Mitochondrial optic neuropathies - disease mechanisms and therapeutic strategies

Patrick Yu-Wai-Man, Philip G Griffiths, Patrick F Chinnery, Patrick Yu-Wai-Man, Philip G Griffiths, Patrick F Chinnery

Abstract

Leber hereditary optic neuropathy (LHON) and autosomal-dominant optic atrophy (DOA) are the two most common inherited optic neuropathies in the general population. Both disorders share striking pathological similarities, marked by the selective loss of retinal ganglion cells (RGCs) and the early involvement of the papillomacular bundle. Three mitochondrial DNA (mtDNA) point mutations; m.3460G>A, m.11778G>A, and m.14484T>C account for over 90% of LHON cases, and in DOA, the majority of affected families harbour mutations in the OPA1 gene, which codes for a mitochondrial inner membrane protein. Optic nerve degeneration in LHON and DOA is therefore due to disturbed mitochondrial function and a predominantly complex I respiratory chain defect has been identified using both in vitro and in vivo biochemical assays. However, the trigger for RGC loss is much more complex than a simple bioenergetic crisis and other important disease mechanisms have emerged relating to mitochondrial network dynamics, mtDNA maintenance, axonal transport, and the involvement of the cytoskeleton in maintaining a differential mitochondrial gradient at sites such as the lamina cribosa. The downstream consequences of these mitochondrial disturbances are likely to be influenced by the local cellular milieu. The vulnerability of RGCs in LHON and DOA could derive not only from tissue-specific, genetically-determined biological factors, but also from an increased susceptibility to exogenous influences such as light exposure, smoking, and pharmacological agents with putative mitochondrial toxic effects. Our concept of inherited mitochondrial optic neuropathies has evolved over the past decade, with the observation that patients with LHON and DOA can manifest a much broader phenotypic spectrum than pure optic nerve involvement. Interestingly, these phenotypes are sometimes clinically indistinguishable from other neurodegenerative disorders such as Charcot-Marie-Tooth disease, hereditary spastic paraplegia, and multiple sclerosis, where mitochondrial dysfunction is also thought to be an important pathophysiological player. A number of vertebrate and invertebrate disease models has recently been established to circumvent the lack of human tissues, and these have already provided considerable insight by allowing direct RGC experimentation. The ultimate goal is to translate these research advances into clinical practice and new treatment strategies are currently being investigated to improve the visual prognosis for patients with mitochondrial optic neuropathies.

Copyright © 2010 Elsevier Ltd. All rights reserved.

Figures

Fig. 1
Fig. 1
The mitochondrial respiratory chain and oxidative phosphorylation. Reproduced with permission from Nijtmans et al. (2004).
Fig. 2
Fig. 2
The human mitochondrial genome. Protein coding (yellow), rRNA (red), and tRNA (purple) genes are depicted on the heavy (H-, outer) and light (L-, inner) strands. The 22 tRNAs are indicated by their cognate amino acid letter code and the 2 rRNAs by their sedimentation coefficients (12S and 16S). The origins of mtDNA replication and the direction of synthesis are denoted by OH for the H-strand, and OL for the L-strand.
Fig. 3
Fig. 3
Mitochondrial and nuclear-encoded subunits of the mitochondrial respiratory chain complexes.
Fig. 4
Fig. 4
Skeletal muscle sections illustrating the characteristic histochemical features of mitochondrial dysfunction: (A) Ragged-red muscle fibre identified using the modified Gomori trichome stain. The red component of the staining mixture is selectively sequestered by mitochondria, which have accumulated in the subsarcolemmal region, giving the fibre an irregular red outline, (B) Serial section of the same muscle fibre after SDH staining. This is a more specific assay for detecting the subsarcolemmal accumulation of mitochondria, SDH being a specific marker for complex II activity, (C) Abnormal COX–SDH histochemistry from a patient with chronic progressive external ophthalmoplegia (CPEO) due to a single 5 kb mtDNA deletion, showing normal COX-positive (Brown) and energy deficient, COX-negative (Blue) muscle fibres.
Fig. 5
Fig. 5
The minimum prevalence of inherited optic nerve disorders in the North of England.
Fig. 6
Fig. 6
Summary of linkage studies investigating the existence of putative LHON nuclear modifiers on the X-chromosome. A nonparametric LOD score (NPL) >2 is indicative of significant linkage, and these chromosomal areas possibly harbour susceptibility loci which influence the risk of visual loss among LHON carriers. The different studies are colour coded: red (Hudson et al., 2005), blue (Shankar et al., 2008), and black (Ji et al., 2010). Reproduced with permission from Ji et al. (2010).
Fig. 7
Fig. 7
The complex interaction of genetic, hormonal, and environmental factors in the pathophysiology of LHON.
Fig. 8
Fig. 8
The optic disc appearance of a patient with a confirmed pathogenic OPA1 mutation showing pallor of the neuro-retinal rim, which is more marked temporally. The bottom panels illustrate the pattern of retinal nerve fibre layer (RNFL) thinning seen in patients with OPA1 mutations, with relative sparing of the nasal peripapillary quadrant. The RNFL profile for each eye is superimposed on the normal distribution percentiles, and compared with each other (Bottom left panel). Various measurement parameters are automatically generated by the analysis software including sectorial RNFL thickness for each individual quadrant and clock hour, and an overall value for the average RNFL thickness (Bottom middle panel). The normal distribution indices are colour-coded: (i) red <1%, (ii) yellow 1–5%, (iii) green 5–95%, and (iv) white >95% (Bottom right panel).
Fig. 9
Fig. 9
Age-related decrease in average RNFL thickness, consistent with progressive retinal ganglion cell loss among OPA1 mutational carriers (n = 40). Spearman rank correlation coefficient = −0.2419, P = 0.0307 (PWYM, unpublished data).
Fig. 10
Fig. 10
Optic nerve sections stained for myelin and mitochondrial COX activity: (A) Sudan black staining revealing the presence of myelin posterior to the lamina cribosa, (B) marked differential COX activity in the pre- and post-lamina cribosa segments, with intense COX staining in transverse sections taken from the pre-laminar region (C), and significantly lower levels of COX activity in the pos-tlaminar region (D). Reproduced with permission from Andrews et al. (1999a).
Fig. 11
Fig. 11
Histology, mitochondrial histochemistry and immunohistochemistry (IHC) performed on serial longitudinal optic nerve sections: (A) haematoxylin and eosin, (B) Van Gieson preparation for connective tissue fibres (Red), with the arrow pointing towards the lamina cribosa, (C) Weigert iron haematoxylin preparation for myelin (Dark blue), (D) COX activity, with a darker stain (Brown) evident in the unmyelinated pre-lamina cribosa segment of the optic nerve, (E) IHC for COX subunit IV revealing a pattern consistent with the level of mitochondrial enzyme activity, (F ) IHC for Nav 1.1 showing a greater concentration of these specific voltage gated Na+ channels in the pre-laminar region, (G) IHC for Nav 1.2 demonstrating a uniformly pale labelling pattern in both pre- and post-lamina cribosa areas, (H) IHC for Nav 1.6 with a strong staining reaction observed in the unmyelinated pre-laminar optic nerve, and (I) control optic nerve section with the primary antibody omitted. Reproduced with permission from Barron et al. (2004).
Fig. 12
Fig. 12
The importance of the cytoskeleton in maintaining the differential concentration of mitochondria in the pre- and post-lamina cribosa segments of the optic nerve. The left panel shows a longitudinal section of a human optic nerve stained sequentially for COX and SDH. The pre-lamina, unmyelinated segment has a much darker COX staining consistent with the higher concentration of mitochondria. The right panel is a schematic representation of the cytoskeletal–mitochondrial interactions which facilitate the transport, distribution, and localisation of mitochondria to different areas of the optic nerve.

References

    1. Ahn J., Kim N.J., Choung H.K., Hwang S.W., Sung M., Lee M.J. Frontalis sling operation using silicone rod for the correction of ptosis in chronic progressive external ophthalmoplegia. British Journal of Ophthalmology. 2008;92:1685–1688.
    1. Ahrlich K.G., De Moraes C.G.V., Teng C.C., Prata T.S., Tello C., Ritch R. Visual field progression differences between normal-tension and exfoliative high-tension glaucoma. Investigative Ophthalmology & Visual Science. 2010;51:1458–1463.
    1. Aijaz S., Erskine L., Jeffery G., Bhattacharya S.S., Votruba M. Developmental expression profile of the optic atrophy gene product: OPA1 is not localized exclusively in the mammalian retinal ganglion cell layer. Investigative Ophthalmology & Visual Science. 2004;45:1667–1673.
    1. Akepati V.R. Characterization of OPA1 isoforms isolated from mouse tissues. Journal of Neurochemistry. 2008;106:372–383.
    1. Alavi M.V., Bette S., Schimpf S., Schuettauf F., Schraermeyer U., Wehrl H.F. A splice site mutation in the murine Opa1 gene features pathology of autosomal dominant optic atrophy. Brain. 2007;130:1029–1042.
    1. Alberio S., Mineri R., Tiranti V., Zeviani M. Depletion of mtDNA: syndromes and genes. Mitochondrion. 2007;7:6–12.
    1. Alexander C., Votruba M., Pesch U.E.A., Thiselton D.L., Mayer S., Moore A. OPA1, encoding a dynamin-related GTPase, is mutated in autosomal dominant optic atrophy linked to chromosome 3q28. Nature Genetics. 2000;26:211–215.
    1. Alward W.L., Feldman F., Cashwell L.F., Wilensky J., Geijssen H.C., Greeve E. The effectiveness of intraocular pressure reduction in the treatment of normal-tension glaucoma. American Journal of Ophthalmology. 1998;126:498–505.
    1. Amati-Bonneau P., Guichet A., Olichon A., Chevrollier A., Viala F., Miot S. OPA1 R445H mutation in optic atrophy associated with sensorineural deafness. Annals of Neurology. 2005;58:958–963.
    1. Amati-Bonneau P., Odent S., Derrien C., Pasquier L., Malthiery Y., Reynier P. The association of autosomal dominant optic atrophy and moderate deafness may be due to the R445H mutation in the OPA1 gene. American Journal of Ophthalmology. 2003;136:1170–1171.
    1. Amati-Bonneau P., Valentino M.L., Reynier P., Gallardo M.E., Bornstein B., Boissiere A. OPA1 mutations induce mitochondrial DNA instability and optic atrophy plus phenotypes. Brain. 2008;131:338–351.
    1. Anderson S., Bankier A.T., Barrell B.G., de Bruijn M.H., Coulson A.R., Drouin J. Sequence and organization of the human mitochondrial genome. Nature. 1981;290:457–465.
    1. Andrews R.M., Griffiths P.G., Johnson M.A., Turnbull D.M. Histochemical localisation of mitochondrial enzyme activity in human optic nerve and retina. British Journal of Ophthalmology. 1999;83:231–235.
    1. Andrews R.M., Kubacka I., Chinnery P.F., Lightowlers R.N., Turnbull D.M., Howell N. Reanalysis and revision of the Cambridge reference sequence for human mitochondrial DNA. Nature Genetics. 1999;23:147.
    1. Anikster Y., Kleta R., Shaag A., Gahl W.A., Elpeleg O. Type III 3-methylglutaconic aciduria (optic atrophy plus syndrome, or Costeff optic atrophy syndrome): identification of the OPA3 gene and its founder mutation in Iraqi Jews. American Journal of Human Genetics. 2001;69:1218–1224.
    1. Araie M., Kitazawa M., Koseki N. Intraocular pressure and central visual field of normal tension glaucoma. British Journal of Ophthalmology. 1997;81:852–856.
    1. Arnold A.C. Evolving management of optic neuritis and multiple sclerosis. American Journal of Ophthalmology. 2005;139:1101–1108.
    1. Artal-Sanz M., Tavernarakis N. Prohibitin and mitochondrial biology. Trends in Endocrinology and Metabolism. 2009;20:394–401.
    1. Assink J.J.M., Tijmes N.T., ten Brink J.B., Oostra R.J., Riemslag F.C., deJong P. A gene for X-linked optic atrophy is closely linked to the Xp11.4-Xp11.2 region of the X chromosome. American Journal of Human Genetics. 1997;61:934–939.
    1. Aung T., Ocaka L., Ebenezer N.D., Morris A.G., Krawczak M., Thiselton D.L. A major marker for normal tension glaucoma: association with polymorphisms in the OPA1 gene. Human Genetics. 2002;110:52–56.
    1. Balcer L.J. Optic neuritis. New England Journal of Medicine. 2006;354:1273–1280.
    1. Ban T., Heymann J.A.W., Song Z.Y., Hinshaw J.E., Chan D.C. OPA1 disease alleles causing dominant optic atrophy have defects in cardiolipin-stimulated GTP hydrolysis and membrane tubulation. Human Molecular Genetics. 2010;19:2113–2122.
    1. Baracca A., Solaini G., Sgarbi G., Lenaz G., Baruzzi A., Schapira A.H. Severe impairment of complex I-driven adenosine triphosphate synthesis in leber hereditary optic neuropathy cybrids. Archives of Neurology. 2005;62:730–736. [see comment]
    1. Barbet F., Gerber S., Hakiki S., Perrault I., Hanein S., Ducroq D. A first locus for isolated autosomal recessive optic atrophy (ROA1) maps to chromosome 8q. European Journal of Human Genetics. 2003;11:966–971.
    1. Barbet F., Hakiki S., Orssaud C., Gerber S., Perrault I., Hanein S. A third locus for dominant optic atrophy on chromosome 22q. Journal of Medical Genetics. 2005:42.
    1. Barbiroli B., Montagna P., Cortelli P., Iotti S., Lodi R., Barboni P. Defective brain and muscle energy-metabolism shown by in-vivo P-31 magnetic-resonance spectroscopy in nonaffected carriers of 11778-mtDNA mutation. Neurology. 1995;45:1364–1369.
    1. Barboni P., Carbonelli M., Savini G., Foscarini B., Parisi V., Valentino M.L. OPA1 mutations associated with dominant optic atrophy influence optic nerve head size. Ophthalmology. 2010;117:1547–1553.
    1. Barboni P., Carbonelli M., Savini G., Ramos C.D., Carta A., Berezovsky A. Natural history of Leber’s hereditary optic neuropathy: longitudinal analysis of the retinal nerve fiber layer by optical coherence tomography. Ophthalmology. 2009
    1. Barboni P., Savini G., Valentino M.L., La Morgia C., Bellusci C., De Negri A.M. Leber’s hereditary optic neuropathy with childhood onset. Investigative Ophthalmology & Visual Science. 2006;47:5303–5309.
    1. Barboni P., Savini G., Valentino M.L., Montagna P., Cortelli P., De Negri A.M. Retinal nerve fiber layer evaluation by optical coherence tomography in Leber’s hereditary optic neuropathy. Ophthalmology. 2005;112:120–126.
    1. Barnils N., Mesa E., Munoz S., Ferrer-Artola A., Arruga J. Response to idebenone and multivitamin therapy in Leber’s hereditary optic neuropathy. Archivos de la Sociedad Española de Oftalmología. 2007;82:377–380.
    1. Barron M.J., Griffiths P., Turnbull D.M., Bates D., Nichols P. The distributions of mitochondria and sodium channels reflect the specific energy requirements and conduction properties of the human optic nerve head. British Journal of Ophthalmology. 2004;88:286–290.
    1. Beck R.W., Cleary P.A., Trobe J.D., Kaufman D.I., Kupersmith M.J., Paty D.W. The effect of corticosteroids for acute optic neuritis on the subsequent development of multiple-sclerosis. New England Journal of Medicine. 1993;329:1764–1769.
    1. Beretta S., Mattavelli L., Sala G., Tremolizzo L., Schapira A.H.V., Martinuzzi A. Leber hereditary optic neuropathy mtDNA mutations disrupt glutamate transport in cybrid cell lines. Brain. 2004;127:2183–2192.
    1. Bern C., Philen R.M., Fredman D., Bowman B.A., Newman N.J., Gerr F. Epidemic optic neuropathy in cuba – clinical characterization and risk-factors. New England Journal of Medicine. 1995;333:1176–1182.
    1. Berninger T.A., Jaeger W., Krastel H. Electrophysiology and color perimetry in dominant infantile optic atrophy. British Journal of Ophthalmology. 1991;75:49–52.
    1. Besch D., Leo-Kottler B., Zrenner E., Wissinger B. Leber’s hereditary optic neuropathy: clinical and molecular genetic findings in a patient with a new mutation in the ND6 gene. Graefes Archive for Clinical & Experimental Ophthalmology. 1999;237:745–752.
    1. Bette S., Schlaszus H., Wissinger B., Meyermann R., Mittelbronn M. OPA1, associated with autosomal dominant optic atrophy, is widely expressed in the human brain. Acta Neuropathologica. 2005;109:393–399.
    1. Bhat N.K., Niranjan B.G., Avadhani N.G. The complexity of mitochondrial translation products in mammalian-cells. Biochemical and Biophysical Research Communications. 1981;103:621–628.
    1. Biermann J., Grieshaber P., Goebel U., Martin G., Thanos S., Di Giovanni S. Valproic acid-mediated neuroprotection and regeneration in injured retinal ganglion cells. Investigative Ophthalmology & Visual Science. 2010;51:526–534.
    1. Binder J., Hofmann S., Kreisel S., Wohrle J.C., Bazner H., Krauss J.K. Clinical and molecular findings in a patient with a novel mutation in the deafness-dystonia peptide (DDP1) gene. Brain. 2003;126:1814–1820.
    1. Biousse V., Brown M.D., Newman N.J., Allen J.C., Rosenfeld J., Meola G. De novo 14484 mitochondrial DNA mutation in monozygotic twins discordant for Leber’s hereditary optic neuropathy. Neurology. 1997;49:1136–1138.
    1. Blakely E.L., de Silva R., King A., Schwarzer V., Harrower T., Dawidek G. LHON/MELAS overlap syndrome associated with a mitochondrial MTND1 gene mutation. European Journal of Human Genetics. 2005;13:623–627.
    1. Blesa J.R., Solano A., Briones P., Prieto-Ruiz J.A., Hernandez-Yago J., Coria F. Molecular genetics of a patient with Mohr–Tranebjaerg syndrome due to a new mutation in the DDPI gene. Neuromolecular Medicine. 2007;9:285–291.
    1. Boland M.V., Quigley H.A. Risk factors and open-angle glaucoma: classification and application. Journal of Glaucoma. 2007;16:406–418.
    1. Boldogh I.R., Pon L.A. Mitochondria on the move. Trends in Cell Biology. 2007;17:502–510.
    1. Borchert M., Garcia-Filion P. The syndrome of optic nerve hypoplasia. Current Neurology and Neuroscience Reports. 2008;8:395–403.
    1. Bower S.P., Hawley I., Mackey D.A. Cardiac arrhythmia and Leber’s hereditary optic neuropathy. Lancet. 1992;339:1427–1428. [letter]
    1. Bremner F.D., Tomlin E.A., Shallo-Hoffmann J., Votruba M., Smith S.E. The pupil in dominant optic atrophy. Investigative Ophthalmology & Visual Science. 2001;42:675–678.
    1. Bristow E.A., Griffiths P.G., Andrews R.M., Johnson M.A., Turnbull D.M. The distribution of mitochondrial activity in relation to optic nerve structure. Archives of Ophthalmology. 2002;120:791–796.
    1. Brown D.T., Herbert M., Lamb V.K., Chinnery P.F., Taylor R.W., Lightowlers R.N. Transmission of mitochondrial DNA disorders: possibilities for the future. Lancet. 2006;368:87–89.
    1. Brown M.D., Starikovskaya E., Derbeneva O., Hosseini S., Allen J.C., Mikhailovskaya I.E. The role of mtDNA background in disease expression: a new primary LHON mutation associated with Western Eurasian haplogroup. Journal of Human Genetics. 2002;110:130–138.
    1. Brown M.D., Torroni A., Reckord C.L., Wallace D.C. Phylogenetic analysis of Leber’s hereditary optic neuropathy mitochondrial DNA’s indicates multiple independent occurrences of the common mutations. Human Mutation. 1995;6:311–325.
    1. Brown M.D., Trounce I.A., Jun A.S., Allen J.C., Wallace D.C. Functional analysis of lymphoblast and cybrid mitochondria containing the 3460, 11778, or 14484 Leber’s hereditary optic neuropathy mitochondrial DNA mutation. Journal of Biological Chemistry. 2000;275:39831–39836.
    1. Brown M.D., Zhadanov S., Allen J.C., Hosseini S., Newman N.J., Atamonov V.V. Novel mtDNA mutations and oxidative phosphorylation dysfunction in Russian LHON families. Human Genetics. 2001;109:33–39.
    1. Bu X., Rotter J.I. Leber hereditary optic neuropathy: estimation of number of embryonic precursor cells and disease threshold in heterozygous affected females at the X-linked locus. Clinical Genetics. 1992;42:143–148.
    1. Bu X.D., Rotter J.I. X chromosome-linked and mitochondrial gene control of Leber hereditary optic neuropathy: evidence from segregation analysis for dependence on X chromosome inactivation. Proceedings of the National Academy of Sciences of the United States of America. 1991;88:8198–8202.
    1. Bua E., Johnson J., Herbst A., Delong B., McKenzie D., Salamat S. Mitochondrial DNA-deletion mutations accumulate intracellularly to detrimental levels in aged human skeletal muscle fibers. American Journal of Human Genetics. 2006;79:469–480.
    1. Bunce C., Wormald R. Leading causes of certification for blindness and partial sight in England & Wales. BMC Public Health. 2006;6:58.
    1. Buono L.M., Foroozan R., Sergott R.C., Savino P.J. Is normal tension glaucoma actually an unrecognized hereditary optic neuropathy? New evidence from genetic analysis. Current Opinion in Ophthalmology. 2002;13:362–370.
    1. Burgoyne C.F., Downs J.C., Bellezza A.J., Suh J.K.F., Hart R.T. The optic nerve head as a biomechanical structure: a new paradigm for understanding the role of IOP-related stress and strain in the pathophysiology of glaucomatous optic nerve head damage. Progress in Retinal and Eye Research. 2005;24:39–73.
    1. Campuzano V., Montermini L., Molto M.D., Pianese L., Cossee M., Cavalcanti F. Friedreich’s ataxia: autosomal recessive disease caused by an intronic GAA triplet repeat expansion. Science. 1996;271:1423–1427.
    1. Cann R.L. Genetic clues to dispersal in human populations: retracing the past from the present. Science. 2001;291:1742–1748.
    1. Carelli V., Achilli A., Valentino M.L., Rengo C., Semino O., Pala M. Haplogroup effects and recombination of mitochondrial DNA: novel clues from the analysis of Leber hereditary optic neuropathy pedigrees. American Journal of Human Genetics. 2006;78:564–574.
    1. Carelli V., Bellan M. Myelin, mitochondria, and autoimmunity – what’s the connection? Neurology. 2008;70:1075–1076.
    1. Carelli V., Franceschini F., Venturi S., Barboni P., Savini G., Barbieri G. Grand rounds: could occupational exposure to n-hexane and other solvents precipitate visual failure in Leber hereditary optic neuropathy? Environmental Health Perspectives. 2007;115:113–115.
    1. Carelli V., Ghelli A., Ratta M., Bacchilega E., Sangiorgi S., Mancini R. Leber’s hereditary optic neuropathy: biochemical effect of 11778/ND4 and 3460/ND1 mutations and correlation with the mitochondrial genotype. Neurology. 1997;48:1623–1632.
    1. Carelli V., La Morgia C., Iommarini L., Carroccia R., Mattiazzi M., Sangiorgi S. Mitochondrial optic neuropathies: how two genomes may kill the same cell type? Bioscience Reports. 2007;27:173–184.
    1. Carelli V., La Morgia C., Valentino M.L., Barboni P., Ross-Cisneros F.N., Sadun A.A. Retinal ganglion cell neurodegeneration in mitochondrial inherited disorders. Biochimica et Biophysica Acta-Bioenergetics. 2009;1787:518–528.
    1. Carelli V., Ross-Cisneros F.N., Sadun A.A. Mitochondrial dysfunction as a cause of optic neuropathies. Progress in Retinal and Eye Research. 2004;23:53–89.
    1. Carelli V., Rugolo M., Sgarbi G., Ghelli A., Zanna C., Baracca A. Bioenergetics shapes cellular death pathways in Leber’s hereditary optic neuropathy: a model of mitochondrial neurodegeneration. Biochimica et Biophysica Acta-Bioenergetics. 2004;1658:172–179.
    1. Carelli V., Schimpf S., Valentino M.L., Fuhrmann N., Papke M., Schaich S. Dominant optic atrophy (DOA) and sensorineural hearing loss: clinical, biochemical, spectroscopic and molecular genetic study of a large Italian pedigree linked to a new locus an chromosome 16. Neurology. 2007;68:A42.
    1. Cartoni R., Martinou J.C. Role of mitofusin 2 mutations in the physiopathology of Charcot-Marie-Tooth disease type 2A. Experimental Neurology. 2009;218:268–273.
    1. Casari G., De Fusco M., Ciarmatori S., Zeviani M., Mora M., Fernandez P. Spastic paraplegia and OXPHOS impairment caused by mutations in paraplegin, a nuclear-encoded mitochondrial metalloprotease. Cell. 1998;93:973–983.
    1. Chalasani M.L., Balasubranianian D., Swarup G. Focus on molecules: optineurin. Experimental Eye Research. 2008;87:1–2.
    1. Chalasani M.L., Radha V., Gupta V., Agarwal N., Balasubramanian D., Swarup G. A glaucoma-associated mutant of optineurin selectively induces death of retinal ganglion cells which is inhibited by antioxidants. Investigative Ophthalmology & Visual Science. 2007;48:1607–1614.
    1. Chalmers R.M., Harding A.E. A case-control study of Leber’s hereditary optic neuropathy. Brain. 1996;119:1481–1486.
    1. Chalmers R.M., Robertson N., Kellarwood H., Compston D.A.S., Harding A.E. Sequence of the human homolog of a mitochondrially encoded murine transplantation antigen in patients with multiple-sclerosis. Journal of Neurology. 1995;242:332–334.
    1. Chen H., Chan D.C. Mitochondrial dynamics-fusion, fission, movement, and mitophagy-in neurodegenerative diseases. Human Molecular Genetics. 2009;18:R169–R176.
    1. Chen S.F., Chang Y., Wu J.C. The spatial distribution of macular pigment in humans. Current Eye Research. 2001;23:422–434.
    1. Cheng J.W., Cai J.P., Wei R.L. Meta-analysis of medical intervention for normal tension glaucoma. Ophthalmology. 2009;116:1243–1249.
    1. Chevrollier A., Guillet V., Loiseau D., Gueguen N., de Crescenzo M.A., Verny C. Hereditary optic neuropathies share a common mitochondrial coupling defect. Annals of Neurology. 2008;63:794–798.
    1. Chinnery P.F. Modulating heteroplasmy. Trends in Genetics. 2002;18:173–176.
    1. Chinnery P.F., Andrews R.M., Turnbull D.M., Howell N. Leber hereditary optic neuropathy: does heteroplasmy influence the inheritance and expression of the G11778A mitochondrial DNA mutation? American Journal of Medical Genetics. 2001;98:235–243.
    1. Chinnery P.F., Brown D.T., Andrews R.M., Singh-Kler R., Riordan-Eva P., Lindley J. The mitochondrial ND6 gene is a hot spot for mutations that cause Leber’s hereditary optic neuropathy. Brain. 2001;124:209–218.
    1. Chinnery P.F., Majamaa K., Turnbull D., Thorburn D. Treatment for mitochondrial disorders. Cochrane Database of Systematic Reviews. 2006
    1. Chinnery P.F., Thorburn D.R., Samuels D.C., White S.L., Dahl H.H.M., Turnbull D.M. The inheritance of mitochondrial DNA heteroplasmy: random drift, selection or both? Trends in Genetics. 2000;16:500–505.
    1. Chinnery P.F., Zeviani M. 155th ENMC workshop: polymerase gamma and disorders of mitochondrial DNA synthesis, 21–23 September 2007, Naarden, The Netherlands. Neuromuscul Disord. 2008;18:259–267.
    1. Chinnery P.F., Zwijnenburg P.J.G., Walker M., Howell N., Taylor R.W., Lightowlers R.N. Nonrandom tissue distribution of mutant mtDNA. American Journal of Medical Genetics. 1999;85:498–501.
    1. Chung H., Yoon Y.H., Hwang J.J., Cho K.S., Koh J.Y., Kim J.G. Ethambutol-induced toxicity is mediated by zinc and lysosomal membrane permeabilization in cultured retinal cells. Toxicology and Applied Pharmacology. 2009;235:163–170.
    1. Ciccosanti F., Corazzari M., Soldani F., Matarrese P., Pagliarini V., Iadevaia V. Proteomic analysis identifies prohibitin down-regulation as a crucial event in the mitochondrial damage observed in HIV-infected patients. Antiviral Therapy. 2010;15:377–390.
    1. Cipolat S., Rudka T., Hartmann D., Costa V., Serneels L., Craessaerts K. Mitochondrial rhomboid PARL regulates cytochrome c release during apoptosis via OPA1-dependent cristae remodeling. Cell. 2006;126:163–175.
    1. Cock H.R., Cooper J.M., Schapira A.H. The 14484 ND6 mtDNA mutation in Leber hereditary optic neuropathy does not affect fibroblast complex I activity. American Journal of Human Genetics. 1995;57:1501–1502. [letter]
    1. Cock H.R., Cooper J.M., Schapira A.H. Functional consequences of the 3460-bp mitochondrial DNA mutation associated with Leber’s hereditary optic neuropathy. Journal of the Neurological Sciences. 1999;165:10–17.
    1. Cock H.R., Tabrizi S.J., Cooper J.M., Schapira A.H. The influence of nuclear background on the biochemical expression of 3460 Leber’s hereditary optic neuropathy. Annals of Neurology. 1998;44:187–193.
    1. Cogan D.G., Truman J.T., Smith T.R. Optic neuropathy, chloramphenicol, and infantile genetic agranulocytosis. Investigative Ophthalmology. 1973;12:534–537.
    1. Cohn A.C., Toomes C., Hewitt A.W., Kearns L.S., Inglehearn C.F., Craig J.E. The natural history of OPA1-related autosomal dominant optic atrophy. British Journal of Ophthalmology. 2008;24:24.
    1. Cohn A.C., Toomes C., Potter C., Towns K.V., Hewitt A.W., Inglehearn C.F. Autosomal dominant optic atrophy: penetrance and expressivity in patients with OPA1 mutations. American Journal of Ophthalmology. 2007;143:656–662.
    1. Cordeiro M.F., Guo L., Luong V., Harding G., Wang W., Jones H.E. Real-time imaging of single nerve cell apoptosis in retinal neurodegeneration. Proceedings of the National Academy of Sciences of the United States of America. 2004;101:13352–13356.
    1. Cornille K., Milea D., Amati-Bonneau P., Procaccio V., Zazoun L., Guillet V. Reversible optic neuropathy with OPA1 exon 5b mutation. Annals of Neurology. 2008;63:667–671.
    1. Corral-debrinski M., Horton T., Lott M.T., Shoffner J.M., Beal M.F., Wallace D.C. Mitochondrial-DNA deletions in human brain – regional variability and increase with advanced age. Nature Genetics. 1992;2:324–329.
    1. Cortelli P., Montagna P., Pierangeli G., Lodi R., Barboni P., Liguori R. Clinical and brain bioenergetics improvement with idebenone in a patient with Leber’s hereditary optic neuropathy: a clinical and 31P-MRS study. Journal of the Neurological Sciences. 1997;148:25–31.
    1. Costeff H., Gadoth N., Apter N., Prialnic M., Savir H. A familial syndrome of infantile optic atrophy, movement disorder, and spastic paraplegia. Neurology. 1989;39:595–597.
    1. Cote H.C.F., Brumme Z.L., Craib K.J.P., Alexander C.S., Wynhoven B., Ting L.L. Changes in mitochondrial DNA as a marker of nucleoside toxicity in HIV-infected patients. New England Journal of Medicine. 2002;346:811–820.
    1. Craig J.E., Hewitt A.W., Dimasi D.P., Howell N., Toomes C., Cohn A.C. The role of the Met98Lys optineurin variant in inherited optic nerve diseases. British Journal of Ophthalmology. 2006;90:1420–1424.
    1. Craven L., Tuppen H.A., Greggains G.D., Harbottle S.J., Murphy J.L., Cree L.M. Pronuclear transfer in human embryos to prevent transmission of mitochondrial DNA disease. Nature. 2010;465:82–U89.
    1. Cree L.M., Samuels D.C., Chinnery P.F. The inheritance of pathogenic mitochondrial DNA mutations. Biochimica et Biophysica Acta-Molecular Basis of Disease. 2009;1792:1097–1102.
    1. Cullom M.E., Heher K.L., Miller N.R., Savino P.J., Johns D.R. Lebers hereditary optic neuropathy masquerading as tobacco-alcohol amblyopia. Archives of Ophthalmology. 1993;111:1482–1485.
    1. Das M., Chiron S., Verde F. Microtubule-dependent spatial organization of mitochondria in fission yeast. Methods Cell Biol. 2010;97:203–221.
    1. Daut P.M., Steinemann T.L., Westfall C.T. Chronic exposure keratopathy complicating surgical correction of ptosis in patients with chronic progressive external ophthalmoplegia. American Journal of Ophthalmology. 2000;130:519–521.
    1. Davey K.M., Parboosingh J.S., McLeod D.R., Chan A., Casey R., Ferreira P. Mutation of DNAJC19, a human homologue of yeast inner mitochondrial membrane co-chaperones, causes DCMA syndrome, a novel autosomal recessive Barth syndrome-like condition. Journal of Medical Genetics. 2006;43:385–393.
    1. Davies V., Votruba M. Focus on molecules: the OPA1 protein. Experimental Eye Research. 2006;83:1003–1004.
    1. Davies V.J., Hollins A.J., Piechota M.J., Yip W., Davies J.R., White K.E. Opa1 deficiency in a mouse model of autosomal dominant optic atrophy impairs mitochondrial morphology, optic nerve structure and visual function. Human Molecular Genetics. 2007;16:1307–1318.
    1. Davies V.J., Powell K.A., White K.E., Yip W., Hogan V., Hollins A.J. A missense mutation in the murine Opa3 gene models human Costeff syndrome. Brain. 2008;131:368–380.
    1. De Marco N., Buono M., Troise F., Diez-Roux G. Optineurin increases cell survival and translocates to the nucleus in a Rab8-dependent manner upon an apoptotic stimulus. Journal of Biological Chemistry. 2006;281:16147–16156.
    1. De Marinis M. Optic neuropathy after treatment with antituberculous drugs in a subject with Leber’s hereditary optic neuropathy mutation. Journal of Neurology. 2001;248:818–819.
    1. De Vries D.D., Went L.N., Bruyn G.W., Scholte H.R., Hofstra R.M., Bolhuis P.A. Genetic and biochemical impairment of mitochondrial complex I activity in a family with Leber hereditary optic neuropathy and hereditary spastic dystonia. American Journal of Human Genetics. 1996;58:703–711.
    1. De Vriese A.S., Van Coster R., Smet J., Seneca S., Lovering A., Van Haute L.L. Linezolid-induced inhibition of mitochondrial protein synthesis. Clinical Infectious Diseases. 2006;42:1111–1117.
    1. Decanini-Mancera A., Harrison A.R., Lee M.S. Another case of Leber hereditary optic neuropathy in an octogenarian. Journal of Neuro-Ophthalmology. 2009;29:159–160.
    1. Degli Esposti M., Carelli V., Ghelli A., Ratta M., Crimi M., Sangiorgi S. Functional alterations of the mitochondrially encoded ND4 subunit associated with Leber’s hereditary optic neuropathy. FEBS Letters. 1994;352:375–379.
    1. Del Bo R., Bordoni A., Sciacco M., Di Fonzo A., Galbiati S., Crimi M. Remarkable infidelity of polymerase gamma A associated with mutations in POLG1 exonuclease domain. Neurology. 2003;61:903–908.
    1. del Toro D., Alberch J., Lazaro-Dieguez F., Martin-Ibanez R., Xifro X., Egea G. Mutant huntingtin impairs Post-golgi trafficking to lysosomes by delocalizing optineurin/Rab8 complex from the golgi apparatus. Molecular Biology of the Cell. 2009;20:1478–1492.
    1. Delettre C., Lenaers G., Griffoin J.M., Gigarel N., Lorenzo C., Belenguer P. Nuclear gene OPA1, encoding a mitochondrial dynamin-related protein, is mutated in dominant optic atrophy. Nature Genetics. 2000;26:207–210.
    1. Di Prospero N.A., Baker A., Jeffries N., Fischbeck K.H. Neurological effects of high-dose idebenone in patients with Friedreich’s ataxia: a randomised, placebo-controlled trial. Lancet Neurology. 2007;6:878–886.
    1. DiMauro S., Mancuso M. Mitochondrial diseases: therapeutic approaches. Bioscience Reports. 2007;27:125–137.
    1. DiMauro S., Rustin P. A critical approach to the therapy of mitochondrial respiratory chain and oxidative phosphorylation diseases. Biochimica et Biophysica Acta-Molecular Basis of Disease. 2009;1792:1159–1167.
    1. DiMauro S., Schon E.A. Mechanisms of disease: mitochondrial respiratory-chain diseases. New England Journal of Medicine. 2003;348:2656–2668.
    1. Dotti M.T., Plewnia K., Cardaioli E., Manneschi L., Rufa A., Alema G. A case of ethambutol-induced optic neuropathy harbouring the primary mitochondrial LHON mutation at nt 11778. Journal of Neurology. 1998;245:302–303.
    1. Dudkina N.V., Eubel H., Keegstra W., Boekema E.J., Braun H.P. Structure of a mitochondrial supercomplex formed by respiratory-chain complexes I and III. Proceedings of the National Academy of Sciences of the United States of America. 2005;102:3225–3229.
    1. Dudkina N.V., Kouril R., Peters K., Braun H.P., Boekema E.J. Structure and function of mitochondrial supercomplexes. Biochimica et Biophysica Acta-Bioenergetics. 2010;1797:664–670.
    1. Durham S.E., Samuels D.C., Cree L.M., Chinnery P.F. Normal levels of wild-type mitochondrial DNA maintain cytochrome c oxidase activity for two pathogenic mitochondrial DNA mutations but not for m.3243A->G. American Journal of Human Genetics. 2007;81:189–195.
    1. Durr A., Cossee M., Agid Y., Campuzano V., Mignard C., Penet C. Clinical and genetic abnormalities in patients with Friedreich’s ataxia. New England Journal of Medicine. 1996;335:1169–1175.
    1. Echaniz-Laguna A., Chassagne M., de Seze J., Mohr M., Clerc-Renaud P., Tranchant C. POLG1 variations presenting as multiple sclerosis. Archives of Neurology. 2010;67:1140–1143.
    1. Eliott D., Traboulsi E.I., Maumenee I.H. Visual prognosis in autosomal dominant optic atrophy (Kjer Type) American Journal of Ophthalmology. 1993;115:360–367.
    1. Elliott H.R., Samuels D.C., Eden J.A., Relton C.L., Chinnery P.F. Pathogenic mitochondrial DNA mutations are common in the general population. American Journal of Human Genetics. 2008;83:254–260.
    1. Ellouze S., Augustin S., Bouaita A., Bonnet C., Simonutti M., Forster V. Optimized allotopic expression of the human mitochondrial ND4 prevents blindness in a rat model of mitochondrial dysfunction. American Journal of Human Genetics. 2008;83:373–387.
    1. Fauser S., Leo-Kottler B., Besch D., Luberichs J. Confirmation of the 14568 mutation in the mitochondrial ND6 gene as causative in Leber’s hereditary optic neuropathy. Ophthalmic Genetics. 2002;23:191–197.
    1. Fauser S., Luberichs J., Besch D., Leo-Kottler B. Sequence analysis of the complete mitochondrial genome in patients with Leber’s hereditary optic neuropathy lacking the three most common pathogenic DNA mutations. Biochemical and Biophysical Research Communications. 2002;295:342–347.
    1. Ferraris S., Clark S., Garelli E., Davidzon G., Moore S.A., Kardon R.H. Progressive external ophthalmoplegia and vision and hearing loss in a patient with mutations in POLG2 and OPA1. Archives of Neurology. 2008;65:125–131.
    1. Ferre M., Amati-Bonneau P., Tourmen Y., Malthiery Y., Reynier P. eOPA1: An online database for OPA1 mutations. Human Mutation. 2005;25:423–428.
    1. Ferre M., Bonneau D., Milea D., Chevrollier A., Verny C., Dollfus H. Molecular screening of 980 cases of suspected hereditary optic neuropathy with a report on 77 novel OPA1 mutations. Human Mutation. 2009;30:E692–E705.
    1. Figueroa-Martinez, F., Vazquez-Acevedo, M., Cortes-Hernandez, P., Garcia-Trejo, J.J., Davidson, E., King, M.P., et al., 2010. What limits the allotopic expression of nucleus-encoded mitochondrial genes? The case of the chimeric Cox3 and Atp6 genes. Mitochondrion, Epub ahead of print.
    1. Floreani M., Napoli E., Martinuzzi A., Pantano G., De Riva V., Trevisan R. Antioxidant defences in cybrids harboring mtDNA mutations associated with Leber’s hereditary optic neuropathy. FEBS Journal. 2005;272:1124–1135.
    1. Folliot S., Briot D., Conrath H., Provost N., Cherel Y., Moullier P. Sustained tetracycline-regulated transgene expression in vivo in rat retinal ganglion cells using a single type 2 adeno-associated viral vector. Journal of Gene Medicine. 2003;5:493–501.
    1. Fortuna F., Barboni P., Liguori R., Valentino M.L., Savini G., Gellera C. Visual system involvement in patients with Friedreichs ataxia. Brain. 2009;132:116–123.
    1. Fournier A.V., Damji K.F., Epstein D.L., Pollock S.C. Disc excavation in dominant optic atrophy. Ophthalmology. 2001;108:1595–1602.
    1. Fox C.M., Bensa S., Bray I., Zajicek J.P. The epidemiology of multiple sclerosis in Devon: a comparison of the new and old classification criteria. Journal of Neurology, Neurosurgery, and Psychiatry. 2004;75:56–60.
    1. Fraser J.A., Biousse V., Newman N.J. The neuro-ophthalmology of mitochondrial disease. Survey of Ophthalmology. 2010;55:299–334.
    1. Frerichs F.C.P., Dingemans K.P., Brinkman K. Cardiomyopathy with mitochondrial damage associated with nucleoside reverse-transcriptase inhibitors. New England Journal of Medicine. 2002;347:1895–1896.
    1. Frey T.G., Mannella C.A. The internal structure of mitochondria. Trends in Biochemical Sciences. 2000;25:319–324.
    1. Frezza C., Cipolat S., de Brito O.M., Micaroni M., Beznoussenko G.V., Rudka T. OPA1 controls apoptotic cristae remodeling independently from mitochondrial fusion. Cell. 2006;126:177–189.
    1. Friedmann T. Medical ethics – principles for human gene therapy studies. Science. 2000;287:2163–2165.
    1. Frohman E.M., Frohman T.C., Zee D.S., McColl R., Galetta S. The neuro-ophthalmology of multiple sclerosis. Lancet Neurology. 2005;4:111–121.
    1. Frohman E.M., Fujimoto J.G., Frohman T.C., Calabresi P.A., Cutter G., Balcer L.J. Optical coherence tomography: a window into the mechanisms of multiple sclerosis. Nature Clinical Practice Neurology. 2008;4:664–675.
    1. Fuhrmann N., Alavi M.V., Bitoun P., Woernle S., Auburger G., Leo-Kottler B. Genomic rearrangements in OPA1 are frequent in patients with autosomal dominant optic atrophy. Journal of Medical Genetics. 2009;46:136–144.
    1. Gabaldon T., Huynen M.A. Shaping the mitochondrial proteome. Biochimica et Biophysica Acta-Bioenergetics. 2004;1659:212–220.
    1. Garcin R., Raverdy P., Delthil S., Man H.X., Chimenes H. On a familial hereditary disorder comprising cataracts, optic atrophy, extra-pyramidal signs, and certain stigmata of Friedreich’s ataxia. Revista de Neurologia [Paris] 1961;104:373–379.
    1. Ghelli A., Porcelli A.M., Zanna C., Martinuzzi A., Carelli V., Rugolo M. Protection against oxidant-induced apoptosis by exogenous glutathione in Leber hereditary optic neuropathy cybrids. Investigative Ophthalmology & Visual Science. 2008;49:671–676.
    1. Ghelli A., Porcelli A.M., Zanna C., Vidoni S., Mattioli S., Barbieri A. The background of mitochondrial DNA haplogroup J increases the sensitivity of Leber’s hereditary optic neuropathy cells to 2,5-hexanedione toxicity. PLoS One. 2009:4.
    1. Ghelli A., Zanna C., Porcelli A.M., Schapira A.H.V., Martinuzzi A., Carelli V. Leber’s hereditary optic neuropathy (LHON) pathogenic mutations induce mitochondrial-dependent apoptotic death in transmitochondrial cells incubated with galactose medium. Journal of Biological Chemistry. 2003;278:4145–4150.
    1. Giordano, C., Montopoli, M., Perli, E., Orlandi, M., Fantin, M., Ross-Cisneros, F.N., et al., 2010. Oestrogens ameliorate mitochondrial dysfunction in Leber’s hereditary optic neuropathy. Brain, Epub ahead of print.
    1. Giraudet S., Lamirel C., Amati-Bonneau P., Reynier P., Bonneau D., Milea D. Never too old to harbour a young man’s disease? British Journal of Ophthalmology. 2010;2010:31.
    1. Godel V., Nemet P., Lazar M. Chloramphenicol optic neuropathy. Archives of Ophthalmology. 1980;98:1417–1421.
    1. Gray D.C., Wolfe R., Gee B.P., Scoles D., Geng Y., Masella B.D. In vivo imaging of the fine structure of rhodamine-labeled macaque retinal ganglion cells. Investigative Ophthalmology & Visual Science. 2008;49:467–473.
    1. Gray M.W., Burger G., Lang B.F. Mitochondrial evolution. Science. 1999;283:1476–1481.
    1. Gronlund M.A., Honarvar A.K.S., Andersson S., Moslemi A.R., Oldfors A., Holme E. Ophthalmological findings in children and young adults with genetically verified mitochondrial disease. British Journal of Ophthalmology. 2010;94:121–127.
    1. Gropman A., Chen T.J., Perng C.L., Krasnewich D., Chernoff E., Tifft C. Variable clinical manifestation of homoplasmic G14459A mitochondrial DNA mutation. American Journal of Medical Genetics: Part A. 2004;124:377–382.
    1. Grzybowski A. Tobacco amblyopia: does it really exist? Eye. 2007;21:1448–1449.
    1. Grzybowski, A., Holder, G.E., 2010. Tobacco optic neuropathy (TON) – the historical and present concept of the disease. Acta Ophthalmologica, Epub ahead of print.
    1. Guillet V., Chevrollier A., Cassereau J., Letournel F., Gueguen N., Richard L. Ethambutol-induced optic neuropathy linked to OPA1 mutation and mitochondrial toxicity. Mitochondrion. 2010;10:115–124.
    1. Guo L., Cordeiro M.F. Assessment of neuroprotection in the retina with DARC. Glaucoma: an Open Window to Neurodegeneration and Neuroprotection. 2008;173:437–450.
    1. Gvozdjak J., Gvozdjakova A., Kucharska J., Bada V. The effect of smoking on myocardial metabolism. Czechoslovak Medicine. 1987;10:47–53.
    1. Hanein S., Perrault I., Roche O., Gerber S., Khadom N., Rio M. TMEM126A, encoding a mitochondrial protein, is mutated in autosomal-recessive nonsyndromic optic atrophy. American Journal of Human Genetics. 2009;84:493–498.
    1. Harding A.E. Classification of the hereditary ataxias and paraplegias. Lancet. 1983;1:1151–1155.
    1. Harding A.E., Riordan-Eva P., Govan G.G. Mitochondrial DNA diseases: genotype and phenotype in Leber’s hereditary optic neuropathy. Muscle & Nerve. 1995;3:S82–S84.
    1. Harding A.E., Sweeney M.G., Govan G.G., Riordan-Eva P. Pedigree analysis in Leber hereditary optic neuropathy families with a pathogenic mtDNA mutation. American Journal of Human Genetics. 1995;57:77–86.
    1. Harding A.E., Sweeney M.G., Miller D.H., Mumford C.J., Kellar-Wood H., Menard D. Occurrence of a multiple sclerosis-like illness in women who have a Leber’s hereditary optic neuropathy mitochondrial DNA mutation. Brain. 1992;115:979–989.
    1. Harris M.O., Walsh L.E., Hattab E.M., Golomb M.R. Is it ADEM, POLG, or both? Archives of Neurology. 2010;67:493–496.
    1. Hauser M.A., Sena D.F., Flor J., Walter J., Auguste J., LaRocque-Abramson K. Distribution of optineurin sequence variations in an ethnically diverse population of low-tension glaucoma patients from the United States. Journal of Glaucoma. 2006;15:358–363.
    1. Hayreh S.S. Ischemic optic neuropathy. Progress in Retinal and Eye Research. 2009;28:34–62.
    1. Hayreh S.S. Pathogenesis of nonarteritic anterior ischemic optic neuropathy. Archives of Ophthalmology. 2009;127:1082–1083.
    1. He L.P., Chinnery P.F., Durham S.E., Blakely E.L., Wardell T.M., Borthwick G.M. Detection and quantification of mitochondrial DNA deletions in individual cells by real-time PCR. Nucleic Acids Research. 2002;30
    1. Heiduschka P., Schnichels S., Fuhrmann N., Hofmeister S., Schraermeyer U., Wissinger B. Electrophysiological and histologic assessment of retinal ganglion cell fate in a mouse model for OPA1-associated autosomal dominant optic atrophy. Investigative Ophthalmology & Visual Science. 2010;51:1424–1431.
    1. Heng J.E., Vorwerk C.K., Lessell E., Zurakowski D., Levin L.A., Dreyer E.B. Ethambutol is toxic to retinal ganglion cells via an excitotoxic pathway. Investigative Ophthalmology & Visual Science. 1999;40:190–196.
    1. Herrnstadt C., Elson J.L., Fahy E., Preston G., Turnbull D.M., Anderson C. Reduced-median-network analysis of complete mitochondrial DNA coding-region sequences for the major African, Asian, and European haplogroups. American Journal of Human Genetics. 2002;70:1152–1171.
    1. Ho G., Walter J.H., Christodoulou J. Costeff optic atrophy syndrome: new clinical case and novel molecular findings. Journal of inherited metabolic disease. 2008;7:7.
    1. Hofhaus G., Johns D.R., Hurko O., Attardi G., Chomyn A. Respiration and growth defects in transmitochondrial cell lines carrying the 11778 mutation associated with Leber’s hereditary optic neuropathy. Journal of Biological Chemistry. 1996;271:13155–13161.
    1. Horvath J., Horvath R., Karcagi V., Komoly S., Johns D.R. Sequence analysis of Hungarian LHON patients not carrying the common primary mutations. Journal of Inherited Metabolic Disease. 2002;25:323–324.
    1. Horvath R., Gorman G., Chinnery P.F. How can we treat mitochondrial encephalomyopathies? Approaches to therapy. Neurotherapeutics. 2008;5:558–568.
    1. Horvath R., Hudson G., Ferrari G., Futterer N., Ahola S., Lamantea E. Phenotypic spectrum associated with mutations of the mitochondrial polymerase gamma gene. Brain. 2006;129:1674–1684.
    1. Howell N., Bindoff L.A., McCullough D.A., Kubacka I., Poulton J., Mackey D. Leber hereditary optic neuropathy: identification of the same mitochondrial ND1 mutation in six pedigrees. American Journal of Human Genetics. 1991;49:939–950.
    1. Howell N., Halvorson S., Burns J., McCullough D.A., Paulton J. When does bilateral optic atrophy become Leber hereditary optic neuropathy? American Journal of Human Genetics. 1993;53:959–963.
    1. Howell N., Kubacka I., Xu M., McCullough D.A. Leber hereditary optic neuropathy: involvement of the mitochondrial ND1 gene and evidence for an intragenic suppressor mutation. American Journal of Human Genetics. 1991;48:935–942.
    1. Howell N., Kubacka I., Mackey D.A. How rapidly does the human mitochondrial genome evolve? American Journal of Human Genetics. 1996;59:501–509.
    1. Howell N., Mackey D.A. Low-penetrance branches in matrilineal pedigrees with leber hereditary optic neuropathy. American Journal of Human Genetics. 1998;63:1220–1224.
    1. Howell N., Oostra R.J., Bolhuis P.A., Spruijt L., Clarke L.A., Mackey D.A. Sequence analysis of the mitochondrial genomes from Dutch pedigrees with Leber hereditary optic neuropathy. American Journal of Human Genetics. 2003;72:1460–1469.
    1. Hoyt C.S. Autosomal dominant optic atrophy – a spectrum of disability. Ophthalmology. 1980;87:245–251.
    1. Huang T.S., Santarelli R., Starr A. Mutation of OPA1 gene causes deafness by affecting function of auditory nerve terminals. Brain Research. 2009;1300:97–104.
    1. Hudson G., Amati-Bonneau P., Blakely E.L., Stewart J.D., He L.P., Schaefer A.M. Mutation of OPA1 causes dominant optic atrophy with external ophthalmoplegia, ataxia, deafness and multiple mitochondrial DNA deletions: a novel disorder of mtDNA maintenance. Brain. 2008;131:329–337.
    1. Hudson G., Carelli V., Horvath R., Zeviani M., Smeets H.J., Chinnery P.F. X-Inactivation patterns in females harboring mtDNA mutations that cause Leber hereditary optic neuropathy. Molecular Vision. 2007;13:2339–2343.
    1. Hudson G., Carelli V., Spruijt L., Gerards M., Mowbray C., Achilli A. Clinical expression of Leber hereditary optic neuropathy is affected by the mitochondrial DNA-haplogroup background. American Journal of Human Genetics. 2007;81:228–233.
    1. Hudson G., Keers S., Man P.Y.W., Griffiths P., Huoponen K., Savontaus M.L. Identification of an X-chromosomal locus and haplotype modulating the phenotype of a mitochondrial DNA disorder. American Journal of Human Genetics. 2005;77:1086–1091.
    1. Huizing M., Dorward H., Ly L., Klootwijk E., Kleta R., Skovby F. OPA3, mutated in 3-methylglutaconic aciduria type III, encodes two transcripts targeted primarily to mitochondria. Molecular Genetics and Metabolism. 2010;100:149–154.
    1. Huoponen K., Lamminen T., Juvonen V., Aula P., Nikoskelainen E., Savontaus M.L. The spectrum of mitochondrial DNA mutations in families with Leber hereditary optic neuroretinopathy. Human Genetics. 1993;92:379–384.
    1. Huoponen K., Vilkki J., Aula P., Nikoskelainen E.K., Savontaus M.L. A new mtDNA mutation associated with Leber hereditary optic neuroretinopathy. American Journal of Human Genetics. 1991;48:1147–1153.
    1. Hwang J.M., Kim J., Park S.S. Leber’s hereditary optic neuropathy mutations in ethambutol-induced optic neuropathy. Journal of Neurology. 2003;250:87–89.
    1. Ikeda A., Ikeda T., Ikeda N., Kawakami Y., Mimura O. Leber’s hereditary optic neuropathy precipitated by ethambutol. Japanese Journal of Ophthalmology. 2006;50:280–283.
    1. Inglese M., Rovaris M., Bianchi S., La Mantia L., Mancardi G.L., Ghezzi A. Magnetic resonance imaging, magnetisation transfer imaging, and diffusion weighted imaging correlates of optic nerve, brain, and cervical cord damage in Leber’s hereditary optic neuropathy. Journal of Neurology, Neurosurgery, and Psychiatry. 2001;70:444–449.
    1. Ito Y., Nakamura M., Yamakoshi T., Lin J., Yatsuya H., Terasaki H. Reduction of inner retinal thickness in patients with autosomal dominant optic atrophy associated with OPA1 mutations. Investigative Ophthalmology & Visual Science. 2007;48:4079–4086.
    1. Jansen P.H., van der Knaap M.S., de Coo I.F. Leber’s hereditary optic neuropathy with the 11 778 mtDNA mutation and white matter disease resembling multiple sclerosis: clinical, MRI and MRS findings. Journal of the Neurological Sciences. 1996;135:176–180.
    1. Jaros E., Mahad D.J., Hudson G., Birchall D., Sawcer S.J., Griffiths P.G. Primary spinal cord neurodegeneration in leber hereditary optic neuropathy. Neurology. 2007;69:214–216.
    1. Javaheri M., Khurana R.N., O’Hearn T.M., Lai M.M., Sadun A.A. Linezolid-induced optic neuropathy: a mitochondrial disorder? British Journal of Ophthalmology. 2007;91:111–115.
    1. Jazin E., Soodyall H., Jalonen P., Lindholm E., Stoneking M., Gyllensten U. Mitochondrial mutation rate revisited: hot spots and polymorphism. Nature Genetics. 1998;18:109–110.
    1. Ji Y.L., Jia X.Y., Li S.Q., Xiao X.S., Guo X.M., Zhang Q.J. Evaluation of the X-linked modifier loci for Leber hereditary optic neuropathy with the G11778A mutation in Chinese. Molecular Vision. 2010;16:416–424.
    1. Ji Y.L., Zhang A.M., Jia X.Y., Zhang Y.P., Xiao X.S., Li S.Q. Mitochondrial DNA haplogroups M7b1’2 and M8a affect clinical expression of Leber hereditary optic neuropathy in chinese families with the m.11778G→A mutation. American Journal of Human Genetics. 2008;83:760–768.
    1. Johns D.R., Heher K.L., Miller N.R., Smith K.H. Leber’s hereditary optic neuropathy. Clinical manifestations of the 14484 mutation. Archives of Ophthalmology. 1993;111:495–498.
    1. Johns D.R., Neufeld M.J. Cytochrome c oxidase mutations in Leber hereditary optic neuropathy. Biochemical and Biophysical Research Communications. 1993;196:810–815.
    1. Johns D.R., Neufeld M.J., Park R.D. An ND-6 mitochondrial DNA mutation associated with Leber hereditary optic neuropathy. Biochemical and Biophysical Research Communications. 1992;187:1551–1557.
    1. Johns D.R., Smith K.H., Miller N.R., Sulewski M.E., Bias W.B. Identical twins who are discordant for Leber’s hereditary optic neuropathy. Archives of Ophthalmology. 1993;111:1491–1494.
    1. Ju W.K., Kim K.Y., Angert M., Duong-Polk K.X., Lindsey J.D., Ellisman M.H. Memantine blocks mitochondrial OPA1 and cytochrome c release and subsequent apoptotic cell death in glaucomatous retina. Investigative Ophthalmology & Visual Science. 2009;50:707–716.
    1. Ju W.K., Kim K.Y., Duong-Polk K.X., Lindsey J.D., Ellisman M.H., Weinreb R.N. Increased optic atrophy type 1 expression protects retinal ganglion cells in a mouse model of glaucoma. Molecular Vision. 2010;16:1331–1342.
    1. Ju W.K., Kim K.Y., Lindsey J.D., Angert M., Duong-Polk K.X., Scott R.T. Intraocular pressure elevation induces mitochondrial fission and triggers OPA1 release in glaucomatous optic nerve. Investigative Ophthalmology & Visual Science. 2008;49:4903–4911.
    1. Ju W.K., Kim K.Y., Lindsey J.D., Angert M., Patel A., Scott R.T. Elevated hydrostatic pressure triggers release of OPA1 and cytochrome C, and induces apoptotic cell death in differentiated RGC-5 cells. Molecular Vision. 2009;15:120–134.
    1. Ju W.K., Lindsey J.D., Angert M., Patel A., Weinreb R.N. Glutamate receptor activation triggers OPA1 release and induces apoptotic cell death in ischemic rat retina. Molecular Vision. 2008;14:2629–2638.
    1. Jun A.S., Brown M.D., Wallace D.C. A mitochondrial DNA mutation at nucleotide pair 14459 of the NADH dehydrogenase subunit 6 gene associated with maternally inherited Leber hereditary optic neuropathy and dystonia. Proceedings of the National Academy of Sciences of the United States of America. 1994;91:6206–6210.
    1. Kamei S., Chen-Kuo-Chang M., Cazevieille C., Lenaers G., Olichon A., Belenguer P. Expression of the Opa1 mitochondrial protein in retinal ganglion cells: its downregulation causes aggregation of the mitochondrial network. Investigative Ophthalmology & Visual Science. 2005;46:4288–4294.
    1. Katz B.J., Zhao Y., Warner J.E.A., Tong Z.Z., Yang Z.L., Zhang K. A family with X-linked optic atrophy linked to the OPA2 locus Xp11.4-Xp11.2. American Journal of Medical Genetics: Part A. 2006;140A:2207–2211.
    1. Kaukonen J., Juselius J.K., Tiranti V., Kyttala A., Zeviani M., Comi G.P. Role of adenine nucleotide translocator 1 in mtDNA maintenance. Science. 2000;289:782–785.
    1. Kawasaki A., Herbst K., Sander B., Milea D. Selective wavelength pupillometry in Leber hereditary optic neuropathy. Clinical and Experimental Ophthalmology. 2010;38:322–324.
    1. Kellar-Wood H., Robertson N., Govan G.G., Compston D.A., Harding A.E. Leber’s hereditary optic neuropathy mitochondrial DNA mutations in multiple sclerosis. Annals of Neurology. 1994;36:109–112.
    1. Kerrison J.B., Arnould V.J., Sallum J.M.F., Vagefi M.R., Barmada M.M., Li Y.Y. Genetic heterogeneity of dominant optic atrophy, Kjer type – identification of a second locus on chromosome 18q12.2-12.3. Archives of Ophthalmology. 1999;117:805–810.
    1. Kerrison J.B., Miller N.R., Hsu F., Beaty T.H., Maumenee I.H., Smith K.H. A case-control study of tobacco and alcohol consumption in Leber hereditary optic neuropathy. American Journal of Ophthalmology. 2000;130:803–812.
    1. Khrapko K. Two ways to make an mtDNA bottleneck. Nature Genetics. 2008;40:134–135.
    1. Kim J.Y., Hwang J.M., Park S.S. Mitochondrial DNA C4171A/ND1 is a novel primary causative mutation of Leber’s hereditary optic neuropathy with a good prognosis. Annals of Neurology. 2002;51:630–634.
    1. Kim T.W., Hwang J.M. Stratus OCT in dominant optic atrophy: features differentiating it from glaucoma. Journal of Glaucoma. 2007;16:655–658.
    1. Kim U., Hwang J.M. Early stage ethambutol optic neuropathy: retinal nerve fiber layer and optical coherence tomography. European Journal of Ophthalmology. 2009;19:466–469.
    1. Kirkman M.A., Korsten A., Leonhardt M., Dimitriadis K., De Coo I.F., Klopstock T. Quality of life in patients with leber hereditary optic neuropathy. Investigative Ophthalmology & Visual Science. 2009;50:3112–3115.
    1. Kirkman M.A., Yu-Wai-Man P., Korsten A., Leonhardt M., Dimitriadis K., De Coo I.F. Gene-environment interactions in Leber hereditary optic neuropathy. Brain. 2009;132:2317–2326.
    1. Kjer B. Infantile optic atrophy with dominant transmission. Danish Medical Bulletin. 1956;3:135–141.
    1. Kjer B., Eiberg H., Kjer P., Rosenberg T. Dominant optic atrophy mapped to chromosome 3q region. II. Clinical and epidemiological aspects. Acta Ophthalmologica Scandinavica. 1996;74:3–7.
    1. Kjer P. Infantile optic atrophy with dominant mode of inheritance: a clinical and genetic study of 19 Danish families. Acta Ophthalmologica. 1959:1–146.
    1. Kline L.B., Glaser J.S. Dominant optic atrophy – clinical profile. Archives of Ophthalmology. 1979;97:1680–1686.
    1. Klivenyi P., Karg E., Rozsa C., Horvath R., Komoly S., Nemeth I. alpha-tocopherol/lipid ratio in blood is decreased in patients with Leber’s hereditary optic neuropathy and asymptomatic carriers of the 11778 mtDNA mutation. Journal of Neurology, Neurosurgery, and Psychiatry. 2001;70:359–362.
    1. Kobbert C., Apps R., Bechmann I., Lanciego J.L., Mey J., Thanos S. Current concepts in neuroanatomical tracing. Progress in Neurobiology. 2000;62:327–351.
    1. Koch-Henriksen N., Sorensen P.S. The changing demographic pattern of multiple sclerosis epidemiology. Lancet Neurology. 2010;9:520–532.
    1. Kovacs G.G., Hoftberger R., Horvath R., Barsi P., Komoly S., Lassmann H. Neuropathology of white matter disease in Leber’s hereditary optic neuropathy. Brain. 2005;128:35–41.
    1. Kumaramanickavel G., Aung T., Sripriya S., Waseem N., Okada K., Seah S.K.L. Lack of association of IVS8+4 C/T and IVS8+32 T/C polymorphisms in the OPA1 gene with normal tension glaucoma in patients from Singapore, India and Japan. Investigative Ophthalmology & Visual Science. 2005;46:3811.
    1. Kyriakouli D.S., Boesch P., Taylor R.W., Lightowlers R.N. Progress and prospects: gene therapy for mitochondrial DNA disease. Gene Therapy. 2008;15:1017–1023.
    1. La Morgia C., Achilli A., Iommarini L., Barboni P., Pala M., Olivieri A. Rare mtDNA variants in Leber hereditary optic neuropathy families with recurrence of myoclonus. Neurology. 2008;70:762–770.
    1. La Morgia C., Ross-Cisneros F.N., Sadun A.A., Hannibal J., Munarini A., Mantovani V. Melanopsin retinal ganglion cells are resistant to neurodegeneration in mitochondrial optic neuropathies. Brain. 2010;133:2426–2438.
    1. Lamminen T., Majander A., Juvonen V., Wikstrom M., Aula P., Nikoskelainen E. A mitochondrial mutation at nt 9101 in the ATP synthase 6 gene associated with deficient oxidative phosphorylation in a family with Leber hereditary optic neuroretinopathy. American Journal of Human Genetics. 1995;56:1238–1240. [letter]
    1. Landrum J.T., Bone R.A. Lutein, zeaxanthin, and the macular pigment. Archives of Biochemistry and Biophysics. 2001;385:28–40.
    1. Larsson N.G., Andersen O., Holme E., Oldfors A., Wahlstrom J. Leber’s hereditary optic neuropathy and complex I deficiency in muscle. Annals of Neurology. 1991;30:701–708.
    1. Lascaratos G., Ji D., Wood J.P.M., Osborne N.N. Visible light affects mitochondrial function and induces neuronal death in retinal cell cultures. Vision Research. 2007;47:1191–1201.
    1. Lee E.J., Kim S.J., Choung H.K., Kim J.H., Yu Y.S. Incidence and clinical features of ethambutol-induced optic neuropathy in Korea. Journal of Neuro-Ophthalmology. 2008;28:269–277.
    1. Lenaers G., Reynier P., ElAchouri G., Soukkarieh C., Olichon A., Belenguer P. OPA1 functions in mitochondria and dysfunctions in optic nerve. International Journal of Biochemistry & Cell Biology. 2009;41:1866–1874.
    1. Leo-Kottler B., Luberichs J., Besch D., Christ-Adler M., Fauser S. Leber’s hereditary optic neuropathy: clinical and molecular genetic results in a patient with a point mutation at np T11253C (isoleucine to threonine) in the ND4 gene and spontaneous recovery. Graefes Archive for Clinical & Experimental Ophthalmology. 2002;240:758–764.
    1. Lessell S. Nutritional amblyopia. Journal of Neuro-Ophthalmology. 1998;18:106–111.
    1. Leung C.K.S., Weinreb R.N. Experimental detection of retinal ganglion cell damage in vivo. Experimental Eye Research. 2009;88:831–836.
    1. Li C.H., Cheng Y.W., Liao P.L., Yang Y.T., Kang J.J. Chloramphenicol causes mitochondrial stress, decreases ATP biosynthesis, induces matrix metalloproteinase-13 expression, and solid-tumor cell invasion. Toxicological Sciences. 2010;116:140–150.
    1. Libby R.T., Gould D.B., Anderson M.G., John S.W.M. Complex genetics of glaucoma susceptibility. Annual Review of Genomics and Human Genetics. 2005;6:15–44.
    1. Lightowlers R.N., Chinnery P.F., Turnbull D.M., Howell N. Mammalian mitochondrial genetics: heredity, heteroplasmy and disease. Trends in Genetics. 1997;13:450–455.
    1. Liguori M. A phenotypic variation of dominant optic atrophy and deafness (ADOAD) due to a novel OPA1 mutation. Journal of Neurology. 2008;255:127–129.
    1. Lodi R., Carelli V., Cortelli P., Iotti S., Valentino M.L., Barboni P. Phosphorus MR spectroscopy shows a tissue specific in vivo distribution of biochemical expression of the G3460A mutation in Leber’s hereditary optic neuropathy. Journal of Neurology, Neurosurgery, and Psychiatry. 2002;72:805–807.
    1. Lodi R., Montagna P., Cortelli P., Iotti S., Cevoli S., Carelli V. ’Secondary’ 4216/ND1 and 13708/ND5 Leber’s hereditary optic neuropathy mitochondrial DNA mutations do not further impair in vivo mitochondrial oxidative metabolism when associated with the 11778/ND4 mitochondrial DNA mutation. Brain. 2000;123:1896–1902.
    1. Lodi R., Taylor D.J., Tabrizi S.J., Kumar S., Sweeney M., Wood N.W. In vivo skeletal muscle mitochondrial function in Leber’s hereditary optic neuropathy assessed by P-31 magnetic resonance spectroscopy. Annals of Neurology. 1997;42:573–579.
    1. Lodi R., Tonon C., Valentino M.L., Iotti S., Clementi V., Malucelli E. Deficit of in vivo mitochondrial ATP production in OPA1-related dominant optic atrophy. Annals of Neurology. 2004;56:719–723.
    1. Loiseau D., Chevrollier A., Verny C., Guillet V., Gueguen N., de Crescenzo M.A.P. Mitochondrial coupling defect in Charcot-Marie-Tooth type 2A disease. Annals of Neurology. 2007;61:315–323.
    1. Longley M.J., Clark S., Man C.Y.W., Hudson G., Durham S.E., Taylor R.W. Mutant POLG2 disrupts DNA polymerase gamma subunits and causes progressive external ophthalmoplegia. American Journal of Human Genetics. 2006;78:1026–1034.
    1. Loveland B., Wang C.R., Yonekawa H., Hermel E., Lindahl K.F. Maternally transmitted histocompatibility antigen of mice – a hydrophobic peptide of a mitochondrially encoded protein. Cell. 1990;60:971–980.
    1. Luca C.C., Lam B.L., Moraes C.T. Erythromycin as a potential precipitating agent in the onset of Leber’s hereditary optic neuropathy. Mitochondrion. 2004;4:31–36.
    1. Maagaard A., Holberg-Petersen M., Kvittingen E.A., Sandvik L., Bruun J.N. Depletion of mitochondrial DNA copies/cell in peripheral blood mononuclear cells in HIV-1-infected treatment-naive patients. HIV Medicine. 2006;7:53–58.
    1. Mabuchi F., Tang S., Kashiwagi K., Yamagata Z., Iijima H., Tsukahara S. The OPA1 gene polymorphism is associated with normal tension and high tension glaucoma. American Journal of Ophthalmology. 2007;143:125–130.
    1. MacKenzie J.A., Payne R.M. Mitochondrial protein import and human health and disease. Biochimica et Biophysica Acta-Molecular Basis of Disease. 2007;1772:509–523.
    1. Mackey D., Howell N. A variant of Leber hereditary optic neuropathy characterized by recovery of vision and by an unusual mitochondrial genetic etiology. American Journal of Human Genetics. 1992;51:1218–1228.
    1. Mackey D.A., Buttery R.G. Leber hereditary optic neuropathy in Australia. Australian and New Zealand Journal of Ophthalmology. 1992;20:177–184.
    1. Mackey D.A., Fingert J.H., Luzhansky J.Z., McCluskey P.J., Howell N., Hall A.J.H. Leber’s hereditary optic neuropathy triggered by antiretroviral therapy for human immunodeficiency virus. Eye. 2003;17:312–317.
    1. Mackey D.A., Oostra R.J., Rosenberg T., Nikoskelainen E., Bronte-Stewart J., Poulton J. Primary pathogenic mtDNA mutations in multigeneration pedigrees with Leber hereditary optic neuropathy. American Journal of Human Genetics. 1996;59:481–485.
    1. Mackey D.A., Trounce I. Genetics: optic nerve genetics-more than meets the eye. Nature Reviews Neurology. 2010;6:357–358.
    1. Mahad D.J., Ziabreva I., Campbell G., Lax N., White K., Hanson P.S. Mitochondrial changes within axons in multiple sclerosis. Brain. 2009;132:1161–1174.
    1. Mahad D.J., Ziabreva I., Lassmann H., Turnbull D.M. Mitochondria and primary oligodendrocyte dysfunction in multiple sclerosis. Journal of Neurology, Neurosurgery, and Psychiatry. 2007;78:101.
    1. Majander A., Finel M., Savontaus M.L., Nikoskelainen E., Wikstrom M. Catalytic activity of complex I in cell lines that possess replacement mutations in the ND genes in Leber’s hereditary optic neuropathy. European Journal of Biochemistry. 1996;239:201–207.
    1. Majander A., Huoponen K., Savontaus M.L., Nikoskelainen E., Wikstrom M. Electron transfer properties of NADH:ubiquinone reductase in the ND1/3460 and the ND4/11778 mutations of the Leber hereditary optic neuroretinopathy (LHON) FEBS Letters. 1991;292:289–292.
    1. Man P.Y., Griffiths P.G., Brown D.T., Howell N., Turnbull D.M., Chinnery P.F. The epidemiology of Leber hereditary optic neuropathy in the North East of England. American Journal of Human Genetics. 2003;72:333–339.
    1. Manfredi G., Fu J., Ojaimi J., Sadlock J.E., Kwong J.Q., Guy J. Rescue of a deficiency in ATP synthesis by transfer of MTATP6, a mitochondrial DNA-encoded gene, to the nucleus. Nature Genetics. 2002;30:394–399.
    1. Mao P.Z., Reddy P.H. Is multiple sclerosis a mitochondrial disease? Biochimica et Biophysica Acta-Molecular Basis of Disease. 2010;1802:66–79.
    1. Marchbank N.J., Craig J.E., Leek J.P., Toohey M., Churchill A.J., Markham A.F. Deletion of the OPA1 gene in a dominant optic atrophy family: evidence that haploinsufficiency is the cause of disease. Journal of Medical Genetics. 2002;39:e47.
    1. Marella, M., Seo, B.B., Thomas, B.B., Matsuno-Yagi, A., Yagi, T., 2010. Successful amelioration of mitochondrial optic neuropathy using the yeast NDI1 gene in a rat animal model. PLoS One, 5, e11472.
    1. Margulis L. Symbiosis and evolution. Scientific American. 1971;225:48–57.
    1. Martin W., Muller M. The hydrogen hypothesis for the first eukaryote. Nature. 1998;392:37–41.
    1. Martinelli P., Rugarli E.I. Emerging roles of mitochondrial proteases in neurodegeneration. Biochimica et Biophysica Acta-Bioenergetics. 2010;1797:1–10.
    1. Maruyama H., Morino H., Ito H., Izumi Y., Kato H., Watanabe Y. Mutations of optineurin in amyotrophic lateral sclerosis. Nature. 2010;465:223–226.
    1. Mashima Y., Kigasawa K., Hasegawa H., Tani M., Oguchi Y. High incidence of pre-excitation syndrome in Japanese families with Leber’s hereditary optic neuropathy. Clinical Genetics. 1996;50:535–537.
    1. Mashima Y., Kigasawa K., Wakakura M., Oguchi Y. Do idebenone and vitamin therapy shorten the time to achieve visual recovery in Leber hereditary optic neuropathy? Journal of Neuro-Ophthalmology. 2000;20:166–170.
    1. Mashima Y., Yamada K., Wakakura M., Kigasawa K., Kudoh J., Shimizu N. Spectrum of pathogenic mitochondrial DNA mutations and clinical features in Japanese families with Leber’s hereditary optic neuropathy. Current Eye Research. 1998;17:403–408.
    1. Mayorov V., Biousse V., Newman N.J., Brown M.D. The role of the ND5 gene in LHON: characterization of a new, heteroplasmic LHON mutation. Annals of Neurology. 2005;58:807–811.
    1. McFarland R., Chinnery P.F., Blakely E.L., Schaefer A.M., Morris A.A.M., Foster S.M. Homoplasmy, heteroplasmy, and mitochondrial dystonia. Neurology. 2007;69:911–916.
    1. McKinley S.H., Foroozan R. Optic neuropathy associated with linezolid treatment. Journal of Neuro-Ophthalmology. 2005;25:18–21.
    1. Meire F., Delaey J.J., Debie S., Vanstaey M., Matton M.T. Dominant optic-nerve atrophy with progressive hearing-loss and chronic progressive external ophthalmoplegia (Cpeo) Ophthalmic Paediatrics and Genetics. 1985;5:91–97.
    1. Meire F.M., Van Coster R., Cochaux P., Obermaier-Kusser B., Candaele C., Martin J.J. Neurological disorders in members of families with Leber’s hereditary optic neuropathy (LHON) caused by different mitochondrial mutations. Ophthalmic Genetics. 1995;16:119–126.
    1. Melamud A., Kosmorsky G.S., Lee M.S. Ocular ethambutol toxicity. Mayo Clinic Proceedings. 2003;78:1409–1411.
    1. Meyer E., Michaelides M., Tee L.J., Robson A.G., Rahman F., Pasha S. Nonsense mutation in TMEM126A causing autosomal recessive optic atrophy and auditory neuropathy. Molecular Vision. 2010;16:650–664.
    1. Milea D., Sander B., Wegener M., Jensen H., Kjer B., Jorgensen T.M. Axonal loss occurs early in dominant optic atrophy. Acta Ophthalmologica. 2010;88:342–346.
    1. Miller J.W. Preliminary results of gene therapy for retinal degeneration. New England Journal of Medicine. 2008;358:2282–2284.
    1. Mokranjac D., Bourenkov G., Hell K., Neupert W., Groll M. Structure and function of Tim14 and Tim16, the J and J-like components of the mitochondrial protein import motor. EMBO Journal. 2006;25:4675–4685.
    1. Montagna P., Plazzi G., Cortelli P., Carelli V., Lugaresi E., Barboni P. Abnormal lactate after effort in healthy carriers of Leber’s hereditary optic neuropathy. Journal of Neurology, Neurosurgery, and Psychiatry. 1995;58:640–641. [letter]
    1. Moore B.A., Gonzalez Aviles G.D., Larkins C.E., Hillman M.J., Caspary T. Mitochondrial retention of Opa1 is required for mouse embryogenesis. Mammalian Genome. 2010;21:350–360.
    1. Morfini G.A., Burns M., Binder L.I., Kanaan N.M., LaPointe N., Bosco D.A. Axonal transport defects in neurodegenerative diseases. Journal of Neuroscience. 2009;29:12776–12786.
    1. Morgan J.E. Circulation and axonal transport in the optic nerve. Eye. 2004;18:1089–1095.
    1. Nekhaeva E., Bodyak N.D., Kraytsberg Y., McGrath S.B., Van Orsouw N.J., Pluzhnikov A. Clonally expanded mtDNA point mutations are abundant in individual cells of human tissues. Proceedings of the National Academy of Sciences of the United States of America. 2002;99:5521–5526.
    1. Newman N.J. Leber hereditary optic neuropathy: bad habits, bad vision. Brain. 2009;132:2306–2308.
    1. Newman N.J., Biousse V. Hereditary optic neuropathies. Eye. 2004;18:1144–1160.
    1. Newman N.J., Biousse V., David R., Bhatti M.T., Hamilton S.R., Farris B.K. Prophylaxis for second eye involvement in leber hereditary optic neuropathy: an open-labeled, nonrandomized multicenter trial of topical brimonidine purite. American Journal of Ophthalmology. 2005;140:407–415.
    1. Newman N.J., Torroni A., Brown M.D., Lott M.T., Fernandez M.M., Wallace D.C. Epidemic neuropathy in Cuba not associated with mitochondrial-DNA mutations found in Lebers hereditary optic neuropathy patients. American Journal of Ophthalmology. 1994;118:158–168.
    1. Nijtmans L.G.J., Ugalde C., van den Heuvel L.P., Smeitink J.A.M. Function and dysfunction of the oxidative phosphorylation system. Mitochondrial Function and Biogenesis. 2004;8:149–176.
    1. Nikoskelainen E., Sogg R.L., Rosenthal A.R., Friberg T.R., Dorfman L.J. The early phase in Leber hereditary optic atrophy. Archives of Ophthalmology. 1977;95:969–978.
    1. Nikoskelainen E.K. Clinical picture of LHON. Clinical Neuroscience. 1994;2:115–120.
    1. Nikoskelainen E.K., Huoponen K., Juvonen V., Lamminen T., Nummelin K., Savontaus M.L. Ophthalmologic findings in Leber hereditary optic neuropathy, with special reference to mtDNA mutations. Ophthalmology. 1996;103:504–514.
    1. Nikoskelainen E.K., Marttila R.J., Huoponen K., Juvonen V., Lamminen T., Sonninen P. Leber’s “plus”: neurological abnormalities in patients with Leber’s hereditary optic neuropathy. Journal of Neurology, Neurosurgery, and Psychiatry. 1995;59:160–164.
    1. Nikoskelainen E.K., Savontaus M.L., Huoponen K., Antila K., Hartiala J. Pre-excitation syndrome in Leber’s hereditary optic neuropathy. Lancet. 1994;344:857–858.
    1. Nishigaki Y., Marti R., Copeland W.C., Hirano M. Site-speciflic somatic mitochondrial DNA point mutations in patients with thymidine phosphorylase deficiency. Journal of Clinical Investigation. 2003;111:1913–1921.
    1. Nishino I., Spinazzola A., Hirano M. Thymidine phosphorylase gene mutations in MNGIE, a human mitochondrial disorder. Science. 1999;283:689–692.
    1. Nochez Y., Arsene S., Gueguen N., Chevrollier A., Ferre M., Guillet V. Acute and late-onset optic atrophy due to a novel OPA1 mutation leading to a mitochondrial coupling defect. Molecular Vision. 2009;15:598–608.
    1. Nolan D., Mallal S. Complications associated with NRTI therapy: update on clinical features, and possible pathogenic mechanisms. Antiviral Therapy. 2004;9:849–863.
    1. Olichon A., Guillou E., Delettre C., Landes T., Arnaune-Pelloquin L., Emorine L.J. Mitochondrial dynamics and disease, OPA1. Biochimica et Biophysica Acta-Molecular Cell Research. 2006;1763:500–509.
    1. Olichon A., Landes T., Arnaune-Pelloquin L., Emorine L.J., Mils V., Guichet A. Effects of OPA1 mutations on mitochondrial morphology and apoptosis: relevance to ADOA pathogenesis. Journal of Cellular Physiology. 2007;211:423–430.
    1. Oostra R.J., Kemp S., Bolhuis P.A., Bleeker-Wagemakers E.M. No evidence for ’skewed’ inactivation of the X-chromosome as cause of Leber’s hereditary optic neuropathy in female carriers. Human Genetics. 1996;97:500–505.
    1. Oostra R.J., Van Galen M.J., Bolhuis P.A., Bleeker-Wagemakers E.M., Van den Bogert C. The mitochondrial DNA mutation ND6∗14,484C associated with leber hereditary optic neuropathy, leads to deficiency of complex I of the respiratory chain. Biochemical and Biophysical Research Communications. 1995;215:1001–1005.
    1. Osborne N.N., Li G.Y., Ji D., Mortiboys H.J., Jackson S. Light affects mitochondria to cause apoptosis to cultured cells: possible relevance to ganglion cell death in certain optic neuropathies. Journal of Neurochemistry. 2008;105:2013–2028.
    1. Ostojic J., Jancic J., Kozic D., Semnic R., Koprivsek K., Prvulovic M. Brain white matter 1 H MRS in Leber optic neuropathy mutation carriers. Acta Neurologica Belgica. 2009;109:305–309.
    1. Palace J. Multiple sclerosis associated with Leber’s hereditary optic neuropathy. Journal of the Neurological Sciences. 2009;286:24–27.
    1. Pareyson D., Marchesi C. Diagnosis, natural history, and management of Charcot-Marie-Tooth disease. Lancet Neurology. 2009;8:654–667.
    1. Parker W.D., Jr., Oley C.A., Parks J.K. A defect in mitochondrial electron-transport activity (NADH-coenzyme Q oxidoreductase) in Leber’s hereditary optic neuropathy. New England Journal of Medicine. 1989;320:1331–1333.
    1. Parsons T.J., Muniec D.S., Sullivan K., Woodyatt N., AllistonGreiner R., Wilson M.R. A high observed substitution rate in the human mitochondrial DNA control region. Nature Genetics. 1997;15:363–368.
    1. Payne M., Yang Z.L., Katz B.J., Warner J.E.A., Weight C.J., Zhao Y. Dominant optic atrophy, sensorineural hearing loss, ptosis, and ophthalmoplegia: a syndrome caused by a missense mutation in OPA1. American Journal of Ophthalmology. 2004;138:749–755.
    1. Pegoraro E., Carelli V., Zeviani M., Cortelli P., Montagna P., Barboni P. X-inactivation patterns in female Leber’s hereditary optic neuropathy patients do not support a strong X-linked determinant. American Journal of Medical Genetics. 1996;61:356–362.
    1. Pei W.H., Kratz L.E., Bernardini I., Sood R., Yokogawa T., Dorward H. A model of Costeff Syndrome reveals metabolic and protective functions of mitochondrial OPA3. Development. 2010;137:2587–2596.
    1. Pellegrini L., Scorrano L. A cut short to death: Parl and Opa1 in the regulation of mitochondrial morphology and apoptosis. Cell Death and Differentiation. 2007;14:1275–1284.
    1. Pello R., Martin M.A., Carelli V., Nijtmans L.G., Achilli A., Pala M. Mitochondrial DNA background modulates the assembly kinetics of OXPHOS complexes in a cellular model of mitochondrial disease. Human Molecular Genetics. 2008;17:4001–4011.
    1. Perales-Clemente, E., Fernandez-Silva, P., Acin-Perez, R., Perez-Martos, A., Enriquez, J.A., 2010. Allotopic expression of mitochondrial-encoded genes in mammals: achieved goal, undemonstrated mechanism or impossible task? Nucleic Acids Research, Epub ahead of print.
    1. Perkins G., Renken C., Martone M.E., Young S.J., Ellisman M. Electron tomography of neuronal mitochondria: three-dimensional structure and organization of cristae and membrane contacts. Journal of Structural Biology. 1997;119:260–272.
    1. Pesch U.E.A., Fries J.E., Bette S., Kalbacher H., Wissinger B., Alexander C. OPA1, the disease gene for autosomal dominant optic atrophy, is specifically expressed in ganglion cells and intrinsic neurons of the retina. Investigative Ophthalmology & Visual Science. 2004;45:4217–4225.
    1. Pesch U.E.A., Leo-Kottler B., Mayor S., Jurklies B., Kellner U., Apfelstedt-Sylla E. OPA1 mutations in patients with autosomal dominant optic atrophy and evidence for semi-dominant inheritance. Human Molecular Genetics. 2001;10:1359–1368.
    1. Pfeffer G., Cote H.C.F., Montaner J.S., Li C.C., Jitratkosol M., Mezei M.M. Ophthalmoplegia and ptosis: mitochondrial toxicity in patients receiving HIV therapy. Neurology. 2009;73:71–72.
    1. Phasukkijwatana N., Kunhapan B., Stankovich J., Chuenkongkaew W.L., Thomson R., Thornton T. Genome-wide linkage scan and association study of PARL to the expression of LHON families in Thailand. Human Genetics. 2010;128:39–49.
    1. Pich S., Bach D., Briones P., Liesa M., Camps M., Testar X. The Charcot-Marie-Tooth type 2A gene product, Mfn2, up-regulates fuel oxidation through expression of OXPHOS system. Human Molecular Genetics. 2005;14:1405–1415.
    1. Pommer R., Schoeler S., Mawrin C., Szibor R., Kirches E. The G11778A LHON mutation does not enhance ethambutol cytotoxicity in a cybrid model. Clinical Neuropathology. 2008;27:414–423.
    1. Ponjavic V., Andreasson S., Tranebjaerg L., Lubs H.A. Full-field electroretinograms in a family with Mohr–Tranebjaerg syndrome. Acta Ophthalmologica Scandinavica. 1996;74:632–635.
    1. Powell B., Toomes C., Scott S., Yeung A., Marchbank N., Spry P. Polymorphisms in OPA1 are associated with normal tension glaucoma. Molecular Vision. 2003;9:460–464.
    1. Pradhan M., Sharp D., Best S., Vincent A., Vaphiades M. Drug-induced optic neuropathy-TB or Not TB. Survey of Ophthalmology. 2010;55:378–385.
    1. Puomila A., Hamalainen P., Kivioja S., Savontaus M.L., Koivumaki S., Huoponen K. Epidemiology and penetrance of Leber hereditary optic neuropathy in Finland. European Journal of Human Genetics. 2007;15:1079–1089.
    1. Puomila A., Huoponen K., Mantyjarvi M., Hamalainen P., Paananen R., Sankila E.M. Dominant optic atrophy: correlation between clinical and molecular genetic studies. Acta Ophthalmologica Scandinavica. 2005;83:337–346.
    1. Purohit S.S., Tomsak R.L. Nutritional deficiency amblyopia or Leber’s hereditary optic neuropathy? Neuro-Ophthalmology. 1997;18:111–116.
    1. Qi X.P., Lewin A.S., Hauswirth W.W., Guy J. Suppression of complex I gene expression induces optic neuropathy. Annals of Neurology. 2003;53:198–205.
    1. Qi X.P., Lewin A.S., Sun L., Hauswirth W.W., Guy J. SOD2 gene transfer protects against optic neuropathy induced by deficiency of complex I. Annals of Neurology. 2004;56:182–191.
    1. Qi X.P., Sun L., Hauswirth W.W., Lewin A.S., Guy J. Use of mitochondrial antioxidant defenses for rescue of cells with a Leber hereditary optic neuropathy-causing mutation. Archives of Ophthalmology. 2007;125:268–272.
    1. Qi X.P., Sun L., Lewin A.S., Hauswirth W.W., Guy J. The mutant human ND4 subunit of complex I induces optic neuropathy in the mouse. Investigative Ophthalmology & Visual Science. 2007;48:1–10.
    1. Quigley H.A., Broman A.T. The number of people with glaucoma worldwide in 2010 and 2020. British Journal of Ophthalmology. 2006;90:262–267.
    1. Quiros P.A., Torres R.J., Salomao S., Berezovsky A., Carelli V., Sherman J. Colour vision defects in asymptomatic carriers of the Leber’s hereditary optic neuropathy (LHON) mtDNA 11778 mutation from a large Brazilian LHON pedigree: a case-control study. British Journal of Ophthalmology. 2006;90:150–153.
    1. Raha S., Robinson B.H. Mitochondria, oxygen free radicals, disease and ageing. Trends in Biochemical Sciences. 2000;25:502–508.
    1. Ramos CdVF, Bellusci C., Savini G., Carbonelli M., Berezovsky A., Tamaki C. Association of optic disc size with development and prognosis of Leber’s hereditary optic neuropathy. Investigative Ophthalmology & Visual Science. 2009;50:1666–1674.
    1. Ray K., Mookherjee S. Molecular complexity of primary open angle glaucoma: current concepts. Journal of Genetics. 2009;88:451–467.
    1. Reid E. Science in motion: common molecular pathological themes emerge in the hereditary spastic paraplegias. Journal of Medical Genetics. 2003;40:81–86.
    1. Reynier P., Amati-Bonneau P., Verny C., Olichon A., Simard G., Guichet A. OPA3 gene mutations responsible for autosomal dominant optic atrophy and cataract. Journal of Medical Genetics. 2004:41.
    1. Rezaie T., Child A., Hitchings R., Brice G., Miller L., Coca-Prados M. Adult-onset primary open-angle glaucoma caused by mutations in optineurin. Science. 2002;295:1077–1079.
    1. Richardson C., Smith T., Schaefer A., Turnbull D., Griffiths P. Ocular motility findings in chronic progressive external ophthalmoplegia. Eye. 2005;19:258–263.
    1. Rizzo J.F., Lessell S. Tobacco amblyopia. American Journal of Ophthalmology. 1993;116:84–87.
    1. Rodriguez M.C., MacDonald J.R., Mahoney D.J., Parise G., Beal M.F., Tarnopolsky M.A. Beneficial effects of creatine, CoQ(10), and lipoic acid in mitochondrial disorders. Muscle & Nerve. 2007;35:235–242.
    1. Rotig A., de Lonlay P., Chretien D., Foury F., Koenig M., Sidi D. Frataxin gene expansion causes aconitase and mitochondrial iron-sulphur protein deficiency in Friedreich ataxia. European Journal of Human Genetics. 1998;6:C304.
    1. Rotig A., deLonlay P., Chretien D., Foury F., Koenig M., Sidi D. Aconitase and mitochondrial iron–sulphur protein deficiency in Friedreich ataxia. Nature Genetics. 1997;17:215–217.
    1. Rotig A., Sidi D., Munnich A., Rustin P. Molecular insights into Friedreich’s ataxia and antioxidant-based therapies. Trends in Molecular Medicine. 2002;8:221–224.
    1. Rouault T.A., Tong W.H. Iron-sulphur cluster biogenesis and mitochondrial iron homeostasis. Nature Reviews Molecular Cell Biology. 2005;6:345–351.
    1. Rucker J.C., Hamilton S.R., Bardenstein D., Isada C.M., Lee M.S. Linezolid-associated toxic optic neuropathy. Neurology. 2006;66:595–598.
    1. Rustin P., Bonnet D., Rotig A., Munnich A., Sidi D. Idebenone treatment in Friedreich patients: one-year-long randomized placebo-controlled trial. Neurology. 2004;62:524–525.
    1. Rustin P., von Kleist-Retzow J.C., Chantrel-Groussard K., Sidi D., Munnich A., Rotig A. Effect of idebenone on cardiomyopathy in Friedreich’s ataxia: a preliminary study. Lancet. 1999;354:477–479.
    1. Ryu S.W., Jeong H.J., Choi M., Karbowski M., Choi C. Optic atrophy 3 as a protein of the mitochondrial outer membrane induces mitochondrial fragmentation. Cellular and Molecular Life Sciences. 2010;67:2839–2850.
    1. Sacai P.Y., Salomao S.R., Carelli V., Pereira J.M., Belfort R., Jr., Sadun A.A. Visual evoked potentials findings in non-affected subjects from a large Brazilian pedigree of 11778 Leber’s hereditary optic neuropathy. Documenta Ophthalmologica. 2010;121:147–154.
    1. Sacconi S., Trevisson E., Salviati L., Ayme S., Rigal O., Redondo A.G. Coenzyme Q(10) is frequently reduced in muscle of patients with mitochondrial myopathy. Neuromuscular Disorders. 2010;20:44–48.
    1. Sadun A.A., Carelli V., Salomao S.R., Berezovsky A., Quiros P.A., Sadun F. Extensive investigation of a large Brazilian pedigree of 11778/haplogroup J Leber hereditary optic neuropathy. American Journal of Ophthalmology. 2003;136:231–238.
    1. Sadun A.A., Martone J.F., Mucimendoza R., Reyes L., Dubois L., Silva J.C. Epidemic optic neuropathy in Cuba – eye findings. Archives of Ophthalmology. 1994;112:691–699.
    1. Sadun A.A., Salomao S.R., Berezovsky A., Sadun F., Denegri A.M., Quiros P.A. Subclinical carriers and conversions in Leber hereditary optic neuropathy: a prospective psychophysical study. Transactions of American Ophthalmological Society. 2006;104:51–61.
    1. Sadun A.A., Win P.H., Ross-Cisneros F.N., Walker S.O., Carelli V. Leber’s hereditary optic neuropathy differentially affects smaller axons in the optic nerve. Trans Am Ophthalmol Soc. 2000;98:223–232. discussion 232–5.
    1. Saijo T., Hayashi K., Yamada H., Wakakura M. Linezolid-induced optic neuropathy. American Journal of Ophthalmology. 2005;139:1114–1116.
    1. Sala G., Trombin F., Beretta S., Tremolizzo L., Presutto P., Montopoli M. Antioxidants partially restore glutamate transport defect in Leber hereditary optic neuropathy cybrids. Journal of Neuroscience Research. 2008;86:3331–3337.
    1. Salinas S., Proukakis C., Crosby A., Warner T.T. Hereditary spastic paraplegia: clinical features and pathogenetic mechanisms. Lancet Neurology. 2008;7:1127–1138.
    1. Salomao S.R., Berezovsky A., Andrade R.E., Belfort R., Carelli V., Sadun A.A. Visual electrophysiologic findings in patients from an extensive Brazilian family with Leber’s hereditary optic neuropathy – visual electrophysiology in LHON. Documenta Ophthalmologica. 2004;108:147–155.
    1. Samuels D.C. Mitochondrial AZT metabolism. IUBMB Life. 2006;58:403–408.
    1. Sanchez R.N., Smith A.J., Carelli V., Sadun A.A., Keltner J.L. Leber hereditary optic neuropathy possibly triggered by exposure to tire fire. Journal of Neuro-Ophthalmology. 2006;26:268–272.
    1. Sapey E., Burdon M.A., Nightingale S. Evidence of active demyelination in a man with Leber’s hereditary optic neuropathy mtDNA 14484 genotype. Neuro-Ophthalmology. 2001;26:119–126.
    1. Sato M., Kawase K., Yamamoto T., Dubo S., Shink E.E., Si E. Lack of association between normal-tension glaucoma and intron 8 polymorphisms in the gene causing autosomal dominant optic atrophy, OPA1, in Japan. Investigative Ophthalmology & Visual Science. 2003;44:1124.
    1. Savini G., Barboni P., Valentino M.L., Montagna P., Cortelli P., De Negri A.M. Retinal nerve fiber layer evaluation by optical coherence tomography in unaffected carriers with Leber’s hereditary optic neuropathy mutations. Ophthalmology. 2005;112:127–131.
    1. Schaefer A.M., McFarland R., Blakely E.L., He L., Whittaker R.G., Taylor R.W. Prevalence of mitochondrial DNA disease in adults. Annals of Neurology. 2008;63:35–39.
    1. Schimpf S., Schaich S., Wissinger B. Activation of cryptic splice sites is a frequent splicing defect mechanism caused by mutations in exon and intron sequences of the OPA1 gene. Human Genetics. 2006;118:767–771.
    1. Schmucker S., Puccio H. Understanding the molecular mechanisms of Friedreich’s ataxia to develop therapeutic approaches. Human Molecular Genetics. 2010;19:R103–R110.
    1. Schon E.A., DiMauro S., Hirano M., Gilkerson R.W. Therapeutic prospects for mitochondrial disease. Trends in Molecular Medicine. 2010;16:268–276.
    1. Schulzer M., Alward W.L., Feldman F., Cashwell L.F., Wilensky J., Geijssen H.C. Comparison of glaucomatous progression between untreated patients with normal-tension glaucoma and patients with therapeutically reduced intraocular pressures. American Journal of Ophthalmology. 1998;126:487–497.
    1. Scruggs E.R., Naylor A.J.D. Mechanisms of zidovudine-induced mitochondrial toxicity and myopathy. Pharmacology. 2008;82:83–88.
    1. Seo J.H., Hwang J.M., Park S.S. Comparison of retinal nerve fibre layers between 11778 and 14484 mutations in Leber’s hereditary optic neuropathy. Eye (London) 2009
    1. Seo J.H., Hwang J.M., Park S.S. Antituberculosis medication as a possible epigenetic factor of Leber’s hereditary optic neuropathy. Clinical and Experimental Ophthalmology. 2010;38:363–366.
    1. Shah V.A., Randhawa S., Mizen T., Lee A.G., Foroozan R. You’re too old for that. Survey of Ophthalmology. 2008;53:403–410.
    1. Shahrestani P., Leung H.T., Le P.K., Pak W.L., Tse S., Ocorr K. Heterozygous mutation of Drosophila Opa1 causes the development of multiple organ abnormalities in an age-dependent and organ-specific manner. Plos One. 2009;4
    1. Shaikh S., Ta C., Basham A.A., Mansour S. Leber hereditary optic neuropathy associated with antiretroviral therapy for human immunodeficiency virus infection. American Journal of Ophthalmology. 2001;131:143–145.
    1. Shankar S.P., Fingert J.H., Carelli V., Valentino M.L., King T.M., Daiger S.P. Evidence for a novel x-linked modifier locus for leber hereditary optic neuropathy. Ophthalmic Genetics. 2008;29:17–24.
    1. Shi P., Gal J., Kwinter D.M., Liu X.Y., Zhu H.N. Mitochondrial dysfunction in amyotrophic lateral sclerosis. Biochimica et Biophysica Acta-Molecular Basis of Disease. 2010;1802:45–51.
    1. Shimizu S., Mori N., Kishi M., Sugata H., Tsuda A., Kubota N. A novel mutation in the OPA1 gene in a Japanese patient with optic atrophy. American Journal of Ophthalmology. 2003;135:256–257.
    1. Shindler K.S., Ventura E., Rex T.S., Elliott P., Rostami A. SIRT1 activation confers neuroprotection in experimental optic neuritis. Investigative Ophthalmology & Visual Science. 2007;48:3602–3609.
    1. Shorr N., Christenbury J.D., Goldberg R.A. Management of ptosis in chronic progressive external ophthalmoplegia. Ophthal Plast Reconstr Surg. 1987;3:141–145.
    1. Shoubridge E.A., Karpati G., Hastings K.E.M. Deletion mutants are functionally dominant over wild-type mitochondrial genomes in skeletal-muscle fiber segments in mitochondrial disease. Cell. 1990;62:43–49.
    1. Shy M.E., Garbern J.Y., Kamholz J. Hereditary motor and sensory neuropathies: a biological perspective. Lancet Neurology. 2002;1:110–118.
    1. Sigal I.A., Ethier C.R. Biomechanics of the optic nerve head. Experimental Eye Research. 2009;88:799–807.
    1. Sigal I.A., Flanagan J.G., Tertinegg I., Ethier C.R. 3D morphometry of the human optic nerve head. Experimental Eye Research. 2010;90:70–80.
    1. Siguroardottir S., Helgason A., Gulcher J.R., Stefansson K., Donnelly P. The mutation rate in the human mtDNA control region. American Journal of Human Genetics. 2000;66:1599–1609.
    1. Skinnider L.F., Ghadially F.N. Chloramphenicol-induced mitochondrial and ultrastructural changes in hematopoietic cells. Archives of Pathology & Laboratory Medicine. 1976;100:601–605.
    1. Smith K.H., Johns D.R., Heher K.L., Miller N.R. Heteroplasmy in Leber’s hereditary optic neuropathy. Archives of Ophthalmology. 1993;111:1486–1490.
    1. Smith P.R., Cooper J.M., Govan G.G., Harding A.E., Schapira A.H. Platelet mitochondrial function in Leber’s hereditary optic neuropathy. Journal of the Neurological Sciences. 1994;122:80–83.
    1. Smith U., Smith D.S., Yunis A.A. Chloramphenicol-related changes in mitochondrial ultrastructure. Journal of Cell Science. 1970;7:501–521.
    1. Spelbrink J.N., Li F.Y., Tiranti V., Nikali K., Yuan Q.P., Tariq M. Human mitochondrial DNA deletions associated with mutations in the gene encoding Twinkle, a phage T7 gene LF-like protein localized in mitochondria. Nature Genetics. 2001;28:223–231.
    1. Spinazzi M., Cazzola S., Bortolozzi M., Baracca A., Loro E., Casarin A. A novel deletion in the GTPase domain of OPA1 causes defects in mitochondrial morphology and distribution, but not in function. Human Molecular Genetics. 2008;17:3291–3302.
    1. Spinazzola A., Invernizzi F., Carrara F., Lamantea E., Donati A., DiRocco M. Clinical and molecular features of mitochondrial DNA depletion syndromes. Journal of Inherited Metabolic Disease. 2009;32:143–158.
    1. Spinazzola A., Zeviani M. Mitochondrial diseases: a cross-talk between mitochondrial and nuclear genomes. In: Espinos C.F.V.P.F., editor. vol. 652. 2009. pp. 69–84. (Inherited Neuromuscular Diseases: Translation from Pathomechanisms to Therapies).
    1. Spruijt L., Kolbach D.N., de Coo R.F., Plomp A.S., Bauer N.J., Smeets H.J. Influence of mutation type on clinical expression of Leber hereditary optic neuropathy. American Journal of Ophthalmology. 2006;141:676–682.
    1. Spruijt L., Smeets H.J., Hendrickx A., Bettink-Remeijer M.W., Maat-Kievit A., Schoonderwoerd K.C. A MELAS-associated ND1 mutation causing Leber hereditary optic neuropathy and spastic dystonia. Archives of Neurology. 2007;64:890–893.
    1. Sripriya S., Nirmaladevi J., George R., Hemamalini A., Baskaran M., Prema R. OPTN gene: profile of patients with glaucoma from India. Molecular Vision. 2006;12:816–820.
    1. Stemmler T.L., Lesuisse E., Pain D., Dancis A. Frataxin and mitochondrial FeS cluster biogenesis. Journal of Biological Chemistry. 2010;285:26737–26743.
    1. Stewart J.D., Hudson G., Yu-Wai-Man P., Blakeley E.L., He L., Horvath R. Opa1 in multiple mitochondrial DNA deletion disorders. Neurology. 2008;71:1829–1831.
    1. Stone E.M., Newman N.J., Miller N.R., Johns D.R., Lott M.T., Wallace D.C. Visual recovery in patients with Leber’s hereditary optic neuropathy and the 11778 mutation. Journal of Clinical Neuro-Ophthalmology. 1992;12:10–14.
    1. Suen D.F., Norris K.L., Youle R.J. Mitochondrial dynamics and apoptosis. Genes & Development. 2008;22:1577–1590.
    1. Sugisaka E., Ohde H., Shinoda K., Mashima Y. Woman with atypical unilateral Leber’s hereditary optic neuropathy with visual improvement. Clinical & Experimental Ophthalmology. 2007;35:868–870.
    1. Tachibana M., Sparman M., Sritanaudomchai H., Ma H., Clepper L., Woodward J. Mitochondrial gene replacement in primate offspring and embryonic stem cells. Nature. 2009;461:367–372.
    1. Talman L.S., Bisker E.R., Sackel D.J., Long D.A., Jr., Galetta K.M., Ratchford J.N. Longitudinal study of vision and retinal nerve fiber layer thickness in multiple sclerosis. Annals of Neurology. 2010;67:749–760.
    1. Tang S., Le P.K., Tse S., Wallace D.C., Huang T.S. Heterozygous mutation of Opa1 in Drosophila shortens lifespan mediated through increased reactive oxygen species production. Plos One. 2009;4
    1. Tarnopolsky M.A. The mitochondrial cocktail: rationale for combined nutraceutical therapy in mitochondrial cytopathies. Advanced Drug Delivery Reviews. 2008;60:1561–1567.
    1. Tarnopolsky M.A., Baker S.K., Myint T., Maxner C.E., Robitaille J., Robinson B.H. Clinical variability in maternally inherited leber hereditary optic neuropathy with the G14459A mutation. American Journal of Medical Genetics: Part A. 2004;124:372–376.
    1. Taylor R.W., Jobling M.S., Turnbull D.M., Chinnery P.F. Frequency of rare mitochondrial DNA mutations in patients with suspected Leber’s hereditary optic neuropathy. Journal of Medical Genetics. 2003:40.
    1. Taylor R.W., Schaefer A.M., Barron M.J., McFarland R., Turnbull D.M. The diagnosis of mitochondrial muscle disease. Neuromuscular Disorders. 2004;14:237–245.
    1. Taylor R.W., Turnbull D.M. Mitochondrial DNA mutations in human disease. Nature Reviews Genetics. 2005;6:389–402.
    1. Tharaphan P., Chuenkongkaew W.L., Luangtrakool K., Sanpachudayan T., Suktitipat B., Suphavilai R. Mitochondrial DNA haplogroup distribution in pedigrees of Southeast Asian G11778A Leber hereditary optic neuropathy. Journal of Neuro-Ophthalmology. 2006;26:264–267.
    1. Thomas D.Y., Wilkie D. Inhibition of mitochondrial synthesis in yeast by erythromycin – cytoplasmic and nuclear factors controlling resistance. Genetical Research. 1968;11:33–41.
    1. Tok J.B.H., Bi L.R. Aminoglycoside and its derivatives as ligands to target the ribosome. Current Topics in Medicinal Chemistry. 2003;3:1001–1019.
    1. Tonon C., Lodi R. Idebenone in Friedreich’s ataxia. Expert Opinion on Pharmacotherapy. 2008;9:2327–2337.
    1. Tonska K., Kodron A., Bartnik E. Genotype-phenotype correlations in Leber hereditary optic neuropathy. Biochimica et Biophysica Acta-Bioenergetics. 2010;1797:1119–1123.
    1. Toomes C., Marchbank N.J., Mackey D.A., Craig J.E., Newbury-Ecob R.A., Bennett C.P. Spectrum, frequency and penetrance of OPA1 mutations in dominant optic atrophy. Human Molecular Genetics. 2001;10:1369–1378.
    1. Torroni A., Wallace D.C. Mitochondrial DNA variation in human populations and implications for detection of mitochondrial DNA mutations of pathological significance. Journal of Bioenergetics and Biomembranes. 1994;26:261–271.
    1. Touitou V., Johnson M.A., Miller N.R., Guo Y., Bernstein D.A., Bernstein S.L. Sustained neuroprotection after a single intravitreal injection of PGJ2 in a rodent model of NAION. Neuro-Ophthalmology. 2010;34:271–272.
    1. Tranebjaerg L. Deafness-dystonia-optic neuronopathy syndrome. In: Pagon R.A., Bird T.C., Dolan C.R., Stephens K., editors. GeneReviews. University of Washington, Seattle; Seattle (WA): 2003.
    1. Tranebjaerg L., Jensen P.K., Van Ghelue M., Vnencak-Jones C.L., Sund S., Elgjo K. Neuronal cell death in the visual cortex is a prominent feature of the X-linked recessive mitochondrial deafness-dystonia syndrome caused by mutations in the TIMM8a gene. Ophthalmic Genetics. 2001;22:207–223.
    1. Tranebjaerg L., Schwartz C., Eriksen H., Andreasson S., Ponjavic V., Dahl A. A new X-linked recessive deafness syndrome with blindness, dystonia, fractures, and mental deficiency is linked to Xq22. Journal of Medical Genetics. 1995;32:257–263.
    1. Tranebjaerg L., van Ghelue M., Nilssen O., Hodes M.E., Dlouhy S.R., Farlow M.R. Jensen syndrome is allelic to Mohr–Tranebjaerg syndrome and both are caused by stop mutations in the DDP gene. American Journal of Human Genetics. 1997;61:2043.
    1. Trobe J.D., Glaser J.S., Cassady J., Herschler J., Anderson D.R. Nonglaucomatous excavation of the optic disk. Archives of Ophthalmology. 1980;98:1046–1050.
    1. Tsao K., Aitken P.A., Johns D.R. Smoking as an aetiological factor in a pedigree with Leber’s hereditary optic neuropathy. British Journal of Ophthalmology. 1999;83:577–581.
    1. Tzoulis C., Engelsen B.A., Telstad W., Aasly J., Zeviani M., Winterthun S. The spectrum of clinical disease caused by the A467T and W748SPOLG mutations: a study of 26 cases. Brain. 2006;129:1685–1692.
    1. Ujike H., Tanabe Y., Takehisa Y., Hayabara T., Kuroda S. A family with X-linked dystonia-deafness syndrome with a novel mutation of the DDP gene. Archives of Neurology. 2001;58:1004–1007.
    1. Valentino M.L., Avoni P., Barboni P., Pallotti F., Rengo C., Torroni A. Mitochondrial DNA nucleotide changes C14482G and C14482A in the ND6 gene are pathogenic for Leber’s hereditary optic neuropathy. Annals of Neurology. 2002;51:774–778.
    1. Valentino M.L., Barboni P., Ghelli A., Bucchi L., Rengo C., Achilli A. The ND1 gene of complex I is a mutational hot spot for Leber’s hereditary optic neuropathy. Annals of Neurology. 2004;56:631–641.
    1. Van Goethem G., Dermaut B., Lofgren A., Martin J.J., Van Broeckhoven C. Mutation of POLG is associated with progressive external ophthalmoplegia characterized by mtDNA deletions. Nature Genetics. 2001;28:211–212.
    1. Vanjaarsveld H., Kuyl J.M., Alberts D.W. Exposure of rats to low concentration of cigarette-smoke increases myocardial sensitivity to ischemia reperfusion. Basic Research in Cardiology. 1992;87:393–399.
    1. Vanopdenbosch L., Dubois B., D’Hooghe M.B., Meire F., Carton H. Mitochondrial mutations of Leber’s hereditary optic neuropathy: a risk factor for multiple sclerosis. Journal of Neurology. 2000;247:535–543.
    1. Venegas-Francke P., Fruns-Quintana M., Oporto-Caroca M. Bilateral optic neuritis due to chloramphenicol. Revista De Neurologia. 2000;31:699–700.
    1. Vergani L., Martinuzzi A., Carelli V., Cortelli P., Montagna P., Schievano G. MtDNA mutations associated with Leber’s hereditary optic neuropathy: studies on cytoplasmic hybrid (cybrid) cells. Biochemical and Biophysical Research Communications. 1995;210:880–888.
    1. Verny C., Loiseau D., Scherer C., Lejeune P., Chevrollier A., Gueguen N. Multiple sclerosis-like disorder in OPA1-related autosomal dominant optic atrophy. Neurology. 2008;70:1152–1153.
    1. Votruba M., Fitzke F.W., Holder G.E., Carter A., Bhattacharya S.S., Moore A.T. Clinical features in affected individuals from 21 pedigrees with dominant optic atrophy. Archives of Ophthalmology. 1998;116:351–358.
    1. Votruba M., Moore A.T., Bhattacharya S.S. Clinical features, molecular genetics, and pathophysiology of dominant optic atrophy. Journal of Medical Genetics. 1998;35:793–800.
    1. Votruba M., Thiselton D., Bhattacharya S.S. Optic disc morphology of patients with OPA1 autosomal dominant optic atrophy. British Journal of Ophthalmology. 2003;87:48–53.
    1. Wakakura M., Yokoe J. Evidence for preserved direct pupillary light response in Lebers hereditary optic neuropathy. British Journal of Ophthalmology. 1995;79:442–446.
    1. Wallace D.C., Singh G., Lott M.T., Hodge J.A., Schurr T.G., Lezza A.M. Mitochondrial DNA mutation associated with Leber’s hereditary optic neuropathy. Science. 1988;242:1427–1430.
    1. Walsh M.K., Quigley H.A. In vivo time-lapse fluorescence imaging of individual retinal ganglion cells in mice. Journal of Neuroscience Methods. 2008;169:214–221.
    1. Wang Y., Michikawa Y., Mallidis C., Bai Y., Woodhouse L., Yarasheski K.E. Muscle-specific mutations accumulate with aging in critical human mtDNA control sites for replication. Proceedings of the National Academy of Sciences of the United States of America. 2001;98:4022–4027.
    1. Wanrooij S., Luoma P., van Goethem G., van Broeckhoven C., Suomalainen A., Spelbrink J.N. Twinkle and POLG defects enhance age-dependent accumulation of mutations in the control region of mtDNA. Nucleic Acids Research. 2004;32:3053–3064.
    1. Waterham H.R., Koster J., van Roermund C.W.T., Mooyer P.A.W., Wanders R.J.A., Leonard J.V. A lethal defect of mitochondrial and peroxisomal fission. New England Journal of Medicine. 2007;356:1736–1741.
    1. Weisschuh N., Neumann D., Wolf C., Wissinger B., Gramer E. Prevalence of myocilin and optineurin sequence variants in German normal tension glaucoma patients. Molecular Vision. 2005;11:284–287.
    1. Wheeler L., WoldeMussie E., Lai R. Role of alpha-2 agonists in neuroprotection. Survey of Ophthalmology. 2003;48:S47–S51.
    1. White K.E., Davies V.J., Hogan V.E., Piechota M.J., Nichols P.P., Turnbull D.M. OPA1 deficiency associated with increased autophagy in retinal ganglion cells in a murine model of dominant optic atrophy. Investigative Ophthalmology & Visual Science. 2009;50:2567–2571.
    1. Wiggs J.L. Genetic etiologies of glaucoma. Archives of Ophthalmology. 2007;125:30–37.
    1. Wilkinson P.A., Crosby A.H., Turner C., Bradley L.J., Ginsberg L., Wood N.W. A clinical, genetic and biochemical study of SPG7 mutations in hereditary spastic paraplegia. Brain. 2004;127:973–980.
    1. Williams P.A., Morgan J.E., Votruba M. Opa1 deficiency in a mouse model of dominant optic atrophy leads to retinal. Brain. 2010;133:2942–2951.
    1. Wissinger B., Besch D., Baumann B., Fauser S., Christ-Adler M., Jurklies B. Mutation analysis of the ND6 gene in patients with Lebers hereditary optic neuropathy. Biochemical and Biophysical Research Communications. 1997;234:511–515.
    1. Wolf C., Gramer E., Muller-Myhsok B., Pasutto F., Reinthal E., Wissinger B. Evaluation of nine candidate genes in patients with normal tension glaucoma: a case control study. BMC Medical Genetics. 2009:10.
    1. Wong A., Cavelier L., Collins-Schramm H.E., Seldin M.F., McGrogan M., Savontaus M.L. Differentiation-specific effects of LHON mutations introduced into neuronal NT2 cells. Human Molecular Genetics. 2002;11:431–438.
    1. Woo S.J., Kim D.M., Kim J.Y., Park S.S., Ko H.S., Yoo T. Investigation of the association between OPA1 polymorphisms and normal-tension glaucoma in Korea. Journal of Glaucoma. 2004;13:492–495.
    1. Yaffe M.P., Stuurman N., Vale R.D. Mitochondrial positioning in fission yeast is driven by association with dynamic microtubules and mitotic spindle poles. Proceedings of the National Academy of Sciences of the United States of America. 2003;100:11424–11428.
    1. Yang Z., Harrison C.A., Chuang G.C., Ballinger S.W. The role of tobacco smoke induced mitochondrial damage in vascular dysfunction and atherosclerosis. Mutation Research-Fundamental and Molecular Mechanisms of Mutagenesis. 2007;621:61–74.
    1. Yao W.L., Jiao X.D., Hejtmancik J., Leske M., Hennis A., Nemesure B. Evaluation of the association between OPA1 polymorphisms and primary open-angle glaucoma in Barbados families. Molecular Vision. 2006;12:649–654.
    1. Yarosh W., Monserrate J., Tong J.J., Tse S., Le P.K., Nguyen K. The molecular mechanisms of OPA1-mediated optic atrophy in Drosophila model and prospects for antioxidant treatment. Plos Genetics. 2008;4
    1. Yen M.Y., Kao S.H., Wang A.G., Wei Y.H. Increased 8-hydroxy-2′-deoxyguanosine in leukocyte DNA in Leber’s hereditary optic neuropathy. Investigative Ophthalmology & Visual Science. 2004;45:1688–1691.
    1. Yen M.Y., Wang A.G., Chang W.L., Hsu W.M., Liu J.H., Wei Y.H. Leber’s hereditary optic neuropathy-the spectrum of mitochondrial DNA mutations in Chinese patients. Japanese Journal of Ophthalmology. 2002;46:45–51.
    1. Yoshida M., Muneyuki E., Hisabori T. ATP synthase – a marvellous rotary engine of the cell. Nature Reviews Molecular Cell Biology. 2001;2:669–677.
    1. Yu-Wai-Man P., Bateman D.E., Hudson G., Griffiths P.G., Chinnery P.F. Leber hereditary optic neuropathy presenting in a 75-year-old man. Journal of Neuroophthalmology. 2008;28:155.
    1. Yu-Wai-Man P., Davies V.J., Piechota M.J., Cree L.M., Votruba M., Chinnery P.F. Secondary mtDNA defects do not cause optic nerve dysfunction in a mouse model of dominant optic atrophy. Investigative Ophthalmology & Visual Science. 2009;50:4561–4566.
    1. Yu-Wai-Man P., Griffiths P.G., Hudson G., Chinnery P.F. Inherited mitochondrial optic neuropathies. Journal of Medical Genetics. 2009;46:145–158.
    1. Yu-Wai-Man P., Griffiths P.G., Burke A., Sellar P.W., Clarke M.P., Gnanaraj L. The prevalence and natural history of dominant optic atrophy due to OPA1 mutations. Ophthalmology. 2010;117:1538.
    1. Yu-Wai-Man P., Griffiths P.G., Gorman G.S., Lourenco C.M., Wright A.F., Auer-Grumbach M. Multi-system neurological disease is common in patients with OPA1 mutations. Brain. 2010;133:771–786.
    1. Yu-Wai-Man, P., Shankar, S.P., Biousse, V., Miller, N.R., Bean, L.J.H., Coffee, B., et al., 2010c. Genetic screening for OPA1 and OPA3 mutations in patients with suspected inherited optic neuropathies. Ophthalmology, Epub ahead of print.
    1. Yu-Wai-Man P., Sitarz K.S., Samuels D.C., Griffiths P.G., Reeve A.K., Bindoff L.A. OPA1 mutations cause cytochrome c oxidase deficiency due to loss of wild-type mtDNA molecules. Human Molecular Genetics. 2010;19:3043–3052.
    1. Yu-Wai-Man P., Stewart J.D., Hudson G., Andrews R.M., Griffiths P.G., Birch M.K. OPA1 increases the risk of normal but not high tension glaucoma. Journal of Medical Genetics. 2010;47:120–125.
    1. Yu-Wai-Man, P., Trenell, M.I., Hollingsworth, K.G., Griffiths, P.G., Chinnery, P.F., 2010f. OPA1 mutations impair mitochondrial function in both pure and complicated dominant optic atrophy. Brain, Epub ahead of print.
    1. Yunis A.A. Chloramphenicol toxicity – 25 years of research. American Journal of Medicine. 1989;87:N44–N48.
    1. Yunis A.A., Smith U.S. Reversible bone marrow suppression from chloramphenicol – a consequence of mitochondrial injury. Archives of Internal Medicine. 1970;126:272–275.
    1. Zanna C., Ghelli A., Porcelli A.M., Carelli V., Martinuzzi A., Rugolo M. Apoptotic cell death of cybrid cells bearing Leber’s hereditary optic neuropathy mutations is caspase independent. Apoptosis: From Signaling Pathways to Therapeutic Tools. 2003;1010:213–217.
    1. Zanna C., Ghelli A., Porcelli A.M., Karbowski M., Youle R.J., Schimpf S. OPA1 mutations associated with dominant optic atrophy impair oxidative phosphorylation and mitochondrial fusion. Brain. 2008;131:352–367.
    1. Zaveri M.S., Conger A., Salter A., Frohman T.C., Galetta S.L., Markowitz C.E. Retinal imaging by laser polarimetry and optical coherence tomography evidence of axonal degeneration in multiple sclerosis. Archives of Neurology. 2008;65:924–928.
    1. Zeviani M. OPA1 mutations and mitochondrial DNA damage: keeping the magic circle in shape. Brain. 2008;131:314–317.
    1. Zhadanov S.I., Atamanov V.V., Zhadanov N.I., Oleinikov O.V., Osipova L.P., Schurr T.G. A novel mtDNA ND6 gene mutation associated with LHON in a Caucasian family. Biochemical and Biophysical Research Communications. 2005;332:1115–1121.
    1. Zhang A.M., Jia X., Zhang Q., Yao Y.G. No association between the SNPs (rs3749446 and rs1402000) in the PARL gene and LHON in Chinese patients with m.11778G>A. Human Genetics. 2010;128:465–468.
    1. Zhang X., Jones D., Gonzalez-Lima F. Mouse model of optic neuropathy caused by mitochondrial complex I dysfunction. Neuroscience Letters. 2002;326:97–100.
    1. Zhu G., Wu C.J., Zhao Y., Ashwell J.D. Optineurin negatively regulates TNF alpha-induced NF-kappa B activation by competing with NEMO for ubiquitinated RIP. Current Biology. 2007;17:1438–1443.
    1. Zuchner S., De Jonghe P., Jordanova A., Claeys K.G., Guergueltcheva V., Cherninkova S. Axonal neuropathy with optic atrophy is caused by mutations in mitofusin 2. Annals of Neurology. 2006;59:276–281.
    1. Zuchner S., Mersiyanova I.V., Muglia M., Bissar-Tadmouri N., Rochelle J., Dadali E.L. Mutations in the mitochondrial GTPase mitofusin 2 cause Charcot-Marie-Tooth neuropathy type 2A. Nature Genetics. 2004;36:449–451.
    1. Zuchner S., Vance J.M. Mechanisms of disease: a molecular genetic update on hereditary axonal neuropathies. Nature Clinical Practice Neurology. 2006;2:45–53.

Source: PubMed

3
Se inscrever