondrial sequestration, as well as extrusion via the plasma membrane Ca(2+)-ATPase (PMCA) and Na(+)/Ca(2+) exchanger, to the clearance of voltage-gated Ca(2+) influx, Ca(2+)-induced Ca(2+)-release (CICR), and store-operated Ca(2+) influx. Cultured bag cell neurons were filled with the Ca(2+) indicator, fura-PE3, to image Ca(2+) under whole-cell voltage clamp. A 5 Hz, 1 min train of depolarizing voltage steps elicited voltage-gated Ca(2+) influx followed by EGTA-sensitive CICR from the mitochondria. A compartment model of Ca(2+) indicated the effect of EGTA on CICR was due to buffering of released mitochondrial Ca(2+) rather than uptake competition. Removal of voltage-gated Ca(2+) influx was dominated by the mitochondria and PMCA, with no contribution from the Na(+)/Ca(2+) exchanger or sarcoplasmic/endoplasmic Ca(2+)-ATPase (SERCA). In contrast, CICR recovery was slowed by eliminating the Na(+)/Ca(2+) exchanger and PMCA. Last, store-operated influx, evoked by ER depletion, was removed by the SERCA and depended on the mitochondrial membrane potential. Our results demonstrate that distinct buffering systems are dedicated to particular Ca(2+) sources. In general, this may represent a means to differentially regulate Ca(2+)-dependent processes, and for Aplysia, influence how reproductive behavior is triggered.
Collapse
|
34
|
Zsurka G, Kunz WS. Mitochondrial involvement in neurodegenerative diseases. IUBMB Life 2013; 65:263-72. [PMID: 23341346 DOI: 10.1002/iub.1126] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 12/06/2012] [Accepted: 12/06/2012] [Indexed: 01/12/2023]
Abstract
The classical bioenergetical view of the involvement of mitochondria in neurogeneration is based on the fact that mitochondria are the central players of ATP synthesis in neurons and their failure leads to neuronal dysfunction and eventually to cell death. Mutations in at least 39 genes in inherited neurodegenerative disorders seem to alter directly or indirectly mitochondrial function. Most of these mutations do not directly affect oxidative phosphorylation, but act through disturbed mitochondrial dynamics and quality control. This, however, does not invalidate the bioenergetic hypothesis. Neurodegeneration is not necessarily associated with a gross failure of ATP production, but might rather be a consequence of local insufficiencies of ATP supply in critical compartments of neurons, like the presynaptic terminal. We hypothesize that slow disease progression, at least in a subgroup of neurodegenerative diseases, can be explained by the parallel action of subcellular ATP insufficiency and clonal expansion of somatic mitochondrial DNA mutations, and particularly deletions.
Collapse
Affiliation(s)
- Gábor Zsurka
- Department of Epileptology and Life and Brain Center, University Bonn, Bonn, Germany
| | | |
Collapse
|
35
|
Abstract
Calcium is important in controlling nuclear gene expression through the activation of multiple signal-transduction pathways in neurons. Compared with other voltage-gated calcium channels, CaV1 channels demonstrate a considerable advantage in signalling to the nucleus. In this review, we summarize the recent progress in elucidating the mechanisms involved. CaV1 channels, already advantaged in their responsiveness to depolarization, trigger communication with the nucleus by attracting colocalized clusters of activated CaMKII (Ca2+/calmodulin-dependent protein kinase II). CaV2 channels lack this ability, but must work at a distance of >1 μm from the CaV1-CaMKII co-clusters, which hampers their relative efficiency for a given rise in bulk [Ca2+]i (intracellular [Ca2+]). Moreover, Ca2+ influx from CaV2 channels is preferentially buffered by the ER (endoplasmic reticulum) and mitochondria, further attenuating their effectiveness in signalling to the nucleus.
Collapse
|
36
|
Peroxisome proliferator-activated receptor α mediates acute effects of palmitoylethanolamide on sensory neurons. J Neurosci 2012; 32:12735-43. [PMID: 22972997 DOI: 10.1523/jneurosci.0130-12.2012] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The amplitude of the depolarization-evoked Ca2+ transient is larger in dorsal root ganglion (DRG) neurons from tumor-bearing mice compared with that of neurons from naive mice, and the change is mimicked by coculturing DRG neurons with the fibrosarcoma cells used to generate the tumors (Khasabova et al., 2007). The effect of palmitoylethanolamide (PEA), a ligand for the peroxisome proliferator-activated receptor α (PPARα), was determined on the evoked-Ca2+ transient in the coculture condition. The level of PEA was reduced in DRG cells from tumor-bearing mice as well as those cocultured with fibrosarcoma cells. Pretreatment with PEA, a synthetic PPARα agonist (GW7647), or ARN077, an inhibitor of the enzyme that hydrolyzes PEA, acutely decreased the amplitude of the evoked Ca2+ transient in small DRG neurons cocultured with fibrosarcoma cells. The PPARα antagonist GW6471 blocked the effect of each. In contrast, the PPARα agonist was without effect in the control condition, but the antagonist increased the amplitude of the Ca2+ transient, suggesting that PPARα receptors are saturated by endogenous ligand under basal conditions. Effects of drugs on mechanical sensitivity in vivo paralleled their effects on DRG neurons in vitro. Local injection of ARN077 decreased mechanical hyperalgesia in tumor-bearing mice, and the effect was blocked by GW6471. These data support the conclusion that the activity of DRG neurons is rapidly modulated by PEA through a PPARα-dependent mechanism. Moreover, agents that increase the activity of PPARα may provide a therapeutic strategy to reduce tumor-evoked pain.
Collapse
|
37
|
Wheeler DG, Groth RD, Ma H, Barrett CF, Owen SF, Safa P, Tsien RW. Ca(V)1 and Ca(V)2 channels engage distinct modes of Ca(2+) signaling to control CREB-dependent gene expression. Cell 2012; 149:1112-24. [PMID: 22632974 DOI: 10.1016/j.cell.2012.03.041] [Citation(s) in RCA: 183] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2009] [Revised: 11/11/2011] [Accepted: 03/07/2012] [Indexed: 12/23/2022]
Abstract
Activity-dependent gene expression triggered by Ca(2+) entry into neurons is critical for learning and memory, but whether specific sources of Ca(2+) act distinctly or merely supply Ca(2+) to a common pool remains uncertain. Here, we report that both signaling modes coexist and pertain to Ca(V)1 and Ca(V)2 channels, respectively, coupling membrane depolarization to CREB phosphorylation and gene expression. Ca(V)1 channels are advantaged in their voltage-dependent gating and use nanodomain Ca(2+) to drive local CaMKII aggregation and trigger communication with the nucleus. In contrast, Ca(V)2 channels must elevate [Ca(2+)](i) microns away and promote CaMKII aggregation at Ca(V)1 channels. Consequently, Ca(V)2 channels are ~10-fold less effective in signaling to the nucleus than are Ca(V)1 channels for the same bulk [Ca(2+)](i) increase. Furthermore, Ca(V)2-mediated Ca(2+) rises are preferentially curbed by uptake into the endoplasmic reticulum and mitochondria. This source-biased buffering limits the spatial spread of Ca(2+), further attenuating Ca(V)2-mediated gene expression.
Collapse
Affiliation(s)
- Damian G Wheeler
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305-5345, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Mitochondria, calcium-dependent neuronal death and neurodegenerative disease. Pflugers Arch 2012; 464:111-21. [PMID: 22615071 PMCID: PMC3387496 DOI: 10.1007/s00424-012-1112-0] [Citation(s) in RCA: 138] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 04/29/2012] [Accepted: 05/02/2012] [Indexed: 11/18/2022]
Abstract
Understanding the mechanisms of neuronal dysfunction and death represents a major frontier in contemporary medicine, involving the acute cell death in stroke, and the attrition of the major neurodegenerative diseases, including Parkinson's, Alzheimer's, Huntington's and Motoneuron diseases. A growing body of evidence implicates mitochondrial dysfunction as a key step in the pathogenesis of all these diseases, with the promise that mitochondrial processes represent valuable potential therapeutic targets. Each disease is characterised by the loss of a specific vulnerable population of cells—dopaminergic neurons in Parkinson's disease, spinal motoneurons in Motoneuron disease, for example. We discuss the possible roles of cell type-specific calcium signalling mechanisms in defining the pathological phenotype of each of these major diseases and review central mechanisms of calcium-dependent mitochondrial-mediated cell death.
Collapse
|
39
|
Calì T, Ottolini D, Brini M. Mitochondrial Ca(2+) and neurodegeneration. Cell Calcium 2012; 52:73-85. [PMID: 22608276 PMCID: PMC3396847 DOI: 10.1016/j.ceca.2012.04.015] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2012] [Revised: 04/18/2012] [Accepted: 04/20/2012] [Indexed: 12/16/2022]
Abstract
Mitochondria are essential for ensuring numerous fundamental physiological processes such as cellular energy, redox balance, modulation of Ca2+ signaling and important biosynthetic pathways. They also govern the cell fate by participating in the apoptosis pathway. The mitochondrial shape, volume, number and distribution within the cells are strictly controlled. The regulation of these parameters has an impact on mitochondrial function, especially in the central nervous system, where trafficking of mitochondria is critical to their strategic intracellular distribution, presumably according to local energy demands. Thus, the maintenance of a healthy mitochondrial population is essential to avoid the impairment of the processes they regulate: for this purpose, cells have developed mechanisms involving a complex system of quality control to remove damaged mitochondria, or to renew them. Defects of these processes impair mitochondrial function and lead to disordered cell function, i.e., to a disease condition. Given the standard role of mitochondria in all cells, it might be expected that their dysfunction would give rise to similar defects in all tissues. However, damaged mitochondrial function has pleiotropic effects in multicellular organisms, resulting in diverse pathological conditions, ranging from cardiac and brain ischemia, to skeletal muscle myopathies to neurodegenerative diseases. In this review, we will focus on the relationship between mitochondrial (and cellular) derangements and Ca2+ dysregulation in neurodegenerative diseases, emphasizing the evidence obtained in genetic models. Common patterns, that recognize the derangement of Ca2+ and energy control as a causative factor, have been identified: advances in the understanding of the molecular regulation of Ca2+ homeostasis, and on the ways in which it could become perturbed in neurological disorders, may lead to the development of therapeutic strategies that modulate neuronal Ca2+ signaling.
Collapse
Affiliation(s)
- Tito Calì
- Department of Comparative Biomedicine and Food Science, University of Padova, Padova, Italy
| | | | | |
Collapse
|
40
|
Brittain MK, Brustovetsky T, Sheets PL, Brittain JM, Khanna R, Cummins TR, Brustovetsky N. Delayed calcium dysregulation in neurons requires both the NMDA receptor and the reverse Na+/Ca2+ exchanger. Neurobiol Dis 2012; 46:109-17. [PMID: 22249110 DOI: 10.1016/j.nbd.2011.12.051] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Revised: 12/12/2011] [Accepted: 12/31/2011] [Indexed: 12/20/2022] Open
Abstract
