101
|
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
|
102
|
Aoyama-Ishiwatari S, Hirabayashi Y. Endoplasmic Reticulum-Mitochondria Contact Sites-Emerging Intracellular Signaling Hubs. Front Cell Dev Biol 2021; 9:653828. [PMID: 34095118 PMCID: PMC8172986 DOI: 10.3389/fcell.2021.653828] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/06/2021] [Indexed: 01/04/2023] Open
Abstract
It has become apparent that our textbook illustration of singular isolated organelles is obsolete. In reality, organelles form complex cooperative networks involving various types of organelles. Light microscopic and ultrastructural studies have revealed that mitochondria-endoplasmic reticulum (ER) contact sites (MERCSs) are abundant in various tissues and cell types. Indeed, MERCSs have been proposed to play critical roles in various biochemical and signaling functions such as Ca2+ homeostasis, lipid transfer, and regulation of organelle dynamics. While numerous proteins involved in these MERCS-dependent functions have been reported, how they coordinate and cooperate with each other has not yet been elucidated. In this review, we summarize the functions of mammalian proteins that localize at MERCSs and regulate their formation. We also discuss potential roles of the MERCS proteins in regulating multiple organelle contacts.
Collapse
Affiliation(s)
| | - Yusuke Hirabayashi
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
103
|
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
|
104
|
Bitarafan F, Khodaeian M, Amjadi Sardehaei E, Darvishi FZ, Almadani N, Nilipour Y, Garshasbi M. Identification of a novel MICU1 nonsense variant causes myopathy with extrapyramidal signs in an Iranian consanguineous family. Mol Cell Pediatr 2021; 8:6. [PMID: 33969448 PMCID: PMC8107061 DOI: 10.1186/s40348-021-00116-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 04/14/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ca2+ as a universal second messenger regulates basic biological functions including cell cycle, cell proliferation, cell differentiation, and cell death. Lack of the protein mitochondrial calcium uptake1 (MICU1), which has been regarded as a gatekeeper of Ca ions, leads to the abnormal mitochondrial Ca2+ handling, excessive production of reactive oxygen species (ROS), and increased cell death. Mutations in MICU1 gene causes a very rare neuromuscular disease, myopathy with extrapyramidal signs (MPXPS), due to primary alterations in mitochondrial calcium signaling which demonstrates the key role of mitochondrial Ca2+ uptake. To date, 13 variants have been reported in MICU1 gene in 44 patients presented with the vast spectrum of symptoms. CASE PRESENTATION Here, we report a 44-year-old Iranian patient presented with learning disability, muscle weakness, easy fatigability, reduced tendon reflexes, ataxia, gait disturbance, elevated hepatic transaminases, elevated serum creatine kinase (CK), and elevated lactate dehydrogenase (LDH). We identified a novel nonsense variant c.385C>T; p.(R129*) in MICU1 gene by whole exome sequencing (WES) and segregation analysis. CONCLUSIONS Our finding along with previous studies provides more evidence on the clinical presentation of the disease caused by pathogenic mutations in MICU1. Finding more variants and expanding the spectrum of the disease increases the diagnostic rate of molecular testing in screening of this kind of diseases and in turn improves the quality of counseling for at risk couples and helps them to minimize the risks of having affected children.
Collapse
Affiliation(s)
- Fatemeh Bitarafan
- Department of Biology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | | | | | | | - Navid Almadani
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Yalda Nilipour
- Pediatric Pathology Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Garshasbi
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
105
|
Nakamura T, Ogawa M, Kojima K, Takayanagi S, Ishihara S, Hattori K, Naguro I, Ichijo H. The mitochondrial Ca 2+ uptake regulator, MICU1, is involved in cold stress-induced ferroptosis. EMBO Rep 2021; 22:e51532. [PMID: 33822458 PMCID: PMC8097382 DOI: 10.15252/embr.202051532] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 02/08/2021] [Accepted: 02/15/2021] [Indexed: 12/11/2022] Open
Abstract
Ferroptosis has recently attracted much interest because of its relevance to human diseases such as cancer and ischemia-reperfusion injury. We have reported that prolonged severe cold stress induces lipid peroxidation-dependent ferroptosis, but the upstream mechanism remains unknown. Here, using genome-wide CRISPR screening, we found that a mitochondrial Ca2+ uptake regulator, mitochondrial calcium uptake 1 (MICU1), is required for generating lipid peroxide and subsequent ferroptosis under cold stress. Furthermore, the gatekeeping activity of MICU1 through mitochondrial calcium uniporter (MCU) is suggested to be indispensable for cold stress-induced ferroptosis. MICU1 is required for mitochondrial Ca2+ increase, hyperpolarization of the mitochondrial membrane potential (MMP), and subsequent lipid peroxidation under cold stress. Collectively, these findings suggest that the MICU1-dependent mitochondrial Ca2+ homeostasis-MMP hyperpolarization axis is involved in cold stress-induced lipid peroxidation and ferroptosis.
Collapse
Affiliation(s)
- Toshitaka Nakamura
- Laboratory of Cell SignalingGraduate School of Pharmaceutical SciencesThe University of TokyoTokyoJapan
| | - Motoyuki Ogawa
- Laboratory of Cell SignalingGraduate School of Pharmaceutical SciencesThe University of TokyoTokyoJapan
| | - Kazuki Kojima
- Laboratory of Cell SignalingGraduate School of Pharmaceutical SciencesThe University of TokyoTokyoJapan
| | - Saki Takayanagi
- Laboratory of Cell SignalingGraduate School of Pharmaceutical SciencesThe University of TokyoTokyoJapan
| | - Shunya Ishihara
- Laboratory of Cell SignalingGraduate School of Pharmaceutical SciencesThe University of TokyoTokyoJapan
| | - Kazuki Hattori
- Laboratory of Cell SignalingGraduate School of Pharmaceutical SciencesThe University of TokyoTokyoJapan
| | - Isao Naguro
- Laboratory of Cell SignalingGraduate School of Pharmaceutical SciencesThe University of TokyoTokyoJapan
| | - Hidenori Ichijo
- Laboratory of Cell SignalingGraduate School of Pharmaceutical SciencesThe University of TokyoTokyoJapan
| |
Collapse
|
106
|
Belosludtsev KN, Sharipov RR, Boyarkin DP, Belosludtseva NV, Dubinin MV, Krasilnikova IA, Bakaeva ZV, Zgodova AE, Pinelis VG, Surin AM. The effect of DS16570511, a new inhibitor of mitochondrial calcium uniporter, on calcium homeostasis, metabolism, and functional state of cultured cortical neurons and isolated brain mitochondria. Biochim Biophys Acta Gen Subj 2021; 1865:129847. [PMID: 33453305 DOI: 10.1016/j.bbagen.2021.129847] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/20/2020] [Accepted: 01/11/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Disorders of mitochondrial Ca2+ homeostasis play a key role in the glutamate excitotoxicity of brain neurons. DS16570511 (DS) is a new penetrating inhibitor of mitochondrial Ca2+ uniporter complex (MCUC). The paper examines the effects of DS on the cultivated cortical neurons and isolated mitochondria of the rat brain. METHODS The functions of neurons and mitochondria were examined using fluorescence microscopy, XF24 microplate-based сell respirometry, ion-selective microelectrodes, spectrophotometry, and polarographic technique. RESULTS At the doses of 30 and 45 μM, DS reliably slowed down the onset of glutamate-induced delayed calcium deregulation of neurons and suppressed their death. 30 μM DS caused hyperpolarization of mitochondria of resting neurons, and 45 μM DS temporarily depolarized neuronal mitochondria. It was also demonstrated that 30-60 μM DS stimulated cellular respiration. DS was shown to suppress Ca2+ uptake by isolated brain mitochondria. In addition, DS inhibited ADP-stimulated mitochondrial respiration and ADP-induced decrease in the mitochondrial membrane potential. It was found that DS inhibited the activity of complex II of the respiratory chain. In the presence of Ca2+, high DS concentrations caused a collapse of the mitochondrial membrane potential. CONCLUSIONS The data obtained indicate that, in addition to the inhibition of MCUC, DS affects the main energy-transducing functions of mitochondria. GENERAL SIGNIFICANCE The using DS as a tool for studying MCUC and its functional role in neuronal cells should be done with care, bearing in mind multiple effects of DS, a proper evaluation of which would require multivariate analysis.
Collapse
Affiliation(s)
- Konstantin N Belosludtsev
- Mari State University, pl. Lenina 1, Yoshkar-Ola, 424001, Russia; Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, Pushchino, 142290, Russia.
| | - Rinat R Sharipov
- Institute of General Pathology and Pathophysiology, Baltiyskaya st. 8, Moscow, 125315, Russia
| | - Dmitry P Boyarkin
- National Medical Research Center for Children's Health, Lomonosovsky pr. 2, Moscow, 119296, Russia
| | - Natalia V Belosludtseva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, Pushchino, 142290, Russia
| | | | - Irina A Krasilnikova
- National Medical Research Center for Children's Health, Lomonosovsky pr. 2, Moscow, 119296, Russia
| | - Zanda V Bakaeva
- National Medical Research Center for Children's Health, Lomonosovsky pr. 2, Moscow, 119296, Russia
| | - Arina E Zgodova
- National Medical Research Center for Children's Health, Lomonosovsky pr. 2, Moscow, 119296, Russia
| | - Vsevolod G Pinelis
- National Medical Research Center for Children's Health, Lomonosovsky pr. 2, Moscow, 119296, Russia
| | - Alexander M Surin
- Institute of General Pathology and Pathophysiology, Baltiyskaya st. 8, Moscow, 125315, Russia; National Medical Research Center for Children's Health, Lomonosovsky pr. 2, Moscow, 119296, Russia
| |
Collapse
|
107
|
Ruggiero A, Katsenelson M, Slutsky I. Mitochondria: new players in homeostatic regulation of firing rate set points. Trends Neurosci 2021; 44:605-618. [PMID: 33865626 DOI: 10.1016/j.tins.2021.03.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 02/10/2021] [Accepted: 03/11/2021] [Indexed: 10/21/2022]
Abstract
Neural circuit functions are stabilized by homeostatic processes at long timescales in response to changes in behavioral states, experience, and learning. However, it remains unclear which specific physiological variables are being stabilized and which cellular or neural network components compose the homeostatic machinery. At this point, most evidence suggests that the distribution of firing rates among neurons in a neuronal circuit is the key variable that is maintained around a set-point value in a process called 'firing rate homeostasis.' Here, we review recent findings that implicate mitochondria as central players in mediating firing rate homeostasis. While mitochondria are known to regulate neuronal variables such as synaptic vesicle release or intracellular calcium concentration, the mitochondrial signaling pathways that are essential for firing rate homeostasis remain largely unknown. We used basic concepts of control theory to build a framework for classifying possible components of the homeostatic machinery that stabilizes firing rate, and we particularly emphasize the potential role of sleep and wakefulness in this homeostatic process. This framework may facilitate the identification of new homeostatic pathways whose malfunctions drive instability of neural circuits in distinct brain disorders.
Collapse
Affiliation(s)
- Antonella Ruggiero
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Maxim Katsenelson
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Inna Slutsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel.
| |
Collapse
|
108
|
Bustos G, Ahumada-Castro U, Silva-Pavez E, Puebla A, Lovy A, Cesar Cardenas J. The ER-mitochondria Ca 2+ signaling in cancer progression: Fueling the monster. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 363:49-121. [PMID: 34392932 DOI: 10.1016/bs.ircmb.2021.03.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer is a leading cause of death worldwide. All major tumor suppressors and oncogenes are now recognized to have fundamental connections with metabolic pathways. A hallmark feature of cancer cells is a reprogramming of their metabolism even when nutrients are available. Increasing evidence indicates that most cancer cells rely on mitochondrial metabolism to sustain their energetic and biosynthetic demands. Mitochondria are functionally and physically coupled to the endoplasmic reticulum (ER), the major calcium (Ca2+) storage organelle in mammalian cells, through special domains known as mitochondria-ER contact sites (MERCS). In this domain, the release of Ca2+ from the ER is mainly regulated by inositol 1,4,5-trisphosphate (IP3) receptors (IP3Rs), a family of Ca2+ release channels activated by the ligand IP3. IP3R mediated Ca2+ release is transferred to mitochondria through the mitochondrial Ca2+ uniporter (MCU). Once in the mitochondrial matrix, Ca2+ activates several proteins that stimulate mitochondrial performance. The role of IP3R and MCU in cancer, as well as the other proteins that enable the Ca2+ communication between these two organelles is just beginning to be understood. Here, we describe the function of the main players of the ER mitochondrial Ca2+ communication and discuss how this particular signal may contribute to the rise and development of cancer traits.
Collapse
Affiliation(s)
- Galdo Bustos
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Ulises Ahumada-Castro
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Eduardo Silva-Pavez
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Andrea Puebla
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Alenka Lovy
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile; Department of Neuroscience, Center for Neuroscience Research, Tufts School of Medicine, Boston, MA, United States.
| | - J Cesar Cardenas
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile; Buck Institute for Research on Aging, Novato, CA, United States; Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA, United States.
| |
Collapse
|
109
|
Zhao H, Pan X. Mitochondrial Ca 2+ and cell cycle regulation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 362:171-207. [PMID: 34253295 DOI: 10.1016/bs.ircmb.2021.02.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
It has been demonstrated for more than 40 years that intracellular calcium (Ca2+) controls a variety of cellular functions, including mitochondrial metabolism and cell proliferation. Cytosolic Ca2+ fluctuation during key stages of the cell cycle can lead to mitochondrial Ca2+ uptake and subsequent activation of mitochondrial oxidative phosphorylation and a range of signaling. However, the relationship between mitochondrial Ca2+ and cell cycle progression has long been neglected because the molecule responsible for Ca2+ uptake has been unknown. Recently, the identification of the mitochondrial Ca2+ uniporter (MCU) has led to key advances. With improved Ca2+ imaging and detection, effects of MCU-mediated mitochondrial Ca2+ have been observed at different stages of the cell cycle. Elevated Ca2+ signaling boosts ATP and ROS production, remodels cytosolic Ca2+ pathways and reprograms cell fate-determining networks. These findings suggest that manipulating mitochondrial Ca2+ signaling may serve as a potential strategy in the control of many crucial biological events, such as tumor development and cell division in hematopoietic stem cells (HSCs). In this review, we summarize the current understanding of the role of mitochondrial Ca2+ signaling during different stages of the cell cycle and highlight the potential physiological and pathological significance of mitochondrial Ca2+ signaling.
Collapse
Affiliation(s)
- Haixin Zhao
- State Key Laboratory of Experimental Haematology, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xin Pan
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China.
| |
Collapse
|
110
|
Guo L. Mitochondria and the permeability transition pore in cancer metabolic reprogramming. Biochem Pharmacol 2021; 188:114537. [PMID: 33811907 DOI: 10.1016/j.bcp.2021.114537] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023]
Abstract
Mitochondria are a major source of ATP provision as well as cellular suicidal weapon store. Accumulating evidences demonstrate that mitochondrial bioenergetics, biosynthesis and signaling are important mediators of tumorigenesis. Metabolic plasticity enables cancer cell reprogramming to cope with cellular and environmental alterations, a process requires mitochondria biology. Mitochondrial metabolism emerges to be a promising arena for cancer therapeutic targets. The permeability transition pore (PTP) participates in physiological Ca2+ and ROS homeostasis as well as cell death depending on the open state. The hypothesis that PTP forms from F-ATP synthase provides clues to the potential collaborative role of mitochondrial respiration and PTP in regulating cancer cell fate and metabolic reprogramming.
Collapse
Affiliation(s)
- Lishu Guo
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China.
| |
Collapse
|
111
|
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
|
112
|
Giamogante F, Poggio E, Barazzuol L, Covallero A, Calì T. Apoptotic signals at the endoplasmic reticulum-mitochondria interface. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 126:307-343. [PMID: 34090618 DOI: 10.1016/bs.apcsb.2021.02.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The maintenance of cellular homeostasis involves the participation of multiple organelles, such as the endoplasmic reticulum (ER) and mitochondria. Specifically, ER plays a key role in calcium (Ca2+) storage, lipid synthesis, protein folding, and assembly, while mitochondria are the "energy factories" and provide energy to drive intracellular processes. Hence, alteration in ER or mitochondrial homeostasis has detrimental effects on cell survival, being linked to the triggering of apoptosis, a programmed form of cell death. Besides, ER stress conditions affect mitochondria functionality and vice-versa, as ER and mitochondria communicate via mitochondria-associated ER membranes (MAMs) to carry out a number of fundamental cellular functions. It is not surprising, thus, that also MAMs perturbations are involved in the regulation of apoptosis. This chapter intends to accurately discuss the involvement of MAMs in apoptosis, highlighting their crucial role in controlling this delicate cellular process.
Collapse
Affiliation(s)
- Flavia Giamogante
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Elena Poggio
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Lucia Barazzuol
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Alberto Covallero
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Tito Calì
- Department of Biomedical Sciences, University of Padova, Padova, Italy.
| |
Collapse
|
113
|
Chen LT, Xu TT, Qiu YQ, Liu NY, Ke XY, Fang L, Yan JP, Zhu DY. Homocysteine induced a calcium-mediated disruption of mitochondrial function and dynamics in endothelial cells. J Biochem Mol Toxicol 2021; 35:e22737. [PMID: 33751715 DOI: 10.1002/jbt.22737] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 12/03/2020] [Accepted: 01/20/2021] [Indexed: 12/24/2022]
Abstract
Homocysteine (Hcy) is a sulfur-containing amino acid that originated in methionine metabolism and the elevated level of Hcy in plasma is considered to be an independent risk factor for cardiovascular diseases (CVD). Endothelial dysfunction plays a major role in the development of CVD, while the potential mechanism of Hcy-induced endothelial dysfunction is still unclear. Here, in Hcy-treated endothelial cells, we observed the destruction of mitochondrial morphology and the decline of mitochondrial membrane potential. Meanwhile, the level of ATP was reduced and the reactive oxygen species was increased. The expressions of dynamin-related protein 1 (Drp1) and phosphate-Drp1 (Ser616) were upregulated, whereas the expression of mitofusin 2 was inhibited by Hcy treatment. These findings suggested that Hcy not only triggered mitochondrial dysfunction but also incurred an imbalance of mitochondrial dynamics in endothelial cells. The expression of mitochondrial calcium uniporter (MCU) was activated by Hcy, contributing to calcium transferring into mitochondria. Interestingly, the formation of mitochondria-associated membranes (MAMs) was increased in endothelial cells after Hcy administration. The inositol 1,4,5-triphosphate receptor (IP3R)-glucose-regulated protein 75 (Grp75)-voltage-dependent anion channel (VDAC) complex, which was enriched in MAMs, was also increased. The accumulation of mitochondrial calcium could be blocked by inhibiting with the IP3R inhibitor Xestospongin C (XeC) in Hcy-treated cells. Then, we confirmed that the mitochondrial dysfunction and the increased mitochondrial fission induced by Hcy could be attenuated after Hcy and XeC co-treatment. In conclusion, Hcy-induced mitochondrial dysfunction and dynamics disorder in endothelial cells were mainly related to the increase of calcium as a result of the upregulated expressions of the MCU and the IP3R-Grp75-VDAC complex in MAMs.
Collapse
Affiliation(s)
- Li-Ting Chen
- Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou, China
| | - Ting-Ting Xu
- Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou, China
| | - Ya-Qing Qiu
- Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou, China
| | - Nuo-Ya Liu
- Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou, China
| | - Xin-Yu Ke
- Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou, China
| | - Lu Fang
- Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou, China
| | - Jie-Ping Yan
- Department of Pharmacy, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Dan-Yan Zhu
- Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou, China
| |
Collapse
|
114
|
Di Marco G, Vallese F, Jourde B, Bergsdorf C, Sturlese M, De Mario A, Techer-Etienne V, Haasen D, Oberhauser B, Schleeger S, Minetti G, Moro S, Rizzuto R, De Stefani D, Fornaro M, Mammucari C. A High-Throughput Screening Identifies MICU1 Targeting Compounds. Cell Rep 2021; 30:2321-2331.e6. [PMID: 32075766 PMCID: PMC7034061 DOI: 10.1016/j.celrep.2020.01.081] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/08/2020] [Accepted: 01/22/2020] [Indexed: 01/01/2023] Open
Abstract
Mitochondrial Ca2+ uptake depends on the mitochondrial calcium uniporter (MCU) complex, a highly selective channel of the inner mitochondrial membrane (IMM). Here, we screen a library of 44,000 non-proprietary compounds for their ability to modulate mitochondrial Ca2+ uptake. Two of them, named MCU-i4 and MCU-i11, are confirmed to reliably decrease mitochondrial Ca2+ influx. Docking simulations reveal that these molecules directly bind a specific cleft in MICU1, a key element of the MCU complex that controls channel gating. Accordingly, in MICU1-silenced or deleted cells, the inhibitory effect of the two compounds is lost. Moreover, MCU-i4 and MCU-i11 fail to inhibit mitochondrial Ca2+ uptake in cells expressing a MICU1 mutated in the critical amino acids that forge the predicted binding cleft. Finally, these compounds are tested ex vivo, revealing a primary role for mitochondrial Ca2+ uptake in muscle growth. Overall, MCU-i4 and MCU-i11 represent leading molecules for the development of MICU1-targeting drugs. An HTS identifies MCU-i4 and MCU-i11 as negative modulators of the MCU MCU-i4 and MCU-i11 bind MICU1 MICU1 is required for the activity of MCU-i4 and MCU-i11 MCU-i4 and MCU-i11 impair muscle cell growth
Collapse
Affiliation(s)
- Giulia Di Marco
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
| | - Francesca Vallese
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
| | - Benjamin Jourde
- Novartis Institutes for Biomedical Research, Novartis Campus, 4056 Basel, Switzerland
| | - Christian Bergsdorf
- Novartis Institutes for Biomedical Research, Novartis Campus, 4056 Basel, Switzerland
| | - Mattia Sturlese
- Molecular Modeling Section, Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35131 Padua, Italy
| | - Agnese De Mario
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
| | | | - Dorothea Haasen
- Novartis Institutes for Biomedical Research, Novartis Campus, 4056 Basel, Switzerland
| | - Berndt Oberhauser
- Novartis Institutes for Biomedical Research, Novartis Campus, 4056 Basel, Switzerland
| | - Simone Schleeger
- Novartis Institutes for Biomedical Research, Novartis Campus, 4056 Basel, Switzerland
| | - Giulia Minetti
- Novartis Institutes for Biomedical Research, Novartis Campus, 4056 Basel, Switzerland
| | - Stefano Moro
- Molecular Modeling Section, Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35131 Padua, Italy
| | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
| | - Diego De Stefani
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
| | - Mara Fornaro
- Novartis Institutes for Biomedical Research, Novartis Campus, 4056 Basel, Switzerland.
| | - Cristina Mammucari
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy.
| |
Collapse
|
115
|
Mitochondrial Calcium Signaling in Pancreatic β-Cell. Int J Mol Sci 2021; 22:ijms22052515. [PMID: 33802289 PMCID: PMC7959128 DOI: 10.3390/ijms22052515] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/22/2021] [Accepted: 02/26/2021] [Indexed: 12/13/2022] Open
Abstract
Accumulation of calcium in energized mitochondria of pancreatic β-cells is emerging as a crucial process for pancreatic β-cell function. β-cell mitochondria sense and shape calcium signals, linking the metabolism of glucose and other secretagogues to the generation of signals that promote insulin secretion during nutrient stimulation. Here, we describe the role of mitochondrial calcium signaling in pancreatic β-cell function. We report the latest pharmacological and genetic findings, including the first mitochondrial calcium-targeted intervention strategies developed to modulate pancreatic β-cell function and their potential relevance in the context of diabetes.
