Next Article in Journal
Genotype–Phenotype Relations for the Dystonia-Parkinsonism Genes GLB1, SLC6A3, SLC30A10, SLC39A14, and PLA2G6: MDSGene Systematic Review
Previous Article in Journal
Comparative Genomics, Transcriptome, and Prokaryotic Expression Analysis of alkB1_1 in Acinetobacter vivianii KJ-1: Revealing the Mechanism of Petroleum Hydrocarbon Degradation
Previous Article in Special Issue
Differences in Acute Expression of Matrix Metalloproteinases-9, 3, and 2 Related to the Duration of Brain Ischemia and Tissue Plasminogen Activator Treatment in Experimental Stroke
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

PPARα Genetic Deletion Reveals Global Transcriptional Changes in the Brain and Exacerbates Cerebral Infarction in a Mouse Model of Stroke

1
Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
2
Health Sciences Center, Tulane University, New Orleans, LA 70112, USA
3
School of Medicine, Emory University, Atlanta, GA 30322, USA
4
Bioinformatics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
5
Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(9), 4082; https://doi.org/10.3390/ijms26094082
Submission received: 6 March 2025 / Revised: 11 April 2025 / Accepted: 16 April 2025 / Published: 25 April 2025
(This article belongs to the Special Issue Inflammatory Biomarkers in Ischemic Stroke)

Abstract

Ischemic stroke is a leading cause of death and disability worldwide. Currently, there is an unmet clinical need for pharmacological treatments that can improve ischemic stroke outcomes. In this study, we investigated the role of brain peroxisome proliferator-activated receptor alpha (PPARα) in ischemic stroke pathophysiology. We used a well-established model of cerebral ischemia in PPARα transgenic mice and conducted the RNA sequencing (RNA-seq) of mouse stroke brains harvested 48 h post-middle cerebral artery occlusion (MCAO). PPARα knockout (KO) increased brain infarct size following stroke, indicating a protective role of PPARα in brain ischemia. Our RNA-seq analysis showed that PPARα KO altered the expression of genes in mouse brains with known roles in ischemic stroke pathophysiology. We also identified many other differentially expressed genes (DEGs) upon the loss of PPARα that correlated with increased infarct size in our stroke model. Gene set enrichment analysis (GSEA) and Gene Ontology (GO) analysis revealed the upregulation of gene signatures for the positive regulation of leukocyte proliferation, apoptotic processes, acute-phase response, and cellular component disassembly in mouse stroke brains with PPARα KO. In addition, pathway analysis of our RNA-seq data revealed that TNFα signaling, IL6/STAT3 signaling, and epithelial–mesenchymal transition (EMT) gene signatures were increased in PPARα KO stroke brains. Our study highlights PPARα as an attractive drug target for ischemic stroke due to its transcriptional regulation of inflammation-, apoptosis-, and EMT-related genes in brain tissue following ischemia.

1. Introduction

Ischemic stroke is characterized by the sudden obstruction of blood flow to the brain and remains a leading cause of mortality and long-term disability worldwide. In 2021, stroke accounted for approximately 1 of every 21 deaths in the United States [1]. To date, the only FDA-approved acute treatments for ischemic stroke are thrombolysis with tissue plasminogen activator (tPA) and mechanical thrombectomy. The therapeutic window for these interventions is narrow, and tPA is known to increase the risk of hemorrhagic transformation in stroke patients if administered too late. Thus, many ischemic stroke patients experience irreversible brain damage and long-term disability. There is an unmet clinical need for new pharmacological therapies that can reduce infarct size, limit neurological deficits, and promote recovery in stroke patients. The pathophysiology of ischemic stroke involves cellular energy failure, neural excitotoxicity, oxidative stress, inflammation, and ultimately cell death [2,3,4,5]. Thus, druggable targets that can modulate several of these pathways are attractive candidates for stroke therapy.
Peroxisome proliferator-activated receptor alpha (PPARα) is a member of the nuclear hormone receptor superfamily and plays a central role in lipid metabolism, energy homeostasis, and inflammation [6]. When bound to its ligands, PPARα traffics to the nucleus and functions as a transcription factor to regulate the expression of genes involved in fatty acid oxidation and inflammation [7]. Previous research on PPARα characterized its physiological role in the vasculature, where it regulates lipid metabolism, inflammation, and endothelial function [8,9]. In fact, PPARα agonists such as fenofibrate, gemfibrozil, and bezafibrate are used in the clinic to manage cardiovascular disease by dampening inflammation, reducing triglyceride levels, and increasing beneficial HDL cholesterol [10].
Accumulating evidence suggests that PPARα also plays a neuroprotective role in the brain. Several studies report that pretreatment with PPARα agonists like fenofibrate increases the activity of antioxidant enzymes, decreases oxidative stress, and improves outcomes in rodent models of ischemic stroke [11,12,13]. Some preclinical studies suggest that treatment with PPARα agonists benefits stroke outcome by eliciting anti-inflammatory effects in the CNS and the periphery [11,12,14,15,16], while other studies report that PPARα activation reduces endothelial dysfunction and blood–brain barrier (BBB) breakdown after stroke [17,18]. Altogether, PPARα activation may protect against ischemic stroke through the modulation of several pathophysiological pathways such as oxidative stress, inflammation, and blood vessel dysfunction. However, the detailed transcriptional targets that PPARα regulates in the brain vasculature during ischemia remain largely unknown. In this study, we modeled ischemic stroke in wild-type (WT) and PPARα genetic knockout (KO) mice and utilized the RNA-sequencing of ipsilesional hemisphere brain tissue harvested 48 hours (h) post-stroke to characterize genes and transcriptional pathways associated with PPARα. Our study is the first to use global CNS transcriptome profiling to investigate the role of PPARα in the subacute phase of ischemic stroke.

2. Results

2.1. PPARα KO Increases Infarct Volume in Mouse Stroke Brains

Treatment with PPARα agonists has beneficial effects on the vasculature, but PPARα’s role in the context of ischemic stroke is not well understood. We assessed the effects of PPARα genetic deletion on infarct volume in mouse stroke brains. We used the TTC staining of brain sections to quantify the ischemic lesion volume (percentage of the brain hemisphere) 48 h after middle cerebral artery occlusion followed by reperfusion (MCAO/R). Compared with brains from MCAO/R WT mice, brains from MCAO/R PPARα KO mice had significantly larger infarct sizes, indicating greater stroke severity (Figure A1). The mean infarct volume of the ipsilesional hemisphere in MCAO/R WT stroke brains was 49.4 ± 2.89% (**** p < 0.0001 vs. sham). In MCAO/R PPARα KO brains, the mean infarct volume was 59.74 ± 3.57% (**** p < 0.0001 vs. sham), which was larger than in the MCAO/R WT group (# p < 0.05 vs. MCAO/R WT) (Figure 1). These results suggest that the loss of PPARα worsens ischemic brain injury, indicating a protective role of PPARα in brain ischemia.

