1. Introduction
Ocular diseases, encompassing retinal and corneal disorders alongside ocular surface conditions, such as eyelid pathologies, significantly contribute to the global burden of visual impairment [
1,
2,
3,
4]. These conditions pose significant challenges in prevention and treatment. For instance, diabetic retinopathy (DR) affected over 100 million people globally in 2020, with projections surpassing 160 million by 2045 [
5]. Similarly, glaucoma, a leading cause of irreversible blindness, is estimated to impact 111.8 million people aged 40–80 worldwide by 2040 [
6]. Dry eye syndrome impacts a significant portion of the population, with prevalence rates varying widely between 5% and 50%, underscoring the growing burden of age-related ocular conditions, which are expected to increase with the aging population, projected to double to 2.1 billion by 2050 [
7,
8].
Moreover, lifestyle factors, such as unhealthy eating habits, smoking, and the frequent use of digital devices, have intensified these challenges. Worryingly, projections from the Global Burden of Disease Study suggest that by 2050, approximately 474 million people may experience moderate to severe visual impairments, with 61 million potentially losing their sight entirely [
9].
In recent years, considering these obstacles, there has been growing interest in curcumin as a possible therapeutic agent in managing ocular diseases. Curcumin (C
21H
20O
6), a lipophilic polyphenol derived from the dried rhizome of
Curcuma longa L. and related species, has gained significant attention due to its extensive pharmacological properties, including anti-inflammatory, antioxidant, antimicrobial, and antitumor activities [
10,
11,
12]. Alongside its primary forms—demethoxycurcumin and bis-demethoxycurcumin—turmeric contains over 50 additional curcuminoids, including bisabocurcumin, curcumalongin, cyclocurcumin, and terpecurcumin, as well as volatile oils and resins. These compounds broaden turmeric’s pharmacological profile, offering synergistic effects that enhance its therapeutic versatility and reinforce its global use both as a culinary spice and as a source of health benefits [
13].
Curcumin’s molecular structure, comprising two o-methoxy phenolic aromatic rings linked by a seven-carbon α, β-unsaturated β-diketone chain, underpins its pleiotropic effects. Its properties—anti-inflammatory, antioxidant, antibacterial, anti-angiogenic, and anti-apoptotic—show promise in ophthalmology [
14,
15]. Research indicates its potential for treating corneal and retinal neovascularization, inhibiting lens epithelial cell proliferation, and modulating retinal pigment epithelium-related pathways, making it a valuable candidate for managing inflammatory and degenerative ocular diseases [
16,
17]. Topical formulations, such as hydrogels, creams, and nanocarrier systems, have been developed to enhance their physicochemical properties, including solubility, permeability, and stability. These innovations protect curcumin from degradation and enable sustained release, proving effective in treating dermatological conditions, such as psoriasis, acne, and atopic dermatitis, due to its anti-inflammatory, wound-healing, and antioxidant properties. This success in dermatology has spurred interest in its application for ocular treatments due to similar physicochemical barriers [
18,
19].
Recognized by the FDA as ‘Generally Recognized as Safe’ (GRAS) for human consumption, curcumin demonstrates significant therapeutic potential [
20]. Clinical trials have confirmed their excellent safety, tolerability, and efficacy, even at high oral doses ranging from 4 to 8 g per day and doses up to 12 g per day for curcuminoid formulations containing 95% curcumin, bisdemethoxycurcumin, and demethoxycurcumin [
20,
21]. However, its clinical application is limited by critical pharmacokinetic challenges, including poor aqueous solubility, light sensitivity, low bioavailability, limited absorption, and rapid systemic metabolism and elimination. These factors complicate consistent therapeutic outcomes and pharmacological interpretations, particularly given curcumin’s classification as a PAIN (pan-assay interference compound) and an IMP (invalid metabolic panacea), which highlights its complex bioactivity. Its degradation of products and fluorescence further complicate pharmacological evaluations [
22,
23]. These limitations not only hinder consistent therapeutic outcomes but also complicate the identification of the actual bioactive species responsible for its effects.
