Next Article in Journal
Role of Deubiquitinases in Parkinson’s Disease—Therapeutic Perspectives
Next Article in Special Issue
Selective PPAR-Delta/PPAR-Gamma Activation Improves Cognition in a Model of Alzheimer’s Disease
Previous Article in Journal
The Role of Peroxisome Proliferator-Activated Receptors in Preeclampsia
Previous Article in Special Issue
The Role of PPARs in Breast Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Peroxisome Proliferator-Activated Receptor-Targeted Therapies: Challenges upon Infectious Diseases

1
Department of Microbiology, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
2
Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
3
Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
4
Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
*
Author to whom correspondence should be addressed.
Cells 2023, 12(4), 650; https://doi.org/10.3390/cells12040650
Submission received: 9 December 2022 / Revised: 13 February 2023 / Accepted: 14 February 2023 / Published: 17 February 2023
(This article belongs to the Special Issue The Role of PPARs in Disease II)

Abstract

:
Peroxisome proliferator-activated receptors (PPARs) α, β, and γ are nuclear receptors that orchestrate the transcriptional regulation of genes involved in a variety of biological responses, such as energy metabolism and homeostasis, regulation of inflammation, cellular development, and differentiation. The many roles played by the PPAR signaling pathways indicate that PPARs may be useful targets for various human diseases, including metabolic and inflammatory conditions and tumors. Accumulating evidence suggests that each PPAR plays prominent but different roles in viral, bacterial, and parasitic infectious disease development. In this review, we discuss recent PPAR research works that are focused on how PPARs control various infections and immune responses. In addition, we describe the current and potential therapeutic uses of PPAR agonists/antagonists in the context of infectious diseases. A more comprehensive understanding of the roles played by PPARs in terms of host-pathogen interactions will yield potential adjunctive personalized therapies employing PPAR-modulating agents.

1. Introduction

Peroxisome proliferator-activated receptors (PPARs) are adopted orphan family members of the nuclear receptor group that regulates various biological functions, including glucose and lipid homeostasis, inflammation, and adipose cell differentiation [1,2]. PPARs are ligand-activated transcription factors that are subdivided into three isoforms, termed PPARα (NR1C1), PPARβ/δ (also termed PPARβ or PPARδ, or NR1C2), and PPARγ (NR1C3) [3]. The endogenous PPAR ligands include long-chain polyunsaturated fatty acids and eicosanoids, although the functions of the ligands remain largely unknown [2,4]. Each PPAR isoform evidences a distinct cellular and tissue distribution and biological functions with a focus on energy balance and inflammation [2].
PPARs feature N-terminal DNA-binding and C-terminal ligand-binding domains and form heterodimers with nuclear retinoid X receptor (RXR)-α [5,6]. After interacting with the ligands, PPAR-RXR heterodimers undergo conformational changes that allow them to regulate the transcription of many genes with peroxisome proliferator response elements (PPREs) in their promoter regions [7]. The many PPAR-mediated functions are orchestrated via recruitment of different transcriptional co-activators, including PPAR co-activator-1α, co-activator-associated proteins, and co-repressors [2,5]. Moreover, each PPAR isoform transcriptionally regulates the expression of the other PPAR isoforms via feedback control [2].
PPARα is found principally in the liver and transcriptionally regulates fatty acid oxidation, cholesterol and glycogen metabolism, gluconeogenesis, ketogenesis, and inflammation [8,9]. PPARγ is found in both hematopoietic and non-hematopoietic cells and tissues (adipose tissue and the large intestine) [10]. PPARγ modulates many biological functions, including fatty acid and glucose metabolism and anti-inflammatory signaling via nuclear factor kappa B (NF-κB); it also suppresses oxidative stress and prevents platelet-leukocyte interactions [10,11]. Recent insights into the roles played by PPAR ligands have enabled development of PPAR agonists/antagonists, which serve as candidate drugs for inflammatory, metabolic, and autoimmune diseases, as well as cancers [12]. Several PPARα ligands, including fibrates, helpfully treat dyslipidemia, while the PPARγ ligands pioglitazone and rosiglitazone are well-known anti-diabetic drugs [13]. The three PPARs play critical but distinct roles in regulating the inflammation and metabolic pathways closely associated with immune cell functions [14,15,16]. It is thus essential to understand how PPARs affect antimicrobial actions against diverse infections. Here, we highlight recent insights into how the PPAR isoforms and their agonists regulate antimicrobial host defenses against viral, bacterial, and parasitic diseases.

2. Overview of PPARs

2.1. Molecular Characteristics of PPARs

Peroxisomes, 0.5 μm diameter single-membrane cytoplasmic organelles, play essential roles in the oxidation of various biomolecules [17,18]. Peroxisome proliferators are multiple chemicals that increase the abundance of peroxisomes in cells [19,20]. These molecules also increase gene expression for β-oxidation of long-chain fatty acids and cytochrome P450 (CYP450) [21,22]. Given the gene transcriptional modulation of peroxisome proliferators, PPARs have been identified as nuclear receptors [23,24,25,26,27,28,29]. The PPAR subfamily consists of three isoforms, PPARα, PPARβ/δ, and PPARγ [30]. The three PPARs differ in tissue-specific expression patterns and ligand-biding domains, each performing distinct functions. PPARA, encoding PPARα, is located in chromosome 22q13.31 in humans and is mainly expressed in the liver, intestine, kidney, heart, and muscle [31,32]. PPARγ has four alternative splicing forms from PPARG located in chromosome 3p25.2 and is highly expressed in adipose tissue, the spleen, and intestine [33,34]. PPARδ, encoded by PPARD, is located in chromosome 6q21.31 and presents ubiquitously [29,35]. Thus, it is essential in the study of PPARs to consider their tissue distribution and functions.
PPAR is a nuclear receptor superfamily class II member that heterodimerizes with RXR [36,37]. The PPAR structure includes the A/B, C, D, and E domains from N-terminus to C-terminus [38]. The N-terminal A/B domain (NTD) is a ligand-independent transactivation domain containing the activator function (AF)-1 region. The NTD is targeted for variable post-translational modifications, including SUMOylation, phosphorylation, acetylation, O-GlcNAcylation, and ubiquitination, resulting in transcriptional regulatory activities [39]. DNA-binding C domain (DBD) has two DNA-binding zinc finger motifs containing cysteines, which dock to PPREs. PPARs reside upstream of RXR upon the direct repeat (DR)-1 motifs, which are composed of two hexanucleotide consensus sequences with one spacing nucleotide (AGGTCA N AGGTCA) [40]. The hinge D region is a linker between the C and E domains, which contains a nuclear localization signal, and is the site for post-translational modifications such as phosphorylation, acetylation, and SUMOylation [39]. The ligand-binding E domain (LBD) carries the hydrophobic ligand-binding pocket and the AF-2 region. The absence of agonists enables LBD to recruit co-repressors containing the CoRNR motifs [41]. Engaging agonists to LBD elicits conformational changes of AF-2 to facilitate interaction with LXXLL motifs of many co-activators [42]. Like other nuclear receptor superfamily class II members, such as thyroid hormone receptor (TR), retinoic acid receptor (RAR), and vitamin D receptor (VDR), PPARs function as heterodimers with RXR through LBD [6,43]. LBD is also targeted for SUMOylation and ubiquitination [39]. Advancement of research on the PPAR structure helps thoroughly dissect the roles of PPARs. We will discuss the roles of specific PPAR subtypes in the following subsections.

2.1.1. Roles of PPARα

PPARα is predominantly expressed in the liver but is also found in other tissues, including the heart, muscle, and kidney [4,32]. PPARα regulates the expression of genes involved in metabolism and inflammation. Activation of PPARα leads to the upregulation of genes involved in fatty acid oxidation and the downregulation of genes involved in fatty acid synthesis [8]. PPARα also modulates other genes, including genes involved in the transport and uptake of fatty acids and the synthesis and secretion of lipoproteins [4,8]. In addition, activation of PPARα has been shown to improve insulin sensitivity, reduce oxidative stress, and reduce inflammation in preclinical studies [7,8,44]. PPARα activation has been shown to modify the expression of immune response genes, including those encoding cytokines and chemokines, which are signaling molecules that regulate the immune response [44,45]. PPARα activation has also been demonstrated to reduce the production of pro-inflammatory cytokines, such as tumor necrosis factor-alpha (TNF-α) and interleukin (IL)-6 [46,47]. PPARα has been shown to interfere with the DNA binding of both AP-1 and NF-κB [45,46,48]. Thus, the roles of PPARα in infectious diseases should be studied in wide ranging aspects, including metabolism and inflammation.
In the context of infection, PPARα has been shown to play an essential role in the hepatic metabolic response to infection. During an infectious challenge, the liver coordinates several metabolic changes to support the host defense response, including the mobilization of energy stores, production of acute-phase proteins, and synthesis of new metabolites. Activation of PPARα in the liver leads to the upregulation of genes involved in fatty acid oxidation and ketogenesis with fibroblast growth factor 21 (FGF21) production [49]. FGF21 is a hormone produced by the liver that has been shown to promote ketogenesis and reduce glucose utilization [50,51]. The ketogenesis regulation of PPARα with FGF21 is essential for reacting to microbial or viral sepsis [52,53,54]. In conclusion, the hepatic PPARα metabolic response to infection is crucial to the host defense response.

2.1.2. Roles of PPARβ/δ

PPARβ is expressed in diverse tissues, including adipose tissue, muscle, and the liver [29,55], and is activated by multiple ligands, such as fatty acids and their derivatives [7]. PPARβ is involved in regulating lipid metabolism and energy homeostasis, as well as controlling inflammation and immune function [56]. PPARβ activation has been demonstrated to have pro- and anti-inflammatory effects based on the situation [56]. The role of PPARβ in tumorigenesis is debatable. PPARβ activation has been found in some cases to have anti-tumorigenic effects, such as causing apoptosis and inhibiting cell proliferation [57,58]. In other cases, however, activation of PPARβ has been shown to promote tumorigenesis by enhancing cell survival, promoting angiogenesis, and reducing cellular differentiation [59,60,61,62]. Overall, the role of PPARβ activation in cancer is not entirely known and is complex. Similarly, the function of PPARβ in infection is not well understood. Additional research is required to comprehend the function of PPARβ in the context of immunology against cancers and infectious diseases.

2.1.3. Roles of PPARγ

PPARγ is expressed in a variety of tissues, including adipose tissue, muscle, and the liver [33,34,55], and is activated by diverse ligands, including fatty acids and their derivatives, as well as synthetic chemicals known as thiazolidinediones [4,7]. PPARγ is responsible for regulating lipid metabolism, glucose homeostasis, and inflammation [63,64]. Numerous inflammatory mediators and cytokines are inhibited by PPARγ ligands in various cell types, including monocytes/macrophages, epithelial cells, smooth muscle cells, endothelial cells, dendritic cells, and lymphocytes. In addition, PPARγ diminishes the activities of transcription factors AP-1, STAT, NF-κB, and NFAT to adversely regulate inflammatory gene expression [65,66,67]. As a result, PPARγ has been demonstrated to have a protective function against infections by modulating the immune response and lowering inflammation. However, other researchers have hypothesized that PPARγ activation may impair the function of immune cells, such as macrophages, and contribute to the development of infections. Therefore, the role of PPARγ in disease is complex and context-dependent, and more research is needed to fully understand the molecular mechanisms by which PPARγ regulates the host response to infection.

2.2. Regulatory Mechanisms of PPARs

The PPAR ligand-binding pocket is large and capable of engaging diverse ligands [68,69]. Endogenous ligands vary depending on the PPAR isoform, including n-3 polyunsaturated fatty acids such as docosahexaenoic acid and eicosapentaenoic acid for all PPARs, leukotriene B4 for PPARα, carbaprostacyclin for PPARδ, and prostaglandin J2 for PPARγ [70]. Representative synthetic agonists include fibrates (PPARα agonists) and thiazolidinediones (PPARγ agonists) [7]. Fibrates, such as fenofibrate, clofibrate, and gemfibrozil, are widely used for treating dyslipidemia. Thiazolidinediones, such as rosiglitazone, pioglitazone, and lobeglitazone, improve insulin resistance [7]. Most clinical studies on PPAR actions in infectious diseases have been conducted retrospectively, and no clinical studies currently in progress are listed in ClinicalTrials.gov (https://clinicaltrials.gov/ (accessed on 13 February 2023)). Since widely used PPAR agonists exist, clinical research can be conducted through a deeper understanding of PPAR roles in infectious diseases.
PPAR-RXR heterodimerization occurs ligand-independently [6]. The heterodimer appears to exert transcriptional regulation both ligand-dependently and -independently [7]. Although LBD may interact with either co-repressor or co-activator in the state of not binding with an agonist, binding to a ligand elicits stabilized co-activator-LBD interaction, thus increasing transactivation [7,71]. Further, recent studies have shown that PPARs inhibit other transcription factors, such as NF-κB, activator protein-1 (AP-1), signal transducer and activator of transcription (STAT), and nuclear factor of activated T cells (NFAT) [44,65,66,67]. Recent studies revealed the possibility of forming a protein chaperone complex with PPAR-associated proteins, such as heat shock proteins (HSPs). Similar to interactions between other type I intracellular receptors and heat shock proteins, HSP90 repressed PPARα and PPARβ activities but not that of PPARγ [72]. Instead, HSP90 was required for PPARγ signaling in the nonalcoholic fatty liver disease mouse model [73]. Thus, it is necessary to study the various modes of PPAR actions. The intracellular regulatory mechanisms of PPARs are shown in Figure 1.

3. PPARs and Viral Infections

3.1. PPARs and Respiratory Viral Infections

Many studies have shown that PPARγ controls viral replication and virus-associated inflammation by antagonizing inflammatory signaling pathways such as the NF-κB and STAT pathways [74,75]. In particular, PPARγ of alveolar macrophages critically modulates acute inflammation to promote recovery from respiratory viral infections, most of which are caused by influenza A virus (IAV) and respiratory syncytial virus (RSV) [76]. Several PPAR agonists have shown promise in terms of ameliorating virus-related cytokine storms and the damage caused by severe IAV infection [77]. Macrophage PPARγ is essential for resolving the chronic pulmonary collagen deposition and fibrotic changes that follow influenza infection [78]. Several researchers have sought new therapeutic candidates for IAV disease. A recent screening of traditional Chinese medicines showed that emodin and analogs thereof evidenced excellent anti-IAV activities mediated by activation of the PPARα/γ and adenosine monophosphate (AMP)-activated protein kinase (AMPK) pathways [79]. High-throughput screening of natural compounds and/or synthetic drugs/agents will yield new therapeutics against respiratory viral infections based on drug interactions with PPAR pathways.
A link has been suggested between severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus infection and PPARα activity in the context of lipid uptake, lipotoxicity, and vascular inflammation [80,81,82]. The PPARα agonist fenofibrate is a potential adjunctive coronavirus disease (COVID-19) therapy; the material exhibits anti-inflammatory and anti-thrombotic activities [80,82]. A study employing a public database on subjects with type 2 diabetes and COVID-19, along with animal studies, revealed that the PPARγ agonist pioglitazone may ameliorate acute lung injury and SARS-CoV-2-mediated hyperinflammation [83]. Cannabidiol working via PPARγ is proposed as a therapeutic approach for the severe form of COVID-19 [84]. A recent study demonstrated that cannabidiol attenuated inflammation and epithelial damage in colonic epithelial cells exposed to the SARS-CoV-2 spike protein through a PPARγ-dependent mechanism [85]. The natural compound γ-oryzanol may also serve as an adjunctive therapy to reduce the cytokine storm associated with COVID-19; the material stimulated PPARγ to modulate oxidative stress and the inflammatory response in adipose tissues [86]. The Middle East respiratory syndrome coronavirus (MERS-CoV)-derived S glycoprotein activates PPARγ to suppress the pathologic inflammatory responses of macrophages [87]. Further research on the modulatory roles played by PPAR agonists/antagonists in terms of virus-associated inflammation will yield novel adjunctive therapeutics to counter emerging and re-emerging viral infections. Table 1 summarizes studies on PPARs and their ligands in relation to viral infections.

3.2. PPARs and Virus-Related Inflammation

A recent study showed that the inflammatory responses during infection with Chikungunya virus (CHIKV) involved the renin-angiotensin system (RAS) and PPARγ pathways [88]. The telmisartan-mediated suppression of CHIKV infection is at least partly mediated via activation of PPARγ; a PPARγ antagonist increased the CHIKV viral load [88]. Omeragic et al. showed that PPARγ played a critical role in terms of human immunodeficiency virus (HIV-1) ADA glycoprotein 120 (gp120)-related inflammatory marker generation was observed in primary astrocytes and microglia and also in vivo [89]. The anti-inflammatory activities induced by the PPARγ agonists rosiglitazone and pioglitazone reflected suppression of the NF-κB signaling pathway [89]. These relationships between PPARγ and viral infections are included in Table 1. Δ-9-tetrahydrocannabinol improved epithelial barrier function and thus protected colonic tissues of rhesus macaques chronically infected with simian immunodeficiency virus (SIV). This was at least partly attributable to the upregulation of PPARγ [92]. PPARα signaling is required for restoration of the intestinal barrier by the probiotic Lactobacillus plantarum and amelioration of gut inflammation during SIV infection [93]. Such findings strongly suggest that targeting PPARγ would both prevent and treat virus-associated inflammation of the brain, endothelial system, and intestinal tissues. The PPARγ antagonist GW9662 protected against dengue virus infection and di(2-ethylhexyl) phthalate (DEHP)-induced interleukin (IL)-23 expression, thus suppressing the viral load [94]. Therefore, future clinical trials should explore the protective effects of several possible PPAR agonists/antagonists and combinations thereof with current antivirals in patients with various viral infections.
Zika virus (ZIKV) is a serious arthropod-borne (arbovirus) pathogen that causes congenital defects and neurological diseases in both infants and adults [95]. A recent study showed that ZIKV-induced cellular responses of induced pluripotent stem cell (iPSC)-derived neural progenitor cells involved the PPAR signaling pathways, which may contribute to neurogenesis and viral replication [96]. However, further research is required.

3.3. PPARs and Hepatitis Virus Infection

The roles played by PPAR pathways in terms of hepatitis B virus (HBV) infection elimination are complex. IL-1β production induced by HBV infection of M1-like inflammatory macrophages triggered anti-HBV responses via downregulation of PPARα and forkhead box O3 (FOXO3) expression in hepatocytes [97]. OSS_128167, a sirtuin 6 inhibitor, inhibited HBV transcription and replication in hepatic cells and in vivo by targeting PPARα expression [90]. In the HBV replicative mouse model, PPAR agonists, including bezafibrate, fenofibrate, and rosiglitazone, significantly increased the serum levels of HBV antigens HBsAg, HBeAg, and HBcAg and that of HBV DNA, as well as the viral load in mouse liver [98]. Thus, patients with metabolic diseases taking PPAR-based therapeutics should take care to avoid HBV infection. However, in a retrospective study of HBV-infected patients treated with entecavir and tenofovir-disoproxil-fumarate, the drugs exerted profound extrahepatic effects on lipid metabolism, reducing serum cholesterol levels by inducing the expression of PPARα target genes such as CD36 in liver tissue and cells [99]. Thus, the PPARα-activating nucleoside analogs tenofovir-disoproxil-fumarate may usefully treat atherosclerosis and hepatocarcinogenesis, both of which are associated with dyslipidemia. This would be a new role for an anti-HBV therapeutic. However, the precise functions of PPARs during HBV infection remain unclear. The antiviral, antitumor, and extrahepatic actions of PPAR agonists vary with the clinical condition.
During hepatitis C virus (HCV) infection, PPAR-α/β/γ stimulators/agonists reduce calcitriol-mediated anti-HCV responses, presumably by counteracting the calcitriol-mediated activation of vitamin D receptor signaling and inhibiting nitrative stress [91]. Naringenin, a grapefruit flavonoid, suppressed HCV production by inhibiting viral particle assembly via PPARα activation, suggesting potential roles for PPARα agonists in the resolution of infection [100]. It is essential to perform an in-depth exploration of how the three PPARs and their signaling pathways affect the outcomes of HBV and HCV infections. Studies on PPARs and hepatitis virus infections are summarized in Table 1.

4. PPARs and Bacterial Infections

4.1. PPARs and Post-Influenza Bacterial Infections

PPARs exacerbate the severity of post-influenza bacterial infections. During Staphylococcus aureus superinfection following IAV infection, the levels of CYP450 metabolites, which are PPARα ligands, increase significantly and trigger receptor-interacting serine/threonine-protein kinase 3 (RIPK3)-induced necroptosis, thus exacerbating the lung pathology and increasing mortality from secondary bacterial infection [101]. The PPARγ agonist rosiglitazone reduces bacterial clearance during secondary bacterial pneumonia, which is a frequent complication of primary IAV infection [102]. Diabetic patients treated with rosiglitazone exhibited increased mortality from IAV-associated pneumonia compared to those not treated with rosiglitazone, as revealed by data from the National Health and Nutrition Examination Survey (NHANES) [102]. CYP450 metabolites reduced the protective inflammatory responses via PPARα activation, thereby increasing the susceptibility to secondary bacterial infection following IAV infection [103]. Thus, PPARα or PPARγ drives host protection but reduces bacterial clearance at different stages of IAV infection. The molecular mechanisms by which PPARα/γ mediates immune modulation during a bacterial infection following IAV infection require urgent attention. Better medicines are needed to treat the different stages of IAV-associated disease, which is often fatal in susceptible patients.

4.2. PPARs in Bacterial Infections

PPARs and agonists/antagonists thereof may modulate disease severity and outcomes in patients with bacterial infections and associated inflammation. In a model of intestinal colitis, 5-aminosalicylic acid, a PPARγ agonist, exerted therapeutic anti-inflammatory effects by activating the epithelial PPARγ signaling pathway [104]. After infection with Klebsiella pneumoniae, which is the respiratory Gram-negative bacterium that usually causes pneumonia, PPARγ agonists such as pioglitazone reduced proinflammatory cytokine and myeloperoxidase levels, bacterial growth in lung tissues, and bacterial dissemination to distant organs [105]. The taste receptor type-2 member 138 (TAS2R138) plays a role in neutrophil-associated host innate immune defense after Pseudomonas aeruginosa infection [106]. TAS2R138 mediated the degradation of lipid bodies via competitive binding to the PPARγ antagonist N-(3-oxododecanoyl)-L-homoserine lactone (AHL-12), a mediator of virulence produced by P. aeruginosa [106]. Although the exact roles of PPARγ in antimicrobial responses remain unclear, a study employing a model of P. aeruginosa infection found that the PPARγ agonist pioglitazone increased the levels of certain chemokines (Cxcl1, Cxcl2, and Ccl20) and cytokines (Tnfa, Il6, and Cfs3) in bronchial epithelial cells and suppressed inflammatory responses in bronchoalveolar lavage fluid [107]. Future studies must explore the utility of PPAR agonists/antagonists as adjuvant therapies and determine whether systemic or local treatments improve disease outcomes.
During Chlamydia pneumoniae infection, both PPARα and PPARγ are required to upregulate foam macrophage formation via induction of the scavenger receptor A1 (SR-A1) and the acyl-coenzyme A cholesterol acyltransferase 1 (ACAT1) involved in cholesterol esterification [108]. PPARα and PPARγ agonists, including fenofibrate and rosiglitazone, may suppress atherosclerotic plaque formation in patients with coronary heart disease infected with C. pneumoniae [108]. Activation of both PPARα and PPARγ by PAR5359 protected against Citrobacter rodentium-induced colitis. The dual agonism promoted antibacterial immunity and ameliorated the inflammatory response [109].
In contrast to studies with Gram-negative bacteria, few reports have explored the roles played by PPARs during Gram-positive infections. In a Caenorhabditis elegans model, induction of the gene encoding flavin-containing monooxygenase (FMO) fmo-2/FMO5 by NHR-49/PPAR-α was critical in terms of the establishment of an effective innate host defense against S. aureus infection [110]. Erythropoietin limits infections caused by Gram-negative Escherichia coli and Gram-positive S. aureus; macrophage-mediated clearance of these bacteria is at least partly mediated by a PPARγ-dependent pathway [111]. Inhibition of PPARγ signaling reduced the survival of Rickettsia conorii, an intracellular Gram-positive bacterium, probably by reducing lipid droplet production [112]. Although PPAR-based therapeutics may counter bacterial infections, more preclinical and clinical studies are required. Table 2 summarizes the roles of PPAR ligands in bacterial infections.

4.3. PPARs and Mycobacterial Infections

Many scholars have sought to clarify the effects of PPARs in those infected with Mycobacterium tuberculosis (Mtb) and nontuberculous mycobacteria (NTM), which cause tuberculosis and NTM disease, respectively [113]. Although the relevant bacterial components have not been fully characterized, M. leprae and Mtb lead to activation of PPARs [113,114,115]. PPARα and PPARγ appear to play opposite roles. The virulent Mtb strain H37Rv and cell wall component lipoarabinomannan induced PPARγ expression, in turn activating IL-8 and cyclooxygenase (COX) 2 expression, but the attenuated M. bovis strain, termed Bacillus Calmette-Guérin (BCG), induced less PPARγ expression [115]. PPARγ activation during Mtb or BCG infection upregulates lipid body formation and increases bacterial survival in macrophages [116,117]. Either PPARγ knockdown or PPARγ antagonist GW9662 increased macrophage-mediated Mtb killing [115,117]. PPARγ activation was associated with enhanced cholesterol and triacylglycerol uptake; these materials are required for macrophage lipid body formation during mycobacterial infection [113]. Antagonists of PPARδ or PPARγ significantly inhibited lipid accumulation by cells infected with M. leprae, thus reducing parasitization [114,118]. Together, the data suggest that PPARγ is required for intracellular bacterial survival; PPARγ enhances lipid body formation and foam macrophage development during mycobacterial infection.
In contrast, PPARα appears to enhance defenses against macrophage and lung Mtb or BCG infection in mice. PPARα-mediated antimicrobial responses are at least partly mediated via promotion of lipid catabolism and activation of the transcription factor EB (TFEB), a transcriptional factor required for lysosomal biogenesis [119]. Notably, PPARα agonists GW7647 and Wy14643 protected macrophages against Mtb or BCG infection [119]. Macrophage PPARα expression reduces inflammatory cytokine synthesis during Mtb or BCG infection [119], suggesting that PPARα ameliorates inflammation. PPARα deficiency reduced the antimicrobial response and increased lung tissue damage during pulmonary Mycobacteroides abscessus (Mabc) infection [120]. Gemfibrozil, a PPARα activator, reduced the in vivo Mabc load and lung inflammation during infection [120]. It is important to clarify whether PPARα modulates lipid body formation during infections with Mabc and other NTMs.

5. PPARs and Parasitic Infections

The anti-inflammatory responses of M2 macrophages and Th2 immunity protect against parasitic infections [121]. In allergic patients and those infected with the nematode Heligmosomoides polygyrus, PPARγ is highly expressed in Th2 cells. PPARγ affects the development of Th2-associated pathological immune responses and increases IL-33 receptor levels in Th2 cells [122]. Neospora caninum infection triggers maturation of M2 macrophage development via upregulation of PPARγ activity and downregulation of NF-κB signaling [123]. In a model of eosinophilic meningoencephalitis caused by the rat lungworm Angiostrongylus cantonensis, PPARγ played anti-inflammatory and protective roles by inhibiting NF-κB-mediated pathological inflammatory responses; the PPARγ antagonist GW9662 increased susceptibility to angiostrongyliasis [124]. In a model of cerebral malaria using clinical isolates of Plasmodium falciparum, dimethyl fumarate increased the expression of nuclear factor E2-related factor 2 (NRF2), in turn enhancing PPAR signaling and thus ameliorating the neuroinflammatory responses of primary human brain microvascular endothelial cells [125]. Cerebral malaria susceptibility was associated with a lack of PPARγ nuclear translocation and increased COX-2 levels in brain tissues, which was associated with higher-level parasitemia and poorer survival [126]. PPAR signaling may exert useful antiparasitic functions by attenuating inflammation.
Toxoplasma gondii, one of the most common zoonotic pathogens, infects both immunocompromised patients and healthy individuals and most commonly targets the central nervous system [127]. In T. gondii-infected astroglia, the PPARγ agonist rosiglitazone reduced neuroinflammation, whereas the PPARγ antagonist GW9662 increased levels of matrix metalloprotease (MMP)-2, MMP-9, and inflammatory mediators. These findings suggested that PPARγ signaling protects against T. gondii infection [128]. Proteomic analysis showed that the hepatic protein responses to T. gondii infection modulated the PPAR signaling pathways to dysregulate further liver lipid metabolism [129]. However, it remains unclear how T. gondii-mediated modulation of PPARγ signaling affects such metabolism and the consequence thereof.
Sometimes, PPAR signaling negatively affects host defenses against parasitic infections, particularly when M2 macrophage responses are associated with disease progression. During infection of Balb/c mice and hamsters with Leishmania donovani, a causative agent of visceral leishmaniasis, the mRNA expression levels of IL-4- and IL-10-driven markers increased significantly [130]. Although any IL-4-related PPARγ function remains unclear, the parasitic load correlated with the effects of IL-10 on the hamster spleen [130]. Schistosomiasis (bilharzia), caused by parasitic flatworms of the genus Schistosoma, is associated with inflammatory responses of the intestinal, hepato-splenic, and urogenital systems [131,132]. The Sm16/SPO-1/SmSLP protein from S. mansoni may allow the parasite to escape the actions of the innate immune pathway and cellular metabolism, at least partly via a PPAR-dependent pathway [133]. The Trypanosoma cruzi protozoan causes Chagas disease, a neglected but chronic tropical infection of great concern in Latin America [134]. During T. cruzi infection, both PPARα and PPARγ agonists appear to be involved in macrophage polarization from M1 to M2 types, thereby suppressing inflammation but increasing phagocytosis and macrophage parasitic loads [135]. Thus, PPAR functions may vary by parasite and experimental model. Future studies must explore whether PPARs trigger host defenses or immune evasion during parasitic infections.
Several PPAR ligands may serve as useful adjunct therapies for Chagas disease, although more preclinical and clinical data are required. The new PPARγ ligand HP24, a pyridinecarboxylic acid derivative, evidenced anti-inflammatory and pro-angiogenic activities and might serve as an adjunct therapy for Chagas disease [136]. 15-deoxy-Δ12,14 prostaglandin J2 (15dPGJ2), a natural PPARγ agonist, reduced liver inflammation and fibrosis during T. cruzi infection [137]. However, the use of PPAR agonists/antagonists should be considered in the context of in vivo PPAR expression levels during certain parasitic infections. For example, acute T. cruzi mouse infections trigger significant adipose tissue loss and dysregulation of lipolytic and lipogenic enzymes, which are associated with decreased adipocyte PPAR-γ levels in vivo [138]. Given the robust PPARγ inhibition in this mouse model, PPARγ agonists were minimally effective. However, certain parasites do not specifically affect the host responses depending on PPAR down- or upregulation in target tissues or cells. After infection with the intestinal parasite Giardia muris, rapid PPARα induction did not affect the protective or pathological immune responses; PPARα-deficient mice cleared the parasite as did wild-type controls [139]. It is important to explore whether aberrant PPAR expression induced by different parasites improves disease status or, rather, enhances dysfunctional inflammation and infection progression. Table 3 summarizes the roles of PPAR agonists/antagonists in parasitic infections.

6. Future Perspectives

PPARs play a wide range of roles across host metabolism, inflammation, and immune responses. Recent studies indicate that PPARs modulate the host responses to infections, such as infectious agent clearance and inflammation. Several PPAR ligands have been utilized in infection models and their functions have been investigated. However, there are no clinical trials of well-known, licensed metabolic medicines utilizing PPAR pathways for infectious diseases. PPAR-based future drugs may serve as adjuvants or components of combination therapies against infections. Understanding the fundamental processes of PPAR-mediated host immune regulation is necessary to develop the most effective treatment approaches for infectious diseases. Future research may also benefit from developing synthetic ligands that preferentially target the specific PPAR isoform implicated in immune response modification.

7. Conclusions

Accumulating evidence suggests that PPARs are involved in the host responses to infections caused by bacteria, viruses, and parasites. However, the molecular mechanisms by which PPARs modulate disease progression or protective responses remain unknown. It is essential to further explore the PPAR functions and mechanisms involved in pathogen survival, the pathological responses during different stages of infection, and the associated modulation of the distinct types of infection-associated acute and chronic inflammation. Apart from shaping the inflammatory and metabolic responses during infections, PPARs may impact disease outcomes. The PPAR signaling pathways exert potent immunomodulatory effects; pathway activation or suppression may usefully treat infectious diseases. Infectious pathogens modulate the individual and collaborative activities of PPAR(s) during infection. We speculate that aberrant PPAR expression by various parasites may contribute to inflammation-related dysfunction. It is essential to better understand the possible clinical effects of PPAR-based therapeutics in patients with various infectious diseases.

Author Contributions

Conceptualization, writing—original draft preparation, review and editing, visualization, E.-K.J., I.S.K. and P.S.; supervision, project administration, funding acquisition, E.-K.J. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (grant number: 2017R1A5A2015385). Additionally, this research was supported by the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI) funded by the Ministry of Health & Welfare, Republic of Korea (grant number: HI22C1361).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Peters, J.M.; Shah, Y.M.; Gonzalez, F.J. The role of peroxisome proliferator-activated receptors in carcinogenesis and chemoprevention. Nat. Rev. Cancer 2012, 12, 181–195. [Google Scholar] [CrossRef] [Green Version]
  2. Toobian, D.; Ghosh, P.; Katkar, G.D. Parsing the Role of PPARs in Macrophage Processes. Front. Immunol. 2021, 12, 783780. [Google Scholar] [CrossRef]
  3. Schupp, M.; Lazar, M.A. Endogenous ligands for nuclear receptors: Digging deeper. J. Biol. Chem. 2010, 285, 40409–40415. [Google Scholar] [CrossRef] [Green Version]
  4. Lamas Bervejillo, M.; Ferreira, A.M. Understanding Peroxisome Proliferator-Activated Receptors: From the Structure to the Regulatory Actions on Metabolism. Adv. Exp. Med. Biol. 2019, 1127, 39–57. [Google Scholar] [CrossRef]
  5. Viswakarma, N.; Jia, Y.; Bai, L.; Vluggens, A.; Borensztajn, J.; Xu, J.; Reddy, J.K. Coactivators in PPAR-Regulated Gene Expression. PPAR Res. 2010, 2010, 250126. [Google Scholar] [CrossRef] [Green Version]
  6. Chandra, V.; Huang, P.; Hamuro, Y.; Raghuram, S.; Wang, Y.; Burris, T.P.; Rastinejad, F. Structure of the intact PPAR-gamma-RXR- nuclear receptor complex on DNA. Nature 2008, 456, 350–356. [Google Scholar] [CrossRef] [Green Version]
  7. Grygiel-Górniak, B. Peroxisome proliferator-activated receptors and their ligands: Nutritional and clinical implications--a review. Nutr. J. 2014, 13, 17. [Google Scholar] [CrossRef] [Green Version]
  8. Ye, X.; Zhang, T.; Han, H. PPARα: A potential therapeutic target of cholestasis. Front. Pharmacol. 2022, 13, 916866. [Google Scholar] [CrossRef]
  9. Bordet, R.; Ouk, T.; Petrault, O.; Gelé, P.; Gautier, S.; Laprais, M.; Deplanque, D.; Duriez, P.; Staels, B.; Fruchart, J.C.; et al. PPAR: A new pharmacological target for neuroprotection in stroke and neurodegenerative diseases. Biochem. Soc. Trans. 2006, 34, 1341–1346. [Google Scholar] [CrossRef] [Green Version]
  10. Marion-Letellier, R.; Savoye, G.; Ghosh, S. Fatty acids, eicosanoids and PPAR gamma. Eur. J. Pharmacol. 2016, 785, 44–49. [Google Scholar] [CrossRef]
  11. Croasdell, A.; Duffney, P.F.; Kim, N.; Lacy, S.H.; Sime, P.J.; Phipps, R.P. PPARγ and the Innate Immune System Mediate the Resolution of Inflammation. PPAR Res. 2015, 2015, 549691. [Google Scholar] [CrossRef] [Green Version]
  12. Mirza, A.Z.; Althagafi, I.I.; Shamshad, H. Role of PPAR receptor in different diseases and their ligands: Physiological importance and clinical implications. Eur. J. Med. Chem. 2019, 166, 502–513. [Google Scholar] [CrossRef]
  13. Derosa, G.; Sahebkar, A.; Maffioli, P. The role of various peroxisome proliferator-activated receptors and their ligands in clinical practice. J. Cell. Physiol. 2018, 233, 153–161. [Google Scholar] [CrossRef]
  14. Diskin, C.; Ryan, T.A.J.; O’Neill, L.A.J. Modification of Proteins by Metabolites in Immunity. Immunity 2021, 54, 19–31. [Google Scholar] [CrossRef]
  15. Haas, R.; Cucchi, D.; Smith, J.; Pucino, V.; Macdougall, C.E.; Mauro, C. Intermediates of Metabolism: From Bystanders to Signalling Molecules. Trends Biochem. Sci. 2016, 41, 460–471. [Google Scholar] [CrossRef]
  16. Soto-Heredero, G.; Gómez de Las Heras, M.M.; Gabandé-Rodríguez, E.; Oller, J.; Mittelbrunn, M. Glycolysis—A key player in the inflammatory response. FEBS J. 2020, 287, 3350–3369. [Google Scholar] [CrossRef] [Green Version]
  17. De Duve, C.; Baudhuin, P. Peroxisomes (microbodies and related particles). Physiol. Rev. 1966, 46, 323–357. [Google Scholar] [CrossRef]
  18. Okumoto, K.; Tamura, S.; Honsho, M.; Fujiki, Y. Peroxisome: Metabolic Functions and Biogenesis. Adv. Exp. Med. Biol. 2020, 1299, 3–17. [Google Scholar] [CrossRef]
  19. Reddy, J.K.; Krishnakantha, T.P.; Azarnoff, D.L.; Moody, D.E. 1-methyl-4piperidyl-bis (P-chlorophenoxy) acetate: A new hypolipidemic peroxisome proliferator. Res. Commun. Chem. Pathol. Pharmacol. 1975, 10, 589–592. [Google Scholar]
  20. Reddy, J.K.; Lalwai, N.D. Carcinogenesis by hepatic peroxisome proliferators: Evaluation of the risk of hypolipidemic drugs and industrial plasticizers to humans. Crit. Rev. Toxicol. 1983, 12, 1–58. [Google Scholar] [CrossRef]
  21. Reddy, J.K.; Goel, S.K.; Nemali, M.R.; Carrino, J.J.; Laffler, T.G.; Reddy, M.K.; Sperbeck, S.J.; Osumi, T.; Hashimoto, T.; Lalwani, N.D.; et al. Transcription regulation of peroxisomal fatty acyl-CoA oxidase and enoyl-CoA hydratase/3-hydroxyacyl-CoA dehydrogenase in rat liver by peroxisome proliferators. Proc. Natl. Acad. Sci. USA 1986, 83, 1747–1751. [Google Scholar] [CrossRef] [Green Version]
  22. Hardwick, J.P.; Song, B.J.; Huberman, E.; Gonzalez, F.J. Isolation, complementary DNA sequence, and regulation of rat hepatic lauric acid omega-hydroxylase (cytochrome P-450LA omega). Identification of a new cytochrome P-450 gene family. J. Biol. Chem. 1987, 262, 801–810. [Google Scholar] [CrossRef]
  23. Issemann, I.; Green, S. Activation of a member of the steroid hormone receptor superfamily by peroxisome proliferators. Nature 1990, 347, 645–650. [Google Scholar] [CrossRef]
  24. Zhu, Y.; Alvares, K.; Huang, Q.; Rao, M.S.; Reddy, J.K. Cloning of a new member of the peroxisome proliferator-activated receptor gene family from mouse liver. J. Biol. Chem. 1993, 268, 26817–26820. [Google Scholar] [CrossRef]
  25. Göttlicher, M.; Widmark, E.; Li, Q.; Gustafsson, J.A. Fatty acids activate a chimera of the clofibric acid-activated receptor and the glucocorticoid receptor. Proc. Natl. Acad. Sci. USA 1992, 89, 4653–4657. [Google Scholar] [CrossRef] [Green Version]
  26. Rolls, B.J.; Hammer, V.A. Fat, carbohydrate, and the regulation of energy intake. Am. J. Clin. Nutr. 1995, 62, 1086s–1095s. [Google Scholar] [CrossRef]
  27. Dreyer, C.; Krey, G.; Keller, H.; Givel, F.; Helftenbein, G.; Wahli, W. Control of the peroxisomal beta-oxidation pathway by a novel family of nuclear hormone receptors. Cell 1992, 68, 879–887. [Google Scholar] [CrossRef]
  28. Greene, M.E.; Blumberg, B.; McBride, O.W.; Yi, H.F.; Kronquist, K.; Kwan, K.; Hsieh, L.; Greene, G.; Nimer, S.D. Isolation of the human peroxisome proliferator activated receptor gamma cDNA: Expression in hematopoietic cells and chromosomal mapping. Gene Expr. 1995, 4, 281–299. [Google Scholar]
  29. Schmidt, A.; Endo, N.; Rutledge, S.J.; Vogel, R.; Shinar, D.; Rodan, G.A. Identification of a new member of the steroid hormone receptor superfamily that is activated by a peroxisome proliferator and fatty acids. Mol. Endocrinol. 1992, 6, 1634–1641. [Google Scholar] [CrossRef] [Green Version]
  30. Christofides, A.; Konstantinidou, E.; Jani, C.; Boussiotis, V.A. The role of peroxisome proliferator-activated receptors (PPAR) in immune responses. Metabolism 2021, 114, 154338. [Google Scholar] [CrossRef]
  31. Sher, T.; Yi, H.F.; McBride, O.W.; Gonzalez, F.J. cDNA cloning, chromosomal mapping, and functional characterization of the human peroxisome proliferator activated receptor. Biochemistry 1993, 32, 5598–5604. [Google Scholar] [CrossRef]
  32. Tyagi, S.; Gupta, P.; Saini, A.S.; Kaushal, C.; Sharma, S. The peroxisome proliferator-activated receptor: A family of nuclear receptors role in various diseases. J. Adv. Pharm. Technol. Res. 2011, 2, 236–240. [Google Scholar] [CrossRef]
  33. Qi, J.S.; Desai-Yajnik, V.; Greene, M.E.; Raaka, B.M.; Samuels, H.H. The ligand-binding domains of the thyroid hormone/retinoid receptor gene subfamily function in vivo to mediate heterodimerization, gene silencing, and transactivation. Mol. Cell. Biol. 1995, 15, 1817–1825. [Google Scholar] [CrossRef] [Green Version]
  34. Evans, R.M.; Barish, G.D.; Wang, Y.X. PPARs and the complex journey to obesity. Nat. Med. 2004, 10, 355–361. [Google Scholar] [CrossRef]
  35. Wang, S.; Dougherty, E.J.; Danner, R.L. PPARγ signaling and emerging opportunities for improved therapeutics. Pharmacol. Res. 2016, 111, 76–85. [Google Scholar] [CrossRef] [Green Version]
  36. Bain, D.L.; Heneghan, A.F.; Connaghan-Jones, K.D.; Miura, M.T. Nuclear receptor structure: Implications for function. Annu. Rev. Physiol. 2007, 69, 201–220. [Google Scholar] [CrossRef]
  37. Weikum, E.R.; Liu, X.; Ortlund, E.A. The nuclear receptor superfamily: A structural perspective. Protein Sci. 2018, 27, 1876–1892. [Google Scholar] [CrossRef] [Green Version]
  38. Desvergne, B.; Wahli, W. Peroxisome proliferator-activated receptors: Nuclear control of metabolism. Endocr. Rev. 1999, 20, 649–688. [Google Scholar] [CrossRef] [Green Version]
  39. Brunmeir, R.; Xu, F. Functional Regulation of PPARs through Post-Translational Modifications. Int. J. Mol. Sci. 2018, 19, 1738. [Google Scholar] [CrossRef] [Green Version]
  40. Palmer, C.N.; Hsu, M.H.; Griffin, H.J.; Johnson, E.F. Novel sequence determinants in peroxisome proliferator signaling. J. Biol. Chem. 1995, 270, 16114–16121. [Google Scholar] [CrossRef] [Green Version]
  41. Hu, X.; Lazar, M.A. The CoRNR motif controls the recruitment of corepressors by nuclear hormone receptors. Nature 1999, 402, 93–96. [Google Scholar] [CrossRef]
  42. Poulsen, L.; Siersbæk, M.; Mandrup, S. PPARs: Fatty acid sensors controlling metabolism. Semin. Cell Dev. Biol. 2012, 23, 631–639. [Google Scholar] [CrossRef]
  43. Chan, L.S.; Wells, R.A. Cross-Talk between PPARs and the Partners of RXR: A Molecular Perspective. PPAR Res. 2009, 2009, 925309. [Google Scholar] [CrossRef] [Green Version]
  44. Ricote, M.; Glass, C.K. PPARs and molecular mechanisms of transrepression. Biochim. Biophys. Acta 2007, 1771, 926–935. [Google Scholar] [CrossRef] [Green Version]
  45. Delerive, P.; Gervois, P.; Fruchart, J.C.; Staels, B. Induction of IkappaBalpha expression as a mechanism contributing to the anti-inflammatory activities of peroxisome proliferator-activated receptor-alpha activators. J. Biol. Chem. 2000, 275, 36703–36707. [Google Scholar] [CrossRef] [Green Version]
  46. Staels, B.; Koenig, W.; Habib, A.; Merval, R.; Lebret, M.; Torra, I.P.; Delerive, P.; Fadel, A.; Chinetti, G.; Fruchart, J.C.; et al. Activation of human aortic smooth-muscle cells is inhibited by PPARalpha but not by PPARgamma activators. Nature 1998, 393, 790–793. [Google Scholar] [CrossRef]
  47. Madej, A.; Okopien, B.; Kowalski, J.; Zielinski, M.; Wysocki, J.; Szygula, B.; Kalina, Z.; Herman, Z.S. Effects of fenofibrate on plasma cytokine concentrations in patients with atherosclerosis and hyperlipoproteinemia IIb. Int. J. Clin. Pharmacol. Ther. 1998, 36, 345–349. [Google Scholar]
  48. Delerive, P.; Martin-Nizard, F.; Chinetti, G.; Trottein, F.; Fruchart, J.C.; Najib, J.; Duriez, P.; Staels, B. Peroxisome proliferator-activated receptor activators inhibit thrombin-induced endothelin-1 production in human vascular endothelial cells by inhibiting the activator protein-1 signaling pathway. Circ. Res. 1999, 85, 394–402. [Google Scholar] [CrossRef] [Green Version]
  49. Kersten, S.; Seydoux, J.; Peters, J.M.; Gonzalez, F.J.; Desvergne, B.; Wahli, W. Peroxisome proliferator-activated receptor alpha mediates the adaptive response to fasting. J. Clin. Invest. 1999, 103, 1489–1498. [Google Scholar] [CrossRef] [Green Version]
  50. Montagner, A.; Polizzi, A.; Fouché, E.; Ducheix, S.; Lippi, Y.; Lasserre, F.; Barquissau, V.; Régnier, M.; Lukowicz, C.; Benhamed, F.; et al. Liver PPARα is crucial for whole-body fatty acid homeostasis and is protective against NAFLD. Gut 2016, 65, 1202–1214. [Google Scholar] [CrossRef] [Green Version]
  51. Fougerat, A.; Schoiswohl, G.; Polizzi, A.; Régnier, M.; Wagner, C.; Smati, S.; Fougeray, T.; Lippi, Y.; Lasserre, F.; Raho, I.; et al. ATGL-dependent white adipose tissue lipolysis controls hepatocyte PPARα activity. Cell Rep. 2022, 39, 110910. [Google Scholar] [CrossRef]
  52. Wang, A.; Huen, S.C.; Luan, H.H.; Yu, S.; Zhang, C.; Gallezot, J.D.; Booth, C.J.; Medzhitov, R. Opposing Effects of Fasting Metabolism on Tissue Tolerance in Bacterial and Viral Inflammation. Cell 2016, 166, 1512–1525.e1512. [Google Scholar] [CrossRef] [Green Version]
  53. Huen, S.C.; Wang, A.; Feola, K.; Desrouleaux, R.; Luan, H.H.; Hogg, R.; Zhang, C.; Zhang, Q.J.; Liu, Z.P.; Medzhitov, R. Hepatic FGF21 preserves thermoregulation and cardiovascular function during bacterial inflammation. J. Exp. Med. 2021, 218, e20202151. [Google Scholar] [CrossRef]
  54. Paumelle, R.; Haas, J.T.; Hennuyer, N.; Baugé, E.; Deleye, Y.; Mesotten, D.; Langouche, L.; Vanhoutte, J.; Cudejko, C.; Wouters, K.; et al. Hepatic PPARα is critical in the metabolic adaptation to sepsis. J. Hepatol. 2019, 70, 963–973. [Google Scholar] [CrossRef]
  55. Braissant, O.; Foufelle, F.; Scotto, C.; Dauça, M.; Wahli, W. Differential expression of peroxisome proliferator-activated receptors (PPARs): Tissue distribution of PPAR-alpha, -beta, and -gamma in the adult rat. Endocrinology 1996, 137, 354–366. [Google Scholar] [CrossRef] [Green Version]
  56. Müller, R. PPARβ/δ in human cancer. Biochimie 2017, 136, 90–99. [Google Scholar] [CrossRef]
  57. Yao, P.L.; Chen, L.; Dobrzański, T.P.; Zhu, B.; Kang, B.H.; Müller, R.; Gonzalez, F.J.; Peters, J.M. Peroxisome proliferator-activated receptor-β/δ inhibits human neuroblastoma cell tumorigenesis by inducing p53- and SOX2-mediated cell differentiation. Mol. Carcinog. 2017, 56, 1472–1483. [Google Scholar] [CrossRef]
  58. Foreman, J.E.; Sharma, A.K.; Amin, S.; Gonzalez, F.J.; Peters, J.M. Ligand activation of peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta) inhibits cell growth in a mouse mammary gland cancer cell line. Cancer Lett. 2010, 288, 219–225. [Google Scholar] [CrossRef] [Green Version]
  59. Genini, D.; Garcia-Escudero, R.; Carbone, G.M.; Catapano, C.V. Transcriptional and Non-Transcriptional Functions of PPARβ/δ in Non-Small Cell Lung Cancer. PLoS ONE 2012, 7, e46009. [Google Scholar] [CrossRef]
  60. Pedchenko, T.V.; Gonzalez, A.L.; Wang, D.; DuBois, R.N.; Massion, P.P. Peroxisome proliferator-activated receptor beta/delta expression and activation in lung cancer. Am. J. Respir. Cell Mol. Biol. 2008, 39, 689–696. [Google Scholar] [CrossRef] [Green Version]
  61. Wagner, K.D.; Benchetrit, M.; Bianchini, L.; Michiels, J.F.; Wagner, N. Peroxisome proliferator-activated receptor β/δ (PPARβ/δ) is highly expressed in liposarcoma and promotes migration and proliferation. J. Pathol. 2011, 224, 575–588. [Google Scholar] [CrossRef]
  62. Bishop-Bailey, D. PPARs and angiogenesis. Biochem. Soc. Trans. 2011, 39, 1601–1605. [Google Scholar] [CrossRef]
  63. Daynes, R.A.; Jones, D.C. Emerging roles of PPARs in inflammation and immunity. Nat. Rev. Immunol. 2002, 2, 748–759. [Google Scholar] [CrossRef]
  64. Kostadinova, R.; Wahli, W.; Michalik, L. PPARs in diseases: Control mechanisms of inflammation. Curr. Med. Chem. 2005, 12, 2995–3009. [Google Scholar] [CrossRef]
  65. Ricote, M.; Li, A.C.; Willson, T.M.; Kelly, C.J.; Glass, C.K. The peroxisome proliferator-activated receptor-gamma is a negative regulator of macrophage activation. Nature 1998, 391, 79–82. [Google Scholar] [CrossRef]
  66. Chinetti, G.; Griglio, S.; Antonucci, M.; Torra, I.P.; Delerive, P.; Majd, Z.; Fruchart, J.C.; Chapman, J.; Najib, J.; Staels, B. Activation of proliferator-activated receptors alpha and gamma induces apoptosis of human monocyte-derived macrophages. J. Biol. Chem. 1998, 273, 25573–25580. [Google Scholar] [CrossRef] [Green Version]
  67. Bao, Y.; Li, R.; Jiang, J.; Cai, B.; Gao, J.; Le, K.; Zhang, F.; Chen, S.; Liu, P. Activation of peroxisome proliferator-activated receptor gamma inhibits endothelin-1-induced cardiac hypertrophy via the calcineurin/NFAT signaling pathway. Mol. Cell. Biochem. 2008, 317, 189–196. [Google Scholar] [CrossRef]
  68. Kamata, S.; Oyama, T.; Saito, K.; Honda, A.; Yamamoto, Y.; Suda, K.; Ishikawa, R.; Itoh, T.; Watanabe, Y.; Shibata, T.; et al. PPARα Ligand-Binding Domain Structures with Endogenous Fatty Acids and Fibrates. iScience 2020, 23, 101727. [Google Scholar] [CrossRef]
  69. Nolte, R.T.; Wisely, G.B.; Westin, S.; Cobb, J.E.; Lambert, M.H.; Kurokawa, R.; Rosenfeld, M.G.; Willson, T.M.; Glass, C.K.; Milburn, M.V. Ligand binding and co-activator assembly of the peroxisome proliferator-activated receptor-gamma. Nature 1998, 395, 137–143. [Google Scholar] [CrossRef]
  70. Krey, G.; Braissant, O.; L’Horset, F.; Kalkhoven, E.; Perroud, M.; Parker, M.G.; Wahli, W. Fatty acids, eicosanoids, and hypolipidemic agents identified as ligands of peroxisome proliferator-activated receptors by coactivator-dependent receptor ligand assay. Mol. Endocrinol. 1997, 11, 779–791. [Google Scholar] [CrossRef]
  71. Aagaard, M.M.; Siersbæk, R.; Mandrup, S. Molecular basis for gene-specific transactivation by nuclear receptors. Biochim. Biophys. Acta 2011, 1812, 824–835. [Google Scholar] [CrossRef] [Green Version]
  72. Sumanasekera, W.K.; Tien, E.S.; Davis, J.W., 2nd; Turpey, R.; Perdew, G.H.; Vanden Heuvel, J.P. Heat shock protein-90 (Hsp90) acts as a repressor of peroxisome proliferator-activated receptor-alpha (PPARalpha) and PPARbeta activity. Biochemistry 2003, 42, 10726–10735. [Google Scholar] [CrossRef]
  73. Wheeler, M.C.; Gekakis, N. Hsp90 modulates PPARγ activity in a mouse model of nonalcoholic fatty liver disease. J. Lipid Res. 2014, 55, 1702–1710. [Google Scholar] [CrossRef] [Green Version]
  74. Layrolle, P.; Payoux, P.; Chavanas, S. PPAR Gamma and Viral Infections of the Brain. Int. J. Mol. Sci. 2021, 22, 8876. [Google Scholar] [CrossRef]
  75. Bassaganya-Riera, J.; Song, R.; Roberts, P.C.; Hontecillas, R. PPAR-gamma activation as an anti-inflammatory therapy for respiratory virus infections. Viral Immunol. 2010, 23, 343–352. [Google Scholar] [CrossRef]
  76. Huang, S.; Zhu, B.; Cheon, I.S.; Goplen, N.P.; Jiang, L.; Zhang, R.; Peebles, R.S.; Mack, M.; Kaplan, M.H.; Limper, A.H.; et al. PPAR-γ in Macrophages Limits Pulmonary Inflammation and Promotes Host Recovery following Respiratory Viral Infection. J. Virol. 2019, 93. [Google Scholar] [CrossRef] [Green Version]
  77. Liu, Q.; Zhou, Y.H.; Yang, Z.Q. The cytokine storm of severe influenza and development of immunomodulatory therapy. Cell. Mol. Immunol. 2016, 13, 3–10. [Google Scholar] [CrossRef] [Green Version]
  78. Huang, S.; Goplen, N.P.; Zhu, B.; Cheon, I.S.; Son, Y.; Wang, Z.; Li, C.; Dai, Q.; Jiang, L.; Xiang, M.; et al. Macrophage PPAR-γ suppresses long-term lung fibrotic sequelae following acute influenza infection. PLoS ONE 2019, 14, e0223430. [Google Scholar] [CrossRef] [Green Version]
  79. Bei, Y.; Tia, B.; Li, Y.; Guo, Y.; Deng, S.; Huang, R.; Zeng, H.; Li, R.; Wang, G.F.; Dai, J. Anti-influenza A Virus Effects and Mechanisms of Emodin and Its Analogs via Regulating PPARα/γ-AMPK-SIRT1 Pathway and Fatty Acid Metabolism. Biomed. Res. Int. 2021, 2021, 9066938. [Google Scholar] [CrossRef]
  80. Heffernan, K.S.; Ranadive, S.M.; Jae, S.Y. Exercise as medicine for COVID-19: On PPAR with emerging pharmacotherapy. Med. Hypotheses 2020, 143, 110197. [Google Scholar] [CrossRef]
  81. Dias, S.S.G.; Soares, V.C.; Ferreira, A.C.; Sacramento, C.Q.; Fintelman-Rodrigues, N.; Temerozo, J.R.; Teixeira, L.; Nunes da Silva, M.A.; Barreto, E.; Mattos, M.; et al. Lipid droplets fuel SARS-CoV-2 replication and production of inflammatory mediators. PLoS Pathog. 2020, 16, e1009127. [Google Scholar] [CrossRef]
  82. Alkhayyat, S.S.; Al-Kuraishy, H.M.; Al-Gareeb, A.I.; El-Bouseary, M.M.; AboKamer, A.M.; Batiha, G.E.; Simal-Gandara, J. Fenofibrate for COVID-19 and related complications as an approach to improve treatment outcomes: The missed key for Holy Grail. Inflamm. Res. 2022, 71, 1159–1167. [Google Scholar] [CrossRef]
  83. Jagat, J.M.; Kalyan, K.G.; Subir, R. Use of pioglitazone in people with type 2 diabetes mellitus with coronavirus disease 2019 (COVID-19): Boon or bane? Diabetol. Metab. Syndr. 2020, 14, 829–831. [Google Scholar] [CrossRef]
  84. Nagarkatti, P.; Miranda, K.; Nagarkatti, M. Use of Cannabinoids to Treat Acute Respiratory Distress Syndrome and Cytokine Storm Associated with Coronavirus Disease-2019. Front. Pharmacol. 2020, 11, 589438. [Google Scholar] [CrossRef]
  85. Corpetti, C.; Del Re, A.; Seguella, L.; Palenca, I.; Rurgo, S.; De Conno, B.; Pesce, M.; Sarnelli, G.; Esposito, G. Cannabidiol inhibits SARS-Cov-2 spike (S) protein-induced cytotoxicity and inflammation through a PPARγ-dependent TLR4/NLRP3/Caspase-1 signaling suppression in Caco-2 cell line. Phytother. Res. 2021, 35, 6893–6903. [Google Scholar] [CrossRef]
  86. Francisqueti-Ferron, F.V.; Garcia, J.L.; Ferron, A.J.T.; Nakandakare-Maia, E.T.; Gregolin, C.S.; Silva, J.; Dos Santos, K.C.; Lo, Â.T.C.; Siqueira, J.S.; de Mattei, L.; et al. Gamma-oryzanol as a potential modulator of oxidative stress and inflammation via PPAR-y in adipose tissue: A hypothetical therapeutic for cytokine storm in COVID-19? Mol. Cell. Endocrinol. 2021, 520, 111095. [Google Scholar] [CrossRef]
  87. Al-Qahtani, A.A.; Lyroni, K.; Aznaourova, M.; Tseliou, M.; Al-Anazi, M.R.; Al-Ahdal, M.N.; Alkahtani, S.; Sourvinos, G.; Tsatsanis, C. Middle east respiratory syndrome corona virus spike glycoprotein suppresses macrophage responses via DPP4-mediated induction of IRAK-M and PPARγ. Oncotarget 2017, 8, 9053–9066. [Google Scholar] [CrossRef] [Green Version]
  88. De, S.; Mamidi, P.; Ghosh, S.; Keshry, S.S.; Mahish, C.; Pani, S.S.; Laha, E.; Ray, A.; Datey, A.; Chatterjee, S.; et al. Telmisartan Restricts Chikungunya Virus Infection In Vitro and In Vivo through the AT1/PPAR-γ/MAPKs Pathways. Antimicrob. Agents Chemother. 2022, 66, e0148921. [Google Scholar] [CrossRef]
  89. Omeragic, A.; Hoque, M.T.; Choi, U.Y.; Bendayan, R. Peroxisome proliferator-activated receptor-gamma: Potential molecular therapeutic target for HIV-1-associated brain inflammation. J. Neuroinflamm. 2017, 14, 183. [Google Scholar] [CrossRef]
  90. Jiang, H.; Cheng, S.T.; Ren, J.H.; Ren, F.; Yu, H.B.; Wang, Q.; Huang, A.L.; Chen, J. SIRT6 Inhibitor, OSS_128167 Restricts Hepatitis B Virus Transcription and Replication Through Targeting Transcription Factor Peroxisome Proliferator-Activated Receptors α. Front. Pharmacol. 2019, 10, 1270. [Google Scholar] [CrossRef] [Green Version]
  91. Lin, Y.M.; Sun, H.Y.; Chiu, W.T.; Su, H.C.; Chien, Y.C.; Chong, L.W.; Chang, H.C.; Bai, C.H.; Young, K.C.; Tsao, C.W. Calcitriol Inhibits HCV Infection via Blockade of Activation of PPAR and Interference with Endoplasmic Reticulum-Associated Degradation. Viruses 2018, 10, 57. [Google Scholar] [CrossRef] [Green Version]
  92. Kumar, V.; Mansfield, J.; Fan, R.; MacLean, A.; Li, J.; Mohan, M. miR-130a and miR-212 Disrupt the Intestinal Epithelial Barrier through Modulation of PPARγ and Occludin Expression in Chronic Simian Immunodeficiency Virus-Infected Rhesus Macaques. J. Immunol. 2018, 200, 2677–2689. [Google Scholar] [CrossRef] [Green Version]
  93. Crakes, K.R.; Santos Rocha, C.; Grishina, I.; Hirao, L.A.; Napoli, E.; Gaulke, C.A.; Fenton, A.; Datta, S.; Arredondo, J.; Marco, M.L.; et al. PPARα-targeted mitochondrial bioenergetics mediate repair of intestinal barriers at the host-microbe intersection during SIV infection. Proc. Natl. Acad. Sci. USA 2019, 116, 24819–24829. [Google Scholar] [CrossRef] [Green Version]
  94. Lin, C.Y.; Huang, C.H.; Wang, W.H.; Tenhunen, J.; Hung, L.C.; Lin, C.C.; Chen, Y.C.; Chen, Y.H.; Liao, W.T. Mono-(2-ethylhexyl) phthalate Promotes Dengue Virus Infection by Decreasing IL-23-Mediated Antiviral Responses. Front. Immunol. 2021, 12, 599345. [Google Scholar] [CrossRef]
  95. Mulkey, S.B.; Arroyave-Wessel, M.; Peyton, C.; Bulas, D.I.; Fourzali, Y.; Jiang, J.; Russo, S.; McCarter, R.; Msall, M.E.; du Plessis, A.J.; et al. Neurodevelopmental Abnormalities in Children with In Utero Zika Virus Exposure Without Congenital Zika Syndrome. JAMA Pediatr. 2020, 174, 269–276. [Google Scholar] [CrossRef]
  96. Thulasi Raman, S.N.; Latreille, E.; Gao, J.; Zhang, W.; Wu, J.; Russell, M.S.; Walrond, L.; Cyr, T.; Lavoie, J.R.; Safronetz, D.; et al. Dysregulation of Ephrin receptor and PPAR signaling pathways in neural progenitor cells infected by Zika virus. Emerg. Microbes Infect. 2020, 9, 2046–2060. [Google Scholar] [CrossRef]
  97. Li, Y.; Zhu, Y.; Feng, S.; Ishida, Y.; Chiu, T.P.; Saito, T.; Wang, S.; Ann, D.K.; Ou, J.J. Macrophages activated by hepatitis B virus have distinct metabolic profiles and suppress the virus via IL-1β to downregulate PPARα and FOXO3. Cell Rep. 2022, 38, 110284. [Google Scholar] [CrossRef]
  98. Du, L.; Ma, Y.; Liu, M.; Yan, L.; Tang, H. Peroxisome Proliferators Activated Receptor (PPAR) agonists activate hepatitis B virus replication in vivo. Virol. J. 2017, 14, 96. [Google Scholar] [CrossRef] [Green Version]
  99. Suzuki, K.; Suda, G.; Yamamoto, Y.; Furuya, K.; Baba, M.; Nakamura, A.; Miyoshi, H.; Kimura, M.; Maehara, O.; Yamada, R.; et al. Tenofovir-disoproxil-fumarate modulates lipid metabolism via hepatic CD36/PPAR-alpha activation in hepatitis B virus infection. J. Gastroenterol. 2021, 56, 168–180. [Google Scholar] [CrossRef]
  100. Goldwasser, J.; Cohen, P.Y.; Lin, W.; Kitsberg, D.; Balaguer, P.; Polyak, S.J.; Chung, R.T.; Yarmush, M.L.; Nahmias, Y. Naringenin inhibits the assembly and long-term production of infectious hepatitis C virus particles through a PPAR-mediated mechanism. J. Hepatol. 2011, 55, 963–971. [Google Scholar] [CrossRef] [Green Version]
  101. Tam, V.C.; Suen, R.; Treuting, P.M.; Armando, A.; Lucarelli, R.; Gorrochotegui-Escalante, N.; Diercks, A.H.; Quehenberger, O.; Dennis, E.A.; Aderem, A.; et al. PPARα exacerbates necroptosis, leading to increased mortality in postinfluenza bacterial superinfection. Proc. Natl. Acad. Sci. USA 2020, 117, 15789–15798. [Google Scholar] [CrossRef]
  102. Gopal, R.; Mendy, A.; Marinelli, M.A.; Richwalls, L.J.; Seger, P.J.; Patel, S.; McHugh, K.J.; Rich, H.E.; Grousd, J.A.; Forno, E.; et al. Peroxisome Proliferator-Activated Receptor Gamma (PPARγ) Suppresses Inflammation and Bacterial Clearance during Influenza-Bacterial Super-Infection. Viruses 2019, 11, 505. [Google Scholar] [CrossRef] [Green Version]
  103. Lucarelli, R.; Gorrochotegui-Escalante, N.; Taddeo, J.; Buttaro, B.; Beld, J.; Tam, V. Eicosanoid-Activated PPARα Inhibits NFκB-Dependent Bacterial Clearance During Post-Influenza Superinfection. Front. Cell. Infect. Microbiol. 2022, 12, 881462. [Google Scholar] [CrossRef]
  104. Cevallos, S.A.; Lee, J.Y.; Velazquez, E.M.; Foegeding, N.J.; Shelton, C.D.; Tiffany, C.R.; Parry, B.H.; Stull-Lane, A.R.; Olsan, E.E.; Savage, H.P.; et al. 5-Aminosalicylic Acid Ameliorates Colitis and Checks Dysbiotic Escherichia coli Expansion by Activating PPAR-γ Signaling in the Intestinal Epithelium. mBio 2021, 12. [Google Scholar] [CrossRef]
  105. Ramirez-Moral, I.; Ferreira, B.L.; de Vos, A.F.; van der Poll, T. Post-treatment with the PPAR-γ agonist pioglitazone inhibits inflammation and bacterial growth during Klebsiella pneumonia. Respir. Res. 2021, 22, 230. [Google Scholar] [CrossRef]
  106. Pu, Q.; Guo, K.; Lin, P.; Wang, Z.; Qin, S.; Gao, P.; Combs, C.; Khan, N.; Xia, Z.; Wu, M. Bitter receptor TAS2R138 facilitates lipid droplet degradation in neutrophils during Pseudomonas aeruginosa infection. Signal Transduct. Target. Ther. 2021, 6, 210. [Google Scholar] [CrossRef]
  107. Ferreira, B.L.; Ramirez-Moral, I.; Otto, N.A.; Salomão, R.; de Vos, A.F.; van der Poll, T. The PPAR-γ agonist pioglitazone exerts proinflammatory effects in bronchial epithelial cells during acute Pseudomonas aeruginosa pneumonia. Clin. Exp. Immunol. 2022, 207, 370–377. [Google Scholar] [CrossRef]
  108. Wu, X.; Cheng, B.; Guo, X.; Wu, Q.; Sun, S.; He, P. PPARα/γ signaling pathways are involved in Chlamydia pneumoniae-induced foam cell formation via upregulation of SR-A1 and ACAT1 and downregulation of ABCA1/G1. Microb. Pathog. 2021, 161, 105284. [Google Scholar] [CrossRef]
  109. Katkar, G.D.; Sayed, I.M.; Anandachar, M.S.; Castillo, V.; Vidales, E.; Toobian, D.; Usmani, F.; Sawires, J.R.; Leriche, G.; Yang, J.; et al. Artificial intelligence-rationalized balanced PPARα/γ dual agonism resets dysregulated macrophage processes in inflammatory bowel disease. Commun. Biol. 2022, 5, 231. [Google Scholar] [CrossRef]
  110. Wani, K.A.; Goswamy, D.; Taubert, S.; Ratnappan, R.; Ghazi, A.; Irazoqui, J.E. NHR-49/PPAR-α and HLH-30/TFEB cooperate for C. elegans host defense via a flavin-containing monooxygenase. eLife 2021, 10, e62775. [Google Scholar] [CrossRef]
  111. Liang, F.; Guan, H.; Li, W.; Zhang, X.; Liu, T.; Liu, Y.; Mei, J.; Jiang, C.; Zhang, F.; Luo, B.; et al. Erythropoietin Promotes Infection Resolution and Lowers Antibiotic Requirements in E. coli- and S. aureus-Initiated Infections. Front. Immunol. 2021, 12, 658715. [Google Scholar] [CrossRef]
  112. Allen, P.E.; Noland, R.C.; Martinez, J.J. Rickettsia conorii survival in THP-1 macrophages involves host lipid droplet alterations and active rickettsial protein production. Cell. Microbiol. 2021, 23, e13390. [Google Scholar] [CrossRef]
  113. Tanigawa, K.; Luo, Y.; Kawashima, A.; Kiriya, M.; Nakamura, Y.; Karasawa, K.; Suzuki, K. Essential Roles of PPARs in Lipid Metabolism during Mycobacterial Infection. Int. J. Mol. Sci. 2021, 22, 7597. [Google Scholar] [CrossRef]
  114. Luo, Y.; Tanigawa, K.; Kawashima, A.; Ishido, Y.; Ishii, N.; Suzuki, K. The function of peroxisome proliferator-activated receptors PPAR-γ and PPAR-δ in Mycobacterium leprae-induced foam cell formation in host macrophages. PLoS Negl. Trop. Dis. 2020, 14, e0008850. [Google Scholar] [CrossRef]
  115. Rajaram, M.V.; Brooks, M.N.; Morris, J.D.; Torrelles, J.B.; Azad, A.K.; Schlesinger, L.S. Mycobacterium tuberculosis activates human macrophage peroxisome proliferator-activated receptor gamma linking mannose receptor recognition to regulation of immune responses. J. Immunol. 2010, 185, 929–942. [Google Scholar] [CrossRef] [Green Version]
  116. Mahajan, S.; Dkhar, H.K.; Chandra, V.; Dave, S.; Nanduri, R.; Janmeja, A.K.; Agrewala, J.N.; Gupta, P. Mycobacterium tuberculosis modulates macrophage lipid-sensing nuclear receptors PPARγ and TR4 for survival. J. Immunol. 2012, 188, 5593–5603. [Google Scholar] [CrossRef] [Green Version]
  117. Almeida, P.E.; Silva, A.R.; Maya-Monteiro, C.M.; Töröcsik, D.; D’Avila, H.; Dezsö, B.; Magalhães, K.G.; Castro-Faria-Neto, H.C.; Nagy, L.; Bozza, P.T. Mycobacterium bovis bacillus Calmette-Guérin infection induces TLR2-dependent peroxisome proliferator-activated receptor gamma expression and activation: Functions in inflammation, lipid metabolism, and pathogenesis. J. Immunol. 2009, 183, 1337–1345. [Google Scholar] [CrossRef] [Green Version]
  118. D’Avila, H.; Melo, R.C.; Parreira, G.G.; Werneck-Barroso, E.; Castro-Faria-Neto, H.C.; Bozza, P.T. Mycobacterium bovis bacillus Calmette-Guérin induces TLR2-mediated formation of lipid bodies: Intracellular domains for eicosanoid synthesis in vivo. J. Immunol. 2006, 176, 3087–3097. [Google Scholar] [CrossRef] [Green Version]
  119. Kim, Y.S.; Lee, H.M.; Kim, J.K.; Yang, C.S.; Kim, T.S.; Jung, M.; Jin, H.S.; Kim, S.; Jang, J.; Oh, G.T.; et al. PPAR-α Activation Mediates Innate Host Defense through Induction of TFEB and Lipid Catabolism. J. Immunol. 2017, 198, 3283–3295. [Google Scholar] [CrossRef] [Green Version]
  120. Kim, Y.S.; Kim, J.K.; Hanh, B.T.B.; Kim, S.Y.; Kim, H.J.; Kim, Y.J.; Jeon, S.M.; Park, C.R.; Oh, G.T.; Park, J.W.; et al. The Peroxisome Proliferator-Activated Receptor α-Agonist Gemfibrozil Promotes Defense Against Mycobacterium abscessus Infections. Cells 2020, 9, 648. [Google Scholar] [CrossRef] [Green Version]
  121. Yamanishi, Y.; Miyake, K.; Iki, M.; Tsutsui, H.; Karasuyama, H. Recent advances in understanding basophil-mediated Th2 immune responses. Immunol. Rev. 2017, 278, 237–245. [Google Scholar] [CrossRef]
  122. Chen, T.; Tibbitt, C.A.; Feng, X.; Stark, J.M.; Rohrbeck, L.; Rausch, L.; Sedimbi, S.K.; Karlsson, M.C.I.; Lambrecht, B.N.; Karlsson Hedestam, G.B.; et al. PPAR-γ promotes type 2 immune responses in allergy and nematode infection. Sci. Immunol. 2017, 2. [Google Scholar] [CrossRef]
  123. He, X.; Gong, P.; Wei, Z.; Liu, W.; Wang, W.; Li, J.; Yang, Z.; Zhang, X. Peroxisome proliferator-activated receptor-γ-mediated polarization of macrophages in Neospora caninum infection. Exp. Parasitol. 2017, 178, 37–44. [Google Scholar] [CrossRef]
  124. Chen, K.M.; Peng, C.Y.; Shyu, L.Y.; Lan, K.P.; Lai, S.C. Peroxisome-Proliferator Activator Receptor γ in Mouse Model with Meningoencephalitis Caused by Angiostrongylus cantonensis. J. Parasitol. 2021, 107, 205–213. [Google Scholar] [CrossRef]
  125. Mita-Mendoza, N.K.; Magallon-Tejada, A.; Parmar, P.; Furtado, R.; Aldrich, M.; Saidi, A.; Taylor, T.; Smith, J.; Seydel, K.; Daily, J.P. Dimethyl fumarate reduces TNF and Plasmodium falciparum induced brain endothelium activation in vitro. Malar. J. 2020, 19, 376. [Google Scholar] [CrossRef]
  126. Borges, T.K.; Alves, É.A.; Vasconcelos, H.A.; Carneiro, F.P.; Nicola, A.M.; Magalhães, K.G.; Muniz-Junqueira, M.I. Differences in the modulation of reactive species, lipid bodies, cyclooxygenase-2, 5-lipoxygenase and PPAR-γ in cerebral malaria-susceptible and resistant mice. Immunobiology 2017, 222, 604–619. [Google Scholar] [CrossRef]
  127. Matta, S.K.; Rinkenberger, N.; Dunay, I.R.; Sibley, L.D. Toxoplasma gondii infection and its implications within the central nervous system. Nat. Rev. Microbiol. 2021, 19, 467–480. [Google Scholar] [CrossRef]
  128. Shyu, L.Y.; Chen, K.M.; Lu, C.Y.; Lai, S.C. Regulation of Proinflammatory Enzymes by Peroxisome Proliferator-Activated Receptor Gamma in Astroglia Infected with Toxoplasma gondii. J. Parasitol. 2020, 106, 564–571. [Google Scholar] [CrossRef]
  129. He, J.J.; Ma, J.; Elsheikha, H.M.; Song, H.Q.; Zhou, D.H.; Zhu, X.Q. Proteomic Profiling of Mouse Liver following Acute Toxoplasma gondii Infection. PLoS ONE 2016, 11, e0152022. [Google Scholar] [CrossRef] [Green Version]
  130. Moulik, S.; Karmakar, J.; Joshi, S.; Dube, A.; Mandal, C.; Chatterjee, M. Status of IL-4 and IL-10 driven markers in experimental models of Visceral Leishmaniasis. Parasite Immunol. 2021, 43, e12783. [Google Scholar] [CrossRef]
  131. McManus, D.P.; Dunne, D.W.; Sacko, M.; Utzinger, J.; Vennervald, B.J.; Zhou, X.N. Schistosomiasis. Nat. Rev. Dis. Prim. 2018, 4, 13. [Google Scholar] [CrossRef]
  132. Gryseels, B.; Polman, K.; Clerinx, J.; Kestens, L. Human schistosomiasis. Lancet 2006, 368, 1106–1118. [Google Scholar] [CrossRef]
  133. Shiels, J.; Cwiklinski, K.; Alvarado, R.; Thivierge, K.; Cotton, S.; Gonzales Santana, B.; To, J.; Donnelly, S.; Taggart, C.C.; Weldon, S.; et al. Schistosoma mansoni immunomodulatory molecule Sm16/SPO-1/SmSLP is a member of the trematode-specific helminth defence molecules (HDMs). PLoS Negl. Trop. Dis. 2020, 14, e0008470. [Google Scholar] [CrossRef]
  134. Rassi, A., Jr.; Rassi, A.; Marin-Neto, J.A. Chagas disease. Lancet 2010, 375, 1388–1402. [Google Scholar] [CrossRef]
  135. Penas, F.; Mirkin, G.A.; Vera, M.; Cevey, Á.; González, C.D.; Gómez, M.I.; Sales, M.E.; Goren, N.B. Treatment in vitro with PPARα and PPARγ ligands drives M1-to-M2 polarization of macrophages from T. cruzi-infected mice. Biochim. Biophys. Acta 2015, 1852, 893–904. [Google Scholar] [CrossRef] [Green Version]
  136. Penas, F.N.; Carta, D.; Cevey, Á.C.; Rada, M.J.; Pieralisi, A.V.; Ferlin, M.G.; Sales, M.E.; Mirkin, G.A.; Goren, N.B. Pyridinecarboxylic Acid Derivative Stimulates Pro-Angiogenic Mediators by PI3K/AKT/mTOR and Inhibits Reactive Nitrogen and Oxygen Species and NF-κB Activation Through a PPARγ-Dependent Pathway in T. cruzi-Infected Macrophages. Front. Immunol. 2019, 10, 2955. [Google Scholar] [CrossRef]
  137. Penas, F.N.; Cevey, Á.C.; Siffo, S.; Mirkin, G.A.; Goren, N.B. Hepatic injury associated with Trypanosoma cruzi infection is attenuated by treatment with 15-deoxy-Δ(12,14) prostaglandin J(2). Exp. Parasitol. 2016, 170, 100–108. [Google Scholar] [CrossRef]
  138. González, F.B.; Villar, S.R.; Toneatto, J.; Pacini, M.F.; Márquez, J.; D’Attilio, L.; Bottasso, O.A.; Piwien-Pilipuk, G.; Pérez, A.R. Immune response triggered by Trypanosoma cruzi infection strikes adipose tissue homeostasis altering lipid storage, enzyme profile and adipokine expression. Med. Microbiol. Immunol. 2019, 208, 651–666. [Google Scholar] [CrossRef]
  139. Dreesen, L.; De Bosscher, K.; Grit, G.; Staels, B.; Lubberts, E.; Bauge, E.; Geldhof, P. Giardia muris infection in mice is associated with a protective interleukin 17A response and induction of peroxisome proliferator-activated receptor alpha. Infect. Immun. 2014, 82, 3333–3340. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Roles of peroxisome proliferator-activated receptors (PPARs) and their regulatory mechanisms. PPAR ligands bind to the PPAR ligand-binding domain and activate receptors. PPARs interact with heat shock protein (HSP) in the cytosol. PPARs inhibit inflammation-related gene transcription by interfering with transcription factors such as NF-κB, AP-1, STAT, and NFAT. PPARs form heterodimers with Retinoid X receptor (RXR), a receptor of 9-cis-retinoic acid (9-cis-RA), and bind to direct repeat 1 (DR-1), a peroxisome-proliferator-responsive element. The PPAR-RXR heterodimer complex and co-repressors represses target gene transcription. However, the complex with co-activators promotes target gene transcription. Through these mechanisms, PPARs play significant roles in energy metabolism, inflammatory modulation, and the cell cycle. AP-1, Activator protein 1; NF-κB, Nuclear factor kappa-light-chain-enhancer of activated B cells; NFAT, Nuclear factor of activated T cells; STAT, Signal transducer and activator of transcription.
Figure 1. Roles of peroxisome proliferator-activated receptors (PPARs) and their regulatory mechanisms. PPAR ligands bind to the PPAR ligand-binding domain and activate receptors. PPARs interact with heat shock protein (HSP) in the cytosol. PPARs inhibit inflammation-related gene transcription by interfering with transcription factors such as NF-κB, AP-1, STAT, and NFAT. PPARs form heterodimers with Retinoid X receptor (RXR), a receptor of 9-cis-retinoic acid (9-cis-RA), and bind to direct repeat 1 (DR-1), a peroxisome-proliferator-responsive element. The PPAR-RXR heterodimer complex and co-repressors represses target gene transcription. However, the complex with co-activators promotes target gene transcription. Through these mechanisms, PPARs play significant roles in energy metabolism, inflammatory modulation, and the cell cycle. AP-1, Activator protein 1; NF-κB, Nuclear factor kappa-light-chain-enhancer of activated B cells; NFAT, Nuclear factor of activated T cells; STAT, Signal transducer and activator of transcription.
Cells 12 00650 g001
Table 1. Studies on PPARs and their ligands during viral infections.
Table 1. Studies on PPARs and their ligands during viral infections.
PathogenStudy ModelInterventionPPAR StatusMechanismRef.
IAV, RSVAMs, micePpargΔLyz2 miceRegulation of PPARγ through STAT1 activation following IFN signaling[76]
IAVAMs, human lung macrophages, micePpargΔLyz2 mice, BleomycinIncreased influenza-induced pulmonary collagen deposition in PPARγ-deficient mice[78]
IAVA549 cells, miceEmodin and its analogsActivation of PPARα/γ and AMPK, decreased fatty acid biosynthesis and increased ATP level[79]
MERS-CoVTHP-1 cells, primary human monocytessiRNAsMERS-CoV S glycoprotein interaction with DPP4 leading to IRAK-M and PPARγ expression[87]
CHIKVVero cells, RAW264.7 cellsTelmisatran, PPAR-γ antagonist GW9662Activation of PPAR-γ and inhibition of AT1 by telmisartan[88]
HIVPrimary rat astrocytes, microglia, ratsgp120ADA, Rosiglitazone, PioglitazoneInduction of inflammatory response and decrease in GLT-1 expression in the brain by gp120[89]
HBVHepG2.2.15, Huh7, HepG2-NTCP ellsOS_128167, overexpression and downregulation studies, HBV transgenic mice-Activation of HBV core promoter by SIRT6 through upregulation of PPARα[90]
HCVHuh7.5 cellsCalciterol, Linoleic acid, Ly171883, Wy14643-Activation of VDR but inhibition of PPARα/β/γ by calcitriol [91]
Abbreviations: AMPK, AMP-activated protein kinase; AMs, Alveolar macrophages; AT1, Angiotensin 1; CHIKV, Chikungunya virus; DPP4, Dipeptidyl-peptidase 4; GLT-1, Glutamate transporter 1; HBV, Hepatitis B virus; HCV, Hepatitis C virus; HIV, human immunodeficiency virus; IAV, Influenza A virus; IFN, interferon; IRAK-M, Interleukin-1 receptor-associated kinase 3; MERS-CoV, Middle east respiratory syndrome corona virus; RSV, Respiratory syncytial virus; SIRT6, Sirtuin 6; STAT1, Signal transducer and activator of transcription 1; VDR; Vitamin D receptor; ↑, increase/activation; ↓, decrease/inhibition; -, not reported.
Table 2. Roles of PPAR agonists/antagonists in bacterial infections.
Table 2. Roles of PPAR agonists/antagonists in bacterial infections.
PathogenDrug/ReagentFunctionStudy ModelMechanism of ActionRef.
Escherichia coli5-aminosalicylic acidPPARγ agonistDSS-induced murine colitis model, Pparg-deficient mice, CaCo-2 cellsAmelioration of a respiration-dependent luminal expansion of E. coli[104]
Klebsiella pneumoniaePioglitazonePPARγ agonistIn vivo mouse modelReduction of cytokines and myeloperoxidase levels in the lungs[105]
Pseudomonas aeruginosaPioglitazonePPARγ agonistIn vivo mouse modelIncreased pro-inflammatory cytokines with enhanced expression of genes involved in glycolysis[107]
Chlamydia pneumoniaeRosiglitazonePPARγ agonistTHP-1 macrophages, HEp-2 cellsRegulation of Cpn induced macrophage-derived foam cell formation by upregulating SR-A1 an ACAT1, and downregulating ABCA1/G1 expression via PPARα/γ signaling[108]
FenofibratePPARα agonist
GW9662PPARγ antagonist
MK886PPARα antagonist
Citrobacter rodentiumPAR5359PPARα/γ-dual-agonistCitrobacter rodentium- and DSS-induced murine colitis model, IBD patient-derived PBMCsEnhanced bacterial clearance, controlled production of ROS and cytokines, anti-inflammatory/healing[109]
Rickettsia conoriiGW9662PPARγ antagonistTHP-1 macrophagesIncreased intracellular survival of bacteria[112]
Abbreviations: ABCA1/G1, ATP binding cassette transporters A1/G1; ACAT1, acyl-coenzyme A: cholesterol acyltransferase 1; Cpn, Chlamydia pneumonia; DSS, Dextran sulfate sodium; IBD, Inflammatory bowel disease; PBMCs, Peripheral blood mononuclear cells; ROS, Reactive oxygen species; SR-A1, scavenger receptor A1.
Table 3. Roles of PPAR agonists/antagonists in parasitic infections.
Table 3. Roles of PPAR agonists/antagonists in parasitic infections.
PathogenDrug/ReagentFunctionStudy ModelMechanism of ActionRef.
Angiostrongylus cantonensisGW9662PPARγ antagonistMouse model of angiostrongyliasisNF-κB activation and increase in inflammation and BBB permeability[124]
Plasmodium falciparumDimethyl fumarate-Cerebral cortex derived HBMVECsUpregulation of PPAR pathway, NRF2-mediated oxidative stress responses, ErbB4 signaling to downregulate the neuroinflammation[125]
Toxoplasma gondiiRosiglitazonePPARγ agonistSVG p12 cells, Hs68 cellsDecreased expression of MMP-2, MMP-9, COX-2, PGE2, iNOS and NO[128]
GW9662PPARγ antagonistIncreased expression of MMP-2, MMP-9, COX-2, PGE2, iNOS and NO
Trypanosoma cruziHP24pyridinecarboxylic acid derivativeIn vivo mice infection, mouse peritoneal macrophagesInduction of PI3K/Akt/mTOR signaling (pro-angiogenic), inhibition of NF-κB signaling (anti-inflammatory)[136]
15-deoxy-D12,14 prostaglandin J2PPARγ agonistIn vivo mice infectionReduction of liver inflammatory infiltrates, pro-inflammatory enzymes and cytokine expression through inhibition of NF-kB signaling, No change in parasitic load[137]
Abbreviations: BBB, Blood-brain barrier; COX-2, Cyclooxygenase-2; ErbB4, Erb-b2 receptor tyrosine kinase 4; HP24, 1-methyl-3-hydroxy-4-pyridinecarboxylic acid derivative 24; iNOS, Inducible nitric oxide synthase; MMP, Matrix metalloproteinase; mTOR, Mammalian target of rapamycin; NF-kB, Nuclear factor-κB; NO, Nitric oxide; NRF2, Nuclear factor E2-related factor 2; PGE2, Prostaglandin E2; PI3K, Phosphoinositide 3-kinase.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kim, I.S.; Silwal, P.; Jo, E.-K. Peroxisome Proliferator-Activated Receptor-Targeted Therapies: Challenges upon Infectious Diseases. Cells 2023, 12, 650. https://doi.org/10.3390/cells12040650

AMA Style

Kim IS, Silwal P, Jo E-K. Peroxisome Proliferator-Activated Receptor-Targeted Therapies: Challenges upon Infectious Diseases. Cells. 2023; 12(4):650. https://doi.org/10.3390/cells12040650

Chicago/Turabian Style

Kim, In Soo, Prashanta Silwal, and Eun-Kyeong Jo. 2023. "Peroxisome Proliferator-Activated Receptor-Targeted Therapies: Challenges upon Infectious Diseases" Cells 12, no. 4: 650. https://doi.org/10.3390/cells12040650

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop