Damage-Associated Molecular Patterns in Psoriasis: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: , , , , ,

Psoriasis is a chronic skin disorder that involves both innate and adaptive immune responses in its pathogenesis. Local tissue damage is a hallmark feature of psoriasis and other autoimmune diseases. In psoriasis, damage-associated molecular patterns (DAMPs) released by damaged local tissue act as danger signals and trigger inflammatory responses by recruiting and activating immune cells. They also stimulate the release of pro-inflammatory cytokines and chemokines, which exacerbate the inflammatory response and contribute to disease progression. DAMPs have a dual function. On the one hand, they can regulate cell homeostasis and maintain cell function when present within cells. On the other hand, they can also act as endogenous molecules of cell death or injury and amplify the signal of inflammation through various cell receptors upon release. This leads to the activation of immune cells and the secretion of a large number of inflammatory factors. DAMPs not only play a pro-inflammatory role in acute inflammation, such as sepsis, acute liver injury, or acute pancreatitis, but they also mediate immunity in chronic immune diseases such as rheumatoid arthritis, multiple sclerosis, and inflammatory bowel disease.

  • DAMPs
  • psoriasis
  • drug targets
  • autoimmune diseases

1. HMGB1 and Psoriasis

High mobility group box 1 (HMGB1) is a highly conserved non-histone nucleoprotein with a size of approximately 30 kDa [1]. It comprises two cognate DNA-binding domains and a negatively charged C-terminal domain [2]. HMGB1 is present in all higher eukaryotic cells and assists in DNA replication, transcription, and repair by binding to chromatin in the nucleus [3]. Importantly, it also functions as a DAMP in infection, inflammation, and immune processes, primarily by stimulating TLR2, TLR4, and RAGE receptors. In recent years, increasing evidence suggests that HMGB1 is closely linked to psoriasis. For instance, researchers have detected a significant elevation of serum HMGB1 levels in psoriasis patients compared to healthy individuals, with levels being closely related to disease severity [4]. In addition, the expression of HMGB1 in the lesion sites of psoriasis patients was also significantly increased compared with healthy people [5].
HMGB1 is involved in psoriasis progression through several mechanisms. A study by Zhen Wang et al. revealed that HMGB1 promotes the expression of psoriasis-associated cytokines and antimicrobial peptides by activating autophagy in keratinocytes. HMGB1 also amplifies the IL-23/IL-17 immune cycle by activating dermal γδ T cells to produce IL-17A [6]. Furthermore, HMGB1 upregulates the expression of TLR2, TLR4, and RAGE in the skin lesion area of psoriasis and activates the inflammasome and NF-κB pathway in keratinocytes to promote IL-18 secretion, which would aggravate the condition of IMQ model psoriasis mice [7]. In addition, Lisa Strohbuecker et al. conducted a comprehensive study on the relationship between HMGB1 and immune cells, revealing that HMGB1 expression was markedly elevated in the psoriatic skin lesions, and its receptor RAGE expression was also significantly increased on CD8+ cells and CD4+ Treg cells within the skin lesion area, which was strongly associated with the disease progression in psoriasis [8]. Collectively, HMGB1 plays a crucial role in both downstream keratinocytes and upstream immune cells.
The role of HMGB1 in psoriasis has been extensively studied, and its potential as a therapeutic target has been demonstrated in various preclinical studies. HMGB1 knockdown or inhibition using antibodies or small molecules have shown significant improvements in psoriasis symptoms in animal models. For example, Hmgb1-KD mice and mice treated with HMGB1 antibodies exhibited improved disease outcomes after IMQ-induced psoriasis modeling [6]. Additionally, treatment with the HMGB1 inhibitor glycyrrhizin alleviated the condition of IMQ psoriasis mice [7]. The mechanism of action of HMGB1 blockade involves the inhibition of Th17 cell responses, the reduction in γδ T cells, and the regulation of inflammatory cytokine expression, such as IL-6, TNF-α, IFN-γ, and IL-17, leading to the suppression of clinical and histological evolution of IMQ-treated skin [7][9]. Interestingly, the mechanism of action of HMGB1 has also been implicated in existing psoriasis drugs, such as methotrexate (MTX), which inhibits the interaction between HMGB1/RAGE by binding to the RAGE binding region of HMGB1 [10]. Overall, HMGB1 is an important DAMP in the pathogenesis of psoriasis and a promising diagnostic and therapeutic target.

