Next Article in Journal
Is There a Role for Machine Learning in Liquid Biopsy for Brain Tumors? A Systematic Review
Next Article in Special Issue
Tako-Tsubo Syndrome in Amyotrophic Lateral Sclerosis: Single-Center Case Series and Brief Literature Review
Previous Article in Journal
Metabolic Processes and Biological Macromolecules Defined the Positive Effects of Protein-Rich Biostimulants on Sugar Beet Plant Development
Previous Article in Special Issue
After Ischemic Stroke, Minocycline Promotes a Protective Response in Neurons via the RNA-Binding Protein HuR, with a Positive Impact on Motor Performance
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Inhibition of Galectins and the P2X7 Purinergic Receptor as a Therapeutic Approach in the Neurovascular Inflammation of Diabetic Retinopathy

by
Caterina Claudia Lepre
1,†,
Marina Russo
2,†,
Maria Consiglia Trotta
2,
Francesco Petrillo
3,
Fabiana Anna D’Agostino
4,
Gennaro Gaudino
5,
Giovanbattista D’Amico
6,
Maria Rosaria Campitiello
7,
Erminia Crisci
4,
Maddalena Nicoletti
4,
Carlo Gesualdo
4,*,
Francesca Simonelli
4,
Michele D’Amico
2,‡,
Anca Hermenean
1,‡ and
Settimio Rossi
4,‡
1
“Aurel Ardelean” Institute of Life Sciences, Vasile Goldis Western University of Arad, 310144 Arad, Romania
2
Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
3
Ph.D. Course in Translational Medicine, Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
4
Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
5
School of Anesthesia and Intensive Care, University of Foggia, 71122 Foggia, Italy
6
School of Geriatrics, University of Studies of L’Aquila, 67010 L’Aquila, Italy
7
Department of Obstetrics and Gynecology and Physiopathology of Human Reproduction, ASL Salerno, 84124 Salerno, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(11), 9721; https://doi.org/10.3390/ijms24119721
Submission received: 26 April 2023 / Revised: 30 May 2023 / Accepted: 1 June 2023 / Published: 3 June 2023

Abstract

:
Diabetic retinopathy (DR) is the most frequent microvascular retinal complication of diabetic patients, contributing to loss of vision. Recently, retinal neuroinflammation and neurodegeneration have emerged as key players in DR progression, and therefore, this review examines the neuroinflammatory molecular basis of DR. We focus on four important aspects of retinal neuroinflammation: (i) the exacerbation of endoplasmic reticulum (ER) stress; (ii) the activation of the NLRP3 inflammasome; (iii) the role of galectins; and (iv) the activation of purinergic 2X7 receptor (P2X7R). Moreover, this review proposes the selective inhibition of galectins and the P2X7R as a potential pharmacological approach to prevent the progression of DR.

1. Diabetic Retinopathy (DR): An Overview

Diabetes is known as a chronic disease with multiple complications [1]. One of the most harmful and common complications is diabetic retinopathy (DR) [2,3]. This is a leading cause of blindness worldwide [4], with 103.1 million adults affected by DR in 2020 and a predicted 160.5 million affected in 2045 [5]. Moreover, DR individuals often develop a visual impairment, leading to a poor quality of life for the patient and impacting on the health care system in terms of direct and indirect costs. Indeed, in 2020, there were 28.5 million patients affected by vision-threatening DR, and it is estimated this number will be 44.8 million by 2045 [5].
Historically, DR has been considered as a microangiopathy, and is characterized by increased retinal vascular permeability and endothelial damage [2,6,7]. In particular, the hallmarks of DR onset include the thickening of the basement membrane, the loss of pericytes, the alteration of tight junctions and the endothelial barrier, the formation of microaneurysms and the uncontrolled proliferation of endothelial cells [8,9,10]. These events later translate into capillary occlusion that anticipates retinal ischemia, which is, in turn, associated with vascular endothelial growth factor (VEGF) overexpression and neovascularization [9,11].
Accordingly, DR can be clinically classified as either non-proliferative diabetic retinopathy (NPDR) or proliferative diabetic retinopathy (PDR) [8], which are usually diagnosed by fluorescein angiography (FA) or mydriatic fundus camera examinations [12]. However, other imaging methods such as optical coherence tomography (OCT), Doppler OCT, OCT angiography and retinal imaging of the fundus can be used [12]. While NPDR is the early stage of the disease (which can be further classified as mild, moderate and severe), PDR represents the advanced stage of retinopathy. NPDR is characterized by progressive changes in retinal capillary microcirculation, resulting in microaneurysms, blood–retinal barrier (BRB) breakdown and intraretinal exudates [13]. Specifically, mild NPDR is diagnosed by the presence of few microaneurysms; moderate NPDR is characterized by microaneurysms, venous beading or intraretinal hemorrhages; severe NPDR is defined by the presence of hemorrhages or microaneurysms or both, venous beading or prominent intraretinal microvascular abnormalities (IRMAs) [14]. In contrast, PDR is characterized by the presence of numerous ischemic retinal areas, VEGF overproduction and neoangiogenesis [15,16], and is diagnosed by neovascularization of the disc retina and iris or by tractional retinal detachment or vitreous hemorrhage [14]. Indeed, PDR can evolve into more severe stages such as diabetic macular edema (DME), neovascular glaucoma and, in rare cases, retinal detachment [9,13,15,17].
While the glycemic, lipidic and blood pressure controls are needed for DR primary prevention, the current surgical and pharmacological options for DR management include pan-retinal laser photocoagulation in severe NPDR and PDR patients, focal retinal laser photocoagulation in patients with diabetic DME, anti-VEGF intravitreal injections in PDR and DME patients, and steroid intravitreal injections for non-responder DR patients or naïve patients when appropriate [18]. In particular, the early use of laser photocoagulation or intravitreal injections of glucocorticoids or anti-angiogenics drugs has proven effective in preventing the onset of blindness [19], while anti-VEGF intravitreal injections are considered the actual gold standard for DR therapy. However, DR management is still challenging, with the 40% of inadequately treated NPDR cases evolving to PDR within 12 months [13,20]. Therefore, several studies have been performed to identify new molecular pathways or circulating biomarkers as new potential pharmacological tools in DR prevention and/or treatment. Among these, an important role as predictive and therapeutic targets in DR progression has been proposed for melanocortin receptors, retinol binding protein 3 (RBP3), angiopoietin-like 3 (ANGPTL3), microRNAs, extracellular vesicles, metabolites as 12-hydroxyeicosatetraenoic acid and 2-piperidone, along with several mediators involved in the resolution of inflammation [21,22,23,24,25,26,27].
Currently, intraretinal inflammation and neurodegeneration have emerged as key processes in DR progression [16,28]. Indeed, along with microvascular damage, inflammatory and neurodegenerative processes contribute to alterations in the “retinal neurovascular unit”, a functional unit composed of endothelial cells, glial cells and neurons [29]. In this regard, early retinal neurovascular alterations have been recently associated with specific serum microglial biomarkers such as ionized calcium-binding adapter molecule 1 (Iba-1), glucose transporter 5 (GLUT5), and translocator protein (TSPO) [27].
In this review, we focus on the importance of endoplasmic reticulum stress (ERS) and the consequent activation of the nucleotide-binding oligomerization domain-, leucine-rich repeat- and pyrin domain-containing 3 (NLRP3) inflammasome during retinal neuroinflammation. In particular, ERS modulation by specific proteins binding beta-galactoside residues on glycated proteins known as galectins is detailed. The galectin family consists of 15 identified members, all of which contain conserved carbohydrate-recognition domains (CRDs) of about 130 amino acids [30]. Based on the CRD organization of the polypeptide monomer, the galectins have been classified into three types: proto-type, chimera-type, and tandem-repeat-type [31]. The prototypical galectins have one CRD (galectin-1, -2, -5, -7, -10, -11, -13, -14 and -15); the tandem-repeat-type galectins contain two homologous CRDs in a single polypeptide chain, separated by a linker of up to 70 amino acids (galectin-4, -6, -8, -9 and -12); galectin-3 contains a non-lectin N-terminal region (about 120 amino acids) connected to a CRD [32]. Outside the cell, galectins bind to cell-surface and extracellular matrix glycans; however, galectins are also detectable in the cytosol and nucleus. They may influence cellular signaling pathways, such as cell migration, autophagy, immune response, and inflammation [32].
Moreover, the importance of purinergic 2X7 receptor (P2X7R) in NLRP3 activation underlying neuroinflammation is analyzed, along with the options for their selective inhibition in several clinical settings. Specifically, the state of the art on galectins and P2X7R inhibitors in DR is described, along with their potential use as pharmacological tools to prevent DR progression.

2. The Role of Neuroinflammation in DR

The “retinal neurovascular unit” includes specific retinal glia elements, such as microglial cells and macroglia (Müller cells and astrocytes) [28,33,34]. These express marked ERS [35,36,37] and subsequent activation of NLRP3 inflammasome [38], as a consequence of advanced glycation end products (AGEs) and reactive oxygen species (ROS) formation, strongly induced by chronic hyperglycemia [39]. In this regard, increases in many pro-inflammatory cytokines and chemokines, such as interleukin 1 beta (IL-1β), interleukin 6 (IL-6), interleukin 8 (IL-8), tumor necrosis factor alpha (TNF-α) and monocyte chemoattractant protein-1 (MCP-1), have been found in serum and in ocular samples (vitreous and aqueous humor) of DR patients. The accumulation of these cytokines is mainly mediated by cells from the macroglia and microglia [40].
Microglial cells represent the first defense line in neural retina [41,42,43]. Specifically, the retinal inner layers are characterized by the presence of ramified microglial cells [44,45] controlling the regulation of retinal growth, blood vessel formation and the retinal endothelial cell–glia–neuron interactions [46,47,48,49,50]. When activated by hyperglycemia, retinal hypoxia, ischemia and ER stress [51,52,53,54], these cells are classified as M1 macrophages and release pro-inflammatory mediators such as IL-6 and IL-12, TNF-α and interferon gamma (IFN-γ), known inducers of inflammation, apoptosis and neurotoxicity [55,56,57]. Microglial cells can be alternatively classified as M2 macrophages and release anti-inflammatory mediators, thus exerting anti-inflammatory effects and neuroprotection [42,55,58].
Retinal microglia polarization directly influences Müller cells and macroglia cells, spanning the entire retinal width and connecting neurons and vascular components [59,60]. In healthy retina, Müller cells contribute to the BRB integrity, modulate retinal blood flow and regulate the production of ions, neurotransmitters and metabolites that favor retinal blood vessel dilatation or constriction [61]. As a consequence of hyperglycemia-induced ER stress [35,39], Müller cell activation leads to the release of pro-inflammatory cytokines and chemokines [62] which favor leucocytes recruitment [63], a phenomenon termed gliosis. Moreover, Müller cell inflammatory actions across the retinal laminal structure seems to favor microglia attraction and adhesion by modulating retinal injury response [60].
Lastly, astrocytes are the most abundant central nervous system (CNS) macroglial cell type, forming the inner retinal BRB [64]. These are prevalent in the retinal ganglion cell layer (GCL) and nerve fiber layer (NFL) and modulate the neuronal metabolism, neurotransmission, and the neurorepair process [65,66,67]. Astrocytes can exert either pro- or anti-inflammatory actions, depending on the microenvironment in which they are located and the received signal [68].

3. Mechanisms of DR Neuroinflammation

3.1. Endoplasmic Reticulum (ER) Stress

In the diabetic retina, hyperglycemia leads to elevated ROS production by various mechanisms, including the reduction of antioxidant enzymes [69] and the activation of hypoxia-inducible transcription factor 1 (HIF-1) [70,71].
During DR, the hyperglycemia-induced ER stress (ERS) affects all the components of the “neurovascular unit” and leads to the apoptosis of both vascular and neuronal cells [72,73]. In particular, the abnormal ERS causes the apoptosis of retinal pericytes [74] with BRB impairment and retinal neuroinflammation [75,76]. Among the ERS inducers, key roles are played by glycative stress and activation of the unfolded protein response (UPR) that are stimulated by AGEs and ROS formation, respectively [77]. Glycative stress is used to monitor the status of protein folding and ensure that only properly folded proteins are trafficked to the Golgi [78,79]. UPR is an adaptive signaling pathway which tends to enhance the ER capacity for protein folding and modification to restore an efficient protein-folding environment [77]. The main UPR actors are the inositol-requiring kinase 1 (IRE1), the protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) and the activating transcription factor 6 (ATF6) [77,80]. When the accumulation of protein aggregates exceeds the ER load capacity, the activation of UPR exacerbates ERS [81] (Figure 1).
ERS-induced microglial activation occurs during the different DR stages [27,47]. Indeed, activated microglial cells have been observed in the retinal plexiform layers (RPL) of NPDR patients and around the ischemic areas in PDR patients [82]. Notably, changes in M1/M2 microglia polarization can lead to visual loss by increasing the apoptosis of retinal neurons with consequent retinal NFL thinning [83].
Conversely, ERS-induced Müller cell activation seems to precede the DR vascular alterations [84], with NPDR patients showing Müller cell gliosis and swelling in the retinal inner nuclear layer (INL) and outer plexiform layer (OPL) [85], although PDR neuronal damage and DME cyst formation also characterize gliosis [86]. Lastly, retinal astrocytes are responsible for a sustained production of the pro-inflammatory IL-1β in response to hyperglycemia-induced ERS [87].

