Kjer's optic neuropathy

Last updated
Kjer's optic neuropathy
Other namesAutosomal dominant optic atrophy, Kjer type; Kjer optic atrophy; or, Kjer's autosomal dominant optic atrophy.
Specialty Neurology   OOjs UI icon edit-ltr-progressive.svg

Dominant optic atrophy (DOA), or autosomal dominant optic atrophy (ADOA), (Kjer's type) is an autosomally inherited disease that affects the optic nerves, causing reduced visual acuity and blindness beginning in childhood. However, the disease can seem to re-present a second time with further vision loss due to the early onset of presbyopia symptoms (i.e., difficulty in viewing objects up close). [1] DOA is characterized as affecting neurons called retinal ganglion cells (RGCs). This condition is due to mitochondrial dysfunction mediating the death of optic nerve fibers. The RGCs axons form the optic nerve. Therefore, the disease can be considered of the central nervous system. [2] Dominant optic atrophy was first described clinically by Batten in 1896 and named Kjer’s optic neuropathy in 1959 after Danish ophthalmologist Poul Kjer, who studied 19 families with the disease. [3] Although dominant optic atrophy is the most common autosomally inherited optic neuropathy (i.e., disease of the optic nerves), it is often misdiagnosed.

Contents

Presentation

Autosomal dominant optic atrophy can present clinically as an isolated bilateral optic neuropathy (non-syndromic form) or rather as a complicated phenotype with extra-ocular signs (syndromic form). Dominant optic atrophy usually affects both eyes roughly symmetrically in a slowly progressive pattern of vision loss beginning in childhood and is hence a contributor to childhood blindness.

A Humphrey Visual Field (HVF) can detect where areas of impaired vision have occurred, which usually shows up as central, centrocaecal, or paracentral scotomas for DOA patients. [4] Another visual test—optical coherence tomography (OCT) analyzes the optic nerve head—shows other structural consequences of the disease as retinal nerve fiber layer (RNFL) and ganglion cell layer thinning. Photographs of the fundus shows the typical optic atrophy worse on the temporal side of the optic disc in the shape of a wedge. [5] Cupping of the optic disc was found in 89% of DOA patients in at least one of their eyes. [6]

With loss of the central visual fields, there is impairment of color vision in addition to loss of visual acuity varying from mild to severe, typically ranging from 6/6 (in meters, equivalent to 20/20, ft) to 6/60 (20/200, ft) with a median value of 6/36 (roughly equivalent to 20/125 ft), corrected vision. Vision loss may sometimes be more severe.

Characteristic changes of the fundus (eye) evident on examination includes optic atrophy. In some cases, this may include optic disc cupping similar in appearance to glaucoma of the optic disc. Because the onset of Dominant optic atrophy is insidious, symptoms are often not noticed by the patients in its early stages and are picked up by chance in routine school eye screenings. The first signs of DOA typically present between 6–10 years of age, though presentation at as early as 1 year of age has been reported. In some cases, Dominant optic atrophy may remain subclinical until early adulthood. While symptoms typically begin to present in childhood, adult patients (around the age of 35) commonly complain of new onset loss of vision at near.

The emergence of premature presbyopia occurs from DOA patients being accustomed to holding objects closer to their faces to read. As a result, Donder’s curve should not be used to prescribe them lenses to correct premature presbyopia. Instead of calculating based on individuals reading from about 16 inches from the face, DOA patients should be calculated at around 8 inches due to their shortened reading distance. [7]

Progression of dominant optic atrophy varies even within the same family. Some have mild cases with visual acuity stabilizing in adolescence, others have slowly but constantly progressing cases, and others still have sudden step-like decreases in visual acuity. Generally, the severity of the condition by adolescence reflects the overall level of visual function to be expected throughout most of the patient’s adult life (Votruba, 1998). Slow decline in acuity is known to occur in late middle age in some families.

In complicated cases of autosomal dominant optic atrophy, in addition to bilateral optic neuropathy, several other neurological signs of neurological involvement can rarely be observed: peripheral neuropathy, deafness, cerebellar ataxia, spastic paraparesis, myopathy. [8]

Genetics

Dominant optic atrophy is inherited in an autosomal dominant manner. That is, a heterozygous patient with the disease has a 50% chance of passing on the disease to each offspring, assuming his/her partner does not have the disease. Males and females are affected at the same rate. Although DOA has a high penetrance (98%), severity and progression of DOA are extremely variable even within the same family.