2. S100 Protein and Psoriasis

The S100 protein family consists of 25 members that share a molecular weight of about 10–12 kDa and maintain an amino acid sequence similarity of 25–65% [11][12]. Most S100 proteins are Ca2+ signaling proteins that contain a conserved calcium-binding motif called EF-hand. Upon binding to Ca2+, S100 proteins interact with other proteins to regulate a range of cellular functions, including cell migration, proliferation, apoptosis, and maintenance of Ca2+ homeostasis [13]. When released outside the cell as DAMPs, S100 proteins play a pro-inflammatory role by binding to receptors such as TLRs and RAGE [11]. In psoriasis, S100A7, S100A8, S100A9, S100A12, and S100A5, defined as “antimicrobial peptides”, are the main S100 family members involved. They are released by keratinocytes and activate innate immunity [14]. Among them, S100A7 is overexpressed in the damaged skin of psoriasis patients but tends to decrease in serum with increasing disease severity [15]. In contrast, both S100A8 and S100A9 are significantly increased in the serum and damaged skin of psoriasis patients [16].
The role of S100 family proteins in regulating psoriasis is still controversial. Hu Lei et al. demonstrated that human S100A7 aggravates psoriasis severity by inducing the expression of interleukin 1α in mature epidermal keratinocytes through the RAGE-p38 MAPK-calpain 1 pathway [17]. In a model of psoriasis induced by IMQ, Joan Defrêne et al. found that S100A8 inhibits the proliferation of keratinocytes and regulates their differentiation [18]. However, some scholars suggest that S100 family proteins are mainly secreted by keratinocytes and act on immune cells. Carolin Christmann et al. found that the expression of S100A8/S100A9 did not significantly affect the maturation and inflammatory response patterns of keratinocytes in primary S100a9−/− keratinocytes, indicating that keratinocytes are not the target cells for the pro-inflammatory effect of S100A8/S100A9 [19].
The potential of S100s as targets for psoriasis diagnosis and treatment has been suggested. Henry J. Grantham et al. proposed that S100A8/9 in serum can serve as a biological indicator of atherosclerosis caused by psoriasis [20]. Helia B. Schonthaler et al. identified S100A9 as a chromatin component that regulates C3 expression in mouse and human cells by binding to a region upstream of the C3 initiation site. Knocking out S100A9 in a psoriasis mouse model strongly attenuated psoriasiform skin disease and inflammation, with reduced C3 volume and mild immune infiltration [21]. However, the study by Joan Defrêne et al. showed that S100A8 and S100A9 knockout mice had more severe symptoms after IMQ-induced psoriasis modeling, with increased IL-17 responses in the dermis and lymph nodes, indicating the anti-inflammatory properties of S100A8 and S100A9 [18]. Overall, while the S100 family is involved in regulating multiple immune pathways in psoriasis, its main role requires further investigation.

