1
|
Chanoine JP, Thompson DM, Lehman A. Diabetes Associated With Maternally Inherited Diabetes and Deafness (MIDD): From Pathogenic Variant to Phenotype. Diabetes 2025; 74:153-163. [PMID: 39556456 DOI: 10.2337/db24-0515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024]
Abstract
ARTICLE HIGHLIGHTS Maternally inherited diabetes and deafness (MIDD) is a mitochondrial disorder characterized primarily by hearing impairment and diabetes. m.3243A>G, the most common phenotypic variant, causes a complex rewiring of the cell with discontinuous remodeling of both mitochondrial and nuclear genome expressions. We propose that MIDD depends on a combination of insulin resistance and impaired β-cell function that occurs in the presence of high skeletal muscle heteroplasmy (approximately ≥60%) and more moderate cell heteroplasmy (∼25%-72%) for m.3243A>G. Understanding the complex mechanisms of MIDD is necessary to develop disease-specific management guidelines that are presently lacking.
Collapse
Affiliation(s)
- Jean-Pierre Chanoine
- Endocrinology and Diabetes Unit, Department of Pediatrics, BC Children's Hospital and The University of British Columbia, Vancouver, British Columbia, Canada
| | - David M Thompson
- Division of Endocrinology, Department of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Anna Lehman
- Department of Medical Genetics, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
2
|
Wen H, Deng H, Li B, Chen J, Zhu J, Zhang X, Yoshida S, Zhou Y. Mitochondrial diseases: from molecular mechanisms to therapeutic advances. Signal Transduct Target Ther 2025; 10:9. [PMID: 39788934 PMCID: PMC11724432 DOI: 10.1038/s41392-024-02044-3] [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: 07/02/2024] [Revised: 09/28/2024] [Accepted: 10/31/2024] [Indexed: 01/12/2025] Open
Abstract
Mitochondria are essential for cellular function and viability, serving as central hubs of metabolism and signaling. They possess various metabolic and quality control mechanisms crucial for maintaining normal cellular activities. Mitochondrial genetic disorders can arise from a wide range of mutations in either mitochondrial or nuclear DNA, which encode mitochondrial proteins or other contents. These genetic defects can lead to a breakdown of mitochondrial function and metabolism, such as the collapse of oxidative phosphorylation, one of the mitochondria's most critical functions. Mitochondrial diseases, a common group of genetic disorders, are characterized by significant phenotypic and genetic heterogeneity. Clinical symptoms can manifest in various systems and organs throughout the body, with differing degrees and forms of severity. The complexity of the relationship between mitochondria and mitochondrial diseases results in an inadequate understanding of the genotype-phenotype correlation of these diseases, historically making diagnosis and treatment challenging and often leading to unsatisfactory clinical outcomes. However, recent advancements in research and technology have significantly improved our understanding and management of these conditions. Clinical translations of mitochondria-related therapies are actively progressing. This review focuses on the physiological mechanisms of mitochondria, the pathogenesis of mitochondrial diseases, and potential diagnostic and therapeutic applications. Additionally, this review discusses future perspectives on mitochondrial genetic diseases.
Collapse
Affiliation(s)
- Haipeng Wen
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China
| | - Hui Deng
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Bingyan Li
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Junyu Chen
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Junye Zhu
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Xian Zhang
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Shigeo Yoshida
- Department of Ophthalmology, Kurume University School of Medicine, Kurume, Fukuoka, 830-0011, Japan
| | - Yedi Zhou
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China.
| |
Collapse
|
3
|
Kompatscher M, Gonnella I, Erlacher M. Studying the Function of tRNA Modifications: Experimental Challenges and Opportunities. J Mol Biol 2025:168934. [PMID: 39756793 DOI: 10.1016/j.jmb.2024.168934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/19/2024] [Accepted: 12/31/2024] [Indexed: 01/07/2025]
Abstract
tRNAs are essential molecules in protein synthesis, responsible for translating the four-nucleotide genetic code into the corresponding amino acid sequence. RNA modifications play a crucial role in influencing tRNA folding, structure, and function. These modifications, ranging from simple methylations to complex hypermodified species, are distributed throughout the tRNA molecule. Depending on their type and position, they contribute to the accuracy and efficiency of decoding by participating in a complex network of interactions. The enzymatic processes introducing these modifications are equally intricate and diverse, adding further complexity. As a result, studying tRNA modifications faces limitations at multiple levels. This review addresses the challenges involved in manipulating and studying the function of tRNA modifications and discusses experimental strategies and possibilities to overcome these obstacles.
Collapse
Affiliation(s)
- Maria Kompatscher
- Institute of Genomics and RNomics, Medical University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Isabell Gonnella
- Institute of Genomics and RNomics, Medical University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Matthias Erlacher
- Institute of Genomics and RNomics, Medical University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria.
| |
Collapse
|
4
|
Rackham O, Saurer M, Ban N, Filipovska A. Unique architectural features of mammalian mitochondrial protein synthesis. Trends Cell Biol 2025; 35:11-23. [PMID: 38853081 DOI: 10.1016/j.tcb.2024.05.001] [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: 02/12/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 06/11/2024]
Abstract
Mitochondria rely on coordinated expression of their own mitochondrial DNA (mtDNA) with that of the nuclear genome for their biogenesis. The bacterial ancestry of mitochondria has given rise to unique and idiosyncratic features of the mtDNA and its expression machinery that can be specific to different organisms. In animals, the mitochondrial protein synthesis machinery has acquired many new components and mechanisms over evolution. These include several new ribosomal proteins, new stop codons and ways to recognise them, and new mechanisms to deliver nascent proteins into the mitochondrial inner membrane. Here we describe the mitochondrial protein synthesis machinery in mammals and its unique mechanisms of action elucidated to date and highlight the technologies poised to reveal the next generation of discoveries in mitochondrial translation.
Collapse
Affiliation(s)
- Oliver Rackham
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia; ARC Centre of Excellence in Synthetic Biology, QEII Medical Centre, Nedlands, WA, Australia; Curtin Medical School Curtin University, Bentley, WA, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia; Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, Nedlands, WA, Australia
| | - Martin Saurer
- Institute of Molecular Biology and Biophysics, Department of Biology, ETH Zürich, 8093 Zürich, Switzerland
| | - Nenad Ban
- Institute of Molecular Biology and Biophysics, Department of Biology, ETH Zürich, 8093 Zürich, Switzerland
| | - Aleksandra Filipovska
- ARC Centre of Excellence in Synthetic Biology, QEII Medical Centre, Nedlands, WA, Australia; Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, Nedlands, WA, Australia; The University of Western Australia Centre for Child Health Research, Northern Entrance, Perth Children's Hospital, Nedlands, WA, Australia.
| |
Collapse
|
5
|
Cabané P, Correa C, Bode I, Aguilar R, Elorza AA. Biomarkers in Thyroid Cancer: Emerging Opportunities from Non-Coding RNAs and Mitochondrial Space. Int J Mol Sci 2024; 25:6719. [PMID: 38928426 PMCID: PMC11204084 DOI: 10.3390/ijms25126719] [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: 04/20/2024] [Revised: 06/01/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Thyroid cancer diagnosis primarily relies on imaging techniques and cytological analyses. In cases where the diagnosis is uncertain, the quantification of molecular markers has been incorporated after cytological examination. This approach helps physicians to make surgical decisions, estimate cancer aggressiveness, and monitor the response to treatments. Despite the availability of commercial molecular tests, their widespread use has been hindered in our experience due to cost constraints and variability between them. Thus, numerous groups are currently evaluating new molecular markers that ultimately will lead to improved diagnostic certainty, as well as better classification of prognosis and recurrence. In this review, we start reviewing the current preoperative testing methodologies, followed by a comprehensive review of emerging molecular markers. We focus on micro RNAs, long non-coding RNAs, and mitochondrial (mt) signatures, including mtDNA genes and circulating cell-free mtDNA. We envision that a robust set of molecular markers will complement the national and international clinical guides for proper assessment of the disease.
Collapse
Affiliation(s)
- Patricio Cabané
- Department of Head and Neck Surgery, Clinica INDISA, Santiago 7520440, Chile; (P.C.); (C.C.)
- Faculty of Medicine, Universidad Andres Bello, Santiago 8370071, Chile
- Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Claudio Correa
- Department of Head and Neck Surgery, Clinica INDISA, Santiago 7520440, Chile; (P.C.); (C.C.)
- Faculty of Medicine, Universidad Andres Bello, Santiago 8370071, Chile
| | - Ignacio Bode
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370071, Chile;
| | - Rodrigo Aguilar
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370071, Chile;
| | - Alvaro A. Elorza
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370071, Chile;
| |
Collapse
|
6
|
Ambrogetti R, Kavanagh E, ElTayeb K. Late-onset mitochondrial encephalopathy with lactic acidosis and stroke-like episodes and the role of serial imaging. BMJ Case Rep 2024; 17:e259102. [PMID: 38417938 PMCID: PMC10900402 DOI: 10.1136/bcr-2023-259102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2024] Open
Abstract
Mitochondria are essential for human metabolic function. Over 350 genetic mutations are associated with mitochondrial diseases, which are inherited in a matrilineal fashion. In mitochondrial encephalomyopathy, lactic acidosis and stroke-like episodes (MELAS), defective mitochondrial function and resultant impaired cellular energy production compromise vascular perfusion in affected tissues. Early diagnostic criteria suggested the diagnosis should be considered in those under 40. However, a broader range of phenotypes are now recognised, including those that present for the first time later in life. The primary presenting feature in MELAS is a stroke-like episode invariably resulting in patients undergoing neuroradiological imaging. We present a case of a woman with a first presentation of a stroke-like episode and seizures in her 40s who was eventually diagnosed with MELAS. We detail her clinical presentation, treatment and diagnosis, emphasising the role of serial imaging in her diagnosis.
Collapse
Affiliation(s)
- Robert Ambrogetti
- Internal Medicine, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Ethan Kavanagh
- University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Khalid ElTayeb
- Neurology, University Hospitals of Leicester NHS Trust, Leicester, UK
| |
Collapse
|
7
|
Liu L, Yang J, Otani Y, Shiga T, Yamaguchi A, Oda Y, Hattori M, Goto T, Ishibashi S, Kawashima-Sonoyama Y, Ishihara T, Matsuzaki Y, Akamatsu W, Fujitani M, Taketani T. MELAS-Derived Neurons Functionally Improve by Mitochondrial Transfer from Highly Purified Mesenchymal Stem Cells (REC). Int J Mol Sci 2023; 24:17186. [PMID: 38139018 PMCID: PMC10742994 DOI: 10.3390/ijms242417186] [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: 11/17/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episode (MELAS) syndrome, caused by a single base substitution in mitochondrial DNA (m.3243A>G), is one of the most common maternally inherited mitochondrial diseases accompanied by neuronal damage due to defects in the oxidative phosphorylation system. There is no established treatment. Our previous study reported a superior restoration of mitochondrial function and bioenergetics in mitochondria-deficient cells using highly purified mesenchymal stem cells (RECs). However, whether such exogenous mitochondrial donation occurs in mitochondrial disease models and whether it plays a role in the recovery of pathological neuronal functions is unknown. Here, utilizing induced pluripotent stem cells (iPSC), we differentiated neurons with impaired mitochondrial function from patients with MELAS. MELAS neurons and RECs/mesenchymal stem cells (MSCs) were cultured under contact or non-contact conditions. Both RECs and MSCs can donate mitochondria to MELAS neurons, but RECs are more excellent than MSCs for mitochondrial transfer in both systems. In addition, REC-mediated mitochondrial transfer significantly restored mitochondrial function, including mitochondrial membrane potential, ATP/ROS production, intracellular calcium storage, and oxygen consumption rate. Moreover, mitochondrial function was maintained for at least three weeks. Thus, REC-donated exogenous mitochondria might offer a potential therapeutic strategy for treating neurological dysfunction in MELAS.
Collapse
Affiliation(s)
- Lu Liu
- Department of Pediatrics, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo 693-8501, Japan; (L.L.); (J.Y.); (Y.O.); (M.H.); (T.G.); (Y.K.-S.)
| | - Jiahao Yang
- Department of Pediatrics, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo 693-8501, Japan; (L.L.); (J.Y.); (Y.O.); (M.H.); (T.G.); (Y.K.-S.)
| | - Yoshinori Otani
- Department of Anatomy and Neuroscience, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo 693-8501, Japan; (Y.O.); (M.F.)
| | - Takahiro Shiga
- Center for Genomic and Regenerative Medicine, School of Medicine, Juntendo University, Tokyo 113-8421, Japan; (T.S.); (A.Y.); (W.A.)
| | - Akihiro Yamaguchi
- Center for Genomic and Regenerative Medicine, School of Medicine, Juntendo University, Tokyo 113-8421, Japan; (T.S.); (A.Y.); (W.A.)
| | - Yasuaki Oda
- Department of Pediatrics, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo 693-8501, Japan; (L.L.); (J.Y.); (Y.O.); (M.H.); (T.G.); (Y.K.-S.)
| | - Miho Hattori
- Department of Pediatrics, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo 693-8501, Japan; (L.L.); (J.Y.); (Y.O.); (M.H.); (T.G.); (Y.K.-S.)
| | - Tsukimi Goto
- Department of Pediatrics, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo 693-8501, Japan; (L.L.); (J.Y.); (Y.O.); (M.H.); (T.G.); (Y.K.-S.)
- Clinical Laboratory Division, Shimane University Hospital, 89-1 Enya-cho, Izumo 693-8501, Japan
| | - Shuichi Ishibashi
- Department of Digestive and General Surgery, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo 693-8501, Japan;
| | - Yuki Kawashima-Sonoyama
- Department of Pediatrics, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo 693-8501, Japan; (L.L.); (J.Y.); (Y.O.); (M.H.); (T.G.); (Y.K.-S.)
| | - Takaya Ishihara
- Department of Life Science, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo 693-8501, Japan; (T.I.); (Y.M.)
| | - Yumi Matsuzaki
- Department of Life Science, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo 693-8501, Japan; (T.I.); (Y.M.)
| | - Wado Akamatsu
- Center for Genomic and Regenerative Medicine, School of Medicine, Juntendo University, Tokyo 113-8421, Japan; (T.S.); (A.Y.); (W.A.)
| | - Masashi Fujitani
- Department of Anatomy and Neuroscience, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo 693-8501, Japan; (Y.O.); (M.F.)
| | - Takeshi Taketani
- Department of Pediatrics, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo 693-8501, Japan; (L.L.); (J.Y.); (Y.O.); (M.H.); (T.G.); (Y.K.-S.)
| |
Collapse
|
8
|
Liufu T, Yu H, Yu J, Yu M, Tian Y, Ou Y, Deng J, Xing G, Wang Z. Complex I deficiency in m.3243A>G fibroblasts is alleviated by reducing NADH accumulation. Front Physiol 2023; 14:1164287. [PMID: 37650111 PMCID: PMC10464909 DOI: 10.3389/fphys.2023.1164287] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 08/03/2023] [Indexed: 09/01/2023] Open
Abstract
Introduction: Mitochondrial disease is a spectrum of debilitating disorders caused by mutations in the mitochondrial DNA (mtDNA) or nuclear DNA that compromises the respiratory chain. Mitochondrial 3243A>G (m.3243 A>G) is the most common mutation showing great heterogeneity in phenotype. Previous studies have indicated that NADH: ubiquinone oxidoreductase (complex I) deficiency accompanied by a decreased nicotinamide adenine dinucleotide (NAD+)/reduced NAD+ (NADH) ratio may play a pivotal role in the pathogenesis of m.3243A>G mutation. Methods: To evaluate the potential effects of strategies targeting the imbalanced NAD+/NADH ratio in m.3243A>G mutation, we treated fibroblasts derived from patients with the m.3243 A>G mutation using nicotinamide riboside (NR) or mitochondria-targeted H2O-forming NADH oxidase (mitoLbNOX). Results: M.3243 A>G fibroblasts showed a significant reduction in complex I core subunit 6, complex I enzymatic activity, complex I-dependent oxygen consumption rate (OCR), and adenosine triphosphate (ATP) production compared to the controls. The NAD+/NADH ratio was also significantly reduced in m.3243 A>G fibroblasts, and, using fluorescence lifetime imaging microscopy, we also found that the NADH level was elevated in m.3243 A>G fibroblasts. After NR treatment, the NAD+/NADH ratio, complex I-dependent OCR, and ATP levels increased, whereas NADH levels remained unchanged. More excitingly, after treatment with mitoLbNOX, the NAD+/NADH ratio, complex I-independent OCR, and ATP levels increased more pronouncedly compared with the NR treatment group, accompanied by significantly reduced NADH levels. Discussion: The present study suggests that compared with repletion of NAD+ alone, the combination of this therapeutic modality with alleviation of NADH overload may amplify the treatment effect of restoring NAD+/NADH balance in m.3243A>G fibroblasts.
Collapse
Affiliation(s)
- Tongling Liufu
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Haiyan Yu
- Department of Clinical Laboratory, Peking University First Hospital, Beijing, China
| | - Jiaxi Yu
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Meng Yu
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Yue Tian
- Neuroscience Research Institute, Peking University, Beijing, China
| | - Yichun Ou
- Neuroscience Research Institute, Peking University, Beijing, China
| | - Jianwen Deng
- Department of Neurology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Neurovascular Disease Discovery, Beijing, China
| | - Guogang Xing
- Neuroscience Research Institute, Peking University, Beijing, China
| | - Zhaoxia Wang
- Department of Neurology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Neurovascular Disease Discovery, Beijing, China
| |
Collapse
|
9
|
Tomoda E, Nagao A, Shirai Y, Asano K, Suzuki T, Battersby B, Suzuki T. Restoration of mitochondrial function through activation of hypomodified tRNAs with pathogenic mutations associated with mitochondrial diseases. Nucleic Acids Res 2023; 51:7563-7579. [PMID: 36928678 PMCID: PMC10415153 DOI: 10.1093/nar/gkad139] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/14/2023] [Accepted: 03/08/2023] [Indexed: 03/18/2023] Open
Abstract
Mutations in mitochondrial (mt-)tRNAs frequently cause mitochondrial dysfunction. Mitochondrial myopathy, encephalopathy, lactic acidosis, and stroke-like episodes (MELAS), and myoclonus epilepsy associated with ragged red fibers (MERRF) are major clinical subgroups of mitochondrial diseases caused by pathogenic point mutations in tRNA genes encoded in mtDNA. We previously reported a severe reduction in the frequency of 5-taurinomethyluridine (τm5U) and its 2-thiouridine derivative (τm5s2U) in the anticodons of mutant mt-tRNAs isolated from the cells of patients with MELAS and MERRF, respectively. The hypomodified tRNAs fail to decode cognate codons efficiently, resulting in defective translation of respiratory chain proteins in mitochondria. To restore the mitochondrial activity of MELAS patient cells, we overexpressed MTO1, a τm5U-modifying enzyme, in patient-derived myoblasts. We used a newly developed primer extension method and showed that MTO1 overexpression almost completely restored the τm5U modification of the MELAS mutant mt-tRNALeu(UUR). An increase in mitochondrial protein synthesis and oxygen consumption rate suggested that the mitochondrial function of MELAS patient cells can be activated by restoring the τm5U of the mutant tRNA. In addition, we confirmed that MTO1 expression restored the τm5s2U of the mutant mt-tRNALys in MERRF patient cells. These findings pave the way for epitranscriptomic therapies for mitochondrial diseases.
Collapse
Affiliation(s)
- Ena Tomoda
- Department of Chemistry and Biotechnology, Graduate School of Engineering, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Asuteka Nagao
- Department of Chemistry and Biotechnology, Graduate School of Engineering, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Yuki Shirai
- Department of Chemistry and Biotechnology, Graduate School of Engineering, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Kana Asano
- Department of Chemistry and Biotechnology, Graduate School of Engineering, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Takeo Suzuki
- Department of Chemistry and Biotechnology, Graduate School of Engineering, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | | | - Tsutomu Suzuki
- Department of Chemistry and Biotechnology, Graduate School of Engineering, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
10
|
Ueda S, Yagi M, Tomoda E, Matsumoto S, Ueyanagi Y, Do Y, Setoyama D, Matsushima Y, Nagao A, Suzuki T, Ide T, Mori Y, Oyama N, Kang D, Uchiumi T. Mitochondrial haplotype mutation alleviates respiratory defect of MELAS by restoring taurine modification in tRNA with 3243A > G mutation. Nucleic Acids Res 2023; 51:7480-7495. [PMID: 37439353 PMCID: PMC10415116 DOI: 10.1093/nar/gkad591] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 06/22/2023] [Accepted: 06/28/2023] [Indexed: 07/14/2023] Open
Abstract
The 3243A > G in mtDNA is a representative mutation in mitochondrial diseases. Mitochondrial protein synthesis is impaired due to decoding disorder caused by severe reduction of 5-taurinomethyluridine (τm5U) modification of the mutant mt-tRNALeu(UUR) bearing 3243A > G mutation. The 3243A > G heteroplasmy in peripheral blood reportedly decreases exponentially with age. Here, we found three cases with mild respiratory symptoms despite bearing high rate of 3243A > G mutation (>90%) in blood mtDNA. These patients had the 3290T > C haplotypic mutation in addition to 3243A > G pathogenic mutation in mt-tRNALeu(UUR) gene. We generated cybrid cells of these cases to examine the effects of the 3290T > C mutation on mitochondrial function and found that 3290T > C mutation improved mitochondrial translation, formation of respiratory chain complex, and oxygen consumption rate of pathogenic cells associated with 3243A > G mutation. We measured τm5U frequency of mt-tRNALeu(UUR) with 3243A > G mutation in the cybrids by a primer extension method assisted with chemical derivatization of τm5U, showing that hypomodification of τm5U was significantly restored by the 3290T > C haplotypic mutation. We concluded that the 3290T > C is a haplotypic mutation that suppresses respiratory deficiency of mitochondrial disease by restoring hypomodified τm5U in mt-tRNALeu(UUR) with 3243A > G mutation, implying a potential therapeutic measure for mitochondrial disease associated with pathogenic mutations in mt-tRNAs.
Collapse
Affiliation(s)
- Saori Ueda
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Mikako Yagi
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
- Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Ena Tomoda
- Department of Chemistry and Biotechnology, Graduate School of Engineering, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Shinya Matsumoto
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yasushi Ueyanagi
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yura Do
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Daiki Setoyama
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yuichi Matsushima
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
- Department of Biological Sciences, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Japan
| | - Asuteka Nagao
- Department of Chemistry and Biotechnology, Graduate School of Engineering, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Tsutomu Suzuki
- Department of Chemistry and Biotechnology, Graduate School of Engineering, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Tomomi Ide
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yusuke Mori
- Department of Internal Medicine Kitakyushu City Yahata Hospital, 2-6-2 Ogura, Yahatahigashi-ku, Kitakyushu 805-8534, Japan
| | - Noriko Oyama
- Department of Endocrinology and Metabolism, Fukuoka Children's Hospital, 5-1-1 Kashiiteriha, Higashi-ku, Fukuoka 813-0017, Japan
| | - Dongchon Kang
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Takeshi Uchiumi
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
- Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
11
|
Mullin NK, Voigt AP, Flamme-Wiese MJ, Liu X, Riker MJ, Varzavand K, Stone EM, Tucker BA, Mullins RF. Multimodal single-cell analysis of nonrandom heteroplasmy distribution in human retinal mitochondrial disease. JCI Insight 2023; 8:e165937. [PMID: 37289546 PMCID: PMC10443808 DOI: 10.1172/jci.insight.165937] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 06/02/2023] [Indexed: 06/10/2023] Open
Abstract
Variants within the high copy number mitochondrial genome (mtDNA) can disrupt organelle function and lead to severe multisystem disease. The wide range of manifestations observed in patients with mitochondrial disease results from varying fractions of abnormal mtDNA molecules in different cells and tissues, a phenomenon termed heteroplasmy. However, the landscape of heteroplasmy across cell types within tissues and its influence on phenotype expression in affected patients remains largely unexplored. Here, we identify nonrandom distribution of a pathogenic mtDNA variant across a complex tissue using single-cell RNA-Seq, mitochondrial single-cell ATAC sequencing, and multimodal single-cell sequencing. We profiled the transcriptome, chromatin accessibility state, and heteroplasmy in cells from the eyes of a patient with mitochondrial encephalopathy, lactic acidosis, and stroke-like episodes (MELAS) and from healthy control donors. Utilizing the retina as a model for complex multilineage tissues, we found that the proportion of the pathogenic m.3243A>G allele was neither evenly nor randomly distributed across diverse cell types. All neuroectoderm-derived neural cells exhibited a high percentage of the mutant variant. However, a subset of mesoderm-derived lineage, namely the vasculature of the choroid, was near homoplasmic for the WT allele. Gene expression and chromatin accessibility profiles of cell types with high and low proportions of m.3243A>G implicate mTOR signaling in the cellular response to heteroplasmy. We further found by multimodal single-cell sequencing of retinal pigment epithelial cells that a high proportion of the pathogenic mtDNA variant was associated with transcriptionally and morphologically abnormal cells. Together, these findings show the nonrandom nature of mitochondrial variant partitioning in human mitochondrial disease and underscore its implications for mitochondrial disease pathogenesis and treatment.
Collapse
Affiliation(s)
- Nathaniel K. Mullin
- University of Iowa Institute for Vision Research, Iowa City, Iowa, USA
- Department of Ophthalmology and Visual Sciences and
- Medical Scientist Training Program, University of Iowa, Iowa City, Iowa, USA
| | - Andrew P. Voigt
- University of Iowa Institute for Vision Research, Iowa City, Iowa, USA
- Department of Ophthalmology and Visual Sciences and
- Medical Scientist Training Program, University of Iowa, Iowa City, Iowa, USA
| | - Miles J. Flamme-Wiese
- University of Iowa Institute for Vision Research, Iowa City, Iowa, USA
- Department of Ophthalmology and Visual Sciences and
| | - Xiuying Liu
- University of Iowa Institute for Vision Research, Iowa City, Iowa, USA
- Department of Ophthalmology and Visual Sciences and
| | - Megan J. Riker
- University of Iowa Institute for Vision Research, Iowa City, Iowa, USA
- Department of Ophthalmology and Visual Sciences and
| | - Katayoun Varzavand
- University of Iowa Institute for Vision Research, Iowa City, Iowa, USA
- Department of Ophthalmology and Visual Sciences and
| | - Edwin M. Stone
- University of Iowa Institute for Vision Research, Iowa City, Iowa, USA
- Department of Ophthalmology and Visual Sciences and
| | - Budd A. Tucker
- University of Iowa Institute for Vision Research, Iowa City, Iowa, USA
- Department of Ophthalmology and Visual Sciences and
| | - Robert F. Mullins
- University of Iowa Institute for Vision Research, Iowa City, Iowa, USA
- Department of Ophthalmology and Visual Sciences and
| |
Collapse
|
12
|
Jia YZ, Liu J, Wang GQ, Pan H, Huang TZ, Liu R, Zhang Y. HIG1 domain family member 1A is a crucial regulator of disorders associated with hypoxia. Mitochondrion 2023; 69:171-182. [PMID: 36804467 DOI: 10.1016/j.mito.2023.02.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 02/06/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023]
Abstract
Mitochondria play a central role in cellular energy conversion, metabolism, and cell proliferation. The regulation of mitochondrial function by HIGD1A, which is located on the inner membrane of the mitochondria, is essential to maintain cell survival under hypoxic conditions. In recent years, there have been shown other cellular pathways and mechanisms involving HIGD1A diametrically or through its interaction. As a novel regulator, HIGD1A maintains mitochondrial integrity and enhances cell viability under hypoxic conditions, increasing cell resistance to hypoxia. HIGD1A mainly targets cytochrome c oxidase by regulating downstream signaling pathways, which affects the ATP generation system and subsequently alters mitochondrial respiratory function. In addition, HIGD1A plays a dual role in cell survival in distinct degree hypoxia regions of the tumor. Under mild and moderate anoxic areas, HIGD1A acts as a positive regulator to promote cell growth. However, HIGD1A plays a role in inhibiting cell growth but retaining cellular activity under severe anoxic areas. We speculate that HIGD1A engages in tumor recurrence and drug resistance mechanisms. This review will focus on data concerning how HIGD1A regulates cell viability under hypoxic conditions. Therefore, HIGD1A could be a potential therapeutic target for hypoxia-related diseases.
Collapse
Affiliation(s)
- Yin-Zhao Jia
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jing Liu
- Key Laboratory of Coal Science and Technology of Ministry of Education, Taiyuan University of Technology, Taiyuan 030024, Shanxi, China
| | - Geng-Qiao Wang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hao Pan
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tie-Zeng Huang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ran Liu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yong Zhang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
13
|
Ren B, Guan MX, Zhou T, Cai X, Shan G. Emerging functions of mitochondria-encoded noncoding RNAs. Trends Genet 2023; 39:125-139. [PMID: 36137834 DOI: 10.1016/j.tig.2022.08.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/03/2022] [Accepted: 08/11/2022] [Indexed: 01/27/2023]
Abstract
Mitochondria, organelles that harbor their own circular genomes, are critical for energy production and homeostasis maintenance in eukaryotic cells. Recent studies discovered hundreds of mitochondria-encoded noncoding RNAs (mt-ncRNAs), including novel subtypes of mitochondria-encoded circular RNAs (mecciRNAs) and mitochondria-encoded double-stranded RNAs (mt-dsRNAs). Here, we discuss the emerging field of mt-ncRNAs by reviewing their expression patterns, biogenesis, metabolism, regulatory roles, and functional mechanisms. Many mt-ncRNAs have regulatory roles in cellular physiology, and some are associated with, or even act as, causal factors in human diseases. We also highlight developments in technologies and methodologies and further insights into future perspectives and challenges in studying these noncoding RNAs, as well as their potential biomedical applications.
Collapse
Affiliation(s)
- Bingbing Ren
- Department of Pulmonary and Critical Care Medicine, Regional Medical Center for National Institute of Respiratory Disease, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China; Cancer Center, Zhejiang University, Hangzhou 310058, China; Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Min-Xin Guan
- Division of Medical Genetics and Genomics, The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, China; Zhejiang Provincial Key Lab of Genetic and Developmental Disorder, Institute of Genetics, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Tianhua Zhou
- Cancer Center, Zhejiang University, Hangzhou 310058, China; Department of Cell Biology and Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China; Institute of Gastroenterology, Zhejiang University, Hangzhou 310016, China
| | - Xiujun Cai
- Cancer Center, Zhejiang University, Hangzhou 310058, China; Zhejiang Provincial Key Laboratory of Laparoscopic Technology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China; Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China; Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang University, Hangzhou 310016, China; Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Zhejiang University, Hangzhou 310016, China
| | - Ge Shan
- Department of Pulmonary and Critical Care Medicine, Regional Medical Center for National Institute of Respiratory Disease, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China; Cancer Center, Zhejiang University, Hangzhou 310058, China; Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China; Department of Clinical Laboratory, The First Affiliated Hospital of USTC, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230027, China.
| |
Collapse
|
14
|
Lin DS, Huang YW, Ho CS, Huang TS, Lee TH, Wu TY, Huang ZD, Wang TJ. Impact of Mitochondrial A3243G Heteroplasmy on Mitochondrial Bioenergetics and Dynamics of Directly Reprogrammed MELAS Neurons. Cells 2022; 12:cells12010015. [PMID: 36611807 PMCID: PMC9818214 DOI: 10.3390/cells12010015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/17/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
The MELAS syndrome primarily affecting the CNS is mainly caused by the m.A3243G mutation. The heteroplasmy in different tissues affects the phenotypic spectrum, yet the impact of various levels of m.A3243G heteroplasmy on CNS remains elusive due to the lack of a proper neuronal model harboring m.A3243G mutation. We generated induced neurons (iNs) through the direct reprogramming of MELAS patients, with derived fibroblasts harboring high (>95%), intermediate (68%), and low (20%) m.A3243G mutation. iNs demonstrated neuronal morphology with neurite outgrowth, branching, and dendritic spines. The heteroplasmy and deficiency of respiratory chain complexes were retained in MELAS iNs. High heteroplasmy elicited the elevation in ROS levels and the disruption of mitochondrial membrane potential. Furthermore, high and intermediate heteroplasmy led to the impairment of mitochondrial bioenergetics and a change in mitochondrial dynamics toward the fission and fragmentation of mitochondria, with a reduction in mitochondrial networks. Moreover, iNs derived from aged individuals manifested with mitochondrial fission. These results help us in understanding the impact of various heteroplasmic levels on mitochondrial bioenergetics and mitochondrial dynamics in neurons as the underlying pathomechanism of neurological manifestations of MELAS syndrome. Furthermore, these findings provide targets for further pharmacological approaches of mitochondrial diseases and validate iNs as a reliable platform for studies in neuronal aspects of aging, neurodegenerative disorders, and mitochondrial diseases.
Collapse
Affiliation(s)
- Dar-Shong Lin
- Department of Pediatrics, Mackay Memorial Hospital, Taipei 10449, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei 25245, Taiwan
- Correspondence: ; Tel.: +886-2-2809-4661; Fax: +886-2-2809-4679
| | - Yu-Wen Huang
- Department of Medical Research, Mackay Memorial Hospital, Taipei 10449, Taiwan
| | - Che-Sheng Ho
- Department of Medicine, Mackay Medical College, New Taipei 25245, Taiwan
- Department of Neurology, Mackay Children’s Hospital, Taipei 10449, Taiwan
| | - Tung-Sun Huang
- Department of Surgery, Mackay Memorial Hospital, Taipei 10449, Taiwan
| | - Tsung-Han Lee
- Department of Medical Research, Mackay Memorial Hospital, Taipei 10449, Taiwan
| | - Tsu-Yen Wu
- Department of Medical Research, Mackay Memorial Hospital, Taipei 10449, Taiwan
| | - Zon-Darr Huang
- Department of Medical Research, Mackay Memorial Hospital, Taipei 10449, Taiwan
| | - Tuan-Jen Wang
- Department of Laboratory Medicine, Mackay Memorial Hospital, Taipei 10449, Taiwan
| |
Collapse
|
15
|
Yu T, Zhang Y, Zheng WQ, Wu S, Li G, Zhang Y, Li N, Yao R, Fang P, Wang J, Zhou XL. Selective degradation of tRNASer(AGY) is the primary driver for mitochondrial seryl-tRNA synthetase-related disease. Nucleic Acids Res 2022; 50:11755-11774. [PMID: 36350636 PMCID: PMC9723649 DOI: 10.1093/nar/gkac1028] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/13/2022] [Accepted: 10/21/2022] [Indexed: 11/11/2022] Open
Abstract
Mitochondrial translation is of high significance for cellular energy homeostasis. Aminoacyl-tRNA synthetases (aaRSs) are crucial translational components. Mitochondrial aaRS variants cause various human diseases. However, the pathogenesis of the vast majority of these diseases remains unknown. Here, we identified two novel SARS2 (encoding mitochondrial seryl-tRNA synthetase) variants that cause a multisystem disorder. c.654-14T > A mutation induced mRNA mis-splicing, generating a peptide insertion in the active site; c.1519dupC swapped a critical tRNA-binding motif in the C-terminus due to stop codon readthrough. Both mutants exhibited severely diminished tRNA binding and aminoacylation capacities. A marked reduction in mitochondrial tRNASer(AGY) was observed due to RNA degradation in patient-derived induced pluripotent stem cells (iPSCs), causing impaired translation and comprehensive mitochondrial function deficiencies. These impairments were efficiently rescued by wild-type SARS2 overexpression. Either mutation caused early embryonic fatality in mice. Heterozygous mice displayed reduced muscle tissue-specific levels of tRNASers. Our findings elucidated the biochemical and cellular consequences of impaired translation mediated by SARS2, suggesting that reduced abundance of tRNASer(AGY) is a key determinant for development of SARS2-related diseases.
Collapse
Affiliation(s)
| | | | - Wen-Qiang Zheng
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China
| | - Siqi Wu
- State Key Laboratory of Bioorganic and Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
| | - Guoqiang Li
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Key Laboratory of Clinical Molecular Diagnostics for Pediatrics, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
| | - Yong Zhang
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China
| | - Niu Li
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Key Laboratory of Clinical Molecular Diagnostics for Pediatrics, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
| | - Ruen Yao
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Key Laboratory of Clinical Molecular Diagnostics for Pediatrics, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
| | - Pengfei Fang
- State Key Laboratory of Bioorganic and Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
| | - Jian Wang
- Correspondence may also be addressed to Jian Wang. Tel: +86 21 3808 7371;
| | - Xiao-Long Zhou
- To whom correspondence should be addressed. Tel: +86 21 5492 1247; Fax: +86 21 5492 1011;
| |
Collapse
|
16
|
The Bacterial ClpXP-ClpB Family Is Enriched with RNA-Binding Protein Complexes. Cells 2022; 11:cells11152370. [PMID: 35954215 PMCID: PMC9368063 DOI: 10.3390/cells11152370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/23/2022] [Accepted: 07/28/2022] [Indexed: 11/17/2022] Open
Abstract
In the matrix of bacteria/mitochondria/chloroplasts, Lon acts as the degradation machine for soluble proteins. In stress periods, however, proteostasis and survival depend on the strongly conserved Clp/Hsp100 family. Currently, the targets of ATP-powered unfoldases/disaggregases ClpB and ClpX and of peptidase ClpP heptameric rings are still unclear. Trapping experiments and proteome profiling in multiple organisms triggered confusion, so we analyzed the consistency of ClpP-trap targets in bacteria. We also provide meta-analyses of protein interactions in humans, to elucidate where Clp family members are enriched. Furthermore, meta-analyses of mouse complexomics are provided. Genotype–phenotype correlations confirmed our concept. Trapping, proteome, and complexome data retrieved consistent coaccumulation of CLPXP with GFM1 and TUFM orthologs. CLPX shows broad interaction selectivity encompassing mitochondrial translation elongation, RNA granules, and nucleoids. CLPB preferentially attaches to mitochondrial RNA granules and translation initiation components; CLPP is enriched with them all and associates with release/recycling factors. Mutations in CLPP cause Perrault syndrome, with phenotypes similar to defects in mtDNA/mtRNA. Thus, we propose that CLPB and CLPXP are crucial to counteract misfolded insoluble protein assemblies that contain nucleotides. This insight is relevant to improve ClpP-modulating drugs that block bacterial growth and for the treatment of human infertility, deafness, and neurodegeneration.
Collapse
|
17
|
Belal S, Goudenège D, Bocca C, Dumont F, Chao De La Barca JM, Desquiret-Dumas V, Gueguen N, Geffroy G, Benyahia R, Kane S, Khiati S, Bris C, Aranyi T, Stockholm D, Inisan A, Renaud A, Barth M, Simard G, Reynier P, Letournel F, Lenaers G, Bonneau D, Chevrollier A, Procaccio V. Glutamate-Induced Deregulation of Krebs Cycle in Mitochondrial Encephalopathy Lactic Acidosis Syndrome Stroke-Like Episodes (MELAS) Syndrome Is Alleviated by Ketone Body Exposure. Biomedicines 2022; 10:biomedicines10071665. [PMID: 35884972 PMCID: PMC9312837 DOI: 10.3390/biomedicines10071665] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/19/2022] [Accepted: 07/01/2022] [Indexed: 11/16/2022] Open
Abstract
(1) Background: The development of mitochondrial medicine has been severely impeded by a lack of effective therapies. (2) Methods: To better understand Mitochondrial Encephalopathy Lactic Acidosis Syndrome Stroke-like episodes (MELAS) syndrome, neuronal cybrid cells carrying different mutation loads of the m.3243A > G mitochondrial DNA variant were analysed using a multi-omic approach. (3) Results: Specific metabolomic signatures revealed that the glutamate pathway was significantly increased in MELAS cells with a direct correlation between glutamate concentration and the m.3243A > G heteroplasmy level. Transcriptomic analysis in mutant cells further revealed alterations in specific gene clusters, including those of the glutamate, gamma-aminobutyric acid pathways, and tricarboxylic acid (TCA) cycle. These results were supported by post-mortem brain tissue analysis from a MELAS patient, confirming the glutamate dysregulation. Exposure of MELAS cells to ketone bodies significantly reduced the glutamate level and improved mitochondrial functions, reducing the accumulation of several intermediate metabolites of the TCA cycle and alleviating the NADH-redox imbalance. (4) Conclusions: Thus, a multi-omic integrated approach to MELAS cells revealed glutamate as a promising disease biomarker, while also indicating that a ketogenic diet should be tested in MELAS patients.
Collapse
Affiliation(s)
- Sophie Belal
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
| | - David Goudenège
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
- Biochemistry and Genetics Department, University Hospital of Angers, 49933 Angers, France; (M.B.); (G.S.)
| | - Cinzia Bocca
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
- Biochemistry and Genetics Department, University Hospital of Angers, 49933 Angers, France; (M.B.); (G.S.)
| | - Florent Dumont
- Signalling and Cardiovascular Pathophysiology, INSERM UMR-S 1180, University of Paris-Saclay, 92296 Châtenay-Malabry, France;
| | - Juan Manuel Chao De La Barca
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
- Biochemistry and Genetics Department, University Hospital of Angers, 49933 Angers, France; (M.B.); (G.S.)
| | - Valérie Desquiret-Dumas
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
- Biochemistry and Genetics Department, University Hospital of Angers, 49933 Angers, France; (M.B.); (G.S.)
| | - Naïg Gueguen
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
- Biochemistry and Genetics Department, University Hospital of Angers, 49933 Angers, France; (M.B.); (G.S.)
| | - Guillaume Geffroy
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
| | - Rayane Benyahia
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
| | - Selma Kane
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
| | - Salim Khiati
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
| | - Céline Bris
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
- Biochemistry and Genetics Department, University Hospital of Angers, 49933 Angers, France; (M.B.); (G.S.)
| | - Tamas Aranyi
- Institute of Enzymology, Research Center for Natural Sciences, H-1519 Budapest, Hungary;
- Department of Molecular Biology, Semmelweis University of Medicine, H-1519 Budapest, Hungary
| | - Daniel Stockholm
- Ecole Pratique des Hautes Etudes, PSL Research University, 75014 Paris, France;
- Centre de Recherche Saint-Antoine, UMRS-938, INSERM, Sorbonne Université, F-75012 Paris, France
| | - Aurore Inisan
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
| | - Aurélie Renaud
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
| | - Magalie Barth
- Biochemistry and Genetics Department, University Hospital of Angers, 49933 Angers, France; (M.B.); (G.S.)
| | - Gilles Simard
- Biochemistry and Genetics Department, University Hospital of Angers, 49933 Angers, France; (M.B.); (G.S.)
| | - Pascal Reynier
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
- Biochemistry and Genetics Department, University Hospital of Angers, 49933 Angers, France; (M.B.); (G.S.)
| | - Franck Letournel
- Department of Neurobiology-Neuropathology, Angers Hospital, 49933 Angers, France;
- UMR INSERM 1066-CNRS 6021, MINT Laboratory, 49933 Angers, France
| | - Guy Lenaers
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
- Service de Neurologie, CHU d'Angers, 49933 Angers, France
| | - Dominique Bonneau
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
- Biochemistry and Genetics Department, University Hospital of Angers, 49933 Angers, France; (M.B.); (G.S.)
| | - Arnaud Chevrollier
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
| | - Vincent Procaccio
- MitoLab Team, UMR CNRS 6015-INSERM U1083, Unité MitoVasc, SFR ICAT, Université d’Angers, 49933 Angers, France; (S.B.); (D.G.); (C.B.); (J.M.C.D.L.B.); (V.D.-D.); (N.G.); (G.G.); (R.B.); (S.K.); (S.K.); (C.B.); (A.I.); (A.R.); (P.R.); (G.L.); (D.B.); (A.C.)
- Biochemistry and Genetics Department, University Hospital of Angers, 49933 Angers, France; (M.B.); (G.S.)
- Correspondence:
| |
Collapse
|
18
|
Organization and expression of the mammalian mitochondrial genome. Nat Rev Genet 2022; 23:606-623. [PMID: 35459860 DOI: 10.1038/s41576-022-00480-x] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2022] [Indexed: 02/07/2023]
Abstract
The mitochondrial genome encodes core subunits of the respiratory chain that drives oxidative phosphorylation and is, therefore, essential for energy conversion. Advances in high-throughput sequencing technologies and cryoelectron microscopy have shed light on the structure and organization of the mitochondrial genome and revealed unique mechanisms of mitochondrial gene regulation. New animal models of impaired mitochondrial protein synthesis have shown how the coordinated regulation of the cytoplasmic and mitochondrial translation machineries ensures the correct assembly of the respiratory chain complexes. These new technologies and disease models are providing a deeper understanding of mitochondrial genome organization and expression and of the diseases caused by impaired energy conversion, including mitochondrial, neurodegenerative, cardiovascular and metabolic diseases. They also provide avenues for the development of treatments for these conditions.
Collapse
|
19
|
Tong HF, Lee HCH, Tong TYT, Lam SF, Sheng B, Chan KW, Li JKY, Tam HKV, Ching CK. Neurological manifestations in m.3243A>G-related disease triggered by metformin. J Diabetes Complications 2022; 36:108111. [PMID: 35123869 DOI: 10.1016/j.jdiacomp.2021.108111] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 12/06/2021] [Accepted: 12/18/2021] [Indexed: 12/26/2022]
Abstract
INTRODUCTION m.3243A>G-related disease has multi-systemic manifestations including diabetes mellitus. It is uncertain whether metformin would trigger neurological manifestations of this disease. This study aims to review the diagnosis and management of m.3243A>G-related diabetes genetically confirmed by our laboratory and to evaluate the risk of metformin use triggering neurological manifestations. METHODS Cases with m.3243A>G detected between 2009 and 2020 were reviewed. Cases with diabetes mellitus were included. Cases with mitochondrial encephalomyopathy, lactic acidosis and stroke-like episodes (MELAS) before diabetes onset were excluded. Odds ratio was calculated for association between metformin use and newly developed neurological manifestations. RESULTS Sixteen patients were identified. Odds ratio for metformin use was 3.50 [0.37-33.0; p = 0.3287]. One illustrative case with clear causal relationship between metformin use and neurological manifestations was described in detail. CONCLUSION m.3243A>G-related diabetes mellitus is underdiagnosed. Red flags including positive family history, short stature, low body weight and hearing loss are often overlooked. Early diagnosis allows regular systemic assessment. In the era of precision medicine and novel therapies, it is prudent to avoid metformin as it could trigger neurological manifestations in this condition. Coenzyme Q10, DPP-IV inhibitors, SGLT2 inhibitors and GLP-1 receptor agonists may be considered.
Collapse
Affiliation(s)
- Hok-Fung Tong
- Kowloon West Cluster Laboratory Genetic Service, Department of Pathology, Princess Margaret Hospital, Hong Kong
| | - Han-Chih Hencher Lee
- Kowloon West Cluster Laboratory Genetic Service, Department of Pathology, Princess Margaret Hospital, Hong Kong
| | - Tsz-Yan Tammy Tong
- Kowloon West Cluster Laboratory Genetic Service, Department of Pathology, Princess Margaret Hospital, Hong Kong
| | - Siu-Fung Lam
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong; Health Sense Diabetes & Endocrine Centre, Hong Kong
| | - Bun Sheng
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong
| | - Kin-Wah Chan
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong
| | | | - Ho-Kee Vicki Tam
- Department of Medicine and Geriatrics, Caritas Medical Centre, Hong Kong
| | - Chor-Kwan Ching
- Kowloon West Cluster Laboratory Genetic Service, Department of Pathology, Princess Margaret Hospital, Hong Kong.
| |
Collapse
|
20
|
Tsybrovskyy O, De Luise M, de Biase D, Caporali L, Fiorini C, Gasparre G, Carelli V, Hackl D, Imamovic L, Haim S, Sobrinho‐Simões M, Tallini G. Papillary thyroid carcinoma tall cell variant shares accumulation of mitochondria, mitochondrial DNA mutations, and loss of oxidative phosphorylation complex I integrity with oncocytic tumors. J Pathol Clin Res 2022; 8:155-168. [PMID: 34792302 PMCID: PMC8822387 DOI: 10.1002/cjp2.247] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/14/2021] [Accepted: 09/27/2021] [Indexed: 02/05/2023]
Abstract
Papillary thyroid carcinoma tall cell variant (PTC-TCV), a form of PTC regarded as an aggressive subtype, shares several morphologic features with oncocytic tumors. Pathogenic homoplasmic/highly heteroplasmic somatic mitochondrial DNA (mtDNA) mutations, usually affecting oxidative phosphorylation (OXPHOS) complex I subunits, are hallmarks of oncocytic cells. To clarify the relationship between PTC-TCV and oncocytic thyroid tumors, 17 PTC-TCV and 16 PTC non-TCV controls (cPTC) were subjected to: (1) transmission electron microscopy (TEM) to assess mitochondria accumulation, (2) next-generation sequencing to analyze mtDNA and nuclear genes frequently mutated in thyroid carcinoma, and (3) immunohistochemistry (IHC) for prohibitin and complex I subunit NDUFS4 to evaluate OXPHOS integrity. TEM showed replacement of cytoplasm by mitochondria in PTC-TCV but not in cPTC cells. All 17 PTC-TCV had at least one mtDNA mutation, totaling 21 mutations; 3/16 cPTC (19%) had mtDNA mutations (p < 0.001). PTC-TCV mtDNA mutations were homoplasmic/highly heteroplasmic, 16/21 (76%) mapping within mtDNA-encoded complex I subunits. MtDNA mutations in cPTC were homoplasmic in 2 cases and at low heteroplasmy in the third case, 2/3 mapping to mtDNA-encoded complex I subunits; 2/3 cPTC with mtDNA mutations had small tall cell subpopulations. PTC-TCV showed strong prohibitin expression and cPTC low-level expression, consistent with massive and limited mitochondrial content, respectively. All 17 PTC-TCV showed NDUFS4 loss (partial or complete) and 3 of 16 cPTC (19%) had (partial) NDUFS4 loss, consistent with lack of complex I integrity in PTC-TCV (p < 0.001). IHC loss of NDUFS4 was associated with mtDNA mutations (p < 0.001). Four BRAF V600E mutated PTCs had loss of NDUSF4 expression limited to neoplastic cell subpopulations with tall cell features, indicating that PTCs first acquire BRAF V600E and then mtDNA mutations. Similar to oncocytic thyroid tumors, PTC-TCV is characterized by mtDNA mutations, massive accumulation of mitochondria, and loss of OXPHOS integrity. IHC loss of NDUFS-4 can be used as a surrogate marker for OXPHOS disruption and to reliably diagnose PTC-TCV.
