International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Glaucoma is a group of progressive optic neuropathies characterized by retinal ganglion cell (RGC) loss and optic nerve damage that can lead to irreversible blindness (1,2). Estimates indicate that glaucoma is responsible for ~3.6 million cases of blindness among individuals aged ≥50 years (3). As the global population ages, the prevalence of glaucoma is expected to keep increasing. Therefore, research into this disease is of great significance for public health management and may have a profound socioeconomic impact.
Despite notable advances in medical and surgical treatments, ~12.5% of glaucomatous eyes will show fast vision loss and ~14.3% of patients with glaucoma will be blind in one eye within 20 years (4). The estimates highlight the need for novel and more effective treatments. Molecular biomarkers have been widely studied in glaucoma research (5), and molecular therapy could be a promising new approach (6). Recent advances in gene therapy, RNA interference (RNAi), cell-based strategies and small-molecule inhibitors have demonstrated marked preclinical success in animal models of glaucoma (5,6).
The present review provides a comprehensive overview of the current research progress on molecular therapies for glaucoma, encompassing the multifactorial mechanisms of glaucoma, emerging molecular treatment strategies and future directions.
The central pathological mechanism of glaucoma is trabecular meshwork (TM) dysfunction and increased intraocular pressure (IOP), which exert stress on the retina and neurons, progressively leading to vision loss (5). The TM regulates IOP by facilitating aqueous humor outflow and its dysfunction increases outflow resistance, leading to IOP elevation (5). Beyond IOP and translaminar pressure gradients, anatomical features of the optic nerve head, including the optic disc size and lamina cribrosa morphology, serve an important role in glaucoma (7). Larger discs are more susceptible to lamina cribrosa displacement under pressure gradients, which has been associated with impaired hemodynamics and reduced oxygen availability (7,8). Furthermore, vascular dysregulation may lead to chronic vasoconstriction, restricting nutrient and oxygen delivery to the retina and thereby contributing to glaucomatous damage (9).
Nevertheless, optic nerve degeneration at the optic nerve head has been reported to precede RGC injury, suggesting a window of therapeutic opportunity before actual RGC damage (10). Thus, by identifying and targeting these early molecular mechanisms, it may be possible to intervene before irreversible damage to the RGCs occurs, which could markedly improve the prognosis for patients with glaucoma.
Oxidative stress can directly damage the TM and impair aqueous humor outflow, leading to increased IOP; it also activates the RGCs, triggering inflammatory mediators that worsen damage and oxidative stress (11). Furthermore, both external and mitochondrial reactive oxygen species (ROS) can harm mitochondria, creating a cycle of increasing ROS, lower energy production and increased cell damage (11,12). Oxidative stress also triggers endoplasmic reticulum stress and DNA damage, both of which can harm RGCs (12,13).
A recent Mendelian randomization study highlighted the bidirectional causal effects of oxidative stress and glaucoma (14), supporting the view of a self-sustained vicious circle. Systemic biomarkers of oxidative stress such as total antioxidant capacity are elevated in patients with elevated IOP (15). Notably, oxidative stress is a complex condition that also involves exogenous factors; results from the large-scale National Health and Nutrition Examination Survey (NHANES) study indicated that elevated oxidative balance scores (calculated based on nutritional and lifestyle factors) were associated with glaucoma (16). Physical activity may have favorable impacts on glaucoma progression through decreased oxidative stress (17). Therefore, these results highlight the importance of oxidative stress in glaucoma pathogenesis and pathophysiology.
Mitochondrial abnormalities serve a critical role in the pathogenesis of primary open angle glaucoma (POAG), particularly through their involvement in ROS production, energy metabolism and cell survival (18). Elevated IOP, aging, neuroinflammation, vascular impairment, neurotrophic factor deprivation and oxidative stress collectively disrupt the delicate balance of mitochondrial fission and fusion (18). This imbalance leads to reduced mitochondrial efficiency, increased ROS production and compromised cellular energy supply, all of which exacerbate RGC damage (18).
Inflammation, closely associated with oxidative stress and mitochondrial dysfunction, is a central process in increased IOP and glaucoma (19,20). Intermediate inflammation (or para-inflammation) is an adaptive process that involves stresses and malfunctions in the retina and is involved in preserving tissue homeostasis and function. However, it may be deleterious if sustained over long periods (20). Such inflammation could contribute to impaired aqueous outflow and increased IOP (21). C-reactive protein is a biomarker of systemic low-grade inflammation and has been associated with glaucoma in a recent meta-analysis (22). Another NHANES analysis linked the systemic inflammatory response index with the incidence of glaucoma (23).
Furthermore, RGC injury leads to RGC death (24). As RGCs cannot regenerate and reconnect to the visual pathway, their death will lead to progressive vision loss (24,25). Notably, recent research identified that different subtypes of RGCs have different vulnerability levels in the pathogenesis of glaucoma, which could have implications for glaucoma management (25). Historically, apoptosis was considered the most important RGC death mechanism, but focusing on apoptosis led to disappointing results as saving RGCs through caspase inhibition still ultimately leads to cell death through mitochondrial dysfunction (26).
Ferroptosis is a recently identified iron-dependent programmed cell death triggered by lipid peroxidation and relying on iron-generated ROS, and it is involved in RGC death (27,28). Ferroptosis induction is associated with higher blood iron (which is associated with a higher risk of glaucoma), excitotoxicity, neuroinflammation, ischemia/reperfusion injury and a pathologically high IOP, which also all participate in glaucoma (24,29). Pyroptosis is another programmed cell death path involved in glaucoma RGC death; however, the exact mechanism remains elusive (24). Under high IOP, the glial cells are activated and their recruitment, except for the RGCs, precedes RGC death. Glial cells participate in inflammation, and pyroptosis is associated with inflammation (24).
Mitochondrial fission, fusion and function depend on proteins such as optic atrophy protein 1 (OPA1), dynamin-related protein 1 (DRP1), A-kinase anchoring protein 1 (AKAP1) and nicotinamide nucleotide adenylyltransferase 1 (NMNAT1). OPA1 mutations can cause optic neuropathies, and single nucleotide polymorphisms in OPA1 have been associated with POAG and normal tension glaucoma (30). Acute high IOP raises DRP1 levels, leading to early neurodegenerative events, increased gliosis and RGC apoptosis (31). AKAP1 expression appears decreased or lost in glaucoma, suggesting its role in RGC injury and death (31). NMNAT1 is involved in NAD+ synthesis, mitigating mitochondrial dysfunction and related diseases (32). Decreased NAD+ levels have been associated with glaucoma and retina survival depends upon adequate NAD+ (33).
Mutations in several genes increase the susceptibility to glaucoma. Myocilin (MYOC) is a protein involved in the formation and maintenance of the TM, a structure involved in aqueous humor drainage. Mutations in the MYOC gene are associated with protein misfolding and dysfunctional TM and account for ~5% of POAG cases (34). Mutations in the cytochrome P450 family 1 subfamily B member 1 (CYP1B1) gene are responsible for ~20% of the cases of childhood glaucoma in Japan (35) and are associated with congenital glaucoma (36). The forkhead box C1 (FOXC1) gene encodes a transcription factor, and mutations in FOXC1 lead to ocular drainage dysfunction (35). Optineurin (OPTN) is an adapter protein involved in several metabolic processes. Mutations in the OPTN gene are connected to neurodegenerative disorders and glaucoma (37,38). Recent studies have also implicated the mutations in ataxin 2 (associated with reduced RGC survival and elevated IOP) (39), EGF containing fibulin extracellular matrix protein 1 (linked to juvenile or adult-onset hereditary isolated glaucoma) (40) and paired box 6 (critical for prenatal ocular development, leading to secondary glaucoma) (41). Therefore, the large number of genes associated with glaucoma illustrate the complexity of the disease.
Besides mutations in specific genes, Moazzeni et al (42) identified 18 transcription factors, 195 microRNAs (miRNAs/miRs), 106 long non-coding RNAs and two circular RNAs as being involved in glaucoma pathogenesis, highlighting the multifaceted pathophysiology of glaucoma. A proteomic study identified 176 dysregulated proteins in cells and 7 in extracellular vesicles, suggesting numerous potential glaucoma biomarkers (43). A metabolomic study reported that patients with POAG displayed decreased levels of phenylalanine, phenylacetate, leucine, N-acetylated compounds, formic acid and uridine in their tears, and increased taurine, glycine, urea, glucose and unsaturated fatty acids, allowing the non-invasive detection of POAG with 100% sensitivity and 83% specificity (44). Exosome density and aqueous humor programmed death-ligand 1 levels can also provide information about retinal damage in patients with glaucoma (45).
In conclusion, mitochondria-associated proteins regulate mitochondrial function through complex interactions, and their dysregulation is closely associated with optic neuropathies, including glaucoma. Several gene mutations increase the susceptibility to glaucoma by affecting the function of the TM, metabolic processes and ocular drainage structures, highlighting the complexity of this disease and its therapeutic challenges. Multiple molecules, including transcription factors, non-coding RNAs, dysfunctional proteins and metabolites, are involved in the pathogenesis of glaucoma. These may collectively provide potential biomarkers and non-invasive testing methods for the diagnosis, prognosis and treatment of glaucoma.
An improved understanding of the classical, metabolic and molecular factors involved in glaucoma may help to elucidate several potential treatment targets, several of which are already in the preclinical stages and appear promising.
Mutations in several genes are involved in the pathogenesis of glaucoma, including MYOC, CYP1B1, FOXC1 and OPTN, among others. Although certain treatments can be effective in certain patients with gene mutation-related glaucoma, the only possible curative option is the correction of the mutated allele(s) using gene therapy (46). Several studies have assessed different methods for editing several genes involved in glaucoma (Table I). For instance, gene editing targeting MYOC mutations can reduce endoplasmic reticulum stress caused by the accumulation of misfolded MYOC and restore the MYOC protein function in the TM, leading to reduced IOP in animal models (47–49). Introducing viral vectors that express a functional protein can also be used instead of gene editing, and such an option is being explored for the OPTN gene as OPTN is involved in ~19% of POAG cases (50). Nevertheless, nerve regeneration using gene therapy is an intense area of research, and it could ultimately be used to regenerate RGCs (51). Aquaporin 1 (AQP1) is involved in aqueous humor production and targeting AQP1 decreases aqueous humor production and IOP (52). Elevated transforming growth factor β 2 (TGFβ2) expression is associated with pathological changes in the TM, and POAG shows elevated TGFβ2 expression (53). Interfering with TGFβ2 expression using CRISPR technology could be used to manage POAG (54). Additionally, suppressing the phosphatase and tensin homolog (PTEN) gene also appears neuroprotective in RGCs (55). Therefore, gene editing could also be used to improve surgical outcomes. Lee et al (56) reported that targeting the connective tissue growth factor gene using CRISPR technology could reduce fibrosis after glaucoma filtration surgery. Moreover, a recent study reported that the disruption of the AQP1/β2 adrenergic receptor/rho-associated protein kinase 1/rho-associated protein kinase 2 genes using CRISPR-CasRx technology reduced IOP and RGC damage in mice (57). Therefore, these genes may be targeted to improve glaucoma outcomes.
Gene therapy can also modulate beneficial or harmful protein production in glaucoma; however, its success is limited, potentially due to the complex genetic basis of the disease (46). Nevertheless, the expression of several therapeutic genes have been explored (Table I), including brain-derived neurotrophic factor (BDNF) (58–61), tropomyosin-related kinase receptor-B (TrkB) (58,61), brain-specific homeobox/POU domain protein 3b (Brn3b) (62,63), B-cell lymphoma-xl (Bcl-xl) (64), Myc-associated protein X (MAX) (65), neuroprotective intracellular transcription factor 2 (Nrf2) (60), superoxide dismutase 2 (SOD2) (66), ATP-binding cassette A1 (67), C3 (68), mouse γ-synuclein (mSncg) (69), K-Ras (70) and matrix metalloproteinase-3 (MMP-3) (71). The proposed therapies target mechanisms such as neuroprotection (BDNF, TrkB, MAX, Nrf2, C3, K-Ras and mSncg), apoptosis (Brn3b, Bcl-xl and ATP-binding cassette A1), oxidative stress (SOD2) and aqueous humor outflow (MMP-3). Although several genes can be theoretically modulated to influence IOP and glaucoma progression, compensatory mechanisms from other genes are often observed, and adverse effects of modulating genes must be avoided (46). However, several companies are exploring the use of genes involved in IOP and RGC neuroprotection as drugs (72). NADH-quinone oxidoreductase-based gene therapy can improve mitochondrial function and reduce oxidative stress (73). Additionally, CRISPR/Cas technology can be used to disrupt genes related to high IOP. For example, AQP1 is involved in aqueous humor production, and its disruption decreases IOP (47,51). Such a disruptive approach could also be used for TGFβ2, which is elevated in ~50% of patients with POAG, and participates in extracellular matrix (ECM) remodeling and elevated IOP (46,54). Caveolin-1 also serves a role in the response of RGCs to increased IOP, and ablating caveolin-1 in animal models has been reported to improve the glaucoma phenotype (74).
An alternative to gene editing or introducing a novel copy of a gene is RNAi, which works by interfering with the mRNA of the target gene, offering a non-permanent regulatory approach. Small interfering RNAs (siRNAs) can be delivered using vesicles, and their effect is transient. Although it can necessitate repeated injections, this method avoids the ethical and safety issues associated with manipulating the genome of an individual. Furthermore, siRNAs can be delivered locally (such as in the aqueous humor) to perform their effect in a limited area, such as the retina (75,76). RNAi technology is excellent for short-term or local treatments due to its reversibility and suitability for local applications (75). This allows the selection of the most appropriate strategy based on the specific pathological mechanisms and treatment needs of glaucoma, thereby maximizing treatment effectiveness while minimizing potential risks (76).
Stem cells, particularly mesenchymal stem cells (MSCs), induced pluripotent stem cells (iPSCs), embryonic stem cells (ESCs) and retinal progenitor cells, offer the potential to restore damaged tissue and replace lost or dysfunctional cells (77). Restoring TM function and regenerating damaged RGCs are key goals in glaucoma treatment (77).
Restoration of TM cellularity and function could repopulate the outflow pathway, re-establish physiological aqueous humor drainage and durable IOP control. Native TM stem cells (TMSCs), iPSC-derived TM cells (iPSC-TMs) and MSCs have been investigated for TM regeneration, each with distinct advantages and translational profiles (78–80). Human TMSCs can be isolated from donor TM tissue and display label-retaining, slow-cycling properties, multipotency and the capacity to home specifically to TM tissue after intracameral delivery (78,80). In preclinical mouse models, intracameral injection of expanded human TMSCs resulted in TM engraftment without notable inflammation and the appearance of TM marker expression within days, suggesting in situ differentiation and niche restoration (78,81). Complementary work has reported that transplanted iPSC-TMs not only survive in perfused human anterior segments and animal models, but also stimulate proliferation of endogenous TM cells, increase TM cellularity and improve outflow facility (79,82). These findings support a model in which exogenous cells can both directly replace lost TM cells and act via paracrine or contact-dependent cues to recruit endogenous progenitors.
Several groups have extended these approaches to disease models and shown functional benefit. For instance, in mouse glaucoma models, intracameral transplantation of TMSCs or iPSC-TMs decreased IOP and restored outflow facility, whereas adipose-derived stem cells and MSCs improved TM cellularity and ECM turnover, leading to partial normalization of IOP (80,81). Mechanistically, cell therapies act via the following: i) Direct replacement of TM endothelial-like cells; ii) secretion of matrix-remodeling enzymes and trophic factors that normalize ECM deposition; and iii) activation of endogenous TM progenitor proliferation (78–82). Notably, in certain studies only a small fraction of transplanted cells persisted long-term; nevertheless, the therapeutic effect has often been associated with increased proliferation of resident TM cells, pointing to an important inductive or paracrine role for the graft (79,82).
Despite robust preclinical progress, key translational challenges remain. These include the following: Ensuring the tolerability and safety of intracameral cell delivery (avoiding inflammation, angle obstruction or aberrant neovascularization); defining optimal cell sources and differentiation protocols to yield stable TM phenotypes; scaling good manufacturing practice (GMP)-compliant manufacturing; and establishing long-term functional endpoints in large-animal models and human ex-vivo perfusion systems prior to first-in-human studies (81). In addition, TM regional heterogeneity and disease-associated niche alteration (such as in POAG eyes with pronounced ECM remodeling) may require combinatorial strategies (pairing cell replacement with ECM-modulating enzymes or gene-editing of resident cells) to achieve durable restoration (78–82). In summary, stem-cell based TM regeneration has advanced to compelling preclinical proof-of-concept and the next translational steps (standardized manufacturing, safety testing in large animals and controlled early clinical trials) are now tractable priorities for the field.
Stem cell-based approaches for regenerated damaged RGCs target two complementary therapeutic goals: i) Neuroprotection of surviving RGCs and their axons; and ii) replacement or repopulation of lost RGCs to restore visual function (83). Several cell types have been investigated preclinically, including MSCs, ESCs, neural progenitor cells and iPSC-derived retinal ganglion-like cells (iPSC-RGCs) (83–87). The therapeutic actions of transplanted cells are multifactorial and include paracrine secretion of neurotrophic factors (such as BDNF and ciliary neurotrophic factor), immunomodulation (reduction of microglial activation and inflammatory cytokines), antioxidative effects, delivery of mitochondria or mitochondrial rescue factors and, in certain contexts, direct differentiation or fusion with host retinal neurons (83,84). Over the past decade, attention has also shifted to cell-free therapies based on stem cell-derived extracellular vesicles (EVs; including exosomes), which recapitulate several paracrine benefits whilst reducing risks associated with live-cell transplantation (immune rejection and ectopic growth) (85).
Multiple preclinical studies have reported efficacy in models relevant to glaucoma (84–86). In optic nerve crush models, intravitreal or periocular delivery of human Wharton's jelly MSCs enhanced long-term RGC survival, promoted axonal regeneration and, in a report, enabled partial reconnection to central visual targets up to 120 days post-injury (86). These experiments used quantitative histology, anterograde axon tracing to assess regeneration and functional readouts such as visually evoked potentials. In ocular hypertension models, intravitreal injection of MSC-derived EVs reduced RGC apoptosis, decreased glial reactivity and preserved inner retinal structure and function on electroretinogram/visually evoked potential testing (84,85). Notably, studies have identified miRNA cargoes (such as miR-21 and miR-146a) and protein factors within EVs that mediate antiapoptotic and anti-inflammatory signaling, which can be validated by gain-/loss-of-function experiments (84,85).
Efforts toward RGC replacement have advanced using human PSC technologies (87,88). Protocols now differentiate ESCs or iPSCs into RGC-like neurons expressing canonical markers (such as brain-specific homeobox 3A, RNA-binding protein with multiple splicing and synuclein γ) and functional properties (such as spiking and synaptic proteins) (87,88). Transplantation of iPSC-RGCs into rodent retina results in survival and partial integration within the ganglion cell layer (87). Certain studies have reported axon extension toward the optic nerve head, but long-distance and target-specific reconnection to thalamic or collicular targets remains rare without additional pro-regenerative manipulations (87,88). Recent work emphasizes combinatorial strategies, such as pairing cell replacement with gene or pharmacological enhancement of intrinsic regenerative programs and modulation of the inhibitory extracellular environment, to improve integration and functional outcome (88).
Clinical translation is progressing cautiously. Several early-phase clinical initiatives (such as the Stem Cell Ophthalmology Treatment Study, SCOTS/SCOTS2; clinicaltrials.gov NCT03011541) have investigated autologous bone marrow-derived stem cells for optic nerve and retinal diseases, reporting safety signals (no surgical complications, no need for immunosuppression and no teratoma formation) but mixed efficacy outcomes and methodological heterogeneity. A systematic review and meta-analysis indicated that stem cell therapies show promise in improving surrogate visual outcomes in optic neuropathies (89); however, high-quality randomized data for glaucoma specifically remain lacking and long-term safety requires continued vigilance (89).
Nevertheless, key technical and translational challenges persist, including the following: i) Cell survival and targeted delivery: The vitreous and inner retinal milieu can be hostile and cell engraftment rates are typically low; ii) immune and tumorigenic risks: Allogeneic cells may provoke immune responses and pluripotent cells carry teratoma risk unless rigorously purified; iii) functional integration: Even when transplanted RGC-like cells survive, forming correct synaptic connections with bipolar/amacrine cells and extending axons through the optic nerve to central targets is notably difficult in the adult mammalian central nervous system; and iv) standardization and potency assays: Reproducible manufacturing, potency assays and release criteria for cell therapy products are still being refined (90). To address these, current preclinical trends focus on the following: i) EV-based therapies as a safer, cell-free approach; ii) biomaterial scaffolds and hydrogel matrices that improve cell retention and oriented axon growth; iii) combination approaches that include gene editing (such as CRISPR to modulate PTEN pathways), neurotrophic factor delivery and local immunomodulation; and iv) careful stepwise clinical translation with standardized endpoints, long-term follow-up and registries (84,88,90).
In summary, stem cell therapies provide compelling preclinical evidence for restoration of the TM and neuroprotection in glaucoma models and offer a plausible route toward regenerative strategies. Nevertheless, major biological and translational hurdles must be overcome before routine clinical application. Priorities for the field include rigorous mechanism-of-action studies, standardized manufacturing and potency assays, well-designed early-phase clinical trials with objective functional endpoints and the development of combination strategies to enable true structural and functional restoration of the TM and RGC pathway (91).
Once damaged, RGCs are unable to regenerate or reconnect to the visual pathway (6,24). Therefore, preventing their death is a critical strategy for preserving vision in patients with glaucoma (5,6,24). To achieve this, the disease process can be intervened through certain molecular mechanisms (5,6,24,92–97).
Inhibiting ferroptosis (including using iron chelators such as deferiprone and deferoxamine, lipid ROS scavengers such as ferrostatin-1 and endogenous iron-regulating proteins such as transferrin) (24), blocking pyroptosis (including applying caspase-1 inhibitors such as fluoromethyl ketone and NLR family pyrin domain containing 3 inhibitors such as baicalin extract) (24) and modulating inflammatory responses can help to slow down RGC damage (6).
In addition, a variety of molecules have shown potential in glaucoma treatment. Insulin has been reported to stimulate the regeneration of RGC dendrites and synapses during ocular hypertension (92). However, glucocorticoid treatment, which is commonly associated with increased IOP, involves the activation of glucocorticoid receptors (93). Therefore, blocking these ocular receptors may represent a potential strategy to prevent IOP elevation. Additionally, given that glaucoma involves both histaminergic and nitrergic systems, combining a histamine H3 receptor antagonist with a nitric oxide donor could offer an effective approach to managing IOP (94). Furthermore, NAD+ depletion serves a marked role in several neurodegenerative diseases, including glaucoma. Oral niacinamide treatment has demonstrated the ability to improve visual outcomes in patients with glaucoma, suggesting its potential as a therapeutic option (95). In a preclinical study, stable gastric pentadecapeptide body protection compound 157 therapy has been reported to alleviate signs and symptoms of glaucoma in rat models (96). Another promising avenue involves the activation of σ-1 receptors, as their low expression is associated with RGC degeneration (97).
Moreover, neuroprotection serves an important role in glaucoma treatment. Several growth factors (such as ciliary neurotrophic factor, nerve growth factor and brain-derived growth factor) have notable neuroprotective effects and can protect and repair RGCs (6). In addition, metabolic abnormalities have been reported to be involved in the pathogenesis of glaucoma, particularly in the occurrence and development of RGC damage (6). Therefore, intervening in the progression of glaucoma through metabolic regulation is of great significance. For example, metformin, insulin and glucagon-like peptide-1 receptor agonists are not only effective for type 2 diabetes but also show potential therapeutic value for glaucoma (6). Taken together, these findings suggest that targeting several molecular pathways could pave the way for more effective therapies.
Nanomedicine, the application of nanoscale drug carriers and delivery platforms, improves the ocular penetration of traditional eye drops, provides sustained and controlled release, enables targeted delivery to anterior- or posterior-segment structures and supports co-delivery of multi-modal payloads (such as IOP-lowering agents plus neuroprotectants) (98). Nanomedicine represents promising avenues for glaucoma management, spanning IOP control, neuroprotection and combination approaches that may reduce dosing burden and enhance disease modification in the future (99,100).
Common nanocarrier classes for glaucoma include liposomes, solid lipid nanoparticles (SLNs) and nanostructured lipid carriers, polymeric nanoparticles [such as poly(lactic-co-glycolic acid) (PLGA)], nanoemulsions, niosomes, dendrimers and hybrid/hydrogel embedded nanoparticle systems (98,100). Lipid-based systems are particularly attractive for lipophilic prostaglandin analogs; preclinical work has reported that latanoprost or bimatoprost loaded into liposomes or SLNs can markedly extend ocular residence and prolong IOP lowering after a single administration compared with eye drops (101). For example, Satyanarayana et al (101) developed bimatoprost-loaded SLNs that provide extended in vivo release and tolerability in rabbits, supporting the potential to reduce dosing frequency.
For hydrophilic drugs (such as timolol), mucoadhesive gelatin or polymeric nanoparticles embedded in viscous vehicles have been reported to improve corneal retention and sustain delivery, translating into longer pharmacodynamic effects in preclinical studies (98,102). Polymeric PLGA nanoparticles have been used to encapsulate neuroprotective small molecules (such as memantine) to enhance posterior-segment delivery and RGC protection in experimental glaucoma models (103). Niosomes and proniosomal gels have been explored to sustain the release of brimonidine, increasing ocular residence and potentially enhancing its documented neuroprotective actions independent of IOP lowering (104). Furthermore, nanoemulsions and contact-lens/insert reservoirs represent alternative platforms to achieve prolonged release whilst maintaining patient comfort and compliance (98,100).
Beyond single-agent delivery, nanocarriers enable combination strategies: Co-encapsulation of an IOP-lowering drug with an anti-inflammatory or antioxidant payload can concurrently reduce pressure and modulate pathogenic microenvironments that contribute to RGC loss. Nanocarriers can also be surface-modified (PEGylation, targeting ligands and mucoadhesive coatings) to tune corneal uptake, reduce clearance and direct payloads toward TM, ciliary body or posterior tissues when needed (98,100).
However, despite promising preclinical data, several translational barriers remain. Safety concerns include local ocular irritation, inflammation, corneal toxicity and the long-term fate of non-biodegradable nanomaterials. Manufacturing at GMP scale with batch-to-batch consistency, stability during storage, sterilization without compromising carrier integrity and regulatory pathways for combination products (device + drug) are non-trivial hurdles (98,100). Immunogenicity of certain surface chemistries and the potential for ocular accumulation with repeated dosing require long-term biocompatibility studies. Moreover, economic and commercial considerations (such as cost of goods, patient acceptance and delivery form factors) also influence which nanoplatforms progress to clinical trials (98,99).
In conclusion, conventional glaucoma treatment methods have limitations in efficacy and struggle to meet clinical needs. Therefore, future treatment directions should focus on emerging technologies such as gene therapy, gene editing, stem cell therapy and molecular targeted therapy. These methods offer new possibilities for glaucoma treatment by precisely intervening in disease mechanisms.
Although current gene therapies are unable to reverse vision loss caused by RGC death, they can effectively delay or even halt disease progression, thereby preventing further visual impairment. If applied in the early stages of the disease, such treatments may also completely avoid notable vision loss and achieve improved prognostic outcomes. However, these cutting-edge therapies are still in the developmental stage, and their clinical translation faces certain challenges. Although certain gene-editing-based clinical trials (such as NCT04560790, NCT01949324, NCT02862938, NCT04577300 and NCT03872479) (105) are ongoing or have been completed, most therapeutic strategies remain in the preclinical research phase and have not yet been widely applied in clinical practice. Therefore, further efforts are needed to strengthen basic research and clinical trials to validate the safety and efficacy of these therapies, facilitating their earlier entry into clinical application.
Several genes have been associated with POAG, highlighting the complex and diverse genetic basis of glaucoma (106). However, the specific functions of several of these genes and their roles in the disease mechanism remain incompletely understood, indicating that numerous potential molecular targets still await further investigation. Future research may not only uncover additional gene targets associated with glaucoma but also offer new approaches and strategies for molecular therapy.
Notably, glaucoma may share certain pathophysiological mechanisms with other neurodegenerative diseases, such as Alzheimer's disease and Parkinson's disease (107,108). These shared mechanisms include apoptosis, ferroptosis, pyroptosis, mitochondrial dysfunction, oxidative stress and inflammation (107,108). These common mechanisms suggest that glaucoma may have similar neuroprotective and neuroregenerative requirements as these diseases. Therefore, effective therapeutic approaches for neurodegenerative diseases may also be applicable to the treatment of glaucoma. Exploring these shared mechanisms may uncover new treatment strategies for glaucoma and foster interdisciplinary research.
Not applicable.
National Natural Science Foundation of China (grant no. 81500719), Shaanxi Science and Technology Project (grant no. 2022SF-434) and Xi'an Science and Technology Project (grant no. 21YXYJ0044).
Not applicable.
WW drafted the original article. GC, QZ, SW and LZ edited, validated and revised the original article. All authors read and approved the final version of the manuscript. Data authentication is not applicable.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
Shan S, Wu J, Cao J, Feng Y, Zhou J, Luo Z, Song P and Rudan I; Global Health Epidemiology Research Group (GHERG), : Global incidence and risk factors for glaucoma: A systematic review and meta-analysis of prospective studies. J Glob Health. 14:042522024. View Article : Google Scholar : PubMed/NCBI | |
|
Chen DF, Wang C, Si Y, Lu X, Zhou W, Huang Q, Zuo J, Cheng G, Leung DYL, Wang N, et al: Natural history and risk factors for glaucoma progression in Chinese patients with normal-tension glaucoma. Invest Ophthalmol Vis Sci. 65:282024. View Article : Google Scholar : PubMed/NCBI | |
|
GBD 2019 Blindness and Vision Impairment Collaborators and Vision Loss Expert Group of the Global Burden of Disease Study, . Causes of blindness and vision impairment in 2020 and trends over 30 years, and prevalence of avoidable blindness in relation to VISION 2020: The Right to Sight: An analysis for the Global Burden of Disease Study. Lancet Glob Health. 9:e144–e160. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Jackson AB, Martin KR, Coote MA, Medeiros FA, Girkin CA, Fazio MA, Liebmann JM, De Moraes CG, Weinreb RN, Zangwill LM and Wu Z: Fast progressors in glaucoma: Prevalence based on global and central visual field loss. Ophthalmology. 130:462–468. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang W and Wang H: Understanding the complex genetics and molecular mechanisms underlying glaucoma. Mol Aspects Med. 94:1012202023. View Article : Google Scholar : PubMed/NCBI | |
|
Bou Ghanem GO, Wareham LK and Calkins DJ: Addressing neurodegeneration in glaucoma: Mechanisms, challenges, and treatments. Prog Retin Eye Res. 100:1012612024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang YX, Panda-Jonas S and Jonas JB: Optic nerve head anatomy in myopia and glaucoma, including parapapillary zones alpha, beta, gamma and delta: Histology and clinical features. Prog Retin Eye Res. 83:1009332021. View Article : Google Scholar : PubMed/NCBI | |
|
Chuangsuwanich T, Birgersson KE, Thiery A, Thakku SG, Leo HL and Girard MJ: Factors influencing lamina cribrosa microcapillary hemodynamics and oxygen concentrations. Invest Ophthalmol Vis Sci. 57:6167–6179. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou W and Sabel BA: Vascular dysregulation in glaucoma: Retinal vasoconstriction and normal neurovascular coupling in altitudinal visual field defects. EPMA J. 14:87–99. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Maddineni P, Kasetti RB, Patel PD, Millar JC, Kiehlbauch C, Clark AF and Zode GS: CNS axonal degeneration and transport deficits at the optic nerve head precede structural and functional loss of retinal ganglion cells in a mouse model of glaucoma. Mol Neurodegener. 15:482020. View Article : Google Scholar : PubMed/NCBI | |
|
Fernández-Albarral JA, Ramírez AI, de Hoz R, Matamoros JA, Salobrar-García E, Elvira-Hurtado L, López-Cuenca I, Sánchez-Puebla L, Salazar JJ and Ramírez JM: Glaucoma: From pathogenic mechanisms to retinal glial cell response to damage. Front Cell Neurosci. 18:13545692024. View Article : Google Scholar : PubMed/NCBI | |
|
Hurley DJ, Normile C, Irnaten M and O'Brien C: The intertwined roles of oxidative stress and endoplasmic reticulum stress in glaucoma. Antioxidants (Basel). 11:8862022. View Article : Google Scholar : PubMed/NCBI | |
|
Tanito M, Kaidzu S, Takai Y and Ohira A: Association between systemic oxidative stress and visual field damage in open-angle glaucoma. Sci Rep. 6:257922016. View Article : Google Scholar : PubMed/NCBI | |
|
Shi R, Wu Y, Chen H, Zhang Z, Bao S, Qu J and Zhou M: The causal effect of oxidative stress on the risk of glaucoma. Heliyon. 10:e248522024. View Article : Google Scholar : PubMed/NCBI | |
|
Baris ME, Furundaoturan O, Kocamanoğlu M, Şahin S, Akçay Y and Yılmaz SG: Serum oxidative stress-related biomarkers in ocular hypertension and glaucoma. J Ophthalmic Vis Res. 19:433–439. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Huang J, Zhang Y, Wu C, Wu Y, Wang F, Ning Y and Shi L: Association between oxidative balance score and glaucoma in the National Health and Nutrition Examination Survey. Front Nutr. 12:15281142025. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Q, Jiang Y, Deng C and Wang J: Effects and potential mechanisms of exercise and physical activity on eye health and ocular diseases. Front Med (Lausanne). 11:13536242024. View Article : Google Scholar : PubMed/NCBI | |
|
Ju WK, Liu Q, Perkins GA, Kim KY, Bastola T, Choi WY and Choi SH: Glaucomatous optic neuropathy: Mitochondrial dynamics, dysfunction and protection in retinal ganglion cells. Prog Retin Eye Res. 95:1011362023. View Article : Google Scholar : PubMed/NCBI | |
|
Reinehr S, Rahim Pamuk M, Fuchshofer R, Dick HB and Joachim SC: Increased inflammation in older high-pressure glaucoma mice. Neurobiol Aging. 145:55–64. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Baudouin C, Kolko M, Melik-Parsadaniantz S and Messmer EM: Inflammation in glaucoma: From the back to the front of the eye, and beyond. Prog Retin Eye Res. 83:1009162021. View Article : Google Scholar : PubMed/NCBI | |
|
Sacca SC, Gandolfi S, Bagnis A, Manni G, Damonte G, Traverso CE and Izzotti A: From DNA damage to functional changes of the TM in aging and glaucoma. Ageing Res Rev. 29:26–41. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Al-Namaeh M: A meta-analysis of the association between high-sensitivity C-reactive protein level and glaucoma. Eur J Ophthalmol. 35:29–39. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Li X, Sun YQ, Zhong XD, Zhang ZJ, Tang JF and Luo ZY: Association between systemic inflammatory response index and glaucoma incidence from 2005 to 2008. Front Med (Lausanne). 12:15420732025. View Article : Google Scholar : PubMed/NCBI | |
|
Chen D, Miao S, Chen X, Wang Z, Lin P, Zhang N and Yang N: Regulated necrosis in glaucoma: Focus on ferroptosis and pyroptosis. Mol Neurobiol. 61:2542–2555. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Morgan JE, Bevan RJ and Cimaglia G: Retinal ganglion cell subtypes and their vulnerability in glaucoma. Methods Mol Biol. 2858:191–205. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Qin Q, Yu N, Gu Y, Ke W, Zhang Q, Liu X, Wang K and Chen M: Inhibiting multiple forms of cell death optimizes ganglion cells survival after retinal ischemia reperfusion injury. Cell Death Dis. 13:5072022. View Article : Google Scholar : PubMed/NCBI | |
|
Feng Y, Wang X, Li P, Shi X, Prokosch V and Liu H: Exogenous hydrogen sulfide and NOX2 inhibition mitigate ferroptosis in Pressure-induced retinal ganglion cell damage. Biochim Biophys Acta Mol Basis Dis. 1871:1677052025. View Article : Google Scholar : PubMed/NCBI | |
|
Yao F, Peng J, Zhang E, Ji D, Gao Z, Tang Y, Yao X and Xia X: Pathologically high intraocular pressure disturbs normal iron homeostasis and leads to retinal ganglion cell ferroptosis in glaucoma. Cell Death Differ. 30:69–81. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Huang S, Liu K, Su Y, Wang F and Feng T: Research progress of ferroptosis in glaucoma and optic nerve damage. Mol Cell Biochem. 478:721–727. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Yu-Wai-Man P, Griffiths PG, Gorman GS, Lourenco CM, Wright AF, Auer-Grumbach M, Taylor RW, Turnbull DM and Chinnery PF: OPA1 mutations cause cytochrome c oxidase deficiency due to loss of wild-type mtDNA molecules. Hum Mol Genet. 19:3043–3052. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Edwards G, Gnanasekaran A, Jeong YM, Liu Y, Croft K, Dickey AS, Bernstein SL, Cortopassi GA and Flippo KH: Loss of AKAP1 triggers Drp1 dephosphorylation-mediated mitochondrial fission and loss in retinal ganglion cells. Cell Death Dis. 11:2542020. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang H, Ryu D, Wu Y, Gariani K, Wang X, Luan P, D'Amico D, Ropelle ER, Lutolf MP, Aebersold R, et al: NAD+ repletion improves mitochondrial and stem cell function and enhances life span in mice. Science. 352:1436–1443. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Petriti B, Fernandez-Albarral JA, Salobrar-Garcia E, Lopez-Cuenca I, Ramirez AI, de Hoz R, Trivino A and Ramirez JM: Neuroprotection in Glaucoma: NAD+/NADH redox state as a potential biomarker and therapeutic target. Cells. 10:14022021. View Article : Google Scholar : PubMed/NCBI | |
|
Saccuzzo EG, Youngblood HA and Lieberman RL: Myocilin misfolding and glaucoma: A 20-year update. Prog Retin Eye Res. 95:1011882023. View Article : Google Scholar : PubMed/NCBI | |
|
Fuse N, Kimura M, Shimizu A, Koshiba S, Hamanaka T, Nakamura M, Ishida N, Sakai H, Ikeda Y, Mori K, et al: Mutations of CYP1B1 and FOXC1 genes for childhood glaucoma in Japanese individuals. Jpn J Ophthalmol. 68:688–701. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Faiq MA, Singh HN, Ali M, Dada R, Chan KC, Dada T and Saluja D: Functional genomics of primary congenital glaucoma by pathway analysis and functional characterization of CYP1B1 mutations. Vision Res. 227:1085342025. View Article : Google Scholar : PubMed/NCBI | |
|
Swarup G, Medchalmi S, Ramachandran G and Sayyad Z: Molecular aspects of cytoprotection by Optineurin during stress and disease. Biochim Biophys Acta Mol Cell Res. 1872:1198952025. View Article : Google Scholar : PubMed/NCBI | |
|
Liu D, Webber HC, Bian F, Xu Y, Prakash M, Feng X, Yang M, Yang H, You IJ, Li L, et al: Optineurin-facilitated axonal mitochondria delivery promotes neuroprotection and axon regeneration. Nat Commun. 16:17892025. View Article : Google Scholar : PubMed/NCBI | |
|
Song Rong S, Larson A and Wiggs JL; NEIGHBORHOOD consortium, : ATXN2 loss of function results in glaucoma-related features supporting a role for Ataxin-2 in primary open-angle glaucoma (POAG) pathogenesis. Vision Res. 226:1085082025. View Article : Google Scholar : PubMed/NCBI | |
|
Chacon-Camacho OF, Ordaz-Robles T, Cid-García MA, Yepes-Rodríguez O, Arce-González R, Martínez-Aguilar A and Zenteno JC: A new ocular phenotype combining juvenile glaucoma and Doyne honeycomb retinal dystrophy (Malattia Leventinese) due to a novel EFEMP1 pathogenic variant. Am J Med Genet A. 197:e638692025. View Article : Google Scholar : PubMed/NCBI | |
|
Csidey M, Csorba A, Kormányos K, Náray A, Kéki-Kovács K, Németh O, Knézy K, Bausz M, Szigeti A, Szabó D, et al: Examination of the corneal endothelium in patients with congenital aniridia with a PAX6 mutation using in vivo confocal laser scanning microscopy. Cornea. 44:324–331. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Moazzeni H, Khani M and Elahi E: Insights into the regulatory molecules involved in glaucoma pathogenesis. Am J Med Genet C Semin Med Genet. 184:782–827. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Rejas-González R, Montero-Calle A, Pastora Salvador N, Crespo Carballés MJ, Ausín-González E, Sánchez-Naves J, Pardo Calderón S, Barderas R and Guzman-Aranguez A: Unraveling the nexus of oxidative stress, ocular diseases, and small extracellular vesicles to identify novel glaucoma biomarkers through in-depth proteomics. Redox Biol. 77:1033682024. View Article : Google Scholar : PubMed/NCBI | |
|
Botello-Marabotto M, Martínez-Bisbal MC, Pinazo-Durán MD and Martínez-Máñez R: Tear metabolomics for the diagnosis of primary open angle glaucoma. Talanta. 273:1258262024. View Article : Google Scholar : PubMed/NCBI | |
|
Shin DY, Han JS, Park CK, Lee NY and Jung KI: Parallel analysis of exosomes and cytokines in aqueous humor samples to evaluate biomarkers for glaucoma. Cells. 13:10302024. View Article : Google Scholar : PubMed/NCBI | |
|
Sulak R, Liu X and Smedowski A: The concept of gene therapy for glaucoma: The dream that has not come true yet. Neural Regen Res. 19:92–99. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Sundaresan Y, Yacoub S, Kodati B, Amankwa CE, Raola A and Zode G: Therapeutic applications of CRISPR/Cas9 gene editing technology for the treatment of ocular diseases. FEBS J. 290:5248–5269. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Jain A, Zode G, Kasetti RB, Ran FA, Yan W, Sharma TP, Bugge K, Searby CC, Fingert JH, Zhang F, et al: CRISPR Cas9 based treatment of myocilin associated glaucoma. Proc Natl Acad Sci USA. 114:11199–11204. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Patil SV, Kaipa BR, Ranshing S, Sundaresan Y, Millar JC, Nagarajan B, Kiehlbauch C, Zhang Q, Jain A, Searby CC, et al: Lentiviral mediated delivery of CRISPR/Cas9 reduces intraocular pressure in a mouse model of myocilin glaucoma. Sci Rep. 14:69582024. View Article : Google Scholar : PubMed/NCBI | |
|
Wen D, Ji Y, Li Y, Duan W, Wang Y, Li Z, Tao M and Liu Y: OPTN gene therapy increases autophagy and protects mitochondria in SOD1 G93A expressing transgenic mice and cells. FEBS J. 291:795–813. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Khatib TZ, Osborne A, Yang S, Ali Z, Jia W, Manyakin I, Hall K, Watt R, Widdowson PS and Martin KR: Receptor ligand supplementation via a self-cleaving 2A peptide-based gene therapy promotes CNS axonal transport with functional recovery. Sci Adv. 7:eabd25902021. View Article : Google Scholar : PubMed/NCBI | |
|
Wu J, Bell OH, Copland DA, Young A, Pooley JR, Maswood R, Evans RS, Khaw PT, Ali RR, Dick AD and Chu CJ: Gene therapy for glaucoma by ciliary body aquaporin 1 disruption using CRISPR-Cas9. Mol Ther. 28:820–829. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Wang J, Harris A, Prendes MA, Alshawa L, Gross JC, Wentz SM, Rao AB, Kim NJ, Synder A and Siesky B: Targeting transforming growth factor-beta signaling in primary open-angle glaucoma. J Glaucoma. 26:390–395. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Rayana NP, Sugali CK, Dai J, Peng M, Liu S, Zhang Y, Wan J and Mao W: Using CRISPR Interference as a Therapeutic Approach to Treat TGFβ2-Induced Ocular Hypertension and Glaucoma. Invest Ophthalmol Vis Sci. 62:72021. View Article : Google Scholar : PubMed/NCBI | |
|
Sun F, Park KK, Belin S, Wang D, Lu T, Chen G, Zhang K, Yeung C, Feng G, Yankner BA and He Z: Sustained axon regeneration induced by co-deletion of PTEN and SOCS3. Nature. 480:372–375. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Lee EJ, Han JC, Park DY, Cho J and Kee C: Effect of connective tissue growth factor gene editing using adeno-associated virus-mediated CRISPR-Cas9 on rabbit glaucoma filtering surgery outcomes. Gene Ther. 28:277–286. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Yao M, Zeng Z, Li S, Zou Z, Chen Z, Chen X, Gao Q, Zhao G, Chen A, Li Z, et al: CRISPR-CasRx-mediated disruption of Aqp1/Adrb2/Rock1/Rock2 genes reduces intraocular pressure and retinal ganglion cell damage in mice. Nat Commun. 15:63952024. View Article : Google Scholar : PubMed/NCBI | |
|
Osborne A, Khatib TZ, Songra L, Barber AC, Hall K, Kong GYX, Widdowson PS and Martin KR: Neuroprotection of retinal ganglion cells by a novel gene therapy construct that achieves sustained enhancement of brain-derived neurotrophic factor/tropomyosin-related kinase receptor-B signaling. Cell Death Dis. 9:10072018. View Article : Google Scholar : PubMed/NCBI | |
|
Wójcik-Gryciuk A, Gajewska-Woźniak O, Kordecka K, Boguszewski PM, Waleszczyk W and Skup M: Neuroprotection of retinal ganglion cells with AAV2-BDNF pretreatment restoring normal TrkB receptor protein levels in glaucoma. Int J Mol Sci. 21:62622020. View Article : Google Scholar : PubMed/NCBI | |
|
Fujita K, Nishiguchi KM, Shiga Y and Nakazawa T: Spatially and temporally regulated NRF2 gene therapy using Mcp-1 promoter in retinal ganglion cell injury. Mol Ther Methods Clin Dev. 5:130–141. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Nishijima E, Honda S, Kitamura Y, Namekata K, Kimura A, Guo X, Azuchi Y, Harada C, Murakami A, Matsuda A, et al: Vision protection and robust axon regeneration in glaucoma models by membrane-associated Trk receptors. Mol Ther. 31:810–824. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Phatak NR, Stankowska DL and Krishnamoorthy RR: Bcl-2, Bcl-xL, and p-AKT are involved in neuroprotective effects of transcription factor Brn3b in an ocular hypertension rat model of glaucoma. Mol Vis. 22:1048–1061. 2016.PubMed/NCBI | |
|
Stankowska DL, Minton AZ, Rutledge MA, Mueller BH II, Phatak NR, He S, Ma HY, Forster MJ, Yorio T and Krishnamoorthy RR: Neuroprotective effects of transcription factor Brn3b in an ocular hypertension rat model of glaucoma. Invest Ophthalmol Vis Sci. 56:893–907. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Donahue RJ, Fehrman RL, Gustafson JR and Nickells RW: BCLXL gene therapy moderates neuropathology in the DBA/2J mouse model of inherited glaucoma. Cell Death Dis. 12:7812021. View Article : Google Scholar : PubMed/NCBI | |
|
Lani-Louzada R, Marra C, Dias MS, de Araújo VG, Abreu CA, Ribas VT, Adesse D, Allodi S, Chiodo V, Hauswirth W, et al: Neuroprotective gene therapy by overexpression of the transcription factor MAX in rat models of glaucomatous neurodegeneration. Invest Ophthalmol Vis Sci. 63:52022. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang W, Tang L, Zeng J and Chen B: Adeno-associated virus-mediated SOD gene therapy protects the retinal ganglion cells from chronic intraocular pressure elevation-induced injury via attenuating oxidative stress and improving mitochondrial dysfunction in a rat model. Am J Transl Res. 8:799–810. 2016.PubMed/NCBI | |
|
Luo J, Wang S, Zhou Z and Zhao Y: Ad- and AAV8-mediated ABCA1 gene therapy in a murine model with retinal ischemia/reperfusion injuries. Mol Ther Methods Clin Dev. 20:551–558. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Tan J, Zhang X, Li D, Liu G, Wang Y, Zhang D, Wang X, Tian W, Dong X, Zhou L, et al: scAAV2-mediated C3 Transferase gene therapy in a rat model with retinal ischemia/reperfusion injuries. Mol Ther Methods Clin Dev. 17:894–903. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Q, Zhuang P, Huang H, Li L, Liu L, Webber HC, Dalal R, Siew L, Fligor CM, Chang KC, et al: Mouse gamma-synuclein promoter-mediated gene expression and editing in mammalian retinal ganglion cells. J Neurosci. 40:3896–3914. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Kiyota N, Namekata K, Nishijima E, Guo X, Kimura A, Harada C, Nakazawa T and Harada T: Effects of constitutively active K-Ras on axon regeneration after optic nerve injury. Neurosci Lett. 799:1371242023. View Article : Google Scholar : PubMed/NCBI | |
|
O'Callaghan J, Delaney C, O'Connor M, van Batenburg-Sherwood J, Schicht M, Lütjen-Drecoll E, Hudson N, Ni Dhubhghaill S, Humphries P, Stanley C, et al: Matrix metalloproteinase-3 (MMP-3)-mediated gene therapy for glaucoma. Sci Adv. 9:eadf65372023. View Article : Google Scholar : PubMed/NCBI | |
|
Borras T, Stepankoff M and Danias J: Genes as drugs for glaucoma: Latest advances. Curr Opin Ophthalmol. 35:131–137. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Millington-Ward S, Palfi A, Shortall C, Finnegan LK, Bargroff E, Post IJM, Maguire J, Irnaten MO, Brien C, Kenna PF, et al: AAV-NDI1 therapy provides significant benefit to murine and cellular models of glaucoma. Int J Mol Sci. 25:88762024. View Article : Google Scholar : PubMed/NCBI | |
|
Abbasi M, Gupta VK, Chitranshi N, Gupta VB, Mirzaei M, Dheer Y, Garthwaite L, Zaw T, Parton RG, You Y and Graham SL: Caveolin-1 ablation imparts partial protection against inner retinal injury in experimental glaucoma and reduces apoptotic activation. Mol Neurobiol. 57:3759–3784. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Huang X and Chau Y: Enhanced delivery of siRNA to retinal ganglion cells by intravitreal lipid nanoparticles of positive charge. Mol Pharm. 18:377–385. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang J, Zhang X, Tang Y, Li S and Chen J: Progress on ocular siRNA gene-silencing therapy and drug delivery systems. Fundam Clin Pharmacol. 35:4–24. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Kong AW and Ou Y: The catcher in the eye: Stem cells as a therapeutic for glaucoma. Ophthalmol Glaucoma. 6:1–3. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Du Y, Yun H, Yang E and Schuman JS: Stem cells from trabecular meshwork home to TM tissue in vivo. Invest Ophthalmol Vis Sci. 54:1450–1459. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu W, Gramlich OW, Laboissonniere L, Jain A, Sheffield VC, Trimarchi JM, Tucker BA and Kuehn MH: Transplantation of iPSC-derived TM cells rescues glaucoma phenotypes in vivo. Proc Natl Acad Sci USA. 113:E3492–E3500. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Xiong S, Kumar A, Tian S, Taher EE, Yang E, Kinchington PR, Xia X and Du Y: Stem cell transplantation rescued a primary open-angle glaucoma mouse model. Elife. 10:e636772021. View Article : Google Scholar : PubMed/NCBI | |
|
Coulon SJ, Schuman JS, Du Y, Bahrani Fard MR, Ethier CR and Stamer WD: A novel glaucoma approach: Stem cell regeneration of the trabecular meshwork. Prog Retin Eye Res. 90:1010632022. View Article : Google Scholar : PubMed/NCBI | |
|
Xi G, Feng P, Zhang X, Wu S, Zhang J, Wang X, Xiang A, Xu W, Wang N and Zhu W: iPSC-derived cells stimulate ABCG2+/NES+ endogenous trabecular meshwork cell proliferation and tissue regeneration. Cell Prolif. 57:e136112024. View Article : Google Scholar : PubMed/NCBI | |
|
Hu BY, Xin M, Chen M, Yu P and Zeng LZ: Mesenchymal stem cells for repairing glaucomatous optic nerve. Int J Ophthalmol. 17:748–760. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Ji S, Peng Y, Liu J, Xu P and Tang S: Human adipose tissue-derived stem cell extracellular vesicles attenuate ocular hypertension-induced retinal ganglion cell damage by inhibiting microglia-TLR4/MAPK/NF-kB proinflammatory cascade signaling. Acta Neuropathol Commun. 12:442024. View Article : Google Scholar : PubMed/NCBI | |
|
Yu F, Wang Y, Huang CQ, Lin SJ, Gao RX and Wu RY: Neuroprotective effect of mesenchymal stem cell-derived extracellular vesicles on optic nerve injury in chronic ocular hypertension. Neural Regen Res. 18:2301–2306. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
da Silva-Junior AJ, Mesentier-Louro LA, Nascimento-Dos-Santos G, Teixeira-Pinheiro LC, Vasques JF, Chimeli-Ormonde L, Bodart-Santos V, de Carvalho LRP, Santiago MF and Mendez-Otero R: Human mesenchymal stem cell therapy promotes retinal ganglion cell survival and target reconnection after optic nerve crush in adult rats. Stem Cell Res Ther. 12:692021. View Article : Google Scholar : PubMed/NCBI | |
|
Soucy JR, Aguzzi EA, Cho J, Gilhooley MJ, Keuthan C, Luo Z, Monavarfeshani A, Saleem MA, Wang XW, Wohlschlegel J, et al: Retinal ganglion cell repopulation for vision restoration in optic neuropathy: A roadmap from the RReSTORe Consortium. Mol Neurodegener. 18:642023. View Article : Google Scholar : PubMed/NCBI | |
|
Lei Q, Zhang R, Yuan F and Xiang M: Integration and differentiation of transplanted Human iPSC-Derived retinal ganglion cell precursors in murine retinas. Int J Mol Sci. 25:129472024. View Article : Google Scholar : PubMed/NCBI | |
|
Chaibakhsh S, Azimi F, Shoae-Hassani A, Niknam P, Ghamari A, Dehghan S and Nilforushan N: Evaluating the impact of mesenchymal stem cell therapy on visual acuity and retinal nerve fiber layer thickness in optic neuropathy patients: A comprehensive systematic review and meta-analysis. BMC Ophthalmol. 24:3162024. View Article : Google Scholar : PubMed/NCBI | |
|
Niu Y, Ji J, Yao K and Fu Q: Regenerative treatment of ophthalmic diseases with stem cells: Principles, progress, and challenges. Adv Ophthalmol Pract Res. 4:52–64. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Lee DH, Han JW, Park H, Hong SJ, Kim CS, Kim YS, Lee IS and Kim GJ: Achyranthis radix extract enhances antioxidant effect of placenta-derived mesenchymal stem cell on injured human ocular cells. Cells. 13:12292024. View Article : Google Scholar : PubMed/NCBI | |
|
Lee DH, Han JW, Park H, Hong SJ, Kim CS, Kim YS, Lee IS and Kim GJ: Insulin restores retinal ganglion cell functional connectivity and promotes visual recovery in glaucoma. Sci Adv. 10:eadl57222024. View Article : Google Scholar : PubMed/NCBI | |
|
Patel PD, Kodati B and Clark AF: Role of glucocorticoids and glucocorticoid receptors in glaucoma pathogenesis. Cells. 12:24522023. View Article : Google Scholar : PubMed/NCBI | |
|
Sgambellone S, Khanfar MA, Marri S, Villano S, Nardini P, Frank A, Reiner-Link D, Stark H and Lucarini L: Histamine H3 receptor antagonist/nitric oxide donors as novel promising therapeutic hybrid-tools for glaucoma and retinal neuroprotection. Biomed Pharmacother. 180:1174542024. View Article : Google Scholar : PubMed/NCBI | |
|
Cimaglia G, Tribble JR, Votruba M, Williams PA and Morgan JE: Oral nicotinamide provides robust, dose-dependent structural and metabolic neuroprotection of retinal ganglion cells in experimental glaucoma. Acta Neuropathol Commun. 12:1372024. View Article : Google Scholar : PubMed/NCBI | |
|
Kralj T, Kokot A, Zlatar M, Masnec S, Kasnik Kovac K, Milkovic Perisa M, Batelja Vuletic L, Giljanovic A, Strbe S, Sikiric S, et al: Stable gastric pentadecapeptide BPC 157 therapy of rat glaucoma. Biomedicines. 10:892021. View Article : Google Scholar : PubMed/NCBI | |
|
Geva M, Gershoni-Emek N, Naia L, Ly P, Mota S, Rego AC, Hayden MR and Levin LA: Neuroprotection of retinal ganglion cells by the sigma-1 receptor agonist pridopidine in models of experimental glaucoma. Sci Rep. 11:219752021. View Article : Google Scholar : PubMed/NCBI | |
|
Iqbal H, Razzaq A, Zhou D, Lou J, Xiao R, Lin F and Liang YL: Nanomedicine in glaucoma treatment: Current challenges and future perspectives. Mater Today Bio. 28:1012292024. View Article : Google Scholar : PubMed/NCBI | |
|
Ciociola EC, Fernandez E, Kaufmann M and Klifto MR: Future directions of glaucoma treatment: Emerging gene, neuroprotection, nanomedicine, stem cell, and vascular therapies. Curr Opin Ophthalmol. 35:89–96. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Pei K, Georgi M, Hill D, Lam CFJ, Wei W and Cordeiro MF: Review: Neuroprotective nanocarriers in glaucoma. Pharmaceuticals (Basel). 17:11902024. View Article : Google Scholar : PubMed/NCBI | |
|
Satyanarayana SD, Abu Lila AS, Moin A, Moglad EH, Khafagy ES, Alotaibi HF, Obaidullah AJ and Charyulu RN: Ocular delivery of Bimatoprost-loaded solid lipid nanoparticles for effective management of glaucoma. Pharmaceuticals (Basel). 16:10012023. View Article : Google Scholar : PubMed/NCBI | |
|
Esteban-Pérez S, Andrés-Guerrero V, López-Cano JJ, Molina-Martínez I, Herrero-Vanrell R and Bravo-Osuna I: Gelatin Nanoparticles-HPMC hybrid system for effective ocular topical administration of antihypertensive agents. Pharmaceutics. 12:3062020. View Article : Google Scholar : PubMed/NCBI | |
|
Sánchez-López E, Egea MA, Davis BM, Guo L, Espina M, Silva AM, Calpena AC, Souto EMB, Ravindran N, Ettcheto M, et al: Memantine-Loaded PEGylated biodegradable nanoparticles for the treatment of glaucoma. Small. 14:2018.doi: 10.1002/smll.201701808. View Article : Google Scholar : PubMed/NCBI | |
|
Emad Eldeeb A, Salah S and Ghorab M: Proniosomal gel-derived niosomes: An approach to sustain and improve the ocular delivery of brimonidine tartrate; formulation, in-vitro characterization, and in-vivo pharmacodynamic study. Drug Deliv. 26:509–521. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
He M, Rong R, Ji D and Xia X: From bench to bed: The current genome editing therapies for glaucoma. Front Cell Dev Biol. 10:8799572022. View Article : Google Scholar : PubMed/NCBI | |
|
Danford ID, Verkuil LD, Choi DJ, Collins DW, Gudiseva HV, Uyhazi KE, Lau MK, Kanu LN, Grant GR, Chavali VRM and O'Brien JM: Characterizing the ‘POAGome’: A bioinformatics-driven approach to primary open-angle glaucoma. Prog Retin Eye Res. 58:89–114. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Zeppieri M, Visalli F, Musa M, Avitabile A, Giglio R, Tognetto D, Gagliano C, D'Esposito F and Cappellani F: Beyond the eye: Glaucoma and the Brain. Brain Sci. 15:9342025. View Article : Google Scholar : PubMed/NCBI | |
|
MULTI Consortium, . Boquet-Pujadas A, Anagnostakis F, Duggan MR, Joynes CM, Toga AW, Yang Z, Walker KA, Davatzikos C and Wen J: Brain-heart-eye axis revealed by multi-organ imaging genetics and proteomics. Nat Biomed Eng. September 30–2025.(Epub ahead of print). |