Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 3465 word(s) 3465 2021-01-28 09:33:21 |
2 format change Meta information modification 3465 2021-03-08 04:38:46 | |
3 format change Meta information modification 3465 2021-03-08 04:39:51 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Guarneri, F. RAGE and Oxidative Stress. Encyclopedia. Available online: https://encyclopedia.pub/entry/7793 (accessed on 27 April 2024).
Guarneri F. RAGE and Oxidative Stress. Encyclopedia. Available at: https://encyclopedia.pub/entry/7793. Accessed April 27, 2024.
Guarneri, Fabrizio. "RAGE and Oxidative Stress" Encyclopedia, https://encyclopedia.pub/entry/7793 (accessed April 27, 2024).
Guarneri, F. (2021, March 07). RAGE and Oxidative Stress. In Encyclopedia. https://encyclopedia.pub/entry/7793
Guarneri, Fabrizio. "RAGE and Oxidative Stress." Encyclopedia. Web. 07 March, 2021.
RAGE and Oxidative Stress
Edit

The surface receptor for advanced glycosylation end-products (RAGE) and its soluble (sRAGE) and endogenous secretory (EN-RAGE) forms belong to the superfamily of toll-like receptors and play important roles in inflammation and autoimmunity, directly or through binding with advanced glycosylation end-products (AGE) and advanced oxidation protein products (AOPP).

RAGE sRAGE skin alarmins

1. Introduction

Oxidation is a physiological and necessary part of the complex network of physical and chemical phenomena related to life. Oxidative reactions are involved, for example, in the biological chain of aerobic metabolism, in the biosynthesis of small and big molecules, in signal transduction between the outside and inside of cells as well as between the different cell compartments, and in the defense mechanisms used by immune cells [1][2]. However, quantitatively excessive and too long oxidation phenomena are linked to cellular suffering and destruction, which result, at the organism level, in several inflammatory, neoplastic, and degenerative diseases [3]. In this typically hormetic situation, where the beneficial or detrimental effect of reactive oxygen species (ROS) and electrophiles depends on their amount [4], it is not surprising that cells and living organisms have developed several enzymatic and non-enzymatic homeostatic mechanisms to adaptively maintain the delicate and dynamic oxidoreductive (redox) equilibrium within the limits of the so-called “eustress”, as opposed to the aforementioned detrimental imbalance, to which the term “oxidative stress” is commonly referred [5].

In normal conditions, ROS can be generated by the mitochondrial electron transport chain, proteins of the peroxisomal and endoplasmic reticulum, the Fenton reaction, as well as specific enzymes, like cyclooxygenases, nicotinamide adenine dinucleotide phosphate oxidases, lipoxygenases, and xanthine oxidases [6]. External factors, like smoking, eating/drinking habits or lifestyle, may also influence the production of ROS. Environmental variables, and the control of their effects on the redox status of cells, are particularly important in the case of skin, which is the outermost and largest organ of the human body and is structured to be an active interface structure, with multiple connections with the main homeostatic systems of the organism. The major environmental cause of ROS production in the skin is ultraviolet (UV) radiations, mainly derived from sun exposure. Many cutaneous cromophores may directly absorb UV and generate ROS: DNA and other nucleic acids, urocanic acid, tryptophan, tyrosine, NADH (reduced form of nicotinamide adenine dinucleotide), quinones, flavins, and porphyrins mostly absorb ultraviolet B (UVB), while trans-urocanic acid, melanin precursors, and riboflavin absorb ultraviolet A (UVA). Some authors also demonstrated that electromagnetic radiations with lower energy, i.e., visible and infrared light, may induce ROS formation in the skin [7]. Pollutants, like airborne particulate matter or ozone, may increase ROS and/or deplete cutaneous antioxidants [8]. To counterbalance oxidative stress, many antioxidants are available in the skin, including vitamins C and E, beta-carotene, ferritin, uric acid, coenzyme Q, glutathione, glutathione peroxidases, catalase, heme oxygenase, and superoxide dismutase [9].

However generated, excessive ROS may induce cell and organ suffering through multiple mechanisms, such as telomere shortening, DNA damage, mutations of mitochondrial DNA, microRNA (dys)regulation, alteration of matrix metalloproteinases and signal transduction pathways, modifications of the extracellular matrix, chronic inflammation (clinically evident or subclinical), protein oxidation, vascular alterations, and accumulation of advanced glycation end products (AGEs).

