Open Access

Animal models of dry eye disease: Useful, varied and evolving (Review)

  • Authors:
    • Wei Huang
    • Konstantinos Tourmouzis
    • Henry Perry
    • Robert A. Honkanen
    • Basil Rigas
  • View Affiliations

  • Published online on: October 1, 2021     https://doi.org/10.3892/etm.2021.10830
  • Article Number: 1394
  • Copyright: © Huang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Dry eye disease (DED), which is a prevalent disease that still lacks successful treatment options, remains a major challenge in ophthalmology. Multiple animal models of DED have been used to decipher its pathophysiology and to develop novel treatments. These models use mice, rats, rabbits, cats, dogs and non‑human primates. Each model assesses aspects of DED by focusing on elements of the lacrimal functional unit, which controls the homeostasis of the tear film. The present review outlines representative DED animal models and assesses their contribution to the study of DED. Murine models are the most extensively used, followed by rabbit models; the latter offer the advantage of larger eyes, a favorable biochemical profile for drug studies, experimental ease and relatively low cost, contrasting with non‑human primates, which, although closer to humans, are not as accessible and are expensive. No comprehensive ‘ideal’ animal model encompassing all aspects of human DED exists nor is it feasible. Investigators often choose an animal model based on their experimental needs and the following four features of a given model: The size of the eye, its biochemical composition, the available research reagents and cost. As research efforts in DED expand, more refined animal models are needed to supplement the enormous contribution made to date by existing models.

1. Introduction

Animal models of diseases often help investigators to decipher their pathogenesis or develop new treatments. Dry eye disease (DED), a multifactorial disease of the ocular surface that for years lacked even a precise definition, remains a major research challenge in ophthalmology (1). Its importance derives mainly from its high prevalence, variously reported to affect between 5 and 15% of the world population, and from the lack of treatments that provide, at a minimum, adequate symptomatic control.

Homeostasis of the ocular surface maintains key components of ocular function, notably an optically clear cornea and the normal composition of the tear film, which protects the ocular surface from inflammation, infection, desiccation, and traumatic injury. The lacrimal functional unit, thought to maintain such homeostasis, consists of the conjunctival and corneal epithelia, the lacrimal and Meibomian glands, the interconnecting innervation, and the hormonally responsive and immune cells resident in these tissues. Assessing the anatomical and functional elements of the lacrimal functional unit has contributed to our understanding of DED.

Recent progress in the pathophysiology of DED, classifying it as aqueous-deficient, evaporative or mixed, provides a useful reference point to categorize and assess the animal models that have been developed over the years (Table I). It is perhaps superfluous to state at the outset that no animal model can ever recapitulate the entirety of a human disease. Nevertheless, investigators developing such models aim to capture components of the target disease that determine its pathophysiology and response to novel treatments.

Table I

Animal models of DED.

Table I

Animal models of DED.

A, Mouse model
Model type Aqueous-deficientEvaporativeMixed
MRL/lpr mouseYesNoNo
NoD mouseYesNoNo
Lacrimal gland excision modelYesNoNo
Desiccating environmental stress modelNoYesNo
Models based on the aging of miceNoNoYes
B, Rat model
Model type Aqueous-deficientEvaporativeMixed
Scopolamine modelYesNoNo
Visual display terminal user modelNoYesNo
C, Rabbit model
Model type Aqueous-deficientEvaporativeMixed
Atropine modelYesNoNo
Benzalkonium modelYesNoNo
Autoimmune dacryoadenitis modelYesNoNo
Main lacrimal gland ablation modelYesNoNo
Complete dacryoadenectomy modelYesNoNo
Concanavalin A-induced modelYesNoNo
Acute desiccative stress modelNoYesNo
Closure of Meibomian gland orificesNoYesNo
Evaporative DED model (M. tuberculosis)NoYesNo
D, Cat model
Model type Aqueous-deficientEvaporativeMixed
Main lacrimal gland ablation modelYesNoNo
E, Dog model
Model type Aqueous-deficientEvaporativeMixed
Spontaneous DED modelYesNoNo
Main lacrimal gland ablation modelYesNoNo
F, Non-human primate
Model type Aqueous-deficientEvaporativeMixed
Lacrimal gland excision modelYesNoNo

[i] DED, dry eye disease; NOD, non-obese diabetic.

DED models have employed a wide range of species, extending from mice to nonhuman primates. It is only a matter of time before zebra fish are used in DED studies, given its potential contribution to understanding the function of the anterior chamber (2). The contributions of existing models, along with their limitations, provide the impetus for their further refinement until the medical frontier of DED is finally conquered. Here, we present the main murine, rat, rabbit, cat, dog and non-human primate models of this disease and discuss persistent needs in model development.

2. Mouse models of DED

Mouse models

Mouse models of DED are the most commonly used mainly because mice are small, easy to handle, and inexpensive to house. Additional reasons for their widespread use are the availability of interesting knockout and transgenic strains, and of immunological and molecular reagents for their study. Below, we describe the main types of mouse DED models.

Mouse models of Sjὃgren syndrome

Sjὃgren syndrome, the most common autoimmune disease in humans, is characterized by mononuclear cell infiltration of the salivary and lacrimal glands (LGs) (3,4). There are several mouse models of Sjὃgren syndrome, each recapitulating one or more of its aspects, but none is perfect.

The MRL/lpr mouse, primarily used to study systemic lupus erythematosus, was found to have coexisting Sjὃgren syndrome (5). Similar to humans, inflammation is significantly greater in lacrimal and salivary glands of female MRLP/lpr mice compared to age-matched males (6). The onset of dacryoadenitis is observed at the age of 1 month, and the disease progresses rapidly, with the mice dying at 6 months (7). The MRL/lpr mice spontaneously develop T-cell-derived LG inflammation, mimicking the human disorder. The T cells, most of them CD4 T cells, represent approximately 80% of the inflammatory cells in LG lesions (8,9). The mutation of the lpr gene results alters the Fas protein, causing lymphoproliferation with defective lymphocyte apoptosis, clonal deletion of autoreactive T cells in peripheral lymphoid organs, and elimination of activated T cells.

The non-obese diabetic (NOD) mouse, first established as a model of insulin-dependent diabetes mellitus, shows secondary autoimmune dacryoadenitis, in which predominantly CD4+Th1 cells infiltrate the LGs (10-12). A variant strain of the NOD mouse (NOD.B10.H2b with an altered major histocompatibility complex region) does not develop diabetes. However, because of LG T cell infiltration, it displays a mild Sjὃgren phenotype at the age of 10 weeks, followed by severe dacryoadenitis and DED at 1 year (13). Interestingly, in male mice dacryoadenitis has a higher incidence and develops earlier compared to females. These differences have been attributed to differences in sex steroid hormones (14).

There are several additional Sjὃgren syndrome models, such as the NZB/NZW F1 (15,16), the NFS/sld (17), the neurturin-deficient (18), the TGF-β1 knockout (19-21), the IQI/jic (21), and the Id3-deficient (22) mice. Each mouse model exhibits a unique characteristic of dacryoadenitis making them suitable for the study of particular aspects of Sjὃgren syndrome related DED. Strain-specific characteristics of the disease enable targeted manipulation of mechanisms underlying the autoimmune process.

Lacrimal gland excision model

Shinomiya's group developed an exorbital LG excision mouse model (an aqueous-deficient DED model) with persistent tear volume reduction, superficial punctate keratitis, and increased tear levels of IL-1β. Unfortunately, the inflammatory infiltrate of the ocular surface tissues was not sufficiently pronounced to make this model informative. Thus, the added intraorbital LG excision, which led to severe tear volume reduction and severe inflammatory changes in the corneal surface. They also devised a minimally invasive approach to remove the intraorbital LGs via the subcutaneous tissue of the temporal lid margin, which greatly increased the success rate of the surgery (23,24).

Desiccating environmental stress model

Ocular surface desiccation is considered one of the initiating factors in DED (25). Pflugfelder's group was the first to develop a model in which ocular surface desiccation was an initiating factor of DED (26), an evaporative DED model. The environmental desiccating stress is generated by exposing the eyes of mice to a constant low-humidity air flow aimed at the face for 4 h every day. The longer the exposure to desiccating stress, the more prolonged the decreased tear secretion and the longer the recovery time. For example, if mice were exposed to the stress for 4 h per day for 1, 3 or 10 days, decreased tear production lasted for 2, 6 and 18 days, respectively, after discontinuing the desiccating stress (27). When the environmental stress is combined with muscarinic blockade to further reduce tear secretion the resulting DED is even more pronounced (7,26,28-31). Muscarinic blockade is often achieved by subcutaneous administration of scopolamine, which acts on the LGs to reduce tear secretion (7,25). The desiccation plus scopolamine more faithfully represents features of aqueous-deficient DED, including reductions in tear production, tear film stability, corneal staining, conjunctiva goblet cells, and increases in apoptosis of ocular surface epithelium and tear cytokines levels (32-35).

Modifications of this model have been introduced to generate chronic DED, akin to the clinical condition in humans. DED is induced by applying desiccating stress as above for 14 days and transferring the mice to an environment with normal humidity for 4 months (no muscarinic blockade is needed). The severity of DED peaks on day 14. The chronic phase is characterized by corneal epitheliopathy and inflammation, which persists for a long time, never returning to normal (36).

It is worth mentioning the different mechanisms by which desiccation and muscarinic blockade induce DED. Desiccation induces greater conjunctival CD3 (+) T-cell infiltration, and higher Th17-cell activity and Treg dysfunction than muscarinic blockade, while muscarinic blocked decreases tear volume more than desiccation, attenuates Th17 activity and enhances Th2 and Treg responses without affecting Th1 activity. There is increasing support for the combination of desiccation with muscarinic blockade than for either agent alone (25,37).

Models based on the aging of mice

Since age is a significant risk factor for DED (38,39), investigators have produced DED models based on their aging. C57BL/6 mice have been used successfully to study age-related chronic DED, which develops spontaneously in mice over 1 year old, which is characterized by a combination of DED and Meibomian gland dysfunction (MGD). This is thought to reflect the condition in humans older than 50 years with DED and MGD. Highlights of this model are increased production of MMP-9 and T-cell related cytokines in the ocular surface, and influx of CD4+ and CD8+ T cells into aged LG (40-42). There is also Meibomian gland dropout, increased meibocyte differentiation, and increased expression of cytokines. This model is suitable to the study the pathogenesis of age-related MGD (43-45). Pathophysiologically, they represent mixed models.

3. Rat models of DED

Rats, being bigger than mice, have relatively larger eyes, while at the same time these animals are still easy to handle and relatively inexpensive to maintain. Such difference in size, although not great, is nevertheless enough to comfortably perform functional assessments and morphological and molecular analyses of the eye. Not surprisingly, in many cases mouse models have inspired the corresponding rat models and vice versa.

Scopolamine model

A popular rat model has been based on the scopolamine mouse model presented above, an aqueous-deficient DED model. Rats were given subcutaneous scopolamine and housed in an environmentally controlled room with the standard temperature and low humidity (25±2%) for 28 days (46-48). The experiment rats displayed reduced tear production, reduced tear film stability, increased corneal fluorescent staining, and decreased conjunctiva goblet cells (48,49).

Visual display terminal user model

The computer and visual display terminal syndrome is a constellation of ocular (and extraocular) symptoms associated with prolonged use of visual display terminals (50,51). Evaporative DED is part of this syndrome, which is gaining importance with the widespread use of such technologies. Decreased blink frequency, the result of intense attention to the display screen, is considered central to its pathogenesis.

Investigators have established a rat dry eye model of corneal epithelial disorders by inducing improper tear dynamics and changes in blink frequency (52-54). To simulate the video display terminal, the rats are housed under continuous exposure to low-humidity airflow and placed on a balance swing for 7.5 h per day. These rats showed chronic reduction of tear secretion, impaired LG function with abnormal morphology, and superficial punctate keratopathy similar to that in humans. Potentially protective agents have already been tried, a topic of growing importance in a society dependent on computers for daily life functions (53).

Additional models

Several other rat DED models, some analogous to mouse models, have been reported but experience with them is limited or their technical description lacks detail. They include models generated by: Dacryoadenectomy (55-57); orchiectomy or ovariectomy (58,59); autoimmune dacryoadenitis induced by injecting LG extract (60,61); injection of botulinum B into the LG (62); exposure to tobacco smoke (63); and exposure to urban particulate matter (64).

4. Rabbit models of DED

Rabbits have significant advantages over rats and mice for the study of DED. Their larger globe size entails a larger exposed ocular surface, which makes it much easier to perform an array of DED tests such as the Schirmer tear test, tear break up time, fluorescein, rose Bengal staining, and corneal sensitivity using esthesiometry. In addition, as mentioned, rabbits are suitable to the study of drug molecules susceptible to hydrolysis by ocular surface esterase (65).

At least 12 novel rabbit models of DED have been reported. The majority of them attempt to reduce tear production by either removing the LGs or impeding their function. The most direct approaches include partial surgical resection of the LG (with or without concurrent removal of the nictitating membrane and Harderian gland) (66) or closure of the LG excretory ducts with cautery (67). Impairing LG function has been done by: Irradiation of the LGs (68); induction of dacryoadenitis by injecting the LGs with activated lymphocytes (69) or the plant mitogen Concanavalin A into one (70) or all (71) orbital LGs; injection of botulinum toxin A to the palpebral portion of the superior LG (72); or LG denervation (73). Pharmacological agents such as topical atropine (74) or benzalkonium chloride (75) have also been used to induce primarily aqueous-deficient DED in rabbits. Other methods include closing the Meibomian gland openings by cauterization (76); acute desiccation of the eye by manual prevention of blinking (77); and orchiectomy that depletes androgens, which are required for tear production and for the normal structure and function of the corneal epithelium (78). Below, we highlight some of the most commonly employed models.

Atropine model

Instillation of 1% atropine sulfate three times per day for five days is reported to rapidly produce typical dry eye manifestations including reduced tear production and abnormal fluorescein staining of the ocular surface. The atropine model is useful mainly to initially assess the protective activity and the ocular pharmacological profile of tear substitutes or ocular drugs (74,79-81). Our own experience with this model has been mixed, and we did not employ it for its lack of reproducibility.

Benzalkonium model

Topical administration of the ocular preservative benzalkonium chloride twice per day for 14 days (75) or three times per day for 4 weeks reported later (82,83) leads to DED. In particular, the experimental rabbits show corneal and conjunctiva damage, decreased aqueous tear basal secretion, loss of goblet cells, and deficiency of mucin-5 subtype AC (MUC5AC) (75).

Autoimmune dacryoadenitis model

This rabbit model, which resembles Sjὃgren syndrome, was developed by Gou et al (69) by co-culturing peripheral blood lymphocytes with purified acinar cells obtained from an autologous LG and injecting the activated lymphocytes into the contralateral gland. The injected LG shows an infiltrate dominated by CD4+T cells. The ensuing dacryoadenitis leads to reduced tear production, tear film instability and increased ocular surface staining reflecting disruptions in the ocular epithelium (7,69,84).

