Anti-inflammatory role of fenofibrate in treating diseases

Authors

  • Lv Jin Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
  • Hu Hua Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China; Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
  • Yong Ji Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China; Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
  • Zhanjun Jia Department of Nephrology, Children’s Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing 210008, China; Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
  • Mingqi Peng Nursing Department, Children’s Hospital of Nanjing Medical University, Nanjing, China
  • Songming Huang Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China; Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China

DOI:

https://doi.org/10.17305/bb.2022.8534

Keywords:

Fenofibrate, inflammation, mechanism, PPAR-α, diseases, therapy

Abstract

Inflammation contributes to the pathogenesis of several diseases. Fenofibrate, known as a peroxisome proliferator-activated receptor - α (PPAR-α) agonist, is a classic drug for treating hyperlipidemia. In addition to its lipid-lowering effect, fenofibrate has also been reported to exert anti-inflammatory effects with complicated underlying mechanisms of action. In general, the anti-inflammatory effect of fenofibrate is secondary to its lipid-lowering effect, especially for the inflammation caused by hyperlipidemia in the circulatory system. Some anti-inflammatory actions may also come from its regulatory effects on intracellular lipid metabolism by activating PPAR-α. In addition, some roles in anti-inflammation might be mediated by its direct regulation of inflammatory signaling pathways. In order to understand anti-inflammatory activities and the underlying mechanisms of fenofibrate action in disease better, we herein reviewed and discussed the anti-inflammatory roles and its subserving mechanisms in various diseases of different organ systems. Thus, this review offers insights into the optimal use of fenofibrate in the clinical setting.

Downloads

Download data is not yet available.
Anti-inflammatory role of fenofibrate in treating diseases

Downloads

Additional Files

Published

2023-05-01

How to Cite

1.
Jin L, Hua H, Ji Y, Jia Z, Peng M, Huang S. Anti-inflammatory role of fenofibrate in treating diseases. Biomol Biomed [Internet]. 2023May1 [cited 2023Jun.6];23(3):376–391. Available from: https://www.bjbms.org/ojs/index.php/bjbms/article/view/8534

Issue

Section

Reviews

Introduction

Fenofibrate is a peroxisome proliferator-activated receptor-α (PPAR-α) agonist approved by the Food and Drug Administration (FDA) to routinely treat hyperlipidemia with elevated triglycerides (TGs), or mixed hyperlipidemia with elevated TGs and reduced high-density lipoprotein cholesterol (HDL-C) levels [1]. In addition to lowering blood lipids, fenofibrate can significantly reduce the risk of major cardiovascular events [2]. Researchers have demonstrated that fenofibrate exerts protective effects against diabetes and diabetes-associated pathologies [3]. Fenofibrate safeguards epithelial function and reduces vascular complications, including diabetic retinopathy (DR), nephropathy, and cardiopathy [4–6], in addition to being used to rescue diabetes-related organ impairment [7]. Fenofibrate also inhibits the development of liver diseases, including non-alcoholic fatty liver disease (NAFLD), steatohepatitis, hepatitis C virus (HCV), and cholestatic hepatitis [8–11]. Recent evidence shows that fenofibrate can suppress many types of human cancers [12], and even drug-induced disorders [13, 14]. All these therapeutic effects of fenofibrate might arise from its anti-inflammatory properties.

Although inflammation is a defensive response of the body to stimulation, continued or excessive inflammation may result in adverse events [15]. Inflammation has been demonstrated to promote the progression of several diseases, such as obesity, diabetes [16], cancer [17], cardiovascular disease [18], eye disorders [19], arthritis [20], neuroinflammation [21], autoimmune diseases [22], and inflammatory bowel disease [23]. Furthermore, several studies have revealed that fenofibrate exhibits a robust anti-inflammatory effect on diseases of various systems. This work presents the latest research on anti-inflammatory and other similar effects of fenofibrate.

Indirect and direct anti-inflammatory effects of fenofibrate

The anti-inflammatory effects and mechanisms of fenofibrate vary across disease types. Roughly, its anti-inflammatory effects can be divided into indirect and direct. Lipid metabolism is disrupted in an inflammatory environment in vivo [24], and fenofibrate may repress inflammation to maintain a normal metabolic status. Fenofibrate upregulates the expression of HDL and downregulates that of low-density lipoprotein (LDL) [1]. The expression of HDL apolipoprotein A-I (ApoA-I) can be upregulated by fenofibrate to inhibit atherosclerotic progression [25]. By reducing cholesterol accumulation in the liver, fenofibrate attenuates hepatic inflammation and inhibits the progression of non-alcoholic steatohepatitis (NASH) [26]. Very-long-chain sphingolipid can also be raised by fenofibrate to relieve inflammation [3, 27]. These studies prove that fenofibrate can protect against inflammation in an indirect manner.

Fenofibrate also has a direct anti-inflammatory effect. Independent of its lipid-lowering effects, fenofibrate counters inflammation primarily by activating PPAR-α which plays a key role in both lipid metabolism and inflammation. Upon PPAR-α activation, fenofibrate suppresses inflammatory pathways involving nuclear factor kappa-B (NF-κB) [28], sirtuin 1 (SIRT1) [29], toll-like receptor 4 (TLR4) [30], adenosine mitogen-activated protein kinase (AMPK) [31], or interleukin IL-1 or IL-6 [32]. Fenofibrate also directly inhibits the expression of inflammation-related genes in inflammatory pathways. In addition, the attenuation of mitochondrial disfunction [33] and the differentiation of T helper cells 17 (Th17) also assist in combating inflammation [34]. In summary, fenofibrate exerts its anti-inflammatory effects either directly or indirectly.

Anti-inflammatory mechanism of fenofibrate

Fenofibrate has been reported to have anti-inflammatory effects in several diseases. In this part, the mechanism of how fenofibrate works against inflammation will be introduced in various diseases of different organ systems.

Endocrine system diseases

Obesity

Fenofibrate can attenuate the inflammation involved in obesity. A positive correlation between inflammation or reactive oxygen species (ROS) production and peroxisomal dysfunction has been observed in mice with high-fat diet (HFD)-induced obesity. Relying on PPAR-α to repair peroxisomal function, fenofibrate inhibited inflammation in these obese mice [35]. Moreover, in 3T3-L1 adipocytes, fenofibrate increased SIRT1 expression and suppressed the NF-κB pathway by activating PPAR-α and boosting AMPK phosphorylation. Subsequently, fenofibrate-inhibited tumor necrosis factor-α (TNF-α)-induced CD40 expression [29]. Fenofibrate-upregulated SIRT1 expression is associated with a diminution in markers of inflammation, including high-sensitivity C reactive protein (hs-CRP), IL-6, and fetuin-A, in obese patients with or without type 2 diabetes [36]. In addition, fenofibrate increased the expression of adiponectin, which was secreted by adipocytes and able to mediate the interrelationship among adiposity, insulin resistance, and inflammation [37, 38]. At present, fenofibrate is not the main drug to treat obesity, but in some obese patients with hyperlipidemia, fenofibrate may achieve evident effects.

In general, fenofibrate can inhibit the inflammation in obesity, a mechanism that often involves the activation of PPAR-α.

Diabetes

In diabetes, the abnormal glucose metabolism may cause systematic inflammation. In patients with pre-diabetes, the plasma markers of inflammation and monocytic secretory function increase significantly [39]. Fenofibrate shows favorable effects in reducing the cytokine-provoked release of TNF-α, IL-1, IL-6, monocyte chemoattractant protein 1 (MCP-1), and plasma hs-CRP from monocytes in patients with pre-diabetes [39, 40]. Further, the anti-inflammatory effects of fenofibrate improve diabetic patients’ sensitivity to insulin [39]. In patients with metabolic syndrome, the reduction of systemic inflammation markers by fenofibrate is more dependent on the activation of PPAR—than on the regulation of lipid and glucose metabolism and insulin sensitivity [41].

Fenofibrate enhances anti-inflammatory processes in type 1 and type 2 diabetes patients [27, 42]. In type 2 diabetes, β-cell function is damaged by inflammation. A recent study has revealed that fenofibrate ameliorates lipotoxicity-induced β-cell disfunction and apoptosis in lipoprotein lipase (LPL)+/– mice and palmitate (PA)-treated stable mouse insulinoma 6 (MIN6) cells, which is achieved by the inhibition of NF-κB/MIF (macrophage-inhibitory factor) inflammatory pathway [28]. Abnormal metabolism of sphingolipids in type 1 diabetes can also lead to β-cell dysfunction. Fenofibrate upregulates the expression of very-long-chain sphingolipids in non-obese diabetic (NOD) mice and remodels pancreatic lipidome into a more anti-inflammatory state [3, 27].

In type 2 diabetes patients with hypertriglyceridemia, fenofibrate decreased the levels of reduced upon activation, normal T cell expressed and secreted (RANTES) and inhibited inflammatory responses [43]. In addition, both fenofibrate and its phase-I bio-transformed metabolite fenofibric acid weakened COX-2 enzymatic activity, thus generating anti-inflammatory effects both in vivo and in vitro. Moreover, fenofibric acid exhibited potent time-dependent anti-inflammatory effects [44].

Interestingly, fenofibrate reduced systematic inflammation in diabetes by protecting β-cell functions. The inhibition of the COX-2 enzyme and the decrease in RANTES cooperated to repress inflammatory responses. Whether insulin sensitivity is linked to the anti-inflammatory effects of fenofibrate remains to be verified by further studies. At present, fenofibrate is not routinely used to treat diabetes in clinic. Whether it can be combined with other drugs to treat diabetes or hyperlipidemic diabetes patients deserves more in-depth research.

In summary, fenofibrate can significantly improve diabetes-related inflammation through different mechanisms.

Hyperlipidemia

Abnormal blood lipid metabolism may lead to hyperlipidemia. Hyperlipidemia induces an elevation in inflammatory factors and subsequently a chronic inflammation state. Fenofibrate can reduce the plasma concentrations of inflammatory markers, including (hs-CRP) and fibrinogen, in hyperlipidemic patients [45]. This reduction may involve the upregulation of adiponectin by fenofibrate [46]. In hypercholesterolemic rabbits, fenofibrate lowers the levels of plasma acute-phase proteins, a process associated with the reduction in TNF-α in adipocytes [47]. In addition, fenofibrate regulates DNA methylation to control inflammatory response. Downregulation of hs-CRP, IL-2, and IL-6 is associated with multiple cytosine-guanine (CpG) sites [48].

Fenofibrate improved both HFD-induced insulin resistance in skeletal muscle and palmitic acid-induced insulin resistance in myotube cells, thus reduced ER stress-induced inflammation via inhibiting TLR4/NF-κB pathway [30]. Moreover, by activating PPAR-α, fenofibrate suppresses the NF-κB P65 pathway and renders anti-inflammatory effects in patients with acute hypertriglyceridemic pancreatitis [49]. Fenofibrate is commonly used for hyperlipidemia in clinic.

In summary, fenofibrate can significantly curb hyperlipidemia-related inflammation. This effect is partially linked to fenofibrate-induced reduction of lipids in the serum (Table 1).

Table 1: Anti-inflammatory targets and mechanisms of fenofibrate in endocrine system diseases
Diseases Cells/Models Targets and mechanisms Implications References
Obesity Mouse PPAR-α↑-peroxisomal dysfunction Reduce inflammation [35]
3T3-L1 adipocytes PPAR-α↑-AMPK phosphorylation-SIRT1-NF-κB-TNF-α-induced CD40 Reduce proinflammatory factor CD40 [29]
Human SIRT1 Regulate markers of inflammation [36]
Mouse, 3T3-L1 adipocytes Adiponectin Reduce inflammation [37, 38]
Diabetes Human Insulin sensitivity Have early anti-inflammatory effect [39]
Human PPAR-α↑ Reduce inflammatory responses [41]
LPL+/– mouse and PA-treated MIN6 cells NF-κB/MIF inflammatory pathway Ameliorate lipotoxicity-induced β-cell dysfunction and apoptosis [28]
NOD mouse Very-long-chain sphingolipid Alter the pancreatic lipidome to a more anti-inflammatory state [3, 27]
Human RANTES Restrain the inflammatory responses [43]
Rat COX-2 enzyme Have anti-inflammatory effects both in vivo and in vitro [44]
Dyslipidemia Human Adiponectin Reduce inflammation [46]
Rabbit TNF-α in adipocytes-Acute-phase proteins Reduce inflammatory responses [47]
CD4+ T cells DNA methylation Reduce inflammatory responses [48]
Rat, Differentiated C2C12 myotubes TLR4/NF-κB pathway Reduce ER stress-induced inflammation [30]
Human PPAR-α↑-NF-κB P65 Reduce inflammatory responses [49]

PRAR-α: Peroxisome proliferator-activated receptors; AMPK: Adenosine monophosphate activated protein kinase; SIRT1: Sirtuin 1; NF-κB: Nuclear factor κB; TNF-α: Tumor necrosis factor α; MIF: Macrophage migration inhibitory factor; RANTES: Regulated upon activation, normal T cell expressed and secreted; COX-2: Cyclooxygenase-2; TLR4: Toll-like receptor-4; LPL: Lipoprotein lipase; NOD: Non-obese diabetic.

Circulatory system diseases

Atherosclerosis

Inflammation is closely associated with atherosclerosis and its complications [50]. Fenofibrate has demonstrated the ability to effectively attenuate inflammation in atherosclerosis [51], which may be associated with its lipid-lowering effects [52].

Fenofibrate reduces the levels of CRP and the CRP-induced cytokines, including MCP-1 [53], as well as the expression of CD40 and CD40L [54]. The downregulation of the CD40–CD40L-signaling pathway inactivates matrix metalloproteinase 2 (MMP-2) and MMP-9 in human umbilical vein endothelial cells (HUVECs) and reduces inflammatory responses in atherosclerosis [54]. Fenofibrate weakens the proinflammatory effects of cytokines in vascular cells, thus preventing the progression of atherosclerosis [53]. In ApoE*3Leiden transgenic mice fed with high levels of cholesterol, investigators have discerned a reduction in the levels of proinflammatory chemokine MCP-1, intercellular cell adhesion molecule 1 (ICAM-1), and monocyte/macrophage differentiation factor granulocyte-macrophage colony-stimulating factor (GM-CSF) after fenofibrate treatment, indicating that fenofibrate can inhibit the adhesion, recruitment, and maturation of monocytes/macrophages and the inflammation of vascular components. The inhibition on p65-NF-κB signaling is possibly associated with a reduction in monocyte adhesion induced by fenofibrate [55].

Fenofibrate can activate PPAR-α to inhibit atherosclerotic progression. Inflammatory cytokines induce an increase in serum amyloid (SAA) and a decrease in ApoA-I and paraoxonase 1 (PON-1) mRNA expression by suppressing PPAR-α activity. These effects can be reversed by fenofibrate in a PPAR-α-dependent manner, thereby relieving atherosclerosis [25]. By activating PPAR-α, fenofibrate also induces heme oxygenase-1 (HO-1, an anti-inflammatory enzyme) expression in human vascular endothelial and smooth muscle cells [56]. Platelet-activating factor (PAF) is a proinflammatory molecule intimately involved in the progression of atherosclerosis [57]. Fenofibrate decreases the level of PAF receptors in human monocytes and macrophages in a PPAR-α-dependent fashion [58], a process mediated by β-defensin 1. The activation of PPAR-α upregulates the secretion of β-defensin 1 and suppresses TLR4. Through PPAR-α/β-defensin1/TLR4 pathways, fenofibrate inhibits the activation of macrophages induced by lipopolysaccharide (LPS) [59]. Fenofibrate also suppresses TLR4 during angiotensin (Ang) II-induced inflammation in vascular smooth muscle cells (VSMCs). In addition, fenofibrate reduces inflammatory responses of VSMCs by interfering with the TLR4-dependent-signaling pathway (TLR4/IP-10/PKC/NF-kB) [60]. Moreover, the inhibition of another interferon-β (TRIF)-dependent-signaling pathway (TLR4/TRIF/IRF3/IP-10) by fenofibrate was found to assist in reducing inflammatory responses in LPS-induced VSMCs [61]. PPAR-α activation induced by fenofibrate leads to the high expression of SIRT1 and SIRT1-mediated deacetylation of forkhead box O1 (FoxO1). PPAR-α, SIRT1, and FoxO1 co-work to inhibit cellular apoptosis induced by TNF-α in vascular adventitial fibroblasts (VAFs), indicating that fenofibrate can regulate atherosclerosis-associated inflammation to counterstrike cellular apoptosis [62].

The aforementioned results collectively reveal that fenofibrate can attenuate inflammation to block the progression of atherosclerosis. These anti-inflammatory effects are associated with its induction on PPAR-α and subsequent inhibition on inflammatory pathways, rather than fenofibrate’s lipid-lowering activities. However, whether fenofibrate can be used for clinical management of atherosclerosis-related inflammation needs human studies or clinical trials.

Abdominal aortic aneurysm

Abdominal aortic aneurysm (AAA) is a serious condition associated with aortic inflammation. Fenofibrate induces the activation of the sphingosine 1 phosphate (S1P) pathway and the nitric oxide (NO) pathway in mice with Ang II-induced AAA, thereby reducing aortic inflammation [63]. This mechanism also acts to inhibit the progression of atherosclerosis.

Diabetic retinopathy

DR is a microvascular disease associated with inflammation of blood vessels. Fenofibrate reduces the inflammation in DR via both PPAR-α-dependent and -independent mechanisms [31, 64].

The overexpression of PPAR-α in the retina mitigates retinal vascular leakage and retinal inflammation induced by diabetes [65]. By activating PPAR-α, fenofibrate induced direct effects on inhibiting proinflammatory MCP-1, ICAM-1, and transcription factor NF-kB [66]. Studies have revealed that the inhibition on NF-κB reduced the production of the inflammatory chemokines MCP-1, fractalkine (FKN), and ICAM-1, as well as the oxidative products in DR [67, 68]. Enright et al. [69] have demonstrated that fenofibrate activates PPAR-α in the liver, but not in the retina, to modulate circulating cytokines, growth factors, and/or lipids, suggesting that fenofibrate indirectly affects the retina by inducing the expression of genes related to the local inflammatory response.

Fenofibrate also directly activates AMPK in human glomerular microvascular endothelial cells (HGMECs), thus elevating NO production and reducing inflammation [31]. Angiopoietin-like 3 (ANGPTL3) and its accompanying inflammation were closely linked to the pathogenesis of DR [70]. Moreover, fenofibrate attenuates oxidative stress by upregulating nuclear factor erythroid-2-related factor 2 (Nrf2) signaling in Müller cells. Fenofibrate also downregulates the NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome to reduced caspase-1 and pro-IL-1β, thus generating anti-inflammatory effects in DR. However, whether PPAR-α plays a role in this mechanism remains unknown [71]. In addition, adiponectin was demonstrated to mediate proinflammatory effects in DR. Fenofibrate generated significant anti-inflammatory effects and inhibited the progression of DR by reducing the expression of adiponectin and its receptors in DR [72]. Fenofibric acid, a metabolite of fenofibrate, inhibits the expression of COX-2 and inflammatory factors in DR [73]. At present, fenofibrate is not used for diabetes in clinic. In general, fenofibrate employs its anti-inflammatory effects to protect from DR.

Cardiac dysfunction

Inflammation is involved in cardiac disfunction induced by primary diseases. Suppressing inflammation slows down the progression of heart diseases, such as heart failure [74] and myocardial infarction [75].

In cardiac dysfunction induced by high glucose, fenofibrate represses inflammatory responses by upregulating fibroblast growth factor 21 (FGF21) and promoting SIRT1-mediated autophagy in heart tissues [76]. The function of high-mobility group box 1 (HMGB1) protein depends upon its location, and extracellular HMGB1 works as a delayed mediator of proinflammatory cytokines in the initiation and amplification of inflammatory responses, whereas nuclear HMGB1 prevents cardiac hypertrophy and heart failure. Indirectly, fenofibrate activates PPAR-α to modulate both the expression and location of HMGB1, thus arousing anti-inflammatory response against cardiac hypertrophy [77]. In addition, fenofibrate attenuates doxorubicin (DOX)-induced cardiac inflammation in mice by activating the endothelial NOS/EPC pathway [78] and quelling the NF-κB pathway [79]. Fenofibrate also stimulates PPAR-α to relieve myocardial inflammation induced by Ang II. The expression level of NF-kB is downregulated and those of vascular cell adhesion molecule-1 (VCAM-1), platelet endothelial cell adhesion molecule (PECAM), and ICAM-1 are upregulated after fenofibrate intervention [80]. Fenofibrate blunts myocarditis by increasing the expression of the anti-inflammatory cytokine IL-10 [81].

In patients with chronic heart failure (CHF), fenofibrate directly blocks the interaction between monocytes and human aortic endothelial cells (HAECs) as TNF-α is activated. Fenofibrate also reduces VCAM-1 and ICAM-1 expression. It appears that fenofibrate decreases the expression level of cell adhesion molecules to inhibit inflammation in vascular tissues of patients with CHF [82]. Furthermore, fenofibrate hinders the infiltration of macrophages and T lymphocytes into the left ventricle of the heart. These anti-inflammatory effects contribute to the prevention of heart failure [83]. These studies collectively show that fenofibrate counters inflammation to safeguard cardiac function; however, whether it can be applied to clinical practice needs further exploration (Table 2).

Table 2: Anti-inflammatory targets and mechanisms underlying fenofibrate action in circulatory system diseases
Diseases Cells/Models Targets and mechanisms Implications References
Atherosclerosis HUVECs MCP-1 Reduce the inflammation of atherosclerosis [53]
HUVECs CD40-CD40L-MMP-2, MMP-9 Reduce the inflammation of atherosclerosis [54]
Mouse p65-NF-κB Reduce monocyte adhesion [55]
Murine hepatoma cells PPAR-α↑- SAA, apoA-I and PON-1 Reverse the inflammatory cytokine effects [25]
Human vascular endothelial and smooth muscle cells PPAR-α↑-HO-1 Reduce the inflammation of atherosclerosis [56]
Human monocytes and macrophages PPAR-α↑, PAF receptor gene Reduce proinflammatory molecule and inhibit progression of atherosclerosis [58]
The murine macrophage-like cell line J774 PPAR-α↑-β-defensin1-TLR4 Inhibit the inflammatory activation of macrophages [59]
Rat, VSMCs TLR4/IP-10/PKC/NF-κB pathway Reduce the inflammation of atherosclerosis [60]
VAMCs TLR4/TRIF/IRF3/IP-10 pathway Reduce the inflammation of atherosclerosis [61]
Mouse, rat VAFs PPAR-α↑-SIRT1-deacetylation of FoxO1s Reduce cellular apoptosis and regulate the inflammation associated with atherosclerosis [62]
Abdominal aortic aneurysm Mouse S1P pathway-NO pathway Reduce aortic inflammation [63]
Diabetic retinopathy Rat PPAR-α↑ Mitigate retinal vascular leakage and retinal inflammation [65]
Rat, human retinal pigment epithelium cells PPAR-α↑-NF-κB Reduce the production of the inflammatory chemokines and inhibit oxidative products in DR [67, 68]
Mouse PPAR-α in the liver Modulate circulating cytokines, growth factors, and/or lipids [69]
HGMECs AMPK Elevate NO production and reduce inflammation [31]
Rat, HRMECs ANGPTL3 pathway Reduce its accompanying inflammation [70]
Mouse Nrf2 expression, NLRP3 inflammasome activation Attenuate oxidative stress, reduced caspase-1 and pro-IL-1β [71]
Rat, RAW264.7 cells, RGC-5 cells Adiponectin and its receptors Have anti-inflammatory effects [72]
RRECs COX-2 Have anti-inflammatory effects [73]
Cardiac dysfunction Mouse FGF21-SIRT1-mediated autophagy Reduce cardiac inflammatory responses [76]
Rat cardiomyocytes, mouse HMGB1, Extracellular HMGB1 Facilitate anti-inflammatory responses [77]
Mouse eNOS/EPC pathway Attenuate DOX-induced cardiac inflammation [78]
Mouse NF-κB pathway Attenuate DOX-induced cardiac inflammation [79]
Rat PPAR-α↑ Have anti-inflammatory effects [80]
Rat IL-10 Inhibit the progression of myocarditis [81]
PBMCs, HAECs VCAM-1 and ICAM-1- monocyte binding HAECs Attenuate the inflammation seen in vascular tissues [82]
Rat Infiltration of macrophages and T lymphocytes Abatement of heart failure progression [83]

HUVEC: Human umbilical vein endothelial cell; MCP-1: Monocyte chemoattractant protein 1; MMP-2: Matrix metalloproteinase 2; MMP-9 Matrix metalloproteinase 9; NF-κB: Nuclear factor κ-B; PPAR-α: Peroxisome proliferator-activated receptor-α; SAA: Serum amyloid; ApoA-I: Apolipoprotein A-I; PON-1: Paraoxonase 1; HO-1: Heme oxygenase-1; PAF: Platelet-activating factor; TLR4: Toll-like receptor 4; VSMC: Vascular smooth muscle cell; SIRT1: Sirtuin 1; FoxO1: Forkhead box O1; S1P: Sphingosine 1 phosphate; NO: Nitric oxide; AMPK: Adenosine mitogen-activated protein kinase; HGMEC: Human glomerular microvascular endothelial cell; ANGPTL3: Angiopoietin-like 3; Nrf2: Nuclear factor erythroid-2-related factor 2; NLRP3: NOD-like receptor thermal protein domain associated protein 3; FGF21: Fibroblast growth factor 21; HMGB1: High-mobility group box 1; DOX: Doxorubicin; IL: Interleukin; VCAM-1: Vascular cell adhesion molecule-1; ICAM-1: Intercellular cell adhesion molecule 1; HAEC: Human aortic endothelial cell.

Digestive system diseases

Liver diseases

The liver is susceptible to inflammation. Fenofibrate can treat inflammation in NASH, cholestatic liver injury, liver ischemia–reperfusion (I/R) injury, etc.

NASH is a chronic liver disease that is characterized by hepatic steatosis, lobular inflammation, hepatocyte ballooning, and fibrosis. The liver is the site where multiple metabolic pathways occur. Cholesterol is primarily metabolized in the liver and the accumulation of cholesterol in the liver could lead to NASH. Fenofibrate can reverse the downregulation of sterol-regulatory element binding proteins (SREBPs), liver X receptors (LXRs), and their target genes, thereafter contributing to cholesterol catabolism [84]. In this manner, fenofibrate can reduce the cholesterol accumulation in the liver and inhibit inflammation in NASH [26]. The downregulation of PPAR-α is a feature of NASH, and PPAR-α activation prevents the accumulation of lipid and the secretion of modulating inflammatory chemokines [85]. Fenofibrate activates PPAR-α and its target genes to prevent NASH in a murine model of 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)-induced steatohepatitis-like hepatocellular damage [86] and a mouse model of HFD-fed NASH [87]. In addition, fenofibrate exerted a direct anti-inflammation effect in NASH by regulating NF-kB and its target inflammatory genes [9]. These results suggest that fenofibrate induces anti-inflammatory effects through directly regulating lipid metabolism and inflammatory pathways.

Sedimentation of bile acids in the liver can lead to liver toxicity followed by inflammatory responses. Relying on PPAR-α, fenofibrate reduces the production of bile acids and keeps them nontoxic in patients with primary biliary cholangitis (PBC) or primary sclerosing cholangitis (PSC). The proinflammatory cytokines induced by bile acids are then reduced after fenofibrate treatment [88]. In rats with bile duct ligation (BDL), fenofibrate effectively downregulates serum cytokine to ameliorating hepatic inflammation. However, proliferation of the bile duct becomes more marked after fenofibrate treatment [89], but whether this effect leads to side effects in patients remains unknown. In addition, fenofibrate prevents cholestatic liver injury induced by α-naphthyl isothiocyanate (ANIT) by upregulating the expression of genes associated with fatty acid β-oxidation (β-FAO), including carnitine palmitoyltransferase 1b (Cpt1b), carnitine palmitoyltransferase 2 (Cpt2), and medium-chain acyl-CoA dehydrogenase (Mcad) [90]. The anti-inflammatory effects of fenofibrate also demonstrated to recover liver dysfunction in liver I/R injury. Besides, it restores serum levels of alanine aminotransferase (ALT) and TNF-α and suppresses oxidative stress, necrosis, and apoptosis in ischemic liver tissues [91].

Fenofibrate shows evident efficacy in treating multiple typical inflammatory hepatocellular adenomas. Given that inflammatory hepatocellular adenoma is characterized by the activation of the IL-6/JAK/STAT pathway, we suspect that this efficacy may be associated with the suppression of IL-6 [92].

Adiponectin receptors are closely linked with hepatic inflammation. In a model of HCV-induced steatosis, fenofibrate rescues the expression of pAMPK and adiponectin receptor 2 (AdipoR2) by reducing ER stress and inflammatory proteins [93]. In addition to liver inflammation, fenofibrate was reported to reduce systemic inflammation in the systemic acute-phase response in mice by activating PPAR-α and reducing the expression of IL-6 [32, 94]. In conclusion, fenofibrate can significantly relieve liver disease-related inflammation, but its clinical application lacks sufficient evidence.

Intestinal inflammation

Enteritis and colitis, usually caused by bacteria, viruses, fungi, and parasites, involve the abnormal Th17 activities in the intestines. Fenofibrate suppresses the differentiation of Th17 cells independent of PPAR-α [95]. In T and B lymphocytes and colonic epithelial cells, fenofibrate decreases the secretion of interferon-γ (IFN-γ) and IL-17 from Th1 and Th17 cells in colitis by activating PPAR-α. Moreover, fenofibrate represses the expression of chemokines C-X-C motif chemokine ligand 10 (CXCL10), MCP-1, and macrophage inflammatory protein 3 (MIP-3) in intestinal epithelial cells. These effects then allow a dramatic reduction in infiltrating inflammatory cells and lymphocytes in colitis [96]. However, whether fenofibrate can be applied to clinical treatment needs further exploration. In summary, fenofibrate protects against intestinal inflammation through the suppression of lymphocytes in the intestine (Table 3).

Table 3: Anti-inflammatory targets and mechanisms of fenofibrate action in digestive system diseases
Diseases Cells/Models Targets and mechanisms Implications References
NASH Mouse SREBP and LXRs and their target genes Inhibit the accumulation of cholesterol in the liver [26, 84]
Mouse PPAR-α↑ Reduce lipid accumulation and the secretion of modulating inflammatory chemokines [86], [87]
Mouse NF-κB Have anti-inflammatory effects [9]
Cholestatic liver diseases Human PPAR-α↑ Affect bile acids production, make the components of bile acids healthier [88]
Rat PPAR-α↑ Reduce serum cytokines, improve hepatic inflammation [89]
Mouse PPAR-α↑-Cpt1b, Cpt2, Mcad-β-FAO Reduce inflammation [90]
Liver ischemia/reperfusion injury Rat TNF-α↓ Reduce oxidative stress, necrosis, and apoptosis [91]
Hepatocellular adenomas Human IL-6/JAK/STAT pathway Reduce inflammation [92]
HCV-induced steatosis Huh7 cells, Huh.8 cells ER stress and inflammatory proteins- pAMPK and AdipoR2; Reduce inflammation [93]
Acute-phase response Mouse PPAR-α↑-IL-1- IL-6-ARP genes Reduce systemic inflammation in the systemic APR [32]
Mouse PPAR-α↑-IL-6 signaling pathway Reduce systemic inflammation in the systemic APR [94]
Intestinal inflammation Mouse Differentiation of Th17 Reduce inflammation [95]
Mouse, HT-29 Colorectal Cancer Cells Th1,Th17- IFN-γ and IL-17; CXCL10, CCL2, CCL20 Reduce inflammatory cell infiltration and lymphocytes in colitis [96]

NASH: Non-alcoholic steatohepatitis; SREBP: Sterol-regulatory element binding protein; LXR: Liver X receptor; PPAR-α: Peroxisome proliferator-activated receptor-α; NF-κB: Nuclear factor κ-B; Cpt1b: Carnitine palmitoyltransferase 1b; TNF-α: Tumor necrosis factor-α; IL: Interleukin; HCV: Hepatitis C virus; AdipoR2: Adiponectin receptor 2; IFN-γ: Interferon-γ; CXCL10: C-X-C motif chemokine ligand 10; APR: Acute phase response.

Urinary system diseases

The anti-inflammatory effects of fenofibrate in urinary system diseases have been evaluated in studies focusing on kidney diseases, including diabetic nephropathy (DN), hypertensive renal injury, age-related kidney diseases, renal I/R injury, and lipotoxicity-induced renal injury.

DN, a most serious complication of diabetes, involves inflammatory processes. Researchers in the field showed that fenofibrate reduces inflammatory responses in DN by the inhibition of the NF-kB and transforming growth factor-β1 (TGF-β1)/SMAD family member 3 (Smad3)-signaling pathways independent of PPAR-α [97, 98]. Fenofibrate also exhibits protective effects against DN by upregulating the expression of (FGF21) which then induces the activation of the Akt2/GSK-3β/Fyn/Nrf2 and the AMPK pathways, both of which prevent renal inflammation [99, 100]. PPAR-α activation induced by fenofibrate is known to inhibit the inflammatory responses in DN by decreasing the expression of IL-18 [101] and the level of leukocytes adhering to mesangial cells [102].

In hypertensive renal injury, fenofibrate induces PPAR-α activation to reduce renal inflammatory responses in salt-loaded spontaneously hypertensive stroke-prone rats by protecting mitochondrial function [103]. In addition, fenofibrate can reduce oxidative stress and phosphorylation of MAPK to inhibit inflammation in hypertensive renal injury [104].

The kidney is sensitive to aging-related inflammation. Fenofibrate activates PPAR-α to evoke activated AMPK-SIRT1 signaling, thus attenuating age-related renal inflammation [105].

In renal I/R injury, fenofibrate plays a protective role in reducing inflammation via PPAR-α activation [106] that upregulates PI3K/Akt signaling and reduces the secretion of proinflammatory cytokines induced by IR injury [107].

Renal lipotoxicity occurs when the kidneys undergo excessive lipid accumulation. Fenofibrate inhibits this pathogenic mechanism in rats by upregulating the PPAR-α–FoxO3a–PGC-1α signal-transduction pathway [108, 109]. In addition, fenofibrate counteracts renal inflammation induced by lipotoxicity in HFD-fed mice by activating AMPK and autophagy, apparently independent of PPAR-α [110]. At present, the potential of fenofibrate for treating nephritic inflammation has not been clinically assessed. In summary, fenofibrate can significantly be used to diminish inflammation associated with nephropathy (Table 4).

Table 4: Anti-inflammatory targets and mechanisms of fenofibrate action in urinary system diseases
Diseases Cells/Models Targets and mechanisms Implications References
Diabetic nephropathy Rat NF-κB pathway, TGF-β1/Smad3 signaling pathways Reduce inflammation [97, 98]
Rat FGF21-Akt2/GSK-3β/Fyn/Nrf2 pathway, AMPK pathway Prevent renal inflammation [99, 100]
Rat PPAR-α↑-IL-18 Inhibit inflammatory responses [101]
Mouse PPAR-α↑-leukocytes adherent to mesangial cells Inhibit inflammatory responses [102]
Hypertensive renal injury Rat PPAR-α↑ Reduce renal inflammatory responses [103]
Rat Mitochondrial function Reduce renal inflammatory responses [33]
Rat Oxidative stress, phosphorylation of MAPK Inhibit inflammation [104]
Age-related renal injury Mouse PPAR-α↑, AMPK-SIRT1 signaling Attenuate inflammation [105]
Renal ischemia/reperfusion injury Mouse PPAR-α↑ Reduce inflammation [106]
Mouse PPAR-α↑-PI3K/Akt signaling Reduce the secretion of proinflammatory cytokines [107]
Lipotoxicity-induced renal injury Rat PPAR-α↑ Reduce lipid accumulation [108]
Rat PPAR-α–FoxO3a–PGC-1α pathway Reduce lipid accumulation [109]
Mouse AMPK-autophagy, FAO enzymes, and antioxidants Reduce renal inflammation [110]

NF-κB: Nuclear factor κ-B; TGF-β1: Transforming growth factor-β1; Smad3: SMAD family member 3; FGF21: Fibroblast growth factor 21; Nrf2: Nuclear factor erythroid-2-related factor 2; AMPK: Adenosine mitogen-activated protein kinase; PPAR-α: Peroxisome proliferator-activated receptor-α; IL: Interleukin; SIRT1: Sirtuin 1; MAPK: Mitogen-activated protein kinase; PI3K: Phosphoinositide 3-kinase; Akt: Protein kinase B; FoxO3a: Forkhead transcription factor O subfamily member 3a; PGC-1α: Peroxisome proliferator-activated receptor-γ coactivator -1α; FAO: Fatty acid oxidation.

Immune system diseases

Fenofibrate exerts direct anti-inflammatory effects in immune diseases, and most of these effects are associated with PPAR-α [111].

In HIV-infected patients with hypertriglyceridemia, fenofibrate regulates chemokine gene expression in circulating leukocytes, thereby reducing the expression of C-C motif chemokine receptor 2 (CCR2) and C-X3-C motif chemokine ligand 1 (CX3CL1) to mitigate inflammatory responses [112]. This indicates that fenofibrate can suppress systemic inflammation in HIV-infected patients.

Rheumatoid arthritis manifests as acute or chronic connective tissue inflammation. Fenofibrate counters the inflammation in rheumatoid arthritis to improve its clinical symptoms [113–115]. PPAR-α is downregulated in inflammatory responses in arthritis; however, fenofibrate increases PPAR-α expression to inhibit arthritic inflammation [116].

Fenofibrate also reduces Müller cell proliferation and inflammatory cytokines secretion by Müller cells in rats with experimental autoimmune uveoretinitis. This effect is potentially associated with PPAR-α receptor stimulation [117]. In addition, in rats with experimental autoimmune myocarditis, fenofibrate restores Treg/Th17 balance by activating PPAR-α and inhibiting the NF-κB pathway [34]. The translation of fenofibrate into clinical use remains to be explored. In brief, fenofibrate principally counteracts inflammation in autoimmune diseases via PPAR-α activation (Table 5).

Table 5: Anti-inflammatory targets and mechanisms of fenofibrate action in immune system diseases
Diseases Cells/Models Targets and mechanisms Implications References
HIV-infected patients with hypertriglyceridemia Human CCR2 and CX3CL1 Attenuate inflammatory responses [112]
Rheumatoid arthritis Human IL-6, CRP Reduce arthritic inflammation [115]
Rat PPAR-α↑ Reduce arthritic inflammation [116]
Experimental autoimmune disease Rat PPAR-α↑-IL-6, IL-17 and VEGFs, Müller cell Reduce inflammatory responses [117]
Rat PPAR-α↑- NF-κB-Treg/Th17 Reduce inflammatory responses [34]

CCR2: C-C motif chemokine receptor 2; CX3CL1: C-X3-C motif chemokine ligand 1; IL: Interleukin; PPAR-α: Peroxisome proliferator-activated receptor-α; NF-κB: Nuclear factor kappa-B; Th17: T helper cells 17; CRP: C-reactive protein; VEGF: Vascular endothelial growth factor.

Nervous system diseases

In neurodegenerative diseases, fenofibrate decreases the expression of the two neuro-inflammatory genes—inducible NO synthase (iNOS) and COX-2, activating PPARs to exert neuroprotective effects [118]. The activation of PPAR-α induced by fenofibrate also improves peroxisomal function and reduces NO-induced neuroinflammation [119].

Traumatic brain injury (TBI) causes neuroinflammatory responses. The activation of PPAR-α by fenofibrate may counteract the deleterious inflammatory responses after TBI [120]. The expression of iNOS, COX-2, and MMP-9 is upregulated in TBI, but then reversed by fenofibrate; but whether these effects are associated with the activation of PPAR-α requires further examination [121].

Ischemic stroke can lead to neuroinflammation and cerebral injury. Chronic use of fenofibrate activates PPAR-α to diminish the expression of ICAM-1 and VCAM-1 [122]. Acute fenofibrate treatment also counters inflammation by reducing the infiltration of polymorphonuclear leukocytes (PMNs) and microglial activation after ischemic stroke. ICAM-1 is downregulated after fenofibrate intervention [123]. Fenofibrate improves stroke outcomes and resolves neuroinflammation more effectively in male than in female mice, which may be due to the lower PPAR-α expression in cells and tissues of females [124]. In addition, in rats with global cerebral I/R, fenofibrate suppresses p65 NF-κB and p38 MAPK activities, resulting in an anti-inflammatory effect [125].

Fenofibrate attenuates the neuroinflammation induced by intracerebral LPS injection and the expression of inflammation-related cytokines. The activities of macrophages and leukocytes are also weakened by fenofibrate through PPAR-α activation [126].

Multiple sclerosis is a chronic inflammatory demyelinating disease of the central nervous system. Astrocytes and microglia play crucial roles in the inflammatory responses typical of multiple sclerosis. Fenofibrate inhibits the production of NO induced by LPS, as well as the NF-κB DNA-binding activity and the secretion of the proinflammatory cytokines TNF-α, IL-1β, and IL-6 by astrocytes [127]. Moreover, NF-κB activity is suppressed by fenofibrate by activating SIRT1, but not PPAR-α [128]. Similar effects are also observed in microglia. Further, the inhibition on MCP-1 secretion after fenofibrate intervention impedes the migration of peripheral immune cells into the central nervous system [129].

A high-glucose environment favors the development of sciatic nerve inflammation, which may be attenuated by fenofibrate by triggering the PPAR-α-AMPK-PGC-1α-eNOS pathway, followed by the activation of the PI3K-Akt-eNOS-signaling pathway [130].

In a PPAR-α-dependent manner, fenofibrate also leads to the underproduction of IL-1β and TNF-α in the brains of patients with Huntington’s disease [131]. In summary, fenofibrate can effectively reduce neuroinflammation, which endows it with clinical therapeutic potential (Table 6).

Table 6: Anti-inflammatory targets and mechanisms of fenofibrate action in nervous system diseases
Diseases Cells/Models Targets and mechanisms Implications References
Neurodegenerative diseases Mouse PPAR-α↑- iNOS, COX-2 Generate neuroprotective effects [118]
Cortical neurons PPAR-α↑- peroxisomal activity Reduce neuroinflammation [119]
Traumatic brain injury Rat PPAR-α↑ Reduce inflammatory responses [120]
Rat iNOS, COX-2 and matrix metalloproteinase-9 (MMP-9) Reduce inflammatory responses [121]
Ischemic strokes Mouse PPAR-α↑- ICAM-1,VCAM-1 Have anti-inflammatory effects [122]
Rat, mouse PMNs infiltration and microglial activation, ICAM-1 Reduce inflammatory responses [123]
Global cerebral ischemia/reperfusion Rat P65 NF-κB, p38 MAPK Have anti-inflammatory effects [125]
Neuroinflammation induce by intracerebral LPS injection Mouse PPAR-α↑ Attenuate neuroinflammation [126]
Multiple sclerosis Murine astrocytes NO,NF-κB DNA binding activity Reduce inflammatory responses [127]
Murine microglia SIRT1- NF-κB Reduce inflammatory responses [128]
Murine microglia MCP-1 Impede the migration of peripheral immune cells into the central nervous system [129]
Diabetic peripheral neuropathy Mouse, HUVECs, HSCs PPAR-α-AMPK-PGC-1a-eNOS pathway-PI3K-Akt-eNOS signaling Reduce inflammatory responses [130]
Huntington’s disease Rat PPAR-α↑- IL-1β↓, TNF-α↓ Reduce inflammatory responses [131]

PPAR-α: Peroxisome proliferator-activated receptor α; iNOS: Inducible nitric oxide synthase; ICAM-1: Intercellular cell adhesion molecule 1; PMN: Polymorphonuclear leukocyte; NF-κB: Nuclear factor κ-B; NO: Nitric oxide; SIRT1: Sirtuin 1; HUVEC: Human umbilical vein endothelial cell; AMPK: Adenosine mitogen-activated protein kinase; COX-2: Cyclooxygenase-2; VCAM - 1: Vascular cell adhesion protein 1; MAPK: Mitogen-activated protein kinase; MPC -1: Monocyte chemoattractant protein 1; PGC: Primordial germ cell; eNOS: Endothelial nitric oxide synthase; PI3K: Phosphoinositide 3-kinase; IL-1: Interleukin 1; TNF-α: Tumor necrosis factor α; LPS: Lipopolysaccharide; CNS: Central nervous system; HCS: Human chorionic somatomammotropin.

Respiratory system diseases

Fenofibrate inhibits inflammation in the respiratory system by upregulating PPAR-α [132]. By means of PPAR-α, fenofibrate downregulates cellular infiltration and chemoattractant production, thus enhancing MMP activity triggered by LPS in the mouse lung [133]. Moreover, fenofibrate lowers the level of IL-1β to reduce neutrophilic inflammation [134].

In treating bronchial asthma, fenofibrate is more effective than dexamethasone, whose effect is partially mediated by suppression of Th17-IL-23/IL-17 axis. This superiority is achieved by modulation of both Th2- and Th17-cell-derived cytokines [135]. Fenofibrate dose-dependently blocks inflammatory cell infiltration in asthmatic airways. Fenofibrate also dramatically reduces inflammatory responses, including infiltration of eosinophils, neutrophils, lymphocytes, and macrophages, and increases the release of IL-4, IL-5, TNF-α, MIP-2, and MCP-1 [136]. These beneficial effects may bring a possibility of using fenofibrate to design anti-asthma drug. In conclusion, fenofibrate can significantly improve the inflammation of the respiratory system (Table 7).

Table 7: Anti-inflammatory targets and mechanisms of fenofibrate action in respiratory system diseases
Diseases Cells/Models Targets and mechanisms Implications References
Airway inflammation Mouse PPAR-α↑ Inhibit inflammation [132, 133]
Human small airway epithelial cells IL-1β production Reduce neutrophilic inflammation [134]
Asthma Rat TH17-IL-23/IL-17 axis Have anti-inflammatory effects [135]
Mouse Infiltration of eosinophils, neutrophils, lymphocytes, macrophages, IL-4, IL-5, TNF-α, MIP-2, and MCP-1 Reduce inflammatory responses [136]

PPAR-α: Peroxisome proliferator-activated receptor α; IL: Interleukin; TNF-α: Tumor necrosis factor-α; MCP-1: Monocyte chemoattractant protein 1; MIP-2: Macrophage inflammatory protein 2.

Table 8: Anti-inflammatory targets and mechanisms of fenofibrate action in bacterial sepsis
Diseases Cells/Models Targets and mechanisms Implications References
Bacterial sepsis Mouse ERK phosphorylation- GRK2- CXCR2 Promote neutrophil accumulation at infection sites, quick eliminate pathogens [137]
Mouse, bone marrow-derived macrophages LKB1-pAMPK-SHP- NF-κB, UCP2 Reduce inflammatory responses [138]
Rabbit Monocyte tissue factor Block endothelial inflammation [139]
Mouse TNF Decrease endotoxin action [140]

ERK: Extracellular-signal-regulated kinase; GRK2: G protein-coupled receptor kinases 2; CXCR2: C-X-C motif chemokine receptor 2; LKB1: Liver kinase B1; SHP: Small heterodimer partner; NF-κB: Nuclear factor κ-B; UCP2: Uncoupling protein 2; TNF: Tumor necrosis factor.

Bacterial sepsis

Bacterial sepsis is a systemic inflammation induced by infection. Fenofibrate reduces this inflammation by enhancing neutrophil chemotaxis. Independent of PPAR-α, fenofibrate inhibits the downregulation of C-X-C motif chemokine receptor 2 (CXCR2) by blocking extracellular-signal-regulated kinase (ERK) phosphorylation and subsequent expression of G protein-coupled receptor kinases 2 (GRK2) and promotes neutrophil accumulation at infection sites and quick elimination of pathogens [137]. Moreover, fenofibrate activates the liver kinase B1 (LKB1)/AMPK pathway and increases small heterodimer partner (SHP) expression. SHP then downregulates NF-kB and upregulates mitochondrial uncoupling protein 2 (UCP2) to reduce ROS production, providing the inhibitory effects on inflammatory responses [138]. Fenofibrate also represses monocyte tissue factor expression to block endothelial inflammation in endotoxin-induced shock by the activation of PPAR-α [139]. In addition, by activating PPAR-α, fenofibrate also blunted the upregulation of TNF, which is proved to be a proximal mediator of endotoxin action [140].

Although fenofibrate effectively reduces the systematic inflammation in bacterial sepsis, this effect is limited to inflammation induced by pathogens; in contrast, neutrophil-dependent sterile inflammation does not subside after fenofibrate treatment [137] (Table 8).

Figure 1.: A schematic figure of the mechanisms governing the anti-inflammatory effects (direct or indirect) of fenofibrate in various diseases. HDL: High-density lipoprotein; ApoA-I: Apolipoprotein A-I; SREBP: Sterol-regulatory element binding protein; LXR: Liver X receptors; TLR4: Toll-like receptor 4; PPAR-α: Peroxisome proliferator-activated receptor α; FGF21: Fibroblast growth factor 21; NF-κB: Nuclear factor κ-B; AMPK: Adenosine mitogen-activated protein kinase; SIRT1: Sirtuin 1.

Discussion

Fenofibrate, a PPAR-α agonist, has been primarily used to control diseases related to metabolism, and its anti-inflammatory properties have been observed in various diseases. Its action against inflammation is chiefly achieved via the activation of PPAR-α and the regulation of associated inflammatory pathways. Fenofibrate also functions independently of PPAR-α. Fenofibrate suppresses inflammatory responses by modulating cytokines secretion, cellular infiltration, cell death, and organ dysfunction. The advantage of fenofibrate treatment lies in simultaneous regulation of various metabolic disorders and few side effects. The direct or indirect anti-inflammatory effects of fenofibrate have been observed in atherosclerosis, diabetic retinopathy, NASH, etc. However, most of these studies are at the animal or cell level. In clinical practice, many patients with abnormal lipid metabolism also suffer from these diseases. Therefore, whether fenofibrate can be used to treat these diseases requires human or clinical trials.

Although fenofibrate shows extensive anti-inflammatory effects in many diseases, it is not always protective against inflammation. In some studies, fenofibrate exerts no effects [141], or even exacerbates the inflammatory response [142–144]. For example, fenofibrate, at a dose commonly used for lipid abnormalities and heart disease, does not suppress inflammation in response to low-dose endotoxin in healthy humans. These results suggest that the anti-inflammatory effects of fenofibrate in systemic inflammation are limited [145] compared to those in local inflammation.

Conclusion

Here we summarized the anti-inflammatory effects and mechanisms of fenofibrate in various systemic diseases (Figure 1). As a blood lipid-lowering drug used clinically, fenofibrate exerts an anti-inflammatory effect, which is secondary to the decrease of blood lipid, to reduce the inflammation induced by hyperlipidemia. As a PPAR-α agonist, fenofibrate plays most of its roles through activating PPAR-α and inhibiting downstream inflammatory signaling pathways. In addition to these indirect effects, fenofibrate can also directly regulate inflammation-related signaling pathways. Fenofibrate is not the first choice for many diseases we discussed in this review. Moreover, many studies on its efficacy are still at the animal or cellular level. Thus, the therapeutic potential of fenofibrate should be validated with more basic research and clinical trials.

Acknowledgements

The study was supported by grants from the National Natural Science Foundation of China (No. 82000642) and the Nanjing Health Science and Technology Development Foundation (No.YKK22168).

References

  1. , , , , , (). Mechanism of action of fibrates on lipid and lipoprotein metabolism. Circulation. https://doi.org/10.1161/01.CIR.98.19.2088
  2. , , , , (). Use of fenofibrate on cardiovascular outcomes in statin users with metabolic syndrome: propensity matched cohort study. BMJ. https://doi.org/10.1136/bmj.l5125
  3. , , , , , (). Fenofibrate increases very-long-chain sphingolipids and improves blood glucose homeostasis in NOD mice. Diabetologia. https://doi.org/10.1007/s00125-019-04973-z
  4. , (). Fenofibrate for diabetic retinopathy. Asia Pac J Ophthalmol (Phila). https://doi.org/10.22608/APO.2018288
  5. , , , , , (). Fenofibrate effects on arterial endothelial function in adults with type 2 diabetes mellitus: a FIELD substudy. Atherosclerosis. https://doi.org/10.1016/j.atherosclerosis.2015.07.038
  6. , , , , , (). Fenofibrate improves vascular endothelial function and contractility in diabetic mice. Redox Biol. https://doi.org/10.1016/j.redox.2018.09.024
  7. , , , , , (). Fenofibrate rescues diabetes-related impairment of ischemia-mediated angiogenesis by PPARα-independent modulation of thioredoxin-interacting protein. Diabetes. https://doi.org/10.2337/db17-0926
  8. , , , , , (). PARP1-mediated PPARα poly(ADP-ribosyl)ation suppresses fatty acid oxidation in non-alcoholic fatty liver disease. J Hepatol. https://doi.org/10.1016/j.jhep.2016.11.020
  9. , , , , , (). Early diet-induced non-alcoholic steatohepatitis in APOE2 knock-in mice and its prevention by fibrates. J Hepatol. https://doi.org/10.1016/j.jhep.2005.10.033
  10. , , , , , (). Impaired expression of the peroxisome proliferator–activated receptor alpha during hepatitis C virus infection. Gastroenterology. https://doi.org/10.1053/j.gastro.2004.11.016
  11. , , (). Fibrates and cholestasis. Hepatology (Baltimore, MD). https://doi.org/10.1002/hep.27744
  12. , , , , , (). Anticancer properties of fenofibrate: a repurposing use. J Cancer. https://doi.org/10.7150/jca.24488
  13. , , , (). Fenofibrate reduces cisplatin-induced apoptosis of renal proximal tubular cells via inhibition of JNK and p38 pathways. J Toxicol Sci. https://doi.org/10.2131/jts.41.339
  14. , , , (). Protective roles of fenofibrate against cisplatin-induced ototoxicity by the rescue of peroxisomal and mitochondrial dysfunction. Toxicol Appl Pharmacol. https://doi.org/10.1016/j.taap.2018.06.010
  15. (). Anti-inflammatory agents: present and future. Cell. https://doi.org/10.1016/j.cell.2010.02.043
  16. , , , (). Inflammation in obesity, diabetes, and related disorders. Immunity. https://doi.org/10.1016/j.immuni.2021.12.013
  17. , , , , , (). The relation between systemic inflammation and incident cancer in patients with stable cardiovascular disease: a cohort study. Eur Heart J. https://doi.org/10.1093/eurheartj/ehz587
  18. , (). Interplay between inflammation and thrombosis in cardiovascular pathology. Nat Rev Cardiol. https://doi.org/10.1038/s41569-021-00552-1
  19. , (). The eye in systemic inflammatory diseases. Lancet (London, England). https://doi.org/10.1016/S0140-6736(04)17554-5
  20. , , , , , (). Distinct fibroblast subsets drive inflammation and damage in arthritis. Nature. https://doi.org/10.1038/s41586-019-1263-7
  21. , , , (). Fenofibrate reduces mortality and precludes neurological deficits in survivors in murine model of Japanese encephalitis viral infection. PLoS One. https://doi.org/10.1371/journal.pone.0035427
  22. , , (). Anti-colony-stimulating factor therapies for inflammatory and autoimmune diseases. Nat Rev Drug Discov. https://doi.org/10.1038/nrd.2016.231
  23. , , , , , (). Role of antioxidants and natural products in inflammation. Oxid Med Cell Longev. https://doi.org/10.1155/2016/5276130
  24. , (). Regulation of low-density lipoprotein cholesterol by intestinal inflammation and the acute phase response. Cardiovasc Res. https://doi.org/10.1093/cvr/cvx237
  25. , , , , , (). Reciprocal and coordinate regulation of serum amyloid A versus apolipoprotein A-I and paraoxonase-1 by inflammation in murine hepatocytes. Arterioscler Thromb Vasc Biol. https://doi.org/10.1161/01.ATV.0000227472.70734.ad
  26. , , , , , (). Intrahepatic cholesterol influences progression, inhibition and reversal of non-alcoholic steatohepatitis in hyperlipidemic mice. FEBS Lett. https://doi.org/10.1016/j.febslet.2010.01.046
  27. , , , , , (). Abnormal islet sphingolipid metabolism in type 1 diabetes. Diabetologia. https://doi.org/10.1007/s00125-018-4614-2
  28. , , , , , (). Fenofibrate attenuates fatty acid-induced islet beta-cell dysfunction and apoptosis via inhibiting the NF-kappaB/MIF dependent inflammatory pathway. Metabolism. https://doi.org/10.1016/j.metabol.2017.09.001
  29. , , , , , (). PPARα agonist fenofibrate attenuates TNF-α-induced CD40 expression in 3T3-L1 adipocytes via the SIRT1-dependent signaling pathway. Exp Cell Res. https://doi.org/10.1016/j.yexcr.2013.04.007
  30. , , , , , (). Fenofibrate improves high-fat diet-induced and palmitate-induced endoplasmic reticulum stress and inflammation in skeletal muscle. Life Sci. https://doi.org/10.1016/j.lfs.2016.06.008
  31. , , , , (). Fenofibrate suppresses microvascular inflammation and apoptosis through adenosine monophosphate–activated protein kinase activation. Metabolism. https://doi.org/10.1016/j.metabol.2010.04.020
  32. , , , (). Systemic and distal repercussions of liver-specific peroxisome proliferator-activated receptor-α control of the acute-phase response. Endocrinology. https://doi.org/10.1210/en.2007-1339
  33. , , , , , (). Fenofibrate attenuates cardiac and renal alterations in young salt-loaded spontaneously hypertensive stroke-prone rats through mitochondrial protection. J Hypertens. https://doi.org/10.1097/HJH.0000000000001651
  34. , , , , (). Fenofibrate treatment of rats with experimental autoimmune myocarditis by alleviating Treg/Th17 disorder. Cent Eur J Immunol. https://doi.org/10.5114/ceji.2016.58817
  35. , , , , , (). Impaired peroxisomal fitness in obese mice, a vicious cycle exacerbating adipocyte dysfunction via oxidative stress. Antioxid Redox Signal. https://doi.org/10.1089/ars.2018.7614
  36. , , , (). Fenofibrate reduces inflammation in obese patients with or without type 2 diabetes mellitus via sirtuin 1/fetuin A axis. Diabetes Res Clin Pract. https://doi.org/10.1016/j.diabres.2015.05.043
  37. , (). Adiponectin: a manifold therapeutic target for metabolic syndrome, diabetes, and coronary disease?. Cardiovasc Diabetol. https://doi.org/10.1186/1475-2840-13-103
  38. , , , , , (). Effects of peroxisome proliferator-activated receptor ligands, bezafibrate and fenofibrate, on adiponectin level. Arterioscler Thromb Vasc Biol. https://doi.org/10.1161/01.ATV.0000256469.06782.d5
  39. , , (). Effects of short-term fenofibrate treatment on circulating markers of inflammation and hemostasis in patients with impaired glucose tolerance. J Clin Endocrinol Metab. https://doi.org/10.1210/jc.2005-1615
  40. , , , (). Pleiotropic effects of atorvastatin and fenofibrate in metabolic syndrome and different types of pre-diabetes. Diabetes Care. https://doi.org/10.2337/dc10-0272
  41. , , , (). Fenofibrate reduces systemic inflammation markers independent of its effects on lipid and glucose metabolism in patients with the metabolic syndrome. J Clin Endocrinol Metab. https://doi.org/10.1210/jc.2009-1487
  42. , , (). Pleiotropic action of short-term metformin and fenofibrate treatment, combined with lifestyle intervention, in type 2 diabetic patients with mixed dyslipidemia. Diabetes Care. https://doi.org/10.2337/dc08-2335
  43. , , , , , (). PPAR-α agonist fenofibrate decreased RANTES levels in type 2 diabetes patients with hypertriglyceridemia. Med Sci Monit. https://doi.org/10.12659/MSM.897307
  44. , , , , (). Anti-inflammatory activity of anti-hyperlipidemic drug, fenofibrate, and its phase-I metabolite fenofibric acid: in silico, in vitro, and in vivo studies. Inflammopharmacology. https://doi.org/10.1007/s10787-017-0428-y
  45. , , , , , (). Effect of fenofibrate on serum inflammatory markers in patients with high triglyceride values. Atheroscler. Suppl.. https://doi.org/10.1177/107424840400900i105
  46. (). Effect of fenofibrate on adiponectin and inflammatory biomarkers in metabolic syndrome patients. Obesity (Silver Spring). https://doi.org/10.1038/oby.2008.530
  47. , (). Fenofibrate reduces tumor necrosis factor-α serum concentration and adipocyte secretion of hypercholesterolemic rabbits. Clin Chim Acta. https://doi.org/10.1016/j.cccn.2004.04.001
  48. , , , , , (). An epigenome-wide association study of inflammatory response to fenofibrate in the genetics of lipid lowering drugs and diet network. Pharmacogenomics. https://doi.org/10.2217/pgs-2017-0037
  49. , , , , (). PPAR-α agonist fenofibrate combined with octreotide acetate in the treatment of acute hyperlipidemia pancreatitis. PPAR Res. https://doi.org/10.1155/2021/6629455
  50. , (). Targeting inflammation in atherosclerosis—from experimental insights to the clinic. Nat Rev Drug Discov. https://doi.org/10.1038/s41573-021-00198-1
  51. , , , , , (). The reduction of inflammatory biomarkers by statin, fibrate, and combination therapy among diabetic patients with mixed dyslipidemia: the DIACOR (diabetes and combined lipid therapy regimen) study. J Am Coll Cardiol. https://doi.org/10.1016/j.jacc.2006.05.009
  52. , , , , , (). Lipid lowering and imaging protease activation in atherosclerosis. J Nucl Cardiol. https://doi.org/10.1007/s12350-013-9843-7
  53. , , , (). Modulation of C-reactive protein–mediated monocyte chemoattractant protein-1 induction in human endothelial cells by anti-atherosclerosis drugs. Circulation Official J Am Heart Assoc. https://doi.org/10.1161/01.CIR.103.21.2531
  54. , , , (). C-reactive protein-induced expression of CD40–CD40L and the effect of lovastatin and fenofibrate on it in human vascular endothelial cells. Biol Pharm Bull. https://doi.org/10.1248/bpb.27.1537
  55. , , , , , (). Fenofibrate reduces atherogenesis in ApoE*3Leiden mice: evidence for multiple antiatherogenic effects besides lowering plasma cholesterol. Arterioscler Thromb Vasc Biol. https://doi.org/10.1161/01.ATV.0000238348.05028.14
  56. , , , , , (). Expression of heme oxygenase-1 in human vascular cells is regulated by peroxisome proliferator-activated receptors. Arterioscler Thromb Vasc Biol. https://doi.org/10.1161/ATVBAHA.107.142638
  57. (). Atherosclerosis—an inflammatory disease. N Engl J Med. https://doi.org/10.1056/NEJM199901143400207
  58. , , , , , (). Oxidized low-density lipoprotein and peroxisome-proliferator-activated receptor α down-regulate platelet-activating-factor receptor expression in human macrophages. Biochem J. https://doi.org/10.1042/bj3540225
  59. , , , , , (). PPARα agonists inhibit inflammatory activation of macrophages through upregulation of beta-defensin 1. Atherosclerosis. https://doi.org/10.1016/j.atherosclerosis.2015.04.005
  60. , , , , (). PPARalpha activator fenofibrate modulates angiotensin II-induced inflammatory responses in vascular smooth muscle cells via the TLR4-dependent signaling pathway. Biochem Pharmacol. https://doi.org/10.1016/j.bcp.2009.06.095
  61. , , , (). Modulation of LPS-mediated inflammation by fenofibrate via the TRIF-dependent TLR4 signaling pathway in vascular smooth muscle cells. Cell Physiol Biochem. https://doi.org/10.1159/000315082
  62. , , , , , (). Activation of PPAR α by fenofibrate inhibits apoptosis in vascular adventitial fibroblasts partly through SIRT1-mediated deacetylation of FoxO1. Exp Cell Res. https://doi.org/10.1016/j.yexcr.2015.07.027
  63. , , , , , (). Fenofibrate increases high-density lipoprotein and sphingosine 1 phosphate concentrations limiting abdominal aortic aneurysm progression in a mouse model. Am J Pathol. https://doi.org/10.1016/j.ajpath.2012.04.015
  64. , , , , , (). Therapeutic effects of PPARα agonists on diabetic retinopathy in type 1 diabetes models. Diabetes. https://doi.org/10.2337/db11-0413
  65. , , , , , (). Pathogenic role of diabetes-induced PPAR-α down-regulation in microvascular dysfunction. Proc Natl Acad Sci USA. https://doi.org/10.1073/pnas.1307211110
  66. (). Direct effects of PPARalpha agonists on retinal inflammation and angiogenesis may explain how fenofibrate lowers risk of severe proliferative diabetic retinopathy. Diabetes. https://doi.org/10.2337/db12-1223
  67. , , , , , (). Effect of fenofibrate on the expression of inflammatory mediators in a diabetic rat model. Curr Eye Res. https://doi.org/10.1080/02713683.2019.1622020
  68. , , , , (). Fenofibrate prevents the disruption of the outer blood retinal barrier through downregulation of NF-kappaB activity. Acta Diabetol. https://doi.org/10.1007/s00592-015-0759-3
  69. , , , , , (). Fenofibrate reduces the severity of neuroretinopathy in a type 2 model of diabetes without inducing peroxisome proliferator-activated receptor alpha-dependent retinal gene expression. J Clin Med. https://doi.org/10.3390/jcm10010126
  70. , , , , , (). Fenofibrate exerts protective effects in diabetic retinopathy via inhibition of the ANGPTL3 pathway. Invest Ophthalmol Vis Sci. https://doi.org/10.1167/iovs.18-24155
  71. , , , , , (). Fenofibrate ameliorates diabetic retinopathy by modulating Nrf2 signaling and NLRP3 inflammasome activation. Mol Cell Biochem. https://doi.org/10.1007/s11010-017-3256-x
  72. , , , , (). Effects of fenofibrate on adiponectin expression in retinas of streptozotocin-induced diabetic rats. J Diabetes Res. https://doi.org/10.1155/2014/540326
  73. , , , , (). Beneficial effects of fenofibric acid on overexpression of extracellular matrix components, COX-2, and impairment of endothelial permeability associated with diabetic retinopathy. Exp Eye Res. https://doi.org/10.1016/j.exer.2015.08.010
  74. , , , (). Reappraising the role of inflammation in heart failure. Nat Rev Cardiol. https://doi.org/10.1038/s41569-019-0315-x
  75. , , , , (). Loss of protease-activated receptor 4 prevents inflammation resolution and predisposes the heart to cardiac rupture after myocardial infarction. Circulation. https://doi.org/10.1161/CIRCULATIONAHA.119.044340
  76. , , , , , (). Fenofibrate increases cardiac autophagy via FGF21/SIRT1 and prevents fibrosis and inflammation in the hearts of type 1 diabetic mice. Clin Sci (Lond). https://doi.org/10.1042/CS20150623
  77. , , , , , (). HMGB1 is involved in the protective effect of the PPAR alpha agonist fenofibrate against cardiac hypertrophy. PPAR Res. https://doi.org/10.1155/2014/541394
  78. , , , , , (). Fenofibrate attenuates doxorubicin-induced cardiac dysfunction in mice via activating the eNOS/EPC pathway. Sci Rep. https://doi.org/10.1038/s41598-021-80984-4
  79. , , , , , (). Treatment with fenofibrate plus a low dose of benznidazole attenuates cardiac dysfunction in experimental Chagas disease. Int J Parasitol Drugs Drug Resist. https://doi.org/10.1016/j.ijpddr.2017.10.003
  80. , , , , , (). PPAR alpha activator fenofibrate inhibits myocardial inflammation and fibrosis in angiotensin II-infused rats. J Mol Cell Cardiol. https://doi.org/10.1016/j.yjmcc.2003.11.004
  81. , , , , , (). Fenofibrate, a peroxisome proliferator-activated receptor alpha activator, suppresses experimental autoimmune myocarditis by stimulating the interleukin-10 pathway in rats. J Atheroscler Thromb. https://doi.org/10.5551/jat.9.87
  82. , , , , , (). Fenofibrate attenuates endothelial monocyte adhesion in chronic heart failure: an in vitro study. Eur J Clin Invest. https://doi.org/10.1111/j.1365-2362.2009.02176.x
  83. , , , , , (). Attenuation of cardiac dysfunction by a PPAR-alpha agonist is associated with down-regulation of redox-regulated transcription factors. J Mol Cell Cardiol. https://doi.org/10.1016/j.yjmcc.2006.05.013
  84. , , , , (). Novel putative SREBP and LXR target genes identified by microarray analysis in liver of cholesterol-fed mice. J Lipid Res. https://doi.org/10.1194/jlr.M300203-JLR200
  85. , , , , , (). Human hepatic in vitro models reveal distinct anti-NASH potencies of PPAR agonists. Cell Biol Toxicol. https://doi.org/10.1007/s10565-020-09544-2
  86. , , , , , (). The PPARα agonist fenofibrate prevents formation of protein aggregates (mallory-denk bodies) in a murine model of steatohepatitis-like hepatotoxicity. Sci Rep. https://doi.org/10.1038/s41598-018-31389-3
  87. , , , , , (). Fenofibrate regulates visceral obesity and nonalcoholic steatohepatitis in obese female ovariectomized C57BL/6J mice. Int J Mol Sci. https://doi.org/10.3390/ijms22073675
  88. , , , , , (). Fenofibrate improves liver function and reduces the toxicity of the bile acid pool in patients with primary biliary cholangitis and primary sclerosing cholangitis who are partial responders to ursodiol. Clin Pharmacol Ther. https://doi.org/10.1002/cpt.1930
  89. , , , , , (). Peroxisome proliferators-activated alpha agonist treatment ameliorates hepatic damage in rats with obstructive jaundice: an experimental study. BMC Gastroenterol. https://doi.org/10.1186/1471-230X-7-44
  90. , , , , (). PPARα activation protects against cholestatic liver injury. Sci Rep. https://doi.org/10.1038/s41598-017-10524-6
  91. , (). Effect of preischemic treatment with fenofibrate, a peroxisome proliferator-activated receptor-alpha ligand, on hepatic ischemia–reperfusion injury in rats. J Mol Histol. https://doi.org/10.1007/s10735-011-9313-y
  92. , , (). Fenofibrate-induced massive regression of multiple inflammatory hepatocellular adenoma. Clin Res Hepatol Gastroenterol. https://doi.org/10.1016/j.clinre.2015.09.009
  93. , , , (). Fenofibrate and PBA prevent fatty acid-induced loss of adiponectin receptor and pAMPK in human hepatoma cells and in hepatitis C virus-induced steatosis. J Lipid Res. https://doi.org/10.1194/jlr.M800633-JLR200
  94. , , , , , (). Global suppression of IL-6-induced acute phase response gene expression after chronic in vivo treatment with the peroxisome proliferator-activated receptor-alpha activator fenofibrate. J Biol Chem. https://doi.org/10.1074/jbc.M400346200
  95. , , , , , (). Fenofibrate inhibited the differentiation of T helper 17 cells in vitro. PPAR Res. https://doi.org/10.1155/2012/145654
  96. , , , , , (). Fenofibrate represses interleukin-17 and interferon-gamma expression and improves colitis in interleukin-10-deficient mice. Gastroenterology. https://doi.org/10.1053/j.gastro.2007.03.113
  97. , , , (). Fenofibrate attenuates tubulointerstitial fibrosis and inflammation through suppression of nuclear factor-kappaB and transforming growth factor-beta1/Smad3 in diabetic nephropathy. Exp Biol Med (Maywood). https://doi.org/10.1258/ebm.2009.009218
  98. , , , , (). Improvement of inflammatory responses associated with NF-kappa B pathway in kidneys from diabetic rats. Inflamm Res. https://doi.org/10.1007/s00011-006-6190-z
  99. , , , , , (). Up-regulation of Nrf2 is involved in FGF21-mediated fenofibrate protection against type 1 diabetic nephropathy. Free Radic Biol Med. https://doi.org/10.1016/j.freeradbiomed.2016.02.002
  100. , , , , , (). The role of Akt2 in the protective effect of fenofibrate against diabetic nephropathy. Int J Biol Sci. https://doi.org/10.7150/ijbs.40643
  101. , , , (). Fenofibrate improves renal function by amelioration of NOX-4, IL-18, and p53 expression in an experimental model of diabetic nephropathy. J Cell Biochem. https://doi.org/10.1002/jcb.27055
  102. , , , , , (). Pathophysiology of apolipoprotein E deficiency in mice: relevance to apo E-related disorders in humans. FASEB J. https://doi.org/10.1096/fj.01-0463com
  103. , , , , , (). Peroxisome proliferator-activated receptor {alpha} agonism prevents renal damage and the oxidative stress and inflammatory processes affecting the brains of stroke-prone rats. J Pharmacol Exp Ther. https://doi.org/10.1124/jpet.110.171090
  104. , , , , , (). PPARalpha agonist fenofibrate protects the kidney from hypertensive injury in spontaneously hypertensive rats via inhibition of oxidative stress and MAPK activity. Biochem Biophys Res Commun. https://doi.org/10.1016/j.bbrc.2010.03.043
  105. , , , , , (). PPARα agonist, fenofibrate, ameliorates age-related renal injury. Exp Gerontol. https://doi.org/10.1016/j.exger.2016.04.021
  106. , , , , , (). Peroxisome proliferator-activated receptor-alpha contributes to the resolution of inflammation after renal ischemia/reperfusion injury. J Pharmacol Exp Ther. https://doi.org/10.1124/jpet.108.146191
  107. , , (). Fenofibrate pre-treatment suppressed inflammation by activating phosphoinositide 3 kinase/protein kinase B (PI3K/Akt) signaling in renal ischemia-reperfusion injury. J Huazhong Univ Sci Technolog Med Sci. https://doi.org/10.1007/s11596-015-1389-2
  108. , , , , , (). Peroxisome proliferator-activated receptor-α activator fenofibrate prevents high-fat diet-induced renal lipotoxicity in spontaneously hypertensive rats. Hypertens Res. https://doi.org/10.1038/hr.2009.107
  109. , , , , , (). High-fat diet-induced renal cell apoptosis and oxidative stress in spontaneously hypertensive rat are ameliorated by fenofibrate through the PPARα-FoxO3a-PGC-1α pathway. Nephrol Dial Transplant. https://doi.org/10.1093/ndt/gfr613
  110. , , , , , (). Delayed treatment with fenofibrate protects against high-fat diet-induced kidney injury in mice: the possible role of AMPK autophagy. Am J Physiol Renal Physiol. https://doi.org/10.1152/ajprenal.00596.2015
  111. , , , (). The role of PPAR alpha in the modulation of innate immunity. Int J Mol Sci. https://doi.org/10.3390/ijms221910545
  112. , , , , , (). Treatment of hypertriglyceridemia and HIV: fenofibrate-induced changes in the expression of chemokine genes in circulating leukocytes. AIDS Res Ther. https://doi.org/10.1186/1742-6405-6-26
  113. (). A comparative study of anti-inflammatory and antidyslipidemic effects of fenofibrate and statins on rheumatoid arthritis. Mod Rheumatol. https://doi.org/10.1007/s10165-009-0261-2
  114. , , (). Protective effects of fenofibrate and resveratrol in an aggressive model of rheumatoid arthritis in rats. Pharm Biol. https://doi.org/10.3109/13880209.2015.1125931
  115. , , , (). The effects of fenofibrate on inflammation and cardiovascular markers in patients with active rheumatoid arthritis: a pilot study. Rheumatol Int. https://doi.org/10.1007/s00296-012-2613-z
  116. , , , , , (). Fenofibrate, a PPAR α agonist, decreases atrogenes and myostatin expression and improves arthritis-induced skeletal muscle atrophy. Am J Physiol Endocrinol Metab. https://doi.org/10.1152/ajpendo.00590.2010
  117. , , , , (). Treatment of experimental autoimmune uveoretinitis with peroxisome proliferator-activated receptor α agonist fenofibrate. Mol Vis. http://www.molvis.org/molvis/v20/1518/.
  118. , , , , , (). Preferential PPAR-α activation reduces neuroinflammation, and blocks neurodegeneration in vivo. Hum Mol Genet. https://doi.org/10.1093/hmg/ddv477
  119. , , , , (). Peroxisome proliferator-activated receptor-αagonists protect cortical neurons from inflammatory mediators and improve peroxisomal function. Eur J Neurosci. https://doi.org/10.1111/j.1460-9568.2011.07637.x
  120. , , , (). Fenofibrate, a peroxisome proliferator-activated receptor α agonist, exerts neuroprotective effects in traumatic brain injury. Neurosci Lett. https://doi.org/10.1016/j.neulet.2005.06.019
  121. , , , , , (). Neurological recovery-promoting, anti-inflammatory, and anti-oxidative effects afforded by fenofibrate, a PPAR alpha agonist, in traumatic brain injury. J Neurotrauma. https://doi.org/10.1089/neu.2006.0216
  122. , , , , , (). Peroxisome proliferator-activated receptor-alpha activation as a mechanism of preventive neuroprotection induced by chronic fenofibrate treatment. J Neurosci Official J Soci Neurosci. https://doi.org/10.1523/JNEUROSCI.23-15-06264.2003
  123. , , , , , (). Effects of the PPAR-α agonist fenofibrate on acute and short-term consequences of brain ischemia. J Cereb Blood Flow Metab. https://doi.org/10.1038/jcbfm.2013.233
  124. , , , , , (). Sex differences and the role of PPAR alpha in experimental stroke. Metab Brain Dis. https://doi.org/10.1007/s11011-015-9766-x
  125. , , , , , (). PPARα agonist fenofibrate ameliorates learning and memory deficits in rats following global cerebral ischemia. Mol Neurobiol. https://doi.org/10.1007/s12035-014-8882-7
  126. , (). Effects of chronic systemic treatment with peroxisome proliferator-activated receptor αactivators on neuroinflammation induced by intracerebral injection of lipopolysaccharide in adult mice. Neurosci Res. https://doi.org/10.1016/j.neures.2011.02.001
  127. , , , (). Peroxisome proliferator-activated receptor-αand retinoid X receptor agonists inhibit inflammatory responses of astrocytes. J Neuroimmunol. https://doi.org/10.1016/j.jneuroim.2006.04.019
  128. , , , , , (). Pemafibrate, a selective PPARα modulator, and fenofibrate suppress microglial activation through distinct PPARα and SIRT1-dependent pathways. Biochem Biophys Res Commun. https://doi.org/10.1016/j.bbrc.2020.01.118
  129. , , , , (). Agonists for the peroxisome proliferator-activated receptor-α and the retinoid X receptor inhibit inflammatory responses of microglia. J Neurosci Res. https://doi.org/10.1002/jnr.20518
  130. , , , , , (). Therapeutic effects of fenofibrate on diabetic peripheral neuropathy by improving endothelial and neural survival in db/db mice. PLoS One. https://doi.org/10.1371/journal.pone.0083204
  131. , , , , , (). Peroxisome proliferator-activated receptor-α activation attenuates 3-nitropropionic acid induced behavioral and biochemical alterations in rats: possible neuroprotective mechanisms. Eur J Pharmacol. https://doi.org/10.1016/j.ejphar.2011.10.029
  132. , , , (). Regulation of peroxisome proliferator-activated receptor-α expression during lung inflammation. Pulm Pharmacol Ther. https://doi.org/10.1016/j.pupt.2007.08.001
  133. , , , , , (). PPARα downregulates airway inflammation induced by lipopolysaccharide in the mouse. Respir Res. https://doi.org/10.1186/1465-9921-6-91
  134. , , , , (). Fenofibrate attenuates neutrophilic inflammation in airway epithelia: potential drug repurposing for cystic fibrosis. Clin Transl Sci. https://doi.org/10.1111/cts.12310
  135. , , , , (). Modulation of the IL-23/IL-17 axis by fenofibrate ameliorates the ovalbumin/lipopolysaccharide-induced airway inflammation and bronchial asthma in rats. Naunyn Schmiedebergs Arch Pharmacol. https://doi.org/10.1007/s00210-017-1459-z
  136. , , , , (). Suppression of allergen-induced airway inflammation and immune response by the peroxisome proliferator-activated receptor-alpha agonist fenofibrate. Eur J Pharmacol. https://doi.org/10.1016/j.ejphar.2007.11.040
  137. , , , , , (). Fibrates ameliorate the course of bacterial sepsis by promoting neutrophil recruitment via CXCR2. EMBO Mol Med. https://doi.org/10.1002/emmm.201303415
  138. , , , , , (). Small heterodimer partner-targeting therapy inhibits systemic inflammatory responses through mitochondrial uncoupling protein 2. PLoS One. https://doi.org/10.1371/journal.pone.0063435
  139. , , , , , (). Pretreatment with peroxysome proliferator-activated receptor α agonist fenofibrate protects endothelium in rabbit Escherichia coli endotoxin-induced shock. Intensive Care Med. https://doi.org/10.1007/s00134-005-2730-1
  140. , , , , , (). Effect of peroxisome proliferator-activated receptor alpha activators on tumor necrosis factor expression in mice during endotoxemia. Infection and immunity. https://doi.org/10.1128/IAI.67.7.3488-3493.1999
  141. , , , , (). Apolipoprotein CIII overexpression-induced hypertriglyceridemia increases nonalcoholic fatty liver disease in association with inflammation and cell death. Oxid Med Cell Longev. https://doi.org/10.1155/2017/1838679
  142. , , , (). Fenofibrate, a peroxisome proliferator-activated receptor-alpha agonist, blocks steatosis and alters the inflammatory response in a mouse model of inflammation-dioxin interaction. Chem Biol Interact. https://doi.org/10.1016/j.cbi.2021.109521
  143. , , , , , (). GC-MS metabolomics on PPARα-dependent exacerbation of colitis. Mol Biosyst. https://doi.org/10.1039/C5MB00048C
  144. , , , , , (). Hepatotoxic effects of fenofibrate in spontaneously hypertensive rats expressing human C-reactive protein. Physiol Res. https://doi.org/10.33549/physiolres.933304
  145. , , , , , (). Peroxisome proliferator–activated receptor-α agonism with fenofibrate does not suppress inflammatory responses to evoked endotoxemia. J Am Heart Assoc. https://doi.org/10.1161/JAHA.112.002923

Conflicts of interest: Authors declare no conflicts of interest.

Funding: The study was supported by grants from the National Natural Science Foundation of China (No. 82000642) and the Nanjing Health Science and Technology Development Foundation (No.YKK22168).