Collapse
Affiliation(s)
- Oleksiy Tsybrovskyy
- Diagnostic and Research Institute of PathologyMedical University of GrazGrazAustria
- Department of Clinical PathologyOrdensklinikum/Hospital of the Sisters of CharityLinzAustria
| | - Monica De Luise
- Department of Medical and Surgical Sciences (DIMEC) and Center for Applied Biomedical Research (CRBA)University of BolognaBolognaItaly
| | - Dario de Biase
- Department of Pharmacy and Biotechnology (FABIT)University of BolognaBolognaItaly
| | - Leonardo Caporali
- Programma di NeurogeneticaIRCCS Istituto delle Scienze NeurologicheBolognaItaly
| | - Claudio Fiorini
- Programma di NeurogeneticaIRCCS Istituto delle Scienze NeurologicheBolognaItaly
| | - Giuseppe Gasparre
- Department of Medical and Surgical Sciences (DIMEC) and Center for Applied Biomedical Research (CRBA)University of BolognaBolognaItaly
| | - Valerio Carelli
- Programma di NeurogeneticaIRCCS Istituto delle Scienze NeurologicheBolognaItaly
- Department of Biomedical and Neuromotor Sciences (DIBINEM)University of BolognaBolognaItaly
| | - Dominik Hackl
- Department of General and Visceral SurgeryOrdensklinikum/Hospital of the Sisters of CharityLinzAustria
| | - Larisa Imamovic
- Department of Nuclear Medicine & Endocrinology, PET‐CT Center LinzOrdensklinikum/Hospital of the Sisters of CharityLinzAustria
| | - Silke Haim
- Department of Nuclear Medicine & Endocrinology, PET‐CT Center LinzOrdensklinikum/Hospital of the Sisters of CharityLinzAustria
| | - Manuel Sobrinho‐Simões
- IpatimupInstitute of Molecular Pathology and Immunology of the University of PortoPortoPortugal
| | - Giovanni Tallini
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES)University of BolognaBolognaItaly
| |
Collapse
|
21
|
Haast RAM, De Coo IFM, Ivanov D, Khan AR, Jansen JFA, Smeets HJM, Uludağ K. OUP accepted manuscript. Brain Commun 2022; 4:fcac024. [PMID: 35187487 PMCID: PMC8853728 DOI: 10.1093/braincomms/fcac024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 11/26/2021] [Accepted: 02/01/2022] [Indexed: 11/14/2022] Open
Abstract
Mutations of the mitochondrial DNA are an important cause of inherited diseases that can severely affect the tissue’s homeostasis and integrity. The m.3243A > G mutation is the most commonly observed across mitochondrial disorders and is linked to multisystemic complications, including cognitive deficits. In line with in vitro experiments demonstrating the m.3243A > G’s negative impact on neuronal energy production and integrity, m.3243A > G patients show cerebral grey matter tissue changes. However, its impact on the most neuron dense, and therefore energy-consuming brain structure—the cerebellum—remains elusive. In this work, we used high-resolution structural and functional data acquired using 7 T MRI to characterize the neurodegenerative and functional signatures of the cerebellar cortex in m.3243A > G patients. Our results reveal altered tissue integrity within distinct clusters across the cerebellar cortex, apparent by their significantly reduced volume and longitudinal relaxation rate compared with healthy controls, indicating macroscopic atrophy and microstructural pathology. Spatial characterization reveals that these changes occur especially in regions related to the frontoparietal brain network that is involved in information processing and selective attention. In addition, based on resting-state functional MRI data, these clusters exhibit reduced functional connectivity to frontal and parietal cortical regions, especially in patients characterized by (i) a severe disease phenotype and (ii) reduced information-processing speed and attention control. Combined with our previous work, these results provide insight into the neuropathological changes and a solid base to guide longitudinal studies aimed to track disease progression.
Collapse
Affiliation(s)
- Roy A. M. Haast
- Correspondence to: Roy A. M. Haast Centre for Functional and Metabolic Mapping Robarts Research Institute Western University 1151 Richmond St N., London ON, Canada N6A 5B7 E-mail:
| | - Irenaeus F. M. De Coo
- Department of Toxicogenomics, Unit Clinical Genomics, Maastricht University, MHeNs School for Mental Health and Neuroscience, Maastricht, the Netherlands
| | - Dimo Ivanov
- Department of Cognitive Neuroscience, Faculty of Psychology and Neuroscience, Maastricht University, PO Box 616, 6200 MD, Maastricht, the Netherlands
| | - Ali R. Khan
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, Western University, London, ON, Canada, N6A 5B7
- Brain and Mind Institute, Western University, London, ON, Canada, N6A 3K7
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada, N6A 5B7
| | - Jacobus F. A. Jansen
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- School for Mental Health & Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Hubert J. M. Smeets
- Department of Toxicogenomics, Unit Clinical Genomics, Maastricht University, MHeNs School for Mental Health and Neuroscience, Maastricht, the Netherlands
- School for Mental Health & Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Kâmil Uludağ
- IBS Center for Neuroscience Imaging Research, Sungkyunkwan University, Seobu-ro, 2066, Jangan-gu, Suwon, South Korea
- Department of Biomedical Engineering, N Center, Sungkyunkwan University, Seobu-ro, 2066, Jangan-gu, Suwon, South Korea
- Techna Institute and Koerner Scientist in MR Imaging, University Health Network, Toronto, ON, Canada, M5G 1L5
| |
Collapse
|
22
|
Canavero I, Rifino N, Montano V, Pantoni L, Gatti L, Pollaci G, Potenza A, Carrozzini T, Finsterer J, Bersano A. Cognitive aspects of MELAS and CARASAL. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2022; 3:100139. [PMID: 36324419 PMCID: PMC9616374 DOI: 10.1016/j.cccb.2022.100139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 11/18/2022]
Abstract
MELAS and CARASAL have been associated with clinical evidence of cognitive impairment and should be considered as possible causes of early onset Vascular Dementia (VaD), particularly in patients with a familial history of dementia or cerebrovascular disease. Cognitive deterioration in MELAS involves executive function, attention, language, memory, visuospatial, and motor functioning and may correlate with the total Stroke-like episodes (SLEs) lesion load. CARASIL is characterized by late and slow cognition disorders, involving episodic memory, executive functions and facial recognition.
Monogenic diseases, although rare, should be always considered in the diagnostic work up of vascular dementia (VaD), particularly in patients with early onset and a familial history of dementia or cerebrovascular disease. They include, other than CADASIL, Fabry disease, Col4A1-A2 related disorders, which are well recognized causes of VaD, other heritable diseases such as mitochondrial encephalopathy, lactic acidosis, and stroke-like episodes (MELAS) and cathepsin-A related arteriopathy strokes and leukoencephalopathy (CARASAL). MELAS, caused by mtDNA (80% of adult cases m.3243A>G mutations) and more rarely POLG1 mutations, has minimum prevalence of 3.5/100,000. CARASAL, which is caused by mutations in the CTSA gene, has been described in about 19 patients so far. In both these two disorders cognitive features have not been fully explored and are described only in case series or families. This review paper is aimed at providing an update on the clinical manifestations, with particular focus on cognitive aspects, but also neuroradiological and genetic features of these less frequent monogenic diseases associated with VaD.
Collapse
Affiliation(s)
- I Canavero
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - N Rifino
- University of Milano-Bicocca, Milan, Italy
| | - V Montano
- Department of Clinical and Experimental Medicine, Neurological Institute, University of Pisa, Italy
| | - L Pantoni
- Luigi Sacco Department of Biomedical and Clinical Sciences, Stroke and Dementia Lab, University of Milan, Italy
| | - L Gatti
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - G Pollaci
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - A Potenza
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - T Carrozzini
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - J Finsterer
- Krankenanstalt Rudolfstiftung, Messerli Institute, Vienna, Austria
| | - A Bersano
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
- Corresponding author.
| |
Collapse
|
23
|
Klein Gunnewiek TM, Verboven AHA, Pelgrim I, Hogeweg M, Schoenmaker C, Renkema H, Beyrath J, Smeitink J, de Vries BBA, Hoen PBAC', Kozicz T, Nadif Kasri N. Sonlicromanol improves neuronal network dysfunction and transcriptome changes linked to m.3243A>G heteroplasmy in iPSC-derived neurons. Stem Cell Reports 2021; 16:2197-2212. [PMID: 34329596 PMCID: PMC8452519 DOI: 10.1016/j.stemcr.2021.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 07/01/2021] [Accepted: 07/02/2021] [Indexed: 12/13/2022] Open
Abstract
Mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS) is often caused by an adenine to guanine variant at m.3243 (m.3243A>G) of the MT-TL1 gene. To understand how this pathogenic variant affects the nervous system, we differentiated human induced pluripotent stem cells (iPSCs) into excitatory neurons with normal (low heteroplasmy) and impaired (high heteroplasmy) mitochondrial function from MELAS patients with the m.3243A>G pathogenic variant. We combined micro-electrode array (MEA) measurements with RNA sequencing (MEA-seq) and found reduced expression of genes involved in mitochondrial respiration and presynaptic function, as well as non-cell autonomous processes in co-cultured astrocytes. Finally, we show that the clinical phase II drug sonlicromanol can improve neuronal network activity when treatment is initiated early in development. This was intricately linked with changes in the neuronal transcriptome. Overall, we provide insight in transcriptomic changes in iPSC-derived neurons with high m.3243A>G heteroplasmy, and show the pathology is partially reversible by sonlicromanol.
Collapse
Affiliation(s)
- Teun M Klein Gunnewiek
- Department of Medical Imaging, Anatomie, Radboud University Medical Center, Geert Grooteplein 10, Nijmegen, 6525 GA, the Netherlands; Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen, 6500 HB, the Netherlands
| | - Anouk H A Verboven
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen, 6500 HB, the Netherlands; Centre for Molecular and Biomolecular Informatics, Radboudumc, Nijmegen, the Netherlands
| | - Iris Pelgrim
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen, 6500 HB, the Netherlands; Khondrion B.V., Nijmegen, the Netherlands
| | - Mark Hogeweg
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen, 6500 HB, the Netherlands
| | - Chantal Schoenmaker
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen, 6500 HB, the Netherlands
| | | | | | | | - Bert B A de Vries
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen, 6500 HB, the Netherlands
| | - Peter-Bram A C 't Hoen
- Centre for Molecular and Biomolecular Informatics, Radboudumc, Nijmegen, the Netherlands
| | - Tamas Kozicz
- Department of Medical Imaging, Anatomie, Radboud University Medical Center, Geert Grooteplein 10, Nijmegen, 6525 GA, the Netherlands; Department of Laboratory Medicine and Pathology. Mayo Clinic, Rochester, MN 55905, USA; Department of Clinical Genomics, Mayo Clinic, 55905 Rochester, MN, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, 55905 Rochester, MN, USA.
| | - Nael Nadif Kasri
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen, 6500 HB, the Netherlands.
| |
Collapse
|
24
|
Zhong BR, Zhou GF, Song L, Wen QX, Deng XJ, Ma YL, Hu LT, Chen GJ. TUFM is involved in Alzheimer's disease-like pathologies that are associated with ROS. FASEB J 2021; 35:e21445. [PMID: 33774866 DOI: 10.1096/fj.202002461r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/20/2021] [Accepted: 02/01/2021] [Indexed: 12/14/2022]
Abstract
Mitochondrial Tu translation elongation factor (TUFM or EF-Tu) is part of the mitochondrial translation machinery. It is reported that TUFM expression is reduced in the brain of Alzheimer's disease (AD), suggesting that TUFM might play a role in the pathophysiology. In this study, we found that TUFM protein level was decreased in the hippocampus and cortex especially in the aged APP/PS1 mice, an animal model of AD. In HEK cells that stably express full-length human amyloid-β precursor protein (HEK-APP), TUFM knockdown or overexpression increased or reduced the protein levels of β-amyloid protein (Aβ) and β-amyloid converting enzyme 1 (BACE1), respectively. TUFM-mediated reduction of BACE1 was attenuated by translation inhibitor cycloheximide (CHX) or α-[2-[4-(3,4-Dichlorophenyl)-2-thiazolyl]hydrazinylidene]-2-nitro-benzenepropanoic acid (4EGI1), and in cells overexpressing BACE1 constructs deleting the 5' untranslated region (5'UTR). TUFM silencing increased the half-life of BACE1 mRNA, suggesting that RNA stability was affected by TUFM. In support, transcription inhibitor Actinomycin D (ActD) and silencing of nuclear factor κB (NFκB) failed to abolish TUFM-mediated regulation of BACE1 protein and mRNA. We further found that the mitochondria-targeted antioxidant TEMPO diminished the effects of TUFM on BACE1, suggesting that reactive oxygen species (ROS) played an important role. Indeed, cellular ROS levels were affected by TUFM knockdown or overexpression, and TUFM-mediated regulation of apoptosis and Tau phosphorylation at selective sites was attenuated by TEMPO. Collectively, TUFM protein levels were decreased in APP/PS1 mice. TUFM is involved in AD pathology by regulating BACE1 translation, apoptosis, and Tau phosphorylation, in which ROS plays an important role.
Collapse
Affiliation(s)
- Bi-Rou Zhong
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Gui-Feng Zhou
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Li Song
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Qi-Xin Wen
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Xiao-Juan Deng
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Yuan-Lin Ma
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Li-Tian Hu
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China.,Department of Neurology, Nanchong Central Hospital, the Second Clinical College of North Sichuan Medical College, Nanchong, China
| | - Guo-Jun Chen
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| |
Collapse
|
25
|
Partially modified tRNAs for the study of tRNA maturation and function. Methods Enzymol 2021; 658:225-250. [PMID: 34517948 DOI: 10.1016/bs.mie.2021.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Transfer RNA (tRNA) is the most highly and diversely modified class of RNA in all domains of life. However, we still have only a limited understanding of the concerted action of the many enzymes that modify tRNA during tRNA maturation and the synergistic functions of tRNA modifications for protein synthesis. Here, we describe the preparation of in vitro transcribed tRNAs with a partial set of defined modifications and the use of partially modified tRNAs in biochemical assays. By comparing the affinity and activity of tRNA modification enzymes for partially modified and unmodified tRNAs, we gain insight into the preferred pathways of tRNA maturation. Additionally, partially modified tRNAs will be highly useful to investigate the importance of tRNA modifications for tRNA function during translation including the interaction with aminoacyl-tRNA synthases, translation factors and the ribosome. Thereby, the methods described here lay the foundation for understanding the mechanistic function of tRNA modifications.
Collapse
|
26
|
Exploring the Ability of LARS2 Carboxy-Terminal Domain in Rescuing the MELAS Phenotype. Life (Basel) 2021; 11:life11070674. [PMID: 34357047 PMCID: PMC8303833 DOI: 10.3390/life11070674] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/06/2021] [Accepted: 07/08/2021] [Indexed: 12/25/2022] Open
Abstract
The m.3243A>G mutation within the mitochondrial mt-tRNALeu(UUR) gene is the most prevalent variant linked to mitochondrial encephalopathy with lactic acidosis and stroke-like episodes (MELAS) syndrome. This pathogenic mutation causes severe impairment of mitochondrial protein synthesis due to alterations of the mutated tRNA, such as reduced aminoacylation and a lack of post-transcriptional modification. In transmitochondrial cybrids, overexpression of human mitochondrial leucyl-tRNA synthetase (LARS2) has proven effective in rescuing the phenotype associated with m.3243A>G substitution. The rescuing activity resides in the carboxy-terminal domain (Cterm) of the enzyme; however, the precise molecular mechanisms underlying this process have not been fully elucidated. To deepen our knowledge on the rescuing mechanisms, we demonstrated the interactions of the Cterm with mutated mt-tRNALeu(UUR) and its precursor in MELAS cybrids. Further, the effect of Cterm expression on mitochondrial functions was evaluated. We found that Cterm ameliorates de novo mitochondrial protein synthesis, whilst it has no effect on mt-tRNALeu(UUR) steady-state levels and aminoacylation. Despite the complete recovery of cell viability and the increase in mitochondrial translation, Cterm-overexpressing cybrids were not able to recover bioenergetic competence. These data suggest that, in our MELAS cell model, the beneficial effect of Cterm may be mediated by factors that are independent of the mitochondrial bioenergetics.
