Next Article in Journal
The Targeting of Native Proteins to the Endoplasmic Reticulum-Associated Degradation (ERAD) Pathway: An Expanding Repertoire of Regulated Substrates
Next Article in Special Issue
Pathophysiological Roles of Histamine Receptors in Cancer Progression: Implications and Perspectives as Potential Molecular Targets
Previous Article in Journal
Development and Evaluation of Gellan Gum/Silk Fibroin/Chondroitin Sulfate Ternary Injectable Hydrogel for Cartilage Tissue Engineering
Previous Article in Special Issue
TRPV1 and TRPA1 Channels Are Both Involved Downstream of Histamine-Induced Itch
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Novel Insight of Histamine and Its Receptor Ligands in Glaucoma and Retina Neuroprotection

1
Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy
2
Toxicology Unit, Emergency Department, Careggi University Hospital, 50139 Florence, Italy
*
Author to whom correspondence should be addressed.
Biomolecules 2021, 11(8), 1186; https://doi.org/10.3390/biom11081186
Submission received: 19 July 2021 / Revised: 6 August 2021 / Accepted: 9 August 2021 / Published: 11 August 2021
(This article belongs to the Special Issue New Developments in Histamine Research)

Abstract

:
Glaucoma is a multifactorial neuropathy characterized by increased intraocular pressure (IOP), and it is the second leading cause of blindness worldwide after cataracts. Glaucoma combines a group of optic neuropathies characterized by the progressive degeneration of retinal ganglionic cells (RGCs). Increased IOP and short-term IOP fluctuation are two of the most critical risk factors in glaucoma progression. Histamine is a well-characterized neuromodulator that follows a circadian rhythm, regulates IOP and modulates retinal circuits and vision. This review summarizes findings from animal models on the role of histamine and its receptors in the eye, focusing on the effects of histamine H3 receptor antagonists for the future treatment of glaucomatous patients.

1. Introduction

Glaucoma is a group of optic multifactorial neuropathies characterized by the progressive degeneration of retinal ganglionic cells (RGCs). These are central nervous system (CNS) neurons with cell bodies in the inner retina and axons in the optic nerve. Degeneration of the optic nerve results in progressive, permanent vision loss, starting with hidden blind spots at the edges of the visual field, progressing to tunnel vision, and finally to blindness [1]. The biological basis of glaucoma is poorly understood, and the factors contributing to its progression have not been fully characterized [2].
Glaucoma represents a significant public health problem. It is the second leading cause of blindness worldwide after cataracts, and the global prevalence of glaucoma in the population aged 40 to 80 years is 3.5% [3].
There are different types of glaucoma: hypertensive and normotensive glaucoma are the most common. In the first, elevated intraocular pressure (IOP) is caused by an imbalance between the production and outflow of aqueous humors (AH). In the second, the optic nerve head becomes damaged even though IOP is within the normal range. We assist with a reduction in ocular perfusion through less oxygen and nutrients supplied to the optic nerve. Physiologic IOP is generally within a range between 10 and 20 mmHg to ensure constant corneal curvature. This pressure is determined by the volume of AH within the eye exerting an outward pressure, scleral compliance and extraocular muscle tone exerting inward pressure [4].
The increased intraocular pressure with altered IOP circadian waves, if untreated, can reduce local perfusion, recurrent ischemic insults, and ultimately reduces the number of RGCs by autophagy and apoptosis [5]. When the number of RGCs is no longer suitable for neural transmission, the visual field becomes more and more narrow. Several mechanisms have been proposed to explain nerve head damage in glaucoma. Elevation of IOP and axial shallowing of the anterior chamber from posterior pressure characterize AH misdirection; however, the precise mechanism of increased resistance to AH outflow remains unclear, and is currently an active focus of research [6,7,8,9].
The histaminergic system in the eye may exert modulatory functions on visual activity and modulate IOP homeostasis and circadian fluctuations. Along with other factors, the dysregulation of histamine secretion may thus contribute to the pathophysiology of glaucoma.

2. Mechanism of Damage of Optic Nerve

Raised IOP is thought to damage the optic nerve head via induced mechanical changes at the lamina cribrosa or via vascular dysfunction and resultant ischemia. Several mechanisms are postulated to cause elevated IOP, the majority of which are related to reduced AH outflow. Structural changes include:
  • Trabecular meshwork obstruction by foreign material (e.g., glycosaminoglycans, pigments, red blood cells);
  • Trabecular cell loss of phagocytic activity and death;
  • Loss of giant vacuoles from Schlemm’s canal endothelium and reduced pore size and density in the wall of this canal.
These changes could be brought on by altered endogenous corticosteroid metabolism with secondary trabecular meshwork changes and IOP rise [10].
Following isolation of the Myocilin (MYOC) gene, the clinical manifestation of glaucoma has been associated with a genetic predisposition. This gene, previously known as the trabecular meshwork inducible glucocorticoid response (TIGR) gene [11], was identified in pedigrees with juvenile open-angle glaucoma (OAG), a term used to refer to primary open-angle glaucoma (POAG) earlier age at onset, and an autosomal dominant mode of inheritance [12]. In recent years, the role of the MYOC gene has been extensively studied. The sequence variations in the gene account for approximately 2% to 4% of glaucoma cases. One particular MYOC mutation, Gln368Stop (dbSNP accession number: rs74315329), is the most common genetic mutation, causing glaucoma by increasing IOP [13].
It has recently been demonstrated that the integrity of Schlemm’s canal is maintained by coordinated functions of angiopoietin-Tie2 (Angpt-Tie2) signaling, and TIE2 mutations have been identified in patients with POAG [14]. The baro-protection remains the most important therapeutic approach for preserving visual function in glaucoma patients; however, in recent years, a normal retinal and choroidal vascular perfusion and neurotrophic factor preservation have arisen as important [15].
Multiple treatments for reducing ocular hypertension and glaucoma exist and are chiefly separated into pharmacological, laser, and surgical therapies [10,16,17]. The pharmacological treatment of glaucoma includes drugs of various classes, generally administered as eye drops: prostaglandin analogues, β-blockers, carbonic anhydrase inhibitors, α-adrenergic agonists, miotics, hyperosmotic agents, and recently, Rho-kinase inhibitors [18]. These drugs can be administered singularly or in association, but a significant number of patients are “non-responders” to these treatments even when they are administered in combination. The therapeutic options available for lowering IOP, especially relating to the enhancement of AH outflow through the trabecular pathway, are limited, and in addressing this challenge, novel therapeutic approaches have been the focus. The current review presents an overview of the role of histamine in glaucoma and the future use of histaminergic drugs to treat glaucomatous patients.

