Fibroblasts-specific p16INK4a exacerbates inflammageing-mediated post-infarction ventricular remodelling through interacting with STAT3 to regulate NLRP3 transcription

Xin Gu , Yingqiang Du , Jin'ge Zhang , Jiyu Li , Haiyun Chen , Yujie Lin , Yue Wang , Chunli Zhang , Shiyu Lin , Nannan Hao , Chengyi Peng , Jiacheng Ge , Jin Liu , Yan Liang , Yongjie Zhang , Xiaoyan Wang , Fang Wang , Jianliang Jin

Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (6) : e70344

PDF
Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (6) : e70344 DOI: 10.1002/ctm2.70344
RESEARCH ARTICLE

Fibroblasts-specific p16INK4a exacerbates inflammageing-mediated post-infarction ventricular remodelling through interacting with STAT3 to regulate NLRP3 transcription

Author information +
History +
PDF

Abstract

Background and Aims: Inflammageing represents both a critical pathophysiological hallmark and independent risk factor for myocardial infarction (MI), with age-related increases observed in MI incidence and severity of post-MI ventricular remodelling. Novel therapeutic strategies targeting inflammageing-driven mechanisms are urgently required to attenuate adverse ventricular remodelling following MI. This investigation was designed to elucidate the impact of fibroblast-specific p16INK4a on inflammageing-associated ventricular remodelling after MI and to develop a targeted nanotherapy to mitigate this process.

Methods and Results: We found that p16-mediated inflammageing positively correlated with the severity of post-infarction ventricular remodelling in patients. POSTN-driven p16INK4a knockout improved cardiac function, and reduced ventricular remodelling, myocardial inflammation and NLRP3 signalling activation following MI through downregulating STAT3-mediated NLRP3 inflammasome and upregulating glutathione metabolism pathway in fibroblasts. P16INK4a overexpression induced NLRP3 signalling activation through upregulating NLRP3 transcribed by STAT3 in fibroblasts. In terms of mechanisms, p16INK4a interacted with STAT3, which depended on the SH2 domain of STAT3; P16INK4a promoted the interaction of EZH2 and STAT3, increased the di-methylation on K49 and phosphorylation on Y705 of STAT3 by EZH2, and promoted NLRP3 transcription through regulating histone modification in the NLRP3 promoter by interfering the formation of Bmi-1-EZH2 or Bmi-1-BCL6 complex in fibroblasts. Injection of p16INK4a-accumulated ageing cardiac fibroblasts, or p16INK4a overexpression adenovirus aggravated profibrosis and proinflammation in MI area. However, a novel FH peptide ‘FHKHKSPALSPV’-neutrophil membrane proteins (NMPs)-artificial lipid (Li) membranes-mesoporous silica nanoparticle (MSN) core (FNLM)-nanocaged p16INK4a-siRNA, as a newly constructed nanomaterial drug, could prevent post-infarction ventricular remodelling through inhibiting NLRP3 transcription in targeted cardiac fibroblasts and ameliorating proinflammation and profibrosis.

Conclusions: P16INK4a drives inflammageing-mediated post-MI ventricular remodeling by activating STAT3/NLRP3 signaling in fibroblasts. Targeting p16INK4a via FNLM-siRNA nanotherapy represents a novel strategy to ameliorate adverse cardiac remodelling, offering translational potential for clinical intervention.

Keywords

nanotherapy / NLRP3 inflammasome / p16INK4a / post-infarction ventricular remodelling / STAT3

Cite this article

Download citation ▾
Xin Gu, Yingqiang Du, Jin'ge Zhang, Jiyu Li, Haiyun Chen, Yujie Lin, Yue Wang, Chunli Zhang, Shiyu Lin, Nannan Hao, Chengyi Peng, Jiacheng Ge, Jin Liu, Yan Liang, Yongjie Zhang, Xiaoyan Wang, Fang Wang, Jianliang Jin. Fibroblasts-specific p16INK4a exacerbates inflammageing-mediated post-infarction ventricular remodelling through interacting with STAT3 to regulate NLRP3 transcription. Clinical and Translational Medicine, 2025, 15(6): e70344 DOI:10.1002/ctm2.70344

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Perrone MA, Aimo A, Bernardini S, Clerico A. Inflammageing and cardiovascular system: focus on cardiokines and cardiac-specific biomarkers. Int J Mol Sci. 2023; 24: 844.

[2]

Andonian BJ, Hippensteel JA, Abuabara K, et al. Inflammation and aging-related disease: a transdisciplinary inflammaging framework. Geroscience. 2025; 47: 515-542.

[3]

Liu D, Richardson G, Benli FM, et al. Inflammageing in the cardiovascular system: mechanisms, emerging targets, and novel therapeutic strategies. Clin Sci (Lond). 2020; 134: 2243-2262.

[4]

Uriel N, Sayer G, Annamalai S, Kapur NK, Burkhoff D. Mechanical unloading in heart failure. J Am Coll Cardiol. 2018; 72: 569-580.

[5]

Suetomi T, Miyamoto S, Brown JH. Inflammation in nonischemic heart disease: initiation by cardiomyocyte CaMKII and NLRP3 inflammasome signaling. Am J Physiol Heart Circ Physiol. 2019; 317: H877-H890.

[6]

D'Arcangelo D, Tinaburri L, Dellambra E. The Role of p16(INK4a) pathway in human epidermal stem cell self-renewal, aging and cancer. Int J Mol Sci. 2017; 18: 1591.

[7]

Li J, Karim MA, Che H, Geng Q, Miao D. Deletion of p16 prevents estrogen deficiency-induced osteoporosis by inhibiting oxidative stress and osteocyte senescence. Am J Transl Res. 2020; 12: 672-683.

[8]

Gu X, Meng H, Peng C, et al. Inflammasome activation and metabolic remodelling in p16-positive aging cells aggravates high-fat diet-induced lung fibrosis by inhibiting NEDD4L-mediated K48-polyubiquitin-dependent degradation of SGK1. Clin Transl Med. 2023; 13: e1308.

[9]

Anderson R, Lagnado A, Maggiorani D, et al. Length-independent telomere damage drives post-mitotic cardiomyocyte senescence. EMBO J. 2019; 38: e100492.

[10]

Redgrave R, Dookun E, Booth L, et al. Senescent cardiomyocytes contribute to cardiac dysfunction following myocardial infarction. Heart. 2023; 109: A249-A249.

[11]

Piber D, Olmstead R, Cho JH, et al. Inflammaging: age and systemic, cellular, and nuclear inflammatory biology in older adults. J Gerontol A Biol Sci Med Sci. 2019; 74: 1716-1724.

[12]

Xia S, Zhang X, Zheng S, et al. An update on inflamm-aging: mechanisms, prevention, and treatment. J Immunol Res. 2016; 2016: 8426874.

[13]

Turner NA. Inflammatory and fibrotic responses of cardiac fibroblasts to myocardial damage associated molecular patterns (DAMPs). J Mol Cell Cardiol. 2016; 94: 189-200.

[14]

Tang X, Li PH, Chen HZ. Cardiomyocyte senescence and cellular communications within myocardial microenvironments. Front Endocrinol (Lausanne). 2020; 11: 280.

[15]

Latz E, Duewell P. NLRP3 inflammasome activation in inflammaging. Semin Immunol. 2018; 40: 61-73.

[16]

Wang Z, Hu W, Lu C, et al. Targeting NLRP3 (nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3) inflammasome in cardiovascular disorders. Arterioscler Thromb Vasc Biol. 2018; 38: 2765-2779.

[17]

Samad MA, Ahmad I, Hasan A, et al. STAT3 signaling pathway in health and disease. MedComm. 2025; 6: e70152.

[18]

Rao T, Tong H, Li J, Huang J, Yin Y, Zhang J. Exploring the role and mechanism of hyperoside against cardiomyocyte injury in mice with myocardial infarction based on JAK2/STAT3 signaling pathway. Phytomedicine. 2024; 128: 155319.

[19]

Wang L, Zhan X, Shen X, et al. P16 promotes the growth and mobility potential of breast cancer both in vitro and in vivo: the key role of the activation of IL-6/JAK2/STAT3 signaling. Mol Cell Biochem. 2018; 446: 137-148.

[20]

Guarda G, Braun M, Staehli F, et al. Type I interferon inhibits interleukin-1 production and inflammasome activation. Immunity. 2011; 34: 213-223.

[21]

Chen H, Chen H, Liang J, et al. TGF-beta1/IL-11/MEK/ERK signaling mediates senescence-associated pulmonary fibrosis in a stress-induced premature senescence model of Bmi-1 deficiency. Exp Mol Med. 2020; 52: 130-151.

[22]

Choi BY, Choi HS, Ko K, et al, The tumor suppressor p16(INK4a) prevents cell transformation through inhibition of c-Jun phosphorylation and AP-1 activity. Nat Struct Mol Biol. 2005; 12: 699-707.

[23]

Zhou W, Huang W, You H, et al. EZH2 knockout in mice activates STAT3 signalling via STAT3 methylation and modulates ferroptosis in pulpitis-affected dental pulp vascular endothelial cells: a laboratory investigation. Int Endod J. 2025.

[24]

Chen Q, Hu Q, Chen Y, et al. PRMT6 methylation of STAT3 regulates tumor metastasis in breast cancer. Cell Death Dis. 2023; 14: 655.

[25]

Kim E, Kim M, Woo DH, et al. Phosphorylation of EZH2 activates STAT3 signaling via STAT3 methylation and promotes tumorigenicity of glioblastoma stem-like cells. Cancer Cell. 2013; 23: 839-852.

[26]

Dasgupta M, Dermawan JK, Willard B, Stark GR. STAT3-driven transcription depends upon the dimethylation of K49 by EZH2. Proc Natl Acad Sci USA. 2015; 112: 3985-3990.

[27]

Bejerano T, Etzion S, Elyagon S, Etzion Y, Cohen S. Nanoparticle delivery of miRNA-21 mimic to cardiac macrophages improves myocardial remodeling after myocardial infarction. Nano Lett. 2018; 18: 5885-5891.

[28]

Kwon SP, Hwang BH, Park EH, et al. Nanoparticle-mediated blocking of excessive inflammation for prevention of heart failure following myocardial infarction. Small. 2021; 17: e2101207.

[29]

Wang Q, Song Y, Chen J, et al. Direct in vivo reprogramming with non-viral sequential targeting nanoparticles promotes cardiac regeneration. Biomaterials. 2021; 276: 121028.

[30]

Ruiz-Villalba A, Romero JP, Hernandez SC, et al. Single-cell RNA sequencing analysis reveals a crucial role for CTHRC1 (collagen triple helix repeat containing 1) cardiac fibroblasts after myocardial infarction. Circulation. 2020; 142: 1831-1847.

[31]

Liu CC, Huang ZX, Li X, et al. Upregulation of NLRP3 via STAT3-dependent histone acetylation contributes to painful neuropathy induced by bortezomib. Exp Neurol. 2018; 302: 104-111.

[32]

Kruczyk M, Przanowski P, Dabrowski M, et al. Integration of genome-wide of Stat3 binding and epigenetic modification mapping with transcriptome reveals novel Stat3 target genes in glioma cells. Biochim Biophys Acta. 2014; 1839: 1341-1350.

[33]

Fang RH, Hu CM, Luk BT, et al. Cancer cell membrane-coated nanoparticles for anticancer vaccination and drug delivery. Nano Lett. 2014; 14: 2181-2188.

[34]

Mehdizadeh M, Aguilar M, Thorin E, Ferbeyre G, Nattel S. The role of cellular senescence in cardiac disease: basic biology and clinical relevance. Nat Rev Cardiol. 2022; 19: 250-264.

[35]

Kura B, Szeiffova Bacova B, Kalocayova B, Sykora M, Slezak J. Oxidative Stress-responsive microRNAs in heart injury. Int J Mol Sci. 2020; 21: 358.

[36]

Zhu W, Wu RD, Lv YG, Liu YM, Huang H, Xu JQ. BRD4 blockage alleviates pathological cardiac hypertrophy through the suppression of fibrosis and inflammation via reducing ROS generation. Biomed Pharmacother. 2020; 121: 109368.

[37]

Ma ZG, Yuan YP, Wu HM, Zhang X, Tang QZ. Cardiac fibrosis: new insights into the pathogenesis. Int J Biol Sci. 2018; 14: 1645-1657.

[38]

Anzai T. Inflammatory mechanisms of cardiovascular remodeling. Circ J. 2018; 82: 629-635.

[39]

Ridker PM, Everett BM, Thuren T, et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N Engl J Med. 2017; 377: 1119-1131.

[40]

Morton AC, Rothman AM, Greenwood JP, et al. The effect of interleukin-1 receptor antagonist therapy on markers of inflammation in non-ST elevation acute coronary syndromes: the MRC-ILA Heart Study. Eur Heart J. 2015; 36: 377-384.

[41]

Bueno V, Sant'Anna OA, Lord JM. Ageing and myeloid-derived suppressor cells: possible involvement in immunosenescence and age-related disease. Age. 2014; 36: 9729.

[42]

Kanisicak O, Khalil H, Ivey MJ, et al. Genetic lineage tracing defines myofibroblast origin and function in the injured heart. Nat Commun. 2016; 7: 12260.

[43]

Wu JB, Subbaiah KCV, Xie LHT, et al. Glutamyl-prolyl-tRNA synthetase regulates proline-rich pro-fibrotic protein synthesis during cardiac fibrosis. Circ Res. 2020; 127: 827-846.

[44]

Xue K, Chen S, Chai JY, et al. Upregulation of periostin through CREB participates in myocardial infarction-induced myocardial fibrosis. J Cardiovasc Pharm. 2022; 79: 687-697.

[45]

Johnson DE, O'Keefe RA, Grandis JR. Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nat Rev Clin Oncol. 2018; 15: 234-248.

[46]

Banerjee K, Resat H. Constitutive activation of STAT3 in breast cancer cells: a review. Int J Cancer. 2016; 138: 2570-2578.

[47]

Decker T, Kovarik P. Serine phosphorylation of STATs. Oncogene. 2000; 19: 2628-2637.

[48]

Bhattacharya R, Mustafi SB, Street M, Dey A, Dwivedi SK. Bmi-1: at the crossroads of physiological and pathological biology. Genes Dis. 2015; 2: 225-239.

[49]

Zhao J, Chen A, Wang R, et al. Bmi-1 epigenetically orchestrates osteogenic and adipogenic differentiation of bone marrow mesenchymal stem cells to delay bone aging. Adv Sci (Weinh). 2024; 11: e2404518.

[50]

Huai W, Zhao R, Song H, et al. Aryl hydrocarbon receptor negatively regulates NLRP3 inflammasome activity by inhibiting NLRP3 transcription. Nat Commun. 2014; 5: 4738.

[51]

Chen D, Xiong XQ, Zang YH, et al. BCL6 attenuates renal inflammation via negative regulation of NLRP3 transcription. Cell Death Dis. 2017; 8: e3156.

[52]

Beguelin W, Teater M, Gearhart MD, et al. EZH2 and BCL6 cooperate to assemble CBX8-BCOR complex to repress bivalent promoters, mediate germinal center formation and lymphomagenesis. Cancer Cell. 2016; 30: 197-213.

[53]

Mosteiro L, Pantoja C, de Martino A, Serrano M. Senescence promotes in vivo reprogramming through p16(INK)(4a) and IL-6. Aging Cell. 2018; 17: e12711.

[54]

Nacarelli T, Lau L, Fukumoto T, et al. NAD(+) metabolism governs the proinflammatory senescence-associated secretome. Nat Cell Biol. 2019; 21: 397-407.

[55]

Neelakantan H, Brightwell CR, Graber TG, et al. Small molecule nicotinamide N-methyltransferase inhibitor activates senescent muscle stem cells and improves regenerative capacity of aged skeletal muscle. Biochem Pharmacol. 2019; 163: 481-492.

[56]

Linton PJ, Thoman ML. Immunosenescence in monocytes, macrophages, and dendritic cells: lessons learned from the lung and heart. Immunol Lett. 2014; 162: 290-297.

[57]

Sapochnik M, Fuertes M, Arzt E. Programmed cell senescence: role of IL-6 in the pituitary. J Mol Endocrinol. 2017; 58: R241-R253.

[58]

Grosse L, Wagner N, Emelyanov A, et al. Defined p16(high) senescent cell types are indispensable for mouse healthspan. Cell Metab. 2020; 32: 87-99e86.

[59]

Shi J, Sun J, Liu L, et al. P16ink4a overexpression ameliorates cardiac remodeling of mouse following myocardial infarction via CDK4/pRb pathway. Biochem Biophys Res Commun. 2022; 595: 62-68.

RIGHTS & PERMISSIONS

2025 The Author(s). Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

6

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/