Collapse
|
27
|
Wang F, Zhang D, Zhang D, Li P, Gao Y. Mitochondrial Protein Translation: Emerging Roles and Clinical Significance in Disease. Front Cell Dev Biol 2021; 9:675465. [PMID: 34277617 PMCID: PMC8280776 DOI: 10.3389/fcell.2021.675465] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/09/2021] [Indexed: 12/28/2022] Open
Abstract
Mitochondria are one of the most important organelles in cells. Mitochondria are semi-autonomous organelles with their own genetic system, and can independently replicate, transcribe, and translate mitochondrial DNA. Translation initiation, elongation, termination, and recycling of the ribosome are four stages in the process of mitochondrial protein translation. In this process, mitochondrial protein translation factors and translation activators, mitochondrial RNA, and other regulatory factors regulate mitochondrial protein translation. Mitochondrial protein translation abnormalities are associated with a variety of diseases, including cancer, cardiovascular diseases, and nervous system diseases. Mutation or deletion of various mitochondrial protein translation factors and translation activators leads to abnormal mitochondrial protein translation. Mitochondrial tRNAs and mitochondrial ribosomal proteins are essential players during translation and mutations in genes encoding them represent a large fraction of mitochondrial diseases. Moreover, there is crosstalk between mitochondrial protein translation and cytoplasmic translation, and the imbalance between mitochondrial protein translation and cytoplasmic translation can affect some physiological and pathological processes. This review summarizes the regulation of mitochondrial protein translation factors, mitochondrial ribosomal proteins, mitochondrial tRNAs, and mitochondrial aminoacyl-tRNA synthetases (mt-aaRSs) in the mitochondrial protein translation process and its relationship with diseases. The regulation of mitochondrial protein translation and cytoplasmic translation in multiple diseases is also summarized.
Collapse
Affiliation(s)
- Fei Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Deyu Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Dejiu Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yanyan Gao
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China.,Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
| |
Collapse
|
28
|
Klein Gunnewiek TM, Van Hugte EJH, Frega M, Guardia GS, Foreman K, Panneman D, Mossink B, Linda K, Keller JM, Schubert D, Cassiman D, Rodenburg R, Vidal Folch N, Oglesbee D, Perales-Clemente E, Nelson TJ, Morava E, Nadif Kasri N, Kozicz T. m.3243A > G-Induced Mitochondrial Dysfunction Impairs Human Neuronal Development and Reduces Neuronal Network Activity and Synchronicity. Cell Rep 2021; 31:107538. [PMID: 32320658 DOI: 10.1016/j.celrep.2020.107538] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 02/13/2020] [Accepted: 03/30/2020] [Indexed: 12/11/2022] Open
Abstract
Epilepsy, intellectual and cortical sensory deficits, and psychiatric manifestations are the most frequent manifestations of mitochondrial diseases. How mitochondrial dysfunction affects neural structure and function remains elusive, mostly because of a lack of proper in vitro neuronal model systems with mitochondrial dysfunction. Leveraging induced pluripotent stem cell technology, we differentiated excitatory cortical neurons (iNeurons) with normal (low heteroplasmy) and impaired (high heteroplasmy) mitochondrial function on an isogenic nuclear DNA background from patients with the common pathogenic m.3243A > G variant of mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS). iNeurons with high heteroplasmy exhibited mitochondrial dysfunction, delayed neural maturation, reduced dendritic complexity, and fewer excitatory synapses. Micro-electrode array recordings of neuronal networks displayed reduced network activity and decreased synchronous network bursting. Impaired neuronal energy metabolism and compromised structural and functional integrity of neurons and neural networks could be the primary drivers of increased susceptibility to neuropsychiatric manifestations of mitochondrial disease.
Collapse
Affiliation(s)
- Teun M Klein Gunnewiek
- Department of Anatomy, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, 6500 HB Nijmegen, the Netherlands; Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, 6500 HB Nijmegen, the Netherlands
| | - Eline J H Van Hugte
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, 6500 HB Nijmegen, the Netherlands
| | - Monica Frega
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, 6500 HB Nijmegen, the Netherlands; Department of Clinical Neurophysiology, University of Twente, 7522 NB Enschede, the Netherlands
| | - Gemma Solé Guardia
- Department of Anatomy, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, 6500 HB Nijmegen, the Netherlands; Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, 6500 HB Nijmegen, the Netherlands
| | - Katharina Foreman
- Department of Cognitive Neuroscience, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, 6500 HB Nijmegen, the Netherlands
| | - Daan Panneman
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Britt Mossink
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, 6500 HB Nijmegen, the Netherlands
| | - Katrin Linda
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, 6500 HB Nijmegen, the Netherlands
| | - Jason M Keller
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, 6500 HB Nijmegen, the Netherlands
| | - Dirk Schubert
- Department of Cognitive Neuroscience, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, 6500 HB Nijmegen, the Netherlands
| | - David Cassiman
- Department of Hepatology, UZ Leuven, 3000 Leuven, Belgium
| | - Richard Rodenburg
- Radboud Center for Mitochondrial Disorders, Radboudumc, 6500 HB Nijmegen, the Netherlands
| | - Noemi Vidal Folch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Devin Oglesbee
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Timothy J Nelson
- Division of General Internal Medicine, Division of Pediatric Cardiology, Departments of Medicine, Molecular Pharmacology, and Experimental Therapeutics, Mayo Clinic Center for Regenerative Medicine, Rochester, MN 55905, USA
| | - Eva Morava
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
| | - Nael Nadif Kasri
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, 6500 HB Nijmegen, the Netherlands; Department of Cognitive Neuroscience, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, 6500 HB Nijmegen, the Netherlands.
| | - Tamas Kozicz
- Department of Anatomy, Radboudumc, Donders Institute for Brain, Cognition, and Behaviour, 6500 HB Nijmegen, the Netherlands; Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, 55905 Rochester, MN, USA.
| |
Collapse
|
29
|
Oh JK, Lima de Carvalho JR, Nuzbrokh Y, Ryu J, Chemudupati T, Mahajan VB, Sparrow JR, Tsang SH. Retinal Manifestations of Mitochondrial Oxidative Phosphorylation Disorders. Invest Ophthalmol Vis Sci 2021; 61:12. [PMID: 33049060 PMCID: PMC7571321 DOI: 10.1167/iovs.61.12.12] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Purpose The purpose of this paper was to discuss manifestations of primary mitochondrial dysfunctions and whether the retinal pigment epithelium or the photoreceptors are preferentially affected. Methods A retrospective analysis was performed of patients with clinically and laboratory confirmed diagnoses of maternally inherited diabetes and deafness (MIDD) or Kearns–Sayre syndrome (KSS). Patients underwent full ophthalmic examination, full-field electroretinogram, and multimodal imaging studies, including short-wavelength autofluorescence, spectral domain-optical coherence tomography, and color fundus photography. Results A total of five patients with MIDD and four patients with KSS were evaluated at two tertiary referral centers. Mean age at initial evaluation was 50.3 years old. Nascent outer retinal tubulations corresponding with faint foci of autofluorescence were observed in two patients with MIDD. Characteristic features of this cohort included a foveal sparing phenotype observed in 13 of 18 eyes (72%), global absence of intraretinal pigment migration, and preserved retinal function on full-field electroretinogram testing in 12 of 16 eyes (75%). One patient diagnosed with MIDD presented with an unusual pattern of atrophy surrounding the parapapillary region and one patient with KSS presented with an atypical choroideremia-like phenotype. Conclusions MIDD and KSS are phenotypically heterogeneous disorders. Several features of disease suggest that primary mitochondrial dysfunction may first affect the retinal pigment epithelium followed by secondary photoreceptor loss. Similarities between primary mitochondrial degenerations and retinal disorders, such as age-related macular degeneration may suggest a primary role of mitochondria in the pathogenesis of these oligogenic disorders.
Collapse
Affiliation(s)
- Jin Kyun Oh
- Jonas Children's Vision Care, Department of Ophthalmology, Columbia University Irving Medical Center, New York, New York, United States.,State University of New York at Downstate Medical Center, Brooklyn, New York, United States
| | - Jose Ronaldo Lima de Carvalho
- Jonas Children's Vision Care, Department of Ophthalmology, Columbia University Irving Medical Center, New York, New York, United States.,Department of Ophthalmology, Empresa Brasileira de Servicos Hospitalares (EBSERH) - Hospital das Clinicas de Pernambuco (HCPE), Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil.,Department of Ophthalmology, Federal University of São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Yan Nuzbrokh
- Jonas Children's Vision Care, Department of Ophthalmology, Columbia University Irving Medical Center, New York, New York, United States.,Renaissance School of Medicine at Stony Brook University, Stony Brook, New York, United States
| | - Joseph Ryu
- Jonas Children's Vision Care, Department of Ophthalmology, Columbia University Irving Medical Center, New York, New York, United States
| | - Teja Chemudupati
- Molecular Surgery Laboratory, Byers Eye Institute, Stanford University, Palo Alto, California, United States
| | - Vinit B Mahajan
- Molecular Surgery Laboratory, Byers Eye Institute, Stanford University, Palo Alto, California, United States.,Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States
| | - Janet R Sparrow
- Jonas Children's Vision Care, Department of Ophthalmology, Columbia University Irving Medical Center, New York, New York, United States.,Department of Pathology and Cell Biology and Columbia Stem Cell Initiative (CSCI), Columbia University Irving Medical Center, New York, New York, United States
| | - Stephen H Tsang
- Jonas Children's Vision Care, Department of Ophthalmology, Columbia University Irving Medical Center, New York, New York, United States.,Department of Pathology and Cell Biology and Columbia Stem Cell Initiative (CSCI), Columbia University Irving Medical Center, New York, New York, United States
| |
Collapse
|
30
|
Shu L, Hu C, Xu M, Yu J, He H, Lin J, Sha H, Lu B, Engelender S, Guan M, Song Z. ATAD3B is a mitophagy receptor mediating clearance of oxidative stress-induced damaged mitochondrial DNA. EMBO J 2021; 40:e106283. [PMID: 33665835 PMCID: PMC8047441 DOI: 10.15252/embj.2020106283] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 01/22/2021] [Accepted: 02/01/2021] [Indexed: 12/11/2022] Open
Abstract
Mitochondrial DNA (mtDNA) encodes several key components of respiratory chain complexes that produce cellular energy through oxidative phosphorylation. mtDNA is vulnerable to damage under various physiological stresses, especially oxidative stress. mtDNA damage leads to mitochondrial dysfunction, and dysfunctional mitochondria can be removed by mitophagy, an essential process in cellular homeostasis. However, how damaged mtDNA is selectively cleared from the cell, and how damaged mtDNA triggers mitophagy, remain mostly unknown. Here, we identified a novel mitophagy receptor, ATAD3B, which is specifically expressed in primates. ATAD3B contains a LIR motif that binds to LC3 and promotes oxidative stress-induced mitophagy in a PINK1-independent manner, thus promoting the clearance of damaged mtDNA induced by oxidative stress. Under normal conditions, ATAD3B hetero-oligomerizes with ATAD3A, thus promoting the targeting of the C-terminal region of ATAD3B to the mitochondrial intermembrane space. Oxidative stress-induced mtDNA damage or mtDNA depletion reduces ATAD3B-ATAD3A hetero-oligomerization and leads to exposure of the ATAD3B C-terminus at the mitochondrial outer membrane and subsequent recruitment of LC3 for initiating mitophagy. Furthermore, ATAD3B is little expressed in m.3243A > G mutated cells and MELAS patient fibroblasts showing endogenous oxidative stress, and ATAD3B re-expression promotes the clearance of m.3243A > G mutated mtDNA. Our findings uncover a new pathway to selectively remove damaged mtDNA and reveal that increasing ATAD3B activity is a potential therapeutic approach for mitochondrial diseases.
Collapse
Affiliation(s)
- Li Shu
- Hubei Key Laboratory of Cell HomeostasisCollege of Life SciencesFrontier Science Center for Immunology and MetabolismWuhan UniversityWuhanChina
| | - Chao Hu
- Hubei Key Laboratory of Cell HomeostasisCollege of Life SciencesFrontier Science Center for Immunology and MetabolismWuhan UniversityWuhanChina
| | - Meng Xu
- Hubei Key Laboratory of Cell HomeostasisCollege of Life SciencesFrontier Science Center for Immunology and MetabolismWuhan UniversityWuhanChina
| | - Jianglong Yu
- Hubei Key Laboratory of Cell HomeostasisCollege of Life SciencesFrontier Science Center for Immunology and MetabolismWuhan UniversityWuhanChina
| | - He He
- Hubei Key Laboratory of Cell HomeostasisCollege of Life SciencesFrontier Science Center for Immunology and MetabolismWuhan UniversityWuhanChina
| | - Jie Lin
- Department of NeurologyHuashan HospitalFudan UniversityShanghaiChina
| | - Hongying Sha
- State Key Laboratory of Medical NeurobiologyInstitute of Brain ScienceShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Bin Lu
- Attardi Institute of Mitochondrial BiomedicineSchool of Life SciencesWenzhou Medical UniversityWenzhouChina
| | - Simone Engelender
- Department of BiochemistryRappaport Faculty of MedicineTechnion‐Israel Institute of TechnologyHaifaIsrael
| | - Minxin Guan
- Institute of GeneticsZhejiang University School of MedicineHangzhouChina
| | - Zhiyin Song
- Hubei Key Laboratory of Cell HomeostasisCollege of Life SciencesFrontier Science Center for Immunology and MetabolismWuhan UniversityWuhanChina
| |
Collapse
|
31
|
Richter U, McFarland R, Taylor RW, Pickett SJ. The molecular pathology of pathogenic mitochondrial tRNA variants. FEBS Lett 2021; 595:1003-1024. [PMID: 33513266 PMCID: PMC8600956 DOI: 10.1002/1873-3468.14049] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/14/2021] [Accepted: 01/18/2021] [Indexed: 12/16/2022]
Abstract
Mitochondrial diseases are clinically and genetically heterogeneous disorders, caused by pathogenic variants in either the nuclear or mitochondrial genome. This heterogeneity is particularly striking for disease caused by variants in mitochondrial DNA-encoded tRNA (mt-tRNA) genes, posing challenges for both the treatment of patients and understanding the molecular pathology. In this review, we consider disease caused by the two most common pathogenic mt-tRNA variants: m.3243A>G (within MT-TL1, encoding mt-tRNALeu(UUR) ) and m.8344A>G (within MT-TK, encoding mt-tRNALys ), which together account for the vast majority of all mt-tRNA-related disease. We compare and contrast the clinical disease they are associated with, as well as their molecular pathologies, and consider what is known about the likely molecular mechanisms of disease. Finally, we discuss the role of mitochondrial-nuclear crosstalk in the manifestation of mt-tRNA-associated disease and how research in this area not only has the potential to uncover molecular mechanisms responsible for the vast clinical heterogeneity associated with these variants but also pave the way to develop treatment options for these devastating diseases.
Collapse
Affiliation(s)
- Uwe Richter
- Wellcome Centre for Mitochondrial ResearchThe Medical SchoolNewcastle UniversityUK
- Molecular and Integrative Biosciences Research ProgrammeFaculty of Biological and Environmental SciencesUniversity of HelsinkiFinland
- Newcastle University Biosciences InstituteNewcastle UniversityUK
| | - Robert McFarland
- Wellcome Centre for Mitochondrial ResearchThe Medical SchoolNewcastle UniversityUK
- Newcastle University Translational and Clinical Research InstituteNewcastle UniversityUK
| | - Robert W. Taylor
- Wellcome Centre for Mitochondrial ResearchThe Medical SchoolNewcastle UniversityUK
- Newcastle University Translational and Clinical Research InstituteNewcastle UniversityUK
| | - Sarah J. Pickett
- Wellcome Centre for Mitochondrial ResearchThe Medical SchoolNewcastle UniversityUK
- Newcastle University Translational and Clinical Research InstituteNewcastle UniversityUK
| |
Collapse
|
32
|
Basu U, Bostwick AM, Das K, Dittenhafer-Reed KE, Patel SS. Structure, mechanism, and regulation of mitochondrial DNA transcription initiation. J Biol Chem 2020; 295:18406-18425. [PMID: 33127643 PMCID: PMC7939475 DOI: 10.1074/jbc.rev120.011202] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 10/29/2020] [Indexed: 12/14/2022] Open
Abstract
Mitochondria are specialized compartments that produce requisite ATP to fuel cellular functions and serve as centers of metabolite processing, cellular signaling, and apoptosis. To accomplish these roles, mitochondria rely on the genetic information in their small genome (mitochondrial DNA) and the nucleus. A growing appreciation for mitochondria's role in a myriad of human diseases, including inherited genetic disorders, degenerative diseases, inflammation, and cancer, has fueled the study of biochemical mechanisms that control mitochondrial function. The mitochondrial transcriptional machinery is different from nuclear machinery. The in vitro re-constituted transcriptional complexes of Saccharomyces cerevisiae (yeast) and humans, aided with high-resolution structures and biochemical characterizations, have provided a deeper understanding of the mechanism and regulation of mitochondrial DNA transcription. In this review, we will discuss recent advances in the structure and mechanism of mitochondrial transcription initiation. We will follow up with recent discoveries and formative findings regarding the regulatory events that control mitochondrial DNA transcription, focusing on those involved in cross-talk between the mitochondria and nucleus.
