47 results on '"Euro, L."'
Search Results
2. P416 Systemic NAD+ deficiency reveals a potential therapeutic target for RYR1-related myopathies
- Author
-
Lawal, T., primary, Riekhof, W., additional, Groom, L., additional, Varma, P., additional, Chrismer, I., additional, Kokkinis, A., additional, Grunseich, C., additional, Witherspoon, J., additional, Razaqyar, M., additional, Meilleur, K., additional, Bönnemann, C., additional, Xiang, L., additional, Euro, L., additional, Jansson, S., additional, Mohassel, P., additional, Dirksen, R., additional, and Todd, J., additional
- Published
- 2023
- Full Text
- View/download PDF
3. Cys377 residue in NqrF subunit confers Ag+ sensitivity of Na+-translocating NADH:quinone oxidoreductase from Vibrio harveyi
- Author
-
Fadeeva, M. S., Bertsova, Y. V., Euro, L., and Bogachev, A. V.
- Published
- 2011
- Full Text
- View/download PDF
4. Twinkle is not the mitochondrial DNA replicative helicase in C. elegans, but may have alternate mitochondrial functions
- Author
-
Anu Suomalainen, Henderson Hr, Euro L, and Andrew Dillin
- Subjects
0303 health sciences ,Mitochondrial DNA ,biology ,Helicase ,biology.organism_classification ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,Mitochondrial unfolded protein response ,biology.protein ,Replisome ,Mitochondrial fission ,Inner mitochondrial membrane ,030217 neurology & neurosurgery ,Caenorhabditis elegans ,030304 developmental biology ,Mitochondrial DNA replication - Abstract
Dysfunction of mitochondrial DNA replication machinery is a common cause of mitochondrial diseases. The minimal mammalian replisome is made up of DNA polymerase gamma, replicative helicase Twinkle, and single-stranded DNA binding protein. The replisome is localized to the inner mitochondrial membrane and serves as the site of mitochondrial DNA replication and mitochondrial fission. Recently, a sequence homolog of Twinkle was uncovered in the nematode Caenorhabditis elegans. Here, we characterized this homolog, twnk-1, and report that twnk-1 does not function as the primary mitochondrial DNA replicative helicase in this species, as loss of twnk-1 does not result in reduce mitochondrial DNA levels, or result in other expected mitochondrial dysfunctions such as reduced oxygen consumption rates, increased sensitivity to metabolic perturbations, or reduced muscle function. Instead, twnk-1 mutants have increased mitochondrial DNA as they age, and exhibit phenotypes associated with mitochondrial stress, including reduced fecundity, an activation of the mitochondrial unfolded protein response, and mitochondrial fragmentation. Our results suggest in Caenorhabditis elegans, twnk-1 does not function as the mitochondrial DNA replicative helicase, but has an alternative function in regulating mitochondrial function.
- Published
- 2019
- Full Text
- View/download PDF
5. Structural modeling of tissue-specific mitochondrial alanyl-tRNA synthetase (AARS2) defects predicts differential effects on aminoacylation
- Author
-
Euro, L, Konovalova, S, Asin-Cayuela, J, Tulinius, M, Griffin, H, Horvath, R, Taylor, RW, Chinnery, PF, Schara, U, Thorburn, DR, Suomalainen, A, Chihade, J, Tyynismaa, H, Euro, L, Konovalova, S, Asin-Cayuela, J, Tulinius, M, Griffin, H, Horvath, R, Taylor, RW, Chinnery, PF, Schara, U, Thorburn, DR, Suomalainen, A, Chihade, J, and Tyynismaa, H
- Abstract
The accuracy of mitochondrial protein synthesis is dependent on the coordinated action of nuclear-encoded mitochondrial aminoacyl-tRNA synthetases (mtARSs) and the mitochondrial DNA-encoded tRNAs. The recent advances in whole-exome sequencing have revealed the importance of the mtARS proteins for mitochondrial pathophysiology since nearly every nuclear gene for mtARS (out of 19) is now recognized as a disease gene for mitochondrial disease. Typically, defects in each mtARS have been identified in one tissue-specific disease, most commonly affecting the brain, or in one syndrome. However, mutations in the AARS2 gene for mitochondrial alanyl-tRNA synthetase (mtAlaRS) have been reported both in patients with infantile-onset cardiomyopathy and in patients with childhood to adulthood-onset leukoencephalopathy. We present here an investigation of the effects of the described mutations on the structure of the synthetase, in an effort to understand the tissue-specific outcomes of the different mutations. The mtAlaRS differs from the other mtARSs because in addition to the aminoacylation domain, it has a conserved editing domain for deacylating tRNAs that have been mischarged with incorrect amino acids. We show that the cardiomyopathy phenotype results from a single allele, causing an amino acid change R592W in the editing domain of AARS2, whereas the leukodystrophy mutations are located in other domains of the synthetase. Nevertheless, our structural analysis predicts that all mutations reduce the aminoacylation activity of the synthetase, because all mtAlaRS domains contribute to tRNA binding for aminoacylation. According to our model, the cardiomyopathy mutations severely compromise aminoacylation whereas partial activity is retained by the mutation combinations found in the leukodystrophy patients. These predictions provide a hypothesis for the molecular basis of the distinct tissue-specific phenotypic outcomes.
- Published
- 2015
6. Numerical Modeling of the Dynamical Interaction Between Slug Flow and Vortex Induced Vibration in Horizontal Submarine Pipelines
- Author
-
Bossio V., Boris M., primary, Blanco A., Armando J., additional, and Casanova M., Euro L., additional
- Published
- 2014
- Full Text
- View/download PDF
7. Mitochondrial EFTs defects in juvenile-onset Leigh disease, ataxia, neuropathy, and optic atrophy
- Author
-
Ahola, S., primary, Isohanni, P., additional, Euro, L., additional, Brilhante, V., additional, Palotie, A., additional, Pihko, H., additional, Lonnqvist, T., additional, Lehtonen, T., additional, Laine, J., additional, Tyynismaa, H., additional, and Suomalainen, A., additional
- Published
- 2014
- Full Text
- View/download PDF
8. POLG1 manifestations in childhood
- Author
-
Isohanni, P., primary, Hakonen, A.H., additional, Euro, L., additional, Paetau, I., additional, Linnankivi, T., additional, Liukkonen, E., additional, Wallden, T., additional, Luostarinen, L., additional, Valanne, L., additional, Paetau, A., additional, Uusimaa, J., additional, Lönnqvist, T., additional, Suomalainen, A., additional, and Pihko, H., additional
- Published
- 2011
- Full Text
- View/download PDF
9. The Cys-377 in NqrF subunit of Na+-translocating NADH: Quinone oxidoreductase from Vibrio harveyi confers its sensitivity to low concentrations of Ag+ ions
- Author
-
Fadeeva, M.S., primary, Euro, L., additional, Bertsova, Yu. V., additional, and Bogachev, A.V., additional
- Published
- 2010
- Full Text
- View/download PDF
10. Analysis of an Accelerating Rotor-Bearing System With Flexible Damped Supports
- Author
-
Casanova, Euro L., primary and Medina, Luis U., additional
- Published
- 1998
- Full Text
- View/download PDF
11. POLG1manifestations in childhood
- Author
-
Isohanni, P., Hakonen, A.H., Euro, L., Paetau, I., Linnankivi, T., Liukkonen, E., Wallden, T., Luostarinen, L., Valanne, L., Paetau, A., Uusimaa, J., Lönnqvist, T., Suomalainen, A., and Pihko, H.
- Abstract
Mitochondrial DNA polymerase (POLG1) mutations in children often manifest as Alpers syndrome, whereas in adults, a common manifestation is mitochondrial recessive ataxia syndrome (MIRAS) with severe epilepsy. Because some patients with MIRAS have presented with ataxia or epilepsy already in childhood, we searched for POLG1mutations in neurologic manifestations in childhood.
- Published
- 2011
- Full Text
- View/download PDF
12. ATpase-deficient mitochondrial inner membrane protein ATAD3a disturbs mitochondrial dynamics in dominant hereditary spastic paraplegia
- Author
-
Cooper H., Yang Y., Ylikallio E., Khairullin R., Woldegebriel R., Lin K., Euro L., Palin E., Wolf A., Trokovic R., Isohanni P., Kaakkola S., Auranen M., Lonqvist T., Wanrooij S., Tyynismaa H., Cooper H., Yang Y., Ylikallio E., Khairullin R., Woldegebriel R., Lin K., Euro L., Palin E., Wolf A., Trokovic R., Isohanni P., Kaakkola S., Auranen M., Lonqvist T., Wanrooij S., and Tyynismaa H.
- Abstract
© The Author 2017. Published by Oxford University Press. All rights reserved.De novo mutations in ATAD3A (ATPase family AAA-domain containing protein 3A) were recently found to cause a neurological syndrome with developmental delay, hypotonia, spasticity, optic atrophy, axonal neuropathy, and hypertrophic cardiomyopathy. Using whole-exome sequencing, we identified a dominantly inherited heterozygous variant c.1064G>A (p. G355D) in ATAD3A in a mother presenting with hereditary spastic paraplegia (HSP) and axonal neuropathy and her son with dyskinetic cerebral palsy, both with disease onset in childhood. HSP is a clinically and genetically heterogeneous disorder of the upper motor neurons. Symptoms beginning in early childhood may resemble spastic cerebral palsy. The function of ATAD3A, a mitochondrial inner membrane AAA ATPase, is yet undefined. AAA ATPases form hexameric rings, which are catalytically dependent on the co-operation of the subunits. The dominant-negative patient mutation affects the Walker A motif, which is responsible for ATP binding in the AAA module of ATAD3A, and we show that the recombinant mutant ATAD3A protein has a markedly reduced ATPase activity. We further show that overexpression of the mutant ATAD3A fragments the mitochondrial network and induces lysosome mass. Similarly, we observed altered dynamics of the mitochondrial network and increased lysosomes in patient fibroblasts and neurons derived through differentiation of patient-specific induced pluripotent stem cells. These alterations were verified in patient fibroblasts to associate with upregulated basal autophagy through mTOR inactivation, resembling starvation. Mutations in ATAD3A can thus be dominantly inherited and underlie variable neurological phenotypes, including HSP, with intrafamiliar variability. This finding extends the group of mitochondrial inner membrane AAA proteins associated with spasticity.
