10
Thymidine kinase 2 defects can cause multi-tissue mtDNA depletion syndrome Alexandra Go« tz, 1, Pirjo Isohanni, 1,2, Helena Pihko, 2 Anders Paetau, 3 Riitta Herva, 4 Outi Saarenpa« a«- Heikkila« , 5 Leena Valanne, 6 Sanna Marjavaara 1 and Anu Suomalainen 1,7 1 Research Programme of Molecular Neurology, Biomedicum-Helsinki, University of Helsinki, Helsinki, 2 Department of Paediatric Neurology, Hospital for Children and Adolescents, Helsinki University Central Hospital, Helsinki, 3 Department of Pathology, University of Helsinki and Helsinki University Central Hospital, Helsinki, 4 Department of Pathology, Oulu University Hospital, Oulu, 5 Paediatric Clinics, Tampere University Hospital, Tampere, 6 Helsinki Medical Imaging Center, University of Helsinki and 7 Department of Neurology, Helsinki University Central Hospital, Helsinki, Finland These authors contributed equally to this work. Correspondence to: Prof. Anu Suomalainen, Research Programme of Molecular Neurology, Biomedicum-Helsinki, room c523a, Haartmaninkatu 8, University of Helsinki, 00290 Helsinki, Finland E-mail: [email protected] Mitochondrial DNA depletion syndrome (MDS) is a severe recessively inherited disease of childhood. It mani- fests most often in infancy, is rapidly progressive and leads to early death. MDS is caused by an increasing number of nuclear genes leading to multisystemic or tissue-specific decrease in mitochondrial DNA (mtDNA) copy number. Thymidine kinase 2 (TK2) has been reported to cause a myopathic form of MDS. We report here the clinical, autopsy and molecular genetic findings of rapidly progressive fatal infantile mitochondrial syndrome. All of our seven patients had rapidly progressive myopathy/encephalomyopathy, leading to respiratory failure within the first 3 years of life, with high creatine kinase values and dystrophic changes in the muscle with cyto- chrome c oxidase-negative fibres. In addition, two patients also had terminal-phase seizures, one had epilepsia partialis continua and one had cortical laminar necrosis. We identified two different homozygous or compound heterozygous mutations in theTK2 gene in all the patients: c.739 C s 2`Tand c.898 C 2` T, leading to p.R172W and p.R225W changes at conserved protein sites. R172W mutation led to myopathy or encephalomyopathy with the onset during the first months of life, and was associated with severe mtDNA depletion in the muscle, brain and liver. Homozygosity for R225Wmutation manifested during the second year of life as a myopathy, and showed muscle-specific mtDNA depletion. Both mutations originated from single ancient founders, with Finnish origin and enrichment for the new R172W mutation, and possibly Scandinavian ancestral origin for the R225W.We conclude that TK2 mutations may manifest as infantile-onset fatal myopathy with dystrophic features, but should be considered also in infantile progressive encephalomyopathy with wide-spread mtDNA depletion. Keywords: mitochondrial DNA depletion syndrome; thymidine kinase 2; myopathy; encephalomyopathy; mtDNA Abbreviations: APP = amyloid beta precursor protein; CK = creatine kinase; COX = cytochrome c oxidase; CytB = cytochrome B; dNTP = deoxy-nucleotidetriphosphate; EEG = electroencephalography; ENMG = electroneuromyography; GT = glutamyl transpeptidase; Mb = mega-basepair; MDS = mitochondrial DNA depletion syndrome; mtDNA = mitochondrial DNA; SDH = succinate dehydrogenase; SMA I = spinal muscular atrophy type I; TK2 = thymidine kinase 2. Received May 23, 2008. Revised August 21, 2008. Accepted September 3, 2008 Introduction Mitochondrial DNA depletion syndrome (MDS) is a group of severe autosomal recessive disorders affecting the skeletal muscle, liver, brain or several tissues (Moraes et al., 1991). MDS manifests often in infancy, with average fatality by the age of 3 years, irrespective of the presentation (McFarland et al., 2002). However, some children manifest later with a slowly progressive phenotype and survive into adulthood (Vu et al., 1998). MDS is characterized by severe reduction of mitochondrial DNA (mtDNA) copy number in the affected doi:10.1093/brain/awn236 Brain (2008) Page 1 of 10 ß The Author (2008). Published by Oxford University Press on behalf of the Guarantors of Brain. All rights reserved. For Permissions, please email: [email protected] Brain Advance Access published September 26, 2008 by guest on February 11, 2016 http://brain.oxfordjournals.org/ Downloaded from

Thymidine kinase 2 defects can cause multi-tissue mtDNA depletion syndrome

Embed Size (px)

Citation preview

Thymidine kinase 2 defects can cause multi-tissuemtDNA depletion syndromeAlexandra Go« tz,1,� Pirjo Isohanni,1,2,� Helena Pihko,2 Anders Paetau,3 Riitta Herva,4 Outi Saarenpa« a« -Heikkila« ,5 LeenaValanne,6 Sanna Marjavaara1 and Anu Suomalainen1,7

1Research Programme of Molecular Neurology, Biomedicum-Helsinki, University of Helsinki, Helsinki, 2Department ofPaediatric Neurology, Hospital for Children and Adolescents, Helsinki University Central Hospital, Helsinki, 3Department ofPathology, University of Helsinki and Helsinki University Central Hospital, Helsinki, 4Department of Pathology, OuluUniversity Hospital, Oulu, 5Paediatric Clinics, Tampere University Hospital, Tampere, 6Helsinki Medical Imaging Center,University of Helsinki and 7Department of Neurology, Helsinki University Central Hospital, Helsinki, Finland�These authors contributed equally to this work.

