Chchd10, a Novel Bi-Organellar Regulator of Cellular Metabolism: Implications in Neurodegeneration

Total Page:16

File Type:pdf, Size:1020Kb

Chchd10, a Novel Bi-Organellar Regulator of Cellular Metabolism: Implications in Neurodegeneration Wayne State University Wayne State University Dissertations January 2018 Chchd10, A Novel Bi-Organellar Regulator Of Cellular Metabolism: Implications In Neurodegeneration Neeraja Purandare Wayne State University, [email protected] Follow this and additional works at: https://digitalcommons.wayne.edu/oa_dissertations Part of the Molecular Biology Commons Recommended Citation Purandare, Neeraja, "Chchd10, A Novel Bi-Organellar Regulator Of Cellular Metabolism: Implications In Neurodegeneration" (2018). Wayne State University Dissertations. 2125. https://digitalcommons.wayne.edu/oa_dissertations/2125 This Open Access Dissertation is brought to you for free and open access by DigitalCommons@WayneState. It has been accepted for inclusion in Wayne State University Dissertations by an authorized administrator of DigitalCommons@WayneState. CHCHD10, A NOVEL BI-ORGANELLAR REGULATOR OF CELLULAR METABOLISM: IMPLICATIONS IN NEURODEGENERATION by NEERAJA PURANDARE DISSERTATION Submitted to the Graduate School of Wayne State University, Detroit, Michigan in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY 2018 MAJOR: MOLECULAR BIOLOGY AND GENETICS Approved By: Advisor Date © COPYRIGHT BY NEERAJA PURANDARE 2018 All Rights Reserved ACKNOWLEDGEMENTS First, I would I like to express the deepest gratitude to my mentor Dr. Grossman for the advice and support and most importantly your patience. Your calm and collected approach during our discussions provided me much needed perspective towards prioritizing and planning my work and I hope to carry this composure in my future endeavors. Words cannot describe my gratefulness for the support of Dr. Siddhesh Aras. You epitomize the scientific mind. I hope that I have inculcated a small fraction of your scientific thought process and I will carry this forth not just in my career, but for everything else that I do. None of this would have been possible without your guidance and constant encouragement. I would also like to thank my graduate committee members - Dr. Russell Finley, Dr. Kezhong Zhang, and Dr. Miriram Greenberg for their insightful questions, constructive criticism, and valuable advice. I would like to acknowledge my lab members Stephanie Gladyck, Marissa Petitpas and Mohsen Mohktari for all their help and support. A special thank you to Dr. Mallika Somayajulu- Nitu for her advice and help with imaging and analysis of the microscopy data for this work. I would also like to thank Dr. Maik Hüttemann and members of his lab - Jenney Liu, Asmita Vaishnav, and Hasini Kalpage for all the help and advise both in the lab and during our shared lab meetings. I would also like to express my gratitude for the help from the support personnel of the Center for Molecular Genetics and Genomics for their help and advice. Lastly, but not the least, I would like to thank my friends and family members. I have been blessed with two sets of parents. My parents Mr. Aniruddha Purandare and Mrs. Tejaswini Purandare have always been supportive of my career. But this journey would not have been possible without my uncle; Mr. Ashutosh Kale, and my aunt; Mrs. Kalyani Kale, in the US. I consider myself fortunate indeed to be your third child. Thanks to your constant encouragement and understanding, I have never felt like I was away from home. ii TABLE OF CONTENTS Acknowledgements ......................................................................................................... ii List of Figures ....…….….………………..……….……………..............……....................... vii List of Tables ..…......….........….….…….…………………….……………........................... ix List of AbbreviAtions ...….................……………................……….….…….….................. x ChApter I: Introduction ............…...……………..............….………….....………............... 1 1. The structure and origin of mitochondria ….....………….……………......…................ 3 2. Mitochondria and their role in cellular physiology …….………..…….......................... 3 2.1. Energy production ………….…….………..........................………….................... 3 2.2. Apoptosis ……….……………...……….…......……...….…….……………...….….. 4 2.3. Generation of reactive oxygen species (ROS) ……….........…….………............ 5 2.4. Calcium homeostasis .......……….….…………......……….........…....................... 5 2.5. Lipid homeostasis ......….................……...........………...…………......…............. 6 2.6. Iron homeostasis ……....……….…..…...…….....……….………........................... 7 3. Electron Transport Chain .…….......…………..…....………....…….............................. 7 3.1. Complex I ….………….......………...…..…….…...……......................................... 8 3.2. Complex II ….