3.2. NLRP3 Inflammasome

ROS-induced ER stress leads to the activation of the NLRP3 inflammasome, a multiproteic complex found as monomer when inactive [88,89,90,91] (Figure 1). In particular, ERS exacerbation leads to activation of the NLRP3 inflammasome [92] through the binding of thioredoxin-interacting protein (TXNIP) to NLRP3 [93,94].
The NLRP3 complex consists of a sensor component (NLRP3 protein), an effector component (caspase-1) and, in some cases, an adapter protein (known as apoptosis-associated speck-like protein—ASC) linking the sensor and the effector [93,95,96]. The NLRP3 sensor is characterized by three different domains known as cytosolic pattern recognition receptors (PRRs): an amino-terminal pyrin domain (PYD), a central nucleotide-binding and oligomerization domain (NOD domain), and a C-terminal leucine-rich repeat (LRR) domain. Through these, NLRP3 is known to sense an extremely broad range of both exogenous and endogenous stimuli, known as pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs), including changes in the ion gradient across the cell, cellular stress mechanisms such as higher ROS production after lysosomal rupture, mitochondrial stress and ERS in response to the accumulation of misfolded proteins [97,98,99,100]. When activated, the pyrin domain of NLRP3 interacts with the ASC pyrin domain to initiate inflammasome assembly [100,101] (Figure 2).
The pyrene domain connects to the NLRP3 pyrene domain through an oligomerization process; in addition, the adaptor protein ASC brings pro-caspase 1 monomers near to each other through the CARD domain by inducing a proximity-mediated caspase-1 autoactivation [101]. Active caspase-1 can induce the release of pro-inflammatory cytokines, such as IL-1β and interleukin 18 (IL-18), favoring both apoptosis and pyroptosis, a cell death process during which some pro-inflammatory cytokines are released to attract and activate immune cells [93,95,102,103]. Moreover, active caspase-1 is also necessary for the proteolytic cleavage of Gasdermin D (GSDMD) and the consequent release of the GSDMD N-terminal fragment, which is necessary to mediate pyroptosis [104].
Similarly, NLRP3 is pivotal in DR progression [89,91,105,106,107]. Indeed, retinal NLRP3 levels were increased in serum and vitreous samples from patients affected by PDR compared with NPDR patients [40,106,107].
NLRP3 activation is shown by retinal epithelial [108,109,110,111] and endothelial cells [112,113,114]. However, it is evident also in retinal neuronal layers, such as retinal ganglion cells (RGC) in mouse models of retinal degeneration and in photoreceptors from a genetic mouse DR model [115,116,117]. Moreover, NLRP3 levels were increased in retinal microglial cells exposed to high glucose [118] and in retinal macroglia from a mouse model of ocular hypertension [119].

4. New Actors in Neuroinflammation

4.1. Galectins and ERS

Overall, galectin dysregulation has been linked to different pathological conditions, such as fibrosis, heart disease, cancer, and diabetes [32]. In this regard, Gal-1 has been associated with type 2 diabetes [120], while Gal-3 has been correlated to both type 1 and type 2 diabetes [121,122,123], with a suggested role for this galectin in mediating the chronic inflammation underlying the progression from prediabetes to the diabetic stage [124]. Moreover, in diabetic patients, diabetic nephropathy, diabetic foot, diabetic microvascular complications, and diabetic cardiomyopathy have been all related to changes in Gal-3 serum levels [125]. In particular, Galectin 1 (Gal-1) and Galectin 3 (Gal-3) seem to initiate the inflammatory response by acting as chemotactic agents towards the inflammatory site for the neutrophils, facilitating their binding to the endothelium and their trafficking through the extracellular matrix [126].
Galectins, induced by AGEs and considered as receptors for AGEs (RAGE), have emerged as ERS regulators. For example, the elevation of Galectin 9 (Gal-9) has been linked to inflammatory processes in both type 1 and type 2 diabetes [127,128]. However, an additional protective role against ERS has been described for Gal-9 [129], along with its importance in the facilitation of NLRP3 autophagic degradation [130].
In addition, Gal-1 and Gal-3 have also emerged as ERS regulators (Figure 1). Specifically, Gal-1 is upregulated in hypoxic microenvironments [131], resulting in increased ROS production and activation of the nuclear factor kappa B (NF-κB) signaling pathway [132]. This protein is considered a key regulator of endothelial cells functions and shows potent proangiogenic properties [133,134].
Gal-3 is localized at the ER-mitochondria interface and regulates the UPR [135]. It is involved in several processes underlying retinopathies, such as oxidative stress, proliferation, phagocytosis, apoptosis, oxidative stress, and angiogenesis [136]. Interestingly, Gal-3 may favor adaptive UPR following ERS by acting as both a pro- and anti-apoptotic regulator [135]. Tian and colleagues demonstrated that Gal-3 is also implicated in the process of activating the NLRP3 inflammasome, discovering a direct link of the N-terminal domain of Gal-3 to the NLRP3 inflammasome [137]. Several studies have shown alterations in the concentration of galectins in the brain and blood of patients with neurodegenerative diseases compared with healthy subjects. In particular, Gal-1 was found in neurofilamentous lesions of patients affected by amyotrophic lateral sclerosis (ALS) [138], while patients with multiple sclerosis showed a higher concentration of Galectin 4 (Gal-4) in chronic lesions of the brain [139]. However, Gal-3 can be considered as an indicator of prognosis, mortality or remission in neurodegenerative diseases [140] since its expression was found to be increased in patients with ALS, Alzheimer’s and Parkinson’s diseases (AD and PD, respectively) [141].
This could be firstly due to the high affinity showed by galectins for β-galactosides [142,143], which are involved in neuroprotection and neuroinflammation [144,145]. Moreover, galectins are expressed by glial cells. In particular, Gal-1 expression was reported in astrocytes and Müller cells, participating in the protection from axonal damage as it mediates T-cell activation and differentiation, whereas Gal-3 expression was mainly observed in M1 microglial cells and was associated with microglial activation in cell damage, ischemia, and encephalitis [30,140,146]. Particularly astrocyte-derived Gal-1 seems to play a key role in the modulation of inflammation, phagocytosis, axon growth and gliosis after spinal cord injury [147,148]. It was also found to be important in the modulation of microglia polarization and neuromodulation in a multiple sclerosis model [149], as well as modulation of axonal degeneration in a transgenic mouse model of ALS [150]. Regarding Gal-3, in primary rat microglia and macroglia, Gal-3 exposure increases the expression of TNF-α, IL-1β, IL-6 and INF-γ [151]. Accordingly, in a mouse model of Huntington’s disease, Gal-3 increased the microglial expression of IL-1β [152]. Moreover, the suppression of Gal-3 in AD mice improved their cognitive performance, reducing amyloid plaques [153], as well as cognitive impairment, neuroinflammation and oxidative stress associated with diabetes in rats caused by modified citrus pectin (MCP) [154].

4.2. Purinergic 2X7 Receptor (P2X7R) and NLRP3 Inflammasome

P2X7R is a trimeric ion channel that belongs to the P2X family of ionotropic receptors preferably permeable to sodium, potassium and calcium, which are exclusively triggered by extracellular adenosine triphosphate (ATP) [96]. P2X receptors are widespread in different tissues and exert various functions: in some smooth muscle cells, activated P2X receptors mediate depolarization and contraction; in the CNS, activated P2X receptors allow calcium to enter neurons, leading to slower neuromodulatory responses; and in the cells of the immune system, activated P2X receptors trigger the release of pro-inflammatory cytokines [155].
P2X7R presents five domains: one extracellular domain; two transmembrane domains (classified as transmembrane 1 and 2); and two intracellular domains (N- and C-terminus), which form homotrimeric receptors after their activation [156,157]. Its signaling is important for regulating both the innate and adaptive immune response [156] and also inflammatory processes [96].
P2X7R is highly expressed in different cell types and tissues, such as retinal neural cells and the retinal vasculature [158], but also in immune cells [159]. In particular, in the human retina, P2X7R is localized in INL, OPL and GCL and is expressed by human retinal Müller cells, the native retinal pigment epithelium (RPE) and adult retinal pigment epithelial cell line-19 (ARPE-19) [160].
P2X7R has a key role in linking inflammation with purinergic signaling: this receptor and the NLRP3 inflammasome interact at distinct cytoplasmic sites, where changes in P2X7R-dependent ion concentrations (specifically a K+ efflux) occur [156,161,162], probably mediated by the cytoplasmic kinase never-in-mitosis A- related kinases (NEK) [156] (Figure 2). In particular, during NLRP3 inflammasome activation, the K+ efflux seems to be mediated by P2X7R following an increase in extracellular ATP [163]. This allows the release of pro-inflammatory cytokines associated with NLRP3 inflammasome activation [156]. In this regard, P2X7R is upregulated [162] in pathological conditions as a consequence of increased extracellular microglial ATP concentration [164]. This is indicative of the pro-inflammatory M1 phenotype microglia activation [164]. P2X7R is widely expressed in the CNS regions and is associated with neuroinflammation and neurodegeneration [164,165,166]. Therefore, P2X7R has been extensively investigated in order to develop small molecules that could act as potent blockers of the receptor [155].
Indeed, P2X7R activation in glial cells overall results in the release of the pro-inflammatory cytokines, thereby triggering or potentiating neuroinflammation [167]. This contributes to neurodegeneration by inducing microglia-mediated neuronal death [168], glutamate-mediated excitotoxicity and NLRP3 inflammasome activation, with the consequent release of IL-1β and IL-18 [96,164,169].
Several studies evidenced that P2X7R expression is upregulated in the activated microglia of AD patients and is concentrated in amyloid plaques [170,171]. Similarly, P2X7R was upregulated in the hippocampus of an AD animal model [172,173]. Furthermore, preclinical evidence suggests that P2X7R may have a role in the pathogenesis of Huntington’s disease [174]. P2X7R also has a possible involvement in PD by mediating activation of the NLRP3 inflammasome [175]. This aspect is under investigation in an observational prospective study at the University of Pisa (Italy). This study is evaluating changes in P2X7R levels and their association with changes in NLRP3 inflammasome levels in patients with newly diagnosed PD or AD receiving routine treatment in comparison with an age- and gender-matched group [176]. P2X7R was also recently identified as a key contributor to cognitive impairment in a mouse model of migraine; activation of the NLRP3 inflammasome and P2X7R upregulation led to gliosis, neuronal loss and neuroinflammation [177]. Astrogliosis related to P2X7R has also been reported in a rat model of autoimmune encephalomyelitis [178].

5. Inhibition of Galectins and P2X7R and Its Potential Therapeutic Application for DR Neuroinflammation

It is well known that galectins and P2X7R are two key mediators in DR pathology and progression [84,157,179,180,181,182,183,184,185,186,187,188,189]. Therefore, these neuroinflammatory actors represent two potential pharmacological targets to prevent the onset of DR by their selective inhibition.
In this regard, different compounds inhibiting galectins have been evaluated in clinical trials. In particular, although Gal-1 inhibitor OTX0008 has been tested in patients with advanced solid tumors [190], Gal-3 inhibition has gained a wider application in several ongoing clinical trials. Indeed, both the Gal-3 inhibitors GR-MD-02 and GB1211 are in evaluation for liver fibrosis in non-alcoholic steatohepatitis (NASH) [143,191,192]. Administration of GB1211 alone has also been investigated for hepatic impairment [193], and its combination with atezolizumab (a monoclonal antibody targeting the Programmed Death Ligand-1) has been considered in non-small cell lung cancer patients [194]. Furthermore, the co-administration of GR-MD-02 with ipilimumab (a monoclonal antibody targeting the Cytotoxic T-Lymphocyte Antigen 4) or pembrolizumab (a monoclonal human antibody targeting the Programmed Cell Death protein 1) is undergoing evaluation in patients with metastatic melanoma [195] and in patients with advanced melanoma, non-small cell lung cancer and head and neck squamous cell cancer, respectively [196,197].
Gal-3 inhibition by GB0139 and TD139 compounds is under evaluation in idiopathic pulmonary fibrosis (IPF) [143,198,199], while MCP as a Gal-3 inhibitor has been considered for both hypertension and osteoarthritis [200,201].
Regarding P2X7R, different compounds have been tested on healthy volunteers to assess their safety and tolerability, bioavailability, pharmacokinetics, and pharmacodynamics as P2X7R antagonists. These include GSK1482160 [202], AZD9056 [203,204], and ce-224,535 [205,206]. In particular, ce-224,535 and AZD9056 have both been in evaluation for patients with rheumatoid arthritis (RA) [207,208], with ce-224,535 considered also for patients with knee osteoarthritis pain [209].
To date, none of these inhibitors has been considered for DR patients. However, several pre-clinical studies evidenced the potential role of galectin and P2X7R inhibition strategies in modulating diabetic retinal damage [179,181,183,184].

5.1. Galectin Inhibition in DR

Gal-1 and Gal-3 have been recently associated with the insurgence and progression of DR pathology (Table 1).
In particular, Gal-1 levels were increased in the vitreous fluid and epiretinal fibrovascular membrane of PDR patients compared with non-diabetic controls [179]. This was probably due to the higher Gal-1 secretion induced in retinal Müller cells and astrocytes by hyperglycemic conditions [136]. The upregulation of Gal-1 protein levels in vitreous samples rose substantially with DR progression, being present from the pre-ischemic inflammatory stage [186]. Increased Gal-1 plasma levels were also detected in PDR patients compared with non-diabetic controls, along with a Gal-1 correlation with AGEs and IL-1β [185]. In line with this evidence, Gal-1 was also found to be upregulated in neovascular ocular tissues surgically excised from PDR patients, where it exhibited a co-localization with Vascular Endothelial Growth Factor Receptor 2 (VEGFR2) [186]. While in this study, Gal-1 exhibited no correlation with VEGFA, in a mouse model of oxygen induced-retinopathy (OIR), Gal-1 was shown to mediate vascular alterations concomitantly to VEGF up-regulation [187].
Gal-1 also seems to be involved in the regulation of the RPE that forms the BRB. Indeed, ARPE-19 cells exposed to high glucose exhibited high Gal-1 levels, which was associated with epithelial fibrosis and epithelial-mesenchymal transition [181]. Both processes were reduced by selective Gal-1 blocking with OTX008 [181].
Although Gal-1 has been associated with neuronal alterations in a mouse OIR model, and particularly with neuroglial injuries at post-natal day 26 [187], retinal neuroinflammatory actions have been specifically shown for Gal-3, whose pro-inflammatory role in diabetic optic neuropathy occurs through ROS-induced ERS [84]. Accordingly, within the neuronal retina, Gal-3 knockout resulted in reduced microglial activation and led to better preservation of RGC, nerve fibers, axons and cell bodies in diabetic mice [180]. This evidence was recently confirmed by Mendonça and colleagues [188], who showed that Gal-3 knockout in diabetic mice attenuated neuroinflammation in the retina and optic nerve. This effect was exerted by reducing the activation of retinal microglia and macroglia, and by increasing the number of myelinated fibers. Gal-3 knockout has also been associated with AGEs and VEGF reduction, along with the amelioration of BRB dysfunction, during short-term diabetes in mice [189].

5.2. P2X7R Inhibition in DR

Recent evidence suggests a role of the P2X7 receptor in controlling the BRB function and integrity [210,211]. Once P2X7R is activated, it mediates inflammatory vascular reactions induced by cytokines which degrade the BRB integrity and lead to retinal vascular occlusion and ischemia [182,210,212]. Moreover, P2X7R seems to mediate the accumulation of microglia and macrophages in the subretina [213].
A specific role for the P2X7R-NLRP3 inflammasome pathway in retinal endothelial inflammation and pericyte loss during the NPDR stage has been also described [182,183,184] (Table 2). Moreover, P2X7R stimulation or overexpression positively regulates VEGF secretion and accumulation, thus promoting DR neo-angiogenesis [157].
Therefore, a P2X7R inhibition strategy could represent a useful therapeutic tool to manage the early phase of DR. In this regard, the P2X7R selective inhibitor A74003 reduced the apoptosis of mice retinal endothelial cells (mRECs) stimulated with high glucose and lipopolysaccharide (LPS) [183]. Furthermore, Platania and colleagues analyzed the possible anti-inflammatory role of the selective P2X7R antagonist JNJ47965567 in diabetic human retinal pericytes [184]. While high glucose levels induced pericyte cell damage and a significant release of IL-1β, the treatment with JNJ47965567 decreased the IL-1β release by blocking P2X7R and consequently, NLRP3 inflammasome activation [184].

6. Conclusions

Due to the role of galectins in ERS modulation and the role of P2X7R in NLRP3 inflammasome activation, both these mediators could represent two new potential targets, whose specific inhibition could help counteract the inflammatory process underlying DR progression, although evidence for a specific molecular relation and interlink between them is not available yet (Figure 3).

