NLRP3 Inflammasome Involvement in Heart, Liver, and Lung Diseases—A Lesson from Cytokine Storm Syndrome
Abstract
:1. Receptors of Innate Immune Response
2. Mechanisms of Inflammasome Activation and Regulation
3. Pyroptosis: The Death Side of Inflammasomes
4. Inflammasome Involvement in Cardiovascular Diseases
5. Inflammasomes and Liver Disease
6. Inflammasome Involvement in Pulmonary Diseases
7. The Inflammasome in the Pathogenesis of Cytokine Storm Syndrome
8. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Deets, K.A.; Vance, R.E. Inflammasomes and adaptive immune responses. Nat. Immunol. 2021, 22, 412–422. [Google Scholar] [CrossRef]
- Lamkanfi, M.; Dixit, V.M. Inflammasomes: Guardians of cytosolic sanctity. Immunol. Rev. 2009, 227, 95–105. [Google Scholar] [CrossRef]
- Thompson, M.R.; Kaminski, J.J.; Kurt-Jones, E.A.; Fitzgerald, K.A. Pattern recognition receptors and the innate immune response to viral infection. Viruses 2011, 3, 920–940. [Google Scholar] [CrossRef]
- Rhodes, J.; Zipfel, C.; Jones, J.D.G.; Ngou, B.P.M. Concerted actions of PRR- and NLR-mediated immunity. Essays Biochem. 2022, 66, 501–511. [Google Scholar] [CrossRef]
- Li, D.; Wu, M. Pattern recognition receptors in health and diseases. Signal Transduct. Target. Ther. 2021, 6, 291. [Google Scholar] [CrossRef]
- Veeranki, S.; Choubey, D. Interferon-inducible p200-family protein IFI16, an innate immune sensor for cytosolic and nuclear double-stranded DNA: Regulation of subcellular localization. Mol. Immunol. 2012, 49, 567–571. [Google Scholar] [CrossRef]
- Kanneganti, T.D.; Lamkanfi, M.; Núñez, G. Intracellular NOD-like receptors in host defense and disease. Immunity 2007, 27, 549–559. [Google Scholar] [CrossRef]
- Awad, F.; Assrawi, E.; Louvrier, C.; Jumeau, C.; Georgin-Lavialle, S.; Grateau, G.; Amselem, S.; Giurgea, I.; Karabina, S.A. Inflammasome biology, molecular pathology and therapeutic implications. Pharmacol. Ther. 2018, 187, 133–149. [Google Scholar] [CrossRef]
- Janeway, C.A.; Medzhitov, R. Innate immune recognition. Annu. Rev. Immunol. 2002, 20, 197–216. [Google Scholar] [CrossRef]
- Arnoult, D.; Soares, F.; Tattoli, I.; Castanier, C.; Philpott, D.J.; Girardin, S.E. An N-terminal addressing sequence targets NLRX1 to the mitochondrial matrix. J. Cell Sci. 2009, 122 Pt 17, 3161–3168. [Google Scholar] [CrossRef]
- Moore, C.B.; Bergstralh, D.T.; Duncan, J.A.; Lei, Y.; Morrison, T.E.; Zimmermann, A.G.; Accavitti-Loper, M.A.; Madden, V.J.; Sun, L.; Ye, Z.; et al. NLRX1 is a regulator of mitochondrial antiviral immunity. Nature 2008, 451, 573–577. [Google Scholar] [CrossRef] [PubMed]
- Taniguchi, S.; Sagara, J. Regulatory molecules involved in inflammasome formation with special reference to a key mediator protein, ASC. Semin. Immunopathol. 2007, 29, 231–238. [Google Scholar] [CrossRef] [PubMed]
- Kumar, M.; Roe, K.; Orillo, B.; Muruve, D.A.; Nerurkar, V.R.; Gale, M., Jr.; Verma, S. Inflammasome Adaptor Protein Apoptosis-Associated Speck-Like Protein Containing CARD (ASC) Is Critical for the Immune Response and Survival in West Nile Virus Encephalitis. J. Virol. 2013, 87, 3655–3667. [Google Scholar] [CrossRef] [PubMed]
- Martinon, F.; Burns, K.; Tschopp, J. The inflammasome: A molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol. Cell 2002, 10, 417–426. [Google Scholar] [CrossRef]
- Fernandes-Alnemri, T.; Yu, J.W.; Datta, P.; Wu, J.; Alnemri, E.S. AIM2 activates the inflammasome and cell death in response to cytoplasmic DNA. Nature 2009, 458, 509–513. [Google Scholar] [CrossRef]
- Kerur, N.; Veettil, M.V.; Sharma-Walia, N.; Bottero, V.; Sadagopan, S.; Otageri, P.; Chandran, B. IFI16 acts as a nuclear pathogen sensor to induce the inflammasome in response to Kaposi Sarcoma-associated herpesvirus infection. Cell Host Microbe 2011, 9, 363–375. [Google Scholar] [CrossRef]
- Pothlichet, J.; Meunier, I.; Davis, B.K.; Ting, J.P.-Y.; Skamene, E.; von Messling, V.; Vidal, S.M. Type I IFN triggers RIG-I/TLR3/NLRP3-dependent inflammasome activation in influenza A virus infected cells. PLoS Pathog. 2013, 9, e1003256. [Google Scholar] [CrossRef]
- Triantafilou, K. Enigmatic inflammasomes. Immunology 2021, 162, 249–251. [Google Scholar] [CrossRef]
- Kelley, N.; Jeltema, D.; Duan, Y.; He, Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int. J. Mol. Sci. 2019, 20, 3328. [Google Scholar] [CrossRef]
- Sharma, B.R.; Kanneganti, T.-D. NLRP3 inflammasome in cancer and metabolic diseases. Nat. Immunol. 2021, 22, 550–559. [Google Scholar] [CrossRef]
- Paerewijck, O.; Lamkanfi, M. The human inflammasomes. Mol. Asp. Med. 2022, 88, 101100. [Google Scholar] [CrossRef] [PubMed]
- Miao, E.A.; Leaf, I.A.; Treuting, P.M.; Mao, D.P.; Dors, M.; Sarkar, A.; Warren, S.E.; Wewers, M.D.; Aderem, A. Caspase-1-induced pyroptosis is an innate immune effector mechanism against intracellular bacteria. Nat. Immunol. 2010, 11, 1136–1142. [Google Scholar] [CrossRef] [PubMed]
- Kesavardhana, S.; Malireddi, R.K.S.; Kanneganti, T.D. Caspases in Cell Death, Inflammation, and Pyroptosis. Annu. Rev. Immunol. 2020, 38, 567–595. [Google Scholar] [CrossRef]
- Shi, J.; Gao, W.; Shao, F. Pyroptosis: Gasdermin-Mediated Programmed Necrotic Cell Death. Trends Biochem. Sci. 2017, 42, 245–254. [Google Scholar] [CrossRef] [PubMed]
- Ding, J.; Wang, K.; Liu, W.; She, Y.; Sun, Q.; Shi, J.; Sun, H.; Wang, D.C.; Shao, F. Pore-forming activity and structural autoinhibition of the gasdermin family. Nature 2016, 535, 111–116. [Google Scholar] [CrossRef] [PubMed]
- Lamkanfi, M.; Dixit, V.M. Manipulation of host cell death pathways during microbial infections. Cell Host Microbe 2010, 8, 44–54. [Google Scholar] [CrossRef]
- Tsuchiya, K. Switching from Apoptosis to Pyroptosis: Gasdermin-Elicited Inflammation and Antitumor Immunity. Int. J. Mol. Sci. 2021, 22, 426. [Google Scholar] [CrossRef]
- Swanson, K.V.; Deng, M.; Ting, J.P. The NLRP3 inflammasome: Molecular activation and regulation to therapeutics. Nat. Rev. Immunol. 2019, 19, 477–489. [Google Scholar] [CrossRef]
- Carrillo-Salinas, F.J.; Ngwenyama, N.; Anastasiou, M.; Kaur, K.; Alcaide, P. Heart Inflammation: Immune Cell Roles and Roads to the Heart. Am. J. Pathol. 2019, 189, 1482–1494. [Google Scholar] [CrossRef]
- Toldo, S.; Abbate, A. The NLRP3 inflammasome in acute myocardial infarction. Nat. Rev. Cardiol. 2018, 15, 203–214. [Google Scholar] [CrossRef]
- Frangogiannis, N.G. Cardiac fibrosis: Cell biological mechanisms, molecular pathways and therapeutic opportunities. Mol. Asp. Med. 2019, 65, 70–99. [Google Scholar] [CrossRef] [PubMed]
- Marino, M.; Scuderi, F.; Mannella, F.; Bartoccioni, E. TGF-beta 1 and IL-10 modulate IL-1 beta-induced membrane and soluble ICAM-1 in human myoblasts. J. Neuroimmunol. 2003, 134, 151–157. [Google Scholar] [CrossRef] [PubMed]
- Morimoto, H.; Takahashi, M.; Izawa, A.; Ise, H.; Hongo, M.; Kolattukudy, P.E.; Ikeda, U. Cardiac overexpression of monocyte chemoattractant protein-1 in transgenic mice prevents cardiac dysfunction and remodeling after myocardial infarction. Circ. Res. 2006, 99, 891–899. [Google Scholar] [CrossRef] [PubMed]
- Bartoccioni, E.; Scuderi, F.; Marino, M.; Provenzano, C. IL-6, monocyte infiltration and parenchymal cells. Trends Immunol. 2003, 24, 299–300. [Google Scholar] [CrossRef] [PubMed]
- Zhaolin, Z.; Guohua, L.; Shiyuan, W.; Zuo, W. Role of pyroptosis in cardiovascular disease. Cell Prolif. 2019, 52, e12563. [Google Scholar] [CrossRef] [PubMed]
- Abbate, A. The heart on fire: Inflammasome and cardiomyopathy. Exp. Physiol. 2013, 98, 385. [Google Scholar] [CrossRef]
- Van Tassell, B.W.; Toldo, S.; Mezzaroma, E.; Abbate, A. Targeting interleukin-1 in heart disease. Circulation 2013, 128, 1910–1923. [Google Scholar] [CrossRef]
- Toldo, S.; Mauro, A.G.; Cutter, Z.; Abbate, A. Inflammasome, pyroptosis, and cytokines in myocardial ischemia-reperfusion injury. Am. J. Physiol. Heart Circ. Physiol. 2018, 315, H1553–H1568. [Google Scholar] [CrossRef]
- Bracey, N.A.; Beck, P.L.; Muruve, D.A.; Hirota, S.A.; Guo, J.; Jabagi, H.; Wright, J.R., Jr.; Macdonald, J.A.; Lees-Miller, J.P.; Roach, D.; et al. The Nlrp3 inflammasome promotes myocardial dysfunction in structural cardiomyopathy through interleukin-1β. Exp. Physiol. 2013, 98, 462–472. [Google Scholar] [CrossRef]
- Luan, Y.; Guo, Y.; Li, S.; Yu, B.; Zhu, S.; Li, S.; Li, N.; Tian, Z.; Peng, C.; Cheng, J.; et al. Interleukin-18 among atrial fibrillation patients in the absence of structural heart disease. Europace 2010, 12, 1713–1718. [Google Scholar] [CrossRef]
- Cheng, T.; Wang, X.F.; Hou, Y.T.; Zhang, L. Correlation between atrial fibrillation, serum amyloid protein A and other inflammatory cytokines. Mol. Med. Rep. 2012, 6, 581–584. [Google Scholar] [CrossRef] [PubMed]
- Yao, C.; Veleva, T.; Scott, L., Jr.; Cao, S.; Li, L.; Chen, G.; Jeyabal, P.; Pan, X.; Alsina, K.M.; Abu-Taha, I.; et al. Enhanced Cardiomyocyte NLRP3 Inflammasome Signaling Promotes Atrial Fibrillation. Circulation 2018, 138, 2227–2242. [Google Scholar] [CrossRef] [PubMed]
- Wei, J.; Wang, H.; Wang, H.; Wang, B.; Meng, L.; Xin, Y.; Jiang, X. The role of NLRP3 inflammasome activation in radiation damage. Biomed. Pharmacother. 2019, 118, 109217. [Google Scholar] [CrossRef] [PubMed]
- Stewart, F.A.; Seemann, I.; Hoving, S.; Russell, N.S. Understanding radiation-induced cardiovascular damage and strategies for intervention. Clin. Oncol. 2013, 25, 617–624. [Google Scholar] [CrossRef] [PubMed]
- Aleman, B.M.P.; van den Belt-Dusebout, A.W.; Bruin, M.L.d.; van’t Veer, M.B.; Baaijens, M.H.A.; de Boer, J.P.; Hart, A.A.M.; Klokman, W.J.; Kuenen, M.A.; Ouwens, G.M.; et al. Late cardiotoxicity after treatment for Hodgkin lymphoma. Blood 2007, 109, 1878–1886. [Google Scholar] [CrossRef]
- Brosius, F.C., III; Waller, B.F.; Roberts, W.C. Radiation heart disease. analysis of 16 young (aged 15 to 33 years) necropsy patients who received over 3500 rads to the heart. Am. J. Med. 1981, 70, 519–530. [Google Scholar] [CrossRef]
- Virmani, R.; Farb, A.; Carter, A.J.; Jones, R.M. Pathology of radiation- induced coronary artery disease in human and pig. Cardiovasc. Radiat. Med. 1999, 1, 98–101. [Google Scholar] [CrossRef]
- Wang, H.; Wei, J.; Zheng, Q.; Meng, L.; Xin, Y.; Yin, X.; Jiang, X. Radiation-induced heart disease: A review of classification, mechanism and prevention. Int. J. Biol. Sci. 2019, 15, 2128–2138. [Google Scholar] [CrossRef]
- Todd, D.G.; Mikkelsen, R.B. Ionizing radiation induces a transient increase in cytosolic free [Ca2+] in human epithelial tumor cells. Cancer Res. 1994, 54, 5224–5230. [Google Scholar]
- Yaron, J.R.; Gangaraju, S.; Rao, M.Y.; Kong, X.; Zhang, L.; Su, F.; Tian, Y.; Glenn, H.L.; Meldrum, D.R. K+ regulates Ca2+ to drive inflammasome signaling: Dynamic visualization of ion flux in live cells. Cell Death Dis. 2015, 6, e1954. [Google Scholar] [CrossRef]
- Sharma, D.; Czarnota, G.J. Role of acid sphingomyelinase-induced ceramide generation in response to radiation. Oncotarget 2019, 10, 6–7. [Google Scholar] [CrossRef] [PubMed]
- Cheng, H.; Chen, L.; Huang, M.; Hou, J.; Chen, Z.; Yang, X. Hunting down NLRP3 inflammasome: An executioner of radiation-induced injury. Front. Immunol. 2022, 13, 967989. [Google Scholar] [CrossRef]
- Schaftenaar, F.; Frodermann, V.; Kuiper, J.; Lutgens, E. Atherosclerosis: The interplay between lipids and immune cells. Curr Opin Lipidol. 2016, 27, 209–215. [Google Scholar] [CrossRef] [PubMed]
- Zheng, Y.; Xu, L.; Dong, N.; Li, F. NLRP3 inflammasome: The rising star in cardiovascular diseases. Front. Cardiovasc. Med. 2022, 9, 927061. [Google Scholar] [CrossRef]
- Paramel Varghese, G.; Folkersen, L.; Strawbridge, R.J.; Halvorsen, B.; Yndestad, A.; Ranheim, T.; Krohg-Sørensen, K.; Skjelland, M.; Espevik, T.; Aukrust, P.; et al. NLRP3 Inflammasome Expression and Activation in Human Atherosclerosis. J. Am. Hear. Assoc. 2016, 5, e003031. [Google Scholar] [CrossRef] [PubMed]
- Olofsson, P.S.; Sheikine, Y.; Jatta, K.; Ghaderi, M.; Samnegård, A.; Eriksson, P.; Sirsjö, A. A functional interleukin-1 receptor antagonist polymorphism influences atherosclerosis development. The interleukin-1beta: Interleukin-1 receptor antagonist balance in atherosclerosis. Circ. J. 2009, 73, 1531–1536. [Google Scholar] [CrossRef]
- Mallat, Z.; Corbaz, A.; Scoazec, A.; Graber, P.; Alouani, S.; Esposito, B.; Humbert, Y.; Chvatchko, Y.; Tedgui, A. Interleukin-18/interleukin-18 binding protein signaling modulates atherosclerotic lesion development and stability. Circ. Res. 2001, 89, E41–E45. [Google Scholar] [CrossRef]
- Elhage, R.; Jawien, J.; Rudling, M.; Ljunggren, H.G.; Takeda, K.; Akira, S.; Bayard, F.; Hansson, G.K. Reduced atherosclerosis in interleukin-18 deficient apolipoprotein E-knockout mice. Cardiovasc. Res. 2003, 59, 234–240. [Google Scholar] [CrossRef]
- Liberale, L.; Badimon, L.; Montecucco, F.; Lüscher, T.F.; Libby, P.; Camici, G.G. Inflammation, Aging, and Cardiovascular Disease: JACC Review Topic of the Week. J. Am. Coll. Cardiol. 2022, 79, 837–847. [Google Scholar] [CrossRef]
- Puzianowska-Kuźnicka, M.; Owczarz, M.; Wieczorowska-Tobis, K.; Nadrowski, P.; Chudek, J.; Slusarczyk, P.; Skalska, A.; Jonas, M.; Franek, E.; Mossakowska, M. Interleukin-6 and C-reactive protein, successful aging, and mortality: The PolSenior study. Immun. Ageing 2016, 13, 21. [Google Scholar] [CrossRef]
- Sanada, F.; Taniyama, Y.; Muratsu, J.; Otsu, R.; Shimizu, H.; Rakugi, H.; Morishita, R. Source of Chronic Inflammation in Aging. Front. Cardiovasc. Med. 2018, 5, 12. [Google Scholar] [CrossRef] [PubMed]
- Dhalla, N.S.; Shah, A.K.; Tappia, P.S. Role of Oxidative Stress in Metabolic and Subcellular Abnormalities in Diabetic Cardiomyopathy. Int. J. Mol. Sci. 2020, 21, 2413. [Google Scholar] [CrossRef] [PubMed]
- Marín-Aguilar, F.; Lechuga-Vieco, A.V.; Alcocer-Gómez, E.; Castejón-Vega, B.; Lucas, J.; Garrido, C.; Peralta-Garcia, A.; Pérez-Pulido, A.J.; Varela-López, A.; Quiles, J.L.; et al. NLRP3 inflammasome suppression improves longevity and prevents cardiac aging in male mice. Aging Cell 2020, 19, e13050. [Google Scholar] [CrossRef] [PubMed]