………...………...……........….….....……….................................... 8 3.3. Complex III …..………………...............…….......…...………….......….................. 9 3.4. Complex V (FOF1 ATP synthase) ........………...…...…………...…….….…........... 9 4. Complex IV (Cytochrome c oxidase) ….……………...................................…....……. 10 5. Twin CX9C Proteins ….………....……..……...…......….……..........................….......... 12 6. Orthologs for CHCHD10 and MNRR1 ………......…………...………........................... 16 7. Similarities and differences between CHCHD10 and MNRR1 ................................... 18 8. MNRR1 ….…….……………........….……….......…...……........................................... 23 8.1. MNRR1 and its role in the nucleus and mitochondria …….….…...…................... 23 iii 8.2. MNRR1 and disease ..……….………....……....……...….....…............................ 28 9. CHCHD10 ………...………….…………..............……...............………....................... 32 9.1. CHCHD10’s role in the nucleus and mitochondria ...………...……..................... 32 9.2. CHCHD10 and its role in disease ……….………………..................................... 33 ChApter II: Results ……...…………………………...………....…...……............................. 41 1. Preliminary characterization of CHCHD10 localization and function ………..…........ 41 1.1. CHCHD10 is localized to the nucleus and the mitochondria ……….....……....... 41 1.2. CHCHD10 is a hypoxia-sensitive gene …...................………............................. 42 1.3. Knockdown of CHCHD10 has pleiotropic effects in cells ………............…......... 44 2. CHCHD10 regulates transcription in the nucleus …….........…………..........….......... 45 2.1. CHCHD10 functions as a repressor at the oxygen responsive element (ORE) ____ in the nucleus ...……………….………….............…….......................................……. 45 2.2. CHCHD10 functions as a repressor by interacting with the inhibitory CXXC5 at ___ the ORE ….…………...…….…………....…….………................................................. 46 3. CHCHD10’s regulates oxygen consumption in the mitochondria .............................. 48 3.1. CHCHD10 interacts with COX ............................................................................ 48 3.2. CHCHD10 stimulates oxygen consumption in the mitochondria …........……...... 50 3.3. Defective mitochondrial oxygen consumption in CHCHD10-KD cells arises via ___ defective phosphorylation of MNRR1 ....……………...……….…….....…..…............... 50 4. CHCHD10’s effects in the nucleus and mitochondria under stress .…....….............….. 54 4.1. CHCHD10 function is enhanced at 8% hypoxia ……………............................... 54 4.2. Point mutations in CHCHD10 abrogate CHCHD10’s function in the nucleus ___ 56 adds and mitochondria ...................….………….…….....…….……….…........……............. 4.2.1. Point mutations in CHCHD10 fail to repress ORE-mediated transcription _____ the nucleus ………...……….………....….………………...…...………..................... 57 4.2.2. Point mutations in CHCHD10 are defective in maintaining optimal ETC ______ in function in the mitochondria ……...…....….…........................………................. 58 iv ChApter III: Discussion …………..…...…......................………...……….......................... 62 1. CHCHD10 and MNRR1’s effects in the mitochondria ….……………..……................. 63 2. CHCHD10 and MNRR1’s effects in the nucleus …………………….....….…............... 65 3. The hypoxia sensitivity of MNRR1 and CHCHD10 ……………........….……............... 68 4. The mechanism of mitochondrial dysfunction for CHCHD10 mutations .....…..…....... 70 5. Summary ……...........................………………………………...................................... 72 6. Future Directions ……….…….…….…………….…………...…..…….......................... 73 ChApter IV: MAteriAls And Methods …………..…….....……....….…………….................. 76 1. Cell culture ……….……….…………….......…….….........…....…………..................... 76 2. Effector and reporter plasmids …….……………......……….….....…………............... 77 3. Transient transfection of HEK293 cells ……….….………...............…………............ 77 4. Real-time polymerase chain reaction ……...….….........……………….………........... 77 5. Hypoxia assays …............……….……….…...………....…………...……..................... 78 6. Luciferase reporter assays ………........…………….......…………...………................. 78 7. DNA binding assays ……..................…....……………….………...………................... 78 8. Cell proliferation assay ……...……...……........….……….……...………...................... 79 9. Cell counting assay …...………........……….…...…......…....…………..…................... 79 10. Intact cellular oxygen consumption ……….………..........…….…...………................ 79 11. Cytochrome c oxidase assay ……….…..….……......…..……………...….................. 79 12. ROS measurement …......………….......…………...……….…...…………….................. 79 13. Confocal microscopy .…………….........…………...……….…...………...…..................
