Abstract
Dry eye disease (DED) is a highly prevalent multifactorial disorder of the ocular surface that extends beyond local tear film pathology to involve systemic immune, neuroendocrine, and neurosensory mechanisms. Increasing evidence reveals a strong and bidirectional association between DED and psychiatric disorders, particularly depression, anxiety, post-traumatic stress disorder (PTSD), and sleep disturbances. This review synthesises the current knowledge on shared molecular, neuroimmune, and neuropathic pathways that underlie this comorbidity. Key mechanisms include hypothalamic–pituitary–adrenal (HPA) axis dysregulation, systemic and ocular inflammation, oxidative stress, mitochondrial dysfunction, and impaired neurotrophic signaling, especially reduced brain-derived neurotrophic factor (BDNF). Dysregulation of monoaminergic neurotransmitters such as serotonin and norepinephrine not only contributes to mood disturbances but also alters tear secretion and corneal pain perception. Corneal nerve changes and trigeminal–limbic sensitisation further reinforce the overlap between neuropathic ocular pain and affective dysregulation. Psychotropic medications, while essential for psychiatric care, may exacerbate ocular surface dysfunction through anticholinergic effects, altered neurotransmission, and tear film instability, highlighting the iatrogenic dimension of this interface. Conversely, tear-based biomarkers, including cytokines, serotonin, and BDNF, offer promising translational tools for patient stratification, diagnosis, and treatment monitoring across ocular and psychiatric domains. Recognising DED as part of a systemic, biopsychosocial continuum is critical for effective management. Multidisciplinary strategies that integrate ophthalmologic and psychiatric care, alongside novel therapies targeting shared molecular pathways, provide a framework for improving outcomes. Future research should prioritise longitudinal studies, biomarker validation, and personalised interventions to address this complex comorbidity.
1. Introduction
Dry eye disease (DED) is a chronic, multifactorial disorder of the ocular surface characterised by loss of tear film homeostasis, ocular discomfort, visual disturbance, and inflammation. Affecting up to 50% of individuals in certain populations, especially older adults and postmenopausal women, DED has become a significant global public health concern. While traditionally considered a local ocular surface pathology, a growing body of evidence highlights its systemic dimensions and interrelationships with other chronic conditions, particularly psychiatric disorders [,,].
Over the past decade, studies have shown a strong comorbidity between DED and psychiatric conditions, especially depression, anxiety, post-traumatic stress disorder (PTSD), and sleep disturbances. Epidemiological evidence confirms that psychiatric symptoms are significantly more common in DED patients than in the general population [,,,,,,,,]. These psychiatric comorbidities may exacerbate symptom perception, especially ocular pain and dryness, while chronic ocular symptoms themselves can contribute to psychological distress, creating a bidirectional feedback loop [,,,,,,,,,]. Discrepancies between subjective symptoms and objective signs in DED are common and are frequently attributed to somatisation, alexithymia, and central sensitisation mechanisms [,,,,,].
Epidemiological studies reveal that up to 54% of patients with DED suffer from depression, and 64% report anxiety symptoms [,]. Sleep disturbance is even more prevalent, affecting more than 80% of DED patients [,,]. These psychiatric comorbidities not only exacerbate the subjective experience of ocular symptoms, especially pain and dryness, but also contribute to disease chronification, diminished treatment response, and impaired quality of life. Conversely, chronic ocular discomfort, blurred vision, and visual fatigue serve as persistent stressors that may induce or worsen psychiatric symptoms, creating a vicious cycle of somatic and psychological burden [,,,,,,].
The symptom–sign discordance in DED highlights its bidirectional link with psychiatric disorders [,,,]. Molecular and neurobiological studies suggest common pathways that may explain this overlap [,,,]. Both DED and psychiatric disorders are linked to chronic systemic inflammation, hypothalamic–pituitary–adrenal (HPA) axis dysregulation, oxidative stress, mitochondrial dysfunction, and reduced levels of neurotrophic factors like brain-derived neurotrophic factor (BDNF). Dysregulated neurotransmitter systems, particularly serotonin and norepinephrine, play vital roles in mood regulation, ocular surface stability, tear secretion, and pain perception [,,]. Tear serotonin levels have been correlated with DED severity and ocular pain, suggesting potential as a biomarker [].
From a neuropathic perspective, in vivo confocal microscopy studies reveal structural changes in the corneal nerve plexus of DED patients, including decreased nerve density, increased tortuosity, and formation of microneuroma. These alterations are associated with symptoms of hyperalgesia and allodynia, and mirror the central sensitisation phenomena seen in chronic pain syndromes and mood disorders [,,,]. Additionally, several neuropeptides involved in pain signalling and emotional regulation, such as substance P, Calcitonin Gene-Related Peptide (CGRP), and Transient Receptor Vanilloid 1 (TRPV1), are dysregulated in DED [,,].
Adding further complexity, psychotropic medications commonly prescribed for depression and anxiety, including selective serotonin reuptake inhibitors (SSRIs) and selective serotonin and norepinephrine reuptake inhibitors (SNRIs), tricyclic antidepressants (TCAs), and antipsychotics, may impair ocular surface function through anticholinergic effects, tear reflex suppression, or immune response modulation. This creates an important iatrogenic aspect to the DED-psychiatry interface, especially among elderly patients and those on polypharmacy [,,,,,,].
Despite the accumulating evidence, the connection between DED and psychiatric disorders remains under-recognised in clinical practice [,]. Ophthalmologists often overlook psychiatric contributors to symptom severity, while mental health professionals may misattribute ocular symptoms to medication side effects. A biopsychosocial approach and interdisciplinary collaboration are essential for optimising diagnosis and treatment in this patient population [,,,,].
This review synthesises the current knowledge on the shared molecular, neuroimmune, and neuropathic mechanisms underlying the connection between DED and psychiatric disorders. It presents emerging insights into the involvement of BDNF, oxidative stress, HPA axis dysregulation, and serotonin signalling as potential links between ocular and mental health. By integrating molecular findings with clinical observations, this review proposes a biopsychosocial framework to support more effective diagnosis, biomarker identification, and multidisciplinary treatment strategies. Emphasising a translational perspective, this review aims to facilitate the integration of ocular and psychiatric care, ultimately improving patient outcomes.
Data Collection
A comprehensive narrative literature review was conducted to provide an integrative overview of the bidirectional relationship between DED and psychiatric disorders. The specific objectives were to summarise the epidemiological associations, shared aetiological and pathophysiological mechanisms, overlapping symptomatology, clinical implications, emerging therapeutic strategies, and future research perspectives. A systematic literature search was conducted using the PubMed, MEDLINE, and Google Scholar databases, encompassing all publications available up to June 2025. The search strategy incorporated a combination of the following keywords: “dry eye disease”, “dry eye”, “ocular surface”, “tear”, “psych*”, “psychiatry*”, “psychiatric disorders”, “psychiatric symptoms”, “psychological”, “depress*”, “depression”, “anxiety”, “sleep”, “stress”, “mechanism”, “neuroimmune”, “neuroimmune mechanisms”, “molecular”, “pain”, “medications”, “psychotropic”, “drugs”, “biomarkers”, “therapy”, “treatment”, and “interventions”. The search was limited to studies published in English. After the initial identification of relevant publications, duplicates were removed, and the titles and abstracts were screened to assess eligibility. Full-text articles that met the inclusion criteria were subsequently reviewed in detail. The reference lists of selected studies were manually examined (snowballing method) to identify additional relevant articles not captured in the initial database search. All types of peer-reviewed scientific publications written in English were considered, including original preclinical and clinical research, systematic reviews, and meta-analyses. To ensure inclusion of the most up-to-date evidence, particular emphasis was placed on studies published within the past five years. Articles without English translations, those not aligned with this review’s objectives, and those failing to meet higher methodological or publication standards were excluded. No quantitative data synthesis was performed, as the aim was to provide a comprehensive narrative discussion on the topic. This review is therefore designed as a narrative synthesis aimed at integrating mechanistic and clinical evidence rather than performing quantitative meta-analysis.
2. Epidemiological and Psychosomatic Links Between Dry Eye Disease and Psychiatric Disorders
DED is closely linked with psychiatric disorders, influencing symptom severity, quality of life, and treatment outcomes [,,,,,,,,,,,,,]. Understanding these interactions is essential for comprehensive patient care and the development of personalised therapeutic strategies, which is key to holistic patient management.
2.1. Psychiatric Comorbidities and Symptom–Sign Discordance
Patients with DED commonly exhibit psychiatric symptoms, with significantly higher rates of depression, anxiety, and sleep disturbances compared to the general population. Meta-analyses estimate that 40–54% of DED patients experience depression, while 39–64% report anxiety symptoms [,]. Conversely, nearly one-third of individuals with depression and anxiety are diagnosed with DED, and 22% report DED-like symptoms without a formal diagnosis []. A large meta-analysis of approximately three million participants showed that DED patients have a threefold higher risk of both depression and anxiety than controls [].
Sleep disturbances are prevalent in DED, affecting up to 84% of patients []. These include insomnia, sleep fragmentation, and obstructive sleep apnea [], with women particularly affected [,]. Among ocular conditions, DED is linked to the worst sleep outcomes, with over 42% of patients reporting disturbed sleep []. Depression is especially pronounced in subgroups such as patients with Sjögren’s syndrome [].
Emerging data also point to a potential association between PTSD and DED. In a U.S. Veterans Affairs cohort, women with PTSD had a 2.4-fold increased risk of DED compared to men []. Additionally, obsessive compulsive disorder (OCD) has been associated with heightened DED symptomatology []. These findings underscore the multidimensional burden of DED and the need for integrated psychiatric screening.
A hallmark feature of DED is the poor and inconsistent correlation between subjective symptom severity and objective clinical signs [,]. This disconnection resembles patterns seen in other chronic diseases like rheumatoid arthritis and glaucoma, where patients may exhibit marked signs without symptoms, delaying diagnosis and treatment [,]. Approximately 42% of patients show this discrepancy, either reporting significant symptoms without objective evidence or presenting with clinical signs in the absence of symptoms [].
This disconnection is particularly prominent in patients with anxiety or depression. Psychiatric symptoms correlate strongly with subjective complaints but not with tear break-up time (TBUT), Schirmer test results, or fluorescein staining. This may reflect altered perception due to disrupted neurochemical signalling in mood disorders, which amplifies perception of somatic symptoms []. Patients often report higher depressive symptoms on self-rating scales, yet structured psychiatric interviews reveal fewer actual diagnoses, suggesting emotional hypersensitivity or somatisation [,].
A similar dissociation is seen with sleep disturbances. Tools like the Insomnia Severity Index (ISI) and the Pittsburgh Sleep Quality Index (PSQI) correlate with subjective symptoms but not with objective ocular surface metrics []. Pain perception appears central to this disconnection. Ong et al. identified pain as a key factor underlying the mismatch between subjective symptoms and objective signs in DED, particularly in patients with arthritis, extraocular pain, or central sensitisation syndromes []. Other contributing factors include female sex, likely due to hormonal influences, and sensitivity to low humidity []. This discrepancy may, at least in part, be attributed to the inherent limitations in sensitivity, specificity, and reproducibility of current diagnostic tests. Many objective tests show poor reproducibility and low intercorrelation, and are most effective only in advanced DED. Tear osmolarity remains the most consistent measure across all disease severities []. Symptom variability during the day, often worse in the evening, further complicates assessment. Patients with primary Sjögren’s syndrome may report fewer symptoms despite severe objective findings, possibly due to altered pain signalling involving TRPV1 and substance P [].
Older patients may underreport symptoms due to age-related reductions in corneal sensitivity and a stoic personality profile [,]. Notably, individuals with the greatest symptom–sign discrepancies tend to have lower corneal nerve density and worse ocular-related quality of life [,,]. As in other chronic illnesses, this discrepancy may impair treatment response []. Notably, up to 18% of pilots demonstrate objective signs of DED without symptoms, raising safety concerns and highlighting the need for improved diagnostic protocols [].
2.2. Psychological Factors in Symptom Amplification
In addition to biological mechanisms, psychological factors such as somatisation significantly influence symptom perception in DED [,]. Somatisation, defined as the experience of physical symptoms without objective findings, affects up to 80% of individuals with depression and frequently co-occurs with anxiety and depressive symptoms [,,]. DED patients with pronounced symptoms but minimal signs often exhibit high levels of somatisation and alexithymia, suggesting that emotional dysregulation may manifest as ocular discomfort [,].
2.2.1. Somatisation and Central Pain Sensitisation
Somatisation plays a key role in pain amplification and treatment resistance, contributing to the subjective burden of DED, particularly ocular pain, which strongly correlates with anxiety, depression, and sleep disturbances [,]. Health anxiety has also been identified as a mediator linking psychiatric symptoms with dry eye complaints []. These findings underscore the importance of assessing psychological dimensions during DED evaluations.
While somatic symptoms are part of depression’s diagnostic criteria, they should not be equated with somatisation []. In some cases, DED may be a somatic expression of depression, given overlapping mechanisms such as pain sensitisation [,]. However, although pain sensitisation and somatisation are related, they remain distinct entities []. It is therefore plausible that somatisation amplifies rather than causes DED symptoms [].
2.2.2. Personality Traits and Emotional Vulnerability
Several personality traits and psychological factors, such as neuroticism, low conscientiousness, emotional dysregulation, and reduced subjective well-being, have been associated with increased DED symptom severity [,,,]. Anxiety sensitivity is another contributor, particularly under emotional stress [].
Lifestyle factors such as excessive screen time contribute to both DED and poor sleep quality [,,], while poor sleep exacerbates stress, anxiety, and DED symptoms [,]. Typical DED symptoms, such as ocular pain, foreign body sensation, and blurred vision, negatively affect quality of life and may precipitate depressive symptoms []. Frequent medical visits, low treatment satisfaction, and the financial burden of care further reinforce emotional distress [,].
Evidence supports a strong, bidirectional association between DED and psychiatric symptoms [,,,]. Shared risk factors include female sex, menopause, ageing, and an elevated omega-6:omega-3 ratio [,,,]. Patients with moderate-to-severe DED often report a higher psychiatric burden, particularly in postmenopausal women with Sjögren’s syndrome [,,,].
2.2.3. Sleep Disturbances and Lifestyle Risk Factors
Sleep disturbances are both a consequence and a driver of psychiatric and ocular symptoms. Individuals sleeping five or fewer hours per night report significantly worse DED symptoms []. Among DED symptoms, ocular pain shows the strongest association with depression, anxiety, and sleep disorders [], while blurred vision is more closely related to depression than anxiety [,]. In severe cases, sleep quality appears to be the strongest predictor of symptom burden. Meibomian gland dysfunction and ocular surface inflammation are both associated with poor sleep [].
2.2.4. Implications for Clinical Management and Research
An observational study on DED treatment over 3–6 months showed reduced therapeutic efficacy in patients with comorbid anxiety or depression. Patients without psychiatric comorbidity experienced greater symptom relief, and while DED therapy somewhat improved depressive symptoms, it had no significant effect on anxiety []. These findings support routine screening for dry eye symptoms in individuals with psychiatric conditions, with early referral to ophthalmology when needed [].
Despite growing interest, longitudinal studies examining causal links between psychiatric disorders and DED remain limited [,]. Many reports are based on small, cross-sectional samples. They should therefore be interpreted cautiously, as high prevalence estimates may reflect selection bias or subjective symptom reporting rather than true population effects [,]. Psychiatric–ocular research is particularly prone to methodological variability, including inconsistent diagnostic criteria, heterogeneous assessment tools, small sample size and the lack of a control group in many studies [,,,,,]. Selection bias and reliance on self-reported symptoms further challenge the validity of observed associations [,]. Moreover, most studies do not clearly distinguish between primary DED and secondary forms related to psychopharmacologic treatment, which may confound interpretation of pathophysiological mechanisms and therapeutic outcomes. Geographic, sociocultural, and epigenetic factors also influence disease expression and likely contribute to the heterogeneity reported in meta-analyses []. Sociocultural and epigenetic influences may further shape the clinical expression and perception of DED–psychiatric comorbidity [,,,,,]. Studies from Asian cohorts often show higher DED prevalence, especially among women, reflecting differences in lifestyle, diet, and cultural norms affecting symptom reporting and healthcare-seeking behaviour. Further, data from U.S. Veterans highlight occupational stress, trauma exposure, and psychotropic medication use as major contributing factors [,,,,,,,,,,]. At the molecular level, stress, sleep deprivation, and inflammation can trigger epigenetic modifications in genes via regulation of the HPA axis and neurotrophic signaling such as BDNF and NR3C1, leading to population-specific vulnerability patterns [,,,,]. Integrating these sociocultural and biological determinants into future research frameworks will improve comparability between studies and strengthen causal inference.
Future research should therefore employ standardised diagnostic protocols for both ocular and psychiatric evaluation, include well-defined control groups, and use validated, clinician-administered instruments []. Large, multicentre, longitudinal studies are needed to minimise cultural and regional variability and strengthen causal inference. Given the complex interaction of ocular, psychiatric, and somatic factors, future work should also utilise advanced statistical approaches, such as cluster or latent class analyses, to identify high-risk patient phenotypes and guide the development of personalised and integrated therapeutic strategies [].
4. Neurosensory Dysfunction and Central Pain Mechanisms
Emerging evidence suggests that the link between DED and psychiatric disorders extends beyond epidemiological associations and shared symptomatology, encompassing deeply interconnected neuroimmune and neuropathic mechanisms [,,,,,,,,]. Figure 1 illustrates the conceptual framework of these shared pathways, highlighting how peripheral ocular surface inflammation and neurosensory alterations converge with central mechanisms of stress regulation, emotional processing, and pain modulation. While systemic inflammation, neurotransmitter dysregulation, and hormonal imbalances significantly contribute to both conditions, recent studies highlight the role of structural and functional changes in the peripheral and central nervous systems that further reinforce this bidirectional relationship [,,,,,,]. Specifically, alterations in corneal innervation, dysregulated neuropeptide signalling, and sensitisation of the trigeminal–limbic pain axis create a complex neurobiological interface through which ocular and emotional symptoms interact [,,]. These changes are paralleled by central mechanisms including microglial activation, HPA axis dysregulation, and impaired neurotrophic signalling, all of which are implicated in the pathophysiology of depression, anxiety, and chronic pain [,,,,].
Figure 1.
Shared neuroimmune and neuropathic pathways linking dry eye disease and psychiatric disorders. DED: Dry Eye Disease; PTSD: Posttraumatic Stress Disorder; HPA: Hypothalamic–pituitary–adrenal axis; BDNF: Brain-derived Neurotrophic Factor; Trk: tyrosine kinase; IL: interleukin; QoL: Quality of Life.
4.1. Corneal Nerve Alterations and Neuropathic Ocular Pain
Neurosensory dysfunction is a key contributor to the pathogenesis of DED. Reduced tear secretion and tear film instability lead to inflammation and damage of peripheral corneal nerves, which may result in neuropathic pain []. The cornea, particularly its subbasal nerve plexus, is densely innervated, and in vivo confocal microscopy enables visualisation of nerve alterations [,].
DED patients frequently exhibit reduced corneal nerve fibre density, increased tortuosity, and morphological changes consistent with nerve damage and regeneration [,,]. These alterations correlate with symptoms such as hypoesthesia, dysesthesia, and heightened sensitivity, particularly in Sjögren’s syndrome []. Both main and branching nerve fibres are decreased in DED, with more pronounced reductions observed in aqueous-deficient subtypes compared to evaporative forms []. Aqueous-deficient DED is also associated with reduced epithelial cell density, corneal thinning, and marked stromal inflammation [].
Increased nerve tortuosity, an indicator of aberrant nerve regeneration, is positively correlated with DED severity and negatively with tear film stability [,]. A meta-analysis confirmed reductions in corneal nerve length and density in DED patients but highlighted the need for further validation of IVCM as a diagnostic tool [,]. Microneuromas, localised swellings of subbasal nerve fibres, are frequently observed in patients with neuropathic corneal pain (NCP), especially in cases of DED linked to Meibomian gland dysfunction [].
Depression and neuropathy are bidirectionally linked, with anxiety and depressive symptoms more prevalent in individuals with neuropathic pain, particularly those exhibiting severe symptoms and high levels of pain sensation []. Depressive symptoms are significantly more common in patients with diabetic neuropathy []. Shared pathophysiological mechanisms between depression and neuropathic pain include chronic psychosocial stress, systemic and neuroinflammation, autoimmune responses, and maladaptive neuroplasticity [].
Neurotransmitter systems implicated in both conditions include monoamines and BDNF, with reduced BDNF levels and GABAergic dysfunction contributing to heightened pain sensitivity and depressive symptoms []. In the murine model, sciatic nerve injury induced depressive-like behaviours within eight weeks, accompanied by microglial activation and inflammation in the prefrontal cortex, hippocampus, and amygdala, regions central to mood regulation []. Accordingly, patients with DED and comorbid NCP are likely to exhibit more severe depressive symptoms [].
4.2. Neuropeptides in Pain and Emotion
CGRP is a neuropeptide abundantly expressed in the peripheral and central nervous systems, particularly in the dorsal horn and trigeminal ganglion [,,]. It plays a key role in pain transmission and central sensitisation, with elevated levels observed in various pain conditions []. CGRP is also found in brain regions involved in emotional processing, including the hippocampus, amygdala, hypothalamus, and thalamus []. Following nerve injury or inflammation, CGRP synthesis increases, partly via nerve growth factor (NGF) released by activated macrophages. Upon peripheral nerve stimulation, CGRP induces neurogenic inflammation through vasodilation [].
Furthermore, a preclinical study shows that CGRP is also involved in mood regulation. In a murine stress model, reduced hippocampal CGRP mRNA levels were associated with depressive-like behaviour, whereas intracerebral CGRP administration before stress prevented these symptoms by enhancing NGF signalling []. Clinical evidence further indicates that anti-CGRP monoclonal antibodies may alleviate depressive symptoms in migraine patients, independent of headache relief; however, additional studies are required to confirm these findings [].
In the ocular system, CGRP is released from corneal sensory nerves and stimulates lacrimal gland secretion, suggesting a protective role in tear production []. Dysregulated CGRP expression is noted in autoimmune DED, such as Sjögren’s syndrome and ocular cicatricial pemphigoid []. A recent study showed reduced tear levels of CGRP and substance P in DED patients, especially in severe cases, reinforcing the role of neuropeptides in DED pathophysiology. These neuropeptides, secreted from nerve endings and ocular surface tissues, contribute to inflammation, reflex tearing, and ocular discomfort, emphasising the importance of the neuroimmune interface in DED [].
Transient Receptor Potential Vanilloid 1 (TRPV1) is a polymodal receptor expressed in small and medium-sized nociceptive neurons, including C- and Aδ-fibers of the dorsal root and trigeminal ganglia. In the cornea, TRPV1 is present on cold-sensitive nerve fibres, contributing to cold-induced pain and regulation of the tear reflex []. In DED, TRPV1 expression is upregulated in the trigeminal ganglion, enhancing cold allodynia and pain through the release of substance P []. Preclinical studies in adult male mice demonstrate that the TRPV1 antagonist capsazepine reduces corneal sensitivity to noxious stimuli, alleviates pain, and diminishes DED-related anxiety, highlighting its therapeutic potential []. While substance P promotes corneal epithelial healing, its overexpression induces hyperalgesia, inflammation, and neovascularisation. Altered substance P levels in DED and its interaction with the neurokinin-1 receptor (NK1R) underscore NK1R antagonists as a promising target for future therapeutic strategies [].
TRPVI is increasingly recognised for its role in higher brain functions, including learning, memory, and emotional regulation []. TRPV1-deficient mice exhibit reduced fear and anxiety; however, the effects of TRPV1 modulation in humans remain poorly understood []. In mice fed a high-fat diet, depressive-like behaviour has been linked to TRPV1 dysfunction in brain regions such as the prefrontal cortex, hippocampus, hypothalamus, and amygdala []. Preclinical studies in mice and rats further demonstrate that TRPV1 antagonism in the hippocampus and amygdala produces anxiolytic and antidepressant effects, likely through enhanced serotonergic activity [].
TRPV1 also contributes to neuropathic pain through microglial activation and glutamatergic signalling at the synaptic level. However, conflicting evidence suggests that TRPV1 may not modulate synaptic plasticity in the anterior cingulate cortex, as demonstrated in adult male mice []. Substance P, a key mediator of pain and emotion, is released during nociceptive signalling and amplifies pain via NMDA receptor sensitisation. Elevated substance P levels have been observed in individuals with depression and anxiety, and are associated with structural brain changes and hyperalgesia []. Acting through NK1 receptors, substance P contributes to mood and anxiety disorders, making NK1 receptor antagonists a promising therapeutic strategy for depression, anxiety, and PTSD, as suggested by findings from a limited number of clinical trials [].
4.3. Trigeminal–Limbic Pathways and Central Sensitisation
Central sensitisation refers to heightened pain perception caused by repeated activation of peripheral nociceptors, particularly C-fibres, leading to excessive NMDA receptor activation in the dorsal horn of the spinal cord and reduced pain thresholds [,,,]. This mechanism is closely linked to depressive disorders through limbic system involvement. Pain activates the kappa opioid system, which reduces dopamine release in the nucleus accumbens, a pathway implicated in the emotional burden of chronic pain and the development of depressive symptoms [,].
In DED, nociceptive pain arises from ocular surface stimuli such as tear film instability and inflammation, while neuropathic pain results from dysfunction in the sensory nervous system. Corneal pain is mediated by Aδ and C fibres of the ophthalmic branch of the trigeminal nerve, with cell bodies in the trigeminal ganglion projecting to the spinal trigeminal nucleus [,]. From there, pain signals ascend to the thalamus and somatosensory cortex, where they are integrated within the limbic system, including the amygdala, insula, anterior cingulate cortex, and prefrontal cortex, regions responsible for the emotional aspect of pain [,].
Central sensitisation in DED often involves hyperexcitability of neurons in the subnucleus caudalis of the spinal trigeminal complex [,]. Glutamatergic transmission via AMPA, NMDA, and mGluR5 receptors enhances pain signalling, while reduced GABAergic inhibition contributes to persistent hypersensitivity. Supporting this mechanism, a preclinical study in rats demonstrated that activation of GABA A receptor agonists may reduce pain in this region, highlighting the role of excitation/inhibition imbalance in chronic ocular pain [].
Resting-state fMRI studies in patients with DED reveal abnormal regional neural activity (ReHo) within limbic–cortical circuits, supporting a link between central neural alterations, chronic pain, and mood disorders []. Central sensitisation is further exacerbated by microglial activation and neuroinflammation []. Persistent ocular nerve injury induces changes in the expression of ion channels and receptors, including NMDA receptors, within the trigeminal system, promoting the transition to neuropathic pain. Consistently, a preclinical study on rats indicates that NMDA receptor antagonists may show therapeutic potential in alleviating centrally mediated ocular pain [].
These mechanisms explain why DED patients may report severe discomfort despite normal clinical findings and why subjective symptoms often correlate more strongly with psychiatric comorbidities than with objective ocular signs []. Elevated serotonin levels in tears may serve as a biomarker of peripheral pain sensitivity in DED, but not in cases dominated by central sensitisation [].
4.4. Microglial Activation and Neuroinflammation
Psychosocial stress in depression activates the NLRP3 inflammasome via endogenous damage-associated molecular patterns (DAMPs), triggering the release of pro-inflammatory mediators that stimulate microglial activation [,]. Activated microglia secrete prostaglandins, leukotrienes, and chemokines, such as CXCL4, CXCL7, and CXCL8, that promote monocyte recruitment and sustain a neuroinflammatory feedback loop contributing to neuronal damage, particularly in the limbic system []. Elevated levels of Iba1, a marker of microglial activation, are commonly observed in depression. Chronic glucocorticoid exposure further amplifies NLRP3 inflammasome activity and microglial proliferation, linking prolonged stress to immune and affective dysregulation [].
Monoaminergic dysfunction in depression also contributes to excessive glial activation [,]. Preclinical rat studies have demonstrated that norepinephrine exerts anti-inflammatory effects via β-adrenergic receptors on glial cells. Serotonin effects are concentration- and receptor-dependent: 5-HT1A and 5-HT7 receptor activation is anti-inflammatory, whereas 5-HT2A/2B/2C receptors promote microglial activation and NF-κB signalling []. Additionally, mouse models indicate that activation of astrocytic dopamine D2 receptors suppresses inflammation through alpha B-crystallin (CRYAB)-mediated pathways, whereas receptor antagonism enhances glial activation and inflammation []. These findings underscore the critical role of glial–immune interactions in the pathophysiology of mood disorders [,,,].
Functional parainflammation is a protective, regulated immune response mediated by factors such as Nrf2 and NF-κB, which promote the production of antioxidants and growth factors, thereby maintaining homeostasis []. However, when such inflammation becomes chronic, it contributes to diseases such as glaucoma and age-related macular degeneration (AMD) []. In DED, persistent inflammation reflects dysregulated immune mechanisms that involve both innate and adaptive responses []. Neuronal release of proinflammatory mediators activates microglia and astrocytes, which then release cytokines, including IL-1β, IL-6, TNF-α, chemokines, BDNF, and excitatory molecules such as glutamate and ATP, leading to increased pain sensitisation []. Microglia are activated first, followed by prolonged astrocyte activation, which sustains neuropathic pain in DED []. In a mouse model with lacrimal gland excision, enhanced corneal sensitivity, trigeminal synaptic activity, elevated inflammatory markers, and increased expression of glial markers (Iba1, GFAP, CD68, Itgam) further confirmed the role of neuroinflammation in pain generation []. Other mouse models similarly demonstrate heightened microglial activity in the trigeminal nucleus, which subsides once corneal inflammation resolves, indicating reversibility of both peripheral and central sensitisation []. Glial activation also contributes to retinal diseases such as AMD through the activation of microglia and the complement system []. Collectively, these findings indicate that neurogenic inflammation, mediated by both peripheral and central sensitisation, plays a key role in the pathophysiology of DED [,,,].
5. Psychotropic Medications and Ocular Surface Health
Psychotropic medications, including TCAs, SSRIs and SNRIs, benzodiazepines, pregabalin, gabapentin, antipsychotics, and mood stabilisers, can compromise ocular surface health through anticholinergic activity, tear film destabilisation, and altered corneal innervation [,,,]. The eye’s rich vascular supply facilitates systemic drug penetration, making ocular side effects relatively common []. Many of these agents are associated with measurable declines in tear production or stability, pro-inflammatory changes, and structural alterations in the cornea, underscoring the need for vigilance in patients with pre-existing DED [,,,].
Table 2 summarises the ocular surface impact of frequently prescribed psychotropic medications, including mechanisms, clinical symptoms, and evidence-based management strategies [,,,,,,,,,,,,,,,,,,,,,,,,,].
Table 2.
Ocular Surface Effects and Management of Common Psychotropic Medications.
5.1. Antidepressants and Anxiolytics
TCAs exert potent anticholinergic effects, leading to reduced lacrimal and goblet cell secretion, and are frequently associated with mydriasis, blurred vision, and an increased risk of acute angle-closure [,]. Although SSRIs and SNRIs possess weaker anticholinergic properties, they have been associated with elevated intraocular pressure and, in rare cases, acute glaucoma [,]. Benzodiazepines, pregabalin, and gabapentin may cause blurred vision, diplopia, or acute glaucoma episodes [].
Multiple studies have demonstrated that antidepressants and anxiolytics significantly increase the risk of developing DED, independent of the presence of depression [,,]. Large population-based analyses rank the use of these agents as a DED risk factor on par with glaucoma, blepharitis, and the use of antihypertensive medications []. TCAs cause the greatest reduction in Schirmer I test scores, whereas SSRIs reduce both Schirmer and Tear Break-up time Test (TBUT) values. In contrast, SNRIs primarily decrease TBUT, indicating impaired tear film quality without significant loss of aqueous volume [,]. Although anticholinergic mechanisms are predominant, a rat study by Dankis et al. demonstrated that even agents with minimal anticholinergic activity, such as escitalopram, can suppress reflex tear secretion, suggesting that disrupted neural regulation also contributes to the pathophysiology of DED [].
SSRIs may further impair ocular surface integrity by modulating tear serotonin concentrations and activating NF-κB–mediated inflammation [,]. In one study, 75% of individuals receiving SSRIs exhibited a TBUT of less than 10 s accompanied by corneal epithelial damage, despite normal Schirmer test values, indicating a selective impairment of tear film stability rather than aqueous tear production []. SNRIs such as duloxetine and venlafaxine may reduce ocular pain perception through central pain-modulation pathways [,]; however, elevated IL-17 and TNF-α in tears of depressed DED patients suggest a pro-inflammatory milieu influenced by drug selection []. While dry eye is typically labelled a rare adverse effect, clinicians should consider drug class, anti-inflammatory properties, and neural impact when managing patients with coexisting psychiatric disease and DED [].
5.2. Antipsychotics
Antipsychotic medications, particularly atypical agents, are increasingly recognised as contributors to ocular surface alterations, including reduced tear secretion, tear film instability, epithelial damage, corneal thinning, and pigment deposition []. Severe DED can develop within the first 1–5 years of treatment, with prevalence up to 20% after one year []. Antipsychotic-induced neuromuscular effects, such as blepharospasm and tics, reported with second-generation agents, can exacerbate evaporative DED [].
Many antipsychotics’ anticholinergic effects contribute to tear film dysfunction [,]. Clozapine, a strong muscarinic M3 receptor antagonist critical for tear secretion [,], is especially implicated, alone or with quetiapine [,]. It is associated with decreased corneal thickness and markedly reduced Schirmer and TBUT scores compared to controls [,]. Clozapine’s antagonism of 5-HT2C receptors also lowers blink rate by suppressing nigrostriatal dopamine, further impairing tear film renewal and worsening dry eye [].
Importantly, DED often goes undiagnosed in patients with schizophrenia due to their typically minimal subjective symptoms []. Consequently, clinicians should maintain a high level of vigilance, particularly in cases of polypharmacy and long-term antipsychotic use, and consider routine ophthalmologic screening for this vulnerable group.
5.3. Mood Stabilisers
Mood stabilisers such as lithium and valproate have been linked to tear film instability and the development of evaporative DED [,]. Lithium, which is excreted in tears, increases tear osmolarity and reduces tear production, causing ocular irritation [,]. Rarely, lamotrigine may trigger Sjögren’s-like symptoms through autoimmune pathways involving IL-4 and IL-5 []. Similarly, lithium may promote autoimmunity in salivary glands, while valproate induces oxidative stress, damaging lacrimal glands [,]. Although lithium does not affect the blink reflex, its ocular side effects are often underrecognized, potentially leading to treatment nonadherence [,]. Current evidence on antiepileptic and mood stabilisers in DED is limited to small studies and case reports, highlighting the need for further research []. Notably, longer duration of psychotropic use is consistently associated with higher DED prevalence across antidepressants, anxiolytics, and antipsychotics [,].
5.4. Polypharmacy and Cumulative Risk
Polypharmacy, typically defined as the use of five or more medications, is a significant iatrogenic risk factor for DED, especially in older adults, increasing the likelihood of ocular or oral dryness by 88% []. The additive anticholinergic burden and neurotransmitter interactions can impair parasympathetic regulation of lacrimal and salivary secretion, a critical consideration in suspected Sjögren’s syndrome, where medications may mimic or exacerbate glandular dysfunction []. Along with systemic diseases and age-related structural changes, polypharmacy is a leading cause of DED in the elderly []. Moreover, antipsychotic polytherapy confers a higher risk of DED compared to monotherapy [], underscoring the importance of thorough medication review to optimise ocular surface health.
Although it remains challenging to clearly differentiate primary from secondary, psychopharmacologically induced DED, the existing literature allows for a pragmatic clinical distinction [,,,,,,,,,,,,,,,,,,,,,,,,]. Secondary DED is typically associated with the use of medications that impair the blink reflex, reduce lacrimal secretion, or disrupt ocular surface homeostasis. This is particularly evident in patients receiving polypharmacy, especially drugs with anticholinergic properties or long-term psychotropic therapy. In such cases, additional ocular side effects, including blurred vision, pupillary changes, or ocular surface toxicity, are often present alongside DED manifestations [,,,,,,,,,,,,,,,,,,,,,,,].
6. Clinical and Translational Perspectives
In some patients, severe dry eye symptoms may primarily reflect underlying psychological distress rather than ocular surface damage. If overlooked, this can result in ineffective, surface-focused treatment. Routine screening for depression and anxiety in DED is essential, as comorbid psychological disorders can impair treatment response and prolong symptoms []. Integrated care between ophthalmologists and mental health professionals supports personalised management and improves outcomes [].
Severe symptoms may also indicate neuropathic pain, often associated with chronic illness, central sensitisation, and overlapping pain conditions [,]. These patients may not benefit from standard therapies and may require treatments targeting neural dysfunction. Corneal neuropathic pain often mimics severe DED, leading to potential misdiagnosis.
Emerging therapies targeting corneal nerve dysfunction include topical nerve growth factor (NGF), already approved for neurotrophic keratitis, and may aid in regenerating damaged corneal nerves in CNP. Other promising drugs include lacosamide, low-dose naltrexone, enkephalin modulators, and libvatrep [,]. Accurate diagnosis of psychogenic or neuropathic DED requires careful history-taking, awareness of neuropathic symptoms such as burning, allodynia, refractory pain, and advanced imaging, such as in vivo confocal microscopy [,].
6.1. Tear-Based Biomarkers for Diagnosis and Monitoring
Tear fluid contains cytokines, chemokines, and growth factors that indicate ocular surface inflammation and disease severity, providing key insights for diagnosing and monitoring DED []. Tears are readily accessible and can be collected noninvasively, making them ideal for biomarker analysis. Inflammatory markers such as IL-1β and IL-6 indicate epithelial damage, while EGF reflects the function of the lacrimal gland [,,,]. Tear biomarkers are more sensitive and correlate better with disease severity than traditional tests like Schirmer’s and corneal staining. Their noninvasive nature and easy accessibility make tears an attractive medium for clinical assessment. Despite this, their routine use is limited by high costs, sampling variability, diurnal fluctuations, low specificity, and inherent biological heterogeneity [,,]. Notably, tear composition is influenced by sex, as sex steroids differentially modulate tear secretion []. In addition, standardised reference ranges for key molecules, including serotonin, remain unavailable across different body fluids []. To overcome these limitations, the establishment of standardised sampling protocols and the development of composite biomarker indices are critical, promising to enhance both the accuracy and translational potential of tear-based diagnostics [].
6.2. Emerging Molecular and Neuromodulatory Therapies
Emerging treatments targeting inflammatory and neuroregenerative pathways show promise for DED. The JAK-STAT pathway, activated by cytokines such as IL-2, IL-6, IL-17, and TNF-α, along with the SYK, which facilitates antigen presentation and immune cell recruitment, represents a key mediator of ocular surface inflammation [,]. In preclinical models, topical JAK inhibitors, such as tofacitinib, have demonstrated notable anti-inflammatory effects, including reduced expression of conjunctival HLA-DR. However, authors emphasise that larger clinical trials are needed to confirm these findings and establish therapeutic efficacy [,].
Recombinant human nerve growth factor (rhNGF, Cenegermin), approved for neurotrophic keratitis, promotes epithelial healing, nerve regeneration, tear secretion, and tear film stability, making it a promising option for severe DED []. ST-100, a collagen-mimetic peptide, supports corneal nerve regeneration and ECM repair, particularly in patients with corneal hypoesthesia [].
Targeting TRPV1 also offers therapeutic potential by reducing aberrant pain signaling and associated discomfort, thereby addressing the emotional component of chronic ocular pain [,]. TRPV1 antagonists, such as capsazepine, show promise for neuropathic DED, while broader TRP channel modulators enhance tear secretion, increase tear meniscus height, reduce conjunctival hyperemia, and improve both quality of life and daily functioning [].
6.3. Multidisciplinary and Lifestyle Interventions
A multidisciplinary approach is increasingly recognised as essential for effective DED management. In the mouse model, combined treatment with the anti-inflammatory lifitegrast and antioxidant tocopherol outperformed monotherapy by reducing inflammation, oxidative stress, and corneal damage while improving tear volume and stability []. A 10-week exercise program integrating cognitive-behavioural therapy also improved dry eye symptoms and psychological well-being in healthy office workers, highlighting the importance of lifestyle and stress management [].
New therapies targeting neurosensory pathways show promise in DED. Varenicline nasal spray, acting on nicotinic receptors of trigeminal nerves, significantly increased tear production over 12 weeks in clinical trials without serious adverse effects []. In a rabbit study, topical gabapentin demonstrated analgesic, anti-inflammatory, and tear-enhancing effects, likely through increased expression of acetylcholine, norepinephrine, and aquaporin 5 in lacrimal glands []. A cross-sectional retrospective study suggests that botulinum toxin A (BoNT-A) alleviates migraine, photophobia, and dry eye symptoms by modulating CGRP-mediated neural pathways [].
7. Integrated Management Strategies
DED is a multifactorial disease involving tear instability, inflammation, neurosensory dysfunction, and psychological factors. Effective treatment requires a comprehensive, personalised approach that addresses both ocular and systemic causes, particularly when symptoms exceed clinical findings [,,,].
7.1. Personalised Approaches and Phenotyping
DED exhibits considerable heterogeneity, with some patients experiencing severe symptoms despite minimal signs [,]. Psychiatric comorbidities and the use of psychotropic medications can further destabilise the tear film and exacerbate ocular surface inflammation, contributing to persistent symptoms and suboptimal therapeutic outcomes [,,,].
A stepwise, personalised approach should begin with conventional therapies including preservative-free artificial tears, topical anti-inflammatories such as cyclosporine A or lifitegrast, and environmental adjustments to reduce evaporative stress []. As discussed, emerging neuromodulatory treatments, including topical nerve growth factor (NGF), TRPV1 antagonists, and nicotinic agonists, such as varenicline nasal spray, show promise in restoring corneal innervation and enhancing tear secretion [,,].
In cases of neuropathic ocular pain, pharmacological agents such as gabapentinoids, low-dose naltrexone, or botulinum toxin A may reduce pain sensitivity and modulate aberrant trigeminal signalling [,,,]. When symptoms and signs differ markedly, mental health screening tools such as Patient Health Questionnaire-9 (PHQ-9) and Generalised Anxiety Disorder-7 (GAD-7) can support identification of psychological distress [,]. Referral to mental health specialists is advised if psychiatric factors impair treatment response. This integrated approach targets both ocular and central emotional factors, enhancing therapeutic effectiveness and long-term outcomes.
7.2. Ocular Impact of Psychotropic Medications
The association between DED and psychiatric disorders is well established. Shared mechanisms likely involve immune dysregulation, chronic inflammation, and genetic pleiotropy, as supported by genome-wide association studies [].
Combining standard DED treatments with psychiatric therapy, pharmacological or psychotherapeutic, can improve symptom control and overall well-being []. However, many psychotropic drugs, particularly those with anticholinergic or sympathomimetic effects, may worsen DED by reducing tear secretion and damaging the ocular surface [,,,,]. This risk increases with polypharmacy, underscoring the need for careful medication review and close collaboration between ophthalmologists and mental health professionals to optimise care [,].
7.3. Patient Education, Adherence and Psychosocial Support
Effective DED management extends beyond pharmacotherapy, requiring patient education, adherence, and psychosocial support. Patients should be informed about non-modifiable risk factors, including age, female sex, East Asian ethnicity, as well as modifiable behaviours such as screen overuse, poor sleep, contact lens wear, and exposure to low humidity [,,].
Education enhances adherence, sets realistic expectations, and supports lifestyle changes. Chronic ocular discomfort can severely impact quality of life, particularly in vulnerable groups like those with Sjögren’s syndrome [,,]. Integrating psychological support and routine mental health screening enables more holistic, patient-centred DED care [].
7.4. Integrated Ophthalmologic–Psychiatric Care in Dry Eye Disease
Symptom–sign discrepancy in DED often reflects underlying psychological factors such as anxiety, emotional dysregulation, or somatisation [,,], which, if unaddressed, can lead to persistent discomfort, poor adherence, and reduced treatment efficacy.
Integrated care between ophthalmologists and psychiatrists is essential, involving routine mental health screening, awareness of psychotropic ocular effects, and timely referral. Unaddressed psychological distress can amplify symptoms and promote central sensitisation [,]. Interdisciplinary coordination supports early detection of comorbidities, personalised treatment, and better quality of life. By targeting both peripheral and central drivers, it offers a more effective, holistic approach to managing DED in patients with psychiatric comorbidities [].
8. Clinical Implementation and Care Pathways
Recognising the bidirectional link between DED and psychiatric disorders is essential for effective, patient-centred care. Psychiatric comorbidities such as depression, anxiety, PTSD, and sleep disturbances are common in DED and amplify symptom perception, impair adherence, and reduce treatment response [,,,,,,,,,,,,,,]. Integrating ophthalmologic and psychiatric assessment enables early identification of these comorbidities and guides more personalised therapy. Routine use of brief screening instruments (e.g., PHQ-9, GAD-7) is recommended, particularly in patients who exhibit symptom–sign discrepancy or poor response to conventional treatment [,,,]. Interdisciplinary collaboration facilitates timely referral, optimises psychotropic medication choice, and enhances overall outcomes.
Emerging translational tools, including tear-based biomarkers (serotonin, BDNF, IL-6, TNF-α), offer potential for early diagnosis, patient stratification, and monitoring of treatment response. Combined with multimodal imaging and psychological profiling, they may support precision medicine approaches that bridge ophthalmology and psychiatry [,,,,,,,,,]. Implementing integrated care pathways, linking ophthalmologists, psychiatrists, and primary care providers, can improve quality of life, reduce chronicity, and address the full biopsychosocial spectrum of DED.
9. Future Perspectives
Despite increasing evidence supporting the interplay between DED and psychiatric disorders, significant knowledge gaps remain [,,,,,,,,,,]. To translate current insights into clinical practice, several key areas warrant further investigation. Table 3 outlines strategic research directions aimed at advancing our understanding and management of DED, as well as its intersection with psychiatric disorders [,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,,].
Table 3.
Strategic Research Priorities to Bridge Dry Eye Disease and Psychiatric Disorders.
From a translational perspective, there is great promise in the development of tear-based biomarkers such as BDNF, serotonin, IL-6, TNF-α and cortisol. These biomarkers may help stratify patients, predict disease severity, monitor treatment response, and guide personalised therapeutic approaches. The integration of tear proteomics and metabolomics into clinical practice may eventually enable point-of-care diagnostics that bridge the ophthalmologic and psychiatric domains [,,,,,,,,,,].
Future research should prioritise large-scale, longitudinal cohort studies to clarify the temporal dynamics and causal relationships between DED and psychiatric disorders. Most current studies are cross-sectional, limiting the ability to determine whether psychological symptoms precede ocular changes or arise as a consequence of chronic ocular discomfort [,,,,,,,,,,]. Elucidating this directionality is essential for timely prevention, risk stratification, and early intervention strategies. Standardised diagnostic tools, both ophthalmological (such as tear osmolarity, IVCM, and tear proteomics) and psychiatric (including clinician-administered diagnostic interviews), should be employed consistently across studies to enable meaningful comparisons and facilitate subgroup analysis [,,,,,,]. Combining these modalities may significantly enhance early detection of high-risk phenotypes and inform tailored therapeutic approaches.
Moreover, future investigations should explore neuroimaging correlates of central sensitisation in DED, particularly using fMRI to assess alterations in limbic and pain-processing circuits. Parallel studies of corneal nerve structure using IVCM and brain activity may illuminate novel neuro-ocular signatures of comorbidity. Combining in vivo confocal microscopy, tear proteomics, optical coherence tomography, and standardised psychiatric assessment tools could improve early detection and stratification of high-risk patients [,,,,,,,,,,].
Furthermore, establishing composite biomarker panels and threshold values for clinical decision-making may advance the field toward precision medicine. Tear fluid, as a non-invasive and information-rich biological sample, represents a unique translational bridge between ocular surface disease and central nervous system dysfunction [,,,,,,,,,,].
Emerging evidence highlights the significance of the gut–eye–brain axis in regulating both ocular surface homeostasis and neuropsychiatric function. Gut microbiota dysbiosis can disrupt systemic and central immune responses through the production of microbial metabolites, including short-chain fatty acids, tryptophan catabolites, and lipopolysaccharides. These molecules influence HPA axis activity, alter blood–brain barrier permeability, and modulate cytokine release, thereby contributing to both dry eye symptoms and psychiatric morbidity. In particular, gut-derived signals affect central serotonin and GABA pathways, linking microbial composition to emotional regulation, pain perception, and ocular surface inflammation. Furthermore, recent studies suggest that microbiota-driven immune dysregulation may impair lacrimal gland function and promote tear film instability, reinforcing the gut’s role in ocular disease [,,,,,].
In parallel, artificial intelligence (AI) and digital phenotyping are emerging as powerful tools for early detection, stratification, and personalised management of DED-psychiatric comorbidity. Machine learning models trained on multimodal datasets, including tear biomarker profiles, neuroimaging, patient-reported outcomes, and wearable sensor data, can uncover novel phenotypic clusters, predict therapeutic response, and support precision diagnostics. Smartphone-based passive monitoring and natural language processing of subjective symptom reports further enable real-time detection of emotional distress in DED patients. Integrating AI technologies with molecular diagnostics and brain–ocular correlates may accelerate the development of predictive models and multidisciplinary treatment strategies [,,,].
At the therapeutic level, interventions targeting shared mechanisms, such as anti-inflammatory agents with central effects, neurotrophic modulators, and antioxidant strategies, hold promise for dual benefit. Clinical studies should evaluate whether therapies targeting one domain, such as antidepressants and psychotherapy, affect outcomes in the other (ocular pain, inflammation), and vice versa. Agents that modulate the BDNF–TrkB pathway or regulate serotonin signalling may simultaneously alleviate ocular and psychiatric symptoms. Clinical trials assessing such agents should include ocular endpoints as secondary outcomes [,,,,,,,,,,,,].
Clinically, a multidisciplinary model of care is essential. Ophthalmologists should routinely screen for psychiatric comorbidities in refractory DED cases, particularly when symptoms outweigh clinical findings. Mental health professionals should consider DED a potential somatic manifestation of mood disorders or a side effect of therapy. Patient-centred communication and cross-speciality collaboration are key to improving outcomes [,,,,,,,,,,,].
Finally, public health strategies should emphasise awareness campaigns, clinical guidelines, and patient empowerment initiatives to promote early screening, reduce diagnostic delays, and encourage interdisciplinary care. Specific at-risk populations such as postmenopausal women, patients with Sjögren’s syndrome, patients with PTSD, and elderly individuals on multiple medications should be prioritised for screening and multidisciplinary intervention [,,,,,,,,,].
While future research directions highlight promising avenues for translational and clinical advancement, several limitations of the present review should be acknowledged. As a narrative synthesis, this review is inherently qualitative and cannot establish causal relationships; instead, it outlines associations and mechanistic hypotheses linking DED and psychiatric disorders. Many cited studies are preclinical, cross-sectional, or based on small cohorts, which limits the generalizability of the findings. Consequently, distinctions between correlation, causation, and theoretical assumptions are sometimes unclear.
Research at the interface of psychiatry and ocular health is also susceptible to selection bias and relies heavily on subjective symptom reporting, while the lack of standardised diagnostic criteria for both DED and psychiatric conditions further restricts comparability across studies. Evidence regarding tear biomarkers, particularly serotonin and BDNF, remains preliminary, largely stemming from preclinical or small-sample investigations. Inconsistent sampling protocols and diurnal variability further complicate interpretation. Moreover, human translational data are still limited relative to preclinical findings, and the differentiation between primary DED and secondary DED induced by psychopharmacological treatments has not been adequately addressed. Future research should therefore prioritise large, longitudinal, and well-controlled clinical and translational studies that employ standardised methodologies to clarify causal mechanisms and enhance the clinical relevance of neuroimmune and neuropsychiatric findings in DED.
10. Conclusions
DED should be recognised not merely as a local ocular disorder but as a systemic condition with profound neuroimmune, psychosomatic, and psychiatric dimensions. Adopting a systems-level, biopsychosocial model that integrates ophthalmology and psychiatry provides a promising framework for both clinical practice and scientific progress. Routine mental health screening, interdisciplinary collaboration, and patient-centred communication can enhance diagnostic precision, optimise therapeutic outcomes, and substantially improve quality of life. Translating molecular and neuroimmune insights into patient-specific interventions remains crucial. Shared mechanisms, including systemic inflammation, HPA axis dysregulation, oxidative stress, mitochondrial dysfunction, and reduced BDNF expression, represent both pathophysiological drivers and therapeutic targets. Ultimately, bridging ophthalmology and psychiatry through integrated research and clinical innovation will foster the development of targeted, multimodal strategies that address both peripheral and central disease pathways. Such an integrated approach has the potential to transform patient care and improve quality of life in this complex and frequently under-recognised patient population. Recognising dry eye disease as a systemic, neuroimmune-driven disorder with strong psychiatric interplay underscores the need for integrated, multidisciplinary approaches that bridge ophthalmology and psychiatry in both research and patient care.
Author Contributions
Conceptualisation, S.K. and L.K.; data curation, S.K., I.B., T.M., Z.T. and D.V.; original draft preparation and writing, S.K., L.K., I.B. and Z.T.; review and editing, S.K., Z.T. and L.K.; visualisation, Z.T., D.V. and T.M.; supervision, S.K. and L.K. All authors have read and agreed to the published version of the manuscript.
Funding
This research received no external funding.
Institutional Review Board Statement
Not applicable.
Informed Consent Statement
Not applicable.
Data Availability Statement
No new data were created or analyzed in this study. Data sharing is not applicable to this article.
Acknowledgments
We acknowledge Angela Budimir, University Hospital Dubrava, for proofreading the manuscript.
Conflicts of Interest
The authors declare no conflicts of interest.
References
- Stapleton, F.; Argüeso, P.; Asbell, P.; Azar, D.; Bosworth, C.; Chen, W.; Ciolino, J.B.; Craig, J.P.; Gallar, J.; Galor, A.; et al. TFOS DEWS III: Digest. Arch. Ophthalmol. 2025, 279, 451–553. [Google Scholar] [CrossRef]
- Kaštelan, S.; Gabrić, K.; Mikuličić, M.; Zimak, D.M.; Karabatić, M.; Antunica, A.G. The Influence of Tear Film Quality on Visual Function. Vision 2024, 8, 8. [Google Scholar] [CrossRef] [PubMed]
- Huang, R.; Su, C.; Fang, L.; Lu, J.; Chen, J.; Ding, Y. Dry eye syndrome: Comprehensive etiologies and recent clinical trials. Int. Ophthalmol. 2022, 42, 3253–3272. [Google Scholar] [CrossRef]
- Kaštelan, S.; Tomić, M.; Salopek-Rabatić, J.; Novak, B. Diagnostic Procedures and Management of Dry Eye. BioMed Res. Int. 2013, 2013, 309723. [Google Scholar] [CrossRef] [PubMed]
- He, Q.; Chen, Z.; Xie, C.; Liu, L.; Wei, R. The Association Between Dry Eye Disease with Depression, Anxiety and Sleep Disturbance During COVID-19. Front. Psychiatry 2022, 12, 802302. [Google Scholar] [CrossRef]
- Basilious, A.; Xu, C.Y.; Malvankar-Mehta, M.S. Dry eye disease and psychiatric disorders: A systematic review and meta-analysis. Eur. J. Ophthalmol. 2021, 32, 1872–1889. [Google Scholar] [CrossRef]
- Wan, K.H.; Chen, L.J.; Young, A.L. Depression and anxiety in dry eye disease: A systematic review and meta-analysis. Eye 2016, 30, 1558–1567. [Google Scholar] [CrossRef]
- Galor, A.; Feuer, W.; Lee, D.J.; Florez, H.; Faler, A.L.; Zann, K.L.; Perez, V.L. Depression, Post-traumatic Stress Disorder, and Dry Eye Syndrome: A Study Utilizing the National United States Veterans Affairs Administrative Database. Arch. Ophthalmol. 2012, 154, 340–346.e2. [Google Scholar] [CrossRef] [PubMed]
- Ayaki, M.; Tsubota, K.; Kawashima, M.; Kishimoto, T.; Mimura, M.; Negishi, K. Sleep Disorders are a Prevalent and Serious Comorbidity in Dry Eye. Investig. Opthalmology Vis. Sci. 2018, 59, DES143–DES150. [Google Scholar] [CrossRef]
- Galor, A.; Feuer, W.; Lee, D.J.; Florez, H.; Carter, D.; Pouyeh, B.; Prunty, W.J.; Perez, V.L. Prevalence and Risk Factors of Dry Eye Syndrome in a United States Veterans Affairs Population. Arch. Ophthalmol. 2011, 152, 377–384.e2. [Google Scholar] [CrossRef]
- Eroglu, F.C.; Gediz, B.S.; Ozturk, M.; Kazancı, B. Association Between Dry Eye Disease and Newly Diagnosed Obsessive-Compulsive Disorder. Cornea 2021, 40, 817–821. [Google Scholar] [CrossRef]
- Bartlett, J.D.; Keith, M.S.; Sudharshan, L.; Snedecor, S.J. Associations between signs and symptoms of dry eye disease: A systematic review. Clin. Ophthalmol. 2015, 9, 1719–1730. [Google Scholar] [CrossRef]
- Ong, E.S.; Felix, E.R.; Levitt, R.C.; Feuer, W.J.; Sarantopoulos, C.D.; Galor, A. Epidemiology of discordance between symptoms and signs of dry eye. Br. J. Ophthalmol. 2017, 102, 674–679. [Google Scholar] [CrossRef]
- Bitar, M.S.; Olson, D.J.; Li, M.; Davis, R.M. The Correlation Between Dry Eyes, Anxiety and Depression: The Sicca, Anxiety and Depression Study. Cornea 2019, 38, 684–689. [Google Scholar] [CrossRef]
- Hua, R.; Yao, K.; Hu, Y.; Chen, L. Discrepancy between subjectively reported symptoms and objectively measured clinical findings in dry eye: A population based analysis. BMJ Open 2014, 4, e005296. [Google Scholar] [CrossRef] [PubMed]
- Kitazawa, M.; Sakamoto, C.; Yoshimura, M.; Kawashima, M.; Inoue, S.; Mimura, M.; Tsubota, K.; Negishi, K.; Kishimoto, T. The Relationship of Dry Eye Disease with Depression and Anxiety: A Naturalistic Observational Study. Transl. Vis. Sci. Technol. 2018, 7, 35. [Google Scholar] [CrossRef] [PubMed]
- Ayoubi, M.; Cabrera, K.; Mangwani-Mordani, S.; Locatelli, E.V.T.; Galor, A. Associations between dry eye disease and sleep quality: A cross-sectional analysis. BMJ Open Ophthalmol. 2024, 9, e001584. [Google Scholar] [CrossRef] [PubMed]
- Kim, M.; Chun, Y.S.; Kim, K.W. Different perception of dry eye symptoms between patients with and without primary Sjogren’s syndrome. Sci. Rep. 2022, 12, 2172. [Google Scholar] [CrossRef]
- Fachinetti, A.; Marelli, E.; Velati, P.; Minoretti, P.; De Palma, G.; Sigurtà, C. Subjective Dry Eye Symptoms and Objective Ocular Surface Signs in a Civil Air Crew Population: A Cross-Sectional Study. Cureus 2024, 16, e51447. [Google Scholar] [CrossRef]
- Han, S.B.; Yang, H.K.; Hyon, J.Y.; Wee, W.R. Association of dry eye disease with psychiatric or neurological disorders in elderly patients. Clin. Interv. Aging 2017, ume 12, 785–792. [Google Scholar] [CrossRef]
- Michelsen, B.; Kristianslund, E.K.; Hammer, H.B.; Fagerli, K.M.; Lie, E.; Wierød, A.; Kalstad, S.; Rødevand, E.; Krøll, F.; Haugeberg, G.; et al. Discordance between tender and swollen joint count as well as patient’s and evaluator’s global assessment may reduce likelihood of remission in patients with rheumatoid arthritis and psoriatic arthritis: Data from the prospective multicentre NOR-DMARD study. Ann. Rheum. Dis. 2017, 76, 708–711. [Google Scholar] [CrossRef]
- Yu, H.; Zeng, W.; Xu, M.; Wu, W.; Feng, Y. Experience of Dry Eye Patients with Anxiety and Depression: A Qualitative Study. Front. Med. 2022, 9, 830986. [Google Scholar] [CrossRef]
- Kaštelan, S.; Bakija, I.; Bogadi, M.; Antunica, A.G.; Gotovac, M.; Filipčić, I.Š. Mental Disorders as Influencing Factors for Discordances in the Signs and Symptoms of Dry Eye Disease. Psychiatr Danub. 2021, 33, 588–595. [Google Scholar]
- Ma, S.; Yang, J.; Cheng, H.; Wang, W.; Chen, G.; Bai, H.; Yao, L.; Liu, Z. The central symptoms of depression, anxiety, and somatization: A network analysis. All Life 2022, 15, 933–941. [Google Scholar] [CrossRef]
- Ran, L.; Wang, W.; Ai, M.; Kong, Y.; Chen, J.; Kuang, L. Psychological resilience, depression, anxiety, and somatization symptoms in response to COVID-19: A study of the general population in China at the peak of its epidemic. Soc. Sci. Med. 2020, 262, 113261. [Google Scholar] [CrossRef] [PubMed]
- Yavrum, F.; Ozates, M.E.; Ozates, S. An important contributing factor in dry eye disease: Somatization. Int. Ophthalmol. 2024, 44, 190. [Google Scholar] [CrossRef] [PubMed]
- McMonnies, C.W. Why the symptoms and objective signs of dry eye disease may not correlate. J. Optom. 2021, 14, 3–10. [Google Scholar] [CrossRef]
- Hassamal, S. Chronic stress, neuroinflammation, and depression: An overview of pathophysiological mechanisms and emerging anti-inflammatories. Front. Psychiatry 2023, 14, 1130989. [Google Scholar] [CrossRef] [PubMed]
- Chhadva, P.; Lee, T.; Sarantopoulos, C.D.; Hackam, A.S.; McClellan, A.L.; Felix, E.R.; Levitt, R.C.; Galor, A. Human Tear Serotonin Levels Correlate with Symptoms and Signs of Dry Eye. Ophthalmology 2015, 122, 1675–1680. [Google Scholar] [CrossRef]
- Benítez-Del-Castillo, J.M.; Acosta, M.C.; Wassfi, M.A.; Díaz-Valle, D.; Gegúndez, J.A.; Fernandez, C.; García-Sánchez, J. Relation between Corneal Innervation with Confocal Microscopy and Corneal Sensitivity with Noncontact Esthesiometry in Patients with Dry Eye. Investig. Opthalmology Vis. Sci. 2007, 48, 173–181. [Google Scholar] [CrossRef]
- Ma, B.; Xie, J.; Yang, T.; Su, P.; Liu, R.; Sun, T.; Zhou, Y.; Wang, H.; Feng, X.; Ma, S.; et al. Quantification of Increased Corneal Subbasal Nerve Tortuosity in Dry Eye Disease and Its Correlation with Clinical Parameters. Transl. Vis. Sci. Technol. 2021, 10, 26. [Google Scholar] [CrossRef]
- Cox, S.M.; Kheirkhah, A.; Aggarwal, S.; Abedi, F.; Cavalcanti, B.M.; Cruzat, A.; Hamrah, P. Alterations in corneal nerves in different subtypes of dry eye disease: An in vivo confocal microscopy study. Ocul. Surf. 2021, 22, 135–142. [Google Scholar] [CrossRef]
- Jastrzębska-Miazga, I.; Machna, B.; Wyględowska-Promieńska, D.; Smędowski, A. The Role of Substance P in Corneal Homeostasis. Biomolecules 2025, 15, 729. [Google Scholar] [CrossRef]
- Zhang, Y.; Zhang, H.; Zhu, X.; Ye, H.; Yang, K.; Zhou, X.; Hong, J. Tear neuropeptides are associated with clinical symptoms and signs of dry eye patients. Ann. Med. 2025, 57, 2451194. [Google Scholar] [CrossRef]
- Aleixandre-Carrera, F.; Engelmayer, N.; Ares-Suárez, D.; Acosta, M.d.C.; Belmonte, C.; Gallar, J.; Meseguer, V.; Binshtok, A.M. Optical Assessment of Nociceptive TRP Channel Function at the Peripheral Nerve Terminal. Int. J. Mol. Sci. 2021, 22, 481. [Google Scholar] [CrossRef] [PubMed]
- He, Q.; Chen, Z.; Xie, C.; Liu, L.; Yang, H.; Wei, R. Relationship Between Dry Eye Disease and Emotional Disorder: The Mediating Effect of Health Anxiety. Front. Public Health 2022, 10, 771554. [Google Scholar] [CrossRef]
- Li, X.; Zhang, H.; Han, X.; Guo, L.; Ceban, F.; Liao, Y.; Shi, J.; Wang, W.; Liu, Y.; Song, W.; et al. Predictive potential of somatic symptoms for the identification of subthreshold depression and major depressive disorder in primary care settings. Front. Psychiatry 2023, 14, 999047. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Y.; Murrough, J.; Yu, Y.; Roy, N.; Sayegh, R.; Asbell, P.; Maguire, M.G.; Ying, G.S.; DREAM Study Research Group. Association Between Depression and Severity of Dry Eye Symptoms, Signs, and Inflammatory Markers in the DREAM Study. JAMA Ophthalmol. 2022, 140, 392–399. [Google Scholar] [CrossRef]
- Bakija, I.; Filipčić, I.; Bogadi, M.; Filipčić, I.Š.; Gotovac, M.; Kaštelan, S. Comparison of the Schein and Osdi Questionnaire as Indicator of Tear Film Stability in Patients with Schizophrenia. Psychiatr Danub. 2021, 33, 596–603. [Google Scholar] [PubMed]
- Takeuchi, T.; Hashimoto, K.; Koyama, A.; Asakura, K.; Hashizume, M. The Association of Central Sensitisation with Depression, Anxiety, and Somatic Symptoms: A Cross-Sectional Study of a Mental Health Outpatient Clinic in Japan. Life 2024, 14, 612. [Google Scholar] [CrossRef]
- Magno, M.S.; Wolpert, L.E.; Morthen, M.K.; Utheim, T.P.; Hammond, C.J.; Vehof, J. The association between personality and dry eye disease: A large cross-sectional study. Ocul. Surf. 2025, 37, 132–139. [Google Scholar] [CrossRef] [PubMed]
- Toth, M.; Jokić-Begić, N.; Krašić, S. The Relationship Between Anxiety Sensitivity, Emotional States, and Dry Eye Disease Symptom Severity: A Cross-Sectional Study. Vision 2025, 9, 36. [Google Scholar] [CrossRef] [PubMed]
- Tong, L.; Waduthantri, S.; Wong, T.Y.; Saw, S.M.; Wang, J.J.; Rosman, M.; Lamoureux, E. Impact of symptomatic dry eye on vision-related daily activities: The Singapore Malay Eye Study. Eye 2010, 24, 1486–1491. [Google Scholar] [CrossRef]
- Kastelan, S.; Bakija, I.; Bogadi, M.; Oreškovic, I.; Kasun, B.; Gotovac, M.; Antunica, A.G. Psychiatric Disorders and Dry Eye Disease—A Transdisciplinary Approach. Psychiatr Danub. 2021, 33, S580–S587. [Google Scholar]
- van der Vaart, R.; Weaver, M.A.; Lefebvre, C.; Davis, R.M. The Association Between Dry Eye Disease and Depression and Anxiety in a Large Population-Based Study. Arch. Ophthalmol. 2015, 159, 470–474. [Google Scholar] [CrossRef]
- Liang, C.-Y.; Cheang, W.-M.; Wang, C.-Y.; Lin, K.-H.; Wei, L.-C.; Chen, Y.-Y.; Shen, Y.-C. The association of dry eye syndrome and psychiatric disorders: A nationwide population-based cohort study. BMC Ophthalmol. 2020, 20, 123. [Google Scholar] [CrossRef]
- Lee, W.; Lim, S.-S.; Won, J.-U.; Roh, J.; Lee, J.-H.; Seok, H.; Yoon, J.-H. The association between sleep duration and dry eye syndrome among Korean adults. Sleep. Med. 2015, 16, 1327–1331. [Google Scholar] [CrossRef] [PubMed]
- Liyue, H.; Chiang, P.P.; Sung, S.C.; Tong, L. Dry eye-related visual blurring and irritative symptoms and their association with depression and anxiety in eye clinic patients. Curr. Eye Res. 2016, 41, 590–599. [Google Scholar] [CrossRef]
- Vehof, J.; Snieder, H.; Jansonius, N.; Hammond, C.J. Prevalence and risk factors of dry eye in 79,866 participants of the population-based Lifelines cohort study in the Netherlands. Ocul. Surf. 2021, 19, 83–93. [Google Scholar] [CrossRef]
- Herman, J.P.; McKlveen, J.M.; Ghosal, S.; Kopp, B.; Wulsin, A.; Makinson, R.; Scheimann, J.; Myers, B. Regulation of the hypothalamic-pituitary-adrenocortical stress response. Compr. Physiol. 2016, 6, 603–621. [Google Scholar] [CrossRef]
- Lei, A.A.; Phang, V.W.X.; Lee, Y.Z.; Kow, A.S.F.; Tham, C.L.; Ho, Y.-C.; Lee, M.T. Chronic Stress-Associated Depressive Disorders: The Impact of HPA Axis Dysregulation and Neuroinflammation on the Hippocampus—A Mini Review. Int. J. Mol. Sci. 2025, 26, 2940. [Google Scholar] [CrossRef]
- Dressle, R.J.; Feige, B.; Spiegelhalder, K.; Schmucker, C.; Benz, F.; Mey, N.C.; Riemann, D. HPA axis activity in patients with chronic insomnia: A systematic review and meta-analysis of case–control studies. Sleep Med. Rev. 2022, 62, 101588. [Google Scholar] [CrossRef]
- Bertollo, A.G.; Santos, C.F.; Bagatini, M.D.; Ignácio, Z.M. Hypothalamus-pituitary-adrenal and gut-brain axes in biological interaction pathway of the depression. Front. Neurosci. 2025, 19, 1541075. [Google Scholar] [CrossRef] [PubMed]
- Iob, E.; Kirschbaum, C.; Steptoe, A. Persistent depressive symptoms, HPA-axis hyperactivity, and inflammation: The role of cognitive-affective and somatic symptoms. Mol. Psychiatry 2019, 25, 1130–1140. [Google Scholar] [CrossRef]
- Galor, A.; Levitt, R.C.; McManus, K.T.; Kalangara, J.P.; Seiden, B.E.; Park, J.J.; Covington, D.B.; Sarantopoulos, C.D.; Felix, E.R. Assessment of Somatosensory Function in Patients with Idiopathic Dry Eye Symptoms. JAMA Ophthalmol. 2016, 134, 1290–1298. [Google Scholar] [CrossRef] [PubMed]
- Hyon, J.Y.; Yang, H.K.; Han, S.B. Association between Dry Eye Disease and Psychological Stress among Paramedical Workers in Korea. Sci. Rep. 2019, 9, 3783. [Google Scholar] [CrossRef]
- Glavina, A.; Zurak, P.; Martinović, D.; Gotovac, M.; Šupe-Domić, D.; Lugović-Mihić, L. Evaluation of the Hypothalamic–Pituitary–Adrenal Axis in Patients with Primary Sjögren’s Disease. Medicina 2024, 60, 1886. [Google Scholar] [CrossRef]
- Alkozi, H.A.A.H.A.; Alhudhayf, H.; Alawad, N.M.A. Association Between Dry Eye Disease with Anxiety and Depression Among Medical Sciences Students in Qassim Region: Cortisol Levels in Tears as a Stress Biomarker. J. Multidiscip. Health 2024, 17, 4549–4557. [Google Scholar] [CrossRef]
- Kumari, M.; Badrick, E.; Ferrie, J.; Perski, A.; Marmot, M.; Chandola, T. Self-Reported Sleep Duration and Sleep Disturbance Are Independently Associated with Cortisol Secretion in the Whitehall II Study. J. Clin. Endocrinol. Metab. 2009, 94, 4801–4809. [Google Scholar] [CrossRef] [PubMed]
- Fries, G.R.; Saldana, V.A.; Finnstein, J.; Rein, T. Molecular pathways of major depressive disorder converge on the synapse. Mol. Psychiatry 2023, 28, 284–297. [Google Scholar] [CrossRef]
- Elhwuegi, A.S. Central monoamines and their role in major depression. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2004, 28, 435–451. [Google Scholar] [CrossRef]
- Hao, S.; Shi, W.; Liu, W.; Chen, Q.-Y.; Zhuo, M. Multiple modulatory roles of serotonin in chronic pain and injury-related anxiety. Front. Synaptic Neurosci. 2023, 15, 1122381. [Google Scholar] [CrossRef] [PubMed]
- International Classification of Disease (ICD), 11th Edition. Depressive Disorders. 2025. Available online: https://icd.who.int/browse/2025-01/mms/en#578635574 (accessed on 25 August 2025).
- Han, C.; Pae, C.-U. Pain and Depression: A Neurobiological Perspective of Their Relationship. Psychiatry Investig. 2015, 12, 1–8. [Google Scholar] [CrossRef]
- Nijs, J.; Leysen, L.; Vanlauwe, J.; Logghe, T.; Ickmans, K.; Polli, A.; Malfliet, A.; Coppieters, I.; Huysmans, E. Treatment of central sensitization in patients with chronic pain: Time for change? Expert. Opin. Pharmacother. 2019, 20, 1961–1970. [Google Scholar] [CrossRef]
- Ohashi, Y.; Uchida, K.; Fukushima, K.; Inoue, G.; Takaso, M. Mechanisms of Peripheral and Central Sensitization in Osteoarthritis Pain. Cureus 2023, 15, e35331. [Google Scholar] [CrossRef]
- Desch, S.; Schweinhardt, P.; Seymour, B.; Flor, H.; Becker, S. Evidence for dopaminergic involvement in endogenous modulation of pain relief. eLife 2023, 12, e81436. [Google Scholar] [CrossRef] [PubMed]
- Strobel, C.; Hunt, S.; Sullivan, R.; Sun, J.; Sah, P. Emotional regulation of pain: The role of noradrenaline in the amygdala. Sci. China Life Sci. 2014, 57, 384–390. [Google Scholar] [CrossRef] [PubMed]
- Galor, A.; Covington, D.; Levitt, A.E.; McManus, K.T.; Seiden, B.; Felix, E.R.; Kalangara, J.; Feuer, W.; Patin, D.J.; Martin, E.R.; et al. Neuropathic Ocular Pain due to Dry Eye Is Associated with Multiple Comorbid Chronic Pain Syndromes. J. Pain. 2016, 17, 310–318. [Google Scholar] [CrossRef]
- Hole, C.; Dhamsania, A.; Brown, C.; Ryznar, R. Immune Dysregulation in Depression and Anxiety: A Review of the Immune Response in Disease and Treatment. Cells 2025, 14, 607. [Google Scholar] [CrossRef]
- Saggu, S.; Pless, A.; Dew, E.; Ware, D.; Jiao, K.; Wang, Q. Monoamine signaling and neuroinflammation: Mechanistic connections and implications for neuropsychiatric disorders. Front. Immunol. 2025, 16, 1543730. [Google Scholar] [CrossRef]
- Mrugacz, M.; Ostrowska, L.; Bryl, A.; Szulc, A.; Zelazowska-Rutkowska, B.; Mrugacz, G. Pro-inflammatory cytokines associated with clinical severity of dry eye disease of patients with depression. Adv. Med. Sci. 2017, 62, 338–344. [Google Scholar] [CrossRef]
- Yang, H.; Tian, Q.; Luan, L.; Yang, M.; Li, C.; Zhang, X. NGF-β and BDNF levels are altered in male patients with chronic schizophrenia: Effects on clinical symptoms. BMC Psychiatry 2025, 25, 240. [Google Scholar] [CrossRef]
- Alqahtani, S.M.; Al-Kuraishy, H.M.; Al Gareeb, A.I.; Albuhadily, A.K.; Alexiou, A.; Papadakis, M.; Hemeda, L.R.; Faheem, S.A.; Batiha, G.E.-S. Unlocking Alzheimer’s Disease: The Role of BDNF Signaling in Neuropathology and Treatment. Neuromolecular Med. 2025, 27, 36. [Google Scholar] [CrossRef]
- Yu, Z.; Joy, S.; Mi, T.; Yazdanpanah, G.; Burgess, K.; de Paiva, C.S. New, potent, small molecule agonists of tyrosine kinase receptors attenuate dry eye disease. Front. Med. 2022, 9, 937142. [Google Scholar] [CrossRef]
- Sano, K.; Kawashima, M.; Imada, T.; Suzuki, T.; Nakamura, S.; Mimura, M.; Tanaka, K.F.; Tsubota, K. Enriched environment alleviates stress-induced dry-eye through the BDNF axis. Sci. Rep. 2019, 9, 3422. [Google Scholar] [CrossRef]
- Malvasi, M.; Pascale, E.; Locuratolo, N.; Fattapposta, F.; Pauletti, C.; Finoia, M.G.; Conti, M.E.; Pacella, E. Implementing potential biomarkers for early detection of retinal changes in Parkinson’s disease: Preliminary study on BDNF role. Eur. J. Ophthalmol. 2025, 35, 1746–1760. [Google Scholar] [CrossRef] [PubMed]
- Hallak, J.A.; Tibrewal, S.; Mohindra, N.; Gao, X.; Jain, S. Single Nucleotide Polymorphisms in the BDNF, VDR, and DNASE 1 Genes in Dry Eye Disease Patients: A Case-Control Study. Investig. Ophthalmol. Vis. Sci. 2015, 56, 5990–5996. [Google Scholar] [CrossRef] [PubMed]
- Han, Y.; Chen, X.; Tang, G.; Peng, Q.; Liu, J.; Liu, Y.; Zhou, Q.; Long, L. A study on factors related to sleep disorders and serum BDNF expression levels in patients with primary Sjögren’s syndrome. Mod. Rheumatol. 2024, 35, 332–338. [Google Scholar] [CrossRef]
- Kan, H.-W.; Peng, W.-H.; Wu, C.-C.; Wang, D.-W.; Lee, M.T.; Lee, Y.-K.; Chu, T.-H.; Ho, Y.-C. Rapid antidepressant-like effects of muscarinic receptor antagonists require BDNF-dependent signaling in the ventrolateral periaqueductal gray. Psychopharmacology 2022, 239, 3805–3818. [Google Scholar] [CrossRef] [PubMed]
- Lee, Y.-R.; Park, K.-M.; Lee, N.-K.; Paik, H.-D. Neuroprotective effects of Lactiplantibacillus plantarum and Pediococcus pentosaceus strains against oxidative stress via modulation of Nrf2-mediated antioxidation and anti-apoptosis. Brain Res. 2025, 1866, 149925. [Google Scholar] [CrossRef]
- Tayeb, A.E.K.A.; Poinsignon, V.; Chappell, K.; Bouligand, J.; Becquemont, L.; Verstuyft, C. Major Depressive Disorder and Oxidative Stress: A Review of Peripheral and Genetic Biomarkers According to Clinical Characteristics and Disease Stages. Antioxidants 2023, 12, 942. [Google Scholar] [CrossRef]
- Black, C.N.; Bot, M.; Scheffer, P.G.; Cuijpers, P.; Penninx, B.W. Is depression associated with increased oxidative stress? A systematic review and meta-analysis. Psychoneuroendocrinology 2015, 51, 164–175. [Google Scholar] [CrossRef]
- Palta, P.P.; Samuel, L.J.P.; Miller, E.R.I.; Szanton, S.L.P. Depression and Oxidative Stress. Psychosom. Med. 2014, 76, 12–19. [Google Scholar] [CrossRef] [PubMed]
- Fu, Q.; Qiu, R.; Liang, J.; Wu, S.; Huang, D.; Qin, Y.; Li, Q.; Shi, X.; Xiong, X.; Jiang, Z.; et al. Sugemule-7 alleviates oxidative stress, neuroinflammation, and cell death, promoting synaptic plasticity recovery in mice with postpartum depression. Sci. Rep. 2025, 15, 1426. [Google Scholar] [CrossRef]
- Good, K.V.; Kalani, L.; Vincent, J.B.; Ausió, J. Multifaceted roles of MeCP2 in cellular regulation and phase separation: Implications for neurodevelopmental disorders, depression, and oxidative stress. Biochem. Cell Biol. 2025, 103, 1–12. [Google Scholar] [CrossRef]
- Su, M.; Hong, J.; Zhao, Y.; Liu, S.; Xue, X. MeCP2 controls hippocampal brain-derived neurotrophic factor expression via homeostatic interactions with microRNA-132 in rats with depression. Mol. Med. Rep. 2015, 12, 5399–5406. [Google Scholar] [CrossRef]
- Navel, V.; Sapin, V.; Henrioux, F.; Blanchon, L.; Labbé, A.; Chiambaretta, F.; Baudouin, C.; Dutheil, F. Oxidative and antioxidative stress markers in dry eye disease: A systematic review and meta-analysis. Acta Ophthalmol. 2021, 100, 45–57. [Google Scholar] [CrossRef]
- Seen, S.; Tong, L. Dry eye disease and oxidative stress. Acta Ophthalmol. 2018, 96, e412–e420. [Google Scholar] [CrossRef]
- Bu, J.; Liu, Y.; Zhang, R.; Lin, S.; Zhuang, J.; Sun, L.; Zhang, L.; He, H.; Zong, R.; Wu, Y.; et al. Potential New Target for Dry Eye Disease—Oxidative Stress. Antioxidants 2024, 13, 422. [Google Scholar] [CrossRef] [PubMed]
- Simpson, C.L.; Asbell, P.A.; Roy, N.S. Review of the Role of Oxidative Stress in Ocular Surface Disease and Use of MDA and 4-HNE in Tears as Potential Markers. J. Ophthalmic Res. Vis. Care 2022, 2, 1–12. [Google Scholar] [CrossRef]
- Liu, Z.; Li, Y.; Bao, J.; Li, S.; Wen, Y.; Zhang, P.; Feng, J.; Wang, Y.; Tian, L.; Jie, Y. Astaxanthin ameliorates benzalkonium chloride–induced dry eye disease through suppressing inflammation and oxidative stress via Keap1-Nrf2/HO-1 signaling pathways. Anim. Model. Exp. Med. 2025, 8, 1056–1079. [Google Scholar] [CrossRef]
- Chen, H.; Shi, J.; Tang, Y.; Chen, X.; Wang, Z.; Liu, Q.; Wu, K.; Yao, X. Exploring the effect of chlorogenic acid on oxidative stress and autophagy in dry eye mice via the AMPK/ULK1 pathway. Eur. J. Pharmacol. 2025, 991, 177311. [Google Scholar] [CrossRef]
- Arslan, M.A.; Rousseau, A.; Liang, H.; Chardonnet, S.; Pionneau, C.; Rabut, G.; Le Goazigo, A.R.; Labetoulle, M.; Baudouin, C.; Brignole-Baudouin, F.; et al. Deficiency of antioxidant enzyme PRDX6 and mucosal chemokine CXCL17: Unraveling the factors behind oxidative stress and mucosal homeostasis disruption in dry eye disease. Biochim. Et Biophys. Acta-Mol. Basis Dis. 2025, 1871, 167911. [Google Scholar] [CrossRef]
- Ouyang, W.; Yan, D.; Hu, J.; Liu, Z. Multifaceted mitochondrial as a novel therapeutic target in dry eye: Insights and interventions. Cell Death Discov. 2024, 10, 398. [Google Scholar] [CrossRef]
- Huang, B.; Zhang, N.; Qiu, X.; Zeng, R.; Wang, S.; Hua, M.; Li, Q.; Nan, K.; Lin, S. Mitochondria-targeted SkQ1 nanoparticles for dry eye disease: Inhibiting NLRP3 inflammasome activation by preventing mitochondrial DNA oxidation. J. Control. Release 2023, 365, 1–15. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.; Zhang, W.; Zheng, Y.; Xu, Y. Ameliorative Potential of Resveratrol in Dry Eye Disease by Restoring Mitochondrial Function. Evid.-Based Complement. Altern. Med. 2022, 2022, 013444. [Google Scholar] [CrossRef] [PubMed]
- Tan, X.; Chen, Q.; Chen, Z.; Sun, Z.; Chen, W.; Wei, R. Mitochondrial DNA-Activated cGAS-STING Signaling in Environmental Dry Eye. Investig. Ophthalmol. Vis. Sci. 2024, 65, 33. [Google Scholar] [CrossRef]
- Allen, J.; Romay-Tallon, R.; Brymer, K.J.; Caruncho, H.J.; Kalynchuk, L.E. Mitochondria and Mood: Mitochondrial Dysfunction as a Key Player in the Manifestation of Depression. Front. Neurosci. 2018, 12, 386. [Google Scholar] [CrossRef] [PubMed]
- Li, W.; Zhu, L.; Chen, Y.; Zhuo, Y.; Wan, S.; Guo, R. Association between mitochondrial DNA levels and depression: A systematic review and meta-analysis. BMC Psychiatry 2023, 23, 866. [Google Scholar] [CrossRef]
- Liu, L.; Cheng, S.; Qi, X.; Meng, P.; Yang, X.; Pan, C.; Chen, Y.; Zhang, H.; Zhang, Z.; Zhang, J.; et al. Mitochondria-wide association study observed significant interactions of mitochondrial respiratory and the inflammatory in the development of anxiety and depression. Transl. Psychiatry 2023, 13, 216. [Google Scholar] [CrossRef]
- Fernström, J.; Mellon, S.H.; McGill, M.A.; Picard, M.; Reus, V.I.; Hough, C.M.; Lin, J.; Epel, E.S.; Wolkowitz, O.M.; Lindqvist, D. Blood-based mitochondrial respiratory chain function in major depression. Transl. Psychiatry 2021, 11, 593. [Google Scholar] [CrossRef]
- Casaril, A.M.; Dantzer, R.; Bas-Orth, C. Neuronal Mitochondrial Dysfunction and Bioenergetic Failure in Inflammation-Associated Depression. Front. Neurosci. 2021, 15, 725547. [Google Scholar] [CrossRef]
- Hensler, J.G. Serotonin in Mood and Emotion. Handb. Behav. Neurosci. 2010, 21, 367–378. [Google Scholar] [CrossRef]
- Bamalan, O.A.; Moore, M.J.; Al Khalili, Y. Physiology, Serotonin. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2025. Available online: https://www.ncbi.nlm.nih.gov/books/NBK545168/ (accessed on 19 June 2025).
- Arnone, D.; Wise, T.; Fitzgerald, P.B.; Harmer, C.J. The involvement of serotonin in major depression: Nescience in disguise? Mol. Psychiatry 2024, 29, 200–202. [Google Scholar] [CrossRef]
- Lin, J.; Liu, W.; Guan, J.; Cui, J.; Shi, R.; Wang, L.; Chen, D.; Liu, Y. Latest updates on the serotonergic system in depression and anxiety. Front. Synaptic Neurosci. 2023, 15, 1124112. [Google Scholar] [CrossRef] [PubMed]
- Moncrieff, J.; Cooper, R.E.; Stockmann, T.; Amendola, S.; Hengartner, M.P.; Horowitz, M.A. The serotonin theory of depression: A systematic umbrella review of the evidence. Mol. Psychiatry 2022, 28, 3243–3256. [Google Scholar] [CrossRef] [PubMed]
- Howard, D.M.; Adams, M.J.; Clarke, T.-K.; Hafferty, J.D.; Gibson, J.; Shirali, M.; Coleman, J.R.I.; Hagenaars, S.P.; Ward, J.; Wigmore, E.M.; et al. Genome-wide meta-analysis of depression identifies 102 independent variants and highlights the importance of the prefrontal brain regions. Nat. Neurosci. 2019, 22, 343–352. [Google Scholar] [CrossRef]
- Bîlc, M.I.; Iacob, A.; Szekely-Copîndean, R.D.; Kiss, B.; Ștefan, M.-G.; Mureșan, R.C.; Pop, C.F.; Pițur, S.; Szentágotai-Tătar, A.; Vulturar, R.; et al. Serotonin and emotion regulation: The impact of tryptophan depletion on emotional experience, neural and autonomic activity. Cogn. Affect. Behav. Neurosci. 2023, 23, 1414–1427. [Google Scholar] [CrossRef]
- Park, I.; Choi, M.; Kim, J.; Jang, S.; Kim, D.; Kim, J.; Choe, Y.; Geum, D.; Yu, S.-W.; Choi, J.-W.; et al. Role of the circadian nuclear receptor REV-ERBα in dorsal raphe serotonin synthesis in mood regulation. Commun. Biol. 2024, 7, 998. [Google Scholar] [CrossRef]
- Liu, H.N.; Nakamura, M.; Kawashima, H. New Role of the Serotonin as a Biomarker of Gut–Brain Interaction. Life 2024, 14, 1280. [Google Scholar] [CrossRef] [PubMed]
- Imada, T.; Nakamura, S.; Hisamura, R.; Izuta, Y.; Jin, K.; Ito, M.; Kitamura, N.; Tanaka, K.F.; Mimura, M.; Shibuya, I.; et al. Serotonin hormonally regulates lacrimal gland secretory function via the serotonin type 3a receptor. Sci. Rep. 2017, 7, 6965. [Google Scholar] [CrossRef]
- Dartt, D.A.; Hodges, R.R.; Zoukhri, D. Tears and their secretion. Adv. Organ Biol. 2005, 10, 21–82. [Google Scholar] [CrossRef]
- Sharif, N.A.; Senchyna, M. Serotonin receptor subtype mRNA expression in human ocular tissues, determined by RT-PCR. Mol Vis. 2006, 12, 1040–1047. [Google Scholar]
- Crider, J.Y.; Williams, G.W.; Drace, C.D.; Katoli, P.; Senchyna, M.; Sharif, N.A. Pharmacological Characterization of a Serotonin Receptor (5-HT7) Stimulating cAMP Production in Human Corneal Epithelial Cells. Investig. Ophthalmol. Vis. Sci. 2003, 44, 4837–4844. [Google Scholar] [CrossRef] [PubMed]
- Pavlenko, T.А.; Chesnokova, N.B.; Nodel, M.R.; Kim, A.R.; Ugrumov, M.V.; Вениаминович, У.M. Molecular Mechanisms and Clinical Manifestations of Catecholamine Dysfunction in the Eye in Parkinson’s Disease as a Basis for Developing Early Diagnosis. Acta Naturae 2020, 12, 52–62. [Google Scholar] [CrossRef]
- Zhou, X.; Dai, Y.; Zhai, Z.; Hong, J. WAY-100635 Alleviates Corneal Lesions Through 5-HT1A Receptor-ROS-Autophagy Axis in Dry Eye. Front. Med. 2021, 8, 799949. [Google Scholar] [CrossRef] [PubMed]
- Gureje, O.; Von Korff, M.; Simon, G.E.; Gater, R. Persistent Pain and Well-being. JAMA 1998, 280, 147–151. [Google Scholar] [CrossRef]
- Hermesdorf, M.; Berger, K.; Baune, B.T.; Wellmann, J.; Ruscheweyh, R.; Wersching, H. Pain Sensitivity in Patients with Major Depression: Differential Effect of Pain Sensitivity Measures, Somatic Cofactors, and Disease Characteristics. J. Pain. 2016, 17, 606–616. [Google Scholar] [CrossRef]
- Machalińska, A.; Kuligowska, A.; Ziontkowska-Wrzałek, A.; Stroynowska, B.; Pius-Sadowska, E.; Safranow, K.; Machaliński, J.; Mozolewska-Piotrowska, K.; Machaliński, B. The Severity of Diabetic Retinopathy Corresponds with Corneal Nerve Alterations and Ocular Discomfort of the Patient. Int. J. Mol. Sci. 2024, 25, 6072. [Google Scholar] [CrossRef]
- Tanase, D.M.; Valasciuc, E.; Gosav, E.M.; Floria, M.; Buliga-Finis, O.N.; Ouatu, A.; Cucu, A.I.; Botoc, T.; Costea, C.F. Enhancing Retinal Resilience: The Neuroprotective Promise of BDNF in Diabetic Retinopathy. Life 2025, 15, 263. [Google Scholar] [CrossRef]
- Zloh, M.; Kutilek, P.; Hejda, J.; Fiserova, I.; Kubovciak, J.; Murakami, M.; Stofkova, A. Visual stimulation and brain-derived neurotrophic factor (BDNF) have protective effects in experimental autoimmune uveoretinitis. Life Sci. 2024, 355, 122996. [Google Scholar] [CrossRef]
- Nagino, K.; Sung, J.; Oyama, G.; Hayano, M.; Hattori, N.; Okumura, Y.; Fujio, K.; Akasaki, Y.; Huang, T.; Midorikawa-Inomata, A.; et al. Prevalence and characteristics of dry eye disease in Parkinson’s disease: A systematic review and meta-analysis. Sci. Rep. 2022, 12, 18348. [Google Scholar] [CrossRef] [PubMed]
- Davinelli, S.; Medoro, A.; Siracusano, M.; Savino, R.; Saso, L.; Scapagnini, G.; Mazzone, L. Oxidative stress response and NRF2 signaling pathway in autism spectrum disorder. Redox Biol. 2025, 83, 103661. [Google Scholar] [CrossRef] [PubMed]
- Guerrero-Moreno, A.; Liang, H.; Moreau, N.; Luzu, J.; Rabut, G.; Parsadaniantz, S.M.; Labbé, A.; Baudouin, C.; Goazigo, A.R.-L. Corneal Nerve Abnormalities in Painful Dry Eye Disease Patients. Biomedicines 2021, 9, 1424. [Google Scholar] [CrossRef] [PubMed]
- Bucsan, R.; Coroleucă, R.; Garhöfer, G.; Popa-Cherecheanu, A.; Schmetterer, L.; Iancu, R. Confocal Microscopy of the Cornea in Aqueous-Deficient Dry Eye Disease—A Literature Review. Diagnostics 2024, 14, 1613. [Google Scholar] [CrossRef]
- Chiang, J.C.B.; Tran, V.; Wolffsohn, J.S. The impact of dry eye disease on corneal nerve parameters: A systematic review and meta-analysis. Ophthalmic Physiol. Opt. 2023, 43, 1079–1091. [Google Scholar] [CrossRef]
- Damci, A.; Schruers, K.R.J.; Leue, C.; Faber, C.G.; Hoeijmakers, J.G.J. Anxiety and depression in small fiber neuropathy. J. Peripher. Nerv. Syst. 2022, 27, 291–301. [Google Scholar] [CrossRef]
- Alghafri, R.M.; Gatt, A.; Formosa, C. Depression Symptoms in Patients with Diabetic Peripheral Neuropathy. Rev. Diabet. Stud. 2020, 16, 35–40. [Google Scholar] [CrossRef]
- Fülöp, B.; Borbély, É.; Helyes, Z. How does chronic psychosocial distress induce pain? Focus on neuroinflammation and neuroplasticity changes. Brain Behav. Immun.-Health 2025, 47, 100964. [Google Scholar] [CrossRef]
- Sheng, J.; Liu, S.; Wang, Y.; Cui, R.; Zhang, X. The Link between Depression and Chronic Pain: Neural Mechanisms in the Brain. Neural Plast. 2017, 2017, 9724371. [Google Scholar] [CrossRef]
- Barcelon, E.E.; Cho, W.-H.; Jun, S.B.; Lee, S.J. Brain Microglial Activation in Chronic Pain-Associated Affective Disorder. Front. Neurosci. 2019, 13, 213. [Google Scholar] [CrossRef] [PubMed]
- Tsai, C.-Y.; Jiesisibieke, Z.L.; Tung, T.-H. Association between dry eye disease and depression: An umbrella review. Front. Public Health 2022, 10, 910608. [Google Scholar] [CrossRef]
- Malon, J.T.; Cao, L. Calcitonin gene-related peptide contributes to peripheral nerve injury-induced mechanical hypersensitivity through CCL5 and p38 pathways. J. Neuroimmunol. 2016, 297, 68–75. [Google Scholar] [CrossRef] [PubMed]
- Iyengar, S.; Ossipov, M.H.; Johnson, K.W. The role of calcitonin gene–related peptide in peripheral and central pain mechanisms including migraine. PAIN® 2017, 158, 543–559. [Google Scholar] [CrossRef]
- Russell, F.A.; King, R.; Smillie, S.-J.; Kodji, X.; Brain, S.D. Calcitonin Gene-Related Peptide: Physiology and Pathophysiology. Physiol. Rev. 2014, 94, 1099–1142. [Google Scholar] [CrossRef]
- Hashikawa-Hobara, N.; Ogawa, T.; Sakamoto, Y.; Matsuo, Y.; Ogawa, M.; Zamami, Y.; Hashikawa, N. Calcitonin gene-related peptide pre-administration acts as a novel antidepressant in stressed mice. Sci. Rep. 2015, 5, srep12559. [Google Scholar] [CrossRef]
- Lentsch, S.d.V.; van der Arend, B.W.H.; de Boer, I.; van Zwet, E.W.; MaassenVanDenBrink, A.; Terwindt, G.M. Depression and treatment with anti-calcitonin gene related peptide (CGRP) (ligand or receptor) antibodies for migraine. Eur. J. Neurol. 2024, 31, e16106. [Google Scholar] [CrossRef]
- Ma, L.; Yang, L.; Wang, X.; Zhao, L.; Bai, X.; Qi, X.; Chen, Q.; Li, Y.; Zhou, Q. CGRP Released by Corneal Sensory Nerve Maintains Tear Secretion of the Lacrimal Gland. Investig. Ophthalmol. Vis. Sci. 2024, 65, 30. [Google Scholar] [CrossRef]
- Li, F.; Yang, W.; Jiang, H.; Guo, C.; Huang, A.J.W.; Hu, H.; Liu, Q. TRPV1 activity and substance P release are required for corneal cold nociception. Nat. Commun. 2019, 10, 5678. [Google Scholar] [CrossRef]
- Fakih, D.; Guerrero-Moreno, A.; Baudouin, C.; Goazigo, A.R.-L.; Parsadaniantz, S.M. Capsazepine decreases corneal pain syndrome in severe dry eye disease. J. Neuroinflammation 2021, 18, 111. [Google Scholar] [CrossRef]
- Meza, R.C.; Ancatén-González, C.; Chiu, C.Q.; Chávez, A.E. Transient Receptor Potential Vanilloid 1 Function at Central Synapses in Health and Disease. Front. Cell. Neurosci. 2022, 16, 864828. [Google Scholar] [CrossRef]
- Chahl, L.A. TRPV1 Channels in the Central Nervous System as Drug Targets. Pharmaceuticals 2024, 17, 756. [Google Scholar] [CrossRef] [PubMed]
- Lin, Y.-W.; Cheng, S.-W.; Liu, W.-C.; Zailani, H.; Wu, S.-K.; Hung, M.-C.; Su, K.-P. Chemogenetic targeting TRPV1 in obesity-induced depression: Unveiling therapeutic potential of eicosapentaenoic acid and acupuncture. Brain Behav. Immun. 2024, 123, 771–783. [Google Scholar] [CrossRef] [PubMed]
- Liu, M.-G.; Zhuo, M. No requirement of TRPV1 in long-term potentiation or long-term depression in the anterior cingulate cortex. Mol. Brain 2014, 7, 27. [Google Scholar] [CrossRef] [PubMed]
- Won, E.; Kang, J.; Choi, S.; Kim, A.; Han, K.-M.; Yoon, H.-K.; Cho, S.-H.; Tae, W.-S.; Lee, M.-S.; Joe, S.-H.; et al. The association between substance P and white matter integrity in medication-naive patients with major depressive disorder. Sci. Rep. 2017, 7, 9707. [Google Scholar] [CrossRef]
- Humes, C.; Sic, A.; Knezevic, N.N. Substance P’s Impact on Chronic Pain and Psychiatric Conditions—A Narrative Review. Int. J. Mol. Sci. 2024, 25, 5905. [Google Scholar] [CrossRef]
- Galor, A.; Levitt, R.C.; Felix, E.R.; Martin, E.R.; Sarantopoulos, C.D. Neuropathic ocular pain: An important yet underevaluated feature of dry eye. Eye 2014, 29, 301–312. [Google Scholar] [CrossRef]
- Puja, G.; Sonkodi, B.; Bardoni, R. Mechanisms of Peripheral and Central Pain Sensitization: Focus on Ocular Pain. Front. Pharmacol. 2021, 12, 764396. [Google Scholar] [CrossRef]
- Yan, H.; Shan, X.; Wei, S.; Liu, F.; Li, W.; Lei, Y.; Guo, W.; Luo, S. Abnormal Spontaneous Brain Activities of Limbic-Cortical Circuits in Patients with Dry Eye Disease. Front. Hum. Neurosci. 2020, 14, 574758. [Google Scholar] [CrossRef]
- Launay, P.-S.; Reboussin, E.; Liang, H.; Kessal, K.; Godefroy, D.; Rostene, W.; Sahel, J.-A.; Baudouin, C.; Parsadaniantz, S.M.; Le Goazigo, A.R. Ocular inflammation induces trigeminal pain, peripheral and central neuroinflammatory mechanisms. Neurobiol. Dis. 2016, 88, 16–28. [Google Scholar] [CrossRef]
- Belmonte, C.; Nichols, J.J.; Cox, S.M.; Brock, J.A.; Begley, C.G.; Bereiter, D.A.; Dartt, D.A.; Galor, A.; Hamrah, P.; Ivanusic, J.J.; et al. TFOS DEWS II pain and sensation report. Ocul. Surf. 2017, 15, 404–437. [Google Scholar] [CrossRef]
- Barabino, S.; Aragona, P.; Baudouin, C.; Boboridis, K.; Salgado-Borges, J.; Benitez-Del-Castillo, J.M.; Messmer, E.M.; Stanila, A.; Horwath-Winter, J.; Wylegala, E.; et al. Parainflammation in the Ocular System: Considerations on the Underlying Mechanisms and Treatment of Dry Eye Disease. Ophthalmol. Ther. 2025, 14, 883–895. [Google Scholar] [CrossRef]
- Ganesalingam, K.; Ismail, S.; Sherwin, T.; Craig, J.P. Molecular evidence for the role of inflammation in dry eye disease. Clin. Exp. Optom. 2019, 102, 446–454. [Google Scholar] [CrossRef]
- Asiedu, K. Role of ocular surface neurobiology in neuronal-mediated inflammation in dry eye disease. Neuropeptides 2022, 95, 102266. [Google Scholar] [CrossRef] [PubMed]
- Fakih, D.; Zhao, Z.; Nicolle, P.; Reboussin, E.; Joubert, F.; Luzu, J.; Labbé, A.; Rostène, W.; Baudouin, C.; Parsadaniantz, S.M.; et al. Chronic dry eye induced corneal hypersensitivity, neuroinflammatory responses, and synaptic plasticity in the mouse trigeminal brainstem. J. Neuroinflammation 2019, 16, 268. [Google Scholar] [CrossRef] [PubMed]
- Ismayilov, A.S.; Celikel, G. Effects of tricyclic antidepressants, selective serotonin reuptake inhibitors, and selective serotonin-norepinephrine reuptake inhibitors on the ocular surface. Arq. Bras. De Oftalmol. 2023, 86, e20230068. [Google Scholar] [CrossRef]
- Constable, P.A.; Al-Dasooqi, D.; Bruce, R.; Prem-Senthil, M. A Review of Ocular Complications Associated with Medications Used for Anxiety, Depression, and Stress. Clin. Optom. 2022, ume 14, 13–25. [Google Scholar] [CrossRef] [PubMed]
- Rakofsky, J.J.; Rakofsky, S.I.; Dunlop, B.W. Dry Those Crying Eyes: The Role of Depression and Antidepressants in Dry Eye Disease. J. Clin. Psychopharmacol. 2021, 41, 295–303. [Google Scholar] [CrossRef]
- Benítez-Del-Castillo, J.M.; Burgos-Blasco, B. Prevalence of dry eye disease in Spain: A population-based survey (PrevEOS). Ocul. Surf. 2025, 36, 126–133. [Google Scholar] [CrossRef]
- Koçer, E.; Koçer, A.; Özsütçü, M.; Dursun, A.E.; Kirpinar, I. Dry Eye Related to Commonly Used New Antidepressants. J. Clin. Psychopharmacol. 2015, 35, 411–413. [Google Scholar] [CrossRef]
- Dankis, M.; Aydogdu, O.; Tobin, G.; Winder, M. Acute Inhibitory Effects of Antidepressants on Lacrimal Gland Secretion in the Anesthetized Rat. Investig. Opthalmology Vis. Sci. 2021, 62, 8. [Google Scholar] [CrossRef] [PubMed]
- Acan, D.; Kurtgoz, P. Influence of selective serotonin reuptake inhibitors on ocular surface. Clin. Exp. Optom. 2017, 100, 83–86. [Google Scholar] [CrossRef]
- Chen, Q.; Wei, Z.; Wang, L.; Xu, X.; Wei, Z.; Zheng, P.; Cao, K.; Zhang, Z.; Chen, K.; Liang, Q. Dry Eye Disease in Patients with Schizophrenia: A Case-Control Study. Front. Med. 2022, 9, 831337. [Google Scholar] [CrossRef]
- Balogun, M.M.; Coker, A.O. Dry Eyes Illness in Psychiatric Patients Placed on Antipsychotic Medications. J. Behav. Health Psychol. 2023, 12, 1–6. [Google Scholar] [CrossRef]
- Mu, C.; Chen, L. Characteristics of eye disorders induced by atypical antipsychotics: A real-world study from 2016 to 2022 based on Food and Drug Administration Adverse Event Reporting System. Front. Psychiatry 2024, 15, 1322939. [Google Scholar] [CrossRef]
- Dixitha, V.; Abhilash, B.; Bhavya, H.U.; Mahesh, B.; Sudhakar, N.A. Quantitative Analysis of Tear Film in Patients on Atypical Antipsychotics. Ocul. Immunol. Inflamm. 2023, 32, 566–571. [Google Scholar] [CrossRef] [PubMed]
- Musayeva, A.; Jiang, S.; Ruan, Y.; Zadeh, J.K.; Chronopoulos, P.; Pfeiffer, N.; Müller, W.E.; Ackermann, M.; Xia, N.; Li, H.; et al. Aged Mice Devoid of the M3 Muscarinic Acetylcholine Receptor Develop Mild Dry Eye Disease. Int. J. Mol. Sci. 2021, 22, 6133. [Google Scholar] [CrossRef]
- Haidary, H.A.; Padhy, R.K. Clozapine. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2025. Available online: https://www.ncbi.nlm.nih.gov/books/NBK535399/ (accessed on 19 September 2025).
- Ceylan, E.; Ozer, M.D.; Yilmaz, Y.C.; Kartal, B.; Ekinci, D.Y.; Çinici, E.; Ceylan, N.A. The ocular surface side effects of an anti-psychotic drug, clozapine. Cutan. Ocul. Toxicol. 2015, 35, 62–66. [Google Scholar] [CrossRef]
- Dibajnia, P.; Mohammadinia, M.; Moghadasin, M.; Amiri, M.A. Tear Film Break-up Time in Bipolar Disorder. Iran. J. Psychiatry 2012, 7, 191–193. [Google Scholar]
- Fraunfelder, F.W.; Jefferson, J.W. The Effects of Lithium on the Human Visual System. J. Toxicol. Cutan. Ocul. Toxicol. 1992, 11, 97–169. [Google Scholar] [CrossRef]
- Park, J.S.; Sharma, R.A.; Sharma, V. Ophthalmic adverse effects of lithium. Int. Clin. Psychopharmacol. 2020, 35, 69–73. [Google Scholar] [CrossRef]
- Chuang, J.-Y.; Tzeng, N.-S. Sjögren’s syndrome precipiated by lamotrigine treatment. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2009, 33, 1075–1076. [Google Scholar] [CrossRef]
- Alev-Tuzuner, B.; Oktay, S.; Cergel, E.; Elik, G.; Magaji, U.F.; Sacan, O.; Yanardag, R.; Yarat, A. Moringa oleifera hydroalcoholic leaf extracts mitigate valproate-induced oxidative status in the extraorbital lacrimal gland in a rat model. Exp. Eye Res. 2024, 248, 110104. [Google Scholar] [CrossRef]
- Guo, M.; Diaz, G.M.; Yu, Y.; Patel, C.A.; Farrar, J.T.; Asbell, P.A.; Ying, G.-S. Association between systemic medication use and severity of dry eye signs and symptoms in the DRy eye assessment and management (DREAM) study. Ocul. Surf. 2024, 32, 112–119. [Google Scholar] [CrossRef]
- Wen, W.; Wu, Y.; Chen, Y.; Gong, L.; Li, M.; Chen, X.; Yan, M.; Xiao, Z.; Sun, X. Dry Eye Disease in Patients with Depressive and Anxiety Disorders in Shanghai. Cornea 2012, 31, 686–692. [Google Scholar] [CrossRef] [PubMed]
- Checa-Caratachea, X.A.; Pineda, Á.E.G.-A.; Valera-Mota, M.M.; Bautista-Delgado, A.; Moreno-Altamirano, G.A.; Cruz-Hervert, L.P. Polypharmacy Associated with the Simultaneous Prevalence of Dry Eye due to Aqueous Deficiency and Hyposalivation in Adults over 50 Years Old. Cureus 2024, 16, e76439. [Google Scholar] [CrossRef] [PubMed]
- Fraunfelder, F.T.; Sciubba, J.J.; Mathers, W.D. The Role of Medications in Causing Dry Eye. J. Ophthalmol. 2012, 2012, 285851, Erratum in J. Ophthalmol. 2019, 2019, 2989680. https://doi.org/10.1155/2019/2989680. [Google Scholar] [CrossRef]
- Suárez-Cortés, T.; Herrera, I. Emerging Age-Specific Therapeutic Approaches for Dry Eye Disease. J. Clin. Med. 2025, 14, 4147. [Google Scholar] [CrossRef] [PubMed]
- Chang, V.S.; Rose, T.P.O.; Karp, C.L.; Levitt, R.C.; Sarantopoulos, C.M.; Galor, A.M. Neuropathic-Like Ocular Pain and Nonocular Comorbidities Correlate with Dry Eye Symptoms. Eye Contact Lens 2018, 44, S307–S313. [Google Scholar] [CrossRef]
- Watson, S.L.; Le, D.T.-M. Corneal neuropathic pain: A review to inform clinical practice. Eye 2024, 38, 2350–2358. [Google Scholar] [CrossRef]
- Lee, C.J.; Levitt, R.C.; Felix, E.R.; Sarantopoulos, C.D.; Galor, A. Evidence that dry eye is a comorbid pain condition in a U.S. veteran population. PAIN Rep. 2017, 2, e629. [Google Scholar] [CrossRef]
- Dieckmann, G.; Goyal, S.; Hamrah, P. Neuropathic Corneal Pain. Ophthalmology 2017, 124, S34–S47. [Google Scholar] [CrossRef]
- Kumar, N.R.; Praveen, M.; Narasimhan, R.; Khamar, P.; D’sOuza, S.; Sinha-Roy, A.; Sethu, S.; Shetty, R.; Ghosh, A. Tear biomarkers in dry eye disease: Progress in the last decade. Indian J. Ophthalmol. 2023, 71, 1190–1202. [Google Scholar] [CrossRef] [PubMed]
- Shih, K.; Fong, P.; Lam, P.; Chan, T.Y.; Jhanji, V.; Tong, L. Role of tear film biomarkers in the diagnosis and management of dry eye disease. Taiwan J. Ophthalmol. 2019, 9, 150–159. [Google Scholar] [CrossRef] [PubMed]
- Kheirkhah, A.; Di Zazzo, A.; Satitpitakul, V.; Fernandez, M.; Magilavy, D.; Dana, R. A Pilot Randomized Trial on Safety and Efficacy of a Novel Topical Combined Inhibitor of Janus Kinase 1/3 and Spleen Tyrosine Kinase for GVHD-Associated Ocular Surface Disease. Cornea 2017, 36, 799–804. [Google Scholar] [CrossRef]
- Huang, J.-F.; Yafawi, R.; Zhang, M.; McDowell, M.; Rittenhouse, K.D.; Sace, F.; Liew, S.H.; Cooper, S.R.; Pickering, E.H. Immunomodulatory Effect of the Topical Ophthalmic Janus Kinase Inhibitor Tofacitinib (CP-690,550) in Patients with Dry Eye Disease. Ophthalmology 2012, 119, e43–e50. [Google Scholar] [CrossRef]
- Sacchetti, M.; Lambiase, A.; Schmidl, D.; Schmetterer, L.; Ferrari, M.; Mantelli, F.; Allegretti, M.; Garhoefer, G. Effect of recombinant human nerve growth factor eye drops in patients with dry eye: A phase IIa, open label, multiple-dose study. Br. J. Ophthalmol. 2020, 104, 127–135. [Google Scholar] [CrossRef] [PubMed]
- Baratta, R.O.; Schlumpf, E.; Del Buono, B.J.; DeLorey, S.; Ousler, G.; Calkins, D.J. A Phase 2 Trial to Test Safety and Efficacy of ST-100, a Unique Collagen Mimetic Peptide Ophthalmic Solution for Dry Eye Disease. Ophthalmol. Sci. 2023, 4, 100451. [Google Scholar] [CrossRef]
- Gou, Q.; Song, Z.; Gong, Y.; Li, J. TRPV1 in Dry Eye Disease. Front. Biosci. 2024, 29, 175. [Google Scholar] [CrossRef]
- Duan, X.; Ju, M.; Liu, X.; Hu, J. Efficacy and safety of transient receptor potential channel modulators for dry eye: A systematic review and meta-analysis. Contact Lens Anterior Eye 2025, 48, 102347. [Google Scholar] [CrossRef]
- Moon, J.; Jiang, E.; Liu, J.; Jin, H.; Yoon, H.S.; Choi, H.-I.; Choi, J.S.; Qi, H.; Yoon, H.-J.; Yoon, K.C. Therapeutic Potential of Combined 5% Lifitegrast and Tocopherol Eye Drops in Managing Inflammation and Oxidative Stress in Murine Dry Eye. Pharmaceuticals 2025, 18, 38. [Google Scholar] [CrossRef]
- Sano, K.; Kawashima, M.; Takechi, S.; Mimura, M.; Tsubota, K. Exercise program improved subjective dry eye symptoms for office workers. Clin. Ophthalmol. 2018, 12, 307–311. [Google Scholar] [CrossRef]
- Quiroz-Mercado, H.; Hernandez-Quintela, E.; Chiu, K.H.; Henry, E.; Nau, J.A. A phase II randomized trial to evaluate the long-term (12-week) efficacy and safety of OC-01 (varenicline solution) nasal spray for dry eye disease: The MYSTIC study. Ocul. Surf. 2022, 24, 15–21. [Google Scholar] [CrossRef]
- Cammalleri, M.; Amato, R.; Olivieri, M.; Pezzino, S.; Bagnoli, P.; Monte, M.D.; Rusciano, D. Effects of Topical Gabapentin on Ocular Pain and Tear Secretion. Front. Pharmacol. 2021, 12, 671238. [Google Scholar] [CrossRef]
- Diel, R.J.; Kroeger, Z.A.; Levitt, R.C.; Sarantopoulos, C.; Sered, H.; Martinez-Barrizonte, J.; Galor, A. Botulinum Toxin A for the Treatment of Photophobia and Dry Eye. Ophthalmology 2018, 125, 139–140. [Google Scholar] [CrossRef] [PubMed]
- Katipoğlu, Z.; Abay, R.N. The relationship between dry eye disease and anticholinergic burden. Eye 2023, 37, 2921–2925. [Google Scholar] [CrossRef] [PubMed]
- Kumar, V.; Prakash, U.; Mohammad, Y. Psycho Ophthalmology: The Interface Between Psychiatry and Ophthalmology. Int. J. Med. Biomed. Stud. 2024, 8, 87–93. [Google Scholar] [CrossRef]
- Chan, C.C.; Prokopich, C.L. Lifitegrast Ophthalmic Solution 5.0% for Treatment of Dry Eye Disease: Overview of Clinical Trial Program. J. Pharm. Pharm. Sci. 2019, 22, 49–56. [Google Scholar] [CrossRef]
- Kroenke, K.; Spitzer, R.L.; Williams, J.B. The PHQ-9: Validity of a brief depression severity measure. J. Gen. Intern. Med. 2001, 16, 606–613. [Google Scholar] [CrossRef]
- Spitzer, R.L.; Kroenke, K.; Williams, J.B.W.; Löwe, B. A Brief Measure for Assessing Generalized Anxiety Disorder: The GAD-7: The GAD-7. Arch. Intern. Med. 2006, 166, 1092–1097. [Google Scholar] [CrossRef] [PubMed]
- Chang, K.-J.; Wu, H.-Y.; Chiang, P.-H.; Hsu, Y.-T.; Weng, P.-Y.; Yu, T.-H.; Chen, Y.-H.; Dai, H.-J.; Tsai, H.-Y.; Chang, Y.-J.; et al. Decoding and reconstructing disease relations between dry eye and depression: A multimodal investigation comprising meta-analysis, genetic pathways and Mendelian randomization. J. Adv. Res. 2024, 69, 197–213. [Google Scholar] [CrossRef]
- Britten-Jones, A.C.; Wang, M.T.M.; Samuels, I.; Jennings, C.; Stapleton, F.; Craig, J.P. Epidemiology and Risk Factors of Dry Eye Disease: Considerations for Clinical Management. Medicina 2024, 60, 1458. [Google Scholar] [CrossRef]
- Pili, K.; Kaštelan, S.; Karabatić, M.; Kasun, B.; Čulig, B. Dry eye in contact lens wearers as a growing public health problem. Psychiatr Danub. 2014, 26 (Suppl. S3), 528–532. [Google Scholar]
- Kaštelan, S.; Pjevač, N.; Braš, M.; Đorđević, V.; Keleminić, N.P.; Mezzich, J.E. Person-centered care in ophthalmology: Current knowledge and perspectives. Croat. Med. J. 2024, 65, 156–160. [Google Scholar] [CrossRef]
- Kaštelan, S.; Braš, M.; Pjevač, N.; Bakija, I.; Tomić, Z.; Keleminić, N.P.; Antunica, A.G. Tear Biomarkers and Alzheimer’s Disease. Int. J. Mol. Sci. 2023, 24, 13429. [Google Scholar] [CrossRef]
- Ong, E.S.; Alghamdi, Y.A.; Levitt, R.C.; McClellan, A.L.; Lewis, G.; Sarantopoulos, C.D.; Felix, E.R.; Galor, A. Longitudinal Examination of Frequency of and Risk Factors for Severe Dry Eye Symptoms in US Veterans. JAMA Ophthalmol. 2017, 135, 116–123. [Google Scholar] [CrossRef] [PubMed]
- Donthineni, P.R.; Shanbhag, S.S.; Basu, S. An Evidence-Based Strategic Approach to Prevention and Treatment of Dry Eye Disease, a Modern Global Epidemic. Healthcare 2021, 9, 89. [Google Scholar] [CrossRef] [PubMed]
- Labetoulle, M.; Baudouin, C.; del Castillo, J.M.B.; Rolando, M.; Rescigno, M.; Messmer, E.M.; Aragona, P. How gut microbiota may impact ocular surface homeostasis and related disorders. Prog. Retin. Eye Res. 2024, 100, 101250. [Google Scholar] [CrossRef]
- Kammoun, S.; Rekik, M.; Dlensi, A.; Aloulou, S.; Smaoui, W.; Sellami, S.; Trigui, K.; Gargouri, R.; Chaari, I.; Sellami, H.; et al. The gut-eye axis: The retinal/ocular degenerative diseases and the emergent therapeutic strategies. Front. Cell. Neurosci. 2024, 18, 1468187. [Google Scholar] [CrossRef] [PubMed]
- Campagnoli, L.I.M.; Varesi, A.; Barbieri, A.; Marchesi, N.; Pascale, A. Targeting the Gut–Eye Axis: An Emerging Strategy to Face Ocular Diseases. Int. J. Mol. Sci. 2023, 24, 13338. [Google Scholar] [CrossRef]
- Jin, Q.; Wang, S.; Yao, Y.; Jiang, Q.; Li, K. The gut–eye axis: From brain neurodegenerative diseases to age-related macular degeneration. Neural Regen. Res. 2024, 20, 2741–2757. [Google Scholar] [CrossRef] [PubMed]
- Oudin, A.; Maatoug, R.; Bourla, A.; Ferreri, F.; Bonnot, O.; Millet, B.; Schoeller, F.; Mouchabac, S.; Adrien, V. Digital Phenotyping: Data-Driven Psychiatry to Redefine Mental Health. J. Med. Internet Res. 2023, 25, e44502. [Google Scholar] [CrossRef] [PubMed]
- Parvin, N.; Joo, S.W.; Jung, J.H.; Mandal, T.K. Multimodal AI in Biomedicine: Pioneering the Future of Biomaterials, Diagnostics, and Personalized Healthcare. Nanomaterials 2025, 15, 895. [Google Scholar] [CrossRef]
- Liu, Y.; Wang, B. Advanced applications in chronic disease monitoring using IoT mobile sensing device data, machine learning algorithms and frame theory: A systematic review. Front. Public Health 2025, 13, 1510456. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).