MPV17 Loss Causes Deoxynucleotide Insufficiency and Slow DNA Replication in Mitochondria

Ilaria Dalla Rosa, Yolanda Cámara, Romina Durigon, Chloe F Moss, Sara Vidoni, Gokhan Akman, Lilian Hunt, Mark A Johnson, Sarah Grocott, Liya Wang, David R Thorburn, Michio Hirano, Joanna Poulton, Robert W Taylor, Greg Elgar, Ramon Martí, Peter Voshol, Ian J Holt, Antonella Spinazzola, Ilaria Dalla Rosa, Yolanda Cámara, Romina Durigon, Chloe F Moss, Sara Vidoni, Gokhan Akman, Lilian Hunt, Mark A Johnson, Sarah Grocott, Liya Wang, David R Thorburn, Michio Hirano, Joanna Poulton, Robert W Taylor, Greg Elgar, Ramon Martí, Peter Voshol, Ian J Holt, Antonella Spinazzola

Abstract

MPV17 is a mitochondrial inner membrane protein whose dysfunction causes mitochondrial DNA abnormalities and disease by an unknown mechanism. Perturbations of deoxynucleoside triphosphate (dNTP) pools are a recognized cause of mitochondrial genomic instability; therefore, we determined DNA copy number and dNTP levels in mitochondria of two models of MPV17 deficiency. In Mpv17 ablated mice, liver mitochondria showed substantial decreases in the levels of dGTP and dTTP and severe mitochondrial DNA depletion, whereas the dNTP pool was not significantly altered in kidney and brain mitochondria that had near normal levels of DNA. The shortage of mitochondrial dNTPs in Mpv17-/- liver slows the DNA replication in the organelle, as evidenced by the elevated level of replication intermediates. Quiescent fibroblasts of MPV17-mutant patients recapitulate key features of the primary affected tissue of the Mpv17-/- mice, displaying virtual absence of the protein, decreased dNTP levels and mitochondrial DNA depletion. Notably, the mitochondrial DNA loss in the patients' quiescent fibroblasts was prevented and rescued by deoxynucleoside supplementation. Thus, our study establishes dNTP insufficiency in the mitochondria as the cause of mitochondrial DNA depletion in MPV17 deficiency, and identifies deoxynucleoside supplementation as a potential therapeutic strategy for MPV17-related disease. Moreover, changes in the expression of factors involved in mitochondrial deoxynucleotide homeostasis indicate a remodeling of nucleotide metabolism in MPV17 disease models, which suggests mitochondria lacking functional MPV17 have a restricted purine mitochondrial salvage pathway.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Fig 1. Mpv17 ablation in CFW mice…
Fig 1. Mpv17 ablation in CFW mice causes liver specific mtDNA depletion and deficiency of all respiratory chain complexes that contain mtDNA-encoded subunits.
(A-C) Mpv17 expression (A), mtDNA copy number (B) and steady state levels of OXPHOS subunits (C) in the liver, kidney and brain of wild-type (WT, Mpv17+/+) and knockout (KO, Mpv17-/-) mice. (B) Quantification of mtDNA in WT and KO mice. Data are expressed as mean ± SEM of n = 6. (Student test, *** P<0.001, NS, not significant, p>0.05). (D) BN-PAGE analysis of OXPHOS complexes in the liver of WT and KO mice. Sup-C, supercomplexes, Sub-C, subcomplexes of the OXPHOS system. Vdac levels were used as a loading control on a 12% SDS-PAGE gel.
Fig 2. Mpv17 -/- mouse liver mitochondria…
Fig 2. Mpv17-/- mouse liver mitochondria have significantly reduced levels of two precursors of DNA synthesis, dGTP and dTTP.
The abundance of dNTPs was determined by means of an extension assay after extraction of nucleotides from liver (A), kidney (B) or brain (C) of Mpv17+/+ and Mpv17-/- mice (see Methods). All mice (n = 5) were sacrificed between 8 and 10 weeks of age. P values were obtained using Mann-Whitney test (* P<0.05, NS, not significant P>0.05).
Fig 3. Mpv17 ablation results in a…
Fig 3. Mpv17 ablation results in a marked increase of mtDNA replication intermediates.
Analysis of mtDNA replication intermediates in the liver of WT and Mpv17-/- mice. Mitochondrial DNAs from six Mpv17-/- and six control livers were isolated, digested with BclI and fractionated by 2D-AGE and blot hybridized to a probe to the major non-coding region (NCR) of the murine mitochondrial genome.
Fig 4. MPV17 is upregulated in non-dividing…
Fig 4. MPV17 is upregulated in non-dividing cells and quiescence induces mtDNA depletion in MPV17-deficient human fibroblasts, unless supplemented with deoxynucleosides.
(A) Steady state levels of MPV17 protein in proliferating and quiescent control fibroblasts. (B) Relative mtDNA levels in proliferating control (black) and MPV17 mutant fibroblasts (gray). (C) MtDNA quantification in quiescent fibroblasts. The mtDNA amount is expressed relative to the mean of the controls. (D) mtDNA copy number of quiescent control and MPV17-mutant fibroblasts. Data are expressed as mean ± SEM of n = 8. (Student’s t test: ***P<0.001). (E) Mitochondrial dNTP levels in quiescent control and MPV17-mutant fibroblasts. dATP levels were disregarded owing to the low values obtained for the controls. dCTP and dGTP were measured in 3 independent experiments in 3 patients and 3 control cell lines. *** P<0.001—Mann-Whitney test. (F) Relative mtDNA copy number in quiescent fibroblasts supplemented with deoxynucleosides. Quiescent fibroblasts were cultured for 14 days in the absence or presence of 50 μM or 100 μM deoxyadenosine (AdR), deoxycytosine (CdR), deoxyguanosine (GdR), deoxythymidine (TdR) alone or in the different combinations as indicated. The amount of mtDNA is expressed relative to its amount in proliferating cells (Student’s t test: *P<0.05; **P<0.01; ***P<0.001); C1 and C2, control fibroblasts; P1-P5, fibroblasts derived from patients with pathological mutations in MPV17 (S2 Table).
Fig 5. Deoxynucleoside supplementation enables full mtDNA…
Fig 5. Deoxynucleoside supplementation enables full mtDNA recovery in MPV17-deficient fibroblasts after drug-induced transient depletion.
(A) Slow recovery of mtDNA copy number in quiescent MPV17-mutant fibroblasts after depletion with ethidium bromide (EB). Two control (black) and four MPV17 patient-derived cell lines (red) were tested in two or three independent experiments. (Student’s t test: ***P<0.001) (B) MtDNA recovery in control (black) and MPV17-deficient fibroblasts (red) with or without deoxynucleoside supplementation (open and closed circles, respectively) of 100 μM AdR, CdR, and GdR. Data are expressed as mean ± SEM of two control and three MPV17 patient-derived cell lines, tested in two independent experiments (Student’s t test: *P<0.05). (C) MtDNA copy number was monitored via Q-PCR during and after ethidium bromide (EB)-induced mtDNA depletion (as per Fig 5B). C1 –control (black) and P4 MPV17-deficient fibroblast (red), with or without (open and closed circles, respectively) 50 μM of the indicated deoxynucleosides.
Fig 6. The abundance of the mitochondrial…
Fig 6. The abundance of the mitochondrial deoxynucleotide (di- and tri- phosphate) transporters is increased in the absence of MPV17.
Representative immunoblot of the equilibrative nucleoside transporter (ENT1) in (A) control and MPV17-mutant fibroblasts in dividing and quiescent cells, and (B) in the liver of wild-type (WT) and knockout (KO) mice. (C) Steady state levels of the mitochondrial deoxynucleotide transporters 1 and 2 (PNC1 and PNC2) in control and MPV17 deficient fibroblasts in proliferating and quiescent conditions. (D) Pnc1 and Pnc2 steady state levels in the liver, of type (WT) and knockout (KO) mice. Vinculin (VCL), and Tom20 were used as loading control.
Fig 7. MPV17 loss of function affects…
Fig 7. MPV17 loss of function affects the purine branch of mitochondrial salvage pathway.
Representative immunoblot thymidine kinase 2 (TK2) in (A) control and MPV17-mutant fibroblasts in dividing and quiescent cells, and (B) in the liver of wild-type (WT) and knockout (KO) mice. (C) Steady state levels of adenylate kinase 2 and 3 (AK2 and AK3) and Deoxyguanosine Kinase (Dguok) in the liver of wild-type (WT) and knockout (KO) mice. The arrow indicates the Dguok isoform downregulated in KO mouse liver. The samples were from 2 month-old mice unless indicated. (D) AK2, AK3 and DGUOK steady state levels in control and MPV17 mutant fibroblasts in proliferating and quiescent cells. Vinculin, GAPDH, and Tom20 were used as loading control.

References

    1. Spinazzola A, Zeviani M (2009) Disorders from perturbations of nuclear-mitochondrial intergenomic cross-talk. J Intern Med 265: 174–192. 10.1111/j.1365-2796.2008.02059.x
    1. Mandel H, Szargel R, Labay V, Elpeleg O, Saada A, et al. (2001) The deoxyguanosine kinase gene is mutated in individuals with depleted hepatocerebral mitochondrial DNA. Nat Genet 29: 337–341.
    1. Saada A, Shaag A, Mandel H, Nevo Y, Eriksson S, et al. (2001) Mutant mitochondrial thymidine kinase in mitochondrial DNA depletion myopathy. Nat Genet 29: 342–344.
    1. Bourdon A, Minai L, Serre V, Jais JP, Sarzi E, et al. (2007) Mutation of RRM2B, encoding p53-controlled ribonucleotide reductase (p53R2), causes severe mitochondrial DNA depletion. Nat Genet 39: 776–780.
    1. Naviaux RK, Nguyen KV (2004) POLG mutations associated with Alpers' syndrome and mitochondrial DNA depletion. Ann Neurol 55: 706–712.
    1. Alston CL, Schaefer AM, Raman P, Solaroli N, Krishnan KJ, et al. (2013) Late-onset respiratory failure due to TK2 mutations causing multiple mtDNA deletions. Neurology 81: 2051–2053. 10.1212/01.wnl.0000436931.94291.e6
    1. Nishino I, Spinazzola A, Hirano M (1999) Thymidine phosphorylase gene mutations in MNGIE, a human mitochondrial disorder. Science 283: 689–692.
    1. Ronchi D, Garone C, Bordoni A, Gutierrez Rios P, Calvo SE, et al. (2012) Next-generation sequencing reveals DGUOK mutations in adult patients with mitochondrial DNA multiple deletions. Brain 135: 3404–3415. 10.1093/brain/aws258
    1. Spelbrink JN, Li FY, Tiranti V, Nikali K, Yuan QP, et al. (2001) Human mitochondrial DNA deletions associated with mutations in the gene encoding Twinkle, a phage T7 gene 4-like protein localized in mitochondria. Nat Genet 28: 223–231.
    1. Tyynismaa H, Sun R, Ahola-Erkkila S, Almusa H, Poyhonen R, et al. (2012) Thymidine kinase 2 mutations in autosomal recessive progressive external ophthalmoplegia with multiple mitochondrial DNA deletions. Hum Mol Genet 21: 66–75. 10.1093/hmg/ddr438
    1. Van Goethem G, Dermaut B, Lofgren A, Martin JJ, Van Broeckhoven C (2001) Mutation of POLG is associated with progressive external ophthalmoplegia characterized by mtDNA deletions. Nat Genet 28: 211–212.
    1. Tyynismaa H, Ylikallio E, Patel M, Molnar MJ, Haller RG, et al. (2009) A heterozygous truncating mutation in RRM2B causes autosomal-dominant progressive external ophthalmoplegia with multiple mtDNA deletions. Am J Hum Genet 85: 290–295. 10.1016/j.ajhg.2009.07.009
    1. Sarzi E, Goffart S, Serre V, Chretien D, Slama A, et al. (2007) Twinkle helicase (PEO1) gene mutation causes mitochondrial DNA depletion. Ann Neurol 62: 579–587.
    1. Spinazzola A, Viscomi C, Fernandez-Vizarra E, Carrara F, D'Adamo P, et al. (2006) MPV17 encodes an inner mitochondrial membrane protein and is mutated in infantile hepatic mitochondrial DNA depletion. Nat Genet 38: 570–575.
    1. Blakely EL, Butterworth A, Hadden RD, Bodi I, He L, et al. (2012) MPV17 mutation causes neuropathy and leukoencephalopathy with multiple mtDNA deletions in muscle. Neuromuscul Disord 22: 587–591. 10.1016/j.nmd.2012.03.006
    1. Garone C, Rubio JC, Calvo SE, Naini A, Tanji K, et al. (2012) MPV17 Mutations Causing Adult-Onset Multisystemic Disorder With Multiple Mitochondrial DNA Deletions. Arch Neurol 69: 1648–1651. 10.1001/archneurol.2012.405
    1. Pontarin G, Ferraro P, Hakansson P, Thelander L, Reichard P, et al. (2007) p53R2-dependent ribonucleotide reduction provides deoxyribonucleotides in quiescent human fibroblasts in the absence of induced DNA damage. J Biol Chem 282: 16820–16828.
    1. Hakansson P, Hofer A, Thelander L (2006) Regulation of mammalian ribonucleotide reduction and dNTP pools after DNA damage and in resting cells. J Biol Chem 281: 7834–7841.
    1. Tanaka H, Arakawa H, Yamaguchi T, Shiraishi K, Fukuda S, et al. (2000) A ribonucleotide reductase gene involved in a p53-dependent cell-cycle checkpoint for DNA damage. Nature 404: 42–49.
    1. Pontarin G, Ferraro P, Bee L, Reichard P, Bianchi V (2012) Mammalian ribonucleotide reductase subunit p53R2 is required for mitochondrial DNA replication and DNA repair in quiescent cells. Proc Natl Acad Sci U S A 109: 13302–13307. 10.1073/pnas.1211289109
    1. Besse A, Wu P, Bruni F, Donti T, Graham BH, et al. (2015) The GABA transaminase, ABAT, is essential for mitochondrial nucleoside metabolism. Cell Metab 21: 417–427. 10.1016/j.cmet.2015.02.008
    1. Spinazzola A, Marti R, Nishino I, Andreu AL, Naini A, et al. (2002) Altered thymidine metabolism due to defects of thymidine phosphorylase. J Biol Chem 277: 4128–4133.
    1. Nishigaki Y, Marti R, Copeland WC, Hirano M (2003) Site-specific somatic mitochondrial DNA point mutations in patients with thymidine phosphorylase deficiency. J Clin Invest 111: 1913–1921.
    1. Gonzalez-Vioque E, Torres-Torronteras J, Andreu AL, Marti R (2011) Limited dCTP availability accounts for mitochondrial DNA depletion in mitochondrial neurogastrointestinal encephalomyopathy (MNGIE). PLoS Genet 7: e1002035 10.1371/journal.pgen.1002035
    1. Song S, Wheeler LJ, Mathews CK (2003) Deoxyribonucleotide pool imbalance stimulates deletions in HeLa cell mitochondrial DNA. J Biol Chem 278: 43893–43896.
    1. Gandhi VV, Samuels DC (2011) Enzyme kinetics of the mitochondrial deoxyribonucleoside salvage pathway are not sufficient to support rapid mtDNA replication. PLoS Comput Biol 7: e1002078 10.1371/journal.pcbi.1002078
    1. Young P, Leeds JM, Slabaugh MB, Mathews CK (1994) Ribonucleotide reductase: evidence for specific association with HeLa cell mitochondria. Biochem Biophys Res Commun 203: 46–52.
    1. Chimploy K, Song S, Wheeler LJ, Mathews CK (2013) Ribonucleotide reductase association with mammalian liver mitochondria. J Biol Chem 288: 13145–13155. 10.1074/jbc.M113.461111
    1. Anderson DD, Quintero CM, Stover PJ (2011) Identification of a de novo thymidylate biosynthesis pathway in mammalian mitochondria. Proc Natl Acad Sci U S A 108: 15163–15168. 10.1073/pnas.1103623108
    1. Carrozzo R, Wittig I, Santorelli FM, Bertini E, Hofmann S, et al. (2006) Subcomplexes of human ATP synthase mark mitochondrial biosynthesis disorders. Ann Neurol 59: 265–275.
    1. Viscomi C, Spinazzola A, Maggioni M, Fernandez-Vizarra E, Massa V, et al. (2009) Early-onset liver mtDNA depletion and late-onset proteinuric nephropathy in Mpv17 knockout mice. Hum Mol Genet 18: 12–26. 10.1093/hmg/ddn309
    1. Weiher H, Noda T, Gray DA, Sharpe AH, Jaenisch R (1990) Transgenic mouse model of kidney disease: insertional inactivation of ubiquitously expressed gene leads to nephrotic syndrome. Cell 62: 425–434.
    1. Ashley N, Adams S, Slama A, Zeviani M, Suomalainen A, et al. (2007) Defects in maintenance of mitochondrial DNA are associated with intramitochondrial nucleotide imbalances. Hum Mol Genet 16: 1400–1411.
    1. Han Z, Stachow C (1994) Analysis of Schizosaccharomyces pombe mitochondrial DNA replication by two dimensional gel electrophoresis. Chromosoma 103: 162–170.
    1. Reyes A, Kazak L, Wood SR, Yasukawa T, Jacobs HT, et al. (2013) Mitochondrial DNA replication proceeds via a 'bootlace' mechanism involving the incorporation of processed transcripts. Nucleic Acids Res 41: 5837–5850. 10.1093/nar/gkt196
    1. Saada A (2009) Fishing in the (deoxyribonucleotide) pool. Biochem J 422: e3–6. 10.1042/BJ20091194
    1. Eriksson S, Wang L (2008) Molecular mechanisms of mitochondrial DNA depletion diseases caused by deficiencies in enzymes in purine and pyrimidine metabolism. Nucleosides Nucleotides Nucleic Acids 27: 800–808. 10.1080/15257770802146197
    1. Saada A (2008) Mitochondrial deoxyribonucleotide pools in deoxyguanosine kinase deficiency. Mol Genet Metab 95: 169–173. 10.1016/j.ymgme.2008.07.007
    1. Pontarin G, Ferraro P, Rampazzo C, Kollberg G, Holme E, et al. (2011) Deoxyribonucleotide metabolism in cycling and resting human fibroblasts with a missense mutation in p53R2, a subunit of ribonucleotide reductase. J Biol Chem 286: 11132–11140. 10.1074/jbc.M110.202283
    1. Camara Y, Gonzalez-Vioque E, Scarpelli M, Torres-Torronteras J, Caballero A, et al. (2014) Administration of deoxyribonucleosides or inhibition of their catabolism as a pharmacological approach for mitochondrial DNA depletion syndrome. Hum Mol Genet.
    1. Uusimaa J, Evans J, Smith C, Butterworth A, Craig K, et al. (2014) Clinical, biochemical, cellular and molecular characterization of mitochondrial DNA depletion syndrome due to novel mutations in the MPV17 gene. Eur J Hum Genet 22: 184–191. 10.1038/ejhg.2013.112
    1. Wiseman A, Attardi G (1978) Reversible tenfod reduction in mitochondria DNA content of human cells treated with ethidium bromide. Mol Gen Genet 167: 51–63.
    1. Song S, Pursell ZF, Copeland WC, Longley MJ, Kunkel TA, et al. (2005) DNA precursor asymmetries in mammalian tissue mitochondria and possible contribution to mutagenesis through reduced replication fidelity. Proc Natl Acad Sci U S A 102: 4990–4995.
    1. Mathews CK (2006) DNA precursor metabolism and genomic stability. FASEB J 20: 1300–1314.
    1. Lai Y, Tse CM, Unadkat JD (2004) Mitochondrial expression of the human equilibrative nucleoside transporter 1 (hENT1) results in enhanced mitochondrial toxicity of antiviral drugs. J Biol Chem 279: 4490–4497.
    1. Lee EW, Lai Y, Zhang H, Unadkat JD (2006) Identification of the mitochondrial targeting signal of the human equilibrative nucleoside transporter 1 (hENT1): implications for interspecies differences in mitochondrial toxicity of fialuridine. J Biol Chem 281: 16700–16706.
    1. Villarroya J, de Bolos C, Meseguer A, Hirano M, Vila MR (2009) Altered gene transcription profiles in fibroblasts harboring either TK2 or DGUOK mutations indicate compensatory mechanisms. Exp Cell Res 315: 1429–1438. 10.1016/j.yexcr.2009.02.018
    1. Di Noia MA, Todisco S, Cirigliano A, Rinaldi T, Agrimi G, et al. (2014) The human SLC25A33 and SLC25A36 genes of solute carrier family 25 encode two mitochondrial pyrimidine nucleotide transporters. J Biol Chem 289: 33137–33148. 10.1074/jbc.M114.610808
    1. Elpeleg O, Miller C, Hershkovitz E, Bitner-Glindzicz M, Bondi-Rubinstein G, et al. (2005) Deficiency of the ADP-forming succinyl-CoA synthase activity is associated with encephalomyopathy and mitochondrial DNA depletion. Am J Hum Genet 76: 1081–1086.
    1. Ostergaard E (2008) Disorders caused by deficiency of succinate-CoA ligase. J Inherit Metab Dis 31: 226–229. 10.1007/s10545-008-0828-7
    1. Ostergaard E, Christensen E, Kristensen E, Mogensen B, Duno M, et al. (2007) Deficiency of the alpha subunit of succinate-coenzyme A ligase causes fatal infantile lactic acidosis with mitochondrial DNA depletion. Am J Hum Genet 81: 383–387.
    1. Dallabona C, Marsano RM, Arzuffi P, Ghezzi D, Mancini P, et al. (2010) Sym1, the yeast ortholog of the MPV17 human disease protein, is a stress-induced bioenergetic and morphogenetic mitochondrial modulator. Hum Mol Genet 19: 1098–1107. 10.1093/hmg/ddp581
    1. Krauss J, Astrinidis P, Frohnhofer HG, Walderich B, Nusslein-Volhard C (2013) transparent, a gene affecting stripe formation in Zebrafish, encodes the mitochondrial protein Mpv17 that is required for iridophore survival. Biol Open 2: 703–710. 10.1242/bio.20135132
    1. Antonenkov VD, Isomursu A, Mennerich D, Vapola MH, Weiher H, et al. (2015) The Human Mitochondrial DNA Depletion Syndrome Gene MPV17 Encodes a Non-selective Channel That Modulates Membrane Potential. J Biol Chem 290: 13840–13861. 10.1074/jbc.M114.608083
    1. Reinhold R, Kruger V, Meinecke M, Schulz C, Schmidt B, et al. (2012) The channel-forming Sym1 protein is transported by the TIM23 complex in a presequence-independent manner. Mol Cell Biol 32: 5009–5021. 10.1128/MCB.00843-12
    1. Da-Re C, Franzolin E, Biscontin A, Piazzesi A, Pacchioni B, et al. (2014) Functional characterization of drim2, the Drosophila melanogaster homolog of the yeast mitochondrial deoxynucleotide transporter. J Biol Chem 289: 7448–7459. 10.1074/jbc.M113.543926
    1. Van Dyck E, Jank B, Ragnini A, Schweyen RJ, Duyckaerts C, et al. (1995) Overexpression of a novel member of the mitochondrial carrier family rescues defects in both DNA and RNA metabolism in yeast mitochondria. Mol Gen Genet 246: 426–436.
    1. Marobbio CM, Di Noia MA, Palmieri F (2006) Identification of a mitochondrial transporter for pyrimidine nucleotides in Saccharomyces cerevisiae: bacterial expression, reconstitution and functional characterization. Biochem J 393: 441–446.
    1. Marcelino LA, Thilly WG (1999) Mitochondrial mutagenesis in human cells and tissues. Mutat Res 434: 177–203.
    1. Richter C, Park JW, Ames BN (1988) Normal oxidative damage to mitochondrial and nuclear DNA is extensive. Proc Natl Acad Sci U S A 85: 6465–6467.
    1. Kazak L, Reyes A, Holt IJ (2012) Minimizing the damage: repair pathways keep mitochondrial DNA intact. Nat Rev Mol Cell Biol 13: 659–671. 10.1038/nrm3439
    1. Ferraro P, Pontarin G, Crocco L, Fabris S, Reichard P, et al. (2005) Mitochondrial deoxynucleotide pools in quiescent fibroblasts: a possible model for mitochondrial neurogastrointestinal encephalomyopathy (MNGIE). J Biol Chem 280: 24472–24480.
    1. Garone C, Garcia-Diaz B, Emmanuele V, Lopez LC, Tadesse S, et al. (2014) Deoxypyrimidine monophosphate bypass therapy for thymidine kinase 2 deficiency. EMBO Mol Med 6: 1016–1027. 10.15252/emmm.201404092
    1. Dalla Rosa I, Durigon R, Pearce SF, Rorbach J, Hirst EM, et al. (2014) MPV17L2 is required for ribosome assembly in mitochondria. Nucleic Acids Res 42: 8500–8515. 10.1093/nar/gku513
    1. Schmittgen TD, Livak KJ (2008) Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc 3: 1101–1108.
    1. Yasukawa T, Reyes A, Cluett TJ, Yang MY, Bowmaker M, et al. (2006) Replication of vertebrate mitochondrial DNA entails transient ribonucleotide incorporation throughout the lagging strand. EMBO J 25: 5358–5371.
    1. Reyes A, Yasukawa T, Holt IJ (2007) Analysis of replicating mitochondrial DNA by two-dimensional agarose gel electrophoresis. Methods Mol Biol 372: 219–232. 10.1007/978-1-59745-365-3_16
    1. Friedman KL, Brewer BJ (1995) Analysis of replication intermediates by two-dimensional agarose gel electrophoresis. Methods Enzymol 262: 613–627.
    1. Li H (2013) Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM. arXiv preprint arXiv:13033997.
    1. Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, et al. (2009) The Sequence Alignment/Map format and SAMtools. Bioinformatics 25: 2078–2079. 10.1093/bioinformatics/btp352

Source: PubMed

3
Se inscrever