[1]吴浩亮,周文杰,崔胜宇,等.NLRP3炎症小体在病理性心肌肥厚中的研究进展[J].心血管病学进展,2022,(10):923.[doi:10.16806/j.cnki.issn.1004-3934.2022.10.014]
 WU Haoliang,ZHOU Wenjie,CUI Shengyu,et al.NLRP3 Inflammasome in Pathological Cardiac Hypertrophy[J].Advances in Cardiovascular Diseases,2022,(10):923.[doi:10.16806/j.cnki.issn.1004-3934.2022.10.014]
点击复制

NLRP3炎症小体在病理性心肌肥厚中的研究进展()
分享到:

《心血管病学进展》[ISSN:51-1187/R/CN:1004-3934]

卷:
期数:
2022年10期
页码:
923
栏目:
综述
出版日期:
2022-10-25

文章信息/Info

Title:
NLRP3 Inflammasome in Pathological Cardiac Hypertrophy
作者:
吴浩亮1周文杰1崔胜宇1陶波1冯高科1易欣1吴青青1孟祥平2浦湧3范稣圳1徐林1
(1.武汉大学人民医院心内科 武汉大学心血管病研究所 心血管病湖北省重点实验室,湖北 武汉 430060; 2.武汉市第四医院,湖北 武汉 430030;3.武汉大学人民医院汉南医院(汉南区人民医院)
Author(s):
WU Haoliang1 ZHOU Wenjie 1 CUI Shengyu1 TAO Bo 1 FENG Gaoke1 YI Xin1 WU Qingqing1 MENG Xiangping2 PU Yong3 FAN Suzhen1 XU Lin1
(1.Department of Cardiology,Renmin Hospital of Wuhan University;Cardiovascular Research Institute,Wuhan University; Hubei Key Laboratory of Cardiology,Hubei 430060 ,Wuhan,China; 2. Wuhan Fourth Hospital , Hubei 4300 30,Wuhan,China;3. Renmin Hospital of Hannan,Renmin Hospital of Wuhan University,Hubei 430090 Wuhan,China)
关键词:
病理性心肌肥厚NOD样受体蛋白3炎症小体
Keywords:
Pathological cardiac hypertrophyNOD-like?receptor?protein?3 Inflammasome
DOI:
10.16806/j.cnki.issn.1004-3934.2022.10.014
摘要:
病理性心肌肥厚是指在各种病理因素作用下导致的心脏体积增大,质量增加和功能下降的一种病理状态。先前研究普遍认为病理性心肌肥厚是心脏在各种病理因素刺激下做出的适应性反应。而随着对机制的进一步深入了解,人们发现炎症反应、转录翻译、免疫调节、表观遗传都在病理性心肌肥厚中发挥重要的调控作用。因此不能简单将病理性心肌肥厚理解为心肌的代偿性反应。在众多作用因素中,NOD样受体蛋白3(NLRP3)调控的无菌性炎症反应是病理性心肌肥厚发生发展过程中的关键驱动因素。现总结NLRP3炎症小体在促进病理性心肌肥厚发生发展过程中潜在分子机制以及与NLRP3炎症小体相关的靶向药物在心肌肥厚中的应用。
Abstract:
Pathological cardiac hypertrophy is a state stimulated by various pathological factors ,which is characterized by the increase of heart volume and mass and the decrease of cardiac function. Previous studies generally accepted that cardiac hypertrophy is the adaptive response of the heart to the stimulation of various physiological and pathological factors. However,with the in-depth understanding of cardiac hypertrophy,people have found that in pathological cardiac hypertrophy,inflammatory response,transcription and translation,immune regulation and epigenetics all play an important regulatory role and thus pathological cardiac hypertrophy can not be simply understood as the compensatory response of myocardium. Among these many factors,the aseptic inflammatory response regulated by NOD-like?receptor?protein?3 ( NLRP3) is the key driving factor in the occurrence and development of pathological cardiac hypertrophy. This review summarized the potential molecular mechanism of NLRP3 inflammasome in promoting the occurrence and development of pathological cardiac hypertrophy and the application of therapeutic measures related to NLRP3 inflammasome in pathological cardiac hypertrophy.

参考文献/References:


[1] Nakamura M,Sadoshima J. Mechanisms of physiological and pathological cardiac hypertrophy[J]. Nat Rev Cardiol,2018,15(7):387-407.

[2] Lin HB,Naito K,Oh Y,et al. Innate immune Nod1/RIP2 signaling is essential for cardiac hypertrophy but requires mitochondrial antiviral signaling protein for signal transductions and energy balance[J]. Circulation,2020,142(23):2240-2258.

[3] Tupik JD,Nagai-Singer MA,Allen IC. To protect or adversely affect? The dichotomous role of the NLRP1 inflammasome in human disease[J]. Mol Aspects Med,2020,76:100858.

[4] He Y,Hara H,Nú?ez G. Mechanism and regulation of NLRP3 inflammasome activation[J]. Trends Biochem Sci,2016,41(12):1012-1021.

[5] Fritz JH,Ferrero RL,Philpott DJ,et al. Nod-like proteins in immunity,inflammation and disease[J]. Nat Immunol,2006,7(12):1250-1257.

[6] Franchi L,Mu?oz-Planillo R,Nú?ez G. Sensing and reacting to microbes through the inflammasomes[J]. Nat Immunol,2012,13(4):325-332.

[7] Travassos LH,Carneiro LA,Ramjeet M,et al. Nod1 and Nod2 direct autophagy by recruiting ATG16L1 to the plasma membrane at the site of bacterial entry[J]. Nat Immunol,2010,11(1):55-62.

[8] Suzuki T,Franchi L,Toma C,et al. Differential regulation of caspase-1 activation,pyroptosis,and autophagy via Ipaf and ASC in Shigella-infected macrophages[J]. PLoS Pathog,2007,3(8):e111.

[9] da Silva Correia J,Miranda Y,Austin-Brown N,et al. Nod1-dependent control of tumor growth[J]. Proc Natl Acad Sci U S A,2006,103(6):1840-1845.

[10] Richardson WM,Sodhi CP,Russo A,et al. Nucleotide-binding oligomerization domain-2 inhibits toll-like receptor-4 signaling in the intestinal epithelium[J]. Gastroenterology,2010,139(3):904-917.e6.

[11] Zamboni DS,Kobayashi KS,Kohlsdorf T,et al. The Birc1e cytosolic pattern-recognition receptor contributes to the detection and control of Legionella pneumophila infection[J]. Nat Immunol,2006,7(3):318-325.

[12] Wright EK,Goodart SA,Growney JD,et al. Naip5 affects host susceptibility to the intracellular pathogen Legionella pneumophila [J]. Curr Biol,2003,13(1):27-36.

[13] Koonin EV,Aravind L. The NACHT family—A new group of predicted NTPases implicated in apoptosis and MHC transcription activation[J]. Trends Biochem Sci,2000,25(5):223-224.

[14] Bouskra D,Brezillon C,Berard M,et al. Lymphoid tissue genesis induced by commensals through NOD1 regulates intestinal homeostasis[J]. Nature,2008,456(7221):507-510.

[15] Halle A,Hornung V,Petzold GC,et al. The NALP3 inflammasome is involved in the innate immune response to amyloid-beta[J]. Nat Immunol,2008,9(8):857-865.

[16] Mezzaroma E,Toldo S,Farkas D,et al. The inflammasome promotes adverse cardiac remodeling following acute myocardial infarction in the mouse[J]. Proc Natl Acad Sci U S A,2011,108(49):19725-19730.

[17] Kawaguchi M,Takahashi M,Hata T,et al. Inflammasome activation of cardiac fibroblasts is essential for myocardial ischemia/reperfusion injury[J]. Circulation,2011,123(6):594-604.

[18] Sandanger O,Ranheim T,Vinge LE,et al. The NLRP3 inflammasome is up-regulated in cardiac fibroblasts and mediates myocardial ischaemia-reperfusion injury[J]. Cardiovasc Res,2013,99(1):164-174.

[19] Guo H,Callaway JB,Ting JP. Inflammasomes:mechanism of action,role in disease,and therapeutics[J]. Nat Med,2015,21(7):677-687.

[20] Mangan MSJ,Olhava EJ,Roush WR,et al. Targeting the NLRP3 inflammasome in inflammatory diseases[J]. Nat Rev Drug Discov,2018,17(9):688.

[21] Franchi L,Eigenbrod T,Munoz-Planillo R,et al. The inflammasome:a caspase-1-activation platform that regulates immune responses and disease pathogenesis[J]. Nat Immunol,2009,10(3):241-247.

[22] Dinarello CA. Immunological and inflammatory functions of the interleukin-1 family[J]. Annu Rev Immunol,2009,27:519-550.

[23] Fink SL,Cookson BT. Caspase-1-dependent pore formation during pyroptosis leads to osmotic lysis of infected host macrophages[J]. Cell Microbiol,2006,8(11):1812-1825.

[24] Xiao H,Li H,Wang JJ,et al. IL-18 cleavage triggers cardiac inflammation and fibrosis upon beta-adrenergic insult[J]. Eur Heart J,2018,39(1):60-69.

[25] Dang S,Zhang ZY,Li KL,et al. Blockade of β-adrenergic signaling suppresses inflammasome and alleviates cardiac fibrosis[J]. Ann Transl Med,2020,8(4):127.

[26] Suetomi T,Willeford A,Brand CS,et al. Inflammation and NLRP3 inflammasome activation initiated in response to pressure overload by Ca2+/calmodulin-dependent protein kinase Ⅱ δ Signaling in cardiomyocytes are essential for adverse cardiac remodeling[J]. Circulation,2018,138(22):2530-2544.

[27] Willeford A,Suetomi T,Nickle A,et al. CaMKⅡδ-mediated inflammatory gene expression and inflammasome activation in cardiomyocytes initiate inflammation and induce fibrosis[J]. JCI Insight,2018,3(12):e97054.

[28] Wang Y,Liu X,Shi H,et al. NLRP3 inflammasome,an immune-inflammatory target in pathogenesis and treatment of cardiovascular diseases[J]. Clin Transl Med,2020,10(1):91-106.

[29] Anker SD,von Haehling S. Inflammatory mediators in chronic heart failure:an overview[J]. Heart,2004,90(4):464-470.

[30] Dick SA,Epelman S. Chronic heart failure and inflammation:what do we really know?[J]. Circ Res,2016,119(1):159-176.

[31] Chen Y,Zeng M,Zhang Y,et al. Nlrp3 deficiency alleviates angiotensin Ⅱ-induced cardiomyopathy by inhibiting mitochondrial dysfunction[J]. Oxid Med Cell Longev,2021,2021:6679100.

[32] Hornung V,Bauernfeind F,Halle A,et al. Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization[J]. Nat Immunol,2008,9(8):847-856.

[33] Gaidt MM,Ebert TS,Chauhan D,et al. The DNA inflammasome in human myeloid cells is initiated by a STING-cell death program upstream of NLRP3[J]. Cell,2017,171(5):1110-1124.e18.

[34] Li N,Zhou H,Wu H,et al. STING-IRF3 contributes to lipopolysaccharide-induced cardiac dysfunction,inflammation,apoptosis and pyroptosis by activating NLRP3[J]. Redox Biol,2019,24:101215.

[35] Lim S,Lee ME,Jeong J,et al. sRAGE attenuates angiotensin Ⅱ-induced cardiomyocyte hypertrophy by inhibiting RAGE-NFkappaB-NLRP3 activation[J]. Inflamm Res,2018,67(8):691-701.

[36] Li X,Zhu Q,Wang Q,et al. Protection of sacubitril/valsartan against pathological cardiac remodeling by inhibiting the NLRP3 inflammasome after relief of pressure overload in mice[J]. Cardiovasc Drugs Ther,2020,34(5):629-640.

[37] Xu L,Balzarolo M,Robinson EL,et al. NOX1 mediates metabolic heart disease in mice and is upregulated in monocytes of humans with diastolic dysfunction[J]. Cardiovasc Res,2021:cvab349.

[38] Byrne NJ,Soni S,Takahara S,et al. Chronically elevating circulating ketones can reduce cardiac inflammation and blunt the development of heart failure[J]. Circ Heart Fail,2020,13(6):e006573.

[39] Ding YY,Li JM,Guo FJ,et al. Triptolide upregulates myocardial forkhead helix transcription factor p3 expression and attenuates cardiac hypertrophy[J]. Front Pharmacol,2016,7:471.

[40] Pan XC,Liu Y,Cen YY,et al. Dual role of triptolide in interrupting the NLRP3 inflammasome pathway to attenuate cardiac fibrosis[J]. Int J Mol Sci,2019,20(2):360.

[41] Coll RC,Robertson AA,Chae JJ,et al. A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases[J]. Nat Med,2015,21(3) :248-255.

[42] Gan W,Ren J,Li T,et al. The SGK1 inhibitor EMD638683,prevents angiotensinⅡ-induced cardiac inflammation and fibrosis by blocking NLRP3 inflammasome activation[J]. Biochim Biophys Acta Mol Basis Dis,2018,1864(1):1-10.

[43] Sano S,Oshima K,Wang Y,et al. Tet2-mediated clonal hematopoiesis accelerates heart failure through a mechanism involving the IL-1β/NLRP3 inflammasome[J]. J Am Coll Cardiol,2018,71(8):875-886.

[44] Zhao M,Zhang J,Xu Y,et al. Selective inhibition of NLRP3 inflammasome reverses pressure overload-induced pathological cardiac remodeling by attenuating hypertrophy,fibrosis,and inflammation[J]. Int Immunopharmacol,2021,99:108046.

[45] Tang X,Pan L,Zhao S,et al. SNO-MLP (S-nitrosylation of muscle LIM protein) facilitates myocardial hypertrophy through TLR3 (Toll-like receptor 3)-mediated RIP3 (receptor-interacting protein kinase 3) and NLRP3 (NOD-like receptor pyrin domain containing 3) inflammasome activation[J]. Circulation,2020,141(12):984-1000.

[46] Wang F,Liang Q,Ma Y,et al. Silica nanoparticles induce pyroptosis and cardiac hypertrophy via ROS/NLRP3/Caspase-1 pathway[J]. Free Radic Biol Med,2022,182:171-181.

[47] Ridker PM,Everett BM,Thuren T,et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease[J]. N Engl J Med,2017,377(12):1119-1131.

[48] Everett BM,Cornel JH,Lainscak M,et al. Anti-inflammatory therapy with canakinumab for the prevention of hospitalization for heart failure[J]. Circulation,2019,139(10):1289-1299.

更新日期/Last Update: 2022-12-26