Discover millions of ebooks, audiobooks, and so much more with a free trial

Only $11.99/month after trial. Cancel anytime.

Essentials of Veterinary Ophthalmology
Essentials of Veterinary Ophthalmology
Essentials of Veterinary Ophthalmology
Ebook2,576 pages22 hours

Essentials of Veterinary Ophthalmology

Rating: 0 out of 5 stars

()

Read preview

About this ebook

Essentials of Veterinary Ophthalmology, Third Edition offers an updated introduction to the diagnosis and clinical management of ocular conditions in veterinary patients, providing trusted information in a user-friendly format.  The content of the book is distilled from the fifth edition of the gold-standard reference Veterinary Ophthalmology, emphasizing the information most relevant to veterinary students and general practitioners.  Fully updated throughout, the Third Edition focuses more strongly on small animals and horses, with streamlined coverage of other species, and new chapters have been added on morphology, physiology, and pharmacology.

Carefully designed to be equally useful for learning and in practice, the book offers a streamlined, practical approach, with bolded terms to enhance comprehension.  High-quality color photographs provide visual depictions of the conditions discussed.  Essentials of Veterinary Ophthalmology, Third Edition is an indispensable resource for veterinary students or clinicians wishing to hone their ophthalmology knowledge and skills.

LanguageEnglish
PublisherWiley
Release dateAug 6, 2014
ISBN9781118771877
Essentials of Veterinary Ophthalmology

Related to Essentials of Veterinary Ophthalmology

Related ebooks

Medical For You

View More

Related articles

Reviews for Essentials of Veterinary Ophthalmology

Rating: 0 out of 5 stars
0 ratings

0 ratings0 reviews

What did you think?

Tap to rate

Review must be at least 10 words

    Book preview

    Essentials of Veterinary Ophthalmology - Kirk N. Gelatt

    Table of Contents

    Title page

    Copyright page

    Preface

    Acknowledgments

    About the Companion Website

    Section 1: Basics for Clinical Veterinary Ophthalmology

    Chapter 1: Development of the Eye

    Gastrulation and Neurulation

    Formation of the Optic Vesicle and Optic Cup

    Lens Formation

    Vascular Development

    Development of the Cornea and Anterior Chamber

    Development of the Iris, Ciliary Body, and Iridocorneal Angle

    Retina and Optic Nerve Development

    Sclera, Choroid, and Tapetum

    Vitreous

    Optic Nerve

    Eyelids

    Extraocular Muscles

    Chapter 2: Ophthalmic Structures

    Adnexa: Protective Apparatus

    Upper and Lower Eyelids

    Conjunctivae

    Third Eyelid: Nictitating Membrane

    Lacrimal and Nasolacrimal System

    Globe

    Cornea

    Sclera

    Uvea

    Lens

    Vitreous

    Retina

    Optic Nerve

    Vasculature of the Eye and Orbit

    Chapter 3: Physiology of the Eye

    Anterior Segment of the Eye

    Tear Production and Drainage

    Cornea

    Drug Transport Through the Cornea, Limbus, Bulbar Conjunctiva, and Sclera

    Iris and Pupil

    Nutrition of Intraocular Tissues

    Ocular Circulation

    Ocular Barriers

    Aqueous Humor and Intraocular Pressure

    Lens

    Vitreous

    Ocular Mobility

    Chapter 4: Optics and Physiology of Vision

    Light and Vision

    Refractive Structures of the Eye

    Visual Processing: From Photoreceptors to Cortex

    Visual Perception

    Chapter 5: Ocular Pharmacology and Therapeutics

    Drug Delivery and Pharmacokinetics

    Barriers to Ocular Drug Delivery

    Topical Administration

    Drug Disposition after Eye Drop Application

    Systemic Absorption

    Topical Drug Delivery to the Posterior Eye Segment

    Drug Delivery Kinetics and Ocular Bioavailability

    Periocular Administration

    Intraocular Administration

    Sustained Drug Delivery to Intraocular Tissues

    Systemic Administration

    Other Methods of Ocular Drug Delivery

    Antimicrobial Agents

    Antibacterial Agents

    Antifungal Agents

    Antiviral Agents

    Anti-Inflammatory and Immunosuppressant Drugs

    Anti-Inflammatory Agents

    Nonsteroidal Anti-Inflammatory Drugs

    Immunosuppressant Drugs

    Mydriatics/Cycloplegics, Anesthetics, and Tear Substitutes and Stimulators

    Mydriatics/Cycloplegics

    Local Anesthetics

    Tear Substitutes and Stimulators

    Drugs That Affect Aqueous Humor Dynamics and Intraocular Pressure

    Parasympathomimetics (or Cholinomimetics)

    Drugs Acting on Adrenoceptors

    Carbonic Anhydrase Inhibitors

    Prostaglandin Analogues

    Osmotic Agents

    Section 2: Ophthalmic Examination and Imaging

    Chapter 6: Eye Examination and Diagnostics

    History

    General (Basic and Advanced) Ocular Examination

    Restraint

    Eyelid Akinesia

    Regional Anesthesia/Analgesia

    Ophthalmic Examination in Ambient Lighting

    Tear Tests

    Close Examination of the Adnexa and Globe

    Intraocular Pressure Measurement and Pupil Dilation

    Anterior Segment Examination after Pupil Dilation: Lens Examination

    Posterior Segment Examination

    Ophthalmic Diagnostic Procedures

    Basic Ophthalmic Diagnostics

    Advanced Ophthalmic Diagnostics

    Ocular Imaging: Basic and Advanced Diagnostics

    Basic Imaging Systems

    Advanced Imaging Systems

    Ophthalmic Imaging by Ultrasonography

    Physics and Basic Principles

    Examination Technique

    Standard (10–12 MHz) Ultrasonographic Imaging

    Advanced Ultrasound Imaging (20–100 MHz)

    Color Doppler Ultrasound

    Electrodiagnostic Evaluation of Vision

    Flash Electroretinogram

    Equipment for Recording and Light Stimulation

    Patient Preparation

    Anesthetic Protocol

    ERG Results

    Section 3: Canine Ophthalmology

    Chapter 7: Canine Orbit: Diseases and Surgery

    Clinical Signs/Examination

    Ancillary Diagnostic Tests

    Congenital Anomalies of the Orbit and Globe

    Exophthalmos

    Acquired Orbital Diseases

    Surgery of the Globe and the Orbit

    Chapter 8: Canine Eyelids: Diseases And Surgery

    Structure and Function

    Diagnostic Tests for Eyelids

    Principles of Eyelid Surgery

    Congenital and Presumed Heredity Structural Abnormalities

    Entropion

    Lid Trauma

    Inflammations

    Other Eyelid Diseases

    Reconstructive Blepharoplasty

    Other Eyelid Procedures

    Chapter 9: Canine Nasolacrimal Duct and Lacrimal Secretory Systems: Diseases and Surgery

