Role of necroptosis in infection-related, immune-mediated, and autoimmune skin diseases
Lulu Liu
Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
Search for more papers by this authorZiting Tang
Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
Search for more papers by this authorYilan Zeng
Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
Search for more papers by this authorYuanhong Liu
Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
Search for more papers by this authorLu Zhou
Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
Search for more papers by this authorCorresponding Author
Shengbo Yang
Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
Correspondence
Shengbo Yang and Dan Wang, Department of Dermatology, The Third Xiangya Hospital, Central South University, 308 Tongzipo Road, Changsha, Hunan Province 410013, China.
Emails: [email protected]; [email protected]
Search for more papers by this authorCorresponding Author
Dan Wang
Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
Correspondence
Shengbo Yang and Dan Wang, Department of Dermatology, The Third Xiangya Hospital, Central South University, 308 Tongzipo Road, Changsha, Hunan Province 410013, China.
Emails: [email protected]; [email protected]
Search for more papers by this authorLulu Liu
Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
Search for more papers by this authorZiting Tang
Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
Search for more papers by this authorYilan Zeng
Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
Search for more papers by this authorYuanhong Liu
Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
Search for more papers by this authorLu Zhou
Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
Search for more papers by this authorCorresponding Author
Shengbo Yang
Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
Correspondence
Shengbo Yang and Dan Wang, Department of Dermatology, The Third Xiangya Hospital, Central South University, 308 Tongzipo Road, Changsha, Hunan Province 410013, China.
Emails: [email protected]; [email protected]
Search for more papers by this authorCorresponding Author
Dan Wang
Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, China
Correspondence
Shengbo Yang and Dan Wang, Department of Dermatology, The Third Xiangya Hospital, Central South University, 308 Tongzipo Road, Changsha, Hunan Province 410013, China.
Emails: [email protected]; [email protected]
Search for more papers by this authorAbstract
Regulated necrosis, also termed necroptosis, is another programmed cell death depending on a unique molecular pathway that does not overlap with apoptosis. Tumor necrosis factor and Toll-like receptor family members, interferon, and other mediators are the factors that mainly cause necroptosis. Activating necroptosis by ligands of death receptors requires the kinase activity of receptor-interacting proteins 1 and 3, and a mixed lineage kinase domain-like protein, which is a critical downstream mediator of necroptosis. Increasing evidence has revealed that necroptosis does not only involve physiological regulation but also the occurrence, development, and prognosis of certain diseases, such as septicemia, neurodegenerative diseases, and ischemic–reperfusion injury. Many excellent documented systematic discussions of necroptosis and its role in various skin diseases. In this review, we summarize the molecular mechanism of necroptosis, as well as the current knowledge on the contribution of necroptosis, in infection-related, immune-mediated, autoimmune skin diseases, and malignant skin tumors.
CONFLICT OF INTEREST
None declared.
REFERENCES
- 1Zhang S, Tang M-B, Luo H-Y, Shi C-H, Xu Y-M. Necroptosis in neurodegenerative diseases: a potential therapeutic target. Cell Death Dis. 2017; 8(6):e2905.
- 2Gong Y, Fan Z, Luo G, Yang C, Huang Q, Fan K, et al. The role of necroptosis in cancer biology and therapy. Mol Cancer. 2019; 18(1): 100.
- 3Orzalli MH, Kagan JC. Apoptosis and necroptosis as host defense strategies to prevent viral infection. Trends Cell Biol. 2017; 27(11): 800–9.
- 4Shi ZW, Ge LS, Li YC. The role of necroptosis in cardiovascular disease. Front Pharmacol. 2018; 9: 721.
- 5Gupta K, Phan N, Wang Q, Liu BO. Necroptosis in cardiovascular disease - a new therapeutic target. J Mol Cell Cardiol. 2018; 118: 26–35.
- 6Li J, Yang Z, Li YI, Xia J, Li D, Li H, et al. Cell apoptosis, autophagy and necroptosis in osteosarcoma treatment. Oncotarget. 2016; 7(28): 44763–78.
- 7Thapa RJ, Nogusa S, Chen P, Maki JL, Lerro A, Andrake M, et al. Interferon-induced RIP1/RIP3-mediated necrosis requires PKR and is licensed by FADD and caspases. Proc Natl Acad Sci USA. 2013; 110(33): E3109–E3118.
- 8Wu J, Huang Z, Ren J, Zhang Z, He P, Li Y, et al. Mlkl knockout mice demonstrate the indispensable role of Mlkl in necroptosis. Cell Res. 2013; 23(8): 994–1006.
- 9Vercammen D, Beyaert R, Denecker G, Goossens V, Van Loo G, Declercq W, et al. Inhibition of caspases increases the sensitivity of L929 cells to necrosis mediated by tumor necrosis factor. J Exp Med. 1998; 9(187): 1477–85.
- 10Vandenabeele P, Galluzzi L, Vanden Berghe T, Kroemer G. Molecular mechanisms of necroptosis: an ordered cellular explosion. Nat Rev Mol Cell Biol. 2010; 11(10): 700–14.
- 11Zhou W, Yuan J. Necroptosis in health and diseases. Semin Cell Dev Biol. 2014; 35: 14–23.
- 12Christofferson DE, Li Y, Yuan J. Control of life-or-death decisions by RIP1 kinase. Annu Rev Physiol. 2014; 76(1): 129–50.
- 13Wright A, Reiley WW, Chang M, Jin W, Lee AJ, Zhang M, et al. Regulation of early wave of germ cell apoptosis and spermatogenesis by deubiquitinating enzyme CYLD. Dev Cell. 2007; 13: 705–16.
- 14Fritsch M, Günther SD, Schwarzer R, Albert M-C, Schorn F, Werthenbach JP, et al. Caspase-8 is the molecular switch for apoptosis, necroptosis and pyroptosis. Nature. 2019; 575(7784): 683–7.
- 15Tenev T, Bianchi K, Darding M, Broemer M, Langlais C, Wallberg F, et al. The Ripoptosome, a signaling platform that assembles in response to genotoxic stress and loss of IAPs. Mol Cell. 2011; 3(43): 432–48.
- 16Hitomi J, Christofferson DE, Ng A, Yao J, Degterev A, Xavier RJ, et al. Identification of a molecular signaling network that regulates a cellular necrotic cell death pathway. Cell. 2009; 135(7): 1311–23.
- 17Chen J, Kos R, Garssen J, Redegeld F. Molecular insights into the mechanism of necroptosis: the necrosome as a potential therapeutic target. Cells. 2019; 8(12): 1486.
- 18Zhang Y, Chen X, Gueydan C, Han J. Plasma membrane changes during programmed cell deaths. Cell Res. 2018; 28(1): 9–21.
- 19Vanden Berghe T, Hassannia B, Vandenabeele P. An outline of necrosome triggers. Cell Mol Life Sci. 2016; 73(11): 2137–52.
- 20Zhang T, Zhang Y, Cui M, Jin LI, Wang Y, Lv F, et al. CaMKII is a RIP3 substrate mediating ischemia- and oxidative stress-induced myocardial necroptosis. Nat Med. 2016; 22(2): 175–82.
- 21Cai Z, Jitkaew S, Zhao J, Chiang H-C, Choksi S, Liu J, et al. Plasma membrane translocation of trimerized MLKL protein is required for TNF-induced necroptosis. Nat Cell Biol. 2014; 1(16): 55–65.
- 22Wang Z, Jiang H, Chen S, Du F, Wang X. The mitochondrial phosphatase PGAM5 functions at the convergence point of multiple necrotic death pathways. Cell. 2012; 148(1–2): 228–43.
- 23Tait S, Oberst A, Quarato G, Milasta S, Haller M, Wang R, et al. Widespread mitochondrial depletion via mitophagy does not compromise necroptosis. Cell Rep. 2013; 4(5): 878–85.
- 24González-Juarbe N, Gilley RP, Hinojosa CA, Bradley KM, Kamei A, Gao G, et al. Pore-forming toxins induce macrophage necroptosis during acute bacterial pneumonia. PLoS Pathog. 2015; 12(11):e1005337.
- 25Kitur K, Wachtel S, Brown A, Wickersham M, Paulino F, Peñaloza HF, et al. Necroptosis promotes staphylococcus aureus clearance by inhibiting excessive inflammatory signaling. Cell Rep. 2016; 16(8): 2219–30.
- 26Wen S-H, Lin L-N, Wu H-J, Yu LU, Lin LI, Zhu L-L, et al. TNF-α increases Staphylococcus aureus-induced death of human alveolar epithelial cell line A549 associated with RIP3-mediated necroptosis. Life Sci. 2018; 195: 81–6.
- 27Tam VC, Suen R, Treuting PM, Armando A, Lucarelli R, Gorrochotegui-Escalante N, et al. PPARα exacerbates necroptosis, leading to increased mortality in postinfluenza bacterial superinfection. Proc Natl Acad Sci U S A. 2020; 117(27): 15789–98.
- 28Paudel S, Ghimire L, Jin L, Baral P, Cai S, Jeyaseelan S. NLRC4 suppresses IL-17A-mediated neutrophil-dependent host defense through upregulation of IL-18 and induction of necroptosis during Gram-positive pneumonia. Mucosal Immunol. 2019; 12(1): 247–57.
- 29Wang X, Yousefi S, Simon HU. Necroptosis and neutrophil-associated disorders. Cell Death Dis. 2018; 9(2): 111.
- 30Wong Fok Lung T, Monk IR, Acker KP, Mu A, Wang N, Riquelme SA, et al. Staphylococcus aureus small colony variants impair host immunity by activating host cell glycolysis and inducing necroptosis. Nat Microbiol. 2020; 5(1): 141–53.
- 31Akhtar J, Shukla D. Viral entry mechanisms: cellular and viral mediators of herpes simplex virus entry. FEBS J. 2010; 276(24): 7228–36.
- 32Guo H, Omoto S, Harris P, Finger J, Bertin J, Gough P, et al. Herpes simplex virus suppresses necroptosis in human cells. Cell Host Microbe. 2015; 2(17): 243–51.
- 33Mack C, Sickmann A, Lembo D, Brune W. Inhibition of proinflammatory and innate immune signaling pathways by a cytomegalovirus RIP1-interacting protein. Proc Natl Acad Sci U S A. 2008; 105(8): 3094–9.
- 34Upton JW, Kaiser WJ, Mocarski ES. Cytomegalovirus M45 cell death suppression requires Receptor-Interacting Protein (RIP) Homotypic Interaction Motif (RHIM)-dependent interaction with RIP1. J Biol Chem. 2008; 283(25): 16966–70.
- 35Wang X, Li Y, Liu S, Yu X, Li L, Shi C, et al. Direct activation of RIP3/MLKL-dependent necrosis by herpes simplex virus 1 (HSV-1) protein ICP6 triggers host antiviral defense. Proc Natl Acad U S A. 2014; 111(43): 15438–43.
- 36Nailwal H, Chan FK. Necroptosis in anti-viral inflammation. Cell Death Differ. 2019; 26(1): 4–13.
- 37Guo H, Gilley RP, Fisher A, Lane R, Landsteiner VJ, Ragan KB, et al. Species-independent contribution of ZBP1/DAI/DLM-1-triggered necroptosis in host defense against HSV1. Cell Death Dis. 2018; 8(9): 816.
- 38Moos S, Mohebiany AN, Waisman A, Kurschus FC. Imiquimod-induced psoriasis in mice depends on the IL-17 signaling of keratinocytes. J Invest Dermatol. 2019; 139(5): 1110–7.
- 39Gijbels MJJ, Zurcher C, Kraal G, Elliott GR, HogenEsch H, Schijff G, et al. Pathogenesis of skin lesions in mice with chronic proliferative dermatitis (cpdm/cpdm). Am J Pathol. 1996; 148(3): 941–50.
- 40Douglas T, Champagne C, Morizot A, Lapointe J-M, Saleh M, et al. The inflammatory caspases-1 and -11 mediate the pathogenesis of dermatitis in Sharpin-deficient mice. J Immunol. 2015; 195(5): 2365–73.
- 41Duan X, Liu X, Liu N, Huang Y, Jin Z, Zhang S, et al. Inhibition of keratinocyte necroptosis mediated by RIPK1/RIPK3/MLKL provides a protective effect against psoriatic inflammation. Cell Death Dis. 2020; 11(2): 134.
- 42Honda T, Yamamoto O, Sawada YU, Egawa G, Kitoh A, Otsuka A, et al. Receptor-interacting protein kinase 3 controls keratinocyte activation in a necroptosis-independent manner and promotes psoriatic dermatitis in mice. J Allergy Clin Immunol. 2017; 140(2): 619–22.e6.
- 43Jiang B-W, Zhang W-J, Wang Y, Tan L-P, Bao Y-L, Song Z-B, et al. Convallatoxin induces HaCaT cell necroptosis and ameliorates skin lesions in psoriasis-like mouse models. Biomed Pharmacother. 2019; 121: 109615.
- 44Zhang W-J, Song Z-B, Bao Y-L, Li W-L, Yang X-G, Wang QI, et al. Periplogenin induces necroptotic cell death through oxidative stress in HaCaT cells and ameliorates skin lesions in the TPA- and IMQ-induced psoriasis-like mouse models. Biochem Pharmacol. 2016; 105: 66–79.
- 45Saito N, Qiao H, Yanagi T, Shinkuma S, Nishimura K, Suto A, et al. An annexin A1-FPR1 interaction contributes to necroptosis of keratinocytes in severe cutaneous adverse drug reactions. Sci Transl Med. 2014; 6(245):245ra95.
- 46Jensen S, Seidelin JB, LaCasse EC, Nielsen OH. SMAC mimetics and RIPK inhibitors as therapeutics for chronic inflammatory diseases. Sci Signal. 2020; 13(619):eaax8295.
- 47Panayotova-Dimitrova D, Feoktistova M, Leverkus M. RIPping the skin apart: necroptosis signaling in toxic epidermal necrolysis. J Invest Dermatol. 2015; 135(8): 1940–3.
- 48Hasegawa A, Shinkuma S, Hayashi R, Hama N, Watanabe H, Kinoshita M, et al. RIP3 as a diagnostic and severity marker for Stevens-Johnson syndrome and toxic epidermal necrolysis. J Allergy Clin Immunol Pract. 2020; 8(5): 1768–71.e7.
- 49Kim SK, Kim W-J, Yoon J-H, Ji J-H, Morgan MJ, Cho H, et al. Upregulated RIP3 expression potentiates MLKL phosphorylation-mediated programmed necrosis in toxic epidermal necrolysis. J Invest Dermatol. 2015; 135(8): 2021–30.
- 50Perretti M, D’Acquisto F. Annexin A1 and glucocorticoids as effectors of the resolution of inflammation. Nat Rev Immunol. 2009; 9(1): 62–70.
- 51Choi S-W, Park H-H, Kim S, Chung JM, Noh H-J, Kim SK, et al. PELI1 selectively targets kinase-active RIP3 for ubiquitylation-dependent proteasomal degradation. Mol Cell. 2018; 70(5): 920–35.e7.
- 52Wang H, Meng H, Li X, Zhu K, Dong K, Mookhtiar AK, et al. PELI1 functions as a dual modulator of necroptosis and apoptosis by regulating ubiquitination of RIPK1 and mRNA levels of c-FLIP. Proc Natl Acad U S A. 2017; 114(45): 11944–9.
- 53Grootjans S, Berghe TV, Vandenabeele P. Initiation and execution mechanisms of necroptosis: an overview. Cell Death Differ. 2017; 24(7): 1184–95.
- 54Du Q, Xie J, Kim H-J, Ma X. Type I interferon: the mediator of bacterial infection-induced necroptosis. Cell Mol Immunol. 2013; 10(001): 4–6.
- 55Bengtsson A, Rönnblom L. Role of interferons in SLE. Best Pract Res Clin Rheumatol. 2017; 31(3): 415–28.
- 56Sarhan J, Liu BC, Muendlein HI, Weindel CG, Smirnova I, Tang AY, et al. Constitutive interferon signaling maintains critical threshold of MLKL expression to license necroptosis. Cell Death Differ. 2019; 26(2): 332–47.
- 57Fan H, Liu F, Dong G, Ren D, Xu Y, Dou J, et al. Activation-induced necroptosis contributes to B-cell lymphopenia in active systemic lupus erythematosus. Cell Death Dis. 2014; 5(9):e1416.
- 58Mohsen H, Paulina K, von Rauchhaupt E, Lech M. The involvement of microRNAs in modulation of innate and adaptive immunity in systemic lupus erythematosus and lupus nephritis. J Immunol Res. 2018; 2018: 1–15.
- 59Mangan MSJ, Olhava EJ, Roush WR, Seidel HM, Glick GD, Latz E, et al. Targeting the NLRP3 inflammasome in inflammatory diseases. Nat Rev Drug Discovery. 2018; 17(8): 588–606.
- 60Guo C, Fu R, Zhou M, Wang S, Huang Y, Hu H, et al. Pathogenesis of lupus nephritis: RIP3 dependent necroptosis and NLRP3 inflammasome activation. J Autoimmun. 2019; 103: 102286.
- 61Yang F, He YI, Zhai Z, Sun E. Programmed cell death pathways in the pathogenesis of systemic lupus erythematosus. Immunol Res. 2019; 2019: 3638562.
10.1155/2019/3638562 Google Scholar
- 62Bumiller-Bini V, Cipolla GA, Spadoni MB, Augusto DG, Petzl-Erler ML, Beltrame MH, et al. Condemned or not to die? Gene polymorphisms associated with cell death in pemphigus foliaceus. Front Immunol. 2019; 18(10): 2416.
- 63Lauffer F, Jargosch M, Krause L, Garzorz-Stark N, Franz R, Roenneberg S, et al. Type I immune response induces keratinocyte necroptosis and is associated with interface dermatitis. J Invest Dermatol. 2018; 54(1): 147–76.
- 64Kong Q, Lv J, Yan S, Chang K-J, Wang G. A novel naphthyridine derivative, 3u, induces necroptosis at low concentrations and apoptosis at high concentrations in human melanoma A375 cells. Int J Mol Sci. 2018; 19(10): 2975.
- 65Zhang Y, Zhan X, Xiong J, Peng S, Huang W, Joshi R, et al. Temperature-dependent cell death patterns induced by functionalized gold nanoparticle photothermal therapy in melanoma cells. Sci Rep. 2018; 8(1): 8720.
- 66Mohammadalipour Z, Rahmati M, Khataee A, Moosavi MA. Differential effects of N-TiO2 nanoparticle and its photo-activated form on autophagy and necroptosis in human melanoma A375 cells. J Cell Physiol. 2020; 235(11): 8246–59.
- 67Pawlikowska M, Piotrowski J, Jędrzejewski T, Kozak W, Slominski AT, Brożyna AA. Coriolus versicolor-derived protein-bound polysaccharides trigger the caspase-independent cell death pathway in amelanotic but not melanotic melanoma cells. Phytother Res. 2020; 34(1): 173–83.
- 68Basit F, van Oppen LMPE, Schöckel L, Bossenbroek HM, van Emst-de Vries SE, Hermeling JCW, et al. Mitochondrial complex I inhibition triggers a mitophagy-dependent ROS increase leading to necroptosis and ferroptosis in melanoma cells. Cell Death Dis. 2017; 8(3):e2716.
- 69Van Hoecke L, Raes L, Stremersch S, Brans T, Fraire JC, Roelandt R, et al. Delivery of mixed-lineage kinase domain-like protein by vapor nanobubble photoporation induces necroptotic-like cell death in tumor cells. Int J Mol Sci. 2019; 20(17): 4254.
- 70Geserick P, Wang J, Schilling R, Horn S, Harris PA, Bertin J, et al. Absence of RIPK3 predicts necroptosis resistance in malignant melanoma. Cell Death Dis. 2015; 6(9):e1884.
- 71Van Hoecke L, Van Lint S, Roose K, Van Parys A, Vandenabeele P, Grooten J, et al. Treatment with mRNA coding for the necroptosis mediator MLKL induces antitumor immunity directed against neo-epitopes. Nat Commun. 2018; 9(1): 3417.
- 72Strilic B, Yang L, Albarrán-Juárez J, Wachsmuth L, Han K, Müller UC, et al. Tumour-cell-induced endothelial cell necroptosis via death receptor 6 promotes metastasis. Nature. 2016; 536(7615): 215–8.
- 73Hänggi K, Vasilikos L, Valls AF, Yerbes R, Knop J, Spilgies LM, et al. RIPK1/RIPK3 promotes vascular permeability to allow tumor cell extravasation independent of its necroptotic function. Cell Death Dis. 2017; 8(2):e2588.
- 74Hou J, Ju J, Zhang Z, Zhao C, Li Z, Zheng J, et al. Discovery of potent necroptosis inhibitors targeting RIPK1 kinase activity for the treatment of inflammatory disorder and cancer metastasis. Cell Death Dis. 2019; 10(7): 493.
- 75Li YS, Xiong Y, Zhang G, Zhang L, Yang W, Yang J, et al. Identification of 5-(2,3-dihydro-1H-indol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine Derivatives as a New Class of Receptor-Interacting Protein Kinase 1 (RIPK1) Inhibitors, Which Showed Potent Activity in a Tumor Metastasis Model. J Med Chem. 2018; 61(24): 11398–414.
- 76Que SKT, Zwald FO, Schmults CD. Cutaneous squamous cell carcinoma: Incidence, risk factors, diagnosis, and staging. J Am Acad Dermatol. 2018; 78(2): 237–47.
- 77Baba N, Higashi Y, Kanekura T. Japanese black vinegar “Izumi” inhibits the proliferation of human squamous cell carcinoma cells via necroptosis. Nutr Cancer. 2013; 65(7): 1093–7.
- 78Kadigamuwa C, Choksi S, Xu Q, Cataisson C, Greenbaum SS, Yuspa SH, et al. Role of retinoic acid receptor-γ in DNA damage-induced necroptosis. iScience. 2019; 17: 74–86.