3. HSP and Psoriasis

Heat shock proteins (HSPs) are a ubiquitous class of proteins that are synthesized in response to conditions such as heat shock, heavy metals, infection, or an abnormal physiological state. They have intracellular and extracellular functions. Intracellular HSPs are involved in protein folding, recognizing and binding nascent polypeptide chains and partially folded intermediates of proteins to prevent their aggregation and misfolding [22]. In contrast, extracellular HSPs function as DAMPs. Among them, HSP60, HSP70, and HSP90 are mainly involved in psoriasis and act through receptors such as TLR2, TLR4, and CD91 [23][24]. Studies have shown that the average IRID (immunoreactivity intensity distribution index) score of HSP60 expression in the basal, suprabasal, and superficial epidermal layers of psoriatic skin is higher than that of healthy skin [25]. HSP70 is also expressed in the skin of psoriasis sufferers but primarily in the basal layer [26]. HSP90 expression increases throughout the year with the frequency of psoriasis exacerbations, and psoriasis-related hyperlipidemia or diabetes also upregulates HSP90 expression [27]. These studies have shown that the expression levels of HSP family proteins are closely related to the severity of psoriasis.
Recent studies have shed light on the mechanisms underlying the involvement of HSP family proteins in the regulation of psoriasis. For example, O. Boyman et al. reported that dendritic antigen-presenting cells expressing the HSP receptor CD91 accumulate during the occurrence of psoriasis lesions, accompanied by activation of NF-κB and increased production of TNF. This suggests that HSP family proteins may affect the migration of dendritic cells through CD91 [28]. Furthermore, Jonathan L. et al. demonstrated that HSP can induce the maturation of blood-derived DCs, accompanied by increased IL-12 production and enhanced antigen presentation function [29]. The anti-inflammatory pain drug luteolin can regulate the ratio of immune cells by inhibiting the expression of HSP90 and exosome secretion and relieving the lesions and symptoms of psoriasis [30]. In addition, some drugs treat psoriasis by targeting the MAPK pathway and inhibiting the expression of HSP90 and HSP60 in keratinocytes [31]. HSP90 inhibitors have been shown to significantly improve psoriasis symptoms in a xenograft mouse model [32]. Specifically, Rikke S. Hansen et al. demonstrated that HSP90 inhibitors can reduce the expression of pro-inflammatory cytokines IL-23, IL-6, and TNF caused by activation of TLR3 in keratinocytes, leading to a reduction in the inflammatory response [33].

4. Other DAMPs and Psoriasis

Other DAMPs, such as cytosolic DNA, ATP, self-RNA, the LL37 complex, and IL-33, are also implicated in psoriasis. For instance, the release of cytosolic DNA as DAMP induces the synergistic activation of STING in immune cells and keratinocytes, amplifying the inflammatory response in psoriasis [34]. ATP released from keratinocytes triggers the Koebner phenomenon in psoriasis [35]. The self-RNA and LL37 complex can activate DC to generate inflammatory cytokines and aggravate psoriasis [36]. IL-33 can worsen the condition of IMQ psoriasis model mice, reduce CD4 + T and CD8 + T cells in the spleen of psoriatic mice, inhibit autophagy in the skin, and promote STAT3 tyrosine phosphorylation [37]. In summary, despite the critical roles of various DAMPs in regulating inflammation in psoriasis (Table 1), further studies are required to explore their specific functions and mechanisms.
Table 1. The role of DAMP in psoriasis.
DAMP Psoriasis Patients Experimental Psoriasis
HMGB1 It is increased in the serum and skin of psoriasis patients [5][8]. Its inhibitor, antibody, or deficiency reduces inflammation of IMQ-induced mice [6][7][9].
S100s S100A7: It is strongly expressed in psoriatic lesions and decreased in the serum of patients with psoriasis [15].
S100A8/A9: It is increased in the serum and lesion skin of psoriasis patients [16].
S100A8/A9: Serum S100A8/A9 levels may act as biomarker of atherosclerosis severity in psoriasis; deletion of them enhances inflammation in imiquimod-induced psoriasis mice [18][20].
HSPs HSP60: The mean IRIDI scores for its expression in the basal, suprabasal, and superficial epidermal layers of psoriasis are higher than those of normal skin [25].
HSP70: The expression of it has no obvious differences between lesion and normal skin [26].
HSP90: The expression of it increases with the frequency of exacerbations of psoriasis throughout the year [27].
HSP70: It prevents imiquimod-induced, psoriasis-like inflammation in mice [38].
HSP90: Its inhibitor alleviates psoriasis in a xenograft transplantation model [33].