13. ATpase-deficient mitochondrial inner membrane protein ATAD3a disturbs mitochondrial dynamics in dominant hereditary spastic paraplegia
- Author
-
Cooper H., Yang Y., Ylikallio E., Khairullin R., Woldegebriel R., Lin K., Euro L., Palin E., Wolf A., Trokovic R., Isohanni P., Kaakkola S., Auranen M., Lonqvist T., Wanrooij S., Tyynismaa H., Cooper H., Yang Y., Ylikallio E., Khairullin R., Woldegebriel R., Lin K., Euro L., Palin E., Wolf A., Trokovic R., Isohanni P., Kaakkola S., Auranen M., Lonqvist T., Wanrooij S., and Tyynismaa H.
- Abstract
© The Author 2017. Published by Oxford University Press. All rights reserved.De novo mutations in ATAD3A (ATPase family AAA-domain containing protein 3A) were recently found to cause a neurological syndrome with developmental delay, hypotonia, spasticity, optic atrophy, axonal neuropathy, and hypertrophic cardiomyopathy. Using whole-exome sequencing, we identified a dominantly inherited heterozygous variant c.1064G>A (p. G355D) in ATAD3A in a mother presenting with hereditary spastic paraplegia (HSP) and axonal neuropathy and her son with dyskinetic cerebral palsy, both with disease onset in childhood. HSP is a clinically and genetically heterogeneous disorder of the upper motor neurons. Symptoms beginning in early childhood may resemble spastic cerebral palsy. The function of ATAD3A, a mitochondrial inner membrane AAA ATPase, is yet undefined. AAA ATPases form hexameric rings, which are catalytically dependent on the co-operation of the subunits. The dominant-negative patient mutation affects the Walker A motif, which is responsible for ATP binding in the AAA module of ATAD3A, and we show that the recombinant mutant ATAD3A protein has a markedly reduced ATPase activity. We further show that overexpression of the mutant ATAD3A fragments the mitochondrial network and induces lysosome mass. Similarly, we observed altered dynamics of the mitochondrial network and increased lysosomes in patient fibroblasts and neurons derived through differentiation of patient-specific induced pluripotent stem cells. These alterations were verified in patient fibroblasts to associate with upregulated basal autophagy through mTOR inactivation, resembling starvation. Mutations in ATAD3A can thus be dominantly inherited and underlie variable neurological phenotypes, including HSP, with intrafamiliar variability. This finding extends the group of mitochondrial inner membrane AAA proteins associated with spasticity.
14. The Cys-377 in NqrF subunit of Na+-translocating NADH: Quinone oxidoreductase from Vibrio harveyi confers its sensitivity to low concentrations of Ag+ ions
- Author
-
Fadeeva, M.S., Euro, L., Bertsova, Yu. V., and Bogachev, A.V.
- Published
- 2010
- Full Text
- View/download PDF
15. Preferential binding of ADP-bound mitochondrial HSP70 to the nucleotide exchange factor GRPEL1 over GRPEL2.
- Author
-
Manjunath P, Stojkovič G, Euro L, Konovalova S, Wanrooij S, Koski K, and Tyynismaa H
- Subjects
- Humans, Protein Binding, Mitochondrial Proteins metabolism, Mitochondrial Proteins chemistry, Mitochondrial Proteins genetics, Models, Molecular, Mitochondria metabolism, HSP70 Heat-Shock Proteins metabolism, HSP70 Heat-Shock Proteins chemistry, HSP70 Heat-Shock Proteins genetics, Adenosine Diphosphate metabolism
- Abstract
Human nucleotide exchange factors GRPEL1 and GRPEL2 play pivotal roles in the ADP-ATP exchange within the protein folding cycle of mitochondrial HSP70 (mtHSP70), a crucial chaperone facilitating protein import into the mitochondrial matrix. Studies in human cells and mice have indicated that while GRPEL1 serves as an essential co-chaperone for mtHSP70, GRPEL2 has a role regulated by stress. However, the precise structural and biochemical mechanisms underlying the distinct functions of the GRPEL proteins have remained elusive. In our study, we present evidence revealing that ADP-bound mtHSP70 exhibits remarkably higher affinity for GRPEL1 compared to GRPEL2, with the latter experiencing a notable decrease in affinity upon ADP binding. Additionally, Pi assay showed that GRPEL1, but not GRPEL2, enhanced the ATPase activity of mtHSP70. Utilizing Alphafold modeling, we propose that the interaction between GRPEL1 and mtHSP70 can induce the opening of the nucleotide binding cleft of the chaperone, thereby facilitating the release of ADP, whereas GRPEL2 lacks this capability. Additionally, our findings suggest that the redox-regulated Cys87 residue in GRPEL2 does not play a role in dimerization but rather reduces its affinity for mtHSP70. Our findings on the structural and functional disparities between GRPEL1 and GRPEL2 may have implications for mitochondrial protein folding and import processes under varying cellular conditions., (© 2024 The Author(s). Protein Science published by Wiley Periodicals LLC on behalf of The Protein Society.)
- Published
- 2024
- Full Text
- View/download PDF
16. Niacin supplementation in a child with novel MTTN variant m.5670A>G causing early onset mitochondrial myopathy and NAD + deficiency.
- Author
-
Aaltio J, Euro L, Tynninen O, Vu HS, Ni M, DeBerardinis RJ, Suomalainen A, and Isohanni P
- Subjects
- Humans, Male, Child, Preschool, Muscle, Skeletal pathology, Muscle, Skeletal drug effects, Mutation, Dietary Supplements, DNA, Mitochondrial genetics, Child, Niacin, Mitochondrial Myopathies genetics, Mitochondrial Myopathies drug therapy, NAD metabolism
- Abstract
Myopathy is a common manifestation in mitochondrial disorders, but the pathomechanisms are still insufficiently studied in children. Here, we report a severe, progressive mitochondrial myopathy in a four-year-old child, who died at eight years. He developed progressive loss of muscle strength with nocturnal hypoventilation and dilated cardiomyopathy. Skeletal muscle showed ragged red fibers and severe combined respiratory chain deficiency. Mitochondrial DNA sequencing revealed a novel m.5670A>G mutation in mitochondrial tRNA
Asn (MTTN) with 88 % heteroplasmy in muscle. The proband also had systemic NAD+ deficiency but rescuing this with the NAD+ precursor niacin did not stop disease progression. Targeted metabolomics revealed an overall shift of metabolism towards controls after niacin supplementation, with normalized tryptophan metabolites and lipid-metabolic markers, but most amino acids did not respond to niacin therapy. To conclude, we report a new MTTN mutation, secondary NAD+ deficiency in childhood-onset mitochondrial myopathy with metabolic but meager clinical response to niacin supplementation., Competing Interests: Declaration of competing interest The authors declare that they have known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper. A.S. and L.E. are co-founders of NADMED Ltd., (Copyright © 2024. Published by Elsevier B.V.)- Published
- 2024
- Full Text
- View/download PDF
17. Genetic and functional correction of argininosuccinate lyase deficiency using CRISPR adenine base editors.
- Author
-
Jalil S, Keskinen T, Juutila J, Sartori Maldonado R, Euro L, Suomalainen A, Lapatto R, Kuuluvainen E, Hietakangas V, Otonkoski T, Hyvönen ME, and Wartiovaara K
- Subjects
- Humans, Clustered Regularly Interspaced Short Palindromic Repeats, RNA, Guide, CRISPR-Cas Systems, Urea, Gene Editing methods, Argininosuccinate Lyase genetics, Argininosuccinic Aciduria genetics, Argininosuccinic Aciduria therapy
- Abstract
Argininosuccinate lyase deficiency (ASLD) is a recessive metabolic disorder caused by variants in ASL. In an essential step in urea synthesis, ASL breaks down argininosuccinate (ASA), a pathognomonic ASLD biomarker. The severe disease forms lead to hyperammonemia, neurological injury, and even early death. The current treatments are unsatisfactory, involving a strict low-protein diet, arginine supplementation, nitrogen scavenging, and in some cases, liver transplantation. An unmet need exists for improved, efficient therapies. Here, we show the potential of a lipid nanoparticle-mediated CRISPR approach using adenine base editors (ABEs) for ASLD treatment. To model ASLD, we first generated human-induced pluripotent stem cells (hiPSCs) from biopsies of individuals homozygous for the Finnish founder variant (c.1153C>T [p.Arg385Cys]) and edited this variant using the ABE. We then differentiated the hiPSCs into hepatocyte-like cells that showed a 1,000-fold decrease in ASA levels compared to those of isogenic non-edited cells. Lastly, we tested three different FDA-approved lipid nanoparticle formulations to deliver the ABE-encoding RNA and the sgRNA targeting the ASL variant. This approach efficiently edited the ASL variant in fibroblasts with no apparent cell toxicity and minimal off-target effects. Further, the treatment resulted in a significant decrease in ASA, to levels of healthy donors, indicating restoration of the urea cycle. Our work describes a highly efficient approach to editing the disease-causing ASL variant and restoring the function of the urea cycle. This method relies on RNA delivered by lipid nanoparticles, which is compatible with clinical applications, improves its safety profile, and allows for scalable production., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
18. Compressive stress-mediated p38 activation required for ERα + phenotype in breast cancer.
- Author
-
Munne PM, Martikainen L, Räty I, Bertula K, Nonappa, Ruuska J, Ala-Hongisto H, Peura A, Hollmann B, Euro L, Yavuz K, Patrikainen L, Salmela M, Pokki J, Kivento M, Väänänen J, Suomi T, Nevalaita L, Mutka M, Kovanen P, Leidenius M, Meretoja T, Hukkinen K, Monni O, Pouwels J, Sahu B, Mattson J, Joensuu H, Heikkilä P, Elo LL, Metcalfe C, Junttila MR, Ikkala O, and Klefström J
- Subjects
- Breast Neoplasms metabolism, Breast Neoplasms pathology, Case-Control Studies, Cell Line, Tumor, Cinnamates pharmacology, Collagen chemistry, Collagen pharmacology, Drug Combinations, Enhancer of Zeste Homolog 2 Protein genetics, Enhancer of Zeste Homolog 2 Protein metabolism, Estradiol pharmacology, Estrogen Receptor alpha metabolism, Female, Fulvestrant pharmacology, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Histones genetics, Histones metabolism, Humans, Indazoles pharmacology, Laminin chemistry, Laminin pharmacology, Mammary Glands, Human drug effects, Mammary Glands, Human metabolism, Mammary Glands, Human pathology, Phenotype, Proteoglycans chemistry, Proteoglycans pharmacology, Tamoxifen pharmacology, Tissue Culture Techniques, p38 Mitogen-Activated Protein Kinases metabolism, Breast Neoplasms genetics, Estrogen Receptor alpha genetics, Mechanotransduction, Cellular genetics, Transcriptome, p38 Mitogen-Activated Protein Kinases genetics
- Abstract
Breast cancer is now globally the most frequent cancer and leading cause of women's death. Two thirds of breast cancers express the luminal estrogen receptor-positive (ERα + ) phenotype that is initially responsive to antihormonal therapies, but drug resistance emerges. A major barrier to the understanding of the ERα-pathway biology and therapeutic discoveries is the restricted repertoire of luminal ERα + breast cancer models. The ERα + phenotype is not stable in cultured cells for reasons not fully understood. We examine 400 patient-derived breast epithelial and breast cancer explant cultures (PDECs) grown in various three-dimensional matrix scaffolds, finding that ERα is primarily regulated by the matrix stiffness. Matrix stiffness upregulates the ERα signaling via stress-mediated p38 activation and H3K27me3-mediated epigenetic regulation. The finding that the matrix stiffness is a central cue to the ERα phenotype reveals a mechanobiological component in breast tissue hormonal signaling and enables the development of novel therapeutic interventions. Subject terms: ER-positive (ER + ), breast cancer, ex vivo model, preclinical model, PDEC, stiffness, p38 SAPK., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
19. SUCLA2 mutations cause global protein succinylation contributing to the pathomechanism of a hereditary mitochondrial disease.