Recommended publications
  • Tomo-Seq Identifies SOX9 As a Key Regulator of Cardiac Fibrosis During Ischemic Injury
    myocardial myocardial Eva van ◼ osis fibr SOX9 transcription ◼ PhD* PhD PhD PhD MSc, PhD naarden, MSc, PhD naarden, PhD* ventricular remodeling Correspondence to: Correspondence Rooij, MSc, PhD, Hubrecht Department of Institute, KNAW University Medical Cardiology, Uppsalalaan 8, Center Utrecht, The Netherlands. 3584CT Utrecht, E-mail [email protected] of Funding, see page 1408 Sources Key Words: ischemia ◼ © 2017 American Heart Association, Inc. *Drs. Lacraz and Junker contributed equally. Grégory P.A. Lacraz, MSc, P.A. Grégory MSc, Jan Philipp Junker, Monika M. Gladka, MSc, MSc Bas Molenaar, Scholman, MSc Koen T. MSc Marta Vigil-Garcia, BS Danielle Versteeg, BS Hesther de Ruiter, MSc, Vermunt, Marit W. MSc, Creyghton, Menno P. Manon M.H. Huibers, Nicolaas de Jonge, MD Alexander van Oude- Eva van Rooij, MSc, PhD 2017;136:1396–1409. DOI: 10.1161/CIRCULATIONAHA.117.027832 DOI: 2017;136:1396–1409. Circulation. blunted the cardiac fibrotic fibrotic blunted the cardiac Sox9 ). Subsequent correlation analysis allowed). Subsequent correlation Serca2 Editorial, see p 1410 , and Nppa Based on the exact local expression cues, tomo-seq can Based on the exact local expression Cardiac ischemic injury induces a pathological remodeling ischemic injury induces a pathological remodeling Cardiac , Although genome-wide transcriptome analysis on diseased tissues Tracing transcriptional differences with a high spatial resolution with a high spatial resolution transcriptional differences Tracing Col1a2 October 10, 2017 October 10, 1396 CONCLUSIONS: novel genes and key transcription factors involved in specific serve to reveal able to unveil the Using tomo-seq, we were remodeling. aspects of cardiac pointing fibrosis, of cardiac of SOX9 as a key regulator unknown relevance fibrosis.
    [Show full text]
  • Neurodevelopmental Signatures of Narcotic and Neuropsychiatric Risk Factors in 3D Human-Derived Forebrain Organoids
    Molecular Psychiatry www.nature.com/mp ARTICLE OPEN Neurodevelopmental signatures of narcotic and neuropsychiatric risk factors in 3D human-derived forebrain organoids 1 1 1 1 2 2 3 Michael Notaras , Aiman Lodhi , Estibaliz✉ Barrio-Alonso , Careen Foord , Tori Rodrick , Drew Jones , Haoyun Fang , David Greening 3,4 and Dilek Colak 1,5 © The Author(s) 2021 It is widely accepted that narcotic use during pregnancy and specific environmental factors (e.g., maternal immune activation and chronic stress) may increase risk of neuropsychiatric illness in offspring. However, little progress has been made in defining human- specific in utero neurodevelopmental pathology due to ethical and technical challenges associated with accessing human prenatal brain tissue. Here we utilized human induced pluripotent stem cells (hiPSCs) to generate reproducible organoids that recapitulate dorsal forebrain development including early corticogenesis. We systemically exposed organoid samples to chemically defined “enviromimetic” compounds to examine the developmental effects of various narcotic and neuropsychiatric-related risk factors within tissue of human origin. In tandem experiments conducted in parallel, we modeled exposure to opiates (μ-opioid agonist endomorphin), cannabinoids (WIN 55,212-2), alcohol (ethanol), smoking (nicotine), chronic stress (human cortisol), and maternal immune activation (human Interleukin-17a; IL17a). Human-derived dorsal forebrain organoids were consequently analyzed via an array of unbiased and high-throughput analytical approaches, including state-of-the-art TMT-16plex liquid chromatography/mass- spectrometry (LC/MS) proteomics, hybrid MS metabolomics, and flow cytometry panels to determine cell-cycle dynamics and rates of cell death. This pipeline subsequently revealed both common and unique proteome, reactome, and metabolome alterations as a consequence of enviromimetic modeling of narcotic use and neuropsychiatric-related risk factors in tissue of human origin.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Whole Exome Should Be Preferred Over Sanger Sequencing in Suspected Mitochondrial Myopathy
    Neurobiology of Aging 78 (2019) 166e167 Contents lists available at ScienceDirect Neurobiology of Aging journal homepage: www.elsevier.com/locate/neuaging Letter to the editor Whole exome should be preferred over Sanger sequencing in suspected mitochondrial myopathy With interest we read the article by Rubino et al. about Sanger X-linked trait of inheritance, whole exome sequencing rather than sequencing of the genes CHCHD2 and CHCHD10 in 62 Italian pa- Sanger sequencing of single genes is recommended to detect the tients with a mitochondrial myopathy without a genetic defect underlying genetic defect. In case of a maternal trait of inheritance, (Rubino et al., 2018). The authors found the previously reported however, sequencing of the mtDNA is recommended. Whole exome variant c.307C>A in the CHCHD10 gene (Perrone et al., 2017)in1of sequencing is preferred over Sanger sequencing as myopathies or the 62 patients (Rubino et al., 2018). We have the following com- phenotypes in general that resemble an MID are in fact due to ments and concerns. mutations in genes not involved in mitochondrial functions, rep- If no mutation was found in 61 of the 62 included myopathy resenting genotypic heterogeneity. patients, how can the authors be sure that these patients had We do not agree that application of SIFT and polyphem 2 is indeed a mitochondrial disorder (MID). We should be informed on sufficient to confirm pathogenicity of a variant. Confirmation of the which criteria and by which means the diagnosis of an MID was pathogenicity requires documentation of the variant in other established in the 61 patients, who did not carry a mutation in the populations, segregation of the phenotype with the genotype CHCHD2 and CHCHD10 genes, respectively.
    [Show full text]
  • Supplementary Materials
    Supplementary materials Supplementary Table S1: MGNC compound library Ingredien Molecule Caco- Mol ID MW AlogP OB (%) BBB DL FASA- HL t Name Name 2 shengdi MOL012254 campesterol 400.8 7.63 37.58 1.34 0.98 0.7 0.21 20.2 shengdi MOL000519 coniferin 314.4 3.16 31.11 0.42 -0.2 0.3 0.27 74.6 beta- shengdi MOL000359 414.8 8.08 36.91 1.32 0.99 0.8 0.23 20.2 sitosterol pachymic shengdi MOL000289 528.9 6.54 33.63 0.1 -0.6 0.8 0 9.27 acid Poricoic acid shengdi MOL000291 484.7 5.64 30.52 -0.08 -0.9 0.8 0 8.67 B Chrysanthem shengdi MOL004492 585 8.24 38.72 0.51 -1 0.6 0.3 17.5 axanthin 20- shengdi MOL011455 Hexadecano 418.6 1.91 32.7 -0.24 -0.4 0.7 0.29 104 ylingenol huanglian MOL001454 berberine 336.4 3.45 36.86 1.24 0.57 0.8 0.19 6.57 huanglian MOL013352 Obacunone 454.6 2.68 43.29 0.01 -0.4 0.8 0.31 -13 huanglian MOL002894 berberrubine 322.4 3.2 35.74 1.07 0.17 0.7 0.24 6.46 huanglian MOL002897 epiberberine 336.4 3.45 43.09 1.17 0.4 0.8 0.19 6.1 huanglian MOL002903 (R)-Canadine 339.4 3.4 55.37 1.04 0.57 0.8 0.2 6.41 huanglian MOL002904 Berlambine 351.4 2.49 36.68 0.97 0.17 0.8 0.28 7.33 Corchorosid huanglian MOL002907 404.6 1.34 105 -0.91 -1.3 0.8 0.29 6.68 e A_qt Magnogrand huanglian MOL000622 266.4 1.18 63.71 0.02 -0.2 0.2 0.3 3.17 iolide huanglian MOL000762 Palmidin A 510.5 4.52 35.36 -0.38 -1.5 0.7 0.39 33.2 huanglian MOL000785 palmatine 352.4 3.65 64.6 1.33 0.37 0.7 0.13 2.25 huanglian MOL000098 quercetin 302.3 1.5 46.43 0.05 -0.8 0.3 0.38 14.4 huanglian MOL001458 coptisine 320.3 3.25 30.67 1.21 0.32 0.9 0.26 9.33 huanglian MOL002668 Worenine
    [Show full text]
  • © 2019 Jan C. Lumibao
    © 2019 Jan C. Lumibao CHCHD2 AND THE TUMOR MICROENVIRONMENT IN GLIOBLASTOMA BY JAN C. LUMIBAO DISSERTATION Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in Nutritional Sciences in the Graduate College of the University of Illinois at Urbana-Champaign, 2019 Urbana, Illinois Doctoral Committee: Professor Brendan A. Harley, Chair Professor H. Rex Gaskins, Director of Research Assistant Professor Andrew J. Steelman Professor Rodney W. Johnson Professor Emeritus John W. Erdman ABSTRACT Glioblastoma (GBM) is the most common, aggressive, and deadly form of primary brain tumor in adults, with a median survival time of only 14.6 months. GBM tumors present with chemo- and radio-resistance and rapid, diffuse invasion, making complete surgical resection impossible and resulting in nearly universal recurrence. While investigating the genomic landscape of