Correspondence to: Prof. Anu Suomalainen, Research Programme of Molecular Neurology, Biomedicum-Helsinki, roomc523a, Haartmaninkatu 8, University of Helsinki, 00290 Helsinki, FinlandE-mail: [email protected]

Mitochondrial DNA depletion syndrome (MDS) is a severe recessively inherited disease of childhood. It mani-fests most often in infancy, is rapidly progressive and leads to early death. MDS is caused by an increasingnumber of nuclear genes leading to multisystemic or tissue-specific decrease in mitochondrial DNA (mtDNA)copy number.Thymidine kinase 2 (TK2) has been reported to cause a myopathic form of MDS.We report herethe clinical, autopsy andmolecular genetic findings of rapidly progressive fatal infantilemitochondrial syndrome.All of our seven patients had rapidly progressive myopathy/encephalomyopathy, leading to respiratory failurewithin the first 3 years of life, with high creatine kinase values and dystrophic changes in the muscle with cyto-chrome c oxidase-negative fibres. In addition, two patients also had terminal-phase seizures, one had epilepsiapartialis continua and one had cortical laminar necrosis.We identified two different homozygous or compoundheterozygousmutations in theTK2 gene in all the patients: c.739C s 2`Tand c.898C 2` T, leading to p.R172Wand p.R225Wchanges at conserved protein sites. R172Wmutation led to myopathy or encephalomyopathy withthe onset during the first months of life, and was associated with severe mtDNA depletion in the muscle, brainand liver.Homozygosity for R225Wmutationmanifestedduring the secondyearof life as amyopathy, andshowedmuscle-specificmtDNAdepletion.Bothmutationsoriginated fromsingleancient founders,withFinnishoriginandenrichment for the new R172Wmutation, and possibly Scandinavian ancestral origin for the R225W.We concludethat TK2 mutations may manifest as infantile-onset fatal myopathy with dystrophic features, but should beconsideredalso in infantile progressive encephalomyopathy withwide-spreadmtDNAdepletion.

Keywords: mitochondrial DNA depletion syndrome; thymidine kinase 2; myopathy; encephalomyopathy; mtDNA

Abbreviations: APP=amyloid beta precursor protein; CK=creatine kinase; COX=cytochrome c oxidase;CytB=cytochrome B; dNTP=deoxy-nucleotidetriphosphate; EEG=electroencephalography;ENMG=electroneuromyography; GT=glutamyl transpeptidase; Mb=mega-basepair; MDS=mitochondrial DNAdepletion syndrome; mtDNA=mitochondrial DNA; SDH=succinate dehydrogenase; SMA I= spinal muscular atrophy type I;TK2=thymidine kinase 2.

Received May 23, 2008. Revised August 21, 2008. Accepted September 3, 2008

IntroductionMitochondrial DNA depletion syndrome (MDS) is a group ofsevere autosomal recessive disorders affecting the skeletalmuscle, liver, brain or several tissues (Moraes et al., 1991).MDS manifests often in infancy, with average fatality by the

age of 3 years, irrespective of the presentation (McFarlandet al., 2002). However, some children manifest later with aslowly progressive phenotype and survive into adulthood (Vuet al., 1998). MDS is characterized by severe reduction ofmitochondrial DNA (mtDNA) copy number in the affected

doi:10.1093/brain/awn236 Brain (2008) Page 1 of 10

� The Author (2008). Published by Oxford University Press on behalf of the Guarantors of Brain. All rights reserved. For Permissions, please email: [email protected]

Brain Advance Access published September 26, 2008 by guest on February 11, 2016

http://brain.oxfordjournals.org/D

ownloaded from

tissue(s). Patients typically present with progressive myopathy,hepatopathy and/or encephalopathy with lactic acidosis(McFarland et al., 2002). Three main MDS forms have beendescribed: myopathic, encephalomyopathic and hepatocere-bral form (OMIM #609560 - #251880) (Alberio et al., 2007).

MDS is quite common among mitochondrial diseases inchildren (Sarzi et al., 2007a), but the causative nucleargenes are only starting to unravel. Eight MDS genes areknown, all encoding proteins involved in deoxyribonucleo-tide triphosphate metabolism and mtDNA maintenance:thymidine kinase 2 (TK2) causing myopathic MDS (Saadaet al., 2001), deoxyguanosine kinase, mitochondrial twinklehelicase, a mitochondrial inner membrane protein MPV17and polymerase gamma causing hepatocerebral MDS(Ferrari et al., 2005; Nguyen et al., 2005; Spinazzola et al.,2006; Hakonen et al., 2007; Sarzi et al., 2007b), ADP-formingbeta and alpha-subunits of the succinate-coenzyme-A-ligase(SUCLA2 and SUCLG1) and cytoplasmic p53-inducible smallsubunit of ribonucleotide reductase (RRM2B) causinggeneralized encephalomyopathic MDS (Elpeleg et al., 2005;Bourdon et al., 2007; Ostergaard et al., 2007).

We studied the clinical, morphological and molecularbackground of seven patients with severe, infantile dys-trophic myopathy and early death, reminiscent of spinalmuscular atrophy type I (SMA I) in clinical severity. Twoindex patients were found to have severe muscle-specificmtDNA depletion and TK2 mutations. Search for TK2mutations in similar phenotypes led to identification of fivemore patients, including encephalomyopathies with multi-tissue mtDNA depletion.

Patients, Materials and MethodsPatientsSeven infants of Finnish origin presented rapidly progressive muscle

weakness leading to death of six of them and permanent ventilatorsupport in one. Their parents were not known to be consanguineous,but three families originated from the same rural area. Informationof ancestors’ birthplaces was obtained by parent interviews. Patients

5, 6 and 7 were previously described in a series of congenitaldystrophy, before molecular knowledge of their disease (Pihko et al.,1992). The patients arose from a material of 41 patients with severe

childhood muscle weakness, and were investigated at HelsinkiUniversity Central Hospital between the years 1977 and 2003. Allsamples were taken for diagnostic purposes, and used for researchwith oral informed consent from the parents.

Patient 1Patient 1 was the second child of the family, born after in vitrofertilization at term. Her mother has hypothyroidism and has hadcardiac arrhythmias, whereas father and elder brother are healthy.The patient had normal early development and could walk

without support at 12 months of age. Progressive muscle weaknesswas noticed at 18 months: she could not crawl up the stairs andhad to push herself with hands up from the floor. On examinationat 1 year 9 months of age, she had severe generalized muscle

weakness and absent deep tendon reflexes, and her cognitive

development was normal. Her disease progressed rapidly duringthe following months, within 6 months she lost all antigravitymovements and after a year she needed gastrostomy. She died ofrespiratory failure 18 months after the disease onset.

