Next Article in Journal
The Pleiotropic Effect of ANRIL in Glaucoma and Cardiovascular Disease
Previous Article in Journal
Long COVID and Its Impacts: A Case–Control Study in Brazil
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

PPARgamma Modulates CD4+ T-Cell Differentiation and Allergic Inflammation in Allergic Rhinitis: A Potential Therapeutic Target

Department of Otolaryngology-Head and Neck Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
*
Author to whom correspondence should be addressed.
Biomedicines 2025, 13(7), 1616; https://doi.org/10.3390/biomedicines13071616
Submission received: 22 May 2025 / Revised: 26 June 2025 / Accepted: 29 June 2025 / Published: 1 July 2025
(This article belongs to the Section Cell Biology and Pathology)

Abstract

Objectives: Given the emerging role of peroxisome proliferator-activated receptor gamma (PPARgamma) in immune regulation and the increasing prevalence of allergic rhinitis (AR), we sought to understand how modulation of the PPARgamma pathway impacts the balance of CD4+ T-cell subsets, particularly regulatory T cells (Tregs) and T helper (TH)1, TH2, and TH17 cells, which are key players in the pathogenesis of AR. This knowledge is crucial for developing novel therapeutic strategies targeting the PPARgamma-CD4+ T-cell axis to manage AR more effectively. Methods: We used PPARgammaf/fLyz2-Cre mice for PPARgamma deletion. In an ovalbumin (OVA)-induced AR mouse model, PPARgamma+/-f/fLyz2-Cre mice were assessed for allergic symptoms, splenic Tregs, and nasal eosinophils. Additionally, the effects of a PPARgamma agonist on the polarization of naïve CD4+ T cells were examined. Results: PPARgamma+/-f/fLyz2-Cre mice showed worsened allergic symptoms, reduced splenic Tregs, and increased nasal mucosa eosinophilic infiltration. PPARgamma agonist treatment promoted naïve CD4+ T-cell polarization into Tregs and inhibited their differentiation into TH1, TH2, and TH17 subsets. Conclusions: Our findings indicate that PPARgamma plays a crucial role in regulating TH-cell subsets in AR. PPARgamma agonists could be a potential therapeutic strategy to mitigate allergic inflammation in AR by promoting Treg development and suppressing pathogenic TH-cell responses.

Graphical Abstract

1. Introduction

Allergic rhinitis (AR) is a prevalent chronic nasal condition characterized by symptoms such as paroxysmal sneezing, rhinorrhea, nasal congestion, and nasal pruritus [1]. Its global prevalence ranges from 10% to 40% of the population [2]. Current treatment approaches for AR emphasize a “four in one” strategy that includes environmental control, pharmacotherapy, immunotherapy, and health education [3]. However, existing therapeutic options often fail to fully alleviate patients’ clinical symptoms, may have associated side effects, and are characterized by a high recurrence rate [4]. Therefore, there is an urgent need to explore safer and more effective treatment methods for AR.
Peroxisome proliferator-activated receptor gamma (PPARgamma) is a receptor transcription factor belonging to the nuclear hormone receptor superfamily [5]. The biological functions of PPARgamma are multifaceted, primarily involving the regulation of lipid and glucose metabolism [6]. Additionally, PPARgamma has a significant role in the regulation of inflammatory responses and immune modulation [7,8]. Studies have proven that PPARgamma is widely present in immune cells and can regulate inflammation [9]. It can influence immune cell differentiation and secretion, inhibit the expression of pro-inflammatory cytokines in macrophages, and modulate the type 1/type 2 immunity balance, thus playing an important role in inflammation and immune regulation [10,11]. In our previous study, we observed that the PPARgamma agonist significantly alleviated nasal allergy symptoms, reduced levels of interleukin (IL)-4, IL-5, and IL-13 in nasal lavage fluid, and inhibited the infiltration of inflammatory cells in the nasal mucosa [12].
The activation of naïve CD4+ T cells marks the beginning of their differentiation into mature, functionally specialized T helper (TH) cell subsets [13]. To date, several TH-cell subsets have been identified, including TH1, TH2, TH17, and regulatory T cells (Tregs). The balance between type 1 and type 2 immunity is crucial in the pathogenesis of allergic inflammation [14]. In parallel, extensive research has established that Tregs and associated cytokines play a fundamental role in maintaining immune homeostasis in allergic diseases [15,16]. Studies indicate that Tregs in cord blood exhibit impairments in quantity, expression, and function at birth, even before the development of type 1/type 2 immunity imbalances in the offspring of atopic mothers [17]. Furthermore, Tregs interact with TH17 cells to mediate allergic inflammation, as evidenced by their co-regulation in pediatric asthma and rhinitis. Prenatal exposure and genetic susceptibility contribute to early TH17 immune maturation, influencing childhood asthma development [18].
Despite evidence linking PPARgamma to T-cell regulation, two critical questions remain unaddressed: (i) Does myeloid PPARgamma deficiency exacerbate allergic inflammation by altering TH-subset dynamics? (ii) Can PPARgamma activation therapeutically reprogram CD4+ T-cell fate in AR? To investigate this, we first assessed symptoms and immune responses in ovalbumin (OVA)-induced AR using PPARgamma conditional knockout (cKO) mice (PPARgamma+/-f/fLyz2-Cre) versus wild-type controls (PPARgamma-/-f/fLyz2-Cre, wild type [WT]). We then evaluated splenic Treg proportions and nasal eosinophilia, and finally examined how PPARgamma agonism modulates naïve CD4+ T-cell polarization into Tregs, TH1, TH2, and TH17 lineages.

2. Materials and Methods

2.1. Mice

A total of 24 male C57BL/6 mice (approximately 8 weeks old) were utilized: 9 mice for in vivo studies comprising WT controls (n = 3), OVA-sensitized WT (n = 3), and myeloid-specific PPARgamma knockout mice (n = 3), and 15 WT mice for in vitro naïve CD4+ T-cell isolation. PPARgammaf/fLyz2-Cre mice were generated to achieve myeloid-specific PPARgamma deletion by crossbreeding PPARgamma-floxed mice (PPARgammaf/f; Ppargtm2Rev/J) with Lyz2-Cre transgenic mice expressing Cre recombinase under the lysozyme 2 promoter. Both parental strains (C57BL/6 background; Stock No: C001003) were obtained from Cyagen Biosciences (Suzhou, China). The mice were housed under the standard conditions at a temperature of 22–24 °C with 50–60% humidity and a 12 h dark–light cycle. They were provided unrestricted access to OVA-free food and water. The study was approved by the Animal Research Ethics Board of Shanghai East Hospital (No. 2024-002).

2.2. OVA Sensitization, Antigen Challenge

Sensitization and antigen challenge were performed in mice using a modified established protocol [12]. Under pathogen-free conditions, mice in the AR model group were sensitized by the intraperitoneal (i.p.) injection of 150 μg OVA (Sigma-Aldrich, St. Louis, MO, USA) emulsified in 6 mg aluminum hydroxide [Al(OH)3] (Sango Biotech, Shanghai, China) in 0.3 mL sterile saline on days 0, 7, and 14. Control mice received i.p. injections of 0.3 mL sterile saline containing 6 mg Al(OH)3 (without OVA) on identical days. For nasal challenge, mice were administered 20 μL of 40 mg/mL OVA solution (800 μg/dose) via intranasal instillation (i.n.) daily from days 21 to 28. Control mice received 20 μL sterile saline using the same protocol (Figure 1).

2.3. Symptom Assessment

Each mouse was challenged with an intranasal OVA instillation 24 h after the last intranasal saline or OVA challenge. Two observers blinded to experimental group assignments independently recorded the frequencies of sneezing and nasal rubbing events during the 15 min period immediately post-challenge. The final symptom counts represent the consensus value between observers, with discrepancies >15% resolved by video re-evaluation. This blinded protocol ensured the objective quantification of AR inflammatory parameters.

2.4. Measures of Tregs by Flow Cytometry

After OVA sensitization and allergen challenge, mice were deeply anesthetized with isoflurane overdose (5% induction, 3% maintenance) followed by cervical dislocation. Single-cell suspensions of spleens were prepared by passage through nylon mesh cell strainers (BD Biosciences, Franklin Lakes, NJ, USA). Single-cell suspensions from the spleen were prepared, and 1 × 106 cells were used for flow cytometry. The cells were washed twice and resuspended in 100 μL of phosphate-buffered saline (PBS) and then stained with Percp/Cy5.5-conjugated anti-CD4 antibodies (Biolengend, San Diego, CA, USA) and incubated at 4 °C for 30 min. The resuspended cells were fixed and permeabilized using a Foxp3 Fix/Perm Fixation/Permeabilization Kit (Biolengend) under the manufacturer’s instructions. The cells were then stained with FITC-conjugated anti-Foxp3 antibodies (Biolengend) and incubated at 4 °C for 30 min. Flow cytometric analysis was performed using CytoFLEX and CytExpert software v2.4 (Beckman Coulter, Miami, FL, USA). The fluorescence strength was represented as a percentage. FoxP3+ CD4+ T cells were defined as Tregs.

2.5. Nasal Mucosa Histopathology Analysis

Following euthanasia, spleens were immediately harvested for flow cytometric analysis. Systemic perfusion fixation was then conducted via transcardial perfusion: approximately 40 mL of physiological saline followed by 20 mL of 4% paraformaldehyde (PFA) were administered through the left ventricle after puncturing the right auricle. Mouse heads were subsequently collected, trimmed of extraneous tissue, and post-fixed in 10% neutral-buffered formalin at 20 °C for 48 h. Fixed specimens underwent decalcification in 0.5 M EDTA solution for 28 days, with weekly solution changes and periodic tissue trimming to maintain optimal morphology. Following complete decalcification, specimens were paraffin-embedded. Serial 3 μm sections were stained with hematoxylin and eosin (H&E) to assess eosinophilic infiltration. Eosinophil quantification was performed by counting cells per high-power field (HPF) across three representative sections per mouse, with final values expressed as mean eosinophils/HPF.

2.6. Isolation of Naïve CD4+ T Cells and Measure of Treg/Th1/Th2/Th17 Cells by Flow Cytometric Analysis

Single-cell suspensions prepared from the spleens of untreated WT mice (genotype: PPARgamma-/-f/fLyz2-Cre) were subjected to magnetic separation using a Naïve CD4+ T Cell Isolation kit (Miltenyi Biotech, Bergisch Gladbach, Germany; cat# 130-104-453), achieving >90% purity post-sorting; 6 × 106 naïve CD4+ T cells were polarized for 72 h in anti-CD3ε-coated 96-well plates (5 μg/mL overnight) with soluble anti-CD28 (5 μg/mL) in 200 μL complete RPMI-1640 (10% FBS, 1 mM sodium pyruvate, 55 μM β-mercaptoethanol) under TH1 conditions (IL-2 [20 ng/mL] + IL-12 [20 ng/mL] + anti-IL-4 [10 μg/mL]); TH2 condition (IL-2 [20 ng/mL] + IL-4 [100 ng/mL] + anti-IFN-γ [10 μg/mL] + anti-IL-12 [10 μg/mL]); TH17 conditions (IL-2 [20 ng/mL] + IL-6 [100 ng/mL] + TGF-β [5 ng/mL] + anti-IL-4 [10 μg/mL] + anti-IFN-γ [10 μg/mL]), or Treg conditions (IL-2 [20 ng/mL] + TGF-β [5 ng/mL]), with pioglitazone (PIO) added at culture initiation and maintained throughout the 72 h polarization period. Cells were washed twice post-culture, surface-stained with anti-CD4 (BioLegend) at 4 °C for 30 min, fixed/permeabilized (Cytofix/Cytoperm Buffer Set, BD), intracellularly stained (BioLegend), and analyzed using CytoFLEX (CytExpert software v2.4); proportions were calculated as follows: TH1 = IFN-γ+CD4+, TH2 = IL-4+CD4+, TH17 = IL-17+CD4+, Tregs = FoxP3+CD4+.

2.7. Statistical Analysis

All quantitative data are presented as the mean ± standard deviation (SD). Statistical analyses were rigorously selected based on experimental design: one-way analysis of variance (ANOVA) with Tukey’s multiple comparisons test was employed for multi-group comparisons of independent samples, and Paired t-tests were used for two-group comparisons of related samples. Normality and homogeneity of variance assumptions were verified using Shapiro–Wilk and Brown–Forsythe tests, respectively. A probability value of p < 0.05 was considered statistically significant for all analyses, which were performed using GraphPad Prism 9.0 (GraphPad Software, San Diego, CA, USA).

3. Results

3.1. Myeloid PPARgamma Deficiency Exacerbates AR Pathology and Lowers Splenic Treg Proportion

Following OVA-induced AR modeling, the sneezing frequencies (15 min observation) were 6.00 ± 1.00 (WT Control), 37.33 ± 11.72 (WT AR), and 100.33 ± 10.97 (cKO-AR), while the nasal rubbing frequencies were 18.33 ± 6.66 (WT Control), 61.00 ± 17.56 (WT AR), and 83.33 ± 3.51 (cKO-AR). Statistical analysis using one-way ANOVA with Tukey’s post hoc test revealed significant group differences for sneezing (F = 80.31, p < 0.0001) and nasal rubbing (F = 79.38, p < 0.0001), with post hoc comparisons confirming increased sneezing in WT AR versus the control (p = 0.0145), cKO-AR versus the control (p < 0.0001), and cKO-AR versus WT AR (p = 0.0004), while nasal rubbing was elevated in both WT AR and cKO-AR versus the control (p < 0.001), and significantly higher in cKO-AR versus WT AR (p = 0.0126). Histopathological analysis demonstrated a disordered ciliated epithelium and significantly increased eosinophil infiltration in the nasal mucosa of cKO-AR mice (5.50 ± 0.89 eosinophils/HPF) versus WT AR (2.70 ± 0.78; p = 0.0102) and the control (0.63 ± 0.61; p < 0.0006) using ANOVA (F = 30.28, p = 0007) with Turkey’s multiple comparisons test. Flow cytometry confirmed reduced splenic Treg proportions in cKO-AR mice (6.16 ± 0.85%) versus WT AR (9.52 ± 1.47%; p = 0.0335) and the control (14.53 ± 1.23%; p = 0004), with overall significance indicated by ANOVA (F = 36.26, p = 0.0004) (Figure 2).

3.2. PPARgamma Agonist Promotes the Polarization of Naïve CD4+ T Cells into Tregs

Mouse spleen naïve CD4+ T cells were isolated via magnetic separation, achieving >90% purity (Figure 3A), and subsequently stimulated with CD3/CD28 in the presence of PIO (0, 1, 10, 20 μM) or a 0.1% DMSO vehicle. Flow cytometry analysis demonstrated concentration-dependent enhancement of Treg polarization (CD4+Foxp3+ cells), with significant overall concentration effects indicated by one-way ANOVA (F = 14.34, p = 0.0004), where Treg proportions at 1 μM (40.38 ± 6.28%; p = 0.019 vs. 0 μM), 10 μM (50.46 ± 3.04%; p < 0.0001 vs. 0 μM), and 20 μM (42.40 ± 5.94%; p = 0.003 vs. 0 μM) were all significantly elevated versus the 0 μM control (25.72 ± 0.67%) according to Tukey’s post hoc test, confirming maximal efficacy at 10 μM. This optimal effect was validated in expanded replicates, where a Paired t-test confirmed substantially higher Treg proportions at 10 μM versus the vehicle control (40.15 ± 4.13% vs. 28.20 ± 2.85%; t = 5.47, p = 0.0028) (Figure 3B,D).

3.3. PPARgamma Agonist Inhibits the Polarization of Naïve CD4+ T Cells Toward TH1/TH2/TH17 Cells

Naïve CD4+ T lymphocytes isolated from mouse spleens were stimulated with CD3/CD28 and polarized for 72 h under TH1, TH2, and TH17 conditions, with parallel treatment using 10 µM PIO or the DMSO vehicle. Paired t-test analysis revealed significantly reduced proportions of IFN-γ+ CD4+ T cells (TH1) (22.54 ± 2.41% vs. 28.12 ± 0.90%; t = 6.04, p = 0.0264) (Figure 4A,B) and IL-4+ CD4+ T cells (TH2) (2.73 ± 1.27% vs. 5.90 ± 1.33%; t = 14.83, p = 0.0045) (Figure 5A,B) in PIO-treated cultures versus the controls. While IL-17+ CD4+ T cells (TH17) showed no statistical significance (8.59 ± 4.03% vs. 12.16 ± 2.92%; t = 3.18, p = 0.0864), all three biological replicates demonstrated consistent downward trends (mean reduction: 3.57%) (Figure 6A,B).