- Author
-
Gut P, Matilainen S, Meyer JG, Pällijeff P, Richard J, Carroll CJ, Euro L, Jackson CB, Isohanni P, Minassian BA, Alkhater RA, Østergaard E, Civiletto G, Parisi A, Thevenet J, Rardin MJ, He W, Nishida Y, Newman JC, Liu X, Christen S, Moco S, Locasale JW, Schilling B, Suomalainen A, and Verdin E
- Subjects
- Animals, Cells, Cultured, Female, Humans, Infant, Lysine metabolism, Male, Mice, Mice, Knockout, Mitochondria metabolism, Mitochondrial Diseases genetics, Mitochondrial Diseases metabolism, Mutation, Proteomics, Sirtuins deficiency, Sirtuins genetics, Sirtuins metabolism, Succinate-CoA Ligases deficiency, Succinate-CoA Ligases metabolism, Survival Analysis, Zebrafish, Acyl Coenzyme A metabolism, Mitochondrial Diseases pathology, Succinate-CoA Ligases genetics
- Abstract
Mitochondrial acyl-coenzyme A species are emerging as important sources of protein modification and damage. Succinyl-CoA ligase (SCL) deficiency causes a mitochondrial encephalomyopathy of unknown pathomechanism. Here, we show that succinyl-CoA accumulates in cells derived from patients with recessive mutations in the tricarboxylic acid cycle (TCA) gene succinyl-CoA ligase subunit-β (SUCLA2), causing global protein hyper-succinylation. Using mass spectrometry, we quantify nearly 1,000 protein succinylation sites on 366 proteins from patient-derived fibroblasts and myotubes. Interestingly, hyper-succinylated proteins are distributed across cellular compartments, and many are known targets of the (NAD
+ )-dependent desuccinylase SIRT5. To test the contribution of hyper-succinylation to disease progression, we develop a zebrafish model of the SCL deficiency and find that SIRT5 gain-of-function reduces global protein succinylation and improves survival. Thus, increased succinyl-CoA levels contribute to the pathology of SCL deficiency through post-translational modifications.- Published
- 2020
- Full Text
- View/download PDF
20. Fibroblast Growth Factor 21 Drives Dynamics of Local and Systemic Stress Responses in Mitochondrial Myopathy with mtDNA Deletions.
- Author
-
Forsström S, Jackson CB, Carroll CJ, Kuronen M, Pirinen E, Pradhan S, Marmyleva A, Auranen M, Kleine IM, Khan NA, Roivainen A, Marjamäki P, Liljenbäck H, Wang L, Battersby BJ, Richter U, Velagapudi V, Nikkanen J, Euro L, and Suomalainen A
- Subjects
- Activating Transcription Factors metabolism, Animals, Cell Line, DNA, Mitochondrial genetics, Escherichia coli, Female, Fibroblast Growth Factors genetics, Growth Differentiation Factor 15 metabolism, Humans, Male, Mice, Mitochondria genetics, Mitochondrial Myopathies genetics, Sequence Deletion, Stress, Physiological genetics, DNA, Mitochondrial metabolism, Fibroblast Growth Factors physiology, Mitochondria metabolism, Mitochondrial Myopathies metabolism, Stress, Physiological physiology
- Abstract
Mitochondrial dysfunction elicits stress responses that safeguard cellular homeostasis against metabolic insults. Mitochondrial integrated stress response (ISR
mt ) is a major response to mitochondrial (mt)DNA expression stress (mtDNA maintenance, translation defects), but the knowledge of dynamics or interdependence of components is lacking. We report that in mitochondrial myopathy, ISRmt progresses in temporal stages and development from early to chronic and is regulated by autocrine and endocrine effects of FGF21, a metabolic hormone with pleiotropic effects. Initial disease signs induce transcriptional ISRmt (ATF5, mitochondrial one-carbon cycle, FGF21, and GDF15). The local progression to 2nd metabolic ISRmt stage (ATF3, ATF4, glucose uptake, serine biosynthesis, and transsulfuration) is FGF21 dependent. Mitochondrial unfolded protein response marks the 3rd ISRmt stage of failing tissue. Systemically, FGF21 drives weight loss and glucose preference, and modifies metabolism and respiratory chain deficiency in a specific hippocampal brain region. Our evidence indicates that FGF21 is a local and systemic messenger of mtDNA stress in mice and humans with mitochondrial disease., (Copyright © 2019 Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
21. Instability of the mitochondrial alanyl-tRNA synthetase underlies fatal infantile-onset cardiomyopathy.
- Author
-
Sommerville EW, Zhou XL, Oláhová M, Jenkins J, Euro L, Konovalova S, Hilander T, Pyle A, He L, Habeebu S, Saunders C, Kelsey A, Morris AAM, McFarland R, Suomalainen A, Gorman GS, Wang ED, Thiffault I, Tyynismaa H, and Taylor RW
- Subjects
- Alanine-tRNA Ligase genetics, Cardiomyopathies genetics, Diseases in Twins genetics, Enzyme Stability, Fibroblasts metabolism, Genes, Recessive, Humans, Infant, Lactic Acid, Male, Mitochondria metabolism, Mitochondrial Proteins biosynthesis, Muscle, Skeletal metabolism, Myocardium metabolism, Pedigree, Respiratory Insufficiency enzymology, Alanine-tRNA Ligase metabolism, Cardiomyopathies enzymology
- Abstract
Recessively inherited variants in AARS2 (NM_020745.2) encoding mitochondrial alanyl-tRNA synthetase (mt-AlaRS) were first described in patients presenting with fatal infantile cardiomyopathy and multiple oxidative phosphorylation defects. To date, all described patients with AARS2-related fatal infantile cardiomyopathy are united by either a homozygous or compound heterozygous c.1774C>T (p.Arg592Trp) missense founder mutation that is absent in patients with other AARS2-related phenotypes. We describe the clinical, biochemical and molecular investigations of two unrelated boys presenting with fatal infantile cardiomyopathy, lactic acidosis and respiratory failure. Oxidative histochemistry showed cytochrome c oxidase-deficient fibres in skeletal and cardiac muscle. Biochemical studies showed markedly decreased activities of mitochondrial respiratory chain complexes I and IV with a mild decrease of complex III activity in skeletal and cardiac muscle. Using next-generation sequencing, we identified a c.1738C>T (p.Arg580Trp) AARS2 variant shared by both patients that was in trans with a loss-of-function heterozygous AARS2 variant; a c.1008dupT (p.Asp337*) nonsense variant or an intragenic deletion encompassing AARS2 exons 5-7. Interestingly, our patients did not harbour the p.Arg592Trp AARS2 founder mutation. In silico modelling of the p.Arg580Trp substitution suggested a deleterious impact on protein stability and folding. We confirmed markedly decreased mt-AlaRS protein levels in patient fibroblasts, skeletal and cardiac muscle, although mitochondrial protein synthesis defects were confined to skeletal and cardiac muscle. In vitro data showed that the p.Arg580Trp variant had a minimal effect on activation, aminoacylation or misaminoacylation activities relative to wild-type mt-AlaRS, demonstrating that instability of mt-AlaRS is the biological mechanism underlying the fatal cardiomyopathy phenotype in our patients.
- Published
- 2019
- Full Text
- View/download PDF
22. Redox regulation of GRPEL2 nucleotide exchange factor for mitochondrial HSP70 chaperone.
- Author
-
Konovalova S, Liu X, Manjunath P, Baral S, Neupane N, Hilander T, Yang Y, Balboa D, Terzioglu M, Euro L, Varjosalo M, and Tyynismaa H
- Subjects
- Cell Line, HSP70 Heat-Shock Proteins analysis, Humans, Intracellular Signaling Peptides and Proteins analysis, Mitochondrial Proteins analysis, Mitochondrial Proteins metabolism, Oxidation-Reduction, Protein Folding, Protein Multimerization, Protein Transport, HSP70 Heat-Shock Proteins metabolism, Intracellular Signaling Peptides and Proteins metabolism, Mitochondria metabolism, Oxidative Stress
- Abstract
Mitochondria are central organelles to cellular metabolism. Their function relies largely on nuclear-encoded proteins that must be imported from the cytosol, and thus the protein import pathways are important for the maintenance of mitochondrial proteostasis. Mitochondrial HSP70 (mtHsp70) is a key component in facilitating the translocation of proteins through the inner membrane into the mitochondrial matrix. Its protein folding cycle is regulated by the nucleotide-exchange factor GrpE, which triggers the release of folded proteins by ATP rebinding. Vertebrates have two mitochondrial GrpE paralogs, GRPEL1 and 2, but without clearly defined roles. Using BioID proximity labeling to identify potential binding partners of the GRPELs in the mitochondrial matrix, we obtained results supporting a model where both GRPELs regulate mtHsp70 as homodimers. We show that GRPEL2 is not essential in human cultured cells, and its absence does not prevent mitochondrial protein import. Instead we find that GRPEL2 is redox regulated in oxidative stress. In the presence of hydrogen peroxide, GRPEL2 forms dimers through intermolecular disulfide bonds in which Cys87 is the thiol switch. We propose that the dimerization of GRPEL2 may activate the folding machinery responsible for protein import into mitochondrial matrix or enhance the chaperone activity of mtHSP70, thus protecting mitochondrial proteostasis in oxidative stress., (Copyright © 2018 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
23. Simultaneous measurement of folate cycle intermediates in different biological matrices using liquid chromatography-tandem mass spectrometry.