This entry is adapted from the peer-reviewed paper 10.3390/ijms25020771

References

  1. Goodwin, G.H.; Johns, E.W. The isolation and purification of the high mobility group (HMG) nonhistone chromosomal proteins. Methods Cell. Biol. 1977, 16, 257–267.
  2. Bustin, M.; Reeves, R. High-mobility-group chromosomal proteins: Architectural components that facilitate chromatin function. Prog. Nucleic Acid Res. Mol. Biol. 1996, 54, 35–100.
  3. Chen, G.; Ward, M.F.; Sama, A.E.; Wang, H. Extracellular HMGB1 as a proinflammatory cytokine. J. Interferon Cytokine Res. 2004, 24, 329–333.
  4. Bergmann, C.; Strohbuecker, L.; Lotfi, R.; Sucker, A.; Joosten, I.; Koenen, H.; Korber, A. High mobility group box 1 is increased in the sera of psoriatic patients with disease progression. J. Eur. Acad. Dermatol. Venereol. 2016, 30, 435–441.
  5. Watanabe, T.; Yamaguchi, Y.; Watanabe, Y.; Takamura, N.; Aihara, M. Increased level of high mobility group box 1 in the serum and skin in patients with generalized pustular psoriasis. J. Dermatol. 2020, 47, 1033–1036.
  6. Wang, Z.; Zhou, H.; Zheng, H.; Zhou, X.; Shen, G.; Teng, X.; Liu, X.; Zhang, J.; Wei, X.; Hu, Z.; et al. Autophagy-based unconventional secretion of HMGB1 by keratinocytes plays a pivotal role in psoriatic skin in fl ammation. Autophagy 2021, 17, 529–552.
  7. Zhang, W.; Guo, S.; Li, B.; Liu, L.; Ge, R.; Cao, T.; Wang, H.; Gao, T.; Wang, G.; Li, C. Proinflammatory effect of high-mobility group protein B1 on keratinocytes: An autocrine mechanism underlying psoriasis development. J. Pathol. 2017, 241, 392–404.
  8. Strohbuecker, L.; Koenen, H.; van Rijssen, E.; van Cranenbroek, B.; Fasse, E.; Joosten, I.; Korber, A.; Bergmann, C. Increased dermal expression of chromatin-associated protein HMGB1 and concomitant T-cell expression of the DNA RAGE in patients with psoriasis vulgaris. Psoriasis 2019, 9, 7–17.
  9. Chen, T.; Fu, L.X.; Guo, Z.P.; Yin, B.; Cao, N.; Qin, S. Involvement of high mobility group box-1 in imiquimod-induced psoriasis-like mice model. J. Dermatol. 2017, 44, 573–581.
  10. Kuroiwa, Y.; Takakusagi, Y.; Kusayanagi, T.; Kuramochi, K.; Imai, T.; Hirayama, T.; Ito, I.; Yoshida, M.; Sakaguchi, K.; Sugawara, F. Identification and characterization of the direct interaction between methotrexate (MTX) and high-mobility group box 1 (HMGB1) protein. PLoS ONE 2013, 8, e63073.
  11. Gonzalez, L.L.; Garrie, K.; Turner, M.D. Role of S100 proteins in health and disease. Biochim. Biophys. Acta Mol. Cell Res. 2020, 1867, 118677.
  12. Schafer, B.W.; Heizmann, C.W. The S100 family of EF-hand calcium-binding proteins: Functions and pathology. Trends Biochem. Sci. 1996, 21, 134–140.
  13. Donato, R.; Cannon, B.R.; Sorci, G.; Riuzzi, F.; Hsu, K.; Weber, D.J.; Geczy, C.L. Functions of S100 proteins. Curr. Mol. Med. 2013, 13, 24–57.