Collapse
|
116
|
Tanwar J, Singh JB, Motiani RK. Molecular machinery regulating mitochondrial calcium levels: The nuts and bolts of mitochondrial calcium dynamics. Mitochondrion 2021; 57:9-22. [PMID: 33316420 PMCID: PMC7610953 DOI: 10.1016/j.mito.2020.12.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 11/18/2020] [Accepted: 12/03/2020] [Indexed: 02/06/2023]
Abstract
Mitochondria play vital role in regulating the cellular energetics and metabolism. Further, it is a signaling hub for cell survival and apoptotic pathways. One of the key determinants that calibrate both cellular energetics and survival functions is mitochondrial calcium (Ca2+) dynamics. Mitochondrial Ca2+ regulates three Ca2+-sensitive dehydrogenase enzymes involved in tricarboxylic acid cycle (TCA) cycle thereby directly controlling ATP synthesis. On the other hand, excessive Ca2+ concentration within the mitochondrial matrix elevates mitochondrial reactive oxygen species (mROS) levels and causes mitochondrial membrane depolarization. This leads to opening of the mitochondrial permeability transition pore (mPTP) and release of cytochrome c into cytosol eventually triggering apoptosis. Therefore, it is critical for cell to maintain mitochondrial Ca2+ concentration. Since cells can neither synthesize nor metabolize Ca2+, it is the dynamic interplay of Ca2+ handling proteins involved in mitochondrial Ca2+ influx and efflux that take the center stage. In this review we would discuss the key molecular machinery regulating mitochondrial Ca2+ concentration. We would focus on the channel complex involved in bringing Ca2+ into mitochondrial matrix i.e. Mitochondrial Ca2+ Uniporter (MCU) and its key regulators Mitochondrial Ca2+ Uptake proteins (MICU1, 2 and 3), MCU regulatory subunit b (MCUb), Essential MCU Regulator (EMRE) and Mitochondrial Ca2+ Uniporter Regulator 1 (MCUR1). Further, we would deliberate on major mitochondrial Ca2+ efflux proteins i.e. Mitochondrial Na+/Ca2+/Li+ exchanger (NCLX) and Leucine zipper EF hand-containing transmembrane1 (Letm1). Moreover, we would highlight the physiological functions of these proteins and discuss their relevance in human pathophysiology. Finally, we would highlight key outstanding questions in the field.
Collapse
Affiliation(s)
- Jyoti Tanwar
- CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi 10025, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Jaya Bharti Singh
- Laboratory of Calciomics and Systemic Pathophysiology (LCSP), Regional Centre for Biotechnology (RCB), Faridabad, Delhi-NCR, India
| | - Rajender K Motiani
- Laboratory of Calciomics and Systemic Pathophysiology (LCSP), Regional Centre for Biotechnology (RCB), Faridabad, Delhi-NCR, India.
| |
Collapse
|
117
|
Docampo R, Vercesi AE, Huang G, Lander N, Chiurillo MA, Bertolini M. Mitochondrial Ca 2+ homeostasis in trypanosomes. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 362:261-289. [PMID: 34253297 PMCID: PMC10424509 DOI: 10.1016/bs.ircmb.2021.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Mitochondrial calcium ion (Ca2+) uptake is important for buffering cytosolic Ca2+ levels, for regulating cell bioenergetics, and for cell death and autophagy. Ca2+ uptake is mediated by a mitochondrial Ca2+ uniporter (MCU) and the discovery of this channel in trypanosomes has been critical for the identification of the molecular nature of the channel in all eukaryotes. However, the trypanosome uniporter, which has been studied in detail in Trypanosoma cruzi, the agent of Chagas disease, and T. brucei, the agent of human and animal African trypanosomiasis, has lineage-specific adaptations which include the lack of some homologues to mammalian subunits, and the presence of unique subunits. Here, we review newly emerging insights into the role of mitochondrial Ca2+ homeostasis in trypanosomes, the composition of the uniporter, its functional characterization, and its role in general physiology.
Collapse
Affiliation(s)
- Roberto Docampo
- Center for Tropical and Emerging Global Diseases and Department of Cellular Biology, University of Georgia, Athens, GA, United States.
| | - Anibal E Vercesi
- Departamento de Patologia Clinica, Universidade Estadual de Campinas, São Paulo, Brazil
| | - Guozhong Huang
- Center for Tropical and Emerging Global Diseases and Department of Cellular Biology, University of Georgia, Athens, GA, United States
| | - Noelia Lander
- Center for Tropical and Emerging Global Diseases and Department of Cellular Biology, University of Georgia, Athens, GA, United States
| | - Miguel A Chiurillo
- Center for Tropical and Emerging Global Diseases and Department of Cellular Biology, University of Georgia, Athens, GA, United States
| | - Mayara Bertolini
- Center for Tropical and Emerging Global Diseases and Department of Cellular Biology, University of Georgia, Athens, GA, United States
| |
Collapse
|
118
|
Mitochondrial metabolism and calcium homeostasis in the development of NAFLD leading to hepatocellular carcinoma. Mitochondrion 2021; 58:24-37. [PMID: 33581332 DOI: 10.1016/j.mito.2021.01.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 01/21/2021] [Accepted: 01/25/2021] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a metabolic syndrome characterized by excessive accumulation of hepatic lipid droplets. The disease progresses with steatosis as the premise for hepatocytic damage and tissue scarring, often culminating in hepatocellular carcinoma (HCC). Perturbations in mitochondrial metabolism and energetics were found to be associated with, and often instrumental in various stages of this progression. Functional impairment of the mitochondria affects all aspects of cellular functioning and a particularly important one is calcium signalling. Changes in mitochondrial calcium specifically in hepatocytes of a fatty liver, is reflected by alterations in calcium signalling as well as calcium transporter activities. This deranged Ca2+ homeostasis aids in even more uptake of lipids into the mitochondria and a shift in equilibrium, both metabolically as well as in terms of energy production, leading to completely altered cellular states. These alterations have been reviewed as a perspective to understand the disease progression through NAFLD leading to HCC.
Collapse
|
119
|
Abstract
Mitochondria are responsible for ATP production but are also known as regulators of cell death, and mitochondrial matrix Ca2+ is a key modulator of both ATP production and cell death. Although mitochondrial Ca2+ uptake and efflux have been studied for over 50 years, it is only in the past decade that the proteins responsible for mitochondrial Ca2+ uptake and efflux have been identified. The identification of the mitochondrial Ca2+ uniporter (MCU) led to an explosion of studies identifying regulators of the MCU. The levels of these regulators vary in a tissue- and disease-specific manner, providing new insight into how mitochondrial Ca2+ is regulated. This review focuses on the proteins responsible for mitochondrial transport and what we have learned from mouse studies with genetic alterations in these proteins.
Collapse
Affiliation(s)
- Elizabeth Murphy
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;
| | - Charles Steenbergen
- Department of Pathology, Johns Hopkins Medicine, Baltimore, Maryland 21287, USA
| |
Collapse
|
120
|
Boyman L, Greiser M, Lederer WJ. Calcium influx through the mitochondrial calcium uniporter holocomplex, MCU cx. J Mol Cell Cardiol 2021; 151:145-154. [PMID: 33147447 PMCID: PMC7880866 DOI: 10.1016/j.yjmcc.2020.10.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 10/20/2020] [Accepted: 10/28/2020] [Indexed: 12/11/2022]
Abstract
Ca2+ flux into the mitochondrial matrix through the MCU holocomplex (MCUcx) has recently been measured quantitatively and with milliseconds resolution for the first time under physiological conditions in both heart and skeletal muscle. Additionally, the dynamic levels of Ca2+ in the mitochondrial matrix ([Ca2+]m) of cardiomyocytes were measured as it was controlled by the balance between influx of Ca2+ into the mitochondrial matrix through MCUcx and efflux through the mitochondrial Na+ / Ca2+ exchanger (NCLX). Under these conditions [Ca2+]m was shown to regulate ATP production by the mitochondria at only a few critical sites. Additional functions attributed to [Ca2+]m continue to be reported in the literature. Here we review the new findings attributed to MCUcx function and provide a framework for understanding and investigating mitochondrial Ca2+ influx features, many of which remain controversial. The properties and functions of the MCUcx subunits that constitute the holocomplex are challenging to tease apart. Such distinct subunits include EMRE, MCUR1, MICUx (i.e. MICU1, MICU2, MICU3), and the pore-forming subunits (MCUpore). Currently, the specific set of functions of each subunit remains non-quantitative and controversial. The more contentious issues are discussed in the context of the newly measured native MCUcx Ca2+ flux from heart and skeletal muscle. These MCUcx Ca2+ flux measurements have been shown to be a highly-regulated, tissue-specific with femto-Siemens Ca2+ conductances and with distinct extramitochondrial Ca2+ ([Ca2+]i) dependencies. These data from cardiac and skeletal muscle mitochondria have been examined quantitatively for their threshold [Ca2+]i levels and for hypothesized gatekeeping function and are discussed in the context of model cell (e.g. HeLa, MEF, HEK293, COS7 cells) measurements. Our new findings on MCUcx dependent matrix [Ca2+]m signaling provide a quantitative basis for on-going and new investigations of the roles of MCUcx in cardiac function ranging from metabolic fuel selection, capillary blood-flow control and the pathological activation of the mitochondrial permeability transition pore (mPTP). Additionally, this review presents the use of advanced new methods that can be readily adapted by any investigator to enable them to carry out quantitative Ca2+ measurements in mitochondria while controlling the inner mitochondrial membrane potential, ΔΨm.
Collapse
Affiliation(s)
- Liron Boyman
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, USA; The Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Maura Greiser
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, USA; The Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - W Jonathan Lederer
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, USA; The Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
121
|
Márta K, Hasan P, Rodríguez-Prados M, Paillard M, Hajnóczky G. Pharmacological inhibition of the mitochondrial Ca 2+ uniporter: Relevance for pathophysiology and human therapy. J Mol Cell Cardiol 2021; 151:135-144. [PMID: 33035551 PMCID: PMC7880870 DOI: 10.1016/j.yjmcc.2020.09.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 09/26/2020] [Accepted: 09/28/2020] [Indexed: 12/20/2022]
Abstract
Mitochondrial Ca2+ uptake has long been considered crucial for meeting the fluctuating energy demands of cells in the heart and other tissues. Increases in mitochondrial matrix [Ca2+] drive mitochondrial ATP production via stimulation of Ca2+-sensitive dehydrogenases. Mitochondria-targeted sensors have revealed mitochondrial matrix [Ca2+] rises that closely follow the cytoplasmic [Ca2+] signals in many paradigms. Mitochondrial Ca2+ uptake is mediated by the Ca2+ uniporter (mtCU). Pharmacological manipulation of the mtCU is potentially key to understanding its physiological significance, but no specific, cell-permeable inhibitors were identified. In the past decade, as the molecular identity of the mtCU was brought to light, efforts have focused on genetic targeting. However, in the cells/animals that are able to survive impaired mtCU function, robust compensatory changes were found in the mtCU as well as other mechanisms. Thus, the discovery, through chemical library screens on normal and mtCU-deficient cells, of new small-molecule inhibitors with improved cell permeability and specificity might offer a better chance to test the relevance of mitochondrial Ca2+ uptake. Success with the development of small molecule mtCU inhibitors is also expected to have clinical impact, considering the growing evidence for the role of mitochondrial Ca2+ uptake in a variety of diseases, including heart attack, stroke and various neurodegenerative disorders. Here, we review the progress in pharmacological targeting of mtCU and illustrate the challenges in this field using data obtained with MCU-i11, a new small molecule inhibitor.
Collapse
Affiliation(s)
- Katalin Márta
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Prottoy Hasan
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Macarena Rodríguez-Prados
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Melanie Paillard
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA; Univ-Lyon, CarMeN Laboratory, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, HCL, 69500 Bron, France
| | - György Hajnóczky
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
122
|
Saxena R, Saribas S, Jadiya P, Tomar D, Kaminski R, Elrod JW, Safak M. Human neurotropic polyomavirus, JC virus, agnoprotein targets mitochondrion and modulates its functions. Virology 2021; 553:135-153. [PMID: 33278736 PMCID: PMC7847276 DOI: 10.1016/j.virol.2020.11.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 11/12/2020] [Indexed: 01/18/2023]
Abstract
JC virus encodes an important regulatory protein, known as Agnoprotein (Agno). We have recently reported Agno's first protein-interactome with its cellular partners revealing that it targets various cellular networks and organelles, including mitochondria. Here, we report further characterization of the functional consequences of its mitochondrial targeting and demonstrated its co-localization with the mitochondrial networks and with the mitochondrial outer membrane. The mitochondrial targeting sequence (MTS) of Agno and its dimerization domain together play major roles in this targeting. Data also showed alterations in various mitochondrial functions in Agno-positive cells; including a significant reduction in mitochondrial membrane potential, respiration rates and ATP production. In contrast, a substantial increase in ROS production and Ca2+ uptake by the mitochondria were also observed. Finally, findings also revealed a significant decrease in viral replication when Agno MTS was deleted, highlighting a role for MTS in the function of Agno during the viral life cycle.
Collapse
Affiliation(s)
- Reshu Saxena
- Department of Neuroscience, Laboratory of Molecular Neurovirology, MERB-757, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Sami Saribas
- Department of Neuroscience, Laboratory of Molecular Neurovirology, MERB-757, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Pooja Jadiya
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, USA
| | - Dhanendra Tomar
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, USA
| | - Rafal Kaminski
- Department of Neuroscience, Laboratory of Molecular Neurovirology, MERB-757, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - John W Elrod
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, USA
| | - Mahmut Safak
- Department of Neuroscience, Laboratory of Molecular Neurovirology, MERB-757, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA, 19140, USA.
| |
Collapse
|
123
|
Kohlschmidt N, Elbracht M, Czech A, Häusler M, Phan V, Töpf A, Huang KT, Bartok A, Eggermann K, Zippel S, Eggermann T, Freier E, Groß C, Lochmüller H, Horvath R, Hajnóczky G, Weis J, Roos A. Molecular pathophysiology of human MICU1 deficiency. Neuropathol Appl Neurobiol 2021; 47:840-855. [PMID: 33428302 DOI: 10.1111/nan.12694] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 09/07/2020] [Accepted: 09/14/2020] [Indexed: 12/20/2022]
Abstract
AIMS MICU1 encodes the gatekeeper of the mitochondrial Ca2+ uniporter, MICU1 and biallelic loss-of-function mutations cause a complex, neuromuscular disorder in children. Although the role of the protein is well understood, the precise molecular pathophysiology leading to this neuropaediatric phenotype has not been fully elucidated. Here we aimed to obtain novel insights into MICU1 pathophysiology. METHODS Molecular genetic studies along with proteomic profiling, electron-, light- and Coherent anti-Stokes Raman scattering microscopy and immuno-based studies of protein abundances and Ca2+ transport studies were employed to examine the pathophysiology of MICU1 deficiency in humans. RESULTS We describe two patients carrying MICU1 mutations, two nonsense (c.52C>T; p.(Arg18*) and c.553C>T; p.(Arg185*)) and an intragenic exon 2-deletion presenting with ataxia, developmental delay and early onset myopathy, clinodactyly, attention deficits, insomnia and impaired cognitive pain perception. Muscle biopsies revealed signs of dystrophy and neurogenic atrophy, severe mitochondrial perturbations, altered Golgi structure, vacuoles and altered lipid homeostasis. Comparative mitochondrial Ca2+ transport and proteomic studies on lymphoblastoid cells revealed that the [Ca2+ ] threshold and the cooperative activation of mitochondrial Ca2+ uptake were lost in MICU1-deficient cells and that 39 proteins were altered in abundance. Several of those proteins are linked to mitochondrial dysfunction and/or perturbed Ca2+ homeostasis, also impacting on regular cytoskeleton (affecting Spectrin) and Golgi architecture, as well as cellular survival mechanisms. CONCLUSIONS Our findings (i) link dysregulation of mitochondrial Ca2+ uptake with muscle pathology (including perturbed lipid homeostasis and ER-Golgi morphology), (ii) support the concept of a functional interplay of ER-Golgi and mitochondria in lipid homeostasis and (iii) reveal the vulnerability of the cellular proteome as part of the MICU1-related pathophysiology.
Collapse
Affiliation(s)
| | - Miriam Elbracht
- Institute of Human Genetics, RWTH Aachen University Hospital, Aachen, Germany
| | - Artur Czech
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V, Dortmund, Germany
| | - Martin Häusler
- Division of Neuropediatrics and Social Pediatrics, Department of Pediatrics, RWTH Aachen University Hospital, Aachen, Germany
| | - Vietxuan Phan
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V, Dortmund, Germany
| | - Ana Töpf
- Institute of Genetic Medicine, International Centre for Life, Central Parkway, Newcastle upon Tyne, UK
| | - Kai-Ting Huang
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Adam Bartok
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Katja Eggermann
- Institute of Human Genetics, RWTH Aachen University Hospital, Aachen, Germany
| | | | - Thomas Eggermann
- Institute of Human Genetics, RWTH Aachen University Hospital, Aachen, Germany
| | - Erik Freier
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V, Dortmund, Germany
| | - Claudia Groß
- Institute of Clinical Genetics and Tumour Genetics, Bonn, Germany
| | - Hanns Lochmüller
- Department of Neuropediatrics and Muscle Disorders, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany.,Centro Nacional de Análisis Genómico, Center for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain.,Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada.,Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, Ontario, Canada
| | - Rita Horvath
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, UK
| | - György Hajnóczky
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Joachim Weis
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Andreas Roos
- Department of Neuropediatrics, Centre for Neuromuscular Disorders in Children, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
124
|
Inoshita T, Imai Y. Cytosolic and Mitochondrial Ca 2+ Imaging in Drosophila Dopaminergic Neurons. Methods Mol Biol 2021; 2322:207-214. [PMID: 34043206 DOI: 10.1007/978-1-0716-1495-2_20] [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] [Indexed: 06/12/2023]
Abstract
The ATP-producing organelle mitochondrion controls cellular or synaptic Ca2+ concentrations through temporal uptake of Ca2+ outside of the mitochondria. Although intracellular Ca2+ influx occurs during neuronal activity, a persistently higher concentration of intracellular Ca2+ is neurotoxic. Healthy mitochondria ensure rapid Ca2+ uptake, which is necessary for proper neuronal activity. Mitochondrial Ca2+ buffering activity decreases in aged or sick neurons. In this chapter, we will introduce our protocol for evaluating Ca2+ buffering activity through the mitochondria during neuronal activity of dopaminergic neurons.
Collapse
Affiliation(s)
- Tsuyoshi Inoshita
- Department of Neurodegenerative and Demented Disorders, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuzuru Imai
- Department of Research for Parkinson's Disease, Juntendo University Graduate School of Medicine, Tokyo, Japan.
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan.
| |
Collapse
|
125
|
TSUBOI M, HIRABAYASHI Y. New insights into the regulation of synaptic transmission and plasticity by the endoplasmic reticulum and its membrane contacts. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2021; 97:559-572. [PMID: 34897182 PMCID: PMC8687855 DOI: 10.2183/pjab.97.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 10/18/2021] [Indexed: 06/14/2023]
Abstract
Mammalian neurons are highly compartmentalized yet very large cells. To provide each compartment with its distinct properties, metabolic homeostasis and molecular composition need to be precisely coordinated in a compartment-specific manner. Despite the importance of the endoplasmic reticulum (ER) as a platform for various biochemical reactions, such as protein synthesis, protein trafficking, and intracellular calcium control, the contribution of the ER to neuronal compartment-specific functions and plasticity remains elusive. Recent advances in the development of live imaging and serial scanning electron microscopy (sSEM) analysis have revealed that the neuronal ER is a highly dynamic organelle with compartment-specific structures. sSEM studies also revealed that the ER forms contacts with other membranes, such as the mitochondria and plasma membrane, although little is known about the functions of these ER-membrane contacts. In this review, we discuss the mechanisms and physiological roles of the ER structure and ER-mitochondria contacts in synaptic transmission and plasticity, thereby highlighting a potential link between organelle ultrastructure and neuronal functions.
Collapse
Affiliation(s)
- Masafumi TSUBOI
- Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | | |
Collapse
|
126
|
Hamilton J, Brustovetsky T, Brustovetsky N. The effect of mitochondrial calcium uniporter and cyclophilin D knockout on resistance of brain mitochondria to Ca 2+-induced damage. J Biol Chem 2021; 296:100669. [PMID: 33864812 PMCID: PMC8131324 DOI: 10.1016/j.jbc.2021.100669] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/10/2021] [Accepted: 04/13/2021] [Indexed: 12/14/2022] Open
Abstract
The mitochondrial calcium uniporter (MCU) and cyclophilin D (CyD) are key players in induction of the permeability transition pore (PTP), which leads to mitochondrial depolarization and swelling, the major signs of Ca2+-induced mitochondrial damage. Mitochondrial depolarization inhibits ATP production, whereas swelling results in the release of mitochondrial pro-apoptotic proteins. The extent to which simultaneous deletion of MCU and CyD inhibits PTP induction and prevents damage of brain mitochondria is not clear. Here, we investigated the effects of MCU and CyD deletion on the propensity for PTP induction using mitochondria isolated from the brains of MCU-KO, CyD-KO, and newly created MCU/CyD-double knockout (DKO) mice. Neither deletion of MCU nor of CyD affected respiration or membrane potential in mitochondria isolated from the brains of these mice. Mitochondria from MCU-KO and MCU/CyD-DKO mice displayed reduced Ca2+ uptake and diminished extent of PTP induction. The Ca2+ uptake by mitochondria from CyD-KO mice was increased compared with mitochondria from WT mice. Deletion of CyD prevented mitochondrial swelling and resulted in transient depolarization in response to Ca2+, but it did not prevent Ca2+-induced delayed mitochondrial depolarization. Mitochondria from MCU/CyD-DKO mice did not swell in response to Ca2+, but they did exhibit mild sustained depolarization. Dibucaine, an inhibitor of the Ca2+-activated mitochondrial phospholipase A2, attenuated and bovine serum albumin completely eliminated the sustained depolarization. This suggests the involvement of phospholipase A2 and free fatty acids. Thus, in addition to induction of the classical PTP, alternative deleterious mechanisms may contribute to mitochondrial damage following exposure to elevated Ca2+.
Collapse
Affiliation(s)
- James Hamilton
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Tatiana Brustovetsky
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Nickolay Brustovetsky
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| |
Collapse
|
127
|
Jung H, Kim SY, Canbakis Cecen FS, Cho Y, Kwon SK. Dysfunction of Mitochondrial Ca 2+ Regulatory Machineries in Brain Aging and Neurodegenerative Diseases. Front Cell Dev Biol 2020; 8:599792. [PMID: 33392190 PMCID: PMC7775422 DOI: 10.3389/fcell.2020.599792] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/06/2020] [Indexed: 12/20/2022] Open
Abstract
Calcium ions (Ca2+) play critical roles in neuronal processes, such as signaling pathway activation, transcriptional regulation, and synaptic transmission initiation. Therefore, the regulation of Ca2+ homeostasis is one of the most important processes underlying the basic cellular viability and function of the neuron. Multiple components, including intracellular organelles and plasma membrane Ca2+-ATPase, are involved in neuronal Ca2+ control, and recent studies have focused on investigating the roles of mitochondria in synaptic function. Numerous mitochondrial Ca2+ regulatory proteins have been identified in the past decade, with studies demonstrating the tissue- or cell-type-specific function of each component. The mitochondrial calcium uniporter and its binding subunits are major inner mitochondrial membrane proteins contributing to mitochondrial Ca2+ uptake, whereas the mitochondrial Na+/Ca2+ exchanger (NCLX) and mitochondrial permeability transition pore (mPTP) are well-studied proteins involved in Ca2+ extrusion. The level of cytosolic Ca2+ and the resulting characteristics of synaptic vesicle release properties are controlled via mitochondrial Ca2+ uptake and release at presynaptic sites, while in dendrites, mitochondrial Ca2+ regulation affects synaptic plasticity. During brain aging and the progress of neurodegenerative disease, mitochondrial Ca2+ mishandling has been observed using various techniques, including live imaging of Ca2+ dynamics. Furthermore, Ca2+ dysregulation not only disrupts synaptic transmission but also causes neuronal cell death. Therefore, understanding the detailed pathophysiological mechanisms affecting the recently discovered mitochondrial Ca2+ regulatory machineries will help to identify novel therapeutic targets. Here, we discuss current research into mitochondrial Ca2+ regulatory machineries and how mitochondrial Ca2+ dysregulation contributes to brain aging and neurodegenerative disease.
Collapse
Affiliation(s)
- Hyunsu Jung
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul, South Korea.,Division of Life Sciences, Korea University, Seoul, South Korea
| | - Su Yeon Kim
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Fatma Sema Canbakis Cecen
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul, South Korea.,Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, South Korea
| | - Yongcheol Cho
- Division of Life Sciences, Korea University, Seoul, South Korea
| | - Seok-Kyu Kwon
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul, South Korea.,Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, South Korea
| |
Collapse
|
128
|
Faizan MI, Ahmad T. Altered mitochondrial calcium handling and cell death by necroptosis: An emerging paradigm. Mitochondrion 2020; 57:47-62. [PMID: 33340710 DOI: 10.1016/j.mito.2020.12.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 11/24/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022]
Abstract
The classical necroptosis signaling is mediated by death receptors (DRs) that work in synergy with traditional caspase inhibitory signals. Currently, potential therapeutic molecules are in various phases of clinical trials for a spectrum of pathological conditions associated with necroptosis. However, a non-classical model of necroptosis has also emerged over the last decade with a relatively unexplored molecular mechanism. Although in vitro studies and preclinical models have shown its close association with mitochondrial dysfunction (mito-dysfunction), contradictory reports have emerged which complicate its definitiveness. Though impaired mitochondrial calcium ([Ca2+]m) handling is established in necrotic cell death, how this interplay regulates necroptosis is yet to be elucidated. Taking these questions into consideration, we have discussed various molecular aspects of necroptosis with the emerging role of mito-dysfunction. Based on the central role of altered [Ca2+]m handling in mito-dysfunction mediated necroptosis, we have provided a comprehensive molecular insight into this emerging paradigm. Potential reasons for the contradictory findings regarding the role of mito-dysfunction in necroptosis in general and mitochondrial-dependent necroptosis in specific are discussed. We also provide insights into the current understanding of how [Ca2+]m can be a critical determinant in deciding the cell fate under certain pathological conditions, while under others it may be dispensable. Lastly, we have highlighted the key molecular targets which have a direct implication for therapeutic intervention in conditions that are associated with impaired [Ca2+]m handling and cell death by necroptosis.
Collapse
Affiliation(s)
- Md Imam Faizan
- Multidisciplinary Centre for Advanced Research & Studies, Jamia Millia Islamia, New Delhi 110025 India
| | - Tanveer Ahmad
- Multidisciplinary Centre for Advanced Research & Studies, Jamia Millia Islamia, New Delhi 110025 India.
| |
Collapse
|
129
|
Zhang L, Wang Z, Lu T, Meng L, Luo Y, Fu X, Hou Y. Mitochondrial Ca 2+ Overload Leads to Mitochondrial Oxidative Stress and Delayed Meiotic Resumption in Mouse Oocytes. Front Cell Dev Biol 2020; 8:580876. [PMID: 33384990 PMCID: PMC7770107 DOI: 10.3389/fcell.2020.580876] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/11/2020] [Indexed: 12/12/2022] Open
Abstract
Overweight or obese women seeking pregnancy is becoming increasingly common. Human maternal obesity gives rise to detrimental effects during reproduction. Emerging evidence has shown that these abnormities are likely attributed to oocyte quality. Oxidative stress induces poor oocyte conditions, but whether mitochondrial calcium homeostasis plays a key role in oocyte status remains unresolved. Here, we established a mitochondrial Ca2+ overload model in mouse oocytes. Knockdown gatekeepers of the mitochondrial Ca2+ uniporters Micu1 and Micu2 as well as the mitochondrial sodium calcium exchanger NCLX in oocytes both increased oocytes mitochondrial Ca2+ concentration. The overload of mitochondria Ca2+ in oocytes impaired mitochondrial function, leaded to oxidative stress, and changed protein kinase A (PKA) signaling associated gene expression as well as delayed meiotic resumption. Using this model, we aimed to determine the mechanism of delayed meiosis caused by mitochondrial Ca2+ overload, and whether oocyte-specific inhibition of mitochondrial Ca2+ influx could improve the reproductive abnormalities seen within obesity. Germinal vesicle breakdown stage (GVBD) and extrusion of first polar body (PB1) are two indicators of meiosis maturation. As expected, the percentage of oocytes that successfully progress to the germinal vesicle breakdown stage and extrude the first polar body during in vitro culture was increased significantly, and the expression of PKA signaling genes and mitochondrial function recovered after appropriate mitochondrial Ca2+ regulation. Additionally, some indicators of mitochondrial performance-such as adenosine triphosphate (ATP) and reactive oxygen species (ROS) levels and mitochondrial membrane potential-recovered to normal. These results suggest that the regulation of mitochondrial Ca2+ uptake in mouse oocytes has a significant role during oocyte maturation as well as PKA signaling and that proper mitochondrial Ca2+ reductions in obese oocytes can recover mitochondrial performance and improve obesity-associated oocyte quality.
Collapse
Affiliation(s)
- Luyao Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Zichuan Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Tengfei Lu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Lin Meng
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yan Luo
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xiangwei Fu
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yunpeng Hou
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| |
Collapse
|
130
|
Bertolini MS, Docampo R. Different Sensitivity of Control and MICU1- and MICU2-Ablated Trypanosoma cruzi Mitochondrial Calcium Uniporter Complex to Ruthenium-Based Inhibitors. Int J Mol Sci 2020; 21:ijms21239316. [PMID: 33297372 PMCID: PMC7730205 DOI: 10.3390/ijms21239316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/03/2020] [Accepted: 12/05/2020] [Indexed: 11/20/2022] Open
Abstract
The mitochondrial Ca2+ uptake in trypanosomatids shares biochemical characteristics with that of animals. However, the composition of the mitochondrial Ca2+ uniporter complex (MCUC) in these parasites is quite peculiar, suggesting lineage-specific adaptations. In this work, we compared the inhibitory activity of ruthenium red (RuRed) and Ru360, the most commonly used MCUC inhibitors, with that of the recently described inhibitor Ru265, on Trypanosoma cruzi, the agent of Chagas disease. Ru265 was more potent than Ru360 and RuRed in inhibiting mitochondrial Ca2+ transport in permeabilized cells. When dose-response effects were investigated, an increase in sensitivity for Ru360 and Ru265 was observed in TcMICU1-KO and TcMICU2-KO cells as compared with control cells. In the presence of RuRed, a significant increase in sensitivity was observed only in TcMICU2-KO cells. However, application of Ru265 to intact cells did not affect growth and respiration of epimastigotes, mitochondrial Ca2+ uptake in Rhod-2-labeled intact cells, or attachment to host cells and infection by trypomastigotes, suggesting a low permeability for this compound in trypanosomes.
Collapse
|
131
|
O'Rourke B, Ashok D, Liu T. Mitochondrial Ca 2+ in heart failure: Not enough or too much? J Mol Cell Cardiol 2020; 151:126-134. [PMID: 33290770 DOI: 10.1016/j.yjmcc.2020.11.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/18/2020] [Accepted: 11/28/2020] [Indexed: 01/04/2023]
Abstract
Ca2+ serves as a ubiquitous second messenger mediating a variety of cellular processes including electrical excitation, contraction, gene expression, secretion, cell death and others. The identification of the molecular components of the mitochondrial Ca2+ influx and efflux pathways has created a resurgent interest in the regulation of mitochondrial Ca2+ balance and its physiological and pathophysiological roles. While the pace of discovery has quickened with the availability of new cellular and animal models, many fundamental questions remain to be answered regarding the regulation and functional impact of mitochondrial Ca2+ in health and disease. This review highlights several experimental observations pertaining to key aspects of mitochondrial Ca2+ homeostasis that remain enigmatic, particularly whether mitochondrial Ca2+ signaling is depressed or excessive in heart failure, which will determine the optimal approach to therapeutic intervention.
Collapse
Affiliation(s)
- Brian O'Rourke
- The Johns Hopkins University, Division of Cardiology, Department of Medicine, Baltimore, MD 21205, USA.
| | - Deepthi Ashok
- The Johns Hopkins University, Division of Cardiology, Department of Medicine, Baltimore, MD 21205, USA
| | - Ting Liu
- The Johns Hopkins University, Division of Cardiology, Department of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
132
|
Tomar D, Elrod JW. Metabolite regulation of the mitochondrial calcium uniporter channel. Cell Calcium 2020; 92:102288. [PMID: 32956979 PMCID: PMC8017895 DOI: 10.1016/j.ceca.2020.102288] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/07/2020] [Accepted: 09/07/2020] [Indexed: 01/26/2023]
Abstract
Calcium (Ca2+) is known to stimulate mitochondrial bioenergetics through the modulation of TCA cycle dehydrogenases and electron transport chain (ETC) complexes. This is hypothesized to be an essential pathway of energetic control to meet cellular ATP demand. While regulatory mechanisms of mitochondrial calcium uptake have been reported, it remains unknown if metabolite flux itself feedsback to regulate mitochondrial calcium (mCa2+) uptake. This hypothesis was recently tested by Nemani et al. (Sci. Signal. 2020) where the authors report that TCA cycle substrate flux regulates the mitochondrial calcium uniporter channel gatekeeper, mitochondrial calcium uptake 1 (MICU1), gene transcription in an early growth response protein 1 (EGR1) dependent fashion. They posit this is a regulatory feedback mechanism to control ionic homeostasis and mitochondrial bioenergetics with changing fuel availability. Here, we provide a historical overview of mitochondrial calcium exchange and comprehensive appraisal of these results in the context of recent literature and discuss possible regulatory pathways of mCa2+ uptake and mitochondrial bioenergetics.
Collapse
Affiliation(s)
- Dhanendra Tomar
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.
| | - John W Elrod
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|
133
|
Mechanisms of EMRE-Dependent MCU Opening in the Mitochondrial Calcium Uniporter Complex. Cell Rep 2020; 33:108486. [PMID: 33296646 PMCID: PMC7764451 DOI: 10.1016/j.celrep.2020.108486] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 10/09/2020] [Accepted: 11/13/2020] [Indexed: 12/02/2022] Open
Abstract
The mitochondrial calcium uniporter is a multi-subunit Ca2+-activated Ca2+ channel, made up of the pore-forming MCU protein, a metazoan-specific EMRE subunit, and MICU1/MICU2, which mediate Ca2+ activation. It has been established that metazoan MCU requires EMRE binding to conduct Ca2+, but how EMRE promotes MCU opening remains unclear. Here, we demonstrate that EMRE controls MCU activity via its transmembrane helix, while using an N-terminal PKP motif to strengthen binding with MCU. Opening of MCU requires hydrophobic interactions mediated by MCU residues near the pore’s luminal end. Enhancing these interactions by single mutation allows human MCU to transport Ca2+ without EMRE. We further show that EMRE may facilitate MCU opening by stabilizing the open state in a conserved MCU gating mechanism, present also in non-metazoan MCU homologs. These results provide insights into the evolution of the uniporter machinery and elucidate the mechanism underlying the physiologically crucial EMRE-dependent MCU activation process. The mitochondrial calcium uniporter is a multi-subunit ion channel that imports cytoplasmic Ca2+ into mitochondria to regulate cell energy production and death. In this work, Van Keuren et al. report a key activation mechanism of the uniporter, mediated by an auxiliary EMRE subunit in the channel complex.
Collapse
|
134
|
Dia M, Gomez L, Thibault H, Tessier N, Leon C, Chouabe C, Ducreux S, Gallo-Bona N, Tubbs E, Bendridi N, Chanon S, Leray A, Belmudes L, Couté Y, Kurdi M, Ovize M, Rieusset J, Paillard M. Reduced reticulum-mitochondria Ca 2+ transfer is an early and reversible trigger of mitochondrial dysfunctions in diabetic cardiomyopathy. Basic Res Cardiol 2020; 115:74. [PMID: 33258101 PMCID: PMC7704523 DOI: 10.1007/s00395-020-00835-7] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 11/13/2020] [Indexed: 12/17/2022]
Abstract
Type 2 diabetic cardiomyopathy features Ca2+ signaling abnormalities, notably an altered mitochondrial Ca2+ handling. We here aimed to study if it might be due to a dysregulation of either the whole Ca2+ homeostasis, the reticulum-mitochondrial Ca2+ coupling, and/or the mitochondrial Ca2+ entry through the uniporter. Following a 16-week high-fat high-sucrose diet (HFHSD), mice developed cardiac insulin resistance, fibrosis, hypertrophy, lipid accumulation, and diastolic dysfunction when compared to standard diet. Ultrastructural and proteomic analyses of cardiac reticulum-mitochondria interface revealed tighter interactions not compatible with Ca2+ transport in HFHSD cardiomyocytes. Intramyocardial adenoviral injections of Ca2+ sensors were performed to measure Ca2+ fluxes in freshly isolated adult cardiomyocytes and to analyze the direct effects of in vivo type 2 diabetes on cardiomyocyte function. HFHSD resulted in a decreased IP3R-VDAC interaction and a reduced IP3-stimulated Ca2+ transfer to mitochondria, with no changes in reticular Ca2+ level, cytosolic Ca2+ transients, and mitochondrial Ca2+ uniporter function. Disruption of organelle Ca2+ exchange was associated with decreased mitochondrial bioenergetics and reduced cell contraction, which was rescued by an adenovirus-mediated expression of a reticulum-mitochondria linker. An 8-week diet reversal was able to restore cardiac insulin signaling, Ca2+ transfer, and cardiac function in HFHSD mice. Therefore, our study demonstrates that the reticulum-mitochondria Ca2+ miscoupling may play an early and reversible role in the development of diabetic cardiomyopathy by disrupting primarily the mitochondrial bioenergetics. A diet reversal, by counteracting the MAM-induced mitochondrial Ca2+ dysfunction, might contribute to restore normal cardiac function and prevent the exacerbation of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Maya Dia
- Laboratoire CarMeN-Équipe 5 Cardioprotection, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, U1060 CARMEN, Equipe 5- Cardioprotection, Groupement Hospitalier Est, Bâtiment B13, 59 boulevard Pinel, 69500, Bron, France.,Laboratory of Experimental and Clinical Pharmacology, Faculty of Sciences, Lebanese University-Beirut, Beirut, Lebanon
| | - Ludovic Gomez
- Laboratoire CarMeN-Équipe 5 Cardioprotection, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, U1060 CARMEN, Equipe 5- Cardioprotection, Groupement Hospitalier Est, Bâtiment B13, 59 boulevard Pinel, 69500, Bron, France
| | - Helene Thibault
- Laboratoire CarMeN-Équipe 5 Cardioprotection, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, U1060 CARMEN, Equipe 5- Cardioprotection, Groupement Hospitalier Est, Bâtiment B13, 59 boulevard Pinel, 69500, Bron, France.,IHU OPERA, Hospices Civils de Lyon, 69500, Bron, France
| | - Nolwenn Tessier
- Laboratoire CarMeN-Équipe 5 Cardioprotection, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, U1060 CARMEN, Equipe 5- Cardioprotection, Groupement Hospitalier Est, Bâtiment B13, 59 boulevard Pinel, 69500, Bron, France
| | - Christelle Leon
- Laboratoire CarMeN-Équipe 5 Cardioprotection, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, U1060 CARMEN, Equipe 5- Cardioprotection, Groupement Hospitalier Est, Bâtiment B13, 59 boulevard Pinel, 69500, Bron, France
| | - Christophe Chouabe
- Laboratoire CarMeN-Équipe 5 Cardioprotection, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, U1060 CARMEN, Equipe 5- Cardioprotection, Groupement Hospitalier Est, Bâtiment B13, 59 boulevard Pinel, 69500, Bron, France
| | - Sylvie Ducreux
- Laboratoire CarMeN-Équipe 5 Cardioprotection, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, U1060 CARMEN, Equipe 5- Cardioprotection, Groupement Hospitalier Est, Bâtiment B13, 59 boulevard Pinel, 69500, Bron, France
| | - Noelle Gallo-Bona
- Laboratoire CarMeN-Équipe 5 Cardioprotection, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, U1060 CARMEN, Equipe 5- Cardioprotection, Groupement Hospitalier Est, Bâtiment B13, 59 boulevard Pinel, 69500, Bron, France
| | - Emily Tubbs
- Laboratoire CarMeN-Équipe 3, INSERM, INRA, Université Claude Bernard Lyon-1, INSA Lyon, Univ-Lyon, 69921, Oullins, France
| | - Nadia Bendridi
- Laboratoire CarMeN-Équipe 3, INSERM, INRA, Université Claude Bernard Lyon-1, INSA Lyon, Univ-Lyon, 69921, Oullins, France
| | - Stephanie Chanon
- Laboratoire CarMeN-Équipe 3, INSERM, INRA, Université Claude Bernard Lyon-1, INSA Lyon, Univ-Lyon, 69921, Oullins, France
| | - Aymeric Leray
- Laboratoire Interdisciplinaire Carnot de Bourgogne, UMR 6303 CNRS, Université de Bourgogne Franche Comté, 21078, Dijon, France
| | - Lucid Belmudes
- Univ. Grenoble Alpes, CEA, Inserm, IRIG, BGE, 38000, Grenoble, France
| | - Yohann Couté
- Univ. Grenoble Alpes, CEA, Inserm, IRIG, BGE, 38000, Grenoble, France
| | - Mazen Kurdi
- Laboratory of Experimental and Clinical Pharmacology, Faculty of Sciences, Lebanese University-Beirut, Beirut, Lebanon
| | - Michel Ovize
- Laboratoire CarMeN-Équipe 5 Cardioprotection, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, U1060 CARMEN, Equipe 5- Cardioprotection, Groupement Hospitalier Est, Bâtiment B13, 59 boulevard Pinel, 69500, Bron, France.,IHU OPERA, Hospices Civils de Lyon, 69500, Bron, France
| | - Jennifer Rieusset
- Laboratoire CarMeN-Équipe 3, INSERM, INRA, Université Claude Bernard Lyon-1, INSA Lyon, Univ-Lyon, 69921, Oullins, France
| | - Melanie Paillard
- Laboratoire CarMeN-Équipe 5 Cardioprotection, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, U1060 CARMEN, Equipe 5- Cardioprotection, Groupement Hospitalier Est, Bâtiment B13, 59 boulevard Pinel, 69500, Bron, France.
| |
Collapse
|
135
|
Feno S, Rizzuto R, Raffaello A, Vecellio Reane D. The molecular complexity of the Mitochondrial Calcium Uniporter. Cell Calcium 2020; 93:102322. [PMID: 33264708 DOI: 10.1016/j.ceca.2020.102322] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/19/2020] [Accepted: 11/19/2020] [Indexed: 12/20/2022]
Abstract
The role of mitochondria in regulating cellular Ca2+ homeostasis is crucial for the understanding of different cellular functions in physiological and pathological conditions. Nevertheless, the study of this aspect was severely limited by the lack of the molecular identity of the proteins responsible for mitochondrial Ca2+ uptake. In 2011, the discovery of the gene encoding for the Mitochondrial Calcium Uniporter (MCU), the selective channel responsible for mitochondrial Ca2+ uptake, gave rise to an explosion of studies aimed to characterize the composition, the regulation of the channel and its pathophysiological roles. Here, we summarize the recent discoveries on the molecular structure and composition of the MCU complex by providing new insights into the mechanisms that regulate MCU channel activity.
Collapse
Affiliation(s)
- Simona Feno
- Department of Biomedical Science, University of Padua, via G. Colombo 3, 35100 Padua, Italy
| | - Rosario Rizzuto
- Department of Biomedical Science, University of Padua, via G. Colombo 3, 35100 Padua, Italy
| | - Anna Raffaello
- Department of Biomedical Science, University of Padua, via G. Colombo 3, 35100 Padua, Italy; Myology Center, University of Padua, via G. Colombo 3, 35100 Padova, Italy.
| | - Denis Vecellio Reane
- Department of Biomedical Science, University of Padua, via G. Colombo 3, 35100 Padua, Italy.
| |
Collapse
|
136
|
Therapeutic Strategies to Target Calcium Dysregulation in Alzheimer's Disease. Cells 2020; 9:cells9112513. [PMID: 33233678 PMCID: PMC7699688 DOI: 10.3390/cells9112513] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 12/31/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia, affecting millions of people worldwide. Unfortunately, none of the current treatments are effective at improving cognitive function in AD patients and, therefore, there is an urgent need for the development of new therapies that target the early cause(s) of AD. Intracellular calcium (Ca2+) regulation is critical for proper cellular and neuronal function. It has been suggested that Ca2+ dyshomeostasis is an upstream factor of many neurodegenerative diseases, including AD. For this reason, chemical agents or small molecules aimed at targeting or correcting this Ca2+ dysregulation might serve as therapeutic strategies to prevent the development of AD. Moreover, neurons are not alone in exhibiting Ca2+ dyshomeostasis, since Ca2+ disruption is observed in other cell types in the brain in AD. In this review, we examine the distinct Ca2+ channels and compartments involved in the disease mechanisms that could be potential targets in AD.
Collapse
|
137
|
Yang CH, Lee KH, Ho WK, Lee SH. Inter-spike mitochondrial Ca 2+ release enhances high frequency synaptic transmission. J Physiol 2020; 599:1567-1594. [PMID: 33140422 DOI: 10.1113/jp280351] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 10/27/2020] [Indexed: 01/03/2023] Open
Abstract
KEY POINTS Presynaptic mitochondria not only absorb but also release Ca2+ during high frequency stimulation (HFS) when presynaptic [Ca2+ ] is kept low (<500 nm) by high cytosolic Ca2+ buffer or strong plasma membrane calcium clearance mechanisms under physiological external [Ca2+ ]. Mitochondrial Ca2+ release (MCR) does not alter the global presynaptic Ca2+ transients. MCR during HFS enhances short-term facilitation and steady state excitatory postsynaptic currents by increasing vesicular release probability. The intra-train MCR may provide residual calcium at interspike intervals, and thus support high frequency neurotransmission at central glutamatergic synapses. ABSTRACT Emerging evidence indicates that mitochondrial Ca2+ buffering contributes to local regulation of synaptic transmission. It is unknown, however, whether mitochondrial Ca2+ release (MCR) occurs during high frequency synaptic transmission. Confirming the previous notion that 2 μm tetraphenylphosphonium (TPP+ ) is a specific inhibitor of the mitochondrial Na+ /Ca2+ exchanger (mNCX), we studied the role of MCR via mNCX in short-term plasticity during high frequency stimulation (HFS) at the calyx of Held synapse of the rat. TPP+ reduced short-term facilitation (STF) and steady state excitatory postsynaptic currents during HFS at mature calyx synapses under physiological extracellular [Ca2+ ] ([Ca2+ ]o = 1.2 mm), but not at immature calyx or at 2 mm [Ca2+ ]o . The inhibitory effects of TPP+ were stronger at synapses with morphologically complex calyces harbouring many swellings and at 32°C than at simple calyx synapses and at room temperature. These effects of TPP+ on STF were well correlated with those on the presynaptic mitochondrial [Ca2+ ] build-up during HFS. Mitochondrial [Ca2+ ] during HFS was increased by TPP+ at mature calyces under 1.2 mm [Ca2+ ]o , and further enhanced at 32°C, but not under 2 mm [Ca2+ ]o or at immature calyces. The close correlation of the effects of TPP+ on mitochondrial [Ca2+ ] with those on STF suggests that mNCX contributes to STF at the calyx of Held synapses. The intra-train MCR enhanced vesicular release probability without altering global presynaptic [Ca2+ ]. Our results suggest that MCR during HFS elevates local [Ca2+ ] near synaptic sites at interspike intervals to enhance STF and to support stable synaptic transmission under physiological [Ca2+ ]o .
Collapse
Affiliation(s)
- Che Ho Yang
- Department of Physiology, Cell Physiology Lab., Seoul National University College of Medicine and Neuroscience Research Institute, Seoul National University Medical Research Centre, Seoul, Republic of Korea
| | - Kyu-Hee Lee
- Department of Physiology, Cell Physiology Lab., Seoul National University College of Medicine and Neuroscience Research Institute, Seoul National University Medical Research Centre, Seoul, Republic of Korea
| | - Won-Kyung Ho
- Department of Physiology, Cell Physiology Lab., Seoul National University College of Medicine and Neuroscience Research Institute, Seoul National University Medical Research Centre, Seoul, Republic of Korea.,Department of Brain and Cognitive Science, Seoul National University, Seoul, Republic of Korea
| | - Suk-Ho Lee
- Department of Physiology, Cell Physiology Lab., Seoul National University College of Medicine and Neuroscience Research Institute, Seoul National University Medical Research Centre, Seoul, Republic of Korea.,Department of Brain and Cognitive Science, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
138
|
Georgiadou E, Rutter GA. Control by Ca 2+ of mitochondrial structure and function in pancreatic β-cells. Cell Calcium 2020; 91:102282. [PMID: 32961506 PMCID: PMC7116533 DOI: 10.1016/j.ceca.2020.102282] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/25/2020] [Accepted: 08/25/2020] [Indexed: 12/11/2022]
Abstract
Mitochondria play a central role in glucose metabolism and the stimulation of insulin secretion from pancreatic β-cells. In this review, we discuss firstly the regulation and roles of mitochondrial Ca2+ transport in glucose-regulated insulin secretion, and the molecular machinery involved. Next, we discuss the evidence that mitochondrial dysfunction in β-cells is associated with type 2 diabetes, from a genetic, functional and structural point of view, and then the possibility that these changes may in part be mediated by dysregulation of cytosolic Ca2+. Finally, we review the importance of preserved mitochondrial structure and dynamics for mitochondrial gene expression and their possible relevance to the pathogenesis of type 2 diabetes.
Collapse
Affiliation(s)
- Eleni Georgiadou
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, du Cane Road, London, W12 0NN, UK
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
139
|
Abstract
New cryo-electron microscopy structures of the mitochondrial Ca2+ uniporter ion channel complex in various conformations reveal channel gating regulation by Ca2+-dependent unblock of the channel pore by MICU1.
Collapse
Affiliation(s)
- J Kevin Foskett
- Departments of Physiology and Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States.
| |
Collapse
|
140
|
A Novel Biomarker Driving Poor-Prognosis Liver Cancer: Overexpression of the Mitochondrial Calcium Gatekeepers. Biomedicines 2020; 8:biomedicines8110451. [PMID: 33114428 PMCID: PMC7693594 DOI: 10.3390/biomedicines8110451] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/24/2022] Open
Abstract
Several studies have indicated the biological role of mitochondrial Ca2+ uptake in cancer pathophysiology; however, its implications in predicting the prognosis of hepatocellular carcinoma (HCC) are not yet fully understood. Here, we collected tumor specimens and adjacent normal liver tissues from 354 confirmed HCC patients and analyzed the levels of cyclic adenosine monophosphate (cAMP) responsive element binding protein 1 (CREB), mitochondrial calcium uniporter (MCU), mitochondrial calcium uptake 1 and 2 (MICU1, MICU2) using bioinformatics, qRT-PCR, and immunohistochemistry (IHC), and their relationship with clinicopathological characteristics and prognosis. HCC patients with low CREB/MICU1 and high MCU/MICU2 expression exhibited poor survival rate and prognosis in overall survival (OS) and disease-free survival (DFS) analyses. Low CREB/MICU1 and low MICU1 alone indicated poor prognosis in stage I/II and III/IV patients, respectively. In the poor differentiation/undifferentiation group, low expression of MICU1 indicated poor clinical outcomes. Low CREB/MICU1 expression suggested poor outcomes in patients with or without hepatitis B virus (HBV) infection and poor prognosis in the HCV infection group. In the non- hepatitis C virus (HCV) infection group, low MCU1 indicated a poor prognosis. Multivariate analysis demonstrated that CREB and MICU1 expression showed prognostic significance. This study demonstrates the prognostic significance of CREB, MCU, MICU1, and MICU2, in predicting HCC outcomes. Low CREB/MICU1 and high MCU/MICU2 in HCC tissues are associated with poor prognosis, thus offering a novel perspective in the clinical management for HCC patients.
Collapse
|
141
|
Li A, Yi J, Li X, Zhou J. Physiological Ca 2+ Transients Versus Pathological Steady-State Ca 2+ Elevation, Who Flips the ROS Coin in Skeletal Muscle Mitochondria. Front Physiol 2020; 11:595800. [PMID: 33192612 PMCID: PMC7642813 DOI: 10.3389/fphys.2020.595800] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/05/2020] [Indexed: 12/20/2022] Open
Abstract
Mitochondria are both the primary provider of ATP and the pivotal regulator of cell death, which are essential for physiological muscle activities. Ca2+ plays a multifaceted role in mitochondrial function. During muscle contraction, Ca2+ influx into mitochondria activates multiple enzymes related to tricarboxylic acid (TCA) cycle and oxidative phosphorylation, resulting in increased ATP synthesis to meet the energy demand. Pathophysiological conditions such as skeletal muscle denervation or unloading also lead to elevated Ca2+ levels inside mitochondria. However, the outcomes of this steady-state elevation of mitochondrial Ca2+ level include exacerbated reactive oxygen species (ROS) generation, sensitized opening of mitochondrial permeability transition pore (mPTP), induction of programmed cell death, and ultimately muscle atrophy. Previously, both acute and long-term endurance exercises have been reported to activate certain signaling pathways to counteract ROS production. Meanwhile, electrical stimulation is known to help prevent apoptosis and alleviate muscle atrophy in denervated animal models and patients with motor impairment. There are various mechanistic studies that focus on the excitation-transcription coupling framework to understand the beneficial role of exercise and electrical stimulation. Interestingly, a recent study has revealed an unexpected role of rapid mitochondrial Ca2+ transients in keeping mPTP at a closed state with reduced mitochondrial ROS production. This discovery motivated us to contribute this review article to inspire further discussion about the potential mechanisms underlying differential outcomes of physiological mitochondrial Ca2+ transients and pathological mitochondrial Ca2+ elevation in skeletal muscle ROS production.
Collapse
Affiliation(s)
- Ang Li
- Department of Kinesiology, College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX, United States
| | - Jianxun Yi
- Department of Kinesiology, College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX, United States
| | - Xuejun Li
- Department of Kinesiology, College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX, United States
| | - Jingsong Zhou
- Department of Kinesiology, College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX, United States
| |
Collapse
|
142
|
Naumova N, Šachl R. Regulation of Cell Death by Mitochondrial Transport Systems of Calcium and Bcl-2 Proteins. MEMBRANES 2020; 10:E299. [PMID: 33096926 PMCID: PMC7590060 DOI: 10.3390/membranes10100299] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/16/2020] [Accepted: 10/19/2020] [Indexed: 12/13/2022]
Abstract
Mitochondria represent the fundamental system for cellular energy metabolism, by not only supplying energy in the form of ATP, but also by affecting physiology and cell death via the regulation of calcium homeostasis and the activity of Bcl-2 proteins. A lot of research has recently been devoted to understanding the interplay between Bcl-2 proteins, the regulation of these interactions within the cell, and how these interactions lead to the changes in calcium homeostasis. However, the role of Bcl-2 proteins in the mediation of mitochondrial calcium homeostasis, and therefore the induction of cell death pathways, remain underestimated and are still not well understood. In this review, we first summarize our knowledge about calcium transport systems in mitochondria, which, when miss-regulated, can induce necrosis. We continue by reviewing and analyzing the functions of Bcl-2 proteins in apoptosis. Finally, we link these two regulatory mechanisms together, exploring the interactions between the mitochondrial Ca2+ transport systems and Bcl-2 proteins, both capable of inducing cell death, with the potential to determine the cell death pathway-either the apoptotic or the necrotic one.
Collapse
Affiliation(s)
| | - Radek Šachl
- J. Heyrovský Institute of Physical Chemistry, Czech Academy of Sciences, 182 23 Prague, Czech Republic;
| |
Collapse
|
143
|
Gherardi G, Monticelli H, Rizzuto R, Mammucari C. The Mitochondrial Ca 2+ Uptake and the Fine-Tuning of Aerobic Metabolism. Front Physiol 2020; 11:554904. [PMID: 33117189 PMCID: PMC7575740 DOI: 10.3389/fphys.2020.554904] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 09/14/2020] [Indexed: 12/13/2022] Open
Abstract
Recently, the role of mitochondrial activity in high-energy demand organs and in the orchestration of whole-body metabolism has received renewed attention. In mitochondria, pyruvate oxidation, ensured by efficient mitochondrial pyruvate entry and matrix dehydrogenases activity, generates acetyl CoA that enters the TCA cycle. TCA cycle activity, in turn, provides reducing equivalents and electrons that feed the electron transport chain eventually producing ATP. Mitochondrial Ca2+ uptake plays an essential role in the control of aerobic metabolism. Mitochondrial Ca2+ accumulation stimulates aerobic metabolism by inducing the activity of three TCA cycle dehydrogenases. In detail, matrix Ca2+ indirectly modulates pyruvate dehydrogenase via pyruvate dehydrogenase phosphatase 1, and directly activates isocitrate and α-ketoglutarate dehydrogenases. Here, we will discuss the contribution of mitochondrial Ca2+ uptake to the metabolic homeostasis of organs involved in systemic metabolism, including liver, skeletal muscle, and adipose tissue. We will also tackle the role of mitochondrial Ca2+ uptake in the heart, a high-energy consuming organ whose function strictly depends on appropriate Ca2+ signaling.
Collapse
Affiliation(s)
- Gaia Gherardi
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | | | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | | |
Collapse
|
144
|
Rao G, Dwivedi SKD, Zhang Y, Dey A, Shameer K, Karthik R, Srikantan S, Hossen MN, Wren JD, Madesh M, Dudley JT, Bhattacharya R, Mukherjee P. MicroRNA-195 controls MICU1 expression and tumor growth in ovarian cancer. EMBO Rep 2020; 21:e48483. [PMID: 32851774 PMCID: PMC7534609 DOI: 10.15252/embr.201948483] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/17/2020] [Accepted: 07/27/2020] [Indexed: 12/12/2022] Open
Abstract
MICU1 is a mitochondrial inner membrane protein that inhibits mitochondrial calcium entry; elevated MICU1 expression is characteristic of many cancers, including ovarian cancer. MICU1 induces both glycolysis and chemoresistance and is associated with poor clinical outcomes. However, there are currently no available interventions to normalize aberrant MICU1 expression. Here, we demonstrate that microRNA-195-5p (miR-195) directly targets the 3' UTR of the MICU1 mRNA and represses MICU1 expression. Additionally, miR-195 is under-expressed in ovarian cancer cell lines, and restoring miR-195 expression reestablishes native MICU1 levels and the associated phenotypes. Stable expression of miR-195 in a human xenograft model of ovarian cancer significantly reduces tumor growth, increases tumor doubling times, and enhances overall survival. In conclusion, miR-195 controls MICU1 levels in ovarian cancer and could be exploited to normalize aberrant MICU1 expression, thus reversing both glycolysis and chemoresistance and consequently improving patient outcomes.
Collapse
Affiliation(s)
- Geeta Rao
- Department of PathologyThe University of Oklahoma Health Sciences CenterOklahoma CityOKUSA
| | | | - Yushan Zhang
- Department of PathologyThe University of Oklahoma Health Sciences CenterOklahoma CityOKUSA
| | - Anindya Dey
- Department of Obstetrics and GynecologyThe University of Oklahoma Health Sciences CenterOklahoma CityOKUSA
| | - Khader Shameer
- Institute of Next Generation Healthcare (INGH)Icahn Institute for Data Science and Genomic TechnologyDepartment of Genetics and Genomic SciencesMount Sinai Health SystemNew YorkNYUSA
| | - Ramachandran Karthik
- Department of MedicineCardiology DivisionUniversity of Texas Health San AntonioSan AntonioTXUSA
| | - Subramanya Srikantan
- Department of MedicineCardiology DivisionUniversity of Texas Health San AntonioSan AntonioTXUSA
| | - Md Nazir Hossen
- Department of PathologyThe University of Oklahoma Health Sciences CenterOklahoma CityOKUSA
| | - Jonathan D Wren
- Genes & Human Disease Research ProgramOklahoma Medical Research FoundationOklahoma CityOKUSA
| | - Muniswamy Madesh
- Department of MedicineCardiology DivisionUniversity of Texas Health San AntonioSan AntonioTXUSA
| | - Joel T Dudley
- Institute of Next Generation Healthcare (INGH)Icahn Institute for Data Science and Genomic TechnologyDepartment of Genetics and Genomic SciencesMount Sinai Health SystemNew YorkNYUSA
| | - Resham Bhattacharya
- Department of Obstetrics and GynecologyThe University of Oklahoma Health Sciences CenterOklahoma CityOKUSA
- Peggy and Charles Stephenson Cancer CenterThe University of Oklahoma Health Sciences CenterOklahoma CityOKUSA
| | - Priyabrata Mukherjee
- Department of PathologyThe University of Oklahoma Health Sciences CenterOklahoma CityOKUSA
- Peggy and Charles Stephenson Cancer CenterThe University of Oklahoma Health Sciences CenterOklahoma CityOKUSA
| |
Collapse
|
145
|
MacEwen MJ, Markhard AL, Bozbeyoglu M, Bradford F, Goldberger O, Mootha VK, Sancak Y. Evolutionary divergence reveals the molecular basis of EMRE dependence of the human MCU. Life Sci Alliance 2020; 3:e202000718. [PMID: 32769116 PMCID: PMC7425227 DOI: 10.26508/lsa.202000718] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 01/18/2023] Open
Abstract
The mitochondrial calcium uniporter (MCU) is a calcium-activated calcium channel critical for signaling and bioenergetics. MCU, the pore-forming subunit of the uniporter, contains two transmembrane domains and is found in all major eukaryotic taxa. In amoeba and fungi, MCU homologs are sufficient to form a functional calcium channel, whereas human MCU exhibits a strict requirement for the metazoan protein essential MCU regulator (EMRE) for conductance. Here, we exploit this evolutionary divergence to decipher the molecular basis of human MCU's dependence on EMRE. By systematically generating chimeric proteins that consist of EMRE-independent Dictyostelium discoideum MCU and Homo sapiens MCU (HsMCU), we converged on a stretch of 10 amino acids in D. discoideum MCU that can be transplanted to HsMCU to render it EMRE independent. We call this region in human MCU the EMRE dependence domain (EDD). Crosslinking experiments show that EMRE directly interacts with HsMCU at its transmembrane domains as well as the EDD. Our results suggest that EMRE stabilizes the EDD of MCU, permitting both channel opening and calcium conductance, consistent with recently published structures of MCU-EMRE.
Collapse
Affiliation(s)
| | - Andrew L Markhard
- Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Mert Bozbeyoglu
- Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Forrest Bradford
- Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Olga Goldberger
- Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Vamsi K Mootha
- Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Yasemin Sancak
- Department of Pharmacology, University of Washington, Seattle, WA, USA
| |
Collapse
|
146
|
Gibhardt CS, Ezeriņa D, Sung HM, Messens J, Bogeski I. Redox regulation of the mitochondrial calcium transport machinery. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2020.07.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
147
|
Bisbach CM, Hutto RA, Poria D, Cleghorn WM, Abbas F, Vinberg F, Kefalov VJ, Hurley JB, Brockerhoff SE. Mitochondrial Calcium Uniporter (MCU) deficiency reveals an alternate path for Ca 2+ uptake in photoreceptor mitochondria. Sci Rep 2020; 10:16041. [PMID: 32994451 PMCID: PMC7525533 DOI: 10.1038/s41598-020-72708-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 09/04/2020] [Indexed: 01/18/2023] Open
Abstract
Rods and cones use intracellular Ca2+ to regulate many functions, including phototransduction and neurotransmission. The Mitochondrial Calcium Uniporter (MCU) complex is thought to be the primary pathway for Ca2+ entry into mitochondria in eukaryotes. We investigate the hypothesis that mitochondrial Ca2+ uptake via MCU influences phototransduction and energy metabolism in photoreceptors using a mcu-/- zebrafish and a rod photoreceptor-specific Mcu-/- mouse. Using genetically encoded Ca2+ sensors to directly examine Ca2+ uptake in zebrafish cone mitochondria, we found that loss of MCU reduces but does not eliminate mitochondrial Ca2+ uptake. Loss of MCU does not lead to photoreceptor degeneration, mildly affects mitochondrial metabolism, and does not alter physiological responses to light, even in the absence of the Na+/Ca2+, K+ exchanger. Our results reveal that MCU is dispensable for vertebrate photoreceptor function, consistent with its low expression and the presence of an alternative pathway for Ca2+ uptake into photoreceptor mitochondria.
Collapse
Affiliation(s)
- Celia M Bisbach
- Biochemistry Department, University of Washington, Seattle, WA, USA
| | - Rachel A Hutto
- Biochemistry Department, University of Washington, Seattle, WA, USA
| | - Deepak Poria
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Fatima Abbas
- Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT, USA
| | - Frans Vinberg
- Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT, USA
| | - Vladimir J Kefalov
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - James B Hurley
- Biochemistry Department, University of Washington, Seattle, WA, USA
- Ophthalmology Department, University of Washington, Seattle, WA, USA
| | - Susan E Brockerhoff
- Biochemistry Department, University of Washington, Seattle, WA, USA.
- Ophthalmology Department, University of Washington, Seattle, WA, USA.
| |
Collapse
|
148
|
Diaz-Juarez J, Suarez JA, Dillmann WH, Suarez J. Mitochondrial calcium handling and heart disease in diabetes mellitus. Biochim Biophys Acta Mol Basis Dis 2020; 1867:165984. [PMID: 33002576 DOI: 10.1016/j.bbadis.2020.165984] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 01/23/2023]
Abstract
Diabetes mellitus-induced heart disease, including diabetic cardiomyopathy, is an important medical problem and is difficult to treat. Diabetes mellitus increases the risk for heart failure and decreases cardiac myocyte function, which are linked to changes in cardiac mitochondrial energy metabolism. The free mitochondrial calcium concentration ([Ca2+]m) is fundamental in activating the mitochondrial respiratory chain complexes and ATP production and is also known to regulate the activity of key mitochondrial dehydrogenases. The mitochondrial calcium uniporter complex (MCUC) plays a major role in mediating mitochondrial Ca2+ import, and its expression and function therefore may have a marked impact on cardiac myocyte metabolism and function. Here, we summarize the pathophysiological role of [Ca2+]m handling and MCUC in the diabetic heart. In addition, we evaluate potential therapeutic targets, directed to the machinery that regulates mitochondrial calcium handling, to alleviate diabetes-related cardiac disease.
Collapse
Affiliation(s)
- Julieta Diaz-Juarez
- Department of Pharmacology, Instituto Nacional de Cardiología, Juan Badiano No. 1, Col. Seccion XVI, 14080 Tlalpan, Ciudad de Mexico, Mexico
| | - Jorge A Suarez
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Wolfgang H Dillmann
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jorge Suarez
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
149
|
Debattisti V, Horn A, Singh R, Seifert EL, Hogarth MW, Mazala DA, Huang KT, Horvath R, Jaiswal JK, Hajnóczky G. Dysregulation of Mitochondrial Ca 2+ Uptake and Sarcolemma Repair Underlie Muscle Weakness and Wasting in Patients and Mice Lacking MICU1. Cell Rep 2020; 29:1274-1286.e6. [PMID: 31665639 PMCID: PMC7007691 DOI: 10.1016/j.celrep.2019.09.063] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 08/07/2019] [Accepted: 09/20/2019] [Indexed: 01/29/2023] Open
Abstract
Muscle function is regulated by Ca2+, which mediates excitation-contraction coupling, energy metabolism, adaptation to exercise, and sarcolemmal repair. Several of these actions rely on Ca2+ delivery to the mitochondrial matrix via the mitochondrial Ca2+ uniporter, the pore of which is formed by mitochondrial calcium uniporter (MCU). MCU's gatekeeping and cooperative activation are controlled by MICU1. Loss-of-protein mutation in MICU1 causes a neuromuscular disease. To determine the mechanisms underlying the muscle impairments, we used MICU1 patient cells and skeletal muscle-specific MICU1 knockout mice. Both these models show a lower threshold for MCU-mediated Ca2+ uptake. Lack of MICU1 is associated with impaired mitochondrial Ca2+ uptake during excitation-contraction, aerobic metabolism impairment, muscle weakness, fatigue, and myofiber damage during physical activity. MICU1 deficit compromises mitochondrial Ca2+ uptake during sarcolemmal injury, which causes ineffective repair of the damaged myofibers. Thus, dysregulation of mitochondrial Ca2+ uptake hampers myofiber contractile function, likely through energy metabolism and membrane repair.
Collapse
Affiliation(s)
- Valentina Debattisti
- MitoCare Center for Mitochondrial Imaging Research and Diagnostics, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Adam Horn
- Center for Genetic Medicine Research, Children's National Health System, 111 Michigan Avenue Northwest, Washington, DC 20010, USA; Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
| | - Raghavendra Singh
- MitoCare Center for Mitochondrial Imaging Research and Diagnostics, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Erin L Seifert
- MitoCare Center for Mitochondrial Imaging Research and Diagnostics, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Marshall W Hogarth
- Center for Genetic Medicine Research, Children's National Health System, 111 Michigan Avenue Northwest, Washington, DC 20010, USA; Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
| | - Davi A Mazala
- Center for Genetic Medicine Research, Children's National Health System, 111 Michigan Avenue Northwest, Washington, DC 20010, USA; Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
| | - Kai Ting Huang
- MitoCare Center for Mitochondrial Imaging Research and Diagnostics, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Rita Horvath
- Wellcome Centre for Mitochondrial Research, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Jyoti K Jaiswal
- Center for Genetic Medicine Research, Children's National Health System, 111 Michigan Avenue Northwest, Washington, DC 20010, USA; Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA.
| | - György Hajnóczky
- MitoCare Center for Mitochondrial Imaging Research and Diagnostics, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
150
|
Marchi S, Giorgi C, Galluzzi L, Pinton P. Ca 2+ Fluxes and Cancer. Mol Cell 2020; 78:1055-1069. [PMID: 32559424 DOI: 10.1016/j.molcel.2020.04.017] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/14/2020] [Accepted: 04/15/2020] [Indexed: 02/06/2023]
Abstract
Ca2+ ions are key second messengers in both excitable and non-excitable cells. Owing to the rather pleiotropic nature of Ca2+ transporters and other Ca2+-binding proteins, however, Ca2+ signaling has attracted limited attention as a potential target of anticancer therapy. Here, we discuss cancer-associated alterations of Ca2+ fluxes at specific organelles as we identify novel candidates for the development of drugs that selectively target Ca2+ signaling in malignant cells.
Collapse
Affiliation(s)
- Saverio Marchi
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Carlotta Giorgi
- Department of Medical Sciences, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA; Sandra and Edward Meyer Cancer Center, New York, NY, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA; Department of Dermatology, Yale School of Medicine, New Haven, CT, USA; Université de Paris, Paris, France.
| | - Paolo Pinton
- Department of Medical Sciences, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy.
| |
Collapse
|