Author Contributions

Conceptualization, C.C.L., M.R. and M.C.T.; methodology, F.P. and F.A.D.; software, G.G. and G.D.; writing—original draft preparation, C.C.L., M.R., M.C.T. and C.G.; writing—review and editing, M.D., A.H. and S.R.; visualization, M.R.C., E.C. and M.N.; supervision, F.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Italian Ministry of Education, University and Research, grant number PRIN 2020FR7TCL_002 and by the Roman Ministry of Research, Innovation and Digitalization, CNCS/CCCDI-UEFISCDI, project number PN-III-P4-ID-PCE-2020-1772, within PNCDI III.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Harding, J.L.; Pavkov, M.E.; Magliano, D.J.; Shaw, J.E.; Gregg, E.W. Global Trends in Diabetes Complications: A Review of Current Evidence. Diabetologia 2019, 62, 3–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Ahmed, R.; Khalil, S.; Al-Qahtani, M. Diabetic Retinopathy and the Associated Risk Factors in Diabetes Type 2 Patients in Abha, Saudi Arabia. J. Fam. Commun. Med. 2016, 23, 18. [Google Scholar] [CrossRef] [PubMed]
  3. Mahobia, A.; Sahoo, S.; Maiti, N.; Sathyanarayanan, R.; Aravinth, R.; Pandraveti, R.; Tiwari, H. Diabetic Retinopathy and Its Effect on Quality of Life: An Original Research. J. Pharm. Bioall. Sci. 2021, 13, 1365. [Google Scholar] [CrossRef] [PubMed]
  4. Magliah, S.; Bardisi, W.; Al Attah, M.; Khorsheed, M. The Prevalence and Risk Factors of Diabetic Retinopathy in Selected Primary Care Centers during the 3-Year Screening Intervals. J. Family Med. Prim. Care 2018, 7, 975. [Google Scholar] [CrossRef]
  5. Teo, Z.L.; Tham, Y.-C.; Yu, M.; Chee, M.L.; Rim, T.H.; Cheung, N.; Bikbov, M.M.; Wang, Y.X.; Tang, Y.; Lu, Y.; et al. Global Prevalence of Diabetic Retinopathy and Projection of Burden through 2045. Ophthalmology 2021, 128, 1580–1591. [Google Scholar] [CrossRef]
  6. Zhang, X.; Saaddine, J.B.; Chou, C.-F.; Cotch, M.F.; Cheng, Y.J.; Geiss, L.S.; Gregg, E.W.; Albright, A.L.; Klein, B.E.K.; Klein, R. Prevalence of Diabetic Retinopathy in the United States, 2005–2008. JAMA 2010, 304, 649. [Google Scholar] [CrossRef]
  7. Moreno, A.; Lozano, M.; Salinas, P. Retinopatia Diabetica. Nutr. Hosp. 2013, 28 (Suppl. S2), 53–56. [Google Scholar] [CrossRef]
  8. Kollias, A.N.; Ulbig, M.W. Diabetic Retinopathy. Dtsch. Arztebl Int. 2010, 107, 75–84. [Google Scholar] [CrossRef]
  9. Beltramo, E.; Porta, M. Pericyte Loss in Diabetic Retinopathy: Mechanisms and Consequences. CMC 2013, 20, 3218–3225. [Google Scholar] [CrossRef] [Green Version]
  10. Bernabeu, M.O.; Lu, Y.; Abu-Qamar, O.; Aiello, L.P.; Sun, J.K. Estimation of Diabetic Retinal Microaneurysm Perfusion Parameters Based on Computational Fluid Dynamics Modeling of Adaptive Optics Scanning Laser Ophthalmoscopy. Front. Physiol. 2018, 9, 989. [Google Scholar] [CrossRef] [Green Version]
  11. Adornetto, A.; Gesualdo, C.; Laganà, M.L.; Trotta, M.C.; Rossi, S.; Russo, R. Autophagy: A Novel Pharmacological Target in Diabetic Retinopathy. Front. Pharmacol. 2021, 12, 695267. [Google Scholar] [CrossRef] [PubMed]
  12. Das, D.; Biswas, S.K.; Bandyopadhyay, S. A Critical Review on Diagnosis of Diabetic Retinopathy Using Machine Learning and Deep Learning. Multimed Tools Appl. 2022, 81, 25613–25655. [Google Scholar] [CrossRef] [PubMed]
  13. Wang, W.; Lo, A. Diabetic Retinopathy: Pathophysiology and Treatments. Int. J. Mol. Sci. 2018, 19, 1816. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Wu, L. Classification of Diabetic Retinopathy and Diabetic Macular Edema. World J. Diabetes 2013, 4, 290. [Google Scholar] [CrossRef]
  15. Nentwich, M.M. Diabetic Retinopathy—Ocular Complications of Diabetes Mellitus. World J. Diabetes 2015, 6, 489. [Google Scholar] [CrossRef]
  16. Trotta, M.C.; Gesualdo, C.; Petrillo, F.; Lepre, C.C.; Della Corte, A.; Cavasso, G.; Maggiore, G.; Hermenean, A.; Simonelli, F.; D’Amico, M.; et al. Resolution of Inflammation in Retinal Disorders: Briefly the State. Int. J. Mol. Sci. 2022, 23, 4501. [Google Scholar] [CrossRef] [PubMed]
  17. Cheung, N.; Mitchell, P.; Wong, T.Y. Diabetic Retinopathy. Lancet 2010, 376, 124–136. [Google Scholar] [CrossRef]
  18. Gale, M.J.; Scruggs, B.A.; Flaxel, C.J. Diabetic Eye Disease: A Review of Screening and Management Recommendations. Clin. Exp. Ophthalmol. 2021, 49, 128–145. [Google Scholar] [CrossRef]
  19. Romero-Aroca, P. Managing Diabetic Macular Edema: The Leading Cause of Diabetes Blindness. World J. Diabetes 2011, 2, 98. [Google Scholar] [CrossRef]
  20. Hendrick, A.M.; Gibson, M.V.; Kulshreshtha, A. Diabetic Retinopathy. Prim. Care Clin. Off. Pract. 2015, 42, 451–464. [Google Scholar] [CrossRef]
  21. Rossi, S.; Maisto, R.; Gesualdo, C.; Trotta, M.C.; Ferraraccio, F.; Kaneva, M.K.; Getting, S.J.; Surace, E.; Testa, F.; Simonelli, F.; et al. Corrigendum to “Activation of Melanocortin Receptors MC1 and MC5 Attenuates Retinal Damage in Experimental Diabetic Retinopathy”. Mediat. Inflamm. 2021, 2021, 9861434. [Google Scholar] [CrossRef] [PubMed]
  22. Kaushik, V.; Gessa, L.; Kumar, N.; Fernandes, H. Towards a New Biomarker for Diabetic Retinopathy: Exploring RBP3 Structure and Retinoids Binding for Functional Imaging of Eyes In Vivo. Int. J. Mol. Sci. 2023, 24, 4408. [Google Scholar] [CrossRef] [PubMed]
  23. Yu, C.-G.; Yuan, S.-S.; Yang, L.-Y.; Ke, J.; Zhang, L.-J.; Lang, J.-N.; Zhang, D.-W.; Zhao, S.-Z.; Zhao, D.; Feng, Y.-M. Angiopoietin-like 3 Is a Potential Biomarker for Retinopathy in Type 2 Diabetic Patients. Am. J. Ophthalmol. 2018, 191, 34–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Trotta, M.C.; Gesualdo, C.; Platania, C.B.M.; De Robertis, D.; Giordano, M.; Simonelli, F.; D’Amico, M.; Drago, F.; Bucolo, C.; Rossi, S. Circulating MiRNAs in Diabetic Retinopathy Patients: Prognostic Markers or Pharmacological Targets? Biochem. Pharmacol. 2021, 186, 114473. [Google Scholar] [CrossRef]
  25. Xie, Z.; Xiao, X. Novel Biomarkers and Therapeutic Approaches for Diabetic Retinopathy and Nephropathy: Recent Progress and Future Perspectives. Front. Endocrinol. 2022, 13, 1065856. [Google Scholar] [CrossRef]
  26. Xuan, Q.; Ouyang, Y.; Wang, Y.; Wu, L.; Li, H.; Luo, Y.; Zhao, X.; Feng, D.; Qin, W.; Hu, C.; et al. Multiplatform Metabolomics Reveals Novel Serum Metabolite Biomarkers in Diabetic Retinopathy Subjects. Adv. Sci. 2020, 7, 2001714. [Google Scholar] [CrossRef]
  27. Trotta, M.C.; Gesualdo, C.; Petrillo, F.; Cavasso, G.; Corte, A.D.; D’Amico, G.; Hermenean, A.; Simonelli, F.; Rossi, S. Serum Iba-1, GLUT5, and TSPO in Patients With Diabetic Retinopathy: New Biomarkers for Early Retinal Neurovascular Alterations? A Pilot Study. Trans. Vis. Sci. Tech. 2022, 11, 16. [Google Scholar] [CrossRef]
  28. Bianco, L.; Arrigo, A.; Aragona, E.; Antropoli, A.; Berni, A.; Saladino, A.; Battaglia Parodi, M.; Bandello, F. Neuroinflammation and Neurodegeneration in Diabetic Retinopathy. Front. Aging Neurosci. 2022, 14, 937999. [Google Scholar] [CrossRef]
  29. Sorrentino, F.S.; Allkabes, M.; Salsini, G.; Bonifazzi, C.; Perri, P. The Importance of Glial Cells in the Homeostasis of the Retinal Microenvironment and Their Pivotal Role in the Course of Diabetic Retinopathy. Life Sci. 2016, 162, 54–59. [Google Scholar] [CrossRef] [Green Version]
  30. Nio-Kobayashi, J.; Itabashi, T. Galectins and Their Ligand Glycoconjugates in the Central Nervous System Under Physiological and Pathological Conditions. Front. Neuroanat. 2021, 15, 767330. [Google Scholar] [CrossRef]
  31. Vasta, G.R. Lectins as Innate Immune Recognition Factors: Structural, Functional, and Evolutionary Aspects. In The Evolution of the Immune System; Elsevier: Amsterdam, The Netherlands, 2016; pp. 205–224. ISBN 978-0-12-801975-7. [Google Scholar]
  32. Johannes, L.; Jacob, R.; Leffler, H. Galectins at a Glance. J. Cell Sci. 2018, 131, jcs208884. [Google Scholar] [CrossRef] [Green Version]
  33. Rathnasamy, G.; Foulds, W.S.; Ling, E.-A.; Kaur, C. Retinal Microglia—A Key Player in Healthy and Diseased Retina. Prog. Neurobiol. 2019, 173, 18–40. [Google Scholar] [CrossRef] [PubMed]
  34. Vecino, E.; Rodriguez, F.D.; Ruzafa, N.; Pereiro, X.; Sharma, S.C. Glia–Neuron Interactions in the Mammalian Retina. Prog. Retin. Eye Res. 2016, 51, 1–40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Yang, J.; Chen, C.; McLaughlin, T.; Wang, Y.; Le, Y.-Z.; Wang, J.J.; Zhang, S.X. Loss of X-Box Binding Protein 1 in Müller Cells Augments Retinal Inflammation in a Mouse Model of Diabetes. Diabetologia 2019, 62, 531–543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Fernández, D.; Geisse, A.; Bernales, J.I.; Lira, A.; Osorio, F. The Unfolded Protein Response in Immune Cells as an Emerging Regulator of Neuroinflammation. Front. Aging Neurosci. 2021, 13, 682633. [Google Scholar] [CrossRef]
  37. Sims, S.G.; Cisney, R.N.; Lipscomb, M.M.; Meares, G.P. The Role of Endoplasmic Reticulum Stress in Astrocytes. Glia 2022, 70, 5–19. [Google Scholar] [CrossRef]
  38. Zheng, Q.; Ren, Y.; Reinach, P.S.; Xiao, B.; Lu, H.; Zhu, Y.; Qu, J.; Chen, W. Reactive Oxygen Species Activated NLRP3 Inflammasomes Initiate Inflammation in Hyperosmolarity Stressed Human Corneal Epithelial Cells and Environment-Induced Dry Eye Patients. Exp. Eye Res. 2015, 134, 133–140. [Google Scholar] [CrossRef]
  39. Kang, Q.; Dai, H.; Jiang, S.; Yu, L. Advanced Glycation End Products in Diabetic Retinopathy and Phytochemical Therapy. Front. Nutr. 2022, 9, 1037186. [Google Scholar] [CrossRef]
  40. Wu, H.; Hwang, D.-K.; Song, X.; Tao, Y. Association between Aqueous Cytokines and Diabetic Retinopathy Stage. J. Ophthalmol. 2017, 2017, 9402198. [Google Scholar] [CrossRef] [Green Version]
  41. Sarlus, H.; Heneka, M.T. Microglia in Alzheimer’s Disease. J. Clin. Investig. 2017, 127, 3240–3249. [Google Scholar] [CrossRef] [Green Version]
  42. Guo, S.; Wang, H.; Yin, Y. Microglia Polarization From M1 to M2 in Neurodegenerative Diseases. Front. Aging Neurosci. 2022, 14, 815347. [Google Scholar] [CrossRef]
  43. Wake, H.; Moorhouse, A.J.; Nabekura, J. Functions of Microglia in the Central Nervous System—Beyond the Immune Response. Neuron. Glia Biol. 2011, 7, 47–53. [Google Scholar] [CrossRef]
  44. Santiago, A.R.; Baptista, F.I.; Santos, P.F.; Cristóvão, G.; Ambrósio, A.F.; Cunha, R.A.; Gomes, C.A. Role of Microglia Adenosine A 2A Receptors in Retinal and Brain Neurodegenerative Diseases. Mediators Inflamm. 2014, 2014, 465694. [Google Scholar] [CrossRef] [PubMed]
  45. Santos, A.M.; MartÃ-n-Oliva, D.; Ferrer-MartÃ-n, R.M.; Tassi, M.; Calvente, R.; Sierra, A.; Carrasco, M.-C.; MarÃ-n-Teva, J.L.; Navascués, J.; Cuadros, M.A. Microglial Response to Light-Induced Photoreceptor Degeneration in the Mouse Retina. J. Comp. Neurol. 2010, 518, 477–492. [Google Scholar] [CrossRef] [PubMed]
  46. Huang, T.; Cui, J.; Li, L.; Hitchcock, P.F.; Li, Y. The Role of Microglia in the Neurogenesis of Zebrafish Retina. Biochem. Biophy. Res. Commun. 2012, 421, 214–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Zeng, H. Microglial Activation in Human Diabetic Retinopathy. Arch. Ophthalmol. 2008, 126, 227. [Google Scholar] [CrossRef] [Green Version]
  48. Jiao, H.; Natoli, R.; Valter, K.; Provis, J.M.; Rutar, M. Spatiotemporal Cadence of Macrophage Polarisation in a Model of Light-Induced Retinal Degeneration. PLoS ONE 2015, 10, e0143952. [Google Scholar] [CrossRef]
  49. Arroba, A.I.; Álvarez-Lindo, N.; van Rooijen, N.; de la Rosa, E.J. Microglia-Mediated IGF-I Neuroprotection in the Rd10 Mouse Model of Retinitis Pigmentosa. Investig. Ophthalmol. Vis. Sci. 2011, 52, 9124. [Google Scholar] [CrossRef] [Green Version]
  50. Kettenmann, H.; Kirchhoff, F.; Verkhratsky, A. Microglia: New Roles for the Synaptic Stripper. Neuron 2013, 77, 10–18. [Google Scholar] [CrossRef] [Green Version]
  51. Zhang, W.; Liu, H.; Al-Shabrawey, M.; Caldwell, R.W.; Caldwell, R.B. Inflammation and Diabetic Retinal Microvascular Complications. J. Cardiovasc. Dis. Res. 2011, 2, 96–103. [Google Scholar] [CrossRef] [Green Version]
  52. Tang, J.; Kern, T.S. Inflammation in Diabetic Retinopathy. Prog. Retin. Eye Res. 2011, 30, 343–358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Fumagalli, S.; Perego, C.; Pischiutta, F.; Zanier, E.R.; De Simoni, M.-G. The Ischemic Environment Drives Microglia and Macrophage Function. Front. Neurol. 2015, 6, 81. [Google Scholar] [CrossRef] [Green Version]