Pathophysiology

Vision loss in dominant optic atrophy is due to optic nerve fiber loss from mitochondria dysfunction. Dominant optic atrophy is associated with mutation of the OPA1 gene [9] found on chromosome 3, region q28-qter. Also, 5 other chromosomal genes are described as causing optic atrophy: OPA2 (x-linked), OPA3 (dominant), OPA4 (dominant), OPA5 (dominant) and OPA6 (recessive) (see OMIM 165500).

The OPA1 gene codes for a dynamin-related GTPase protein targeted to the mitochondrial inner membrane. OPA1 has distinct roles in the fusion of mitochondrial inner membranes during mitochondrial fusion events, and in regulation of cell death. [10]

Mitochondria are subcellular structures that generate and transform energy from metabolism into discrete usable units (ATP) for the cell’s functions (See oxidative phosphorylation, electron transport chain). Retinal ganglion cells (neurons) make up the optic nerve and have a long unmyelinated portion, hence the high energy demand and sensitivity to mitochondrial dysfunction. This is especially the case for smaller axons such as those found in the papillomacular bundle of the retina, which transmit information corresponding to the central visual field. The surface ratios of these smaller axons make them even more vulnerable to mitochondrial impairments. Biochemical and mitochondrial morphological studies on cells from patients affected by autosomal dominant optic atrophy have shown a severe defect in the shape (with a very remarkable fragmentation of the mitochondrial tubules in small spheres) and distribution of mitochondria, occurring independently from a bioenergetic defect (respiratory chain function, ATP synthesis, and reactive oxygen species production) or apoptosis, indicating that the mitochondrial fusion defect is the primary pathogenetic mechanism, [11] although variable bioenergetic defects can also occur as a secondary phenomenon, especially in severe cases with complicated phenotypes and accumulation of multiple mitochondrial-DNA deletions.

Over 200 different mutations of the OPA1 gene causing DOA have been reported, most of which occur in the catalytic domain of the protein.

Mutations at the OPA1 gene are also associated with normal tension glaucoma (OMIM 606657) and deafness (OMIM 125250).

Management

Currently, there are no curative therapies available for dominant optic atrophy. Some studies have found usage of idebenone to be associated with mild improvement in visual acuity for DOA patients with OPA1 mutation. [12] Idebenone works as an antioxidant by shuttling electrons affected by reduced ATP synthesis and defective oxidative phosphorylation in complex I directly to complex III. [13] Typically, idebenone is prescribed to be taken three times a day. [14]

Patients should be monitored for changes in vision by their eye-care professional. Children of patients should be screened regularly for visual changes related to dominant optic atrophy. Research is underway to further characterize the disease so that therapies may be developed.

Since November 2018, Cure ADOA Foundation has been focusing on fellow patients and their families. They have the following goals: scientific research, disease awareness, interaction between all parties involved and a trustworthy place for the patients.

Incidence

The incidence of dominant optic atrophy has been estimated to be 1:50,000 with prevalence as high as 1:10,000 in the Danish population (Votruba, 1998).

See also

Related Research Articles

<span class="mw-page-title-main">Optic nerve</span> Second cranial nerve, which connects the eyes to the brain

In neuroanatomy, the optic nerve, also known as the second cranial nerve, cranial nerve II, or simply CN II, is a paired cranial nerve that transmits visual information from the retina to the brain. In humans, the optic nerve is derived from optic stalks during the seventh week of development and is composed of retinal ganglion cell axons and glial cells; it extends from the optic disc to the optic chiasma and continues as the optic tract to the lateral geniculate nucleus, pretectal nuclei, and superior colliculus.

<span class="mw-page-title-main">Retinitis pigmentosa</span> Gradual retinal degeneration leading to progressive sight loss

Retinitis pigmentosa (RP) is a genetic disorder of the eyes that causes loss of vision. Symptoms include trouble seeing at night and decreasing peripheral vision. As peripheral vision worsens, people may experience "tunnel vision". Complete blindness is uncommon. Onset of symptoms is generally gradual and often begins in childhood.

<span class="mw-page-title-main">Leber's hereditary optic neuropathy</span> Mitochondrially inherited degeneration of retinal cells in human

Leber's hereditary optic neuropathy (LHON) is a mitochondrially inherited degeneration of retinal ganglion cells (RGCs) and their axons that leads to an acute or subacute loss of central vision; it predominantly affects young adult males. LHON is transmitted only through the mother, as it is primarily due to mutations in the mitochondrial genome, and only the egg contributes mitochondria to the embryo. Men cannot pass on the disease to their offspring. LHON is usually due to one of three pathogenic mitochondrial DNA (mtDNA) point mutations. These mutations are at nucleotide positions 11778 G to A, 3460 G to A and 14484 T to C, respectively in the ND4, ND1 and ND6 subunit genes of complex I of the oxidative phosphorylation chain in mitochondria.