  14. Buchau, A.S.; Gallo, R.L. Innate immunity and antimicrobial defense systems in psoriasis. Clin. Dermatol. 2007, 25, 616–624.
  15. Anderson, K.S.; Wong, J.; Polyak, K.; Aronzon, D.; Enerback, C. Detection of psoriasin/S100A7 in the sera of patients with psoriasis. Br. J. Dermatol. 2009, 160, 325–332.
  16. Benoit, S.; Toksoy, A.; Ahlmann, M.; Schmidt, M.; Sunderkotter, C.; Foell, D.; Pasparakis, M.; Roth, J.; Goebeler, M. Elevated serum levels of calcium-binding S100 proteins A8 and A9 reflect disease activity and abnormal differentiation of keratinocytes in psoriasis. Br. J. Dermatol. 2006, 155, 62–66.
  17. Lei, H.; Li, X.; Jing, B.; Xu, H.; Wu, Y. Human S100A7 Induces Mature Interleukin1alpha Expression by RAGE-p38 MAPK-Calpain1 Pathway in Psoriasis. PLoS ONE 2017, 12, e0169788.
  18. Defrene, J.; Berrazouane, S.; Esparza, N.; Page, N.; Cote, M.F.; Gobeil, S.; Aoudjit, F.; Tessier, P.A. Deletion of S100a8 and S100a9 Enhances Skin Hyperplasia and Promotes the Th17 Response in Imiquimod-Induced Psoriasis. J. Immunol. 2021, 206, 505–514.
  19. Christmann, C.; Zenker, S.; Martens, L.; Hubner, J.; Loser, K.; Vogl, T.; Roth, J. Interleukin 17 Promotes Expression of Alarmins S100A8 and S100A9 During the Inflammatory Response of Keratinocytes. Front. Immunol. 2020, 11, 599947.
  20. Grantham, H.J.; Hussain, A.B.; Reynolds, N.J. Serum S100A8/A9 May Act as Biomarker of Atherosclerosis Severity in Psoriasis. J. Investig. Dermatol. 2022, 142, 2848–2850.
  21. Schonthaler, H.B.; Guinea-Viniegra, J.; Wculek, S.K.; Ruppen, I.; Ximenez-Embun, P.; Guio-Carrion, A.; Navarro, R.; Hogg, N.; Ashman, K.; Wagner, E.F. S100A8-S100A9 protein complex mediates psoriasis by regulating the expression of complement factor C3. Immunity 2013, 39, 1171–1181.
  22. Tsan, M.F.; Gao, B. Heat shock protein and innate immunity. Cell. Mol. Immunol. 2004, 1, 274–279.
  23. Asea, A.; Rehli, M.; Kabingu, E.; Boch, J.A.; Bare, O.; Auron, P.E.; Stevenson, M.A.; Calderwood, S.K. Novel signal transduction pathway utilized by extracellular HSP70: Role of toll-like receptor (TLR) 2 and TLR4. J. Biol. Chem. 2002, 277, 15028–15034.
  24. Basu, S.; Binder, R.J.; Ramalingam, T.; Srivastava, P.K. CD91 is a common receptor for heat shock proteins gp96, hsp90, hsp70, and calreticulin. Immunity 2001, 14, 303–313.
  25. Bayramgurler, D.; Ozkara, S.K.; Apaydin, R.; Ercin, C.; Bilen, N. Heat shock proteins 60 and 70 expression of cutaneous lichen planus: Comparison with normal skin and psoriasis vulgaris. J. Cutan. Pathol. 2004, 31, 586–594.
  26. Boehncke, W.H.; Dahlke, A.; Zollner, T.M.; Sterry, W. Differential expression of heat shock protein 70 (HSP70) and heat shock cognate protein 70 (HSC70) in human epidermis. Arch. Dermatol. Res. 1994, 287, 68–71.