- Author
-
Nandania J, Kokkonen M, Euro L, and Velagapudi V
- Subjects
- Animals, Brain Chemistry, Folic Acid blood, Folic Acid chemistry, Folic Acid isolation & purification, Linear Models, Muscles chemistry, Myocardium chemistry, Organ Specificity, Rats, Reproducibility of Results, Sensitivity and Specificity, Chromatography, High Pressure Liquid methods, Folic Acid analysis, Tandem Mass Spectrometry methods
- Abstract
The folate cycle is an essential metabolic pathway in the cell, involved in nucleotide synthesis, maintenance of the redox balance in the cell, methionine metabolism and re-methylation reactions. Standardised methods for the measurement of folate cycle intermediates in different biological matrices are in great demand. Here we describe a rapid, sensitive, precise and accurate liquid chromatographic-tandem mass spectrometric (LC-MS/MS) method with a wide calibration curve range and a short run time for the simultaneous determination of folate cycle metabolites, including tetrahydrofolic acid (THF), 5‑methyl THF, 5‑formyl THF, 5,10‑methenyl THF, 5,10‑methylene THF, dihydrofolic acid (DHF) and folic acid in different biological matrices. Extraction of folate derivatives from soft and hard tissue samples as well as from adherent cells was achieved using homogenisation in buffer, while extraction from the whole blood and plasma relied on the anion exchange solid-phase extraction (SPE) method. Chromatographic separation was completed using a Waters Atlantis dC
18 2.0 × 100 mm, 3-μ column with a gradient elution using formic acid in water (0.1% v/v) and acetonitrile as the mobile phases. LC gradient started with 95% of the aqueous phase which was gradually changed to 95% of the organic phase during 2.70 min in order to separate the selected metabolites. The analytes were separated with a run time of 5 min at a flow rate of 0.300 mL/min and detected using a Waters Xevo-TQS triple quadrupole mass spectrometer in the multiple reaction monitoring mode (MRM) at positive polarity. The instrument response was linear over a calibration range of 0.5 to 2500 ng/mL (r2 > 0.980). The developed bioanalytical method was thoroughly validated in terms of accuracy, precision, linearity, recovery, sensitivity and stability for tissue and blood samples. The matrix effect was compensated by using structurally similar isotope labelled internal standard (IS),13 C5 ‑methyl THF, for all folate metabolites. However, not all folate metabolites can be accurately quantified using this method due to their high interconversion rates especially at low pH. This applies to 5,10‑methylene THF which interconverts into THF, and 5,10‑methenyl‑THF interconverting into 5‑formyl‑THF. Using this method, we measured folate cycle intermediates in mouse bone marrow cells and plasma, in human whole blood; in mouse muscle, liver, heart and brain samples., (Copyright © 2018 Elsevier B.V. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
24. Editing activity for eliminating mischarged tRNAs is essential in mammalian mitochondria.
- Author
-
Hilander T, Zhou XL, Konovalova S, Zhang FP, Euro L, Chilov D, Poutanen M, Chihade J, Wang ED, and Tyynismaa H
- Subjects
- Alanine-tRNA Ligase metabolism, Amino Acid Sequence, Animals, Cells, Cultured, Embryo, Mammalian cytology, Fibroblasts cytology, Fibroblasts metabolism, Humans, Mammals, Mice, Inbred C57BL, Mice, Knockout, Mitochondria metabolism, Mutation, Protein Biosynthesis genetics, RNA, Transfer metabolism, Sequence Homology, Amino Acid, Alanine-tRNA Ligase genetics, Mitochondria genetics, RNA Editing, RNA, Transfer genetics, Transfer RNA Aminoacylation genetics
- Abstract
Accuracy of protein synthesis is enabled by the selection of amino acids for tRNA charging by aminoacyl-tRNA synthetases (ARSs), and further enhanced by the proofreading functions of some of these enzymes for eliminating tRNAs mischarged with noncognate amino acids. Mouse models of editing-defective cytoplasmic alanyl-tRNA synthetase (AlaRS) have previously demonstrated the importance of proofreading for cytoplasmic protein synthesis, with embryonic lethal and progressive neurodegeneration phenotypes. Mammalian mitochondria import their own set of nuclear-encoded ARSs for translating critical polypeptides of the oxidative phosphorylation system, but the importance of editing by the mitochondrial ARSs for mitochondrial proteostasis has not been known. We demonstrate here that the human mitochondrial AlaRS is capable of editing mischarged tRNAs in vitro, and that loss of the proofreading activity causes embryonic lethality in mice. These results indicate that tRNA proofreading is essential in mammalian mitochondria, and cannot be overcome by other quality control mechanisms., (© The Author(s) 2017. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2018
- Full Text
- View/download PDF
25. Loss of mtDNA activates astrocytes and leads to spongiotic encephalopathy.
- Author
-
Ignatenko O, Chilov D, Paetau I, de Miguel E, Jackson CB, Capin G, Paetau A, Terzioglu M, Euro L, and Suomalainen A
- Subjects
- Animals, Brain metabolism, Brain ultrastructure, DNA Helicases genetics, DNA Helicases metabolism, DNA, Mitochondrial metabolism, Mice, Knockout, Microscopy, Electron, Mitochondrial Proteins genetics, Mitochondrial Proteins metabolism, Mutation, Neurons metabolism, Astrocytes metabolism, Brain Diseases genetics, DNA, Mitochondrial genetics, Mitochondrial Diseases genetics
- Abstract
Mitochondrial dysfunction manifests as different neurological diseases, but the mechanisms underlying the clinical variability remain poorly understood. To clarify whether different brain cells have differential sensitivity to mitochondrial dysfunction, we induced mitochondrial DNA (mtDNA) depletion in either neurons or astrocytes of mice, by inactivating Twinkle (TwKO), the replicative mtDNA helicase. Here we show that astrocytes, the most abundant cerebral cell type, are chronically activated upon mtDNA loss, leading to early-onset spongiotic degeneration of brain parenchyma, microgliosis and secondary neurodegeneration. Neuronal mtDNA loss does not, however, cause symptoms until 8 months of age. Findings in astrocyte-TwKO mimic neuropathology of Alpers syndrome, infantile-onset mitochondrial spongiotic encephalopathy caused by mtDNA maintenance defects. Our evidence indicates that (1) astrocytes are dependent on mtDNA integrity; (2) mitochondrial metabolism contributes to their activation; (3) chronic astrocyte activation has devastating consequences, underlying spongiotic encephalopathy; and that (4) astrocytes are a potential target for interventions.
- Published
- 2018
- Full Text
- View/download PDF
26. Defective mitochondrial RNA processing due to PNPT1 variants causes Leigh syndrome.
- Author
-
Matilainen S, Carroll CJ, Richter U, Euro L, Pohjanpelto M, Paetau A, Isohanni P, and Suomalainen A
- Subjects
- Child, Preschool, Exome, Exoribonucleases chemistry, Female, Gene Expression, Humans, Infant, Infant, Newborn, Leigh Disease metabolism, Mitochondria metabolism, Mitochondrial Diseases genetics, Polyribonucleotide Nucleotidyltransferase, RNA metabolism, RNA, Messenger metabolism, RNA, Mitochondrial, Exoribonucleases genetics, Exoribonucleases metabolism, Leigh Disease genetics
- Abstract
Leigh syndrome is a severe infantile encephalopathy with an exceptionally variable genetic background. We studied the exome of a child manifesting with Leigh syndrome at one month of age and progressing to death by the age of 2.4 years, and identified novel compound heterozygous variants in PNPT1, encoding the polynucleotide phosphorylase (PNPase). Expression of the wild type PNPT1 in the subject's myoblasts functionally complemented the defects, and the pathogenicity was further supported by structural predictions and protein and RNA analyses. PNPase is a key enzyme in mitochondrial RNA metabolism, with suggested roles in mitochondrial RNA import and degradation. The variants were predicted to locate in the PNPase active site and disturb the RNA processing activity of the enzyme. The PNPase trimer formation was not affected, but specific RNA processing intermediates derived from mitochondrial transcripts of the ND6 subunit of Complex I, as well as small mRNA fragments, accumulated in the subject's myoblasts. Mitochondrial RNA processing mediated by the degradosome consisting of hSUV3 and PNPase is poorly characterized, and controversy on the role and location of PNPase within human mitochondria exists. Our evidence indicates that PNPase activity is essential for the correct maturation of the ND6 transcripts, and likely for the efficient removal of degradation intermediates. Loss of its activity will result in combined respiratory chain deficiency, and a classic respiratory chain-deficiency-associated disease, Leigh syndrome, indicating an essential role for the enzyme for normal function of the mitochondrial respiratory chain., (© The Author 2017. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2017
- Full Text
- View/download PDF
27. ATPase-deficient mitochondrial inner membrane protein ATAD3A disturbs mitochondrial dynamics in dominant hereditary spastic paraplegia.
- Author
-
Cooper HM, Yang Y, Ylikallio E, Khairullin R, Woldegebriel R, Lin KL, Euro L, Palin E, Wolf A, Trokovic R, Isohanni P, Kaakkola S, Auranen M, Lönnqvist T, Wanrooij S, and Tyynismaa H
- Subjects
- ATPases Associated with Diverse Cellular Activities, Adenosine Triphosphatases biosynthesis, Adolescent, Adult, Axons metabolism, Axons pathology, Cerebral Palsy pathology, Child, Preschool, Female, Gene Expression Regulation, Humans, Male, Membrane Proteins biosynthesis, Mitochondria metabolism, Mitochondria pathology, Mitochondrial Dynamics genetics, Mitochondrial Membranes metabolism, Mitochondrial Membranes pathology, Mitochondrial Proteins biosynthesis, Mutation, Spastic Paraplegia, Hereditary pathology, TOR Serine-Threonine Kinases genetics, Adenosine Triphosphatases genetics, Cerebral Palsy genetics, Membrane Proteins genetics, Mitochondrial Proteins genetics, Spastic Paraplegia, Hereditary genetics
- Abstract
De novo mutations in ATAD3A (ATPase family AAA-domain containing protein 3A) were recently found to cause a neurological syndrome with developmental delay, hypotonia, spasticity, optic atrophy, axonal neuropathy, and hypertrophic cardiomyopathy. Using whole-exome sequencing, we identified a dominantly inherited heterozygous variant c.1064G > A (p.G355D) in ATAD3A in a mother presenting with hereditary spastic paraplegia (HSP) and axonal neuropathy and her son with dyskinetic cerebral palsy, both with disease onset in childhood. HSP is a clinically and genetically heterogeneous disorder of the upper motor neurons. Symptoms beginning in early childhood may resemble spastic cerebral palsy. The function of ATAD3A, a mitochondrial inner membrane AAA ATPase, is yet undefined. AAA ATPases form hexameric rings, which are catalytically dependent on the co-operation of the subunits. The dominant-negative patient mutation affects the Walker A motif, which is responsible for ATP binding in the AAA module of ATAD3A, and we show that the recombinant mutant ATAD3A protein has a markedly reduced ATPase activity. We further show that overexpression of the mutant ATAD3A fragments the mitochondrial network and induces lysosome mass. Similarly, we observed altered dynamics of the mitochondrial network and increased lysosomes in patient fibroblasts and neurons derived through differentiation of patient-specific induced pluripotent stem cells. These alterations were verified in patient fibroblasts to associate with upregulated basal autophagy through mTOR inactivation, resembling starvation. Mutations in ATAD3A can thus be dominantly inherited and underlie variable neurological phenotypes, including HSP, with intrafamiliar variability. This finding extends the group of mitochondrial inner membrane AAA proteins associated with spasticity., (© The Author 2017. Published by Oxford University Press.)