Patient 2Patient 2 was the first child of healthy parents. The pregnancy, herbirth and early developmental milestones were normal. At 7 monthsof age, she could raise herself up to standing position, but thereafterher motor development slowed down. At the age of 1 year 4 months,upon a respiratory tract infection, her condition deteriorated duringa couple of weeks: she stopped crawling, could not rise up and heldher head with difficulty. Her disease progressed rapidly. At the ageof two she went into respiratory failure and needed permanentventilator support. Her cognitive skills could not be formallyassessed, but appeared normal. She has had epileptic seizures sincethe age of 2 years and the seizures evolved to epilepsia partialiscontinua. Repeated EEGs showed slow background activity andfocal spikes, multi-spikes and spike-slow wave discharges. BrainMRI was normal at 16 months of age, but at 5 years showed corticaland central atrophy. Presently, she is 5 years of age, hospitalizedand ventilator-dependent, with extreme muscle weakness andatrophy, cardiomyopathy and has gastrostomy. She has hadfractures of large bones, e.g. femoral bone, during daily routinecare, possibly caused by inactivity. She has retained minimalmovement in her eyelids and finger tips, and she communicates byopening and closing her eyes.

Patient 3Patient 3 was the third child of healthy parents with two healthysiblings. The pregnancy and delivery were normal. She wasreferred for investigations because of poor head control since theage of 5 months. On examination at 6 months of age she was analert baby, with poor muscle tone and severe proximal muscleweakness and absent reflexes. The muscle weakness progressedrapidly and she died of respiratory failure at the age of 12 months.

Patient 4Patient 4 was born as the first child of healthy parents afternormal pregnancy and delivery. On examination at 3 months ofage she was alert, but showed severe proximal muscle weakness,hypotonia and absent reflexes. Her muscle weakness progressedrapidly. She had short seizures with focal epileptic discharges inEEG from the age of 6 months, and she died of respiratory failureat the age of 7 months.

Patient 5Patient 5 was the second child of healthy parents, born afternormal pregnancy and delivery. Her brother was healthy.Her weight gain was poor, and her motor development wasslow from 3 months of age. On examination at 5 months she wasalert, but had severe muscle weakness and hypotonia, with absentreflexes. Her muscle weakness progressed rapidly, and shedeveloped short focal seizures. She died of pneumonia andrespiratory failure at the age of 7 months.

Patient 6Patient 6 was the second child of a healthy father and a motherwith aortic stenosis with two healthy daughters. The pregnancy

Page 2 of 10 Brain (2008) A.Go« tz et al.

by guest on February 11, 2016http://brain.oxfordjournals.org/

Dow

nloaded from

and delivery were normal. Severe progressive muscle weakness wasnoticed at 3 months of age, and he died of pneumonia at theage of 6 months. The diagnosis of congenital dystrophy wasmade based on the high creatine kinase (CK)-value and dystrophicchanges in the muscle.

Patient 7Patient 7 was the brother of Patient 6. He was born at term afternormal pregnancy. His hypotonia raised concern at 3 months of age.He had frequent respiratory infections. On examination at 5 monthsof age he was alert but had muscle weakness and hypotonia, with nohead control. He had high CK and dystrophic changes in themuscle. His muscle weakness progressed, and he died at the age of 8months of pneumonia, with the diagnosis of congenital dystrophy.

All studied patients (P1–5) had liver transaminases 2- to 4-foldthe normal range while CK was elevated 5- to 10-fold. Transaminaseincrease is generally associated with high CK, and therefore thisfinding cannot be interpreted as direct evidence for liver dysfunc-tion. However, Patient 2 has had consistently increased gammaglutamyl transpeptidase (GT) values (71–893 U/l; normal 550 U/l)from the age of 3 years. As her gamma GT values were over 300 U/lalready when antiepileptic medication included clobazam, oxcarba-zepine or gabapentine that often do not cause liver dysfunction, theincrease of gamma-GT may be a sign of disease-associated liverdysfunction.

Control subjectsAs ‘muscle controls’ we used biopsy samples of vastus lateralis fromage-matched subjects, who were deemed not to have a muscledisease. As ‘brain and liver controls’ we used paraffin-embedded orfrozen post-mortem samples from age-matched subjects, with nomitochondrial disease. In addition to age-matching, each paraffin-embedded patient sample was matched with controls with similarpost-mortem (524 h) and formalin-fixation times, and frozensamples were matched with frozen control samples. The matchedcontrol groups were the following: C1, frozen muscle biopsyspecimens of children of 3, 10 and 43 months of age; C2, cerebralcortex, basal ganglia and cerebellum autopsy samples from childrenof 2, 3 and 7 months of age, and C3, liver autopsy sample of a patientof 512 months of age, all matched for similar post-mortem andformalin-fixation times; C4, frozen autopsy samples of cerebralcortex and cerebellum of children of 8 and 9 months of age, and C5,frozen liver biopsy specimens of children of 3, 9, 11, 30, 38 and 41months of age.

Morphologic analysisOpen surgical biopsies from vastus lateralis muscle were takenunder generalized anaesthesia. The samples were snap-frozen inisopentane and processed for histochemical analyses for cytochromec oxidase (COX) and succinate dehydrogenase (SDH) activities, aswell as for the following stainings: haematoxylin–eosin, Van Gieson,modified Gomori trichrome, periodic acid-Schiff, oil red-O,NADH-tetrazolium reductase and adenosine triphosphatase atpH 10.4, 4.6 and 4.3 preincubations. Samples for ultrastructuralstudies were fixed by glutaraldehyde and examined with a Jeol JEM-1200EX electron microscope. Autopsy information with neuro-pathological examination on formalin-fixed brain was available ofPatients 1, 3, 4 and 5. Autopsy samples of Patients 1, 4 and 5 wereavailable to us.

Biochemical analysis of respiratory chainenzyme activitiesThe mitochondria were isolated from a fresh musclebiopsy specimen, and the respiratory chain enzyme activities,rotenone-sensitive NADH: cytochrome c oxidoreductase (CI + III),antimycin-sensitive succinate: cytochrome c oxidoreductase(CII + III), SDH (CII), COX (CIV) and citrate synthase, wereanalysed as previously described (Majander et al., 1995).

DNA analysesThe Homo sapiens chromosome 16 reference assembly sequence,which contains the human TK2 genomic DNA sequence(NC_000016) and TK2 mRNA (NM_004614), and newly revisedTK2 protein sequence (NP_004605.3) were used as referencesequences. Of note, the recent revision of TK2 sequence describedadditional N-terminal amino acids, changing the amino acidnumbering compared with that used in previous publications,utilizing old reference sequence XM_007855 (Galbiati et al., 2006).

Samples for DNA-analysis were snap-frozen in liquidnitrogen and stored at �80�C. Total DNA from frozen muscle,brain and liver samples (either autopsy or muscle biopsy-derived),as well as from paraffin-embedded formalin-fixed autopsy sampleswas extracted by standard methods (Shibata et al., 1988).

We amplified and sequenced the TK2 exons and exon–intronboundaries as in (Saada et al., 2001), using the BigDye Terminatorv3.1 sequencing kit (Applied Biosystems, Warrington, Cheshire,UK) on an ABI 3730XL DNA Analyzer (Applied Biosystems, FosterCity, CA, USA). Sequence processing was done with Sequencher 4.5software (GeneCodes). For single-nucleotide detection of mutantnucleotides we utilized solid-phase minisequencing, according to(Suomalainen and Syvanen, 2000). Specific primers were designedfor both the mutations:

Exon6_F: Biotin 50-CTCCTTTTCCCCTGAGTTAGTGATTCTCTT-30

Exon6_R: 50-CCTTCTATACAGGTTTTCTACAAAAATGTATCTTGCG-30

Exon6_R Detection: 50–AATCGACCTCTCCATCAACC-30

Exon8_F: Biotin 50-GAGCAGAGTCCTGAGAGCCAC-30

Exon8_R: 50–CCTCTTCCCTGCATCTCTTCTTTAACCTC-30

Exon8_R Detection: 50-ACAAGTCTCAGGATTGGTCC-30

For mtDNA quantification, the mitochondrial cytochrome b(Cytb) in mtDNA and the nuclear amyloid beta precursor protein(APP) genes were simultaneously amplified by quantitativeTaqMan real-time PCR assay in the ABI Prism 7000 DetectionSystem Cycler, using the following primers and probes:

hCytb_F: 50-GCCTGCCTGATCCTCCAAAT-30

hCytb_R: 50-AAGGTAGCGGATGATTCAGCC-30

hAPP_F1: 50-TGTGTGCTCTCCCAGGTCTA-30

hAPP_R1: 50-CAGTTCTGGATGGTCACTGG-30

h_cytB-FAM: 6-FAM 50-CACCAGACGCCTCAACCGCCTT-30 TAMRA

h_APP_VIC: VIC 50-CCCTGAACTGCAGATCACCAATGTGGTAG-30 TAMRA

As amplification standards we used human APP or CytbcDNAs cloned in PCR-TOPO vector (Invitrogen, Carlsbad,California, USA), amplified parallel to the samples and used atconcentration dilutions of 103–106 copies. Of the genomic DNA,25 ng was amplified using TaqMan Universal PCR Master Mix

TK2 mutations in encephalomyopathy Brain (2008) Page 3 of 10

by guest on February 11, 2016http://brain.oxfordjournals.org/

Dow

nloaded from

(Applied Biosystems, Roche, Branchburg, New Jersey, USA),

900 nM of each primer and 250 nM of each probe in 30 ml

volume in following conditions: an enzyme activation step for

2 min at 50�C, followed by denaturing for 10 min at 95�C, and 40

amplification cycles of 15 s at 95�C, and 1 min at 60�C.

Fluorescence acquisition was done at the end of each annealing

step, and data were analysed using the ABI Prism 7000 SDS

software (Applied Biosystems, FosterCity, CA, USA). The ratio of

mtDNA to nuclear DNA obtained by comparison to the standard

curve, created using known copy numbers of standard plasmids

with CytB or APP genes as amplification templates, was used as a

measure of mtDNA content (Livak and Schmittgen, 2001), with

typically 2–5 replicate analyses per one sample. The mtDNA

sequence was determined as in (Pietilainen et al., 2008), and

multiple mtDNA deletion analysis was done as in (Van Goethem

et al., 2004).

Haplotype analysisWe analysed five polymorphic dinucleotide markers flanking the

TK2 gene (D16S3021, D16S3019, D16S397, D16S421 and

D16S3025). PCR was performed in 0.2 mM of dNTPs, 0.4 mM of

PCR primers, 0.8 mCi of [33P]dATP and 0.6 U of thermostable DNA

polymerase (Dynazyme II, Finnzymes, Espoo, Finland), in 30 ml of its

buffer, with an initial cycle at 95�C for 3 min, followed by 30–35

cycles at 94�C for 30 s, 55–56�C (optimized for each primer pair

separately) for 30 s, and 72�C for 1 min and a final elongation at 72�C

for 10 min. The radioactive PCR products were electrophoresed

through non-denaturing 8% polyacrylamide gels, and autoradio-

graphy was done on Biomax MR films (Kodak, Rochester, NY, USA)

for 1–2 days.

BioinformaticsHuman TK2 wild-type protein sequence (NP_004605) and the

sequence introducing the TK2 mutations were analysed for post-

translational modifications and protein structure using the

PredictProtein analysing tool (www.predictprotein.org).

ResultsClinical featuresTable 1 summarizes the symptoms and signs of the patients.No patient showed pseudohypertrophy of muscles, andmuscle atrophy came late in the disease course. The disease-onset was acute or subacute in all patients and led to loss ofall motor skills rapidly from one to few months fromdisease-onset, as in SMA I. Cognitive state was consistent tothe patients’ ages. In electroneuromyography (ENMG), allstudied patients had polyphasic motor units as a sign ofmyopathic changes. Nerve conduction velocities werenormal. Patient 1 had also spontaneous electrical activity,fibrillations, as a sign of lower motor neuron involvement.EEGs were performed on patients 2, 4 and 5, and it showednormal or slow background activity with localized orsecondarily generalized spike and slow-wave activity.

Muscle morphology and histochemistryTable 1 summarizes the light microscopic findings in thepatients’ muscle biopsy samples. Two representative biopsyfindings are described in detail:

Patient 2At 21 months of age, the muscle showed marked fibre sizevariation and atrophy, slight fibrosis, increased neutrallipids and up to 60–70% COX-negative fibres. Scatterednecrotic fibres were seen. Electron microscopy demon-strated increased lipid vacuoles, but mitochondria appearednormal (not shown).

Patient 4At 5 months of age, the muscle showed severe dystrophicchanges with marked fibre size variation and fibrosisrendering a preliminary diagnosis of congenital muscledystrophy (Fig. 1A). A few centronuclear, possibly regen-erating fibres were seen, as well as occasional necroticfibres. Most muscle fibres were of ragged-red type and470% COX-negative fibres were detected (Fig. 1B). Inelectron microscopy, mitochondria were increased innumber especially subsarcolemmally and often abnormalin shape with concentric cristae (not shown).

Autopsy findingsThe macroscopic and light microscopic findings ofthoroughly sampled brains of Patients 1 and 3 werewithin normal range, except for slight terminal capillarycongestion and marginal oedema. Patient 4 displayed afocal mid-layer laminar necrosis of the visual cortex withneuronal loss and gliosis (Fig. 1C). Patient 5 showedpyknotic neurons in the neocortex and to some extent inthe basal ganglia and thalamus, consistent with ischaemicinjury. Similar mild changes were also seen in the dentategyrus of hippocampus. Purkinje cell damage was detectedin the cerebellar cortex (Fig. 1D and E). Patients 4 and 5had mild to moderate, mainly macrovesicular steatosisperiportally in the liver (Fig. 1F). The spinal cords ofPatients 1, 3 and 4 showed no changes in histologicalanalysis.

TK2 sequence analysisOf a total of 41 screened patients with infantile muscle hypo-tonia we could diagnose seven with mutations in the TK2. Wefound two different missense mutations in TK2 (Fig. 2). Inexon 8, C4T transversion at nucleotide 898 (Genbankacc.no: NM_004614), causing an amino acid change fromarginine to tryptophane, and in exon 6 at nucleotide739 C4T, which also changed an arginine to tryptophane.Both amino acids are highly conserved in species (Fig. 2).Table 1 indicates the mutations found in each patient.

Page 4 of 10 Brain (2008) A.Go« tz et al.

by guest on February 11, 2016http://brain.oxfordjournals.org/

Dow

nloaded from

mtDNA analysisQuantification of mtDNA by real-time PCR revealed severemtDNA depletion in the skeletal muscle (Fig. 3A) in allstudied patients. In addition, severe mtDNA depletion inthe brain and liver of Patients 4 and 5 were detected,whereas the brain of Patient 1 showed normal mtDNAamount and the liver of Patient 1 showed only slightlyreduced mtDNA amount compared with the liver and brainsamples of the age-matched controls (Fig. 3B and C). Thisresult was verified by Southern blotting (data not shown).Complete mtDNA sequence of Patients 1 and 2 wasanalysed from muscle DNA, and no pathogenic mutationswere found. Number of polymorphic variants was notincreased. In PCR analysis, no large-scale mtDNA deletionswere found.

Biochemical analysisTable 2 shows the respiratory chain enzyme activities infresh muscle homogenates of Patients 1 and 2. The CI + IIIdependent activities were reduced to 20% or 25% ofcontrols per citrate synthase, in P1 and P2, respectively, andCII + CIII activities were reduced to 30% and 60% ofcontrols per citrate synthase. COX was most severelyaffected of the enzyme complexes, with 17% and 19%,respectively, remaining activities compared with the

controls. Also a moderate reduction of CII, which is notdependent on mtDNA-encoded subunits, was seen in bothpatients. In Patient 2, citrate synthase activity was increased,as a sign of mitochondrial proliferation.

Haplotype analysis and ancestral originsFigure 4 summarizes the haplotype analysis. The patientsand the heterozygote carriers of R172W shared a commoncore haplotype of three polymorphic dinucleotide markers,extending over a chromosomal region of �1.1 Mb.However, in some patients with R172W the sharedhaplotype extended over 3.3 Mb. The R225W patientsshared a common haplotype of two polymorphic dinucleo-tide markers extending over a chromosomal region of�700 kb. Figure 5 shows the birthplaces of the grandparentsand great-grandparents of the patients.

Bioinformatic analysis of the mutant TK2Secondary structure analysis showed that the missense changeR225W is located at the first position of a putative tyrosinekinase phosphorylation site (residue 225–232, -RTNPETCY-)abolishing it. The R172W missense change at the thirdposition of a putative protein kinase c phosphorylation site(residue 170–172, -SVR-) abolishes the site.

Table 1 Clinical features of our patients with infantile dystrophic myopathy and TK2 mutations

Patient P1 P2 P3 P4 P5 P6a P7a

Gender F F F F F M MAge at onset(months)

18 7 5 2 3 3 3

Age at death(months)

38 alive at 5 y 12 7 7 6 8

Cause of death RF alive RF RF P P RFSeizures � ++ � + + � �

Gastrostomy/nasogastric tube

+ + + + + NA +

Serum creatinekinase (U/l)b

1315 1270 3670 3015 1996 NA 520

Plasma lactate(mmol/l)c

3.4 4.6 NE 2.3 NE NE NE

ENMG Myopathicand neuropathic

Myopathic Myopathic Myopathic Myopathic NA Myopathic

Muscle biopsy(age at biopsy)

Myopathic(21mo)

Myopathic(21mo)

Dystrophic(7mo)

Dystrophic(5mo)

Dystrophic(6mo)

Dystrophic(5mo)

Dystrophic(6mo)

COX-negative fibres 50% 70% 20% 70% NE NE NEBrain MRI(age)

Normal(23mo)

Normal (16mo),brainatrophy (5y)

Ultrasonographynormal

Normal(5mo)

NE NE NE

TK2 mutation R225W/R225W

R172W/R225W

R172W/R225W

R172W/R172W

R172W/R172W

R172W/R172W

R172W/R172W

Residual mtDNA inmuscle (biopsy sample)

5% 10% NA 10% (12%)(autopsy sample)

NA NA

F= female; M=male; mo=months; MRI=magnetic resonance imaging; NA=not available; NE=not evaluated; P=pneumonia;RF=respiratory failure; y=years.aP6 and P7 were siblings. bCreatine kinase reference value (5270) for other samples except for P7 (550), the latter analysed in 1970’s withdifferent methodology. cLactate reference value (0.5^2.2).

TK2 mutations in encephalomyopathy Brain (2008) Page 5 of 10

by guest on February 11, 2016http://brain.oxfordjournals.org/

Dow

nloaded from

DiscussionWe describe here mitochondrial TK2 defects in sevenFinnish infants, and the first autopsy studies of suchpatients. All our patients showed severe dystrophic myo-pathy with mtDNA depletion, and two were also verified tohave multi-tissue manifestation with mtDNA depletion inthe skeletal muscle, brain and the liver. Four patients werehomozygous for a novel TK2 mutation leading to aminoacid change R172W, one patient was homozygous forR225W change, previously described in Swedish patients(note: R225W has been previously reported as R183W)(Tulinius et al., 2005), and two patients were compoundheterozygotes for both. Since no other TK2 mutations werefound in our representative material of 41 childhoodmyopathies, these two mutations are likely to explain themolecular background of myopathic MDS in Finland.

No previous patients homozygous for either mutationhave been reported, allowing us a unique opportunity forgenotype–phenotype correlation. The phenotype of R172Wwas among the most severe reported in TK2 deficiency,with disease-onset in the first months of life, rapid loss ofall motor skills and death within few months. Compoundheterozygosity for R172W/R225W resulted in almost equaldisease severity. The R225W homozygosity mimicked thedisease course often reported with TK2–MDS (Oskouiet al., 2006), with normal motor development up to 1.5years, rapid progressive myopathy, leading to loss of allantigravity muscle power and respiratory failure at 3 yearsof age. Interestingly, the R225W homozygosity lead tomuscle-specific manifestation, whereas R172W lead tomulti-tissue mtDNA depletion involving the muscle, brainand the liver.

Fig. 1 Histological findings inTK2 deficiency. Muscle biopsy sample from Patient 4 shows (A) marked fibre variation, atrophic fibres, endo-mysial fibrosis and fatty degeneration (frozen section, haematoxylin^ eosin �400, original magnification) and (B) high proportion of darkblue cytochrome c oxidase-negative and SDH-positive fibres (frozen section, COX-SDH �200; original magnification). (C) Visual cortexfrom Patient 4. Between arrows: a spongiotic zone of laminar necrosis with neuronal loss and gliosis. (paraffin section, HE�40 originalmagnification). (D) Neocortical mid-laminar pyramidal layers of Patient 5 harboured many pyknotic red neurons consistent with ischaemicinjury (arrow). (Paraffin section, HE�400 original magnification). (E) In the cerebellar cortex of Patient 5 pyknotic red Purkinje cells(arrow) could easily be found. (paraffin section, HE�200 original magnification). (F) In the liver parenchyma of Patient 5 periportal, mainlymacrovesicular steatotic vacuolization (arrows) could be seen. (paraffin section, HE� 200 original magnification).

Page 6 of 10 Brain (2008) A.Go« tz et al.

by guest on February 11, 2016http://brain.oxfordjournals.org/

Dow

nloaded from

All our patients had severe muscle weakness resemblingSMA I. Previously, two patients with TK2 mutations havebeen described as ‘SMA-like’, with loss of motor units andfibrillations (Mancuso et al., 2002) or with‘neuropathic’ find-ings (Oskoui et al., 2006) in ENMG. Our patients had myo-pathic ENMG except for Patient 1, who also had fibrillationsas a sign of lower motor neuron involvement. However, noneof our TK2 patients had giant motor units, reflectingdenervation–reinnervation and typical for SMA. In ourpatients, the disease was sometimes accompanied by enceph-alopathy with partial seizures, but none of our patients

showed progressive cognitive impairment or ocular myopa-thy, as reported in (Galbiati et al., 2006). Three patients hadseizures as a late manifestation, and one of them showedlaminar cortical necrosis in the occipital region. Lack ofmolecular diagnosis and rapidly developing respiratory insuf-ficiency led one patient to ventilator treatment (compoundheterozygous for R172W/R225W), which has kept her alive tothe present age of 5 years. She has minimal muscle power,epilepsia partialis continua—a typical seizure type in mito-chondrial diseases—and severe cortical atrophy. The R172Wpatients showed clear clinical signs consistent with CNS

Fig. 2 Sequence analysis of TK2 gene. (A) A newTK2 mutation in exon 6 C739T (R172W) occurs in P4 as homozygous. (B) The exon 8C898T (R225W) TK2 mutation, for which P1 is homozygous. P2 and P3 were heterozygous for both mutations. Both mutations are highlyconserved in species and result in a change of a tryptophan for an arginine.

Table 2 Mitochondrial respiratory chain activities in muscle (nmol/min/mg) (per citrate synthase)

P1 (22 months) P2 (21months) controls (0^17 years)

C I+ III (NADH:cyt c reductase) 100 (0.04) 93 (0.05) 273�134 (0.2)C II+ III (succinate: cyt c reductase) 93 (0.03) 120 (0.06) 131�72 (0.1)C II (SDH) 101 (0.04) 72 (0.04) 150� 50 (0.11)C IV (COX) 722 (0.26) 540 (0.29) 2068� 888 (1.50)citrate synthase 2807 1849 1375� 494

NADH = reduced nicotinamide adenine dinucleotide; Cyt c = cytochrome c.

TK2 mutations in encephalomyopathy Brain (2008) Page 7 of 10

by guest on February 11, 2016http://brain.oxfordjournals.org/

Dow

nloaded from

manifestation of the disease. Our findings also show that theaggressive skeletal muscle involvement of R172W patientswould likely be followed by manifestations in other post-mitotic tissues, if the patients did not die of respiratorymuscle weakness.

Recently, prominent vacuolar changes in spinal cordneurons were found in TK2 knock-in mice (Akman et al.,2008), but the spinal cords of three of our patients wereinvestigated and found to be normal. As the TK2 mice didnot have prominent muscle involvement, the differencesbetween the TK2 disease in humans and mice may reflectdifferent tissue-specific dNTP pool regulatory pathways.

Demonstration of mtDNA depletion in the brain in twochildren with R172W change strongly suggested that theCNS symptoms and signs are MDS associated. R172W also

Fig. 3 The mtDNA copy number analysis by quantitative real-time PCR (Q-PCR) in patients withTK2 mutations. (A) Skeletal musclemtDNA copy number is dramatically decreased in all TK2-patients. Frozen muscle biopsy samples of patients P1 (21months), P2(21months), P4 (5 months) and pooled controls C1 (3,10 and 43 months; as specified in methods); autopsy-derived formalin-fixed muscleof patient P5 (7 months). (B) The brain (cortex, cerebellum, basal ganglia) and liver of TK2-patients with the R172Wmutation showsevere mtDNA depletion. Brain and cerebellum samples of patient P4 (7 months), the pooled controls C2 (2, 3 and 7 months), the liversample of P4 and the control C3 (512 months) were autopsy-derived, formalin-fixed paraffin samples, matched with similar post-mortemand formalin-fixation times. (C) Patient P1homozygous for the R225W TK2 mutation has high amounts of mtDNA in the brain and slightlyreduced in the liver, at 3 years of age. Pooled controls C4 (8 and 9 months; frozen brain samples) and C5 (3, 9, 11, 30, 38 and 41monthsof age; frozen liver biopsy specimens). The mtDNA copy number of one of the controls in each pool (10-month old in pool C1, 7-monthold in C2 and 9-month old in C4) was set as 100% reference point. The results are average values of two to five Q-PCR experiments,each with triplicate samples.

Fig. 4 Haplotypes of the FinnishTK2 patients. The DNAmarker names are shown without ‘D16S’. P1^4 areTK2 patientsand C1^2 are heterozygote carriers of eitherTK2 mutation.The haplotype associated with the R172W change is shown ingray, and the haplotype associated with the R225W change isshown in black.

Page 8 of 10 Brain (2008) A.Go« tz et al.

by guest on February 11, 2016http://brain.oxfordjournals.org/

Dow

nloaded from

led to partial mtDNA depletion in the liver, withmacrovesicular steatosis, but to no clinical signs. This isthe first report of TK2-associated mtDNA depletion in non-muscle tissues and clearly confirms the clinical suspicion ofmulti-organ involvement. Our single homozygous patientfor R225W, with severe muscle-restricted mtDNA deple-tion, manifested later and lived longer than those homo-zygous or compound heterozygous for R172W change. Wedemonstrate here unique mutation-specific tissue manifes-tations of TK2-MDS. As both R225W and R172W affectpotential phosphorylation sites, our findings suggest thatsingle residues can differentially modulate tissue-specificfunctions of mitochondrial TK2, possibly through mod-ification of phosphorylation status. Mechanisms involvedmay give important insights into tissue specificity of MDS.

The haplotype analysis showed that the R172W andR225W changes were associated with single specific hap-lotypes, indicating that they originated from single ancestralfounders. The grandparents and great-grandparents of ourpatients with R172W originated from north-easternFinland, which is a sub-isolate within the genetically iso-lated country, with its own disease heritage (Norio, 2003a,b). The grandparent clustering and common haplotypeextending over 3 Mbp suggest that R172W has a Finnishorigin. The R225W carriers originated from south-westernFinland, with strong Swedish impact to their gene pool.Interestingly, R225W has previously been published inSwedish patients (Tulinius et al., 2005), suggesting acommon Scandinavian founder. The shared R225W-associated haplotype was short, suggesting distant related-ness, tens of generations, between the present families.

Both TK2 mutations described here changed a highly con-served hydrophilic arginine to hydrophobic tryptophan in theTK2 polypeptide. R172W change is novel, although the sameamino acid changed to glutamine, combined with Y154Nchange, was mentioned in a recent survey of myopathic MDS,without further clinical details of this specific patient (Sarziet al., 2007a). R225W and R225G have been described ascompound heterozygous in pure childhood myopathies(Carrozzo et al., 2003; Vila et al., 2003; Tulinius et al.,2005), and R225G, combined with a truncating mutation, ininfantile-onset encephalomyopathy (Carrozzo et al., 2003).The causative role of both mutations is further supportedby the ancestral mutation-specific haplotypes and their fullco-segregation with the disease in multiple families withsimilar disease. Further, TK2 mutations were not found in 68chromosomes of Finnish patients with other types ofchildhood myopathies.

All our patients studied showed just 5–10% residualmtDNA amount in muscle compared with age-matchedcontrols, at the time of their muscle biopsy and diagnosis.The longest surviving R225W homozygote patient showed95% depletion at 21 months of age, whereas the patient withthe most severe phenotype, R172W homozygote, had 90%depletion already at 5 months of age. This shows that at thetime of diagnosis, the muscle has already lost the majority ofits mtDNA. Quite surprisingly, the low mtDNA amount wasstill capable to maintain 20–30% of residual activities of therespiratory chain enzyme complexes I, III and IV, containingmtDNA-encoded subunits, which suggests compensatorymechanisms, possibly involving increased mitochondrialtranscript or protein half-lives (Vila et al., 2008). We detectedalso downregulation of complex II, with no mtDNA-encodedsubunits, probably reflecting a secondary effect due to thesevere dystrophy of the muscle.

In conclusion, our study shows that TK2 mutationsshould be considered when searching for the molecularbackground of progressive muscle disease with SMA I-likeclinical severity. Furthermore, we show that TK2 gene canalso underlie MDS with brain and liver involvement andthat the tissue specificity of the manifestation may bemodified by single missense mutations.

AcknowledgementsWe thank the patients’ families for their cooperation, IlseLappalainen for her skilful technical assistance, KatriVuopala for autopsy and samples of patient 4.

FundingAcademy of Finland (to A.S. and S.K.M.); HelsinkiUniversity; Sigrid Juselius Foundation (to A.S. and H.P.);Graduate School of Biotechnology and Molecular Biology(to A.G.); Foundation for Paediatric Research and HelsinkiUniversity Central Hospital EVO grant (to H.P.); HumanFrontier Science Program (LT00341/2001-M, to S.K.M.).

Fig. 5 TK2 genealogy. Birthplaces of the Finnish grandparents (bigdots) and great-grandparents (small dots) of the patients withR172Wmutation (left), of patients with R225Wmutation (right).

TK2 mutations in encephalomyopathy Brain (2008) Page 9 of 10

by guest on February 11, 2016http://brain.oxfordjournals.org/

Dow

nloaded from

ReferencesAkman HO, Dorado B, Lopez LC, Garcia-Cazorla A, Vila MR, Tanabe LM,

et al. Thymidine kinase 2 (H126N) knockin mice show the essential role

of balanced deoxynucleotide pools for mitochondrial DNA maintenance.

Hum Mol Genet 2008; 17: 2433–40.

Alberio S, Mineri R, Tiranti V, Zeviani M. Depletion of mtDNA:

syndromes and genes. Mitochondrion 2007; 7: 6–12.

Bourdon A, Minai L, Serre V, Jais JP, Sarzi E, Aubert S, et al. Mutation of

RRM2B, encoding p53-controlled ribonucleotide reductase (p53R2),

causes severe mitochondrial DNA depletion. Nat Genet 2007; 39:

776–80.

Carrozzo R, Bornstein B, Lucioli S, Campos Y, de la Pena P, Petit N, et al.

Mutation analysis in 16 patients with mtDNA depletion. Hum Mutat

2003; 21: 453–4.