  27. Czajkowski, R.; Kaszewski, S.; Tadrowski, T.; Grzanka, D.; Ziandarska, J.; Drewa, G.; Marek-Jozefowicz, L. Does HSP90 play an important role in psoriasis? Postep. Dermatol. Alergol. 2021, 38, 319–326.
  28. Boyman, O.; Conrad, C.; Dudli, C.; Kielhorn, E.; Nickoloff, B.J.; Nestle, F.O. Activation of dendritic antigen-presenting cells expressing common heat shock protein receptor CD91 during induction of psoriasis. Br. J. Dermatol. 2005, 152, 1211–1218.
  29. Curry, J.L.; Qin, J.Z.; Bonish, B.; Carrick, R.; Bacon, P.; Panella, J.; Robinson, J.; Nickoloff, B.J. Innate immune-related receptors in normal and psoriatic skin. Arch. Pathol. Lab. Med. 2003, 127, 178–186.
  30. Lv, J.; Zhou, D.; Wang, Y.; Sun, W.; Zhang, C.; Xu, J.; Yang, H.; Zhou, T.; Li, P. Effects of luteolin on treatment of psoriasis by repressing HSP90. Int. Immunopharmacol. 2020, 79, 106070.
  31. Xiang, X.; Tu, C.; Li, Q.; Wang, W.; Huang, X.; Zhao, Z.; Xiong, H.; Mei, Z. Oxymatrine ameliorates imiquimod-induced psoriasis pruritus and inflammation through inhibiting heat shock protein 90 and heat shock protein 60 expression in keratinocytes. Toxicol. Appl. Pharmacol. 2020, 405, 115209.
  32. Stenderup, K.; Rosada, C.; Gavillet, B.; Vuagniaux, G.; Dam, T.N. Debio 0932, a new oral Hsp90 inhibitor, alleviates psoriasis in a xenograft transplantation model. Acta Derm. Venereol. 2014, 94, 672–676.
  33. Hansen, R.S.; Thuesen, K.K.H.; Bregnhoj, A.; Moldovan, L.I.; Kristensen, L.S.; Grek, C.L.; Ghatnekar, G.S.; Iversen, L.; Johansen, C. The HSP90 inhibitor RGRN-305 exhibits strong immunomodulatory effects in human keratinocytes. Exp. Dermatol. 2021, 30, 773–781.
  34. Yu, Y.; Xue, X.; Tang, W.; Su, L.; Zhang, L.; Zhang, Y. Cytosolic DNA-Mediated STING-Dependent Inflammation Contributes to the Progression of Psoriasis. J. Investig. Dermatol. 2022, 142, 898–906.e4.
  35. Okamoto, T.; Ogawa, Y.; Kinoshita, M.; Ihara, T.; Shimada, S.; Koizumi, S.; Kawamura, T. Mechanical stretch-induced ATP release from keratinocytes triggers Koebner phenomenon in psoriasis. J. Dermatol. Sci 2021, 103, 60–62.
  36. Ganguly, D.; Chamilos, G.; Lande, R.; Gregorio, J.; Meller, S.; Facchinetti, V.; Homey, B.; Barrat, F.J.; Zal, T.; Gilliet, M. Self-RNA-antimicrobial peptide complexes activate human dendritic cells through TLR7 and TLR8. J. Exp. Med. 2009, 206, 1983–1994.
  37. Duan, Y.; Dong, Y.; Hu, H.; Wang, Q.; Guo, S.; Fu, D.; Song, X.; Kalvakolanu, D.V.; Tian, Z. IL-33 contributes to disease severity in Psoriasis-like models of mouse. Cytokine 2019, 119, 159–167.
  38. Seifarth, F.G.; Lax, J.E.; Harvey, J.; DiCorleto, P.E.; Husni, M.E.; Chandrasekharan, U.M.; Tytell, M. Topical heat shock protein 70 prevents imiquimod-induced psoriasis-like inflammation in mice. Cell Stress Chaperones 2018, 23, 1129–1135.
More
This entry is offline, you can click here to edit this entry!
Video Production Service