  54. Trotta, M.C.; Gesualdo, C.; Herman, H.; Gharbia, S.; Balta, C.; Lepre, C.C.; Russo, M.; Itro, A.; D’Amico, G.; Peluso, L.; et al. Systemic Beta-Hydroxybutyrate Affects BDNF and Autophagy into the Retina of Diabetic Mice. Int. J. Mol. Sci. 2022, 23, 10184. [Google Scholar] [CrossRef] [PubMed]
  55. Yunna, C.; Mengru, H.; Lei, W.; Weidong, C. Macrophage M1/M2 Polarization. Eur. J. Pharmacol. 2020, 877, 173090. [Google Scholar] [CrossRef]
  56. Wang, L.; Shang, X.; Qi, X.; Ba, D.; Lv, J.; Zhou, X.; Wang, H.; Shaxika, N.; Wang, J.; Ma, X. Clinical Significance of M1/M2 Macrophages and Related Cytokines in Patients with Spinal Tuberculosis. Dis. Markers 2020, 2020, 2509454. [Google Scholar] [CrossRef] [PubMed]
  57. Colonna, M.; Butovsky, O. Microglia Function in the Central Nervous System During Health and Neurodegeneration. Annu. Rev. Immunol. 2017, 35, 441–468. [Google Scholar] [CrossRef]
  58. Grigsby, J.G.; Cardona, S.M.; Pouw, C.E.; Muniz, A.; Mendiola, A.S.; Tsin, A.T.C.; Allen, D.M.; Cardona, A.E. The Role of Microglia in Diabetic Retinopathy. J. Ophthalmol. 2014, 2014, 705783. [Google Scholar] [CrossRef] [Green Version]
  59. Coughlin, B.A.; Feenstra, D.J.; Mohr, S. Müller Cells and Diabetic Retinopathy. Vision Res. 2017, 139, 93–100. [Google Scholar] [CrossRef]
  60. Wang, M.; Ma, W.; Zhao, L.; Fariss, R.N.; Wong, W.T. Adaptive Müller Cell Responses to Microglial Activation Mediate Neuroprotection and Coordinate Inflammation in the Retina. J. Neuroinflamm. 2011, 8, 173. [Google Scholar] [CrossRef] [Green Version]
  61. Carpi-Santos, R.; De Melo Reis, R.A.; Gomes, F.C.A.; Calaza, K.C. Contribution of Müller Cells in the Diabetic Retinopathy Development: Focus on Oxidative Stress and Inflammation. Antioxidants 2022, 11, 617. [Google Scholar] [CrossRef]
  62. Liu, X.; Ye, F.; Xiong, H.; Hu, D.; Limb, G.A.; Xie, T.; Peng, L.; Yang, W.; Sun, Y.; Zhou, M.; et al. IL-1β Upregulates IL-8 Production in Human Müller Cells Through Activation of the P38 MAPK and ERK1/2 Signaling Pathways. Inflammation 2014, 37, 1486–1495. [Google Scholar] [CrossRef] [PubMed]
  63. Bringmann, A.; Wiedemann, P. Müller Glial Cells in Retinal Disease. Ophthalmologica 2012, 227, 1–19. [Google Scholar] [CrossRef] [PubMed]
  64. Tang, Y.; Chen, Y.; Chen, D. The Heterogeneity of Astrocytes in Glaucoma. Front. Neuroanat. 2022, 16, 995369. [Google Scholar] [CrossRef] [PubMed]
  65. Yu, Y.; Chen, H.; Su, S.B. Neuroinflammatory Responses in Diabetic Retinopathy. J. Neuroinflamm. 2015, 12, 141. [Google Scholar] [CrossRef] [Green Version]
  66. Chiareli, R.A.; Carvalho, G.A.; Marques, B.L.; Mota, L.S.; Oliveira-Lima, O.C.; Gomes, R.M.; Birbrair, A.; Gomez, R.S.; Simão, F.; Klempin, F.; et al. The Role of Astrocytes in the Neurorepair Process. Front. Cell Dev. Biol. 2021, 9, 665795. [Google Scholar] [CrossRef]
  67. García-Cáceres, C.; Balland, E.; Prevot, V.; Luquet, S.; Woods, S.C.; Koch, M.; Horvath, T.L.; Yi, C.-X.; Chowen, J.A.; Verkhratsky, A.; et al. Role of Astrocytes, Microglia, and Tanycytes in Brain Control of Systemic Metabolism. Nat. Neurosci. 2019, 22, 7–14. [Google Scholar] [CrossRef] [Green Version]
  68. Giovannoni, F.; Quintana, F.J. The Role of Astrocytes in CNS Inflammation. Trends Immunol. 2020, 41, 805–819. [Google Scholar] [CrossRef]
  69. Maisto, R.; Gesualdo, C.; Trotta, M.C.; Grieco, P.; Testa, F.; Simonelli, F.; Barcia, J.M.; D’Amico, M.; Di Filippo, C.; Rossi, S. Melanocortin Receptor Agonists MCR1–5 Protect Photoreceptors from High-Glucose Damage and Restore Antioxidant Enzymes in Primary Retinal Cell Culture. J. Cell Mol. Med. 2017, 21, 968–974. [Google Scholar] [CrossRef]
  70. Costagliola, C.; Romano, V.; De Tollis, M.; Aceto, F.; dell’Omo, R.; Romano, M.R.; Pedicino, C.; Semeraro, F. TNF-Alpha Levels in Tears: A Novel Biomarker to Assess the Degree of Diabetic Retinopathy. Mediators Inflamm. 2013, 2013, 629529. [Google Scholar] [CrossRef]
  71. Lazzara, F.; Trotta, M.C.; Platania, C.B.M.; D’Amico, M.; Petrillo, F.; Galdiero, M.; Gesualdo, C.; Rossi, S.; Drago, F.; Bucolo, C. Stabilization of HIF-1α in Human Retinal Endothelial Cells Modulates Expression of MiRNAs and Proangiogenic Growth Factors. Front. Pharmacol 2020, 11, 1063. [Google Scholar] [CrossRef]
  72. Kong, D.-Q.; Li, L.; Liu, Y.; Zheng, G.-Y. Association between Endoplasmic Reticulum Stress and Risk Factors of Diabetic Retinopathy. Int. J. Ophthalmol. 2018, 11, 1704–1710. [Google Scholar] [CrossRef]
  73. Rinaldi, C.; Donato, L.; Alibrandi, S.; Scimone, C.; D’Angelo, R.; Sidoti, A. Oxidative Stress and the Neurovascular Unit. Life 2021, 11, 767. [Google Scholar] [CrossRef]
  74. Zhong, Y.; Wang, J.J.; Zhang, S.X. Intermittent but Not Constant High Glucose Induces ER Stress and Inflammation in Human Retinal Pericytes. Adv. Exp. Med. Biol. 2012, 723, 285–292. [Google Scholar] [CrossRef] [Green Version]
  75. Shi, X.; Li, P.; Liu, H.; Prokosch, V. Oxidative Stress, Vascular Endothelium, and the Pathology of Neurodegeneration in Retina. Antioxidants 2022, 11, 543. [Google Scholar] [CrossRef] [PubMed]
  76. Yang, X.; Yu, X.-W.; Zhang, D.-D.; Fan, Z.-G. Blood-Retinal Barrier as a Converging Pivot in Understanding the Initiation and Development of Retinal Diseases. Chin. Med. J. 2020, 133, 2586–2594. [Google Scholar] [CrossRef]
  77. Cao, S.S.; Kaufman, R.J. Endoplasmic Reticulum Stress and Oxidative Stress in Cell Fate Decision and Human Disease. Antioxid. Redox Signal. 2014, 21, 396–413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Araki, K.; Nagata, K. Protein Folding and Quality Control in the ER. Cold Spring Harb. Perspec. Biol. 2011, 3, a007526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. The Plant Endoplasmic Reticulum: Methods and Protocols; Hawes, C.; Kriechbaumer, V. (Eds.) Methods in Molecular Biology; Springer: New York, NY, USA, 2018; Volume 1691, ISBN 978-1-4939-7388-0. [Google Scholar]
  80. Martino, E.; Balestrieri, A.; Mele, L.; Sardu, C.; Marfella, R.; D’Onofrio, N.; Campanile, G.; Balestrieri, M.L. Milk Exosomal MiR-27b Worsen Endoplasmic Reticulum Stress Mediated Colorectal Cancer Cell Death. Nutrients 2022, 14, 5081. [Google Scholar] [CrossRef]
  81. Li, W.; Cao, T.; Luo, C.; Cai, J.; Zhou, X.; Xiao, X.; Liu, S. Crosstalk between ER Stress, NLRP3 Inflammasome, and Inflammation. Appl. Microbiol. Biotechnol. 2020, 104, 6129–6140. [Google Scholar] [CrossRef]
  82. Karlstetter, M.; Scholz, R.; Rutar, M.; Wong, W.T.; Provis, J.M.; Langmann, T. Retinal Microglia: Just Bystander or Target for Therapy? Prog. Retin. Eye Res. 2015, 45, 30–57. [Google Scholar] [CrossRef]
  83. Altmann, C.; Schmidt, M.H.H. The Role of Microglia in Diabetic Retinopathy: Inflammation, Microvasculature Defects and Neurodegeneration. Int. J. Mol. Sci. 2018, 19, 110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Mendonca, H.; Carpi-Santos, R.; da Costa Calaza, K.; Blanco Martinez, A. Neuroinflammation and Oxidative Stress Act in Concert to Promote Neurodegeneration in the Diabetic Retina and Optic Nerve: Galectin-3 Participation. Neural Regen. Res. 2020, 15, 625. [Google Scholar] [CrossRef] [PubMed]
  85. Rübsam, A.; Parikh, S.; Fort, P. Role of Inflammation in Diabetic Retinopathy. Int. J. Mol. Sci. 2018, 19, 942. [Google Scholar] [CrossRef] [Green Version]
  86. Udaondo, P.; Adan, A.; Arias-Barquet, L.; Ascaso, F.J.; Cabrera-López, F.; Castro-Navarro, V.; Donate-López, J.; García-Layana, A.; Lavid, F.J.; Rodríguez-Maqueda, M.; et al. Challenges in Diabetic Macular Edema Management: An Expert Consensus Report. OPTH 2021, 15, 3183–3195. [Google Scholar] [CrossRef] [PubMed]
  87. Liu, Y.; Biarnés Costa, M.; Gerhardinger, C. IL-1β Is Upregulated in the Diabetic Retina and Retinal Vessels: Cell-Specific Effect of High Glucose and IL-1β Autostimulation. PLoS ONE 2012, 7, e36949. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Kim, Y.K.; Shin, J.-S.; Nahm, M.H. NOD-Like Receptors in Infection, Immunity, and Diseases. Yonsei Med. J. 2016, 57, 5. [Google Scholar] [CrossRef] [Green Version]
  89. Trotta, M.C.; Maisto, R.; Guida, F.; Boccella, S.; Luongo, L.; Balta, C.; D’Amico, G.; Herman, H.; Hermenean, A.; Bucolo, C.; et al. The Activation of Retinal HCA2 Receptors by Systemic Beta-Hydroxybutyrate Inhibits Diabetic Retinal Damage through Reduction of Endoplasmic Reticulum Stress and the NLRP3 Inflammasome. PLoS ONE 2019, 14, e0211005. [Google Scholar] [CrossRef] [PubMed]
  90. Abrishami, M.; Tohidinezhad, F.; Daneshvar, R.; Omidtabrizi, A.; Amini, M.; Sedaghat, A.; Amini, S.; Reihani, H.; Allahyari, A.; Seddigh-Shamsi, M.; et al. Ocular Manifestations of Hospitalized Patients with COVID-19 in Northeast of Iran. Ocul. Immunol. Inflamm. 2020, 28, 739–744. [Google Scholar] [CrossRef]
  91. Kuo, C.Y.-J.; Maran, J.J.; Jamieson, E.G.; Rupenthal, I.D.; Murphy, R.; Mugisho, O.O. Characterization of NLRP3 Inflammasome Activation in the Onset of Diabetic Retinopathy. Int. J. Mol. Sci. 2022, 23, 14471. [Google Scholar] [CrossRef]
  92. Chen, X.; Guo, X.; Ge, Q.; Zhao, Y.; Mu, H.; Zhang, J. ER Stress Activates the NLRP3 Inflammasome: A Novel Mechanism of Atherosclerosis. Oxidative Med. Cell. Longev. 2019, 2019, 3462530. [Google Scholar] [CrossRef]
  93. Zheng, D.; Liwinski, T.; Elinav, E. Inflammasome Activation and Regulation: Toward a Better Understanding of Complex Mechanisms. Cell Discov. 2020, 6, 36. [Google Scholar] [CrossRef] [PubMed]
  94. Zhou, R.; Tardivel, A.; Thorens, B.; Choi, I.; Tschopp, J. Thioredoxin-Interacting Protein Links Oxidative Stress to Inflammasome Activation. Nat. Immunol. 2010, 11, 136–140. [Google Scholar] [CrossRef] [PubMed]
  95. Lu, A.; Wu, H. Structural Mechanisms of Inflammasome Assembly. FEBS J. 2015, 282, 435–444. [Google Scholar] [CrossRef] [PubMed]
  96. Pelegrin, P. P2X7 Receptor and the NLRP3 Inflammasome: Partners in Crime. Biochem. Pharmacol. 2021, 187, 114385. [Google Scholar] [CrossRef] [PubMed]
  97. Abais, J.M.; Xia, M.; Zhang, Y.; Boini, K.M.; Li, P.-L. Redox Regulation of NLRP3 Inflammasomes: ROS as Trigger or Effector? Antioxid. Redox Signal. 2015, 22, 1111–1129. [Google Scholar] [CrossRef] [Green Version]
  98. Snezhkina, A.V.; Kudryavtseva, A.V.; Kardymon, O.L.; Savvateeva, M.V.; Melnikova, N.V.; Krasnov, G.S.; Dmitriev, A.A. ROS Generation and Antioxidant Defense Systems in Normal and Malignant Cells. Oxidative Med. Cell. Longev. 2019, 2019, 6175804. [Google Scholar] [CrossRef] [Green Version]
  99. Swanson, K.V.; Deng, M.; Ting, J.P.-Y. The NLRP3 Inflammasome: Molecular Activation and Regulation to Therapeutics. Nat. Rev. Immunol. 2019, 19, 477–489. [Google Scholar] [CrossRef]
  100. Kelley, N.; Jeltema, D.; Duan, Y.; He, Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int. J. Mol. Sci. 2019, 20, 3328. [Google Scholar] [CrossRef] [Green Version]
  101. Prather, E.R.; Gavrilin, M.A.; Wewers, M.D. The Central Inflammasome Adaptor Protein ASC Activates the Inflammasome after Transition from a Soluble to an Insoluble State. J. Biol. Chem. 2022, 298, 102024. [Google Scholar] [CrossRef]
  102. Yu, P.; Zhang, X.; Liu, N.; Tang, L.; Peng, C.; Chen, X. Pyroptosis: Mechanisms and Diseases. Sig. Transduct. Target. Ther. 2021, 6, 128. [Google Scholar] [CrossRef]
  103. de Zoete, M.R.; Palm, N.W.; Zhu, S.; Flavell, R.A. Inflammasomes. Cold Spring Harb. Perspect. Biol. 2014, 6, a016287. [Google Scholar] [CrossRef]
  104. He, W.; Wan, H.; Hu, L.; Chen, P.; Wang, X.; Huang, Z.; Yang, Z.-H.; Zhong, C.-Q.; Han, J. Gasdermin D Is an Executor of Pyroptosis and Required for Interleukin-1β Secretion. Cell Res. 2015, 25, 1285–1298. [Google Scholar] [CrossRef]
  105. Raman, K.S.; Matsubara, J.A. Dysregulation of the NLRP3 Inflammasome in Diabetic Retinopathy and Potential Therapeutic Targets. Ocul. Immunol. Inflamm. 2022, 30, 470–478. [Google Scholar] [CrossRef] [PubMed]
  106. Loukovaara, S.; Piippo, N.; Kinnunen, K.; Hytti, M.; Kaarniranta, K.; Kauppinen, A. NLRP3 Inflammasome Activation Is Associated with Proliferative Diabetic Retinopathy. Acta Ophthalmol. 2017, 95, 803–808. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Kuo, C.Y.J.; Murphy, R.; Rupenthal, I.D.; Mugisho, O.O. Correlation between the Progression of Diabetic Retinopathy and Inflammasome Biomarkers in Vitreous and Serum—A Systematic Review. BMC Ophthalmol. 2022, 22, 238. [Google Scholar] [CrossRef] [PubMed]
