1
|
Wang J, Jiang J, Hu H, Chen L. MCU complex: Exploring emerging targets and mechanisms of mitochondrial physiology and pathology. J Adv Res 2025; 68:271-298. [PMID: 38417574 PMCID: PMC11785567 DOI: 10.1016/j.jare.2024.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/16/2024] [Accepted: 02/17/2024] [Indexed: 03/01/2024] Open
Abstract
BACKGROUND Globally, the onset and progression of multiple human diseases are associated with mitochondrial dysfunction and dysregulation of Ca2+ uptake dynamics mediated by the mitochondrial calcium uniporter (MCU) complex, which plays a key role in mitochondrial dysfunction. Despite relevant studies, the underlying pathophysiological mechanisms have not yet been fully elucidated. AIM OF REVIEW This article provides an in-depth analysis of the current research status of the MCU complex, focusing on its molecular composition, regulatory mechanisms, and association with diseases. In addition, we conducted an in-depth analysis of the regulatory effects of agonists, inhibitors, and traditional Chinese medicine (TCM) monomers on the MCU complex and their application prospects in disease treatment. From the perspective of medicinal chemistry, we conducted an in-depth analysis of the structure-activity relationship between these small molecules and MCU and deduced potential pharmacophores and binding pockets. Simultaneously, key structural domains of the MCU complex in Homo sapiens were identified. We also studied the functional expression of the MCU complex in Drosophila, Zebrafish, and Caenorhabditis elegans. These analyses provide a basis for exploring potential treatment strategies targeting the MCU complex and provide strong support for the development of future precision medicine and treatments. KEY SCIENTIFIC CONCEPTS OF REVIEW The MCU complex exhibits varying behavior across different tissues and plays various roles in metabolic functions. It consists of six MCU subunits, an essential MCU regulator (EMRE), and solute carrier 25A23 (SLC25A23). They regulate processes, such as mitochondrial Ca2+ (mCa2+) uptake, mitochondrial adenosine triphosphate (ATP) production, calcium dynamics, oxidative stress (OS), and cell death. Regulation makes it a potential target for treating diseases, especially cardiovascular diseases, neurodegenerative diseases, inflammatory diseases, metabolic diseases, and tumors.
Collapse
Affiliation(s)
- Jin Wang
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical College, University of South China, Hengyang 421001, China
| | - Jinyong Jiang
- Department of Pharmacy, The First Affiliated Hospital of Jishou University, Jishou 416000, China
| | - Haoliang Hu
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical College, University of South China, Hengyang 421001, China; College of Medicine, Hunan University of Arts and Science, Changde 415000, China.
| | - Linxi Chen
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical College, University of South China, Hengyang 421001, China.
| |
Collapse
|
2
|
Boto T, Tomchik SM. Functional Imaging of Learning-Induced Plasticity in the Central Nervous System with Genetically Encoded Reporters in Drosophila. Cold Spring Harb Protoc 2024; 2024:pdb.top107799. [PMID: 37197830 DOI: 10.1101/pdb.top107799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Learning and memory allow animals to adjust their behavior based on the predictive value of their past experiences. Memories often exist in complex representations, spread across numerous cells and synapses in the brain. Studying relatively simple forms of memory provides insights into the fundamental processes that underlie multiple forms of memory. Associative learning occurs when an animal learns the relationship between two previously unrelated sensory stimuli, such as when a hungry animal learns that a particular odor is followed by a tasty reward. Drosophila is a particularly powerful model to study how this type of memory works. The fundamental principles are widely shared among animals, and there is a wide range of genetic tools available to study circuit function in flies. In addition, the olfactory structures that mediate associative learning in flies, such as the mushroom body and its associated neurons, are anatomically organized, relatively well-characterized, and readily accessible to imaging. Here, we review the olfactory anatomy and physiology of the olfactory system, describe how plasticity in the olfactory pathway mediates learning and memory, and explain the general principles underlying calcium imaging approaches.
Collapse
Affiliation(s)
- Tamara Boto
- Department of Physiology, Trinity College Dublin, Dublin 2, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Seth M Tomchik
- Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA
- Stead Family Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA
| |
Collapse
|
3
|
Rose HM, Ferrán B, Ranjit R, Masingale AM, Owen DB, Hussong S, Kinter MT, Galvan V, Logan S, Díaz-García CM. Mitochondrial calcium uniporter deficiency in dentate granule cells remodels neuronal metabolism and impairs reversal learning. J Neurochem 2024; 168:592-607. [PMID: 37415312 PMCID: PMC10770303 DOI: 10.1111/jnc.15901] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/27/2023] [Accepted: 06/12/2023] [Indexed: 07/08/2023]
Abstract
The mitochondrial calcium uniporter (MCU) is the main route of calcium (Ca2+) entry into neuronal mitochondria. This channel has been linked to mitochondrial Ca2+ overload and cell death under neurotoxic conditions, but its physiologic roles for normal brain function remain poorly understood. Despite high expression of MCU in excitatory hippocampal neurons, it is unknown whether this channel is required for learning and memory. Here, we genetically down-regulated the Mcu gene in dentate granule cells (DGCs) of the hippocampus and found that this manipulation increases the overall respiratory activity of mitochondrial complexes I and II, augmenting the generation of reactive oxygen species in the context of impaired electron transport chain. The metabolic remodeling of MCU-deficient neurons also involved changes in the expression of enzymes that participate in glycolysis and the regulation of the tricarboxylic acid cycle, as well as the cellular antioxidant defenses. We found that MCU deficiency in DGCs does not change circadian rhythms, spontaneous exploratory behavior, or cognitive function in middle-aged mice (11-13 months old), when assessed with a food-motivated working memory test with three choices. DGC-targeted down-regulation of MCU significantly impairs reversal learning assessed with an 8-arm radial arm water maze but does not affect their ability to learn the task for the first time. Our results indicate that neuronal MCU plays an important physiologic role in memory formation and may be a potential therapeutic target to develop interventions aimed at improving cognitive function in aging, neurodegenerative diseases, and brain injury.
Collapse
Affiliation(s)
- Hadyn M Rose
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Center for Geroscience and Healthy Brain Aging, Oklahoma City, Oklahoma, USA
| | - Beatriz Ferrán
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Center for Geroscience and Healthy Brain Aging, Oklahoma City, Oklahoma, USA
| | - Rojina Ranjit
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Center for Geroscience and Healthy Brain Aging, Oklahoma City, Oklahoma, USA
| | - Anthony M Masingale
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Center for Geroscience and Healthy Brain Aging, Oklahoma City, Oklahoma, USA
| | - Daniel B Owen
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Center for Geroscience and Healthy Brain Aging, Oklahoma City, Oklahoma, USA
| | - Stacy Hussong
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Oklahoma City Veterans Health Care System, Oklahoma City, Oklahoma, USA
| | - Michael T Kinter
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Veronica Galvan
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Center for Geroscience and Healthy Brain Aging, Oklahoma City, Oklahoma, USA
- Oklahoma City Veterans Health Care System, Oklahoma City, Oklahoma, USA
| | - Sreemathi Logan
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Center for Geroscience and Healthy Brain Aging, Oklahoma City, Oklahoma, USA
| | - Carlos Manlio Díaz-García
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Center for Geroscience and Healthy Brain Aging, Oklahoma City, Oklahoma, USA
| |
Collapse
|
4
|
Petersen EN, Pavel MA, Hansen SS, Gudheti M, Wang H, Yuan Z, Murphy KR, Ja W, Ferris HA, Jorgensen E, Hansen SB. Mechanical activation of TWIK-related potassium channel by nanoscopic movement and rapid second messenger signaling. eLife 2024; 12:RP89465. [PMID: 38407149 PMCID: PMC10942622 DOI: 10.7554/elife.89465] [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] [Indexed: 02/27/2024] Open
Abstract
Rapid conversion of force into a biological signal enables living cells to respond to mechanical forces in their environment. The force is believed to initially affect the plasma membrane and then alter the behavior of membrane proteins. Phospholipase D2 (PLD2) is a mechanosensitive enzyme that is regulated by a structured membrane-lipid site comprised of cholesterol and saturated ganglioside (GM1). Here we show stretch activation of TWIK-related K+ channel (TREK-1) is mechanically evoked by PLD2 and spatial patterning involving ordered GM1 and 4,5-bisphosphate (PIP2) clusters in mammalian cells. First, mechanical force deforms the ordered lipids, which disrupts the interaction of PLD2 with the GM1 lipids and allows a complex of TREK-1 and PLD2 to associate with PIP2 clusters. The association with PIP2 activates the enzyme, which produces the second messenger phosphatidic acid (PA) that gates the channel. Co-expression of catalytically inactive PLD2 inhibits TREK-1 stretch currents in a biological membrane. Cellular uptake of cholesterol inhibits TREK-1 currents in culture and depletion of cholesterol from astrocytes releases TREK-1 from GM1 lipids in mouse brain. Depletion of the PLD2 ortholog in flies results in hypersensitivity to mechanical force. We conclude PLD2 mechanosensitivity combines with TREK-1 ion permeability to elicit a mechanically evoked response.
Collapse
Affiliation(s)
- E Nicholas Petersen
- Departments of Molecular Medicine, The Scripps Research Institute, ScrippsJupiterUnited States
- Scripps Research Skaggs Graduate School of Chemical and Biological Science, The Scripps Research Institute, Scripps,JupiterUnited States
| | - Mahmud Arif Pavel
- Departments of Molecular Medicine, The Scripps Research Institute, ScrippsJupiterUnited States
| | - Samuel S Hansen
- Departments of Molecular Medicine, The Scripps Research Institute, ScrippsJupiterUnited States
| | - Manasa Gudheti
- Division of Endocrinology and Metabolism, Center for Brain Immunology and Glia, Department of Neuroscience, University of VirginiaCharlottesvilleUnited States
| | - Hao Wang
- Departments of Molecular Medicine, The Scripps Research Institute, ScrippsJupiterUnited States
- Scripps Research Skaggs Graduate School of Chemical and Biological Science, The Scripps Research Institute, Scripps,JupiterUnited States
| | - Zixuan Yuan
- Departments of Molecular Medicine, The Scripps Research Institute, ScrippsJupiterUnited States
- Scripps Research Skaggs Graduate School of Chemical and Biological Science, The Scripps Research Institute, Scripps,JupiterUnited States
| | - Keith R Murphy
- Department of Neuroscience, The Scripps Research Institute, ScrippsJupiterUnited States
- Center on Aging,The Scripps Research Institute, ScrippsJupiterUnited States
| | - William Ja
- Department of Neuroscience, The Scripps Research Institute, ScrippsJupiterUnited States
- Center on Aging,The Scripps Research Institute, ScrippsJupiterUnited States
| | - Heather A Ferris
- Division of Endocrinology and Metabolism, Center for Brain Immunology and Glia, Department of Neuroscience, University of VirginiaCharlottesvilleUnited States
| | - Erik Jorgensen
- Department of Biology, Howard Hughes Medical Institute, University of UtahSalt Lake CityUnited States
| | - Scott B Hansen
- Departments of Molecular Medicine, The Scripps Research Institute, ScrippsJupiterUnited States
| |
Collapse
|
5
|
Davis RL. Learning and memory using Drosophila melanogaster: a focus on advances made in the fifth decade of research. Genetics 2023; 224:iyad085. [PMID: 37212449 PMCID: PMC10411608 DOI: 10.1093/genetics/iyad085] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 05/03/2023] [Indexed: 05/23/2023] Open
Abstract
In the last decade, researchers using Drosophila melanogaster have made extraordinary progress in uncovering the mysteries underlying learning and memory. This progress has been propelled by the amazing toolkit available that affords combined behavioral, molecular, electrophysiological, and systems neuroscience approaches. The arduous reconstruction of electron microscopic images resulted in a first-generation connectome of the adult and larval brain, revealing complex structural interconnections between memory-related neurons. This serves as substrate for future investigations on these connections and for building complete circuits from sensory cue detection to changes in motor behavior. Mushroom body output neurons (MBOn) were discovered, which individually forward information from discrete and non-overlapping compartments of the axons of mushroom body neurons (MBn). These neurons mirror the previously discovered tiling of mushroom body axons by inputs from dopamine neurons and have led to a model that ascribes the valence of the learning event, either appetitive or aversive, to the activity of different populations of dopamine neurons and the balance of MBOn activity in promoting avoidance or approach behavior. Studies of the calyx, which houses the MBn dendrites, have revealed a beautiful microglomeruluar organization and structural changes of synapses that occur with long-term memory (LTM) formation. Larval learning has advanced, positioning it to possibly lead in producing new conceptual insights due to its markedly simpler structure over the adult brain. Advances were made in how cAMP response element-binding protein interacts with protein kinases and other transcription factors to promote the formation of LTM. New insights were made on Orb2, a prion-like protein that forms oligomers to enhance synaptic protein synthesis required for LTM formation. Finally, Drosophila research has pioneered our understanding of the mechanisms that mediate permanent and transient active forgetting, an important function of the brain along with acquisition, consolidation, and retrieval. This was catalyzed partly by the identification of memory suppressor genes-genes whose normal function is to limit memory formation.
Collapse
Affiliation(s)
- Ronald L Davis
- Department of Neuroscience, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, 130 Scripps Way, Jupiter, FL 33458, USA
| |
Collapse
|
6
|
Duan W, Liu C, Zhou J, Yu Q, Duan Y, Zhang T, Li Y, Fu G, Sun Y, Tian J, Xia Z, Yang Y, Liu Y, Xu S. Upregulation of mitochondrial calcium uniporter contributes to paraquat-induced neuropathology linked to Parkinson's disease via imbalanced OPA1 processing. JOURNAL OF HAZARDOUS MATERIALS 2023; 453:131369. [PMID: 37086674 DOI: 10.1016/j.jhazmat.2023.131369] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/18/2023] [Accepted: 04/03/2023] [Indexed: 05/03/2023]
Abstract
Paraquat (PQ) is the most widely used herbicide in agriculture worldwide and has been considered a high-risk environmental factor for Parkinson's disease (PD). Chronic PQ exposure selectively induces dopaminergic neuron loss, the hallmark pathologic feature of PD, resulting in Parkinson-like movement disorders. However, the underlying mechanisms remain unclear. Here, we demonstrated that repetitive PQ exposure caused dopaminergic neuron loss, dopamine deficiency and motor deficits dose-dependently in mice. Accordingly, mitochondrial calcium uniporter (MCU) was highly expressed in PQ-exposed mice and neuronal cells. Importantly, MCU knockout (KO) effectively rescued PQ-induced dopaminergic neuron loss and motor deficits in mice. Genetic and pharmacological inhibition of MCU alleviated PQ-induced mitochondrial dysfunction and neuronal death in vitro. Mechanistically, PQ exposure triggered mitochondrial fragmentation via imbalance of the optic atrophy 1 (OPA1) processing manifested by cleavage of L-OPA1 to S-OPA1, which was reversed by inhibition of MCU. Notably, the upregulation of MCU was mediated by miR-129-1-3p posttranscriptionally, and overexpression of miR-129-1-3p could rebalance OPA1 processing and attenuate mitochondrial dysfunction and neuronal death induced by PQ exposure. Consequently, our work uncovers an essential role of MCU and a novel molecular mechanism, miR-MCU-OPA1, in PQ-induced pathogenesis of PD, providing a potential target and strategy for environmental neurotoxins-induced PD treatment.
Collapse
Affiliation(s)
- Weixia Duan
- Center of Laboratory Medicine, Chongqing Prevention and Treatment Center for Occupational Diseases, Chongqing 400060, China; Chongqing Key Laboratory of Prevention and Treatment for Occupational Diseases and Poisoning, Chongqing 400060, China
| | - Cong Liu
- Center of Laboratory Medicine, Chongqing Prevention and Treatment Center for Occupational Diseases, Chongqing 400060, China; Chongqing Key Laboratory of Prevention and Treatment for Occupational Diseases and Poisoning, Chongqing 400060, China
| | - Jie Zhou
- Center of Laboratory Medicine, Chongqing Prevention and Treatment Center for Occupational Diseases, Chongqing 400060, China; Chongqing Key Laboratory of Prevention and Treatment for Occupational Diseases and Poisoning, Chongqing 400060, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Qin Yu
- Center of Laboratory Medicine, Chongqing Prevention and Treatment Center for Occupational Diseases, Chongqing 400060, China; Chongqing Key Laboratory of Prevention and Treatment for Occupational Diseases and Poisoning, Chongqing 400060, China
| | - Yu Duan
- Center of Laboratory Medicine, Chongqing Prevention and Treatment Center for Occupational Diseases, Chongqing 400060, China; Chongqing Key Laboratory of Prevention and Treatment for Occupational Diseases and Poisoning, Chongqing 400060, China
| | - Tian Zhang
- Center of Laboratory Medicine, Chongqing Prevention and Treatment Center for Occupational Diseases, Chongqing 400060, China; Chongqing Key Laboratory of Prevention and Treatment for Occupational Diseases and Poisoning, Chongqing 400060, China; Bioengineering College of Chongqing University, Chongqing 400044, China
| | - Yuanyuan Li
- Center of Laboratory Medicine, Chongqing Prevention and Treatment Center for Occupational Diseases, Chongqing 400060, China; Chongqing Key Laboratory of Prevention and Treatment for Occupational Diseases and Poisoning, Chongqing 400060, China
| | - Guanyan Fu
- Center of Laboratory Medicine, Chongqing Prevention and Treatment Center for Occupational Diseases, Chongqing 400060, China; Chongqing Key Laboratory of Prevention and Treatment for Occupational Diseases and Poisoning, Chongqing 400060, China
| | - Yapei Sun
- Center of Laboratory Medicine, Chongqing Prevention and Treatment Center for Occupational Diseases, Chongqing 400060, China; Chongqing Key Laboratory of Prevention and Treatment for Occupational Diseases and Poisoning, Chongqing 400060, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Jiacheng Tian
- Center of Laboratory Medicine, Chongqing Prevention and Treatment Center for Occupational Diseases, Chongqing 400060, China; Chongqing Key Laboratory of Prevention and Treatment for Occupational Diseases and Poisoning, Chongqing 400060, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Zhiqin Xia
- Center of Laboratory Medicine, Chongqing Prevention and Treatment Center for Occupational Diseases, Chongqing 400060, China; Chongqing Key Laboratory of Prevention and Treatment for Occupational Diseases and Poisoning, Chongqing 400060, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yingli Yang
- Center of Laboratory Medicine, Chongqing Prevention and Treatment Center for Occupational Diseases, Chongqing 400060, China; Chongqing Key Laboratory of Prevention and Treatment for Occupational Diseases and Poisoning, Chongqing 400060, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yongseng Liu
- Center of Laboratory Medicine, Chongqing Prevention and Treatment Center for Occupational Diseases, Chongqing 400060, China; Chongqing Key Laboratory of Prevention and Treatment for Occupational Diseases and Poisoning, Chongqing 400060, China
| | - Shangcheng Xu
- Center of Laboratory Medicine, Chongqing Prevention and Treatment Center for Occupational Diseases, Chongqing 400060, China; Chongqing Key Laboratory of Prevention and Treatment for Occupational Diseases and Poisoning, Chongqing 400060, China.
| |
Collapse
|
7
|
Skupio U, Welte J, Serrat R, Eraso-Pichot A, Julio-Kalajzić F, Gisquet D, Cannich A, Delcasso S, Matias I, Fundazuri UB, Pouvreau S, Pagano Zottola AC, Lavanco G, Drago F, Ruiz de Azua I, Lutz B, Bellocchio L, Busquets-Garcia A, Chaouloff F, Marsicano G. Mitochondrial cannabinoid receptors gate corticosterone impact on novel object recognition. Neuron 2023; 111:1887-1897.e6. [PMID: 37098353 DOI: 10.1016/j.neuron.2023.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 02/21/2023] [Accepted: 03/30/2023] [Indexed: 04/27/2023]
Abstract
Corticosteroid-mediated stress responses require the activation of complex brain circuits involving mitochondrial activity, but the underlying cellular and molecular mechanisms are scantly known. The endocannabinoid system is implicated in stress coping, and it can directly regulate brain mitochondrial functions via type 1 cannabinoid (CB1) receptors associated with mitochondrial membranes (mtCB1). In this study, we show that the impairing effect of corticosterone in the novel object recognition (NOR) task in mice requires mtCB1 receptors and the regulation of mitochondrial calcium levels in neurons. Different brain circuits are modulated by this mechanism to mediate the impact of corticosterone during specific phases of the task. Thus, whereas corticosterone recruits mtCB1 receptors in noradrenergic neurons to impair NOR consolidation, mtCB1 receptors in local hippocampal GABAergic interneurons are required to inhibit NOR retrieval. These data reveal unforeseen mechanisms mediating the effects of corticosteroids during different phases of NOR, involving mitochondrial calcium alterations in different brain circuits.
Collapse
Affiliation(s)
- Urszula Skupio
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France
| | - Julia Welte
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France
| | - Roman Serrat
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France
| | - Abel Eraso-Pichot
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France
| | - Francisca Julio-Kalajzić
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France
| | - Doriane Gisquet
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France
| | - Astrid Cannich
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France
| | | | - Isabelle Matias
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France
| | - Unai B Fundazuri
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France
| | - Sandrine Pouvreau
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France
| | - Antonio C Pagano Zottola
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France; Institute for Cellular Biochemistry and Genetics, UMR 5095, Bordeaux 33077, France
| | - Gianluca Lavanco
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France
| | - Filippo Drago
- Department of Clinical and Molecular Biomedicine, Section of Pharmacology and Biochemistry, University of Catania, Catania, Italy
| | - Inigo Ruiz de Azua
- Institute of Physiological Chemistry, University Medical Center, Mainz 55131 Germany; Leibniz Institute for Resilience Research (LIR), Mainz 55122, Germany
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center, Mainz 55131 Germany; Leibniz Institute for Resilience Research (LIR), Mainz 55122, Germany
| | - Luigi Bellocchio
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France
| | - Arnau Busquets-Garcia
- Cell-Type Mechanisms in Normal and Pathological Behavior Research Group, Neuroscience Program, IMIM Hospital del Mar Medical Research Institute, Barcelona 08003, Spain
| | - Francis Chaouloff
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France
| | - Giovanni Marsicano
- INSERM, U1215 NeuroCentre Magendie, Bordeaux 33077, France; University of Bordeaux, Bordeaux 33077, France.
| |
Collapse
|
8
|
Walters GC, Usachev YM. Mitochondrial calcium cycling in neuronal function and neurodegeneration. Front Cell Dev Biol 2023; 11:1094356. [PMID: 36760367 PMCID: PMC9902777 DOI: 10.3389/fcell.2023.1094356] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/12/2023] [Indexed: 01/26/2023] Open
Abstract
Mitochondria are essential for proper cellular function through their critical roles in ATP synthesis, reactive oxygen species production, calcium (Ca2+) buffering, and apoptotic signaling. In neurons, Ca2+ buffering is particularly important as it helps to shape Ca2+ signals and to regulate numerous Ca2+-dependent functions including neuronal excitability, synaptic transmission, gene expression, and neuronal toxicity. Over the past decade, identification of the mitochondrial Ca2+ uniporter (MCU) and other molecular components of mitochondrial Ca2+ transport has provided insight into the roles that mitochondrial Ca2+ regulation plays in neuronal function in health and disease. In this review, we discuss the many roles of mitochondrial Ca2+ uptake and release mechanisms in normal neuronal function and highlight new insights into the Ca2+-dependent mechanisms that drive mitochondrial dysfunction in neurologic diseases including epilepsy, Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. We also consider how targeting Ca2+ uptake and release mechanisms could facilitate the development of novel therapeutic strategies for neurological diseases.
Collapse
Affiliation(s)
- Grant C. Walters
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, United States
| | - Yuriy M. Usachev
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
9
|
Kashif M, Sivaprakasam P, Vijendra P, Waseem M, Pandurangan AK. A Recent Update on Pathophysiology and Therapeutic Interventions of Alzheimer's Disease. Curr Pharm Des 2023; 29:3428-3441. [PMID: 38038007 DOI: 10.2174/0113816128264355231121064704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 10/09/2023] [Accepted: 10/19/2023] [Indexed: 12/02/2023]
Abstract
AIM Alzheimer's disease (AD) has been identified as a progressive brain disorder associated with memory dysfunction and the accumulation of β-amyloid plaques and neurofibrillary tangles of τ protein. Mitochondria is crucial in maintaining cell survival, cell death, calcium regulation, and ATP synthesis. Mitochondrial dysfunction and linked calcium overload have been involved in the pathogenesis of AD. CRM2 (Collapsin response mediator protein-2) is involved in endosomal lysosomal trafficking as well as autophagy, and their reduced level is also a primary culprit in the progression of AD. In addition, Cholinergic neurotransmission and neuroinflammation are two other mechanisms implicated in AD onset and might be protective targets to attenuate disease progression. The microbiota-gut-brain axis (MGBA) is another crucial target for AD treatment. Crosstalk between gut microbiota and brain mutually benefitted each other, dysbiosis in gut microbiota affects the brain functions and leads to AD progression with increased AD-causing biomarkers. Despite the complexity of AD, treatment is only limited to symptomatic management. Therefore, there is an urgent demand for novel therapeutics that target associated pathways responsible for AD pathology. This review explores the role of different mechanisms involved in AD and possible therapeutic targets to protect against disease progression. BACKGROUND Amidst various age-related diseases, AD is the most deleterious neurodegenerative disorder that affects more than 24 million people globally. Every year, approximately 7.7 million new cases of dementia have been reported. However, to date, no novel disease-modifying therapies are available to treat AD. OBJECTIVE The aim of writing this review is to highlight the role of key biomarker proteins and possible therapeutic interventions that could play a crucial role in mitigating the ongoing prognosis of Alzheimer's disease. MATERIALS AND METHODS The available information about the disease was collected through multiple search engines, including PubMed, Science Direct, Clinical Trials, and Google Scholar. RESULTS Accumulated pieces of evidence reveal that extracellular aggregation of β-amyloid plaques and intracellular tangles of τ protein are peculiar features of perpetuated Alzheimer's disease (AD). Further, the significant role of mitochondria, calcium, and cholinergic pathways in the pathogenesis of AD makes the respiratory cell organelle a crucial therapeutic target in this neurodegenerative disease. All currently available drugs either delay the clinical damage to cells or temporarily attenuate some symptoms of Alzheimer's disease. CONCLUSION The pathological features of AD are extracellular deposition of β-amyloid, acetylcholinesterase deregulation, and intracellular tangles of τ protein. The multifactorial heterogeneity of disease demands more research work in this field to find new therapeutic biological targets.
Collapse
Affiliation(s)
- Mohd Kashif
- School of Life Sciences, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai, India
| | - Prathibha Sivaprakasam
- School of Life Sciences, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai, India
| | - Poornima Vijendra
- Department of Studies in Food Technology, Davangere University, Davangere, Karnataka, India
| | - Mohammad Waseem
- Department of Pharmaceutical Science, University of Maryland, Eastern Shore, Baltimore, USA
| | - Ashok Kumar Pandurangan
- School of Life Sciences, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai, India
| |
Collapse
|
10
|
Delgado BD, Long SB. Mechanisms of ion selectivity and throughput in the mitochondrial calcium uniporter. SCIENCE ADVANCES 2022; 8:eade1516. [PMID: 36525497 PMCID: PMC9757755 DOI: 10.1126/sciadv.ade1516] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 11/17/2022] [Indexed: 06/17/2023]
Abstract
The mitochondrial calcium uniporter, which regulates aerobic metabolism by catalyzing mitochondrial Ca2+ influx, is arguably the most selective ion channel known. The mechanisms for this exquisite Ca2+ selectivity have not been defined. Here, using a reconstituted system, we study the electrical properties of the channel's minimal Ca2+-conducting complex, MCU-EMRE, from Tribolium castaneum to probe ion selectivity mechanisms. The wild-type TcMCU-EMRE complex recapitulates hallmark electrophysiological properties of endogenous Uniporter channels. Through interrogation of pore-lining mutants, we find that a ring of glutamate residues, the "E-locus," serves as the channel's selectivity filter. Unexpectedly, a nearby "D-locus" at the mouth of the pore has diminutive influence on selectivity. Anomalous mole fraction effects indicate that multiple Ca2+ ions are accommodated within the E-locus. By facilitating ion-ion interactions, the E-locus engenders both exquisite Ca2+ selectivity and high ion throughput. Direct comparison with structural information yields the basis for selective Ca2+ conduction by the channel.
Collapse
Affiliation(s)
- Bryce D. Delgado
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
- Graduate Program in Biochemistry and Structural Biology, Cell and Developmental Biology, and Molecular Biology, Weill Cornell Medicine Graduate School of Medical Sciences, New York, NY 10065, USA
| | - Stephen B. Long
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| |
Collapse
|
11
|
Bourouliti A, Skoulakis EMC. Anesthesia Resistant Memories in Drosophila, a Working Perspective. Int J Mol Sci 2022; 23:ijms23158527. [PMID: 35955662 PMCID: PMC9369046 DOI: 10.3390/ijms23158527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 12/10/2022] Open
Abstract
Memories are lasting representations over time of associations between stimuli or events. In general, the relatively slow consolidation of memories requires protein synthesis with a known exception being the so-called Anesthesia Resistant Memory (ARM) in Drosophila. This protein synthesis-independent memory type survives amnestic shocks after a short, sensitive window post training, and can also emerge after repeated cycles of training in a negatively reinforced olfactory conditioning task, without rest between cycles (massed conditioning—MC). We discussed operational and molecular mechanisms that mediate ARM and differentiate it from protein synthesis-dependent long-term memory (LTM) in Drosophila. Based on the notion that ARM is unlikely to specifically characterize Drosophila, we examined protein synthesis and MC-elicited memories in other species and based on intraspecies shared molecular components and proposed potential relationships of ARM with established memory types in Drosophila and vertebrates.
Collapse
Affiliation(s)
- Anna Bourouliti
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center “Alexander Fleming”, 16674 Vari, Greece;
- Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Efthimios M. C. Skoulakis
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center “Alexander Fleming”, 16674 Vari, Greece;
- Correspondence:
| |
Collapse
|
12
|
Cold Shock Disrupts Massed Training-Elicited Memory in Drosophila. Int J Mol Sci 2022; 23:ijms23126407. [PMID: 35742852 PMCID: PMC9224184 DOI: 10.3390/ijms23126407] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/03/2022] [Accepted: 06/06/2022] [Indexed: 02/04/2023] Open
Abstract
Memory consolidation is a time-dependent process occurring over hours, days, or longer in different species and requires protein synthesis. An apparent exception is a memory type in Drosophila elicited by a single olfactory conditioning episode, which ostensibly consolidates quickly, rendering it resistant to disruption by cold anesthesia a few hours post-training. This anesthesia-resistant memory (ARM), is independent of protein synthesis. Protein synthesis independent memory can also be elicited in Drosophila by multiple massed cycles of olfactory conditioning, and this led to the prevailing notion that both of these operationally distinct training regimes yield ARM. Significantly, we show that, unlike bona fide ARM, massed conditioning-elicited memory remains sensitive to the amnestic treatment two hours post-training and hence it is not ARM. Therefore, there are two protein synthesis-independent memory types in Drosophila.
Collapse
|
13
|
Mitochondrial calcium uniporter stabilization preserves energetic homeostasis during Complex I impairment. Nat Commun 2022; 13:2769. [PMID: 35589699 PMCID: PMC9120069 DOI: 10.1038/s41467-022-30236-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 04/20/2022] [Indexed: 12/15/2022] Open
Abstract
Calcium entering mitochondria potently stimulates ATP synthesis. Increases in calcium preserve energy synthesis in cardiomyopathies caused by mitochondrial dysfunction, and occur due to enhanced activity of the mitochondrial calcium uniporter channel. The signaling mechanism that mediates this compensatory increase remains unknown. Here, we find that increases in the uniporter are due to impairment in Complex I of the electron transport chain. In normal physiology, Complex I promotes uniporter degradation via an interaction with the uniporter pore-forming subunit, a process we term Complex I-induced protein turnover. When Complex I dysfunction ensues, contact with the uniporter is inhibited, preventing degradation, and leading to a build-up in functional channels. Preventing uniporter activity leads to early demise in Complex I-deficient animals. Conversely, enhancing uniporter stability rescues survival and function in Complex I deficiency. Taken together, our data identify a fundamental pathway producing compensatory increases in calcium influx during Complex I impairment.
Collapse
|
14
|
Garbincius JF, Elrod JW. Mitochondrial calcium exchange in physiology and disease. Physiol Rev 2022; 102:893-992. [PMID: 34698550 PMCID: PMC8816638 DOI: 10.1152/physrev.00041.2020] [Citation(s) in RCA: 204] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 08/16/2021] [Accepted: 10/19/2021] [Indexed: 12/13/2022] Open
Abstract
The uptake of calcium into and extrusion of calcium from the mitochondrial matrix is a fundamental biological process that has critical effects on cellular metabolism, signaling, and survival. Disruption of mitochondrial calcium (mCa2+) cycling is implicated in numerous acquired diseases such as heart failure, stroke, neurodegeneration, diabetes, and cancer and is genetically linked to several inherited neuromuscular disorders. Understanding the mechanisms responsible for mCa2+ exchange therefore holds great promise for the treatment of these diseases. The past decade has seen the genetic identification of many of the key proteins that mediate mitochondrial calcium uptake and efflux. Here, we present an overview of the phenomenon of mCa2+ transport and a comprehensive examination of the molecular machinery that mediates calcium flux across the inner mitochondrial membrane: the mitochondrial uniporter complex (consisting of MCU, EMRE, MICU1, MICU2, MICU3, MCUB, and MCUR1), NCLX, LETM1, the mitochondrial ryanodine receptor, and the mitochondrial permeability transition pore. We then consider the physiological implications of mCa2+ flux and evaluate how alterations in mCa2+ homeostasis contribute to human disease. This review concludes by highlighting opportunities and challenges for therapeutic intervention in pathologies characterized by aberrant mCa2+ handling and by summarizing critical unanswered questions regarding the biology of mCa2+ flux.
Collapse
Affiliation(s)
- Joanne F Garbincius
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - John W Elrod
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
15
|
Serrat R, Oliveira-Pinto A, Marsicano G, Pouvreau S. Imaging mitochondrial calcium dynamics in the central nervous system. J Neurosci Methods 2022; 373:109560. [PMID: 35320763 DOI: 10.1016/j.jneumeth.2022.109560] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 03/04/2022] [Accepted: 03/06/2022] [Indexed: 12/28/2022]
Abstract
Mitochondrial calcium handling is a particularly active research area in the neuroscience field, as it plays key roles in the regulation of several functions of the central nervous system, such as synaptic transmission and plasticity, astrocyte calcium signaling, neuronal activity… In the last few decades, a panel of techniques have been developed to measure mitochondrial calcium dynamics, relying mostly on photonic microscopy, and including synthetic sensors, hybrid sensors and genetically encoded calcium sensors. The goal of this review is to endow the reader with a deep knowledge of the historical and latest tools to monitor mitochondrial calcium events in the brain, as well as a comprehensive overview of the current state of the art in brain mitochondrial calcium signaling. We will discuss the main calcium probes used in the field, their mitochondrial targeting strategies, their key properties and major drawbacks. In addition, we will detail the main roles of mitochondrial calcium handling in neuronal tissues through an extended report of the recent studies using mitochondrial targeted calcium sensors in neuronal and astroglial cells, in vitro and in vivo.
Collapse
Affiliation(s)
- Roman Serrat
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1215 NeuroCentre Magendie, France; University of Bordeaux, Bordeaux 33077, France
| | - Alexandre Oliveira-Pinto
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1215 NeuroCentre Magendie, France; University of Bordeaux, Bordeaux 33077, France
| | - Giovanni Marsicano
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1215 NeuroCentre Magendie, France; University of Bordeaux, Bordeaux 33077, France
| | - Sandrine Pouvreau
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1215 NeuroCentre Magendie, France; University of Bordeaux, Bordeaux 33077, France.
| |
Collapse
|
16
|
Sapian S, Taib IS, Latip J, Katas H, Chin KY, Mohd Nor NA, Jubaidi FF, Budin SB. Therapeutic Approach of Flavonoid in Ameliorating Diabetic Cardiomyopathy by Targeting Mitochondrial-Induced Oxidative Stress. Int J Mol Sci 2021; 22:11616. [PMID: 34769045 PMCID: PMC8583796 DOI: 10.3390/ijms222111616] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/19/2021] [Accepted: 10/22/2021] [Indexed: 12/26/2022] Open
Abstract
Diabetes cardiomyopathy is one of the key factors of mortality among diabetic patients around the globe. One of the prior contributors to the progression of diabetic cardiomyopathy is cardiac mitochondrial dysfunction. The cardiac mitochondrial dysfunction can induce oxidative stress in cardiomyocytes and was found to be the cause of majority of the heart morphological and dynamical changes in diabetic cardiomyopathy. To slow down the occurrence of diabetic cardiomyopathy, it is crucial to discover therapeutic agents that target mitochondrial-induced oxidative stress. Flavonoid is a plentiful phytochemical in plants that shows a wide range of biological actions against human diseases. Flavonoids have been extensively documented for their ability to protect the heart from diabetic cardiomyopathy. Flavonoids' ability to alleviate diabetic cardiomyopathy is primarily attributed to their antioxidant properties. In this review, we present the mechanisms involved in flavonoid therapies in ameliorating mitochondrial-induced oxidative stress in diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Syaifuzah Sapian
- Centre for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (S.S.); (I.S.T.); (N.A.M.N.); (F.F.J.)
| | - Izatus Shima Taib
- Centre for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (S.S.); (I.S.T.); (N.A.M.N.); (F.F.J.)
| | - Jalifah Latip
- School of Chemical Sciences and Food Technology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi 46300, Malaysia;
| | - Haliza Katas
- Centre for Drug Delivery Research, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia;
| | - Kok-Yong Chin
- Department of Pharmacology, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur 56000, Malaysia;
| | - Nor Anizah Mohd Nor
- Centre for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (S.S.); (I.S.T.); (N.A.M.N.); (F.F.J.)
| | - Fatin Farhana Jubaidi
- Centre for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (S.S.); (I.S.T.); (N.A.M.N.); (F.F.J.)
| | - Siti Balkis Budin
- Centre for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (S.S.); (I.S.T.); (N.A.M.N.); (F.F.J.)
| |
Collapse
|
17
|
Pallafacchina G, Zanin S, Rizzuto R. From the Identification to the Dissection of the Physiological Role of the Mitochondrial Calcium Uniporter: An Ongoing Story. Biomolecules 2021; 11:biom11060786. [PMID: 34071006 PMCID: PMC8224590 DOI: 10.3390/biom11060786] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/14/2021] [Accepted: 05/20/2021] [Indexed: 12/16/2022] Open
Abstract
The notion of mitochondria being involved in the decoding and shaping of intracellular Ca2+ signals has been circulating since the end of the 19th century. Despite that, the molecular identity of the channel that mediates Ca2+ ion transport into mitochondria remained elusive for several years. Only in the last decade, the genes and pathways responsible for the mitochondrial uptake of Ca2+ began to be cloned and characterized. The gene coding for the pore-forming unit of the mitochondrial channel was discovered exactly 10 years ago, and its product was called mitochondrial Ca2+ uniporter or MCU. Before that, only one of its regulators, the mitochondria Ca2+ uptake regulator 1, MICU1, has been described in 2010. However, in the following years, the scientific interest in mitochondrial Ca2+ signaling regulation and physiological role has increased. This shortly led to the identification of many of its components, to the description of their 3D structure, and the characterization of the uniporter contribution to tissue physiology and pathology. In this review, we will summarize the most relevant achievements in the history of mitochondrial Ca2+ studies, presenting a chronological overview of the most relevant and landmarking discoveries. Finally, we will explore the impact of mitochondrial Ca2+ signaling in the context of muscle physiology, highlighting the recent advances in understanding the role of the MCU complex in the control of muscle trophism and metabolism.
Collapse
Affiliation(s)
- Giorgia Pallafacchina
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
- Neuroscience Institute, Italian National Research Council (CNR), 35131 Padua, Italy
- Correspondence: (G.P.); (R.R.); Tel.: +39-049-827-6029 (G.P.); +39-049-827-3001 (R.R.)
| | - Sofia Zanin
- Department of Immunology, Infectiology and Haematology, Institut Necker-Enfants Malades (INEM), INSERM U1151-CNRS UMR 8253, 75015 Paris, France;
| | - Rosario Rizzuto
- Neuroscience Institute, Italian National Research Council (CNR), 35131 Padua, Italy
- Correspondence: (G.P.); (R.R.); Tel.: +39-049-827-6029 (G.P.); +39-049-827-3001 (R.R.)
| |
Collapse
|
18
|
Gherardi G, De Mario A, Mammucari C. The mitochondrial calcium homeostasis orchestra plays its symphony: Skeletal muscle is the guest of honor. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 362:209-259. [PMID: 34253296 DOI: 10.1016/bs.ircmb.2021.03.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
Skeletal muscle mitochondria are placed in close proximity of the sarcoplasmic reticulum (SR), the main intracellular Ca2+ store. During muscle activity, excitation of sarcolemma and of T-tubule triggers the release of Ca2+ from the SR initiating myofiber contraction. The rise in cytosolic Ca2+ determines the opening of the mitochondrial calcium uniporter (MCU), the highly selective channel of the inner mitochondrial membrane (IMM), causing a robust increase in mitochondrial Ca2+ uptake. The Ca2+-dependent activation of TCA cycle enzymes increases the synthesis of ATP required for SERCA activity. Thus, Ca2+ is transported back into the SR and cytosolic [Ca2+] returns to resting levels eventually leading to muscle relaxation. In recent years, thanks to the molecular identification of MCU complex components, the role of mitochondrial Ca2+ uptake in the pathophysiology of skeletal muscle has been uncovered. In this chapter, we will introduce the reader to a general overview of mitochondrial Ca2+ accumulation. We will tackle the key molecular players and the cellular and pathophysiological consequences of mitochondrial Ca2+ dyshomeostasis. In the second part of the chapter, we will discuss novel findings on the physiological role of mitochondrial Ca2+ uptake in skeletal muscle. Finally, we will examine the involvement of mitochondrial Ca2+ signaling in muscle diseases.
Collapse
Affiliation(s)
- Gaia Gherardi
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Agnese De Mario
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | | |
Collapse
|
19
|
Datta S, Jaiswal M. Mitochondrial calcium at the synapse. Mitochondrion 2021; 59:135-153. [PMID: 33895346 DOI: 10.1016/j.mito.2021.04.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 03/28/2021] [Accepted: 04/13/2021] [Indexed: 12/15/2022]
Abstract
Mitochondria are dynamic organelles, which serve various purposes, including but not limited to the production of ATP and various metabolites, buffering ions, acting as a signaling hub, etc. In recent years, mitochondria are being seen as the central regulators of cellular growth, development, and death. Since neurons are highly specialized cells with a heavy metabolic demand, it is not surprising that neurons are one of the most mitochondria-rich cells in an animal. At synapses, mitochondrial function and dynamics is tightly regulated by synaptic calcium. Calcium influx during synaptic activity causes increased mitochondrial calcium influx leading to an increased ATP production as well as buffering of synaptic calcium. While increased ATP production is required during synaptic transmission, calcium buffering by mitochondria is crucial to prevent faulty neurotransmission and excitotoxicity. Interestingly, mitochondrial calcium also regulates the mobility of mitochondria within synapses causing mitochondria to halt at the synapse during synaptic transmission. In this review, we summarize the various roles of mitochondrial calcium at the synapse.
Collapse
Affiliation(s)
- Sayantan Datta
- Tata Institute of Fundamental Research, Hyderabad, India
| | - Manish Jaiswal
- Tata Institute of Fundamental Research, Hyderabad, India.
| |
Collapse
|
20
|
Alevriadou BR, Patel A, Noble M, Ghosh S, Gohil VM, Stathopulos PB, Madesh M. Molecular nature and physiological role of the mitochondrial calcium uniporter channel. Am J Physiol Cell Physiol 2021; 320:C465-C482. [PMID: 33296287 PMCID: PMC8260355 DOI: 10.1152/ajpcell.00502.2020] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/23/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023]
Abstract
Calcium (Ca2+) signaling is critical for cell function and cell survival. Mitochondria play a major role in regulating the intracellular Ca2+ concentration ([Ca2+]i). Mitochondrial Ca2+ uptake is an important determinant of cell fate and governs respiration, mitophagy/autophagy, and the mitochondrial pathway of apoptosis. Mitochondrial Ca2+ uptake occurs via the mitochondrial Ca2+ uniporter (MCU) complex. This review summarizes the present knowledge on the function of MCU complex, regulation of MCU channel, and the role of MCU in Ca2+ homeostasis and human disease pathogenesis. The channel core consists of four MCU subunits and essential MCU regulators (EMRE). Regulatory proteins that interact with them include mitochondrial Ca2+ uptake 1/2 (MICU1/2), MCU dominant-negative β-subunit (MCUb), MCU regulator 1 (MCUR1), and solute carrier 25A23 (SLC25A23). In addition to these proteins, cardiolipin, a mitochondrial membrane-specific phospholipid, has been shown to interact with the channel core. The dynamic interplay between the core and regulatory proteins modulates MCU channel activity after sensing local changes in [Ca2+]i, reactive oxygen species, and other environmental factors. Here, we highlight the structural details of the human MCU heteromeric assemblies and their known roles in regulating mitochondrial Ca2+ homeostasis. MCU dysfunction has been shown to alter mitochondrial Ca2+ dynamics, in turn eliciting cell apoptosis. Changes in mitochondrial Ca2+ uptake have been implicated in pathological conditions affecting multiple organs, including the heart, skeletal muscle, and brain. However, our structural and functional knowledge of this vital protein complex remains incomplete, and understanding the precise role for MCU-mediated mitochondrial Ca2+ signaling in disease requires further research efforts.
Collapse
Affiliation(s)
- B Rita Alevriadou
- Department of Biomedical Engineering, Jacobs School of Medicine and Biomedical Sciences and School of Engineering and Applied Sciences, University at Buffalo-State University of New York, Buffalo, New York
| | - Akshar Patel
- Department of Biomedical Engineering, Jacobs School of Medicine and Biomedical Sciences and School of Engineering and Applied Sciences, University at Buffalo-State University of New York, Buffalo, New York
| | - Megan Noble
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Sagnika Ghosh
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas
| | - Vishal M Gohil
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas
| | - Peter B Stathopulos
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Muniswamy Madesh
- Department of Medicine/Cardiology Division, Center for Precision Medicine, University of Texas Health San Antonio, San Antonio, Texas
| |
Collapse
|
21
|
GWAS reveal a role for the central nervous system in regulating weight and weight change in response to exercise. Sci Rep 2021; 11:5144. [PMID: 33664357 PMCID: PMC7933348 DOI: 10.1038/s41598-021-84534-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/17/2021] [Indexed: 01/16/2023] Open
Abstract
Body size and weight show considerable variation both within and between species. This variation is controlled in part by genetics, but also strongly influenced by environmental factors including diet and the level of activity experienced by the individual. Due to the increasing obesity epidemic in much of the world, there is considerable interest in the genetic factors that control body weight and how weight changes in response to exercise treatments. Here, we address this question in the Drosophila model system, utilizing 38 strains of the Drosophila Genetics Reference Panel. We use GWAS to identify the molecular pathways that control weight and weight changes in response to exercise. We find that there is a complex set of molecular pathways controlling weight, with many genes linked to the central nervous system (CNS). The CNS also plays a role in the weight change with exercise, in particular, signaling from the CNS. Additional analyses revealed that weight in Drosophila is driven by two factors, animal size, and body composition, as the amount of fat mass versus lean mass impacts the density. Thus, while the CNS appears to be important for weight and exercise-induced weight change, signaling pathways are particularly important for determining how exercise impacts weight.
Collapse
|
22
|
Bai J, Liu C, Zhu P, Li Y. Novel Insights Into Molecular Mechanism of Mitochondria in Diabetic Cardiomyopathy. Front Physiol 2021; 11:609157. [PMID: 33536936 PMCID: PMC7849834 DOI: 10.3389/fphys.2020.609157] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/21/2020] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular complication is one of the significant causes of death in diabetic mellitus (DM) in which diabetic cardiomyopathy, independent of hypertension, cardiac valvular disease, and coronary atherosclerosis, occupies an important position. Although the detailed pathogenesis of diabetic cardiomyopathy remains unclear currently, mitochondrial morphological abnormality and dysfunction were observed in diabetic cardiomyopathy animal models according to much research, suggesting that mitochondrial structural and functional impairment played an integral role in the formation of diabetic cardiomyopathy. Thus, we have summarized the effect of mitochondria on the process of diabetic cardiomyopathy, including abnormal mitochondrial morphology, mitochondrial energy metabolism disorder, enhanced mitochondrial oxidative stress, mitochondrial unbalanced calcium homeostasis, and mitochondrial autophagy. Based on the above mechanisms and the related evidence, more therapeutic strategies targeting mitochondria in diabetic cardiomyopathy have been and will be proposed to delay the progression of the disease.
Collapse
Affiliation(s)
- Jing Bai
- Medical School of Chinese PLA, Beijing, China.,Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Chuanbin Liu
- Medical School of Chinese PLA, Beijing, China.,Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Pingjun Zhu
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yang Li
- Medical School of Chinese PLA, Beijing, China.,Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
23
|
Wang X, Davis RL. Early Mitochondrial Fragmentation and Dysfunction in a Drosophila Model for Alzheimer's Disease. Mol Neurobiol 2021; 58:143-155. [PMID: 32909149 PMCID: PMC7704861 DOI: 10.1007/s12035-020-02107-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/27/2020] [Indexed: 12/24/2022]
Abstract
Many different cellular systems and molecular processes become compromised in Alzheimer's disease (AD) including proteostasis, autophagy, inflammatory responses, synapse and neuronal circuitry, and mitochondrial function. We focused in this study on mitochondrial dysfunction owing to the toxic neuronal environment produced by expression of Aβ42, and its relationship to other pathologies found in AD including increased neuronal apoptosis, plaque deposition, and memory impairment. Using super-resolution microscopy, we have assayed mitochondrial status in the three distinct neuronal compartments (somatic, dendritic, axonal) of mushroom body neurons of Drosophila expressing Aβ42. The mushroom body neurons comprise a major center for olfactory memory formation in insects. We employed calcium imaging to measure mitochondrial function, immunohistochemical and staining techniques to measure apoptosis and plaque formation, and olfactory classical conditioning to measure learning. We found that mitochondria become fragmented at a very early age along with decreased function measured by mitochondrial calcium entry. Increased apoptosis and plaque deposition also occur early, yet interestingly, a learning impairment was found only after a much longer period of time-10 days, which is a large fraction of the fly's lifespan. This is similar to the pronounced delay between cellular pathologies and the emergence of a memory dysfunction in humans. Our studies are consistent with the model that mitochondrial dysfunction and/or other cellular pathologies emerge at an early age and lead to much later learning impairments. The results obtained further develop this Drosophila model as a useful in vivo system for probing the mechanisms by which Aβ42 produces mitochondrial and other cellular toxicities that produce memory dysfunction.
Collapse
Affiliation(s)
- Xingjun Wang
- Department of Neuroscience, Scripps Research Institute Florida, Jupiter, Florida, 33458, USA
| | - Ronald L Davis
- Department of Neuroscience, Scripps Research Institute Florida, Jupiter, Florida, 33458, USA.
| |
Collapse
|
24
|
Morris O, Deng H, Tam C, Jasper H. Warburg-like Metabolic Reprogramming in Aging Intestinal Stem Cells Contributes to Tissue Hyperplasia. Cell Rep 2020; 33:108423. [PMID: 33238124 PMCID: PMC8011352 DOI: 10.1016/j.celrep.2020.108423] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/28/2020] [Accepted: 11/02/2020] [Indexed: 12/28/2022] Open
Abstract
In many tissues, stem cell (SC) proliferation is dynamically adjusted to regenerative needs. How SCs adapt their metabolism to meet the demands of proliferation and how changes in such adaptive mechanisms contribute to age-related dysfunction remain poorly understood. Here, we identify mitochondrial Ca2+ uptake as a central coordinator of SC metabolism. Live imaging of genetically encoded metabolite sensors in intestinal SCs (ISCs) of Drosophila reveals that mitochondrial Ca2+ uptake transiently adapts electron transport chain flux to match energetic demand upon proliferative activation. This tight metabolic adaptation is lost in ISCs of old flies, as declines in mitochondrial Ca2+ uptake promote a "Warburg-like" metabolic reprogramming toward aerobic glycolysis. This switch mimics metabolic reprogramming by the oncogene RasV12 and enhances ISC hyperplasia. Our data identify a critical mechanism for metabolic adaptation of tissue SCs and reveal how its decline sets aging SCs on a metabolic trajectory reminiscent of that seen upon oncogenic transformation.
Collapse
Affiliation(s)
- Otto Morris
- Immunology Discovery, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Hansong Deng
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 20092, China; Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945-1400, USA
| | - Christine Tam
- Department of Biomolecular Resources, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Heinrich Jasper
- Immunology Discovery, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA; Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945-1400, USA.
| |
Collapse
|
25
|
Rodríguez LR, Calap-Quintana P, Lapeña-Luzón T, Pallardó FV, Schneuwly S, Navarro JA, Gonzalez-Cabo P. Oxidative stress modulates rearrangement of endoplasmic reticulum-mitochondria contacts and calcium dysregulation in a Friedreich's ataxia model. Redox Biol 2020; 37:101762. [PMID: 33128998 PMCID: PMC7585950 DOI: 10.1016/j.redox.2020.101762] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 10/07/2020] [Accepted: 10/12/2020] [Indexed: 12/13/2022] Open
Abstract
Friedreich ataxia (FRDA) is a neurodegenerative disorder characterized by neuromuscular and neurological manifestations. It is caused by mutations in the FXN gene, which results in loss of the mitochondrial protein frataxin. Endoplasmic Reticulum-mitochondria associated membranes (MAMs) are inter-organelle structures involved in the regulation of essential cellular processes, including lipid metabolism and calcium signaling. In the present study, we have analyzed in both, unicellular and multicellular models of FRDA, calcium management and integrity of MAMs. We observed that function of MAMs is compromised in our cellular model of FRDA, which was improved upon treatment with antioxidants. In agreement, promoting mitochondrial calcium uptake was sufficient to restore several defects caused by frataxin deficiency in Drosophila Melanogaster. Remarkably, our findings describe for the first time frataxin as a member of the protein network of MAMs, where interacts with two of the main proteins implicated in endoplasmic reticulum-mitochondria communication. These results suggest a new role of frataxin, indicate that FRDA goes beyond mitochondrial defects and highlight MAMs as novel therapeutic candidates to improve patient's conditions.
Collapse
Affiliation(s)
- Laura R Rodríguez
- Department of Physiology, Faculty of Medicine and Dentistry. University of Valencia-INCLIVA, Valencia, 46010, Spain; Associated Unit for Rare Diseases INCLIVA-CIPF, Valencia, Spain
| | - Pablo Calap-Quintana
- Department of Physiology, Faculty of Medicine and Dentistry. University of Valencia-INCLIVA, Valencia, 46010, Spain; Associated Unit for Rare Diseases INCLIVA-CIPF, Valencia, Spain; CIBER de Enfermedades Raras (CIBERER), Valencia, Spain
| | - Tamara Lapeña-Luzón
- Department of Physiology, Faculty of Medicine and Dentistry. University of Valencia-INCLIVA, Valencia, 46010, Spain; Associated Unit for Rare Diseases INCLIVA-CIPF, Valencia, Spain; CIBER de Enfermedades Raras (CIBERER), Valencia, Spain
| | - Federico V Pallardó
- Department of Physiology, Faculty of Medicine and Dentistry. University of Valencia-INCLIVA, Valencia, 46010, Spain; Associated Unit for Rare Diseases INCLIVA-CIPF, Valencia, Spain; CIBER de Enfermedades Raras (CIBERER), Valencia, Spain
| | - Stephan Schneuwly
- Institute of Zoology, Universitaetsstrasse 31, University of Regensburg, 93040, Regensburg, Germany
| | - Juan A Navarro
- Institute of Zoology, Universitaetsstrasse 31, University of Regensburg, 93040, Regensburg, Germany; INCLIVA Biomedial Research Institute, Valencia, Spain
| | - Pilar Gonzalez-Cabo
- Department of Physiology, Faculty of Medicine and Dentistry. University of Valencia-INCLIVA, Valencia, 46010, Spain; Associated Unit for Rare Diseases INCLIVA-CIPF, Valencia, Spain; CIBER de Enfermedades Raras (CIBERER), Valencia, Spain.
| |
Collapse
|
26
|
Filadi R, Pizzo P. Mitochondrial calcium handling and neurodegeneration: when a good signal goes wrong. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2020.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
27
|
Mitochondrial Calcium Deregulation in the Mechanism of Beta-Amyloid and Tau Pathology. Cells 2020; 9:cells9092135. [PMID: 32967303 PMCID: PMC7564294 DOI: 10.3390/cells9092135] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/18/2020] [Accepted: 09/19/2020] [Indexed: 02/06/2023] Open
Abstract
Aggregation and deposition of β-amyloid and/or tau protein are the key neuropathological features in neurodegenerative disorders such as Alzheimer's disease (AD) and other tauopathies including frontotemporal dementia (FTD). The interaction between oxidative stress, mitochondrial dysfunction and the impairment of calcium ions (Ca2+) homeostasis induced by misfolded tau and β-amyloid plays an important role in the progressive neuronal loss occurring in specific areas of the brain. In addition to the control of bioenergetics and ROS production, mitochondria are fine regulators of the cytosolic Ca2+ homeostasis that induce vital signalling mechanisms in excitable cells such as neurons. Impairment in the mitochondrial Ca2+ uptake through the mitochondrial Ca2+ uniporter (MCU) or release through the Na+/Ca2+ exchanger may lead to mitochondrial Ca2+ overload and opening of the permeability transition pore inducing neuronal death. Recent evidence suggests an important role for these mechanisms as the underlying causes for neuronal death in β-amyloid and tau pathology. The present review will focus on the mechanisms that lead to cytosolic and especially mitochondrial Ca2+ disturbances occurring in AD and tau-induced FTD, and propose possible therapeutic interventions for these disorders.
Collapse
|
28
|
Zhang B, Jia K, Tian J, Du H. Cyclophilin D counterbalances mitochondrial calcium uniporter-mediated brain mitochondrial calcium uptake. Biochem Biophys Res Commun 2020; 529:314-320. [PMID: 32703429 PMCID: PMC7481651 DOI: 10.1016/j.bbrc.2020.05.204] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 05/27/2020] [Indexed: 12/20/2022]
Abstract
Mitochondria play an essential role in maintaining intraneuronal calcium homeostasis. Mitochondrial calcium uniporter (MCU) is a determined major brain mitochondrial calcium entry pathway. Activated MCU-mediated mitochondrial calcium overloading has been linked with brain mitochondrial pathology in disease conditions. Cyclophilin D (CypD)-mediated mitochondrial permeability transition (mPT) favors mitochondrial calcium efflux. The physiological function of CypD-mediated mPT has received increasing recognition. However, the regulatory role of CypD-mediated mPT in brain mitochondrial calcium dynamics in response to mitochondrial calcium accumulation via MCU has not been comprehensively studied. Here, by adopting purified brain mitochondria, we have determined an effect of CypD and CypD-mediated mPT against mitochondrial calcium overloading. In addition, blockade of CypD pharmaceutically or genetically blunts brain mitochondrial MCU's sensitivity to its inhibitor. Therefore, our findings suggest that CypD-mediated mPT is a mitochondrial compensatory response to MCU-mediated excess mitochondrial calcium accumulation. Moreover, CypD may potentially modulate MCU function in calcium-stressed mitochondria.
Collapse
Affiliation(s)
- Bei Zhang
- Department of Biological Sciences, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, USA
| | - Kun Jia
- Department of Biological Sciences, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, USA
| | - Jing Tian
- Department of Biological Sciences, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, USA
| | - Heng Du
- Department of Biological Sciences, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, USA.
| |
Collapse
|
29
|
Tang NH, Kim KW, Xu S, Blazie SM, Yee BA, Yeo GW, Jin Y, Chisholm AD. The mRNA Decay Factor CAR-1/LSM14 Regulates Axon Regeneration via Mitochondrial Calcium Dynamics. Curr Biol 2020; 30:865-876.e7. [PMID: 31983639 DOI: 10.1016/j.cub.2019.12.061] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 11/26/2019] [Accepted: 12/19/2019] [Indexed: 12/11/2022]
Abstract
mRNA decay factors regulate mRNA turnover by recruiting non-translating mRNAs and targeting them for translational repression and mRNA degradation. How mRNA decay pathways regulate cellular function in vivo with specificity is poorly understood. Here, we show that C. elegans mRNA decay factors, including the translational repressors CAR-1/LSM14 and CGH-1/DDX6, and the decapping enzymes DCAP-1/DCP1, function in neurons to differentially regulate axon development, maintenance, and regrowth following injury. In neuronal cell bodies, CAR-1 fully colocalizes with CGH-1 and partially colocalizes with DCAP-1, suggesting that mRNA decay components form at least two types of cytoplasmic granules. Following axon injury in adult neurons, loss of CAR-1 or CGH-1 results in increased axon regrowth and growth cone formation, whereas loss of DCAP-1 or DCAP-2 results in reduced regrowth. To determine how CAR-1 inhibits regrowth, we analyzed mRNAs bound to pan-neuronally expressed GFP::CAR-1 using a crosslinking and immunoprecipitation-based approach. Among the putative mRNA targets of CAR-1, we characterized the roles of micu-1, a regulator of the mitochondrial calcium uniporter MCU-1, in axon injury. We show that loss of car-1 results increased MICU-1 protein levels, and that enhanced axon regrowth in car-1 mutants is dependent on micu-1 and mcu-1. Moreover, axon injury induces transient calcium influx into axonal mitochondria, dependent on MCU-1. In car-1 loss-of-function mutants and in micu-1 overexpressing animals, the axonal mitochondrial calcium influx is more sustained, which likely underlies enhanced axon regrowth. Our data uncover a novel pathway that controls axon regrowth through axonal mitochondrial calcium uptake.
Collapse
Affiliation(s)
- Ngang Heok Tang
- Section of Neurobiology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kyung Won Kim
- Section of Neurobiology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Suhong Xu
- Section of Neurobiology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Stephen M Blazie
- Section of Neurobiology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Brian A Yee
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Gene W Yeo
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yishi Jin
- Section of Neurobiology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Andrew D Chisholm
- Section of Neurobiology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
30
|
Abstract
Drosophila melanogaster, colloquially known as the fruit fly, is one of the most commonly used model organisms in scientific research. Although the final architecture of a fly and a human differs greatly, most of the fundamental biological mechanisms and pathways controlling development and survival are conserved through evolution between the two species. For this reason, Drosophila has been productively used as a model organism for over a century, to study a diverse range of biological processes, including development, learning, behavior and aging. Ca2+ signaling comprises complex pathways that impact on virtually every aspect of cellular physiology. Within such a complex field of study, Drosophila offers the advantages of consolidated molecular and genetic techniques, lack of genetic redundancy and a completely annotated genome since 2000. These and other characteristics provided the basis for the identification of many genes encoding Ca2+ signaling molecules and the disclosure of conserved Ca2+ signaling pathways. In this review, we will analyze the applications of Ca2+ imaging in the fruit fly model, highlighting in particular their impact on the study of normal brain function and pathogenesis of neurodegenerative diseases.
Collapse
|
31
|
The Role of Intracellular Calcium in Changing of ElectricalCharacteristics of Premotor Interneurons in Intact Snails and Snails During Various Forms of Plasticity. BIONANOSCIENCE 2019. [DOI: 10.1007/s12668-019-00669-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
32
|
de-Souza-Ferreira E, Rios-Neto IM, Martins EL, Galina A. Mitochondria-coupled glucose phosphorylation develops after birth to modulate H 2 O 2 release and calcium handling in rat brain. J Neurochem 2019; 149:624-640. [PMID: 31001830 DOI: 10.1111/jnc.14705] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 03/20/2019] [Accepted: 04/11/2019] [Indexed: 12/17/2022]
Abstract
The adult brain is a high-glucose and oxygen-dependent organ, with an extremely organized network of cells and large energy-consuming synapses. To reach this level of organization, early stages in development must include an efficient control of cellular events and regulation of intracellular signaling molecules and ions such as hydrogen peroxide (H2 O2 ) and calcium (Ca2+ ), but in cerebral tissue, these mechanisms of regulation are still poorly understood. Hexokinase (HK) is the first enzyme in the metabolism of glucose and, when bound to mitochondria (mtHK), it has been proposed to have a role in modulation of mitochondrial H2 O2 generation and Ca2+ handling. Here, we have investigated how mtHK modulates these signals in the mitochondrial context during postnatal development of the mouse brain. Using high-resolution respirometry, western blot analysis, spectrometry and resorufin, and Calcium Green fluorescence assays with brain mitochondria purified postnatally from day 1 to day 60, we demonstrate that brain HK increases its coupling to mitochondria and to oxidative phosphorylation to induce a cycle of ADP entry/ATP exit of the mitochondrial matrix that leads to efficient control over H2 O2 generation and Ca2+ uptake during development until reaching plateau at day 21. This contrasts sharply with the antioxidant enzymes, which do not increase as mitochondrial H2 O2 generation escalates. These results suggest that, as its use of glucose increases, the brain couples HK to mitochondria to improve glucose metabolism, redox balance and Ca2+ signaling during development, positioning mitochondria-bound hexokinase as a hub for intracellular signaling control.
Collapse
Affiliation(s)
- Eduardo de-Souza-Ferreira
- Laboratory of Bioenergetics and Mitochondrial Physiology, Institute of Medical Biochemistry Leopoldo de Meis, Center for Health Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Izac Miranda Rios-Neto
- Laboratory of Bioenergetics and Mitochondrial Physiology, Institute of Medical Biochemistry Leopoldo de Meis, Center for Health Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eduarda Lopes Martins
- Laboratory of Bioenergetics and Mitochondrial Physiology, Institute of Medical Biochemistry Leopoldo de Meis, Center for Health Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Antonio Galina
- Laboratory of Bioenergetics and Mitochondrial Physiology, Institute of Medical Biochemistry Leopoldo de Meis, Center for Health Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
33
|
Rohith BN, Shyamala BV. Developmental Deformity Due to
scalloped
Non‐Function in
Drosophila
Brain Leads to Cognitive Impairment. Dev Neurobiol 2019; 79:236-251. [DOI: 10.1002/dneu.22668] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 11/07/2018] [Accepted: 01/18/2019] [Indexed: 11/10/2022]
|
34
|
Liu SM, Li XZ, Zhang SN, Yang ZM, Wang KX, Lu F, Wang CZ, Yuan CS. Acanthopanax senticosus Protects Structure and Function of Mesencephalic Mitochondria in A Mouse Model of Parkinson's Disease. Chin J Integr Med 2018; 24:835-843. [PMID: 30090975 DOI: 10.1007/s11655-018-2935-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2016] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To investigate the neuro-protective effects of Acanthopanax senticosus Harms (EAS) on mesencephalic mitochondria and the mechanism of action, using a mouse model of Parkinson's disease (PD). METHODS The chemical fingerprint analysis of the extract of Acanthopanax senticosus Harms (EAS) was performed using the ultra performance liquid chromatograph and time of flight mass spectrometry. Thirty mice were randomly divided into the control group, the MPTP model group, and the EAS treated group with MPTP (MPTP+EAS group, 10 in each group). The MPTP model group and the MPTP+EAS group received MPTP-HCl (30 mg/kg i.p) once a day for 5 days. The control group received an equal volume of saline (20 mL/kg i.p) once a day for 5 days. Induced by 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine hydrochloride daily (MPTP-HCl, 30 mg/kg) for 5 days, the PD mice were treated with EAS at 45.5 mg/kg daily for 20 days. The behavioral testing of mice was carried out using the pole-climbing test. The integrity and functions of neurons were examined in mesencephalic mitochondria in a PD mouse model, including nicotinamide adenine dinucleotide dehydrogenase ubiquinone flavoprotein 2 (NDUFV2), mitochondrially encoded nicotinamide adenine dinucleotide dehydrogenase 1 (MT-ND1), succinate dehydrogenase complex subunit A (SDHA), and succinate dehydrogenase cytochrome b560 subunit (SDHC). RESULTS After treatment with EAS, the behavioral changes induced by MPTP were attenuated significantly (P<0.05). EAS protected the mesencephalic mitochondria from swelling and attenuated the decreases in their membrane potential (both P<0.05), which was supported by an ultra-structural level analysis. The changes in reactive oxygen species (ROS), malonic dialdehyde (MDA), oxidative phosphorylation (OXPHOS) system 4 subunits levels and PD-related proteins expressions (parkin, Pink1, DJ-1, α-synuclein, and Lrrk2) reverted to near normal levels (all P<0.05), based on the results of immune-histological and Western blotting observations. CONCLUSIONS The neuro-protective effects of EAS are linked to protecting mice against MPTP-induced mitochondrial dysfunction and structural damage. Therefore, EAS is a promising candidate for the prevention or treatment of mitochondrial neurodegenerative disorders, such as PD.
Collapse
Affiliation(s)
- Shu-Min Liu
- Drug Safety Evaluation Center, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Xu-Zhao Li
- Drug Safety Evaluation Center, Heilongjiang University of Chinese Medicine, Harbin, 150040, China.,Department of Pharmacy, Guiyang College of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Shuai-Nan Zhang
- Drug Safety Evaluation Center, Heilongjiang University of Chinese Medicine, Harbin, 150040, China.,Department of Pharmacy, Guiyang College of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Zhi-Ming Yang
- Drug Safety Evaluation Center, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Ke-Xin Wang
- Drug Safety Evaluation Center, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Fang Lu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| | - Chong-Zhi Wang
- Tang Center for Herbal Medicine Research, and Department of Anesthesia and Critical Care, University of Chicago, Chicago, IL, 60637, USA
| | - Chun-Su Yuan
- Tang Center for Herbal Medicine Research, and Department of Anesthesia and Critical Care, University of Chicago, Chicago, IL, 60637, USA
| |
Collapse
|
35
|
Abstract
Mitochondrial Ca2+ regulation is crucial for bioenergetics and cellular signaling. The mechanisms controlling mitochondrial calcium homeostasis have been recently unraveled with the discovery of mitochondrial inner membrane proteins that regulate mitochondrial Ca2+ uptake and extrusion. Mitochondrial Ca2+ uptake depends on a large complex of proteins centered around the Ca2+ channel protein, mitochondrial Ca2+ uniporter (MCU) in close interactions with several regulatory subunits (MCUb, EMRE, MICU1, MICU2). Mitochondrial Ca2+ extrusion is mainly mediated by the mitochondrial Na+/Ca2+/Li+ exchanger (NCLX). Here, we review the major players of mitochondrial Ca2+ homeostasis and their physiological functions.
Collapse
Affiliation(s)
- Trayambak Pathak
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Mohamed Trebak
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA, United States.
| |
Collapse
|
36
|
Mammucari C, Raffaello A, Vecellio Reane D, Gherardi G, De Mario A, Rizzuto R. Mitochondrial calcium uptake in organ physiology: from molecular mechanism to animal models. Pflugers Arch 2018. [PMID: 29541860 PMCID: PMC6060757 DOI: 10.1007/s00424-018-2123-2] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mitochondrial Ca2+ is involved in heterogeneous functions, ranging from the control of metabolism and ATP production to the regulation of cell death. In addition, mitochondrial Ca2+ uptake contributes to cytosolic [Ca2+] shaping thus impinging on specific Ca2+-dependent events. Mitochondrial Ca2+ concentration is controlled by influx and efflux pathways: the former controlled by the activity of the mitochondrial Ca2+ uniporter (MCU), the latter by the Na+/Ca2+ exchanger (NCLX) and the H+/Ca2+ (mHCX) exchanger. The molecular identities of MCU and of NCLX have been recently unraveled, thus allowing genetic studies on their physiopathological relevance. After a general framework on the significance of mitochondrial Ca2+ uptake, this review discusses the structure of the MCU complex and the regulation of its activity, the importance of mitochondrial Ca2+ signaling in different physiological settings, and the consequences of MCU modulation on organ physiology.
Collapse
Affiliation(s)
| | - Anna Raffaello
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | | | - Gaia Gherardi
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - Agnese De Mario
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padova, Padua, Italy.
| |
Collapse
|
37
|
Arduino DM, Perocchi F. Pharmacological modulation of mitochondrial calcium homeostasis. J Physiol 2018; 596:2717-2733. [PMID: 29319185 DOI: 10.1113/jp274959] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 12/13/2017] [Indexed: 12/26/2022] Open
Abstract
Mitochondria are pivotal organelles in calcium (Ca2+ ) handling and signalling, constituting intracellular checkpoints for numerous processes that are vital for cell life. Alterations in mitochondrial Ca2+ homeostasis have been linked to a variety of pathological conditions and are critical in the aetiology of several human diseases. Efforts have been taken to harness mitochondrial Ca2+ transport mechanisms for therapeutic intervention, but pharmacological compounds that direct and selectively modulate mitochondrial Ca2+ homeostasis are currently lacking. New avenues have, however, emerged with the breakthrough discoveries on the genetic identification of the main players involved in mitochondrial Ca2+ influx and efflux pathways and with recent hints towards a deep understanding of the function of these molecular systems. Here, we review the current advances in the understanding of the mechanisms and regulation of mitochondrial Ca2+ homeostasis and its contribution to physiology and human disease. We also introduce and comment on the recent progress towards a systems-level pharmacological targeting of mitochondrial Ca2+ homeostasis.
Collapse
Affiliation(s)
- Daniela M Arduino
- Gene Center, Department of Biochemistry, Ludwig-Maximilians Universität München, Munich, 81377, Germany.,Institute for Diabetes and Obesity, Helmholtz Diabetes Center (HDC), Helmholtz Zentrum München and German National Diabetes Center (DZD), Neuherberg, 85764, Germany
| | - Fabiana Perocchi
- Gene Center, Department of Biochemistry, Ludwig-Maximilians Universität München, Munich, 81377, Germany.,Institute for Diabetes and Obesity, Helmholtz Diabetes Center (HDC), Helmholtz Zentrum München and German National Diabetes Center (DZD), Neuherberg, 85764, Germany
| |
Collapse
|
38
|
Liu JC, Parks RJ, Liu J, Stares J, Rovira II, Murphy E, Finkel T. The In Vivo Biology of the Mitochondrial Calcium Uniporter. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:49-63. [PMID: 28551781 DOI: 10.1007/978-3-319-55330-6_3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The identification of the molecular composition of the mitochondrial calcium uniporter has allowed for the genetic manipulation of its components and the creation of various in vivo genetic models. Here, we review the initial attempts to modulate the expression of components of the calcium uniporter in a range of organisms from plants to mammals. This analysis has confirmed the strict requirement for the uniporter for in vivo mitochondrial calcium uptake and for maintaining mitochondrial calcium homeostasis. We further discuss the physiological effects following genetic manipulation of the uniporter on tissue bioenergetics and the threshold for cell death. Finally, we analyze the limited information regarding the role of various uniporter components in human disease.
Collapse
Affiliation(s)
- Julia C Liu
- Center for Molecular Medicine, National Heart Lung and Blood Institute, Bethesda, MD, 20892, USA
| | - Randi J Parks
- Systems Biology Center, National Heart Lung and Blood Institute, Bethesda, MD, 20892, USA
| | - Jie Liu
- Center for Molecular Medicine, National Heart Lung and Blood Institute, Bethesda, MD, 20892, USA
| | - Justin Stares
- Center for Molecular Medicine, National Heart Lung and Blood Institute, Bethesda, MD, 20892, USA
| | - Ilsa I Rovira
- Center for Molecular Medicine, National Heart Lung and Blood Institute, Bethesda, MD, 20892, USA
| | - Elizabeth Murphy
- Systems Biology Center, National Heart Lung and Blood Institute, Bethesda, MD, 20892, USA
| | - Toren Finkel
- Center for Molecular Medicine, National Heart Lung and Blood Institute, Bethesda, MD, 20892, USA. .,NIH, Bldg 10/CRC 5-3330, Bethesda, MD, 20892, USA.
| |
Collapse
|
39
|
Choi S, Quan X, Bang S, Yoo H, Kim J, Park J, Park KS, Chung J. Mitochondrial calcium uniporter in Drosophila transfers calcium between the endoplasmic reticulum and mitochondria in oxidative stress-induced cell death. J Biol Chem 2017; 292:14473-14485. [PMID: 28726639 DOI: 10.1074/jbc.m116.765578] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 07/13/2017] [Indexed: 12/16/2022] Open
Abstract
Mitochondrial calcium plays critical roles in diverse cellular processes ranging from energy metabolism to cell death. Previous studies have demonstrated that mitochondrial calcium uptake is mainly mediated by the mitochondrial calcium uniporter (MCU) complex. However, the roles of the MCU complex in calcium transport, signaling, and dysregulation by oxidative stress still remain unclear. Here, we confirmed that Drosophila MCU contains evolutionarily conserved structures and requires essential MCU regulator (EMRE) for its calcium channel activities. We generated Drosophila MCU loss-of-function mutants, which lacked mitochondrial calcium uptake in response to caffeine stimulation. Basal metabolic activities were not significantly affected in these MCU mutants, as observed in examinations of body weight, food intake, body sugar level, and starvation-induced autophagy. However, oxidative stress-induced increases in mitochondrial calcium, mitochondrial membrane potential depolarization, and cell death were prevented in these mutants. We also found that inositol 1,4,5-trisphosphate receptor genetically interacts with Drosophila MCU and effectively modulates mitochondrial calcium uptake upon oxidative stress. Taken together, these results support the idea that Drosophila MCU is responsible for endoplasmic reticulum-to-mitochondrial calcium transfer and for cell death due to mitochondrial dysfunction under oxidative stress.
Collapse
Affiliation(s)
- Sekyu Choi
- From the National Creative Research Initiatives Center for Energy Homeostasis Regulation, Institute of Molecular Biology and Genetics and School of Biological Sciences, Seoul National University, Seoul 08826, Korea and
| | - Xianglan Quan
- the Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Gangwon-Do 26426, Korea
| | - Sunhoe Bang
- From the National Creative Research Initiatives Center for Energy Homeostasis Regulation, Institute of Molecular Biology and Genetics and School of Biological Sciences, Seoul National University, Seoul 08826, Korea and
| | - Heesuk Yoo
- From the National Creative Research Initiatives Center for Energy Homeostasis Regulation, Institute of Molecular Biology and Genetics and School of Biological Sciences, Seoul National University, Seoul 08826, Korea and
| | - Jiyoung Kim
- From the National Creative Research Initiatives Center for Energy Homeostasis Regulation, Institute of Molecular Biology and Genetics and School of Biological Sciences, Seoul National University, Seoul 08826, Korea and
| | - Jiwon Park
- From the National Creative Research Initiatives Center for Energy Homeostasis Regulation, Institute of Molecular Biology and Genetics and School of Biological Sciences, Seoul National University, Seoul 08826, Korea and
| | - Kyu-Sang Park
- the Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Gangwon-Do 26426, Korea
| | - Jongkyeong Chung
- From the National Creative Research Initiatives Center for Energy Homeostasis Regulation, Institute of Molecular Biology and Genetics and School of Biological Sciences, Seoul National University, Seoul 08826, Korea and
| |
Collapse
|
40
|
Mammucari C, Gherardi G, Rizzuto R. Structure, Activity Regulation, and Role of the Mitochondrial Calcium Uniporter in Health and Disease. Front Oncol 2017; 7:139. [PMID: 28740830 PMCID: PMC5502327 DOI: 10.3389/fonc.2017.00139] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 06/19/2017] [Indexed: 01/09/2023] Open
Abstract
Mitochondrial Ca2+ uptake plays a pivotal role both in cell energy balance and in cell fate determination. Studies on the role of mitochondrial Ca2+ signaling in pathophysiology have been favored by the identification of the genes encoding the mitochondrial calcium uniporter (MCU) and its regulatory subunits. Thus, research carried on in the last years on one hand has determined the structure of the MCU complex and its regulation, on the other has uncovered the consequences of dysregulated mitochondrial Ca2+ signaling in cell and tissue homeostasis. Whether mitochondrial Ca2+ uptake can be exploited as a weapon to counteract cancer progression is debated. In this review, we summarize recent research on the molecular structure of the MCU, the regulatory mechanisms that control its activity and its relevance in pathophysiology, focusing in particular on its role in cancer progression.
Collapse
Affiliation(s)
| | - Gaia Gherardi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
41
|
Ruggiero A, Aloni E, Korkotian E, Zaltsman Y, Oni-Biton E, Kuperman Y, Tsoory M, Shachnai L, Levin-Zaidman S, Brenner O, Segal M, Gross A. Loss of forebrain MTCH2 decreases mitochondria motility and calcium handling and impairs hippocampal-dependent cognitive functions. Sci Rep 2017; 7:44401. [PMID: 28276496 PMCID: PMC5343590 DOI: 10.1038/srep44401] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 02/07/2017] [Indexed: 12/23/2022] Open
Abstract
Mitochondrial Carrier Homolog 2 (MTCH2) is a novel regulator of mitochondria metabolism, which was recently associated with Alzheimer’s disease. Here we demonstrate that deletion of forebrain MTCH2 increases mitochondria and whole-body energy metabolism, increases locomotor activity, but impairs motor coordination and balance. Importantly, mice deficient in forebrain MTCH2 display a deficit in hippocampus-dependent cognitive functions, including spatial memory, long term potentiation (LTP) and rates of spontaneous excitatory synaptic currents. Moreover, MTCH2-deficient hippocampal neurons display a deficit in mitochondria motility and calcium handling. Thus, MTCH2 is a critical player in neuronal cell biology, controlling mitochondria metabolism, motility and calcium buffering to regulate hippocampal-dependent cognitive functions.
Collapse
Affiliation(s)
- Antonella Ruggiero
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Etay Aloni
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Eduard Korkotian
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Yehudit Zaltsman
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Efrat Oni-Biton
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Yael Kuperman
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Michael Tsoory
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Liat Shachnai
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Smadar Levin-Zaidman
- Department of Chemical research Support, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ori Brenner
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Menahem Segal
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Atan Gross
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|