1
|
Balderas E, Lee SHJ, Rai NK, Mollinedo DM, Duron HE, Chaudhuri D. Mitochondrial Calcium Regulation of Cardiac Metabolism in Health and Disease. Physiology (Bethesda) 2024; 39:0. [PMID: 38713090 PMCID: PMC11460536 DOI: 10.1152/physiol.00014.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/02/2024] [Accepted: 05/02/2024] [Indexed: 05/08/2024] Open
Abstract
Oxidative phosphorylation is regulated by mitochondrial calcium (Ca2+) in health and disease. In physiological states, Ca2+ enters via the mitochondrial Ca2+ uniporter and rapidly enhances NADH and ATP production. However, maintaining Ca2+ homeostasis is critical: insufficient Ca2+ impairs stress adaptation, and Ca2+ overload can trigger cell death. In this review, we delve into recent insights further defining the relationship between mitochondrial Ca2+ dynamics and oxidative phosphorylation. Our focus is on how such regulation affects cardiac function in health and disease, including heart failure, ischemia-reperfusion, arrhythmias, catecholaminergic polymorphic ventricular tachycardia, mitochondrial cardiomyopathies, Barth syndrome, and Friedreich's ataxia. Several themes emerge from recent data. First, mitochondrial Ca2+ regulation is critical for fuel substrate selection, metabolite import, and matching of ATP supply to demand. Second, mitochondrial Ca2+ regulates both the production and response to reactive oxygen species (ROS), and the balance between its pro- and antioxidant effects is key to how it contributes to physiological and pathological states. Third, Ca2+ exerts localized effects on the electron transport chain (ETC), not through traditional allosteric mechanisms but rather indirectly. These effects hinge on specific transporters, such as the uniporter or the Na+/Ca2+ exchanger, and may not be noticeable acutely, contributing differently to phenotypes depending on whether Ca2+ transporters are acutely or chronically modified. Perturbations in these novel relationships during disease states may either serve as compensatory mechanisms or exacerbate impairments in oxidative phosphorylation. Consequently, targeting mitochondrial Ca2+ holds promise as a therapeutic strategy for a variety of cardiac diseases characterized by contractile failure or arrhythmias.
Collapse
Affiliation(s)
- Enrique Balderas
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States
| | - Sandra H J Lee
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States
| | - Neeraj K Rai
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States
| | - David M Mollinedo
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States
| | - Hannah E Duron
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States
| | - Dipayan Chaudhuri
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States
- Division of Cardiovascular Medicine, Department of Internal Medicine, Biochemistry, Biomedical Engineering, University of Utah, Salt Lake City, Utah, United States
| |
Collapse
|
2
|
Bastioli G, Piccirillo S, Graciotti L, Carone M, Sprega G, Taoussi O, Preziuso A, Castaldo P. Calcium Deregulation in Neurodegeneration and Neuroinflammation in Parkinson's Disease: Role of Calcium-Storing Organelles and Sodium-Calcium Exchanger. Cells 2024; 13:1301. [PMID: 39120330 PMCID: PMC11311461 DOI: 10.3390/cells13151301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 08/10/2024] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder that lacks effective treatment strategies to halt or delay its progression. The homeostasis of Ca2+ ions is crucial for ensuring optimal cellular functions and survival, especially for neuronal cells. In the context of PD, the systems regulating cellular Ca2+ are compromised, leading to Ca2+-dependent synaptic dysfunction, impaired neuronal plasticity, and ultimately, neuronal loss. Recent research efforts directed toward understanding the pathology of PD have yielded significant insights, particularly highlighting the close relationship between Ca2+ dysregulation, neuroinflammation, and neurodegeneration. However, the precise mechanisms driving the selective loss of dopaminergic neurons in PD remain elusive. The disruption of Ca2+ homeostasis is a key factor, engaging various neurodegenerative and neuroinflammatory pathways and affecting intracellular organelles that store Ca2+. Specifically, impaired functioning of mitochondria, lysosomes, and the endoplasmic reticulum (ER) in Ca2+ metabolism is believed to contribute to the disease's pathophysiology. The Na+-Ca2+ exchanger (NCX) is considered an important key regulator of Ca2+ homeostasis in various cell types, including neurons, astrocytes, and microglia. Alterations in NCX activity are associated with neurodegenerative processes in different models of PD. In this review, we will explore the role of Ca2+ dysregulation and neuroinflammation as primary drivers of PD-related neurodegeneration, with an emphasis on the pivotal role of NCX in the pathology of PD. Consequently, NCXs and their interplay with intracellular organelles may emerge as potentially pivotal players in the mechanisms underlying PD neurodegeneration, providing a promising avenue for therapeutic intervention aimed at halting neurodegeneration.
Collapse
Affiliation(s)
- Guendalina Bastioli
- Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy;
| | - Silvia Piccirillo
- Department of Biomedical Sciences and Public Health, School of Medicine, University “Politecnica Delle Marche”, Via Tronto 10/A, 60126 Ancona, Italy; (L.G.); (M.C.); (G.S.); (O.T.); (A.P.)
| | - Laura Graciotti
- Department of Biomedical Sciences and Public Health, School of Medicine, University “Politecnica Delle Marche”, Via Tronto 10/A, 60126 Ancona, Italy; (L.G.); (M.C.); (G.S.); (O.T.); (A.P.)
| | - Marianna Carone
- Department of Biomedical Sciences and Public Health, School of Medicine, University “Politecnica Delle Marche”, Via Tronto 10/A, 60126 Ancona, Italy; (L.G.); (M.C.); (G.S.); (O.T.); (A.P.)
- Institute of Chemical and Bioengineering, Department of Chemistry and Applied Biosciences, ETH Zurich, 8092 Zürich, Switzerland
| | - Giorgia Sprega
- Department of Biomedical Sciences and Public Health, School of Medicine, University “Politecnica Delle Marche”, Via Tronto 10/A, 60126 Ancona, Italy; (L.G.); (M.C.); (G.S.); (O.T.); (A.P.)
| | - Omayema Taoussi
- Department of Biomedical Sciences and Public Health, School of Medicine, University “Politecnica Delle Marche”, Via Tronto 10/A, 60126 Ancona, Italy; (L.G.); (M.C.); (G.S.); (O.T.); (A.P.)
| | - Alessandra Preziuso
- Department of Biomedical Sciences and Public Health, School of Medicine, University “Politecnica Delle Marche”, Via Tronto 10/A, 60126 Ancona, Italy; (L.G.); (M.C.); (G.S.); (O.T.); (A.P.)
| | - Pasqualina Castaldo
- Department of Biomedical Sciences and Public Health, School of Medicine, University “Politecnica Delle Marche”, Via Tronto 10/A, 60126 Ancona, Italy; (L.G.); (M.C.); (G.S.); (O.T.); (A.P.)
| |
Collapse
|
3
|
Bulthuis EP, Adjobo-Hermans MJW, de Potter B, Hoogstraten S, Wezendonk LHT, Tutakhel OAZ, Wintjes LT, van den Heuvel B, Willems PHGM, Kamsteeg EJ, Gozalbo MER, Sallevelt SCEH, Koudijs SM, Nicolai J, de Bie CI, Hoogendijk JE, Koopman WJH, Rodenburg RJ. SMDT1 variants impair EMRE-mediated mitochondrial calcium uptake in patients with muscle involvement. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166808. [PMID: 37454773 DOI: 10.1016/j.bbadis.2023.166808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/26/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Ionic calcium (Ca2+) is a key messenger in signal transduction and its mitochondrial uptake plays an important role in cell physiology. This uptake is mediated by the mitochondrial Ca2+ uniporter (MCU), which is regulated by EMRE (essential MCU regulator) encoded by the SMDT1 (single-pass membrane protein with aspartate rich tail 1) gene. This work presents the genetic, clinical and cellular characterization of two patients harbouring SMDT1 variants and presenting with muscle problems. Analysis of patient fibroblasts and complementation experiments demonstrated that these variants lead to absence of EMRE protein, induce MCU subcomplex formation and impair mitochondrial Ca2+ uptake. However, the activity of oxidative phosphorylation enzymes, mitochondrial morphology and membrane potential, as well as routine/ATP-linked respiration were not affected. We hypothesize that the muscle-related symptoms in the SMDT1 patients result from aberrant mitochondrial Ca2+ uptake.
Collapse
Affiliation(s)
- Elianne P Bulthuis
- Department of Biochemistry (286), Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands
| | - Merel J W Adjobo-Hermans
- Department of Biochemistry (286), Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands
| | - Bastiaan de Potter
- Department of Biochemistry (286), Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands
| | - Saskia Hoogstraten
- Department of Biochemistry (286), Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands; Human and Animal Physiology, Wageningen University & Research, 6700 AH Wageningen, the Netherlands
| | - Lisanne H T Wezendonk
- Department of Biochemistry (286), Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands
| | - Omar A Z Tutakhel
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands
| | - Liesbeth T Wintjes
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands
| | - Bert van den Heuvel
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands
| | - Peter H G M Willems
- Department of Biochemistry (286), Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands
| | - Erik-Jan Kamsteeg
- Department of Human Genetics, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands
| | - M Estela Rubio Gozalbo
- Department of Pediatrics, Maastricht University Medical Centre, 6229 HX Maastricht, the Netherlands; Department of Clinical Genetics, Maastricht University Medical Centre, 6229 HX Maastricht, the Netherlands
| | - Suzanne C E H Sallevelt
- Department of Clinical Genetics, Maastricht University Medical Centre, 6229 HX Maastricht, the Netherlands
| | - Suzanne M Koudijs
- Department of Neurology, Maastricht University Medical Centre, 6229 HX Maastricht, the Netherlands
| | - Joost Nicolai
- Department of Neurology, Maastricht University Medical Centre, 6229 HX Maastricht, the Netherlands
| | - Charlotte I de Bie
- Department of Genetics, University Medical Centre Utrecht, 3508 AB Utrecht, the Netherlands
| | - Jessica E Hoogendijk
- Rudolf Magnus Institute of Neuroscience, University Medical Centre Utrecht, 3584 CG Utrecht, the Netherlands
| | - Werner J H Koopman
- Human and Animal Physiology, Wageningen University & Research, 6700 AH Wageningen, the Netherlands; Department of Pediatrics, Amalia Children's Hospital, Radboud Center for Mitochondrial Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands.
| | - Richard J Rodenburg
- Department of Pediatrics, Amalia Children's Hospital, Radboud Center for Mitochondrial Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands.
| |
Collapse
|
4
|
Di Martino R, Sisalli MJ, Sirabella R, Della Notte S, Borzacchiello D, Feliciello A, Annunziato L, Scorziello A. Ncx3-Induced Mitochondrial Dysfunction in Midbrain Leads to Neuroinflammation in Striatum of A53t-α-Synuclein Transgenic Old Mice. Int J Mol Sci 2021; 22:ijms22158177. [PMID: 34360942 PMCID: PMC8347885 DOI: 10.3390/ijms22158177] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/22/2021] [Accepted: 07/27/2021] [Indexed: 12/24/2022] Open
Abstract
The exact mechanism underlying selective dopaminergic neurodegeneration is not completely understood. The complex interplay among toxic alpha-synuclein aggregates, oxidative stress, altered intracellular Ca2+-homeostasis, mitochondrial dysfunction and disruption of mitochondrial integrity is considered among the pathogenic mechanisms leading to dopaminergic neuronal loss. We herein investigated the molecular mechanisms leading to mitochondrial dysfunction and its relationship with activation of the neuroinflammatory process occurring in Parkinson’s disease. To address these issues, experiments were performed in vitro and in vivo in mice carrying the human mutation of α-synuclein A53T under the prion murine promoter. In these models, the expression and activity of NCX isoforms, a family of important transporters regulating ionic homeostasis in mammalian cells working in a bidirectional way, were evaluated in neurons and glial cells. Mitochondrial function was monitored with confocal microscopy and fluorescent dyes to measure mitochondrial calcium content and mitochondrial membrane potential. Parallel experiments were performed in 4 and 16-month-old A53T-α-synuclein Tg mice to correlate the functional data obtained in vitro with mitochondrial dysfunction and neuroinflammation through biochemical analysis. The results obtained demonstrated: 1. in A53T mice mitochondrial dysfunction occurs early in midbrain and later in striatum; 2. mitochondrial dysfunction occurring in the midbrain is mediated by the impairment of NCX3 protein expression in neurons and astrocytes; 3. mitochondrial dysfunction occurring early in midbrain triggers neuroinflammation later into the striatum, thus contributing to PD progression during mice aging.
Collapse
Affiliation(s)
- Rossana Di Martino
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.D.M.); (M.J.S.); (R.S.); (S.D.N.)
| | - Maria Josè Sisalli
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.D.M.); (M.J.S.); (R.S.); (S.D.N.)
| | - Rossana Sirabella
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.D.M.); (M.J.S.); (R.S.); (S.D.N.)
| | - Salvatore Della Notte
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.D.M.); (M.J.S.); (R.S.); (S.D.N.)
| | - Domenica Borzacchiello
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (D.B.); (A.F.)
| | - Antonio Feliciello
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (D.B.); (A.F.)
| | | | - Antonella Scorziello
- Department of Neuroscience, Division of Pharmacology, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Via Pansini 5, 80131 Naples, Italy; (R.D.M.); (M.J.S.); (R.S.); (S.D.N.)
- Correspondence:
| |
Collapse
|
5
|
Mitochondrial DNA A3243G variant-associated retinopathy: Current perspectives and clinical implications. Surv Ophthalmol 2021; 66:838-855. [PMID: 33610586 DOI: 10.1016/j.survophthal.2021.02.008] [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: 06/20/2020] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/12/2022]
Abstract
Cellular function and survival are critically dependent on the proper functionality of the mitochondrion. Neurodegenerative cellular processes including cellular adenosine triphosphate production, intermediary metabolism control, and apoptosis regulation are all mitochondrially mediated. The A to G transition at position 3243 in the mitochondrial MTTL1 gene that encodes for the leucine transfer RNA (m.3243A>G) causes a variety of diseases, including maternally inherited loss of hearing and diabetes syndrome (MIDD), mitochondrial encephalopathy, lactic acidosis, and stroke-like episodes syndrome (MELAS). Ophthalmological findings-including posterior sub-capsular cataract, ptosis, external ophthalmoplegia, and pigmentary retinopathy- have all been associated with the m.3243A>G variant. Pigmentary retinopathy is, however, the most common ocular finding, occurring in 38% to 86% of cases. To date, little is known about the pathogenesis, natural history, and heteroplasmic and phenotypic correlations of m.3243A>G-associated pigmentary retinopathy. We summarize the current understanding of mitochondrial genetics and pathogenesis of some associated diseases. We then review the pathophysiology, histology, clinical features, treatment, and important ocular and systemic phenotypic manifestations of m.3243A>G variant associated retinopathy. Mitochondrial diseases require a multidisciplinary team approach to ensure effective treatment, regular follow-up, and accurate genetic counseling.
Collapse
|
6
|
Jaña F, Bustos G, Rivas J, Cruz P, Urra F, Basualto-Alarcón C, Sagredo E, Ríos M, Lovy A, Dong Z, Cerda O, Madesh M, Cárdenas C. Complex I and II are required for normal mitochondrial Ca 2+ homeostasis. Mitochondrion 2019; 49:73-82. [PMID: 31310854 DOI: 10.1016/j.mito.2019.07.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 04/01/2019] [Accepted: 07/12/2019] [Indexed: 01/03/2023]
Abstract
Cytosolic calcium (cCa2+) entry into mitochondria is facilitated by the mitochondrial membrane potential (ΔΨm), an electrochemical gradient generated by the electron transport chain (ETC). Is has been assumed that as long as mutations that affect the ETC do not affect the ΔΨm, the mitochondrial Ca2+ (mCa2+) homeostasis remains normal. We show that knockdown of NDUFAF3 and SDHB reduce ETC activity altering mCa2+ efflux and influx rates while ΔΨm remains intact. Shifting the equilibrium toward lower [Ca2+]m accumulation renders cells resistant to death. Our findings reveal an unexpected relationship between complex I and II with the mCa2+ homeostasis independent of ΔΨm.
Collapse
Affiliation(s)
- Fabian Jaña
- Departamento de Ciencias de la Salud, Universidad de Aysén, Coyhaique, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Galdo Bustos
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile; Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
| | - José Rivas
- Departamento de Ciencias de la Salud, Universidad de Aysén, Coyhaique, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Pablo Cruz
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Felix Urra
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile; Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Carla Basualto-Alarcón
- Departamento de Ciencias de la Salud, Universidad de Aysén, Coyhaique, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile; Anatomy and Legal Medicine Department, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Eduardo Sagredo
- Centro de Investigación y Tratamiento del Cáncer, Facultad de Medicina, Universidad de Chile, Chile
| | - Melany Ríos
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Alenka Lovy
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile; Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile; Center for Neuroscience Research, Tufts University School of Medicine, Boston, MA, USA
| | - Zhiwei Dong
- Department of Medicine, Center for Precision Medicine, University of Texas Health San Antonio, TX, USA
| | - Oscar Cerda
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile; Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Santiago, Chile.; The Wound Repair, Treatment and Health (WoRTH), Chile
| | - Muniswamy Madesh
- Department of Medicine, Center for Precision Medicine, University of Texas Health San Antonio, TX, USA.
| | - César Cárdenas
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile; Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile; Buck Institute for Research on Aging, Novato, CA, USA; Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA, United States.
| |
Collapse
|
7
|
Theunissen TEJ, Nguyen M, Kamps R, Hendrickx AT, Sallevelt SCEH, Gottschalk RWH, Calis CM, Stassen APM, de Koning B, Mulder-Den Hartog ENM, Schoonderwoerd K, Fuchs SA, Hilhorst-Hofstee Y, de Visser M, Vanoevelen J, Szklarczyk R, Gerards M, de Coo IFM, Hellebrekers DMEI, Smeets HJM. Whole Exome Sequencing Is the Preferred Strategy to Identify the Genetic Defect in Patients With a Probable or Possible Mitochondrial Cause. Front Genet 2018; 9:400. [PMID: 30369941 PMCID: PMC6194163 DOI: 10.3389/fgene.2018.00400] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 09/03/2018] [Indexed: 01/03/2023] Open
Abstract
Mitochondrial disorders, characterized by clinical symptoms and/or OXPHOS deficiencies, are caused by pathogenic variants in mitochondrial genes. However, pathogenic variants in some of these genes can lead to clinical manifestations which overlap with other neuromuscular diseases, which can be caused by pathogenic variants in non-mitochondrial genes as well. Mitochondrial pathogenic variants can be found in the mitochondrial DNA (mtDNA) or in any of the 1,500 nuclear genes with a mitochondrial function. We have performed a two-step next-generation sequencing approach in a cohort of 117 patients, mostly children, in whom a mitochondrial disease-cause could likely or possibly explain the phenotype. A total of 86 patients had a mitochondrial disorder, according to established clinical and biochemical criteria. The other 31 patients had neuromuscular symptoms, where in a minority a mitochondrial genetic cause is present, but a non-mitochondrial genetic cause is more likely. All patients were screened for pathogenic variants in the mtDNA and, if excluded, analyzed by whole exome sequencing (WES). Variants were filtered for being pathogenic and compatible with an autosomal or X-linked recessive mode of inheritance in families with multiple affected siblings and/or consanguineous parents. Non-consanguineous families with a single patient were additionally screened for autosomal and X-linked dominant mutations in a predefined gene-set. We identified causative pathogenic variants in the mtDNA in 20% of the patient-cohort, and in nuclear genes in 49%, implying an overall yield of 68%. We identified pathogenic variants in mitochondrial and non-mitochondrial genes in both groups with, obviously, a higher number of mitochondrial genes affected in mitochondrial disease patients. Furthermore, we show that 31% of the disease-causing genes in the mitochondrial patient group were not included in the MitoCarta database, and therefore would have been missed with MitoCarta based gene-panels. We conclude that WES is preferable to panel-based approaches for both groups of patients, as the mitochondrial gene-list is not complete and mitochondrial symptoms can be secondary. Also, clinically and genetically heterogeneous disorders would require sequential use of multiple different gene panels. We conclude that WES is a comprehensive and unbiased approach to establish a genetic diagnosis in these patients, able to resolve multi-genic disease-causes.
Collapse
Affiliation(s)
- Tom E J Theunissen
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands.,Research Institute GROW, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Minh Nguyen
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands.,Research Institute GROW, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Rick Kamps
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Alexandra T Hendrickx
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Suzanne C E H Sallevelt
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Ralph W H Gottschalk
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Chantal M Calis
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Alphons P M Stassen
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Bart de Koning
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | | | | | - Sabine A Fuchs
- Department of Metabolic Disorders, University Medical Centre Utrecht, Utrecht, Netherlands
| | | | - Marianne de Visser
- Department of Neurology, Academic Medical Centre Amsterdam, Amsterdam, Netherlands
| | - Jo Vanoevelen
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Radek Szklarczyk
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands.,Research Institute GROW, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Mike Gerards
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands.,Maastricht Center for Systems Biology (MaCSBio), Maastricht University Medical Centre, Maastricht, Netherlands
| | - Irenaeus F M de Coo
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands.,Department of Pediatric Neurology, Erasmus MC Sophia Children's Hospital, Rotterdam, Netherlands
| | - Debby M E I Hellebrekers
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Hubert J M Smeets
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, Netherlands.,Research Institute GROW, Maastricht University Medical Centre, Maastricht, Netherlands
| |
Collapse
|
8
|
Fiedorczuk K, Sazanov LA. Mammalian Mitochondrial Complex I Structure and Disease-Causing Mutations. Trends Cell Biol 2018; 28:835-867. [PMID: 30055843 DOI: 10.1016/j.tcb.2018.06.006] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 06/14/2018] [Accepted: 06/22/2018] [Indexed: 12/31/2022]
Abstract
Complex I has an essential role in ATP production by coupling electron transfer from NADH to quinone with translocation of protons across the inner mitochondrial membrane. Isolated complex I deficiency is a frequent cause of mitochondrial inherited diseases. Complex I has also been implicated in cancer, ageing, and neurodegenerative conditions. Until recently, the understanding of complex I deficiency on the molecular level was limited due to the lack of high-resolution structures of the enzyme. However, due to developments in single particle cryo-electron microscopy (cryo-EM), recent studies have reported nearly atomic resolution maps and models of mitochondrial complex I. These structures significantly add to our understanding of complex I mechanism and assembly. The disease-causing mutations are discussed here in their structural context.
Collapse
Affiliation(s)
- Karol Fiedorczuk
- Institute of Science and Technology Austria, Am Campus 1, Klosterneuburg 3400, Austria; Present address: The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Leonid A Sazanov
- Institute of Science and Technology Austria, Am Campus 1, Klosterneuburg 3400, Austria.
| |
Collapse
|
9
|
Patel M, McElroy PB. Mitochondrial Dysfunction in Parkinson’s Disease. OXIDATIVE STRESS AND REDOX SIGNALLING IN PARKINSON’S DISEASE 2017. [DOI: 10.1039/9781782622888-00061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Parkinson’s disease (PD) is one of the most common neurodegenerative disorders where oxidative stress and mitochondrial dysfunction have been implicated as etiological factors. Mitochondria are the major producers of reactive oxygen species (ROS) that can have damaging effects to cellular macromolecules leading to neurodegeneration. The most compelling evidence for the role of mitochondria in the pathogenesis of PD has been derived from toxicant-induced models of parkinsonism. Over the years, epidemiological studies have suggested a link between exposure to environmental toxins such as pesticides and the risk of developing PD. Data from human and experimental studies involving the use of chemical agents like paraquat, diquat, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, rotenone and maneb have provided valuable insight into the underlying mitochondrial mechanisms contributing to PD and associated neurodegeneration. In this review, we have discussed the role of mitochondrial ROS and dysfunction in the pathogenesis of PD with a special focus on environmental agent-induced parkinsonism. We have described the various mitochondrial mechanisms by which such chemicals exert neurotoxicity, highlighting some landmark epidemiological and experimental studies that support the role of mitochondrial ROS and oxidative stress in contributing to these effects. Finally, we have discussed the significance of these studies in understanding the mechanistic underpinnings of PD-related dopaminergic neurodegeneration.
Collapse
Affiliation(s)
- Manisha Patel
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus Aurora CO 80045 USA
| | - Pallavi Bhuyan McElroy
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus Aurora CO 80045 USA
| |
Collapse
|
10
|
Crombie DE, Curl CL, Raaijmakers AJA, Sivakumaran P, Kulkarni T, Wong RCB, Minami I, Evans-Galea MV, Lim SY, Delbridge L, Corben LA, Dottori M, Nakatsuji N, Trounce IA, Hewitt AW, Delatycki MB, Pera MF, Pébay A. Friedreich's ataxia induced pluripotent stem cell-derived cardiomyocytes display electrophysiological abnormalities and calcium handling deficiency. Aging (Albany NY) 2017; 9:1440-1452. [PMID: 28562313 PMCID: PMC5472743 DOI: 10.18632/aging.101247] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 05/27/2017] [Indexed: 01/12/2023]
Abstract
We sought to identify the impacts of Friedreich's ataxia (FRDA) on cardiomyocytes. FRDA is an autosomal recessive degenerative condition with neuronal and non-neuronal manifestations, the latter including progressive cardiomyopathy of the left ventricle, the leading cause of death in FRDA. Little is known about the cellular pathogenesis of FRDA in cardiomyocytes. Induced pluripotent stem cells (iPSCs) were derived from three FRDA individuals with characterized GAA repeats. The cells were differentiated into cardiomyocytes to assess phenotypes. FRDA iPSC- cardiomyocytes retained low levels of FRATAXIN (FXN) mRNA and protein. Electrophysiology revealed an increased variation of FRDA- cardiomyocyte beating rates which was prevented by addition of nifedipine, suggestive of a calcium handling deficiency. Finally, calcium imaging was performed and we identified small amplitude, diastolic and systolic calcium transients confirming a deficiency in calcium handling. We defined a robust FRDA cardiac-specific electrophysiological profile in patient-derived iPSCs which could be used for high throughput compound screening. This cell-specific signature will contribute to the identification and screening of novel treatments for this life-threatening disease.
Collapse
Affiliation(s)
- Duncan E. Crombie
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
- Ophthalmology, Department of Surgery, the University of Melbourne, Melbourne, Australia
| | - Claire L. Curl
- Department of Physiology, the University of Melbourne, Melbourne, Australia
| | | | | | - Tejal Kulkarni
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
- Ophthalmology, Department of Surgery, the University of Melbourne, Melbourne, Australia
- Centre for Neural Engineering & Department of Electrical and Electronic Engineering, The University of Melbourne, Melbourne, Australia
| | - Raymond CB Wong
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
- Ophthalmology, Department of Surgery, the University of Melbourne, Melbourne, Australia
| | - Itsunari Minami
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, Japan
| | - Marguerite V. Evans-Galea
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Childrens Research Institute, and Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| | - Shiang Y. Lim
- Ophthalmology, Department of Surgery, the University of Melbourne, Melbourne, Australia
- O'Brien Institute Department, St Vincent Institute of Medical Research, Fitzroy, Australia
| | - Lea Delbridge
- O'Brien Institute Department, St Vincent Institute of Medical Research, Fitzroy, Australia
| | - Louise A. Corben
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Childrens Research Institute, and Department of Paediatrics, The University of Melbourne, Melbourne, Australia
- School of Psychological Sciences, Monash University, Frankston, Australia
| | - Mirella Dottori
- Centre for Neural Engineering & Department of Electrical and Electronic Engineering, The University of Melbourne, Melbourne, Australia
| | - Norio Nakatsuji
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, Japan
| | - Ian A. Trounce
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
- Ophthalmology, Department of Surgery, the University of Melbourne, Melbourne, Australia
| | - Alex W. Hewitt
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
- Ophthalmology, Department of Surgery, the University of Melbourne, Melbourne, Australia
- Menzies Institute for Medical Research, School of Medicine, University of Tasmania, Hobart, Australia
| | - Martin B. Delatycki
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Childrens Research Institute, and Department of Paediatrics, The University of Melbourne, Melbourne, Australia
- School of Psychological Sciences, Monash University, Frankston, Australia
- Victorian Clinical Genetics Services, Parkville, Australia
| | - Martin F. Pera
- Department of Anatomy and Neurosciences, the University of Melbourne, Florey Neuroscience & Mental Health Institute, Walter and Eliza Hall Institute of Medical Research, Australia
| | - Alice Pébay
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
- Ophthalmology, Department of Surgery, the University of Melbourne, Melbourne, Australia
| |
Collapse
|
11
|
Coherent and Contradictory Facts, Feats and Fictions Associated with Metal Accumulation in Parkinson's Disease: Epicenter or Outcome, Yet a Demigod Question. Mol Neurobiol 2016; 54:4738-4755. [PMID: 27480264 DOI: 10.1007/s12035-016-0016-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 07/12/2016] [Indexed: 01/30/2023]
Abstract
Unwarranted exposure due to liberal use of metals for maintaining the lavish life and to achieve the food demand for escalating population along with an incredible boost in the average human life span owing to orchestrated progress in rejuvenation therapy have gradually increased the occurrence of Parkinson's disease (PD). Etiology is albeit elusive; association of PD with metal accumulation has never been overlooked due to noteworthy similitude between metal-exposure symptoms and a few cardinal features of disease. Even though metals are entailed in the vital functions, a hysterical shift, primarily augmentation, escorts the stern nigrostriatal dopaminergic neurodegeneration. An increase in the passage of metals through the blood brain barrier and impaired metabolic activity and elimination system could lead to metal accumulation in the brain, which eventually makes dopaminergic neurons quite susceptible. In the present article, an update on implication of metal accumulation in PD/Parkinsonism has been provided. Moreover, encouraging and paradoxical facts and fictions associated with metal accumulation in PD/Parkinsonism have also been compiled. Systematic literature survey of PD is performed to describe updated information if metal accumulation is an epicenter or merely an outcome. Finally, a perspective on the association of metal accumulation with pesticide-induced Parkinsonism has been explained to unveil the likely impact of the former in the latter.
Collapse
|
12
|
Mitochondrial complex I-linked disease. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2016; 1857:938-45. [DOI: 10.1016/j.bbabio.2016.02.012] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 02/16/2016] [Accepted: 02/18/2016] [Indexed: 11/22/2022]
|
13
|
Involvement of mitochondrial proteins in calcium signaling and cell death induced by staurosporine in Neurospora crassa. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2015; 1847:1064-74. [DOI: 10.1016/j.bbabio.2015.05.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Revised: 04/23/2015] [Accepted: 05/15/2015] [Indexed: 12/20/2022]
|
14
|
Alam MT, Manjeri GR, Rodenburg RJ, Smeitink JAM, Notebaart RA, Huynen M, Willems PHGM, Koopman WJH. Skeletal muscle mitochondria of NDUFS4-/- mice display normal maximal pyruvate oxidation and ATP production. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2015; 1847:526-33. [PMID: 25687896 DOI: 10.1016/j.bbabio.2015.02.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 02/03/2015] [Accepted: 02/07/2015] [Indexed: 10/24/2022]
Abstract
Mitochondrial ATP production is mediated by the oxidative phosphorylation (OXPHOS) system, which consists of four multi-subunit complexes (CI-CIV) and the FoF1-ATP synthase (CV). Mitochondrial disorders including Leigh Syndrome often involve CI dysfunction, the pathophysiological consequences of which still remain incompletely understood. Here we combined experimental and computational strategies to gain mechanistic insight into the energy metabolism of isolated skeletal muscle mitochondria from 5-week-old wild-type (WT) and CI-deficient NDUFS4-/- (KO) mice. Enzyme activity measurements in KO mitochondria revealed a reduction of 79% in maximal CI activity (Vmax), which was paralleled by 45-72% increase in Vmax of CII, CIII, CIV and citrate synthase. Mathematical modeling of mitochondrial metabolism predicted that these Vmax changes do not affect the maximal rates of pyruvate (PYR) oxidation and ATP production in KO mitochondria. This prediction was empirically confirmed by flux measurements. In silico analysis further predicted that CI deficiency altered the concentration of intermediate metabolites, modestly increased mitochondrial NADH/NAD+ ratio and stimulated the lower half of the TCA cycle, including CII. Several of the predicted changes were previously observed in experimental models of CI-deficiency. Interestingly, model predictions further suggested that CI deficiency only has major metabolic consequences when its activity decreases below 90% of normal levels, compatible with a biochemical threshold effect. Taken together, our results suggest that mouse skeletal muscle mitochondria possess a substantial CI overcapacity, which minimizes the effects of CI dysfunction on mitochondrial metabolism in this otherwise early fatal mouse model.
Collapse
Affiliation(s)
- Mohammad T Alam
- Department of Biochemistry, RIMLS, Radboud University Medical Center, Nijmegen, The Netherlands; Centre for Systems Biology and Bioenergetics, Radboud University Medical Centre, Nijmegen, The Netherlands.
| | - Ganesh R Manjeri
- Department of Biochemistry, RIMLS, Radboud University Medical Center, Nijmegen, The Netherlands; Centre for Systems Biology and Bioenergetics, Radboud University Medical Centre, Nijmegen, The Netherlands.
| | - Richard J Rodenburg
- Centre for Systems Biology and Bioenergetics, Radboud University Medical Centre, Nijmegen, The Netherlands; Department of Pediatrics, NCMD, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Jan A M Smeitink
- Centre for Systems Biology and Bioenergetics, Radboud University Medical Centre, Nijmegen, The Netherlands; Department of Pediatrics, NCMD, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Richard A Notebaart
- Centre for Systems Biology and Bioenergetics, Radboud University Medical Centre, Nijmegen, The Netherlands; Centre for Molecular and Biomolecular Informatics, RIMLS, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Martijn Huynen
- Centre for Systems Biology and Bioenergetics, Radboud University Medical Centre, Nijmegen, The Netherlands; Centre for Molecular and Biomolecular Informatics, RIMLS, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Peter H G M Willems
- Department of Biochemistry, RIMLS, Radboud University Medical Center, Nijmegen, The Netherlands; Centre for Systems Biology and Bioenergetics, Radboud University Medical Centre, Nijmegen, The Netherlands.
| | - Werner J H Koopman
- Department of Biochemistry, RIMLS, Radboud University Medical Center, Nijmegen, The Netherlands; Centre for Systems Biology and Bioenergetics, Radboud University Medical Centre, Nijmegen, The Netherlands.
| |
Collapse
|
15
|
Bongard RD, Townsley MI, Merker MP. The effects of mitochondrial complex I blockade on ATP and permeability in rat pulmonary microvascular endothelial cells in culture (PMVEC) are overcome by coenzyme Q1 (CoQ1). Free Radic Biol Med 2015; 79:69-77. [PMID: 25452141 DOI: 10.1016/j.freeradbiomed.2014.09.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 09/22/2014] [Accepted: 09/26/2014] [Indexed: 12/29/2022]
Abstract
In isolated rat lung perfused with a physiological saline solution (5.5mM glucose), complex I inhibitors decrease lung tissue ATP and increase endothelial permeability (Kf), effects that are overcome using an amphipathic quinone (CoQ1) [Free Radic. Biol. Med.65:1455-1463; 2013]. To address the microvascular endothelial contribution to these intact lung responses, rat pulmonary microvascular endothelial cells in culture (PMVEC) were treated with the complex I inhibitor rotenone and ATP levels and cell monolayer permeability (PS) were measured. There were no detectable effects on ATP or permeability in experimental medium that, like the lung perfusate, contained 5.5mM glucose. To unmask a potential mitochondrial contribution, the glucose concentration was lowered to 0.2mM. Under these conditions, rotenone decreased ATP from 18.4±1.6 (mean±SEM) to 4.6±0.8nmol/mg protein, depolarized the mitochondrial membrane potential (Δψm) from -129.0±3.7 (mean±SEM) to -92.8±5.5mV, and decreased O2 consumption from 2.0±0.1 (mean±SEM) to 0.3±0.1nmol/min/mg protein. Rotenone also increased PMVEC monolayer permeability (reported as PS in nl/min) to FITC-dextran (~40kDa) continually over a 6 h time course. When CoQ1 was present with rotenone, normal ATP (17.4±1.4nmol/mg protein), O2 consumption (1.5±0.1nmol/min/mg protein), Δψm (-125.2±3.3mV), and permeability (PS) were maintained. Protective effects of CoQ1 on rotenone-induced changes in ATP, O2 consumption rate, Δψm, and permeability were blocked by dicumarol or antimycin A, inhibitors of the quinone-mediated cytosol-mitochondria electron shuttle [Free Radic. Biol. Med.65:1455-1463; 2013]. Key rotenone effects without and with CoQ1 were qualitatively reproduced using the alternative complex I inhibitor, piericidin A. We conclude that, as in the intact lung, PMVEC ATP supply is linked to the permeability response to complex I inhibitors. In contrast to the intact lung, the association in PMVEC was revealed only after decreasing the glucose concentration in the experimental medium from 5.5 to 0.2mM.
Collapse
Affiliation(s)
- Robert D Bongard
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mary I Townsley
- Department of Physiology, University of South Alabama College of Medicine, Mobile, AL 36688, USA; Department of Medicine, University of South Alabama College of Medicine, Mobile, AL 36688, USA
| | - Marilyn P Merker
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Zablocki VAMC, Anesthesia Research, Milwaukee, WI 53295, USA.
| |
Collapse
|
16
|
Distelmaier F, Valsecchi F, Liemburg-Apers DC, Lebiedzinska M, Rodenburg RJ, Heil S, Keijer J, Fransen J, Imamura H, Danhauser K, Seibt A, Viollet B, Gellerich FN, Smeitink JAM, Wieckowski MR, Willems PHGM, Koopman WJH. Mitochondrial dysfunction in primary human fibroblasts triggers an adaptive cell survival program that requires AMPK-α. Biochim Biophys Acta Mol Basis Dis 2014; 1852:529-40. [PMID: 25536029 DOI: 10.1016/j.bbadis.2014.12.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 12/11/2014] [Accepted: 12/15/2014] [Indexed: 01/06/2023]
Abstract
Dysfunction of complex I (CI) of the mitochondrial electron transport chain (ETC) features prominently in human pathology. Cell models of ETC dysfunction display adaptive survival responses that still are poorly understood but of relevance for therapy development. Here we comprehensively examined how primary human skin fibroblasts adapt to chronic CI inhibition. CI inhibition triggered transient and sustained changes in metabolism, redox homeostasis and mitochondrial (ultra)structure but no cell senescence/death. CI-inhibited cells consumed no oxygen and displayed minor mitochondrial depolarization, reverse-mode action of complex V, a slower proliferation rate and futile mitochondrial biogenesis. Adaptation was neither prevented by antioxidants nor associated with increased PGC1-α/SIRT1/mTOR levels. Survival of CI-inhibited cells was strictly glucose-dependent and accompanied by increased AMPK-α phosphorylation, which occurred without changes in ATP or cytosolic calcium levels. Conversely, cells devoid of AMPK-α died upon CI inhibition. Chronic CI inhibition did not increase mitochondrial superoxide levels or cellular lipid peroxidation and was paralleled by a specific increase in SOD2/GR, whereas SOD1/CAT/Gpx1/Gpx2/Gpx5 levels remained unchanged. Upon hormone stimulation, fully adapted cells displayed aberrant cytosolic and ER calcium handling due to hampered ATP fueling of ER calcium pumps. It is concluded that CI dysfunction triggers an adaptive program that depends on extracellular glucose and AMPK-α. This response avoids cell death by suppressing energy crisis, oxidative stress induction and substantial mitochondrial depolarization.
Collapse
Affiliation(s)
- Felix Distelmaier
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands; Department of Pediatrics, Nijmegen Center for Mitochondrial Disorders, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands; Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Federica Valsecchi
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands; Department of Pediatrics, Nijmegen Center for Mitochondrial Disorders, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands
| | - Dania C Liemburg-Apers
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands
| | | | - Richard J Rodenburg
- Department of Pediatrics, Nijmegen Center for Mitochondrial Disorders, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands
| | - Sandra Heil
- Department of Human and Animal Physiology, Wageningen University, 6708 WD Wageningen, The Netherlands
| | - Jaap Keijer
- Department of Human and Animal Physiology, Wageningen University, 6708 WD Wageningen, The Netherlands
| | - Jack Fransen
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands
| | - Hiromi Imamura
- The Hakubi Project, Kyoto University, 606-8501 Kyoto, Japan
| | - Katharina Danhauser
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Annette Seibt
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Benoit Viollet
- Institut Cochin, NSERM U1016, Université Paris Descartes, 75014 Paris, France
| | - Frank N Gellerich
- Department of Stereotactic Neurosurgery, Otto-von-Guericke-Universität, 39120 Magdeburg, Germany
| | - Jan A M Smeitink
- Department of Pediatrics, Nijmegen Center for Mitochondrial Disorders, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands
| | | | - Peter H G M Willems
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands
| | - Werner J H Koopman
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands.
| |
Collapse
|
17
|
Scarpelli M, Todeschini A, Rinaldi F, Rota S, Padovani A, Filosto M. Strategies for treating mitochondrial disorders: an update. Mol Genet Metab 2014; 113:253-60. [PMID: 25458518 DOI: 10.1016/j.ymgme.2014.09.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Revised: 09/30/2014] [Accepted: 09/30/2014] [Indexed: 12/12/2022]
Abstract
Mitochondrial diseases are a heterogeneous group of disorders resulting from primary dysfunction of the respiratory chain due to both nuclear and mitochondrial DNA mutations. The wide heterogeneity of biochemical dysfunctions and pathogenic mechanisms typical of this group of diseases has hindered therapy trials; therefore, available treatment options remain limited. Therapeutic strategies aimed at increasing mitochondrial functions (by enhancing biogenesis and electron transport chain function), improving the removal of reactive oxygen species and noxious metabolites, modulating aberrant calcium homeostasis and repopulating mitochondrial DNA could potentially restore the respiratory chain dysfunction. The challenge that lies ahead is the translation of some promising laboratory results into safe and effective therapies for patients. In this review we briefly update and discuss the most feasible therapeutic approaches for mitochondrial diseases.
Collapse
Affiliation(s)
- Mauro Scarpelli
- Section of Neurology, Department of Neurological and Movement Sciences, University of Verona, Verona, Italy
| | - Alice Todeschini
- Clinical Neurology, Section for Neuromuscular Diseases and Neuropathies, University Hospital "Spedali Civili", Brescia, Italy
| | - Fabrizio Rinaldi
- Clinical Neurology, Section for Neuromuscular Diseases and Neuropathies, University Hospital "Spedali Civili", Brescia, Italy
| | - Silvia Rota
- Clinical Neurology, Section for Neuromuscular Diseases and Neuropathies, University Hospital "Spedali Civili", Brescia, Italy
| | - Alessandro Padovani
- Clinical Neurology, Section for Neuromuscular Diseases and Neuropathies, University Hospital "Spedali Civili", Brescia, Italy
| | - Massimiliano Filosto
- Clinical Neurology, Section for Neuromuscular Diseases and Neuropathies, University Hospital "Spedali Civili", Brescia, Italy.
| |
Collapse
|
18
|
Morató L, Bertini E, Verrigni D, Ardissone A, Ruiz M, Ferrer I, Uziel G, Pujol A. Mitochondrial dysfunction in central nervous system white matter disorders. Glia 2014; 62:1878-94. [DOI: 10.1002/glia.22670] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Revised: 03/20/2014] [Accepted: 03/21/2014] [Indexed: 12/26/2022]
Affiliation(s)
- Laia Morató
- Neurometabolic Diseases Laboratory; Bellvitge Biomedical Research Institute (IDIBELL); L'Hospitalet de Llobregat Barcelona Spain
- Center for Biomedical Research on Rare Diseases (CIBERER); ISCIII Spain
| | - Enrico Bertini
- Unit for Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Bambino Gesù Children's Hospital; IRCCS Rome Italy
| | - Daniela Verrigni
- Unit for Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Bambino Gesù Children's Hospital; IRCCS Rome Italy
| | - Anna Ardissone
- Department of Child Neurology The Foundation “Carlo Besta” Neurological Institute (IRCCS); Milan Italy
| | - Montse Ruiz
- Neurometabolic Diseases Laboratory; Bellvitge Biomedical Research Institute (IDIBELL); L'Hospitalet de Llobregat Barcelona Spain
- Center for Biomedical Research on Rare Diseases (CIBERER); ISCIII Spain
| | - Isidre Ferrer
- Institute of Neuropathology, University of Barcelona, L'Hospitalet de Llobregat; Barcelona Spain
- Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED); ISCIII Spain
| | - Graziella Uziel
- Department of Child Neurology The Foundation “Carlo Besta” Neurological Institute (IRCCS); Milan Italy
| | - Aurora Pujol
- Neurometabolic Diseases Laboratory; Bellvitge Biomedical Research Institute (IDIBELL); L'Hospitalet de Llobregat Barcelona Spain
- Center for Biomedical Research on Rare Diseases (CIBERER); ISCIII Spain
- Catalan Institution of Research and Advanced Studies (ICREA); Barcelona Spain
| |
Collapse
|
19
|
Groschner LN, Alam MR, Graier WF. Metabolism-secretion coupling and mitochondrial calcium activities in clonal pancreatic β-cells. VITAMINS AND HORMONES 2014; 95:63-86. [PMID: 24559914 DOI: 10.1016/b978-0-12-800174-5.00003-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pancreatic β-cells are the only cells capable of lowering blood glucose by secreting insulin. The β-cell continuously adjusts its secretory activity to substrate availability in order to keep blood glucose levels within the physiological range--a process called metabolism-secretion coupling. Glucose is readily taken up by the β-cell and broken down into intermediates that fuel oxidative metabolism inside the mitochondria to generate ATP. The resulting increase in the ATP/ADP ratio causes closure of plasma membrane KATP channels, thereby depolarizing the cell and triggering the opening of voltage-gated Ca²⁺ channels. Consequential oscillations of cytosolic Ca²⁺ not only mediate the exocytosis of insulin granules but are also relayed to other subcellular compartments including the mitochondria, where Ca²⁺ is required to accelerate mitochondrial metabolism in response to nutrient stimulation. The mitochondrial Ca²⁺ uptake machinery plays a fundamental role in this feed-forward mechanism that guarantees sustained insulin secretion and, thus, represents a promising therapeutic target for type 2 diabetes.
Collapse
Affiliation(s)
- Lukas N Groschner
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, Graz, Austria
| | - Muhammad Rizwan Alam
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, Graz, Austria
| | - Wolfgang F Graier
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, Graz, Austria.
| |
Collapse
|
20
|
Pienaar IS, Chinnery PF. Existing and emerging mitochondrial-targeting therapies for altering Parkinson's disease severity and progression. Pharmacol Ther 2013; 137:1-21. [DOI: 10.1016/j.pharmthera.2012.08.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 08/07/2012] [Indexed: 02/07/2023]
|
21
|
Distelmaier F, Valsecchi F, Forkink M, van Emst-de Vries S, Swarts HG, Rodenburg RJT, Verwiel ETP, Smeitink JAM, Willems PHGM, Koopman WJH. Trolox-sensitive reactive oxygen species regulate mitochondrial morphology, oxidative phosphorylation and cytosolic calcium handling in healthy cells. Antioxid Redox Signal 2012; 17:1657-69. [PMID: 22559215 PMCID: PMC3474189 DOI: 10.1089/ars.2011.4294] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
AIMS Cell regulation by signaling reactive oxygen species (sROS) is often incorrectly studied through extracellular oxidant addition. Here, we used the membrane-permeable antioxidant Trolox to examine the role of sROS in mitochondrial morphology, oxidative phosphorylation (OXPHOS), and cytosolic calcium (Ca(2+)) handling in healthy human skin fibroblasts. RESULTS AND INNOVATION Trolox treatment reduced the levels of 5-(and-6)-chloromethyl-2',7'-dichlorodihydro-fluorescein (CM-H(2)DCF) oxidizing ROS, lowered cellular lipid peroxidation, and induced a less oxidized mitochondrial thiol redox state. This was paralleled by increased glutathione- and mitofusin-dependent mitochondrial filamentation, increased expression of fully assembled mitochondrial complex I, elevated activity of citrate synthase and OXPHOS enzymes, and a higher cellular O(2) consumption. In contrast, Trolox did not alter hydroethidium oxidation, cytosolic thiol redox state, mitochondrial NAD(P)H levels, or mitochondrial membrane potential. Whole genome expression profiling revealed that Trolox did not trigger significant changes in gene expression, suggesting that Trolox acts downstream of this process. Cytosolic Ca(2+) transients, induced by the hormone bradykinin, were of a higher amplitude and decayed faster in Trolox-treated cells. These effects were dose-dependently antagonized by hydrogen peroxide. CONCLUSIONS Our findings suggest that Trolox-sensitive sROS are upstream regulators of mitochondrial mitofusin levels, morphology, and function in healthy human skin fibroblasts. This information not only facilitates the interpretation of antioxidant effects in cell models (of oxidative-stress), but also contributes to a better understanding of ROS-related human pathologies, including mitochondrial disorders.
Collapse
Affiliation(s)
- Felix Distelmaier
- Department of Biochemistry, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
During the past two decades calcium (Ca(2+)) accumulation in energized mitochondria has emerged as a biological process of utmost physiological relevance. Mitochondrial Ca(2+) uptake was shown to control intracellular Ca(2+) signalling, cell metabolism, cell survival and other cell-type specific functions by buffering cytosolic Ca(2+) levels and regulating mitochondrial effectors. Recently, the identity of mitochondrial Ca(2+) transporters has been revealed, opening new perspectives for investigation and molecular intervention.
Collapse
|
23
|
Valsecchi F, Monge C, Forkink M, de Groof AJC, Benard G, Rossignol R, Swarts HG, van Emst-de Vries SE, Rodenburg RJ, Calvaruso MA, Nijtmans LGJ, Heeman B, Roestenberg P, Wieringa B, Smeitink JAM, Koopman WJH, Willems PHGM. Metabolic consequences of NDUFS4 gene deletion in immortalized mouse embryonic fibroblasts. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2012; 1817:1925-36. [PMID: 22430089 DOI: 10.1016/j.bbabio.2012.03.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Revised: 03/01/2012] [Accepted: 03/04/2012] [Indexed: 12/17/2022]
Abstract
Human mitochondrial complex I (CI) deficiency is associated with progressive neurological disorders. To better understand the CI pathomechanism, we here studied how deletion of the CI gene NDUFS4 affects cell metabolism. To this end we compared immortalized mouse embryonic fibroblasts (MEFs) derived from wildtype (wt) and whole-body NDUFS4 knockout (KO) mice. Mitochondria from KO cells lacked the NDUFS4 protein and mitoplasts displayed virtually no CI activity, moderately reduced CII, CIII and CIV activities and normal citrate synthase and CV (F(o)F(1)-ATPase) activity. Native electrophoresis of KO cell mitochondrial fractions revealed two distinct CI subcomplexes of ~830kDa (enzymatically inactive) and ~200kDa (active). The level of fully-assembled CII-CV was not affected by NDUFS4 gene deletion. KO cells exhibited a moderately reduced maximal and routine O(2) consumption, which was fully inhibited by acute application of the CI inhibitor rotenone. The aberrant CI assembly and reduced O(2) consumption in KO cells were fully normalized by NDUFS4 gene complementation. Cellular [NAD(+)]/[NADH] ratio, lactate production and mitochondrial tetramethyl rhodamine methyl ester (TMRM) accumulation were slightly increased in KO cells. In contrast, NDUFS4 gene deletion did not detectably alter [NADP(+)]/[NADPH] ratio, cellular glucose consumption, the protein levels of hexokinases (I and II) and phosphorylated pyruvate dehydrogenase (P-PDH), total cellular adenosine triphosphate (ATP) level, free cytosolic [ATP], cell growth rate, and reactive oxygen species (ROS) levels. We conclude that the NDUFS4 subunit is of key importance in CI stabilization and that, due to the metabolic properties of the immortalized MEFs, NDUFS4 gene deletion has only modest effects at the live cell level. This article is part of a special issue entitled: 17th European Bioenergetics Conference (EBEC 2012).
Collapse
Affiliation(s)
- Federica Valsecchi
- Department of Biochemistry, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Calcium Signaling in Mast Cells: Focusing on L-Type Calcium Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 740:955-77. [DOI: 10.1007/978-94-007-2888-2_44] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
25
|
Ma HT, Beaven MA. Regulators of Ca(2+) signaling in mast cells: potential targets for treatment of mast cell-related diseases? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 716:62-90. [PMID: 21713652 DOI: 10.1007/978-1-4419-9533-9_5] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
A calcium signal is essential for degranulation, generation of eicosanoids and optimal production of cytokines in mast cells in response to antigen and other stimulants. The signal is initiated by phospholipase C-mediated production of inositol1,4,5-trisphosphate resulting in release of stored Ca(2+) from the endoplasmic reticulum (ER) and Golgi. Depletion of these stores activates influx of extracellular Ca(2+), usually referred to as store-operated calcium entry (SOCE), through the interaction of the Ca(2+)-sensor, stromal interacting molecule-1 (STIM1 ), in ER with Orai1(CRACM1) and transient receptor potential canonical (TRPC) channel proteins in the plasma membrane (PM). This interaction is enabled by microtubular-directed reorganization of ER to form ER/PM contact points or "punctae" in which STIM1 and channel proteins colocalize. The ensuing influx of Ca(2+) replenishes Ca(2+) stores and sustains elevated levels of cytosolic Ca(2+) ions-the obligatory signal for mast-cell activation. In addition, the signal can acquire spatial and dynamic characteristics (e.g., calcium puffs, waves, oscillations) that encode signals for specific functional outputs. This is achieved by coordinated regulation of Ca(2+) fluxes through ATP-dependent Ca(2+)-pumps and ion exchangers in mitochondria, ER and PM. As discussed in this chapter, studies in mast cells revealed much about the mechanisms described above but little about allergic and autoimmune diseases although studies in other types of cells have exposed genetic defects that lead to aberrant calcium signaling in immune diseases. Pharmacologic agents that inhibit or activate the regulatory components of calcium signaling in mast cells are also discussed along with the prospects for development of novel SOCE inhibitors that may prove beneficial in the treatment inflammatory mast-cell related diseases.
Collapse
Affiliation(s)
- Hong-Tao Ma
- Laboratory of Molecular Immunology, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | |
Collapse
|
26
|
Giorgi C, Agnoletto C, Bononi A, Bonora M, De Marchi E, Marchi S, Missiroli S, Patergnani S, Poletti F, Rimessi A, Suski JM, Wieckowski MR, Pinton P. Mitochondrial calcium homeostasis as potential target for mitochondrial medicine. Mitochondrion 2011; 12:77-85. [PMID: 21798374 PMCID: PMC3281195 DOI: 10.1016/j.mito.2011.07.004] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Revised: 06/10/2011] [Accepted: 07/11/2011] [Indexed: 11/28/2022]
Abstract
Mitochondria are crucial in different intracellular pathways of signal transduction. Mitochondria are capable of decoding a variety of extracellular stimuli into markedly different intracellular actions, ranging from energy production to cell death. The fine modulation of mitochondrial calcium (Ca2+) homeostasis plays a fundamental role in many of the processes involving this organelle. When mitochondrial Ca2+ homeostasis is compromised, different pathological conditions can occur, depending on the cell type involved. Recent data have shed light on the molecular identity of the main proteins involved in the handling of mitochondrial Ca2+ traffic, opening fascinating and ambitious new avenues for mitochondria-based pharmacological strategies.
Collapse
Affiliation(s)
- Carlotta Giorgi
- Department of Experimental and Diagnostic Medicine, Section of General Pathology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Chiara Agnoletto
- Department of Experimental and Diagnostic Medicine, Section of General Pathology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Angela Bononi
- Department of Experimental and Diagnostic Medicine, Section of General Pathology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Massimo Bonora
- Department of Experimental and Diagnostic Medicine, Section of General Pathology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Elena De Marchi
- Department of Experimental and Diagnostic Medicine, Section of General Pathology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Saverio Marchi
- Department of Experimental and Diagnostic Medicine, Section of General Pathology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Sonia Missiroli
- Department of Experimental and Diagnostic Medicine, Section of General Pathology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Simone Patergnani
- Department of Experimental and Diagnostic Medicine, Section of General Pathology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Federica Poletti
- Department of Experimental and Diagnostic Medicine, Section of General Pathology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Alessandro Rimessi
- Department of Experimental and Diagnostic Medicine, Section of General Pathology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Jan M. Suski
- Department of Experimental and Diagnostic Medicine, Section of General Pathology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
- Nencki Institute of Experimental Biology, Warsaw, Poland
| | | | - Paolo Pinton
- Department of Experimental and Diagnostic Medicine, Section of General Pathology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
- Corresponding author at: Department of Experimental and Diagnostic Medicine, Section of General Pathology, Via Borsari, 46 44100 Ferrara, Italy.
| |
Collapse
|
27
|
Roestenberg P, Manjeri GR, Valsecchi F, Smeitink JAM, Willems PHGM, Koopman WJH. Pharmacological targeting of mitochondrial complex I deficiency: the cellular level and beyond. Mitochondrion 2011; 12:57-65. [PMID: 21757032 DOI: 10.1016/j.mito.2011.06.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Revised: 01/20/2011] [Accepted: 06/25/2011] [Indexed: 12/20/2022]
Abstract
Complex I (CI) represents a major entry point of electrons in the mitochondrial electron transport chain (ETC). It consists of 45 different subunits, encoded by the mitochondrial (mtDNA) and nuclear DNA (nDNA). In humans, mutations in nDNA-encoded subunits cause severe neurodegenerative disorders like Leigh Syndrome with onset in early childhood. The pathophysiological mechanism of these disorders is still poorly understood. Here we summarize the current knowledge concerning the consequences of nDNA-encoded CI mutations in patient-derived cells, present mouse models for human CI deficiency, and discuss potential treatment strategies for CI deficiency.
Collapse
Affiliation(s)
- Peggy Roestenberg
- Department of Biochemistry, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | | | | | | | |
Collapse
|
28
|
Effect of selenite on basic mitochondrial function in human osteosarcoma cells with chronic mitochondrial stress. Mitochondrion 2011; 12:149-55. [PMID: 21742063 DOI: 10.1016/j.mito.2011.06.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 06/20/2011] [Accepted: 06/24/2011] [Indexed: 11/22/2022]
Abstract
Mitochondrial chronic stress that originates from defective mitochondria is implicated in a growing list of human diseases. To enhance understanding of pathophysiology of chronic mitochondrial dysfunction we investigated human osteosarcoma cells with 2 types of chronic stress: corresponding to the mutation in ATP synthase subunit 6 encoded by mtDNA (NARP syndrome-mild stress) and to a total lack of mtDNA (Rho0 cells-heavy stress). We previously found that selenium influenced mitochondrial stress response and lowered ROS production. Therefore, in this study effect of selenite on other mitochondrial parameters was investigated. We showed that presence of selenium improved survival of starved cells, modified organization of mitochondrial network in NARP cybrids and decreased cytosolic calcium level in NARP and Rho0 cells. Selenium did not affect mitochondrial membrane potential, ATP level, activity of ATP synthase and activity of complex II of the respiratory chain.
Collapse
|
29
|
Heeman B, Van den Haute C, Aelvoet SA, Valsecchi F, Rodenburg RJ, Reumers V, Debyser Z, Callewaert G, Koopman WJH, Willems PHGM, Baekelandt V. Depletion of PINK1 affects mitochondrial metabolism, calcium homeostasis and energy maintenance. J Cell Sci 2011; 124:1115-25. [DOI: 10.1242/jcs.078303] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Loss-of-function mutations in the gene encoding the mitochondrial PTEN-induced putative kinase 1 (PINK1) are a major cause of early-onset familial Parkinson's disease (PD). Recent studies have highlighted an important function for PINK1 in clearing depolarized mitochondria by mitophagy. However, the role of PINK1 in mitochondrial and cellular functioning in physiological conditions is still incompletely understood. Here, we investigate mitochondrial and cellular calcium (Ca2+) homeostasis in PINK1-knockdown and PINK1-knockout mouse cells, both in basal metabolic conditions and after physiological stimulation, using unbiased automated live single-cell imaging in combination with organelle-specific fluorescent probes. Our data reveal that depletion of PINK1 induces moderate fragmentation of the mitochondrial network, mitochondrial membrane depolarization and increased production of reactive oxygen species. This results in reduced uptake of Ca2+ by mitochondria after physiological stimulation. As a consequence, cells with knockdown or knockout of PINK1 display impaired mitochondrial ATP synthesis, which is exacerbated under conditions of increased ATP demand, thereby affecting cytosolic Ca2+ extrusion. The impairment in energy maintenance was confirmed in the brain of PINK1-knockout mice by in vivo bioluminescence imaging. Our findings demonstrate a key role for PINK1 in the regulation of mitochondrial homeostasis and energy metabolism under physiological conditions.
Collapse
Affiliation(s)
- Bavo Heeman
- Laboratory for Neurobiology and Gene Therapy, Molecular Medicine, Katholieke Universiteit Leuven, 3000 Leuven, Flanders, Belgium
| | - Chris Van den Haute
- Laboratory for Neurobiology and Gene Therapy, Molecular Medicine, Katholieke Universiteit Leuven, 3000 Leuven, Flanders, Belgium
| | - Sarah-Ann Aelvoet
- Laboratory for Neurobiology and Gene Therapy, Molecular Medicine, Katholieke Universiteit Leuven, 3000 Leuven, Flanders, Belgium
| | - Federica Valsecchi
- Department of Biochemistry (286), Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, 6500 Nijmegen, the Netherlands
- Department of Pediatrics, Nijmegen Centre for Mitochondrial Disorders, Radboud University Nijmegen Medical Centre, 6500 Nijmegen, the Netherlands
| | - Richard J. Rodenburg
- Department of Pediatrics, Nijmegen Centre for Mitochondrial Disorders, Radboud University Nijmegen Medical Centre, 6500 Nijmegen, the Netherlands
| | - Veerle Reumers
- Laboratory for Neurobiology and Gene Therapy, Molecular Medicine, Katholieke Universiteit Leuven, 3000 Leuven, Flanders, Belgium
| | - Zeger Debyser
- Laboratory for Molecular Virology and Gene Therapy, Molecular Medicine, Katholieke Universiteit Leuven and Interdisciplinary Research Centre, Katholieke Universiteit Leuven Campus Kortrijk, 8500 Kortrijk, Flanders, Belgium
| | - Geert Callewaert
- Research Group Neurodegeneration, Interdisciplinary Research Centre, Katholieke Universiteit Leuven Campus Kortrijk, 8500 Kortrijk, Flanders, Belgium
| | - Werner J. H. Koopman
- Department of Biochemistry (286), Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, 6500 Nijmegen, the Netherlands
| | - Peter H. G. M. Willems
- Department of Biochemistry (286), Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, 6500 Nijmegen, the Netherlands
| | - Veerle Baekelandt
- Laboratory for Neurobiology and Gene Therapy, Molecular Medicine, Katholieke Universiteit Leuven, 3000 Leuven, Flanders, Belgium
| |
Collapse
|
30
|
Koene S, Willems PHGM, Roestenberg P, Koopman WJH, Smeitink JAM. Mouse models for nuclear DNA-encoded mitochondrial complex I deficiency. J Inherit Metab Dis 2011; 34:293-307. [PMID: 20107904 DOI: 10.1007/s10545-009-9005-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2009] [Revised: 09/17/2009] [Accepted: 10/08/2009] [Indexed: 02/08/2023]
Abstract
Mitochondrial diseases are a group of heterogeneous pathologies with decreased cellular energy production as a common denominator. Defects in the oxidative phosphorylation (OXPHOS) system, the most frequent one in humans being isolated complex I deficiency (OMIM 252010), underlie this disturbed-energy generation. As biogenesis of OXPHOS complexes is under dual genetic control, with complex II being the sole exception, mutations in both nuclear DNA (nDNA) and mitochondrial DNA (mtDNA) are found. Increasing knowledge is becoming available with respect to the pathophysiology and cellular consequences of OXPHOS dysfunction. This aids the rational design of new treatment strategies. Recently, the first successful treatment trials were carried out in patient-derived cell lines. In these studies chemical compounds were used that target cellular aberrations induced by complex I dysfunction. Before the field of human clinical trials is entered, it is necessary to study the effects of these compounds with respect to toxicity, pharmacokinetics and therapeutic potential in suitable animal models. Here, we discuss two recent mouse models for nDNA-encoded complex I deficiency and their tissue-specific knock-outs.
Collapse
Affiliation(s)
- Saskia Koene
- Department of Paediatrics, Nijmegen Centre for Mitochondrial Disorders, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | | | | | |
Collapse
|
31
|
Koene S, Smeitink J. Metabolic manipulators: a well founded strategy to combat mitochondrial dysfunction. J Inherit Metab Dis 2011; 34:315-25. [PMID: 20668944 PMCID: PMC3063543 DOI: 10.1007/s10545-010-9162-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2010] [Revised: 06/11/2010] [Accepted: 06/23/2010] [Indexed: 01/27/2023]
Abstract
Whilst the pathophysiology and genetics of mitochondrial disease are slowly being unraveled, currently no effective remedy for mitochondrial disorders is available. One particular strategy in mitochondrial medicine presently under study is metabolic manipulation. This approach is aimed at counteracting the deranged cell biological homeostasis caused by mitochondrial dysfunction, using dietary modifications or small molecule therapy. Cell biological alterations caused by mitochondrial dysfunction include increased reactive oxygen species production, enhanced lipid peroxidation and altered cellular calcium homeostasis. This review covers the five principles of metabolic manipulation: (1) prevention of oxidative damage by reactive oxygen species, (2) amelioration of lipid peroxidation, (3) correction of altered membrane potential, (4) restoration of calcium homeostasis, and (5) transcription regulation interference. We hypothesize that a combination of compounds targeting different metabolic pathways will abolish cellular disturbance arising as a consequences of mitochondrial dysfunction, and thereby improve or stabilize clinical features. However, only a handful of compounds have reached efficacy testing in mammals, and it remains unknown to what extent metabolic manipulation will affect the whole organism. Until a potent remedy is found, patients will remain dependent on supportive, not curative, interventions.
Collapse
Affiliation(s)
- Saskia Koene
- Nijmegen Centre for Mitochondrial Disorders, Radboud University Nijmegen Medical Centre, Geert Grooteplein 10, 6500 HB PO BOX 9101, Nijmegen, The Netherlands
| | - Jan Smeitink
- Nijmegen Centre for Mitochondrial Disorders, Radboud University Nijmegen Medical Centre, Geert Grooteplein 10, 6500 HB PO BOX 9101, Nijmegen, The Netherlands
| |
Collapse
|
32
|
Coussee E, De Smet P, Bogaert E, Elens I, Van Damme P, Willems P, Koopman W, Van Den Bosch L, Callewaert G. G37R SOD1 mutant alters mitochondrial complex I activity, Ca(2+) uptake and ATP production. Cell Calcium 2011; 49:217-25. [PMID: 21388680 DOI: 10.1016/j.ceca.2011.02.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Revised: 02/10/2011] [Accepted: 02/14/2011] [Indexed: 02/06/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by selective death of motor neurons. Mutations in Cu/Zn superoxide dismutase-1 (SOD1) cause familial ALS but the molecular mechanisms whereby these mutations induce motor neuron death remain controversial. Here, we show that stable overexpression of mutant human SOD1 (G37R) - but not wild-type SOD1 (wt-SOD1) - in mouse neuroblastoma cells (N2a) results in morphological abnormalities of mitochondria accompanied by several dysfunctions. Activity of the oxidative phosphorylation complex I was significantly reduced in G37R cells and correlated with lower mitochondrial membrane potential and reduced levels of cytosolic ATP. Using targeted chimeric aequorin we further analyzed the consequences of mitochondrial dysfunction on cellular Ca(2+) handling. Mitochondrial Ca(2+) uptake, elicited by IP(3)-induced Ca(2+) release from endoplasmic reticulum (ER) was significantly reduced in G37R cells, while uptake induced by a brief Ca(2+) pulse was not affected in permeabilized cells. The decreased mitochondrial Ca(2+) uptake resulted in increased cytosolic Ca(2+) transients, whereas ER Ca(2+) load and resting cytosolic Ca(2+) levels were not affected. Together, these findings suggest that the mechanism linking mutant G37R SOD1 and ALS involves mitochondrial respiratory chain deficiency resulting in ATP loss and impairment of mitochondrial and cytosolic Ca(2+) homeostasis.
Collapse
Affiliation(s)
- Evelyne Coussee
- Research Group Neurodegeneration, Campus Kortrijk, K.U. Leuven, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Wenz T, Williams SL, Bacman SR, Moraes CT. Emerging therapeutic approaches to mitochondrial diseases. ACTA ACUST UNITED AC 2011; 16:219-29. [PMID: 20818736 DOI: 10.1002/ddrr.109] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Mitochondrial diseases are very heterogeneous and can affect different tissues and organs. Moreover, they can be caused by genetic defects in either nuclear or mitochondrial DNA as well as by environmental factors. All of these factors have made the development of therapies difficult. In this review article, we will discuss emerging approaches to the therapy of mitochondrial disorders, some of which are targeted to specific conditions whereas others may be applicable to a more diverse group of patients.
Collapse
Affiliation(s)
- Tina Wenz
- Department of Neurology, University of Miami School of Medicine, 1095 NW 14th Terrace, Miami, FL 33136, USA
| | | | | | | |
Collapse
|
34
|
Nugent S, Mothersill CE, Seymour C, McClean B, Lyng FM, Murphy JEJ. Altered mitochondrial function and genome frequency post exposure to γ-radiation and bystander factors. Int J Radiat Biol 2010; 86:829-41. [DOI: 10.3109/09553002.2010.486019] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
35
|
Valsecchi F, Koopman WJ, Manjeri GR, Rodenburg RJ, Smeitink JA, Willems PH. Complex I disorders: Causes, mechanisms, and development of treatment strategies at the cellular level. ACTA ACUST UNITED AC 2010; 16:175-82. [DOI: 10.1002/ddrr.107] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
36
|
Koopman WJH, Nijtmans LGJ, Dieteren CEJ, Roestenberg P, Valsecchi F, Smeitink JAM, Willems PHGM. Mammalian mitochondrial complex I: biogenesis, regulation, and reactive oxygen species generation. Antioxid Redox Signal 2010; 12:1431-70. [PMID: 19803744 DOI: 10.1089/ars.2009.2743] [Citation(s) in RCA: 308] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Virtually every mammalian cell contains mitochondria. These double-membrane organelles continuously change shape and position and contain the complete metabolic machinery for the oxidative conversion of pyruvate, fatty acids, and amino acids into ATP. Mitochondria are crucially involved in cellular Ca2+ and redox homeostasis and apoptosis induction. Maintenance of mitochondrial function and integrity requires an inside-negative potential difference across the mitochondrial inner membrane. This potential is sustained by the electron-transport chain (ETC). NADH:ubiquinone oxidoreductase or complex I (CI), the first and largest protein complex of the ETC, couples the oxidation of NADH to the reduction of ubiquinone. During this process, electrons can escape from CI and react with ambient oxygen to produce superoxide and derived reactive oxygen species (ROS). Depending on the balance between their production and removal by antioxidant systems, ROS may function as signaling molecules or induce damage to a variety of biomolecules or both. The latter ultimately leads to a loss of mitochondrial and cellular function and integrity. In this review, we discuss (a) the role of CI in mitochondrial functioning; (b) the composition, structure, and biogenesis of CI; (c) regulation of CI function; (d) the role of CI in ROS generation; and (e) adaptive responses to CI deficiency.
Collapse
Affiliation(s)
- Werner J H Koopman
- Department of Biochemistry, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
37
|
Frantz MC, Wipf P. Mitochondria as a target in treatment. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2010; 51:462-75. [PMID: 20175113 PMCID: PMC2920596 DOI: 10.1002/em.20554] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Mitochondria are key organelles that perform essential cellular functions and play pivotal roles in cell death and survival signaling. Hence, they represent an attractive target for drugs to treat metabolic, degenerative, and hyperproliferative diseases. Targeting mitochondria with organelle-specific agents or prodrugs has proven to be an effective therapeutic strategy. More specifically, controlling the cellular ROS balance via selective delivery of an antioxidant "payload" into mitochondria is an elegant emerging therapeutic concept. Herein, we review the recent medicinal chemistry and clinical data of these exploratory strategies, which should point the way for future generations of therapeutics.
Collapse
Affiliation(s)
- Marie-Céline Frantz
- Correspondence to: Dr. Peter Wipf, Department of Chemistry, Parkman Ave. CSC 1301, University of Pittsburgh, Pittsburgh, PA 15260, USA, ; or Dr. Marie-Céline Frantz, Department of Chemistry, Parkman Ave. CSC 1310, University of Pittsburgh, Pittsburgh, PA 15260, USA,
| | - Peter Wipf
- Correspondence to: Dr. Peter Wipf, Department of Chemistry, Parkman Ave. CSC 1301, University of Pittsburgh, Pittsburgh, PA 15260, USA, ; or Dr. Marie-Céline Frantz, Department of Chemistry, Parkman Ave. CSC 1310, University of Pittsburgh, Pittsburgh, PA 15260, USA,
| |
Collapse
|
38
|
Abstract
Until even only a few years ago, the idea that effective therapies for human mitochondrial disorders resulting from the dysfunction of the respiratory chain/oxidative phosphorylation system (OxPhos) could be developed was unimaginable. The obstacles to treating diseases caused by mutations in either mitochondrial DNA (mtDNA) or nuclear DNA (nDNA), and which had the potential to affect nearly every organ system, seemed overwhelming. However, although clinically applicable therapies remain largely in the future, the landscape has changed dramatically and we can now envision the possibility of treating some of these disorders. Among these are techniques to upregulate mitochondrial biogenesis, enhance organellar fusion and fission, "shift heteroplasmy" and eliminate the burden of mutant mtDNAs via cytoplasmic transfer.
Collapse
Affiliation(s)
- Eric A Schon
- Department of Neurology, Columbia University Medical Center, New York, NY, USA.
| | | | | | | |
Collapse
|
39
|
Carafoli E. The fateful encounter of mitochondria with calcium: how did it happen? BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2010; 1797:595-606. [PMID: 20385096 DOI: 10.1016/j.bbabio.2010.03.024] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2010] [Revised: 03/29/2010] [Accepted: 03/30/2010] [Indexed: 01/09/2023]
Abstract
A number of findings in the 1950s had offered indirect indications that mitochondria could accumulate Ca2+. In 1961, the phenomenon was directly demonstrated using isolated mitochondria: the uptake process was driven by respiratory chain activity or by the hydrolysis of added ATP. It could be accompanied by the simultaneous uptake of inorganic phosphate, in which case precipitates of hydroxyapatite were formed in the matrix, buffering its free Ca2+ concentration. The properties of the uptake process were established in the 1960s and 1970s: the uptake of Ca2+ occurred electrophoretically on a carrier that has not yet been molecularly identified, and was released from mitochondria via a Na+/Ca2+ antiporter. A H+/Ca2+ release exchanger was also found to operate in some mitochondrial types. The permeability transition pore was later also found to mediate the efflux of Ca2+ from mitochondria. In the mitochondrial matrix two TCA cycle dehydrogenases and pyruvate dehydrogenase phosphate phosphatase were found to be regulated in the matrix by the cycling of Ca2+ across the inner membrane. In conditions of cytoplasmic Ca2+ overload mitochondria could store for a time large amounts of precipitated Ca2+-phosphate, thus permitting cells to survive situations of Ca2+ emergency. The uptake process was found to have very low affinity for Ca2+: since the bulk concentration of Ca2+ in the cytoplasm is in the low to mid-nM range, it became increasingly difficult to postulate a role of mitochondria in the regulation of cytoplsmic Ca2+. A number of findings had nevertheless shown that energy linked Ca2+ transport occurred efficiently in mitochondria of various tissues in situ. The paradox was only solved in the 1990s, when it was found that the concentration of Ca2+ in the cytoplasm is not uniform: perimitochondrial micropools are created by the agonist-promoted discharge of Ca2+ from vicinal stores in which the concentration of Ca2+ is high enough to activate the low affinity mitochondrial uniporter. Mitochondria thus regained center stage as important regulators of cytoplasmic Ca2+ (not only of their own internal Ca2+). Their Ca2+ uptake systems was found to react very rapidly to cytoplasmic Ca2+ demands, even in the 150-200 msec time scale of processes like the contraction and relaxation of heart. An important recent development in the area of mitochondrial Ca2+ transport is its involvement in the disease process. Ca2+ signaling defects are now gaining increasing importance in the pathogenesis of diseases, e.g., neurodegenerative diseases. Since mitochondria have now regained a central role in the regulation of cytoplasmic Ca2+, dysfunctions of their Ca2+ controlling systems have expectedly been found to be involved in the pathogenesis of numerous disease processes.
Collapse
Affiliation(s)
- Ernesto Carafoli
- Department of Biochemistry and Venetian Institute of Molecular Medicine, University of Padova, Italy.
| |
Collapse
|
40
|
Trevelyan AJ, Kirby DM, Smulders-Srinivasan TK, Nooteboom M, Acin-Perez R, Enriquez JA, Whittington MA, Lightowlers RN, Turnbull DM. Mitochondrial DNA mutations affect calcium handling in differentiated neurons. ACTA ACUST UNITED AC 2010; 133:787-96. [PMID: 20207702 PMCID: PMC2842518 DOI: 10.1093/brain/awq023] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Mutations in the mitochondrial genome are associated with a wide range of neurological symptoms, but many aspects of the basic neuronal pathology are not understood. One candidate mechanism, given the well-established role of mitochondria in calcium buffering, is a deficit in neuronal calcium homoeostasis. We therefore examined calcium responses in the neurons derived from various ‘cybrid’ embryonic stem cell lines carrying different mitochondrial DNA mutations. Brief (∼50 ms), focal glutamatergic stimuli induced a transient rise in intracellular calcium concentration, which was visualized by bulk loading the cells with the calcium dye, Oregon Green BAPTA-1. Calcium entered the neurons through N-methyl-d-aspartic acid and voltage-gated calcium channels, as has been described in many other neuronal classes. Intriguingly, while mitochondrial mutations did not affect the calcium transient in response to single glutamatergic stimuli, they did alter the responses to repeated stimuli, with each successive calcium transient decaying ever more slowly in mitochondrial mutant cell lines. A train of stimuli thus caused intracellular calcium in these cells to be significantly elevated for many tens of seconds. These results suggest that calcium-handling deficits are likely to contribute to the pathological phenotype seen in patients with mitochondrial DNA mutations.
Collapse
Affiliation(s)
- Andrew J Trevelyan
- Mitochondrial Research Group, Institute for Ageing and Health Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, England.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Mitochondrial Ca2+ cycling facilitates activation of the transcription factor NFAT in sensory neurons. J Neurosci 2009; 29:12101-14. [PMID: 19793968 DOI: 10.1523/jneurosci.3384-09.2009] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Ca(2+)-dependent gene regulation controls many aspects of neuronal plasticity. Significant progress has been made toward understanding the roles of voltage- and ligand-gated Ca(2+) channels in triggering specific transcriptional responses. In contrast, the functional importance of Ca(2+) buffers and Ca(2+) transporters in neuronal gene regulation is less clear despite their critical contribution to the spatiotemporal control of Ca(2+) signals. Here we examined the role of mitochondrial Ca(2+) uptake and release in regulating the Ca(2+)-dependent transcription factor NFAT (nuclear factor of activated T-cells), which has been implicated in synaptic plasticity, axonal growth, and neuronal survival. Intense stimulation of sensory neurons by action potentials or TRPV1 agonists induced rapid activation and nuclear import of NFAT. Nuclear translocation of NFAT was associated with a characteristic prolonged [Ca(2+)](i) elevation (plateau) that resulted from Ca(2+) uptake by, and its subsequent release from, mitochondria. Measurements using a mitochondrial Ca(2+) indicator, mtPericam, showed that this process recruited mitochondria throughout the cell body, including the perinuclear region. [Ca(2+)](i) levels attained during the plateau phase were similar to or higher than those required for NFAT activation (200-300 nm). The elimination of the [Ca(2+)](i) plateau by blocking either mitochondrial Ca(2+) uptake via the uniporter or Ca(2+) release via the mitochondrial Na(+)/Ca(2+) exchanger strongly reduced nuclear import of NFAT. Furthermore, preventing Ca(2+) mobilization via the mitochondrial Na(+)/Ca(2+) exchanger diminished NFAT-mediated transcription. Collectively, these data implicate activity-induced Ca(2+) uptake and prolonged release from mitochondria as a novel regulatory mechanism in neuronal excitation-transcription coupling.
Collapse
|
42
|
Abstract
Calcium signals mediate diverse cellular functions in immunological cells. Early studies with mast cells, then a preeminent model for studying Ca2+-dependent exocytosis, revealed several basic features of calcium signaling in non-electrically excitable cells. Subsequent studies in these and other cells further defined the basic processes such as inositol 1,4,5-trisphosphate-mediated release of Ca2+ from Ca2+ stores in the endoplasmic reticulum (ER); coupling of ER store depletion to influx of external Ca2+ through a calcium-release activated calcium (CRAC) channel now attributed to the interaction of the ER Ca2+ sensor, stromal interacting molecule-1 (STIM1), with a unique Ca2+-channel protein, Orai1/CRACM1, and subsequent uptake of excess Ca2+ into ER and mitochondria through ATP-dependent Ca2+ pumps. In addition, transient receptor potential channels and ion exchangers also contribute to the generation of calcium signals that may be global or have dynamic (e.g., waves and oscillations) and spatial resolution for specific functional readouts. This review discusses past and recent developments in this field of research, the pharmacologic agents that have assisted in these endeavors, and the mast cell as an exemplar for sorting out how calcium signals may regulate multiple outputs in a single cell.
Collapse
Affiliation(s)
- Hong-Tao Ma
- Laboratory of Molecular Immunology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | | |
Collapse
|
43
|
Gauthier BR, Wiederkehr A, Baquié M, Dai C, Powers AC, Kerr-Conte J, Pattou F, MacDonald RJ, Ferrer J, Wollheim CB. PDX1 deficiency causes mitochondrial dysfunction and defective insulin secretion through TFAM suppression. Cell Metab 2009; 10:110-8. [PMID: 19656489 PMCID: PMC4012862 DOI: 10.1016/j.cmet.2009.07.002] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2009] [Revised: 06/22/2009] [Accepted: 07/01/2009] [Indexed: 01/23/2023]
Abstract
Mutations in the transcription factor Pdx1 cause maturity-onset diabetes of the young 4 (MODY4). Islet transduction with dominant-negative Pdx1 (RIPDN79PDX1) impairs mitochondrial metabolism and glucose-stimulated insulin secretion (GSIS). Transcript profiling revealed suppression of nuclear-encoded mitochondrial factor A (TFAM). Herein, we show that Pdx1 suppression in adult mice reduces islet TFAM expression coinciding with hyperglycemia. We define TFAM as a direct target of Pdx1 both in rat INS1 cells and human islets. Adenoviral overexpression of TFAM along with RIPDN79PDX1 in isolated rat islets rescued mitochondrial DNA (mtDNA) copy number and restored respiratory chain activity as well as glucose-induced ATP synthesis and insulin secretion. CGP37157, which blocks the mitochondrial Na(+)/Ca(2+) exchanger, restored ATP generation and GSIS in RIPDN79PDX1 islets, thereby bypassing the transcriptional defect. Thus, the genetic control by the beta cell-specific factor Pdx1 of the ubiquitous gene TFAM maintains beta cell mtDNA vital for ATP production and normal GSIS.
Collapse
Affiliation(s)
- Benoit R Gauthier
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Willems PHGM, Swarts HG, Hink MA, Koopman WJH. Chapter 16 The use of fluorescence correlation spectroscopy to probe mitochondrial mobility and intramatrix protein diffusion. Methods Enzymol 2009; 456:287-302. [PMID: 19348895 DOI: 10.1016/s0076-6879(08)04416-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Abstract
Within cells, functional changes in mitochondrial metabolic state are associated with alterations in organelle mobility, shape, and configuration of the mitochondrial matrix. Fluorescence correlation spectroscopy (FCS) is a technique that measures intensity fluctuations caused by single fluorescent molecules moving through a small detection volume. By mathematically correlating these fluctuations, information can be obtained about the concentration and rate of diffusion of the fluorescent molecules. Here we present an FCS-based approach for determining the mobility of enhanced yellow fluorescent protein (mitoEYFP) in the mitochondrial matrix of primary human skin fibroblasts. This protocol allows simultaneous quantification of intramatrix EYFP concentration and its diffusion constant, as well as the fraction of moving mitochondria and their velocity.
Collapse
Affiliation(s)
- Peter H G M Willems
- Department of Biochemistry, Nijmegen Centre for Molecular Life Sciences, Nijmegen, The Netherlands
| | | | | | | |
Collapse
|
45
|
de Mattia F, Gubser C, van Dommelen MMT, Visch HJ, Distelmaier F, Postigo A, Luyten T, Parys JB, de Smedt H, Smith GL, Willems PHGM, van Kuppeveld FJM. Human Golgi antiapoptotic protein modulates intracellular calcium fluxes. Mol Biol Cell 2009; 20:3638-45. [PMID: 19553469 DOI: 10.1091/mbc.e09-05-0385] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Golgi antiapoptotic protein (GAAP) is a novel regulator of cell death that is highly conserved in eukaryotes and present in some poxviruses, but its molecular mechanism is unknown. Given that alterations in intracellular Ca(2+) homeostasis play an important role in determining cell sensitivity to apoptosis, we investigated if GAAP affected Ca(2+) signaling. Overexpression of human (h)-GAAP suppressed staurosporine-induced, capacitative Ca(2+) influx from the extracellular space. In addition, it reduced histamine-induced Ca(2+) release from intracellular stores through inositol trisphosphate receptors. h-GAAP not only decreased the magnitude of the histamine-induced Ca(2+) fluxes from stores to cytosol and mitochondrial matrices, but it also reduced the induction and frequency of oscillatory changes in cytosolic Ca(2+). Overexpression of h-GAAP lowered the Ca(2+) content of the intracellular stores and decreased the efficacy of IP(3), providing possible explanations for the observed results. Opposite effects were obtained when h-GAAP was knocked down by siRNA. Thus, our data demonstrate that h-GAAP modulates intracellular Ca(2+) fluxes induced by both physiological and apoptotic stimuli.
Collapse
Affiliation(s)
- Fabrizio de Mattia
- Department of Medical Microbiology, Radboud University Nijmegen Medical Centre, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Vogtländer NPJ, Visch HJ, Bakker MAH, Berden JHM, van der Vlag J. Ligation of alpha-dystroglycan on podocytes induces intracellular signaling: a new mechanism for podocyte effacement? PLoS One 2009; 4:e5979. [PMID: 19543532 PMCID: PMC2695560 DOI: 10.1371/journal.pone.0005979] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2008] [Accepted: 05/15/2009] [Indexed: 12/20/2022] Open
Abstract
Background α-Dystroglycan is a negatively charged glycoprotein that covers the apical and basolateral membrane of the podocyte. Its transmembrane binding to the cytoskeleton is regulated via tyrosine phosphorylation (pY892) of β-dystroglycan. At the basolateral side α-dystroglycan binds the glomerular basement membrane. At the apical membrane, it plays a role in the maintenance of the filtration slit. In this study, we evaluated whether ligation of α-dystroglycan with specific antibodies or natural ligands induces intracellular signaling, and whether there is an effect on podocyte architecture. Methodology/Principal Findings Conditionally immortalized podocytes were exposed in vitro to antibodies to α-dystroglycan, and to fibronectin, biglycan, laminin and agrin. Intracellular calcium fluxes, phosphorylation of β-dystroglycan and podocyte architecture were studied. Antibodies to α-dystroglycan could specifically induce calcium signaling. Fibronectin also induced calcium signaling, and led to dephosphorylation of pY892 in β-dystroglycan. Ligation of α-dystroglycan resulted in an altered actin architecture, a decreased number of podocyte pedicles and a more flattened appearance of the podocyte. Conclusions/Significance We conclude that ligation of α-dystroglycan on podocytes induces intracellular calcium signaling, which leads to an altered cytoskeleton architecture akin to the situation of foot process effacement. In particular the ability of fibronectin to induce intracellular signaling events is of interest, since the expression and excretion of this protein is upregulated in several proteinuric diseases. Therefore, fibronectin-induced signaling via dystroglycan may be a novel mechanism for foot process effacement in proteinuric diseases.
Collapse
Affiliation(s)
- Nils P. J. Vogtländer
- Nephrology Research Laboratory, Nijmegen Centre for Molecular Life Sciences, Division of Nephrology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Henk Jan Visch
- Department of Biochemistry, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Marinka A. H. Bakker
- Nephrology Research Laboratory, Nijmegen Centre for Molecular Life Sciences, Division of Nephrology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Jo H. M. Berden
- Nephrology Research Laboratory, Nijmegen Centre for Molecular Life Sciences, Division of Nephrology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Johan van der Vlag
- Nephrology Research Laboratory, Nijmegen Centre for Molecular Life Sciences, Division of Nephrology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
- * E-mail:
| |
Collapse
|
47
|
Janssen RJ, Distelmaier F, Smeets R, Wijnhoven T, Ostergaard E, Jaspers NG, Raams A, Kemp S, Rodenburg RJ, Willems PH, van den Heuvel LP, Smeitink JA, Nijtmans LG. Contiguous gene deletion of ELOVL7, ERCC8 and NDUFAF2 in a patient with a fatal multisystem disorder. Hum Mol Genet 2009; 18:3365-74. [DOI: 10.1093/hmg/ddp276] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
48
|
Celsi F, Pizzo P, Brini M, Leo S, Fotino C, Pinton P, Rizzuto R. Mitochondria, calcium and cell death: a deadly triad in neurodegeneration. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2009; 1787:335-44. [PMID: 19268425 DOI: 10.1016/j.bbabio.2009.02.021] [Citation(s) in RCA: 223] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Revised: 02/23/2009] [Accepted: 02/24/2009] [Indexed: 12/17/2022]
Abstract
Mitochondrial Ca(2+) accumulation is a tightly controlled process, in turn regulating functions as diverse as aerobic metabolism and induction of cell death. The link between Ca(2+) (dys)regulation, mitochondria and cellular derangement is particularly evident in neurodegenerative disorders, in which genetic models and environmental factors allowed to identify common traits in the pathogenic routes. We will here summarize: i) the current view of mechanisms and functions of mitochondrial Ca(2+) homeostasis, ii) the basic principles of organelle Ca(2+) transport, iii) the role of Ca(2+) in neuronal cell death, and iv) the new information on the pathogenesis of Alzheimer's, Huntington's and Parkinson's diseases, highlighting the role of Ca(2+) and mitochondria.
Collapse
Affiliation(s)
- Fulvio Celsi
- Department of Experimental and Diagnostic Medicine, Interdisciplinary Center for the Study of Inflammation, Italy
| | | | | | | | | | | | | |
Collapse
|
49
|
Distelmaier F, Visch HJ, Smeitink JAM, Mayatepek E, Koopman WJH, Willems PHGM. The antioxidant Trolox restores mitochondrial membrane potential and Ca2+ -stimulated ATP production in human complex I deficiency. J Mol Med (Berl) 2009; 87:515-22. [PMID: 19255735 PMCID: PMC2836243 DOI: 10.1007/s00109-009-0452-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2008] [Revised: 01/22/2009] [Accepted: 02/10/2009] [Indexed: 11/29/2022]
Abstract
Malfunction of mitochondrial complex I caused by nuclear gene mutations causes early-onset neurodegenerative diseases. Previous work using cultured fibroblasts of complex-I-deficient patients revealed elevated levels of reactive oxygen species (ROS) and reductions in both total Ca(2+) content of the endoplasmic reticulum (ER(Ca)) and bradykinin(Bk)-induced increases in cytosolic and mitochondrial free Ca(2+) ([Ca(2+)](C); [Ca(2+)](M)) and ATP ([ATP](C); [ATP](M)) concentration. Here, we determined the mitochondrial membrane potential (Delta psi) in patient skin fibroblasts and show significant correlations with cellular ROS levels and ER(Ca), i.e., the less negative Delta psi, the higher these levels and the lower ER(Ca). Treatment with 6-hydroxy-2,5,7,8-tetramethylchromane-2-carboxylic acid (Trolox) normalized Delta psi and Bk-induced increases in [Ca(2+)](M) and [ATP](M). These effects were accompanied by an increase in ER(Ca) and Bk-induced increase in [Ca(2+)](C). Together, these results provide evidence for an integral role of increased ROS levels in complex I deficiency and point to the potential therapeutic value of antioxidant treatment.
Collapse
Affiliation(s)
- Felix Distelmaier
- Department of Biochemistry (286), Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | | | | | | | | | | |
Collapse
|
50
|
Griffiths EJ, Rutter GA. Mitochondrial calcium as a key regulator of mitochondrial ATP production in mammalian cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2009; 1787:1324-33. [PMID: 19366607 DOI: 10.1016/j.bbabio.2009.01.019] [Citation(s) in RCA: 272] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2008] [Revised: 01/23/2009] [Accepted: 01/27/2009] [Indexed: 12/30/2022]
Abstract
Mitochondrial Ca(2+) transport was initially considered important only in buffering of cytosolic Ca(2+) by acting as a "sink" under conditions of Ca(2+) overload. The main regulator of ATP production was considered to be the relative concentrations of high energy phosphates. However, work by Denton and McCormack in the 1970s and 1980s showed that free intramitochondrial Ca(2+) ([Ca(2+)](m)) activated dehydrogenase enzymes in mitochondria, leading to increased NADH and hence ATP production. This leads them to propose a scheme, subsequently termed a "parallel activation model" whereby increases in energy demand, such as hormonal stimulation or increased workload in muscle, produced an increase in cytosolic [Ca(2+)] that was relayed by the mitochondrial Ca(2+) transporters into the matrix to give an increase in [Ca(2+)](m). This then stimulated energy production to meet the increased energy demand. With the development of methods for measuring [Ca(2+)](m) in living cells that proved [Ca(2+)](m) changed over a dynamic physiological range rather than simply soaking up excess cytosolic [Ca(2+)], this model has now gained widespread acceptance. However, work by ourselves and others using targeted probes to measure changes in both [Ca(2+)] and [ATP] in different cell compartments has revealed variations in the interrelationships between these two in different tissues, suggesting that metabolic regulation by Ca(2+) is finely tuned to the demands and function of the individual organ.
Collapse
Affiliation(s)
- Elinor J Griffiths
- Department of Biochemistry, School of Medical Sciences, University of Bristol, Bristol BS8 1TD, UK.
| | | |
Collapse
|