This site uses tracking information. Visit our privacy policy. Click to agree to this policy and not see this again.

Ophthalmology and Visual Sciences

Fuchs’ Endothelial Corneal Dystrophy

Fuchs’ Endothelial Corneal Dystrophy

Matthew G. Field, MD, PhD; Aaron D. Dotson, MD; Arnulfo Garza Reyes, MD; Christopher R. Fortenbach, MD, PhD; Emily M. Witsberger, MD; Christopher S. Sales, MD; Mark A. Greiner, MD
August 8, 2023

INITIAL PRESENTATION

Chief Complaint: Worsening blurry vision in both eyes

History of Present Illness: A 77-year-old woman was referred for worsening vision. Following an uncomplicated cataract surgery, her vision initially improved. However, her vision gradually became blurry and distorted again over the next few months. She denied any acute changes such as pain, discomfort, irritation, redness, glare, dryness, or photophobia. She is pseudophakic in her right eye, has a cataract in the left eye, and has epiretinal membranes in both eyes.

Past Ocular History

  • Cataract extraction with intraocular lens implantation, right eye
  • Age-related nuclear cataract, left eye
  • High myopia, both eyes

Past Medical History

  • GERD

Systemic Medications

  • Calcium carbonate
  • Lansoprazole

Ocular Medications

  • Sodium chloride (Muro 128) 2% ophthalmic suspension, both eyes, 3-4 times daily

Allergies

  • Erythromycin

Family History

  • Mother - macular degeneration

Social History

  • Denies tobacco, alcohol, or recreational drug use

Review of Systems

  • As noted in HPI, otherwise negative


OCULAR EXAMINATION

  • Visual Acuity with correction
    • Right eye (OD): 20/30-1 (20/25-3 with pinhole)
    • Left eye (OS): 20/50-2 (20/30+2 with pinhole)
  • Ocular Motility
    • Both eyes (OU): Full
  • Intraocular Pressure (IOP)
    • OD: 21 mmHg by Tonopen
    • OS: 18 mmHg by Tonopen
  • Pachymetry
    • OD: 670 µm
    • OS: 667 µm
  • Pupils
    • OD: 4 mm in dark, 3 mm in light, no relative afferent pupillary defect (RAPD)
    • OS: 4 mm in dark, 3 mm in light, no RAPD
  • Confrontation visual fields
    • OU: Full
  • External
    • OU: Normal
  • Slit lamp exam
    Right eye Left eye
    Lids/Lashes Normal Normal
    Conunctiva/Sclera Clear and quiet Clear and quiet
    Cornea 2+ Descemet membrane folds, 4+ guttae, 1+ edema, anterior stromal haze temporal paracentral Trace Descemet membrane folds, 4+ guttae
    Anterior Chamber Deep and quiet Deep and quiet
    Iris Normal architecture Normal architecture
    Lens Posterior chamber intraocular lens, clear 2+ nuclear sclerosis, 1+ cortical cataract
    Vitreous Posterior vitreous detachment (PVD), no anterior vitreous cell (AVC) Normal, no AVC, no PVD
  • Slit lamp photographs of the right (A, C, E) and left (B, D, F) eye demonstrating diffuse central and peripheral guttae by retroillumination (A, B), corneal haze and edema with 2+ Descemet’s folds (C) and trace Descemet’s folds (D), and corneal edema with deposition of endothelial guttae (E, F).
    Figure 1: Slit lamp photographs of the right (A, C, E) and left (B, D, F) eye demonstrating diffuse central and peripheral guttae by retroillumination (A, B), corneal haze and edema with 2+ Descemet’s folds (C) and trace Descemet’s folds (D), and corneal edema with deposition of endothelial guttae (E, F).
  • Dilated fundus examination (DFE)
    Right eye Left eye
    Disc Normal, healthy rims Normal, healthy rims
    Cup-to-disc ratio 0.2 0.2
    Macula Mild epiretinal membrane Trace epiretinal membrane
    Vessels Normal Normal
    Periphery Normal Normal
  • Additional Testing

    Specular Microscopy:

  • Specular microscopy of the right (OD) and left eye (OS) on presentation.  Most adults over age 40 have an endothelial cell density between 2000-3000 cells/mm2 with a uniform, hexagonal endothelial monoloayer.  In this patient, the endothelial cell density is significantly reduced in both eyes. Confluent guttae are seen in both eyes with variability in endothelial cell size (polymegethism) and shape (pleomorphism).
    Figure 2: Specular microscopy of the right (OD) and left eye (OS) on presentation. Most adults over age 40 have an endothelial cell density between 2000-3000 cells/mm2 with a uniform, hexagonal endothelial monoloayer. In this patient, the endothelial cell density is significantly reduced in both eyes. Confluent guttae are seen in both eyes with variability in endothelial cell size (polymegethism) and shape (pleomorphism).

    Corneal Tomography:

    Scheimpflug tomography of the right eye on presentation (Oculus Pentacam), 4 Maps Refractive view. Imaging demonstrated (A) irregular contour; (B) increased corneal thickness, displacement of the thinnest point, and loss of isopach concentricity; and (C) focal areas of posterior depression, representing corneal edema.
    Figure 3: Scheimpflug tomography of the right eye on presentation (Oculus Pentacam), 4 Maps Refractive view. Imaging demonstrated (A) irregular contour; (B) increased corneal thickness, displacement of the thinnest point, and loss of isopach concentricity; and (C) focal areas of posterior depression, representing corneal edema.

    Differential Diagnosis


    DIAGNOSIS: Fuchs’ Endothelial Corneal Dystrophy (FECD)


    CLINICAL COURSE

    The patient was diagnosed with decompensated Fuchs’ endothelial corneal dystrophy with significant corneal edema in both eyes, worse in the right eye than the left. She had initially preferred medical management and had been on Muro 128 for two years with regular follow-up. Despite this, her vision had continued to worsen, and the disease had progressed to the point where the patient was ready to consider surgical options.

    The decision was made to proceed with a partial thickness corneal transplant, Descemet Membrane Endothelial Keratoplasty (DMEK), in her right eye. Three months following the procedure, her visual acuity in the right eye improved from 20/30-1 to 20/25+1 (with pinhole correction to 20/20-1). Her central corneal thickness also decreased from 670 µm to 557 µm, indicating improved corneal endothelial cell function. However, her endothelial function in the left eye continued to decompensate with visual acuity worsening to 20/200 (pinhole 20/70+2). The decision was made to first undergo cataract surgery followed by DMEK a month later. A week after her cataract extraction, her visual acuity in the left eye was 20/60-1. At her one-month post-operative appointment following DMEK, her best corrected visual acuity had improved to 20/30-2 (pinhole correction to 20/20-2). Her corneal thickness map and specular microscopy normalized, indicating a successful DMEK with improved endothelial function.

  • Additional Imaging (Pre-operative vs. Post-operative)

    Specular Microscopy:

    Specular microscopy of the right eye preoperatively (Left) and 3 months post-DMEK (Right). Preoperative specular microscopy shows confluence of the guttae and variability in corneal cell size (polymegathism) and shape (pleomorphism). In contrast, postoperative specular microscopy demonstrates a healthy, hexagonal endothelial cell morphology with improved endothelial cell count.
    Figure 4: Specular microscopy of the right eye preoperatively (Left) and 3 months post-DMEK (Right). Preoperative specular microscopy shows confluence of the guttae and variability in corneal cell size (polymegathism) and shape (pleomorphism). In contrast, postoperative specular microscopy demonstrates a healthy, hexagonal endothelial cell morphology with improved endothelial cell count.

    Corneal Tomography:

    . Corneal tomography of the right eye pre-DMEK (A-C) and post-DMEK (D-F).  Postoperative corneal tomography demonstrates normalization and improved contour of the corneal thickness plot (A,D) and anterior (B, E) and posterior (C,F) elevation maps.
    Figure 5: Corneal tomography of the right eye pre-DMEK (A-C) and post-DMEK (D-F). Postoperative corneal tomography demonstrates normalization and improved contour of the corneal thickness plot (A,D) and anterior (B, E) and posterior (C,F) elevation maps.

  • DISCUSSION

    Etiology/Epidemiology

    Fuchs’ endothelial corneal dystrophy (FECD) is the most common primary corneal endothelial dystrophy, affecting 4-7% of individuals over 40 years old [1,2]. FECD can be divided into two subtypes: early-onset and late-onset (Table 1). Early-onset FECD usually presents in the 2nd or 3rd decade of life and is highly associated with an autosomal dominant COL8A2 mutation [3,4]. Late-onset FECD has a greater predilection for females than males (3-4:1 ratio) and is associated with trinucleotide repeats in the intronic region of TCF4 or with mutations in SLC4A11 or ZEB1 [5]. TCF4 trinucleotide repeats have been reported occur in up to 70% of FECD cases and are believed to cause RNA toxicity through alteration of splicing machinery [5]. In addition to genetic predisposition, exogenous factors including smoking, diabetes, UV light, and hormones, specifically estrogen, have all been shown to increase the risk and progression of FECD [6].

    Early-Onset (Rare) Late-Onset (Common)
    Female: Male 1 : 1 3-4 : 1
    Onset 2nd or 3rd decade 5th or 6th decade
    Genetics
    • COL8A2 mutation [4]]
    • Autosomal dominant
    • 100% penetrance
    • TCF4 trinucleotide repeat [5]
    • SLC4A11 or ZEB1 mutation [5]]
    • Autosomal dominant
    • Variable penetrance
    Exogenous Risk Factors Smoking, diabetes, UV light, hormones [6]]

    Table 1: Characteristics of Early-Onset vs. Late-Onset FECD

    Pathophysiology

    The corneal endothelium provides nutritional support and appropriate hydration to the cornea through active regulation of various transport channels. In particular, the endothelium contains Na+/K+ ATPase pumps that transport water out of the corneal stroma into the aqueous humor in order to maintain transparency and prevent corneal edema [7]. In FECD, this endothelial layer becomes dysfunctional due to accumulations of extracellular matrix material that collect in guttae within Descemet membrane and extend posteriorly, disrupting the endothelium. Pathogenic genetic mutations and exogenous stressors promote FECD through improper protein function and misfolding, endoplasmic reticulum and oxidative stress, and mitochondrial dysfunction that lead to endothelial cell death and senescence and improper functioning of the transport channels [8].

    Signs & Symptoms

    Most patients with FECD are completely asymptomatic and are diagnosed during routine clinical examination through visualization of guttae on slit lamp examination. As the disease progresses, patients will complain about reduced vision and halos, worse in the morning upon waking. Diurnal fluctuation in visual acuity results from buildup of corneal edema while the eyelids are closed with symptomatic eyes have substantially thicker corneas than asymptomatic ones (9). In more advanced disease, patients will have decreased vision, photophobia, epiphora, and pain due to rupture of epithelial bullae leading to epithelial defects and ulcers.

    The modified Krachmer Grading Scale (Table 2) is routinely used in clinical practice to grade the severity of FECD and is largely based on the number and confluence of guttae visible on retroillumination [10]. Guttae are composed of spindle-shaped bundles of wide-spaced collagen, which give the cornea a beaten metal appearance with dark round areas on Descemet membrane that disrupt the endothelium [11]. Guttae first accumulate in the center of the cornea and spread peripherally as the disease progresses, increasing in number an average of 29% over a 30 month period [12].

    Grade Retroillumination or slit-lamp biomicroscopy appearance Symptoms Treatment
    Mild 1 ≤ 12 central nonconfluent guttae None Observation
    2 < 12 central nonconfluent guttae Mild reduction of vision
    • Observation
    • Topical 5% NaCl 4 times per day
    • NaCl ointment at night
    Moderate 3 1-2 mm central confluent guttae Reduced vision and halos in the morning
    4 2-5 mm central confluent guttae
    Severe 5 > 5 mm central confluent guttae without edema Reduction in vision, photophobia, epiphora, pain if rupture of bullae
    • Ruptured bullae should be treated as corneal abrasions
    • Surgery if pain or reduction in daily function
    6 >5 mm central confluent guttae with edema

    Table 2: The modified Krachmer Grading Scale for the clinical assessment of disease severity in FECD with associated signs, symptoms, and treatments for each grade [8,13]

    Additional testing

    Disease progression can be further assessed using specular or confocal microscopy to visualize endothelial cell dropout, accumulation of guttae, and polymegathism/pleomorphism of the remaining corneal endothelial cells. The average endothelial cell density for most adults over 40 is between 2000-3000 cells/mm [2].

    Scheimpflug tomography pachymetry and posterior elevation map patterns can be used to assess disease and predict progression. This can be especially helpful when there is not clinically appreciable edema present [13]. Patel et al. determined that findings predictive of progression in FECD include loss of isopach concentricity, displacement of the thinnest point of the cornea, and focal posterior surface depression. In isolation, central corneal thickness is not a helpful prognostic tool [14].

    Treatment & Management

    Initial management in asymptomatic individuals is often observation. In those with mild to moderate visual disability, medical management can include topical NaCl 5% drops up to 4 times per day and ointment at night to increase external osmolality, thereby dehydrating the corneal epithelium. In those with ruptured bullae, the epithelial defect should be treated as a corneal abrasion.

    With progression of disease and further visual disability, surgical treatment via corneal transplant should be considered. While penetrating keratoplasty (PK) surgery was historically the mainstay of treatment, endothelial keratoplasty with Descemet membrane endothelial keratoplasty (DMEK) or Descemet stripping automated endothelial keratoplasty (DSAEK) have surpassed this over the past two decades. Patients undergoing endothelial keratoplasty tend to have greater improvement of visual acuity, less postoperative astigmatism, faster visual recovery, and greater refractive stability in comparison to patients undergoing PK (15 ,16). DSAEK and DMEK also have superior graft survival with 96.2% and 98.7%, respectively, compared to a 73.5% survival rate of PK grafts [17] and do not require as potent of immunosuppressive treatment. A more detailed description of DSAEK and DMEK can be found here.

    Treatment alternatives to corneal transplantation are actively being investigated. Descemet stripping without endothelial keratoplasty, otherwise known as “Descemet Stripping Only” (DSO) has shown promising outcomes in select patient groups. In DSO, the central Descemet membrane and corneal endothelium are surgically removed. Postoperatively, peripheral endothelial cells migrate to the region that was originally stripped, creating a new barrier which helps to treat and prevent edema [18]. Disadvantages of DSO include longer visual recovery compared to EK, need for healthy peripheral endothelial cells, and lack of long-term reported outcomes. Other studies have shown encouraging data demonstrating accelerated endothelial cell migration following DSO with the use of topical rho-associated coiled-coil-containing protein kinase (ROCK) inhibitors, like ripasudil [19 ,20].

    Injection of donor cultured endothelial and pluripotent/multipotent stem cells is also actively being studied via human clinical trials. Gene therapy is another potential avenue for treating FECD, and it is actively being investigated via the CRISPR/Cas9 system. Pharmacologic intervention for FECD is also on the rise to slow FECD progression and reduce the need for surgery, with several molecules under investigation in preclinical and early phase clinical trials [21 ,22]. While various treatment modalities for FECD currently exist, further investigation may provide better outcomes, safety protocols for patients, and better overall access to care.

    ETIOLOGY/EPIDEMIOLOGY

    • Affects 4-7% of individuals over age 40
    • Early Onset - F:M ratio is 1:1. Occurs in 2nd or 3rd decade of life. Autosomal Dominant. 100% penetrance.
    • Late Onset - F:M ratio is 3-4:1. Occurs in 5th to 6th decade of life. Autosomal Dominant. Variable penetrance.

    SIGNS

    • Corneal guttae, stromal edema, Descemet’s folds, bullae, and scarring
    • Increasing pachymetry
    • Low endothelial cell counts on specular microscopy
    • Worsening corneal thickness plot and irregular anterior and posterior elevation maps on corneal tomography

    SYMPTOMS

    • Glare, blurred vision
    • Diurnal fluctuation in vision (typically worse in the morning)
    • Pain with severe corneal decompensation and bullae

    TREATMENT/MANAGEMENT

    • Topical NaCl 5% (Muro 128) drops and ointment at night to help dehydrate the cornea.
    • Ruptured bullae should be treated as a corneal abrasion.
    • Caution with intraocular surgeries that may injure the corneal endothelium, e.g. cataract surgery.
    • Partial-thickness corneal transplant with DMEK/DSEK when surgery indicated.
References
  1. Eghrari AO, Gottsch JD. Fuchs' corneal dystrophy. Expert Rev Ophthalmol 2010;5(2):147-159. [PMID 20625449]
  2. Vedana G, Villarreal G, Jr., Jun AS. Fuchs endothelial corneal dystrophy: current perspectives. Clin Ophthalmol 2016;10:321-330. [PMID 26937169]
  3. Stipanitzova H, Gerinec A. [Corneal dystrophies]. Cesk Slov Oftalmol 2003;59(5):359-367. [PMID 14518365]
  4. Biswas S, Munier FL, Yardley J, Hart-Holden N, Perveen R, Cousin P, Sutphin JE, Noble B, Batterbury M, Kielty C, Hackett A, Bonshek R, Ridgway A, McLeod D, Sheffield VC, Stone EM, Schorderet DF, Black GC. Missense mutations in COL8A2, the gene encoding the alpha2 chain of type VIII collagen, cause two forms of corneal endothelial dystrophy. Hum Mol Genet 2001;10(21):2415-2423. [PMID 11689488]
  5. Fautsch MP, Wieben ED, Baratz KH, Bhattacharyya N, Sadan AN, Hafford-Tear NJ, Tuft SJ, Davidson AE. TCF4-mediated Fuchs endothelial corneal dystrophy: Insights into a common trinucleotide repeat-associated disease. Prog Retin Eye Res 2021;81:100883. [PMID 32735996]
  6. Zhang X, Igo RP, Jr., Fondran J, Mootha VV, Oliva M, Hammersmith K, Sugar A, Lass JH, Iyengar SK, Fuchs' Genetics Multi-Center Study G. Association of smoking and other risk factors with Fuchs' endothelial corneal dystrophy severity and corneal thickness. Invest Ophthalmol Vis Sci 2013;54(8):5829-5835. [PMID 23882692]
  7. Bennett DB, Taylor ZD, Tewari P, Singh RS, Culjat MO, Grundfest WS, Sassoon DJ, Johnson RD, Hubschman JP, Brown ER. Terahertz sensing in corneal tissues. J Biomed Opt 2011;16(5):057003. [PMID 21639581]
  8. Ong Tone S, Kocaba V, Bohm M, Wylegala A, White TL, Jurkunas UV. Fuchs endothelial corneal dystrophy: The vicious cycle of Fuchs pathogenesis. Prog Retin Eye Res 2021;80:100863. [PMID 32438095]
  9. Fritz M, Grewing V, Maier P, Lapp T, Bohringer D, Reinhard T, Wacker K. Diurnal Variation in Corneal Edema in Fuchs Endothelial Corneal Dystrophy. Am J Ophthalmol 2019;207:351-355. [PMID 31415734]
  10. Matthaei M, Hribek A, Clahsen T, Bachmann B, Cursiefen C, Jun AS. Fuchs Endothelial Corneal Dystrophy: Clinical, Genetic, Pathophysiologic, and Therapeutic Aspects. Annu Rev Vis Sci 2019;5:151-175. [PMID 31525145]
  11. Adamis AP, Filatov V, Tripathi BJ, Tripathi RC. Fuchs' endothelial dystrophy of the cornea. Surv Ophthalmol 1993;38(2):149-168. [PMID 8235998]
  12. Iliff BW, Riazuddin SA, Gottsch JD. The genetics of Fuchs' corneal dystrophy. Expert Rev Ophthalmol 2012;7(4):363-375. [PMID 23585771]
  13. Sun SY, Wacker K, Baratz KH, Patel SV. Determining Subclinical Edema in Fuchs Endothelial Corneal Dystrophy: Revised Classification using Scheimpflug Tomography for Preoperative Assessment. Ophthalmology 2019;126(2):195-204. [PMID 30153944]
  14. Patel SV, Hodge DO, Treichel EJ, Spiegel MR, Baratz KH. Predicting the Prognosis of Fuchs Endothelial Corneal Dystrophy by Using Scheimpflug Tomography. Ophthalmology 2020;127(3):315-323. [PMID 31685256]
  15. Nanavaty MA, Wang X, Shortt AJ. Endothelial keratoplasty versus penetrating keratoplasty for Fuchs endothelial dystrophy. Cochrane Database Syst Rev 2014;2(2):CD008420. [PMID 24526345]
  16. Trousdale ER, Hodge DO, Baratz KH, Maguire LJ, Bourne WM, Patel SV. Vision-related quality of life before and after keratoplasty for Fuchs' endothelial dystrophy. Ophthalmology 2014;121(11):2147-2152. [PMID 25015214]
  17. Woo JH, Ang M, Htoon HM, Tan D. Descemet Membrane Endothelial Keratoplasty Versus Descemet Stripping Automated Endothelial Keratoplasty and Penetrating Keratoplasty. Am J Ophthalmol 2019;207:288-303. [PMID 31228467]
  18. Borkar DS, Veldman P, Colby KA. Treatment of Fuchs Endothelial Dystrophy by Descemet Stripping Without Endothelial Keratoplasty. Cornea 2016;35(10):1267-1273. [PMID 27310885]
  19. Macsai MS, Shiloach M. Use of Topical Rho Kinase Inhibitors in the Treatment of Fuchs Dystrophy After Descemet Stripping Only. Cornea 2019;38(5):529-534. [PMID 30720541]
  20. Moloney G, Petsoglou C, Ball M, Kerdraon Y, Hollhumer R, Spiteri N, Beheregaray S, Hampson J, D'Souza M, Devasahayam RN. Descemetorhexis Without Grafting for Fuchs Endothelial Dystrophy-Supplementation With Topical Ripasudil. Cornea 2017;36(6):642-648. [PMID 28476048]
  21. Kim EC, Toyono T, Berlinicke CA, Zack DJ, Jurkunas U, Usui T, Jun AS. Screening and Characterization of Drugs That Protect Corneal Endothelial Cells Against Unfolded Protein Response and Oxidative Stress. Invest Ophthalmol Vis Sci 2017;58(2):892-900. [PMID 28159976]
  22. Saha S, Skeie JM, Schmidt GA, Eggleston T, Shevalye H, Sales CS, Phruttiwanichakun P, Field M, Rinkoski TA, Fautsch MP, Baratz KH, Roy M, Jun AS, Salem AK, Greiner MA. TCF4 trinucleotide repeat expansions and UV irradiation are associated with ferroptosis susceptibility in Fuchs endothelial corneal dystrophy. bioRxiv 2022;10.1101/2022.06.27.497862:2022.2006.2027.497862
Suggested citation format:

Field MG, Garza Reyes G, Dotson AD, Witsberger EM, Sales CS, Greiner MA. Fuchs’ Endothelial Corneal Dystrophy. EyeRounds.org. Posted August 7, 2023; Available from https://EyeRounds.org/cases/343-fuchs-endothelial-dystrophy.htm


last updated: 08/08/2023

Image Permissions:
Creative Commons

Ophthalmic Atlas Images by EyeRounds.org, The University of Iowa are licensed under a Creative Commons Attribution-NonCommercial-NoDerivs 3.0 Unported License.