- Published
- 2017
- Full Text
- View/download PDF
28. Atomistic Molecular Dynamics Simulations of Mitochondrial DNA Polymerase γ: Novel Mechanisms of Function and Pathogenesis.
- Author
-
Euro L, Haapanen O, Róg T, Vattulainen I, Suomalainen A, and Sharma V
- Subjects
- Catalytic Domain, DNA metabolism, DNA Polymerase gamma, Humans, Mutation, Neurodegenerative Diseases enzymology, Neurodegenerative Diseases genetics, Protein Structure, Secondary, Rotation, DNA-Directed DNA Polymerase chemistry, DNA-Directed DNA Polymerase metabolism, Mitochondria enzymology, Molecular Dynamics Simulation
- Abstract
DNA polymerase γ (Pol γ) is a key component of the mitochondrial DNA replisome and an important cause of neurological diseases. Despite the availability of its crystal structures, the molecular mechanism of DNA replication, the switch between polymerase and exonuclease activities, the site of replisomal interactions, and functional effects of patient mutations that do not affect direct catalysis have remained elusive. Here we report the first atomistic classical molecular dynamics simulations of the human Pol γ replicative complex. Our simulation data show that DNA binding triggers remarkable changes in the enzyme structure, including (1) completion of the DNA-binding channel via a dynamic subdomain, which in the apo form blocks the catalytic site, (2) stabilization of the structure through the distal accessory β-subunit, and (3) formation of a putative transient replisome-binding platform in the "intrinsic processivity" subdomain of the enzyme. Our data indicate that noncatalytic mutations may disrupt replisomal interactions, thereby causing Pol γ-associated neurodegenerative disorders.
- Published
- 2017
- Full Text
- View/download PDF
29. Mitochondrial encephalomyopathy and retinoblastoma explained by compound heterozygosity of SUCLA2 point mutation and 13q14 deletion.
- Author
-
Matilainen S, Isohanni P, Euro L, Lönnqvist T, Pihko H, Kivelä T, Knuutila S, and Suomalainen A
- Published
- 2017
- Full Text
- View/download PDF
30. SDHA mutation with dominant transmission results in complex II deficiency with ocular, cardiac, and neurologic involvement.
- Author
-
Courage C, Jackson CB, Hahn D, Euro L, Nuoffer JM, Gallati S, and Schaller A
- Subjects
- Adolescent, Alleles, Amino Acid Substitution, Biomarkers, Codon, DNA Mutational Analysis, Fatal Outcome, Female, Genes, Mitochondrial, Genotype, Humans, Male, Models, Molecular, Pedigree, Protein Conformation, Succinate Dehydrogenase chemistry, Abnormalities, Multiple diagnosis, Abnormalities, Multiple genetics, Electron Transport Complex II deficiency, Mutation, Phenotype, Succinate Dehydrogenase genetics
- Abstract
Isolated defects of the mitochondrial respiratory complex II (succinate dehydrogenase, SDH) are rare, accounting for approximately 2% of all respiratory chain deficiency diagnoses. Here, we report clinical and molecular investigations of three family members with a heterozygous mutation in the large flavoprotein subunit SDHA previously described to cause complex II deficiency. The index patient presented with bilateral optic atrophy and ocular movement disorder, a progressive polyneuropathy, psychiatric involvement, and cardiomyopathy. Two of his children presented with cardiomyopathy and methylglutaconic aciduria in early childhood. The daughter deceased at the age of 7 months due to cardiac insufficiency. The 30-year old son presents with cardiomyopathy and developed bilateral optic atrophy in adulthood. Of the four nuclear encoded proteins composing complex II (SDHA, SDHB, SDHC, SDHD) and currently known assembly factors SDHAF1 and SDHAF2 mainly recessively inherited mutations have been described in SDHA, SDHB, SDHD, and SDHAF1 to be causative for mitochondrial disease phenotypes. This is the second report presenting autosomal dominant inheritance of a SDHA mutation.© 2016 Wiley Periodicals, Inc., (© 2016 Wiley Periodicals, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
31. The rare Costello variant HRAS c.173C>T (p.T58I) with severe neonatal hypertrophic cardiomyopathy.
- Author
-
Hiippala A, Vasilescu C, Tallila J, Alastalo TP, Paetau A, Tyni T, Suomalainen A, Euro L, and Ojala T
- Subjects
- Abnormalities, Multiple diagnosis, Abnormalities, Multiple genetics, Alleles, Biomarkers, DNA Mutational Analysis, Echocardiography, Genetic Association Studies, Genetic Testing, Genotype, Humans, Infant, Newborn, Male, Radiography, Thoracic, Severity of Illness Index, Cardiomyopathy, Hypertrophic diagnosis, Cardiomyopathy, Hypertrophic genetics, Costello Syndrome diagnosis, Costello Syndrome genetics, Mutation, Phenotype, Proto-Oncogene Proteins p21(ras) genetics
- Abstract
We report a 10-year-old girl presenting with severe neonatal hypertrophic cardiomyopathy (HCM), feeding difficulties, mildly abnormal facial features, and progressive skeletal muscle symptoms but with normal cognitive development. Targeted oligonucleotide-selective sequencing of 101 cardiomyopathy genes revealed the genetic diagnosis, and the mutation was verified by Sanger sequencing in the patient and her parents. To offer insights into the potential mechanism of patient mutation, protein structural analysis was performed using the resolved structure of human activated HRAS protein with bound GTP analogue (PDB id 5P21) in Discovery Studio 4.5 (Dassault Systèmes Biovia, San Diego, CA). The patient with hypertrophic cardiomyopathy and normal cognitive development was diagnosed with an HRAS mutation c.173C>T (p.T58I), a milder variant of Costello syndrome affecting a highly conserved amino acid, threonine 58. Our analysis suggests that the p.G12 mutations slow GTP hydrolysis rendering HRAS unresponsive to GTPase activating proteins, and resulting in permanently active state. The p.T58I mutation likely affects binding of guanidine-nucleotide-exchange factors, thereby promoting the active state but also allowing for slow inactivation. Patients with the HRAS mutation c.173C>T (p.T58I) might go undiagnosed because of the milder phenotype compared with other mutations causing Costello syndrome. We expand the clinical and molecular picture of the rare HRAS mutation by reporting the first case in Europe and the fourth case in the literature. Our protein structure analysis offers insights into the mechanism of the mildly activating p.T58I mutation. © 2016 Wiley Periodicals, Inc., (© 2016 Wiley Periodicals, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
32. Mitochondrial DNA Replication Defects Disturb Cellular dNTP Pools and Remodel One-Carbon Metabolism.
- Author
-
Nikkanen J, Forsström S, Euro L, Paetau I, Kohnz RA, Wang L, Chilov D, Viinamäki J, Roivainen A, Marjamäki P, Liljenbäck H, Ahola S, Buzkova J, Terzioglu M, Khan NA, Pirnes-Karhu S, Paetau A, Lönnqvist T, Sajantila A, Isohanni P, Tyynismaa H, Nomura DK, Battersby BJ, Velagapudi V, Carroll CJ, and Suomalainen A
- Subjects
- Adult, Animals, Carbon metabolism, DNA Helicases genetics, DNA Helicases metabolism, DNA Replication, DNA, Mitochondrial genetics, Female, Folic Acid metabolism, Glucose metabolism, Glutathione metabolism, Humans, Male, Mice, Mice, Inbred C57BL, Mitochondria genetics, Mitochondria pathology, Mitochondrial Myopathies genetics, Mitochondrial Myopathies pathology, Mitochondrial Proteins genetics, Mitochondrial Proteins metabolism, Models, Molecular, Mutation, Serine metabolism, Spinocerebellar Degenerations genetics, Spinocerebellar Degenerations pathology, DNA, Mitochondrial metabolism, Mitochondria metabolism, Mitochondrial Myopathies metabolism, Nucleotides metabolism, Spinocerebellar Degenerations metabolism
- Abstract
Mitochondrial dysfunction affects cellular energy metabolism, but less is known about the consequences for cytoplasmic biosynthetic reactions. We report that mtDNA replication disorders caused by TWINKLE mutations-mitochondrial myopathy (MM) and infantile onset spinocerebellar ataxia (IOSCA)-remodel cellular dNTP pools in mice. MM muscle shows tissue-specific induction of the mitochondrial folate cycle, purine metabolism, and imbalanced and increased dNTP pools, consistent with progressive mtDNA mutagenesis. IOSCA-TWINKLE is predicted to hydrolyze dNTPs, consistent with low dNTP pools and mtDNA depletion in the disease. MM muscle also modifies the cytoplasmic one-carbon cycle, transsulfuration, and methylation, as well as increases glucose uptake and its utilization for de novo serine and glutathione biosynthesis. Our evidence indicates that the mitochondrial replication machinery communicates with cytoplasmic dNTP pools and that upregulation of glutathione synthesis through glucose-driven de novo serine biosynthesis contributes to the metabolic stress response. These results are important for disorders with primary or secondary mtDNA instability and offer targets for metabolic therapy., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
33. Mitochondrial encephalomyopathy and retinoblastoma explained by compound heterozygosity of SUCLA2 point mutation and 13q14 deletion.