2.2. Genetic Deletion of PPARα Elicits Global Transcriptional Changes in Mouse Brains After Stroke

We performed RNA-seq on ipsilesional hemisphere brain tissue harvested from mice at 48 h post-MCAO to investigate the effects of PPARα KO on gene expression during the subacute stroke phase. We further analyzed differentially expressed genes (DEGs) between PPARα KO brains and WT brains to better characterize molecular pathways through which the loss of PPARα exacerbated ischemic stroke. We first compared the overall transcriptomes of PPARα KO mouse stroke brains with those of WT stroke brains (PPARα KO MCAO vs. WT MCAO). Principal component analysis (PCA) showed the clear separation of PPARα KO MCAO and WT MCAO brain transcriptomes along PC1, which accounted for the greatest variability (27.26%) in the data (Figure 2A). These PCA results suggested widespread transcriptomic differences in brain tissue due to PPARα deletion. Overall, we found 166 downregulated and 140 upregulated genes in PPARα KO stroke brains with a fold change >1.5 in either direction (FDR < 0.05) (Figure 2B). PPARα KO brains had upregulated expression of several genes already implicated in neural injury following stroke such as Mmp19, Il6, Saa3, and Il1r2 (Figure 2B). We also observed the downregulation of genes well known to be neuroprotective following stroke such as Sod3 and Bdnf (Figure 2B). To further characterize cellular processes affected in stroke brains by the genetic deletion of PPARα, we conducted gene set enrichment analysis (GSEA) on DEGs between PPARα KO MCAO and WT MCAO brain transcriptomes. In PPARα KO stroke brains, we observed the enrichment of Gene Ontology (GO) Biological Processes and Molecular Functions for the following gene sets: the positive regulation of leukocyte proliferation (−log10 (p-value) = 4.501, Enrichment = 7.871), positive regulation of apoptotic process (−log10 (p-value) = 3.345, Enrichment = 3.349), acute-phase response (−log10 (p-value) = 3.137, Enrichment = 16.962), and cellular component disassembly (−log10 (p-value) = 2.235, Enrichment = 3.699) (Figure 2C). Heatmaps comparing the expression of key genes for each respective GO pathway between PPARα KO MCAO brains and WT MCAO brains are presented in Figure 2D.

2.3. PPARα Deletion Alters Gene Expression Signatures for TNFα Signaling, IL6 Signaling, and Epithelial–Mesenchymal Transition (EMT) in Stroke Brains

We further analyzed our RNA-seq data to identify enriched or depleted cellular pathways resulting from PPARα KO in mouse stroke brains. GSEA revealed positive normalized enrichment scores (NES), indicating the upregulation of Hallmark gene sets for TNFα signaling via NFKB (NES = 1.442), IL6/JAK/STAT3 signaling (NES = 1.435), and epithelial–mesenchymal transition (EMT) (NES = 1.308) in the brains of PPARα KO MCAO mice (Figure 3A). The positive enrichment scores across these pathways indicate their upregulation in ischemic brain tissue from the loss of PPARα and point to a potential protective or modulatory effect of PPARα against ischemia-induced pathological changes in gene expression. Key TNFα signaling-related genes that were upregulated in PPARα KO MCAO brains included Il6, Tnfsf9, Cxcl1, Cxcl2, Tnfaip6, Tnfaip2, Il23a, Nfkbia, and Tnf. Key genes related to IL6/JAK/STAT3 signaling that were upregulated in PPARα KO MCAO brains included Il6, Il1r2, Il12rb1, Il4ra, Socs3, Fas, Tnfrsf12a, and Tnf. Key EMT-related genes upregulated in PPARα KO MCAO brains included Il6, Mmp3, Fbln2, Fbn2, Wnt5a, Itga2, Itga5, Col4a1, Fstl3, Fstl1, and Fgf2 (Figure 3B and Figure A2). Clinical evidence shows that blood and cerebrospinal levels of IL6 are significantly elevated in patients following stroke [19,20,21]; the JAK/STAT3 pathway is highly implicated in post-stroke inflammation by inducing the transcription of inflammatory mediators like cytokines and chemokines [22]. Similarly, TNFα levels increase in the brain and blood following stroke and increase neuroinflammation [23,24]. Previous research links TNFα levels to worse stroke patient outcomes, and preclinical studies demonstrate that TNFα signaling recruits immune cells to the CNS, disrupts BBB function, and promotes brain cell apoptosis during ischemic stroke [21,25]. In addition to inflammation, epithelial-to-mesenchymal transition (EMT) and endothelial-to-mesenchymal transition (EndMT) are processes whereby epithelial or endothelial cells lose their typical morphology and function and gain mesenchymal properties. Following the acute phase of stroke, the inflammatory and hypoxic environment in the brain can trigger EMT/EndMT in cells that form the BBB, which promotes its breakdown, thereby exacerbating edema and allowing the infiltration of immune cells into the brain [26]. The resulting neuroinflammation can worsen tissue damage and impede stroke recovery. Additionally, EndMT may promote fibrosis in the brain vasculature, further impairing blood flow and contributing to long-term injury following stroke [27]. Overall, our results suggest that PPARα signaling may blunt signaling pathways that promote inflammation and BBB breakdown in the subacute stroke phase to reduce the infarct volume.

2.4. Ingenuity Pathway Analysis Reveals TNFα as an Upstream Regulator Affected by PPARα KO in Mouse Stroke Brains

We conducted Ingenuity Pathway Analysis (IPA) on our RNA-seq data to further interrogate the cellular pathways affected by PPARα KO in mouse stroke brains. Similar to GSEA, IPA revealed significant transcriptional reprogramming of TNFα signaling gene signatures in PPARα KO stroke brains 48 h post-MCAO. We observed decreased expression of several genes known to be suppressed by TNFα in PPARα KO brains such as Cxcl12, Scd1, Ucp3, Abcd2, Arc, Etv5, Per2, Adrb1, Ccr5, and Rxrg (Figure 4). In agreement, we also observed an increased expression profile for many genes known to be upregulated by TNFα in stroke brains from PPARα KO mice. Specifically, PPARα KO led to increased expression levels of Gadd45b, Nppa, Hdc, Lcn2, Il6, Sox4, Marcksl1, Saa3, Ucn2, Met, Dusp10, Elovl7, Ngf, C3, Il11, Hmox1, Alox5, Tnfsf9, Angptl4, Plin2, H19, Csf2rb, Atf3, and Col7a1 following stroke (Figure 4). The overall expression pattern of the downstream targets indicates that TNFα is an activated upstream regulator in PPARα KO stroke brains compared with WT stroke brains (Figure A3). The predominance of upregulated downstream targets (red nodes) suggests that TNFα drives a pro-inflammatory transcriptional program following brain ischemia and that PPARα normally suppresses this response.

3. Discussion

Ischemic stroke pathophysiology is complex and involves blood–brain barrier (BBB) breakdown, oxidative stress, neuroinflammation, and the ultimate death of CNS resident cells [28]. Thus, future pharmacological therapies for stroke should prioritize druggable targets that can limit brain tissue damage through multiple mechanisms. PPARα is well known for its role in vascular function, and PPARα agonists such as clofibrate, fenofibrate, and gemfibrozil are used in the clinic as antilipemic agents to manage cardiovascular disease. PPARα is expressed in endothelial cells (ECs) [29], vascular smooth muscle cells (VSMCs) [30], and immune cells such as monocytes and macrophages [31], where it functions as a ligand-activated transcription factor to regulate gene expression. Ligand-activated PPARα dimerizes with the retinoid X receptor (RXR) [7] and associates with PPAR response elements (PPREs) in the promoter regions of genes to regulate their transcription. PPARα can also repress gene expression by binding and inhibiting other transcription factors like nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) and activator protein-1 (AP1) [32,33]. The anti-inflammatory effects of fibrates are mostly attributed to the PPARα-mediated suppression of inflammatory gene expression in vascular and immune cells.
The activation of PPARα improves outcomes in rodent models of ischemic stroke. In Apolipoprotein E (ApoE)-deficient mice, pretreatment with fenofibrate for 14 days (d) prior to MCAO resulted in reduced infarct size and increased antioxidant activity from superoxide dismutase, glutathione reductase, glutathione peroxidase, and glutathione S-transferase [11]. Another study using male Wistar rats reported that 14 d pretreatment with fenofibrate reduced the infarct volume and markers of both neuroinflammation and oxidative stress following ischemic stroke [12]. Other researchers reported that daily treatment with fenofibrate for 7 d prior to 1 h MCAO resulted in upregulated gene expression of all three superoxide dismutase (SOD) isoforms in the brain microvasculature and reduced levels of reactive oxygen species (ROS) and oxidative damage in adult male mice [13]. Together, these studies highlight PPARα agonists as potential pharmacological candidates for stroke therapy. However, the transcriptional pathways that PPARα regulates to protect the brain from ischemia are not yet fully understood.
To address these gaps in our knowledge, we performed the RNA-sequencing of ipsilesional hemisphere brain tissue harvested from WT and PPARα KO adult male mice at 48 h post-MCAO. Through bioinformatic analysis on our RNA-sequencing data, we were able to identify several PPARα KO-regulated transcriptional pathways associated with increased stroke infarct sizes. Our results agree with previous studies that implicate PPARα in regulating superoxide dismutase enzyme expression. In our study, PPARα KO significantly downregulated the brain expression of superoxide dismutase 3 (Sod3) by 48 h post-MCAO. SOD3 is an extracellular antioxidant enzyme that protects against ischemic stroke by scavenging ROS produced during ischemia–reperfusion injury [34]. Increased SOD3 is also associated with reduced BBB damage, smaller infarct volumes, and improved neurological outcomes following ischemic stroke [35]. Thus, our data also suggest a link between PPARα and the expression of antioxidant genes post-stroke. We observed a decreased expression of other well-known neuroprotective genes like brain-derived neurotrophic factor (Bdnf) 48 h post-MCAO in PPARα KO mouse brains. BDNF promotes the survival and growth of neurons, enhances synaptic plasticity, and promotes the repair and regeneration of brain tissue following ischemic injury [36]. Overall, our RNA-seq analysis identified several well-established neuroprotective genes regulated by PPARα that may explain the increased infarct size observed in our PPARα KO mice.
Interestingly, we also observed that the genetic deletion of PPARα increased the expression of several genes that are strongly implicated in ischemic stroke pathophysiology. This highlights a potential neuroprotective role of PPARα-mediated gene repression in the brain following stroke. Specifically, we observed the upregulation of several inflammatory genes such as C3, Il6, and Il1r2 48 h post-MCAO in PPARα KO mouse brains, which correlated with increased infarct size. Gene Ontology (GO) analysis also identified the enrichment of gene signatures for the positive regulation of leukocyte proliferation and the acute-phase response in the brains of PPARα KO mice. Thus, our data agree with previous studies in which fenofibrate treatment reduced markers of neuroinflammation in rodent models of stroke. Of note, IL1 signaling and C3 protein levels are known to be increased following stroke and correlate with worse clinical outcomes in ischemic stroke patients [37,38,39]. Similarly, blood and cerebrospinal fluid levels of IL6 increase following stroke and correlate with stroke severity and worse stroke outcomes [19,20,21]. Specifically, IL6 signaling through the Janus kinase (JAK) and signal transducer and activator of transcription 3 (STAT3) pathway plays a major role in post-stroke inflammatory responses through the transcription of various inflammatory genes [22]. In the acute phase of stroke, high levels of IL6 can contribute to an inflammatory response by activating immune cells and promoting the release of other pro-inflammatory cytokines. Further bioinformatic analysis of our RNA-seq data revealed the upregulation of other genes in the IL6/JAK/STAT3 expression signatures in mouse stroke brains upon the loss of PPARα, as well. For instance, we found that PPARα KO increased the expression of Il12rb1 (Interleukin-12 receptor beta 1) by 48 h post-stroke; signaling through IL12Rβ1 promotes interferon-gamma (IFNγ) production [40], which worsens neuroinflammation post-stroke by activating immune cells to produce more inflammatory cytokines and chemokines. We also noted a higher expression of Socs3 (suppressor of cytokine signaling 3) in PPARα KO brains after stroke. Reducing SOCS3 levels in the brain is reported to polarize macrophages towards the M2 phenotype and promote the resolution of post-stroke inflammation [41]. Therefore, PPARα likely restrains the expression of genes linked to IL6/JAK/STAT3 signaling to prevent rampant neuroinflammation in the subacute stroke phase.
We also observed the upregulation of a pro-inflammatory tumor necrosis factor alpha (TNFα) gene signature in the brains of PPARα KO mice following stroke. Key genes in this signature included Tnfsf9, Cxcl1, Cxcl2, Il23a, Nfkbia, and Tnf. TNFα is a pro-inflammatory cytokine that is rapidly upregulated in the brain following ischemic stroke and promotes the recruitment of immune cells to the site of injury [24]. In addition, TNFα can induce apoptosis in neurons and other brain cells and contribute to BBB dysfunction, thus further increasing tissue damage after stroke [23,24]. CXCL1/CXCL2 are chemokines that promote the migration of neutrophils and other immune cells to the infarcted area, which causes further tissue loss [25,42]. Additionally, TNFSF9 (tumor necrosis factor superfamily member 9) is upregulated in response to cerebral ischemia–reperfusion injury and can exacerbate inflammation and cell death by promoting ferroptosis and apoptosis in brain microvascular endothelial cells [43]. Of note, elevated TNFα levels are linked to worse outcomes in stroke patients, and overwhelming preclinical evidence shows that TNFα signaling increases brain damage following stroke through sustained neuroinflammation, the disruption of BBB function, and the promotion of neural apoptosis [21,25]. Thus, PPARα likely curtails neuroinflammation following stroke through the regulation of TNFα signaling-related genes.
As mentioned previously, BBB disruption and brain cell death coincide with neuroinflammation and are key hallmarks of stroke that lead to long-term neurological disability. Reperfusion injury following stroke damages the BBB and ultimately results in the loss of tight junctions between endothelial cells that form this crucial neurovascular structure. EMT/EndMT (epithelial–mesenchymal transition/endothelial–mesenchymal transition) is a process whereby endothelial cells lose their characteristics and gain mesenchymal properties. In the context of stroke, EMT/EndMT contributes to BBB breakdown by promoting endothelial cell dysfunction and increasing vascular permeability [27]. Other processes such as apoptosis, necroptosis, and the destruction of cellular components that form tight junctions can cause BBB breakdown and worsen stroke outcomes [44,45]. The apoptosis of neurons, glial cells, and brain microvascular cells post-stroke can occur intrinsically due to mitochondrial energy failure, oxidative stress, and calcium imbalance [46]. Immediately following stroke, the acute-phase response is activated and initiates an innate immune response to the ischemic lesion to clear dead and damaged cells. Thus, brain cell loss also occurs through the extrinsic apoptotic pathway mediated by death receptor signaling from immune cells that infiltrate the brain following stroke [46]. While this plays a role in wound healing and repair, prolonged activation and neuroinflammation contribute to extensive brain cell loss and worsen stroke outcomes. Interestingly, we observed the transcriptional enrichment of several of these pathways in the brains of our PPARα KO mice during the subacute stroke phase. Specifically, the GO analysis of our RNA-seq data revealed that the loss of PPARα in the brain led to the enrichment of gene sets related to the acute-phase response, apoptosis, and cellular component disassembly 48 h post-MCAO. Thus, in addition to limiting neuroinflammation, PPARα activation likely protects the brain following stroke by also modulating genes involved in apoptosis, EMT, and the disassembly of cellular components.
PPARα functions as a ligand-activated transcription factor, and we identified hundreds of genes that were differentially regulated in the post-stroke brain upon its genetic deletion. Although we delineated several transcriptional pathways modulated by PPARα, it remains unknown which of these genes are directly regulated by PPARα’s transcription factor activity at regulatory elements upstream of transcription start sites or whether PPARα modulates the expression of some of our identified genes in an indirect manner by modulating the expression of other upstream regulators and transcription factors. Although previous studies in animal models of stroke have reported neuroprotective effects from treatment with PPARα agonists like fenofibrate, further genetic approaches are needed to corroborate these findings. A complementary positive approach using gain-of-function studies with constitutive PPARα signaling in different types of brain cells will help to validate our findings and identify additional PPARα-regulated pathways that may benefit stroke outcomes. Future studies on the precise functions of PPARα in the brain during ischemic stroke will benefit from the use of epigenetic profiling in parallel with RNA-seq or other gene expression assays, to determine the genes that are directly regulated by PPARα’s transcription factor binding activity.
Previous studies using transgenic PPARα KO mice confirmed that PPARα regulates lipid metabolism and cellular energy balance [47,48]. These disruptions may lead to conditions like insulin resistance, obesity, or fatty liver disease, all of which can negatively impact the body’s response to injury and ability to recover. The pathophysiology of ischemic stroke involves energy failure resulting from oxygen and glucose deprivation. Thus, any alterations to cellular metabolism in the CNS are likely to impact stroke outcomes. Future studies using PPARα transgenic mice to model ischemic stroke may consider monitoring physiological and biochemical variables, such as lipid profiles, glucose levels, and liver function, to better assess how the metabolic consequences of PPARα modulation may affect stroke pathophysiology. In addition, we used male transgenic mice for our rodent model of stroke. Although we observed PPARα KO-associated modulation of several pathways linked to increased infarct volume in male stroke brains, it is possible that PPARα modulates transcriptional pathways in the CNS differently between males and females. Future studies would benefit from a comparison of the effect of PPARα KO on stroke outcomes in both male and female transgenic mice and the use of ChIP-seq and proteomics to interrogate whether PPARα cooperates with androgen and estrogen receptors in the nucleus of brain cells to influence gene expression differentially according to biological sex. Nevertheless, our study highlights PPARα as an attractive candidate for pharmacological stroke therapy that warrants further research.

4. Materials and Methods

4.1. Animals

Transgenic PPARα KO mice and littermate controls (8–12 weeks) were obtained from Jackson Laboratories (B6; 129S4-Ppara tm1Gonz/J, Bar Harbor, ME, USA). Mice were kept at 18–22 °C on a 12 h light–dark cycle. Mice were given food and water ad libitum.

4.2. Mouse Model of Stroke

Tulane University (New Orleans, LA, USA) and the University of California, Riverside (Riverside, CA, USA) Institutional Animal Care and Use Committees reviewed and approved animal use for this study. Animals were cared for and treated according to the guidelines of Tulane University and UCR animal protocols, the National Institutes of Health Guide for the Care and Use of Laboratory Animals, and the American Veterinary Medical Association.
We used middle cerebral artery occlusion followed by reperfusion to model transient focal cerebral ischemia in mice [49]. Stroke surgery was performed as previously published [50,51,52,53]. MCAO was performed for 1 h using a 6-0 nylon monofilament (Doccol Corporation, Sharon, MA, USA), and then the filament was removed to allow for reperfusion. For sham surgical controls, mouse cerebral arteries were inserted with the filament followed by immediate removal. Successful MCAO was defined as a reduction in regional cerebral blood flow (rCBF) by >80% and was assessed with a transcranial laser Doppler (Perimed Inc., Las Vegas, NV, USA). Blood reperfusion to >90% of baseline rCBF was the criterion used to determine successful post-MCAO/R recovery.

4.3. Infarct Volume Quantification

Brain ischemic lesions were visualized by sectioning mouse brains and staining with 2,3,5-triphenyl tetrazolium chloride (TTC, Sigma-Aldrich, St. Louis, MO, USA). Mouse brains were harvested 48 h after MCAO and sliced into 1 mm coronal sections. Brain sections were incubated in 2% TTC solution as previously described [50,51,52]. ImageJ v4 software (National Institutes of Health, Bethesda, MD, USA) was used to calculate the infarct area of stained brain sections. The cerebral infarct volume was calculated as a percentage of the contralateral hemisphere using the following formula: [volume of contralateral hemisphere − (volume of total ipsilesional hemisphere − volume of infarct area)]/volume of contralateral hemisphere × 100. This calculation method was able to compensate for post-stroke edema [54].

4.4. RNA Sequencing (RNA-Seq)

Cellular mRNA was isolated using the NEBNext® Poly(A) mRNA Magnetic Isolation Kit. The NEBNext® Ultra™ Directional RNA Library Prep Kit for Illumina® (NEB, Ipswich, MA, USA) was used to construct libraries for RNA sequencing. Libraries were pooled and sequenced as single-end 75 bp on an Illumina NextSeq 500 sequencer with a sequencing depth of 18–30 million reads.

4.4.1. RNA-Seq Data Processing

RNA-seq read trimming, alignment, gene quantification, normalization, and QC were performed using the nf-core rnaseq pipeline v3.14.0 [55] with the --aligner star_rsem parameter. The Mus musculus (house mouse) genome assembly GRCm38 (mm10) and Gencode vM25 gene annotations [56] were used for alignment.

4.4.2. Quality Control and Assurance (QC/QA) of RNA-Seq Data

RNA sequencing alignment and quantification quality was assessed by FastQC and MultiQC using the nf-core rnaseq pipeline described above. Biological replicate transcriptome concordance was assessed using principal component analysis (PCA).

4.4.3. Differential Gene Expression and Gene Set Enrichment Analysis (GSEA)

Differential gene expression analysis and GSEA were performed using the nf-core differential abundance pipeline v1.5.0 using the following parameters: --filtering_min_abundance 4 --filtering_min_proportion 0.50 --gsea_run true --gsea_permute “gene_set” --gsea_nperm 1000. The mouse gene sets v2023.2.Mm from MSigDB were used for GSEA. Differentially expressed genes (DEGs) were defined by a minimum fold-change of 1.25 and FDR < 0.05.

4.4.4. Pathway Analysis with Metascape and IPA

Pathway and Upstream Regulator Analyses on differentially expressed genes were performed using Metascape [57] and QIAGEN Ingenuity Pathway Analysis (IPA) software 24.0.1 (QIAGEN Inc., Germantown, MD, USA) [58].

4.5. Statistical Analysis

Statistical analysis was conducted using R software 4.4.1 (R Foundation for Statistical Computing, Vienna, Austria) and GraphPad Prism 10 (GraphPad Software, LLC, Boston, MA, USA). Unless stated otherwise, one-way ANOVA with Fisher’s LSD post hoc test was used to assess differences between multiple groups. Results were considered statistically significant at p < 0.05. Data are presented as mean ± SEM.

Author Contributions

J.-P.L. conceived the study, designed the experiments, and analyzed and interpreted data. M.H.H. designed and performed the experiments and analyzed and interpreted data. R.M. analyzed data and prepared figures; A.C.B. analyzed data; and A.C.B., M.H.H. and J.-P.L. performed experiments and drafted, edited, and revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

Jean-Pyo Lee is supported by the National Institute of Neurological Disorders and Stroke (Grant 7R01NS110370-06). Jean-Pyo Lee and Milton Hamblin are both supported by startup funds from the Division of Biomedical Sciences, School of Medicine at the University of California, Riverside.

Institutional Review Board Statement

The animal study protocol was approved by the Institutional Animal Care and Use Committee (IACUC) of Tulane University and the University of California, Riverside, for studies involving animals.

Informed Consent Statement

Not applicable.

Data Availability Statement

The raw data supporting the conclusions of this article will be made available by the authors on reasonable request.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the study design, data collection, analysis, interpretation, manuscript writing, or decision to publish the results.

Appendix A

Figure A1. PPARα knockout increases infarct volume after stroke. TTC-stained mouse brain sections 48 h post-stroke show infarcted tissue (white) and viable tissue (red). Ischemic damage is shown in both the caudoputamen and cortex. Panels show representative coronal sections: (A) Sham WT, (B) Sham PPARα KO, (C) MCAO/R WT, and (D) MCAO/R PPARα KO. Each panel shows sections from two different brains. MCAO/R, middle cerebral artery occlusion with reperfusion. Scale bar = 2 mm.
Figure A1. PPARα knockout increases infarct volume after stroke. TTC-stained mouse brain sections 48 h post-stroke show infarcted tissue (white) and viable tissue (red). Ischemic damage is shown in both the caudoputamen and cortex. Panels show representative coronal sections: (A) Sham WT, (B) Sham PPARα KO, (C) MCAO/R WT, and (D) MCAO/R PPARα KO. Each panel shows sections from two different brains. MCAO/R, middle cerebral artery occlusion with reperfusion. Scale bar = 2 mm.
Ijms 26 04082 g0a1
Figure A2. Clustering analysis of mouse Hallmark gene sets related to (A) TNFα signaling via NFKB, (B) IL6/JAK/STAT3 signaling, and (C) Epithelial–Mesenchymal Transition in PPARα KO MCAO (n = 4) compared with WT MCAO (n = 4) mouse brains harvested 48 h post-stroke. Row-wise z-scores were computed using transcripts per million (TPM). MCAO, middle cerebral artery occlusion with reperfusion.
Figure A2. Clustering analysis of mouse Hallmark gene sets related to (A) TNFα signaling via NFKB, (B) IL6/JAK/STAT3 signaling, and (C) Epithelial–Mesenchymal Transition in PPARα KO MCAO (n = 4) compared with WT MCAO (n = 4) mouse brains harvested 48 h post-stroke. Row-wise z-scores were computed using transcripts per million (TPM). MCAO, middle cerebral artery occlusion with reperfusion.
Ijms 26 04082 g0a2
Figure A3. Predicted activation of TNFα as an upstream regulator in PPARα KO stroke brains. Upstream Regulator Analysis in Ingenuity Pathway Analysis (IPA) predicts TNFα activation based on differentially expressed target genes in PPARα KO MCAO (n = 4) vs. WT MCAO (n = 4) mouse brains 48 h post-stroke. Upregulated targets (red) and downregulated targets (green) indicate TNFα-driven transcriptional changes in PPARα KO stroke vs. WT stroke. TNFα is predicted to be activated (center, orange) based on the collective expression patterns of its downstream targets. (includes others)* indicates that the regulator is part of a broader group or network. MCAO, middle cerebral artery occlusion with reperfusion.
Figure A3. Predicted activation of TNFα as an upstream regulator in PPARα KO stroke brains. Upstream Regulator Analysis in Ingenuity Pathway Analysis (IPA) predicts TNFα activation based on differentially expressed target genes in PPARα KO MCAO (n = 4) vs. WT MCAO (n = 4) mouse brains 48 h post-stroke. Upregulated targets (red) and downregulated targets (green) indicate TNFα-driven transcriptional changes in PPARα KO stroke vs. WT stroke. TNFα is predicted to be activated (center, orange) based on the collective expression patterns of its downstream targets. (includes others)* indicates that the regulator is part of a broader group or network. MCAO, middle cerebral artery occlusion with reperfusion.
Ijms 26 04082 g0a3

References

  1. Martin, S.S.; Aday, A.W.; Almarzooq, Z.I.; Anderson, C.A.M.; Arora, P.; Avery, C.L.; Baker-Smith, C.M.; Barone Gibbs, B.; Beaton, A.Z.; Boehme, A.K.; et al. 2024 Heart Disease and Stroke Statistics: A Report of US and Global Data from the American Heart Association. Circulation 2024, 149, e347–e913. [Google Scholar] [CrossRef]
  2. Gelderblom, M.; Leypoldt, F.; Steinbach, K.; Behrens, D.; Choe, C.U.; Siler, D.A.; Arumugam, T.V.; Orthey, E.; Gerloff, C.; Tolosa, E.; et al. Temporal and spatial dynamics of cerebral immune cell accumulation in stroke. Stroke 2009, 40, 1849–1857. [Google Scholar] [CrossRef]
  3. Wang, J.; Fields, J.; Zhao, C.; Langer, J.; Thimmulappa, R.K.; Kensler, T.W.; Yamamoto, M.; Biswal, S.; Doré, S. Role of Nrf2 in protection against intracerebral hemorrhage injury in mice. Free Radic. Biol. Med. 2007, 43, 408–414. [Google Scholar] [CrossRef] [PubMed]
  4. Qureshi, A.I.; Ali, Z.; Suri, M.F.; Shuaib, A.; Baker, G.; Todd, K.; Guterman, L.R.; Hopkins, L.N. Extracellular glutamate and other amino acids in experimental intracerebral hemorrhage: An in vivo microdialysis study. Crit. Care Med. 2003, 31, 1482–1489. [Google Scholar] [CrossRef]
  5. Sims, N.R.; Muyderman, H. Mitochondria, oxidative metabolism and cell death in stroke. Biochim. Biophys. Acta 2010, 1802, 80–91. [Google Scholar] [CrossRef] [PubMed]
  6. Pyper, S.R.; Viswakarma, N.; Yu, S.; Reddy, J.K. PPARα: Energy combustion, hypolipidemia, inflammation and cancer. Nucl. Recept. Signal. 2010, 8, e002. [Google Scholar] [CrossRef] [PubMed]
  7. Miyata, K.S.; McCaw, S.E.; Marcus, S.L.; Rachubinski, R.A.; Capone, J.P. The peroxisome proliferator-activated receptor interacts with the retinoid X receptor in vivo. Gene 1994, 148, 327–330. [Google Scholar] [CrossRef]
  8. Hamblin, M.; Chang, L.; Fan, Y.; Zhang, J.; Chen, Y.E. PPARs and the cardiovascular system. Antioxid. Redox Signal. 2009, 11, 1415–1452. [Google Scholar] [CrossRef]
  9. Lefebvre, P.; Chinetti, G.; Fruchart, J.C.; Staels, B. Sorting out the roles of PPAR alpha in energy metabolism and vascular homeostasis. J. Clin. Investig. 2006, 116, 571–580. [Google Scholar] [CrossRef]
  10. Khuchua, Z.; Glukhov, A.I.; Strauss, A.W.; Javadov, S. Elucidating the Beneficial Role of PPAR Agonists in Cardiac Diseases. Int. J. Mol. Sci. 2018, 19, 3464. [Google Scholar] [CrossRef]
  11. Deplanque, D.; Gelé, P.; Pétrault, O.; Six, I.; Furman, C.; Bouly, M.; Nion, S.; Dupuis, B.; Leys, D.; Fruchart, J.C.; et al. Peroxisome proliferator-activated receptor-alpha activation as a mechanism of preventive neuroprotection induced by chronic fenofibrate treatment. J. Neurosci. 2003, 23, 6264–6271. [Google Scholar] [CrossRef] [PubMed]
  12. Ouk, T.; Laprais, M.; Bastide, M.; Mostafa, K.; Gautier, S.; Bordet, R. Withdrawal of fenofibrate treatment partially abrogates preventive neuroprotection in stroke via loss of vascular protection. Vascul. Pharmacol. 2009, 51, 323–330. [Google Scholar] [CrossRef] [PubMed]
  13. Wang, G.; Liu, X.; Guo, Q.; Namura, S. Chronic treatment with fibrates elevates superoxide dismutase in adult mouse brain microvessels. Brain Res. 2010, 1359, 247–255. [Google Scholar] [CrossRef]
  14. Matzen, J.S.; Krogh, C.L.; Forman, J.L.; Garred, P.; Møller, K.; Bache, S. Lectin complement pathway initiators after subarachnoid hemorrhage—An observational study. J. Neuroinflamm. 2020, 17, 338. [Google Scholar] [CrossRef] [PubMed]
  15. Xuan, A.G.; Chen, Y.; Long, D.H.; Zhang, M.; Ji, W.D.; Zhang, W.J.; Liu, J.H.; Hong, L.P.; He, X.S.; Chen, W.L. PPARα Agonist Fenofibrate Ameliorates Learning and Memory Deficits in Rats Following Global Cerebral Ischemia. Mol. Neurobiol. 2015, 52, 601–609. [Google Scholar] [CrossRef]
  16. Collino, M.; Aragno, M.; Mastrocola, R.; Benetti, E.; Gallicchio, M.; Dianzani, C.; Danni, O.; Thiemermann, C.; Fantozzi, R. Oxidative stress and inflammatory response evoked by transient cerebral ischemia/reperfusion: Effects of the PPAR-α agonist WY14643. Free Radic. Biol. Med. 2006, 41, 579–589. [Google Scholar] [CrossRef]
  17. Ouk, T.; Gautier, S.; Pétrault, M.; Montaigne, D.; Maréchal, X.; Masse, I.; Devedjian, J.C.; Deplanque, D.; Bastide, M.; Nevière, R.; et al. Effects of the PPAR-α agonist fenofibrate on acute and short-term consequences of brain ischemia. J. Cereb. Blood Flow Metab. 2014, 34, 542–551. [Google Scholar] [CrossRef]
  18. Mysiorek, C.; Culot, M.; Dehouck, L.; Derudas, B.; Staels, B.; Bordet, R.; Cecchelli, R.; Fenart, L.; Berezowski, V. Peroxisome-proliferator-activated receptor-alpha activation protects brain capillary endothelial cells from oxygen-glucose deprivation-induced hyperpermeability in the blood-brain barrier. Curr. Neurovasc. Res. 2009, 6, 181–193. [Google Scholar] [CrossRef]
  19. Jenny, N.S.; Callas, P.W.; Judd, S.E.; McClure, L.A.; Kissela, B.; Zakai, N.A.; Cushman, M. Inflammatory cytokines and ischemic stroke risk: The REGARDS cohort. Neurology 2019, 92, e2375–e2384. [Google Scholar] [CrossRef]
  20. Yao, H.; Zhang, Y.; Shu, H.; Xie, B.; Tao, Y.; Yuan, Y.; Shang, Y.; Yuan, S.; Zhang, J. Hyperforin Promotes Post-stroke Neuroangiogenesis via Astrocytic IL-6-Mediated Negative Immune Regulation in the Ischemic Brain. Front. Cell. Neurosci. 2019, 13, 201. [Google Scholar] [CrossRef]
  21. Băcilă, C.-I.; Vlădoiu, M.-G.; Văleanu, M.; Moga, D.-F.-C.; Pumnea, P.-M. The Role of IL-6 and TNF-Alpha Biomarkers in Predicting Disability Outcomes in Acute Ischemic Stroke Patients. Life 2025, 15, 47. [Google Scholar] [CrossRef] [PubMed]
  22. Erta, M.; Quintana, A.; Hidalgo, J. Interleukin-6, a major cytokine in the central nervous system. Int. J. Biol. Sci. 2012, 8, 1254–1266. [Google Scholar] [CrossRef] [PubMed]
  23. Watters, O.; O’Connor, J.J. A role for tumor necrosis factor-α in ischemia and ischemic preconditioning. J. Neuroinflamm. 2011, 8, 87. [Google Scholar] [CrossRef]
  24. Clausen, B.H.; Wirenfeldt, M.; Høgedal, S.S.; Frich, L.H.; Nielsen, H.H.; Schrøder, H.D.; Østergaard, K.; Finsen, B.; Kristensen, B.W.; Lambertsen, K.L. Characterization of the TNF and IL-1 systems in human brain and blood after ischemic stroke. Acta Neuropathol. Commun. 2020, 8, 81. [Google Scholar] [CrossRef] [PubMed]
  25. Denorme, F.; Portier, I.; Rustad, J.L.; Cody, M.J.; de Araujo, C.V.; Hoki, C.; Alexander, M.D.; Grandhi, R.; Dyer, M.R.; Neal, M.D.; et al. Neutrophil extracellular traps regulate ischemic stroke brain injury. J. Clin. Investig. 2022, 132, e154225. [Google Scholar] [CrossRef]
  26. Hansen, C.E.; Hollaus, D.; Kamermans, A.; de Vries, H.E. Tension at the gate: Sensing mechanical forces at the blood–brain barrier in health and disease. J. Neuroinflamm. 2024, 21, 325. [Google Scholar] [CrossRef]
  27. Chen, D.; Li, L.; Wang, Y.; Xu, R.; Peng, S.; Zhou, L.; Deng, Z. Ischemia-reperfusion injury of brain induces endothelial-mesenchymal transition and vascular fibrosis via activating let-7i/TGF-βR1 double-negative feedback loop. FASEB J 2020, 34, 7178–7191. [Google Scholar] [CrossRef]
  28. Qin, C.; Yang, S.; Chu, Y.-H.; Zhang, H.; Pang, X.-W.; Chen, L.; Zhou, L.-Q.; Chen, M.; Tian, D.-S.; Wang, W. Signaling pathways involved in ischemic stroke: Molecular mechanisms and therapeutic interventions. Signal Transduct. Target. Ther. 2022, 7, 215. [Google Scholar] [CrossRef]
  29. Inoue, I.; Shino, K.; Noji, S.; Awata, T.; Katayama, S. Expression of peroxisome proliferator-activated receptor α (PPAR α) in primary cultures of human vascular endothelial cells. Biochem. Biophys. Res. Commun. 1998, 246, 370–374. [Google Scholar] [CrossRef]
  30. 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]
  31. 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] [PubMed]
  32. Chinetti, G.; Fruchart, J.C.; Staels, B. Peroxisome proliferator-activated receptors (PPARs): Nuclear receptors at the crossroads between lipid metabolism and inflammation. Inflamm. Res. 2000, 49, 497–505. [Google Scholar] [CrossRef]
  33. Delerive, P.; De Bosscher, K.; Besnard, S.; Vanden Berghe, W.; Peters, J.M.; Gonzalez, F.J.; Fruchart, J.C.; Tedgui, A.; Haegeman, G.; Staels, B. Peroxisome proliferator-activated receptor alpha negatively regulates the vascular inflammatory gene response by negative cross-talk with transcription factors NF-κB and AP-1. J. Biol. Chem. 1999, 274, 32048–32054. [Google Scholar] [CrossRef] [PubMed]
  34. Maier, C.M.; Chan, P.H. Superoxide Dismutases in Stroke. In Handbook of Neurochemistry and Molecular Neurobiology: Acute Ischemic Injury and Repair in the Nervous System; Lajtha, A., Chan, P.H., Eds.; Springer US: New York, NY, USA, 2007; pp. 121–144. [Google Scholar]
  35. Mai, N.; Miller-Rhodes, K.; Prifti, V.; Kim, M.; O’Reilly, M.A.; Halterman, M.W. Lung-Derived SOD3 Attenuates Neurovascular Injury After Transient Global Cerebral Ischemia. J. Am. Heart Assoc. 2019, 8, e011801. [Google Scholar] [CrossRef]
  36. Liu, W.; Wang, X.; O’Connor, M.; Wang, G.; Han, F. Brain-Derived Neurotrophic Factor and Its Potential Therapeutic Role in Stroke Comorbidities. Neural Plast. 2020, 2020, 1969482. [Google Scholar] [CrossRef]
  37. Zhao, M.; Liu, A.; Wu, J.; Mo, L.; Lu, F.; Wan, G. Il1r2 and Tnfrsf12a in transcranial magnetic stimulation effect of ischemic stroke via bioinformatics analysis. Medicine 2024, 103, e36109. [Google Scholar] [CrossRef]
  38. Oh, S.H.; Kim, O.J.; Shin, D.A.; Song, J.; Yoo, H.; Kim, Y.K.; Kim, J.K. Alteration of immunologic responses on peripheral blood in the acute phase of ischemic stroke: Blood genomic profiling study. J. Neuroimmunol. 2012, 249, 60–65. [Google Scholar] [CrossRef] [PubMed]
  39. Yang, P.; Zhu, Z.; Zang, Y.; Bu, X.; Xu, T.; Zhong, C.; Wang, A.; Peng, H.; Guo, D.; Zheng, X.; et al. Increased Serum Complement C3 Levels Are Associated With Adverse Clinical Outcomes After Ischemic Stroke. Stroke 2021, 52, 868–877. [Google Scholar] [CrossRef]
  40. Eriksson, U.; Kurrer, M.O.; Sebald, W.; Brombacher, F.; Kopf, M. Dual Role of the IL-12/IFN-γ Axis in the Development of Autoimmune Myocarditis: Induction by IL-12 and Protection by IFN-γ1. J. Immunol. 2001, 167, 5464–5469. [Google Scholar] [CrossRef]
  41. Ji, X.C.; Shi, Y.J.; Zhang, Y.; Chang, M.Z.; Zhao, G. Reducing Suppressors of Cytokine Signaling-3 (SOCS3) Expression Promotes M2 Macrophage Polarization and Functional Recovery After Intracerebral Hemorrhage. Front. Neurol. 2020, 11, 586905. [Google Scholar] [CrossRef]
  42. De Filippo, K.; Dudeck, A.; Hasenberg, M.; Nye, E.; van Rooijen, N.; Hartmann, K.; Gunzer, M.; Roers, A.; Hogg, N. Mast cell and macrophage chemokines CXCL1/CXCL2 control the early stage of neutrophil recruitment during tissue inflammation. Blood 2013, 121, 4930–4937. [Google Scholar] [CrossRef] [PubMed]
  43. Liang, S.; Wu, Y.; Zhang, R.; Xu, L.; Xie, F. TNFSF9 Silence Impedes Cerebral Ischemia–Reperfusion Injury via Modulating SLC3A2 Expression in Brain Microvascular Endothelial Cells. J. Mol. Neurosci. 2025, 75, 12. [Google Scholar] [CrossRef]
  44. Chen, A.-Q.; Fang, Z.; Chen, X.-L.; Yang, S.; Zhou, Y.-F.; Mao, L.; Xia, Y.-P.; Jin, H.-J.; Li, Y.-N.; You, M.-F.; et al. Microglia-derived TNF-α mediates endothelial necroptosis aggravating blood brain–barrier disruption after ischemic stroke. Cell Death Dis. 2019, 10, 487. [Google Scholar] [CrossRef]
  45. Xue, S.; Zhou, X.; Yang, Z.-H.; Si, X.-K.; Sun, X. Stroke-induced damage on the blood–brain barrier. Front. Neurol. 2023, 14, 1248970. [Google Scholar] [CrossRef] [PubMed]
  46. Broughton, B.R.S.; Reutens, D.C.; Sobey, C.G. Apoptotic Mechanisms After Cerebral Ischemia. Stroke 2009, 40, e331–e339. [Google Scholar] [CrossRef] [PubMed]
  47. 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]
  48. Leone, T.C.; Weinheimer, C.J.; Kelly, D.P. A critical role for the peroxisome proliferator-activated receptor alpha (PPARalpha) in the cellular fasting response: The PPARalpha-null mouse as a model of fatty acid oxidation disorders. Proc. Natl. Acad. Sci. USA 1999, 96, 7473–7478. [Google Scholar] [CrossRef]
  49. Engel, O.; Kolodziej, S.; Dirnagl, U.; Prinz, V. Modeling Stroke in Mice—Middle Cerebral Artery Occlusion with the Filament Model. J. Vis. Exp. 2011, 47, e2423. [Google Scholar] [CrossRef]
  50. Huang, L.; Wong, S.; Snyder, E.Y.; Hamblin, M.H.; Lee, J.P. Human neural stem cells rapidly ameliorate symptomatic inflammation in early-stage ischemic-reperfusion cerebral injury. Stem Cell Res. Ther. 2014, 5, 129. [Google Scholar] [CrossRef]
  51. Eckert, A.; Huang, L.; Gonzalez, R.; Kim, H.S.; Hamblin, M.H.; Lee, J.P. Bystander Effect Fuels Human Induced Pluripotent Stem Cell-Derived Neural Stem Cells to Quickly Attenuate Early Stage Neurological Deficits After Stroke. Stem Cells Transl. Med. 2015, 4, 841–851. [Google Scholar] [CrossRef]
  52. Boese, A.C.; Eckert, A.; Hamblin, M.H.; Lee, J.P. Human neural stem cells improve early stage stroke outcome in delayed tissue plasminogen activator-treated aged stroke brains. Exp. Neurol. 2020, 329, 113275. [Google Scholar] [CrossRef] [PubMed]
  53. Hamblin, M.H.; Boese, A.C.; Murad, R.; Lee, J.P. MMP-3 Knockout Induces Global Transcriptional Changes and Reduces Cerebral Infarction in Both Male and Female Models of Ischemic Stroke. Int. J. Mol. Sci. 2024, 25, 7383. [Google Scholar] [CrossRef] [PubMed]
  54. Swanson, R.A.; Morton, M.T.; Tsao-Wu, G.; Savalos, R.A.; Davidson, C.; Sharp, F.R. A semiautomated method for measuring brain infarct volume. J. Cereb. Blood Flow Metab. 1990, 10, 290–293. [Google Scholar] [CrossRef]
  55. Ewels, P.A.; Peltzer, A.; Fillinger, S.; Patel, H.; Alneberg, J.; Wilm, A.; Garcia, M.U.; Di Tommaso, P.; Nahnsen, S. The nf-core framework for community-curated bioinformatics pipelines. Nat. Biotechnol. 2020, 38, 276–278. [Google Scholar] [CrossRef]
  56. Mudge, J.M.; Carbonell-Sala, S.; Diekhans, M.; Martinez, J.G.; Hunt, T.; Jungreis, I.; Loveland, J.E.; Arnan, C.; Barnes, I.; Bennett, R.; et al. GENCODE 2025: Reference gene annotation for human and mouse. Nucleic Acids Res. 2025, 53, D966–D975. [Google Scholar] [CrossRef] [PubMed]
  57. Zhou, Y.; Zhou, B.; Pache, L.; Chang, M.; Khodabakhshi, A.H.; Tanaseichuk, O.; Benner, C.; Chanda, S.K. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat. Commun. 2019, 10, 1523. [Google Scholar] [CrossRef]
  58. Krämer, A.; Green, J.; Pollard, J., Jr.; Tugendreich, S. Causal analysis approaches in Ingenuity Pathway Analysis. Bioinformatics 2014, 30, 523–530. [Google Scholar] [CrossRef]
Figure 1. PPARα KO increases stroke infarct volume. Infarct volume calculated by TTC staining of male mouse brains harvested 48 h post-MCAO (PPARα KO vs. WT). **** p < 0.0001 vs. sham; # p < 0.05 vs. MCAO/R WT. (n = 10, sham WT; n = 9, sham PPARα KO; n = 11, MCAO/R WT; n = 9, MCAO/R PPARα KO). Data are presented as mean ± SEM. MCAO/R, middle cerebral artery occlusion with reperfusion.
Figure 1. PPARα KO increases stroke infarct volume. Infarct volume calculated by TTC staining of male mouse brains harvested 48 h post-MCAO (PPARα KO vs. WT). **** p < 0.0001 vs. sham; # p < 0.05 vs. MCAO/R WT. (n = 10, sham WT; n = 9, sham PPARα KO; n = 11, MCAO/R WT; n = 9, MCAO/R PPARα KO). Data are presented as mean ± SEM. MCAO/R, middle cerebral artery occlusion with reperfusion.
Ijms 26 04082 g001
Figure 2. PPARα KO elicits global transcriptional changes in the brains of male mice by 48 h post-stroke. (A) Principal component analysis (PCA) of transcriptomes from PPARα KO MCAO (n = 4, red) and WT MCAO (n = 4, blue) mouse brains harvested at 48 h post-stroke. Each dot represents a biological replicate of an RNA-seq sample, and principal component 1 (PC1; 27.26% variance) splits the samples according to PPARα genetic status. (B) Volcano plot of differentially expressed genes (DEGs) between PPARα KO MCAO and WT MCAO samples. Genes downregulated by PPARα KO in stroke brains are colored blue. Genes upregulated by PPARα deletion in stroke brains are colored red. Representative DEGs are labeled in black. (C) Gene Ontology (GO) analysis results of DEGs between PPARα KO MCAO and WT MCAO brains. (D) Heatmaps of gene sets corresponding to positive regulation of apoptotic process, positive regulation of leukocyte proliferation, cellular component disassembly, and acute-phase response. Row-wise z-scores were computed using transcripts per million (TPM). Core enriched genes of interest in each gene set are labeled in black. Genes are clustered based on their expression patterns, with red indicating higher expression and blue indicating lower expression relative to the mean. MCAO, middle cerebral artery occlusion with reperfusion.
Figure 2. PPARα KO elicits global transcriptional changes in the brains of male mice by 48 h post-stroke. (A) Principal component analysis (PCA) of transcriptomes from PPARα KO MCAO (n = 4, red) and WT MCAO (n = 4, blue) mouse brains harvested at 48 h post-stroke. Each dot represents a biological replicate of an RNA-seq sample, and principal component 1 (PC1; 27.26% variance) splits the samples according to PPARα genetic status. (B) Volcano plot of differentially expressed genes (DEGs) between PPARα KO MCAO and WT MCAO samples. Genes downregulated by PPARα KO in stroke brains are colored blue. Genes upregulated by PPARα deletion in stroke brains are colored red. Representative DEGs are labeled in black. (C) Gene Ontology (GO) analysis results of DEGs between PPARα KO MCAO and WT MCAO brains. (D) Heatmaps of gene sets corresponding to positive regulation of apoptotic process, positive regulation of leukocyte proliferation, cellular component disassembly, and acute-phase response. Row-wise z-scores were computed using transcripts per million (TPM). Core enriched genes of interest in each gene set are labeled in black. Genes are clustered based on their expression patterns, with red indicating higher expression and blue indicating lower expression relative to the mean. MCAO, middle cerebral artery occlusion with reperfusion.
Ijms 26 04082 g002
Figure 3. PPARα KO transcriptionally upregulates EMT, TNFα signaling, and IL6/JAK/STAT3 signaling in the subacute stroke phase. (A) Normalized enrichment scores (NESs) comparing PPARα KO MCAO (n = 4) vs. WT MCAO (n = 4) brain transcriptomes for HALLMARK_TNFA_VIA_NFKB, HALLMARK_IL6_JAK_STAT3_SIGNALING, and HALLMARK_EPITHELIAL_MESENCHYMAL_TRANSITION gene set signatures. p < 0.05. (B) Expression heat maps of core enriched genes from panel A hallmark gene sets. Row-wise z-scores were computed using normalized transcripts per million (TPM) expression values. Core enriched genes of interest in each gene set are labeled in black. MCAO, middle cerebral artery occlusion with reperfusion.
Figure 3. PPARα KO transcriptionally upregulates EMT, TNFα signaling, and IL6/JAK/STAT3 signaling in the subacute stroke phase. (A) Normalized enrichment scores (NESs) comparing PPARα KO MCAO (n = 4) vs. WT MCAO (n = 4) brain transcriptomes for HALLMARK_TNFA_VIA_NFKB, HALLMARK_IL6_JAK_STAT3_SIGNALING, and HALLMARK_EPITHELIAL_MESENCHYMAL_TRANSITION gene set signatures. p < 0.05. (B) Expression heat maps of core enriched genes from panel A hallmark gene sets. Row-wise z-scores were computed using normalized transcripts per million (TPM) expression values. Core enriched genes of interest in each gene set are labeled in black. MCAO, middle cerebral artery occlusion with reperfusion.
Ijms 26 04082 g003
Figure 4. PPARα knockout alters TNFα-mediated gene expression in mouse stroke brains. Heatmap of genes downstream of TNFα signaling differentially expressed in PPARα KO MCAO (n = 4) vs. WT MCAO (n = 4) brains. Genes in blue are downregulated and genes in red are upregulated in PPARα KO stroke brains compared with WT stroke brains. TNFα signaling activity is inferred to be increased based on the expression patterns of its downstream targets. Row-wise z-scores were computed using normalized transcripts per million (TPM) expression values. MCAO, middle cerebral artery occlusion with reperfusion.
Figure 4. PPARα knockout alters TNFα-mediated gene expression in mouse stroke brains. Heatmap of genes downstream of TNFα signaling differentially expressed in PPARα KO MCAO (n = 4) vs. WT MCAO (n = 4) brains. Genes in blue are downregulated and genes in red are upregulated in PPARα KO stroke brains compared with WT stroke brains. TNFα signaling activity is inferred to be increased based on the expression patterns of its downstream targets. Row-wise z-scores were computed using normalized transcripts per million (TPM) expression values. MCAO, middle cerebral artery occlusion with reperfusion.
Ijms 26 04082 g004
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

Hamblin, M.H.; Boese, A.C.; Murad, R.; Lee, J.-P. PPARα Genetic Deletion Reveals Global Transcriptional Changes in the Brain and Exacerbates Cerebral Infarction in a Mouse Model of Stroke. Int. J. Mol. Sci. 2025, 26, 4082. https://doi.org/10.3390/ijms26094082

AMA Style

Hamblin MH, Boese AC, Murad R, Lee J-P. PPARα Genetic Deletion Reveals Global Transcriptional Changes in the Brain and Exacerbates Cerebral Infarction in a Mouse Model of Stroke. International Journal of Molecular Sciences. 2025; 26(9):4082. https://doi.org/10.3390/ijms26094082

Chicago/Turabian Style

Hamblin, Milton H., Austin C. Boese, Rabi Murad, and Jean-Pyo Lee. 2025. "PPARα Genetic Deletion Reveals Global Transcriptional Changes in the Brain and Exacerbates Cerebral Infarction in a Mouse Model of Stroke" International Journal of Molecular Sciences 26, no. 9: 4082. https://doi.org/10.3390/ijms26094082

APA Style

Hamblin, M. H., Boese, A. C., Murad, R., & Lee, J.-P. (2025). PPARα Genetic Deletion Reveals Global Transcriptional Changes in the Brain and Exacerbates Cerebral Infarction in a Mouse Model of Stroke. International Journal of Molecular Sciences, 26(9), 4082. https://doi.org/10.3390/ijms26094082

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