AGEs are biomolecules of various nature (proteins, lipids, nucleic acids) that are abnormally covalently bound by glucose or fructose, with the final effect of inhibition of their function. The term “glycation” is used to distinguish this process from glycosilation, a physiological enzyme-mediated binding of sugars that occurs at specific sites of target molecules to enable their functions [10]. Scientific interest in AGEs has grown rapidly in the past decades: published papers went from less than 10 per year in 1991 to about one thousand per year in the last seven years (2014–2020). The reason for this interest is their involvement in a wide and yet incompletely explored range of diseases, as well as biologic processes that are physiological but can be significantly influenced by extrinsic factors [11]. In the latter group, a process of dermatological interest is skin aging, which depends on time but can be enhanced by ROS production, UV exposure, pollutants, lifestyle, and/or personal habits. Among cutaneous molecules, glycated elastin is subject to abnormal aggregation and unusually interacts with lysozyme, glycated collagen resists to the proteolytic action of matrix metalloproteinases, and vimentin undergoes perinuclear accumulation, reducing the cellular contractile capacity; altogether, these modifications result in the tissue stiffening and reduced elasticity that are typically observed in aged skin [11]. Other than having direct detrimental effects, AGEs induce further alterations typical of skin aging through interaction with their receptor, RAGE (Receptor for Advanced Glycation End products) [12].

Discovered by Neeper et al. in 1992 [13], RAGE is a receptor that belongs to the superfamily of toll-like receptors and consists of two cytoplasmatic domains and a variable extracellular domain. Both domains are present in the surface receptor form, while the former domain is missing in the soluble form of the receptor (sRAGE) and the endogenous secretory form (EN-RAGE). In addition to AGEs, which are its canonical ligands, RAGE is also able to bind a number of other endogenous and exogenous molecules. Endogenous ligands belong to the group of DAMPs (Damage-Associated Molecular Patterns) or alarmins, molecules that are released by cells under stress conditions and after their death, are involved in the host’s defense immune system and act, through RAGE, as promoters of inflammation and cytokine production [14]. These alternative RAGE ligands include Advanced Oxidation Protein Products (AOPP) [15], markers of oxidative stress generated by the interaction of plasma proteins, mainly albumin, with chlorinated oxidants, like chloramines and hypochlorous acid [16]. This group of markers includes dityrosine, pentosidine, and carbonyl-containing protein products [17]. AOPP seem to play a role mostly in acute processes of response to inflammatory insults and are emerging as crucial factors in the development of a great deal of skin diseases with acute manifestations, such as systemic lupus erythematosus, vitiligo, psoriasis, and Behcet’s disease, even though other studies involving chronic and tumoral diseases (such as mastocytosis and melanoma) have shown an increase of their circulating levels in patients’ sera [18].

Other known alternative RAGE ligands are S100A12 (S100 calcium-binding protein A12, which is also known as “extracellular newly identified RAGE binding protein” or calgranulin C), High-Mobility Group Box 1 (HMGB1), phosphatidylserine on apoptotic cells, the complement protein C1q, DNA, and Gram negative bacterial lipopolysaccharide (LPS) [15]. Additionally, in other studies, binding to RAGE has been inferred, although without direct demonstration, for diesel particulate matter, oxidized low density lipoprotein, and extracellular heat shock protein 70 [15].

The membrane isoform of RAGE is implicated in the pathways of NFAT (nuclear factor of activated T-cells), NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells), STAT3 (signal transducer and activator of transcription 3), and CREB (cAMP response element-binding protein) transcription factors, which are involved in the autophagy and apoptosis processes. RAGE has been found in a large number of cell types, which include, among others, endothelial cells, neural cells, cardiac myocytes, mesangial cells, as well as cells of the immune system such as monocytes, macrophages, and T lymphocytes [19]. In the skin, RAGE is highly expressed, with variable patterns that depend on age and sun exposure [20]. In detail, in fetal skin, RAGE is located in the upper epidermis and in few endothelial cells within the dermis; in young skin, it is present in the superficial and middle epidermis and in the papillary dermis; in aged skin, it can be found in the middle and basal part of the epidermis and in the reticular dermis. Chronically sun-exposed skin shows patchy distribution of RAGE in the epidermis and increased expression of RAGE on cells of the papillary dermis. For which concerns the type of cells, RAGE was mainly expressed on fibroblasts, dendrocytes, and endothelial cells, while the number of RAGE-positive CD45 lymphocytes is low [20].

The soluble form of RAGE should act as a decoy receptor, preventing the furthering of the pro-inflammatory cascade, in which RAGE plays a role. The role of sRAGE as a diagnostic marker for disease activity has been debated, since its levels might increase (as a way for the body itself to try to shut down inflammatory processes) in several diseases, but results have been inconclusive.

As of now, we know that RAGE plays a role in a plethora of other physiological and pathological processes, including diabetes, osteoporosis and cardiovascular diseases. Over the course of time, many different factors, including oxidative stress, were studied to explain the pathophysiology of many skin diseases characterized by chronic inflammation and frequent relapses, such as atopic dermatitis [21][22][23], psoriasis [24][25], lichen planus [26][27], vitiligo [28][29], or discoid lupus erythematosus [30]. It is well known that the clinical expression of such diseases depends on a variety of factors, both inner and outer, but, as of yet, one of these factors, namely RAGE, seems to play an important role in a context of continuous inflammatory and autoimmune stimuli, thus prompting further studies in order to complete the picture we have of such diseases.

As outlined before, there is great scientific interest and research about the importance of RAGE in all fields of medicine. Dermatology is a peculiar discipline, which includes purely cutaneous diseases and cutaneous manifestations of internal or multi-organ diseases, and dermatological research on RAGE reflects this fact, because available resources were spread over multiple studies concerning different conditions within the discipline, producing a remarkable but polyhedric amount of data. The aim of this review is to organize the currently available knowledge about the role of RAGE in the macrocosm represented by skin diseases to better understand immunopathological pathways involved and possible therapeutic options.

2. Research Data

This entry was performed by browsing the PubMed database (https://www.pubmed.gov), searching for articles labeled with both the MeSH (Medical Subject Headings) terms “RAGE” and “Skin”, with the search string (“skin”[MeSH]) AND “Receptor for Advanced Glycation End Products”[MeSH]. The initial search produced 39 articles, of which nine were excluded from this review by title and abstract only, and three were excluded after reading them entirely. One additional article was found and included after a search in the PubMed database with the two MeSH terms “Vitiligo” and “Receptor for Advanced Glycation End Products”, for a grand total of 28 articles. The articles included in this review showed a direct link between RAGE and various skin conditions, because RAGE was either a prominent feature in prolonging the inflammatory response or a primary cause of it. The excluded articles were reviews, did not concern a direct link between RAGE and skin, or did not provide enough information about materials and methods. Due to the great heterogeneity of materials, methods, and outcomes of the above cited studies, a meta-analysis was not deemed appropriate.

3. RAGE as an Inflammation Receptor

A study by Riehl et al. [31] noted that even though RAGE is not essential for an inflammation process to start, it sustains a concerted increase of other transcriptional factors such as the Rb (retinoblastoma tumor suppressor protein)-E2F pathway, demonstrated by a more prominent staining in wild type mice than in RAGE−/− ones upon stimulation with tetradecanoylphorbol acetate (TPA).

Another remarkable characteristic, demonstrated by Leibold et al. [32], is that mice with a mutation or deletion of RAGE on keratinocytes show an earlier resolution process after an initial inflammatory one, partially because of the role of RAGE in the production of TNF-α by keratinocytes. Nienhuis et al. [33] found a significant role of RAGE in lupus: sRAGE levels were significantly higher in lupus lesional skin.

Another prominent study by Wolf et al. [34] on mice demonstrated that in TPA-induced skin psoriasis lesions, proinflammatory cytokines such as MIP-2 (macrophage-inflammatory protein 2), IL-1α, and TNF-α were significantly lower if RAGE was absent. On the other hand, RAGE null mice showed a higher level of IL-1Ra (interleukin 1 receptor antagonist), as an anti-inflammatory effect, even though only in vitro.

Wolf et al. also demonstrated that in vivo administration of RAGE-blocking antibodies ameliorated the clinical score of atopic dermatitis by reducing the levels of pro-inflammatory cytokines, even though the levels of IL-1α did not drop significantly [34]. Szczepanski et al. [35] studied a very specific skin district, i.e., the ear, and the correlation between the HMGB1/RAGE pathway and the pathogenesis of cholesteatoma. Through such a chain reaction, they noted an increase of RAGE levels on skin cells, a more consistent production of IL-8, and better protection from apoptosis of keratinocytes by phosphorylation of MEK (meiotic chromosome-axis-associated kinase) 1/2, STAT3, MAPK (mitogen-activated protein kinase) p44/p22, and NF-κB. All these effects were lost when the cell medium was enriched with RAGE-binding antibodies [35]. NF-κB and the subsequent production of IL-6 were also shown to be downregulated in mice with TPA-induced atopic dermatitis, when they were treated with Glycyrrhiza glabra, a direct inhibitor of HMGB1, one of the main ligands of RAGE [36]. Concerning the same mechanism, it is noteworthy that de Carvalho et al. investigated the role of RAGE in lichen planus, observing that its levels were more prominent in the upper layers of the dermis of patients compared with healthy controls, even though the levels of its mRNA were lower: they suggested that this might be due to the rapid translocation of HMGB1 by affected keratinocytes [37]. On the same note, but regarding atopic dermatitis, there is a study focusing on the role of quercetin in inhibiting the RAGE pathway through HMGB1 [38]: in this study, the authors observed downregulation of cytoplasmic HMGB1, RAGE, nuclear p-NFκB, p-ERK1/2, COX2, TNFα, IL-1β, IL-2Rα, IFNγ, and IL-4 and upregulation of nuclear Nrf2, which ultimately resulted in an attenuation of the atopic dermatitis-like lesions induced by a house dust mite extract applied to the dorsal skin of NC/Nga transgenic mice. Two more studies focused, respectively, on the difference between a physiological and pathological skin conditions such as skin thickness and inflammation, finding that RAGE expression directly correlated with epidermal thickness [39], and a possible relationship with levels of oxidative stress and artery diseases, finding that skin autofluorescence is increased in stable coronary artery disease and associated with soluble RAGE [40].

4. RAGE in Infectious Diseases

One of the ligands of RAGE, namely EN-RAGE, is implicated in the inflammatory cascade involved in the pathogenesis of leprosy. The levels of both RAGE and EN-RAGE show a significant increase in the multibacillary form of leprosy, specifically in the formation of the granuloma and perpetration of the inflammatory process [41]. Another study, by Na et al., investigated the correlation between disease severity and the levels of RAGE in Staphylococcus aureus-induced skin lesions. In this study, the authors observed that in RAGE-deficient mice, the lesion size was significantly smaller than in wild-type mice, and the occurrence of abscesses in the skin was significantly higher in the RAGE−/− population, with a consequent delay of the healing process. This interesting contrast led them to hypothesize that in a RAGE-deficient environment, there is a more rapid inflammatory response to external stimuli and lower levels of inflammatory cytokines over time. In vitro, the authors of this study observed an enhanced phagocytic activity in RAGE−/− mice compared to controls, resulting in lower bacterial load at the site of infection. However, two studies appear insufficient to draw any definitive conclusions and further studies should be conducted [42].

5. RAGE in Diabetic Skin and Repairing Processes

Lohwasser et al. demonstrated that the levels of RAGE increase over time, in particular in the basal layer of the epidermis and the fibroblasts of the upper layer of the dermis (where skin regeneration occurs) of older patients more than in fetal skin, and more in sun exposed than in not exposed skin [20].

Another dermal effect on fibroblasts was demonstrated by Niu et al. [43], who studied cell turnover in diabetic wounds: the authors let dermal cells grow on both a normal and an AGE-rich extra-cellular matrix, noticing that the interaction between AGE and RAGE produced a slow-down in cellular replication. As counterproof, the authors added anti-RAGE antibodies to the culture, producing more actively replicating cells [43].

The effect of RAGE on diabetic skin is not restricted to repairing processes: the interaction of RAGE and its ligands is also supposed to have a role in microscopic vasculopathy and keratinocyte turnover, producing the typical dryness and vascular lesions over time, by accumulation of ROS [44], and the AGE–RAGE interaction may act on endothelial cells as an inhibiting stimulus for the production of prostacyclins and a positive stimulus for the production of plasminogen activator inhibitor-1, thus increasing the thrombogenic effect on larger vessels and, perhaps, on smaller ones too, through the production of reactive oxygen species [45][46][47][48][49]. These observations led some other authors to propose topical treatments and a possible role of RAGE in estimating diabetic wounds [50][51] with promising results.

6. RAGE and Systemic Sclerosis and Ulcers

There seems to also be a prominent role of the RAGE pathway in sclerotic and ulcerative processes. Yoshizaki et al. investigated the relation between levels of HMGB1 and sRAGE and disease severity in patients affected by systemic sclerosis. In detail, correlation was observed with skin fibrosis, scars, ulcers, pulmonary fibrosis, and, at a molecular level, serum levels of anti-topoisomerase 1 antibodies, IgG, CRP (C reactive protein), and ESR (erythrocyte sedimentation rate), but not TNF-α, IL-1β, and IL-6 [52], even though another study, concerning calcinotic deposits, did not produce such clear cut results [53].

Another role of RAGE as a structure-oriented receptor was demonstrated by Zhao et al. [54]: in this study, the authors delved into the role of S100A12 as a factor activated by skin dryness and its regulation during scar formation and dermal fibrosis, showing that, in fact, in case of reduced water levels it acts as a hypertrophic stimulus through the activation of RAGE. The authors used the S100A12 molecule on ear lobes in mice, showing a significant production of hypertrophic fibrous tissue through the activation of RAGE [54].

7. RAGE and Melanocytes

The RAGE induced pathway has also an important role in melanogenesis, through ligation with AGEs. The phosphorylated form of CREB, which is part of the RAGE downstream cascade, activates the CRE (cAMP response elements) consensus motifs of MITF (microphthalmia-associated transcription factor) and, consequently, tyrosinases, boosting melanin production in melanocytes [55]. On the other hand, Cui et al. demonstrated that RAGE plays an important role in the pathogenesis of vitiligo [56] and ubiquinone-induced irritation of melanocytes [57]. In vitiligo-affected patients, RAGE, out of the three PRRs (pattern recognition receptors) that bind HMGB1 (the others being the toll-like receptors TLR2 and TLR4), is the only one upregulated, and its blockade significantly reduces the levels of IL-8 and CXCL16 [chemokine (C-X-C motif) ligand 16] in keratinocytes. The authors investigated the in vitro levels of RAGE and HMGB1 after hydrogen peroxide was added to the culture: indeed, the levels of both HMGB1 and RAGE increased. Because of these results, questions arise about the possible effects of pro-oxidative therapies such as UVB phototherapy, in which there is an activation of melanocytic stem cells and down-regulation of self-directed T cells, opposed to the UV-induced, pro-inflammatory release of HMGB1 by keratinocytes [56].

8. Conclusions

The current knowledge about the RAGE receptor is very limited. Given the great deal of roles that this molecule plays in different types of skin diseases, what emerges from these studies is that it would be convenient to push forward the research using live models, in order to better understand the role of RAGE in a more complete and complex environment. The great deal of skin diseases which involve both AGEs and AOPPs can provide different opportunities to study the different roles played by RAGE. It is our proposition that the two most interesting diseases that can prove useful on such note would be systemic lupus erythematosus and systemic scleroderma, since both diseases have characteristic phases (acute, subacute, and chronic), which can better define the already discussed roles of AGE (mostly in chronic conditions) and AOPP (most prominent in acute processes) and their interaction with RAGE.

Another key feature emerging from these studies is that the soluble form of RAGE (sRAGE) can prove to be a potential diagnostic tool to better asses the disease activity in various skin diseases, mostly chronic, and its topical form could be used as an alternative treatment option when other topicals are not available or could provide significant side effects. Indeed, it could be interesting to perform compared studies with the current golden standard, topical therapy and sRAGE. On the other hand, based on the current knowledge about this receptor, a systemic sRAGE treatment, although fascinating, is likely to be difficult, since RAGE does not play a single role as an inflammatory receptor but as a remodeling one as well.

Finally, a potential target therapy with antibodies, anti-RAGE, as a systemic treatment, seems to be an interesting perspective for a plethora of diseases, even though further studies must be conducted. This approach could prove useful especially in those diseases where the accumulation of ROS, presence of alarmins, and overall inflammatory state could be a greater management challenge than the first pathogenetic cause, or in those acute diseases where there is a need of a preemptive stop to a self-directed harmful process (such as small vessels diseases).

References

  1. Halliwell, B.; Gutteridge, J.M. Free Radicals in Biology and Medicine; Oxford University Press: Oxford, UK, 2015.
  2. Sies, H.; Jones, D.P. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat. Rev. Mol. Cell Biol. 2020, 21, 363–383.
  3. Sies, H.; Berndt, C.; Jones, D.P. Oxidative stress. Annu. Rev. Biochem. 2017, 86, 715–748.
  4. Mattson, M.P. Hormesis defined. Ageing Res. Rev. 2008, 7, 1–7.
  5. Ron-Doitch, S.; Kohen, R. The cutaneous physiological redox: Essential to maintain but difficult to define. Antioxidants 2020, 9, 942.
  6. Rinnerthaler, M.; Bischof, J.; Streubel, M.K.; Trost, A.; Richter, K. Oxidative stress in aging human skin. Biomolecules 2015, 5, 545–589.
  7. McDaniel, D.; Farris, P.; Valacchi, G. Atmospheric skin aging-contributors and inhibitors. J. Cosmet. Dermatol. 2018, 17, 124–137.
  8. Fussell, J.C.; Kelly, F.J. Oxidative contribution of air pollution to extrinsic skin ageing. Free Radic. Biol. Med. 2020, 151, 111–122.
  9. Gu, Y.; Han, J.; Jiang, C.; Zhang, Y. Biomarkers, oxidative stress and autophagy in skin aging. Ageing Res. Rev. 2020, 59, 101036.
  10. Danby, F.W. Nutrition and aging skin: Sugar and glycation. Clin. Dermatol. 2010, 28, 409–411.
  11. Farrar, M.D. Advanced glycation end products in skin ageing and photoageing: What are the implications for epidermal function? Exp. Dermatol. 2016, 25, 947–948.
  12. Nguyen, H.P.; Katta, R. Sugar sag: Glycation and the role of diet in aging skin. Skin Therapy Lett. 2015, 20, 1–5.
  13. Neeper, M.; Schmidt, A.M.; Brett, J.; Yan, S.D.; Wang, F.; Pan, Y.C.; Elliston, K.; Stern, D.; Shaw, A. Cloning and expression of a cell surface receptor for advanced glycosylation end products of proteins. J. Biol. Chem. 1992, 267, 14998–15004.
  14. De Martinis, M.; Ginaldi, L.; Sirufo, M.M.; Pioggia, G.; Calapai, G.; Gangemi, S.; Mannucci, C. Alarmins in osteoporosis, RAGE, IL-1, and IL-33 Pathways: A literature review. Medicina (Kaunas) 2020, 56, 138.
  15. Prantner, D.; Nallar, S.; Vogel, S.N. The role of RAGE in host pathology and crosstalk between RAGE and TLR4 in innate immune signal transduction pathways. FASEB J. 2020, 34, 15659–15674.
  16. Selmeci, L. Advanced oxidation protein products (AOPP): Novel uremic toxins, or components of the non-enzymatic antioxidant system of the plasma proteome? Free Radic. Res. 2011, 45, 1115–1123.
  17. Capeillère-Blandin, C.; Gausson, V.; Descamps-Latscha, B.; Witko-Sarsat, V. Biochemical and spectrophotometric significance of advanced oxidized protein products. Biochim. Biophys. Acta. 2004, 1689, 91–102.
  18. Cristani, M.; Speciale, A.; Saija, A.; Gangemi, S.; Minciullo, P.L.; Cimino, F. Circulating advanced oxidation protein products as oxidative stress biomarkers and progression mediators in pathological conditions related to inflammation and immune dysregulation. Curr. Med. Chem. 2016, 23, 3862–3882.
  19. Brett, J.; Schmidt, A.M.; Yan, S.D.; Zou, Y.S.; Weidman, E.; Pinsky, D.; Nowygrod, R.; Neeper, M.; Przysiecki, C.; Shaw, A.; et al. Survey of the distribution of a newly characterized receptor for advanced glycation end products in tissues. Am. J. Pathol. 1993, 143, 1699–1712.
  20. Lohwasser, C.; Neureiter, D.; Weigle, B.; Kirchner, T.; Schuppan, D. The receptor for advanced glycation end products is highly expressed in the skin and upregulated by advanced glycation end products and tumor necrosis factor-alpha. J. Investig. Dermatol. 2006, 126, 291–299.
  21. Ji, H.; Li, X.K. Oxidative stress in atopic dermatitis. Oxid. Med. Cell Longev. 2016, 2016, 2721469.
  22. Bertino, L.; Guarneri, F.; Cannavò, S.P.; Casciaro, M.; Pioggia, G.; Gangemi, S. Oxidative stress and atopic dermatitis. Antioxidants 2020, 9, 196.
  23. Guarneri, F.; Costa, C.; Foti, C.; Hansel, K.; Guarneri, C.; Guarneri, B.; Lisi, P.; Stingeni, L. Frequency of autoallergy to manganese superoxide dismutase in patients with atopic dermatitis: Experience of three Italian dermatology centres. Br. J. Dermatol. 2015, 173, 559–562.
  24. Lin, X.; Huang, T. Oxidative stress in psoriasis and potential therapeutic use of antioxidants. Free Radic. Res. 2016, 50, 585–595.
  25. Guarneri, F.; Sapienza, D.; Papaianni, V.; Marafioti, I.; Guarneri, C.; Mondello, C.; Roccuzzo, S.; Asmundo, A.; Cannavò, S.P. Association between genetic polymorphisms of glutathione S-transferase M1/T1 and psoriasis in a population from the area of the strict of messina (Southern Italy). Free Radic. Res. 2020, 54, 57–63.
  26. Upadhyay, R.B.; Carnelio, S.; Shenoy, R.P.; Gyawali, P.; Mukherjee, M. Oxidative stress and antioxidant defense in oral lichen planus and oral lichenoid reaction. Scand. J. Clin. Lab. Investig. 2010, 70, 225–228.
  27. Guarneri, F.; Guarneri, C.; Marini, H. Oral lichen planus and neurogenic inflammation: New observations and therapeutic implications from four clinical cases. Dermatol. Ther. 2014, 27, 206–210.
  28. Boniface, K.; Seneschal, J.; Picardo, M.; Taïeb, A. Vitiligo: Focus on clinical aspects, immunopathogenesis, and therapy. Clin. Rev. Allergy Immunol. 2018, 54, 52–67.
  29. Vaccaro, M.; Bagnato, G.; Cristani, M.; Borgia, F.; Spatari, G.; Tigano, V.; Saja, A.; Guarneri, F.; Cannavò, S.P.; Gangemi, S. Oxidation products are increased in patients affected by non-segmental generalized vitiligo. Arch. Dermatol. Res. 2017, 309, 485–490.
  30. Jafari, S.M.; Salimi, S.; Nakhaee, A.; Kalani, H.; Tavallaie, S.; Farajian-Mashhadi, F.; Zakeri, Z.; Sandoughi, M. Prooxidant-antioxidant balance in patients with systemic lupus erythematosus and its relationship with clinical and laboratory findings. Autoimmune Dis. 2016, 2016, 4343514.
  31. Riehl, A.; Bauer, T.; Brors, B.; Busch, H.; Mark, R.; Németh, J.; Gebhardt, C.; Bierhaus, A.; Nawroth, P.; Eils, R.; et al. Identification of the RAGE-dependent gene regulatory network in a mouse model of skin inflammation. BMC Genom. 2010, 11, 537.
  32. Leibold, J.S.; Riehl, A.; Hettinger, J.; Durben, M.; Hess, J.; Angel, P. Keratinocyte-specific deletion of the receptor RAGE modulates the kinetics of skin inflammation in vivo. J. Investig. Dermatol. 2013, 133, 2400–2406.
  33. Nienhuis, H.L.; de Leeuw, K.; Bijzet, J.; Smit, A.; Schalkwijk, C.G.; Graaff, R.; Kallenberg, C.G.; Bijl, M. Skin autofluorescence is increased in systemic lupus erythematosus but is not reflected by elevated plasma levels of advanced glycation endproducts. Rheumatology (Oxford) 2008, 47, 1554–1558.
  34. Wolf, R.; Mascia, F.; Dharamsi, A.; Howard, O.M.; Cataisson, C.; Bliskovski, V.; Winston, J.; Feigenbaum, L.; Lichti, U.; Ruzicka, T.; et al. Gene from a psoriasis susceptibility locus primes the skin for inflammation. Sci. Transl. Med. 2010, 2, 61ra90.
  35. Szczepanski, M.J.; Luczak, M.; Olszewska, E.; Molinska-Glura, M.; Zagor, M.; Krzeski, A.; Skarzynski, H.; Misiak, J.; Dzaman, K.; Bilusiak, M.; et al. Molecular signaling of the HMGB1/RAGE axis contributes to cholesteatoma pathogenesis. J. Mol. Med. (Berl.) 2015, 93, 305–314.
  36. Wang, Y.; Zhang, Y.; Peng, G.; Han, X. Glycyrrhizin ameliorates atopic dermatitis-like symptoms through inhibition of HMGB1. Int. Immunopharmacol. 2018, 60, 9–17.
  37. de Carvalho, G.C.; Hirata, F.Y.A.; Domingues, R.; Figueiredo, C.A.; Zaniboni, M.C.; Pereira, N.V.; Sotto, M.N.; Aoki, V.; da Silva Duarte, A.J.; Sato, M.N. Up-regulation of HMGB1 and TLR4 in skin lesions of lichen planus. Arch. Dermatol. Res. 2018, 310, 523–528.
  38. Karuppagounder, V.; Arumugam, S.; Thandavarayan, R.A.; Pitchaimani, V.; Sreedhar, R.; Afrin, R.; Harima, M.; Suzuki, H.; Nomoto, M.; Miyashita, S.; et al. Modulation of HMGB1 translocation and RAGE/NFkappaB cascade by quercetin treatment mitigates atopic dermatitis in NC/Nga transgenic mice. Exp. Dermatol. 2015, 24, 418–423.
  39. Iwamura, M.; Yamamoto, Y.; Kitayama, Y.; Higuchi, K.; Fujimura, T.; Hase, T.; Yamamoto, H. Epidermal expression of receptor for advanced glycation end products (RAGE) is related to inflammation and apoptosis in human skin. Exp. Dermatol. 2016, 25, 235–237.
  40. Mulder, D.J.; van Haelst, P.L.; Gross, S.; de Leeuw, K.; Bijzet, J.; Graaff, R.; Gans, R.O.; Zijlstra, F.; Smit, A.J. Skin autofluorescence is elevated in patients with stable coronary artery disease and is associated with serum levels of neopterin and the soluble receptor for advanced glycation end products. Atherosclerosis 2008, 197, 217–223.
  41. Kim, M.H.; Choi, Y.W.; Choi, H.Y.; Myung, K.B.; Cho, S.N. The expression of RAGE and EN-RAGE in leprosy. Br. J. Dermatol. 2006, 154, 594–601.
  42. Na, M.; Mohammad, M.; Fei, Y.; Wang, W.; Holdfeldt, A.; Forsman, H.; Ali, A.; Pullerits, R.; Jin, T. Lack of receptor for advanced glycation end products leads to less severe staphylococcal skin infection but more skin abscesses and prolonged wound healing. J. Infect. Dis. 2018, 218, 791–800.
  43. Niu, Y.; Xie, T.; Ge, K.; Lin, Y.; Lu, S. Effects of extracellular matrix glycosylation on proliferation and apoptosis of human dermal fibroblasts via the receptor for advanced glycosylated end products. Am. J. Dermatopathol. 2008, 30, 344–351.
  44. Park, H.Y.; Kim, J.H.; Jung, M.; Chung, C.H.; Hasham, R.; Park, C.S.; Choi, E.H. A long-standing hyperglycaemic condition impairs skin barrier by accelerating skin ageing process. Exp. Dermatol. 2011, 20, 969–974.
  45. Yamagishi, S.; Fujimori, H.; Yonekura, H.; Yamamoto, Y.; Yamamoto, H. Advanced glycation endproducts inhibit prostacyclin production and induce plasminogen activator inhibitor-1 in human microvascular endothelial cells. Diabetologia 1998, 41, 1435–1441.
  46. Bekircan-Kurt, C.E.; Uçeyler, N.; Sommer, C. Cutaneous activation of RAGE in nonsystemic vasculitic and diabetic neuropathy. Muscle Nerve 2014, 50, 377–383.
  47. Bakker, S.F.; Tushuizen, M.E.; Gözütok, E.; Çiftci, A.; Gelderman, K.A.; Mulder, C.J.; Simsek, S. Advanced glycation end products (AGEs) and the soluble receptor for AGE (sRAGE) in patients with type 1 diabetes and coeliac disease. Nutr. Metab. Cardiovasc. Dis. 2015, 25, 230–235.
  48. Kanková, K.; Márová, I.; Záhejský, J.; Muzík, J.; Stejskalová, A.; Znojil, V.; Vácha, J. Polymorphisms 1704G/T and 2184A/G in the RAGE gene are associated with antioxidant status. Metabolism 2001, 50, 1152–1160.
  49. Kanková, K.; Záhejský, J.; Márová, I.; Muzík, J.; Kuhrová, V.; Blazková, M.; Znojil, V.; Beránek, M.; Vácha, J. Polymorphisms in the RAGE gene influence susceptibility to diabetes-associated microvascular dermatoses in NIDDM. J. Diabetes Complicat. 2001, 15, 185–192.
  50. Wear-Maggitti, K.; Lee, J.; Conejero, A.; Schmidt, A.M.; Grant, R.; Breitbart, A. Use of topical sRAGE in diabetic wounds increases neovascularization and granulation tissue formation. Ann. Plast. Surg. 2004, 52, 519–521.
  51. Ji, X.Y.; Chen, Y.; Ye, G.H.; Dong, M.W.; Lin, K.Z.; Han, J.G.; Feng, X.P.; Li, X.B.; Yu, L.S.; Fan, Y.Y. Detection of RAGE expression and its application to diabetic wound age estimation. Int. J. Legal Med. 2017, 131, 691–698.
  52. Yoshizaki, A.; Komura, K.; Iwata, Y.; Ogawa, F.; Hara, T.; Muroi, E.; Takenaka, M.; Shimizu, K.; Hasegawa, M.; Fujimoto, M.; et al. Clinical significance of serum HMGB-1 and sRAGE levels in systemic sclerosis: Association with disease severity. J. Clin. Immunol. 2009, 29, 180–189.
  53. Davies, C.A.; Herrick, A.L.; Cordingley, L.; Freemont, A.J.; Jeziorska, M. Expression of advanced glycation end products and their receptor in skin from patients with systemic sclerosis with and without calcinosis. Rheumatology (Oxford) 2009, 48, 876–882.
  54. Zhao, J.; Zhong, A.; Friedrich, E.E.; Jia, S.; Xie, P.; Galiano, R.D.; Mustoe, T.A.; Hong, S.J. S100A12 induced in the epidermis by reduced hydration activates dermal fibroblasts and causes dermal fibrosis. J. Investig. Dermatol. 2017, 137, 650–659.
  55. Lee, E.J.; Kim, J.Y.; Oh, S.H. Advanced glycation end products (AGEs) promote melanogenesis through receptor for AGEs. Sci. Rep. 2016, 6, 27848.
  56. Cui, T.; Zhang, W.; Li, S.; Chen, X.; Chang, Y.; Yi, X.; Kang, P.; Yang, Y.; Chen, J.; Liu, L.; et al. Oxidative stress-induced HMGB1 release from melanocytes: A paracrine mechanism underlying the cutaneous inflammation in vitiligo. J. Investig. Dermatol. 2019, 139, 2174–2184.e4.
  57. Cheong, K.A.; Noh, M.; Kim, C.H.; Lee, A.Y. S100B as a potential biomarker for the detection of cytotoxicity of melanocytes. Exp. Dermatol. 2014, 23, 165–171.
More
Information
Subjects: Dermatology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 503
Revisions: 3 times (View History)
Update Date: 08 Mar 2021
1000/1000