4. Discussion

PPARgamma, a nuclear receptor superfamily member, has been studied in many inflammatory contexts [19,20]. Recent research shows that PPARgamma can suppress pro-inflammatory genes such as NF—κB [21] and MUC5AC [22,23,24]. PPARgamma agonists may also modulate inflammation by regulating miR expression; for example, PPARgamma upregulates miR—124 to inhibit pro-inflammatory cytokines [25]. Our experimental specifically addressed the functional role of myeloid PPARgamma in OVA-induced allergic inflammation through direct comparison of littermate-controlled PPARgamma cKO vs. WT mice under identical AR induction conditions. This design enables the definitive attribution of observed phenotypic differences—exacerbated symptoms, reduced Tregs, and enhanced eosinophil infiltration—to PPARgamma deficiency within the AR pathological context.
While our previous work demonstrated that systemic PPARgamma agonism enhances Tregs and ameliorates AR in murine models [12], and Park et al. reported that PPARgamma agonists inhibit TH1/TH2/TH17 differentiation with sex-dependent efficacy [26], these pharmacological approaches could not delineate the cellular origin of PPARgamma’s immunomodulatory effects. Our study resolves this gap through genetic dissection. This directly demonstrates that PPARgamma within myeloid lineages orchestrates T-cell polarization independently of systemic effects. Thus, our mechanistic insight advances prior pharmacological observations by identifying myeloid cells as the primary therapeutic target for PPARgamma-based interventions in AR.
We acknowledge that baseline characterization of PPARgamma cKO phenotypes was not included in the current study. However, this absence does not compromise our central finding that myeloid-specific PPARgamma deficiency exacerbates AR pathology, as all pathological and immunological endpoints were rigorously compared against genotype-matched, OVA-sensitized controls (cKO-AR vs. WT AR). Our experimental design specifically interrogated PPARgamma’s role within established allergic inflammation—not homeostatic immunity—making baseline cKO characterization non-essential for addressing the primary research question. Future studies examining steady-state immune profiles in unchallenged cKO mice would provide valuable mechanistic context, though such characterization extends beyond this work’s focus on PPARgamma-directed immunomodulation during active allergic responses.
The TH17-PPARgamma axis critically influences AR pathogenesis, where IL-17A elevation directly correlates with clinical severity in seasonal AR patients [27]. Our data align with PPARgamma’s immunomodulatory role: while PIO significantly suppressed TH1/TH2 polarization, it induced a consistent downward trend in TH17 proportions despite statistical non-significance, reinforcing PPARgamma’s capacity to broadly temper pro-inflammatory T-cell responses. Mechanistically, PPARgamma activation counteracts IL-17A-driven inflammation—amplified by pollutant-induced myeloid epigenetic dysregulation [28] and TLR4/NF-κB signaling [29,30]—while promoting Treg expansion. Therapeutically, PPARgamma agonism may rebalance TH17/Treg dynamics, addressing IL-17A-mediated neutrophil/eosinophil recruitment [31] unmitigated by current biologics.
Although our study did not directly assess epigenetic modifications, the observed effects of PPARgamma agonism on naïve CD4+T-cell polarization—promoting Treg differentiation while suppressing TH1/TH2/TH17 subsets—may involve epigenetic reprogramming. This possibility is supported by evidence that environmental exposure (e.g., air toxics) remodel the nasal mucosal epigenome, dysregulating genes controlling myeloid immune cell function and TGF-β1 signaling, which, in turn, drive aberrant TH cell responses in AR [16,27]. Moreover, multi-omics studies in AR have demonstrated coordinated alterations in DNA methylation and gene expression patterns in leukocytes during disease progression [28]. Future research should integrate transcriptomic and epigenomic profiling to determine whether PPARgamma activation durably recalibrates T-cell homeostasis through mechanisms such as the stable epigenetic silencing of pro-inflammatory pathways or enhancing regulatory circuitries. Such insights could inspire novel interventions targeting metabolic–epigenetic crosstalk in allergic inflammation.
Several limitations should be considered when interpreting our results. Firstly, the modest sample size in animal groups constrains the statistical power of our findings, particularly given the biological variability inherent in immune responses, necessitating expanded cohorts in future work. Secondly, murine AR models may not fully recapitulate human disease heterogeneity; the validation of PPARgamma expression in clinical nasal mucosa specimens and functional testing on human peripheral blood mononuclear cell (PBMC)-derived CD4+ T cells from AR patients are warranted to improve translational relevance. Finally, while IL-17+ cells were quantified, downstream mediators (e.g., CXCL chemokines) and functional consequences—including neutrophil recruitment and epithelial remodeling—remain unexamined and merit investigation in future work.

5. Conclusions

Myeloid PPARgamma deficiency exacerbates AR pathology and reduces splenic Tregs, while PPARgamma agonism enhances Treg polarization and suppresses TH1/TH2/TH17 differentiation. These results establish PPARgamma as a key regulator of allergic inflammation, supporting its therapeutic targeting to rebalance T-cell responses, warranting further preclinical and clinical validation.

Author Contributions

Conceptualization, W.W. and X.R.; methodology, S.R. and R.F.; software, Y.Z. and P.S.; validation, D.L., S.R. and W.W.; formal analysis, X.R.; investigation, X.R.; resources, W.W.; data curation, W.W.; writing—original draft preparation, X.R.; writing—review and editing, W.W.; visualization, X.R.; supervision, W.W.; project administration, W.W.; funding acquisition, W.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Shanghai Pudong New Area Health Commission, grant number PW2023A-46.