In addition, the metabolism of curcumin and its interaction with the intestinal microbiota play a crucial role in determining its bioavailability and therapeutic efficacy. After oral administration, curcumin exhibits poor solubility and limited gastrointestinal absorption. The absorbed fraction undergoes rapid metabolism in the liver and intestine via reduction (yielding dihydrocurcumin and tetrahydrocurcumin) and conjugation (forming glucuronides and sulfates), leading to its swift elimination. The intestinal microbiota further converts curcumin into more stable and, in some cases, bioactive metabolites. Certain bacterial genera, such as
Bifidobacterium and
Lactobacillus, promote its reduction and demethylation, potentially enhancing biological activity. Conversely, curcumin modulates the gut microbiota, fostering beneficial species while inhibiting pathogens. These interactions have significant implications for curcumin’s therapeutic applications in ocular diseases, especially given the distinct metabolic pathways associated with oral and topical administration [
24].
Additionally, the wide range of commercially available formulations—ranging from turmeric powder to curcuminoid-enriched products and purified curcumin—adds complexity, raising concerns about reproducibility and efficacy in clinical trials [
2,
25,
26,
27,
28]. Despite these challenges, curcumin’s diverse therapeutic potential emphasizes the need for innovative delivery systems.
Nanocarrier technologies, particularly vesicular systems, such as liposomes and proniosomes, have addressed some of these challenges by improving curcumin’s bioavailability, solubility, and stability [
29]. These systems encapsulate curcumin within surfactant vesicles, protecting it from enzymatic degradation and extending its therapeutic presence on ocular surfaces. Moreover, the sustained drug delivery provided by these systems reduces systemic side effects while targeting disease-specific sites [
30].
In ocular inflammation, curcumin has demonstrated efficacy in reducing complications, such as corneal opacity, cataract formation, and retinal detachment [
3]. It also holds promise as a prophylactic agent in proliferative vitreoretinopathy (PVR), with studies reporting reduced rates of retinal detachment following surgery [
31]. Furthermore, curcumin demonstrates therapeutic benefits in DR by modulating hyperglycemia-induced endothelial dysfunction [
32].
Topical drug delivery systems, such as eye drops, i.e., aqueous solutions and suspensions, and oil-based formulations, remain widely used for ocular treatment. These formulations are intended for direct application to the ocular surface, typically in the form of drops. However, they often face significant limitations, including excessive tear production, rapid drainage, and systemic absorption, leading to inefficient drug distribution and the loss of over 95% of the administered dose [
33].
This review explores curcumin’s therapeutic potential in ophthalmology, focusing on its molecular mechanisms, challenges in clinical application, and advanced strategies for optimized delivery. By addressing these barriers, curcumin could transform ocular disease management, highlighting the need for robust randomized trials to confirm its safety and efficacy.
2. Therapeutic Applications of Curcumin in Ophthalmology
Delivering medications to the eye effectively remains a significant challenge due to its distinct pharmacokinetic and pharmacodynamic environment. The eye’s natural defense mechanisms—such as tear production, blinking, and the intricate clearance processes on the ocular surface—serve to protect it but simultaneously hinder drug retention and absorption. These barriers, coupled with the anatomical complexity of anterior and posterior segments, result in low bioavailability for many conventional therapies [
34,
35]. Frequent application of traditional eye drops is often necessary to achieve therapeutic outcomes, but this practice can inadvertently lead to systemic side effects through absorption via the nasolacrimal pathway [
30,
36].
Recent studies suggest that curcumin can be applied to various ophthalmic conditions, offering significant therapeutic potential for a wide range of ocular diseases and addressing many limitations of conventional approaches (
Figure 1). Curcumin’s antimicrobial and immunomodulatory properties make it particularly effective in targeting the complex interplay of infection, inflammation, and oxidative stress underlying many ocular pathologies. It directly disrupts bacterial cell walls and inhibits enzymatic processes critical for bacterial survival while also downregulating pro-inflammatory cytokines and mitigating oxidative stress. These dual antibacterial and anti-inflammatory actions position curcumin as a versatile therapeutic agent, particularly when integrated into advanced drug delivery systems [
37,
38].
As shown in
Table 1, the development of advanced drug delivery systems, including in situ gels, nanostructured lipid carriers, and hydrogels, has emerged as a promising strategy to enhance the solubility, stability, and ocular bioavailability of curcumin.
An in vitro study, supported by ex vivo assays using rabbit corneas, showed that polyethylene glycol-distearoylphosphatidylethanolamine (PEG-DSPE)/Solutol HS 15 mixed micelle-based in situ gels improve corneal penetration, ocular retention, and stability. This system also supports sustained drug release, reduces dosing frequency, and avoids ocular irritation, offering a promising alternative to conventional eye drops [
39].
Thiolated chitosan-coated nanostructured lipid carriers, characterized in vitro and further evaluated in vivo using animal models, enhance corneal contact through covalent bonding with mucus glycoproteins. This interaction ensures sustained release over 72 h without irritation, improving ocular distribution and therapeutic efficacy [
40].
Hydroxypropyl methylcellulose methacrylate hydrogels, tested in vivo, demonstrated strong bioadhesion and controlled curcumin release, contributing to the reduction of oxidative damage in trabecular meshwork cells. This effect helps mitigate the inflammatory and apoptotic processes that are key in glaucoma progression, confirming the safety and potential of this formulation for controlled ocular drug delivery [
41].
These studies highlight the potential of these technologies to improve ocular retention, reduce dosing frequency, and minimize systemic side effects. Such advancements enable sustained drug release, improve therapeutic outcomes, and alleviate the burden of frequent applications associated with traditional formulations in ocular diseases.
To better understand the impact of delivery methods on curcumin’s bioavailability and therapeutic potential,
Table 1 provides a comparison of different routes of administration, including topical ocular systems, oral administration (including trial clinical phase I), and parenteral routes.
Ocular drug delivery systems offer a distinct advantage by bypassing the first-pass metabolism typical of oral administration and directly targeting the eye.
These formulations improve curcumin’s retention, permeability, and controlled release, leading to enhanced therapeutic efficacy when compared to oral or intravenous routes. Additionally, ocular systems minimize systemic exposure, reducing the likelihood of side effects and providing a more focused and controlled therapeutic approach.
Table 1.
Curcumin’s ocular therapeutic potential.
Table 1.
Curcumin’s ocular therapeutic potential.
Route of Administration | Formulation/Technology | Key Findings | Bioavailability/Retention | Refs. |
---|
Topical | Curcumin-loaded in situ gel (eye drops) | Enhanced solubility and corneal penetration | Increased bioavailability (not quantified) | [39] |
Topical | Chitosan–curcumin–nano-lipid carriers (eye drops) | Improved permeability and ocular retention | 6.4–18.8-fold increase | [40] |
Topical | Hydroxypropyl methylcellulose hydrogels | Sustained release, reduced oxidative damage | Sustained release over 6–8 h | [41] |
Oral | Conventional oral curcumin (3600–8000 mg/day) | Low bioavailability, rapid metabolism | Poor systemic bioavailability | [42,43,44] |
Oral | Theracurmin(enhanced oral curcumin) | Increased systemic exposure (18.4–20.5 times higher) | Higher plasma concentration | [45] |
Intravenous | Intravenous administration in rats (10 mg/kg) | Rapid plasma peak concentration (0.36 µg/mL) | High but short-lived bioavailability | [28] |
Intramuscular | Intramuscular injection in rats(50 mg/kg) | Sustained plasma concentration | Bioavailability ~30% | [46] |
Intraperitoneal | Intraperitoneal administration in rats (20 mg/kg) | Higher bioavailability | Bioavailability 35.07% | [47] |
Curcumin serves as a central modulator in multiple molecular systems involved in ocular health. It promotes a dynamic equilibrium between cellular processes that sustain ocular tissue integrity (
Figure 2). This bioactive compound uniquely interacts with key signaling pathways, adjusting their intensity and function to reach an ideal homeostatic state.