Elpeleg O, Miller C, Hershkovitz E, Bitner-Glindzicz M, Bondi-

Rubinstein G, Rahman S, et al. Deficiency of the ADP-forming

succinyl-CoA synthase activity is associated with encephalomyopathy

and mitochondrial DNA depletion. Am J Hum Genet 2005; 76: 1081–6.

Ferrari G, Lamantea E, Donati A, Filosto M, Briem E, Carrara F, et al.

Infantile hepatocerebral syndromes associated with mutations in the

mitochondrial DNA polymerase-gammaA. Brain 2005; 128: 723–31.

Galbiati S, Bordoni A, Papadimitriou D, Toscano A, Rodolico C,

Katsarou E, et al. New mutations in TK2 gene associated with

mitochondrial DNA depletion. Pediatr Neurol 2006; 34: 177–85.

Hakonen AH, Isohanni P, Paetau A, Herva R, Suomalainen A,

Lonnqvist T. Recessive Twinkle mutations in early onset encephalopathy

with mtDNA depletion. Brain 2007; 130: 3032–40.

Livak KJ, Schmittgen TD. Analysis of relative gene expression data using

real-time quantitative PCR and the 2(-Delta Delta C(T)) method.

Methods 2001; 25: 402–8.

Majander A, Rapola J, Sariola H, Suomalainen A, Pohjavuori M, Pihko H.

Diagnosis of fatal infantile defects of the mitochondrial respiratory

chain: age dependence and postmortem analysis of enzyme activities.

J Neurol Sci 1995; 134: 95–102.

Mancuso M, Salviati L, Sacconi S, Otaegui D, Camano P, Marina A, et al.

Mitochondrial DNA depletion: mutations in thymidine kinase gene with

myopathy and SMA. Neurology 2002; 59: 1197–202.

McFarland R, Taylor RW, Turnbull DM. The neurology of mitochondrial

DNA disease. Lancet Neurol 2002; 1: 343–51.

Moraes CT, Shanske S, Tritschler HJ, Aprille JR, Andreetta F, Bonilla E,

et al. mtDNA depletion with variable tissue expression: a novel genetic

abnormality in mitochondrial diseases. Am J Hum Genet 1991; 48:

492–501.

Nguyen KV, Ostergaard E, Ravn SH, Balslev T, Danielsen ER, Vardag A,

et al. POLG mutations in Alpers syndrome. Neurology 2005; 65: 1493–5.

Norio R. Finnish disease heritage I: characteristics, causes, background.

Hum Genet 2003a; 112: 441–56.

Norio R. The finnish disease heritage III: the individual diseases. Hum

Genet 2003b; 112: 470–526.

Oskoui M, Davidzon G, Pascual J, Erazo R, Gurgel-Giannetti J, Krishna S,

et al. Clinical spectrum of mitochondrial DNA depletion due to

mutations in the thymidine kinase 2 gene. Arch Neurol 2006; 63:

1122–6.

Ostergaard E, Christensen E, Kristensen E, Mogensen B, Duno M,

Shoubridge EA, et al. Deficiency of the alpha subunit of succinate-

coenzyme A ligase causes fatal infantile lactic acidosis with mitochon-

drial DNA depletion. Am J Hum Genet 2007; 81: 383–7.

Pietilainen KH, Naukkarinen J, Rissanen A, Saharinen J, Ellonen P,

Keranen H, et al. Global transcript profiles of fat in monozygotic

twins discordant for BMI: pathways behind acquired obesity. PLoS Med

2008; 5: e51.

Pihko H, Louhimo T, Valanne L, Donner M. CNS in congenital muscular

dystrophy without mental retardation. Neuropediatrics 1992; 23:

116–22.

Saada A, Shaag A, Mandel H, Nevo Y, Eriksson S, Elpeleg O. Mutant

mitochondrial thymidine kinase in mitochondrial DNA depletion

myopathy. Nat Genet 2001; 29: 342–4.

Sarzi E, Bourdon A, Chretien D, Zarhrate M, Corcos J, Slama A, et al.

Mitochondrial DNA depletion is a prevalent cause of multiple

respiratory chain deficiency in childhood. J Pediatr 2007a; 150: 531–4,

534 e1–6.

Sarzi E, Goffart S, Serre V, Chretien D, Slama A, Munnich A, et al.

Twinkle helicase (PEO1) gene mutation causes mitochondrial DNA

depletion. Ann Neurol 2007b; 62: 579–87.

Shibata DK, Arnheim N, Martin WJ. Detection of human papilloma virus

in paraffin-embedded tissue using the polymerase chain reaction. J Exp

Med 1988; 167: 225–30.

Spinazzola A, Viscomi C, Fernandez-Vizarra E, Carrara F, D’Adamo P,

Calvo S, et al. MPV17 encodes an inner mitochondrial membrane

protein and is mutated in infantile hepatic mitochondrial DNA

depletion. Nat Genet 2006; 38: 570–5.

Suomalainen A, Syvanen AC. Quantitative analysis of human DNA

sequences by PCR and solid-phase minisequencing. Mol Biotechnol

2000; 15: 123–31.

Tulinius M, Moslemi AR, Darin N, Holme E, Oldfors A. Novel mutations

in the thymidine kinase 2 gene (TK2) associated with fatal mitochon-

drial myopathy and mitochondrial DNA depletion. Neuromuscul Disord

2005; 15: 412–5.

Van Goethem G, Luoma P, Rantamaki M, Al Memar A, Kaakkola S,

Hackman P, et al. POLG mutations in neurodegenerative disorders with

ataxia but no muscle involvement. Neurology 2004; 63: 1251–7.

Vila MR, Segovia-Silvestre T, Gamez J, Marina A, Naini AB, Meseguer A,

et al. Reversion of mtDNA depletion in a patient with TK2 deficiency.

Neurology 2003; 60: 1203–5.

Vila MR, Villarroya J, Garcia-Arumi E, Castellote A, Meseguer A,

Hirano M, et al. Selective muscle fiber loss and molecular compensation

in mitochondrial myopathy due to TK2 deficiency. J Neurol Sci 2008;

267: 137–41.

Vu TH, Tanji K, Valsamis H, DiMauro S, Bonilla E. Mitochondrial

DNA depletion in a patient with long survival. Neurology 1998; 51:

1190–3.

Page 10 of 10 Brain (2008) A.Go« tz et al.

by guest on February 11, 2016http://brain.oxfordjournals.org/

Dow

nloaded from