Institutional Review Board Statement

The animal study protocol used in this study was approved by the Institutional Review Board of Shanghai East Hospital (No. 2024-002, approved on 25 January 2024).

Informed Consent Statement

Not applicable.

Data Availability Statement

The original contributions presented in this study are included in the article material. Further inquiries can be directed to the corresponding author.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ARAllergic rhinitis
PPARgammaPeroxisome proliferator-activated receptor gamma
WTWild type
THT helper
OVAOvalbumin
PIOPioglitazone
TregsRegulatory T cells
cKOConditional knockout

References

  1. Bousquet, J.; Anto, J.M.; Bachert, C.; Baiardini, I.; Bosnic-Anticevich, S.; Walter Canonica, G.; Melén, E.; Palomares, O.; Scadding, G.K.; Togias, A.; et al. Allergic rhinitis. Nat. Rev. Dis. Primers 2020, 6, 95. [Google Scholar] [CrossRef] [PubMed]
  2. Brożek, J.L.; Bousquet, J.; Agache, I.; Agarwal, A.; Bachert, C.; Bosnic-Anticevich, S.; Brignardello-Petersen, R.; Canonica, G.W.; Casale, T.; Chavannes, N.H.; et al. Allergic Rhinitis and its Impact on Asthma (ARIA) guidelines-2016 revision. J. Allergy Clin. Immunol. 2017, 140, 950–958. [Google Scholar] [CrossRef]
  3. Seidman, M.D.; Gurgel, R.K.; Lin, S.Y.; Schwartz, S.R.; Baroody, F.M.; Bonner, J.R.; Dawson, D.E.; Dykewicz, M.S.; Hackell, J.M.; Han, J.K.; et al. Clinical practice guideline: Allergic rhinitis. Otolaryngol Head Neck Surg. 2015, 152 (Suppl. S1), S1–S43. [Google Scholar] [CrossRef]
  4. Bousquet, J.; Schünemann, H.J.; Togias, A.; Bachert, C.; Erhola, M.; Hellings, P.W.; Klimek, L.; Pfaar, O.; Wallace, D.; Ansotegui, I.; et al. Next-generation Allergic Rhinitis and Its Impact on Asthma (ARIA) guidelines for allergic rhinitis based on Grading of Recommendations Assessment, Development and Evaluation (GRADE) and real-world evidence. J. Allergy Clin. Immunol. 2020, 145, 70–80.e3. [Google Scholar] [CrossRef]
  5. Vella, V.; Nicolosi, M.L.; Giuliano, S.; Bellomo, M.; Belfiore, A.; Malaguarnera, R. PPAR-γ Agonists as Antineoplastic Agents in Cancers with Dysregulated IGF Axis. Front. Endocrinol. 2017, 8, 31. [Google Scholar] [CrossRef] [PubMed]
  6. Montaigne, D.; Butruille, L.; Staels, B. PPAR control of metabolism and cardiovascular functions. Nat. Rev. Cardiol. 2021, 18, 809–823. [Google Scholar] [CrossRef] [PubMed]
  7. Stark, J.M.; Coquet, J.M.; Tibbitt, C.A. The Role of PPAR-γ in Allergic Disease. Curr. Allergy Asthma Rep. 2021, 21, 45. [Google Scholar] [CrossRef]
  8. Xiong, Z.; Chan, S.L.; Zhou, J.; Vong, J.S.L.; Kwong, T.T.; Zeng, X.; Wu, H.; Cao, J.; Tu, Y.; Feng, Y.; et al. Targeting PPAR-gamma counteracts tumour adaptation to immune-checkpoint blockade in hepatocellular carcinoma. Gut 2023, 72, 1758–1773. [Google Scholar] [CrossRef]
  9. Scheen, A.J.; Esser, N.; Paquot, N. Antidiabetic agents: Potential anti-inflammatory activity beyond glucose control. Diabetes Metab. 2015, 41, 183–194. [Google Scholar] [CrossRef]
  10. Nagata, K.; Kasakura, K.; Miura, R.; Yashiro, T.; Nishiyama, C. Suppressive role of PPARγ in the IgE-dependent activation of mast cells. Int. Immunol. 2020, 32, 143–150. [Google Scholar] [CrossRef]
  11. Liu, Y.; Wang, J.; Luo, S.; Zhan, Y.; Lu, Q. The roles of PPARγ and its agonists in autoimmune diseases: A comprehensive review. J. Autoimmun. 2020, 113, 102510. [Google Scholar] [CrossRef] [PubMed]
  12. Wang, W.; Zhu, Z.; Zhu, B.; Ma, Z. Peroxisome proliferator-activated receptor-gamma agonist induces regulatory T cells in a murine model of allergic rhinitis. Otolaryngol. Head Neck Surg. 2011, 144, 506–513. [Google Scholar] [CrossRef]
  13. Wang, W.; Sung, N.; Gilman-Sachs, A.; Kwak-Kim, J. T Helper (Th) Cell Profiles in Pregnancy and Recurrent Pregnancy Losses: Th1/Th2/Th9/Th17/Th22/Tfh Cells. Front. Immunol. 2020, 11, 2025. [Google Scholar] [CrossRef] [PubMed]
  14. Shamji, M.H.; Sharif, H.; Layhadi, J.A.; Zhu, R.; Kishore, U.; Renz, H. Diverse immune mechanisms of allergen immunotherapy for allergic rhinitis with and without asthma. J. Allergy Clin. Immunol. 2022, 149, 791–801. [Google Scholar] [CrossRef]
  15. Yang, C.H.; Tian, J.J.; Ko, W.S.; Shih, C.J.; Chiou, Y.L. Oligo-fucoidan improved unbalance the Th1/Th2 and Treg/Th17 ratios in asthmatic patients: An ex vivo study. Exp. Ther. Med. 2019, 17, 3–10. [Google Scholar] [CrossRef]
  16. Potaczek, D.P.; Harb, H.; Michel, S.; Alhamwe, B.A.; Renz, H.; Tost, J. Epigenetics and allergy: From basic mechanisms to clinical applications. Epigenomics 2017, 9, 539–571. [Google Scholar] [CrossRef] [PubMed]
  17. Ponsonby, A.L.; Collier, F.; O’Hely, M.; Tang, M.L.K.; Ranganathan, S.; Gray, L.; Morwitch, E.; Saffery, R.; Burgner, D.; Dwyer, T.; et al. Household size, T regulatory cell development, and early allergic disease: A birth cohort study. Pediatr Allergy Immunol. 2022, 33, e13810. [Google Scholar] [CrossRef]
  18. Lluis, A.; Ballenberger, N.; Illi, S.; Schieck, M.; Kabesch, M.; Illig, T.; Schleich, I.; von Mutius, E.; Schaub, B. Regulation of TH17 markers early in life through maternal farm exposure. J. Allergy Clin. Immunol. 2014, 133, 864–871. [Google Scholar] [CrossRef]
  19. Yang, Y.; Wei, C.; Liu, J.; Ma, D.; Xiong, C.; Lin, D.; Wu, A. Atorvastatin protects against postoperative neurocognitive disorder via a peroxisome proliferator-activated receptor-gamma signaling pathway in mice. J. Int. Med. Res. 2020, 48, 300060520924251. [Google Scholar] [CrossRef]
  20. Schulte, R.; Wohlleber, D.; Unrau, L.; Geers, B.; Metzger, C.; Erhardt, A.; Tiegs, G.; van Rooijen, N.; Heukamp, L.C.; Klotz, L.; et al. Pioglitazone-Mediated Peroxisome Proliferator-Activated Receptor γ Activation Aggravates Murine Immune-Mediated Hepatitis. Int. J. Mol. Sci. 2020, 21, 2523. [Google Scholar] [CrossRef]
  21. Korbecki, J.; Bobiński, R.; Dutka, M. Self-regulation of the inflammatory response by peroxisome proliferator-activated receptors. Inflamm. Res. 2019, 68, 443–458. [Google Scholar] [CrossRef] [PubMed]
  22. Lakshmi, S.P.; Reddy, A.T.; Banno, A.; Reddy, R.C. Airway Epithelial Cell Peroxisome Proliferator-Activated Receptor γ Regulates Inflammation and Mucin Expression in Allergic Airway Disease. J. Immunol. 2018, 201, 1775–1783. [Google Scholar] [CrossRef] [PubMed]
  23. Lv, J.; Su, W.; Yu, Q.; Zhang, M.; Di, C.; Lin, X.; Wu, M.; Xia, Z. Heme oxygenase-1 protects airway epithelium against apoptosis by targeting the proinflammatory NLRP3-RXR axis in asthma. J. Biol. Chem. 2018, 293, 18454–18465. [Google Scholar] [CrossRef]
  24. Yang, P.; Chen, S.; Zhong, G.; Kong, W.; Wang, Y. Agonist of PPAR-γ Reduced Epithelial-Mesenchymal Transition in Eosinophilic Chronic Rhinosinusitis with Nasal Polyps via Inhibition of High Mobility Group Box1. Int. J. Med. Sci. 2019, 16, 1631–1641. [Google Scholar] [CrossRef]
  25. Wang, D.; Shi, L.; Xin, W.; Xu, J.; Xu, J.; Li, Q.; Xu, Z.; Wang, J.; Wang, G.; Yao, W.; et al. Activation of PPARγ inhibits pro-inflammatory cytokines production by upregulation of miR-124 in vitro and in vivo. Biochem. Biophys. Res. Commun. 2017, 486, 726–731. [Google Scholar] [CrossRef]
  26. Park, H.J.; Park, H.S.; Lee, J.U.; Bothwell, A.L.; Choi, J.M. Sex-Based Selectivity of PPARγ Regulation in Th1, Th2, and Th17 Differentiation. Int. J. Mol. Sci. 2016, 17, 1347. [Google Scholar] [CrossRef]
  27. Zhao, Y.; Mei, S.; Yao, S.; Cai, S.; Zhang, P.; Lou, H.; Zhang, L. IL-17-Related Pathways and Myeloid Cell Function are Involved in the Mechanism of Sublingual Immunotherapy with Artemisia annua for Seasonal Allergic Rhinitis. Clin. Rev. Allergy Immunol. 2025, 68, 53. [Google Scholar] [CrossRef] [PubMed]
  28. Irizar, H.; Chun, Y.; Hsu, H.L.; Li, Y.C.; Zhang, L.; Arditi, Z.; Grishina, G.; Grishin, A.; Vicencio, A.; Pandey, G.; et al. Multi-omic integration reveals alterations in nasal mucosal biology that mediate air pollutant effects on allergic rhinitis. Allergy 2024, 79, 3047–3061. [Google Scholar] [CrossRef]
  29. Yang, Q.; Sun, Y.; An, J.; Wang, L.; Zhang, C.; Fei, Y.; Wu, K.; Zhai, X.; Wu, Y. Xiaoqinglong Decoction Attenuates Inflammatory Response and Mitochondrial Injury by Regulating the MyD88/NF-κB Pathway Dependent of TLR4 in Allergic Rhinitis. Int. Arch. Allergy Immunol. 2025, in press. [CrossRef]
  30. Zhang, J.; Gao, L.; Yu, D.; Song, Y.; Zhao, Y.; Feng, Y. Three Artemisia pollens trigger the onset of allergic rhinitis via TLR4/MyD88 signaling pathway. Mol. Biol. Rep. 2024, 51, 319. [Google Scholar] [CrossRef]
  31. Deng, X.H.; Huang, L.X.; Sun, Q.; Li, C.G.; Xie, Y.C.; Liu, X.Q.; Fu, Q.L. Increased circulating LOX-1+ neutrophils activate T cells and demonstrate a pro-inflammatory role in allergic rhinitis. Heliyon 2024, 10, e36218. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic overview of in vivo experimental timeline for OVA-induced AR in myeloid-specific PPARgamma-deficient mice. Abbreviations: OVA, ovalbumin; i.p., intraperitoneal; i.n., intranasal; AR, allergic rhinitis; cKO, conditional knockout.
Figure 1. Schematic overview of in vivo experimental timeline for OVA-induced AR in myeloid-specific PPARgamma-deficient mice. Abbreviations: OVA, ovalbumin; i.p., intraperitoneal; i.n., intranasal; AR, allergic rhinitis; cKO, conditional knockout.
Biomedicines 13 01616 g001
Figure 2. Myeloid PPARgamma deletion aggravates allergic responses in an OVA-induced AR model (n = 3 per group). (A,B) Allergic symptom frequencies in saline-treated controls, OVA-sensitized AR mice, and myeloid-specific PPARgamma cKO mice with OVA-induced AR (cKO-AR): (A) sneezing episodes; (B) nasal rubbing episodes. (C,D) Histopathological evaluation of nasal mucosa: (C) representative H&E-stained sections (100× and 400×; black arrows indicate eosinophils); (D) eosinophil quantification. Data represent mean eosinophil counts per mouse across three high-power fields (HPF, 400× magnification). (E,F) Splenic Treg analysis: (E) representative flow cytometry density plots (color gradient: red = high cell density, blue = low density) of Tregs (CD4+Foxp3+); (F) quantitative Treg proportion. Data represent mean ± SD. Statistical analysis: one-way ANOVA with Tukey’s post hoc test; * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001. Abbreviations: AR, allergic rhinitis; WT, wild type; OVA, ovalbumin; cKO, conditional knockout; H&E, hematoxylin and eosin; HPF, high-power field; SD, standard deviation.
Figure 2. Myeloid PPARgamma deletion aggravates allergic responses in an OVA-induced AR model (n = 3 per group). (A,B) Allergic symptom frequencies in saline-treated controls, OVA-sensitized AR mice, and myeloid-specific PPARgamma cKO mice with OVA-induced AR (cKO-AR): (A) sneezing episodes; (B) nasal rubbing episodes. (C,D) Histopathological evaluation of nasal mucosa: (C) representative H&E-stained sections (100× and 400×; black arrows indicate eosinophils); (D) eosinophil quantification. Data represent mean eosinophil counts per mouse across three high-power fields (HPF, 400× magnification). (E,F) Splenic Treg analysis: (E) representative flow cytometry density plots (color gradient: red = high cell density, blue = low density) of Tregs (CD4+Foxp3+); (F) quantitative Treg proportion. Data represent mean ± SD. Statistical analysis: one-way ANOVA with Tukey’s post hoc test; * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001. Abbreviations: AR, allergic rhinitis; WT, wild type; OVA, ovalbumin; cKO, conditional knockout; H&E, hematoxylin and eosin; HPF, high-power field; SD, standard deviation.
