Skip to main content

Inflammatory bowel disease and the associated risk of dry eye and ocular surface injury: a nationwide matched cohort study

Abstract

Background

Inflammatory bowel disease (IBD) is associated with lacrimal gland dysfunction and ocular inflammation. The objective of this research was to elucidate the temporal relationships between IBD, dry eye disease (DED), and corneal surface damage.

Methods

In a matched nationwide cohort study, we evaluated the risk of DED and corneal surface damage associated with IBD. Multivariable Cox proportional hazards regression analyses were implemented to estimate the risk of ocular complications.

Results

A total of 54,293 matched pairs were included for analyses. The median follow-up time was 8.3 years (interquartile range: 5.5 – 10.5). The period incidence of DED was 8.18 and 5.42 per 1000 person-years in the IBD and non-IBD groups, respectively. After adjusting for confounders, statistically significant associations were found between IBD and DED [adjusted hazard ratio (aHR): 1.43, 95% confidence interval (CI): 1.35 – 1.51, p < 0.0001], Sjögren’s syndrome-related (aHR: 1.67, 95% CI:1.46 – 1.90, p < 0.0001) and non-Sjögren’s syndrome-related subtypes (aHR: 1.38, 95% CI: 1.30 – 1.46, p < 0.0001). Furthermore, increased risks of corneal surface damage (aHR: 1.13, 95% CI: 1.03 – 1.24, p = 0.0094) among the patients with IBD were observed when compared with the controls. Other independent factors associated with corneal surface damage were age (aHR: 1.003), sex (male vs. female, aHR: 0.85), and monthly insurance premium (501–800 vs. 0–500 U.S. dollars, aHR: 1.45; ≥ 801 vs. 0–500 U.S. dollars, aHR: 1.32).

Conclusions

Our results suggested that IBD was an independent risk factor for DED and ocular surface damage. Clinical strategies are needed to prevent visual impairment or losses in these susceptible patients.

Peer Review reports

Background

Dry eye disease (DED) is a highly prevalent disorder and characterized by increased osmolarity of the tear film and inflammation of the ocular surface [1]. Epidemiological studies demonstrated that the prevalence of DED ranged from 5 to 50% worldwide [2]. In Taiwan, the crude incidence rate of DED was reported to be 4.26 per 1000 population in 2015 [3]. In the elderly, the prevalence rate was as high as 33.7% in a Taiwan’s community questionnaire survey [4]. DED can be classified into aqueous-deficient and evaporative subtypes by the Dry Eye Workshop of Tear Film and Ocular Surface (TFOS) Society [5]. The aqueous tear-deficient DED can be further divided into Sjögren's syndrome (SS)-related and non-SS-related subtypes [5, 6]. The evaporative DED presents normal lacrimal secretory functions but excessive water loss from the exposed ocular surface [5, 7]. The symptom of DED has a potential adverse impact on patients' physical function and quality of life [8, 9]. The increased treatment utilization and productivity loss exert a heavy economic burden on patients with DED [9].

Inflammatory bowel disease (IBD) is an immune-related chronic gastrointestinal inflammation condition, which includes two major types, Crohn's disease (CD) and ulcerative colitis (UC) [10]. The prevalence rate of IBD ranged from 2.1% to 12.8% globally, and the incidence rate has been rising in recent years [11]. The pathogenesis of IBD remains unclear. Genetic factors, gut microbial, environment and immunological abnormalities are considered as possible causes [12]. IBD primarily involves the gastrointestinal wall, with continuous lesions of the mucosal and submucosal layer in UC and skipped lesions of the whole layer in CD [13]. In addition to intestinal wall damages and manifestations, IBD may also have a deleterious effect on extraintestinal systems, such as the eye [14]. Previous studies have revealed that nearly 2% to 7% patients with IBD had ocular morbidities with episcleritis, scleritis and uveitis in majority [14]. Czompa et al. reported that patients with IBD had higher rates of dry eye and thin cornea with reduced tear quantity compared with non-IBD controls [15]. However, the relationship between IBD and DED remains unclear due to multiple methodological drawbacks of preceding studies, including small patient sample (< 1000 IBD subjects) [15, 16], single-institution settings [15,16,17], and inadequate adjustment for confounding [15,16,17]. Importantly, the long-term risk and epidemiological statistics of ocular surface damage in IBD have not been estimated in previous studies.

In a nationwide matched cohort study, we aimed to examine the association between IBD, DED, and corneal surface damage using the administrative data from Taiwan's National Health Insurance (NHI) research database. Based on existing evidence [14,15,16,17], we hypothesized that IBD was significantly associated with more DED and corneal surface damage compared with non-IBD people.

Methods

Data source

This study was evaluated and approved by Taipei Medical University – Joint Institutional Review Board (TMU-JIRB-N202210011). This study was conducted in accordance with the Helsinki Declaration and the STROBE study guidelines [18]. Written informed consent was waived due to the use of decoded and scrambled beneficiary identifications. The Taiwan government launched a single-payer NHI program in March 1995. Currently, there are more than 23 million Taiwanese residents covered by this program, representing approximately 99.6% of Taiwan’s entire population. A comprehensive description of the NHI research database has been given in previous articles and government’s official websites [19,20,21,22,23].

Subject eligibility criteria

The participants were considered as having developed an IBD only if the diagnosis was established by board-certified physicians, and the condition occurred at ≥ 2 outpatient visits between January 1, 2002 and June 30, 2013. The diagnostic codes used for this study were based on the International Classification of Diseases, 9th Revision, Clinical Modification (ICD-9-CM) (Supplementary Table S1). The index date for the IBD group was the date when IBD was diagnosed for the first time, whereas the index date for the non-IBD group was the IBD-diagnosed date of the matched IBD subject. Participants with a previous diagnosis of dry eye or corneal diseases were excluded from the analysis, including interstitial and deep keratitis, corneal neovascularization, ocular adnexal burns, open wound of eyeball, corneal ulcers, recurrent corneal erosion, and corneal opacity. Subjects who had used eye lubricants before the index date or died during the study period were also excluded.

Ocular outcomes

The primary outcome assessed was DED, which was defined as the diagnosis established at least twice in conjunction with prescriptions of cyclosporine ophthalmic emulsion (Restasis®) treatment by board-certified ophthalmologists (Supplementary Table S1). In the NHI regulations, ophthalmic cyclosporine can be reimbursed when the Schirmer test score was < 5 mm in 5 min [11]. DED was further classified into Sjögren’s syndrome (SS)-related or non-SS-related subtypes. The secondary outcomes assessed were serious types of ocular surface damages (corneal ulcers, recurrent corneal erosion, and corneal opacity), which were defined as the diagnosis made twice in the ophthalmology care service. Survival times were the corresponding censored observations in subjects without the ocular outcomes. Patient’s status was followed up until December 31, 2013.

Patient and clinical characteristics

Insurance premium was categorized into 0 to 500, 501 to 800, and ≥ 801 U.S. dollars per month. The ICD-9-CM codes of medical diagnoses within 2 years before the index date were used to ascertain coexisting diseases potentially related to corneal diseases (Supplementary Table S1) [24]. The Charlson comorbidity index score was assessed for clinical prognosis and comorbidity adjustment [25]. The prescription of systemic steroids within 6 months after the index date was also analyzed. The numbers of hospital admissions and emergency room visits within 2 years before the index date were calculated to evaluate the level of healthcare resource use.

Statistical analysis

Each IBD subject was matched to a non-IBD subject using the greedy matching methodology with a caliper width of 0.2 SDs of the log odds of the calculated propensity score and without replacement to adjust for the distribution of age, sex, and monthly insurance premium between subjects with and without IBD [26]. An absolute standardized mean difference (ASMD) was used to evaluate the baseline patient characteristics between the matched pairs [27]. Imbalance was defined as an ASMD value higher than 0.1. To clarify the independent relationship between IBD, DED, and corneal surface damage, multivariable Cox regression models were utilized to estimate the adjusted hazard ratio (aHR) for the ocular outcomes. The variables controlled in the multivariable model were age, sex, insurance premium, collected coexisting diseases, Charlson comorbidity index score, use of systemic corticosteroids, number of hospitalizations, and number of emergency room visits. In addition, the potential differences in cumulative incidences of DED and corneal surface damage between the two groups were evaluated using the Kaplan–Meier method and log rank tests. A two-sided significance level of 0.05 was used to define a statistically significant difference. All the statistical analyses were implemented using SAS V.9.4 (SAS Institute Inc., Cary, NC, USA).

Results

Altogether, a total of 54,293 matched pairs with 841,752 person-years of follow-up were included in this cohort study (Supplementary Figure S1). The median follow-up time of the entire cohort was 8.3 years (interquartile range: 5.5 – 10.5). The distributions of age, sex, and monthly insurance premium were well matched without between-group differences (Table 1). Compared with non-IBD subjects, IBD patients had more comorbidities, uses of systemic steroids, and greater emergence room visits.

Table 1 Baseline characteristics of subjects with and without inflammatory bowel disease

In the study period, 3421 patients with IBD developed DED, with a period incidence rate of 8.18 cases per 1000 person-years, whereas 2295 non-IBD controls were diagnosed with DED, with an overall incidence rate of 5.42 cases per 1000 person-years. After adjusting for covariates, patients with IBD had an aHR of 1.43 [95% confidence interval (CI): 1.35 – 1.51, p < 0.0001] for DED compared with non-IBD controls (Table 2; Fig. 1A), SS-associated (aHR: 1.67, 95% CI:1.46 – 1.90, p < 0.0001; Fig. 1B) and non-SS-associated subtypes (aHR: 1.38, 95% CI: 1.30 – 1.46, p < 0.0001; Fig. 1C). The median interval between index date and DED diagnosis was median 4.3 (interquartile range: 2.2 – 6.8) years in the IBD subjects and 4.6 (2.2 – 6.9) years in the non-IBD controls (p = 0.0871). Other variables associated with DED were shown in Table 3. Stratified analyses demonstrated that the higher DED risk associated with IBD was significant, independently of different age groups, sex, use of systemic corticosteroids or not, and different comorbidity levels (Table 4).

Table 2 Risk of dry eye and ocular surface damage for subjects with and without inflammatory bowel disease
Fig. 1
figure 1

Cumulative risk of dry eye disease (DED) (A), Sjögren's syndrome (SS)-related DED (B), non-SS-related DED (C), and corneal surface damage (D) between patients with and without inflammatory bowel disease with number of subjects at risk

Table 3 Univariate and multivariable analyses for dry eye disease
Table 4 Stratified analyses of dry eye disease for subjects with and without inflammatory bowel disease

A total of 1003 patients with IBD were diagnosed with corneal surface damage, with an overall incidence rate of 2.34 cases per 1000 person-years, whereas 866 non-IBD controls developed corneal surface damage, with an incidence rate of 2.02 cases per 1000 person-years (Table 2). The multivariable models showed that IBD was significantly associated with increased corneal surface damage (aHR: 1.13, 95% CI: 1.03 – 1.24, p = 0.0094; Table 5 and Fig. 1D), especially for recurrent corneal erosion (aHR: 1.52, 95% CI: 1.22 – 1.88, p = 0.0002). The median time to corneal surface damage was 4.2 years (interquartile range: 2.0 – 7.0) in the IBD patients and 4.3 years (interquartile range: 1.8 – 7.0) in the non-IBD subjects (p = 0.7855). In addition, age (aHR: 1.003), sex (male vs. female, aHR: 0.85), and monthly insurance premium (501–800 vs. 0–500 U.S. dollars, aHR: 1.45; ≥ 801 vs. 0–500 U.S. dollars, aHR: 1.32) were independent factors for corneal surface damage.

Table 5 Univariate and multivariable analyses for corneal surface damage

Discussion

In this cohort study, patients with IBD had a significantly higher risk of DED and secondary SS than non-IBD individuals. Additionally, we also observed a significant association between IBD and ocular surface damage, especially for recurrent corneal erosion. Our analyses demonstrated several clinical factors associated with corneal injury, providing an important implication in early identification and intervention of severe ocular morbidities in IBD patients. To the best of our knowledge, this is the first large population-based study to evaluate the association between IBD and corneal diseases. Our findings highlight an urgent need for regular ophthalmology surveillance and timely referral to prevent potential vision-threatening complications among patients with IBD.

Although ocular involvement is not uncommon among patients with IBD, few studies have evaluated the long-term temporal relationship between IBD, dry eye, and corneal damage. Most previous studies restricted their study population to single-hospital settings with small patient samples [15,16,17]. In a single-center prospective study, Czompa et al. reported that 30 CD patients and 36 UC patients had thinner cornea with reduced tear quantity compared with 80 controls, while the corneal power did not decrease significantly [15]. In addition, the corneal volume and anterior chamber volume were lower in the UC group [15]. Another study reported that the dry eye rate assessed using the Schimer’s test and tear breakup time was three times higher in the IBD group with increased Ocular Surface Disease Index scores compared with the controls [16]. Furthermore, the ocular surface had squamous metaplasia and absence of goblet cells in patients with IBD [16]. In a retrospective study, Cuny et al. reported that the prevalence of ocular morbidity was 2.6% in IBD, and DED affected 1% of IBD patients [17], which was relatively lower than our results (approximately 9% in the 12-year follow-up). The previous studies had small patient samples with cross-sectional design, making it difficult to accurately evaluate the long-term risk estimates and to clearly delineate the temporal relationship of DED and corneal injury related to IBD. In the present study, our sample size was relatively large with a nationwide coverage, and our analyses included a variety of patient and clinical factors, which have not been examined previously.

The pathological pathway of DED in IBD was still unclear. In an animal study, Sekijima et al. showed that reduced tear secretion, inflammatory cell infiltration and destruction of lacrimal gland were discovered in a mice model with UC, indicating that the inflammation and functional decline of lacrimal gland are potentially responsible for dry eye in UC [28]. In humans, Dogan et al. reported that both tear production and tear-film quality were lower among IBD patients; goblet cell losses and squamous metaplasia on the ocular surface were evident [16]. These pathological findings might be the results of ocular inflammation [16]. Furthermore, T cells, interleukins and interferons are involved in the inflammatory and apoptosis reactions of lacrimal gland, cornea and conjunctiva, contributing to the development and progress of DED [7]. Some researchers hypothesized that the mechanism of the ocular manifestations in IBD includes two main pathways: the expansion of immune responses from the intestine or an independent ocular inflammatory event [14]. Inflammatory damage of intestinal mucosa facilitates the passage of microorganisms and proteins and activates lymphoid tissue responses, antibody production, antigen–antibody complex formation, and induces systemic inflammation [29]. Microbial pathogens may also play a pathogenic role via molecular mimicry although few studies have focused on this mechanism [30]. Other studies suggested that genetic factors potentially contribute to the ocular morbidities of IBD. Mallas et al. reported that HLA-B27 type leukocytes were more common among CD patients with extraintestinal manifestations compared with the normal population [31]. However, sparse evidence was available to give a definite explanation for the association between IBD and corneal damage. More studies are warranted to elucidate the biological mechanism and immunological processes of corneal diseases in IBD.

Epidemiological studies estimated that the rate of ocular morbidities in IBD ranged from 2 to 7%, occurring more frequently in CD than UC [14, 32]. Some risk factors were reported, including presence of both colitis and ileocolitis and multi-organ involvement (e.g., arthralgia) [33, 34]. Our results suggested that older age and female sex were also risk factors for corneal surface damage, which have not been reported previously. Our results highlight the importance of regular ophthalmology follow-up for potential corneal surface damage in patients with IBD. Early diagnoses and intervention for ocular complications are pivotal in improving the quality of life and reducing the economic burden in IBD patients.

Our study showed that IBD patients had an increased long-term risk of secondary SS compared with general population. Few studies have investigated the relationship between IBD and SS. There were only case series on SS as an extraintestinal manifestations in IBD [35,36,37]. The etiology of SS in IBD remains unknown nowadays. Mandl et al. has shown that severe dysbiosis was more common in patients with primary SS and was associated with disease activity [30]. In a small cohort, Palm et al. did not find an association between SS and IBD, which might be due to insufficient statistical power [35]. Our results suggested that clinicians should be aware of the risk of secondary SS in IBD patients. Ophthalmology and rheumatology consultations may be helpful in preventing SS-associated complications. Future efforts should be put on elucidating the pathogenesis and prophylactic strategy for IBD-associated SS.

There were some limitations in our study. First, the activity and severity of IBD (e.g., affected bowel segments and extraintestinal organ involvement) were unknow due to data unavailability in the NHI database. Therefore, we could not analyze the relationship between the activity of IBD and risk of corneal damage [38]. Second, we had no information about patients’ daily activities (e.g., physical capacity and sleep duration), physical examination findings, biochemical and laboratory data, pharmacological treatment details (e.g., immunomodulators and biologics), and clinical presentations (e.g., subjective symptoms of DED) of the included patients. Therefore, the interaction between IBD-related medications, immune system functions, and ocular diseases could not be evaluated in this study [38, 39]. Third, we only evaluated three forms of ocular surface damages (corneal ulcers, recurrent corneal erosion, and corneal opacity), which was based on physiology plausibility, data availability, and existing literature [40]. Further studies are needed to clarify whether IBD patients were susceptible to other types of ocular surface injury. Fourth, the propensity-score matching process only included the parameters of age, sex, and monthly insurance premium between the two groups to minimize sample losses and to ensure sufficient statistical power of the matched dataset. A large sample is necessary in evaluating the long-term risk of SS and corneal surface damage associated with IBD because the incidence rates were relatively low. Finally, the subjects were followed up only until the end of 2013 due to the NHI regulations.

Conclusions

This large population-based cohort study found that patients with IBD had a significantly higher risk of DED, secondary SS, and corneal damage. Gastroenterologists treating patients with IBD should be aware of these possible sight-threatening complications and refer patients with corneal manifestations to ophthalmologists for evaluation and management. More attention should be paid to patients at high risk of corneal injury, such as older, female patients with both colitis and ileocolitis and multiple organ involvement. Prophylactic and therapeutic strategies should be further developed to promote vision health in these susceptible patients.

Availability of data and materials

The data generated and analyzed during this study are available from the corresponding author on reasonable request.

Abbreviations

aHR:

Adjusted hazard ratio

ASMD:

Absolute standardized mean difference

CD:

Crohn's disease

CI:

Confidence interval

DED:

Dry eye disease

IBD:

Inflammatory bowel disease

ICD-9-CM:

International Classification of Diseases, 9th Revision, Clinical Modification

NHI:

National Health Insurance

SS:

Sjögren’s syndrome

TFOS:

Tear Film and Ocular Surface

UC:

Ulcerative colitis

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. 2007;5:75–92. https://www.sciencedirect.com/science/article/abs/pii/S1542012412700812?via%3Dihub.

  2. Stapleton F, Alves M, Bunya VY, Jalbert I, Lekhanont K, Malet F, et al. TFOS DEWS II Epidemiology Report. Ocul Surf. 2017;15:334–65.

    Article  PubMed  Google Scholar 

  3. Kuo YK, Lin IC, Chien LN, Lin TY, How YT, Chen KH, et al. Dry eye disease: A review of epidemiology in Taiwan, and its clinical treatment and merits. J Clin Med. 2019;8:1227.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Lin PY, Tsai SY, Cheng CY, Liu JH, Chou P, Hsu WM. Prevalence of dry eye among an elderly Chinese population in Taiwan: the Shihpai Eye Study. Ophthalmology. 2003;110:1096–101.

    Article  PubMed  Google Scholar 

  5. Craig JP, Nichols KK, Akpek EK, Caffery B, Dua HS, Joo CK, et al. TFOS DEWS II Definition and Classification Report. Ocul Surf. 2017;15:276–83.

    Article  PubMed  Google Scholar 

  6. Akpek EK, Bunya VY, Saldanha IJ. Sjögren’s syndrome: More than just dry eye. Cornea. 2019;38:658–61.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Pflugfelder SC, de Paiva CS. The pathophysiology of dry eye disease: What we know and future directions for research. Ophthalmology. 2017;124:S4–13.

    Article  PubMed  Google Scholar 

  8. Uchino M, Schaumberg DA. Dry eye disease: Impact on quality of life and vision. Curr Ophthalmol Rep. 2013;1:51–7.

    Article  PubMed  PubMed Central  Google Scholar 

  9. McDonald M, Patel DA, Keith MS, Snedecor SJ. Economic and humanistic burden of dry eye disease in Europe, North America, and Asia: A systematic literature review. Ocul Surf. 2016;14:144–67.

    Article  PubMed  Google Scholar 

  10. Strober W, Fuss I, Mannon P. The fundamental basis of inflammatory bowel disease. J Clin Invest. 2007;117:514–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Mak WY, Zhao M, Ng SC, Burisch J. The epidemiology of inflammatory bowel disease: East meets west. J Gastroenterol Hepatol. 2020;35:380–9.

    Article  PubMed  Google Scholar 

  12. Guan Q. A comprehensive review and update on the pathogenesis of inflammatory bowel disease. J Immunol Res. 2019;2019:7247238.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Seyedian SS, Nokhostin F, Malamir MD. A review of the diagnosis, prevention, and treatment methods of inflammatory bowel disease. J Med Life. 2019;12:113–22.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Rogler G, Singh A, Kavanaugh A, Rubin DT. Extraintestinal manifestations of inflammatory bowel disease: Current concepts, treatment, and implications for disease management. Gastroenterology. 2021;161:1118–32.

    Article  CAS  PubMed  Google Scholar 

  15. Czompa L, Barta Z, Ziad H, Nemeth G, Rentka A, Aszalos Z, et al. Corneal manifestations of inflammatory bowel disease. Semin Ophthalmol. 2019;34:543–50.

    Article  PubMed  Google Scholar 

  16. Doğan M, Özcan S, Acartürk G, Özdemir Ç. Conjunctival impression cytology and tear-film changes in patients with inflammatory bowel disease. Eye Contact Lens. 2018;44:S420–5.

    Article  PubMed  Google Scholar 

  17. Cuny A, Guillo L, Baumann C, Netter P, Danese S, Caron B, et al. Ocular manifestations in patients with inflammatory bowel disease in the biologics era. J Clin Med. 2022;11:4538.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. von Elm E, Altman DG, Egger M, Pocock SJ, Gøtzsche PC, Vandenbroucke JP, STROBE Initiative. Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) statement: Guidelines for reporting observational studies. BMJ. 2007;335:806–8.

    Article  Google Scholar 

  19. Hsieh CY, Su CC, Shao SC, Sung SF, Lin SJ, Kao Yang YH, et al. Taiwan’s National Health Insurance Research Database: Past and future. Clin Epidemiol. 2019;11:349–58.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Ting HC, Ma SH, Tai YH, Dai YX, Chang YT, Chen TJ, et al. Association between alopecia areata and retinal diseases: A nationwide population-based cohort study. J Am Acad Dermatol. 2022;87:771–8.

    Article  CAS  PubMed  Google Scholar 

  21. Tai CY, Liu HY, Cata JP, Dai YX, Chen MH, Chen JT, et al. The association between general anesthesia and new postoperative uses of sedative-hypnotics: A nationwide matched cohort study. J Clin Med. 2022;11:3360.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Lai EW, Tai YH, Wu HL, Dai YX, Chen TJ, Cherng YG, et al. The association between autoimmune thyroid disease and ocular surface damage: A retrospective population-based cohort study. J Clin Med. 2023;12:3203.

    Article  PubMed  PubMed Central  Google Scholar 

  23. National Health Insurance Research Database. Data subsets. Available from: https://nhird.nhri.org.tw/en/Data_Subsets.html. Accessed 28 Mar 2023.

  24. Shanti Y, Shehada R, Bakkar MM, Qaddumi J. Prevalence and associated risk factors of dry eye disease in 16 northern West bank towns in Palestine: A cross-sectional study. BMC Ophthalmol. 2020;20:26.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Li B, Evans D, Faris P, Dean S, Quan H. Risk adjustment performance of Charlson and Elixhauser comorbidities in ICD-9 and ICD-10 administrative databases. BMC Health Serv Res. 2008;8:12.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Austin PC. A comparison of 12 algorithms for matching on the propensity score. Stat Med. 2014;33:1057–69.

    Article  PubMed  Google Scholar 

  27. Austin PC. Balance diagnostics for comparing the distribution of baseline covariates between treatment groups in propensity-score matched samples. Stat Med. 2009;28:3083–107.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Sekijima H, Hiramoto K, Kozawa S. A decrease in the tear secretion volume in a mouse model with ulcerative colitis. Cutan Ocul Toxicol. 2020;39:363–9.

    Article  CAS  PubMed  Google Scholar 

  29. Das KM. Relationship of extraintestinal involvements in inflammatory bowel disease: new insights into autoimmune pathogenesis. Dig Dis Sci. 1999;44:1–13.

    Article  CAS  PubMed  Google Scholar 

  30. Mandl T, Marsal J, Olsson P, Ohlsson B, Andréasson K. Severe intestinal dysbiosis is prevalent in primary Sjögren’s syndrome and is associated with systemic disease activity. Arthritis Res Ther. 2017;19:237.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Mallas EG, Mackintosh P, Asquith P, Cooke WT. Histocompatibility antigens in inflammatory bowel disease. Their clinical significance and their association with arthropathy with special reference to HLA-B27 (W27). Gut. 1976;17:906–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Mady R, Grover W, Butrus S. Ocular complications of inflammatory bowel disease. ScientificWorldJournal. 2015;2015:438402.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Salmon JF, Wright JP, Murray AD. Ocular inflammation in Crohn’s disease. Ophthalmology. 1991;98:480–4.

    Article  CAS  PubMed  Google Scholar 

  34. Hopkins DJ, Horan E, Burton IL, Clamp SE, de Dombal FT, Goligher JC. Ocular disorders in a series of 332 patients with Crohn’s disease. Br J Ophthalmol. 1974;58:732–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Palm Ø, Moum B, Gran JT. Estimation of Sjögren’s syndrome among IBD patients. A six year post-diagnostic prevalence study. Scand J Rheumatol. 2002;31:140–5.

    Article  PubMed  Google Scholar 

  36. Katsanos KH, Saougos V, Kosmidou M, Doukas M, Kamina S, Asproudis I, et al. Sjogren’s syndrome in a patient with ulcerative colitis and primary sclerosing cholangitis: Case report and review of the literature. J Crohns Colitis. 2009;3:200–3.

    Article  PubMed  Google Scholar 

  37. Rhew EY, Ramsey-Goldman R, Buchman AL. Sjogren’s syndrome in association with Crohn’s disease. J Clin Gastroenterol. 2003;37:312–4.

    Article  PubMed  Google Scholar 

  38. Ahluwalia B, Moraes L, Magnusson MK, Öhman L. Immunopathogenesis of inflammatory bowel disease and mechanisms of biological therapies. Scand J Gastroenterol. 2018;53:379–89.

    Article  PubMed  Google Scholar 

  39. McMonnies CW. Dry eye disease immune responses and topical therapy. Eye Vis. 2019;6:12.

    Article  Google Scholar 

  40. Hung N, Kang EY, Lee TW, Chen TH, Shyu YC, Sun CC. The risks of corneal surface damage in aqueous-deficient dry eye disease: A 17-year population-based study in Taiwan. Am J Ophthalmol. 2021;227:231–9.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This research was funded by Taipei Medical University, Taiwan, grant number TMU110-AE1-B11. The APC was funded by Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, Y.-H.T. and H.-L.W.; methodology, Y.-H.T.; software, Y.-H.T.; validation, Y.-M.W. and C.-Y.K.; formal analysis, Y.-H.T.; investigation, Y.-T.K. and S.-C.L.; resources, S.-C.L. and Y.-G.C.; data curation, Y.-X.D. and T.-J.C.; writing—original draft preparation, Y.-T.K.; writing—review and editing, Y.-M.W., H.-L.W., S.-C.L., Y.-X.D., T.-J.C., Y.-G.C., Y.-H.T., and C.-Y.K.; project administration, Y.-H.T.; funding acquisition, Y.-H.T. All authors have read and agreed to the published version of the manuscript.

Corresponding authors

Correspondence to Ying-Hsuan Tai or Chia-Yu Kao.

Ethics declarations

Ethics approval and consent to participate

The study was conducted according to the guidelines of the Helsinki Declaration, and was evaluated and approved by Taipei Medical University – Joint Institutional Review Board (TMU-JIRB-N202210011). Taipei Medical University – Joint Institutional Review Board waived the need for informed consent due to the use of decoded and scrambled beneficiary identifications.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1:

Supplementary Table S1. ICD-9-CM codes of exposure factor, coexisting diseases, and ocular outcomes.

Additional file 2:

Supplementary Figure S1. Flow diagram for patient selection.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ko, YT., Wu, YM., Wu, HL. et al. Inflammatory bowel disease and the associated risk of dry eye and ocular surface injury: a nationwide matched cohort study. BMC Ophthalmol 23, 415 (2023). https://doi.org/10.1186/s12886-023-03165-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12886-023-03165-z

Keywords