- Chen, G.; Chelu, M.G.; Dobrev, D.; Li, N. Cardiomyocyte Inflammasome Signaling in Cardiomyopathies and Atrial Fibrillation: Mechanisms and Potential Therapeutic Implications. Front. Physiol. 2018, 9, 1115. [Google Scholar] [CrossRef]
- Abbate, A.; Toldo, S.; Marchetti, C.; Kron, J.; Van Tassell, B.W.; Dinarello, C.A. Interleukin-1 and the Inflammasome as Therapeutic Targets in Cardiovascular Disease. Circ. Res. 2020, 126, 1260–1280. [Google Scholar] [CrossRef]
- Luan, J.; Ju, D. Inflammasome: A Double-Edged Sword in Liver Diseases. Front. Immunol. 2018, 9, 2201. [Google Scholar] [CrossRef]
- Bauer, R.; Rauch, I. The NAIP/NLRC4 inflammasome in infection and pathology. Mol. Asp. Med. 2020, 76, 100863. [Google Scholar] [CrossRef]
- Chattergoon, M.A.; Latanich, R.; Quinn, J.; Winter, M.E.; Buckheit, R.W., 3rd; Blankson, J.N.; Pardoll, D.; Cox, A.L. HIV and HCV activate the inflammasome in monocytes and macrophages via endosomal Toll-like receptors without induction of type 1 interferon. PLoS Pathog. 2014, 10, e1004082. [Google Scholar] [CrossRef]
- Molyvdas, A.; Georgopoulou, U.; Lazaridis, N.; Hytiroglou, P.; Dimitriadis, A.; Foka, P.; Vassiliadis, T.; Loli, G.; Phillipidis, A.; Zebekakis, P.; et al. The role of the NLRP3 inflammasome and the activation of IL-1β in the pathogenesis of chronic viral hepatic inflammation. Cytokine 2018, 110, 389–396. [Google Scholar] [CrossRef]
- Menzel, C.L.; Sun, Q.; Loughran, P.A.; Pape, H.C.; Billiar, T.R.; Scott, M.J. Caspase-1 is hepatoprotective during trauma and hemorrhagic shock by reducing liver injury and inflammation. Mol. Med. 2011, 17, 1031–1038. [Google Scholar] [CrossRef]
- Sun, Q.; Gao, W.; Loughran, P.; Shapiro, R.; Fan, J.; Billiar, T.R.; Scott, M.J. Caspase 1 activation is protective against hepatocyte cell death by up-regulating beclin 1 protein and mitochondrial autophagy in the setting of redox stress. J. Biol. Chem. 2013, 288, 15947–15958. [Google Scholar] [CrossRef] [PubMed]
- Sun, Q.; Loughran, P.; Shapiro, R.; Shrivastava, I.H.; Antoine, D.J.; Li, T.; Yan, Z.; Fan, J.; Billiar, T.R.; Scott, M.J. Redox-dependent regulation of hepatocyte absent in melanoma 2 inflammasome activation in sterile liver injury in mice. Hepatology 2017, 65, 253–268. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.; He, X.; Yuan, X.; Hong, J.; Bhat, O.; Li, G.; Li, P.L.; Guo, J. NLRP3 Inflammasome Formation and Activation in Nonalcoholic Steatohepatitis: Therapeutic Target for Antimetabolic Syndrome Remedy FTZ. Oxidative Med. Cell. Longev. 2018, 2018, 2901871. [Google Scholar] [CrossRef] [PubMed]
- Mridha, A.R.; Wree, A.; Robertson, A.A.B.; Yeh, M.M.; Johnson, C.D.; Van Rooyen, D.M.; Haczeyni, F.; Teoh, N.C.; Savard, C.; Ioannou, G.N.; et al. NLRP3 inflammasome blockade reduces liver inflammation and fibrosis in experimental NASH in mice. J. Hepatol. 2017, 66, 1037–1046. [Google Scholar] [CrossRef]
- Colak, Y.; Hasan, B.; Erkalma, B.; Tandon, K.; Zervos, X.; Menzo, E.L.; Erim, T. Pathogenetic mechanisms of nonalcoholic fatty liver disease and inhibition of the inflammasome as a new therapeutic target. Clin. Res. Hepatol. Gastroenterol. 2021, 45, 101710. [Google Scholar] [CrossRef]
- Menini, S.; Iacobini, C.; Vitale, M.; Pugliese, G. The Inflammasome in Chronic Complications of Diabetes and Related Metabolic Disorders. Cells 2020, 9, 1812. [Google Scholar] [CrossRef]
- Chen, Y.; Zhang, Y.; Li, N.; Jiang, Z.; Li, X. Role of mitochondrial stress and the NLRP3 inflammasome in lung diseases. Inflamm. Res. 2023, 72, 829–846. [Google Scholar] [CrossRef]
- Liu, H.; Gu, C.; Liu, M.; Liu, G.; Wan, Y. NEK7 mediated assembly and activation of NLRP3 inflammasome downstream of potassium efflux in ventilator-induced lung injury. Biochem. Pharmacol. 2020, 177, 113998. [Google Scholar] [CrossRef]
- Fu, J.; Wu, H. Structural Mechanisms of NLRP3 Inflammasome Assembly and Activation. Annu. Rev. Immunol. 2023, 41, 301–316. [Google Scholar] [CrossRef]
- D’Amico, R.; Fusco, R.; Cordaro, M.; Siracusa, R.; Peritore, A.F.; Gugliandolo, E.; Crupi, R.; Scuto, M.; Cuzzocrea, S.; Di Paola, R.; et al. Modulation of NLRP3 Inflammasome through Formyl Peptide Receptor 1 (Fpr-1) Pathway as a New Therapeutic Target in Bronchiolitis Obliterans Syndrome. Int. J. Mol. Sci. 2020, 21, 2144. [Google Scholar] [CrossRef]
- Liu, Y.; Chen, K.; Wang, C.; Gong, W.; Yoshimura, T.; Liu, M.; Wang, J.M. Cell surface receptor FPR2 promotes antitumor host defense by limiting M2 polarization of macrophages. Cancer Res. 2013, 73, 550–560. [Google Scholar] [CrossRef] [PubMed]
- Kasper, M.; Barth, K. Potential contribution of alveolar epithelial type I cells to pulmonary fibrosis. Biosci. Rep. 2017, 37, BSR20171301. [Google Scholar] [CrossRef] [PubMed]