Anterior ischemic optic neuropathy (AION) is a medical condition involving loss of vision caused by damage to the optic nerve as a result of insufficient blood supply (ischemia). This form of ischemic optic neuropathy is generally categorized as two types: arteritic AION, in which the loss of vision is the result of an inflammatory disease of arteries in the head called temporal arteritis, and non-arteritic AION, which is due to non-inflammatory disease of small blood vessels.

Posterior ischemic optic neuropathy (PION) is a medical condition characterized by damage to the retrobulbar portion of the optic nerve due to inadequate blood flow (ischemia) to the optic nerve. Despite the term posterior, this form of damage to the eye's optic nerve due to poor blood flow also includes cases where the cause of inadequate blood flow to the nerve is anterior, as the condition describes a particular mechanism of visual loss as much as the location of damage in the optic nerve. In contrast, anterior ischemic optic neuropathy (AION) is distinguished from PION by the fact that AION occurs spontaneously and on one side in affected individuals with predisposing anatomic or cardiovascular risk factors.

<span class="mw-page-title-main">Mitochondrial myopathy</span> Medical condition

Mitochondrial myopathies are types of myopathies associated with mitochondrial disease. Adenosine triphosphate (ATP), the chemical used to provide energy for the cell, cannot be produced sufficiently by oxidative phosphorylation when the mitochondrion is either damaged or missing necessary enzymes or transport proteins. With ATP production deficient in mitochondria, there is an over-reliance on anaerobic glycolysis which leads to lactic acidosis either at rest or exercise-induced.

<span class="mw-page-title-main">Behr syndrome</span> Medical condition

Behr syndrome is characterized by the association of early-onset optic atrophy with spinocerebellar degeneration resulting in ataxia, pyramidal signs, peripheral neuropathy and developmental delay.

<span class="mw-page-title-main">Ischemic optic neuropathy</span> Medical condition

Ischemic optic neuropathy (ION) is the loss of structure and function of a portion of the optic nerve due to obstruction of blood flow to the nerve. Ischemic forms of optic neuropathy are typically classified as either anterior ischemic optic neuropathy or posterior ischemic optic neuropathy according to the part of the optic nerve that is affected. People affected will often complain of a loss of visual acuity and a visual field, the latter of which is usually in the superior or inferior field.

Toxic and nutritional optic neuropathy is a group of medical disorders defined by visual impairment due to optic nerve damage secondary to a toxic substance and/or nutritional deficiency. The causes of these disorders are various, but they are linked by shared signs and symptoms, which this article will describe. In several of these disorders, both toxic and nutritional factors play a role, acting synergistically.

Optic neuropathy is damage to the optic nerve from any cause. The optic nerve is a bundle of millions of fibers in the retina that sends visual signals to the brain.

<span class="mw-page-title-main">Optic disc drusen</span> Medical condition

Optic disc drusen (ODD) are globules of mucoproteins and mucopolysaccharides that progressively calcify in the optic disc. They are thought to be the remnants of the axonal transport system of degenerated retinal ganglion cells. ODD have also been referred to as congenitally elevated or anomalous discs, pseudopapilledema, pseudoneuritis, buried disc drusen, and disc hyaline bodies.

<span class="mw-page-title-main">Costeff syndrome</span> Medical condition

Costeff syndrome, or 3-methylglutaconic aciduria type III, is a genetic disorder caused by mutations in the OPA3 gene. It is typically associated with the onset of visual deterioration in early childhood followed by the development of movement problems and motor disability in later childhood, occasionally along with mild cases of cognitive deficiency. The disorder is named after Hanan Costeff, the doctor who first described the syndrome in 1989.

<span class="mw-page-title-main">Dynamin-like 120 kDa protein</span> Protein-coding gene in the species Homo sapiens

Dynamin-like 120 kDa protein, mitochondrial is a protein that in humans is encoded by the OPA1 gene. This protein regulates mitochondrial fusion and cristae structure in the inner mitochondrial membrane (IMM) and contributes to ATP synthesis and apoptosis, and small, round mitochondria. Mutations in this gene have been implicated in dominant optic atrophy (DOA), leading to loss in vision, hearing, muscle contraction, and related dysfunctions.