Collapse
Affiliation(s)
- Urmimala Basu
- Department of Biochemistry and Molecular Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA; Graduate School of Biomedical Sciences, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | | | - Kalyan Das
- Department of Microbiology, Immunology, and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | | | - Smita S Patel
- Department of Biochemistry and Molecular Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA.
| |
Collapse
|
33
|
Patananan AN, Sercel AJ, Wu TH, Ahsan FM, Torres A, Kennedy SAL, Vandiver A, Collier AJ, Mehrabi A, Van Lew J, Zakin L, Rodriguez N, Sixto M, Tadros W, Lazar A, Sieling PA, Nguyen TL, Dawson ER, Braas D, Golovato J, Cisneros L, Vaske C, Plath K, Rabizadeh S, Niazi KR, Chiou PY, Teitell MA. Pressure-Driven Mitochondrial Transfer Pipeline Generates Mammalian Cells of Desired Genetic Combinations and Fates. Cell Rep 2020; 33:108562. [PMID: 33378680 PMCID: PMC7927156 DOI: 10.1016/j.celrep.2020.108562] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 10/29/2020] [Accepted: 12/06/2020] [Indexed: 01/19/2023] Open
Abstract
Generating mammalian cells with desired mitochondrial DNA (mtDNA) sequences is enabling for studies of mitochondria, disease modeling, and potential regenerative therapies. MitoPunch, a high-throughput mitochondrial transfer device, produces cells with specific mtDNA-nuclear DNA (nDNA) combinations by transferring isolated mitochondria from mouse or human cells into primary or immortal mtDNA-deficient (ρ0) cells. Stable isolated mitochondrial recipient (SIMR) cells isolated in restrictive media permanently retain donor mtDNA and reacquire respiration. However, SIMR fibroblasts maintain a ρ0-like cell metabolome and transcriptome despite growth in restrictive media. We reprogrammed non-immortal SIMR fibroblasts into induced pluripotent stem cells (iPSCs) with subsequent differentiation into diverse functional cell types, including mesenchymal stem cells (MSCs), adipocytes, osteoblasts, and chondrocytes. Remarkably, after reprogramming and differentiation, SIMR fibroblasts molecularly and phenotypically resemble unmanipulated control fibroblasts carried through the same protocol. Thus, our MitoPunch "pipeline" enables the production of SIMR cells with unique mtDNA-nDNA combinations for additional studies and applications in multiple cell types.
Collapse
Affiliation(s)
- Alexander N Patananan
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Alexander J Sercel
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | | | - Fasih M Ahsan
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Alejandro Torres
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Stephanie A L Kennedy
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Amy Vandiver
- Division of Dermatology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Amanda J Collier
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | | | | | - Lise Zakin
- NantWorks, LLC, Culver City, CA 90232, USA
| | | | | | | | - Adam Lazar
- NantWorks, LLC, Culver City, CA 90232, USA
| | | | - Thang L Nguyen
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Emma R Dawson
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Daniel Braas
- UCLA Metabolomics Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | | | | | | | - Kathrin Plath
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Shahrooz Rabizadeh
- NanoCav LLC, Culver City, CA 90232, USA; NantWorks, LLC, Culver City, CA 90232, USA
| | - Kayvan R Niazi
- NanoCav LLC, Culver City, CA 90232, USA; NantWorks, LLC, Culver City, CA 90232, USA
| | - Pei-Yu Chiou
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Michael A Teitell
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
34
|
Ding Q, Kucharczyk R, Zhao W, Dautant A, Xu S, Niedzwiecka K, Su X, Giraud MF, Gombeau K, Zhang M, Xie H, Zeng C, Bouhier M, di Rago JP, Liu Z, Tribouillard-Tanvier D, Chen H. Case Report: Identification of a Novel Variant (m.8909T>C) of Human Mitochondrial ATP6 Gene and Its Functional Consequences on Yeast ATP Synthase. Life (Basel) 2020; 10:life10090215. [PMID: 32971864 PMCID: PMC7555451 DOI: 10.3390/life10090215] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/09/2020] [Accepted: 09/17/2020] [Indexed: 11/16/2022] Open
Abstract
With the advent of next generation sequencing, the list of mitochondrial DNA (mtDNA) mutations identified in patients rapidly and continuously expands. They are frequently found in a limited number of cases, sometimes a single individual (as with the case herein reported) and in heterogeneous genetic backgrounds (heteroplasmy), which makes it difficult to conclude about their pathogenicity and functional consequences. As an organism amenable to mitochondrial DNA manipulation, able to survive by fermentation to loss-of-function mtDNA mutations, and where heteroplasmy is unstable, Saccharomyces cerevisiae is an excellent model for investigating novel human mtDNA variants, in isolation and in a controlled genetic context. We herein report the identification of a novel variant in mitochondrial ATP6 gene, m.8909T>C. It was found in combination with the well-known pathogenic m.3243A>G mutation in mt-tRNALeu. We show that an equivalent of the m.8909T>C mutation compromises yeast adenosine tri-phosphate (ATP) synthase assembly/stability and reduces the rate of mitochondrial ATP synthesis by 20-30% compared to wild type yeast. Other previously reported ATP6 mutations with a well-established pathogenicity (like m.8993T>C and m.9176T>C) were shown to have similar effects on yeast ATP synthase. It can be inferred that alone the m.8909T>C variant has the potential to compromise human health.
Collapse
Affiliation(s)
- Qiuju Ding
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing 211166, China; (Q.D.); (W.Z.); (S.X.); (M.Z.); (H.X.); (C.Z.); (Z.L.)
| | - Róża Kucharczyk
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 00090 Warsaw, Poland; (R.K.); (K.N.)
| | - Weiwei Zhao
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing 211166, China; (Q.D.); (W.Z.); (S.X.); (M.Z.); (H.X.); (C.Z.); (Z.L.)
| | - Alain Dautant
- Institut de Biochimie et Génétique Cellulaires, Université de Bordeaux, CNRS, UMR 5095, F-33000 Bordeaux, France; (A.D.); (X.S.); (M.-F.G.); (K.G.); (M.B.); (J.-P.d.R.)
| | - Shutian Xu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing 211166, China; (Q.D.); (W.Z.); (S.X.); (M.Z.); (H.X.); (C.Z.); (Z.L.)
| | - Katarzyna Niedzwiecka
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 00090 Warsaw, Poland; (R.K.); (K.N.)
| | - Xin Su
- Institut de Biochimie et Génétique Cellulaires, Université de Bordeaux, CNRS, UMR 5095, F-33000 Bordeaux, France; (A.D.); (X.S.); (M.-F.G.); (K.G.); (M.B.); (J.-P.d.R.)
| | - Marie-France Giraud
- Institut de Biochimie et Génétique Cellulaires, Université de Bordeaux, CNRS, UMR 5095, F-33000 Bordeaux, France; (A.D.); (X.S.); (M.-F.G.); (K.G.); (M.B.); (J.-P.d.R.)
| | - Kewin Gombeau
- Institut de Biochimie et Génétique Cellulaires, Université de Bordeaux, CNRS, UMR 5095, F-33000 Bordeaux, France; (A.D.); (X.S.); (M.-F.G.); (K.G.); (M.B.); (J.-P.d.R.)
| | - Mingchao Zhang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing 211166, China; (Q.D.); (W.Z.); (S.X.); (M.Z.); (H.X.); (C.Z.); (Z.L.)
| | - Honglang Xie
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing 211166, China; (Q.D.); (W.Z.); (S.X.); (M.Z.); (H.X.); (C.Z.); (Z.L.)
| | - Caihong Zeng
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing 211166, China; (Q.D.); (W.Z.); (S.X.); (M.Z.); (H.X.); (C.Z.); (Z.L.)
| | - Marine Bouhier
- Institut de Biochimie et Génétique Cellulaires, Université de Bordeaux, CNRS, UMR 5095, F-33000 Bordeaux, France; (A.D.); (X.S.); (M.-F.G.); (K.G.); (M.B.); (J.-P.d.R.)
| | - Jean-Paul di Rago
- Institut de Biochimie et Génétique Cellulaires, Université de Bordeaux, CNRS, UMR 5095, F-33000 Bordeaux, France; (A.D.); (X.S.); (M.-F.G.); (K.G.); (M.B.); (J.-P.d.R.)
| | - Zhihong Liu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing 211166, China; (Q.D.); (W.Z.); (S.X.); (M.Z.); (H.X.); (C.Z.); (Z.L.)
| | - Déborah Tribouillard-Tanvier
- Institut de Biochimie et Génétique Cellulaires, Université de Bordeaux, CNRS, UMR 5095, F-33000 Bordeaux, France; (A.D.); (X.S.); (M.-F.G.); (K.G.); (M.B.); (J.-P.d.R.)
- Institut national de la santé et de la recherche médicale, 75000 Paris, France
- Correspondence: (D.T.-T.); (H.C.)
| | - Huimei Chen
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing 211166, China; (Q.D.); (W.Z.); (S.X.); (M.Z.); (H.X.); (C.Z.); (Z.L.)
- Correspondence: (D.T.-T.); (H.C.)
| |
Collapse
|
35
|
Uittenbogaard M, Chiaramello A. Maternally inherited mitochondrial respiratory disorders: from pathogenetic principles to therapeutic implications. Mol Genet Metab 2020; 131:38-52. [PMID: 32624334 PMCID: PMC7749081 DOI: 10.1016/j.ymgme.2020.06.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/22/2020] [Accepted: 06/23/2020] [Indexed: 01/19/2023]
Abstract
Maternally inherited mitochondrial respiratory disorders are rare, progressive, and multi-systemic diseases that remain intractable, with no effective therapeutic interventions. Patients share a defective oxidative phosphorylation pathway responsible for mitochondrial ATP synthesis, in most cases due to pathogenic mitochondrial variants transmitted from mother to child or to a rare de novo mutation or large-scale deletion of the mitochondrial genome. The clinical diagnosis of these mitochondrial diseases is difficult due to exceptionally high clinical variability, while their genetic diagnosis has improved with the advent of next-generation sequencing. The mechanisms regulating the penetrance of the mitochondrial variants remain unresolved with the patient's nuclear background, epigenomic regulation, heteroplasmy, mitochondrial haplogroups, and environmental factors thought to act as rheostats. The lack of animal models mimicking the phenotypic manifestations of these disorders has hampered efforts toward curative therapies. Patient-derived cellular paradigms provide alternative models for elucidating the pathogenic mechanisms and screening pharmacological small molecules to enhance mitochondrial function. Recent progress has been made in designing promising approaches to curtail the negative impact of dysfunctional mitochondria and alleviate clinical symptoms: 1) boosting mitochondrial biogenesis; 2) shifting heteroplasmy; 3) reprogramming metabolism; and 4) administering hypoxia-based treatment. Here, we discuss their varying efficacies and limitations and provide an outlook on their therapeutic potential and clinical application.
Collapse
Affiliation(s)
- Martine Uittenbogaard
- George Washington University School of Medicine and Health Sciences, Department of Anatomy and Cell Biology, 2300 I Street N.W., Washington, DC 20037, USA
| | - Anne Chiaramello
- George Washington University School of Medicine and Health Sciences, Department of Anatomy and Cell Biology, 2300 I Street N.W., Washington, DC 20037, USA.
| |
Collapse
|
36
|
Pioglitazone and Deoxyribonucleoside Combination Treatment Increases Mitochondrial Respiratory Capacity in m.3243A>G MELAS Cybrid Cells. Int J Mol Sci 2020; 21:ijms21062139. [PMID: 32244971 PMCID: PMC7139379 DOI: 10.3390/ijms21062139] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/17/2020] [Accepted: 03/17/2020] [Indexed: 01/05/2023] Open
Abstract
The lack of effective treatments for mitochondrial disease has seen the development of new approaches, including those that aim to stimulate mitochondrial biogenesis to boost ATP generation above a critical disease threshold. Here, we examine the effects of the peroxisome proliferator-activated receptor γ (PPARγ) activator pioglitazone (PioG), in combination with deoxyribonucleosides (dNs), on mitochondrial biogenesis in cybrid cells containing >90% of the m.3243A>G mutation associated with mitochondrial encephalopathy, lactic acidosis, and stroke-like episodes (MELAS). PioG + dNs combination treatment increased mtDNA copy number and mitochondrial mass in both control (CON) and m.3243A>G (MUT) cybrids, with no adverse effects on cell proliferation. PioG + dNs also increased mtDNA-encoded transcripts in CON cybrids, but had the opposite effect in MUT cybrids, reducing the already elevated transcript levels. Steady-state levels of mature oxidative phosphorylation (OXPHOS) protein complexes were increased by PioG + dNs treatment in CON cybrids, but were unchanged in MUT cybrids. However, treatment was able to significantly increase maximal mitochondrial oxygen consumption rates and cell respiratory control ratios in both CON and MUT cybrids. Overall, these findings highlight the ability of PioG + dNs to improve mitochondrial respiratory function in cybrid cells containing the m.3243A>G MELAS mutation, as well as their potential for development into novel therapies to treat mitochondrial disease.
Collapse
|
37
|
Mitochondrial Dynamics Regulation in Skin Fibroblasts from Mitochondrial Disease Patients. Biomolecules 2020; 10:biom10030450. [PMID: 32183225 PMCID: PMC7175126 DOI: 10.3390/biom10030450] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/20/2020] [Accepted: 03/09/2020] [Indexed: 12/19/2022] Open
Abstract
Mitochondria are highly dynamic organelles that constantly fuse, divide, and move, and their function is regulated and maintained by their morphologic changes. Mitochondrial disease (MD) comprises a group of disorders involving mitochondrial dysfunction. However, it is not clear whether changes in mitochondrial morphology are related to MD. In this study, we examined mitochondrial morphology in fibroblasts from patients with MD (mitochondrial myopathy, encephalopathy, lactic acidosis, and stroke-like episodes (MELAS) and Leigh syndrome). We observed that MD fibroblasts exhibited significant mitochondrial fragmentation by upregulation of Drp1, which is responsible for mitochondrial fission. Interestingly, the inhibition of mitochondrial fragmentation by Drp1 knockdown enhanced cellular toxicity and led to cell death in MD fibroblasts. These results suggest that mitochondrial fission plays a critical role in the attenuation of mitochondrial damage in MD fibroblasts.
Collapse
|
38
|
Gong S, Wang X, Meng F, Cui L, Yi Q, Zhao Q, Cang X, Cai Z, Mo JQ, Liang Y, Guan MX. Overexpression of mitochondrial histidyl-tRNA synthetase restores mitochondrial dysfunction caused by a deafness-associated tRNAHis mutation. J Biol Chem 2020. [DOI: 10.1016/s0021-9258(17)49906-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
39
|
Ferreira N, Perks KL, Rossetti G, Rudler DL, Hughes LA, Ermer JA, Scott LH, Kuznetsova I, Richman TR, Narayana VK, Abudulai LN, Shearwood AJ, Cserne Szappanos H, Tull D, Yeoh GC, Hool LC, Filipovska A, Rackham O. Stress signaling and cellular proliferation reverse the effects of mitochondrial mistranslation. EMBO J 2019; 38:e102155. [PMID: 31721250 PMCID: PMC6912024 DOI: 10.15252/embj.2019102155] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 09/17/2019] [Accepted: 09/20/2019] [Indexed: 12/12/2022] Open
Abstract
Translation fidelity is crucial for prokaryotes and eukaryotic nuclear-encoded proteins; however, little is known about the role of mistranslation in mitochondria and its potential effects on metabolism. We generated yeast and mouse models with error-prone and hyper-accurate mitochondrial translation, and found that translation rate is more important than translational accuracy for cell function in mammals. Specifically, we found that mitochondrial mistranslation causes reduced overall mitochondrial translation and respiratory complex assembly rates. In mammals, this effect is compensated for by increased mitochondrial protein stability and upregulation of the citric acid cycle. Moreover, this induced mitochondrial stress signaling, which enables the recovery of mitochondrial translation via mitochondrial biogenesis, telomerase expression, and cell proliferation, and thereby normalizes metabolism. Conversely, we show that increased fidelity of mitochondrial translation reduces the rate of protein synthesis without eliciting a mitochondrial stress response. Consequently, the rate of translation cannot be recovered and this leads to dilated cardiomyopathy in mice. In summary, our findings reveal mammalian-specific signaling pathways that respond to changes in the fidelity of mitochondrial protein synthesis and affect metabolism.