- Author
-
Matilainen S, Isohanni P, Euro L, Lönnqvist T, Pihko H, Kivelä T, Knuutila S, and Suomalainen A
- Subjects
- Adolescent, Brain pathology, Comparative Genomic Hybridization, Fatal Outcome, Gene Frequency, Humans, Infant, Magnetic Resonance Imaging, Male, Mitochondrial Encephalomyopathies complications, Mitochondrial Encephalomyopathies diagnosis, Models, Molecular, Pedigree, Protein Conformation, Retinoblastoma complications, Retinoblastoma diagnosis, Sequence Analysis, DNA, Structure-Activity Relationship, Succinate-CoA Ligases chemistry, Chromosome Deletion, Chromosomes, Human, Pair 13, Heterozygote, Mitochondrial Encephalomyopathies genetics, Point Mutation, Retinoblastoma genetics, Succinate-CoA Ligases genetics
- Abstract
Mutations in SUCLA2, encoding the ß-subunit of succinyl-CoA synthetase of Krebs cycle, are one cause of mitochondrial DNA depletion syndrome. Patients have been reported to have severe progressive childhood-onset encephalomyopathy, and methylmalonic aciduria, often leading to death in childhood. We studied two families, with children manifesting with slowly progressive mitochondrial encephalomyopathy, hearing impairment and transient methylmalonic aciduria, without mtDNA depletion. The other family also showed dominant inheritance of bilateral retinoblastoma, which coexisted with mitochondrial encephalomyopathy in one patient. We found a variant in SUCLA2 leading to Asp333Gly change, homozygous in one patient and compound heterozygous in one. The latter patient also carried a deletion of 13q14 of the other allele, discovered with molecular karyotyping. The deletion spanned both SUCLA2 and RB1 gene regions, leading to manifestation of both mitochondrial disease and retinoblastoma. We made a homology model for human succinyl-CoA synthetase and used it for structure-function analysis of all reported pathogenic mutations in SUCLA2. On the basis of our model, all previously described mutations were predicted to result in decreased amounts of incorrectly assembled protein or disruption of ADP phosphorylation, explaining the severe early lethal manifestations. However, the Asp333Gly change was predicted to reduce the activity of the otherwise functional enzyme. On the basis of our findings, SUCLA2 mutations should be analyzed in patients with slowly progressive encephalomyopathy, even in the absence of methylmalonic aciduria or mitochondrial DNA depletion. In addition, an encephalomyopathy in a patient with retinoblastoma suggests mutations affecting SUCLA2.
- Published
- 2015
- Full Text
- View/download PDF
34. Structural modeling of tissue-specific mitochondrial alanyl-tRNA synthetase (AARS2) defects predicts differential effects on aminoacylation.
- Author
-
Euro L, Konovalova S, Asin-Cayuela J, Tulinius M, Griffin H, Horvath R, Taylor RW, Chinnery PF, Schara U, Thorburn DR, Suomalainen A, Chihade J, and Tyynismaa H
- Abstract
The accuracy of mitochondrial protein synthesis is dependent on the coordinated action of nuclear-encoded mitochondrial aminoacyl-tRNA synthetases (mtARSs) and the mitochondrial DNA-encoded tRNAs. The recent advances in whole-exome sequencing have revealed the importance of the mtARS proteins for mitochondrial pathophysiology since nearly every nuclear gene for mtARS (out of 19) is now recognized as a disease gene for mitochondrial disease. Typically, defects in each mtARS have been identified in one tissue-specific disease, most commonly affecting the brain, or in one syndrome. However, mutations in the AARS2 gene for mitochondrial alanyl-tRNA synthetase (mtAlaRS) have been reported both in patients with infantile-onset cardiomyopathy and in patients with childhood to adulthood-onset leukoencephalopathy. We present here an investigation of the effects of the described mutations on the structure of the synthetase, in an effort to understand the tissue-specific outcomes of the different mutations. The mtAlaRS differs from the other mtARSs because in addition to the aminoacylation domain, it has a conserved editing domain for deacylating tRNAs that have been mischarged with incorrect amino acids. We show that the cardiomyopathy phenotype results from a single allele, causing an amino acid change R592W in the editing domain of AARS2, whereas the leukodystrophy mutations are located in other domains of the synthetase. Nevertheless, our structural analysis predicts that all mutations reduce the aminoacylation activity of the synthetase, because all mtAlaRS domains contribute to tRNA binding for aminoacylation. According to our model, the cardiomyopathy mutations severely compromise aminoacylation whereas partial activity is retained by the mutation combinations found in the leukodystrophy patients. These predictions provide a hypothesis for the molecular basis of the distinct tissue-specific phenotypic outcomes.
- Published
- 2015
- Full Text
- View/download PDF
35. Effective treatment of mitochondrial myopathy by nicotinamide riboside, a vitamin B3.
- Author
-
Khan NA, Auranen M, Paetau I, Pirinen E, Euro L, Forsström S, Pasila L, Velagapudi V, Carroll CJ, Auwerx J, and Suomalainen A
- Subjects
- Adipose Tissue, Brown drug effects, Adipose Tissue, Brown metabolism, Adipose Tissue, Brown pathology, Animals, Energy Metabolism drug effects, Forkhead Box Protein O1, Forkhead Transcription Factors metabolism, Lipid Metabolism drug effects, Liver drug effects, Liver metabolism, Liver pathology, Male, Mice, Mice, Inbred C57BL, Mitochondria metabolism, Mitochondria pathology, Mitochondrial Myopathies metabolism, Mitochondrial Myopathies pathology, Muscle, Skeletal drug effects, Muscle, Skeletal metabolism, Muscle, Skeletal pathology, NAD metabolism, Niacinamide therapeutic use, Pyridinium Compounds, Sirtuin 1 metabolism, Unfolded Protein Response drug effects, Mitochondria drug effects, Mitochondrial Myopathies drug therapy, Niacinamide analogs & derivatives, Vitamin B Complex therapeutic use
- Abstract
Nutrient availability is the major regulator of life and reproduction, and a complex cellular signaling network has evolved to adapt organisms to fasting. These sensor pathways monitor cellular energy metabolism, especially mitochondrial ATP production and NAD(+)/NADH ratio, as major signals for nutritional state. We hypothesized that these signals would be modified by mitochondrial respiratory chain disease, because of inefficient NADH utilization and ATP production. Oral administration of nicotinamide riboside (NR), a vitamin B3 and NAD(+) precursor, was previously shown to boost NAD(+) levels in mice and to induce mitochondrial biogenesis. Here, we treated mitochondrial myopathy mice with NR. This vitamin effectively delayed early- and late-stage disease progression, by robustly inducing mitochondrial biogenesis in skeletal muscle and brown adipose tissue, preventing mitochondrial ultrastructure abnormalities and mtDNA deletion formation. NR further stimulated mitochondrial unfolded protein response, suggesting its protective role in mitochondrial disease. These results indicate that NR and strategies boosting NAD(+) levels are a promising treatment strategy for mitochondrial myopathy., (© 2014 The Authors. Published under the terms of the CC BY license.)
- Published
- 2014
- Full Text
- View/download PDF
36. Whole-exome sequencing identifies a mutation in the mitochondrial ribosome protein MRPL44 to underlie mitochondrial infantile cardiomyopathy.
- Author
-
Carroll CJ, Isohanni P, Pöyhönen R, Euro L, Richter U, Brilhante V, Götz A, Lahtinen T, Paetau A, Pihko H, Battersby BJ, Tyynismaa H, and Suomalainen A
- Subjects
- Adolescent, Amino Acid Sequence, Cardiomyopathy, Hypertrophic congenital, Cyclooxygenase 1, Electron Transport Complex I, Electron Transport Complex IV, Fatal Outcome, Female, Fibroblasts metabolism, Humans, Infant, Mitochondrial Diseases congenital, Molecular Sequence Data, Muscle, Skeletal chemistry, Muscle, Skeletal metabolism, Myocardium chemistry, Myocardium metabolism, Pedigree, Sequence Alignment, Sequence Analysis, DNA, Cardiomyopathy, Hypertrophic genetics, Exome genetics, Mitochondrial Diseases genetics, Mitochondrial Proteins genetics, Mutation, Ribosomal Proteins genetics
- Abstract
Background: The genetic complexity of infantile cardiomyopathies is remarkable, and the importance of mitochondrial translation defects as a causative factor is only starting to be recognised. We investigated the genetic basis for infantile onset recessive hypertrophic cardiomyopathy in two siblings., Methods and Results: Analysis of respiratory chain enzymes revealed a combined deficiency of complexes I and IV in the heart and skeletal muscle. Exome sequencing uncovered a homozygous mutation (L156R) in MRPL44 of both siblings. MRPL44 encodes a protein in the large subunit of the mitochondrial ribosome and is suggested to locate in close proximity to the tunnel exit of the yeast mitochondrial ribosome. We found severely reduced MRPL44 levels in the patient's heart, skeletal muscle and fibroblasts suggesting that the missense mutation affected the protein stability. In patient fibroblasts, decreased MRPL44 affected assembly of the large ribosomal subunit and stability of 16S rRNA leading to complex IV deficiency. Despite this assembly defect, de novo mitochondrial translation was only mildly affected in fibroblasts suggesting that MRPL44 may have a function in the assembly/stability of nascent mitochondrial polypeptides exiting the ribosome. Retroviral expression of wild-type MRPL44 in patient fibroblasts rescued the large ribosome assembly defect and COX deficiency., Conclusions: These findings indicate that mitochondrial ribosomal subunit defects can generate tissue-specific manifestations, such as cardiomyopathy.
- Published
- 2013
- Full Text
- View/download PDF
37. Mitochondrial phenylalanyl-tRNA synthetase mutations underlie fatal infantile Alpers encephalopathy.
- Author
-
Elo JM, Yadavalli SS, Euro L, Isohanni P, Götz A, Carroll CJ, Valanne L, Alkuraya FS, Uusimaa J, Paetau A, Caruso EM, Pihko H, Ibba M, Tyynismaa H, and Suomalainen A
- Subjects
- Amino Acid Sequence, Anticodon metabolism, Base Sequence, Diffuse Cerebral Sclerosis of Schilder enzymology, Diffuse Cerebral Sclerosis of Schilder metabolism, Exome, Female, Humans, Infant, Mitochondria metabolism, Mitochondrial Diseases enzymology, Mitochondrial Diseases metabolism, Mitochondrial Proteins chemistry, Mitochondrial Proteins metabolism, Molecular Sequence Data, Mutation, Phenylalanine-tRNA Ligase chemistry, Phenylalanine-tRNA Ligase metabolism, Protein Folding, RNA, Transfer genetics, RNA, Transfer metabolism, Diffuse Cerebral Sclerosis of Schilder genetics, Mitochondria enzymology, Mitochondrial Diseases genetics, Mitochondrial Proteins genetics, Phenylalanine-tRNA Ligase genetics
- Abstract
Next-generation sequencing has turned out to be a powerful tool to uncover genetic basis of childhood mitochondrial disorders. We utilized whole-exome analysis and discovered novel compound heterozygous mutations in FARS2 (mitochondrial phenylalanyl transfer RNA synthetase), encoding the mitochondrial phenylalanyl transfer RNA (tRNA) synthetase (mtPheRS) in two patients with fatal epileptic mitochondrial encephalopathy. The mutations affected highly conserved amino acids, p.I329T and p.D391V. Recently, a homozygous FARS2 variant p.Y144C was reported in a Saudi girl with mitochondrial encephalopathy, but the pathogenic role of the variant remained open. Clinical features, including postnatal onset, catastrophic epilepsy, lactic acidemia, early lethality and neuroimaging findings of the patients with FARS2 variants, resembled each other closely, and neuropathology was consistent with Alpers syndrome. Our structural analysis of mtPheRS predicted that p.I329T weakened ATP binding in the aminoacylation domain, and in vitro studies with recombinant mutant protein showed decreased affinity of this variant to ATP. Furthermore, p.D391V and p.Y144C were predicted to disrupt synthetase function by interrupting the rotation of the tRNA anticodon stem-binding domain from a closed to an open form. In vitro characterization indicated reduced affinity of p.D391V mutant protein to phenylalanine, whereas p.Y144C disrupted tRNA binding. The stability of p.I329T and p.D391V mutants in a refolding assay was impaired. Our results imply that the three FARS2 mutations directly impair aminoacylation function and stability of mtPheRS, leading to a decrease in overall tRNA charging capacity. This study establishes a new genetic cause of infantile mitochondrial Alpers encephalopathy and reports a new mitochondrial aminoacyl-tRNA synthetase as a cause of mitochondrial disease.
- Published
- 2012
- Full Text
- View/download PDF
38. Carrier frequency of a common mutation of carnitine palmitoyltransferase 1A deficiency and long-term follow-up in Finland.
- Author
-
Roomets E, Polinati PP, Euro L, Eskelin PM, Paganus A, and Tyni T
- Subjects
- Adolescent, Adult, Blotting, Western, Carnitine O-Palmitoyltransferase deficiency, Carnitine O-Palmitoyltransferase metabolism, Child, Child Development, Child, Preschool, Diet Therapy, Female, Finland epidemiology, Follow-Up Studies, Genetic Association Studies, Humans, Male, Young Adult, Carnitine O-Palmitoyltransferase genetics, Heterozygote, Mutation, Missense
- Abstract
Objective: To assess the long-term clinical course of carnitine palmitoyltransferase 1A (CPT1A) deficiency, caused by the c.1364A>C (p.K455T) mutation, and the carrier frequency of this mutation in Finland., Study Design: This was a long-term follow-up of patients in whom the common mutation was detected., Results: Between 1999 and 2010, 6 cases of CPT1A deficiency were diagnosed and treated with a high-carbohydrate, low-fat diet. The patients experienced their first symptoms during the first years of life, provoked by viral illness and/or fasting. The clinical features included hypoketotic hypoglycemia, hepatopathy, and loss of consciousness, ranging from transient unconsciousness to prolonged hyperlipidemic coma. Five cases carried a homozygous c.1364A>C (p.K455T) mutation, whereas 1 case had a compound c.1364A>C/c.1493A>C (p.Y498S) mutation. During dietary therapy, the patients had few transient decompensations. No carriers of mutation c.1364A>C were detected by minisequencing of 150 control samples., Conclusion: Even though CPT1A deficiency may be life-threatening and lead to prolonged coma, the long-term prognosis is good. A genotype-phenotype correlation implies that the mutations detected are disease-causing. Despite Finland's location close to the Arctic polar region, the carrier frequency of the c.1364A>C mutation in Finland is far lower than that of the variants found in Alaskan, Canadian, and Greenland native populations., (Copyright © 2012 Mosby, Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
39. Clustering of Alpers disease mutations and catalytic defects in biochemical variants reveal new features of molecular mechanism of the human mitochondrial replicase, Pol γ.
- Author
-
Euro L, Farnum GA, Palin E, Suomalainen A, and Kaguni LS
- Subjects
- Biocatalysis, Catalytic Domain genetics, DNA Polymerase gamma, DNA-Directed DNA Polymerase metabolism, Humans, Mitochondria enzymology, DNA-Directed DNA Polymerase chemistry, DNA-Directed DNA Polymerase genetics, Diffuse Cerebral Sclerosis of Schilder genetics, Mutation
- Abstract
Mutations in Pol γ represent a major cause of human mitochondrial diseases, especially those affecting the nervous system in adults and in children. Recessive mutations in Pol γ represent nearly half of those reported to date, and they are nearly uniformly distributed along the length of the POLG1 gene (Human DNA Polymerase gamma Mutation Database); the majority of them are linked to the most severe form of POLG syndrome, Alpers-Huttenlocher syndrome. In this report, we assess the structure-function relationships for recessive disease mutations by reviewing existing biochemical data on site-directed mutagenesis of the human, Drosophila and yeast Pol γs, and their homologs from the family A DNA polymerase group. We do so in the context of a molecular model of Pol γ in complex with primer-template DNA, which we have developed based upon the recently solved crystal structure of the apoenzyme form. We present evidence that recessive mutations cluster within five distinct functional modules in the catalytic core of Pol γ. Our results suggest that cluster prediction can be used as a diagnosis-supporting tool to evaluate the pathogenic role of new Pol γ variants.
- Published
- 2011
- Full Text
- View/download PDF
40. Exome sequencing identifies mitochondrial alanyl-tRNA synthetase mutations in infantile mitochondrial cardiomyopathy.
- Author
-
Götz A, Tyynismaa H, Euro L, Ellonen P, Hyötyläinen T, Ojala T, Hämäläinen RH, Tommiska J, Raivio T, Oresic M, Karikoski R, Tammela O, Simola KO, Paetau A, Tyni T, and Suomalainen A
- Subjects
- Base Pairing, Cardiomyopathy, Hypertrophic pathology, DNA Mutational Analysis, DNA, Mitochondrial genetics, Female, Humans, Infant, Infant, Newborn, Mitochondrial Diseases pathology, Pedigree, Protein Structure, Tertiary, Alanine-tRNA Ligase genetics, Cardiomyopathy, Hypertrophic genetics, Mitochondria genetics, Mitochondrial Diseases genetics, Mutation, Missense
- Abstract
Infantile cardiomyopathies are devastating fatal disorders of the neonatal period or the first year of life. Mitochondrial dysfunction is a common cause of this group of diseases, but the underlying gene defects have been characterized in only a minority of cases, because tissue specificity of the manifestation hampers functional cloning and the heterogeneity of causative factors hinders collection of informative family materials. We sequenced the exome of a patient who died at the age of 10 months of hypertrophic mitochondrial cardiomyopathy with combined cardiac respiratory chain complex I and IV deficiency. Rigorous data analysis allowed us to identify a homozygous missense mutation in AARS2, which we showed to encode the mitochondrial alanyl-tRNA synthetase (mtAlaRS). Two siblings from another family, both of whom died perinatally of hypertrophic cardiomyopathy, had the same mutation, compound heterozygous with another missense mutation. Protein structure modeling of mtAlaRS suggested that one of the mutations affected a unique tRNA recognition site in the editing domain, leading to incorrect tRNA aminoacylation, whereas the second mutation severely disturbed the catalytic function, preventing tRNA aminoacylation. We show here that mutations in AARS2 cause perinatal or infantile cardiomyopathy with near-total combined mitochondrial respiratory chain deficiency in the heart. Our results indicate that exome sequencing is a powerful tool for identifying mutations in single patients and allows recognition of the genetic background in single-gene disorders of variable clinical manifestation and tissue-specific disease. Furthermore, we show that mitochondrial disorders extend to prenatal life and are an important cause of early infantile cardiac failure., (Copyright © 2011 The American Society of Human Genetics. Published by Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
41. Functional analysis of H. sapiens DNA polymerase gamma spacer mutation W748S with and without common variant E1143G.
- Author
-
Palin EJ, Lesonen A, Farr CL, Euro L, Suomalainen A, and Kaguni LS
- Subjects
- Amino Acid Substitution, Catalytic Domain genetics, Cells, Cultured, DNA Polymerase gamma, DNA Primers genetics, DNA, Intergenic genetics, DNA, Mitochondrial genetics, DNA-Directed DNA Polymerase chemistry, Genes, Recessive, Humans, In Vitro Techniques, Models, Molecular, Mutagenesis, Site-Directed, Protein Subunits, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Spinocerebellar Degenerations enzymology, Spinocerebellar Degenerations genetics, Syndrome, DNA-Directed DNA Polymerase genetics, DNA-Directed DNA Polymerase metabolism, Genetic Variation, Mutation, Missense
- Abstract
Mitochondrial DNA polymerase, POLG, is the sole DNA polymerase found in animal mitochondria. In humans, POLGalpha W748S in cis with an E1143G mutation has been linked to a new type of recessive ataxia, MIRAS, which is the most common inherited ataxia in Finland. We investigated the biochemical phenotypes of the W748S amino acid change, using recombinant human POLG. We measured processive and non-processive DNA polymerase activity, DNA binding affinity, enzyme processivity, and subunit interaction with recombinant POLGbeta. In addition, we studied the effects of the W748S and E1143G mutations in primary human cell cultures using retroviral transduction. Here, we examined cell viability, mitochondrial DNA copy number, and products of mitochondrial translation. Our results indicate that the W748S mutant POLGalpha does not exhibit a clear biochemical phenotype, making it indistinguishable from wild type POLGalpha and as such, fail to replicate previously published results. Furthermore, results from the cell models were concurrent with the findings from patients, and support our biochemical findings., (Copyright 2010 Elsevier B.V. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
42. High affinity cation-binding sites in Complex I from Escherichia coli.
- Author
-
Euro L, Belevich G, Wikström M, and Verkhovskaya M
- Subjects
- Binding Sites, Cations metabolism, Electron Transport Complex I antagonists & inhibitors, Electron Transport Complex I genetics, Electron Transport Complex I isolation & purification, Escherichia coli genetics, Escherichia coli Proteins antagonists & inhibitors, Escherichia coli Proteins chemistry, Escherichia coli Proteins genetics, Kinetics, Lanthanum metabolism, Lanthanum pharmacology, Mutation, Potassium metabolism, Potassium pharmacology, Protein Binding, Quinone Reductases antagonists & inhibitors, Quinone Reductases genetics, Quinone Reductases metabolism, Electron Transport Complex I metabolism, Escherichia coli enzymology, Escherichia coli Proteins metabolism
- Abstract
Studies on the activity of Complex I from Escherichia coli in the presence of different metal cations revealed at least two high affinity metal-binding sites. Membrane-bound or isolated Complex I was activated by K(+) (apparent binding constant approximately 125 microM) and inhibited by La(3+) (IC(50)= 1 microM). K(+) and La(3+) do not occupy the same site. Possible localization of these metal-binding sites and their implication in catalysis are discussed.
