Downloaded from molecularcasestudies.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press

1 Novel biallelic variants in MSTO1 associated with mitochondrial myopathy

2

3 Laura Schultz-Rogers1, Alejandro Ferrer1, Nikita R. Dsouza2, Michael T. Zimmermann2,3, Benn

4 E. Smith4, Eric W. Klee1,5, Radhika Dhamija6

5

6 1Center for Individualized Medicine, Mayo Clinic, Rochester, MN, 55905 USA

7 2Bioinformatics Research and Development Laboratory, Genomics Sciences and Precision

8 Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA

9 3Clinical and Translational Sciences Institute, Medical College of Wisconsin, Milwaukee, WI

10 53226, USA

11 4Department of Neurology, Mayo Clinic, Scottsdale, AZ, 85259 USA

12 5Department of Clinical Genomics, Mayo Clinic, Rochester, MN, 55905 USA

13 6Department of Medical Genetics, Mayo Clinic, Phoenix, AZ, 85054 USA

14 Corresponding author’s email: [email protected]

15

16 HPO TERMS:

17 Hypotonia, early HP:0008947; Restrictive lung disease HP:0002091; Elevated creatine kinase

18 HP:0003236; Delayed gross motor development HP:0002194; Speech articulation difficulties

19 HP:0009088; Dysphagia HP:0002015; Waddling gait HP:0002515; Pectus excavatum

20 HP:0000767; Lumbar kyphosis HP:0008454; EMG: myopathic abnormalities HP:0003458

21

22 SHORT RUNNING TITLE: Novel biallelic variants in MSTO1 myopathy

23

24

25

26 Downloaded from molecularcasestudies.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press

27 ABSTRACT

28 Mitochondrial disorders are caused by nuclear and mitochondrial pathogenic variants

29 leading to defects in mitochondrial function and cellular respiration. Recently, the nuclear-

30 encoded mitochondrial fusion MSTO1 (Misato 1) has been implicated in mitochondrial

31 myopathy and ataxia. Here we report on a 30 year old man presenting with a maternally

32 inherited NM_018116.3:c.651C>G, p.F217L missense variant as well as a paternally inherited

33 arr[GRCh37] 1q22(155581773_155706887)x1 deletion encompassing exons 7-14 of MSTO1.

34 His phenotype included muscle weakness, hypotonia, early motor developmental delay, pectus

35 excavatum, and scoliosis. Testing revealed elevated plasma creatine kinase, and

36 electromyogram results were consistent with longstanding generalized myopathy. These

37 phenotypic features overlap well with previously reported patients harboring biallelic MSTO1

38 variants. Additionally, our patient presents with dysphagia and restrictive lung disease, not

39 previously reported for MSTO1-associated disorders. The majority of patients with disease

40 associated variants in MSTO1 present with biallelic variants suggesting autosomal recessive

41 inheritance, however one family has been reported with a single variant and presumed

42 autosomal dominant inheritance. The pattern of inheritance we observed is consistent with the

43 majority of previous reports suggesting an autosomal recessive disorder. We add to our

44 knowledge of the syndrome caused by variants in MSTO1 and provide additional evidence

45 supporting autosomal recessive inheritance. We also describe phenotypic features not reported

46 in previous cases, although further research is needed to confirm they are associated with

47 defects in MSTO1.

48

49 INTRODUCTION

50 Mitochondrial dysfunction leads to a diverse array of disorders most often characterized

51 by myopathic and neurologic symptoms (Chinnery 1993). As the mitochondria are essential for

52 normal aerobic metabolism, tissues and organs with high energy demands such as skeletal Downloaded from molecularcasestudies.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press

53 muscle are particularly sensitive to mitochondrial disruption (Wallace 1999). Damaging variants

54 in over 1000 nuclear encoded and mitochondrial can affect respiratory chain complexes,

55 mtDNA replication and maintenance, and dynamic fusion and fission processes. Fusion helps

56 maintain a healthy population of mitochondria by distributing mtDNA and metabolites and

57 eliminating damaged mitochondria through mitophagy (for review see (El-Hattab et al. 2018)).

58 MSTO1 (Misato 1) is a nuclear gene that contains a region homologous to the

59 Tubulin/FtsZ, GTPase superfamily which contains that regulate

60 segregation in eukaryotes and mitochondrial fission in eukaryotic algae (Kimura and Okano

61 2007). It encodes a soluble cytoplasmic that translocates to the mitochondrial outer

62 membrane and promotes normal fusion dynamics (Kimura and Okano 2007; El-Hattab et al.

63 2018; Nasca et al. 2017; Gal et al. 2017). In drosophila knockdown of DML1, the homolog of

64 human MSTO1, leads to defects in chromosome segregation during mitosis (Miklos et al. 1997).

65 In S. cerevisiae, disruption of DML1 causes impaired inheritance of mtDNA and segregation of

66 mitochondria (Gurvitz et al. 2002). Inhibition of MSTO1 in HeLa cells results in nuclear and

67 mitochondrial fragmentation, reduced fusion, and apoptosis, while overexpression results in

68 perinuclear aggregations of mitochondria (Kimura and Okano 2007; Gal et al. 2017).

69 MSTO1 was first associated with human disease in 2017 when Nasca et al. reported on

70 three patients with compound heterozygous variants in MSTO1 presenting with myopathy and

71 cerebellar ataxia (MIM:617675). Functional studies in patient fibroblasts confirmed partially

72 reduced transcript and protein expression as well as fragmentation of the mitochondrial network

73 and decreased mitochondrial fusion (Nasca et al. 2017). To date, a total of 11 patients have

74 been reported in the literature with myopathy and ataxia attributed to pathogenic variants in

75 MSTO1. Consistent phenotypic features observed across patients include onset in the first year

76 of life, severe motor delay, cerebral atrophy on MRI, short stature, myopathic pattern on EMG,

77 and elevated plasma creatine kinase (CK) (Nasca et al. 2017; Iwama et al. 2018; Li, Jin, and Downloaded from molecularcasestudies.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press

78 Wu 2019)(Table 1). Additionally, several patients presented with cognitive deficiencies, skeletal

79 abnormalities, tremors, and retinopathy.

80 Here we present a 30 year old male with phenotypic features consistent with previously

81 reported MSTO1 patients. We report additional phenotypic features seen in our patient including

82 dysphagia and restrictive lung disease which may be the consequence of the associated

83 myopathy. Our patient has a novel partial gene deletion adding to the spectrum of variants

84 associated with MSTO1-associated mitochondrial myopathy.

85

86 RESULTS

87 Clinical Presentation

88 A 30 year old Caucasian male presented with moderate-to-severe proximal and distal

89 muscle weakness and significantly decreased muscle tone. He had congenital hypotonia with

90 poor head control and was diagnosed with “muscular dystrophy of unknown type” at 18 months

91 of age. His gross and fine motor milestones were significantly delayed, including walking at 26

92 months of age and the inability to pedal a bike or run as a child. Learning disability was noted at

93 early childhood. Neurological examination at age 30 was significant for proximal and distal

94 muscle weakness with hypotonia, mild facial weakness with ptosis, diminished muscle bulk, no

95 sensory deficits and preserved muscle reflexes. Gait was wide based and waddling. Swallowing

96 video fluoroscopy revealed mild oropharyngeal dysphagia. Electromyogram (EMG) results were

97 consistent with a longstanding, generalized myopathy, with electrodiagnostic features that would

98 predict fiber splitting, necrosis, inflammation, or vacuolar change. He has persistently elevated

99 CK levels with titers between 1000 and 2000 U/L. Pulmonary function tests showed a chronic

100 restrictive pattern with Forced Vital Capacity (FVC) at 44-49% of predicted leading to a

101 diagnosis of restrictive lung disease (Figure 1A,B).

102 Additional phenotypic features reported in the patient include pectus excavatum that was

103 surgically corrected at age 18, as well as scoliosis characterized by mild reversal of cervical Downloaded from molecularcasestudies.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press

104 lordosis and a trace gentle convex curvature of the lumbar spine to the right (Figure 1C,D). An

105 echocardiogram was normal. The family history was unremarkable with the exception of a

106 maternal grandmother who died of complication from ALS at age 74 (Figure 2).

107

108 Genomic Analysis

109 Initial genetic testing included a 141 gene panel (Neuromuscular Genetics Panel; Mayo

110 Clinic) and dystrophin deletion/duplication sequencing analysis. None of the variants reported

111 were considered relevant (Table 2). Testing for Pompe’s disease was also negative.

112 Mitochondrial DNA NGS testing was performed through GeneDX, and a homoplasmic variant of

113 uncertain significance (m.8045A>G, p.I154V) was reported in MT-CO2. This variant was also

114 considered not diagnostic as the patient’s unaffected mother was homoplasmic for the same

115 variant. Whole exome sequencing performed through GeneDx reported two potentially clinically

116 relevant variants; a maternally inherited NM_018116.3:c.651C>G, p.F217L missense variant of

117 uncertain significance and a likely pathogenic paternally inherited arr[GRCh37]

118 1q22(155581773_155706887)x1 copy number change (Figure 3A). The copy number change

119 was originally detected through GeneDx’s NGS-CNV caller (Retterer et al. 2015) and was

120 confirmed by GeneDx with the GenomeDx v5 chromosomal array. The missense variant was

121 confirmed by GeneDx via direct gene-specific PCR.

122 The p.F217L missense variant lies in exon 7 of 14 in the tubulin domain. The affected

123 amino acid is highly conserved across species (Figure 3B) and in silico predictions agree the

124 variant is likely damaging (Polyphen-2 (Adzhubei et al. 2010), SIFT (Sim et al. 2012), M-CAP

125 (Jagadeesh et al. 2016), PredictSNP2 (Bendl et al. 2016)). The p.F217Lvariant could possibly

126 affect the structural conformation as it is a part of an alpha helix and the only hydrophobic

127 residue within a region of charged and hydrophilic residues. The

128 1q22(155581773_155706887x1) copy number variant is a 125 kb deletion encompassing exons

129 7-14 of MSTO1, exon 7-13 of the MSTO2P pseudogene, and all of DAP3 (death associated Downloaded from molecularcasestudies.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press

130 protein 3) and YY1AP1 (YY1 associated protein 1). While the homology between MSTO1 and

131 its pseudogene could affect the detection of CNV’s in this gene, the fact that the missense

132 variant in exon 7 was called as homozygous supports the presence of a deletion in this gene.

133 MSTO2P is not associated with any human disorders other than potential links to cancer, not

134 reported in our patient. Biallelic loss of function variants in YY1AP1 are associated with Grange

135 syndrome, while missense and small insertion variants in DAP3 have been associated with

136 metabolic disorders and autism spectrum disorder. Since our patient does not have a second

137 damaging variant reported in these genes and his phenotype does not overlap with these

138 syndromes, we concluded that it is not likely that a single copy loss of any of these three genes

139 is contributing to our patient’s phenotype.

140 MSTO1 is not predicted to be intolerant to loss of function or missense variation based

141 on gene constraints in gnomAD (pLI=0, Z=0) (Lek et al. 2016). However, as autosomal

142 recessive inheritance is expected it is possible to see damaging variants in a heterozygous state

143 in the normal population. The p.F217L variant is observed at a very low frequency in gnomAD

144 (0.007%) with no homozygotes reported. This variant has not been previously reported in

145 HGMD or ClinVar. The 1q22(155581773_155706887) deletion has not been previously

146 observed in normal or diseased individuals. Based on the Database of Genomic Variants

147 similarly sized deletions have been detected in heterozygotes in the normal population

148 (nsv436522, nsv3043, nsv48213)(MacDonald et al. 2014).

149

150 DISCUSSION

151 Here we present a patient with novel compound heterozygous variants in MSTO1 and

152 mitochondrial myopathy. His phenotypic presentation overlaps well with previously reported

153 patients including early onset muscle weakness, motor delay, elevated plasma CK, myopathic

154 pattern on EMG, and skeletal abnormalities. At 30 years old, our patient is the oldest reported

155 patient with biallelic variants in MSTO1, adding important knowledge about potential lifespan for Downloaded from molecularcasestudies.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press

156 other similarly affected patients. We report features including restrictive lung disease and

157 dysphagia not previously identified in patients with MSTO1-associated mitochondrial myopathy.

158 These additional clinical findings may be the consequence of the associated myopathy.

159 Awareness of the full phenotypic spectrum associated with this disorder is critical for proper

160 diagnosis of future patients. As the role of tubulins in mitochondrial function becomes more

161 apparent (Mado et al. 2019), it will be important to elucidate the role of MSTO1 in normal

162 mitochondrial dynamics and how disruption leads to myopathy as well as skeletal, retinal, and

163 other phenotypic features observed.

164 The p.F217L missense variant seen in our patient lies in the second tubulin domain on

165 the MSTO1 protein. Five of the six previously reported pathogenic missense variants lie in one

166 of the tubulin domains, suggesting these regions are critical for normal protein function and/or

167 stability. Regions of proteins that bind to other proteins often have a hydrophobic center with a

168 hydrophilic and charged ring surrounding them, much like the region around p.F217. If protein

169 interactions with the MSTO1 tubulin domain occur at p.F217, then p.L217 may be compatible

170 with the overall pattern of protein interfaces, but with a reduced and smaller hydrophobic center,

171 which may not be as specific for its binding partners.

172 Similar to our patient, five of the eleven previously reported patients were compound

173 heterozygotes with one missense variant and one loss of function variant (Figure 3A). The two

174 most severely affected patients were compound heterozygotes for two missense variants.

175 Nasca et al. hypothesized that the reduction in protein expression in these patients was due to

176 instability caused by the missense variants. In the gnomAD database of healthy individuals

177 there is only one individual reported as homozygous for any loss of function allele and this call

178 was flagged as dubious. This suggests that MSTO1 is associated with disease in an autosomal

179 recessive manner wherein biallelic loss of function variants are likely lethal but retention of a

180 partially functional missense allele in conjunction with a loss of function allele results in a

181 disease state. This is supported by studies in drosophila in which null mutations are embryonic Downloaded from molecularcasestudies.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press

182 lethal (Miklos et al. 1997). Here we present an additional patient with a loss of function variant,

183 in this case a partial gene deletion, in combination with a missense variant in MSTO1 resulting

184 in mitochondrial myopathy.

185 Contrary to the other reported cases, Gal et al. reported a heterozygous missense

186 variant in members of a family presenting with myopathy, ataxia, and neurodevelopmental

187 impairments (Gal et al. 2017). A second alteration in MSTO1 was not detected with Sanger

188 sequencing of the genomic DNA or real-time PCR analysis of copy number variation. The

189 authors proposed autosomal dominant inheritance of a multi-systemic clinical phenotype.

190 However, the phenotypic features in this family do not overlap well with other reported patients

191 (Table 1). In particular, age of onset was later (15-53 years old), plasma CK levels were normal,

192 and cerebellar atrophy was not observed. Functional studies in fibroblasts from patients 1 and 2

193 (as labeled in the Gal et al. study) did confirm a decrease in transcript and protein expression,

194 mitochondrial fusion, and fragmented and aggregated mitochondria. These discrepant reports

195 may be explained by a genotype/phenotype correlation in which the mode of inheritance

196 depends on the location of the variant or a separate underlying genetic condition.

197 This case adds to our current knowledge on mitochondrial myopathy associated with

198 biallelic pathogenic variants in MSTO1. We present novel phenotypic features not previously

199 described in association with MSTO1-related disease, and add support to reports of autosomal

200 recessive inheritance. We expect that additional MSTO1 cases and further research will

201 elucidate the full clinical spectrum of this disorder.

202

203 METHODS

204 Whole exome sequencing was performed by GeneDx. The mean depth of coverage was 157x

205 and the quality threshold (percentage of sequence covered by at least 10 reads) was 98.9%.

206 Briefly, a proprietary capture system was used for next-generation sequencing and targets were

207 sequenced with paired-end reads on an Illumina platform. Reads were aligned to Downloaded from molecularcasestudies.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press

208 GRCH37/UCSC hg 19. Sequence variants and deletions or duplications involving three or more

209 coding exons were called with XomeAnalyzer. Variants were reported according to the Human

210 Genome Variation Society (HGVS) guidelines. Missense variants were clinically confirmed with

211 direct gene-specific PCR. Copy number variants were clinically confirmed by chromosomal

212 microarray. Briefly, the GeneDx GenomeDx v5 chromosomal array contains 118,000

213 oligonucleotide probes and 66,000 SNP probes throughout the genome used for detecting copy

214 number variants.

215 Sequencing Coverage:

Mean Depth of Coverage 157x

Quality Threshold 98.9%

216

217 As a separate test from whole exome sequencing, mitochondrial sequencing and

218 deletion testing of the mitochondrial genome was performed by GeneDx. Briefly, the

219 mitochondrial genome was amplified and sequenced using a solid state sequencing by-

220 synthesis process and the sequence was compared to the revised Cambridge Reference

221 Sequence (rCRS). Disease associated variants were confirmed by conventional dideoxy

222 sequence analysis.

223

224 ADDITIONAL INFORMATION

225 Data Deposition and Access— Single nucleotide variants identified and interpreted by

226 GeneDx can be found in the ClinVar database (accession number SCV000965685).

227 Ethics Statement— This study was approved by the Mayo Clinic institutional review board and

228 all participants provided written informed consent for genetic testing.

229 Acknowledgments – We would like to thank the patient and his family for participating in this

230 study. Downloaded from molecularcasestudies.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press

231 Author Contributions— L.S.R, A.F., and E.W.K. analyzed patient variants and wrote

232 manuscript. R.D. and B.S. oversaw patient care and genetic interpretation. N.R.D. and M.T.Z.

233 interpreted protein structure. All authors reviewed and approved the final manuscript.

234 Funding - This study was supported by the Mayo Clinic Center for Individualized Medicine.

235

236 REFERENCES

237 Adzhubei, I. A., S. Schmidt, L. Peshkin, V. E. Ramensky, A. Gerasimova, P. Bork, A. S. Kondrashov, and S. 238 R. Sunyaev. 2010. 'A method and server for predicting damaging missense mutations', Nat 239 Methods, 7: 248-9. 240 Bendl, J., M. Musil, J. Stourac, J. Zendulka, J. Damborsky, and J. Brezovsky. 2016. 'PredictSNP2: A Unified 241 Platform for Accurately Evaluating SNP Effects by Exploiting the Different Characteristics of 242 Variants in Distinct Genomic Regions', PLoS Comput Biol, 12: e1004962. 243 Chinnery, P. F. 1993. 'Mitochondrial Disorders Overview.' in M. P. Adam, H. H. Ardinger, R. A. Pagon, S. E. 244 Wallace, L. J. H. Bean, K. Stephens and A. Amemiya (eds.), GeneReviews((R)) (Seattle (WA)). 245 El-Hattab, A. W., J. Suleiman, M. Almannai, and F. Scaglia. 2018. 'Mitochondrial dynamics: Biological 246 roles, molecular machinery, and related diseases', Mol Genet Metab, 125: 315-21. 247 Gal, A., P. Balicza, D. Weaver, S. Naghdi, S. K. Joseph, P. Varnai, T. Gyuris, A. Horvath, L. Nagy, E. L. 248 Seifert, M. J. Molnar, and G. Hajnoczky. 2017. 'MSTO1 is a cytoplasmic pro-mitochondrial fusion 249 protein, whose mutation induces myopathy and ataxia in humans', EMBO Mol Med, 9: 967-84. 250 Gurvitz, A., A. Hartig, H. Ruis, B. Hamilton, and H. G. de Couet. 2002. 'Preliminary characterisation of 251 DML1, an essential Saccharomyces cerevisiae gene related to misato of Drosophila 252 melanogaster', FEMS Yeast Res, 2: 123-35. 253 Iwama, K., T. Takaori, A. Fukushima, J. Tohyama, A. Ishiyama, C. Ohba, S. Mitsuhashi, S. Miyatake, A. 254 Takata, N. Miyake, S. Ito, H. Saitsu, T. Mizuguchi, and N. Matsumoto. 2018. 'Novel recessive 255 mutations in MSTO1 cause cerebellar atrophy with pigmentary retinopathy', J Hum Genet, 63: 256 263-70. 257 Jagadeesh, K. A., A. M. Wenger, M. J. Berger, H. Guturu, P. D. Stenson, D. N. Cooper, J. A. Bernstein, and 258 G. Bejerano. 2016. 'M-CAP eliminates a majority of variants of uncertain significance in clinical 259 exomes at high sensitivity', Nat Genet, 48: 1581-86. 260 Kimura, M., and Y. Okano. 2007. 'Human Misato regulates mitochondrial distribution and morphology', 261 Exp Cell Res, 313: 1393-404. 262 Lek, M., K. J. Karczewski, E. V. Minikel, K. E. Samocha, E. Banks, T. Fennell, A. H. O'Donnell-Luria, J. S. 263 Ware, A. J. Hill, B. B. Cummings, T. Tukiainen, D. P. Birnbaum, J. A. Kosmicki, L. E. Duncan, K. 264 Estrada, F. Zhao, J. Zou, E. Pierce-Hoffman, J. Berghout, D. N. Cooper, N. Deflaux, M. DePristo, R. 265 Do, J. Flannick, M. Fromer, L. Gauthier, J. Goldstein, N. Gupta, D. Howrigan, A. Kiezun, M. I. 266 Kurki, A. L. Moonshine, P. Natarajan, L. Orozco, G. M. Peloso, R. Poplin, M. A. Rivas, V. Ruano- 267 Rubio, S. A. Rose, D. M. Ruderfer, K. Shakir, P. D. Stenson, C. Stevens, B. P. Thomas, G. Tiao, M. 268 T. Tusie-Luna, B. Weisburd, H. H. Won, D. Yu, D. M. Altshuler, D. Ardissino, M. Boehnke, J. 269 Danesh, S. Donnelly, R. Elosua, J. C. Florez, S. B. Gabriel, G. Getz, S. J. Glatt, C. M. Hultman, S. 270 Kathiresan, M. Laakso, S. McCarroll, M. I. McCarthy, D. McGovern, R. McPherson, B. M. Neale, A. 271 Palotie, S. M. Purcell, D. Saleheen, J. M. Scharf, P. Sklar, P. F. Sullivan, J. Tuomilehto, M. T. 272 Tsuang, H. C. Watkins, J. G. Wilson, M. J. Daly, D. G. MacArthur, and Consortium Exome Downloaded from molecularcasestudies.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press

273 Aggregation. 2016. 'Analysis of protein-coding genetic variation in 60,706 humans', Nature, 536: 274 285-91. 275 Li, K., R. Jin, and X. Wu. 2019. 'Whole-exome sequencing identifies rare compound heterozygous 276 mutations in the MSTO1 gene associated with cerebellar ataxia and myopathy', Eur J Med 277 Genet. 278 MacDonald, J. R., R. Ziman, R. K. Yuen, L. Feuk, and S. W. Scherer. 2014. 'The Database of Genomic 279 Variants: a curated collection of structural variation in the ', Nucleic Acids Res, 280 42: D986-92. 281 Mado, K., V. Chekulayev, I. Shevchuk, M. Puurand, K. Tepp, and T. Kaambre. 2019. 'On the role of 282 tubulin, plectin, desmin and vimentin in the regulation of mitochondrial energy fluxes in muscle 283 cells', Am J Physiol Cell Physiol. 284 Miklos, G. L., M. Yamamoto, R. G. Burns, and R. Maleszka. 1997. 'An essential cell division gene of 285 Drosophila, absent from Saccharomyces, encodes an unusual protein with tubulin-like and 286 myosin-like peptide motifs', Proc Natl Acad Sci U S A, 94: 5189-94. 287 Nasca, A., C. Scotton, I. Zaharieva, M. Neri, R. Selvatici, O. T. Magnusson, A. Gal, D. Weaver, R. Rossi, A. 288 Armaroli, M. Pane, R. Phadke, A. Sarkozy, F. Muntoni, I. Hughes, A. Cecconi, G. Hajnoczky, A. 289 Donati, E. Mercuri, M. Zeviani, A. Ferlini, and D. Ghezzi. 2017. 'Recessive mutations in MSTO1 290 cause mitochondrial dynamics impairment, leading to myopathy and ataxia', Hum Mutat, 38: 291 970-77. 292 Retterer, K., J. Scuffins, D. Schmidt, R. Lewis, D. Pineda-Alvarez, A. Stafford, L. Schmidt, S. Warren, F. 293 Gibellini, A. Kondakova, A. Blair, S. Bale, L. Matyakhina, J. Meck, S. Aradhya, and E. Haverfield. 294 2015. 'Assessing copy number from exome sequencing and exome array CGH based on CNV 295 spectrum in a large clinical cohort', Genet Med, 17: 623-9. 296 Sim, N. L., P. Kumar, J. Hu, S. Henikoff, G. Schneider, and P. C. Ng. 2012. 'SIFT web server: predicting 297 effects of amino acid substitutions on proteins', Nucleic Acids Res, 40: W452-7. 298 Wallace, D. C. 1999. 'Mitochondrial diseases in man and mouse', Science, 283: 1482-8.

299

300 FIGURE LEGENDS

301 Table 1. Phenotypic spectrum of MSTO1-associated myopathy. Comparisons of variants,

302 inheritance, and phenotypic features seen across all currently reported patients with MSTO1-

303 associated mitochondrial myopathy. n/a = not available.

304

305 Table 2. Discarded variants of uncertain significance. Prior to whole exome sequencing, a

306 141 gene Neuromuscular Genetics Panel was performed on patient DNA. Several variants of

307 uncertain significance were reported, but all were discarded as candidates for the reasons

308 described in the table.

309 Downloaded from molecularcasestudies.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press

310 Figure 1. Pulmonary function and skeletal abnormalities in a patient with biallelelic

311 MSTO1 variants. A) Air flow (Liters per second) vs. volume (L) displays severe restriction of

312 pulmonary function. B) The forced expiratory volume in one second (FEV1)/forced vital capacity

313 (FVC) is normal; the FEV1 is moderately to severely reduced; the FVC is reduced; maximal

314 expiratory pressure (MEP) 24% predicted, maximal inspiratory pressure (MIP) 38% predicted.

315 Overall results of the pulmonary function test confirm a chronic restrictive pattern. C) Skeletal

316 radiographs show mild reversal of cervical lordosis and D) trace gentle convex curvature of the

317 lumbar spine to the right.

318

319 Figure 2. Family pedigree. A three generation family pedigree in which the proband is the only

320 affected individual. Each MSTO1 variant was inherited from an unaffected parent.

321

322 Figure 3. Gene location of disease associated MSTO1 variants. A) Map of currently known

323 MSTO1 variants associated with mitochondrial myopathy. Green and blue regions denote the

324 tubulin domains. Most of the variants cluster in the known tubulin domains. Figure generated

325 using ProteinPaint, St. Jude Children’s Research Hospital. B) The amino acid affected by our

326 patient’s missense variant (red box) shows high conservation among orthologs.

327

328 TABLES

329 Table 1.

Nasca Nasca Nasca Iwama Iwama Gal Gal Li 2019; Li 2019; Present Gal 2017; Gal 2017; Patient 2017; 2017; 2017; 2018; 2018; 2017; 2017; Patient 1 Patient 2 study Patient 1 Patient 2 Patient 1 Patient 2 Patient 3 Patient 1 Patient 2 Patient 3 Patient 4 Variant c.1033C c.1033C c.971C > c.836 G > c.836 G > c.836G > c.836G > c.651 c.22G > c.22G > 1 c.22G > A c.22G > A > T > T T A A A A C>G A A (cDNA) Variant 1 p.Arg279 p.Arg279 p.Arg279 p.Arg279 p.Val8Me p.Val8Me p.R345C p.R345C p.T324I p.F217L p.Val8Met p.Val8Met (protein His His His His t t ) Variant 1q22 125 c.1128C c.1128C 966+1G c.1099– c. 79 C > c.1259D c.1259D 2 kb None None None None > A > A > A 1 G > A T elG elG (cDNA) deletion Variant n/a p.Val367 p.Gly420 p.Gly420 p.F376L p.F376L p.Gln27* n/a None None None None 2 (splice) Trpfs *2 ValfsTer ValfsTer Downloaded from molecularcasestudies.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press

(protein 2 2 ) Variant Missense Missense Missense Missense Missense Missense Missense Missense Missense Missense Missense Missense 1 Type Partial Variant Splice Splice Nonsens Frameshi Frameshi Missense Missense gene 2 Type site site e ft ft deletion Inherita AR AR AR AR AR AR AR AR AD AD AD AD nce Early Age of 8-9 8-9 10 Childhoo 1 year 5 months Perinatal 1 year Perinatal 53 years 15 years childhoo onset months months months d onset d Motor Yes Yes Yes Yes Yes Yes Yes Yes Yes No No Yes delay? Frontal atrophy Pituitary Cerebell Cerebell Cerebell Cerebell Cerebell Cerebell and adenoma Brain Cerebella ar ar ar ar ar ar n/a enlarged and mild n/a n/a MRI r atrophy atrophy atrophy atrophy atrophy atrophy atrophy interhemis cerebellar pheric ectopia fissure Growth impair Yes Yes n/a Yes Yes Yes Yes No Yes n/a n/a n/a ment Plasma CK 1200 1872 4520 430 916 n/a 2544 1292 Normal Normal Normal n/a value Tremor Yes Yes Yes Yes Yes No No No No No No No s Speech Develop Cogniti delay, Mild Mild Learning mental Mental Learning Learning ve speech develop develop disability, Developm None None delay, retardatio None difficultie difficultie deficien articulati mental mental speech ental delay slurred n s s cy on delay delay difficulty speech difficulty Bilateral retinal pigmenta Pigmenta Ophthal ry ry Pigmenta mologi Bilateral denaturat Visual retinopat ry retinal cal papillary Normal ion, impairme Myopia No No No No No hy with denaturat finding pallor severe nt papillary ion s hypermet pallor ropia, and esotropia Severe III digit on Bone Pectus asymmetr foot develop excavatu y of the Pes missing, mental Multiple m, Skeletal chest, Asymmet planus bone age problems Laxity of arthrogry Pes reversal abnorm pectus ry of the None None and mild delayed, None , pectus the knee posis, planus of alities excavatu thorax genu joint excavatu joints transient cervical m, and valgus hyperlaxity m, lordosis, marked , pes kyphosco scoliosis scoliosis varus liosis Walked Walkin Able to No Severe at 4 g Unable to Unable walk but walking Able to Able to Waddling Able to episodic Able to Able to years but affecte walk to walk occasion as of 3 walk walk gait walk ataxic gait walk walk loss at d? al falls years repeatedly 13 years Muscle biopsy Yes Yes Yes n/a n/a n/a n/a n/a Yes n/a n/a n/a abnorm alities EMG Myopathi Myopathi Myopathi Myopathi Myopathi Myopathic finding n/a n/a n/a n/a n/a n/a c pattern c pattern c pattern c pattern c pattern pattern s Downloaded from molecularcasestudies.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press

Depressio n, Hypoacusi Neurolo hallucinati s, anxiety Social gical Autism, ons, Social None None None None None None Autism and anxiety, Disorde anxiety schizophre anxiety depressed autism rs nia, mood hypoacusi s Respira tory None None None None None None None Yes None None None None sympto ms Lipomatosi Inflammat s, Dysphagi ory hyperthyro a, lipomas, idism, Other None None None None None None None restrictiv Hyperthyr None None mitral and e lung eosis, trisucpidal disease hyperprola insufficien ctanemia cy 330 Table 2.

HGVS HGVS DNA Protein Variant Predicted dbSNP/dbVA Gene Chromosome Reference Reference Type Effect R ID Genotype Reason Discarded Lack of phenotypic overlap (no lipodystrophy); no CAVIN1 17:40556955 c.923A>G p.Y308C Missense Substitution rs146799286 Heterozygous second variant found Lack of phenotypic overlap; no second SMCHD1 18:2732423 c.3209T>C p.I1070T Missense Substitution rs113434340 Heterozygous variant found in DUX4 Lack of phenotypic overlap (no cardiovascular TMEM43 3:14177385 c.859C>T p.H287Y Missense Substitution rs780299346 Heterozygous symptoms) Autosomal dominant association with disease but previously reported in unaffected TTN 2:179419690 c.80792T>G p.L26931R Missense Substitution rs72648234 Heterozygous individuals Autosomal dominant association with disease but previously reported in unaffected TTN 2:179587094 c.18688G>A p.A6230T Missense Substitution rs759713604 Heterozygous individuals 331

332 Variant Table:

HGVS Parent Chromosom HGVS DNA Protein Variant Predicted dbSNP/dbVA of Gene e Reference Reference Type Effect R ID Genotype origin

Heterozyg MSTO1 1:155581864 c.651C>G p.Phe217Leu Missense Substitution rs776826330 ous Mother Copy 1q22(155581773 number Partial gene Heterozyg MSTO1 1q22 _155706887)x1 n/a change deletion n/a ous Father 333 Downloaded from molecularcasestudies.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press

334 FIGURES 335 336 Figure 1:

337 338 Figure 2:

339 340 341 342 Downloaded from molecularcasestudies.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press

343 Figure 3:

344 Downloaded from molecularcasestudies.cshlp.org on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press

Novel biallelic variants in MSTO1 associated with mitochondrial myopathy

Laura Schultz-Rogers, Alejandro Ferrer, Nikita R. Dsouza, et al.

Cold Spring Harb Mol Case Stud published online October 11, 2019 Access the most recent version at doi:10.1101/mcs.a004309

Published online October 11, 2019 in advance of the full issue.

Accepted Peer-reviewed and accepted for publication but not copyedited or typeset; accepted Manuscript manuscript is likely to differ from the final, published version. Published onlineOctober 11, 2019in advance of the full issue.

Creative This article is distributed under the terms of the Commons http://creativecommons.org/licenses/by-nc/4.0/, which permits reuse and License redistribution, except for commercial purposes, provided that the original author and source are credited. Email Alerting Receive free email alerts when new articles cite this article - sign up in the box at the Service top right corner of the article or click here.

Published by Cold Spring Harbor Laboratory Press