Glutamate-induced delayed calcium dysregulation (DCD) is a causal factor leading to neuronal death. The mechanism of DCD is not clear but Ca2+ influx via N-methyl-d-aspartate receptors (NMDAR) and/or the reverse plasmalemmal Na+/Ca2+ exchanger (NCXrev) could be involved in DCD. However, the extent to which NMDAR and NCX(rev) contribute to glutamate-induced DCD is uncertain. Here, we show that both NMDAR and NCX(rev) are critical for DCD in neurons exposed to excitotoxic glutamate. In rat cultured hippocampal neurons, 25 μM glutamate produced DCD accompanied by sustained increase in cytosolic Na+ ([Na+]c) and plasma membrane depolarization. MK801 and memantine, noncompetitive NMDAR inhibitors, added shortly after glutamate, completely prevented DCD whereas AP-5, a competitive NMDAR inhibitor, failed to protect against DCD. None of the tested inhibitors lowered elevated [Na+]c or restored plasma membrane potential. In the experiments with NCX reversal by gramicidin, MK801 and memantine robustly inhibited NCXrev while AP-5 was much less efficacious. In electrophysiological patch-clamp experiments MK801 and memantine inhibited NCXrev-mediated ion currents whereas AP-5 failed. Thus, MK801 and memantine, in addition to NMDAR, inhibited NCXrev. Inhibition of NCXrev either with KB-R7943, or by collapsing Na+ gradient across the plasma membrane, or by inhibiting Na+/H+ exchanger with 5-(N-ethyl-N-isopropyl) amiloride (EIPA) and thus preventing the increase in [Na+]c failed to preclude DCD. However, NCXrev inhibition combined with NMDAR blockade by AP-5 completely prevented DCD. Overall, our data suggest that both NMDAR and NCXrev are essential for DCD in glutamate-exposed neurons and inhibition of individual mechanism is not sufficient to prevent calcium dysregulation.
Collapse
Affiliation(s)
- Matthew K Brittain
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Dolga AM, Terpolilli N, Kepura F, Nijholt IM, Knaus HG, D'Orsi B, Prehn JHM, Eisel ULM, Plant T, Plesnila N, Culmsee C. KCa2 channels activation prevents [Ca2+]i deregulation and reduces neuronal death following glutamate toxicity and cerebral ischemia. Cell Death Dis 2011; 2:e147. [PMID: 21509037 PMCID: PMC3122061 DOI: 10.1038/cddis.2011.30] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Exacerbated activation of glutamate receptor-coupled calcium channels and subsequent increase in intracellular calcium ([Ca2+]i) are established hallmarks of neuronal cell death in acute and chronic neurological diseases. Here we show that pathological [Ca2+]i deregulation occurring after glutamate receptor stimulation is effectively modulated by small conductance calcium-activated potassium (KCa2) channels. We found that neuronal excitotoxicity was associated with a rapid downregulation of KCa2.2 channels within 3 h after the onset of glutamate exposure. Activation of KCa2 channels preserved KCa2 expression and significantly reduced pathological increases in [Ca2+]i providing robust neuroprotection in vitro and in vivo. These data suggest a critical role for KCa2 channels in excitotoxic neuronal cell death and propose their activation as potential therapeutic strategy for the treatment of acute and chronic neurodegenerative disorders.
Collapse
Affiliation(s)
- A M Dolga
- Institut für Pharmakologie und Klinische Pharmazie, Fachbereich Pharmazie, Philipps-Universität Marburg, Marburg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Endocrine pituitary cells are neuronlike; they express numerous voltage-gated sodium, calcium, potassium, and chloride channels and fire action potentials spontaneously, accompanied by a rise in intracellular calcium. In some cells, spontaneous electrical activity is sufficient to drive the intracellular calcium concentration above the threshold for stimulus-secretion and stimulus-transcription coupling. In others, the function of these action potentials is to maintain the cells in a responsive state with cytosolic calcium near, but below, the threshold level. Some pituitary cells also express gap junction channels, which could be used for intercellular Ca(2+) signaling in these cells. Endocrine cells also express extracellular ligand-gated ion channels, and their activation by hypothalamic and intrapituitary hormones leads to amplification of the pacemaking activity and facilitation of calcium influx and hormone release. These cells also express numerous G protein-coupled receptors, which can stimulate or silence electrical activity and action potential-dependent calcium influx and hormone release. Other members of this receptor family can activate calcium channels in the endoplasmic reticulum, leading to a cell type-specific modulation of electrical activity. This review summarizes recent findings in this field and our current understanding of the complex relationship between voltage-gated ion channels, ligand-gated ion channels, gap junction channels, and G protein-coupled receptors in pituitary cells.
Collapse
Affiliation(s)
- Stanko S Stojilkovic
- Program in Developmental Neuroscience, National Institute of Child Health and Human Development, National Institutes of Health, Building 49, Room 6A-36, 49 Convent Drive, Bethesda, Maryland 20892-4510, USA.
| | | | | |
Collapse
|
43
|
Zhdanov AV, Dmitriev RI, Papkovsky DB. Bafilomycin A1 activates respiration of neuronal cells via uncoupling associated with flickering depolarization of mitochondria. Cell Mol Life Sci 2010; 68:903-17. [PMID: 20820851 PMCID: PMC3037485 DOI: 10.1007/s00018-010-0502-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2010] [Revised: 07/08/2010] [Accepted: 08/06/2010] [Indexed: 10/27/2022]
Abstract
Bafilomycin A1 (Baf) induces an elevation of cytosolic Ca(2+) and acidification in neuronal cells via inhibition of the V-ATPase. Also, Baf uncouples mitochondria in differentiated PC12 ((d)PC12), (d)SH-SY5Y cells and cerebellar granule neurons, and markedly elevates their respiration. This respiratory response in (d)PC12 is accompanied by morphological changes in the mitochondria and decreases the mitochondrial pH, Ca(2+) and ΔΨm. The response to Baf is regulated by cytosolic Ca(2+) fluxes from the endoplasmic reticulum. Inhibition of permeability transition pore opening increases the depolarizing effect of Baf on the ΔΨm. Baf induces stochastic flickering of the ΔΨm with a period of 20 ± 10 s. Under conditions of suppressed ATP production by glycolysis, oxidative phosphorylation impaired by Baf does not provide cells with sufficient ATP levels. Cells treated with Baf become more susceptible to excitation with KCl. Such mitochondrial uncoupling may play a role in a number of (patho)physiological conditions induced by Baf.
Collapse
Affiliation(s)
- Alexander V Zhdanov
- Biochemistry Department, University College Cork, Cavanagh Pharmacy Building, College Road, Cork, Republic of Ireland.
| | | | | |
Collapse
|
44
|
Calcium signaling in intact dorsal root ganglia: new observations and the effect of injury. Anesthesiology 2010; 113:134-46. [PMID: 20526180 DOI: 10.1097/aln.0b013e3181e0ef3f] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Ca is the dominant second messenger in primary sensory neurons. In addition, disrupted Ca signaling is a prominent feature in pain models involving peripheral nerve injury. Standard cytoplasmic Ca recording techniques use high K or field stimulation and dissociated neurons. To compare findings in intact dorsal root ganglia, we used a method of simultaneous electrophysiologic and microfluorimetric recording. METHODS Dissociated neurons were loaded by bath-applied Fura-2-AM and subjected to field stimulation. Alternatively, we adapted a technique in which neuronal somata of intact ganglia were loaded with Fura-2 through an intracellular microelectrode that provided simultaneous membrane potential recording during activation by action potentials (APs) conducted from attached dorsal roots. RESULTS Field stimulation at levels necessary to activate neurons generated bath pH changes through electrolysis and failed to predictably drive neurons with AP trains. In the intact ganglion technique, single APs produced measurable Ca transients that were fourfold larger in presumed nociceptive C-type neurons than in nonnociceptive Abeta-type neurons. Unitary Ca transients summated during AP trains, forming transients with amplitudes that were highly dependent on stimulation frequency. Each neuron was tuned to a preferred frequency at which transient amplitude was maximal. Transients predominantly exhibited monoexponential recovery and had sustained plateaus during recovery only with trains of more than 100 APs. Nerve injury decreased Ca transients in C-type neurons, but increased transients in Abeta-type neurons. CONCLUSIONS Refined observation of Ca signaling is possible through natural activation by conducted APs in undissociated sensory neurons and reveals features distinct to neuronal types and injury state.
Collapse
|
45
|
The dynamics of mitochondrial Ca2+ fluxes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2010; 1797:1727-35. [PMID: 20599532 DOI: 10.1016/j.bbabio.2010.06.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 06/10/2010] [Accepted: 06/17/2010] [Indexed: 01/30/2023]
Abstract
We have investigated the kinetics of mitochondrial Ca(2+) influx and efflux and their dependence on cytosolic [Ca(2+)] and [Na(+)] using low-Ca(2+)-affinity aequorin. The rate of Ca(2+) release from mitochondria increased linearly with mitochondrial [Ca(2+)] ([Ca(2+)](M)). Na(+)-dependent Ca(2+) release was predominant al low [Ca(2+)](M) but saturated at [Ca(2+)](M) around 400muM, while Na(+)-independent Ca(2+) release was very slow at [Ca(2+)](M) below 200muM, and then increased at higher [Ca(2+)](M), perhaps through the opening of a new pathway. Half-maximal activation of Na(+)-dependent Ca(2+) release occurred at 5-10mM [Na(+)], within the physiological range of cytosolic [Na(+)]. Ca(2+) entry rates were comparable in size to Ca(2+) exit rates at cytosolic [Ca(2+)] ([Ca(2+)](c)) below 7muM, but the rate of uptake was dramatically accelerated at higher [Ca(2+)](c). As a consequence, the presence of [Na(+)] considerably reduced the rate of [Ca(2+)](M) increase at [Ca(2+)](c) below 7muM, but its effect was hardly appreciable at 10muM [Ca(2+)](c). Exit rates were more dependent on the temperature than uptake rates, thus making the [Ca(2+)](M) transients to be much more prolonged at lower temperature. Our kinetic data suggest that mitochondria have little high affinity Ca(2+) buffering, and comparison of our results with data on total mitochondrial Ca(2+) fluxes indicate that the mitochondrial Ca(2+) bound/Ca(2+) free ratio is around 10- to 100-fold for most of the observed [Ca(2+)](M) range and suggest that massive phosphate precipitation can only occur when [Ca(2+)](M) reaches the millimolar range.
Collapse
|
46
|
Brustovetsky T, Bolshakov A, Brustovetsky N. Calpain activation and Na+/Ca2+ exchanger degradation occur downstream of calcium deregulation in hippocampal neurons exposed to excitotoxic glutamate. J Neurosci Res 2010; 88:1317-28. [PMID: 19937813 PMCID: PMC2830319 DOI: 10.1002/jnr.22295] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Delayed calcium deregulation (DCD) plays an essential role in glutamate excitotoxicity, a major detrimental factor in stroke, traumatic brain injury, and various neurodegenerations. In the present study, we examined the role of calpain activation and Na(+)/Ca(2+) exchanger (NCX) degradation in DCD and excitotoxic cell death in cultured hippocampal neurons. Exposure of neurons to glutamate caused DCD accompanied by secondary mitochondrial depolarization. Activation of calpain was evidenced by detecting NCX isoform 3 (NCX3) degradation products. Degradation of NCX isoform 1 (NCX1) was below the detection limit of Western blotting. Degradation of NCX3 was detected only after 1 hr of incubation with glutamate, whereas DCD occurred on average within 15 min after glutamate application. Calpeptin, an inhibitor of calpain, significantly attenuated NCX3 degradation but failed to inhibit DCD and excitotoxic neuronal death. Calpain inhibitors I, III, and VI also failed to influence DCD and glutamate-induced neuronal death. On the other hand, MK801, an inhibitor of the NMDA subtype of glutamate receptors, added shortly after the initial glutamate-induced jump in cytosolic Ca(2+), completely prevented DCD and activation of calpain and strongly protected neurons against excitotoxicity. Taken together, our results suggest that, in glutamate-treated hippocampal neurons, the initial increase in cytosolic Ca(2+) that precedes DCD is insufficient for sustained calpain activation, which most likely occurs downstream of DCD.
Collapse
Affiliation(s)
- Tatiana Brustovetsky
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Alexey Bolshakov
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Nickolay Brustovetsky
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
47
|
Mechanisms of axonal injury: internodal nanocomplexes and calcium deregulation. Trends Mol Med 2010; 16:160-70. [PMID: 20207196 DOI: 10.1016/j.molmed.2010.02.002] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2009] [Revised: 02/01/2010] [Accepted: 02/03/2010] [Indexed: 12/14/2022]
Abstract
Axonal degeneration causes morbidity in many neurological conditions including stroke, neurotrauma and multiple sclerosis. The limited ability of central nervous system (CNS) neurons to regenerate, combined with the observation that axonal damage causes clinical disability, has spurred efforts to investigate the mechanisms of axonal degeneration. Ca influx from outside the axon is a key mediator of injury. More recently, substantial pools of intra-axonal Ca sequestered in the 'axoplasmic reticulum' have been reported. These Ca stores are under the control of multimolecular 'nanocomplexes' located along the internodes under the myelin. The overactivation of these complexes during disease can lead to a lethal release of Ca from intra-axonal stores. Rich receptor pharmacology offers tantalizing therapeutic options targeting these nanocomplexes in the many diseases where axonal degeneration is prominent.
Collapse
|
48
|
Scullin CS, Partridge LD. Contributions of SERCA pump and ryanodine-sensitive stores to presynaptic residual Ca2+. Cell Calcium 2010; 47:326-38. [PMID: 20153896 DOI: 10.1016/j.ceca.2010.01.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2009] [Revised: 12/31/2009] [Accepted: 01/20/2010] [Indexed: 11/24/2022]
Abstract
The presynaptic Ca2+ signal, which triggers vesicle release, disperses to a broadly distributed residual [Ca2+] ([Ca2+](res)) that plays an important role in synaptic plasticity. We have previously reported a slowing in the decay timecourse of [Ca2+](res) during the second of paired pulses. In this study, we investigated the contributions of organelle and plasma membrane Ca2+ flux pathways to the reduction of effectiveness of [Ca2+](res) clearance during short-term plasticity in Schaffer collateral terminals in the CA1 field of the hippocampus. We show that the slowed decay timecourse is mainly the result of a transport-dependent Ca2+ clearance process; that presynaptic caffeine-sensitive Ca2+ stores are not functionally loaded in the unstimulated terminal, but that these stores can effectively take up Ca2+ even during high frequency trains of stimuli; and that a rate limiting step of sarco-endoplasmic reticulum Ca2+-ATPase (SERCA) kinetics following the first pulse is responsible for a large portion of the observed slowing of [Ca2+](res) clearance during the second pulse. We were able to accurately fit our [Ca2+](res) data with a kinetic model based on these observations and this model predicted a reduction in availability of unbound SERCA during paired pulses, but no saturation of Ca2+ buffer in the endoplasmic reticulum.
Collapse
Affiliation(s)
- Chessa S Scullin
- Department of Neurosciences, University of New Mexico, School of Medicine, Albuquerque, NM 87131, USA
| | | |
Collapse
|
49
|
Gellerich FN, Gizatullina Z, Trumbeckaite S, Nguyen HP, Pallas T, Arandarcikaite O, Vielhaber S, Seppet E, Striggow F. The regulation of OXPHOS by extramitochondrial calcium. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2010; 1797:1018-27. [PMID: 20144582 DOI: 10.1016/j.bbabio.2010.02.005] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Revised: 02/01/2010] [Accepted: 02/02/2010] [Indexed: 11/29/2022]
Abstract
Despite extensive research, the regulation of mitochondrial function is still not understood completely. Ample evidence shows that cytosolic Ca2+ has a strategic task in co-ordinating the cellular work load and the regeneration of ATP by mitochondria. Currently, the paradigmatic view is that Cacyt2+ taken up by the Ca2+ uniporter activates the matrix enzymes pyruvate dehydrogenase, alpha-ketoglutarate dehydrogenase and isocitrate dehydrogenase. However, we have recently found that Ca2+ regulates the glutamate-dependent state 3 respiration by the supply of glutamate to mitochondria via aralar, a mitochondrial glutamate/aspartate carrier. Since this activation is not affected by ruthenium red, glutamate transport into mitochondria is controlled exclusively by extramitochondrial Ca2+. Therefore, this discovery shows that besides intramitochondrial also extramitochondrial Ca2+ regulates oxidative phosphorylation. This new mechanism acts as a mitochondrial "gas pedal", supplying the OXPHOS with substrate on demand. These results are in line with recent findings of Satrustegui and Palmieri showing that aralar as part of the malate-aspartate shuttle is involved in the Ca2+-dependent transport of reducing hydrogen equivalents (from NADH) into mitochondria. This review summarises results and evidence as well as hypothetical interpretations of data supporting the view that at the surface of mitochondria different regulatory Ca2+-binding sites exist and can contribute to cellular energy homeostasis. Moreover, on the basis of our own data, we propose that these surface Ca2+-binding sites may act as targets for neurotoxic proteins such as mutated huntingtin and others. The binding of these proteins to Ca2+-binding sites can impair the regulation by Ca2+, causing energetic depression and neurodegeneration.
Collapse
Affiliation(s)
- Frank N Gellerich
- KeyNeurotek Pharmaceuticals AG, ZENIT Technology Park, Leipziger Str. 44, D-39120 Magdeburg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Acid-evoked Ca2+ signalling in rat sensory neurones: effects of anoxia and aglycaemia. Pflugers Arch 2010; 459:159-81. [PMID: 19806360 PMCID: PMC2765625 DOI: 10.1007/s00424-009-0715-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2008] [Revised: 07/30/2009] [Accepted: 08/14/2009] [Indexed: 12/11/2022]
Abstract
Ischaemia excites sensory neurones (generating pain) and promotes calcitonin gene-related peptide release from nerve endings. Acidosis is thought to play a key role in mediating excitation via the activation of proton-sensitive cation channels. In this study, we investigated the effects of acidosis upon Ca2+ signalling in sensory neurones from rat dorsal root ganglia. Both hypercapnic (pHo 6.8) and metabolic–hypercapnic (pHo 6.2) acidosis caused a biphasic increase in cytosolic calcium concentration ([Ca2+]i). This comprised a brief Ca2+ transient (half-time approximately 30 s) caused by Ca2+ influx followed by a sustained rise in [Ca2+]i due to Ca2+ release from caffeine and cyclopiazonic acid-sensitive internal stores. Acid-evoked Ca2+ influx was unaffected by voltage-gated Ca2+-channel inhibition with nickel and acid sensing ion channel (ASIC) inhibition with amiloride but was blocked by inhibition of transient receptor potential vanilloid receptors (TRPV1) with (E)-3-(4-t-butylphenyl)-N-(2,3-dihydrobenzo[b][1,4] dioxin-6-yl)acrylamide (AMG 9810; 1 μM) and N-(4-tertiarybutylphenyl)-4-(3-cholorphyridin-2-yl) tetrahydropryazine-1(2H)-carbox-amide (BCTC; 1 μM). Combining acidosis with anoxia and aglycaemia increased the amplitude of both phases of Ca2+ elevation and prolonged the Ca2+ transient. The Ca2+ transient evoked by combined acidosis, aglycaemia and anoxia was also substantially blocked by AMG 9810 and BCTC and, to a lesser extent, by amiloride. In summary, the principle mechanisms mediating increase in [Ca2+]i in response to acidosis are a brief Ca2+ influx through TRPV1 followed by sustained Ca2+ release from internal stores. These effects are potentiated by anoxia and aglycaemia, conditions also prevalent in ischaemia. The effects of anoxia and aglycaemia are suggested to be largely due to the inhibition of Ca2+-clearance mechanisms and possible increase in the role of ASICs.
Collapse
|