GBM tumors has expanded understanding of brain tumor biology, targeted therapies against cellular pathways affected by the most common genetic aberrations have been largely ineffective at producing robust survival benefits. Currently, a major obstacle to more effective therapies is the impact of the surrounding tumor microenvironment on intracellular signaling, which has the potential to undermine targeted treatments and advance tumor malignancy, progression, and resistance to therapy. Additionally, mitochondria, generally regarded as putative energy sensors within cells, also play a central role as signaling organelles. Retrograde signaling occurring from
    [Show full text]
  • Molecular Analyses of Malignant Pleural Mesothelioma
    Molecular Analyses of Malignant Pleural Mesothelioma Shir Kiong Lo National Heart and Lung Institute Imperial College Dovehouse Street London SW3 6LY A thesis submitted for MD (Res) Faculty of Medicine, Imperial College London 2016 1 Abstract Malignant pleural mesothelioma (MPM) is an aggressive cancer that is strongly associated with asbestos exposure. Majority of patients with MPM present with advanced disease and the treatment paradigm mainly involves palliative chemotherapy and best supportive care. The current chemotherapy options are limited and ineffective hence there is an urgent need to improve patient outcomes. This requires better understanding of the genetic alterations driving MPM to improve diagnostic, prognostic and therapeutic strategies. This research aims to gain further insights in the pathogenesis of MPM by exploring the tumour transcriptional and mutational profiles. We compared gene expression profiles of 25 MPM tumours and 5 non-malignant pleura. This revealed differentially expressed genes involved in cell migration, invasion, cell cycle and the immune system that contribute to the malignant phenotype of MPM. We then constructed MPM-associated co-expression networks using weighted gene correlation network analysis to identify clusters of highly correlated genes. These identified three distinct molecular subtypes of MPM associated with genes involved in WNT and TGF-ß signalling pathways. Our results also revealed genes involved in cell cycle control especially the mitotic phase correlated significantly with poor prognosis. Through exome analysis of seven paired tumour/blood and 29 tumour samples, we identified frequent mutations in BAP1 and NF2. Additionally, the mutational profile of MPM is enriched with genes encoding FAK, MAPK and WNT signalling pathways.
    [Show full text]
  • Human CLPB) Is a Potent Mitochondrial Protein Disaggregase That Is Inactivated By
    bioRxiv preprint doi: https://doi.org/10.1101/2020.01.17.911016; this version posted January 18, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Skd3 (human CLPB) is a potent mitochondrial protein disaggregase that is inactivated by 3-methylglutaconic aciduria-linked mutations Ryan R. Cupo1,2 and James Shorter1,2* 1Department of Biochemistry and Biophysics, 2Pharmacology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, U.S.A. *Correspondence: [email protected] 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.01.17.911016; this version posted January 18, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. ABSTRACT Cells have evolved specialized protein disaggregases to reverse toxic protein aggregation and restore protein functionality. In nonmetazoan eukaryotes, the AAA+ disaggregase Hsp78 resolubilizes and reactivates proteins in mitochondria. Curiously, metazoa lack Hsp78. Hence, whether metazoan mitochondria reactivate aggregated proteins is unknown. Here, we establish that a mitochondrial AAA+ protein, Skd3 (human CLPB), couples ATP hydrolysis to protein disaggregation and reactivation. The Skd3 ankyrin-repeat domain combines with conserved AAA+ elements to enable stand-alone disaggregase activity. A mitochondrial inner-membrane protease, PARL, removes an autoinhibitory peptide from Skd3 to greatly enhance disaggregase activity. Indeed, PARL-activated Skd3 dissolves α-synuclein fibrils connected to Parkinson’s disease. Human cells lacking Skd3 exhibit reduced solubility of various mitochondrial proteins, including anti-apoptotic Hax1.
    [Show full text]
  • Mitochondrial Medicine in the Omics Era
    Mitochondrial Medicine in the Omics Era Joyeeta Rahman1 and Shamima Rahman1,2* 1 Mitochondrial Research Group, UCL Great Ormond Street Institute of Child Health and 2 Metabolic Unit, Great Ormond Street Hospital NHS Foundation Trust, London, UK *Correspondence to: Professor Shamima Rahman Mitochondrial Research Group Genetics and Genomic Medicine UCL Great Ormond Street Institute of Child Health London WC1N 1EH, UK. Telephone: +44 (0)2079052608 [email protected] Keywords: Mitochondrial disease, OXPHOS, signalling, omics, genomics, transcriptomics, proteomics, metabolomics, mitochondrial stress response, treatment 1 Abstract Mitochondria are dynamic bioenergetic organelles whose maintenance requires ~1500 proteins from two genomes. Mutations in either the mitochondrial or nuclear genome can disrupt a plethora of cellular metabolic and homeostatic functions. Mitochondrial diseases represent one the most common and severe groups of inherited genetic disorders, characterised by clinical, biochemical, and genetic heterogeneity, diagnostic odysseys, and lack of curative therapies. This review aims to discuss recent advances in mitochondrial biology and medicine arising from widespread utilisation of high-throughput omics technologies, and also includes a broad discussion of emerging therapies for mitochondrial disease. New insights into both bioenergetic and biosynthetic mitochondrial functionalities have expedited the genetic diagnosis of primary mitochondrial disorders, and identified novel mitochondrial pathomechanisms and new targets
    [Show full text]
  • Metabolic and Muscle-Derived Serum Biomarkers Define CHCHD10-Linked Late-Onset Spinal Muscular Atrophy
    medRxiv preprint doi: https://doi.org/10.1101/2021.04.07.21254960; this version posted April 9, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. All rights reserved. No reuse allowed without permission. Metabolic and muscle-derived serum biomarkers define CHCHD10-linked late-onset spinal muscular atrophy Julius Järvilehto, MB1, Sandra Harjuhaahto, MSc1, Edouard Palu, MD2, Mari Auranen, MD, PhD2, Jouni Kvist, PhD1, Henrik Zetterberg, MD, PhD3,4,5,6, Johanna Koskivuori, MSc7, Marko Lehtonen, PhD7, Anna Maija Saukkonen, MD8, Manu Jokela, MD, PhD9,10, Emil Ylikallio, MD, PhD1,2*, Henna Tyynismaa, PhD1,11,12* 1Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland 2Clinical Neurosciences, Neurology, Helsinki University Hospital, Helsinki, Finland 3Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden 4Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden 5Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom 6UK Dementia Research Institute at UCL, London, United Kingdom 7School of Pharmacy, University of Eastern Finland, Kuopio, Finland 8Department of Neurology, Central Hospital of Northern Karelia, Joensuu, Finland 9Division of Clinical Neurosciences, Turku University Hospital and University of Turku, Turku, Finland 10Department of Neurology, Neuromuscular Research Center, Tampere University Hospital and Tampere University, Tampere, Finland 11Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland 12Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland *Corresponding Author: Henna Tyynismaa, Biomedicum Helsinki, Haartmaninkatu 8, 00014 University of Helsinki, Finland.