Main lacrimal gland ablation model

Initially developed to study autologous submandibular gland transfer for treating severe DED in this model, the bilateral lacrimal and Harderian glands and nictitating membrane were removed surgically. Because the aqueous phase of the mammalian tear film is produced by the combined activity of the main and accessory LGs, their removal reduces the total tear volume and protein content (66). However, another paper showed significant dry eye phenotypes associated with elevated ocular surface inflammation observed at 1 month. However, the tear production was not reduced and dry eye phenotypes and ocular surface inflammation gradually improved over a period of 4 months without any additional intervention (66,85,86). To induce severe DED, Li et al surgically removed the LG, Harderian gland and nictitating membrane, combined with burning the bulbar conjunctiva with 50% trichloroacetic acid (87). Similar models were also published, like one reporting closure of the LG excretory duct while removing both the nictitating membrane and Harderian gland (67,88).

Complete dacryoadenectomy model

In rabbit models of partial dacryoadenectomy, the removal of only the inferior lacrimal gland (ILG) (without the removal of the superior lacrimal gland) causes partial suppression of tear production leading to inconsistent results (66,67,85,87,89). In view of these limitations, we developed a practical method to completely remove the entire LG of the rabbit as we have described previously (90).

In brief, following the removal of the nictitating membrane, the orbital portion of the superior lacrimal gland was removed through a transcranial approach on the top of the skull; the palpebral portion of the superior lacrimal gland was removed through a transconjunctival approach after the upper eyelid was everted; and finally, the ILG was removed through a transcutaneous approach below the lower eyelid. Determination of TBUT, STT, tear osmolarity, and rose bengal staining of ocular surface showed that the DED induced by complete dacryoadenectomy was stable, chronic, and predominantly aqueous-deficient, thus recapitulating key clinical and histological features of human DED.

Concanavalin A-induced model

Nagelhout et al first reported a rabbit model of LG inflammation induced by a single injection of the T-cell mitogen Concanavalin A (ConA) into inferior LGs bilaterally (70). ConA injection elicits a heterophilic infiltrate, which causes severe and widespread tissue destruction. The consequent impaired tear production results in local inflammation of the ocular surface and is associated in measurable changes in aqueous-deficient dry eye clinical parameters.

While trying to reproduce this promising model, we observed that induction of DED by ConA was inconsistent. Exploration of this lack of reproducibility revealed that the blind transdermal injections of ConA into the ILG failed about 40% of the time to reach the LG. This was due to the varied anatomical location of the gland and its 4-fold variation in size. Additionally, the reduced function of the ILG following injection of ConA was compensated for by overproduction of tears by the superior lacrimal gland (71).

These observations led us to improve this model in a way that eliminated such variability. Specifically, we introduced ultrasound-guided injection of the ILG and added injection of ConA to both lobes of the superior lacrimal gland. The results were gratifying.

Briefly, under ultrasound guidance, all periorbital LGs of the rabbit receive one ConA injection; they include the ILG, the palpebral portion of the superior lacrimal gland, and the orbital portion of the superior lacrimal gland. It is critical that the success of the injection into the ILG be confirmed by a post-injection sonogram. A single injection causes acute DED, which lasts for about 1 week. To induce chronic DED, akin to that encountered clinically, ConA injections should be repeated weekly at least two more times for a total of three weeks. Additional injections lead to severe DED reflecting the almost fibrotic status of these glands with commensurate loss of tear-producing parenchyma (71,91).

Evaporative DED model (77,92,93). Miyake et al developed a rabbit model of evaporative DED (92) by injecting into the margins of the upper eyelid heat-inactivated Mycobacterium tuberculosis dissolved in complete Freud's adjuvant. Three injections are made one each into the nasal, center, and temporal sections of the eyelid. The result is Meibomian gland dysfunction leading to DED in about two weeks.

Acute desiccative stress model

Fujihara et al developed this model by keeping the rabbit eye open for 1-3 h using an eyelid speculum in controlled temperature and humidity (24˚C, 55% relative humidity) (77). The corneal damage can be severe, depending on the duration of the desiccative stress. The model has been used to screen drug effects or to study the contribution of desiccative stress to the pathophysiology of DED (63,94).

Closure of Meibomian gland orifices

The Meibomian glands provide most of the polar lipids that cover the tear film. Thus, removing the tear lipid layer promotes evaporation of tear water, a key event in evaporative DED. Gilbard et al (76) developed a rabbit model of DED by closing the orifices of the Meibomian glands. To achieve this, they cauterized each of these orifices in the upper and lower eyelids. The result was elevated tear osmolarity, decreased conjunctival goblet cells and corneal glycogen, and reduced corneal wetting (76,95).

Other rabbit models of Meibomian gland dysfunction that feature hyperkeratinized epithelium of the ductal orifices have been produced by topical application of epinephrine or by the systemic administration of 13-cis-retinonic acid (isotretinoin) (96-100). Similar to the rat, DED can be induced in the rabbit by surgical castration or ovariectomy; by injecting botulinum toxin A to the palpebral portion of the superior LG; and by LG denervation (72,73,101,102).

Based on the above, the rabbit appears to be one of the most suitable animals for DED studies. It offers the opportunity to study multiple pathophysiological mechanisms of aqueous-deficient DED in a manner that gives experimental flexibility. For example, the role of the lacrimal gland in DED can be examined from different angles, such as pharmacological intervention, immune damage or complete removal of the glands when tear production is assumed by the accessory lacrimal glands. In addition, the evaporative subtype can be studied probably better than in any other animal (see Table I).

The major drawback of rabbits is the paucity of analytical tools for the detection of proteins in the ocular surface. High-throughput transcriptomic methods offer only a partial compensation for two reasons. Changes in mRNA levels do not necessarily equate changes in the corresponding protein levels, and about one third of the rabbit genome is not yet annotated. In his context, murine models can play a complementary role, especially when genetically modified models are available that can help assess the role of specific pathways in DED.

5. Cat models of DED

Cat models have had limited use in the study of DED. In cats, the removal of the main LG decreased basal tear production as measured by the Schirmer test, but it failed to significantly change ocular surface signs (103). However, McLaughlin et al reported that surgical removal of the LG and third eyelid glands resulted in decreased tear production and clinical signs of DED including abnormal corneal fluorescein staining scores (104,105).

6. Dog models of DED

Considering their cost and the demanding effort of operating on dogs, there are only two dog models of DED, only one of which is the result of surgical intervention (ablation of lacrimal glands).

Spontaneous model of DED

Quimby first recognized in 1979 the similarities between Sjogren's syndrome in humans and severe keratoconjunctivitis sicca (KCS) in dogs displaying xerostomia, vaginal dryness, and having multiple serum antibodies (106). Subsequent studies have confirmed these observations (107,108) and demonstrated decreased apoptosis of the lymphocytes infiltrating the LG and increased apoptosis in lacrimal acinar and conjunctival epithelial cells. The American cocker spaniel is the breed with the highest relative prevalence of KCS (20.6%), followed by Lhasa Apso (12.7%) and Shih Tzu (11.5%). However, the commonly used laboratory dogs such as mixed-breed and beagles dogs have lower rates (109). The spontaneous canine dry eye model has been widely used to develop therapeutic interventions for both veterinary and human populations, exemplified by trials of topical application of cyclosporin A (103,110).

Main lacrimal gland ablation model

Bilateral removal of the orbital and the nictitans LGs in dogs induced KCS after 2 weeks, which lasted 6 weeks post-surgery. Tear production was reduced, and the characteristic clinical features of conjunctival hyperemia and accumulation of tenacious discharge were present (111,112).

7. Nonhuman primate models of DED

Among DED models, the monkey model is the one most similar to human DED. Monkeys have one main LG with an anatomical structure similar to that of humans (7). Notably, unlike other species, humans and nonhuman primates do not have a nictitating membrane. Despite these similarities, it has been challenging to develop satisfactory monkey models of DED.

Removal of the LG decreased tear secretion but was not accompanied by reproducible ocular surface damages (103,113). Qin et al developed a monkey dry eye model by complete removal of the principal LG and application of 50% trichloroacetic acid to the conjunctiva. These interventions decreased tear secretion, induced loss of goblet cells and infiltration by inflammatory cells within the ocular surface (114).

8. Conclusions

As already alluded to, it is utopian to expect the development of the ideal animal model of DED. This fundamental consideration notwithstanding, the existing models underscore three facts: First, no perfect or nearly perfect model exists. Second, each of the available models captures specific aspects of DED, reflecting the marked heterogeneity of this disease. And, third, existing models, limited as they may be, have greatly contributed to our progress in understanding and treating DED.

The choice of an animal model is important, because there are notable differences in the anatomical, biochemical, physiological, and morphological characteristics of the ocular surface between monkey, dog, rabbit, rat, cat, mouse, and humans. Four features determine the choice of a given model: The size of the eye, its biochemical composition, the available research reagents, and cost. The size of the eye can be a major factor for certain experimental needs. In general, the closer in size of the eye of the test animal to the human eye, the more useful this model will be. Smaller eyes can be technically difficult to dissect and obtain ocular tissues of sufficient size to evaluate their response to an intervention or to assay drug levels. In some instances, the size of some ocular tissues is so small that it may require analytical methods of unusually high sensitivity. It is often forgotten that, regardless of species, the eye is one of the smallest and structurally more complex organs.

The biochemical profile of the eye varies between species, and this can be in some cases a deciding factor in choosing a particular animal model. An instructive example is the expression of drug metabolizing enzymes such as esterases in ocular tissues. For drugs susceptible to esterase-catalyzed hydrolysis, rabbits and monkeys are closer to the human than mice and rabbits (115).

The availability of species-specific reagents weighs heavily in the choice of an animal model of DED. Mice have been the species of choice for modern molecular biology and, as a result, an abundance of reagents and research kits are commercially available for murine proteins compared to other species. In addition, the murine genome has a more robust database than, for example, the rabbit's. Similarly, mice are over-represented amongst the genetically engineered animal models.

Finally, cost can at times be prohibitive for small labs, the overwhelming majority of the global biomedical enterprise. For example, the eyes of non-human primates are in many aspects the closest to the human. However, such animals are expensive to acquire and house, and require dedicated personnel for their care and a sophisticated infrastructure to maintain them.

The continuous improvement of animal models for DED is limited by the functional and anatomical complexity of the lacrimal functional unit and by our still limited understanding of DED. The former makes it at times difficult to decipher the contribution of a specific component of this unit to DED. The dacryoadenectomy models presented earlier exemplify this limitation. Even when the periorbital lacrimal glands are removed or surgically denervated, there is residual tear production, whose origin remains uncertain. In reality, DED is not a single entity and, at the very least, its pathophysiology is diverse and perhaps only partially known. Thus, only models addressing specific subtypes of DED have been developed.

The landscape of DED animal models is evolving at a robust pace. Many groups have generated important results using currently available models. The strongest testament to their value is perhaps the many therapeutic agents, approved or under development, against DED whose development depended heavily on animal models. Nonetheless, the complexity of DED and the demands of modern pharmacology require more refined models to address difficult questions such as the identification of dominant mechanistic players or drug molecular targets. Given the talent and dedication of the worldwide ocular research community, continual improvements of these models are likely to occur.

Acknowledgements

The authors would like to acknowledge Dr Ernest Natke (Stony Brook University, Stony Brook, USA) for critically reading the manuscript.

Funding

The present study was supported in part by the National Institutes of Health (grant no. R44EY031193).

Availability of data and materials

Not applicable.

Authors' contributions

WH wrote most of the initial draft with BR, and contributed to revisions. KT reviewed the background literature and contributed to revisions. HP read the manuscript critically and provided general comments. RAH participated in writing and revisions. BR provided general guidance, wrote a number of sections, participated in revisions and finalized the manuscript. All authors contributed to writing and/or critically revising this review. Data authentication is not applicable. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

BR has an equity position in Medicon Pharmaceuticals, Inc., and Apis Therapeutics, LLC.

References

1 

The definition and classification of dry eye disease. Report of the definition and classification subcommittee of the international dry eye workshop (2007). Ocul Surf. 5:75–92. 2007.PubMed/NCBI View Article : Google Scholar

2 

Takamiya M, Weger BD, Schindler S, Beil T, Yang L, Armant O, Ferg M, Schlunck G, Reinhard T, Dickmeis T, et al: Molecular description of eye defects in the zebrafish Pax6b mutant, sunrise, reveals a Pax6b-dependent genetic network in the developing anterior chamber. PLoS One. 10(e0117645)2015.PubMed/NCBI View Article : Google Scholar

3 

Baldassano VF Jr: Ocular manifestations of rheumatic diseases. Curr Opin Ophthalmol. 9:85–88. 1998.PubMed/NCBI View Article : Google Scholar

4 

Kemeny-Beke A and Szodoray P: Ocular manifestations of rheumatic diseases. Int Ophthalmol. 40:503–510. 2020.PubMed/NCBI View Article : Google Scholar

5 

Zoukhri D and Kublin CL: Impaired neurotransmitter release from lacrimal and salivary gland nerves of a murine model of Sjögren's syndrome. Invest Ophthalmol Vis Sci. 42:925–932. 2001.PubMed/NCBI

6 

Toda I, Sullivan BD, Wickham LA and Sullivan DA: Gender- and androgen-related influence on the expression of proto-oncogene and apoptotic factor mRNAs in lacrimal glands of autoimmune and non-autoimmune mice. J Steroid Biochem Mol Biol. 71:49–61. 1999.PubMed/NCBI View Article : Google Scholar

7 

Schrader S, Mircheff AK and Geerling G: Animal models of dry eye. Dev Ophthalmol. 41:298–312. 2008.PubMed/NCBI View Article : Google Scholar

8 

Watanabe-Fukunaga R, Brannan CI, Copeland NG, Jenkins NA and Nagata S: Lymphoproliferation disorder in mice explained by defects in Fas antigen that mediates apoptosis. Nature. 356:314–317. 1992.PubMed/NCBI View Article : Google Scholar

9 

Singer GG and Abbas AK: The fas antigen is involved in peripheral but not thymic deletion of T lymphocytes in T cell receptor transgenic mice. Immunity. 1:365–371. 1994.PubMed/NCBI View Article : Google Scholar

10 

Törnwall J, Lane TE, Fox RI and Fox HS: T cell attractant chemokine expression initiates lacrimal gland destruction in nonobese diabetic mice. Lab Invest. 79:1719–1726. 1999.PubMed/NCBI

11 

Robinson CP, Cornelius J, Bounous DE, Yamamoto H, Humphreys-Beher MG and Peck AB: Characterization of the changing lymphocyte populations and cytokine expression in the exocrine tissues of autoimmune NOD mice. Autoimmunity. 27:29–44. 1998.PubMed/NCBI View Article : Google Scholar

12 

Yamamoto H, Sims NE, Macauley SP, Nguyen KH, Nakagawa Y and HumphreysBeher MG: Alterations in the secretory response of non-obese diabetic (NOD) mice to muscarinic receptor stimulation. Clin Immunol Immunop. 78:245–255. 1996.PubMed/NCBI View Article : Google Scholar

13 

Coursey TG, Bian F, Zaheer M, Pflugfelder SC, Volpe EA and de Paiva CS: Age-related spontaneous lacrimal keratoconjunctivitis is accompanied by dysfunctional T regulatory cells. Mucosal Immunol. 10:743–756. 2017.PubMed/NCBI View Article : Google Scholar

14 

Takahashi M, Ishimaru N, Yanagi K, Haneji N, Saito I and Hayashi Y: High incidence of autoimmune dacryoadenitis in male non-obese diabetic (NOD) mice depending on sex steroid. Clin Exp Immunol. 109:555–561. 1997.PubMed/NCBI View Article : Google Scholar

15 

Vendramini ACL, Soo C and Sullivan DA: Testosterone-induced suppression of autoimmune disease in lacrimal tissue of a mouse model (NZB/NZW F1) of Sjögren's syndrome. Invest Ophthalmol Vis Sci. 32:3002–3006. 1991.PubMed/NCBI

16 

Sullivan DA and Edwards JA: Androgen stimulation of lacrimal gland function in mouse models of Sjögren's syndrome. J Steroid Biochem. 60:237–245. 1997.PubMed/NCBI View Article : Google Scholar

17 

Haneji N, Nakamura T, Takio K, Yanagi K, Higashiyama H, Saito I, Noji S, Sugino H and Hayashi Y: Identification of alpha-fodrin as a candidate autoantigen in primary Sjögren's syndrome. Science. 276:604–607. 1997.PubMed/NCBI View Article : Google Scholar

18 

Song XJ, Li DQ, Farley W, Luo LH, Heuckeroth RO, Milbrandt J and Pflugfelder SC: Neurturin-deficient mice develop dry eye and keratoconjunctivitis sicca. Invest Ophthalmol Vis Sci. 44:4223–4229. 2003.PubMed/NCBI View Article : Google Scholar

19 

McCartney-Francis NL, Mizel DE, Frazier-Jessen M, Kulkarni AB, McCarthy JB and Wahl SM: Lacrimal gland inflammation is responsible for ocular pathology in TGF-beta 1 null mice. Am J Pathol. 151:1281–1288. 1997.PubMed/NCBI

20 

McCartney-Francis NL, Mizel DE, Redman RS, Frazier-Jessen M, Panek RB, Kulkarni AB, Ward JM, McCarthy JB and Wahl SM: Autoimmune Sjögren's-like lesions in salivary glands of TGF-beta1-deficient mice are inhibited by adhesion-blocking peptides. J Immunol. 157:1306–1312. 1996.PubMed/NCBI

21 

Konno A, Takada K, Saegusa J and Takiguchi M: Presence of B7-2+ dendritic cells and expression of Th1 cytokines in the early development of sialodacryoadenitis in the IqI/Jic mouse model of primary Sjörgren's syndrome. Autoimmunity. 36:247–254. 2003.PubMed/NCBI View Article : Google Scholar

22 

Li H, Dai M and Zhuang Y: A T cell intrinsic role of Id3 in a mouse model for primary Sjogren's syndrome. Immunity. 21:551–560. 2004.PubMed/NCBI View Article : Google Scholar

23 

Shinomiya K, Ueta M and Kinoshita S: A new dry eye mouse model produced by exorbital and intraorbital lacrimal gland excision. Sci Rep. 8(1483)2018.PubMed/NCBI View Article : Google Scholar

24 

Nakamura T, Hata Y, Nagata M, Yokoi N, Yamaguchi S, Kaku T and Kinoshita S: JBP485 promotes tear and mucin secretion in ocular surface epithelia. Sci Rep. 5(10248)2015.PubMed/NCBI View Article : Google Scholar

25 

Bron AJ, de Paiva CS, Chauhan SK, Bonini S, Gabison EE, Jain S, Knop E, Markoulli M, Ogawa Y, Perez V, et al: TFOS DEWS II pathophysiology report. Ocul Surf. 15:438–510. 2017.PubMed/NCBI View Article : Google Scholar

26 

Dursun D, Wang M, Monroy D, Li DQ, Lokeshwar BL, Stern ME and Pflugfelder SC: A mouse model of keratoconjunctivitis sicca. Invest Ophthalmol Vis Sci. 43:632–638. 2002.PubMed/NCBI

27 

Sano K, Kawashima M, Imada T, Suzuki T, Nakamura S, Mimura M, Tanaka KF and Tsubota K: Enriched environment alleviates stress-induced dry-eye through the BDNF axis. Sci Rep. 9(3422)2019.PubMed/NCBI View Article : Google Scholar

28 

Stewart P, Chen Z, Farley W, Olmos L and Pflugfelder SC: Effect of experimental dry eye on tear sodium concentration in the mouse. Eye Contact Lens. 31:175–178. 2005.PubMed/NCBI View Article : Google Scholar

29 

Yeh S, Song XJ, Farley W, Li DQ, Stern ME and Pflugfelder SC: Apoptosis of ocular surface cells in experimentally induced dry eye. Invest Ophthalmol Vis Sci. 44:124–129. 2003.PubMed/NCBI View Article : Google Scholar

30 

Niederkorn JY, Stern ME, Pflugfelder SC, De Paiva CS, Corrales RM, Gao J and Siemasko K: Desiccating stress induces T cell-mediated Sjögren's syndrome-like lacrimal keratoconjunctivitis. J Immunol. 176:3950–3957. 2006.PubMed/NCBI View Article : Google Scholar

31 

Barabino S, Shen L, Chen L, Rashid S, Rolando M and Dana MR: The controlled-environment chamber: A new mouse model of dry eye. Invest Ophthalmol Vis Sci. 46:2766–2771. 2005.PubMed/NCBI View Article : Google Scholar

32 

De Paiva CS, Corrales RM, Villarreal AL, Farley WJ, Li DQ, Stern ME and Pflugfelder SC: Corticosteroid and doxycycline suppress MMP-9 and inflammatory cytokine expression, MAPK activation in the corneal epithelium in experimental dry eye. Exp Eye Res. 83:526–535. 2006.PubMed/NCBI View Article : Google Scholar

33 

Corrales RM, de Paiva CS, Li DQ, Farley WJ, Henriksson JT, Bergmanson JP and Pflugfelder SC: Entrapment of conjunctival goblet cells by desiccation-induced cornification. Invest Ophthalmol Vis Sci. 52:3492–3499. 2011.PubMed/NCBI View Article : Google Scholar

34 

De Paiva CS, Yoon KC, Pangelinan SB, Pham S, Puthenparambil LM, Chuang EY, Farley WJ, Stern ME, Li DQ and Pflugfelder SC: Cleavage of functional IL-2 receptor alpha chain (CD25) from murine corneal and conjunctival epithelia by MMP-9. J Inflamm (Lond). 6(31)2009.PubMed/NCBI View Article : Google Scholar

35 

Coursey TG, Tukler Henriksson J, Barbosa FL, de Paiva CS and Pflugfelder SC: Interferon-γ-induced unfolded protein response in conjunctival goblet cells as a cause of mucin deficiency in Sjögren syndrome. Am J Pathol. 186:1547–1558. 2016.PubMed/NCBI View Article : Google Scholar

36 

Chen Y, Zhang X, Yang L, Li M, Li B, Wang W and Sheng M: Decreased PPAR-γ expression in the conjunctiva and increased expression of TNF-α and IL-1β in the conjunctiva and tear fluid of dry eye mice. Mol Med Rep. 9:2015–2023. 2014.PubMed/NCBI View Article : Google Scholar

37 

Chen Y, Chauhan SK, Lee HS, Stevenson W, Schaumburg CS, Sadrai Z, Saban DR, Kodati S, Stern ME and Dana R: Effect of desiccating environmental stress versus systemic muscarinic AChR blockade on dry eye immunopathogenesis. Invest Ophthalmol Vis Sci. 54:2457–2464. 2013.PubMed/NCBI View Article : Google Scholar

38 

Schein OD, Muñoz B, Tielsch JM, Bandeen-Roche K and West S: Prevalence of dry eye among the elderly. Am J Ophthalmol. 124:723–728. 1997.PubMed/NCBI View Article : Google Scholar

39 

Moss SE, Klein R and Klein BE: Incidence of dry eye in an older population. Arch Ophthalmol. 122:369–373. 2004.PubMed/NCBI View Article : Google Scholar

40 

McClellan AJ, Volpe EA, Zhang X, Darlington GJ, Li DQ, Pflugfelder SC and de Paiva CS: Ocular surface disease and dacryoadenitis in aging C57BL/6 mice. Am J Pathol. 184:631–643. 2014.PubMed/NCBI View Article : Google Scholar

41 

de Paiva CS: Effects of aging in dry eye. Int Ophthalmol Clin. 57:47–64. 2017.PubMed/NCBI View Article : Google Scholar

42 

Ding J and Sullivan DA: Aging and dry eye disease. Exp Gerontol. 47:483–490. 2012.PubMed/NCBI View Article : Google Scholar

43 

Jester BE, Nien CJ, Winkler M, Brown DJ and Jester JV: Volumetric reconstruction of the mouse meibomian gland using high-resolution nonlinear optical imaging. Anat Rec (Hoboken). 294:185–192. 2011.PubMed/NCBI View Article : Google Scholar

44 

Nien CJ, Paugh JR, Massei S, Wahlert AJ, Kao WW and Jester JV: Age-related changes in the meibomian gland. Exp Eye Res. 89:1021–1027. 2009.PubMed/NCBI View Article : Google Scholar

45 

Parfitt GJ, Xie Y, Geyfman M, Brown DJ and Jester JV: Absence of ductal hyper-keratinization in mouse age-related meibomian gland dysfunction (ARMGD). Aging (Albany NY). 5:825–834. 2013.PubMed/NCBI View Article : Google Scholar

46 

Ru Y, Huang Y, Liu H, Du J, Meng Z, Dou Z, Liu X, Wei RH, Zhang Y and Zhao S: α-Melanocyte-stimulating hormone ameliorates ocular surface dysfunctions and lesions in a scopolamine-induced dry eye model via PKA-CREB and MEK-Erk pathways. Sci Rep. 5(18619)2015.PubMed/NCBI View Article : Google Scholar

47 

Chen W, Zhao K, Li X and Yoshitomi T: Keratoconjunctivitis sicca modifies epithelial stem cell proliferation kinetics in conjunctiva. Cornea. 26:1101–1106. 2007.PubMed/NCBI View Article : Google Scholar

48 

Viau S, Maire MA, Pasquis B, Grégoire S, Fourgeux C, Acar N, Bretillon L, Creuzot-Garcher CP and Joffre C: Time course of ocular surface and lacrimal gland changes in a new scopolamine-induced dry eye model. Graefes Arch Clin Exp Ophthalmol. 246:857–867. 2008.PubMed/NCBI View Article : Google Scholar

49 

Viau S, Maire MA, Pasquis B, Grégoire S, Acar N, Bron AM, Bretillon L, Creuzot-Garcher CP and Joffre C: Efficacy of a 2-month dietary supplementation with polyunsaturated fatty acids in dry eye induced by scopolamine in a rat model. Graefes Arch Clin Exp Ophthalmol. 247:1039–1050. 2009.PubMed/NCBI View Article : Google Scholar

50 

Nakaishi H and Yamada Y: Abnormal tear dynamics and symptoms of eyestrain in operators of visual display terminals. Occup Environ Med. 56:6–9. 1999.PubMed/NCBI View Article : Google Scholar

51 

Uchino M, Kawashima M, Uchino Y, Tsubota K and Yokoi N: Association between tear film break up time and blink interval in visual display terminal users. Int J Ophthalmol. 11:1691–1697. 2018.PubMed/NCBI View Article : Google Scholar

52 

Nakamura S, Kinoshita S, Yokoi N, Ogawa Y, Shibuya M, Nakashima H, Hisamura R, Imada T, Imagawa T, Uehara M, et al: Lacrimal hypofunction as a new mechanism of dry eye in visual display terminal users. PLoS One. 5(e11119)2010.PubMed/NCBI View Article : Google Scholar

53 

Nakamura S, Shibuya M, Nakashima H, Imagawa T, Uehara M and Tsubota K: D-beta-hydroxybutyrate protects against corneal epithelial disorders in a rat dry eye model with jogging board. Invest Ophthalmol Vis Sci. 46:2379–2387. 2005.PubMed/NCBI View Article : Google Scholar

54 

Imada T, Nakamura S, Kitamura N, Shibuya I and Tsubota K: Oral administration of royal jelly restores tear secretion capacity in rat blink-suppressed dry eye model by modulating lacrimal gland function. PLoS One. 9(e106338)2014.PubMed/NCBI View Article : Google Scholar

55 

Xiao F, Cui H and Zhong X: Beneficial effect of daidzin in dry eye rat model through the suppression of inflammation and oxidative stress in the cornea. Saudi J Biol Sci. 25:832–837. 2018.PubMed/NCBI View Article : Google Scholar

56 

Meng ID, Barton ST, Mecum NE and Kurose M: Corneal sensitivity following lacrimal gland excision in the rat. Invest Ophthalmol Vis Sci. 56:3347–3354. 2015.PubMed/NCBI View Article : Google Scholar

57 

Park B, Jo K, Lee TG, Lee IS, Kim JS and Kim CS: Polygonum cuspidatum stem extract (PSE) ameliorates dry eye disease by inhibiting inflammation and apoptosis. J Exerc Nutrition Biochem. 23:14–22. 2019.PubMed/NCBI View Article : Google Scholar

58 

Peng QH, Yao XL, Wu QL, Tan HY and Zhang JR: Effects of extract of Buddleja officinalis eye drops on androgen receptors of lacrimal gland cells of castrated rats with dry eye. Int J Ophthalmol. 3:43–48. 2010.PubMed/NCBI View Article : Google Scholar

59 

Li L, Jin D, Gao J, Wang L, Liu X, Wang J and Xu Z: Activities of autonomic neurotransmitters in meibomian gland tissues are associated with menopausal dry eye. Neural Regen Res. 7:2761–2769. 2012.PubMed/NCBI View Article : Google Scholar

60 

Liu SH, Sakai F, Prendergast RA and Silverstein AM: Experimental autoimmune dacryoadenitis. II. Harderian gland disease in the rat. Invest Ophthalmol Vis Sci. 28:276–280. 1987.PubMed/NCBI

61 

Hou A, Bose T, Chandy KG and Tong L: A chronic autoimmune dry eye rat model with increase in effector memory T cells in eyeball tissue. J Vis Exp. (55592)2017.PubMed/NCBI View Article : Google Scholar

62 

Zhang FD, Hao ZQ, Gao W and Xing YQ: Effect of topical 0.05% cyclosporine A on the tear protein lacritin in a rat model of dry eye. Int J Ophthalmol. 12:189–193. 2019.PubMed/NCBI View Article : Google Scholar

63 

Higuchi A, Inoue H, Kaneko Y, Oonishi E and Tsubota K: Selenium-binding lactoferrin is taken into corneal epithelial cells by a receptor and prevents corneal damage in dry eye model animals. Sci Rep. 6(36903)2016.PubMed/NCBI View Article : Google Scholar

64 

Lee TG, Hyun SW, Jo K, Park B, Lee IS, Song SJ and Kim CS: Achyranthis radix extract improves urban particulate matter-induced dry eye disease. Int J Environ Res Public Health. 16(3229)2019.PubMed/NCBI View Article : Google Scholar

65 

Schechter JE, Warren DW and Mircheff AK: A lacrimal gland is a lacrimal gland, but rodent's and rabbit's are not human. Ocul Surf. 8:111–134. 2010.PubMed/NCBI View Article : Google Scholar

66 

Chen ZY, Liang QF and Yu GY: Establishment of a rabbit model for keratoconjunctivitis sicca. Cornea. 30:1024–1029. 2011.PubMed/NCBI View Article : Google Scholar

67 

Gilbard JP, Rossi SR and Gray KL: A new rabbit model for keratoconjunctivitis sicca. Invest Ophthalmol Vis Sci. 28:225–228. 1987.PubMed/NCBI

68 

Beutel J, Schroder C, von Hof K, Rades D, Kosmehl H, Wedel T, Sieg P, Geerling G and Hakim SG: Pharmacological prevention of radiation-induced dry eye-an experimental study in a rabbit model. Graefes Arch Clin Exp Ophthalmol. 245:1347–1355. 2007.PubMed/NCBI View Article : Google Scholar

69 

Guo Z, Song D, Azzarolo AM, Schechter JE, Warren DW, Wood RL, Mircheff AK and Kaslow HR: Autologous lacrimal-lymphoid mixed-cell reactions induce dacryoadenitis in rabbits. Exp Eye Res. 71:23–31. 2000.PubMed/NCBI View Article : Google Scholar

70 

Nagelhout TJ, Gamache DA, Roberts L, Brady MT and Yanni JM: Preservation of tear film integrity and inhibition of corneal injury by dexamethasone in a rabbit model of lacrimal gland inflammation-induced dry eye. J Ocul Pharmacol Ther. 21:139–148. 2005.PubMed/NCBI View Article : Google Scholar

71 

Honkanen RA, Huang L, Xie G and Rigas B: Phosphosulindac is efficacious in an improved concanavalin A-based rabbit model of chronic dry eye disease. Transl Res. 198:58–72. 2018.PubMed/NCBI View Article : Google Scholar

72 

Demetriades AM, Leyngold IM, D'Anna S, Eghrari AO, Emmert DG, Grant MP and Merbs SL: Intraglandular injection of botulinum toxin a reduces tear production in rabbits. Ophthalmic Plast Reconstr Surg. 29:21–24. 2013.PubMed/NCBI View Article : Google Scholar

73 

Toshida H, Nguyen DH, Beuerman RW and Murakami A: Evaluation of novel dry eye model: Preganglionic parasympathetic denervation in rabbit. Invest Ophthalmol Vis Sci. 48:4468–4475. 2007.PubMed/NCBI View Article : Google Scholar

74 

Burgalassi S, Panichi L, Chetoni P, Saettone MF and Boldrini E: Development of a simple dry eye model in the albino rabbit and evaluation of some tear substitutes. Ophthalmic Res. 31:229–235. 1999.PubMed/NCBI View Article : Google Scholar

75 

Xiong C, Chen D, Liu J, Liu B, Li N, Zhou Y, Liang X, Ma P, Ye C, Ge J and Wang Z: A rabbit dry eye model induced by topical medication of a preservative benzalkonium chloride. Invest Ophthalmol Vis Sci. 49:1850–1856. 2008.PubMed/NCBI View Article : Google Scholar

76 

Gilbard JP, Rossi SR and Heyda KG: Tear film and ocular surface changes after closure of the meibomian gland orifices in the rabbit. Ophthalmology. 96:1180–1186. 1989.PubMed/NCBI View Article : Google Scholar

77 

Fujihara T, Nagano T, Nakamura M and Shirasawa E: Establishment of a rabbit short-term dry eye model. J Ocul Pharmacol Ther. 11:503–508. 1995.PubMed/NCBI View Article : Google Scholar

78 

Yao XL, Peng QH, Peng J, Tan HY, Wu QL, Wu DL, Chen M, Li CK, Li D and Zhu HA: Effects of extract of Buddleja officinalis on partial inflammation of lacrimal gland in castrated rabbits with dry eye. Int J Ophthalmol. 3:114–119. 2010.PubMed/NCBI View Article : Google Scholar

79 

Karn PR, Kim HD, Kang H, Sun BK, Jin SE and Hwang SJ: Supercritical fluid-mediated liposomes containing cyclosporin A for the treatment of dry eye syndrome in a rabbit model: Comparative study with the conventional cyclosporin A emulsion. Int J Nanomedicine. 9:3791–3800. 2014.PubMed/NCBI View Article : Google Scholar

80 

Bucolo C, Fidilio A, Fresta CG, Lazzara F, Platania CBM, Cantarella G, Di Benedetto G, Burgaletto C, Bernardini R, Piazza C, et al: Ocular pharmacological profile of hydrocortisone in dry eye disease. Front Pharmacol. 10(1240)2019.PubMed/NCBI View Article : Google Scholar

81 

Tampucci S, Monti D, Burgalassi S, Terreni E, Zucchetti E, Baldacci F and Chetoni P: Effect of 5-Oxo-2-pyrrolidinecarboxylic Acid (PCA) as a new topically applied agent for dry eye syndrome treatment. Pharmaceutics. 10(137)2018.PubMed/NCBI View Article : Google Scholar

82 

Chen HC, Chen ZY, Wang TJ, Drew VJ, Tseng CL, Fang HW and Lin FH: Herbal supplement in a buffer for dry eye syndrome treatment. Int J Mol Sci. 18(1697)2017.PubMed/NCBI View Article : Google Scholar

83 

Huang HY, Wang MC, Chen ZY, Chiu WY, Chen KH, Lin IC, Yang WV, Wu CC and Tseng CL: Gelatin-epigallocatechin gallate nanoparticles with hyaluronic acid decoration as eye drops can treat rabbit dry-eye syndrome effectively via inflammatory relief. Int J Nanomedicine. 13:7251–7273. 2018.PubMed/NCBI View Article : Google Scholar

84 

Zhou L, Wei R, Zhao P, Koh SK, Beuerman RW and Ding C: Proteomic analysis revealed the altered tear protein profile in a rabbit model of Sjögren's syndrome-associated dry eye. Proteomics. 13:2469–2481. 2013.PubMed/NCBI View Article : Google Scholar

85 

Bhattacharya D, Ning Y, Zhao F, Stevenson W, Chen R, Zhang J and Wang M: Tear production after bilateral main lacrimal gland resection in rabbits. Invest Ophthalmol Vis Sci. 56:7774–7783. 2015.PubMed/NCBI View Article : Google Scholar

86 

Ning Y, Bhattacharya D, Jones RE, Zhao F, Chen R, Zhang J and Wang M: Evaluating the functionality of conjunctiva using a rabbit dry eye model. J Ophthalmol. 2016(3964642)2016.PubMed/NCBI View Article : Google Scholar

87 

Li N, Deng X, Gao Y, Zhang S, He M and Zhao D: Establishment of the mild, moderate and severe dry eye models using three methods in rabbits. BMC Ophthalmol. 13(50)2013.PubMed/NCBI View Article : Google Scholar

88 

Gilbard JP, Rossi SR, Gray KL, Hanninen LA and Kenyon KR: Tear film osmolarity and ocular surface disease in two rabbit models for keratoconjunctivitis sicca. Invest Ophthalmol Vis Sci. 29:374–378. 1988.PubMed/NCBI

89 

Odaka A, Toshida H, Ohta T, Tabuchi N, Koike D, Suto C and Murakami A: Efficacy of retinol palmitate eye drops for dry eye in rabbits with lacrimal gland resection. Clin Ophthalmol. 6:1585–1593. 2012.PubMed/NCBI View Article : Google Scholar

90 

Honkanen RA, Huang W, Huang L, Kaplowitz K, Weissbart S and Rigas B: A new rabbit model of chronic dry eye disease induced by complete surgical dacryoadenectomy. Curr Eye Res. 44:863–872. 2019.PubMed/NCBI View Article : Google Scholar

91 

Honkanen RA, Huang L and Rigas B: A rabbit model of aqueous-deficient dry eye disease induced by concanavalin A injection into the lacrimal glands: Application to drug efficacy studies. J Vis Exp, 2020.

92 

Miyake H, Oda T, Katsuta O, Seno M and Nakamura M: A novel model of meibomian gland dysfunction induced with complete freund's adjuvant in rabbits. Vision (Basel). 1(10)2017.PubMed/NCBI View Article : Google Scholar

93 

Niamprem P, Teapavarapruk P, Srinivas SP and Tiyaboonchai W: Impact of nanostructured lipid carriers as an artificial tear film in a rabbit evaporative dry eye model. Cornea. 38:485–491. 2019.PubMed/NCBI View Article : Google Scholar

94 

Sher I, Tzameret A, Szalapak AM, Carmeli T, Derazne E, Avni-Zauberman N, Marcovich AL, Simon GB and Rotenstreich Y: Multimodal assessment of corneal erosions using optical coherence tomography and automated grading of fluorescein staining in a rabbit dry eye model. Transl Vis Sci Technol. 8(27)2019.PubMed/NCBI View Article : Google Scholar

95 

Eom Y, Han JY, Kang B, Hwang HS, Lee HK, Kim HM and Song JS: Meibomian glands and ocular surface changes after closure of meibomian gland orifices in rabbits. Cornea. 37:218–226. 2018.PubMed/NCBI View Article : Google Scholar

96 

Lambert RW and Smith RE: Pathogenesis of blepharoconjunctivitis complicating 13-cis-retinoic acid (isotretinoin) therapy in a laboratory model. Invest Ophthalmol Vis Sci. 29:1559–1564. 1988.PubMed/NCBI

97 

Jester JV, Nicolaides N, Kiss-Palvolgyi I and Smith RE: Meibomian gland dysfunction. II. The role of keratinization in a rabbit model of MGD. Invest Ophthalmol Vis Sci. 30:936–945. 1989.PubMed/NCBI

98 

Jester JV, Rife L, Nii D, Luttrull JK, Wilson L and Smith RE: In vivo biomicroscopy and photography of meibomian glands in a rabbit model of meibomian gland dysfunction. Invest Ophthalmol Vis Sci. 22:660–667. 1982.PubMed/NCBI

99 

Lambert R and Smith RE: Hyperkeratinization in a rabbit model of meibomian gland dysfunction. Am J Ophthalmol. 105:703–705. 1988.PubMed/NCBI View Article : Google Scholar

100 

Knop E, Knop N, Millar T, Obata H and Sullivan DA: The international workshop on meibomian gland dysfunction: Report of the subcommittee on anatomy, physiology, and pathophysiology of the meibomian gland. Invest Ophthalmol Vis Sci. 52:1938–1978. 2011.PubMed/NCBI View Article : Google Scholar

101 

Qin G, Zhou Y, Peng J, Zhang Y, Peng X, Peng Q and Yang Y: The effect of Buddleja officinalis maxim eye drops on morphology and apoptosis in lacrimal gland of experimental dry eye rabbit model. J Ophthalmol. 2019(5916243)2019.PubMed/NCBI View Article : Google Scholar

102 

Ma M, Yuan Q, Ye L, Liu K, Ye L, Min YL, Jiang N, Li Q, Shi W, Xu X, et al: An experimental study of amniotic lacrimal duct stents in the treatment of perimenopausal female rabbits with dry eye. Mol Med Rep. 19:1056–1064. 2019.PubMed/NCBI View Article : Google Scholar

103 

Barabino S and Dana MR: Animal models of dry eye: A critical assessment of opportunities and limitations. Invest Ophthalmol Vis Sci. 45:1641–1646. 2004.PubMed/NCBI View Article : Google Scholar

104 

McLaughlin SA, Brightman AH II, Helper LC, Primm ND, Brown MG and Greeley S: Effect of removal of lacrimal and third eyelid glands on Schirmer tear test results in cats. J Am Vet Med Assoc. 193:820–822. 1988.PubMed/NCBI

105 

Dota A, Takaoka-Shichijo Y and Nakamura M: Gefarnate stimulates mucin-like glycoprotein secretion in conjunctival tissue and ameliorates corneal epithelial damage in animal dry-eye models. Clin Ophthalmol. 7:211–217. 2013.PubMed/NCBI View Article : Google Scholar

106 

Quimby FW, Schwartz RS, Poskitt T and Lewis RM: A disorder of dogs resembling Sjögren's syndrome. Clin Immunol Immunopathol. 12:471–476. 1979.PubMed/NCBI View Article : Google Scholar

107 

Kaswan RL, Martin CL and Chapman WL Jr: Keratoconjunctivitis sicca: Histopathologic study of nictitating membrane and lacrimal glands from 28 dogs. Am J Vet Res. 45:112–118. 1984.PubMed/NCBI

108 

Kaswan RL, Martin CL and Dawe DL: Keratoconjunctivitis sicca: Immunological evaluation of 62 canine cases. Am J Vet Res. 46:376–383. 1985.PubMed/NCBI

109 

Kaswan R, Pappas C Jr, Wall K and Hirsh SG: Survey of canine tear deficiency in veterinary practice. Adv Exp Med Biol. 438:931–939. 1998.PubMed/NCBI View Article : Google Scholar

110 

Gao J, Schwalb TA, Addeo JV, Ghosn CR and Stern ME: The role of apoptosis in the pathogenesis of canine keratoconjunctivitis sicca: The effect of topical cyclosporin A therapy. Cornea. 17:654–663. 1998.PubMed/NCBI View Article : Google Scholar

111 

Helper LC, Magrane WG, Koehm J and Johnson R: Surgical induction of keratoconjunctivitis sicca in the dog. J Am Vet Med Assoc. 165:172–174. 1974.PubMed/NCBI

112 

Moore CP, McHugh JB, Thorne JG and Phillips TE: Effect of cyclosporine on conjunctival mucin in a canine keratoconjunctivitis sicca model. Invest Ophthalmol Vis Sci. 42:653–659. 2001.PubMed/NCBI

113 

Maitchouk DY, Beuerman RW, Ohta T, Stern M and Varnell RJ: Tear production after unilateral removal of the main lacrimal gland in squirrel monkeys. Arch Ophthalmol. 118:246–252. 2000.PubMed/NCBI View Article : Google Scholar

114 

Qin Y, Tan X, Zhang Y, Jie Y, Labbe A and Pan Z: A new nonhuman primate model of severe dry eye. Cornea. 33:510–517. 2014.PubMed/NCBI View Article : Google Scholar

115 

Wong CC, Cheng KW, Xie G, Zhou D, Zhu CH, Constantinides PP and Rigas B: Carboxylesterases 1 and 2 hydrolyze phospho-nonsteroidal anti-inflammatory drugs: Relevance to their pharmacological activity. J Pharmacol Exp Ther. 340:422–432. 2012.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

December-2021
Volume 22 Issue 6

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Huang W, Tourmouzis K, Perry H, Honkanen RA and Rigas B: Animal models of dry eye disease: Useful, varied and evolving (Review). Exp Ther Med 22: 1394, 2021
APA
Huang, W., Tourmouzis, K., Perry, H., Honkanen, R.A., & Rigas, B. (2021). Animal models of dry eye disease: Useful, varied and evolving (Review). Experimental and Therapeutic Medicine, 22, 1394. https://doi.org/10.3892/etm.2021.10830
MLA
Huang, W., Tourmouzis, K., Perry, H., Honkanen, R. A., Rigas, B."Animal models of dry eye disease: Useful, varied and evolving (Review)". Experimental and Therapeutic Medicine 22.6 (2021): 1394.
Chicago
Huang, W., Tourmouzis, K., Perry, H., Honkanen, R. A., Rigas, B."Animal models of dry eye disease: Useful, varied and evolving (Review)". Experimental and Therapeutic Medicine 22, no. 6 (2021): 1394. https://doi.org/10.3892/etm.2021.10830