Biomedicines 13 01616 g002
Figure 3. PPARgamma agonist enhances naïve CD4+ T-cell polarization toward Tregs. (A) Purity assessment of sorted naïve CD4+ T cells from mouse spleen. Representative flow cytometry density plots (color gradient: red = high cell density, blue = low density) show pre-sort (left) and post-sort (right) populations, with >90% purity achieved after magnetic-activated cell sorting (MACS). (B) Representative flow cytometry density plots of Treg polarization (CD4+Foxp3+ cells) under increasing PIO concentrations (0, 1, 10, 20 μM). (C) Concentration–response relationship quantifying Treg proportions from (B) (n = 3). (D) Validation of optimal concentration (10μM PIO) with expanded replication (n = 6). Data represent mean ± SD. Statistical analysis: one-way ANOVA with Tukey’s post hoc test (C) and Paired t-tests (D); * p < 0.05, ** p < 0.01, *** p < 0.001. Abbreviations: Tregs, regulatory T cells; PIO, pioglitazone; SD, standard deviation.
Figure 3. PPARgamma agonist enhances naïve CD4+ T-cell polarization toward Tregs. (A) Purity assessment of sorted naïve CD4+ T cells from mouse spleen. Representative flow cytometry density plots (color gradient: red = high cell density, blue = low density) show pre-sort (left) and post-sort (right) populations, with >90% purity achieved after magnetic-activated cell sorting (MACS). (B) Representative flow cytometry density plots of Treg polarization (CD4+Foxp3+ cells) under increasing PIO concentrations (0, 1, 10, 20 μM). (C) Concentration–response relationship quantifying Treg proportions from (B) (n = 3). (D) Validation of optimal concentration (10μM PIO) with expanded replication (n = 6). Data represent mean ± SD. Statistical analysis: one-way ANOVA with Tukey’s post hoc test (C) and Paired t-tests (D); * p < 0.05, ** p < 0.01, *** p < 0.001. Abbreviations: Tregs, regulatory T cells; PIO, pioglitazone; SD, standard deviation.
Biomedicines 13 01616 g003
Figure 4.  PPARgamma agonist suppresses TH1 polarization of naïve CD4+ T cells. (A) Representative flow cytometry density plots (color gradient: red = high cell density, blue = low density) of TH1 (CD4+IFN-γ+ cells) from naïve CD4+ T cells isolated from WT mice (PPARgamma-/-f/fLyz2-Cre), cultured under TH1-polarizing conditions with PIO (10 μM) or DMSO vehicle control. (B) Quantitative analysis of TH1 proportions. Data represent mean ± SD. Statistical analysis: Paired t-test; * p < 0.05. Abbreviations: TH, T helper; PIO, pioglitazone; WT, wild type; SD, standard deviation.
Figure 4.  PPARgamma agonist suppresses TH1 polarization of naïve CD4+ T cells. (A) Representative flow cytometry density plots (color gradient: red = high cell density, blue = low density) of TH1 (CD4+IFN-γ+ cells) from naïve CD4+ T cells isolated from WT mice (PPARgamma-/-f/fLyz2-Cre), cultured under TH1-polarizing conditions with PIO (10 μM) or DMSO vehicle control. (B) Quantitative analysis of TH1 proportions. Data represent mean ± SD. Statistical analysis: Paired t-test; * p < 0.05. Abbreviations: TH, T helper; PIO, pioglitazone; WT, wild type; SD, standard deviation.
Biomedicines 13 01616 g004
Figure 5. PPARgamma agonist suppresses TH2 polarization of naïve CD4+ T cells. (A) Representative flow cytometry density plots (color gradient: red = high cell density, blue = low density) of TH2 (CD4+IL-4+ cells) from naïve CD4+ T cells isolated from WT mice (PPARgamma-/-f/fLyz2-Cre), cultured under TH2-polarizing conditions with PIO (10 μM) or DMSO vehicle control. (B) Quantitative analysis of TH2 proportions. Data represent mean ± SD. Statistical analysis: Paired t-test; ** p < 0.01. Abbreviations: TH, T helper; PIO, pioglitazone; WT, wild type; SD, standard deviation.
Figure 5. PPARgamma agonist suppresses TH2 polarization of naïve CD4+ T cells. (A) Representative flow cytometry density plots (color gradient: red = high cell density, blue = low density) of TH2 (CD4+IL-4+ cells) from naïve CD4+ T cells isolated from WT mice (PPARgamma-/-f/fLyz2-Cre), cultured under TH2-polarizing conditions with PIO (10 μM) or DMSO vehicle control. (B) Quantitative analysis of TH2 proportions. Data represent mean ± SD. Statistical analysis: Paired t-test; ** p < 0.01. Abbreviations: TH, T helper; PIO, pioglitazone; WT, wild type; SD, standard deviation.
Biomedicines 13 01616 g005
Figure 6. PPARgamma agonist suppresses TH17 polarization of naïve CD4+ T cells. (A) Representative flow cytometry density plots (color gradient: red = high cell density, blue = low density) of TH17 (CD4+IL-17+ cells) from naïve CD4+ T cells isolated from WT mice (PPARgamma-/-f/fLyz2-Cre), cultured under TH17-polarizing conditions with PIO (10 μM) or DMSO vehicle control. (B) Quantitative analysis of TH17 proportions. Data represent mean ± SD. Statistical analysis: Paired t-test; ns: not significant. Abbreviations: TH, T helper; PIO, pioglitazone; WT, wild type; SD, standard deviation.
Figure 6. PPARgamma agonist suppresses TH17 polarization of naïve CD4+ T cells. (A) Representative flow cytometry density plots (color gradient: red = high cell density, blue = low density) of TH17 (CD4+IL-17+ cells) from naïve CD4+ T cells isolated from WT mice (PPARgamma-/-f/fLyz2-Cre), cultured under TH17-polarizing conditions with PIO (10 μM) or DMSO vehicle control. (B) Quantitative analysis of TH17 proportions. Data represent mean ± SD. Statistical analysis: Paired t-test; ns: not significant. Abbreviations: TH, T helper; PIO, pioglitazone; WT, wild type; SD, standard deviation.
Biomedicines 13 01616 g006
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

Rui, X.; Ruan, S.; Zhang, Y.; Fu, R.; Sun, P.; Lamu, D.; Wang, W. PPARgamma Modulates CD4+ T-Cell Differentiation and Allergic Inflammation in Allergic Rhinitis: A Potential Therapeutic Target. Biomedicines 2025, 13, 1616. https://doi.org/10.3390/biomedicines13071616

AMA Style

Rui X, Ruan S, Zhang Y, Fu R, Sun P, Lamu D, Wang W. PPARgamma Modulates CD4+ T-Cell Differentiation and Allergic Inflammation in Allergic Rhinitis: A Potential Therapeutic Target. Biomedicines. 2025; 13(7):1616. https://doi.org/10.3390/biomedicines13071616

Chicago/Turabian Style

Rui, Xiaoqing, Suyu Ruan, Yu Zhang, Ranran Fu, Pengfei Sun, Danzeng Lamu, and Weihua Wang. 2025. "PPARgamma Modulates CD4+ T-Cell Differentiation and Allergic Inflammation in Allergic Rhinitis: A Potential Therapeutic Target" Biomedicines 13, no. 7: 1616. https://doi.org/10.3390/biomedicines13071616

APA Style

Rui, X., Ruan, S., Zhang, Y., Fu, R., Sun, P., Lamu, D., & Wang, W. (2025). PPARgamma Modulates CD4+ T-Cell Differentiation and Allergic Inflammation in Allergic Rhinitis: A Potential Therapeutic Target. Biomedicines, 13(7), 1616. https://doi.org/10.3390/biomedicines13071616

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