Damage-associated molecular pattern

Damage-associated molecular patterns (DAMPs),[1] also known as danger-associated molecular patterns, danger signals, and alarmin, are host biomolecules that can initiate and perpetuate a noninfectious inflammatory response. For example, they are released from damaged or dying cells and activate the innate immune system by interacting with pattern recognition receptors (PRRs).[2] In contrast, pathogen-associated molecular patterns (PAMPs) initiate and perpetuate the infectious pathogen-induced inflammatory response.[3] Many DAMPs are nuclear or cytosolic proteins with defined intracellular function that, when released outside the cell after tissue injury, move from a reducing to an oxidizing milieu resulting in their functional denaturation.[4] Above those DAMPs, there are other DAMPs originated from different sources, such as ECM (extracellular matrix), mitochondria, granules, ER (endoplasmic reticulum), and plasma membrane. We can characterize those DAMPs and its receptors as follows : Table1.[2]

Table1. List of DAMPs from several origins and their receptors
origin Major DAMPs Receptors
ECM Biglycan TLR2, TLR4, NLRP3
Decorin TLR2, TLR4
Versican TLR2, TLR6, CD14
LMW hyaluronan TLR2, TLR4, NLRP3
Heparan sulfate TLR4
Fibronectin (EDA domain) TLR4
Fibrinogen TLR4
Tenascin C TLR4
Intracellular compartments Cytosol uric acid NLPR3, P2X7
S100 proteins TLR2, TLR4, RAGE
HSP (Heat Shock Proteins) TLR2, TLR4, CD91
ATP P2X7, P2Y2
F-actin DNGR-1
Cyclophilin A CD147
TLR2, NLRP1, NLRP3, CD36, RAGE
Nuclear Histones TLR2, TLR4
HMGB1 TLR2, TLR4, RAGE
HMGN1 TLR4
IL-1α IL-1R
IL-33 ST2
SAP130 Mincle
DNA TLR9, AIM2
RNA TLR3, TLR7, TLR8, RIG-I, MDAS
Mitochondria mtDNA TLR9
TFAM RAGE
Formyl peptide FPR1
mROS NLRP3
ER Calreticulin CD91
Granule Defensins TLR4
Cathelicidin (LL37) P2X7, FPR2
EDN (eosinophil-derived neurotoxin) TLR2
Granulysin TLR4
Plasma membrane Syndecans TLR4
Glypicans TLR4

As an example of nucleotide molecule, tumor cell DNA is released during necrosis (a kind of cell death), with the potential to be recognized as a DAMP.[5]

History

Two papers appearing in 1994 presaged the deeper understanding of innate immune reactivity, dictating the subsequent nature of the adaptive immune response. The first[6] came from transplant surgeons who conducted a prospective randomized double-blind placebo-controlled trial. Administration of recombinant human superoxide dismutase (rh-SOD) in recipients of cadaveric renal allografts demonstrated prolonged patient and graft survival with improvement in both acute and chronic rejection events. They speculated that the effect was related to its antioxidant action on the initial ischemia/reperfusion injury of the renal allograft, thereby reducing the immunogenicity of the allograft and the "grateful dead" or stressed cells. Thus free radical-mediated reperfusion injury-was seen to contribute to the process of innate and subsequent adaptive immune responses.

The second[7] suggested the possibility that the immune system detected "danger", through a series of what we would now call damage associated molecular pattern molecules (DAMPs), working in concert with both positive and negative signals derived from other tissues. Thus these two papers together presaged the modern sense of the role of DAMPs and redox reviewed here, important apparently for both plant and animal resistance to pathogens and the response to cellular injury or damage. Although many immunologists had earlier noted that various "danger signals" could initiate innate immune responses, the "DAMP" was first described by Seong and Matzinger in 2004.[1]

Examples

DAMPs vary greatly depending on the type of cell (epithelial or mesenchymal) and injured tissue.

- Protein DAMPs include intracellular proteins, such as heat-shock proteins[8] or HMGB1[9], and materials derived from the extracellular matrix that are generated following tissue injury, such as hyaluronan fragments.[10]

- Non-protein DAMPs include ATP,[11][12] uric acid,[13] heparin sulfate and DNA.[5]


1. Protein DAMPs

(1) HMGB1 : HMGB1 (High-mobility group box 1), a member of the HMG protein family, is a prototypical chromatin-associated LSP (leaderless secreted protein), secreted by hematopoietic cells through a lysosome-mediated pathway.[14] HMGB1 is a major mediator of endotoxin shock[15] and is recognized as a DAMP by certain immune cells, triggering an inflammatory response.[9] It is known to induce inflammation by activating NF-kB pathway by binding to TLR, TLR4, TLR9, and RAGE (receptor for advanced glycation end products).[16] HMGB1 can also induce dendritic cell maturation via upregulation of CD80, CD83, CD86 and CD11c, and the production of other pro-inflammatory cytokines in myeloid cells (IL-1, TNF-a, IL-6, IL-8), and it can lead to increased expression of cell adhesion molecules (ICAM-1, VCAM-1) on endothelial cells.[17]

(2) DNA and RNA : The presence of DNA anywhere other than the nucleus or mitochondria is perceived as a DAMP and triggers responses mediated by TLR9 and DAI that drive cellular activation and immunoreactivity. Some tissues such as the gut are inhibited by DNA in their immune response (this needs a reference, and may be a misinterpretation of what the gut does). Similarly, damaged RNAs released from UVB-exposed keratinocytes activate TLR3 on intact keratinocytes. TLR3 activation stimulates TNF-alpha and IL-6 production, which initiate the cutaneous inflammation associated with sunburn.[18]

(3) S100 proteins : S100 is a multigenic family of calcium modulated proteins involved in intracellular and extracellular regulatory activities with a connection to cancer as well as tissue, particularly neuronal, injury.[19][20][21][22][23][16] Their main function is the management of calcium storage and shuffling. Although they have various functions, including cell proliferation, differentiation, migration, and energy metabolism, they also act as DAMPs by interacting with their receptors (TLR2, TLR4, RAGE) after they are released from phagocytes.[2]

(4) Mono and polysaccharides : The ability of the immune system to recognize hyaluronan fragments is one example of how DAMPs can be made of sugars.[24]


2. Non-protein DAMPs

- Purine metabolites : Nucleotides (e.g., ATP) and nucleosides (e.g., adenosine) that have reached the extracellular space can also serve as danger signals by signaling through purinergic receptors.[25] ATP and adenosine are released in high concentrations after catastrophic disruption of the cell, as occurs in necrotic cell death.[26] Extracellular ATP triggers mast cell degranulation by signaling through P2X7 receptors.[27][25][28] Similarly, adenosine triggers degranulation through P1 receptors. Uric acid is also an endogenous danger signal released by injured cells.[24] Adenosine triphosphate (ATP) and uric acid, which are purine metabolites, activate NLR family, pyrin domain containing (NLRP) 3 inflammasomes to induce IL-1β and IL-18.[2]


Clinical targets in various disorders

Theoretically, the application of therapeutics in this area to treat disorders as arthritis, cancer, ischemia-reperfusion, myocardial infarction and stroke could include options as:

- Preventing DAMP release

[proapoptotic therapies; platinums; ethyl pyruvate]

- Neutralizing or blocking DAMPs extracellularly

[anti-HMGB1; rasburicase; sRAGE, etc.]

- Blocking the DAMP receptors or their signaling

[RAGE small molecule antagonists; TLR4 antagonists; antibodies to DAMP-R].

1. DAMPs can be used as biomarkers for inflammatory diseases and potential therapeutic targets. For example, increased S100A8/A9 is associated with osteophyte progression in early human OA (osteoarthritis), suggesting that S100 proteins can be used as biomarkers for the diagnosis of the progressive grade of OA. Furthermore, DAMP can be a useful prognostic factor for cancer. This would improve patient classification, and a suitable therapy would be given to patients by diagnosing with DAMPs. The regulation of DAMPs signaling can be a potential therapeutic target to reduce inflammation and treat diseases. For example, administration of neutralizing HMGB1 antibodies or truncated HMGB1-derived A-box protein ameliorated arthritis in collagen-induced arthritis rodent models. Clinical trials with HSP inhibitors have also been reported. For non-small cell lung cancer (NSCLC), HSP27, HSP70, and HSP90 inhibitors are under investigation in clinical trials. In addition, treatment with dnaJP1, which is a synthetic peptide derived from DnaJ (HSP40), had a curative effect in RA (rheumatoid arthritis) patients without critical side effects. Taken together, DAMPs can be useful therapeutic targets for various human diseases, including cancer and autoimmune diseases.[2]

2. Recent evidence revealed that DAMPs can trigger re-epithelialization upon kidney injury, contributing to epithelial-mesenchymal transition and, potentially, to myofibroblast differentiation and proliferation. Thus, these discoveries suggest that DAMPs drive not only immune injury but also kidney regeneration and renal scarring. For example, TLR2-agonistic DAMPs activate renal progenitor cells to regenerate epithelial defects in injured tubules.Also, TLR4-agonistic DAMPs induce renal dendritic cells to release IL-22, which also accelerates tubule re-epithelialization in AKI[29]. Finally, DAMPs also promote renal fibrosis by inducing NLRP3, which also promotes TGF-β receptor signaling.[30]

References

  1. Seong SY, Matzinger P (June 2004). "Hydrophobicity: an ancient damage-associated molecular pattern that initiates innate immune responses". Nature Reviews. Immunology. 4 (6): 469–78. doi:10.1038/nri1372. PMID 15173835.
  2. Roh JS, Sohn DH (August 2018). "Damage-Associated Molecular Patterns in Inflammatory Diseases". Immune Network. 18 (4): e27. doi:10.4110/in.2018.18.e27. PMC 6117512. PMID 30181915.
  3. Janeway C (September 1989). "Immunogenicity signals 1,2,3 ... and 0". Immunology Today. 10 (9): 283–6. doi:10.1016/0167-5699(89)90081-9. PMID 2590379.
  4. Rubartelli A, Lotze MT (October 2007). "Inside, outside, upside down: damage-associated molecular-pattern molecules (DAMPs) and redox". Trends in Immunology. 28 (10): 429–36. doi:10.1016/j.it.2007.08.004. PMID 17845865.
  5. Farkas AM, Kilgore TM, Lotze MT (December 2007). "Detecting DNA: getting and begetting cancer". Current Opinion in Investigational Drugs. 8 (12): 981–6. PMID 18058568.
  6. Land W, Schneeberger H, Schleibner S, Illner WD, Abendroth D, Rutili G, Arfors KE, Messmer K (January 1994). "The beneficial effect of human recombinant superoxide dismutase on acute and chronic rejection events in recipients of cadaveric renal transplants". Transplantation. 57 (2): 211–7. doi:10.1097/00007890-199401001-00010. PMID 8310510.
  7. Matzinger P (1994). "Tolerance, danger, and the extended family". Annual Review of Immunology. 12: 991–1045. doi:10.1146/annurev.iy.12.040194.005015. PMID 8011301.
  8. Panayi GS, Corrigall VM, Henderson B (August 2004). "Stress cytokines: pivotal proteins in immune regulatory networks; Opinion". Current Opinion in Immunology. 16 (4): 531–4. doi:10.1016/j.coi.2004.05.017. PMID 15245751.
  9. Scaffidi P, Misteli T, Bianchi ME (July 2002). "Release of chromatin protein HMGB1 by necrotic cells triggers inflammation". Nature. 418 (6894): 191–5. doi:10.1038/nature00858. PMID 12110890.
  10. Scheibner KA, Lutz MA, Boodoo S, Fenton MJ, Powell JD, Horton MR (July 2006). "Hyaluronan fragments act as an endogenous danger signal by engaging TLR2". Journal of Immunology. 177 (2): 1272–81. doi:10.4049/jimmunol.177.2.1272. PMID 16818787.
  11. Boeynaems JM, Communi D (May 2006). "Modulation of inflammation by extracellular nucleotides". The Journal of Investigative Dermatology. 126 (5): 943–4. doi:10.1038/sj.jid.5700233. PMID 16619009.
  12. Bours MJ, Swennen EL, Di Virgilio F, Cronstein BN, Dagnelie PC (November 2006). "Adenosine 5'-triphosphate and adenosine as endogenous signaling molecules in immunity and inflammation". Pharmacology & Therapeutics. 112 (2): 358–404. doi:10.1016/j.pharmthera.2005.04.013. PMID 16784779.
  13. Shi Y, Evans JE, Rock KL (October 2003). "Molecular identification of a danger signal that alerts the immune system to dying cells". Nature. 425 (6957): 516–21. Bibcode:2003Natur.425..516S. doi:10.1038/nature01991. PMID 14520412.
  14. Gardella S, Andrei C, Ferrera D, Lotti LV, Torrisi MR, Bianchi ME, Rubartelli A (October 2002). "The nuclear protein HMGB1 is secreted by monocytes via a non-classical, vesicle-mediated secretory pathway". EMBO Reports. 3 (10): 995–1001. doi:10.1093/embo-reports/kvf198. PMC 1307617. PMID 12231511.
  15. Wang H, Bloom O, Zhang M, Vishnubhakat JM, Ombrellino M, Che J, Frazier A, Yang H, Ivanova S, Borovikova L, Manogue KR, Faist E, Abraham E, Andersson J, Andersson U, Molina PE, Abumrad NN, Sama A, Tracey KJ (July 1999). "HMG-1 as a late mediator of endotoxin lethality in mice". Science. 285 (5425): 248–51. doi:10.1126/science.285.5425.248. PMID 10398600.
  16. Ibrahim ZA, Armour CL, Phipps S, Sukkar MB (December 2013). "RAGE and TLRs: relatives, friends or neighbours?". Molecular Immunology. 56 (4): 739–44. doi:10.1016/j.molimm.2013.07.008. PMID 23954397.
  17. Galbiati V, Papale A, Galli CL, Marinovich M, Corsini E (November 2014). "Role of ROS and HMGB1 in contact allergen-induced IL-18 production in human keratinocytes". The Journal of Investigative Dermatology. 134 (11): 2719–2727. doi:10.1038/jid.2014.203. PMID 24780928.
  18. Bernard JJ, Cowing-Zitron C, Nakatsuji T, Muehleisen B, Muto J, Borkowski AW, Martinez L, Greidinger EL, Yu BD, Gallo RL (August 2012). "Ultraviolet radiation damages self noncoding RNA and is detected by TLR3". Nature Medicine. 18 (8): 1286–90. doi:10.1038/nm.2861. PMC 3812946. PMID 22772463.
  19. Diederichs S, Bulk E, Steffen B, Ji P, Tickenbrock L, Lang K, Zänker KS, Metzger R, Schneider PM, Gerke V, Thomas M, Berdel WE, Serve H, Müller-Tidow C (August 2004). "S100 family members and trypsinogens are predictors of distant metastasis and survival in early-stage non-small cell lung cancer". Cancer Research. 64 (16): 5564–9. doi:10.1158/0008-5472.CAN-04-2004. PMID 15313892.
  20. Emberley ED, Murphy LC, Watson PH (2004). "S100A7 and the progression of breast cancer". Breast Cancer Research. 6 (4): 153–9. doi:10.1186/bcr816. PMC 468668. PMID 15217486.
  21. Emberley ED, Murphy LC, Watson PH (August 2004). "S100 proteins and their influence on pro-survival pathways in cancer". Biochemistry and Cell Biology. 82 (4): 508–15. doi:10.1139/o04-052. PMID 15284904.
  22. Lin J, Yang Q, Yan Z, Markowitz J, Wilder PT, Carrier F, Weber DJ (August 2004). "Inhibiting S100B restores p53 levels in primary malignant melanoma cancer cells". The Journal of Biological Chemistry. 279 (32): 34071–7. doi:10.1074/jbc.M405419200. PMID 15178678.
  23. Marenholz I, Heizmann CW, Fritz G (October 2004). "S100 proteins in mouse and man: from evolution to function and pathology (including an update of the nomenclature)". Biochemical and Biophysical Research Communications. 322 (4): 1111–22. doi:10.1016/j.bbrc.2004.07.096. PMID 15336958.
  24. Maverakis E, Kim K, Shimoda M, Gershwin ME, Patel F, Wilken R, Raychaudhuri S, Ruhaak LR, Lebrilla CB (February 2015). "Glycans in the immune system and The Altered Glycan Theory of Autoimmunity: a critical review". Journal of Autoimmunity. 57: 1–13. doi:10.1016/j.jaut.2014.12.002. PMC 4340844. PMID 25578468.
  25. Russo MV, McGavern DB (October 2015). "Immune Surveillance of the CNS following Infection and Injury". Trends in Immunology. 36 (10): 637–50. doi:10.1016/j.it.2015.08.002. PMC 4592776. PMID 26431941.
  26. Zeh HJ, Lotze MT (2005). "Addicted to death: invasive cancer and the immune response to unscheduled cell death". Journal of Immunotherapy. 28 (1): 1–9. doi:10.1097/00002371-200501000-00001. PMID 15614039.
  27. Kurashima Y, Kiyono H (March 2014). "New era for mucosal mast cells: their roles in inflammation, allergic immune responses and adjuvant development". Experimental & Molecular Medicine. 46 (3): e83. doi:10.1038/emm.2014.7. PMC 3972796. PMID 24626169.
  28. Kurashima Y, Amiya T, Nochi T, Fujisawa K, Haraguchi T, Iba H, Tsutsui H, Sato S, Nakajima S, Iijima H, Kubo M, Kunisawa J, Kiyono H (2012). "Extracellular ATP mediates mast cell-dependent intestinal inflammation through P2X7 purinoceptors". Nature Communications. 3: 1034. Bibcode:2012NatCo...3.1034K. doi:10.1038/ncomms2023. PMC 3658010. PMID 22948816.
  29. "Acute kidney injury", Wikipedia, 2020-06-13, retrieved 2020-06-16
  30. Anders HJ, Schaefer L (July 2014). "Beyond tissue injury-damage-associated molecular patterns, toll-like receptors, and inflammasomes also drive regeneration and fibrosis". Journal of the American Society of Nephrology. 25 (7): 1387–400. doi:10.1681/ASN.2014010117. PMC 4073442. PMID 24762401.

Further reading

This article is issued from Wikipedia. The text is licensed under Creative Commons - Attribution - Sharealike. Additional terms may apply for the media files.