  108. Tseng, W.A.; Thein, T.; Kinnunen, K.; Lashkari, K.; Gregory, M.S.; D’Amore, P.A.; Ksander, B.R. NLRP3 Inflammasome Activation in Retinal Pigment Epithelial Cells by Lysosomal Destabilization: Implications for Age-Related Macular Degeneration. Investig. Ophthalmol. Vis. Sci. 2013, 54, 110. [Google Scholar] [CrossRef] [PubMed]
  109. Mohr, L.K.M.; Hoffmann, A.V.; Brandstetter, C.; Holz, F.G.; Krohne, T.U. Effects of Inflammasome Activation on Secretion of Inflammatory Cytokines and Vascular Endothelial Growth Factor by Retinal Pigment Epithelial Cells. Invest. Ophthalmol. Vis. Sci. 2015, 56, 6404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Doktor, F.; Prager, P.; Wiedemann, P.; Kohen, L.; Bringmann, A.; Hollborn, M. Hypoxic Expression of NLRP3 and VEGF in Cultured Retinal Pigment Epithelial Cells: Contribution of P2Y2 Receptor Signaling. Purinergic Signal. 2018, 14, 471–484. [Google Scholar] [CrossRef]
  111. Ebeling, M.C.; Fisher, C.R.; Kapphahn, R.J.; Stahl, M.R.; Shen, S.; Qu, J.; Montezuma, S.R.; Ferrington, D.A. Inflammasome Activation in Retinal Pigment Epithelium from Human Donors with Age-Related Macular Degeneration. Cells 2022, 11, 2075. [Google Scholar] [CrossRef]
  112. Jiang, Y.; Liu, L.; Curtiss, E.; Steinle, J.J. Epac1 Blocks NLRP3 Inflammasome to Reduce IL-1 β in Retinal Endothelial Cells and Mouse Retinal Vasculature. Mediat. Inflamm. 2017, 2017, 2860956. [Google Scholar] [CrossRef] [Green Version]
  113. Zou, W.; Luo, S.; Zhang, Z.; Cheng, L.; Huang, X.; Ding, N.; Pan, Y.; Wu, Z. ASK1/P38-mediated NLRP3 Inflammasome Signaling Pathway Contributes to Aberrant Retinal Angiogenesis in Diabetic Retinopathy. Int. J. Mol. Med. 2020, 47, 732–740. [Google Scholar] [CrossRef] [PubMed]
  114. Ding, X.; Xie, H.; Shan, W.; Li, L. Agonism of GPR120 Prevented High Glucose-Induced Apoptosis of Retinal Endothelial Cells through Inhibiting NLRP3 Inflammasome. Klin. Monbl. Augenheilkd 2022. [Google Scholar] [CrossRef] [PubMed]
  115. Viringipurampeer, I.A.; Metcalfe, A.L.; Bashar, A.E.; Sivak, O.; Yanai, A.; Mohammadi, Z.; Moritz, O.L.; Gregory-Evans, C.Y.; Gregory-Evans, K. NLRP3 Inflammasome Activation Drives Bystander Cone Photoreceptor Cell Death in a P23H Rhodopsin Model of Retinal Degeneration. Hum. Mol. Genet. 2016, 25, 1501–1516. [Google Scholar] [CrossRef] [Green Version]
  116. Wooff, Y.; Fernando, N.; Wong, J.H.C.; Dietrich, C.; Aggio-Bruce, R.; Chu-Tan, J.A.; Robertson, A.A.B.; Doyle, S.L.; Man, S.M.; Natoli, R. Caspase-1-Dependent Inflammasomes Mediate Photoreceptor Cell Death in Photo-Oxidative Damage-Induced Retinal Degeneration. Sci. Rep. 2020, 10, 2263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Chaurasia, S.S.; Lim, R.R.; Parikh, B.H.; Wey, Y.S.; Tun, B.B.; Wong, T.Y.; Luu, C.D.; Agrawal, R.; Ghosh, A.; Mortellaro, A.; et al. The NLRP3 Inflammasome May Contribute to Pathologic Neovascularization in the Advanced Stages of Diabetic Retinopathy. Sci. Rep. 2018, 8, 2847. [Google Scholar] [CrossRef] [Green Version]
  118. Huang, L.; You, J.; Yao, Y.; Xie, M. High Glucose Induces Pyroptosis of Retinal Microglia through NLPR3 Inflammasome Signaling. Arq. Bras. Oftalmol. 2021, 84, 67–73. [Google Scholar]
  119. Pronin, A.; Pham, D.; An, W.; Dvoriantchikova, G.; Reshetnikova, G.; Qiao, J.; Kozhekbaeva, Z.; Reiser, A.E.; Slepak, V.Z.; Shestopalov, V.I. Inflammasome Activation Induces Pyroptosis in the Retina Exposed to Ocular Hypertension Injury. Front. Mol. Neurosci. 2019, 12, 36. [Google Scholar] [CrossRef] [Green Version]
  120. Fryk, E.; Strindberg, L.; Lundqvist, A.; Sandstedt, M.; Bergfeldt, L.; Mattsson Hultén, L.; Bergström, G.; Jansson, P.-A. Galectin-1 Is Inversely Associated with Type 2 Diabetes Independently of Obesity—A SCAPIS Pilot Study. Metabolism Open 2019, 4, 100017. [Google Scholar] [CrossRef]
  121. Bobronnikova, L. Galectin-3 as a Potential Biomarker of Metabolic Disorders and Cardiovascular Remodeling in Patients with Hypertension and Type 2 Diabetes. Vessel Plus. 2017, 1, 61–67. [Google Scholar] [CrossRef]
  122. Vora, A.; de Lemos, J.A.; Ayers, C.; Grodin, J.L.; Lingvay, I. Association of Galectin-3 With Diabetes Mellitus in the Dallas Heart Study. J. Clin. Endocrinol. Metab. 2019, 104, 4449–4458. [Google Scholar] [CrossRef]
  123. Souza, I.D.P.; Rodrigues, K.F.; Pietrani, N.T.; Bosco, A.A.; Gomes, K.B.; Alves, M.T. Evaluation of Galectin-3 Levels in Patients with Type 2 Diabetes Mellitus and Chronic Kidney Disease. J. Bras. De Patol. E Med. Lab. 2021, 57, 1–9. [Google Scholar] [CrossRef]
  124. Atalar, M.N.; Abuşoğlu, S.; Ünlü, A.; Tok, O.; İpekçi, S.H.; Baldane, S.; Kebapcılar, L. Assessment of Serum Galectin-3, Methylated Arginine and Hs-CRP Levels in Type 2 Diabetes and Prediabetes. Life Sci. 2019, 231, 116577. [Google Scholar] [CrossRef] [PubMed]
  125. Li, Y.; Li, T.; Zhou, Z.; Xiao, Y. Emerging Roles of Galectin-3 in Diabetes and Diabetes Complications: A Snapshot. Rev. Endocr. Metab. Disord. 2022, 23, 569–577. [Google Scholar] [CrossRef] [PubMed]
  126. Jeethy Ram, T.; Lekshmi, A.; Somanathan, T.; Sujathan, K. Galectin-3: A Factotum in Carcinogenesis Bestowing an Archery for Prevention. Tumor Biol. 2021, 43, 77–96. [Google Scholar] [CrossRef]
  127. Kanzaki, M.; Wada, J.; Sugiyama, K.; Nakatsuka, A.; Teshigawara, S.; Murakami, K.; Inoue, K.; Terami, T.; Katayama, A.; Eguchi, J.; et al. Galectin-9 and T Cell Immunoglobulin Mucin-3 Pathway Is a Therapeutic Target for Type 1 Diabetes. Endocrinology 2012, 153, 612–620. [Google Scholar] [CrossRef] [Green Version]
  128. Kurose, Y.; Wada, J.; Kanzaki, M.; Teshigawara, S.; Nakatsuka, A.; Murakami, K.; Inoue, K.; Terami, T.; Katayama, A.; Watanabe, M.; et al. Serum Galectin-9 Levels Are Elevated in the Patients with Type 2 Diabetes and Chronic Kidney Disease. BMC Nephrol. 2013, 14, 23. [Google Scholar] [CrossRef] [Green Version]
  129. Sudhakar, J.N.; Lu, H.-H.; Chiang, H.-Y.; Suen, C.-S.; Hwang, M.-J.; Wu, S.-Y.; Shen, C.-N.; Chang, Y.-M.; Li, F.-A.; Liu, F.-T.; et al. Lumenal Galectin-9-Lamp2 Interaction Regulates Lysosome and Autophagy to Prevent Pathogenesis in the Intestine and Pancreas. Nat. Commun. 2020, 11, 4286. [Google Scholar] [CrossRef] [PubMed]
  130. Wang, W.; Qin, Y.; Song, H.; Wang, L.; Jia, M.; Zhao, C.; Gong, M.; Zhao, W. Galectin-9 Targets NLRP3 for Autophagic Degradation to Limit Inflammation. J. Immunol. 2021, 206, 2692–2699. [Google Scholar] [CrossRef] [PubMed]
  131. Kuo, P.; Le, Q.-T. Galectin-1 Links Tumor Hypoxia and Radiotherapy. Glycobiology 2014, 24, 921–925. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Morgan, M.J.; Liu, Z. Crosstalk of Reactive Oxygen Species and NF-ΚB Signaling. Cell Res. 2011, 21, 103–115. [Google Scholar] [CrossRef] [Green Version]
  133. Astorgues-Xerri, L.; Riveiro, M.E.; Tijeras-Raballand, A.; Serova, M.; Rabinovich, G.A.; Bieche, I.; Vidaud, M.; de Gramont, A.; Martinet, M.; Cvitkovic, E.; et al. OTX008, a Selective Small-Molecule Inhibitor of Galectin-1, Downregulates Cancer Cell Proliferation, Invasion and Tumour Angiogenesis. Eur. J. Cancer 2014, 50, 2463–2477. [Google Scholar] [CrossRef]
  134. Griffioen, A.W.; Thijssen, V.L. Galectins in Tumor Angiogenesis. Ann. Transl. Med. 2014, 2, 90. [Google Scholar] [PubMed]
  135. Coppin, L.; Jannin, A.; Ait Yahya, E.; Thuillier, C.; Villenet, C.; Tardivel, M.; Bongiovanni, A.; Gaston, C.; de Beco, S.; Barois, N.; et al. Galectin-3 Modulates Epithelial Cell Adaptation to Stress at the ER-Mitochondria Interface. Cell Death Dis. 2020, 11, 360. [Google Scholar] [CrossRef]
  136. Caridi, B.; Doncheva, D.; Sivaprasad, S.; Turowski, P. Galectins in the Pathogenesis of Common Retinal Disease. Front. Pharmacol. 2021, 12, 687495. [Google Scholar] [CrossRef] [PubMed]
  137. Tian, J.; Yang, G.; Chen, H.; Hsu, D.K.; Tomilov, A.; Olson, K.K.A.; Dehnad, A.; Fish, S.R.; Cortopassi, G.; Zhao, K.B.; et al. Galectin-3 Regulates Inflammasome Activation in Cholestatic Liver Injury. FASEB J. 2016, 30, 4202–4213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Kato, T.; Kurita, K.; Seino, T.; Kadoya, T.; Horie, H.; Wada, M.; Kawanami, T.; Daimon, M.; Hirano, A. Galectin-1 Is a Component of Neurofilamentous Lesions in Sporadic and Familial Amyotrophic Lateral Sclerosis. Biochem. Biophys. Res. Commun. 2001, 282, 166–172. [Google Scholar] [CrossRef]
  139. de Jong, C.G.H.M.; Stancic, M.; Pinxterhuis, T.H.; van Horssen, J.; van Dam, A.-M.; Gabius, H.-J.; Baron, W. Galectin-4, a Negative Regulator of Oligodendrocyte Differentiation, Is Persistently Present in Axons and Microglia/Macrophages in Multiple Sclerosis Lesions. J. Neuropathol. Exp. Neurol. 2018, 77, 1024–1038. [Google Scholar] [CrossRef] [Green Version]
  140. Hara, A.; Niwa, M.; Noguchi, K.; Kanayama, T.; Niwa, A.; Matsuo, M.; Hatano, Y.; Tomita, H. Galectin-3 as a Next-Generation Biomarker for Detecting Early Stage of Various Diseases. Biomolecules 2020, 10, 389. [Google Scholar] [CrossRef] [Green Version]
  141. Ramos-Martínez, E.; Ramos-Martínez, I.; Sánchez-Betancourt, I.; Ramos-Martínez, J.C.; Peña-Corona, S.I.; Valencia, J.; Saucedo, R.; Almeida-Aguirre, E.K.P.; Cerbón, M. Association between Galectin Levels and Neurodegenerative Diseases: Systematic Review and Meta-Analysis. Biomolecules 2022, 12, 1062. [Google Scholar] [CrossRef]
  142. Hermenean, A.; Oatis, D.; Herman, H.; Ciceu, A.; D’Amico, G.; Trotta, M.C. Galectin 1—A Key Player between Tissue Repair and Fibrosis. Int. J. Mol. Sci. 2022, 23, 5548. [Google Scholar] [CrossRef]
  143. Oatis, D.; Simon-Repolski, E.; Balta, C.; Mihu, A.; Pieretti, G.; Alfano, R.; Peluso, L.; Trotta, M.C.; D’Amico, M.; Hermenean, A. Cellular and Molecular Mechanism of Pulmonary Fibrosis Post-COVID-19: Focus on Galectin-1, -3, -8, -9. Int. J. Mol. Sci. 2022, 23, 8210. [Google Scholar] [CrossRef] [PubMed]
  144. Chen, H.-L.; Liao, F.; Lin, T.-N.; Liu, F.-T. Galectins and Neuroinflammation. In Glycobiology of the Nervous System; Yu, R.K., Schengrund, C.-L., Eds.; Advances in Neurobiology; Springer: New York, NY, USA, 2014; Volume 9, pp. 517–542. ISBN 978-1-4939-1153-0. [Google Scholar]
  145. Ramírez Hernández, E.; Alanis Olvera, B.; Carmona González, D.; Guerrero Marín, O.; Pantoja Mercado, D.; Valencia Gil, L.; Hernández-Zimbrón, L.F.; Sánchez Salgado, J.L.; Limón, I.D.; Zenteno, E. Neuroinflammation and Galectins: A Key Relationship in Neurodegenerative Diseases. Glycoconj J. 2022, 39, 685–699. [Google Scholar] [CrossRef]
  146. Bonsack, F.; Sukumari-Ramesh, S. Differential Cellular Expression of Galectin-1 and Galectin-3 After Intracerebral Hemorrhage. Front. Cell. Neurosci. 2019, 13, 157. [Google Scholar] [CrossRef] [Green Version]
  147. Gaudet, A.D.; Sweet, D.R.; Polinski, N.K.; Guan, Z.; Popovich, P.G. Galectin-1 in Injured Rat Spinal Cord: Implications for Macrophage Phagocytosis and Neural Repair. Mol. Cell. Neurosci. 2015, 64, 84–94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Quintá, H.R.; Pasquini, J.M.; Rabinovich, G.A.; Pasquini, L.A. Glycan-Dependent Binding of Galectin-1 to Neuropilin-1 Promotes Axonal Regeneration after Spinal Cord Injury. Cell Death Differ. 2014, 21, 941–955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Rubinstein, N.; Alvarez, M.; Zwirner, N.W.; Toscano, M.A.; Ilarregui, J.M.; Bravo, A.; Mordoh, J.; Fainboim, L.; Podhajcer, O.L.; Rabinovich, G.A. Targeted Inhibition of Galectin-1 Gene Expression in Tumor Cells Results in Heightened T Cell-Mediated Rejection. Cancer Cell 2004, 5, 241–251. [Google Scholar] [CrossRef] [Green Version]
  150. Kobayakawa, Y.; Sakumi, K.; Kajitani, K.; Kadoya, T.; Horie, H.; Kira, J.; Nakabeppu, Y. Galectin-1 Deficiency Improves Axonal Swelling of Motor Neurones in SOD1 G93A Transgenic Mice: Galectin-1 Is Associated with Axonal Degeneration in SOD1 G93A Mice. Neuropathol. Appl. Neurobiol. 2015, 41, 227–244. [Google Scholar] [CrossRef]
  151. Jeon, S.-B.; Yoon, H.J.; Chang, C.Y.; Koh, H.S.; Jeon, S.-H.; Park, E.J. Galectin-3 Exerts Cytokine-Like Regulatory Actions through the JAK–STAT Pathway. J. Immunol. 2010, 185, 7037–7046. [Google Scholar] [CrossRef] [Green Version]
  152. Siew, J.J.; Chen, H.-M.; Chen, H.-Y.; Chen, H.-L.; Chen, C.-M.; Soong, B.-W.; Wu, Y.-R.; Chang, C.-P.; Chan, Y.-C.; Lin, C.-H.; et al. Galectin-3 Is Required for the Microglia-Mediated Brain Inflammation in a Model of Huntington’s Disease. Nat. Commun. 2019, 10, 3473. [Google Scholar] [CrossRef] [Green Version]
  153. Boza-Serrano, A.; Ruiz, R.; Sanchez-Varo, R.; García-Revilla, J.; Yang, Y.; Jimenez-Ferrer, I.; Paulus, A.; Wennström, M.; Vilalta, A.; Allendorf, D.; et al. Galectin-3, a Novel Endogenous TREM2 Ligand, Detrimentally Regulates Inflammatory Response in Alzheimer’s Disease. Acta Neuropathol. 2019, 138, 251–273. [Google Scholar] [CrossRef] [Green Version]
  154. Yin, Q.; Chen, J.; Ma, S.; Dong, C.; Zhang, Y.; Hou, X.; Li, S.; Liu, B. Pharmacological Inhibition of Galectin-3 Ameliorates Diabetes-Associated Cognitive Impairment, Oxidative Stress and Neuroinflammation in Vivo and in Vitro. JIR 2020, 13, 533–542. [Google Scholar] [CrossRef] [PubMed]
  155. North, R.A. P2X Receptors. Philos. Trans. R. Soc. B 2016, 371, 20150427. [Google Scholar] [CrossRef] [Green Version]
  156. Martínez-Cuesta, M.Á.; Blanch-Ruiz, M.A.; Ortega-Luna, R.; Sánchez-López, A.; Álvarez, Á. Structural and Functional Basis for Understanding the Biological Significance of P2X7 Receptor. Int. J. Mol. Sci. 2020, 21, 8454. [Google Scholar] [CrossRef]
  157. Tassetto, M.; Scialdone, A.; Solini, A.; Di Virgilio, F. The P2X7 Receptor: A Promising Pharmacological Target in Diabetic Retinopathy. Int. J. Mol. Sci. 2021, 22, 7110. [Google Scholar] [CrossRef] [PubMed]
  158. Platania, C.B.M.; Lazzara, F.; Fidilio, A.; Fresta, C.G.; Conti, F.; Giurdanella, G.; Leggio, G.M.; Salomone, S.; Drago, F.; Bucolo, C. Blood-Retinal Barrier Protection against High Glucose Damage: The Role of P2X7 Receptor. Biochem. Pharmacol. 2019, 168, 249–258. [Google Scholar] [CrossRef] [PubMed]
  159. Di Virgilio, F.; Sarti, A.C.; Grassi, F. Modulation of Innate and Adaptive Immunity by P2X Ion Channels. Curr. Opin. Immunol. 2018, 52, 51–59. [Google Scholar] [CrossRef] [PubMed]
  160. Yang, D. Targeting the P2X7 Receptor in Age-Related Macular Degeneration. Vision 2017, 1, 11. [Google Scholar] [CrossRef] [Green Version]
  161. Franceschini, A.; Capece, M.; Chiozzi, P.; Falzoni, S.; Sanz, J.M.; Sarti, A.C.; Bonora, M.; Pinton, P.; Di Virgilio, F. The P2X7 Receptor Directly Interacts with the NLRP3 Inflammasome Scaffold Protein. FASEB J. 2015, 29, 2450–2461. [Google Scholar] [CrossRef]
  162. Andrejew, R.; Oliveira-Giacomelli, Á.; Ribeiro, D.E.; Glaser, T.; Arnaud-Sampaio, V.F.; Lameu, C.; Ulrich, H. The P2X7 Receptor: Central Hub of Brain Diseases. Front. Mol. Neurosci. 2020, 13, 124. [Google Scholar] [CrossRef]
  163. Xu, Z.; Chen, Z.; Wu, X.; Zhang, L.; Cao, Y.; Zhou, P. Distinct Molecular Mechanisms Underlying Potassium Efflux for NLRP3 Inflammasome Activation. Front. Immunol. 2020, 11, 609441. [Google Scholar] [CrossRef]
  164. Territo, P.R.; Zarrinmayeh, H. P2X7 Receptors in Neurodegeneration: Potential Therapeutic Applications From Basic to Clinical Approaches. Front. Cell. Neurosci. 2021, 15, 617036. [Google Scholar] [CrossRef] [PubMed]
  165. Monif, M.; Burnstock, G.; Williams, D.A. Microglia: Proliferation and Activation Driven by the P2X7 Receptor. Int. J. Biochem. Cell Biol. 2010, 42, 1753–1756. [Google Scholar] [CrossRef]
  166. Illes, P.; Khan, T.M.; Rubini, P. Neuronal P2X7 Receptors Revisited: Do They Really Exist? J. Neurosci. 2017, 37, 7049–7062. [Google Scholar] [CrossRef]
  167. Sidoryk-Węgrzynowicz, M.; Strużyńska, L. Astroglial and Microglial Purinergic P2X7 Receptor as a Major Contributor to Neuroinflammation during the Course of Multiple Sclerosis. Int. J. Mol. Sci. 2021, 22, 8404. [Google Scholar] [CrossRef]
  168. Leeson, H.; Chan-Ling, T.; Lovelace, M.; Brownlie, J.; Gu, B.; Weible, M. P2X7 Receptor Signaling during Adult Hippocampal Neurogenesis. Neural. Regen. Res. 2019, 14, 1684. [Google Scholar] [CrossRef] [PubMed]
  169. Beamer, E.; Gölöncsér, F.; Horváth, G.; Bekő, K.; Otrokocsi, L.; Koványi, B.; Sperlágh, B. Purinergic Mechanisms in Neuroinflammation: An Update from Molecules to Behavior. Neuropharmacology 2016, 104, 94–104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Lee, H.G.; Won, S.M.; Gwag, B.J.; Lee, Y.B. Microglial P2X 7 Receptor Expression Is Accompanied by Neuronal Damage in the Cerebral Cortex of the APP swe /PS1dE9 Mouse Model of Alzheimer’s Disease. Exp. Mol. Med. 2011, 43, 7. [Google Scholar] [CrossRef] [Green Version]
  171. Francistiová, L.; Bianchi, C.; Di Lauro, C.; Sebastián-Serrano, Á.; de Diego-García, L.; Kobolák, J.; Dinnyés, A.; Díaz-Hernández, M. The Role of P2X7 Receptor in Alzheimer’s Disease. Front. Mol. Neurosci. 2020, 13, 94. [Google Scholar] [CrossRef]
  172. Parvathenani, L.K.; Tertyshnikova, S.; Greco, C.R.; Roberts, S.B.; Robertson, B.; Posmantur, R. P2X7 Mediates Superoxide Production in Primary Microglia and Is Up-Regulated in a Transgenic Mouse Model of Alzheimer’s Disease. J. Biol. Chem. 2003, 278, 13309–13317. [Google Scholar] [CrossRef] [Green Version]
  173. McLarnon, J.G.; Ryu, J.K.; Walker, D.G.; Choi, H.B. Upregulated Expression of Purinergic P2X 7 Receptor in Alzheimer Disease and Amyloid-β Peptide-Treated Microglia and in Peptide-Injected Rat Hippocampus. J. Neuropathol. Exp. Neurol. 2006, 65, 1090–1097. [Google Scholar] [CrossRef] [Green Version]
  174. Ollà, I.; Santos-Galindo, M.; Elorza, A.; Lucas, J.J. P2X7 Receptor Upregulation in Huntington’s Disease Brains. Front. Mol. Neurosci. 2020, 13, 567430. [Google Scholar] [CrossRef] [PubMed]
  175. Solini, A.; Rossi, C.; Santini, E.; Giuntini, M.; Raggi, F.; Parolini, F.; Biancalana, E.; Del Prete, E.; Bonuccelli, U.; Ceravolo, R. P2X7 Receptor/NLRP3 Inflammasome Complex and A-synuclein in Peripheral Blood Mononuclear Cells: A Prospective Study in Neo-diagnosed, Treatment-naïve Parkinson’s Disease. Eur. J. Neurol. 2021, 28, 2648–2656. [Google Scholar] [CrossRef] [PubMed]
  176. University of Pisa. P2X7 Receptor, Inflammation and Neurodegenerative Diseases (NeuroInfiam). Available online: https://clinicaltrials.gov/ct2/show/NCT03918616 (accessed on 2 May 2023).
  177. Wang, Y.; Shan, Z.; Zhang, L.; Fan, S.; Zhou, Y.; Hu, L.; Wang, Y.; Li, W.; Xiao, Z. P2X7R/NLRP3 Signaling Pathway-Mediated Pyroptosis and Neuroinflammation Contributed to Cognitive Impairment in a Mouse Model of Migraine. J. Headache Pain 2022, 23, 75. [Google Scholar] [CrossRef] [PubMed]
  178. Grygorowicz, T.; Wełniak-Kamińska, M.; Strużyńska, L. Early P2X7R-Related Astrogliosis in Autoimmune Encephalomyelitis. Mol. Cell. Neurosci. 2016, 74, 1–9. [Google Scholar] [CrossRef] [PubMed]
  179. Abu El-Asrar, A.M.; Ahmad, A.; Allegaert, E.; Siddiquei, M.M.; Alam, K.; Gikandi, P.W.; De Hertogh, G.; Opdenakker, G. Galectin-1 Studies in Proliferative Diabetic Retinopathy. Acta Ophthalmol. 2020, 98, e1–e12. [Google Scholar] [CrossRef] [PubMed]
  180. Abreu, C.A.; De Lima, S.V.; Mendonça, H.R.; Oliveira Goulart, C.d.; Blanco Martinez, A.M. Absence of Galectin-3 Promotes Neuroprotection in Retinal Ganglion Cells after Optic Nerve Injury. Histol. Histopathol. 2017, 32, 253–262. [Google Scholar] [CrossRef]
  181. Trotta, M.C.; Petrillo, F.; Gesualdo, C.; Rossi, S.; Corte, A.D.; Váradi, J.; Fenyvesi, F.; D’Amico, M.; Hermenean, A. Effects of the Calix[4]Arene Derivative Compound OTX008 on High Glucose-Stimulated ARPE-19 Cells: Focus on Galectin-1/TGF-β/EMT Pathway. Molecules 2022, 27, 4785. [Google Scholar] [CrossRef]
  182. Sugiyama, T.; Kobayashi, M.; Kawamura, H.; Li, Q.; Puro, D.G. Enhancement of P2X7-Induced Pore Formation and Apoptosis: An Early Effect of Diabetes on the Retinal Microvasculature. Investig. Ophthalmol. Vis. Sci. 2004, 45, 1026. [Google Scholar] [CrossRef] [Green Version]
  183. Kong, H.; Zhao, H.; Chen, T.; Song, Y.; Cui, Y. Targeted P2X7/NLRP3 Signaling Pathway against Inflammation, Apoptosis, and Pyroptosis of Retinal Endothelial Cells in Diabetic Retinopathy. Cell Death Dis. 2022, 13, 336. [Google Scholar] [CrossRef]
  184. Platania, C.B.M.; Giurdanella, G.; Di Paola, L.; Leggio, G.M.; Drago, F.; Salomone, S.; Bucolo, C. P2X7 Receptor Antagonism: Implications in Diabetic Retinopathy. Biochem. Pharmacol. 2017, 138, 130–139. [Google Scholar] [CrossRef]
  185. Hase, K.; Kanda, A.; Noda, K.; Ishida, S. Increased Plasma Galectin-1 Correlates with Advanced Glycation End Products and Interleukin-1β in Patients with Proliferative Diabetic Retinopathy. Int. J. Ophthalmol. 2019, 12, 692. [Google Scholar] [CrossRef]
  186. Kanda, A.; Noda, K.; Saito, W.; Ishida, S. Aflibercept Traps Galectin-1, an Angiogenic Factor Associated with Diabetic Retinopathy. Sci. Rep. 2015, 5, 17946. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Ridano, M.E.; Subirada, P.V.; Paz, M.C.; Lorenc, V.E.; Stupirski, J.C.; Gramajo, A.L.; Luna, J.D.; Croci, D.O.; Rabinovich, G.A.; Sánchez, M.C. Galectin-1 Expression Imprints a Neurovascular Phenotype in Proliferative Retinopathies and Delineates Responses to Anti-VEGF. Oncotarget 2017, 8, 32505–32522. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Mendonça, H.R.; Carvalho, J.N.A.; Abreu, C.A.; Mariano de Souza Aguiar dos Santos, D.; Carvalho, J.R.; Marques, S.A.; da Costa Calaza, K.; Martinez, A.M.B. Lack of Galectin-3 Attenuates Neuroinflammation and Protects the Retina and Optic Nerve of Diabetic Mice. Brain Res. 2018, 1700, 126–137. [Google Scholar] [CrossRef]
  189. Canning, P.; Glenn, J.V.; Hsu, D.K.; Liu, F.-T.; Gardiner, T.A.; Stitt, A.W. Inhibition of Advanced Glycation and Absence of Galectin-3 Prevent Blood-Retinal Barrier Dysfunction during Short-Term Diabetes. Exp. Diabetes Res. 2007, 2007, 51837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  190. Oncoethix GmbH. A Phase I, First-in-Man Study of OTX008 Given Subcutaneously as a Single Agent to Patients With Advanced Solid Tumors. Available online: https://clinicaltrials.gov/ct2/show/NCT01724320 (accessed on 2 May 2023).
  191. Galectin Therapeutics Inc. Phase 2 Study to Evaluate Non-Invasive Imaging Methods in Efficacy Assessment of GR-MD-02 for the Treatment of Liver Fibrosis in Patients With NASH With Advanced Fibrosis. 2020. Available online: https://clinicaltrials.gov/ct2/show/NCT02421094 (accessed on 2 May 2023).
  192. Galecto Biotech AB. A Study to Evaluate the Safety, Tolerability, Pharmacokinetics and Pharmacodynamics of Orally Administered GB1211 in Participants With Suspected or Confirmed Non-Alcoholic Steatohepatitis (NASH) and Liver Fibrosis. 2020. Available online: https://clinicaltrials.gov/ct2/show/NCT04607655 (accessed on 2 May 2023).
  193. Galecto Biotech. A Single and Repeat Dose Trial in Participants with Hepatic Impairment. 2021. Available online: https://clinicaltrials.gov/ct2/show/NCT05009680 (accessed on 2 May 2023).
  194. A Study to Investigate the Safety and Efficacy of GB1211 (a Galectin-3 Inhibitor) in Combination with Atezolizumab in Patients With Non-Small Cell Lung Cancer (NSCLC). Available online: https://clinicaltrials.gov/ct2/show/NCT05240131 (accessed on 2 May 2023).
  195. Galectin Inhibitor (GR-MD-02) and Ipilimumab in Patients With Metastatic Melanoma. Available online: https://clinicaltrials.gov/ct2/show/NCT02117362 (accessed on 2 May 2023).
  196. GR-MD-02 + Pembrolizumab Versus Pembrolizumab Monotherapy in Melanoma and Squamous Cell Head and Neck Cancer Patients. Available online: https://clinicaltrials.gov/ct2/show/NCT04987996 (accessed on 2 May 2023).
  197. GR-MD-02 Plus Pembrolizumab in Melanoma, Non-Small Cell Lung Cancer, and Squamous Cell Head and Neck Cancer Patients. Available online: https://clinicaltrials.gov/ct2/show/NCT02575404 (accessed on 2 May 2023).
  198. Galecto Biotech AB. A Study to Test the Efficacy and Safety of Inhaled GB0139 in Subjects With Idiopathic Pulmonary Fibrosis (IPF). 2019. Available online: https://clinicaltrials.gov/ct2/show/NCT03832946 (accessed on 2 May 2023).
  199. Galecto Biotech AB. A Placebo-Controlled RCT in HV’s Investigating the Safety, Tolerability and PK (Pharmacokinetic) of TD139, a Galectin-3 Inhibitor, Followed by an Expansion Cohort Treating Subjects With Idiopathic Pulmonary Fibrosis (IPF). 2021. Available online: https://clinicaltrials.gov/ct2/show/NCT02257177 (accessed on 2 May 2023).
  200. Galectin-3 Blockade in Patients With High Blood Pressure. Available online: https://clinicaltrials.gov/ct2/show/NCT01960946 (accessed on 2 May 2023).
  201. Massachusetts General Hospital. Blocking Extracellular Galectin-3 in Patients With Osteoarthritis. 2016. Available online: https://clinicaltrials.gov/ct2/show/NCT02800629 (accessed on 2 May 2023).
  202. GlaxoSmithKline. First Time in Human Study Evaluating the Safety, Tolerability, Pharmacokinetics, Pharmacodynamics and the Effect of Food of Single Assending Doses of GSK1482160. 2009. Available online: https://clinicaltrials.gov/ct2/show/results/NCT00849134 (accessed on 2 May 2023).
  203. AstraZeneca. A Study to Assess Co-Administered AZD9056 (Steady State) and Simvastatin (Single Dose) in Healthy Volunteers. 2008. Available online: https://www.clinicaltrials.gov/ct2/show/NCT00736606 (accessed on 2 May 2023).
  204. AstraZeneca. Cross-over Study to Investigate Retinal Function Following Administration of a Single Dose of AZD9056. 2008. Available online: https://www.clinicaltrials.gov/ct2/show/NCT00700986 (accessed on 2 May 2023).
  205. Pfizer. Study of Controlled Release Formulations of CE-224,535 Against the Immediate Release Formulation in Normal Volunteers. 2008. Available online: https://clinicaltrials.gov/ct2/show/NCT00782600 (accessed on 2 May 2023).
  206. Pfizer. A Study in Normal Healthy People, Testing Different Versions of a Pill That Will Be Used to Treat Rheumatoid Arthritis. 2009. Available online: https://clinicaltrials.gov/ct2/show/NCT00838058 (accessed on 2 May 2023).
  207. Pfizer. Study of CE-224,535 A Twice Daily Pill to Control Rheumatoid Arthritis in Patients Who Have not Totally Improved with Methotrexate. 2008. Available online: https://clinicaltrials.gov/ct2/show/NCT00628095 (accessed on 2 May 2023).
  208. AstraZeneca. A 6-Month Randomised, Double-Blind, Open Arm Comparator, Phase IIb, with AZD9056, in Patients with Rheumatoid Arthritis (RA). 2007. Available online: https://clinicaltrials.gov/ct2/show/NCT00520572 (accessed on 2 May 2023).
  209. Pfizer. A Study of the Effect of CE-224535 on Knee OA (Osteoarthritis) Pain. Available online: https://clinicaltrials.gov/ct2/show/NCT00418782 (accessed on 2 May 2023).
  210. Liou, G.I. Diabetic Retinopathy: Role of Inflammation and Potential Therapies for Anti-Inflammation. World J. Diabetes 2010, 1, 12–18. [Google Scholar] [CrossRef]
  211. Campbell, M.; Humphries, P. The Blood-Retina Barrier: Tight Junctions and Barrier Modulation. Adv. Exp. Med. Biol. 2012, 763, 70–84. [Google Scholar]
  212. Sugiyama, T. Role of P2X 7 Receptors in the Development of Diabetic Retinopathy. World J. Diabetes 2014, 5, 141–145. [Google Scholar] [CrossRef]
  213. Carver, K.A.; Lin, C.M.; Bowes Rickman, C.; Yang, D. Lack of the P2X7 Receptor Protects against AMD-like Defects and Microparticle Accumulation in a Chronic Oxidative Stress-Induced Mouse Model of AMD. Biochem. Biophys. Res. Commun. 2017, 482, 81–86. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Involvement of galectins in ER stress. ATF6: activating transcription factor; ER: endoplasmic reticulum; Gal-1: galectin 1; Gal-3: galectin 3; IRE1: inositol-requiring kinase 1; NLRP3: nucleotide-binding domain, leucine-rich–containing family, pyrin domain–containing-3 inflammasome; PERK: protein kinase R-like endoplasmic reticulum kinase; ROS: reactive oxygen species. Arrow: increase. Created with BioRender.com, accessed on 2 May 2023.
Figure 1. Involvement of galectins in ER stress. ATF6: activating transcription factor; ER: endoplasmic reticulum; Gal-1: galectin 1; Gal-3: galectin 3; IRE1: inositol-requiring kinase 1; NLRP3: nucleotide-binding domain, leucine-rich–containing family, pyrin domain–containing-3 inflammasome; PERK: protein kinase R-like endoplasmic reticulum kinase; ROS: reactive oxygen species. Arrow: increase. Created with BioRender.com, accessed on 2 May 2023.
Ijms 24 09721 g001
Figure 2. Involvement of P2X7R in NLRP3 activation. ASC: apoptosis-associated speck-like protein; ATP: adenosine triphosphate; Ca2+: Calcium; IL-1β: interleukin 1 beta; IL-18: interleukin 18; P2X7R: purinergic 2X7 receptor; ROS: reactive oxygen species; Na+: sodium; NLRP3: nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 inflammasome; K+: potassium. Arrow: increase. Created with BioRender.com, accessed on 2 May 2023.
Figure 2. Involvement of P2X7R in NLRP3 activation. ASC: apoptosis-associated speck-like protein; ATP: adenosine triphosphate; Ca2+: Calcium; IL-1β: interleukin 1 beta; IL-18: interleukin 18; P2X7R: purinergic 2X7 receptor; ROS: reactive oxygen species; Na+: sodium; NLRP3: nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 inflammasome; K+: potassium. Arrow: increase. Created with BioRender.com, accessed on 2 May 2023.
Ijms 24 09721 g002
Figure 3. Diagram summarizing galectins, P2X7R and their inhibition of the ERS-NLRP3 inflammasome. ERS: endoplasmic reticulum stress; Gal-1: galectin 1; Gal-3: galectin 3; P2X7R: P2X7 receptor; NLRP3: nucleotide-binding domain, leucine-rich–containing family, pyrin domain–containing-3 inflammasome; NPDR: non-proliferative diabetic retinopathy; PDR: proliferative diabetic retinopathy; ROS: reactive oxygen species; UPR: unfolded protein response. Black arrow: progression; green arrow: increase; red arrow: decrease. Created with BioRender.com, accessed on 2 May 2023.
Figure 3. Diagram summarizing galectins, P2X7R and their inhibition of the ERS-NLRP3 inflammasome. ERS: endoplasmic reticulum stress; Gal-1: galectin 1; Gal-3: galectin 3; P2X7R: P2X7 receptor; NLRP3: nucleotide-binding domain, leucine-rich–containing family, pyrin domain–containing-3 inflammasome; NPDR: non-proliferative diabetic retinopathy; PDR: proliferative diabetic retinopathy; ROS: reactive oxygen species; UPR: unfolded protein response. Black arrow: progression; green arrow: increase; red arrow: decrease. Created with BioRender.com, accessed on 2 May 2023.
Ijms 24 09721 g003
Table 1. Galectins in DR studies.
Table 1. Galectins in DR studies.
StudyMain ResultsTreatmentsReference
Clinical
Vitreous fluid samples from 36
patients with PDR
(20 patients undergoing
vitrectomy as controls)
VEGF and Gal-1 levels significantly higher in PDR patients compared with controls-[179]
Clinical
Plasma samples from
20 PDR cases
(20 non-diabetic, idiopathic
macular diseases as controls)
Plasma levels of Gal-1, AGEs and IL-1β significantly
increased in PDR patients
-[185]
Clinical
23 vitreous fluids aspirated from PDR
(non-diabetic control eyes with
idiopathic ERM
and MH)
Neovascular ocular tissues
surgically excised from PDR
patients
(non-diabetic tissues as
controls)
Gal-1 levels were
significantly elevated in the vitreous fluids of PDR eyes compared with
controls
Gal-1 upregulated in PDR
tissues and co-localized with VEGFR2
-[186]
In vivo
STZ-Sprague Dawley rats
(non-diabetic rats as controls)
STZ:55 mg/kg
(single dose, i.v. injection)
Vehicle: 10 mM sodium citrate buffer (single dose i.v. injection)
Retinal Gal-1 levels
increased in diabetic rats compared with controls
-[179]
In vivo
Gal-3 knockout
C57/BL6 mice
(wild-type mice as controls)
Gal-3 knockout mice
exhibited less activated
inflammatory cells within the optic nerve after crush
-[180]
In vivo
Diabetic Gal-3 knockout
C57/BL6 mice
(wild-type diabetic mice
as controls)
STZ:160 mg/kg
(single dose, i.p. injection)
Vehicle: sodium citrate buffer
(single dose i.p. injection)
Gal-3 knockout reduced RGC apoptosis, Iba-1 and GFAP in the distal optic nerve in
diabetic mice; moreover, it
prevented the loss of
myelinated fibers
-[188]
In vivo
Diabetic Gal-3 knockout
C57/BL6 mice
(wild-type diabetic mice
as controls)
STZ:165 mg/kg
(single dose, i.p. injection)
Vehicle: sodium citrate buffer
(single dose i.p. injection)
Gal-3 knockout reduced
AGEs, VEGF and BRB
breakdown in diabetic mice
-[189]
In vitro
Hypoxic (CoCl2) human
retinal Müller glial cells
(non-hypoxic cells as controls)
CoCl2: 300 μM for 24 h
OTX008 attenuated the
upregulation of Gal-1, VEGF and NF-κB in
hypoxic retinal Müller cells
Gal-1 selective inhibitor OTX008
OTX008: 10 μM for 24 h
[179]
In vitro
ARPE-19 cells
exposed to HG
(NG cells as controls)
HG: 35 mM
NG: 5 mM
OTX008 induced a
significant increment in
cell viability;
while Gal-1 protein, ROS and TGF-β1 levels were
reduced after OTX008
Gal-1 selective inhibitor OTX008 (2.5–5–10 µM)[181]
Abbreviations. AGEs: advanced glycation end products; ARPE-19: adult retinal pigment epithelial cell line-19; BRB: blood-retinal barrier; CoCl2: cobalt chloride; ERM: epiretinal membrane; Gal-1: galectin 1; Gal-3: galectin 3; GFAP: glial fibrillary acidic protein as activated macroglia marker; h: hours; HG: high glucose; Iba-1: ionized calcium-binding adapter molecule 1; IL-1β: interleukin 1 beta; i.p.: intraperitoneal; i.v.: intravenous; MH: macular hole; NF-kB: nuclear factor kappa-light-chain-enhancer of activated B cells; NG: normal glucose; OTX008: calixarene 0118; PDR: proliferative diabetic retinopathy; RGC: retinal ganglion cells; ROS: reactive oxygen species; STZ: streptozotocin; TGF-β1: transforming growth factor beta 1; VEGF: vascular-endothelial growth factor; VEGFR2: Vascular Endothelial Growth Factor Receptor 2.
Table 2. P2X7R in DR studies.
Table 2. P2X7R in DR studies.
StudyTreatmentMain ResultsReferences
In vivo
STZ-C57BL/6J mice
(non-diabetic mice as
controls)
STZ:50 mg/kg
(daily, i.p. injection)
for 5 days
Vehicle: 0.1% mol/L citrate buffer
(daily, i.p. injection)
for 5 days
P2X7R selective inhibitor A74003 (100 µg/kg/d);
NLRP3 inflammasome
selective inhibitor MCC950 (100 µg/kg/d)
A74003: 100 µg/kg/d, i.p.
injection
every alternate day
from week 9 to week 12
MCC950: 100 µg/kg/d, i.p.
injection daily for the first 3 days
and then every alternate day
from week 9 to week 12
P2X7R mRNA
Significantly higher in the retinal tissues of STZ-mice; A74003 and MCC950
reduced retinal inflammation and apoptosis in STZ-mice
[183]
In vitro
Pericyte-containing retinal microvessels from
STZ-Long Evans rats
(non-diabetic rats as
controls)
STZ: 75 mg/kg
(single dose, i.p. injection)
Vehicle: 0.8 mL
(single dose,
citrate buffer i.p. injection)
P2X7R agonist
BzATP
BzATP: 100 µM for 24 h
BzATP (100 µM) increased the apoptosis of
pericyte-containing retinal microvessels
from STZ-rats
[182]
In vitro
mRECs exposed to HG, alone or in combination
with LPS,
(NG cells as controls)
HG: 50 mM for 24 and 48 h
LPS: 20 ng/mL for 48 h
NG: 5.5 mM for 24 and 48 h
P2X7R selective inhibitor A74003;
NLRP3 inflammasome
selective inhibitor MCC950
A74003: 100 µmol/L 6 h before
HG or HG + LPS, until 72 h
MCC950: 100 µmol/L,
6 h before
HG or HG + LPS, until 72 h
A74003 reduced apoptosis and pyroptosis in mRECS exposed to
HG combined with LPS
[183]
In vitro
Human retinal pericytes
exposed to HG
(NG cells as controls)
HG: 25 mM for 48 h
NG: 5 mM for 48 h
P2X7R agonist BzATP;
P2X7R novel antagonist JNJ47965567
BzATP: 100 µM for 48 h
JNJ47965567: 10–100 nM
for 48 h
BzATP induced human retinal pericyte cell death
and increased the
pro-inflammatory
cytokines levels;
JNJ47965567
Protected pericytes viability and reduced pro-inflammatory cytokines
[184]
Abbreviations: BzATP: benzoylbenzoyl-adenosine triphosphate; h: hours; HG: high glucose; i.p.: intraperitoneal; LPS: lipopolysaccharide; mRECs: mice retinal endothelial cells; mRNA: messenger ribonucleic acid; NG: normal glucose; NLRP3: nucleotide-binding domain, leucine-rich–containing family, pyrin domain–containing-3 inflammasome; P2X7R: purinergic 2x7 receptors; STZ: streptozotocin.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lepre, C.C.; Russo, M.; Trotta, M.C.; Petrillo, F.; D’Agostino, F.A.; Gaudino, G.; D’Amico, G.; Campitiello, M.R.; Crisci, E.; Nicoletti, M.; et al. Inhibition of Galectins and the P2X7 Purinergic Receptor as a Therapeutic Approach in the Neurovascular Inflammation of Diabetic Retinopathy. Int. J. Mol. Sci. 2023, 24, 9721. https://doi.org/10.3390/ijms24119721

AMA Style

Lepre CC, Russo M, Trotta MC, Petrillo F, D’Agostino FA, Gaudino G, D’Amico G, Campitiello MR, Crisci E, Nicoletti M, et al. Inhibition of Galectins and the P2X7 Purinergic Receptor as a Therapeutic Approach in the Neurovascular Inflammation of Diabetic Retinopathy. International Journal of Molecular Sciences. 2023; 24(11):9721. https://doi.org/10.3390/ijms24119721

Chicago/Turabian Style

Lepre, Caterina Claudia, Marina Russo, Maria Consiglia Trotta, Francesco Petrillo, Fabiana Anna D’Agostino, Gennaro Gaudino, Giovanbattista D’Amico, Maria Rosaria Campitiello, Erminia Crisci, Maddalena Nicoletti, and et al. 2023. "Inhibition of Galectins and the P2X7 Purinergic Receptor as a Therapeutic Approach in the Neurovascular Inflammation of Diabetic Retinopathy" International Journal of Molecular Sciences 24, no. 11: 9721. https://doi.org/10.3390/ijms24119721

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop