HDAC3 in action: Expanding roles in inflammation and inflammatory diseases

Ruyuan He , Zhuokun He , Tianyu Zhang , Bohao Liu , Minglang Gao , Ning Li , Qing Geng

Cell Proliferation ›› 2025, Vol. 58 ›› Issue (1) : e13731

PDF
Cell Proliferation ›› 2025, Vol. 58 ›› Issue (1) : e13731 DOI: 10.1111/cpr.13731
REVIEW

HDAC3 in action: Expanding roles in inflammation and inflammatory diseases

Author information +
History +
PDF

Abstract

Inflammation serves as the foundation for numerous physiological and pathological processes, driving the onset and progression of various diseases. Histone deacetylase 3 (HDAC3), an essential chromatin-modifying protein within the histone deacetylase superfamily, exerts its transcriptional inhibitory role through enzymatic histone modification to uphold normal physiological function, growth, and development of the body. With both enzymatic and non-enzymatic activities, HDAC3 plays a pivotal role in regulating diverse transcription factors associated with inflammatory responses and related diseases. This review examines the involvement of HDAC3 in inflammatory responses while exploring its therapeutic potential as a target for treating inflammatory diseases, thereby offering valuable insights for clinical applications.

Cite this article

Download citation ▾
Ruyuan He, Zhuokun He, Tianyu Zhang, Bohao Liu, Minglang Gao, Ning Li, Qing Geng. HDAC3 in action: Expanding roles in inflammation and inflammatory diseases. Cell Proliferation, 2025, 58(1): e13731 DOI:10.1111/cpr.13731

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

MedzhitovR. The spectrum of inflammatory responses. Science (New York, NY). 2021;374:1070-1075.

[2]

HotamisligilGS. Inflammation, metaflammation and immunometabolic disorders. Nature. 2017;542:177-185.

[3]

MeizlishML, Franklin RA, ZhouX, MedzhitovR. Tissue homeostasis and inflammation. Annu Rev Immunol. 2021;39:557-581.

[4]

HunterP. The inflammation theory of disease. The growing realization that chronic inflammation is crucial in many diseases opens new avenues for treatment. EMBO Rep. 2012;13:968-970.

[5]

CoussensLM, WerbZ. Inflammation and cancer. Nature. 2002;420:860-867.

[6]

VerdinE, OttM. 50 years of protein acetylation: from gene regulation to epigenetics, metabolism and beyond. Nat Rev Mol Cell Biol. 2015;16:258-264.

[7]

GoodsonM, JonasBA, PrivalskyMA. Corepressors: custom tailoring and alterations while you wait. Nucl Recept Signal. 2005;3:e003.

[8]

LiJ, WangJ, WangJ, et al. Both corepressor proteins SMRT and N-CoR exist in large protein complexes containing HDAC3. EMBO J. 2000;19:4342-4350.

[9]

NingL, RuiX, BoW, QingG. The critical roles of histone deacetylase 3 in the pathogenesis of solid organ injury. Cell Death Dis. 2021;12:734.

[10]

NguyenHCB, Adlanmerini M, HauckAK, LazarMA. Dichotomous engagement of HDAC3 activity governs inflammatory responses. Nature. 2020;584:286-290.

[11]

ZhangK, LiuZ, YaoY, et al. Structure-based design of a selective class I Histone Deacetylase (HDAC) near-infrared (NIR) probe for epigenetic regulation detection in triple-negative breast cancer (TNBC). J Med Chem. 2021;64:4020-4033.

[12]

YangX-J, SetoE. The Rpd3/Hda1 family of lysine deacetylases: from bacteria and yeast to mice and men. Nat Rev Mol Cell Biol. 2008;9:206-218.

[13]

SauveAA, Wolberger C, SchrammVL, BoekeJD. The biochemistry of sirtuins. Annu Rev Biochem. 2006;75:435-465.

[14]

YoonS, EomGH. HDAC and HDAC inhibitor: from cancer to cardiovascular diseases. Chonnam Med J. 2016;52:1-11.

[15]

ChenX, HeY, FuW, et al. Histone deacetylases (HDACs) and atherosclerosis: a mechanistic and pharmacological review. Front Cell Dev Biol. 2020;8:581015.

[16]

EmmettMJ, LazarMA. Integrative regulation of physiology by histone deacetylase 3. Nat Rev Mol Cell Biol. 2019;20:102-115.

[17]

MaolanonAR, MadsenAS, OlsenCA. Innovative strategies for selective inhibition of histone deacetylases. Cell Chem Biol. 2016;23:759-768.

[18]

AdhikariN, AminSA, TrivediP, Jha T, GhoshB. HDAC3 is a potential validated target for cancer: an overview on the benzamide-based selective HDAC3 inhibitors through comparative SAR/QSAR/QAAR approaches. Eur J Med Chem. 2018;157:1127-1142.

[19]

CaoF, Zwinderman MRH, DekkerFJ. The process and strategy for developing selective histone deacetylase 3 inhibitors. Molecules (Basel, Switzerland). 2018;23:551.

[20]

ZhangY, LeRoyG, SeeligHP, Lane WS, ReinbergD. The dermatomyositis-specific autoantigen Mi2 is a component of a complex containing histone deacetylase and nucleosome remodeling activities. Cell. 1998;95:279-289.

[21]

YoonH-G, ChanDW, HuangZ-Q, et al. Purification and functional characterization of the human N-CoR complex: the roles of HDAC3, TBL1 and TBLR1. EMBO J. 2003;22:1336-1346.

[22]

ZhangJ, KalkumM, ChaitBT, Roeder RG. The N-CoR-HDAC3 nuclear receptor corepressor complex inhibits the JNK pathway through the integral subunit GPS2. Mol Cell. 2002;9:611-623.

[23]

PerissiV, Rosenfeld MG. Controlling nuclear receptors: the circular logic of cofactor cycles. Nat Rev Mol Cell Biol. 2005;6:542-554.

[24]

HuX, LazarMA. The CoRNR motif controls the recruitment of corepressors by nuclear hormone receptors. Nature. 1999;402:93-96.

[25]

PerissiV, JepsenK, GlassCK, Rosenfeld MG. Deconstructing repression: evolving models of co-repressor action. Nat Rev Genet. 2010;11:109-123.

[26]

BoyerLA, LangerMR, CrowleyKA, Tan S, DenuJM, PetersonCL. Essential role for the SANT domain in the functioning of multiple chromatin remodeling enzymes. Mol Cell. 2002;10:935-942.

[27]

GuentherMG, YuJ, KaoGD, Yen TJ, LazarMA. Assembly of the SMRT-histone deacetylase 3 repression complex requires the TCP-1 ring complex. Genes Dev. 2002;16:3130-3135.

[28]

SunZ, FengD, FangB, et al. Deacetylase-independent function of HDAC3 in transcription and metabolism requires nuclear receptor corepressor. Mol Cell. 2013;52:769-782.

[29]

AdrainC, Freeman M. New lives for old: evolution of pseudoenzyme function illustrated by iRhoms. Nat Rev Mol Cell Biol. 2012;13:489-498.

[30]

YouS-H, LimH-W, SunZ, Broache M, WonK-J, LazarMA. Nuclear receptor co-repressors are required for the histone-deacetylase activity of HDAC3 in vivo. Nat Struct Mol Biol. 2013;20:182-187.

[31]

ShakespearMR, HaliliMA, IrvineKM, Fairlie DP, SweetMJ. Histone deacetylases as regulators of inflammation and immunity. Trends Immunol. 2011;32:335-343.

[32]

ChenG, Goeddel DV. TNF-R1 signaling: a beautiful pathway. Science (New York, NY). 2002;296:1634-1635.

[33]

LundbladLKA, Thompson-Figueroa J, LeclairT, et al. Tumor necrosis factor-alpha overexpression in lung disease: a single cause behind a complex phenotype. Am J Respir Crit Care Med. 2005;171:1363-1370.

[34]

AngiolilliC, KabalaPA, GrabiecAM, et al. Histone deacetylase 3 regulates the inflammatory gene expression programme of rheumatoid arthritis fibroblast-like synoviocytes. Ann Rheum Dis. 2017;76:277-285.

[35]

LiuH, WangJ, HeT, et al. Butyrate: a double-edged sword for health? Adv Nutri. 2018;9:21-29.

[36]

SchioppaT, NguyenHO, TiberioL, et al. Inhibition of class I histone deacetylase activity blocks the induction of TNFAIP3 both directly and indirectly via the suppression of endogenous TNF-α. Int J Mol Sci. 2022;23:9752.

[37]

CantleyMD, HaynesDR. Epigenetic regulation of inflammation: progressing from broad acting histone deacetylase (HDAC) inhibitors to targeting specific HDACs. Inflammopharmacology. 2013;21:301-307.

[38]

RyuY, KeeHJ, SunS, et al. Class I histone deacetylase inhibitor MS-275 attenuates vasoconstriction and inflammation in angiotensin II-induced hypertension. PLoS One. 2019;14:e0213186.

[39]

WangT-Y, ChangM-M, LiY-SJ, Huang T-C, ChienS, WuC-C. Maintenance of HDACs and H3K9me3 prevents arterial flow-induced venous endothelial damage. Front Cell Dev Biol. 2021;9:642150.

[40]

ParkD, KimY, KimH, et al. Hyaluronic acid promotes angiogenesis by inducing RHAMM-TGFβreceptor interaction via CD44-PKCδ. Mol Cells. 2012;33:563-574.

[41]

SunH-J, RenX-S, XiongX-Q, et al. NLRP3 inflammasome activation contributes to VSMC phenotypic transformation and proliferation in hypertension. Cell Death Dis. 2017;8:e3074.

[42]

ChenM, WangH, ChenW, Meng G. Regulation of adaptive immunity by the NLRP3 inflammasome. Int Immunopharmacol. 2011;11:549-554.

[43]

LuH, Ashiqueali R, LinCI, et al. Histone deacetylase 3 inhibition decreases cerebral edema and protects the blood-brain barrier after stroke. Mol Neurobiol. 2023;60:235-246.

[44]

ZhangM-J, ZhaoQ-C, XiaM-X, et al. The HDAC3 inhibitor RGFP966 ameliorated ischemic brain damage by downregulating the AIM2 inflammasome. FASEB J. 2020;34:648-662.

[45]

PoliG, FabiC, BelletMM, et al. Epigenetic mechanisms of inflammasome regulation. Int J Mol Sci. 2020;21:5758.

[46]

KukrejaRC, KontosHA, HessML, Ellis EF. PGH synthase and lipoxygenase generate superoxide in the presence of NADH or NADPH. Circ Res. 1986;59:612-619.

[47]

HeR, LiuB, GengB, Li N, GengQ. The role of HDAC3 and its inhibitors in regulation of oxidative stress and chronic diseases. Cell Death Discov. 2023;9:131.

[48]

PeulenO, Gonzalez A, PeixotoP, et al. The anti-tumor effect of HDAC inhibition in a human pancreas cancer model is significantly improved by the simultaneous inhibition of cyclooxygenase 2. PLoS One. 2013;8:e75102.

[49]

KwonY, KimY, EomS, et al. MicroRNA-26a/−26b-COX-2-MIP-2 loop regulates allergic inflammation and allergic inflammation-promoted enhanced tumorigenic and metastatic potential of cancer cells. J Biol Chem. 2015;290:14245-14266.

[50]

TongX, YinL, GiardinaC. Butyrate suppresses Cox-2 activation in colon cancer cells through HDAC inhibition. Biochem Biophys Res Commun. 2004;317:463-471.

[51]

JungS-B, KimC-S, NaqviA, et al. Histone Deacetylase-3 antagonizes aspirin-stimulated endothelial nitric oxide production by reversing aspirin-induced lysine acetylation of endothelial nitric oxide synthase. Circ Res. 2010;107:877-887.

[52]

ZhangM-X, ZhangC, ShenYH, et al. Effect of 27nt small RNA on endothelial nitric-oxide synthase expression. Mol Biol Cell. 2008;19:3997-4005.

[53]

ChoiW-S, SeoY-B, ShinP-G, et al. Veratric acid inhibits iNOS expression through the regulation of PI3K activation and histone acetylation in LPS-stimulated RAW264.7 cells. Int J Mol Med. 2015;35:202-210.

[54]

de la VegaL, Grishina I, MorenoR, KrügerM, BraunT, SchmitzML. A redox-regulated SUMO/acetylation switch of HIPK2 controls the survival threshold to oxidative stress. Mol Cell. 2012;46:472-483.

[55]

BesongEE, Akhigbe TM, ObimmaJN, ObembeOO, Akhigbe RE. Acetate abates arsenic-induced male reproductive toxicity by suppressing HDAC and uric acid-driven Oxido-inflammatory NFkB/iNOS/NO response in rats. Biol Trace Elem Res. 2023;202:2672-2687.

[56]

CaoM, YangJ, WangX, et al. Sophora subprostrate polysaccharide regulates histone acetylation to inhibit inflammation in PCV2-infected murine splenic lymphocytes in vitro and in vivo. Int J Biol Macromol. 2021;191:668-678.

[57]

GiddayJM, GascheYG, CopinJ-C, et al. Leukocyte-derived matrix metalloproteinase-9 mediates blood-brain barrier breakdown and is proinflammatory after transient focal cerebral ischemia. Am J Physiol Heart Circ Physiol. 2005;289:H558-H568.

[58]

CulleyKL, HuiW, BarterMJ, et al. Class I histone deacetylase inhibition modulates metalloproteinase expression and blocks cytokine-induced cartilage degradation. Arthritis Rheum. 2013;65:1822-1830.

[59]

XiangX, DongG, ZhuJ, ZhangG, DongZ. Inhibition of HDAC3 protects against kidney cold storage/transplantation injury and allograft dysfunction. Clin Sci. 2022;136:45-60.

[60]

LiuJ, LiG, WangX, et al. Droxinostat, a histone deacetylase inhibitor, induces apoptosis in hepatocellular carcinoma cell lines via activation of the mitochondrial pathway and downregulation of FLIP. Transl Oncol. 2016;9:70-78.

[61]

Dinkova-KostovaAT, Abramov AY. The emerging role of Nrf2 in mitochondrial function. Free Radic Biol Med. 2015;88:179-188.

[62]

ZhangL, FeiM, WangH, Zhu Y. Sodium aescinate provides neuroprotection in experimental traumatic brain injury via the Nrf2-ARE pathway. Brain Res Bull. 2020;157:26-36.

[63]

WangB, ZhuX, KimY, et al. Histone deacetylase inhibition activates transcription factor Nrf2 and protects against cerebral ischemic damage. Free Radic Biol Med. 2012;52:928-936.

[64]

GuH-P, WuX-F, GongY-T, et al. RGFP966 exerts neuroprotective effect via HDAC3/Nrf2 pathway after surgical brain injury in rats. Heliyon. 2023;9:e18160.

[65]

ZhaoQ, ZhangF, YuZ, et al. HDAC3 inhibition prevents blood-brain barrier permeability through Nrf2 activation in type 2 diabetes male mice. J Neuroinflammation. 2019;16:103.

[66]

ZhangJ, XuZ, GuJ, et al. HDAC3 inhibition in diabetic mice may activate Nrf2 preventing diabetes-induced liver damage and FGF21 synthesis and secretion leading to aortic protection. Am J Physiol Endocrinol Metab. 2018;315:E150-E162.

[67]

RajendranP, Dashwood W-M, LiL, et al. Nrf2 status affects tumor growth, HDAC3 gene promoter associations, and the response to sulforaphane in the colon. Clin Epigenetics. 2015;7:102.

[68]

SunW, ZhangN, LiuB, et al. HDAC3 inhibitor RGFP966 ameliorated neuroinflammation in the Cuprizone-induced demyelinating mouse model and LPS-stimulated BV2 cells by downregulating the P2X7R/STAT3/NF-κB65/NLRP3 activation. ACS Chem Nerosci. 2022;13:2579-2598.

[69]

FalkenbergKJ, Johnstone RW. Histone deacetylases and their inhibitors in cancer, neurological diseases and immune disorders. Nat Rev Drug Discov. 2014;13:673-691.

[70]

GuL-Z, SunH, ChenJ-H. Histone deacetylases 3 deletion restrains PM2.5-induced mice lung injury by regulating NF-κB and TGF-β/Smad2/3 signaling pathways. Biomed Pharmacother. 2017;85:756-762.

[71]

GhizzoniM, HaismaHJ, MaarsinghH, Dekker FJ. Histone acetyltransferases are crucial regulators in NF-κB mediated inflammation. Drug Discov Today. 2011;16:504-511.

[72]

ZieschéE, Kettner-Buhrow D, WeberA, et al. The coactivator role of histone deacetylase 3 in IL-1-signaling involves deacetylation of p65 NF-κB. Nucleic Acids Res. 2013;41:90-109.

[73]

LeusNGJ, Zwinderman MRH, DekkerFJ. Histone deacetylase 3 (HDAC 3) as emerging drug target in NF-κB-mediated inflammation. Curr Opin Chem Biol. 2016;33:160-168.

[74]

TangJ, ChoNW, CuiG, et al. Acetylation limits 53BP1 association with damaged chromatin to promote homologous recombination. Nat Struct Mol Biol. 2013;20:317-325.

[75]

YangT, ZhangY, ChenL, et al. The potential roles of ATF family in the treatment of Alzheimer’s disease. Biomed Pharmacother. 2023;161:114544.

[76]

ChenM, LiuY, YangY, et al. Emerging roles of activating transcription factor (ATF) family members in tumourigenesis and immunity: implications in cancer immunotherapy. Genes Diseases. 2022;9:981-999.

[77]

SteerJH, Kroeger KM, AbrahamLJ, JoyceDA. Glucocorticoids suppress tumor necrosis factor-alpha expression by human monocytic THP-1 cells by suppressing transactivation through adjacent NF-kappa B and c-Jun-activating transcription factor-2 binding sites in the promoter. J Biol Chem. 2000;275:18432-18440.

[78]

SaccaniS, NatoliG. Dynamic changes in histone H3 Lys 9 methylation occurring at tightly regulated inducible inflammatory genes. Genes Dev. 2002;16:2219-2224.

[79]

MahlknechtU, WillJ, VarinA, Hoelzer D, HerbeinG. Histone deacetylase 3, a class I histone deacetylase, suppresses MAPK11-mediated activating transcription factor-2 activation and represses TNF gene expression. J Immunol. 2004;173:3979-3990.

[80]

YousefniaS, Momenzadeh S, Seyed ForootanF, GhaediK, Nasr Esfahani MH. The influence of peroxisome proliferator-activated receptor γ(PPARγ) ligands on cancer cell tumorigenicity. Gene. 2018;649:14-22.

[81]

GaoQ, WeiA, ChenF, et al. Enhancing PPARγby HDAC inhibition reduces foam cell formation and atherosclerosis in ApoE deficient mice. Pharmacol Res. 2020;160:105059.

[82]

YaoY, LiuQ, AdriantoI, et al. Histone deacetylase 3 controls lung alveolar macrophage development and homeostasis. Nat Commun. 2020;11:3822.

[83]

JiangX, YeX, GuoW, LuH, GaoZ. Inhibition of HDAC3 promotes ligand-independent PPARγactivation by protein acetylation. J Mol Endocrinol. 2014;53:191-200.

[84]

LiuX, JiangC, LiuG, et al. Sodium butyrate protects against oxidative stress in human nucleus pulposus cells via elevating PPARγ-regulated Klotho expression. Int Immunopharmacol. 2020;85:85.

[85]

LiY, LiuC, WangG, et al. HDAC3 inhibitor (BRD3308) modulates microglial pyroptosis and neuroinflammation through PPARγ/NLRP3/GSDMD to improve neurological function after intraventricular hemorrhage in mice. Neuropharmacology. 2023;237:109633.

[86]

DaskalakiMG, Tsatsanis C, KampranisSC. Histone methylation and acetylation in macrophages as a mechanism for regulation of inflammatory responses. J Cell Physiol. 2018;233:6495-6507.

[87]

PooladandaV, Thatikonda S, BaleS, et al. Nimbolide protects against endotoxin-induced acute respiratory distress syndrome by inhibiting TNF-αmediated NF-κB and HDAC-3 nuclear translocation. Cell Death Dis. 2019;10:81.

[88]

MullicanSE, GaddisCA, AlenghatT, et al. Histone deacetylase 3 is an epigenomic brake in macrophage alternative activation. Genes Dev. 2011;25:2480-2488.

[89]

AkiraS, TakedaK. Toll-like receptor signalling. Nat Rev Immunol. 2004;4:499-511.

[90]

GriffettK, HayesME, BoeckmanMP, Burris TP. The role of REV-ERB in NASH. Acta Pharmacol Sin. 2022;43:1133-1140.

[91]

WeiX, YingM, DehainiD, et al. Nanoparticle functionalization with platelet membrane enables multifactored biological targeting and detection of atherosclerosis. ACS Nano. 2018;12:109-116.

[92]

HoeksemaMA, Gijbels MJ, Van den BosscheJ, et al. Targeting macrophage histone deacetylase 3 stabilizes atherosclerotic lesions. EMBO Mol Med. 2014;6:1124-1132.

[93]

ZampetakiA, ZengL, MargaritiA, et al. Histone deacetylase 3 is critical in endothelial survival and atherosclerosis development in response to disturbed flow. Circulation. 2010;121:132-142.

[94]

WangJ, XuX, LiP, ZhangB, ZhangJ. HDAC3 protects against atherosclerosis through inhibition of inflammation via the microRNA-19b/PPARγ/NF-κB axis. Atherosclerosis. 2021;323:1-12.

[95]

KulthineeS, YanoN, ZhuangS, Wang L, ZhaoTC. Critical functions of histone deacetylases (HDACs) in modulating inflammation associated with cardiovascular diseases. Pathophysiology. 2022;29:471-485.

[96]

MathesonR, ChidaK, LuH, et al. Neuroprotective effects of selective inhibition of histone deacetylase 3 in experimental stroke. Transl Stroke Res. 2020;11:1052-1063.

[97]

DavisN, TaylorB, Abelleira-HervasL, et al. Histone deacetylase-3 regulates the expression of the amyloid precursor protein and its inhibition promotes neuroregenerative pathways in Alzheimer’s disease models. FASEB J. 2024;38:e23659.

[98]

MarinhoD, Ferreira IL, LorenzoniR, CardosoSM, Santana I, RegoAC. Reduction of class I histone deacetylases ameliorates ER-mitochondria cross-talk in Alzheimer’s disease. Aging Cell. 2023;22:e13895.

[99]

PriorR, Verschoren S, VintsK, et al. HDAC3 inhibition stimulates myelination in a CMT1A mouse model. Mol Neurobiol. 2022;59:3414-3430.

[100]

KumarV, KunduS, SinghA, Singh S. Understanding the role of histone deacetylase and their inhibitors in neurodegenerative disorders: current targets and future perspective. Curr Neuropharmacol. 2022;20:158-178.

[101]

HecklauK, Mueller S, KochSP, et al. The effects of selective inhibition of histone deacetylase 1 and 3 in Huntington’s disease mice. Front Mol Neurosci. 2021;14:616886.

[102]

SuelvesN, Kirkham-McCarthy L, LahueRS, GinésS. A selective inhibitor of histone deacetylase 3 prevents cognitive deficits and suppresses striatal CAG repeat expansions in Huntington’s disease mice. Sci Rep. 2017;7:6082.

[103]

MeleadyL, Towriss M, KimJ, et al. Histone deacetylase 3 regulates microglial function through histone deacetylation. Ciba F Symp. n.d., 2023;18:2241008.

[104]

DurhamBS, GriggR, WoodIC. Inhibition of histone deacetylase 1 or 2 reduces induced cytokine expression in microglia through a protein synthesis independent mechanism. J Neurochem. 2017;143:214-224.

[105]

WangC, ShenD, HuY, et al. Selective targeting of class I HDAC reduces microglial inflammation in the entorhinal cortex of young APP/PS1 mice. Int J Mol Sci. 2023;24:4805.

[106]

HuL, YangK, MaiX, WeiJ, MaC. Depleted HDAC3 attenuates hyperuricemia-induced renal interstitial fibrosis via miR-19b-3p/SF3B3 axis. Cell Cycle. 2022;21:450-461.

[107]

JaN, McH. Potential application of klotho in human chronic kidney disease. Bone. 2017;100:41-48.

[108]

CorralesP, Izquierdo-Lahuerta A, Medina-GómezG. Maintenance of kidney metabolic homeostasis by PPAR gamma. Int J Mol Sci. 2018;19:2063.

[109]

LinW, ZhangQ, LiuL, YinS, LiuZ, CaoW. Klotho restoration via acetylation of peroxisome proliferation-activated receptor γreduces the progression of chronic kidney disease. Kidney Int. 2017;92:669-679.

[110]

LiuL, LinW, ZhangQ, Cao W, LiuZ. TGF-βinduces miR-30d down-regulation and podocyte injury through Smad2/3 and HDAC3-associated transcriptional repression. J Mol Med (Berl). 2016;94:291-300.

[111]

LiQ, GeC, TanJ, et al. Juglanin protects against high fat diet-induced renal injury by suppressing inflammation and dyslipidemia via regulating NF-κB/HDAC3 signaling. Int Immunopharmacol. 2021;95:107340.

[112]

ZhangL, ChenF, DongJ, et al. HDAC3 aberration-incurred GPX4 suppression drives renal ferroptosis and AKI-CKD progression. Redox Biol. 2023;68:102939.

[113]

AlenghatT, Osborne LC, SaenzSA, et al. Histone deacetylase 3 coordinates commensal-bacteria-dependent intestinal homeostasis. Nature. 2013;504:153-157.

[114]

OhSK, KimD, KimK, et al. RORαis crucial for attenuated inflammatory response to maintain intestinal homeostasis. Proc Natl Acad Sci U S A. 2019;116:21140-21149.

[115]

SunZ, MillerRA, PatelRT, et al. Hepatic Hdac3 promotes gluconeogenesis by repressing lipid synthesis and sequestration. Nat Med. 2012;18:934-942.

[116]

PapazyanR, SunZ, KimYH, et al. Physiological suppression of Lipotoxic liver damage by complementary actions of HDAC3 and SCAP/SREBP. Cell Metab. 2016;24:863-874.

[117]

XiaD, ChenD, CaiT, et al. Nimbolide attenuated the inflammation in the liver of autoimmune hepatitis’s mice through regulation of HDAC3. Toxicol Appl Pharmacol. 2022;434:115795.

[118]

CheN, ZhangY, ZhangS, et al. Macrophagic HDAC3 inhibition ameliorates dextran sulfate sodium induced inflammatory bowel disease through GBP5-NLRP3 pathway. Int J Med Sci. 2024;21:1385-1398.

[119]

ZhangF, QiL, FengQ, et al. HIPK2 phosphorylates HDAC3 for NF-κB acetylation to ameliorate colitis-associated colorectal carcinoma and sepsis. Proc Natl Acad Sci U S A. 2021;118:e2021798118.

[120]

NgI, LukIY, NightingaleR, et al. Intestinal-specific Hdac3 deletion increases susceptibility to colitis and small intestinal tumor development in mice fed a high-fat diet. Am J Physiol Gastrointest Liver Physiol. 2023;325:G508-G517.

[121]

SathishkumarC, PrabuP, BalakumarM, et al. Augmentation of histone deacetylase 3 (HDAC3) epigenetic signature at the interface of proinflammation and insulin resistance in patients with type 2 diabetes. Clin Epigenetics. 2016;8:125.

[122]

FeliceC, LewisA, ArmuzziA, Lindsay JO, SilverA. Review article: selective histone deacetylase isoforms as potential therapeutic targets in inflammatory bowel diseases. Aliment Pharmacol Ther. 2015;41:26-38.

[123]

FuB, ShenJ, ZouX, et al. Matrix stiffening promotes chondrocyte senescence and the osteoarthritis development through downregulating HDAC3. Bone Res. 2024;12:32.

[124]

ChenH, FuX, WuX, et al. Gut microbial metabolite targets HDAC3-FOXK1-interferon axis in fibroblast-like synoviocytes to ameliorate rheumatoid arthritis. Bone Res. 2024;12:31.

[125]

YaoF, ZhaoY, YuQ, et al. Extracellular CIRP induces abnormal activation of fibroblast-like synoviocytes from patients with RA via the TLR4-mediated HDAC3 pathways. Int Immunopharmacol. 2024;128:111525.

[126]

YangQ-B, ZhangM-Y, YangL, Wang J, MiQ-S, ZhouJ-G. Deficiency of histone deacetylases 3 in macrophage alleviates monosodium urate crystals-induced gouty inflammation in mice. Arthritis Res Ther. 2024;26:96.

[127]

JiangY, WangL. Role of histone deacetylase 3 in ankylosing spondylitis via negative feedback loop with microRNA-130a and enhancement of tumor necrosis factor-1αexpression in peripheral blood mononuclear cells. Mol Med Rep. 2016;13:35-40.

RIGHTS & PERMISSIONS

2024 The Author(s). Cell Proliferation published by Beijing Institute for Stem Cell and Regenerative Medicine and John Wiley & Sons Ltd.

AI Summary AI Mindmap
PDF

179

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/