- Kanneganti, T.D.; Lamkanfi, M.; Kim, Y.G.; Chen, G.; Park, J.H.; Franchi, L.; Vandenabeele, P.; Núñez, G. Pannexin-1-mediated recognition of bacterial molecules activates the cryopyrin inflammasome independent of Toll-like receptor signaling. Immunity 2007, 26, 433–443. [Google Scholar] [CrossRef] [PubMed]
- Galam, L.; Rajan, A.; Failla, A.; Soundararajan, R.; Lockey, R.F.; Kolliputi, N. Deletion of P2X7 attenuates hyperoxia-induced acute lung injury via inflammasome suppression. Am. J. Physiol. Lung Cell. Mol. Physiol. 2016, 310, L572–L581. [Google Scholar] [CrossRef] [PubMed]
- Ebeling, G.; Blasche, R.; Hofmann, F.; Augstein, A.; Kasper, M.; Barth, K. Effect of P2X7 receptor knockout on AQP-5 expression of type I alveolar epithelial cells. PLoS ONE 2014, 9, e100282. [Google Scholar] [CrossRef] [PubMed]
- Arulkumaran, N.; Unwin, R.J.; Tam, F.W. A potential therapeutic role for P2X7 receptor (P2X7R) antagonists in the treatment of inflammatory diseases. Expert Opin. Investig. Drugs 2011, 20, 897–915. [Google Scholar] [CrossRef] [PubMed]
- Shao, H.; Huang, L.; Duan, S.; Gao, M.; Zhu, J.; Chen, X.; Zhu, Y.; Wang, W.; Yang, Z.; Wang, X.; et al. Glyburide attenuates ozone-induced pulmonary inflammation and injury by blocking the NLRP3 inflammasome. Environ. Toxicol. 2020, 35, 831–839. [Google Scholar] [CrossRef]
- Allen, I.C.; Scull, M.A.; Moore, C.B.; Holl, E.K.; McElvania-TeKippe, E.; Taxman, D.J.; Guthrie, E.H.; Pickles, R.J.; Ting, J.P.-Y. The NLRP3 inflammasome mediates in vivo innate immunity to influenza A virus through recognition of viral RNA. Immunity 2009, 30, 556–565. [Google Scholar] [CrossRef]
- Cohen, T.S.; Prince, A.S. Activation of inflammasome signaling mediates pathology of acute P. aeruginosa pneumonia. J. Clin. Investig. 2013, 123, 1630–1637. [Google Scholar] [CrossRef]
- Miller, L.S.; Pietras, E.M.; Uricchio, L.H.; Hirano, K.; Rao, S.; Lin, H.; O’connell, R.M.; Iwakura, Y.; Cheung, A.L.; Cheng, G.; et al. Inflammasome-mediated production of IL-1beta is required for neutrophil recruitment against Staphylococcus aureus in vivo. J. Immunol. 2007, 179, 6933–6942. [Google Scholar] [CrossRef]
- Cao, F.; Tian, X.; Li, Z.; Lv, Y.; Han, J.; Zhuang, R.; Cheng, B.; Gong, Y.; Ying, B.; Jin, S.; et al. Suppression of NLRP3 Inflammasome by Erythropoietin via the EPOR/JAK2/STAT3 Pathway Contributes to Attenuation of Acute Lung InjuryinMice. Front. Pharmacol. 2020, 11, 306. [Google Scholar] [CrossRef] [PubMed]
- Cron, R.Q.; Goyal, G.; Chatham, W.W. Cytokine Storm Syndrome. Annu. Rev. Med. 2023, 74, 321–337. [Google Scholar] [CrossRef] [PubMed]
- Henderson, L.A.; Canna, S.W.; Schulert, G.S.; Volpi, S.; Lee, P.Y.; Kernan, K.F.; Caricchio, R.; Mahmud, S.; Hazen, M.M.; Halyabar, O.; et al. On the Alert for Cytokine Storm: Immunopathology in COVID-19. Arthritis Rheumatol. 2020, 72, 1059–1063. [Google Scholar] [CrossRef] [PubMed]
- Pontali, E.; Volpi, S.; Signori, A.; Antonucci, G.; Castellaneta, M.; Buzzi, D.; Montale, A.; Bustaffa, M.; Angelelli, A.; Caorsi, R.; et al. Efficacy of early anti-inflammatory treatment with high doses of intravenous anakinra with or without glucocorticoids in patients with severe COVID-19 pneumonia. J. Allergy Clin. Immunol. 2021, 147, 1217–1225. [Google Scholar] [CrossRef]
- Kishimoto, T. IL-6: From arthritis to CAR-T-cell therapy and COVID-19. Int. Immunol. 2021, 33, 515–519. [Google Scholar] [CrossRef]
- Giavridis, T.; van der Stegen, S.J.C.; Eyquem, J.; Hamieh, M.; Piersigilli, A.; Michel Sadelain, M. CAR T cell-induced cytokine release syndrome is mediated by macrophages and abated by IL-1 blockade. Nat. Med. 2018, 24, 731–738. [Google Scholar] [CrossRef]
- Shakoory, B.; Carcillo, A.J.; Chatham, W.W.; Amdur, R.L.; Zhao, H.; Dinarello, C.A.; Cron, R.Q.; Opal, S.M. Interleukin-1 Receptor Blockade Is Associated With Reduced Mortality in Sepsis Patients With Features of Macrophage Activation Syndrome: Reanalysis of a Prior Phase III Trial. Crit. Care Med. 2016, 44, 275–281. [Google Scholar] [CrossRef]
- Tanaka, T.; Narazaki, M.; Kishimoto, T. Immunotherapeutic implications of IL-6 blockade for cytokine storm. Immunotherapy 2016, 8, 959–970. [Google Scholar] [CrossRef]
- Nouveau, L.; Buatois, V.; Cons, L.; Chatel, L.; Pontini, G.; Pleche, N.; Ferlin, W.G. Immunological analysis of the murine anti-CD3-induced cytokine release syndrome model and therapeutic efficacy of anti-cytokine antibodies. Eur. J. Immunol. 2021, 51, 2074–2085. [Google Scholar] [CrossRef]
- Grom, A.A.; Horne, A.C.; De Benedetti, F. Macrophage activation syndrome in the era of biologic therapy. Nat. Rev. Rheumatol. 2016, 12, 259–268. [Google Scholar] [CrossRef]
- Galli, E.; Sorà, F.; Hohaus, S.; Fresa, A.; Pansini, I.; Autore, F.; Metafuni, E.; Innocenti, I.; Limongiello, M.A.; Giammarco, S.; et al. Endothelial activation predicts disseminated intravascular coagulopathy, cytokine release syndrome and prognosis in patients treated with anti-CD19 CAR-T cells. Br. J. Haematol. 2022, 201, 86–94. [Google Scholar] [CrossRef]
- Fajgenbaum, D.C.; Carl HJune, C.H. Cytokine Storm. N. Engl. J. Med. 2020, 383, 2255–2273. [Google Scholar] [CrossRef]
- Lee, D.W.; Gardner, R.; Porter, D.L.; Louis, C.U.; Ahmed, N.; Jensen, M.; Grupp, S.A.; Mackall, C.L. Current concepts in the diagnosis and management of cytokine release syndrome. Blood 2014, 124, 188–195. [Google Scholar] [CrossRef]
- Caricchio, R.; Gallucci, M.; Chandra, D.; Zhang, X.; Gallucci, S.; Fleece, D.; Bromberg, M.; Criner, G.J.; Temple University COVID-19 Research Group. Preliminary predictive criteria for COVID-19 cytokine storm. Ann. Rheum. Dis. 2021, 80, 88–95. [Google Scholar] [CrossRef]
- Mehta, P.; McAuley, D.F.; Brown, M.; Sanchez, E.; Tattersall, R.S.; Manson, J.J.; HLH Across Speciality Collaboration, UK. COVID-19: Consider cytokine storm syndromes and immunosuppression. Lancet 2020, 395, 1033–1034. [Google Scholar] [CrossRef]
- Liu, B.; Li, M.; Zhou, Z.; Guan, X.; Xiang, Y. Can we use interleukin-6 (IL-6) blockade for coronavirus disease 2019 (COVID-19)-induced cytokine release syndrome (CRS)? J. Autoimmun. 2020, 111, 102452. [Google Scholar] [CrossRef]
- Singh, N.; Hofmann, T.J.; Gershenson, Z.; Levine, B.L.; Grupp, S.A.; Teachey, D.T.; Barrett, D.M. Monocyte lineage-derived IL-6 does not affect chimeric antigen receptor T-cell function. Cytotherapy 2017, 19, 867–880. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Fang, Y.; Chen, X.; Wang, Z.; Liang, X.; Zhang, T.; Liu, M.; Zhou, N.; Lv, J.; Tang, K.; et al. Gasdermin E-mediated target cell pyroptosis by CAR T cells triggers cytokine release syndrome. Sci. Immunol. 2020, 5, eaax7969. [Google Scholar] [CrossRef] [PubMed]
- Topp, M.S.; Gökbuget, N.; Stein, A.S.; Zugmaier, G.; O’Brien, S.; Bargou, R.C.; Dombret, H.; Fielding, A.K.; Heffner, L.; Larson, R.A.; et al. Safety and activity of blinatumomab for adult patients with relapsed or refractory B-precursor acute lymphoblastic leukaemia: A multicentre, single-arm, phase 2 study. Lancet Oncol. 2015, 16, 57–66. [Google Scholar] [CrossRef] [PubMed]
- Suntharalingam, G.; Perry, M.R.; Ward, S.; Brett, S.J.; Castello-Cortes, A.; Brunner, M.D.; Panoskaltsis, N. Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412. N. Engl. J. Med. 2006, 355, 1018–1028. [Google Scholar] [CrossRef]
- Crayne, C.B.; Albeituni, S.; Nichols, K.E.; Cron, R.Q. The Immunology of Macrophage Activation Syndrome. Front. Immunol. 2019, 10, 119. [Google Scholar] [CrossRef] [PubMed]
- Zhang, M.; Behrens, E.M.; Prescott Atkinson, T.; Shakoory, B.; Grom, A.A.; Cron, R.Q. Genetic defects in cytolysis in macrophage activation syndrome. Curr. Rheumatol. Rep. 2014, 16, 439. [Google Scholar] [CrossRef] [PubMed]
- Ravelli, A.; Minoia, F.; Davì, S.; Horne, A.; Bovis, F.; Pistorio, A.; Aricò, M.; Avcin, T.; Behrens, E.M.; De Benedetti, F.; et al. 2016 Classification Criteria for Macrophage Activation Syndrome Complicating Systemic Juvenile Idiopathic Arthritis: A European League Against Rheumatism/American College of Rheumatology/Paediatric Rheumatology International Trials Organisation Collaborative Initiative. Ann. Rheum. Dis. 2016, 75, 481–489. [Google Scholar] [CrossRef] [PubMed]
- La Rosée, P.; Horne, A.; Hines, M.; von Bahr Greenwood, T.; Machowicz, R.; Berliner, N.; Birndt, S.; Gil-Herrera, J.; Girschikofsky, M.; Jordan, M.B.; et al. Recommendations for the management of hemophagocytic lymphohistiocytosis in adults. Blood 2019, 133, 2465–2477. [Google Scholar] [CrossRef]
- Gars, E.; Purington, N.; Scott, G.; Chisholm, K.; Gratzinger, D.; Martin, B.A.; Ohgami, R.S. Bone marrow histomorphological criteria can accurately diagnose hemophagocytic lymphohistiocytosis. Haematologica 2018, 103, 1635–1641. [Google Scholar] [CrossRef]
- Schulert, G.; Canna, S.W. Convergent pathways of the hyperferritinemic syndromes. Int. Immunol. 2018, 30, 195–203. [Google Scholar] [CrossRef]
- Jenkins, M.R.; Rudd-Schmidt, J.A.; Lopez, J.A.; Ramsbottom, K.M.; Mannering, S.I.; Andrews, D.M.; Voskoboinik, I.; Trapani, J.A. Failed CTL/NK cell killing and cytokine hypersecretion are directly linked through prolonged synapse time. J. Exp. Med. 2015, 212, 307–317. [Google Scholar] [CrossRef]
- Masters, S.L.; Simon, A.; Aksentijevich, I.; Kastner, D.L. Horror autoinflammaticus: The molecular pathophysiology of autoinflammatory disease (*). Annu. Rev. Immunol. 2009, 27, 621–668. [Google Scholar] [CrossRef]
- Schulert, G.S.; Cron, R.Q. The genetics of macrophage activation syndrome. Genes Immun. 2020, 21, 169–181. [Google Scholar] [CrossRef]
- Canna, S.W.; de Jesus, A.A.; Gouni, S.; Brooks, S.R.; Marrero, B.; Liu, Y.; DiMattia, M.A.; Zaal, K.J.; Sanchez, G.A.; Kim, H.; et al. An activating NLRC4 inflammasome mutation causes autoinflammation with recurrent macrophage activation syndrome. Nat. Genet. 2014, 46, 1140–1146. [Google Scholar] [CrossRef]
- Kernan, K.F.; Ghaloul-Gonzalez, L.; Shakoory, B.; Kellum, J.A.; Angus, D.C.; Carcillo, J.A. Adults with septic shock and extreme hyperferritinemia exhibit pathogenic immune variation. Genes Immun. 2019, 20, 520–526. [Google Scholar] [CrossRef] [PubMed]
- Teijaro, J.R.; Walsh, K.B.; Rice, S.; Rosen, H.; Oldstone, M.B. Mapping the innate signaling cascade essential for cytokine storm during influenza virus infection. Proc. Natl. Acad. Sci. USA 2014, 111, 3799–3804. [Google Scholar] [CrossRef] [PubMed]
- Srikiatkhachorn, A.; Mathew, A.; Rothman, A.L. Immune-mediated cytokine storm and its role in severe dengue. Semin. Immunopathol. 2017, 39, 563–574. [Google Scholar] [CrossRef]
- Wauquier, N.; Becquart, P.; Padilla, C.; Baize, S.; Leroy, E.M. Human fatal zaire ebola virus infection is associated with an aberrant innate immunity and with massive lymphocyte apoptosis. PLoS Neglected Trop. Dis. 2010, 4, e837. [Google Scholar] [CrossRef] [PubMed]
- Kuriakose, T.; Man, S.M.; Malireddi, R.K.; Karki, R.; Kesavardhana, S.; Place, D.E.; Neale, G.; Vogel, P.; Kanneganti, T.D. ZBP1/DAI is an innate sensor of influenza virus triggering the NLRP3 inflammasome and programmed cell death pathways. Sci. Immunol. 2016, 1, aag2045. [Google Scholar] [CrossRef]
- Pan, P.; Zhang, Q.; Liu, W.; Wang, W.; Lao, Z.; Zhang, W.; Shen, M.; Wan, P.; Xiao, F.; Liu, F.; et al. Dengue Virus M Protein Promotes NLRP3 Inflammasome Activation To Induce Vascular Leakage in Mice. J. Virol. 2019, 93, e00996-19. [Google Scholar] [CrossRef]
- Halfmann, P.; Lindsay Hill-Batorski, L.; Kawaoka, Y. The Induction of IL-1β Secretion Through the NLRP3 Inflammasome During Ebola Virus Infection. J. Infect. Dis. 2018, 218 (Suppl. S5), S504–S507. [Google Scholar] [CrossRef]
- Dharra, R.; Kumar Sharma, A.; Datta, S. Emerging aspects of cytokine storm in COVID-19: The role of proinflammatory cytokines and therapeutic prospects. Cytokine 2023, 169, 156287. [Google Scholar] [CrossRef]
- Liu, X.; Shi, S.; Xiao, J.; Wang, H.; Chen, L.; Li, J.; Han, K. Prediction of the Severity of the Coronavirus Disease and Its Adverse Clinical Outcomes. Jpn. J. Infect. Dis. 2020, 73, 404–410. [Google Scholar] [CrossRef]
- Gallucci, S.; Maffei, M.E. DNA Sensing across the Tree of Life. Trends Immunol. 2017, 38, 719–732. [Google Scholar] [CrossRef]
- Martinez-Quinones, P.; Komic, A.; McCarthy, C.G.; Webb, R.C.; Ferreira Wenceslau, C. Targeting Endothelial Barrier Dysfunction Caused by Circulating Bacterial and Mitochondrial N-Formyl Peptides With Deformylase. Front. Immunol. 2019, 10, 1270. [Google Scholar] [CrossRef] [PubMed]
- Bittner, Z.A.; Schrader, M.; George, S.E.; Amann, R. Pyroptosis and Its Role in SARS-CoV-2 Infection. Cells 2022, 11, 1717. [Google Scholar] [CrossRef] [PubMed]
- Rayamajhi, M.; Zhang, Y.; Miao, E.A. Detection of pyroptosis by measuring released lactate dehydrogenase activity. Methods Mol. Biol. 2013, 1040, 85–90. [Google Scholar] [CrossRef] [PubMed]
- Ghofrani Nezhad, M.; Jami, G.; Kooshkaki, O.; Chamani, S.; Naghizadeh, A. The Role of Inflammatory Cytokines (Interleukin-1 and Interleukin-6) as a Potential Biomarker in the Different Stages of COVID-19 (Mild, Severe, and Critical). J. Interf. Cytokine Res. 2023, 43, 147–163. [Google Scholar] [CrossRef] [PubMed]
- Abers, M.S.; Delmonte, O.M.; Ricotta, E.E.; Fintzi, J.; Fink, D.L.; de Jesus, A.A.A.; Zarember, K.A.; Alehashemi, S.; Oikonomou, V.; Desai, J.V.; et al. An immune-based biomarker signature is associated with mortality in COVID-19 patients. JCI Insight 2021, 6, e144455. [Google Scholar] [CrossRef]
- Huet, T.; Beaussier, H.; Voisin, O.; Jouveshomme, S.; Dauriat, G.; Lazareth, I.; Sacco, E.; Naccache, J.M.; Bézie, Y.; Sophie Laplanche, S.; et al. Anakinra for severe forms of COVID-19: A cohort study. Lancet Rheumatol. 2020, 2, e393–e400. [Google Scholar] [CrossRef]
- Dahms, K.; Mikolajewska, A.; Ansems, K.; Metzendorf, M.I.; Benstoem, C.; Stegemann, M. Anakinra for the treatment of COVID-19 patients: A systematic review and meta-analysis. Eur. J. Med. Res. 2023, 28, 100. [Google Scholar] [CrossRef]
- Lan, S.H.; Hsu, C.K.; Chang, S.P.; Lu, L.C.; Lai, C.C. Clinical efficacy and safety of interleukin-1 blockade in the treatment of patients with COVID-19: A systematic review and meta-analysis of randomized controlled trials. Ann. Med. 2023, 55, 2208872. [Google Scholar] [CrossRef]
- Cavalli, G.; Lorenzo Dagna, L. The right place for IL-1 inhibition in COVID-19. Lancet Respir. Med. 2021, 9, 223–224. [Google Scholar] [CrossRef]
- CORIMUNO-19 Collaborative group. Effect of anakinra versus usual care in adults in hospital with COVID-19 and mild-to-moderate pneumonia (CORIMUNO-ANA-1): A randomised controlled trial. Lancet Respir. Med. 2021, 9, 295–304. [Google Scholar] [CrossRef]
- Caricchio, R.; Abbate, A.; Gordeev, I.; Meng, J.; Hsue, P.Y.; Neogi, T.; Arduino, R.; Fomina, D.; Bogdanov, R.; Stepanenko, T.; et al. Effect of Canakinumab vs Placebo on Survival Without Invasive Mechanical Ventilation in Patients Hospitalized With Severe COVID-19: A Randomized Clinical Trial. JAMA 2021, 326, 230–239. [Google Scholar] [CrossRef] [PubMed]
- Sheng, C.C.; Sahoo, D.; Dugar, S.; Prada, R.A.; Wang, T.K.M.; Abou Hassan, O.K.; Brennan, D.; Culver, D.A.; Rajendram, P.; Duggal, A.; et al. Canakinumab to reduce deterioration of cardiac and respiratory function in SARS-CoV-2 associated myocardial injury with heightened inflammation (canakinumab in COVID-19 cardiac injury: The three C study). Clin. Cardiol. 2020, 43, 1055–1063. [Google Scholar] [CrossRef] [PubMed]
- Kyriazopoulou, E.; Poulakou, G.; Milionis, H.; Metallidis, S.; Adamis, G.; Tsiakos, K.; Fragkou, A.; Rapti, A.; Damoulari, C.; Fantoni, M.; et al. Early treatment of COVID-19 with anakinra guided by soluble urokinase plasminogen receptor plasma levels: A double-blind, randomized controlled phase 3 trial. Nat. Med. 2021, 27, 1752–1760. [Google Scholar] [CrossRef] [PubMed]
- Rasmussen, S.R.; Nielsen, R.V.; Møgelvang, R.; Ostrowski, S.R.; Ravn, H.B. Prognostic value of suPAR and hsCRP on acute kidney injury after cardiac surgery. BMC Nephrol. 2021, 22, 120. [Google Scholar] [CrossRef]
- Velissaris, D.; Zareifopoulos, N.; Koniari, I.; Karamouzos, V.; Bousis, D.; Gerakaris, A.; Platanaki, C.; Kounis, N. Soluble Urokinase Plasminogen Activator Receptor as a Diagnostic and Prognostic Biomarker in Cardiac Disease. J. Clin. Med. Res. 2021, 13, 133–142. [Google Scholar] [CrossRef]
- Kyriazopoulou, E.; Panagopoulos, P.; Metallidis, S.; Dalekos, G.N.; Poulakou, G.; Gatselis, N.; Karakike, E.; Saridaki, M.; Loli, G.; Stefos, A.; et al. An open label trial of anakinra to prevent respiratory failure in COVID-19. eLife 2021, 10, e66125. [Google Scholar] [CrossRef]
- Azam, T.U.; Shadid, H.R.; Blakely, P.; O’Hayer, P.; Berlin, H.; Pan, M.; Zhao, P.; Zhao, L.; Pennathur, S.; Pop-Busui, R.; et al. Soluble Urokinase Receptor (SuPAR) in COVID-19-Related AKI. J. Am. Soc. Nephrol. 2020, 31, 2725–2735. [Google Scholar] [CrossRef]
- Hadjadj, J.; Yatim, N.; Barnabei, L.; Corneau, A.; Boussier, J.; Smith, N.; Péré, H.; Charbit, B.; Bondet, V.; Chenevier-Gobeaux, C.; et al. Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science 2020, 369, 718–724. [Google Scholar] [CrossRef]
- Guarda, G.; Braun, M.; Staehli, F.; Tardivel, A.; Mattmann, C.; Förster, I.; Farlik, M.; Decker, T.; Du Pasquier, R.A.; Romero, P.; et al. Type I interferon inhibits interleukin-1 production and inflammasome activation. Immunity 2011, 34, 213–223. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Napodano, C.; Carnazzo, V.; Basile, V.; Pocino, K.; Stefanile, A.; Gallucci, S.; Natali, P.; Basile, U.; Marino, M. NLRP3 Inflammasome Involvement in Heart, Liver, and Lung Diseases—A Lesson from Cytokine Storm Syndrome. Int. J. Mol. Sci. 2023, 24, 16556. https://doi.org/10.3390/ijms242316556
Napodano C, Carnazzo V, Basile V, Pocino K, Stefanile A, Gallucci S, Natali P, Basile U, Marino M. NLRP3 Inflammasome Involvement in Heart, Liver, and Lung Diseases—A Lesson from Cytokine Storm Syndrome. International Journal of Molecular Sciences. 2023; 24(23):16556. https://doi.org/10.3390/ijms242316556
Chicago/Turabian StyleNapodano, Cecilia, Valeria Carnazzo, Valerio Basile, Krizia Pocino, Annunziata Stefanile, Stefania Gallucci, Patrizia Natali, Umberto Basile, and Mariapaola Marino. 2023. "NLRP3 Inflammasome Involvement in Heart, Liver, and Lung Diseases—A Lesson from Cytokine Storm Syndrome" International Journal of Molecular Sciences 24, no. 23: 16556. https://doi.org/10.3390/ijms242316556
APA StyleNapodano, C., Carnazzo, V., Basile, V., Pocino, K., Stefanile, A., Gallucci, S., Natali, P., Basile, U., & Marino, M. (2023). NLRP3 Inflammasome Involvement in Heart, Liver, and Lung Diseases—A Lesson from Cytokine Storm Syndrome. International Journal of Molecular Sciences, 24(23), 16556. https://doi.org/10.3390/ijms242316556