3. The Histaminergic System

In contrast with other biogenic amines, histamine has garnered less attention because of its moderate action in the CNS. However, recent evidence suggests that histamine plays an essential role in multiple neuronal disorders, including insomnia, narcolepsy, Parkinson’s diseases, schizophrenia, Alzheimer’s disease, and cerebral ischemia [19,20]. In the CNS, histamine is produced within mast cells and neurons [21]. Histidine decarboxylase (HDC) is the rate-limiting enzyme in the formation of histamine, catalyzing the synthesis of histamine from the amino acid L-histidine. Within the CNS, the tuberomamillary nucleus of the hypothalamus is the leading site for the neuronal synthesis of histamine. Four histamine receptors (H1R–H4R) have been identified, and these receptors induce neuronal effects via G-protein coupled signaling mechanisms.
Histamine H1 receptor (H1R) is ubiquitously expressed, particularly in lungs, the CNS and blood vessels. It couples to Gαq/11 proteins, triggering phospholipase C (PLC) and protein kinase C (PKC) activation, together with inositol-1,4–5-trisphosphate (IP3) formation and intracellular Ca2+ release [22].
Histamine H1R activation can provoke a type I allergic reaction, whose typical signs are pruritus, increased vascular permeability and edema. Consequently, the administration of H1R antagonists (antihistamines) is the most important anti-allergic therapeutic intervention [23]. In the CNS, H1R is involved in cognitive functions, locomotor activity, emotions, arousal, circadian rhythm, sleep, or pain perception [24]. The most important side effect of brain-penetrating first-generation antihistamines is sedation, and it is caused by antagonism at H1R in the CNS [23,25].
Histamine H2R is ubiquitously expressed, particularly in the stomach, heart, and CNS [22,25]. In 1977, Sir James Black’s research group released the H2R antagonist cimetidine as the first efficient pharmacological treatment for gastroesophageal reflux disease, dyspepsia, gastric or duodenal ulcers. Agonist binding to H2R results in the activation of Gαs-proteins that stimulate the adenylyl-cyclase-mediated production of the second messenger cAMP. In the brain, functions of the H2R include the modulation of cognitive processes, circadian rhythm, food intake and glucose metabolism [24]. Experiments with knockout mice have shown that the histamine H2R is implicated in modifying the immune responses, specifically in the modulation of Th1- or Th2-cell polarization [26].
Histamine H3R is primarily expressed in neurons and acts as a presynaptic auto- and hetero-receptor. It inhibits the release of histamine [27,28] and neurotransmitters, such as acetylcholine, dopamine, noradrenaline or glutamate [29]. Histamine H3R is considered a potential target for treating cerebral disorders [30]; furthermore, it is expressed post-synaptically in the basal ganglia and within the dorsal and ventral striatum [31]. Histamine H3R is coupled with Gi/o protein. It inhibits adenylyl cyclase and the high-voltage-activated Ca2+-channels, which regulate histamine synthesis and neurotransmitter release. The inverse H3R agonist pitolisant is currently used to treat narcoleptic patients [32].
Histamine H4R, discovered in 2000, is the most recently identified histamine receptor. This discovery culminated with six independent groups reporting the cloning of the histamine H4R [22]. This receptor modulates the migration and activation of a wide spectrum of immune cells (basophils, mast cells, eosinophils, dendritic cells, monocytes, NK, iNK T and γδ cells, CD8+ T cells, Treg, and Th2 cells), and it is thus involved in allergic and immune-mediated disorders.
Histamine H4R antagonists prevent mast cell chemotaxis and submucosal accumulation in the trachea of mice after histamine inhalation [33], lung inflammation and fibrosis induced by bleomycin [34,35]. This receptor is expressed in the hippocampus, granular layer of the cerebellar cortex, thalamus, and spinal cord [36]. Recently, observations conducted on histamine H4R knockout mice [37] have shown that H4R modulates locomotor activity, anxiety, nociception and feeding behavior, confirming the role of the histaminergic regulation of neuronal functions and in neuropathic pain [38]. Dysregulation of brain histamine is a potential contributor to neuropsychiatric [29] and Parkinson’s [19] diseases, as well as in Tourette syndrome, a rare genetic disease [39].

4. The Histaminergic System at Ocular Level

Retina, optic nerve and various brain structures of albino and pigmented rabbits contain histamine in the range of 40–400 ng/g of tissue; choroid tissue of both animal strains is characterized by amine contents several times higher. In the retina, no histamine-forming cells have been identified to date; however, retinopetal axons arising from the tuberomamillary nucleus extend across the inner plexiform layer, eliciting responses in a range of inner retinal neurons [40]. The synthesis and release of histamine are controlled by presynaptic histamine H3 auto-receptors located in the CNS [27,28]. Histamine H1, H2 and H3R have been localized in the inner layer of ganglion cells in rodents and primate retinae [40,41]; in particular, circuits involved in scotopic vision may be altered by histamine release. In the macaque retina, the stimulation of histamine H3R increases the delayed rectifier component of the voltage-dependent potassium conductance in ON bipolar cells [42], and in dark-adapted baboon retinas, histamine decreases the rate of maintained firing and the amplitude of the light responses of ON ganglion cells [43]. The primate retina receives input from histaminergic neurons that are active during the day in the posterior hypothalamus, and they receive input from the brain via axons emerging from the optic nerve. One set of retinopetal axons arises from perikarya in the posterior hypothalamus and uses histamine, whereas in the dorsal raphe serotonin is utilized. These histaminergic and serotonergic neurons are not specialized to supply the retina; they are neurons’ subsets that project via collaterals to many other targets in the CNS. They are components of the ascending arousal system, active when the animal is awake. Many of the effects of histamine and serotonin on light responses suggest that retinopetal axons optimize retinal function at an ambient light intensity during the waking period.
Histamine activates chloride channels, increasing chloride conductance, in the monopolar optic cells in insects, suggesting a role of histamine in photoreceptor response to light in flies [44]; moreover, severe mutation in the gene encoding for histamine receptors causes a defect in the transients of the electroretinogram derived from large monopolar cells of the first optic lamina [45]. Histamine reduces the amplitude of light responses in monkey RGCs, a finding consistent with a role for retinopetal axons in light adaptation in these diurnal animals [46]. These findings suggest that histamine acts primarily via volume transmission in the primate retina, increasing the operating range of cones and conserving ATP in bright, ambient light. The histaminergic system is deeply implicated in circadian rhythm and fulfils a significant role in maintaining waking [47]. During the day, the histaminergic tone could play a role in maintaining the IOP balance. Furthermore, histamine is responsible for ciliary muscle contraction in human eyes and, therefore, in IOP reduction [48].
Ocular administration of histamine triggers local inflammation in a non-specific manner; the severity of conjunctivitis is dose-dependent; however, histamine is well tolerated, although transient blepharitis, aqueous flare, and ocular hypertension occur in some experimental situations [49]. Total protein content and serum albumin levels increase after histamine administration, as are lacrimal albumin levels during naturally acquired conjunctivitis; lacrimal albumin concentration decreases in parallel with the reduction in the conjunctivitis score [49]. In the conjunctival immediate hypersensitivity reaction (type I allergy), histamine is released from degranulated mast cells in the early and late phase [50]; histamine levels in the tears of kerato-conjunctivitis (KCV) patients are higher than in control subjects, and histamine H1 receptors are over-expressed in the active phase of the disease [51].
Histamine H1Rs are localized on horizontal cells and in a small number of amacrine cells, whereas histamine H2Rs appear closely associated with synaptic ribbons inside cone pedicles [41]. Histaminergic H1 and H2 receptors in the iris arterioles and H2 receptors in the iridal venules modulate vascular tone in rats [52]. Moreover, several ocular hypertensive effects have been reported in chronic glaucoma patients following the use of cimetidine and ranitidine, two H2R antagonists, for peptic ulcer treatment [53]; on the contrary, recent studies have failed to demonstrate the significant action of topical administered H2 blockers on IOP in humans [54]. Histamine H1 and H2R antagonists possess anticholinergic activity that may induce glaucoma. Promethazine, an antipsychotic with antihistamine activity, has been shown to produce an idiopathic swelling of the lens that could increase the risk of angle-closure glaucoma. Topical administration of ranitidine produces vasoconstriction in both the arterioles and the venules of the iris, suggesting a predominant role of histamine H2R in the vasculature of the iris [52].
Histamine H4 receptors are expressed by various inflammatory cells, including eosinophils and Th2 cells, in allergic disorders [55], and infiltrating inflammatory cells in subconjunctival tissues of KCV patients strongly express H4R [56]. H4Rs are mainly expressed in immune cells; thus, their down-regulation may lead to a decreased eosinophil infiltration into the conjunctival tissue. Therefore, the expression level of H4R on the ocular surface may be a useful biomarker for atopic KCV in clinical examinations [51].
Histamine tone is decreased at night, and nocturnal IOP is higher than diurnal pressure. IOP results from a balance between secretion and outflow of AH [57]. This balance is entirely controlled in healthy subjects, with three crucial mechanisms: the rate of AH formation, the resistance to the outflow, and episcleral venous pressure. These factors change during the night, the AH production decreases significantly in diurnal mammalians, as does the drainage; thus, the IOP increases [58]. Histamine regulates ciliary muscle contraction in human eyes and, therefore, it controls IOP reduction [48]. Moreover, AH production or outflow rate can be influenced by histamine, which is the neurotransmitter of the ascending arousal system, and the contributions of retinopetal axons to visions can be predicted from the known effects of histamine on retinal neurons.

5. The Role of Histamine H3 Receptors in the Control of Intraocular Pressure

The major drainage pathway of AH consists of structures located in the angular region of the anterior chamber of the eye, such as the trabecular meshwork and the Schlemm’s canal system, which are responsible for about 90% of outflow from the eye. Some accessory pathways include the uveoscleral system, with a small contribution of trans-corneal and vitreal flux [59]. Obstruction in the circulatory pathway of AH causes IOP elevation, which is a significant risk factor for glaucoma. AH production and outflow rate can be influenced by histamine. Moreover, histamine has effects on retinal neurons [46]. Previous work in our laboratory [60] reported the expression pattern of histaminergic receptors in rabbit eyes. In this work, histamine H1R and H4R expression were found in the retina and optic nerve at a higher concentration than that revealed in the trabecular meshwork and stomach, used as a positive control (Figure 1A,C). High histamine H3R protein expression levels were found in the retina, optic nerve and ciliary body (Figure 1B), whereas histamine H2R was found only in the stomach, which resulted in undetectable protein in the ocular tissues.
Several histamine H3R antagonists have been tested in animal models of glaucoma in rabbits. Both imidazole (ciproxifan) and non-imidazole compounds, such as DL-76 (1-[3-(4-tert-butylphenoxy)propyl]piperidine hydrogen oxalate, [60]) and GSK189254, at an equimolar concentration (1%) reduced IOP following a single acute challenge or after repeated doses in transient or stable IOP raise models in rabbits [60]. After 50 µL of 5% hypertonic saline injection in the eye’s anterior chamber, IOP increased from 16.8 ± 5.6 mmHg to 39.63 ± 4.85 mmHg. This value remained stable until 120 min, and decaying after that to reach baseline values at 240 min. All the compounds reduced IOP in a statistically significant manner with a different profile; ciproxifan and DL-76 were more effective than GSK189254 (Table 1A). As observed in the transient ocular hypertensive model, all the compounds, at a 1% dose, caused a significant reduction in IOP in the carbomer-induced chronic model in male New Zealand White (NZW) rabbits after seven days of treatment. Ciproxifan and DL-76 were the most effective compounds. The effect of timolol at a 1% dose, the gold standard treatment, is also reported (Table 1B).
Interestingly, the IOP-lowering activity of ciproxifan and DL-76 were suppressed by the pre-treatment with 1% imetit, a histamine H3R agonist, providing the specificity of histamine H3R antagonism action (Figure 2).
The effect of histamine H3R antagonists on the vascular performance at the posterior pole of the eye was evaluated through Doppler ultrasound studies of the ophthalmic artery. In elevated IOP eyes, the Pourcelot Resistivity Index (RI) was significantly higher compared to physiological IOP eyes [61]; the treatment with histamine H3R antagonists significantly reduced Pourcelot RI, indicating a role of histamine in controlling the vascular tone at the ocular level. It is well known that vascular impairment in the eye is important in the pathogenesis of normotensive glaucoma. Hence, chronic treatments with H3R antagonists could have a significant role in improving this kind of glaucoma [60]. The IOP control with a chronic treatment with H3R antagonists significantly prevented the cell death of neurons in the RGC layer of hypertensive rabbit eye (Figure 3). Moreover, RGC death is the leading cause of visual impairment in glaucomatous patients.
In conclusion, ocular hypertension and IOP fluctuation are the most critical risk factors in glaucoma progression. Histamine receptors are expressed in a neuronal and non-neuronal compartment in the eye, as well as in trabecular-meshwork-derived cells, and histamine H3R antagonists are effective in reducing IOP, controlling ocular vascular performance and preventing RGC death.
Recent studies have identified the importance of endothelial signaling molecules in maintaining the AH conventional outflow pathway and roles for Tie2, a tyrosine kinase receptor located on vascular endothelial cells and its ligands angiopoietin-1 and-2 (Angpt-1 and Angpt-2), have been described. In patients with primary congenital glaucoma, a severe form of the disease characterized by an early onset and a severe neuropathy, a mutation of the gene encoding Angpt-1 has been shown [14]. The deletion of ANGPT-1 and -2 or TIE2 genes in adult mice severely impaired the integrity of Schlemm’s canal, leading to IOP elevation, retinal neuron damage and RGC death, all features of glaucoma in humans [62]. The actions of histamine and H3R antagonists are present at the vascular level; therefore, an interaction between histamine and the Tie2 receptor could be postulated for the control of IOP and glaucoma progression.
In conclusion, the histaminergic system may represent a new site for multiple therapeutic interventions that could hamper the progression of glaucoma damage and visual field loss in patients and histamine H3R antagonists, a new class of IOP lowering agents with an innovative mechanism of action.

Author Contributions

Conceptualization, E.M. and C.L.; formal analysis, L.L. and S.S.; data curation, S.S.; writing—original draft preparation, E.M.; writing—review and editing, L.L and S.S.; visualization, S.S.; supervision, L.L. and S.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

All procedures on animals conformed to the Association for Research in Vision and Ophthalmology Resolution, in agreement with the Good Laboratory Practice for the use of animals, with the European Union Regulations (Directive 2010/63/EU), upon authorization of the National Ethics Committee of the Italian Ministry of Health (Authorization number 318/2018-PR).

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are available on request.

Acknowledgments

We acknowledge Francesco Impagnatiello and Elena Bastia for the critical review of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Weinreb, R.N.; Aung, T.; Medeiros, F.A. The pathophysiology and treatment of glaucoma: A review. JAMA J. Am. Med. Assoc. 2014, 311, 1901–1911. [Google Scholar] [CrossRef] [Green Version]
  2. Nickells, R.W.; Howell, G.R.; Soto, I.; John, S.W.M. Under pressure: Cellular and molecular responses during glaucoma, a common neurodegeneration with axonopathy. Annu. Rev. Neurosci. 2012, 35, 153–179. [Google Scholar] [CrossRef] [Green Version]
  3. Tham, Y.C.; Li, X.; Wong, T.Y.; Quigley, H.A.; Aung, T.; Cheng, C.Y. Global prevalence of glaucoma and projections of glaucoma burden through 2040: A systematic review and meta-analysis. Ophthalmology 2014, 121, 2081–2090. [Google Scholar] [CrossRef]
  4. Murgatroyd, H.; Bembridge, J. Intraocular pressure. Contin. Educ. Anaesth. Crit. Care Pain 2008, 8, 100–103. [Google Scholar] [CrossRef]
  5. Wang, Y.; Huang, C.; Zhang, H.; Wu, R. Autophagy in glaucoma: Crosstalk with apoptosis and its implications. Brain Res. Bull. 2015, 117, 1–9. [Google Scholar] [CrossRef]
  6. Sabanay, I.; Tian, B.B.B.; Gabelt, A.T.; Geiger, B.; Kaufman, P.L. Latrunculin B effects on trabecular meshwork and corneal endothelial morphology in monkeys *. Exp. Eye Res. 2006, 82, 2236–2246. [Google Scholar] [CrossRef] [PubMed]
  7. Stamer, W.D.; Acott, T.S. Current understanding of conventional outflow dysfunction in glaucoma. Curr. Opin. Ophthalmol. 2012, 23, 135–143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Saccà, S.C.; Gandolfi, S.; Bagnis, A.; Manni, G.; Damonte, G.; Traverso, C.E.; Izzotti, A. The Outflow Pathway: A Tissue With Morphological and Functional Unity. J. Cell. Physiol. 2016, 231, 1876–1893. [Google Scholar] [CrossRef] [PubMed]
  9. Rao, P.V.; Pattabiraman, P.P.; Kopczynski, C. Role of the Rho GTPase/Rho kinase signaling pathway in pathogenesis and treatment of glaucoma: Bench to bedside research. Exp. Eye Res. 2017, 158, 23–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Quigley, H.A. Glaucoma. In Proceedings of the Lancet; Elsevier: Amsterdam, The Netherlands, 2011; Volume 377, pp. 1367–1377. [Google Scholar]
  11. Kersey, J.P.; Broadway, D.C. Corticosteroid-induced glaucoma: A review of the literature. Eye 2006, 20, 407–416. [Google Scholar] [CrossRef] [Green Version]
  12. Johnson, A.T.; Drack, A.V.; Kwitek, A.E.; Cannon, R.L.; Stone, E.M.; Alward, W.L.M. Clinical Features and Linkage Analysis of a Family with Autosomal Dominant Juvenile Glaucoma. Ophthalmology 1993, 100, 524–529. [Google Scholar] [CrossRef]
  13. Nag, A.; Lu, H.; Arno, M.; Iglesias, A.I.; Bonnemaijer, P.; Broer, L.; Uitterlinden, A.G.; Klaver, C.C.W.; van Duijn, C.; Hysi, P.G.; et al. Evaluation of the Myocilin Mutation Gln368Stop Demonstrates Reduced Penetrance for Glaucoma in European Populations. Ophthalmology 2017, 124, 547–553. [Google Scholar] [CrossRef] [Green Version]
  14. Souma, T.; Tompson, S.W.; Thomson, B.R.; Siggs, O.M.; Kizhatil, K.; Yamaguchi, S.; Feng, L.; Limviphuvadh, V.; Whisenhunt, K.N.; Maurer-Stroh, S.; et al. Angiopoietin receptor TEK mutations underlie primary congenital glaucoma with variable expressivity. J. Clin. Investig. 2016, 126, 2575–2587. [Google Scholar] [CrossRef]
  15. Ramos, R.F.; Hoying, J.B.; Witte, M.H.; Daniel Stamer, W. Schlemm’s Canal Endothelia, Lymphatic, or Blood Vasculature? J. Glaucoma 2007, 16, 391–405. [Google Scholar] [CrossRef]
  16. Quigley, H.A.; Broman, A.T. The number of people with glaucoma worldwide in 2010 and 2020. Br. J. Ophthalmol. 2006, 90, 262–267. [Google Scholar] [CrossRef] [Green Version]
  17. Hoyng, P.F.J.; Van Beek, L.M. Pharmacological therapy for glaucoma: A review. Drugs 2000, 59, 411–434. [Google Scholar] [CrossRef] [PubMed]
  18. Sgambellone, S.; Durante, M.; Masini, E.; Lucarini, L. Up-To Date of the Pharmacological Therapies for Glaucoma Special Focus on Glaucoma Special Focus on Glaucoma. Spec. Focus Glaucoma 2020, 1, 1–18. [Google Scholar]
  19. Shan, L.; Bossers, K.; Luchetti, S.; Balesar, R.; Lethbridge, N.; Chazot, P.L.; Bao, A.M.; Swaab, D.F. Alterations in the histaminergic system in the substantia nigra and striatum of Parkinson’s patients: A postmortem study. Neurobiol. Aging 2012, 33. [Google Scholar] [CrossRef] [PubMed]
  20. Hu, W.; Chen, Z. The roles of histamine and its receptor ligands in central nervous system disorders: An update. Pharmacol. Ther. 2017, 175, 116–132. [Google Scholar] [CrossRef]
  21. Panula, P.; Nuutinen, S. The histaminergic network in the brain: Basic organization and role in disease. Nat. Rev. Neurosci. 2013, 14, 472–487. [Google Scholar] [CrossRef] [PubMed]
  22. Panula, P.; Chazot, P.L.; Cowart, M.; Gutzmer, R.; Leurs, R.; Liu, W.L.S.; Stark, H.; Thurmond, R.L.; Haas, H.L. International union of basic and clinical pharmacology. XCVIII. histamine receptors. Pharmacol. Rev. 2015, 67, 601–655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Simons, F.E.R.; Simons, K.J. Histamine and H1-antihistamines: Celebrating a century of progress. J. Allergy Clin. Immunol. 2011, 128, 1139–1150. [Google Scholar] [CrossRef] [PubMed]
  24. Schneider, E.H.; Neumann, D.; Seifert, R. Modulation of behavior by the histaminergic system: Lessons from HDC-, H3R- and H4R-deficient mice. Neurosci. Biobehav. Rev. 2014, 47, 101–121. [Google Scholar] [CrossRef]
  25. Neumann, D.; Schneider, E.H.; Seifert, R. Analysis of histamine receptor knockout mice in models of inflammation. J. Pharmacol. Exp. Ther. 2014, 348, 2–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Thangam, E.B.; Jemima, E.A.; Singh, H.; Baig, M.S.; Khan, M.; Mathias, C.B.; Church, M.K.; Saluja, R. The role of histamine and histamine receptors in mast cell-mediated allergy and inflammation: The hunt for new therapeutic targets. Front. Immunol. 2018, 9, 1873. [Google Scholar] [CrossRef] [Green Version]
  27. Arrang, J.M.; Garbarg, M.; Schwartz, J.C. Auto-inhibition of brain histamine release mediated by a novel class (H3) of histamine receptor. Nature 1983, 302, 832–837. [Google Scholar] [CrossRef] [PubMed]
  28. Arrang, J.M.; Garbarg, M.; Schwartz, J.C. Autoregulation of histamine release in brain by presynaptic H3-receptors. Neuroscience 1985, 15, 553–562. [Google Scholar] [CrossRef]
  29. Haas, H.L.; Sergeeva, O.A.; Selbach, O. Histamine in the nervous system. Physiol. Rev. 2008, 88, 1183–1241. [Google Scholar] [CrossRef]
  30. Nuutinen, S.; Panula, P. Histamine in Neurotransmission and Brain Diseases. In Histamine in Inflammation; Advances in Experimental Medicine and Biology; Thurmond, R.L., Ed.; Springer: Boston, MA, USA, 2010; Volume 709, pp. 95–107. [Google Scholar] [CrossRef]
  31. Ellenbroek, B.A.; Ghiabi, B. The other side of the histamine H3 receptor. Trends Neurosci. 2014, 37, 191–199. [Google Scholar] [CrossRef]
  32. Dauvilliers, Y.; Bassetti, C.; Lammers, G.J.; Arnulf, I.; Mayer, G.; Rodenbeck, A.; Lehert, P.; Ding, C.L.; Lecomte, J.M.; Schwartz, J.C. Pitolisant versus placebo or modafinil in patients with narcolepsy: A double-blind, randomised trial. Lancet Neurol. 2013, 12, 1068–1075. [Google Scholar] [CrossRef]
  33. Thurmond, R.L.; Kazerouni, K.; Chaplan, S.R.; Greenspan, A.J. Peripheral Neuronal Mechanism of Itch: Histamine and Itch; CRC Press/Taylor & Francis: Boca Raton, FL, USA, 2014; ISBN 9781466505438. [Google Scholar]
  34. Rosa, A.C.; Pini, A.; Lucarini, L.; Lanzi, C.; Veglia, E.; Thurmond, R.L.; Stark, H.; Masini, E. Prevention of bleomycin-induced lung inflammation and fibrosis in mice by naproxen and JNJ7777120 treatment. J. Pharmacol. Exp. Ther. 2014, 351, 308–316. [Google Scholar] [CrossRef] [Green Version]
  35. Lucarini, L.; Pini, A.; Rosa, A.C.; Lanzi, C.; Durante, M.; Chazot, P.L.; Krief, S.; Schreeb, A.; Stark, H.; Masini, E. Role of histamine H4 receptor ligands in bleomycin-induced pulmonary fibrosis. Pharmacol. Res. 2016, 111, 740–748. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Connelly, W.M.; Shenton, F.C.; Lethbridge, N.; Leurs, R.; Waldvogel, H.J.; Faull, R.L.M.; Lees, G.; Chazot, P.L. The histamine H 4 receptor is functionally expressed on neurons in the mammalian CNS. Br. J. Pharmacol. 2009, 157, 55–63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Sanna, M.D.; Ghelardini, C.; Thurmond, R.L.; Masini, E.; Galeotti, N. Behavioural phenotype of histamine H4 receptor knockout mice: Focus on central neuronal functions. Neuropharmacology 2017, 114, 48–57. [Google Scholar] [CrossRef] [PubMed]
  38. Sanna, M.D.; Stark, H.; Lucarini, L.; Ghelardini, C.; Masini, E.; Galeotti, N. Histamine H 4 receptor activation alleviates neuropathic pain through differential regulation of ERK, JNK, and P38 MAPK phosphorylation. Pain 2015, 156, 2492–2504. [Google Scholar] [CrossRef] [PubMed]
  39. Castellan Baldan, L.; Williams, K.A.; Gallezot, J.D.; Pogorelov, V.; Rapanelli, M.; Crowley, M.; Anderson, G.M.; Loring, E.; Gorczyca, R.; Billingslea, E.; et al. Histidine Decarboxylase Deficiency Causes Tourette Syndrome: Parallel Findings in Humans and Mice. Neuron 2014, 81, 77–90. [Google Scholar] [CrossRef] [Green Version]
  40. Greferath, U.; Kambourakis, M.; Barth, C.; Fletcher, E.L.; Murphy, M. Characterization of histamine projections and their potential cellular targets in the mouse retina. Neuroscience 2009, 158, 932–944. [Google Scholar] [CrossRef] [PubMed]
  41. Vila, A.; Satoh, H.; Rangel, C.; Mills, S.L.; Hoshi, H.; O’Brien, J.; Marshak, D.R.; Macleish, P.R.; Marshak, D.W. Histamine receptors of cones and horizontal cells in Old World monkey retinas. J. Comp. Neurol. 2012, 520, 528–543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Yu, Y.C.; Satoh, H.; Wu, S.M.; Marshak, D.W. Histamine enhances voltage-gated potassium currents of on bipolar cells in macaque retina. Investig. Ophthalmol. Vis. Sci. 2009, 50, 959–965. [Google Scholar] [CrossRef] [Green Version]
  43. Akimov, N.P.; Marshak, D.W.; Frishman, L.J.; Glickman, R.D.; Yusupov, R.G. Histamine reduces flash sensitivity of ON ganglion cells in the primate retina. Investig. Ophthalmol. Vis. Sci. 2010, 51, 3825–3834. [Google Scholar] [CrossRef] [Green Version]
  44. Skingsley, D.R.; Laughlin, S.B.; Hardie, R.C. Properties of histamine-activated chloride channels in the large monopolar cells of the dipteran compound eye: A comparative study. J. Comp. Physiol. A 1995, 176, 611–623. [Google Scholar] [CrossRef]
  45. Coombe, P.E. The large monopolar cells L1 and L2 are responsible for ERG transients in Drosophila. J. Comp. Physiol. A 1986, 159, 655–665. [Google Scholar] [CrossRef]
  46. Gastinger, M.; Tian, N.; Horvath, T.; Marshak, D. Retinopetal axons in mammals: Emphasis on histamine and serotonin. Curr. Eye Res. 2006, 31, 655–667. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Lin, J.S.; Sergeeva, O.A.; Haas, H.L. Histamine H3 receptors and sleep-wake regulation. J. Pharmacol. Exp. Ther. 2011, 336, 17–23. [Google Scholar] [CrossRef] [Green Version]
  48. Markwardt, K.L.; Magnino, P.E.; Pang, I.H. Histamine induced contraction of human ciliary muscle cells. Exp. Eye Res. 1997, 64, 713–717. [Google Scholar] [CrossRef] [PubMed]
  49. Sebbag, L.; Allbaugh, R.A.; Weaver, A.; Seo, Y.J.; Mochel, J.P. Histamine-Induced Conjunctivitis and Breakdown of Blood–Tear Barrier in Dogs: A Model for Ocular Pharmacology and Therapeutics. Front. Pharmacol. 2019, 10. [Google Scholar] [CrossRef]
  50. Montan, P.G.; Van Hage-Hamsten, M. Eosinophil cationic protein in tears in allergic conjunctivitis. Br. J. Ophthalmol. 1996, 80, 556–560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Inada, N.; Shoji, J.; Shiraki, Y.; Aso, H.; Yamagami, S. Histamine H1 and H4 receptor expression on the ocular surface of patients with chronic allergic conjunctival diseases. Allergol. Int. 2017, 66, 586–593. [Google Scholar] [CrossRef]
  52. Luncă, D.-C.; Păunescu, H.; Mușat, O.; Fulga, I. The histaminergic control of the iridal vascular tone in rats and its influencing by topical administration of olopatadine and ranitidine. Rom. J. Ophthalmol. 2019, 63, 23–28. [Google Scholar] [CrossRef]
  53. Lachkar, Y.; Bouassida, W. Drug-induced acute angle closure glaucoma. Curr. Opin. Ophthalmol. 2007, 18, 129–133. [Google Scholar] [CrossRef] [Green Version]
  54. Razeghinejad, M.R.; Myers, J.S.; Katz, L.J. Iatrogenic glaucoma secondary to medications. Am. J. Med. 2011, 124, 20–25. [Google Scholar] [CrossRef]
  55. Zampeli, E.; Tiligada, E. The role of histamine H 4 receptor in immune and inflammatory disorders. Br. J. Pharmacol. 2009, 157, 24–33. [Google Scholar] [CrossRef]
  56. Leonardi, A.; Di Stefano, A.; Vicari, C.; Motterle, L.; Brun, P. Histamine H4 receptors in normal conjunctiva and in vernal keratoconjunctivitis. Allergy Eur. J. Allergy Clin. Immunol. 2011, 66, 1360–1366. [Google Scholar] [CrossRef] [PubMed]
  57. Nau, C.B.; Malihi, M.; McLaren, J.W.; Hodge, D.O.; Sit, A.J. Circadian Variation of Aqueous Humor Dynamics in Older Healthy Adults. Investig. Ophthalmol. Vis. Sci. 2013, 54, 7623. [Google Scholar] [CrossRef] [Green Version]
  58. Smith, S.D.; Gregory, D.S. A Circadian Rhythm of Aqueous Flow Underlies the Circadian Rhythm of IOP in NZW Rabbits. | IOVS | ARVO Journals. Available online: https://iovs.arvojournals.org/article.aspx?articleid=2160758 (accessed on 12 April 2021).
  59. Tripathi, R.C.; Tripathi, B.J. Human trabecular endothelium, corneal endothelium, keratocytes, and scleral fibroblasts in primary cell culture. A comparative study of growth characteristics, morphology, and phagocytic activity by light and scanning electron microscopy. Exp. Eye Res. 1982, 35, 611–624. [Google Scholar] [CrossRef]
  60. Lanzi, C.; Lucarini, L.; Durante, M.; Sgambellone, S.; Pini, A.; Catarinicchia, S.; Łażewska, D.; Kieć-Kononowicz, K.; Stark, H.; Masini, E. Role of Histamine H3 Receptor Antagonists on Intraocular Pressure Reduction in Rabbit Models of Transient Ocular Hypertension and Glaucoma. Int. J. Mol. Sci. 2019, 20, 981. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Impagnatiello, F.; Giambene, B.; Lanzi, C.; Pini, A.; Somma, T.; Bastia, E.; Ongini, E.; Galassi, F.; Masini, E. The nitric oxide donating triamcinolone acetonide NCX 434 does not increase intraocular pressure and reduces endothelin-1 induced biochemical and functional changes in the rabbit eye. Br. J. Ophthalmol. 2012, 96, 757–761. [Google Scholar] [CrossRef]
  62. Kim, J.; Park, D.Y.; Bae, H.; Park, D.Y.; Kim, D.; Lee, C.K.; Song, S.; Chung, T.Y.; Lim, D.H.; Kubota, Y.; et al. Impaired angiopoietin/Tie2 signaling compromises Schlemm’s canal integrity and induces glaucoma. J. Clin. Investig. 2017, 127, 3877–3896. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Western blot analysis of histamine H1R (A), H3R (B) and H4R (C) subtypes in the retina, ciliary body, optic nerve, and stomach samples of New Zealand White (NZW) rabbits. Densitometric data of 6 determinations are reported as relative optical density, corrected for the corresponding β-actin content [60] modified.
Figure 1. Western blot analysis of histamine H1R (A), H3R (B) and H4R (C) subtypes in the retina, ciliary body, optic nerve, and stomach samples of New Zealand White (NZW) rabbits. Densitometric data of 6 determinations are reported as relative optical density, corrected for the corresponding β-actin content [60] modified.
Biomolecules 11 01186 g001
Figure 2. IOP lowering effect of ciproxifan in the transient ocular hypertensive model in NZW rabbits. The effect of ciproxifan is suppressed by pre-treatment with 1% imetit. * p < 0.05 ciproxifan 1% at 60′ and 120′ vs. vehicle and imetit 1% + ciproxifan 1%. All the results are expressed as mean ± SEM (n = 6). Two-way ANOVA followed by Bonferroni post hoc test.
Figure 2. IOP lowering effect of ciproxifan in the transient ocular hypertensive model in NZW rabbits. The effect of ciproxifan is suppressed by pre-treatment with 1% imetit. * p < 0.05 ciproxifan 1% at 60′ and 120′ vs. vehicle and imetit 1% + ciproxifan 1%. All the results are expressed as mean ± SEM (n = 6). Two-way ANOVA followed by Bonferroni post hoc test.
Biomolecules 11 01186 g002
Figure 3. Representative images of hematoxylin/eosin-stained histological sections of retinae from different treated groups. RGCs are visible in the upper layer. The histological sections of Vehicle, Ciproxifan and DL-76 panels were prepared from carbomer-induced glaucoma models in male NZW rabbits with stable elevated IOP [60] modified.
Figure 3. Representative images of hematoxylin/eosin-stained histological sections of retinae from different treated groups. RGCs are visible in the upper layer. The histological sections of Vehicle, Ciproxifan and DL-76 panels were prepared from carbomer-induced glaucoma models in male NZW rabbits with stable elevated IOP [60] modified.
Biomolecules 11 01186 g003
Table 1. (A) Transient ocular hypertensive model and (B) carbomer-induced glaucoma model in male NZW rabbits.
Table 1. (A) Transient ocular hypertensive model and (B) carbomer-induced glaucoma model in male NZW rabbits.
A IOP-Lowering Effect
CompoundBasalAfter
Saline
Post Treatment (60 min)ΔΔIOP
(60 min)
Post Treatment (120 min)ΔΔIOP
(120 min)
IOP, mmHgIOP, mmHgIOP, mmHgmmHgIOP, mmHgmmHg
Vehicle 15 ± 0.336 ± 7.538 ± 3.90 ± 3.928 ± 5.70 ± 5.7
Ciproxifan13 ± 4.338 ± 6.222 ± 5.3−18.9 ± 5.3 **17 ± 5.5−16.4 ± 5.5 **
DL-7615 ± 4.334 ± 6.223 ± 7.4−15.4 ± 7.5 **20 ± 3.0−16.1 ± 3.1 **
GSK18925414 ± 5.637 ± 5.532 ± 4.3−8.5 ± 4.3 *25 ± 6.7−9.9 ± 6.7 *
Timolol14 ± 2.538 ± 5.721 ± 3.8−16.5 ± 3.8 **19 ± 4.6−14.8 ± 4.7 **
B IOP-Lowering Effect
CompoundBasalAfter
Carbomer
Post-Treatment
(7 days)
ΔΔIOP
(7 days)
IOP, mmHgIOP, mmHgIOP, mmHgmmHg
Vehicle 15 ± 0.338 ± 2.841 ± 7.30 ± 2.12
Ciproxifan13 ± 5.136 ± 4.120 ± 2.9−19 ± 2.9 **
DL-7612 ± 3.534 ± 2.823 ± 3.8−15.7 ± 2.7 *
GSK18925414 ± 0.040 ± 1.426 ± 2.8−14.5 ± 3.8 *
Timolol15 ± 0.741 ± 5.627 ± 2.1−13.5 ± 2.1 *
Ocular hypotensive efficacy is expressed in mmHg. The average difference in IOP between drug-treated eyes or vehicle-treated eyes and their respective pre-treatment value are shown in the following formula: Efficacy = (IOPdrug − IOPpredose drug) − (IOPveh − IOP predose veh). (A) ** p < 0.01 ciproxifan, DL-76 1% and Timolol 1% at 60′ and 120′; * p < 0.05 GSK189254 1% at 60′ and 120′ versus vehicle; (B) ** p < 0.01 ciproxifan; * p < 0.05 DL-76, Timolol, GSK189254 at day 7 versus vehicle. All the results are expressed as mean ± SEM (n = 6). The significance of differences was assessed by two-way ANOVA for multiple comparisons followed by the Bonferroni post hoc test [60] modified.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sgambellone, S.; Lucarini, L.; Lanzi, C.; Masini, E. Novel Insight of Histamine and Its Receptor Ligands in Glaucoma and Retina Neuroprotection. Biomolecules 2021, 11, 1186. https://doi.org/10.3390/biom11081186

AMA Style

Sgambellone S, Lucarini L, Lanzi C, Masini E. Novel Insight of Histamine and Its Receptor Ligands in Glaucoma and Retina Neuroprotection. Biomolecules. 2021; 11(8):1186. https://doi.org/10.3390/biom11081186

Chicago/Turabian Style

Sgambellone, Silvia, Laura Lucarini, Cecilia Lanzi, and Emanuela Masini. 2021. "Novel Insight of Histamine and Its Receptor Ligands in Glaucoma and Retina Neuroprotection" Biomolecules 11, no. 8: 1186. https://doi.org/10.3390/biom11081186

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