For instance, its anti-inflammatory action arises from a strategic blockade of pro-inflammatory signals, such as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and NLR family pyrin domain containing 3 (NLRP3), leading to a reduction in the excessive production of cytokines like tumor necrosis factor-alpha (TNF-α), interleukin 1 beta (IL-1β), and interleukin 6 (IL-6). This effect can be likened to the fine-tuning of an inflammatory thermostat, ensuring that inflammation required for tissue repair is preserved while preventing the collateral damage associated with chronic inflammation [
38,
48,
49,
50,
51].
In the antioxidant domain, curcumin stands out by activating nuclear factor erythroid 2-related factor 2 (NRF2), a regulatory protein that coordinates the expression of antioxidant enzymes, including superoxide dismutase (SOD), catalase, and glutathione peroxidase (GPX). This mechanism not only neutralizes reactive oxygen species (ROS) but also preserves mitochondrial metabolic functions, protecting ocular cells from oxidative stress. Thus, curcumin acts as a metabolic sentinel, preventing cumulative damage over time [
52,
53,
54,
55,
56,
57,
58].
Furthermore, its ability to inhibit vascular endothelial growth factor (VEGF) in angiogenic processes is particularly relevant in conditions characterized by pathological neovascularization, such as DR and age-related macular degeneration (AMD). This effect is not merely inhibitory but restorative, realigning angiogenic processes to meet the tissue’s physiological needs. Curcumin’s regulation of the apoptotic balance through modulators like B-cell lymphoma 2 (Bcl-2)/Bcl-2-associated X protein (Bax) ratio further solidifies its role as a cellular protector, preventing uncontrolled cell death while maintaining the selective elimination of damaged cells [
52,
55,
56,
59,
60,
61,
62,
63,
64].
Curcumin also exerts a significant antibacterial effect through its modulation of ROS and its ability to suppress bacterial cell wall synthesis. By neutralizing ROS and interfering with bacterial biofilm formation, curcumin enhances its antibacterial potential. This mechanism reduces bacterial load while maintaining a controlled inflammatory response. Through the modulation of the immune system, curcumin minimizes excessive inflammatory damage, contributing to a more effective defense mechanism against microbial infections [
38,
48].
Finally, curcumin’s immunomodulatory effects ensure a favorable environment for tissue repair and regeneration, particularly in autoimmune and infectious diseases affecting the eye. This comprehensive action, combined with its extracellular matrix stabilization properties and regulation of endoplasmic reticulum stress, positions curcumin as a molecularly orchestrated intervention with therapeutic potential adaptable to a wide range of ocular conditions [
24,
49,
50,
51].
2.1. Retinal Diseases
Retinal diseases, characterized by complex pathological processes, such as inflammation, oxidative stress, and pathological angiogenesis, are major contributors to vision impairment. Leveraging curcumin’s unique ability to target these mechanisms, recent research highlights its potential in mitigating retinal damage and preserving visual function (
Table 2) [
2].
In DR, curcumin alleviates hyperglycemia-induced damage to retinal pigment epithelium (RPE) cells and helps maintain blood–retinal barrier integrity. A recent experimental study in diabetic rats showed that curcumin reduces pro-inflammatory cytokines (TNF-α, IL-1, and IFN-γ) and oxidative stress markers, such as malondialdehyde (MDA), GPX, CAT, and SOD [
48,
65]. These findings align with its broader molecular actions, including modulation of extracellular signal-regulated kinase (ERK) and Akt (protein kinase B, PKB) pathways to protect RPE cells and inhibit retinal neovascularization, inhibition of chronic inflammation via NF-κB suppression, and oxidative stress reduction [
48,
65,
66].
In addition to DR, inflammation is a key driver in retinal diseases such as age-related macular degeneration (AMD) and best vitelliform macular dystrophy (BVMD). Curcumin suppresses pathways like NF-κB, reducing pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6) and mitigating chronic inflammation [
1,
16]. This anti-inflammatory effect, mediated through NF-κB modulation, is a common mechanism also observed in glaucoma, where curcumin regulates the NF-κB pathway to reduce inflammation and oxidative stress, helping protect retinal ganglion cells (RGCs) and improve the optic nerve integrity [
53,
67].
Curcumin’s antioxidant effects are critical in combating oxidative stress, a major contributor to retinal degeneration. By enhancing endogenous antioxidant defenses, such as SOD and catalase, curcumin protects retinal cells from ROS, which are implicated in diseases like retinitis pigmentosa (RP), AMD, and BVMD. It also reduces oxidative stress and light-induced damage, especially relevant in BVMD [
52,
53,
54,
55]. In all these conditions, curcumin also acts in glaucoma, where its ability to reduce ROS levels protects RGCs and modulates antioxidant pathways, such as the activation of Nrf2, which is critical for protecting cells against ROS-induced damage [
53,
56].
Furthermore, curcumin may regulate calcium homeostasis in RPE cells, enhancing their function and survival, especially in BVMD [
55]. Neuroprotective effects of curcumin also extend to glaucoma, where it reduces RGC death by inhibiting caspase-3 activation and modulating factors such as Bcl-2 and Bax, providing additional protection against neuronal degeneration [
56]. Curcumin’s ability to modulate autophagy offers a dual benefit by not only preventing cellular degeneration but also enhancing the clearance of toxic protein aggregates and damaged organelles in RPE cells. This dual action reinforces its potential as a therapeutic agent for retinal diseases, highlighting its capacity to target multiple pathological mechanisms simultaneously, thereby preserving retinal structure and function [
68].
Table 2.
Curcumin: molecular mechanisms and activity potential in retinal diseases.
Table 2.
Curcumin: molecular mechanisms and activity potential in retinal diseases.
Relevant Diseases | Activity Potential | Key Actions and Targets | Refs. |
---|
DR | BRB integrity and mitochondrial protection | Modulates ERK and Akt pathways, preserves mitochondrial function in RPE cells. | [48,65,66] |
AMD, DR, GL | Anti-inflammatory | Suppresses pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6). | [1,16,53,67] |
AMD, RP, BVMD, GL | Antioxidant | Activates Nrf2 pathway, enhances glutathione synthesis and heme oxygenase-1 expression. | [52,53,54,55,56] |
BVMD | Calcium homeostasis | Regulates calcium signaling through SERCA in RPE cells. | [55] |
BVMD, GL | Anti-inflammatory and neuroprotective | Suppresses inflammation and apoptosis in RPE cells; modulates cytokines and NF-κB. | [55,56] |
AMD, DR, RP, GL | Neuroprotective and anti-apoptotic | Modulates apoptotic regulators (Bcl-2 and Bax); suppresses caspase-3 activation. | [55,59,60,61] |
AMD | Autophagy activation | Restores autophagy flux, modulates LC3-II/LC3-I ratio, protects RPE cells from degeneration. | [68] |
Wet AMD, PVR, RVO, GL | Anti-angiogenic | Inhibits VEGF receptor phosphorylation (e.g., VEGFR2), limits endothelial cell migration. | [17,62,63,64,69,70,71,72,73,74] |
PVR, BVMD, GL | Fibrosis regulation | Inhibits EMT via suppression of Smad-dependent TGF-β1 signaling. | [74,75,76,77,78] |
RB | Anti-tumor | Upregulates miR-99a, modulates JAK/STAT. | [79] |
For wet AMD and other vascular-related conditions, PVR and retinal vascular obstruction (RVO), curcumin’s anti-angiogenic activity inhibits VEGF, preventing abnormal blood vessel formation and mitigating retinal damage [
17,
62,
64,
69,
70]. VEGF is essential for new blood vessel formation and vascular permeability, playing a key role in retinal diseases like DR, RVO, and exudative AMD. It is produced by retinal endothelial and pigment epithelial cells and is considered a key target in anti-angiogenic therapies. For instance, VEGF-A, a key driver in wet AMD progression, binds to VEGFR2, promoting angiogenesis and vascular leakage [
64,
71,
72]. Although glaucoma is not primarily an angiogenic disease, curcumin’s inhibition of VEGF may contribute to retinal vascular protection, especially in ischemic or injury contexts, with a positive impact on intraocular pressure regulation [
63,
73,
74].
Curcumin also exhibits neuroprotective and anti-apoptotic effects, reducing ganglion cell death and modulating apoptotic pathways. These effects are particularly significant in DR, AMD, and BVMD, where retinal cell survival is crucial for preserving vision [
55,
59,
60,
61]. This mechanism is also present in glaucoma, where curcumin exerts similar effects to protect optic nerve cells from programmed cell death, an important feature of glaucoma pathogenesis [
53,
67].
Curcumin regulates fibrosis, a hallmark of PVR and potentially BVMD, by inhibiting TGF-β1 activity and suppressing miR-21, a microRNA that promotes fibrogenesis, thereby further inhibiting the fibrotic process [
55,
75,
78]. This inhibition reduces the expression of fibrosis-related proteins, such as α-smooth muscle actin (α-SMA), type I collagen (COL1A1), and type III collagen (COL3A1), and potentially preserving retinal structure [
55,
75,
76,
77]. In glaucoma, curcumin has shown a similar effect by modulating fibrotic processes associated with optic nerve injury through the regulation of TGF-β1 and other fibrosis-related molecular factors [
63,
74].
Lastly, curcumin has demonstrated promising therapeutic potential in retinoblastoma (RB), the most common malignant intraocular tumor in children. It exerts anti-tumor effects by inhibiting cell proliferation, migration, and invasion while promoting apoptosis. These effects are primarily mediated through the upregulation of microRNA (miR-99a), which negatively regulates the Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway, a crucial pathway involved in tumor progression and cell survival. This suggests that curcumin’s modulation of miRNA expression contributes significantly to its anti-cancer properties, making it a candidate for adjunct therapy in RB treatment [
79].
2.2. Corneal Diseases
The cornea faces distinct challenges, including infections, fibrosis, and inflammation, which can compromise its transparency and refractive function. In this context, curcumin’s ability to modulate oxidative stress, inflammation, and angiogenesis has shown promise in addressing these conditions (
Table 3).
Inflammatory processes are central to keratitis and DED. Curcumin effectively inhibits p38 MAPK and NF-κB signaling, reducing pro-inflammatory cytokines like IL-1β, IL-6, and TNF-α. A study showed curcumin (5 μM) completely abolished hyperosmoticity-induced IL-1β elevation in human corneal epithelial cells [
48].
Oxidative stress exacerbates corneal injury and delays healing. Curcumin’s antioxidant capacity neutralizes ROS, protecting corneal cells and enhancing cellular survival under stress conditions. Guo et al. demonstrated that pretreatment with 12.5 µM curcumin enhances antioxidant defenses, including SOD1 and heme oxygenase-1, via the Keap1/Nrf2/ARE pathway, improving cell survival under oxidative stress [
57].
Curcumin also promotes corneal healing by stimulating cell migration and collagen synthesis, which reduces scarring and preserves corneal transparency. A dose-dependent effect was observed, where curcumin (10.0–12.5 mg/L) inhibited keratocyte proliferation and modulated fibrotic markers, upregulating decorin and CD90 (a glycoprotein marker of activated fibroblasts associated with tissue remodeling) while downregulating keratocan and aldehyde dehydrogenase [
80].
Table 3.
Curcumin: molecular mechanisms and activity potential in corneal diseases.
Table 3.
Curcumin: molecular mechanisms and activity potential in corneal diseases.
Relevant Diseases | Activity Potential | Key Actions and Targets | Ref. |
---|
Keratitis, dry eye disease | Anti-inflammatory | Inhibits p38 MAPK and NF-κB signaling, reducing cytokine production. | [48] |
Keratitis, dry eye disease | Oxidative stress reduction | Reduces ROS, protecting epithelial and endothelial cells. | [57] |
Corneal fibrosis, keratitis | Promotes healing andReducing fibrosis | Enhances cell migration and collagen synthesis, reducing scarring. | [80] |
Corneal fibrosis | Anti-angiogenic | Inhibits corneal neovascularization by suppressing VEGF. | [81] |
In corneal neovascularization, a severe complication often associated with corneal diseases, curcumin’s anti-angiogenic properties offer significant therapeutic advantages. By downregulating VEGF, curcumin impedes the formation of abnormal blood vessels, thereby reducing tissue damage and preserving vision. In an alkaline-burned rat model, the topical application of 40 μmol/L curcumin every 12 h for five days significantly reduced the area of new blood vessels compared to controls, showcasing its potential for managing angiogenesis-related corneal pathologies [
81].
2.3. Bacterial Ocular Diseases
Bacterial ocular infections, such as conjunctivitis, keratitis, and endophthalmitis, often involve severe inflammatory responses, biofilm formation, and resistance to conventional antibiotics. Curcumin’s molecular mechanisms address these challenges by targeting inflammation, oxidative stress, and bacterial survival strategies (
Table 4).
Biofilm formation is a critical factor in bacterial persistence and resistance. Curcumin disrupts biofilm matrix integrity and inhibits bacterial efflux pumps, thereby increasing susceptibility to antibiotics. This effect is particularly relevant against multidrug-resistant (MDR) pathogens, including MRSA and
Pseudomonas aeruginosa [
15].
Inflammation, a hallmark of bacterial ocular diseases, is modulated by curcumin through the inhibition of NF-κB and MAPK pathways, reducing cytokine storms and promoting ocular tissue recovery. In conjunctivitis, curcumin-based formulations like the product Haridra
® have shown anti-inflammatory and antibacterial efficacy [
82].
Oxidative stress exacerbates tissue damage during bacterial infections. Curcumin’s ability to neutralize ROS through the activation of the Keap1/Nrf2/ARE pathway preserves epithelial and retinal cell viability under stress conditions [
58].
Additionally, emerging research on the gut-ocular axis has opened new avenues for understanding how systemic factors, such as the gut microbiota, influence ocular immunity. Dietary interventions, including omega-3 fatty acids, carotenoids, and probiotics, have been shown to modulate both systemic and ocular immunity, reducing inflammation and improving overall eye health. Curcumin, with its anti-inflammatory and antioxidant effects, integrates into these strategies, offering a multifaceted approach to managing bacterial ocular diseases [
24].
Table 4.
Curcumin: molecular mechanisms and activity potential in bacterial diseases.
Table 4.
Curcumin: molecular mechanisms and activity potential in bacterial diseases.
Relevant Diseases | Activity Potential | Key Actions and Targets | Ref |
---|
Endophthalmitis | Anti-inflammatory | Reduces cytokine storms, protects retinal cells by modulating MAPK and NF-κB pathways. | [37] |
Conjunctivitis | Anti-inflammatory, antibacterial | Inhibits NF-κB signaling, reduces IL-1β and TNF-α, disrupts bacterial membranes. | [82] |
Keratitis | Antioxidant, antibacterial | Neutralizes ROS, inhibits MMP-9, disrupts biofilm matrix. | [58] |
2.4. Periocular and Ocular Surface Disorders
Chronic inflammation, immune dysregulation, and oxidative stress characterize eyelid diseases, such as blepharitis, blepharospasm, and eyelid dermatitis. Curcumin addresses these through its multi-targeted mechanisms, offering significant therapeutic potential (
Table 5).
Curcumin suppresses NF-κB and TLR4 signaling pathways, reducing pro-inflammatory cytokines like TNF-α and IL-1β. Additionally, it inhibits inflammasome activity, specifically NRLP3 to reduce tissue damage in blepharitis and dermatitis. Its antioxidant activity neutralizes ROS, mitigating oxidative tissue damage, while modulation of Th17 cell activity reduces immune dysregulation in autoimmune eyelid conditions [
49,
50,
51].
Table 5.
Curcumin: molecular mechanisms potential in periocular and ocular surface disorders.
Table 5.
Curcumin: molecular mechanisms potential in periocular and ocular surface disorders.
Relevant Diseases | Activity Potential | Key Actions and Targets | Refs. |
---|
Blepharitis, eyelid dermatitis | NF-κB Suppression | Reduces pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6); Suppresses NRLP3 activity, reducing tissue damage. | [49,50,51] |
Autoimmune eyelid disorders | Immune Modulation | Decreases Th17 activity and dendritic cell response; TLR4, MAPK, NF-κB. | [50] |
Chronic inflammation, dermatitis | Antioxidant Defense | Neutralizes free radicals, enhancing tissue resilience; glutathione, SOD, and catalase. | [83] |
Blepharospasm | Neuroprotective Effects | Modulates neuroinflammation and oxidative stress. | [83] |
AllergicConjunctivitis | Reduction of Inflammatory Markers | Suppresses key mediators, such as IL-4, IL-5, and iNOS, in ocular tissues. | [84] |
Meibomian gland Dysfunction | Anti-inflammatory, Lipid Regulation | Reduces inflammation in meibomian glands; modulates lipid composition in the tear film. | [85,86] |
Dry Eye Disease | Oxidative Stress Modulation, Mucin Enhancement | Reduces oxidative stress, stabilizes tear film, enhances mucin production, and improves ocular comfort. | [85,86] |
In blepharospasm, curcumin exhibits neuroprotective effects by reducing neuroinflammation and oxidative stress, promoting cellular resilience [
49,
51,
83]. Additionally, curcumin demonstrates potential in managing allergic conjunctivitis, an inflammatory condition of the ocular surface driven by Th2 immune responses. Studies indicate that curcumin reduces IgE-mediated inflammation, suppressing eosinophilic infiltration and Th2 cytokine production, such as IL-4 and IL-5, in the conjunctiva [
84]. These actions extend curcumin’s therapeutic scope to inflammatory and immune-related ocular surface disorders.
Further expanding its utility, curcumin shows promise in treating meibomian gland dysfunction (MGD), a leading cause of ocular discomfort, by reducing inflammation and improving the lipid composition of the tear film. In dry eye disease, which is often linked with MGD, curcumin alleviates inflammation and oxidative stress while enhancing mucin production, stabilizing the tear film, and improving ocular comfort [
85,
86].
These combined actions position curcumin as a promising agent for managing complex periocular and ocular surface disorders.
6. Conclusions
Curcumin demonstrates significant therapeutic potential in ophthalmology, particularly for retinal and corneal diseases, due to its anti-inflammatory, antioxidant, antibacterial, and anti-angiogenic properties. Its antibacterial activity could enhance treatment options for ocular surface infections, such as conjunctivitis or keratitis, by directly combating bacterial pathogens. However, challenges related to bioavailability and solubility need to be overcome through advanced drug delivery systems like nanoparticles, niosomes, and cyclodextrin complexes.
Curcumin’s therapeutic value lies in its pleiotropic effects, including anti-inflammatory, antioxidant, and anti-angiogenic activities. In comparison to traditional treatments, curcumin offers a multi-targeted approach that may complement or enhance existing therapies. For example, its ability to prevent inflammation and oxidative damage positions it as a potential adjunct to anti-VEGF treatments for conditions like age-related macular degeneration. However, its clinical application is limited by poor bioavailability, necessitating further research to establish its clinical effectiveness relative to conventional treatments.
In conclusion, curcumin holds promising therapeutic potential for ophthalmology, but further studies, especially clinical trials, are required to confirm its clinical efficacy and overcome existing limitations. The continued exploration of innovative delivery systems will be key to unlocking its full therapeutic potential.