Collapse
Affiliation(s)
- Nicola Ferreira
- Harry Perkins Institute of Medical ResearchNedlandsWAAustralia
- The University of Western Australia Centre for Medical ResearchCrawleyWAAustralia
| | - Kara L Perks
- Harry Perkins Institute of Medical ResearchNedlandsWAAustralia
- The University of Western Australia Centre for Medical ResearchCrawleyWAAustralia
| | - Giulia Rossetti
- Harry Perkins Institute of Medical ResearchNedlandsWAAustralia
- The University of Western Australia Centre for Medical ResearchCrawleyWAAustralia
| | - Danielle L Rudler
- Harry Perkins Institute of Medical ResearchNedlandsWAAustralia
- The University of Western Australia Centre for Medical ResearchCrawleyWAAustralia
| | - Laetitia A Hughes
- Harry Perkins Institute of Medical ResearchNedlandsWAAustralia
- The University of Western Australia Centre for Medical ResearchCrawleyWAAustralia
| | - Judith A Ermer
- Harry Perkins Institute of Medical ResearchNedlandsWAAustralia
- The University of Western Australia Centre for Medical ResearchCrawleyWAAustralia
| | - Louis H Scott
- Harry Perkins Institute of Medical ResearchNedlandsWAAustralia
- The University of Western Australia Centre for Medical ResearchCrawleyWAAustralia
| | - Irina Kuznetsova
- Harry Perkins Institute of Medical ResearchNedlandsWAAustralia
- The University of Western Australia Centre for Medical ResearchCrawleyWAAustralia
| | - Tara R Richman
- Harry Perkins Institute of Medical ResearchNedlandsWAAustralia
- The University of Western Australia Centre for Medical ResearchCrawleyWAAustralia
| | - Vinod K Narayana
- Metabolomics AustraliaBio21 Institute of Molecular Science and BiotechnologyUniversity of MelbourneParkvilleVic.Australia
| | - Laila N Abudulai
- Centre for Microscopy, Characterisation and AnalysisThe University of Western AustraliaPerthWAAustralia
- School of Molecular SciencesThe University of Western Australia, CrawleyWAAustralia
- The School of Biomedical SciencesThe University of Western AustraliaNedlandsWAAustralia
| | - Anne‐Marie J Shearwood
- Harry Perkins Institute of Medical ResearchNedlandsWAAustralia
- The University of Western Australia Centre for Medical ResearchCrawleyWAAustralia
| | | | - Dedreia Tull
- Metabolomics AustraliaBio21 Institute of Molecular Science and BiotechnologyUniversity of MelbourneParkvilleVic.Australia
| | - George C Yeoh
- Harry Perkins Institute of Medical ResearchNedlandsWAAustralia
| | - Livia C Hool
- School of Human Sciences (Physiology)The University of Western AustraliaCrawleyWAAustralia
- Victor Chang Cardiac Research InstituteDarlinghurstNSWAustralia
| | - Aleksandra Filipovska
- Harry Perkins Institute of Medical ResearchNedlandsWAAustralia
- The University of Western Australia Centre for Medical ResearchCrawleyWAAustralia
- School of Molecular SciencesThe University of Western Australia, CrawleyWAAustralia
| | - Oliver Rackham
- Harry Perkins Institute of Medical ResearchNedlandsWAAustralia
- School of Pharmacy and Biomedical SciencesCurtin UniversityBentleyWAAustralia
- Curtin Health Innovation Research InstituteCurtin UniversityBentleyWAAustralia
| |
Collapse
|
40
|
Nagao T, Shintani Y, Hayashi T, Kioka H, Kato H, Nishida Y, Yamazaki S, Tsukamoto O, Yashirogi S, Yazawa I, Asano Y, Shinzawa-Itoh K, Imamura H, Suzuki T, Suzuki T, Goto YI, Takashima S. Higd1a improves respiratory function in the models of mitochondrial disorder. FASEB J 2019; 34:1859-1871. [PMID: 31914602 DOI: 10.1096/fj.201800389r] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/26/2018] [Accepted: 06/12/2018] [Indexed: 12/22/2022]
Abstract
The respiratory chain (RC) transports electrons to form a proton motive force that is required for ATP synthesis in the mitochondria. RC disorders cause mitochondrial diseases that have few effective treatments; therefore, novel therapeutic strategies are critically needed. We previously identified Higd1a as a positive regulator of cytochrome c oxidase (CcO) in the RC. Here, we test that Higd1a has a beneficial effect by increasing CcO activity in the models of mitochondrial dysfunction. We first demonstrated the tissue-protective effects of Higd1a via in situ measurement of mitochondrial ATP concentrations ([ATP]mito) in a zebrafish hypoxia model. Heart-specific Higd1a overexpression mitigated the decline in [ATP]mito under hypoxia and preserved cardiac function in zebrafish. Based on the in vivo results, we examined the effects of exogenous HIGD1A on three cellular models of mitochondrial disease; notably, HIGD1A improved respiratory function that was coupled with increased ATP synthesis and demonstrated cellular protection in all three models. Finally, enzyme kinetic analysis revealed that Higd1a significantly increased the maximal velocity of the reaction between CcO and cytochrome c without changing the affinity between them, indicating that Higd1a is a positive modulator of CcO. These results corroborate that Higd1a, or its mimic, provides therapeutic options for the treatment of mitochondrial diseases.
Collapse
Affiliation(s)
- Takemasa Nagao
- Department of Medical Biochemistry, Graduate School of Frontier Biological Science, Osaka University, Suita, Japan
| | - Yasunori Shintani
- Department of Medical Biochemistry, Graduate School of Frontier Biological Science, Osaka University, Suita, Japan
| | - Takaharu Hayashi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hidetaka Kioka
- Department of Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hisakazu Kato
- Department of Medical Biochemistry, Graduate School of Frontier Biological Science, Osaka University, Suita, Japan
| | - Yuya Nishida
- Department of Medical Biochemistry, Graduate School of Frontier Biological Science, Osaka University, Suita, Japan.,Japan Science and Technology Agency-Core Research for Evolutional Science and Technology (CREST), Kawaguchi, Japan
| | - Satoru Yamazaki
- Department of Cell Biology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Osamu Tsukamoto
- Department of Medical Biochemistry, Graduate School of Frontier Biological Science, Osaka University, Suita, Japan
| | - Shohei Yashirogi
- Department of Medical Biochemistry, Graduate School of Frontier Biological Science, Osaka University, Suita, Japan
| | - Issei Yazawa
- Department of Medical Biochemistry, Graduate School of Frontier Biological Science, Osaka University, Suita, Japan
| | - Yoshihiro Asano
- Department of Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | | | - Hiromi Imamura
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Takeo Suzuki
- Department of Chemistry and Biotechnology, Graduate School of Engineering, University of Tokyo, Tokyo, Japan
| | - Tsutomu Suzuki
- Department of Chemistry and Biotechnology, Graduate School of Engineering, University of Tokyo, Tokyo, Japan
| | - Yu-Ichi Goto
- Department of Child Neurology, National Center Hospital of Neurology and Psychiatry (NCNP), Kodaira, Japan
| | - Seiji Takashima
- Department of Medical Biochemistry, Graduate School of Frontier Biological Science, Osaka University, Suita, Japan.,Japan Science and Technology Agency-Core Research for Evolutional Science and Technology (CREST), Kawaguchi, Japan
| |
Collapse
|
41
|
Gong S, Wang X, Meng F, Cui L, Yi Q, Zhao Q, Cang X, Cai Z, Mo JQ, Liang Y, Guan MX. Overexpression of mitochondrial histidyl-tRNA synthetase restores mitochondrial dysfunction caused by a deafness-associated tRNA His mutation. J Biol Chem 2019; 295:940-954. [PMID: 31819004 DOI: 10.1074/jbc.ra119.010998] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/27/2019] [Indexed: 01/19/2023] Open
Abstract
The deafness-associated m.12201T>C mutation affects the A5-U68 base-pairing within the acceptor stem of mitochondrial tRNAHis The primary defect in this mutation is an alteration in tRNAHis aminoacylation. Here, we further investigate the molecular mechanism of the deafness-associated tRNAHis 12201T>C mutation and test whether the overexpression of the human mitochondrial histidyl-tRNA synthetase gene (HARS2) in cytoplasmic hybrid (cybrid) cells carrying the m.12201T>C mutation reverses mitochondrial dysfunctions. Using molecular dynamics simulations, we demonstrate that the m.12201T>C mutation perturbs the tRNAHis structure and function, supported by decreased melting temperature, conformational changes, and instability of mutated tRNA. We show that the m.12201T>C mutation-induced alteration of aminoacylation tRNAHis causes mitochondrial translational defects and respiratory deficiency. We found that the transfer of HARS2 into the cybrids carrying the m.12201T>C mutation raises the levels of aminoacylated tRNAHis from 56.3 to 75.0% but does not change the aminoacylation of other tRNAs. Strikingly, HARS2 overexpression increased the steady-state levels of tRNAHis and of noncognate tRNAs, including tRNAAla, tRNAGln, tRNAGlu, tRNALeu(UUR), tRNALys, and tRNAMet, in cells bearing the m.12201T>C mutation. This improved tRNA metabolism elevated the efficiency of mitochondrial translation, activities of oxidative phosphorylation complexes, and respiration capacity. Furthermore, HARS2 overexpression markedly increased mitochondrial ATP levels and membrane potential and reduced production of reactive oxygen species in cells carrying the m.12201T>C mutation. These results indicate that HARS2 overexpression corrects the mitochondrial dysfunction caused by the tRNAHis mutation. These findings provide critical insights into the pathophysiology of mitochondrial disease and represent a step toward improved therapeutic interventions for mitochondrial disorders.
Collapse
Affiliation(s)
- Shasha Gong
- Taizhou University Hospital, Taizhou University, Taizhou, Zhejiang 318000, China.,Institute of Genetics, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xiaoqiong Wang
- Department of Otolaryngology, Taizhou Municipal Hospital, Taizhou, Zhejiang 318000, China.,Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Feilong Meng
- Institute of Genetics, Zhejiang University, Hangzhou, Zhejiang 310058, China.,Division of Medical Genetics and Genomics, the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Limei Cui
- Institute of Genetics, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Qiuzi Yi
- Institute of Genetics, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Qiong Zhao
- Institute of Genetics, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xiaohui Cang
- Institute of Genetics, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Zhiyi Cai
- Department of Otolaryngology, Taizhou Municipal Hospital, Taizhou, Zhejiang 318000, China
| | - Jun Qin Mo
- Department of Pathology, Rady Children's Hospital, University of California School of Medicine, San Diego, California 92123
| | - Yong Liang
- Taizhou University Hospital, Taizhou University, Taizhou, Zhejiang 318000, China
| | - Min-Xin Guan
- Institute of Genetics, Zhejiang University, Hangzhou, Zhejiang 310058, China .,Division of Medical Genetics and Genomics, the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.,Key Laboratory of Reproductive Genetics, Ministry of Education of PRC, Zhejiang University, Hangzhou, Zhejiang 310058, China.,Joint Institute of Genetics and Genomic Medicine between Zhejiang University and University of Toronto, Hangzhou, Zhejiang 310058, China
| |
Collapse
|
42
|
Deutschman E, Ward JR, Kumar A, Ray G, Welch N, Lemieux ME, Dasarathy S, Longworth MS. Condensin II protein dysfunction impacts mitochondrial respiration and mitochondrial oxidative stress responses. J Cell Sci 2019; 132:jcs233783. [PMID: 31653782 PMCID: PMC6899004 DOI: 10.1242/jcs.233783] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 10/21/2019] [Indexed: 12/28/2022] Open
Abstract
The maintenance of mitochondrial respiratory function and homeostasis is essential to human health. Here, we identify condensin II subunits as novel regulators of mitochondrial respiration and mitochondrial stress responses. Condensin II is present in the nucleus and cytoplasm. While the effects of condensin II depletion on nuclear genome organization are well studied, the effects on essential cytoplasmic and metabolic processes are not as well understood. Excitingly, we observe that condensin II chromosome-associated protein (CAP) subunits individually localize to different regions of mitochondria, suggesting possible mitochondrial-specific functions independent from those mediated by the canonical condensin II holocomplex. Changes in cellular ATP levels and mitochondrial respiration are observed in condensin II CAP subunit-deficient cells. Surprisingly, we find that loss of NCAPD3 also sensitizes cells to oxidative stress. Together, these studies identify new, and possibly independent, roles for condensin II CAP subunits in preventing mitochondrial damage and dysfunction. These findings reveal a new area of condensin protein research that could contribute to the identification of targets to treat diseases where aberrant function of condensin II proteins is implicated.
Collapse
Affiliation(s)
- Emily Deutschman
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
- Department of Genetics and Genome Sciences, Case Western Reserve University Cleveland, OH 44106, USA
| | - Jacqueline R Ward
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Avinash Kumar
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Greeshma Ray
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Nicole Welch
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | | | - Srinivisan Dasarathy
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Michelle S Longworth
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| |
Collapse
|
43
|
Boggan RM, Lim A, Taylor RW, McFarland R, Pickett SJ. Resolving complexity in mitochondrial disease: Towards precision medicine. Mol Genet Metab 2019; 128:19-29. [PMID: 31648942 DOI: 10.1016/j.ymgme.2019.09.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/12/2019] [Accepted: 09/12/2019] [Indexed: 12/12/2022]
Abstract
Mitochondrial diseases, caused by mutations in either the nuclear or mitochondrial genomes (mtDNA), are the most common form of inherited neurometabolic disorders. They are remarkably heterogeneous, both in their clinical presentation and genetic etiology, presenting challenges for diagnosis, clinical management and elucidation of molecular mechanism. The multifaceted nature of these diseases, compounded by the unique characteristics of mitochondrial genetics, cement their space in the field of complex disease. In this review we examine the m.3243A>G variant, one of the most prevalent mitochondrial DNA mutations, using it as an exemplar to demonstrate the challenges presented by these complex disorders. Disease caused by m.3243A>G is one of the most phenotypically diverse of all mitochondrial diseases; we outline known causes of this heterogeneity including mtDNA heteroplasmy, mtDNA copy number and nuclear genetic factors. We consider the impact that this has in the clinic, discussing the personalized management of common manifestations attributed to this pathogenic mtDNA variant, including hearing impairment, diabetes mellitus, myopathy, cardiac disease, stroke-like episodes and gastrointestinal disturbances. Future research into this complex disorder must account for this heterogeneity, benefitting from the use of large patient cohorts to build upon current clinical expertise. Through multi-disciplinary collaboration, the complexities of this mitochondrial disease can be addressed with the variety of diagnostic, prognostic, and treatment approaches that are moulded to best fit the needs of each individual patient.
Collapse
Affiliation(s)
- Róisín M Boggan
- Wellcome Centre for Mitochondrial Research, Institute of Neuroscience, The Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Albert Lim
- Wellcome Centre for Mitochondrial Research, Institute of Neuroscience, The Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Robert W Taylor
- Wellcome Centre for Mitochondrial Research, Institute of Neuroscience, The Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Robert McFarland
- Wellcome Centre for Mitochondrial Research, Institute of Neuroscience, The Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK.
| | - Sarah J Pickett
- Wellcome Centre for Mitochondrial Research, Institute of Neuroscience, The Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| |
Collapse
|
44
|
Otten ABC, Sallevelt SCEH, Carling PJ, Dreesen JCFM, Drüsedau M, Spierts S, Paulussen ADC, de Die-Smulders CEM, Herbert M, Chinnery PF, Samuels DC, Lindsey P, Smeets HJM. Mutation-specific effects in germline transmission of pathogenic mtDNA variants. Hum Reprod 2019; 33:1331-1341. [PMID: 29850888 DOI: 10.1093/humrep/dey114] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 05/15/2018] [Indexed: 12/31/2022] Open
Abstract
STUDY QUESTION Does germline selection (besides random genetic drift) play a role during the transmission of heteroplasmic pathogenic mitochondrial DNA (mtDNA) mutations in humans? SUMMARY ANSWER We conclude that inheritance of mtDNA is mutation-specific and governed by a combination of random genetic drift and negative and/or positive selection. WHAT IS KNOWN ALREADY mtDNA inherits maternally through a genetic bottleneck, but the underlying mechanisms are largely unknown. Although random genetic drift is recognized as an important mechanism, selection mechanisms are thought to play a role as well. STUDY DESIGN, SIZE, DURATION We determined the mtDNA mutation loads in 160 available oocytes, zygotes, and blastomeres of five carriers of the m.3243A>G mutation, one carrier of the m.8993T>G mutation, and one carrier of the m.14487T>C mutation. PARTICIPANTS/MATERIALS, SETTING, METHODS Mutation loads were determined in PGD samples using PCR assays and analysed mathematically to test for random sampling effects. In addition, a meta-analysis has been performed on mutation load transmission data in the literature to confirm the results of the PGD samples. MAIN RESULTS AND THE ROLE OF CHANCE By applying the Kimura distribution, which assumes random mechanisms, we found that mtDNA segregations patterns could be explained by variable bottleneck sizes among all our carriers (moment estimates ranging from 10 to 145). Marked differences in the bottleneck size would determine the probability that a carrier produces offspring with mutations markedly different than her own. We investigated whether bottleneck sizes might also be influenced by non-random mechanisms. We noted a consistent absence of high mutation loads in all our m.3243A>G carriers, indicating non-random events. To test this, we fitted a standard and a truncated Kimura distribution to the m.3243A>G segregation data. A Kimura distribution truncated at 76.5% heteroplasmy has a significantly better fit (P-value = 0.005) than the standard Kimura distribution. For the m.8993T>G mutation, we suspect a skewed mutation load distribution in the offspring. To test this hypothesis, we performed a meta-analysis on published blood mutation levels of offspring-mother (O-M) transmission for the m.3243A>G and m.8993T>G mutations. This analysis revealed some evidence that the O-M ratios for the m.8993T>G mutation are different from zero (P-value <0.001), while for the m.3243A>G mutation there was little evidence that the O-M ratios are non-zero. Lastly, for the m.14487T>G mutation, where the whole range of mutation loads was represented, we found no indications for selective events during its transmission. LARGE SCALE DATA All data are included in the Results section of this article. LIMITATIONS, REASON FOR CAUTION The availability of human material for the mutations is scarce, requiring additional samples to confirm our findings. WIDER IMPLICATIONS OF THE FINDINGS Our data show that non-random mechanisms are involved during mtDNA segregation. We aimed to provide the mechanisms underlying these selection events. One explanation for selection against high m.3243A>G mutation loads could be, as previously reported, a pronounced oxidative phosphorylation (OXPHOS) deficiency at high mutation loads, which prohibits oogenesis (e.g. progression through meiosis). No maximum mutation loads of the m.8993T>G mutation seem to exist, as the OXPHOS deficiency is less severe, even at levels close to 100%. In contrast, high mutation loads seem to be favoured, probably because they lead to an increased mitochondrial membrane potential (MMP), a hallmark on which healthy mitochondria are being selected. This hypothesis could provide a possible explanation for the skewed segregation pattern observed. Our findings are corroborated by the segregation pattern of the m.14487T>C mutation, which does not affect OXPHOS and MMP significantly, and its transmission is therefore predominantly determined by random genetic drift. Our conclusion is that mutation-specific selection mechanisms occur during mtDNA inheritance, which has implications for PGD and mitochondrial replacement therapy. STUDY FUNDING/COMPETING INTEREST(S) This work has been funded by GROW-School of Oncology and Developmental Biology. The authors declare no competing interests.