Paul Kjer is a Danish ophthalmologist who studied a condition in nineteen families that was characterized by infantile optic atrophy along with a dominant inheritance mode. In 1959, the condition was named Kjer's optic neuropathy in his honor.

<span class="mw-page-title-main">Behçet's disease</span> Inflammatory disorder

Behçet's disease (BD) is a type of inflammatory disorder which affects multiple parts of the body. The most common symptoms include painful sores on the mucous membranes of the mouth and other parts of the body, inflammation of parts of the eye, and arthritis. The sores can last from a few days, up to a week or more. Less commonly there may be inflammation of the brain or spinal cord, blood clots, aneurysms, or blindness. Often, the symptoms come and go.

Mitochondrial optic neuropathies are a heterogenous group of disorders that present with visual disturbances resultant from mitochondrial dysfunction within the anatomy of the Retinal Ganglion Cells (RGC), optic nerve, optic chiasm, and optic tract. These disturbances are multifactorial, their aetiology consisting of metabolic and/or structural damage as a consequence of genetic mutations, environmental stressors, or both. The three most common neuro-ophthalmic abnormalities seen in mitochondrial disorders are bilateral optic neuropathy, ophthalmoplegia with ptosis, and pigmentary retinopathy.

Chronic relapsing inflammatory optic neuropathy (CRION) is a form of recurrent optic neuritis that is steroid responsive and dependent. Patients typically present with pain associated with visual loss. CRION is a clinical diagnosis of exclusion, and other demyelinating, autoimmune, and systemic causes should be ruled out. An accurate antibody test which became available commercially in 2017 has allowed most patients previously diagnosed with CRION to be re-identified as having MOG antibody disease, which is not a diagnosis of exclusion. Early recognition is crucial given risks for severe visual loss and because it is treatable with immunosuppressive treatment such as steroids or B-cell depleting therapy. Relapse that occurs after reducing or stopping steroids is a characteristic feature.

Occult macular dystrophy (OMD) is a rare inherited degradation of the retina, characterized by progressive loss of function in the most sensitive part of the central retina (macula), the location of the highest concentration of light-sensitive cells (photoreceptors) but presenting no visible abnormality. "Occult" refers to the degradation in the fundus being difficult to discern. The disorder is called "dystrophy" instead of "degradation" to distinguish its genetic origin from other causes, such as age. OMD was first reported by Y. Miyake et al. in 1989.

<span class="mw-page-title-main">Alfredo Sadun</span> American ophthalmologist

Alfredo Arrigo Sadun is an American ophthalmologist, academic, author and researcher. He holds the Flora L. Thornton Endowed Chair at Doheny Eye Centers-UCLA and is Vice-Chair of Ophthalmology at UCLA.

<span class="mw-page-title-main">Wolfram-like syndrome</span> Medical condition

Wolfram-like syndrome is a rare autosomal dominant genetic disorder that shares some of the features shown by those affected with the autosomal recessive Wolfram syndrome. It is a type of WFS1-related disorder.

References

  1. The retina and its disorders. San Diego, CA: Academic Press. 2011. ISBN   9780123821997.
  2. Lenaers, Guy; Hamel, Christian P; Delettre, Cecile; Amati-Bonneau, Patrizia; Procaccio, Vincent; Bonneau, Dominique; Reynier, Pascal; Milea, Dan (2012). "Dominant optic atrophy". Orphanet Journal of Rare Diseases. 7 (1): 46. doi: 10.1186/1750-1172-7-46 . PMC   3526509 . PMID   22776096.
  3. Kjer, P (1959). "Infantile optic atrophy with dominant mode of inheritance: a clinical and genetic study of 19 Danish families". Acta Ophthalmologica Supplementum. 164 (Supp 54): 1–147. PMID   13660776.
  4. Advances in Ophthalmology and Optometry. Amsterdam. 2019. ISBN   9780323712002.{{cite book}}: CS1 maint: location missing publisher (link)
  5. Sadun, Alfredo A.; Wang, Michelle Y. (2011). "Abnormalities of the optic disc". Neuro-ophthalmology. Handbook of Clinical Neurology. Vol. 102. pp. 117–157. doi:10.1016/B978-0-444-52903-9.00011-X. ISBN   9780444529039. PMID   21601065.
  6. Fournier, Annick V; Damji, Karim F; Epstein, David L; Pollock, Stephen C (September 2001). "Disc excavation in dominant optic atrophy". Ophthalmology. 108 (9): 1595–1602. doi:10.1016/S0161-6420(01)00696-0. PMID   11535456.
  7. Sadun, A.A.; Chicani, C.F. (2010). "Inherited Optic Neuropathies". Encyclopedia of the Eye: 387–391. doi:10.1016/B978-0-12-374203-2.00288-8. ISBN   9780123742032.
  8. Yu-Wai-Man, P; Griffiths, PG; Gorman, GS; Lourenco, CM; Wright, AF; Auer-Grumbach, M; Toscano, A; Musumeci, O; Valentino, ML; Caporali, L; Lamperti, C; Tallaksen, CM; Duffey, P; Miller, J; Whittaker, RG; Baker, MR; Jackson, MJ; Clarke, MP; Dhillon, B; Czermin, B; Stewart, JD; Hudson, G; Reynier, P; Bonneau, D; Marques, W Jr; Lenaers, G; McFarland, R; Taylor, RW; Turnbull, DM; Votruba, M; Zeviani, M; Carelli, V; Bindoff, LA; Horvath, R; Amati-Bonneau, P; Chinnery, PF (March 2010). "Multi-system neurological disease is common in patients with OPA1 mutations". Brain: A Journal of Neurology. 133 (Pt 3): 771–86. doi:10.1093/brain/awq007. PMC   2842512 . PMID   20157015.
  9. Delettre, C; Lenaers, G; Griffoin, JM; Gigarel, N; Lorenzo, C; Belenguer, P; Pelloquin, L; Grosgeorge, J; Turc-Carel, C; Perret, E; Astarie-Dequeker, C; Lasquellec, L; Arnaud, B; Ducommun, B; Kaplan, J; Hamel, CP (October 2000). "Nuclear gene OPA1, encoding a mitochondrial dynamin-related protein, is mutated in dominant optic atrophy". Nature Genetics. 26 (2): 207–10. doi:10.1038/79936. PMID   11017079. S2CID   24514847.
  10. Frezza, C; Cipolat, S; Martins de Brito, O; Micaroni, M; Beznoussenko, GV; Rudka, T; Bartoli, D; Polishuck, RS; Danial, NN; De Strooper, B; Scorrano, L (Jul 14, 2006). "OPA1 controls apoptotic cristae remodeling independently from mitochondrial fusion". Cell. 126 (1): 177–89. doi: 10.1016/j.cell.2006.06.025 . PMID   16839885.
  11. Spinazzi, M; Cazzola, S; Bortolozzi, M; Baracca, A; Loro, E; Casarin, A; Solaini, G; Sgarbi, G; Casalena, G; Cenacchi, G; Malena, A; Frezza, C; Carrara, F; Angelini, C; Scorrano, L; Salviati, L; Vergani, L (Nov 1, 2008). "A novel deletion in the GTPase domain of OPA1 causes defects in mitochondrial morphology and distribution, but not in function". Human Molecular Genetics. 17 (21): 3291–302. doi: 10.1093/hmg/ddn225 . PMID   18678599.
  12. Romagnoli, Martina; La Morgia, Chiara; Carbonelli, Michele; Di Vito, Lidia; Amore, Giulia; Zenesini, Corrado; Cascavilla, Maria Lucia; Barboni, Piero; Carelli, Valerio (April 2020). "Idebenone increases chance of stabilization/recovery of visual acuity in OPA1 ‐dominant optic atrophy". Annals of Clinical and Translational Neurology. 7 (4): 590–594. doi:10.1002/acn3.51026. hdl: 11585/794071 . PMC   7187718 . PMID   32243103.
  13. Giorgio, Valentina; Petronilli, Valeria; Ghelli, Anna; Carelli, Valerio; Rugolo, Michela; Lenaz, Giorgio; Bernardi, Paolo (February 2012). "The effects of idebenone on mitochondrial bioenergetics". Biochimica et Biophysica Acta (BBA) - Bioenergetics. 1817 (2): 363–369. doi:10.1016/j.bbabio.2011.10.012. PMC   3265671 . PMID   22086148.
  14. Romagnoli, Martina; La Morgia, Chiara; Carbonelli, Michele; Di Vito, Lidia; Amore, Giulia; Zenesini, Corrado; Cascavilla, Maria Lucia; Barboni, Piero; Carelli, Valerio (April 2020). "Idebenone increases chance of stabilization/recovery of visual acuity in OPA1 ‐dominant optic atrophy". Annals of Clinical and Translational Neurology. 7 (4): 590–594. doi:10.1002/acn3.51026. hdl: 11585/794071 . PMC   7187718 . PMID   32243103.

Further reading