- Published
- 2009
- Full Text
- View/download PDF
43. The role of the invariant glutamate 95 in the catalytic site of Complex I from Escherichia coli.
- Author
-
Euro L, Belevich G, Bloch DA, Verkhovsky MI, Wikström M, and Verkhovskaya M
- Subjects
- Catalytic Domain, Electron Transport Complex I genetics, Electron Transport Complex I metabolism, Escherichia coli Proteins genetics, Escherichia coli Proteins metabolism, Kinetics, Membrane Proteins chemistry, Membrane Proteins genetics, Membrane Proteins metabolism, Models, Molecular, Mutation, NAD metabolism, Oxidation-Reduction, Protein Binding, Quinone Reductases chemistry, Quinone Reductases genetics, Quinone Reductases metabolism, Electron Transport Complex I chemistry, Escherichia coli Proteins chemistry, Glutamine genetics
- Abstract
Replacement of glutamate 95 for glutamine in the NADH- and FMN-binding NuoF subunit of E. coli Complex I decreased NADH oxidation activity 2.5-4.8 times depending on the used electron acceptor. The apparent K(m) for NADH was 5.2 and 10.4 microM for the mutant and wild type, respectively. Analysis of the inhibitory effect of NAD(+) on activity showed that the E95Q mutation caused a 2.4-fold decrease of K(i)(NAD+) in comparison to the wild type enzyme. ADP-ribose, which differs from NAD(+) by the absence of the positively charged nicotinamide moiety, is also a competitive inhibitor of NADH binding. The mutation caused a 7.5-fold decrease of K(i)(ADP-ribose) relative to wild type enzyme. Based on these findings we propose that the negative charge of Glu95 accelerates turnover of Complex I by electrostatic interaction with the negatively charged phosphate groups of the substrate nucleotide during operation, which facilitates release of the product NAD(+). The E95Q mutation was also found to cause a positive shift of the midpoint redox potential of the FMN, from -350 mV to -310 mV, which suggests that the negative charge of Glu95 is also involved in decreasing the midpoint potential of the primary electron acceptor of Complex I.
- Published
- 2009
- Full Text
- View/download PDF
44. Conserved lysine residues of the membrane subunit NuoM are involved in energy conversion by the proton-pumping NADH:ubiquinone oxidoreductase (Complex I).
- Author
-
Euro L, Belevich G, Verkhovsky MI, Wikström M, and Verkhovskaya M
- Subjects
- Amino Acid Sequence, Electron Transport Complex I chemistry, Escherichia coli enzymology, Escherichia coli growth & development, Molecular Sequence Data, Mutagenesis, Site-Directed, Mutant Proteins metabolism, Mutation genetics, Phenotype, Protein Structure, Secondary, Protein Subunits chemistry, Protein Subunits metabolism, Sequence Analysis, Protein, Cell Membrane metabolism, Conserved Sequence, Electron Transport Complex I metabolism, Energy Transfer, Escherichia coli Proteins chemistry, Escherichia coli Proteins metabolism, Lysine metabolism, NADH Dehydrogenase chemistry, NADH Dehydrogenase metabolism
- Abstract
Analysis of the amino acid sequences of subunits NuoM and NuoN in the membrane domain of Complex I revealed a clear common pattern, including two lysines that are predicted to be located within the membrane, and which are important for quinone reductase activity. Site-directed mutations of the amino acid residues E144, K234, K265 and W243 in this pattern were introduced into the chromosomal gene nuoM of Escherichia coli Complex I. The activity of mutated Complex I was studied in both membranes and in purified Complex I. The quinone reductase activity was practically lost in K234A, K234R and E144A, decreased in W243A and K265A but unchanged in E144D. Complex I from all these mutants contained 1 mol tightly bound ubiquinone per mol FMN like wild type enzyme. The mutant enzymes E144D, W243A and K265A had wild type sensitivity to rolliniastatin and complete proton-pumping efficiency of Complex I. Remarkably, the subunits NuoL and NuoH in the membrane domain also appear to contain conserved lysine residues in transmembrane helices, which may give a clue of the mechanism of proton translocation. A tentative principle of proton translocation by Complex I is suggested based on electrostatic interactions of lysines in the membrane subunits.
- Published
- 2008
- Full Text
- View/download PDF
45. Real-time electron transfer in respiratory complex I.
- Author
-
Verkhovskaya ML, Belevich N, Euro L, Wikström M, and Verkhovsky MI
- Subjects
- Electron Spin Resonance Spectroscopy, Electron Transport, Kinetics, NAD, Oxidation-Reduction, Thermus thermophilus, Electron Transport Complex I chemistry, Escherichia coli chemistry
- Abstract
Electron transfer in complex I from Escherichia coli was investigated by an ultrafast freeze-quench approach. The reaction of complex I with NADH was stopped in the time domain from 90 mus to 8 ms and analyzed by electron paramagnetic resonance (EPR) spectroscopy at low temperatures. The data show that after binding of the first molecule of NADH, two electrons move via the FMN cofactor to the iron-sulfur (Fe/S) centers N1a and N2 with an apparent time constant of approximately 90 mus, implying that these two centers should have the highest redox potential in the enzyme. The rate of reduction of center N2 (the last center in the electron transfer sequence) is close to that predicted by electron transfer theory, which argues for the absence of coupled proton transfer or conformational changes during electron transfer from FMN to N2. After fast reduction of N1a and N2, we observe a slow, approximately 1-ms component of reduction of other Fe/S clusters. Because all elementary electron transfer rates between clusters are several orders of magnitude higher than this observed rate, we conclude that the millisecond component is limited by a single process corresponding to dissociation of the oxidized NAD(+) molecule from its binding site, where it prevents entry of the next NADH molecule. Despite the presence of approximately one ubiquinone per enzyme molecule, no transient semiquinone formation was observed, which has mechanistic implications, suggesting a high thermodynamic barrier for ubiquinone reduction to the semiquinone radical. Possible consequences of these findings for the proton translocation mechanism are discussed.
- Published
- 2008
- Full Text
- View/download PDF
46. Electrostatic interactions between FeS clusters in NADH:ubiquinone oxidoreductase (Complex I) from Escherichia coli.
- Author
-
Euro L, Bloch DA, Wikström M, Verkhovsky MI, and Verkhovskaya M
- Subjects
- Electron Spin Resonance Spectroscopy, Electron Transport Complex I chemistry, Escherichia coli enzymology, Escherichia coli Proteins chemistry, Flavin Mononucleotide chemistry, Flavin Mononucleotide metabolism, Hydrogen-Ion Concentration, Iron-Sulfur Proteins chemistry, Iron-Sulfur Proteins metabolism, Oxidation-Reduction, Static Electricity, Electron Transport Complex I metabolism, Escherichia coli metabolism, Escherichia coli Proteins metabolism
- Abstract
The redox properties of the cofactors of NADH:ubiquinone oxidoreductase (complex I) from Escherichia coli were studied by following the changes in electron paramagnetic resonance (EPR) and optical spectra upon electrochemical redox titration of the purified protein. At neutral pH, the FMN cofactor had a midpoint redox potential ( E m) approximately -350 mV ( n = 2). Binuclear FeS clusters were well-characterized: N1a was titrated with a single ( n = 1) transition, and E m = -235 mV. In contrast, the titration of N1b can only be fitted with the sum of at least two one-electron Nernstian curves with E m values of -245 and -320 mV. The tetranuclear clusters can also be separated into two groups, either having a single, n = 1, or more complex redox titration curves. The titration curves of the EPR bands attributed to the tetranuclear clusters N2 ( g = 2.045 and g = 1.895) and N6b ( g = 2.089 and g = 1.877) can be presented by the sum of at least two components, each with E m (app) approximately -200/-300 mV and -235/-315 mV, respectively. The titration of the signals at g = 1.956-1.947 (N3 or N7, E m = -315 mV), g = 2.022, and g = 1.932 (Nx, -365 mV) and the low temperature signal at g = 1.929 (N4 or N5, -330 mV) followed Nernstian n = 1 curves. The observed redox titration curves are discussed in terms of intrinsic electrostatic interactions between FeS centers in complex I. A model showing shifts of E m due to the electrostatic interaction between the centers is presented.
- Published
- 2008
- Full Text
- View/download PDF
47. Role of the conserved arginine 274 and histidine 224 and 228 residues in the NuoCD subunit of complex I from Escherichia coli.
- Author
-
Belevich G, Euro L, Wikström M, and Verkhovskaya M
- Subjects
- Amino Acid Substitution, Arginine chemistry, Base Sequence, Conserved Sequence, DNA, Bacterial genetics, Electron Spin Resonance Spectroscopy, Electron Transport Complex I genetics, Electron Transport Complex I metabolism, Escherichia coli enzymology, Escherichia coli genetics, Escherichia coli Proteins genetics, Escherichia coli Proteins metabolism, Histidine chemistry, Kinetics, Mutagenesis, Site-Directed, Oxidation-Reduction, Protein Subunits, Electron Transport Complex I chemistry, Escherichia coli Proteins chemistry
- Abstract
The conserved arginine 274 and histidine 224 and 228 residues in subunit NuoCD of complex I from Escherichia coli were substituted for alanine. The wild-type and mutated NuoCD subunit was expressed on a plasmid in an E. coli strain bearing a nuoCD deletion. Complex I was fully expressed in the H224A and H228A mutants, whereas the R274A mutation yielded approximately 50% expression. Ubiquinone reductase activity of complex I was studied in membranes and with purified enzyme and was 50% and 30% of the wild-type activity in the H224A and H228A mutants, respectively. The activity of R274A was less than 5% of the wild type in membranes but 20% in purified complex I. Rolliniastatin inhibited quinone reductase activity in the mutants with similar affinity as in the wild type, indicating that the quinone-binding site was not significantly altered by the mutations. Ubiquinone-dependent superoxide production by complex I was similar to the wild type in the R274A mutant but slightly higher in the H224A and H228A mutants. The EPR spectra of purified complex I from the H224A and H228A mutants did not differ from the wild type. In contrast, the signals of the N2 cluster and another fast-relaxing [4Fe-4S] cluster, tentatively assigned as N6b, were drastically decreased in the NADH-reduced R274A mutant enzyme but reappeared on further reduction with dithionite. These findings show that the redox potential of the N2 and N6b centers is shifted to more negative values by the R274A mutation. Purified complex I was reconstituted into liposomes, and electric potential was generated across the membrane upon NADH addition in all three mutant enzymes, suggesting that none of the mutations directly affect the proton-pumping machinery.
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.