    Nasolacrimal Duct System

    Embryology

    Anatomy

    Physiology

    Clinical Manifestations of Nasolacrimal Disease

    Diagnostic Procedures

    Congenital Diseases

    Developmental Disorders

    Acquired Diseases

    Lacrimal Secretory System

    Formation and Dynamics of Tear Components

    Pathogenesis of Tear Film Disease

    Tear Deficiency

    Qualitative Abnormalities

    Treatment

    Cysts and Neoplasms

    Chapter 10: Canine Conjunctiva: Diseases and Surgery

    Conjunctiva

    Functional Anatomy and Physiology

    Normal Bacterial and Fungal Flora

    Conjunctival Cytology

    General Response to Disease

    Infectious Conjunctivitis

    Noninfectious Conjunctivitis

    Follicular Conjunctivitis

    Conjunctivitis Associated with Tear Deficiencies

    Ligneous Conjunctivitis

    Conjunctival Neoplasia

    Non-neoplastic Conjunctival Masses

    Conjunctival Hemorrhages

    Foreign Bodies

    Orbital Disease

    Anatomic Abnormalities

    Conjunctival Manifestations of Systemic Disease

    Effects of Radiation

    Surgical Procedures

    Nictitating Membrane

    Anatomy, Histology, and Function

    Anomalous, Congenital, and Developmental Disorders

    Neoplasia

    Inflammatory Conditions

    Trauma, Reconstruction, and Foreign Bodies

    Nictitating Membrane Surgery

    Chapter 11: Canine Cornea: Diseases and Surgery

    Cornea

    Anatomy

    Corneal Clarity

    Corneal Wound Healing

    Corneal Pigmentation

    Corneal Edema

    Corneal Vascularization

    Developmental Abnormalities and Congenital Diseases

    Inflammatory Keratopathies

    Descemetoceles and Corneal Perforations

    Full-Thickness Corneal Lacerations

    Nonulcerative Keratitis

    Noninflammatory Keratopathies

    Corneoscleral Masses and Neoplasms

    Scleral Diseases

    Episcleritis

    Nodular Granulomatous Episcleritis

    Chapter 12: Canine Glaucomas

    Epidemiology of the Glaucomas in the Dog versus Man

    Classification of the Glaucomas

    Diagnostics Tests

    Provocative Tests

    Clinical and Pathologic Effects of Elevated Intraocular Pressure

    Primary and Breed-Predisposed Canine Glaucomas

    Secondary Glaucomas

    Congenital Glaucomas

    Medical and Surgical Treatment of the Canine Glaucomas

    Chapter 13: Canine Anterior Uvea: Diseases and Surgery

    Developmental Conditions

    Degenerative Iridal Changes

    Uveal Inflammation

    Uveal Manifestations of Selected Diseases

    Ocular Manifestations of Miscellaneous Diseases

    Uveal Trauma

    Hyphema

    Non-Neoplastic Iridal Proliferations

    Anterior Uveal Tumors

    Uveal Surgery

    Chapter 14: Canine Lens: Cataract, Luxation, and Surgery

    Congenital and Developmental Abnormalities

    Cataract Formation

    Classification of Canine Cataract

    Nuclear or Lenticular Sclerosis

    Clinical and Biomicroscopic Features of Canine Cataracts

    Cataracts with a Heritable or Presumed Heritable Basis

    Cataracts Associated with Systemic Diseases

    Cataracts Associated with Medications or Other Toxic Substances

    Dietary Deficiencies

    Injury to the Lens

    Age-Related Cataracts

    Cataracts Resulting from Inflammations and Lens-Associated Inflammation

    Nonsurgical Clinical Considerations for Canine Cataracts

    Medical Treatment of Cataracts

    Dislocation of the Crystalline Lens

    Cataract Surgery

    Chapter 15: Canine Posterior Segment: Diseases and Surgery

    Diseases of the Vitreous

    Development and Anatomy

    Morphology

    Diagnostic Procedures

    Therapeutic Procedures

    Vitreal Diseases

    Vitreous in Relation to Other Ophthalmic Disorders

    Diseases of the Canine Ocular Fundus

    Methods of Examination

    Structural Visualization of the Fundus

    Functional Testing of the Retina

    Normal Canine Ocular Fundus

    Developmental Anomalies

    Inherited Retinal Degenerations/Dystrophies

    Late-Onset Photoreceptor Degenerations

    Other Generalized Retinopathies/Retinal Dystrophies

    Retinal Pigment Epithelial Autofluorescent Inclusion Epitheliopathy/Retinal Pigment Epithelial Dystrophy

    Inflammation and Infections Affecting the Ocular Fundus

    Retinal Toxicities

    Retinopathies of Nutritional Causes

    Vascular Disease Processes

    Retinopathies with Immunologic Diseases

    Secondary Retinal Degenerations

    Retinal Detachments: Medical Considerations

    Neoplastic and Proliferative Conditions

    Surgery of the Canine Posterior Segment

    Anatomic Considerations for Vitreoretinal Surgeries

    Retinal Detachment Treated by Surgery

    Surgical Procedures for Treatment of Retinal Detachments

    Gene Therapy

    Diseases of the Canine Optic Nerve

    Intraocular Optic Nerve

    Vascular Supply

    Clinical Examination

    Diagnostics for the Optic Nerve

    Electroretinography

    Acquired Optic Nerve Diseases

    Section 4: Special Species

    Chapter 16: Feline Ophthalmology

    Diseases of the Eyelids

    Diseases of the Nasolacrimal and Tear Systems

    Diseases of the Third Eyelid

    Diseases of the Conjunctiva

    Normal Cornea

    Diseases of the Cornea

    Diseases of the Anterior Uvea

    Glaucoma

    Diseases of the Lens and Cataract Formation

    Diseases of the Posterior Segment

    Diseases of the Optic Nerve and Central Nervous System

    Diseases of the Orbit

    Chapter 17: Equine Ophthalmology

    Ocular Examination

    Special Ophthalmic Considerations in Specific Groups of Horses

    Diseases of the Orbit

    Diseases of the Adnexa

    Diseases of the Cornea

    Diseases of the Uvea

    Glaucoma

    Diseases of the Lens

    Diseases of the Posterior Segment

    Diseases of the Optic Nerve

    Chapter 18: Food Animal Ophthalmology

    Cattle

    Orbit

    Eyelids

    Nasolacrimal System

    Conjunctiva and Cornea

    Uveal Tract

    Lens

    Ocular Fundus

    Sheep and Goats

    Eyelids

    Conjunctiva and Cornea

    Uveal Tract

    Lens

    Ocular Fundus

    Pigs

    Orbit and Globe

    Eyelids

    Conjunctiva and Cornea

    Uveal Tract

    Lens

    Ocular Fundus

    Chapter 19: Exotic Animals: Ophthalmic Diseases and Surgery

    Camelids

    Examination Techniques

    Ocular Medications

    Ocular Anatomy

    Ocular Diseases

    Laboratory Animals

    Examination Techniques

    Lacrimal Gland Biology

    Retinal Vasculature

    Ocular Diseases

    Rabbits

    Ocular Anatomy

    Intraocular Pressure and Tear Measurements

    Ocular Diseases

    Exotic Animals

    Examination Techniques

    Fish

    Amphibians

    Reptiles

    Birds

    Mammals

    Marine and Other Aquatic Mammals

    Section 5: Ophthalmic and Systemic Diseases

    Chapter 20: Comparative Neuro-Ophthalmology

    Neuro-Ophthalmic Examination

    Neuroanatomic Lesion Localization and Neuro-Ophthalmic Syndromes

    Neuro-Ophthalmic Diseases

    Chapter 21: Systemic Disease and the Eye

    Dogs

    Congenital Diseases

    Developmental Diseases

    Acquired Diseases

    Cats

    Congenital Diseases

    Developmental Diseases

    Acquired Diseases

    Horses

    Congenital Diseases

    Developmental Diseases

    Acquired Diseases

    Food Animals

    Congenital Diseases

    Developmental Disorders

    Acquired Diseases

    Appendix A: Adrenergics in Veterinary Ophthalmology

    Appendix B: Artificial Tear Substitutes for Veterinary Ophthalmology

    Appendix C: Topical and Local/Injectable Anesthetics for Veterinary Ophthalmology

    Topical

    Local/Injectable

    Appendix D: Topical and Subconjunctival Antibiotics for Veterinary Ophthalmology

    A.  Available Topical Antibiotics

    B.  Selection of Initial Antibiotics on the Basis of Smear Morphology

    C.  Development of Bacterial Resistance

    D.  Suggested Dosages of Subconjunctival Antibiotics

    Appendix E: Antiviral Drugs for Veterinary Ophthalmology (To Treat FHV-1 Ocular Infections)

    Appendix F: Antifungals for Veterinary Ophthalmology

    A.  Antifungals

    B.  Selected Topical Antifungals Agents for Equine Fungal Keratitis/Fungal Abscesses

    Appendix G: Carbonic Anhydrase Inhibitors (CAIs) for the Dog and Cat

    Systemic (mg/kg): Single-Dose Studies in Dogs

    Topical: 2–3× Daily

    Recommended Clinical Multidoses

    Appendix H: Corticosteroids in Veterinary Ophthalmology

    A.  Topical Corticosteroids

    B.  Depot Glucocorticoids for Subconjunctival Administration

    Appendix I: Nonsteroidal Anti-inflammatory Drugs (NSAIDS) in Veterinary Ophthalmology

    Appendix J: Hyperosmotics for Veterinary Ophthalmology

    Topical

    Systemic

    Appendix K: Miotics in the Dog and Cat

    Appendix L: Mydriatics or Pupil-Dilating Agents for the Dog

    Appendix M: Mydriatics or Pupil-Dilating Agents for the Cat

    Appendix N: Mydriatics or Pupil-Dilating Agents for the Horse

    Appendix O: Mydriatics or Pupil-Dilating Agents for the Cow

    Appendix P: Available Pupil Dilating Agents for Selected Birds

    Appendix Q: Topical Prostaglandins for the Dog

    Appendix R: Other Drugs for Veterinary Ophthalmology

    Appendix S: DNA Tests for Feline and Canine Eye Diseases

    Appendix T: Inherited Eye Diseases in the Dog

    Appendix U: Inherited Eye Diseases in the Cat

    Appendix V: Inherited Eye Diseases in the Horse

    Appendix W: Inherited Eye Diseases in Food Animals

    Appendix X: Lysosomal Storage Diseases in the Dog, Cat, and Food Animals

    Glossary

    Common Ophthalmic Roots

    Common Words

    Index

    End User License Agreement

    List of Tables

    Table 1.1.  Sequence of Ocular Development in the Human, Mouse, and Dog

    Table 1.2.  Sequence of Ocular Development in the Cow

    Table 1.3.  Embryonic Origins of Ocular Tissues

    Table 2.1.  Ratios of the Dimensions of the Globes of Domestic Species

    Table 2.2.  Width and Height (mm) of the Cornea Measured in a Straight Line

    Table 3.1.  Eyelid Reflexes

    Table 3.2.  Adrenergic Receptors in the Iris and Ciliary Body

    Table 3.3.  Estimates of Aqueous Humor Dynamics in Selected Species

    Table 3.4.  Intraocular Pressures (IOP) in Selected Animal Species

    Table 4.1.  Refractive Values in Selected Animal Species

    Table 4.2.  Snellen Acuity in Selected Animal Species

    Table 5.1.  Recommended Ophthalmic Antibiotic Choices Based on In Vitro Susceptibility of Organisms Isolated from Clinical Bacterial Keratitis Cases. Antibiotic (# Resistant/Total # Isolates Evaluated, % Resistant in Reference Indicated). Patient eyes vary for each antibiotic tested

    Table 5.2.  Selected Antifungal Medications Used to Treat Keratomycosis

    Table 5.3.  Summary of Selected Topically Administered Antiviral Drugs for Treatment of Feline Herpesvirus

    Table 5.4.  Commercially Available Corticosteroid Agents for Subconjunctival Injection

    Table 5.5.  Reported Activities of Commonly Utilized Mydratics in Dog, Cats and Horses

    Table 6.1.  Fundus Magnification with Direct Ophthalmoscopy in Animalsa

    Table 6.2.  Light Beam Selections for Direct Ophthalmoscopy

    Table 6.3.  Topical Stains for Veterinary Ophthalmology

    Table 6.4.  Normal Intraocular Pressure (IOP) Reported in Animals

    Table 7.1.  Differential Diagnoses for Exophthalmos in the Dog

    Table 7.2.  Recommended Treatment and Prognosis for Traumatic Proptosis in the Dog

    Table 8.1.  Methods to Treat Distichiasis in the Dog

    Table 8.2.  Site of Entropion in Selected Breeds

    Table 8.3.  Surgical Procedures for Canine Entropion

    Table 8.4.  Histogenic Classification and Frequency of Eyelid Tumors in Dogs

    Table 10.1.  Summary of Cytology in Conjunctivitis

    Table 11.1.  Dynamics of Corneal Healing after Corneal Ulceration

    Table 11.2.  Types of Corneal Ulcerations in the Dog

    Table 11.3.  Antiproteolytic Agents for Topical Treatment of Melting Ulcers

    Table 11.4.  Breed Predisposition to Chronic Superficial Keratitis (Pannus), with Age of Onset Given When Available

    Table 12.1.  Clinical Effects of Elevated Intraocular Pressure (IOP)

    Table 12.2.  Clinical Signs of the Primary Glaucomas in the Dog

    Table 12.3.  Treatments for the Secondary Glaucomas in Dogs

    Table 12.4.  Intraocular Pressure (IOP) Controla

    Table 13.1.  Clinical Signs of Uveitis

    Table 13.2.  Recommended Therapies for Anterior Uveitis in the Dog

    Table 13.3.  Diagnosis and Treatment of the Systemic Mycoses and Uveitis in the Dog

    Table 14.1.  Inherited Cataracts in the Dog

    Table 14.2.  Classification of Canine Cataracts Based on Maturity

    Table 15.1.  Ophthalmoscopic Findings in Collie Eye Anomaly

    Table 15.2.  Ophthalmoscopic Findings in Canine Retinal Dysplasia

    Table 15.3.  Breeds of Dogs Affected with Retinal Dysplasia (RD)

    Table 15.4.  Ophthalmoscopic Changes in Progressive Retinal Atrophy (PRA)

    Table 15.5.  Characteristics of the Canine Retinal Photoreceptor Dysplasias

    Table 15.6.  Characteristics of Canine Retinal Photoreceptor Degenerations

    Table 15.7.  Ophthalmoscopic Changes in Chorioretinitis

    Table 15.8.  Ophthalmoscopic Localization of Vitreal and Retinal Hemorrhages

    Table 16.1.  Reported Eyelid Neoplasms in the Cat

    Table 16.2.  Laboratory Diagnosis of Feline Herpesvirus 1 (FHV-1) in Cats

    Table 16.3.  Laboratory Tests for the Feline Anterior Uveitides

    Table 16.4.  Therapy for the Feline Uveitides

    Table 17.1.  Treatment for Periocular Sarcoids

    Table 17.2.  Treatment for Periocular Squamous Cell Carcinoma

    Table 17.3.  Clinical Classification of Cataracts

    Table 20.1.  Neuroanatomic Localization of Horner's Syndrome

    Table 20.2.  Cavernous Sinus Syndrome

    Table 21.1.  Ocular Toxicities with Selected Systemic Drugs in the Dog

    Table 21.2.  Ophthalmic Diseases Associated with FHV-1 in the Cat

    Table 21.3.  Treatment of FHV-1 Ophthalmic Diseases in the Cat

    Table 21.4.  Selected Toxic Plants Producing Systemic and Ocular Disease in the Horse

    Table 21.5.  Ophthalmic Manifestations of Selected Infectious Diseases in Food Animals

    Table 21.6.  Selected Systemic Toxicities in Food Animals with Ophthalmic Manifestations

    List of Illustrations

    Figure 1.1.  Development of the optic sulci, which are the first sign of eye development. Optic sulci on the inside of the forebrain vesicles consist of neural ectoderm (shaded cells). The optic sulci evaginate toward the surface ectoderm as the forebrain vesicles simultaneously rotate inward to fuse. (Source: Cook C, Sulik K, Wright K. Embryology. In: Wright KW and Spiegel PH, eds. Pediatric Ophthalmology and Strabismus. St. Louis: Mosby–Year Book, 2003:3–53. Reproduced with permission of Elsevier.)

    Figure 1.2.  Formation of the lens vesicle and optic cup. Note that the optic fissure is present, because the optic cup is not yet fused inferiorly. (A) Formation of the lens vesicle and optic cup with inferior choroidal or optic fissure. Mesenchyme (M) surrounds the invaginating lens vesicle. (B) Surface ectoderm forms the lens vesicle with a hollow interior. Note that the optic cup and optic stalk are of surface ectoderm origin. RPE, retinal pigment epithelium. (Source: Cook C, Sulik K, Wright K. Embryology. In: Wright KW and Spiegel PH, eds. Pediatric Ophthalmology and Strabismus. St. Louis: Mosby–Year Book, 2003:3–53. Reproduced with permission of Elsevier.)

    Figure 1.3.  Cross-section through optic cup and optic fissure. The lens vesicle is separated from the surface ectoderm. Mesenchyme (M) surrounds the developing lens vesicle, and the hyaloid artery is seen within the optic fissure. RPE, retinal pigment epithelium. (Source: Cook C, Sulik K, Wright K. Embryology. In: Wright KW and Spiegel PH, eds. Pediatric Ophthalmology and Strabismus. St. Louis: Mosby–Year Book, 2003:3–53. Reproduced with permission of Elsevier.)

    Figure 1.4.  The hyaloid vascular system and tunica vasculosa lentis. M, mesenchyme. (Source: Cook C, Sulik K, Wright K. Embryology. In: Wright KW and Spiegel PH, eds. Pediatric Ophthalmology and Strabismus. St. Louis: Mosby–Year Book, 2003:3–53. Reproduced with permission of Elsevier.)

    Figure 2.1.  Canine orbit. Bones of the orbit shown: F, frontal; L, lacrimal; M, maxilla; S, sphenoid; T, temporal; Z, zygomatic. Orbital formina shown: A, rostral alar; E, ethmoidal; Op, optic; Or, orbital fissure.

    Figure 2.2.  Equine orbit. Bones of the orbit shown: F, frontal; L, lacrimal; S, sphenoid; T, temporal; Z, zygomatic. Orbit foramina shown: A, rostral alar; E, ethmoidal; Op, optic; Or, orbital fissure; So, supraorbital.

    Figure 2.3.  Divisions of the orbital fascia. MF, muscle fascia; OS, orbital septum; P, periorbita; T, Tenon's capsule.

    Figure 2.4.  Orbital apex of the base in the dog, illustrating structures passing through the optic foramen and orbital fissure as well as the extraocular muscle attachments. (Source: Modified from Evans H, Christensen G. Miller's Anatomy of the Dog, 2nd edn. Philadelphia: WB Saunders, 1979. Reproduced with permission of Elsevier.)

    Figure 2.5.  Surface anatomy of the dog's eye and adnexa at rest. A, medial canthus; B, lateral canthus; C, cilia; D, free margin of nonpigmented membrana nicitans; E, ciliary zone of iris; F, pupillary zone of iris; G, pupillary ruff; H, collarette.

    Figure 2.6.  Surface anatomy of the cat's eye and adnexa at rest. Arrows show major arterial circle in the ciliary zone of the iris. A, medial canthus; B, lateral canthus; C, cilia; D, free margin of membrana nicitans.

    Figure 2.7.  Upper eyelid of the dog. CF, cilia follicle; HF, hair follicle; O, orbicularis oculi muscle fibers; PC, palpebral conjunctiva; S, skin; TG, tarsal gland. (Original magnification, 10×.)

    Figure 2.8.  Lower eyelid of the dog. The meibomian glands (MG) line the deep surface of the lid. Ciliary and sebaceous (SG) glands are closely associated with hair follicles (HF). (Original magnification, 100×.)

    Figure 2.9.  Palpebral conjunctiva of a canine eyelid is externally lined by a stratified to pseudostratified columnar epithelium possessing numerous goblet cells (GC) near the fornix. (Original magnification, 250×.)

    Figure 2.10.  Drawing of a histologic section of the mammalian nictitating membrane. (Source: Modified from Evans H, Christensen G. Miller's Anatomy of the Dog, 2nd edn. Philadelphia: WB Saunders, 1979. Reproduced with permission of Elsevier.)

    Figure 2.11.  Nictitating membrane of the horse contains both glandular (G) and lymphoid (L) tissues, with the latter being superficially located within the stroma next to the bulbar surface (BS). C, Cartilage. (Original magnification, 10×.)

    Figure 2.12.  The nasolacrimal system. C, canaliculi; LD, lacrimal ducts; LG, lacrimal gland; LS, lacrimal sac; ND, nasolacrimal duct; P, lacrimal puncta.

    Figure 2.13.  Lacrimal gland of the cat. (Original magnification, 10×.)

    Figure 2.14.  Diagram of the three basic coats or tunics that comprise the mammalian globe. Outermost fibrous tunic (yellow and white), consisting of the cornea (Co) and sclera (S); the middle tunic called the uvea (black), consisting of the choroid (Ch), ciliary body (CB) and the iris (I); and the nervous tunic or coat (gray) consisting of the retina (R) and optic nerve (ON), and epithelial lining (*) of the ciliary body and iris.

    Figure 2.15.  Histologic view of the canine cornea, revealing four layers: anterior epithelium (AE), stroma (S), Descemet's membrane (DM), and endothelium (E). (Original magnification, 100×.)

    Figure 2.16.  Anterior epithelia of (A) canine and (B) ovine corneas. BC, basal cells; SC, squamous cells; WC, wing cells. (Original magnification, 400×.)

    Figure 2.17.  Basement membrane (arrows) of the anterior epithelium of the canine cornea viewed light microscopically with the aid of (A) PAS stain and (B) ultrastructurally. AE, anterior epithelium; HD, hemidesmosomes. (Original magnification: A, 400×; B, 18 000×.)

    Figure 2.18.  Posterior cornea of a horse. PE, posterior epithelium (corneal endothelium); PLM, posterior limiting membrane (Descemet's membrane); PS, posterior stroma. (Original magnification, 1200×.)

    Figure 2.19.  As the cornea ages, the Descemet's membrane continues to expand in width, as seen when comparing (A) a 1-year-old dog with (B) a 6-year-old individual. (Original magnification, 400×.)

    Figure 2.20.  SEM of a 4-year-old canine corneal endothelium reveals occasional variability in cell size (A) and the lateral surface interdigitations between cells (B, arrowheads). The most prominent feature of the endothelial cell in the adult cornea is the nucleus (N), which bulges slightly into the anterior chamber. (Original magnification: A, 960×; B, 3500×.)

    Figure 2.21.  Canine limbus. Overall view shows merging of the irregular connective tissue of the sclera (s) with the highly organized connective tissue of the cornea (c). (Original magnification, 400×.)

    Figure 2.22.  The intrascleral plexus (ISP) of a dog is located within the midsclera (S), being interconnected to the angular aqueous plexus (AAP) by aqueous veins (AV) that are sometimes prominent in size. ESV, episcleral veins. (Original magnification, 125×.)

    Figure 2.23.  Scleral ossicles (SO) in birds vary somewhat in size and shape, (A) being well developed with large interosseal spaces in owls (screech owl) (original magnification, 40×) and (B) comparatively thin in the chicken, with considerable overlap between adjacent ossicles (original magnification, 100×). CM, ciliary body musculature (Crampton's muscle); TM, trabecular meshwork.

    Figure 2.24.  SEM of the canine anterior uvea. C, cornea; CM, ciliary body musculature; CP, ciliary processes; I, iris; S, sclera. (Original magnification, 25×.)

    Figure 2.25.  Iris (I) and anterior ciliary body (CB) of the goat. The arrow points to the granular iridica, which extends posteriorly along the posterior pigment epithelium. C, cornea.

    Figure 2.26.  In many canine irides, melanocytes are concentrated in a wide band anterior to the dilator muscle (DM), as seen in the lower half of this iris. MAC, major arterial circle. (Original magnification, 100×.)

    Figure 2.27.  Sphincter muscle (SM) is located more posteriorly within the stroma of the iris in (A) the dog than (B) in the horse. The sphincter muscle in the young horse is capped by the granula iridica (GI), which is a proliferation of the posterior epithelium (PE). (Original magnification: A, 200×; B, 200×.)

    Figure 2.28.  The canine iridal dilator muscle (DM) consists of a single layer of overlapping smooth muscle fibers. Apically, the nucleus (arrowheads) of each cell is partially surrounded by pigment granules. PE, posterior epithelium. (Plastic section; original magnification, 1000×.)

    Figure 2.29.  Inner surface of the ciliary body of a dog previously treated with α-chymotrypsin to remove the lenticular zonules possesses thin ciliary processes (CP), which posteriorly give rise to smaller secondary folds (arrowheads). These folds flatten and disappear in the region called the pars plana (PP), which ends posteriorly at the adjoining retina, forming a line known as the ora ciliaris retinae (arrows). (Original magnification, 18×.)

    Figure 2.30.  The ciliary epithelium that lines the processes and intervening valleys is bilayered. The outer layer is pigmented; the inner layer is nonpigmented. (A) Cross-section of a canine ciliary process. The bilayered epithelium, which is cuboidal, lines blood vessels (BV), which together form a blood–aqueous barrier. (Plastic section; original magnification, 250×.) (B) Longitudinal section of an equine ciliary epithelium at the base of a process. Both layers are considerably more columnar than those in the dog. (Original magnification, 400×.)

    Figure 2.31.  The ciliary body muscle fibers (CM) are mostly oriented along the meridional plane in the dog. These fibers are interspersed with a variable amount of pigmentation. CP, ciliary process; S, sclera. (Original magnification, 40×.)

    Figure 2.32.  Frontal-view SEM of the canine iridocorneal angle. Fibrous pillars that attach the iris (I) to the limbus form the pectinate ligament (PL). Arrowheads indicate smaller fibrous connections between these pillars and the uveal trabeculae located behind the pectinate ligament. (Original magnification, 160×.)

    Figure 2.33.  The corneoscleral trabecular meshwork (CM) and adjacent angular aqueous plexus (AAP) in the dog. Asterisks indicate intertrabecular spaces. S, sclera. (Original magnification, 400×.)

    Figure 2.34.  The canine choroid consists of the suprachoroidea (1), stroma with large blood vessels (2), stroma with medium-sized vessels and tapetum (3), and choriocapillaris (4). A, artery; R, retina; V, vein. (Original magnification, 40×.)

    Figure 2.35.  The carnivorous tapetum lucidum consists of layers of cells, called iridocytes, which vary in number, size, and composition. (A) The dog. (B) The cat. (Original magnification: Both, 200×.)

    Figure 2.36.  (A) Young horse lens near the equator. Anterior lens capsule (*). Arrows delineate the formation of the lens bow by the nuclei of the newly formed fibers. Open arrow points rostrally. (Original magnification, 500×.) (B) Newly formed canine secondary lens fibers are evenly hexagonal in cross-section. They form small ball-and-socket junctions (arrows) along their six angular edges. SEM. (Original magnification, 6600×.)

    Figure 2.37.  The ringwulst, or annular pad (AP), of a screech owl's lens consists of radially arranged cells (arrows) that can withstand direct pressure placed on them. (Original magnification, ×200.) Insert, Overview of the annular pad. I, iris. (Original magnification, 20×.)

    Figure 2.38.  Caudal-view SEM of ciliary processes and zonular attachments to the lens in a cat. Note the zonular fibers extending from the valleys and producing a cluster of fibers at their lenticular insertion with gaps between bundles. A, Posterior lens; B, ciliary process; C, posterior zonular fibers; D, anterior zonular fibers. (Original magnification, 40×.)

    Figure 2.39.  The sensory retina consists of nine discrete layers and a supportive pigmented epithelium that forms an outer, tenth layer, as demonstrated by (A) the SEM in the pig and (B) the plastic section in the dog. The canine retina is rod dominant, being sparsely populated with cones (arrowheads and arrows). The porcine retina, compared to the dog, has a large population of cones and a smaller ratio of rods to cones. A, amacrine cell nuclei; B, bipolar cell nuclei; C, cone nuclei; G, ganglion cell nuclei; H, horizontal cell nuclei; M, Müller cell nuclei; 1, retinal pigment epithelium; 2, layer of rods' and cones' outer (OS) and inner segments (IS); 3, outer limiting membrane; 4, outer nuclear layer; 5, outer plexiform layer; 6, inner nuclear layer; 7, inner plexiform layer; 8, ganglion cell layer; 9, nerve fiber layer; 10, inner limiting membrane. (Original magnification: Both 250×.)

    Figure 2.40.  The visual cell layer of the pig contains many cones (C) among the rods (R) within the area centralis, making this animal well suited for day vision. (Original magnification, 400×.)

    Figure 2.41.  (A) The avian pecten, as seen here in the chicken, consists of a pleated vascular plexus that lies vitreally atop the optic nerve head (ON). (Original magnification, ×50.) (B) Close-up of the base of the pecten as it internally lines the nerve fibers (NF) that form the optic nerve head. BV, blood vessels of the pecten. (Original magnification, 250×.)

    Figure 2.42.  The optic nerve head and optic nerve of a dog. Arrows indicate lamina cribosa; note the number of astrocytes anterior to it. C, choroid; CMK, central meniscus of Kuhnt (accumulation of astrocytes in physiologic cup); CRV, central retinal vein; PS, pial septa; RV, retinal veins; S, sclera. (Original magnification, 720×.)

    Figure 3.1.  The precorneal film, as proposed by Butovich, Millar and Ham, demonstrates the very close interaction of lipid-binding proteins within the precorneal film and especially in its outer lipid component. (Source: Modified from Butovich, Millar and Ham. Understanding and analyzing meibomian lipids – a review. Current Eye Research 2008;33:405–420.)

    Figure 3.2.  Pupillomotor pathways. 1, A descending, excitatory multisynaptic pathway leaving the hypothalamus and projecting through the sympathetic nervous system. 2, A descending, inhibitory multisynaptic pathway leaving the hypothalamus and projecting to the Edinger–Westphal nucleus. 3, An ascending, inhibitory system leaving the reticular formation and projecting to the Edinger–Westphal nucleus. 4, An ascending, inhibitory system leaving the dorsal horn and projecting to the Edinger–Westphal nucleus. Ant median N, anterior median nucleus; DL, dorsal cochlear nucleus; Ex cun N, external cuneate nucleus; Inf Ol, inferior olivary nucleus; IP, interpeduncular nucleus; Ped, cerebral peduncle; Pyr Tr, pyramidal tract; Red N, red nucleus; Sup col, superior colliculus; Sub nig, substantia nigra. (Source: Modified from Loewy AD, Araujo JC, Kerr FW. Pupillodilator pathways in the brain stem of the cat: anatomical and electrophysiological identification of a central autonomic pathway. Brain Res 1973;60:65. Reproduced with permission of Elsevier.)

    Figure 3.3.  Chemical composition of the aqueous humor and lens. Water and protein are expressed as percentages of lens weight. Na+, Cl−, K+, and Ca²+ ions are expressed in μEq/mL of lens water. Other compounds are expressed in μmol/g of lens weight or μmol/mL of aqueous humor. AA, amino acids; RNA, ribonucleic acid.

    Figure 4.1.  Refraction of light through various lenses. (A) A spherical convex lens with a power of 10 D focuses parallel light rays at a distance of 0.1 m. (B) A flatter, spherical convex lens with a power of 5 D focuses parallel rays at a distance of 0.2 m. (C) Parallel rays passing through a concave spherical lens diverge. A virtual image is formed by tracing back (dashed lines) the diverging rays.

    Figure 4.2.  (A) In emmetropia, parallel light rays are focused on the retina. (B) In a far sighted (hypermetropic or hyperopic) eye, light rays are focused behind the retina. (C) In a near sighted, or myopic, eye, the light is focused in front of the retina.

    Figure 4.3.  Spherical aberrations occur when light passes through the (A) lens and (B) cornea. In both cases, peripheral rays are refracted (bent) more than central rays. Therefore, the focal point of the peripheral rays is closer to the lens/cornea, while the focal point of the central rays is closer to the retina. The result is a blurred image. The distance between the two focal points is called spherical aberration and is measured in diopters.

    Figure 4.4.  Photoreceptor pathways. In the fovea or area centralis region, each cone synapses with a single midget bipolar cell. This nonconvergent pathway provides for the high resolution vision which is characteristic of the cone pathway in the central retina (top right panel). In the more peripheral retina, several cones converge their output on a single diffuse bipolar, resulting in lower visual resolution (bottom right panel). The rod pathway is more complex, with a large number of rods converging on a single rod bipolar cell (RBC) (left panel). Its output is modulated by A17 and AII amacrine cells. DCB and HCB are depolarizing and hyperpolarizing cone bipolar cells, respectively, that output to ganglion cells (GC). Their input and output are modulated by horizontal cells (HC) and AII amacrine cells, respectively. (Source: Goldstein, E. B. (2005) Blackwell Handbook of Sensation and Perception, 2nd edn, Blackwell Publishing, Malden, MA. Reproduced with permission of John Wiley & Sons Ltd.)

    Figure 4.5.  Dorsal view of the left hemisphere of the canine brain. The solid arrow points to the location of the cortical area of central vision in Beagles. The hollow arrow points to the location of the cortical area of central vision in Greyhounds. The vertical axis represents distance (cm) anterior (positive values) and posterior (negative values) to the interaural plane (IAP); the horizontal axis represents distance (cm) lateral to the midline. A, marginal sulcus; B, marginal gyrus; C, endomarginal sulcus; D, endomarginal gyrus. (Source: Ofri R, Dawson WW, Samuelson DA. Mapping of the cortical area of central vision in two dog breeds. Vet Comp Ophthalmol 1994;4:172–178.)

    Figure 5.1.  Disposition of drugs instilled in the eye.

    Figure 5.2.  Differences in kinetic profiles of drugs applied as eye drops or as a sustained-release delivery system. Following application of eye drops, a maximum drug concentration is achieved locally, and then drug levels exponentially decline (first-order kinetics) to subtherapeutic tissue concentrations until the next dose. With sustained-release drug delivery systems (i.e., ophthalmic insert or implant), a plateau level of drug distribution is achieved (zero-order kinetics), allowing constant drug release over time.

    Figure 5.3.  Effect on intraocular pressure (IOP) of 0.005% latanoprost in the Beagle with primary open-angle glaucoma after (A) evening and (B) q12 h dosing.

    Figure 6.1.  Horse skull depicting sites for the auriculopalpebral (a and b), palpebral (c), frontal/supraorbital (d), lacrimal (e), zygomatic (f), and infratrochear (g) nerve blocks.

    Figure 6.2.  Corneal reflex: Corneal sensation is tested by means of a wisp of cotton wool contacting the peripheral cornea.

    Figure 6.3.  (A) To obtain a conjunctival sample, for either microbiology or cytology, the swab is gently rolled in the lower conjunctival sac anterior to the third eyelid. This is facilitated by retropulsion to protrude the third eyelid. Care must be taken to ensure that the swab touches only the area of interest to avoid contamination from nearby structures, e.g. eyelids. (B) Instruments for corneoconjunctival cytology include the Kimura spatula (top), cytobrush (middle), and scalpel blade (bottom).

    Figure 6.4.  Optics of direct ophthalmoscopy. The direct ophthalmoscope allows alignment of the observer's visual axis with a light beam, which is reflected via a mirror to illuminate the patient's fundus. Light rays emanating from the patient's eye form a real image on the observer's retina.

    Figure 6.5.  Close direct ophthalmoscopy. The examiner must be as close as possible to obtain an optimal image with direct ophthalmoscopy. To achieve this, the examiner ideally should use the right eye to examine the patient's right eye and vice versa.

    Figure 6.6.  Direct ophthalmoscope head. (A) Observer's side with viewing aperture, lens dial, quick lens switch, and diopter indicator. (B) Patient side with viewing hole and switches/dials allowing selection of beam size and shape, as well as light filters.

    Figure 6.7.  (A) Fluorescein dye is available as a sterile impregnated paper strip and can be made into a solution by mixing with sterile water, saline, or eyewash. (B) Fluorescein dye stains the exposed hydrophilic corneal stroma in a large superficial corneal ulcer in a cat.

    Figure 6.8.  Application of fluorescein dye. The fluorescein impregnated strip is moistened with, for example, sterile saline or eyewash, and then gently touched once on the dorsal bulbar conjunctiva, with the upper lid retracted. The eyelids are then closed or the animal allowed to blink to distribute the fluorescein across the ocular surface.

    Figure 6.9.  Jones test (fluorescein dye passage test) in a 1-year-old Labrador Retriever with a swelling ventral to the medial canthus of the right eye. (A) Fluorescein dye flows onto the skin at the medial canthus of the right eye. (B) As viewed at the nostrils, the right nasolacrimal apparatus is occluded but the left nasolacrimal system is patent.

    Figure 6.10.  A positive Jones result at the right nostril in a horse.

    Figure 6.11.  Aqueous tear production accessed by the Schirmer tear test 1. The short folded end of the paper strip is placed in the lateral half of the lower conjunctival sac in order to measure both basal and reflex tear production.

    Figure 6.12.  Corneal sensation can be assessed quantitatively by a Cochet–Bonnet esthesiometer. (Source: Courtesy of UW Madison Ophthalmology Service, Madison, WI, USA.)

    Figure 6.13.  Positive Seidel test depicts a leaking corneal wound at 12 o'clock.

    Figure 6.14.  Gonioscopy lenses can be divided into direct and indirect. (A) Direct gonioscopy lenses allow examination of the iridocorneal angle (ICA) opposite to the viewing position of the examiner, and create a real image. (B) Indirect goniolenses form a virtual image, which the observer examines from a frontal position.

    Figure 6.15.  Koeppe lens in situ. The lens is retained by suction on the corneal surface, freeing the examiner's hands.

    Figure 6.16.  Normal iridocorneal angle in a Flat Coated Retriever. (Source: Courtesy of Stuart Ellis, Riverbank Veterinary Centre, Ashton, Preston, Lancashire, UK.)

    Figure 6.17.  Binocular indirect ophthalmoscopy with the Keeler Vantage head-mounted indirect ophthalmoscope in the dog. In this technique, the observer does not only have sterosis, but also one hand free to hold the patient's head and eyelids. (Source: Courtesy of Franck J. Ollivier.)

    Figure 6.18.  Comparison of direct (A,D), panoptic (B,E), and indirect ophthalmoscopy (C,F). These photographs illustrate the different appearances in terms of magnification and orientation of the canine and equine ocular fundus with each of the three methods of ophthalmoscopy.

    Figure 6.19.  The optics of binocular indirect ophthalmoscope allow the light from the indirect ophthalmoscope to illuminate the patient's ocular fundus. The image is split with a prism into both examiner's eyes, permitting stereopsis. The patient's fundus image is virtual and inverted.

    Figure 6.20.  Monocular indirect ophthalmoscopy. The technique can be carried out with minimal equipment but stereopsis is lost and the observer relies on the help of an assistant to stabilize the patient's head.

    Figure 6.21.  The panoptic ophthalmoscope is a good compromise between the direct and indirect methods of ophthalmoscopy proving a real image (neither inverted or reversed) and is intermediate in magnifications between the two. (Source: Courtesy of Franck J. Ollivier, Centre Veterinaire DMV, Montreal QC, Canada.)

    Figure 6.22.  Selection of plastic and metal lacrimal cannulas suitable for performing a nasolacrimal flush in a small animal, e.g., dog and cat. Entry into the lacrimal puncta can be facilitated by cutting the end short and at an oblique angle.

    Figure 6.23.  Nasolacrimal flush in a dog. With a 2–5 mL syringe of sterile saline or eyewash attached to the cannula, and the upper lid everted to expose the upper punctum, the cannula is inserted into the dorsal punctum in a ventromedial direction, following the line of the upper canaliculus.

    Figure 6.24.  Paracentesis. (A) Aqueous paracentesis. The hypodermic needle is inserted (bevel up) through the clear cornea immediately adjacent to the limbus or the subconjunctival limbus. A tunneling motion faciliates passage through the sclera or cornea. The needle must enter the anterior chamber anterior to the iris and be directed parallel with the iris. (B) Vitreous paracentesis. The hypodermic needle is inserted posterior to the limbus (5–7 mm in the dog and 10–12 mm in the horse) and firmly tunneled through the sclera and pars plana of the ciliary body. The needle must be directed toward the posterior pole to avoid the lens. (Source: Courtesy of Simon Scurrell, Willow Referral Service, Shirley, West Midlands, UK.)

    Figure 6.25.  Retinoscopy carried out in a dog. Note the working distance of 67 cm between examiner and patient. The skiascopy bar or rack is positioned in front of the dog's eye; it contains a series of plus and minus spherical lenses in increments of 0.5–1.0D to quantify the eye's refractive error.

    Figure 6.26.  Use of a handheld slit-lamp biomicroscope in a dog.

    Figure 6.27.  Indentation tonometry. (A) Schiotz tonometer with 7.5 and 10.0 g weights. (B) Schiotz tonometer use in a cat.

    Figure 6.28.  Applanation tonometry. (A) TonoPen-Vet. (B) The footplate contains a central pressure-sensitive tip that protrudes from and is surrounded by an insensitive ring. (C) The tip is covered by a disposable latex membrane that protects the sensitive plunger and prevents disease transmission. (D) TonoPen-Vet use in a dog.

    Figure 6.29.  Rebound tonometry with the TonoVet in a dog.

    Figure 6.30.  Reformatted sagittal CT images of a normal feline globe seen in the soft tissue window (W 426, L 55). (Source: Courtesy of Paul Mahoney, Willows Veterinary Centre & Referral Service, Solihull, UK.)

    Figure 6.31.  (A) Dacryocystorhinogram of the normal nasolacrimal duct system of a 9-year-old West Highland white terrier. The cannula placed via the upper punctum into the lacrimal sac is visible (small arrow). Contrast medium is present at the nares and refluxing into the nasal cavity (large arrow). Note that the metallic pulse oximetry device is superimposed over the rostral nares and maxillary region. (B) Dacryocystorhinogram of a 3-year-old Labrador Retriever with chronic dacryocystitis affecting the right side. Obstruction of contrast is evident at the level of the third upper premolar tooth (arrow). Proximal to the obstruction there is an irregular cystic dilation of the duct. No contrast is visible distal (rostral) to the obstruction.

    Figure 6.32.  The appearance of the normal canine globe and orbit on (A) dorsal T1- and (B) T2-weighted MRI. (A) The dorsal T1 image shows hyperintense retrobulbar fat, lens capsule, iris, and ciliary body. The aqueous humor and vitreous humor have a slightly lower signal when compared with muscle, and the normal lens has low signal characteristics. (Source: Courtesy of Paul Mahoney, Willows Veterinary Centre & Referral Service, Solihull, UK.)

    Figure 6.33.  (A) Noncontact specular microscopy being undertaken on an anesthetized Beagle cross-bred dog. (B) Specular microscopy image of the central corneal endothelium of a normal 9-month-old cross-bred dog. (Source: Courtesy of Matthew Chandler, Animal Eye Clinic of North Florida, Jacksonville, FL, USA.)

    Figure 6.34.  Optical coherence tomography (OCT) with 3D reconstruction images of the fundus of a normal, 15-week-old domestic shorthair kitten fundus. (Source: Courtesy of Laurence Occelli and Simon Petersen-Jones, Michigan State University, East Lansing, MI, USA.)

    Figure 6.35.  Normal equine fluorescent antibody image with maximal fluorescence (retinal arterial and venous phase). (Source: Molleda J.M., Cervantes I., Galan A., et al. (2008) Fluorangiographic study of the ocular fundus in normal horses. Veterinary Ophthalmology.11(1), 2–7. Reproduced with permission of John Wiley & Sons Ltd.)

    Figure 6.36.  (A) Standardized diagnostic A-mode probe with tissue model for calibration. (B) Ocular ultrasound probes. Top: Diagnostic 10-MHz probe. The white marker on the probe designates the upper portion of the echogram. Middle: High-frequency (20 MHz) B-mode probe. Bottom: Standardized diagnostic 8-MHz A-mode probe.

    Figure 6.37.  Vertical axial scan. The white marker on the probe points dorsally.

    Figure 6.38.  Normal B-mode/vector A-scan echogram of the canine eye. The integrated vector A-scan is displayed in the bottom half of the echogram. Echoes appear as bright lines or dots of different intensity on B-mode, and as vertical spikes from a horizontal baseline on the A-scan vector. In axial vertical direction (white marker points dorsally), the posterior lens capsule produces a strong echo A-mode and B-mode (arrows). The strong posterior eye wall echo contrasts against the acoustically empty vitreous.

    Figure 6.39.  Ciliary body tumor (T) invading the vitreous and causing a lens subluxation (arrow).

    Figure 6.40.  B-mode ultrasound of a long-standing retinal detachment, which appears as a T-shaped membrane (closed funnel).

    Figure 6.41.  B-mode/vector A-scan echogram of a retrobulbar abscess in a Miniature Schnauzer. The abscess (A) is anechoic in the center and surrounded by a thick, highly reflective wall. It can be clearly distinguished from the surrounding orbital tissues. The corresponding vector A-scan shows the low echogenicity within the cystic abscess and the high posterior wall echo (arrows).

    Figure 6.42.  Ultrasound biomicroscopy (50 MHz) of the iridocorneal angle (ICA) of a dog. A, cornea; B, sclera; C, basal iris; D, pectinate ligaments; and E, ciliary cleft.

    Figure 6.43.  Color Doppler images (top) and pulse waves (bottom) of the short posterior ciliary arteries (SPCA; arrows) of a normal (A) and (B) glaucomatous Beagle. Comparisons of the normal to the glaucomatous Beagle's SPCA Doppler blood flow parameter indicated decreased end-diastolic velocities (EDV) and increased resistive indexes compared to the normal dogs. (Source: Courtesy of Kathleen Gelatt-Nickolson, North Florida Radiology, Gainesville, FL, USA.)

    Figure 6.44.  A dark-adapted canine electroretinogram (ERG) in response to a brief, bright flash. Both rod and cone photoreceptors drive this response. OP, oscillatory potential.

    Figure 6.45.  (A) The corkscrew, needle, and button-type electrodes are reliable and easy to use as reference and ground electrodes, as well as active electrodes for visual evoked potentials (VEPs) in animals. The JET electrode is a monopolar corneal contact lens electrode that can be used in many species, such as the cat and dog. (B) The gold-foil electrode can be used as an active electrode for recording the equine electroretinogram (ERG). The plastic foil is bent over the rim of the lower eyelid so the gold-coated side touches the corneal surface.

    Figure 6.46.  Two types of stimulators for flash electroretinograms (FERGs) and flash visual evoked potentials (FVEPs). (A) Both the stimulus and background light intensities need to be measured to ensure that a known amount of light is delivered to the patient's eye. The detector of the photometer is positioned at the level of the eye of a patient. A xenon flash and a halogen bulb, used for steady adapting light, are built into the aluminum housing on top of this full-field (Ganzfeld) stimulator. (B) A handheld stimulator (mini-Ganzfeld) is easy and convenient to use in large animals, but can be also used in small animals. The stimulator should be held close to the eye without disturbing the corneal electrode.

    Figure 6.47.  Four electroretinogram (ERG) responses from a normal dog. (A) A mixed, dark-adapted rod–cone response. The small positive deflection just below the letter is a stimulus artifact, which shows when a brief, white flash (3.0 cd/m²/s) is delivered. The response has a conspicuous, negative a-wave followed by the large, positive b-wave. Superimposed on the ascending limb are a few OPs (open arrow). The peak of the b-wave is bipartite where the later peak reflects the slower rod b-wave (black arrow). (B) A dark-adapted, mainly rod-driven ERG in response to a dim white flash (0.03 cd/m²/s). Note that there is no obvious a-wave and that the implicit time of the b-wave is similar to the rod-driven peak in A. (C) A cone response to a bright, white flash (3.0 cd/m²/s) presented on a rod-saturating background (40 cd/m²). The amplitude is considerably smaller than that of the dark-adapted responses, but both an a- and a b-wave can be seen. (D) A cone-driven response to a bright, white flickering stimulus (30 Hz) in the presence of the steady, adapting light.

    Figure 7.1.  Canine skull demonstrating the lack of an osseous orbital floor, rendering the orbital contents susceptible to penetrating trauma through the roof of the mouth.

    Figure 7.2.  Ultrasonography of an orbital neoplasm (meningioma) in a 10-year-old Collie. There is marked indentation of the caudonasal globe.

    Figure 7.3.  Ultrasound probe placement for a transoral approach to the orbit.

    Figure 7.4.  Orbital foreign body in a 6-year-old dog. (A) Dog at initial presentation with chronic purulent discharge of the right eye. (B) Fistulous tract in the dorsal conjunctival fornix.

    Figure 7.5.  Transoral abscess drainage technique.

    Figure 7.6.  Distended excretory duct resulting from adenitis of the zygomatic salivary gland.

    Figure 7.7.  Masticatory muscle myositis in a German Shepherd. There is marked swelling of the masticatory muscles and bilateral exophthalmos, which is more marked in the left eye. (Source: Courtesy of Ingo Walde, Faculty of Veterinary Medicine, Vienna, Austria.)

    Figure 7.8.  Bilateral extraocular polymyositis in an 8-month-old Golden Retriever.

    Figure 7.9.  A 12-year-old Rough Collie with nictitans protrusion, exophthalmos, and dorsolateral displacement of the globe. Orbital lymphoma was suspected based on an ultrasound-guided fine-needle aspirate (FNA). A polymerase chain reaction (PCR) assay for antigen receptor rearrangement (PARR) confirmed the diagnosis of lymphoma.

    Figure 7.10.  Proptosis of the right globe in a young Golden Retriever. There is entrapment of the eyelids as well as severe swelling and hyperemia of the bulbar conjunctiva. The cornea has already been protected with an ointment.

    Figure 7.11.  Severe proptosis with avulsion of the optic nerve and several extraocular muscles in a Cocker Spaniel. Despite the clear and intact anterior segment, enucleation was inevitable.

    Figure 7.12.  Enucleation: subconjunctival approach. (A) Incision of the bulbar conjunctiva. (B) After transection of the extraocular muscles close to their scleral attachments, the optic nerve is clamped and severed. (C) The orbit is packed with a gauze sponge and the nictitating membrane resected. (D) Excision of the lid margins. (E,F) Closure of the periorbital and deep fascial layers with a continuous suture pattern, after removal of the gauze sponge. Skin closure with simple interrupted or continuous sutures.

    Figure 7.13.  (A) Dog with intrascleral prostheses in both eyes. (B) Close-up of right eye showing mild corneal neovascularization, pigmentation, and fibrosis of the cornea.

    Figure 8.1.  Cross-section through the canine lid. 1, Eyelash-like hair on the lateral part of the upper lid; 2, Zeis/Moll glands; 3, meibomian gland; 4, mucus cells conjunctiva; 5, fornix; 6, scleral conjunctiva; 7, nictitating membrane gland; 8, orbicularis oculi muscle; 9, tarsal plate.

    Figure 8.2.  Muscles of the lids of the left eye of the dog. 1, Orbicularis oculi muscle; 2, lateral palpebral ligament or retractor anguli oculi lateralis; 3, medial palpebral ligament; 4, malaris muscle; 5, levator palpebrae muscle; 6, levator anguli oculi medialis muscle.

    Figure 8.3.  The ends of the upper lid or lateral canthus sutures can be caught or linked together in an outer-placed suture to prevent irritation of the cornea.

    Figure 8.4.  Pathologic ankyloblepharon. Delayed eyelid opening in a puppy has resulted in ophthalmic neonatorum with pus extruding from the medial canthus.

    Figure 8.5.  Distichiasis (hairs in or on the lid margin) emerging from the meibomian (1), Zeis, or Moll (2) gland openings. 3, Tear film; 4, cornea.

    Figure 8.6.  Cryodestruction of multiple distichiae in the conjunctiva–tarsal plate in a dog.

    Figure 8.7.  Entropion correction by retraction sutures (tacking). The sutures can be maintained for at least 2–3 weeks. The scar tissue tube formed around the suture material will result in moderate permanent correction. (A) Simple, interrupted sutures. (B) U-figure suture, with the disadvantage that the needle points in the direction of the cornea during suturing.

    Figure 8.8.  Celsus–Hotz procedure for the correction of severe lower lid entropion with corneal ulceration. (A) The skin is incised at about 2.5 mm (as near as possible to the margin for better prediction of the entropion correction, but with enough space for skin suturing) from and parallel to the lid margin (B). (C) The skin plus orbicularis muscle are excised [not deeper: canaliculus and (sub)conjunctival tissues should not be damaged]. The lid margin should no longer show spontaneous intention of inward rolling. (D) The skin is sutured with material not exceeding 5-0 (e.g., nonabsorbable silk or absorbable, especially in difficult-to-handle animals, mono- or poly-filament), using a fine, round-body needle with or without a micropoint. Continuous sutures alone are not used because of the risk of rupture of the suture material when rubbed, resulting in dehiscence of the entire wound. The first sutures are placed at the medial and lateral ends, and the rest of the wound is closed by halving the intervals in the following order 1, 2, 3, 4, and so on. The distance between sutures is 2–2.5 mm. Alternatively, the intervals of the simple interrupted sutures can be made at about 4 mm and thereafter the remaining wound intervals closed by a continuous suture. (E) Secondary upper eyelid trichiasis to the lower, caused by postoperative lower lid conjunctival swelling, can be prevented by tacking of the upper lid (5).

    Figure 8.9.  Pronounced macroblepharon–ectropion (diamond-shaped fissure) in a Blood Hound. The stretched fissure length was 48 mm. There is also some medial and lateral entropion, and a notch or kink in the lower lid margin. Note that the lower lid hangs in the middle, about 15 mm away from the third eyelid–cornea and is not really everted in an ectropion position. This results in chronic exposure and secondary conjunctivitis.

    Figure 8.10.  Kuhnt–Szymanowski procedure, modified by Blaskovics and further by Fox and Smith for ectropion–macroblepharon to avoid splitting the lid margin. Use in the dog was first described by Munger–Carter. (A,B) The skin incision is 2–2.5 mm below the eyelid margin and extends 5–10 mm beyond the lateral canthus. The skin flap is dissected from its deeper muscle layers. All bleeding has to be arrested. (C,D) Equal-sized wedges, one of the lid margin conjunctiva and one of the skin are excised with scissors. The lid margin is apposed by a figure-of-eight or figure-U 5-0 to 6-0 suture. If desired, the conjunctival wound can be sutured by a subconjunctival, simple, continuous, 8-0 absorbable suture. The skin defect is apposed by simple, interrupted, 5-0 to 6-0 sutures. Advantage: staggering wound, less damaging to the lid margin. Disadvantage: shortens only the lower lid.

    Figure 8.11.  The Roberts–Jensen pocket method for lid fissure length reduction by permanent medial or lateral tarsorrhaphy. (A) The procedure starts with the removal of the outside lid margin and the inside margin plus meibomian glands. If performed in the medial canthus, the upper lacrimal punctum is incised in the procedure. (B) Flap of upper conjunctiva is pulled downward into the pocket in the lower lid between the conjunctiva and muscle–skin layers, and anchored through the muscle–skin with a simple, interrupted, 5-0 suture. (C) Apposition of the new lateral or medial canthus by a figure-of-eight, 5-0 to 6-0 suture, and further closure by simple interrupted sutures.

    Figure 8.12.  For the treatment of nasal fold trichiasis, the nasal folds can be excised. (A) The nasal folds are clamped and carefully excised with curved Mayo scissors. (B) The wound edges are apposed with simple, interrupted, 5-0 to 6-0 sutures. The first sutures are placed at the upper and lower ends and at the medial canthus; then, the rest of the wound is closed by halving the intervals.

    Figure 8.13.  Chronic staphylococcal blepharitis in an adult dog. (Source: Courtesy KN Gelatt.)

    Figure 8.14.  Eyelid pyogranulomas of the upper lid and lateral canthus in a Miniature Poodle. (Source: Courtesy of KN Gelatt.)

    Figure 8.15.  Immune-mediated medial canthal blepharitis in an adult mongrel German hunting dog. The dog also has bilateral chronic superficial keratitis.

    Figure 8.16.  Preoperative (A) and postoperative (B) appearance of an adenoma in the lateral upper eyelid in a 9-year-old Dalmatian.

    Figure 8.17.  Temporary tarsorrhaphy. After, for example, a canthotomy and repositioning of a luxated or proptosed globe, a temporary tarsorrhaphy can be performed. The lid fissure is closed by two or three U-figure sutures, and these are prevented from cutting into the skin by, for example, infusion tubing.

    Figure 9.1.  Strategy and procedures for diagnosis and treatment of nasolacrimal (drainage) disease. (Character of the ocular discharge is mainly serous or seromucus. If the ocular discharge is mucopurulent or purulent, see Chapter 10.)

    Figure 9.2.  Embryologic development of the canine nasolacrimal system. (A) Note the nasolacrimal groove between the lateral nasal fold and the maxillary process at approximately day 21 of gestation in the dog. (B) The lateral nasal fold fuses with the maxillary process between days 22 and 26. This fusion buries the surface ectoderm cells, which will grow and form the nasolacrimal duct system. (C) The ectodermal cells form a cord with two proximal processes that extend toward the medial upper and lower eyelid, whereas the distal end grows toward the nostril. This ectodermal cord canalizes and becomes a duct and canaliculi shortly after birth.

    Figure 9.3.  Gross anatomy of the canine nasolacrimal duct system. Note the relationship of the eyelids, puncta, canaliculi, lacrimal sac, nasolacrimal duct, and nasal puncta.

    Figure 9.4.  12-week-old Papillion puppy with bilateral inferior punctal atresia, resulting in marked epiphora.

    Figure 9.5.  Ventral punctual atresia. Note the ballooning of the conjunctiva over the aplastic punctum (A) during a normograde nasolacrimal flush through the superior punctum. The ballooning conjunctiva is excised with scissors (B) to create a new punctum.

    Figure 9.6.  Anomalies of the medial canthus of the small-breed of dogs that predispose to epiphora and pigmentary keratitis. Note the caruncular trichiasis, the tight medial canthal ligaments that create a medial canthal trough, and the medial ventral entropion that compress the ventral lacrimal punctum and canaliculus.

    Figure 9.7.  A superior punctual and canalicular foreign body that was causing darcyocystitis in a 2-year-old Lhasa Apso. Note the mucopurulent ocular discharge and conjunctivitis.

    Figure 9.8.  Scanning electron micrograph of the bulbar surface of the canine third eyelid (nictitating membrane). A nictitans ductile opening onto the posterocentral surface of the third eyelid is well visualized. (Original magnification, 800×).

    Figure 9.9.  Acute-onset keratoconjunctivitis sicca in a 5-year-old female Pug with mucopurulent discharge, hyperemic conjunctiva, and an infected axial corneal ulcer, stromal malacia, diffuse corneal edema, and ciliary flush.

    Figure 9.10.  A 4-year-old Boston Terrier with early keratoconjunctivitis sicca. Note the mucoid ocular discharge, moderate conjunctival hyperemia, and mild chemosis. This eye had a Schirmer tear test 1 of 8 mm wetting/min.

    Figure 9.11.  Dog with early keratoconjunctivitis sicca. Note the intense conjunctival hyperemia, thick mucopurulent discharge, and chronic keratitis characteristic of a subacute or chronic tear deficiency.

    Figure 9.12.  A 6-year-old male castrated, mixed-breed dog with chronic keratoconjunctivitis sicca. Note the marked conjunctival hyperemia, lackluster appearance to the corneal surface, and superficial corneal neovascularization.

    Figure 9.13.  Tear film break-up time(TBUT) test can be performed in the dog with some difficulty. Immediately after instillation of one drop of topical fluorescein stain, the eyelids are digitally held open, and the dispersion of fluorescein is observed with a portable slit-lamp biomicroscope using a cobalt blue filter. The TBUT is the mean time for the development of dark (dry) spots in the precorneal film.

    Figure 9.14.  (A) A 10-year-old male castrated Shih Tzu with chronic keratoconjunctivitis sicca. The condition had been treated intermittently with topical antibiotics and corticosteroids and twice-daily topical cyclosporine therapy for 4 years. (B) Same dog after substituting topical 0.02% tacrolimus ointment for the cyclosporine therapy. Patient exhibited marked improvement with less mucopurulent ocular discharge and ocular comfort.

    Figure 9.15.  A 7-year-old female spayed Yorkshire Terrier 3 months after parotid duct transposition, showing precipitates on the ocular surface and eyelids.

    Figure 10.1.  Clinical strategy for the diagnosis and treatment of conjunctivitis of the dog.

    Figure 10.2.  Neutrophils, cocci, and conjunctival epithelial cells in a cytologic specimen from a dog with bacterial conjunctivitis. (Diff Quik; original magnification, 33×.)

    Figure 10.3.  Dog with experimentally-induced recurrent canine herpesvirus-1 conjunctivitis in a dog. (Source: Courtesy of Eric Ledbetter, Cornell University, Ithaca, NY, USA.)

    Figure 10.4.  Chemosis in a dog associated with a bee sting.

    Figure 10.5.  Follicular conjunctivitis involving the bulbar conjunctiva of the nictitating membrane.

    Figure 10.6.  Multiple papillomas on the skin and bulbar conjunctiva. (Source: Courtesy of Nancy McLean, VCA Veterinary Care Animal Hospital and Referral Center, Albuquerque, NM, USA.)

    Figure 10.7.  Nodular fasciitis on the temporal aspect of the globe. An immature cataract is also present.

    Figure 10.8.  Conjunctival dermoid with hair at the temporal aspect of the globe.

    Figure 10.9.  Onchocerca granuloma in the ventral bulbar conjunctiva. (Source: Courtesy of Nancy McLean, VCA Veterinary Care Animal Hospital and Referral Center, Albuquerque, NM, USA.)

    Figure 10.10.  Conjunctival hemorrhage secondary to trauma in a dog. Note the miosis indicating anterior uveitis, which is also a result of the trauma.

    Figure 10.11.  Severe conjunctivitis associated with systemic and retrobulbar blastomycosis.

    Figure 10.12.  Petechial and ecchymotic hemorrhages are present on the conjunctiva of the nictitating membrane in a dog with thrombocytopenia.

    Figure 10.13.  Pedicle bulbar conjunctival graft. A viable conjunctival graft 2 weeks post surgery. Multiple vessels are seen extending to the edges of the graft. The polyglactin 910 sutures are still present.

    Figure 10.14.  Completely encircling nictitating membrane in an American Cocker Spaniel.

    Figure 10.15.  Eversion of the cartilage of the nictitating membrane in a Great Dane.

    Figure 10.16.  Prolapse of the gland of the nictitating membrane (cherry eye) in an English Bulldog.

    Figure 10.17.  Strategy for diagnosis and treatment of protrusion of the nictitating membrane.

    Figure 10.18.  Adenoma of the gland of the nictitating membrane.

    Figure 10.19.  Plasma cell infiltration of the nictitating membrane, plasmoma, in a German Shepherd.

    Figure 11.1.  The canine cornea.

    Figure 11.2.  Keratomalacia in a dog with bacterial keratitis.

    Figure 11.3.  Superficial corneal pigment in a Pug with chronic pigmentary keratitis.

    Figure 11.4.  Pigment deposits on the endothelial surface of the cornea resulting from the rupture of uveal cysts. Several intact uveal cysts remain in the anterior chamber and adjacent to the corneal endothelium.

    Figure 11.5.  Afghan Hound with diffuse corneal edema after CA-1 vaccination.

    Figure 11.6.  Dermoid, or choristoma, at the temporal limbus in a young dog.

    Figure 11.7.  Complete incision superficial keratectomy. (A) The initial corneal incision, which may be round, square, or triangular, should completely surround the lesion to be removed and can be made using a corneal trephine, diamond knife, or microsurgical blade. (B,C) After the initial incision is made, the edge of the tissue to be removed is grasped by a forceps, and a corneal dissector (e.g., Martinez corneal dissector, Beaver #64 microsurgical blade, iris spatula) is introduced and held parallel to the cornea. The dissector is used to separate the corneal lamella without penetrating deeper than the original incision. The cornea is then separated until the opposite incision line, or limbus, is reached. (D) Scissors may be needed to connect the dissection to the opposite incision or to remove the corneal tissue from the limbus.

    Figure 11.8.  Dog with persistent pupillary membranes adherent to the posterior cornea, resulting in a focal corneal opacity.

    Figure 11.9.  Canine herpesvirus-1 dendritic ulcerative keratitis in a dog receiving systemic immunosuppressive therapy (cornea stained with fluorescein).

    Figure 11.10.  Chemical burn or keratitis after mace exposure in a dog.

    Figure 11.11.  A corneal ulcer is diagnosed on the basis of retention of topically applied fluorescein dye by the corneal stroma, as observed in this superficial corneal ulcer.

    Figure 11.12.  A large nonvascularized spontaneous chronic corneal epithelial defect (SCCED) in a 9-year-old Boxer. Note the axial location, ring of loose epithelium, and the decrease in intensity of the fluorescein staining as it migrates under the loose epithelium surrounding the defect.

    Figure 11.13.  Epithelial debridement of a spontaneous chronic corneal epithelial defect (SCCED) in a 9-year-old Golden Retriever. One cotton-tipped applicator is used to remove loose epithelium, while a second is used to prevent prolapse of the third eyelid.

    Figure 11.14.  Central, deep stromal corneal ulcer in a brachycephalic dog.

    Figure 11.15.  Bridge or bipedicle conjunctival graft. (A) The conjunctiva is excised from the limbus for approximately 180° both adjacent and parallel to the linear corneal lesion. This area is extensively undermined, and the underlying fibrous tissue is removed. A second conjunctival incision is made 5–8 mm peripheral and parallel to the original conjunctival incision, thus creating a bridge of conjunctiva. (B,C) The bridge is advanced over the lesion and then sutured, using simple, interrupted sutures into the cornea around the lesion. (D) The original graft-harvesting site is closed by opposing the remaining conjunctiva with a simple, continuous suture.

    Figure 11.16.  Pedicle conjunctival graft. (A) The base of the pedicle flap should be directed toward the area of the limbus nearest to the lesion. Once the location of the base is determined, a site 1.0–1.5 cm temporal to the base is located, at which the flap will be initiated. Through a small slit in the conjunctiva, the entire conjunctival flap site is undermined using blunt dissection. Two parallel cuts are then made to create a strip of conjunctiva. (B) The strip of conjunctiva is rotated to cover the corneal lesion. The flap is sutured to the cornea with simple, interrupted sutures of 7-0 to 9-0 polyglactin 910 or nylon. (C) The sutures are placed first at the distal end of the flap and then 1.0–1.5 mm apart.

    Figure 11.17.  (A) After the corneal ulcer is healed, the blood supply to the conjunctival graft can be trimmed. (B) After applying topical anesthetic, scissors can be placed under the pedicle portion of the graft, which is not adhered to the underlying normal corneal epithelium.

    Figure 11.18.  Central corneal descemetocele in a dog with chronic corneal disease.

    Figure 11.19.  Penetrating keratoplasty using fresh tissue in a dog with diffuse corneal edema, (A) immediately after surgery and (B) 3 years later. Frozen cornea can also be used for perforations, but a clear cornea, as observed here, should not be expected. (Source: Courtesy of Bob English, Animal Eye Care Veterinary Ophthalmology Practice, Cary, NC, USA.)

    Figure 11.20.  Horizontal full-thickness corneal laceration with uveal prolapse and hyphema in a dog.

    Figure 11.21.  (A) Mild and (B) severe chronic superficial keratitis in German Shepherd dogs.

    Figure 11.22.  Dachshund with superficial punctate keratitis.

    Figure 11.23.  Crystalline corneal dystrophy in a Siberian Husky.

    Figure 11.24.  (A) Dog with Cushing's disease with focal lipid keratopathy. (B) Dog with perilimbal lipid keratopathy.

    Figure 11.25.  (A) Dog with focal corneal degeneration with lipid infiltration and vascularization. (B) Diffuse corneal degeneration with a dense white plaque and vascularization.

    Figure 11.26.  Diffuse corneal edema in a Boston Terrier with advanced corneal endothelial

    Enjoying the preview?
    Page 1 of 1