    [Show full text]
  • CHCHD2 (NM 016139) Human Tagged ORF Clone Product Data
    OriGene Technologies, Inc. 9620 Medical Center Drive, Ste 200 Rockville, MD 20850, US Phone: +1-888-267-4436 [email protected] EU: [email protected] CN: [email protected] Product datasheet for RC209806 CHCHD2 (NM_016139) Human Tagged ORF Clone Product data: Product Type: Expression Plasmids Product Name: CHCHD2 (NM_016139) Human Tagged ORF Clone Tag: Myc-DDK Symbol: CHCHD2 Synonyms: C7orf17; MIX17B; MNRR1; NS2TP; PARK22 Vector: pCMV6-Entry (PS100001) E. coli Selection: Kanamycin (25 ug/mL) Cell Selection: Neomycin ORF Nucleotide >RC209806 ORF sequence Sequence: Red=Cloning site Blue=ORF Green=Tags(s) TTTTGTAATACGACTCACTATAGGGCGGCCGGGAATTCGTCGACTGGATCCGGTACCGAGGAGATCTGCC GCCGCGATCGCC ATGCCGCGTGGAAGCCGAAGCCGCACCTCCCGCATGGCCCCTCCGGCCAGCCGGGCCCCTCAGATGAGAG CTGCACCCAGGCCAGCACCAGTCGCTCAGCCACCAGCAGCGGCACCCCCATCTGCAGTTGGCTCTTCTGC TGCTGCGCCCCGGCAGCCAGTTCTGATGGCCCAGATGGCAACCACTGCAGCTGGCGTGGCTGTGGGCTCT GCTGTGGGGCACACATTGGGTCACGCCATTACTGGGGGCTTCAGTGGAGGAAGTAATGCTGAGCCTGCGA GGCCTGACATCACTTACCAGGAGCCTCAGGGAACCCAGCCAGCACAGCAGCAGCAGCCTTGCCTCTATGA GATCAAACAGTTTCTGGAGTGTGCCCAGAACCAGGGTGACATCAAGCTCTGTGAGGGTTTCAATGAGGTG CTGAAACAGTGCCGACTTGCAAACGGATTGGCC ACGCGTACGCGGCCGCTCGAGCAGAAACTCATCTCAGAAGAGGATCTGGCAGCAAATGATATCCTGGATT ACAAGGATGACGACGATAAGGTTTAA Protein Sequence: >RC209806 protein sequence Red=Cloning site Green=Tags(s) MPRGSRSRTSRMAPPASRAPQMRAAPRPAPVAQPPAAAPPSAVGSSAAAPRQPVLMAQMATTAAGVAVGS AVGHTLGHAITGGFSGGSNAEPARPDITYQEPQGTQPAQQQQPCLYEIKQFLECAQNQGDIKLCEGFNEV LKQCRLANGLA TRTRPLEQKLISEEDLAANDILDYKDDDDKV Restriction Sites: SgfI-MluI This product
    [Show full text]
  • Mutation Analysis of CHCHD10 in Different Neurodegenerative Diseases
    Mutation analysis of CHCHD10 in neurodegenerative diseases, including Parkinson’s disease Ekaterina Rogaeva Tanz Centre for Research in Faculty of Medicine, Neurodegenerative Diseases, Neurology Division University of Toronto Objective is to update on recent genetic findings related to CHCHD10 & CHCHD2 International Conference on Parkinson’s disease and Movement disorder Frankfurt, Germany, Aug 11-13 2015 CHCHD10 is novel ALS/FTD gene FTD & ALS: genetic, clinical & histopathology data [Hardy J & Rogaeva E, Experimental Neurology, 2013] Novel disease genes: MATR3 (RNA/DNA-binding protein): ALS [ Johnson et al, Nature Neur, 2014] CHCHD10 (mitochondrial protein): ALS/FTD [Bannwarth et al., Brain, 2014] Patients of the French family presented with a complex phenotype, including: • ALS (main) • ALS/FTLD • mitochondrial myopathy • cerebellar ataxia • parkinsonism Bannwarth et al., Brain 2014 Result of whole exome sequencing of 2 affected family members p.S59L is found in all 8 affected cases Bannwarth et al., Brain 2014 CHCHD10 is located in mitochondrial intermembrane space Bannwarth et al., Brain 2014 Immunoelectron microscopy of CHCHD10 CHCHD10 protein is enriched at cristae junctions mitochondria Destruction of the mitochondrial network in CHCHD10 patients Bannwarth et al., Brain 2014 Muscle biopsy shows respiratory chain deficiency Defect in assembly of mitochondrial Complex V Deletions in mitochondrial DNA MT WT MT WT MT WT MT MT WT MT WT Brain pathology in mutation carriers is unknown CHCHD10 is confirmed as ALS gene: novel p.R15L in
    [Show full text]