Collapse
Affiliation(s)
- Auke B C Otten
- Department of Genetics and Cell Biology, School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, the Netherlands
| | - Suzanne C E H Sallevelt
- Department of Clinical Genetics, Maastricht University Medical Centre+ (MUMC+), Maastricht, the Netherlands
| | - Phillippa J Carling
- Department of Neuroscience, Sheffield institute for translational neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Joseph C F M Dreesen
- Department of Clinical Genetics, Maastricht University Medical Centre+ (MUMC+), Maastricht, the Netherlands
| | - Marion Drüsedau
- Department of Clinical Genetics, Maastricht University Medical Centre+ (MUMC+), Maastricht, the Netherlands
| | - Sabine Spierts
- Department of Clinical Genetics, Maastricht University Medical Centre+ (MUMC+), Maastricht, the Netherlands
| | - Aimee D C Paulussen
- Department of Clinical Genetics, Maastricht University Medical Centre+ (MUMC+), Maastricht, the Netherlands
| | | | - Mary Herbert
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Patrick F Chinnery
- Department of Clinical Neuroscience, School of Clinical Medicine, University of Cambridge, Cambridge, UK.,Medical Research Council Mitochondrial Biology Unit, Cambridge, Biomedical Campus, Cambridge, UK
| | - David C Samuels
- Department of Molecular Physiology and Biophysics, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Patrick Lindsey
- Department of Genetics and Cell Biology, School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, the Netherlands
| | - Hubert J M Smeets
- Department of Genetics and Cell Biology, School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
45
|
Richter U, Ng KY, Suomi F, Marttinen P, Turunen T, Jackson C, Suomalainen A, Vihinen H, Jokitalo E, Nyman TA, Isokallio MA, Stewart JB, Mancini C, Brusco A, Seneca S, Lombès A, Taylor RW, Battersby BJ. Mitochondrial stress response triggered by defects in protein synthesis quality control. Life Sci Alliance 2019; 2:2/1/e201800219. [PMID: 30683687 PMCID: PMC6348486 DOI: 10.26508/lsa.201800219] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 01/16/2019] [Accepted: 01/17/2019] [Indexed: 12/11/2022] Open
Abstract
Quality control defects of mitochondrial nascent chain synthesis trigger a sequential stress response characterized by OMA1 activation and ribosome decay, determining mitochondrial form and function. Mitochondria have a compartmentalized gene expression system dedicated to the synthesis of membrane proteins essential for oxidative phosphorylation. Responsive quality control mechanisms are needed to ensure that aberrant protein synthesis does not disrupt mitochondrial function. Pathogenic mutations that impede the function of the mitochondrial matrix quality control protease complex composed of AFG3L2 and paraplegin cause a multifaceted clinical syndrome. At the cell and molecular level, defects to this quality control complex are defined by impairment to mitochondrial form and function. Here, we establish the etiology of these phenotypes. We show how disruptions to the quality control of mitochondrial protein synthesis trigger a sequential stress response characterized first by OMA1 activation followed by loss of mitochondrial ribosomes and by remodelling of mitochondrial inner membrane ultrastructure. Inhibiting mitochondrial protein synthesis with chloramphenicol completely blocks this stress response. Together, our data establish a mechanism linking major cell biological phenotypes of AFG3L2 pathogenesis and show how modulation of mitochondrial protein synthesis can exert a beneficial effect on organelle homeostasis.
Collapse
Affiliation(s)
- Uwe Richter
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Kah Ying Ng
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Fumi Suomi
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Paula Marttinen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Taina Turunen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Christopher Jackson
- Research Programs Unit-Molecular Neurology, University of Helsinki, Helsinki, Finland
| | - Anu Suomalainen
- Research Programs Unit-Molecular Neurology, University of Helsinki, Helsinki, Finland
| | - Helena Vihinen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Eija Jokitalo
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Tuula A Nyman
- Department of Immunology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | | | - James B Stewart
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Cecilia Mancini
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Alfredo Brusco
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Sara Seneca
- Center for Medical Genetics/Research Center Reproduction and Genetics, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Anne Lombès
- Faculté de médecine Cochin, Institut Cochin Institut national de la santé et de la recherche médicale U1016, Centre national de la recherche scientifique Unités Mixtes de Recherche 8104, Université Paris 5, Paris, France
| | - Robert W Taylor
- Wellcome Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | | |
Collapse
|
46
|
Oxidative Insults and Mitochondrial DNA Mutation Promote Enhanced Autophagy and Mitophagy Compromising Cell Viability in Pluripotent Cell Model of Mitochondrial Disease. Cells 2019; 8:cells8010065. [PMID: 30658448 PMCID: PMC6356288 DOI: 10.3390/cells8010065] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/11/2019] [Accepted: 01/15/2019] [Indexed: 12/12/2022] Open
Abstract
Dysfunction of mitochondria causes defects in oxidative phosphorylation system (OXPHOS) and increased production of reactive oxygen species (ROS) triggering the activation of the cell death pathway that underlies the pathogenesis of aging and various diseases. The process of autophagy to degrade damaged cytoplasmic components as well as dysfunctional mitochondria is essential for ensuring cell survival. We analyzed the role of autophagy inpatient-specific induced pluripotent stem (iPS) cells generated from fibroblasts of patients with mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS) with well-characterized mitochondrial DNA mutations and distinct OXPHOS defects. MELAS iPS cells recapitulated the pathogenesis of MELAS syndrome, and showed an increase of autophagy in comparison with its isogenic normal counterpart, whereas mitophagy is very scarce at the basal condition. Our results indicated that the existence of pathogenic mtDNA alone in mitochondrial disease was not sufficient to elicit the degradation of dysfunctional mitochondria. Nonetheless, oxidative insults induced bulk macroautophagy with the accumulation of autophagosomes and autolysosomes upon marked elevation of ROS, overload of intracellular calcium, and robust depolarization of mitochondrial membrane potential, while mitochondria respiratory function was impaired and widespread mitophagy compromised cell viability. Collectively, our studies provide insights into the dysfunction of autophagy and activation of mitophagy contributing to the pathological mechanism of mitochondrial disease.
Collapse
|
47
|
RNA modification landscape of the human mitochondrial tRNA Lys regulates protein synthesis. Nat Commun 2018; 9:3966. [PMID: 30262910 PMCID: PMC6160436 DOI: 10.1038/s41467-018-06471-z] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 09/06/2018] [Indexed: 01/15/2023] Open
Abstract
Post-transcriptional RNA modifications play a critical role in the pathogenesis of human mitochondrial disorders, but the mechanisms by which specific modifications affect mitochondrial protein synthesis remain poorly understood. Here we used a quantitative RNA sequencing approach to investigate, at nucleotide resolution, the stoichiometry and methyl modifications of the entire mitochondrial tRNA pool, and establish the relevance to human disease. We discovered that a N1-methyladenosine (m1A) modification is missing at position 58 in the mitochondrial tRNALys of patients with the mitochondrial DNA mutation m.8344 A > G associated with MERRF (myoclonus epilepsy, ragged-red fibers). By restoring the modification on the mitochondrial tRNALys, we demonstrated the importance of the m1A58 to translation elongation and the stability of selected nascent chains. Our data indicates regulation of post-transcriptional modifications on mitochondrial tRNAs is finely tuned for the control of mitochondrial gene expression. Collectively, our findings provide novel insight into the regulation of mitochondrial tRNAs and reveal greater complexity to the molecular pathogenesis of MERRF.
Collapse
|
48
|
Tranah GJ, Katzman SM, Lauterjung K, Yaffe K, Manini TM, Kritchevsky S, Newman AB, Harris TB, Cummings SR. Mitochondrial DNA m.3243A > G heteroplasmy affects multiple aging phenotypes and risk of mortality. Sci Rep 2018; 8:11887. [PMID: 30089816 PMCID: PMC6082898 DOI: 10.1038/s41598-018-30255-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 07/20/2018] [Indexed: 12/13/2022] Open
Abstract
Mitochondria contain many copies of a circular DNA molecule (mtDNA), which has been observed as a mixture of normal and mutated states known as heteroplasmy. Elevated heteroplasmy at a single mtDNA site, m.3243A > G, leads to neurologic, sensory, movement, metabolic, and cardiopulmonary impairments. We measured leukocyte mtDNA m.3243A > G heteroplasmy in 789 elderly men and women from the bi-racial, population-based Health, Aging, and Body Composition Study to identify associations with age-related functioning and mortality. Mutation burden for the m.3243A > G ranged from 0-19% and elevated heteroplasmy was associated with reduced strength, cognitive, metabolic, and cardiovascular functioning. Risk of all-cause, dementia and stroke mortality was significantly elevated for participants in the highest tertiles of m.3243A > G heteroplasmy. These results indicate that the accumulation of a rare genetic disease mutation, m.3243A > G, manifests as several aging outcomes and that some diseases of aging may be attributed to the accumulation of mtDNA damage.
Collapse
Grants
- R01-NR012459 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- R01 HL121023 NHLBI NIH HHS
- N01-AG-6-2106 U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- R01-HL121023 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P30 AG028740 NIA NIH HHS
- Z01A6000932 Office of Extramural Research, National Institutes of Health (OER)
- R03-AG032498 U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- R01-AG028050 U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- R01 NR012459 NINR NIH HHS
- R03 AG032498 NIA NIH HHS
- R01 AG028050 NIA NIH HHS
- This research was supported in part by the Intramural Research Program of the NIH, National Institute on Aging, Contracts N01-AG-6-2101, N01-AG-6-2103, and N01-AG-6-2106; National Institutes of Health grants R01-AG028050, R03-AG032498, R01-NR012459, Z01A6000932, R01-HL121023, and a grant from the Research and Education Leadership Committee of the CPMC Foundation and the L. K. Whittier Foundation.
- U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- U.S. Department of Health & Human Services | National Institutes of Health (NIH)
Collapse
Affiliation(s)
- Gregory J Tranah
- California Pacific Medical Center Research Institute, San Francisco, CA, 94107, USA.
| | | | - Kevin Lauterjung
- California Pacific Medical Center Research Institute, San Francisco, CA, 94107, USA
| | - Kristine Yaffe
- Departments of Psychiatry, Neurology, and Epidemiology, University of California, San Francisco and the San Francisco VA Medical Center, San Francisco, CA, 94121, USA
| | - Todd M Manini
- Department of Aging and Geriatric Research, University of Florida, Gainesville, FL, 32601, USA
| | - Stephen Kritchevsky
- Sticht Center on Aging, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Anne B Newman
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Tamara B Harris
- Intramural Research Program, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Bethesda, MD, 20892, USA
| | - Steven R Cummings
- California Pacific Medical Center Research Institute, San Francisco, CA, 94107, USA
| |
Collapse
|
49
|
Zhu P, Liu Y, Zhang F, Bai X, Chen Z, Shangguan F, Zhang B, Zhang L, Chen Q, Xie D, Lan L, Xue X, Liang XJ, Lu B, Wei T, Qin Y. Human Elongation Factor 4 Regulates Cancer Bioenergetics by Acting as a Mitochondrial Translation Switch. Cancer Res 2018; 78:2813-2824. [PMID: 29572227 DOI: 10.1158/0008-5472.can-17-2059] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 01/01/2018] [Accepted: 03/20/2018] [Indexed: 11/16/2022]
Abstract
Mitochondria regulate cellular bioenergetics and redox states and influence multiple signaling pathways required for tumorigenesis. In this study, we determined that the mitochondrial translation elongation factor 4 (EF4) is a critical component of tumor progression. EF4 was ubiquitous in human tissues with localization to the mitochondria (mtEF4) and performed quality control on respiratory chain biogenesis. Knockout of mtEF4 induced respiratory chain complex defects and apoptosis, while its overexpression stimulated cancer development. In multiple cancers, expression of mtEF4 was increased in patient tumor tissues. These findings reveal that mtEF4 expression may promote tumorigenesis via an imbalance in the regulation of mitochondrial activities and subsequent variation of cellular redox. Thus, dysregulated mitochondrial translation may play a vital role in the etiology and development of diverse human cancers.Significance: Dysregulated mitochondrial translation drives tumor development and progression. Cancer Res; 78(11); 2813-24. ©2018 AACR.
Collapse
Affiliation(s)
- Ping Zhu
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang District, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yongzhang Liu
- Attardi Institute of Mitochondrial Biomedicine, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Fenglin Zhang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang District, Beijing, China
| | - Xiufeng Bai
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang District, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zilei Chen
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang District, Beijing, China.,Attardi Institute of Mitochondrial Biomedicine, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Fugen Shangguan
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang District, Beijing, China.,Attardi Institute of Mitochondrial Biomedicine, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Bo Zhang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang District, Beijing, China
| | - Lingyun Zhang
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang District, Beijing, China
| | - Qianqian Chen
- University of Chinese Academy of Sciences, Beijing, China.,National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang District, Beijing, China
| | - Deyao Xie
- Departments of Cardiothoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Linhua Lan
- Attardi Institute of Mitochondrial Biomedicine, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiangdong Xue
- Laboratory of Controllable Nanopharmaceuticals, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Zhongguancun, Beijing, China
| | - Xing-Jie Liang
- Laboratory of Controllable Nanopharmaceuticals, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Zhongguancun, Beijing, China
| | - Bin Lu
- Attardi Institute of Mitochondrial Biomedicine, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Taotao Wei
- University of Chinese Academy of Sciences, Beijing, China. .,National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang District, Beijing, China
| | - Yan Qin
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Chaoyang District, Beijing, China. .,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
50
|
Kandel J, Picard M, Wallace DC, Eckmann DM. Mitochondrial DNA 3243A>G heteroplasmy is associated with changes in cytoskeletal protein expression and cell mechanics. J R Soc Interface 2018; 14:rsif.2017.0071. [PMID: 28592659 DOI: 10.1098/rsif.2017.0071] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 05/09/2017] [Indexed: 12/21/2022] Open
Abstract
Mitochondrial and mechanical alterations in cells have both been shown to be hallmarks of human disease. However, little research has endeavoured to establish connections between these two essential features of cells in both functional and dysfunctional situations. In this work, we hypothesized that a specific genetic alteration in mitochondrial function known to cause human disease would trigger changes in cell mechanics. Using a previously characterized set of mitochondrial cybrid cell lines, we examined the relationship between heteroplasmy for the mitochondrial DNA (mtDNA) 3243A>G mutation, the cell cytoskeleton, and resulting cellular mechanical properties. We found that cells with increasing mitochondrial dysfunction markedly differed from one another in gene expression and protein production of various co-regulated cytoskeletal elements. The intracellular positioning and organization of actin also differed across cell lines. To explore the relationship between these changes and cell mechanics, we then measured cellular mechanical properties using atomic force microscopy and found that cell stiffness correlated with gene expression data for known determinants of cell mechanics, γ-actin, α-actinin and filamin A. This work points towards a mechanism linking mitochondrial genetics to single-cell mechanical properties. The transcriptional and structural regulation of cytoskeletal components by mitochondrial function may explain why energetic and mechanical alterations often coexist in clinical conditions.
Collapse
Affiliation(s)
- Judith Kandel
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Martin Picard
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Douglas C Wallace
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David M Eckmann
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA .,Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|