Authors:
Martina Palatella Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany

Search for other papers by Martina Palatella in
Current site
Google Scholar
PubMed
Close
,
Friederike Kruse Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany

Search for other papers by Friederike Kruse in
Current site
Google Scholar
PubMed
Close
,
Silke Glage Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany

Search for other papers by Silke Glage in
Current site
Google Scholar
PubMed
Close
,
André Bleich Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany

Search for other papers by André Bleich in
Current site
Google Scholar
PubMed
Close
,
Marina Greweling-Pils Mouse Pathology Platform, Helmholtz Centre for Infection Research, Braunschweig, Germany

Search for other papers by Marina Greweling-Pils in
Current site
Google Scholar
PubMed
Close
, and
Jochen Huehn Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
Cluster of Excellence RESIST (EXC2155), Hannover Medical School, Hannover, Germany

Search for other papers by Jochen Huehn in
Current site
Google Scholar
PubMed
Close
https://orcid.org/0000-0001-8071-1379
Open access

Abstract

Epigenetic modifications are critical for the regulation of CD4+ T cell differentiation and function. Previously, we identified Acyl-CoA Synthetase Bubble Gum 1 (Acsbg1), a gene involved in fatty acid metabolism, as part of an epigenetic signature that was selectively demethylated in ex vivo isolated T helper 17 (TH17) cells. However, its functional relevance for CD4+ T cells remains incompletely understood. Here, we used in vitro differentiation assays and the adoptive transfer colitis model to investigate the role of Acsbg1 in the differentiation and function of TH1, TH17, and regulatory T (Treg) cells. In vitro, Acsbg1 was expressed in both TH17 and in vitro-induced Treg (iTreg) cells, whereas TH1 cells lacked Acsbg1 expression. Accordingly, Acsbg1 deficiency resulted in impaired TH17 and iTreg differentiation, whereas TH1 differentiation was unaffected. In vivo, upon adoptive transfer of Acsbg1⁻/⁻ Tnaïve cells, immunodeficient recipient mice exhibited an exacerbated colitis, characterized by an altered balance of TH17 and Treg cells, indicating that Acsbg1 expression is essential for optimal TH17 and Treg cell differentiation and function. Our findings highlight the importance of fatty acid (FA) metabolism in maintaining immune homeostasis by regulating T cell differentiation and provide novel insights into the metabolic targeting of inflammatory diseases.

Abstract

Epigenetic modifications are critical for the regulation of CD4+ T cell differentiation and function. Previously, we identified Acyl-CoA Synthetase Bubble Gum 1 (Acsbg1), a gene involved in fatty acid metabolism, as part of an epigenetic signature that was selectively demethylated in ex vivo isolated T helper 17 (TH17) cells. However, its functional relevance for CD4+ T cells remains incompletely understood. Here, we used in vitro differentiation assays and the adoptive transfer colitis model to investigate the role of Acsbg1 in the differentiation and function of TH1, TH17, and regulatory T (Treg) cells. In vitro, Acsbg1 was expressed in both TH17 and in vitro-induced Treg (iTreg) cells, whereas TH1 cells lacked Acsbg1 expression. Accordingly, Acsbg1 deficiency resulted in impaired TH17 and iTreg differentiation, whereas TH1 differentiation was unaffected. In vivo, upon adoptive transfer of Acsbg1⁻/⁻ Tnaïve cells, immunodeficient recipient mice exhibited an exacerbated colitis, characterized by an altered balance of TH17 and Treg cells, indicating that Acsbg1 expression is essential for optimal TH17 and Treg cell differentiation and function. Our findings highlight the importance of fatty acid (FA) metabolism in maintaining immune homeostasis by regulating T cell differentiation and provide novel insights into the metabolic targeting of inflammatory diseases.

Introduction

CD4+ T cells play a central role in orchestrating immune responses. Among these, T helper 17 (TH17) cells, which are characterized by the expression of RORγt and the production of interleukin-17 (IL-17) and IL-22, play a critical role in the protection of mucosal barriers against pathogens [1–3]. TH17 cells can be classified as non-pathogenic (npTH17) or pathogenic TH17 (pTH17) cells [4]. npTH17 cells play a role in maintaining tissue homeostasis, while pTH17 cells are key drivers of tissue inflammation [4]. In contrast, regulatory T (Treg) cells, defined by the expression of Foxp3 [5, 6], are essential for suppressing excessive inflammation and maintaining immune tolerance [7]. The immune system maintains a delicate balance between pro-inflammatory and regulatory mechanisms to ensure immune and tissue homeostasis, while simultaneously facilitating effective pathogen clearance [8, 9]. Dysregulation of TH17 and Treg cells can lead to the development of inflammatory disorders, the onset of autoimmunity, or impaired immune responses to infection [10–12]. This highlights the need for a comprehensive understanding of the molecular mechanisms that regulate their differentiation and function.

Metabolic reprogramming has been identified as a critical factor influencing T cell activation and differentiation [13–16]. Tnaïve cells rely on oxidative phosphorylation (OXPHOS) to generate energy through the metabolism of glucose, lipids, and amino acids [17]. Upon activation, CD4+ T cells undergo a metabolic shift, shifting from OXPHOS to aerobic glycolysis and glutamine oxidation. This shift results in a more anabolic state, whereas memory T cells revert to a catabolic state, similar to that observed in Tnaïve cells [17]. Notably, TH17 and Treg cells have different metabolic requirements during their differentiation [18]. TH17 cells rely on glycolysis and de novo fatty acid synthesis (FAS), with Acyl-Coenzyme A Carboxylase 1 (ACC1) playing a central role [19]. Increasing ACC1 expression has been shown to facilitate the polarization of TH17 cells, whereas its inhibition has been shown to disrupt de novo FAS and glucose metabolism, thereby shifting the T cell fate from TH17 to Treg cells. This process can be reversed by exogenous long-chain fatty acids (FAs), highlighting the metabolic plasticity of T cells [19]. In contrast, Treg cells have a reduced reliance on glycolysis and glutaminolysis and instead primarily use exogenous FAs and mitochondrial metabolism, including OXPHOS and FAs oxidation (FAO), for energy production [17, 20, 21].

Acyl-CoA synthetases (ACS) are enzymes that activate FAs by forming a thioester bond with CoA [22], thereby enabling their participation in anabolic or catabolic pathways. The Acyl-CoA Synthetase Bubble Gum (Acsbg) family is one of the five subfamilies of ACS enzymes [22]. It includes Acsbg1, a highly conserved enzyme that activates long- and very long-chain FAs [23]. Acsbg1 is predominantly expressed in the brain [24], adrenal gland, gonads, and spleen [25]. It is thought to channel FAs, particularly palmitic acid (C16:0), towards β-oxidation [25], thereby maintaining lipid homeostasis and energy metabolism. This suggests a potential regulatory role in immune cells.

Previously, we identified Acsbg1 as part of an epigenetic signature that was selectively demethylated in ex vivo isolated TH17 cells compared to other ex vivo isolated or in vitro differentiated CD4+ T cell subsets. This finding suggests a potential role for Acsbg1 in TH17 biology [26, 27]. In a recent study, Kanno et al. demonstrated that Acsbg1 is selectively expressed in Treg cells, where it acts as a metabolic checkpoint by maintaining mitochondrial fitness [28]. They also showed that Acsbg1 is essential for the resolution of lung inflammation in mice, through the activation of lung-resident ST2+ Treg cells [28]. However, the functional relevance of Acsbg1 in T cells and its role in immune regulation remain incompletely understood.

The present study elucidates the role of Acsbg1 in CD4+ T cell differentiation and immune homeostasis using in vitro and in vivo approaches. In vitro polarization assays were performed to investigate the impact of Acsbg1 deficiency on TH1, TH17, and Treg differentiation and function. To extend these findings to the in vivo setting, we used the adoptive transfer colitis model to assess the role of Acsbg1 in T cell-driven intestinal inflammation. Our results show that Acsbg1 plays a critical role in the differentiation of TH17 and Treg cells, but not in the differentiation of TH1 cells. Furthermore, the absence of Acsbg1 exacerbates colitis by skewing the immune response towards increased pathogenicity. These findings elucidate the interplay between lipid metabolism and immune regulation, with potential implications for the understanding and treatment of inflammatory diseases.

Material and methods

Mice

Full body Acsbg1 knock out mice (B6-Acsbg1tm1a(Eu-comm)Hmgu mice) were generated from embryonic stem cells harboring the tm1a construct within the Acsbg1 locus, obtained from Eucomm [29, 30]. All mice used in this study (B6-Acsbg1tm1a(Eu-comm)Hmgu, B6; hybrid-Rag2tm1Cgn (Rag2−/−)) were bred and maintained at the Helmholtz Centre for Infection Research (Braunschweig, Germany), which provides state-of-the-art laboratory animal care and service. All mice were housed under specific opportunistic and pathogen-free conditions (SOPF) in isolated, ventilated cages, and handled by personnel appropriately trained as well as dedicated animal care staff to assure the highest possible hygienic standards and animal welfare in compliance with German and European animal welfare guidelines. For all experiments, heterozygous Acsbg1+/− mice were bred to obtain Acsbg1−/− mice and Acsbg1+/+ littermate controls. Before experimental use, all mice were genotyped by subjecting genomic DNA to PCR testing for the integrity of intron 2 (F: 5′-GTCTTTGCACCCAGGCTC; R: 5′-GCTTCCATGCTTGCTGCTAC) and for the presence of the ‘tm1a’ construct (F: 5′- GTCTTTGCACCCAGGCTC; R: 5′-GTGGGAAAGGGTTCGAAGTT).

Cell isolation from organs

To prepare single-cell suspensions from lymphoid organs, mice were sacrificed by CO2 asphyxiation. The spleens were isolated and mechanically disrupted using a plunger and phosphate-buffered saline (PBS, Gibco, Schwerte, Germany) containing 0.2% bovine serum albumin (BSA, Merck, Darmstadt, Germany). Cell suspensions were filtered through a nylon mesh with a pore size of 100 μm. Subsequently, the cell suspensions were washed and splenocytes underwent erythrocyte lysis (0.01 M potassium bicarbonate, 0.155 M ammonium chloride, and 0.1 mM ethylenediaminetetraacetic acid (EDTA), all from Sigma-Aldrich, Hamburg, Germany, at pH 7.5). Following an additional washing step, cell suspensions were filtered through a 30 μm nylon mesh. For the preparation of colonic single-cell suspensions, colons were opened longitudinally and washed with pre-digestion medium (PBS, containing 0.2% BSA and 5 mM EDTA) to remove feces. Subsequently, colons were incubated with pre-digestion medium at 37 °C three times for 15 min with a magnetic stirrer (250–300 rpm). After washing with PBS, pre-digested colons were cut into 1–2 mm pieces and digested in Hanks' balanced salt solution (HBSS) (with Mg2+ and Ca2+; Gibco) containing 1 mg mL−1 Collagenase D (Roche, Penzberg, Germany), 100 μg mL−1 DNaseI (Roche), and 25 mM 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) (Gibco) at 37 °C for 1 h with a magnetic stirrer. Subsequently, cells were subjected to 40%/80% Percoll (GE Healthcare, Braunschweig, Germany) gradient centrifugation (780 g, room temperature, 20 min, acceleration and brake off). Cells from the interphase layer were collected, washed, and resuspended in PBS supplemented with 0.2% BSA.

Antibodies, flow cytometry and cell sorting

Flow cytometric analysis was performed as described recently [31]. Viability staining was carried out in PBS using the LIVE/DEAD™ Fixable Blue Dead Cell Stain Kit (Invitrogen, Langenselbold, Germany) in accordance with the manufacturer's instructions. Surface staining was performed for 15 min on ice in PBS containing 0.2% BSA. Unconjugated anti-FcRγIII/II antibody (BioXcell, Eching, Germany; final concentration 10 μg mL−1) was included in the staining mix to block Fc receptors. Intracellular staining was performed using the Foxp3/Transcription Factor Staining Buffer Set (eBioscience, Langenselbold, Germany) following the manufacturer's instructions. To prevent nonspecific antibody binding, 40 μg mL−1 of rat IgG (Dianova, Hamburg, Germany) was used as a blocking agent. For intracellular cytokine staining, cells were stimulated with phorbol 12-myristate 13-acetate (PMA, 10 ng mL−1), Ionomycin (0.5 μg mL−1), and Brefeldin A (10 μg mL−1) for 4 h at 37 °C (all from Sigma-Aldrich). After staining, cells were washed, resuspended in PBS supplemented with 0.2% BSA, and analyzed using a FACSymphony A5 SE flow cytometer (BD Biosciences, Heidelberg, Germany), with data processed using FlowJo software (FlowJo, Ashland, Oregon, USA). Living cell count was quantified on a MACSQuant Analyzer 10 Flow Cytometer (Miltenyi Biotec, Bergisch Gladbach, Germany).

For fluorescence-activated cell sorting (FACS), single-cell suspensions from the indicated organs were stained with specific antibodies, and Lin cells of interest (unless otherwise specified – Lin: CD8α, CD45R, CD19, CD11c, F4/80) were sorted using a BD FACSAria-II SORP, BD FACSAria-Fusion, or Symphony S6 SE (all from BD Biosciences).

In this study, cells were stained with the following fluorochrome-conjugated antibodies purchased from either BD Horizon (Heidelberg, Germany), BioLegend (Eching, Germany), or eBioscience: CD3ε (145-2C11), CD4 (GK1.5), CD8α (53-6.7), CD11c (N418), CD19 (6D5), CD25 (PC61), CD27 (LG.3A10), CD44 (IM7), CD45R (B220) (RA3-6B2), CD62L (MEL-14), CCR6 (140706), IL-10 (JES5-16E3), IL-17A (eBio17B7), IL-22 (Poly5164), IFN-γ (XMG1.2), F4/80 (BM8), Foxp3 (FJK-16S), NK1.1 (PK136), RORγt (Q31-378), T-bet (4B10), and ST2 (RMST2-2).

In vitro TH cell differentiation assay

Single-cell suspensions were prepared from spleens of Acsbg1−/− mice or Acsbg1+/+ littermate controls as described above. Subsequently, FACS-sorted Tnaïve cells (LinCD4+CD25CD62Lhigh cells) were cultured in Iscove's Modified Dulbecco's Medium (IMDM) containing 10% fetal calf serum, 1 mM sodium pyruvate, 50 U mL−1 penicillin and streptomycin, 25 mM HEPES, 50 μM β-mercaptoethanol, and nonessential amino acids (Thermo Fisher Scientific, Langenselbold, Germany; and Biochrom AG, Berlin, Germany), and 1 × 105 cells per well were seeded into 96-well round-bottom plates together with 1 × 105 Dynabeads Mouse T-Activator CD3/CD28 (Thermo Fisher Scientific). For polarization into different TH cell subsets, the following cytokines and antibodies were added: TH0 – non-polarizing control; in vitro-induced Treg (iTreg) – human transforming growth factor β1 (hTGF-β1) (5 ng mL−1); TH17 – IL-6 (30 ng mL−1), hTGF-β1 (2 ng mL−1), IL-1β (10 ng mL−1), anti-IL-2 (5 μg mL−1, JES6-1A12), anti-interferon-γ (IFN-γ) (10 μg mL−1, XMG1.2); TH1 – IL-12 (20 ng mL−1), anti-IL-4 (10 ng mL−1, 11B11) (all purchased from BioLegend and BioXCell, Eching, Germany; Peprotech, Hamburg, Germany; or R&D, Wiesbaden-Nordenstadt, Germany). In each experiment, technical triplicates for each sample and condition were prepared. After three days of cultivation, cells were split 1:1 and supplemented with: TH0 and TH1 – IL-2 (5 ng mL−1, R&D); iTreg – IL-2 (10 ng mL−1); TH17 – IL-23 (5 ng mL−1, BioLegend). On day 4, cells were analyzed via flow cytometry as described above or harvested for RNA isolation and subsequent assessment of Acsbg1 expression.

Real time quantitative PCR

Acsbg1 expression was assessed in in vitro differentiated TH cell subsets. RNA was isolated from cultured T cells using the RNeasy Plus Mini or Micro Kit (Qiagen, Hilden, Germany). Complementary DNA (cDNA) was generated from equivalent amounts of RNA using Transcriptor First Strand cDNA Synthesis Kit (Roche). For quantification, samples were amplified by real-time quantitative polymerase chain reaction (RT-qPCR) using SYBR Green (Roche). RT-qPCR was performed on a LightCycler®480 System (Roche). 5 µL of DNA template was mixed with 10 µL of LightCycler® 480 SYBR Green I Master (Roche), 10 pmol forward and reverse primers in a final volume of 20 μL. After amplification (95 °C for 5 min; 45 cycles: 95 °C for 10 s, 60 °C for 10 s, 72 °C for 10 s), the Ct value was defined by LightCycler®480 software with default settings. Ct values higher than 42 were considered as no expression. Relative messenger RNA (mRNA) expression was calculated by normalization to the expression of ribosomal protein S9 (Rps9). The following primer pairs were used: Rps9 (F: 5′- CTGGACGAGGGCAAGATGAAGC; R: 5′-TGACTGTGGCGGATGAGCACA); Acsbg1 (F: 5′- CCAAAGAGTCTCCAAGTCACG; R: 5′- GAGTACAGAAAGGTTCCAGGC); Acyl-Coenzyme A Synthetase Long Chain Family Member 6 (Acsl6) (F: 5′-CAGAGGAACTCAACTACTGGACC; R: 5′-CCAATGTCTCCAGTGTGAAGCC); Acyl-Coenzyme A Synthetase Long Chain Family Member 5 (Acsl5) (F: 5′-GCATCATTCGGCGGGACAGTTT; R: 5′-GTCAAGACTGGAGTGGAGATGG).

Adoptive transfer colitis

Tnaïve cells were sorted by FACS as LinCD4+CD25CD62Lhigh from single-cell suspensions of pooled spleens from 8-week-old Acsbg1−/− mice or Acsbg1+/+ littermate controls. Sorted cells were washed twice with PBS, and 3 × 105 Tnaïve cells were injected intraperitoneally in a volume of 100 μL PBS into sex-matched 8 to 10-week-old Rag2−/− recipient mice. Rag2−/− mice injected with only 100 μL PBS served as controls. The body weight and health status of recipient mice were monitored over the course of 8 weeks according to the animal license number 33.19-42502-04-20/3540. At the end of the experiment, mice were sacrificed by CO2 asphyxiation, and either the colon was analyzed by histology or single-cell suspensions from the colon were prepared for flow cytometry.

Histological analyses

Colons and small intestines were collected, weighted and their length was measured. The weight-to-length ratio (mg cm−1) was calculated by dividing the organ weight by its length. The samples were arranged into a ‘Swiss roll’ configuration, fixed in 4% neutrally buffered formaldehyde, and embedded in paraffin using standard histological procedures. Sections approximately 3 μm thick were stained with hematoxylin and eosin (H&E) and analyzed under a light microscope. The evaluation was conducted in a randomized and blinded manner. The severity of inflammation was assessed using a histological scoring system adapted from the Jackson Laboratory score developed for scoring colitis in mice [32].

Software

Flow cytometric samples were acquired by BD FACS Diva Software (Heidelberg, Germany). Flow cytometric data were analyzed by FlowJo® 10.10.0 and Microsoft Excel 2024 (Microsoft Corporation, Munich, Germany), and graphs were produced by GraphPad Prism 10.4.1 (GrapPad Software, Boston, MA, USA). Endnote X9 (Clarivate Analytics, Philadelphia, PA, USA) was used for reference management.

Statistical analysis

Statistical analyses were performed using GraphPad Prism (v10.4.1). If not stated otherwise, data were presented as mean ± SEM. Comparisons between groups were made using two-way ANOVA with Šidák's or Tukey's multiple comparison tests, or the Mann-Whitney U test where applicable. Survival data were analyzed using the Kaplan-Meier method and Log-Rank (Mantel-Cox) test. A P-value below 0.05 was considered significant; *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001. Specific details for each analysis are provided in the Figure legends.

Ethics

According to the German Animal Welfare Act (§4, section 4), sacrificing animals solely to remove organs for scientific purposes is notified to the competent authority. All animal experiments were approved by the Lower Saxony Committee on the Ethics of Animal Experiments as well as the responsible state office (Lower Saxony State Office of Consumer Protection and Food Safety) under the permit number 33.19-42502-04-20/3540. This study was carried out in accordance with the principles of the Basel Declaration as well as recommendations as defined by FELASA (Federation of European Laboratory Animal Science Associations) and the German animal welfare body GV-SOLAS (Gesellschaft für Versuchstierkunde/Society for Laboratory Animal Science).

Results

Acsbg1 deficiency impairs TH17 and Treg cell differentiation in vitro

Acsbg1 has been identified as part of a unique epigenetic signature that distinguishes ex vivo TH17 cells from other TH cell subsets, with selective demethylation correlating with increased gene expression [26]. Recently, Kanno et al. identified Acsbg1 as a metabolic checkpoint for Treg cells that regulates mitochondrial fitness and is expressed in both ex vivo Treg and iTreg cells [28]. These findings suggest that Acsbg1 may play an important role in TH17 and Treg biology.

To assess the role of Acsbg1 in modulating the differentiation of TH cell subsets, we examined the effects of Acsbg1 deficiency on in vitro CD4⁺ T cell differentiation. Lymphocytes were isolated from the spleens of Acsbg1+/+ and Acsbg1−/− mice. Sorted CD4+CD25CD62Lhigh Tnaïve cells (Supplementary Fig. 1A) were cultured for four days under TH0 (non-polarizing), TH1, TH17, or iTreg polarizing conditions, followed by flow cytometric analysis (Supplementary Fig. 1B, Fig. 1A–E). Under TH1-polarizing conditions, both Acsbg1+/+ and Acsbg1−/− cells showed a high frequency of Foxp3T-bet+ and IFN-γ+ cells, consistent with the expected differentiation profile. However, no significant differences were observed between Acsbg1+/+ and Acsbg1−/− cultures (Fig. 1A–B). Instead, under TH17 polarizing conditions, both genotypes generated high frequencies of Foxp3RORγt+ and IL-17A+ cells (Fig. 1C–D). However, the frequency of Foxp3RORγt+ cells was significantly reduced in Acsbg1−/− cultures compared to Acsbg1+/+ controls, whereas the frequency of IL-17A+ cells remained comparable between the two groups (Fig. 1C–D). Similarly, under iTreg polarizing conditions, both Acsbg1+/+ and Acsbg1−/− cultures produced a high frequency of Foxp3+ cells, but the frequency of Foxp3+ cells was significantly lower in Acsbg1−/− cultures compared to Acsbg1+/+ controls (Fig. 1E). Under these conditions, IL-10 expression was barely detectable in both groups, making it difficult to evaluate potential differences in IL-10 production (Fig. 1E).

Fig. 1.
Fig. 1.

Acsbg1 deficiency affects in vitro TH17 and Treg cell differentiation. Tnaïve cells from Acsbg1+/+ and Acsbg1−/− mice were cultured under the indicated polarizing conditions for four days, followed by flow cytometric analysis. Representative flow cytometry plots on the left show the expression of key markers for Acsbg1+/+ and Acsbg1−/− cells, while bar graphs on the right show the frequency of specific cell populations across all conditions for Acsbg1+/+ (blue) and Acsbg1−/− (red) cells: (A) Foxp3⁻T-bet+ (TH1), (B) IFN-γ+ (TH1), (C) Foxp3⁻RORγt+ (TH17), (D) IL-17A+ (TH17), and (E) Foxp3+ (iTreg). Data were pooled from two independent experiments with 4 biological replicates. Each symbol represents the mean of technical triplicates performed for each condition in each experiment. Data are presented as mean ± SEM. Statistical significance was assessed using an ordinary two-way ANOVA with Šidák multiple comparison test (***P < 0.001)

Citation: European Journal of Microbiology and Immunology 15, 1; 10.1556/1886.2025.00003

Taken together, these results demonstrate that Acsbg1 deficiency selectively impairs in vitro TH17 and Treg differentiation, while leaving TH1 differentiation unaffected. These findings highlight the potential role of Acsbg1 in the regulation of specific CD4+ T cell subsets.

Acsbg1 is preferentially expressed in in vitro differentiated TH17 cells

To determine whether Acsbg1 expression is specifically induced during TH17 and iTreg differentiation, we examined its mRNA levels in in vitro differentiated CD4⁺ T cell subsets. Tnaïve cells, sorted from the spleen of Acsbg1+/+ mice as described above (Supplementary Fig. 1A), were cultured for four days under TH0 (non-polarizing), TH1, TH17, or iTreg polarizing conditions (Fig. 2). At the end of the cultures, cells were harvested, total RNA was extracted and converted to cDNA, and Acsbg1 expression was assessed by RT-qPCR. Minimal or no Acsbg1 expression was detected in TH0 and TH1 cells, whereas TH17 and iTreg cells showed significantly higher levels of Acsbg1 expression. Notably, Acsbg1 expression was significantly higher in TH17 cells compared to iTreg cells (Fig. 2A).

Fig. 2.
Fig. 2.

Acsbg1 is preferentially expressed by TH17 cells in vitro. Tnaïve cells from Acsbg1+/+ and Acsbg1−/− mice were cultured under the indicated polarizing conditions for four days, followed by RT-PCR analysis of the relative expression of (A) Acsbg1, (B) Acsl6 and (C) Acls5 in Acsbg1+/+ (blue) and Acsbg1−/− (red) cells cultured under the indicated conditions. Each symbol represents the mean of technical duplicates normalized to Rsp9 expression. Data are presented as mean ± SEM. Results are representative of pooled data from two independent experiments with (A) 8–10 or (B–C) 4–5 biological replicates. Statistical significance was determined by two-way ANOVA with Tukey's multiple comparison test (***P < 0.001, ****P < 0.0001)

Citation: European Journal of Microbiology and Immunology 15, 1; 10.1556/1886.2025.00003

To investigate whether the absence of Acsbg1 triggers compensatory mechanisms, we examined the expression of two paralogs, Acsl6 and Acsl5. RT-qPCR analysis revealed no significant changes in the expression of either Acsl6 or Acsl5 in Acsbg1−/− cultures compared to Acsbg1+/+ controls under any of the in vitro differentiation conditions tested (Fig. 2B and C).

These results indicate that, although Acsbg1 is not demethylated in in vitro differentiated TH17 and iTreg cells [26], it is expressed in both subsets, with the highest expression levels detected in TH17 cells. Furthermore, deletion of Acsbg1 does not induce compensatory expression of related paralogs, such as Acsl6 or Acsl5, highlighting the specific and non-redundant role of Acsbg1 in T cell differentiation.

Acsbg1 deficiency leads to more severe transfer colitis

While in vitro assays have provided insight into the role of Acsbg1 in the differentiation and function of TH17 and Treg cells, they do not fully recapitulate the in vivo behavior of the T cells. To overcome this limitation and to better study the role of Acsbg1 deficiency in vivo, we used an adoptive transfer colitis model based on the transfer of Tnaïve cells into immunodeficient recipients. Lymphocytes were isolated from the spleen of Acsbg1+/+ or Acsbg1−/− mice, and LinCD4+CD25CD62Lhigh Tnaïve cells were sorted as shown in Supplementary Fig. 1A. A total of 3 x 105 Tnaïve cells were transferred into sex-matched Rag2−/− mice by intraperitoneal injection. Rag2−/− mice injected with PBS served as controls. To monitor the development of colitis, the body weight was measured for 8 weeks after adoptive transfer (Fig. 3A). As expected, PBS-injected mice did not develop colitis, as indicated by a steady increase in body weight (Fig. 3A). Instead, mice injected with Acsbg1+/+ Tnaïve cells developed pronounced signs of colitis, as indicated by a loss of body weight starting approximately 3 weeks after adoptive transfer and persisting until 5 weeks post-injection (Fig. 3A). After 5 weeks, these mice began to regain body weight, suggesting partial resolution of colitis symptoms (Fig. 3A). Interestingly, mice injected with Acsbg1−/− Tnaïve cells also showed body weight loss from around 3 weeks post-transfer; however, unlike the Acsbg1+/+ group, these mice did not show any signs of weight recovery. Instead, they continued to lose body weight progressively until the end of the experiment, indicating more severe and prolonged colitis, as well as increased pathogenicity of Acsbg1-deficient CD4+ T cells (Fig. 3A). Consistent with these data, survival analysis revealed a trend towards reduced survival in mice receiving Acsbg1−/− Tnaïve cells compared to Acsbg1+/+ cells (Fig. 3B).

Fig. 3.
Fig. 3.

Transfer of Acsbg1 deficient Tnaïve cells results in greater body weight loss. CD62LhighCD25CD4+Lin Tnaïve cells were sorted from pooled spleens of Acsbg1+/+ (blue) or Acsbg1−/− (red) mice. 3 × 105 Tnaïve cells were adoptively transferred into sex-matched Rag2−/− mice by intraperitoneal injection. Control groups received PBS (white, dotted line). Mice were monitored over 56 days post infection. (A) Percentage change in body weight and (B) Kaplan-Meier survival curves were evaluated over the time course of the experiment. Eight weeks after adoptive transfer of Acsbg1+/+ or Acsbg1−/− Tnaïve cells, mice were sacrificed, and the length and weight of the (C) colon and (D) small intestine were assessed and depicted as weight to length ratio in the plot with bars. (E) Representative haematoxylin and eosin (H&E) staining of colon samples are shown. Scale bars represent 50 µm. Transmural invasion of inflammatory cells (i), epithelial hyperplasia (h), and loss of goblet cells (g) are labeled. (F) Bar plot showing the total histological score as a sum of five histological parameters: area involved, crypt hyperplasia, leukocyte invasion, severity of inflammation, and single cell apoptosis. Each symbol represents a single mouse. Data are presented as mean ± SEM. Data represent pooled results from 5 independent experiments with 7–15 mice or (B) 14–53 mice per experimental group. (A, C and D, F) Significance of differences between means was assessed using an ordinary two-way ANOVA with Tukey's multiple comparison test (**P < 0.01, ***P < 0.001, ****P < 0.0001). In (A) the significance of the difference between Acsbg1+/+ and Acsbg1−/− Tnaïve group means is shown at individual time points after injection. (B) Survival analysis was performed using the Log-Rank Test (Mantel-Cox) between the three survival curves (Chi-square = 8.878, df = 2, P = 0.0118) and between the Acsbg1+/+ or Acsbg1−/− groups (Chi-square = 1,502, df = 1, P = 0.2159)

Citation: European Journal of Microbiology and Immunology 15, 1; 10.1556/1886.2025.00003

At 8 weeks after adoptive transfer, both Acsbg1−/− and Acsbg1+/+ Tnaïve cell-transferred mice exhibited similar colon and small intestine weight-to-length ratios, which were significantly higher than PBS controls (Fig. 3C and D). Consistent with these findings, histological analysis of colon samples confirmed comparable inflammatory changes in both groups, including severity of inflammation, leukocyte invasion, single cell apoptosis, area involved, and crypt hyperplasia (Fig. 3E and F). These findings suggest that while Acsbg1 deficiency exacerbated weight loss and survival outcomes, the overall level of colonic inflammation remained largely unaffected.

To further investigate the impact of Acsbg1 deficiency on T cells in vivo, mice were sacrificed 8 weeks after adoptive transfer and flow cytometric analyses were performed on T cells isolated from the colonic lamina propria (Supplementary Fig. 2A and B). Despite similar frequencies of pro-inflammatory cytokine-producing cells, immunophenotyping revealed an increased absolute number of IFN-γ+IL-17A, IFN-γIL-17A+, IFN-γ+IL-17A+, and IL-22+ CD4+ T cells among colonic lamina propria CD4+ T cells in Acsbg1−/− Tnaïve-transferred mice compared to Acsbg1+/+ controls (Supplementary Fig. 3A–D), supporting the hypothesis that Acsbg1 deficiency may result in an increased pathogenicity of CD4+ T cells. Despite an increased IFN-γ production, the frequency and absolute number of TH1 cells (Foxp3T-bet+) remained unchanged (Fig. 4A). However, the absolute number of TH17 cells (Foxp3RORγt+) was significantly increased in Acsbg1−/− Tnaïve-transferred mice compared to Acsbg1+/+ controls, whereas the frequency of TH17 cells remained comparable between the groups (Fig. 4B). Interestingly, an imbalance between Foxp3- conventional T (Tconv) and Treg cells was observed in Acsbg1−/− Tnaïve cell-transferred mice compared to Acsbg1+/+ controls. The frequencies of Tconv cells were significantly reduced (Supplementary Fig. 3E), whereas the frequencies and absolute numbers of CD25+Foxp3+ Treg cells were significantly increased in Acsbg1−/− Tnaïve cell-transferred mice compared to Acsbg1+/+ controls (Fig. 4C).

Fig. 4.
Fig. 4.

Transfer of Acsbg1 deficient Tnaïve cells results in more severe colitis. CD62LhighCD25CD4+Lin Tnaïve cells were sorted from pooled spleens of Acsbg1+/+ (blue) or Acsbg1−/− (red) mice. 3 × 105 Tnaïve cells were adoptively transferred into sex-matched Rag2−/− mice by intraperitoneal injection. Control groups received PBS (white). Eight weeks after adoptive transfer, mice were sacrificed and flow cytometric analysis was performed on cells isolated from the colonic lamina propria. Summary graphs of the frequencies and absolute cell numbers of (A) Foxp3T-bet+, (B) Foxp3RORγt+, (C) Foxp3+CD25+ cells among CD8CD4+ T cells. Each symbol represents a single mouse. Data are presented as mean ± SEM. Data represent results pooled from 5 independent experiments with 11–15 mice. Significance of differences between means was assessed by Mann-Whitney test (*P < 0.05, ***P < 0.001)

Citation: European Journal of Microbiology and Immunology 15, 1; 10.1556/1886.2025.00003

In conclusion, these results suggest that Acsbg1 deficiency exacerbates colitis by dysregulating TH17 and Treg cell differentiation and function, leading to increased pathogenicity of CD4+ T cells. This highlights the essential role of Acsbg1 in maintaining immune homeostasis and provides a basis for further exploration of its therapeutic potential in intestinal inflammatory diseases.

Discussion

Epigenetic modifications, such as DNA methylation, play a critical role in establishing and maintaining distinct gene expression patterns in TH cells, ultimately influencing their fate and function [33, 34]. In Treg cells, specific epigenetic signature genes imprint a unique transcriptional program, that stabilizes the expression of suppressive molecules [35–37]. Similarly, ex vivo isolated TH17 cells exhibit a unique epigenetic signature characterized by selective demethylation of regions located in seven genes: Il17a, Zfp362, Ccr6, Acsbg1, Rora, Dpp4, and Dclk1 [26]. Of these, Il17a [38], Ccr6 [39], Rora [40] and Dpp4 [41], have already been demonstrated to be associated with TH17 biology, while Zfp362 has been shown to act as a negative regulator of Treg cells [42]. However, the functions of Acsbg1 and the serine/threonine protein kinase Dclk1 in the cell differentiation, maintenance, functional properties, and plasticity of TH17 cells remain to be fully elucidated. Recent evidence suggests that Acsbg1 is critical for Treg homeostasis by maintaining their mitochondrial fitness, and promoting the induction of tissue-protective ST2⁺ Treg cells during lung inflammation [28]. Consequently, we postulated that Acsbg1 may play a role in both Treg and TH17 cells.

The present study provides novel insights into the role of Acsbg1 in regulating T cell differentiation and immune homeostasis through a combination of in vitro assays and in vivo models. In vitro, Acsbg1 deficiency selectively impaired TH17 and iTreg differentiation, as evidenced by reduced frequencies of Foxp3RORγt+ and Foxp3+ cells, respectively. Conversely, TH1 differentiation remained unaffected, with no significant changes observed in Foxp3Tbet+ and IFN-γ+ cells. These findings suggest that Acsbg1 is critical for the programming of TH17 and Treg subsets, possibly through its involvement in lipid metabolism, a key determinant of T cell polarization states [15]. TH1 cells, on the other hand, may rely on alternative metabolic pathways. Supporting this hypothesis, Acsbg1 expression was detected in in vitro differentiated TH17 and iTreg cells, albeit at higher levels in TH17 cells, while being minimal in TH1 cells. Interestingly, Acsbg1 was expressed in these cells, although it has been reported to be fully methylated [26], suggesting that methylation at the Acsbg1 locus does not fully repress its transcription in in vitro differentiated TH17 and iTreg cells. One possible explanation is that the differentially methylated region within the Acsbg1 locus resides in the gene body, modulating rather than silencing its expression [26]. Alternatively, Acsbg1 expression could be regulated by environmental, metabolic or transcriptional factors that override methylation. For example, TGF-β signaling via Smad2/3 has been shown to upregulate Acsbg1 expression during iTreg differentiation by opening the chromatin at the Acsbg1 locus, while IL-33, but not TGF-β, increases Acsbg1 expression in lung Treg cells [28]. Furthermore, neither Acsl6 nor Acsl5, two Acsbg1 paralogs [22], exhibited compensatory expression in Acsbg1-deficient cells, emphasizing the unique role of Acsbg1 in TH17 and Treg differentiation.

Notably, in the adoptive transfer colitis model, despite impaired in vitro differentiation, there was an increased accumulation of both TH17 and Treg cells in the colonic lamina propria of Acsbg1−/− Tnaïve cell-transferred mice compared to Acsbg1+/+ controls. This apparent discrepancy highlights the influence of the in vivo microenvironment, where inflammatory cytokines and accessory signals may override the differentiation defects observed in vitro. Furthermore, compensatory pathways may sustain the expansion and survival of TH17 and Treg cells in vivo despite Acsbg1 deficiency. Notably, Acsbg1-deficient T cells caused significantly greater body weight loss and a trend towards reduced survival compared to Acsbg1+/+ controls, underscoring their increased pathogenicity. While both Acsbg1+/+ and Acsbg1−/− cells led to similar histopathological outcomes, the exaggerated body weight loss in the Acsbg1−/− group likely reflects an imbalance between pro-inflammatory and regulatory T cells. Flow cytometric analysis revealed increased absolute numbers of pro-inflammatory populations, including IFN-γ+IL-17A, IFN-γIL-17A+, IFN-γ+IL-17A+, and IL-22+ cells, as well as TH17 (Foxp3RORγt+) cells in Acsbg1−/− Tnaïve cell-transferred mice compared to controls. These findings suggest that Acsbg1-deficient TH17 cells may adopt a more pathogenic phenotype, promoting inflammation. Unexpectedly, Treg numbers were also increased, however this may reflect a compensatory mechanism in response to enhanced colitis, and a failure to effectively suppress ongoing TH17-driven inflammation due to functional impairments, consistent with prior evidence showing that Acsbg1 supports the suppressive capacity of Treg cells [28]. Collectively, these findings indicate that Acsbg1 deficiency disrupts the balance between pro-inflammatory TH17 cells and immunosuppressive Treg cells, exacerbating colitis.

The mechanism by which Acsbg1 regulates T cell proliferation, differentiation and function remains unclear. Acsbg1, like other ACSs, plays a critical role in lipid metabolism by activating FAs into acyl-Coenzyme A (acyl-CoA) molecules, which fuel anabolic and catabolic processes [22]. Therefore, Acsbg1 deficiency could affect any of these pathways, altering for instance energy production, membrane synthesis, and signaling lipid generation, all processes integral to T cell differentiation and function. Emerging evidence suggests that TH17 cells rely on glycolytic-lipogenic pathways [19] and cholesterol synthesis [43, 44], while Treg cells depend on OXPHOS and FAO for energy. Interestingly, npTH17 and pTH17 cells display distinct metabolic profiles, mostly dependent on OXPHOS or on both glycolysis and OXPHOS, respectively, and the intracellular ratio of saturated to polyunsaturated FAs influences TH17 fate, determining their pro- or anti-inflammatory phenotype [445]. Acsbg1 likely supports these metabolic programs, and its absence may alter key checkpoints, affecting T cell function and stability. Future studies employing metabolomics and lipidomics approaches could further elucidate how Acsbg1 deficiency reshapes T cell metabolism and influences immune regulation.

This study focused on T cell-intrinsic effects, but our previous epigenetic study showed that Acsbg1 is demethylated not only in ex vivo TH17 cells, but also in γδ T cells, B cells, neutrophils, monocytes, macrophages, conventional dendritic cells and natural killer cells [26]. This suggests a broader role for Acsbg1 across immune cell types and warrants further investigation. Moreover, functional assays to assess the suppressive capacity of Acsbg1-deficient Treg cells and additional models to investigate TH17-Treg interplay are warranted to elucidate its precise role in shaping immune responses.

In conclusion, our findings establish Acsbg1 as a critical regulator of T cell differentiation and immune homeostasis. By modulating the balance between TH17 and Treg cells, Acsbg1 deficiency exacerbates colitis, highlighting its essential role in controlling inflammation in mucosal tissues. This study provides a foundation for future research into the mechanistic links between lipid metabolism and immune regulation, with potential implications for the treatment of inflammatory diseases such as IBD.

Funding sources

This project has been funded by the European Union's Horizon 2020 Research and Innovation Programme under the Marie Skłodowska-Curie Grant Agreement No. 955321 and by the Deutsche Forschungsgemeinschaft (DFG) under the grant SFB/TRR 355/1 (project number: 490846870).

Authors' contributions

MP and JH designed the research. MP, FK, SG, and MGP performed the experiments. MP, GS, AB, MGP and JH analyzed, discussed, and interpreted the data. MP and JH wrote the manuscript.

Conflicts of interest

The authors declare no conflict of interest.

Supplementary material

Supplementary data to this article can be found online at https://doi.org/10.1556/1886.2025.00003.

Abbreviations

ACC1

Acyl-Coenzyme A Carboxylase 1

ACS

Acyl-CoA synthetases

Acsbg1

Acyl-CoA Synthetase Bubble Gum 1

Acsl5/6

Acyl-Coenzyme A Synthetase Long Chain Family Member 5/6

BSA

Bovine serum albumin

cDNA

Complementary DNA

CoA

Coenzyme A

EDTA

ethylenediaminetetraacetic acid

FACS

fluorescence-activated cell sorting

FAO

Fatty acid oxidation

FAS

Fatty acid synthesis

FAs

Fatty acids

H&E

hematoxylin and eosin

HBSS

Hanks' balanced salt solution

HEPES

4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid

hTGF-β1

human transforming growth factor β1

IBD

Inflammatory Bowel disease

IFN-γ

interferon-γ

IL

Interleukin

IMDM

Iscove's Modified Dulbecco's Medium

iTreg

in vitro-induced Treg

mRNA

Messenger RNA

npTH17

non-pathogenic TH17 cell

OXPHOS

Oxidative phosphorylation

PBS

Phosphate-buffered saline

pTH17

pathogenic TH17 cells

Rsp9

ribosomal protein S9

RT-qPCR

real-time quantitative polymerase chain reaction

Tconv

Conventional T cell

TH0/1/17

T helper 0/1/17 cell

Treg

Regulatory T cell

Acknowledgements

We thank Lothar Groebe and Beate Pietzsch for cell sorting.

References

  • 1.

    Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, et al. Reciprocal developmental pathways for the generation of pathogenic effector Th17 and regulatory T cells. Nature. 2006;441:2358.

    • Search Google Scholar
    • Export Citation
  • 2.

    Harrington LE, Hatton RD, Mangan PR, Turner H, Murphy TL, Murphy KM, et al. Interleukin 17–producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nat Immunol. 2005;6:112332.

    • Search Google Scholar
    • Export Citation
  • 3.

    Mangan PR, Harrington LE, O’Quinn DB, Helms WS, Bullard DC, Elson CO, et al. Transforming growth factor-β induces development of the Th17 lineage. Nature. 2006;441:2314.

    • Search Google Scholar
    • Export Citation
  • 4.

    Omenetti S, Bussi C, Metidji A, Iseppon A, Lee S, Tolaini M, et al. The intestine harbors functionally distinct homeostatic tissue-resident and inflammatory Th17 Cells. Immunity. 2019;51:7789.e6.

    • Search Google Scholar
    • Export Citation
  • 5.

    Khattri R, Cox T, Yasayko S-A, Ramsdell F. An essential role for Scurfin in CD4+CD25+ T regulatory cells. Nat Immunol. 2003;4:33742.

  • 6.

    Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science. 2003;299:105761.

  • 7.

    Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. The J Immunol. 1995;155:115164.

    • Search Google Scholar
    • Export Citation
  • 8.

    Brucklacher-Waldert V, Carr EJ, Linterman MA, Veldhoen M. Cellular plasticity of CD4+ T cells in the intestine. Front Immunol. 2014;5.

  • 9.

    Littman DR, Rudensky AY. Th17 and regulatory T cells in mediating and restraining inflammation. Cell. 2010;140:84558.

  • 10.

    Yan J, Luo M, Chen Z, He B. The function and role of the Th17/Treg cell balance in inflammatory bowel disease. The J Immunol Res. 2020;2020:8813558.

    • Search Google Scholar
    • Export Citation
  • 11.

    Eisenstein EM, Williams CB. The Treg/Th17 cell balance: a new paradigm for autoimmunity. Pediatr Res. 2009;65:2631.

  • 12.

    Zhang S, Gang X, Yang S, Cui M, Sun L, Li Z, et al. The alterations in and the role of the Th17/Treg balance in metabolic diseases. Front Immunol. 2021;12.

    • Search Google Scholar
    • Export Citation
  • 13.

    Bantug GR, Galluzzi L, Kroemer G, Hess C. The spectrum of T cell metabolism in health and disease. Nat Rev Immunol. 2018;18:1934.

  • 14.

    Buck MD, O’Sullivan D, Pearce EL. T cell metabolism drives immunity. J Exp Med. 2015;212:134560.

  • 15.

    Soriano-Baguet L, Brenner D. Metabolism and epigenetics at the heart of T cell function. Trends Immunol. 2023;44:23144.

  • 16.

    Chapman NM, Boothby MR, Chi H. Metabolic coordination of T cell quiescence and activation. Nat Rev Immunol. 2020;20:5570.

  • 17.

    MacIver NJ, Michalek RD, Rathmell JC. Metabolic regulation of T lymphocytes. Annu Rev Immunol. 2013;31:25983.

  • 18.

    Reilly NA, Lutgens E, Kuiper J, Heijmans BT, Jukema JW. Effects of fatty acids on T cell function: role in atherosclerosis. Nat Rev Cardiol. 2021;18:82437.

    • Search Google Scholar
    • Export Citation
  • 19.

    Berod L, Friedrich C, Nandan A, Freitag J, Hagemann S, Harmrolfs K, et al. Erratum: De novo fatty acid synthesis controls the fate between regulatory T and T helper 17 cells. Nat Med. 2015;21:414414.

    • Search Google Scholar
    • Export Citation
  • 20.

    Michalek RD, Gerriets VA, Jacobs SR, Macintyre AN, MacIver NJ, Mason EF, et al. Cutting Edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets. J Immunol. 2011;186:3299303.

    • Search Google Scholar
    • Export Citation
  • 21.

    Weinberg SE, Singer BD, Steinert EM, Martinez CA, Mehta MM, Martínez-Reyes I, et al. Mitochondrial complex III is essential for suppressive function of regulatory T cells. Nature. 2019;565:4959.

    • Search Google Scholar
    • Export Citation
  • 22.

    Grevengoed TJ, Klett EL, Coleman RA. Acyl-CoA metabolism and partitioning. Annu Rev Nutr. 2014;34:130.

  • 23.

    Fraisl P, Forss-Petter S, Zigman M, Berger J. Murine bubblegum orthologue is a microsomal very long-chain acyl-CoA synthetase. Biochem J. 2004;377:8593.

    • Search Google Scholar
    • Export Citation
  • 24.

    Steinberg SJ, Morgenthaler J, Heinzer AK, Smith KD, Watkins PA. Very long-chain acyl-CoA synthetases. Human “bubblegum” represents a new family of proteins capable of activating very long-chain fatty acids. J Biol Chem. 2000;275:351629.

    • Search Google Scholar
    • Export Citation
  • 25.

    Pei Z, Oey NA, Zuidervaart MM, Jia Z, Li Y, Steinberg SJ, et al. The acyl-CoA synthetase “bubblegum” (lipidosin): further characterization and role in neuronal fatty acid beta-oxidation. J Biol Chem. 2003;278:470708.

    • Search Google Scholar
    • Export Citation
  • 26.

    Yang B-H, Floess S, Hagemann S, Deyneko IV, Groebe L, Pezoldt J, et al. Development of a unique epigenetic signature during in vivo Th17 differentiation. Nucleic Acids Res. 2015;43:153748.

    • Search Google Scholar
    • Export Citation
  • 27.

    Yang B-H, Hagemann S, Mamareli P, Lauer U, Hoffmann U, Beckstette M, et al. Foxp3+ T cells expressing RORγt represent a stable regulatory T-cell effector lineage with enhanced suppressive capacity during intestinal inflammation. Mucosal Immunol. 2016;9:44457.

    • Search Google Scholar
    • Export Citation
  • 28.

    Kanno T, Nakajima T, Kawashima Y, Yokoyama S, Asou HK, Sasamoto S, et al. Acsbg1-dependent mitochondrial fitness is a metabolic checkpoint for tissue Treg cell homeostasis. Cell Rep. 2021;37.

    • Search Google Scholar
    • Export Citation
  • 29.

    Coleman JLJ, Brennan K, Ngo T, Balaji P, Graham RM, Smith NJ. Rapid knockout and reporter mouse line generation and breeding colony establishment using EUCOMM conditional-ready embryonic stem cells: a case study. Front Endocrinol. 2015;6.

    • Search Google Scholar
    • Export Citation
  • 30.

    Skarnes WC, Rosen B, West AP, Koutsourakis M, Bushell W, Iyer V, et al. A conditional knockout resource for the genome-wide study of mouse gene function. Nature .2011;474:33742.

    • Search Google Scholar
    • Export Citation
  • 31.

    Cossarizza A, Chang H-D, Radbruch A, Abrignani S, Addo R, Akdis M, et al. Guidelines for the use of flow cytometry and cell sorting in immunological studies (third edition). Eur J Immunol. 2021;51:27083145.

    • Search Google Scholar
    • Export Citation
  • 32.

    M M, Ij B, Eh L, Ae W, Eh B, Co E, et al. Differential susceptibility of inbred mouse strains to dextran sulfate sodium-induced colitis. Am J Physiol. 1998;274.

    • Search Google Scholar
    • Export Citation
  • 33.

    Wilson CB, Rowell E, Sekimata M. Epigenetic control of T-helper-cell differentiation. Nat Rev Immunol. 2009;9:91105.

  • 34.

    Schmidl C, Delacher M, Huehn J, Feuerer M. Epigenetic mechanisms regulating T-cell responses. J Allergy Clin Immunol. 2018;142:72843.

    • Search Google Scholar
    • Export Citation
  • 35.

    Huehn J, Beyer M. Epigenetic and transcriptional control of Foxp3+ regulatory T cells. Semin Immunol. 2015;27:108.

  • 36.

    Herppich S, Toker A, Pietzsch B, Kitagawa Y, Ohkura N, Miyao T, et al. Dynamic imprinting of the Treg cell-specific epigenetic signature in developing thymic regulatory T cells. Front Immunol. 2019;10:2382.

    • Search Google Scholar
    • Export Citation
  • 37.

    Ohkura N, Hamaguchi M, Morikawa H, Sugimura K, Tanaka A, Ito Y, et al. T cell receptor stimulation-induced epigenetic changes and Foxp3 expression are independent and complementary events required for Treg cell development. Immunity 2012;37:78599.

    • Search Google Scholar
    • Export Citation
  • 38.

    Kuchroo VK, Awasthi A. Emerging new roles of Th17 cells. Eur J Immunol. 2012;42:22114.

  • 39.

    Wang C, Kang SG, Lee J, Sun Z, Kim CH. The roles of CCR6 in migration of Th17 cells and regulation of effector T-cell balance in the gut. Mucosal Immunol. 2009;2:17383.

    • Search Google Scholar
    • Export Citation
  • 40.

    Yang XO, Pappu BP, Nurieva R, Akimzhanov A, Kang HS, Chung Y, et al. T helper 17 lineage differentiation is programmed by orphan nuclear receptors ROR alpha and ROR gamma. Immunity 2008;28:2939.

    • Search Google Scholar
    • Export Citation
  • 41.

    Zhao X, Wang W, Zhang K, Yang J, Fuchs H, Fan H. Involvement of CD26 in differentiation and functions of TH1 and TH17 subpopulations of T lymphocytes. J Immunol Res. 2021;2021:6671410.

    • Search Google Scholar
    • Export Citation
  • 42.

    Herppich S, Hoenicke L, Kern F, Kruse F, Smout J, Greweling-Pils MC, et al. Zfp362 potentiates murine colonic inflammation by constraining Treg cell function rather than promoting TH17 cell differentiation. Eur J Immunol. 2023;53:e2250270.

    • Search Google Scholar
    • Export Citation
  • 43.

    Hu X, Wang Y, Hao L-Y, Liu X, Lesch CA, Sanchez BM, et al. Sterol metabolism controls TH17 differentiation by generating endogenous RORγ agonists. Nat Chem Biol 2015;11:1417.

    • Search Google Scholar
    • Export Citation
  • 44.

    Santori FR, Huang P, van de Pavert SA, Douglass EF, Leaver DJ, Haubrich BA, et al. Identification of natural RORγ ligands that regulate the development of lymphoid cells. Cell Metab. 2015;21:28698.

    • Search Google Scholar
    • Export Citation
  • 45.

    Wang C, Yosef N, Gaublomme J, Wu C, Lee Y, Clish CB, et al. CD5L/AIM regulates lipid biosynthesis and restrains TH17 cell pathogenicity. Cell. 2015;163:141327.

    • Search Google Scholar
    • Export Citation
  • 1.

    Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, et al. Reciprocal developmental pathways for the generation of pathogenic effector Th17 and regulatory T cells. Nature. 2006;441:2358.

    • Search Google Scholar
    • Export Citation
  • 2.

    Harrington LE, Hatton RD, Mangan PR, Turner H, Murphy TL, Murphy KM, et al. Interleukin 17–producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nat Immunol. 2005;6:112332.

    • Search Google Scholar
    • Export Citation
  • 3.

    Mangan PR, Harrington LE, O’Quinn DB, Helms WS, Bullard DC, Elson CO, et al. Transforming growth factor-β induces development of the Th17 lineage. Nature. 2006;441:2314.

    • Search Google Scholar
    • Export Citation
  • 4.

    Omenetti S, Bussi C, Metidji A, Iseppon A, Lee S, Tolaini M, et al. The intestine harbors functionally distinct homeostatic tissue-resident and inflammatory Th17 Cells. Immunity. 2019;51:7789.e6.

    • Search Google Scholar
    • Export Citation
  • 5.

    Khattri R, Cox T, Yasayko S-A, Ramsdell F. An essential role for Scurfin in CD4+CD25+ T regulatory cells. Nat Immunol. 2003;4:33742.

  • 6.

    Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science. 2003;299:105761.

  • 7.

    Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. The J Immunol. 1995;155:115164.

    • Search Google Scholar
    • Export Citation
  • 8.

    Brucklacher-Waldert V, Carr EJ, Linterman MA, Veldhoen M. Cellular plasticity of CD4+ T cells in the intestine. Front Immunol. 2014;5.

  • 9.

    Littman DR, Rudensky AY. Th17 and regulatory T cells in mediating and restraining inflammation. Cell. 2010;140:84558.

  • 10.

    Yan J, Luo M, Chen Z, He B. The function and role of the Th17/Treg cell balance in inflammatory bowel disease. The J Immunol Res. 2020;2020:8813558.

    • Search Google Scholar
    • Export Citation
  • 11.

    Eisenstein EM, Williams CB. The Treg/Th17 cell balance: a new paradigm for autoimmunity. Pediatr Res. 2009;65:2631.

  • 12.

    Zhang S, Gang X, Yang S, Cui M, Sun L, Li Z, et al. The alterations in and the role of the Th17/Treg balance in metabolic diseases. Front Immunol. 2021;12.

    • Search Google Scholar
    • Export Citation
  • 13.

    Bantug GR, Galluzzi L, Kroemer G, Hess C. The spectrum of T cell metabolism in health and disease. Nat Rev Immunol. 2018;18:1934.

  • 14.

    Buck MD, O’Sullivan D, Pearce EL. T cell metabolism drives immunity. J Exp Med. 2015;212:134560.

  • 15.

    Soriano-Baguet L, Brenner D. Metabolism and epigenetics at the heart of T cell function. Trends Immunol. 2023;44:23144.

  • 16.

    Chapman NM, Boothby MR, Chi H. Metabolic coordination of T cell quiescence and activation. Nat Rev Immunol. 2020;20:5570.

  • 17.

    MacIver NJ, Michalek RD, Rathmell JC. Metabolic regulation of T lymphocytes. Annu Rev Immunol. 2013;31:25983.

  • 18.

    Reilly NA, Lutgens E, Kuiper J, Heijmans BT, Jukema JW. Effects of fatty acids on T cell function: role in atherosclerosis. Nat Rev Cardiol. 2021;18:82437.

    • Search Google Scholar
    • Export Citation
  • 19.

    Berod L, Friedrich C, Nandan A, Freitag J, Hagemann S, Harmrolfs K, et al. Erratum: De novo fatty acid synthesis controls the fate between regulatory T and T helper 17 cells. Nat Med. 2015;21:414414.

    • Search Google Scholar
    • Export Citation
  • 20.

    Michalek RD, Gerriets VA, Jacobs SR, Macintyre AN, MacIver NJ, Mason EF, et al. Cutting Edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets. J Immunol. 2011;186:3299303.

    • Search Google Scholar
    • Export Citation
  • 21.

    Weinberg SE, Singer BD, Steinert EM, Martinez CA, Mehta MM, Martínez-Reyes I, et al. Mitochondrial complex III is essential for suppressive function of regulatory T cells. Nature. 2019;565:4959.

    • Search Google Scholar
    • Export Citation
  • 22.

    Grevengoed TJ, Klett EL, Coleman RA. Acyl-CoA metabolism and partitioning. Annu Rev Nutr. 2014;34:130.

  • 23.

    Fraisl P, Forss-Petter S, Zigman M, Berger J. Murine bubblegum orthologue is a microsomal very long-chain acyl-CoA synthetase. Biochem J. 2004;377:8593.

    • Search Google Scholar
    • Export Citation
  • 24.

    Steinberg SJ, Morgenthaler J, Heinzer AK, Smith KD, Watkins PA. Very long-chain acyl-CoA synthetases. Human “bubblegum” represents a new family of proteins capable of activating very long-chain fatty acids. J Biol Chem. 2000;275:351629.

    • Search Google Scholar
    • Export Citation
  • 25.

    Pei Z, Oey NA, Zuidervaart MM, Jia Z, Li Y, Steinberg SJ, et al. The acyl-CoA synthetase “bubblegum” (lipidosin): further characterization and role in neuronal fatty acid beta-oxidation. J Biol Chem. 2003;278:470708.

    • Search Google Scholar
    • Export Citation
  • 26.

    Yang B-H, Floess S, Hagemann S, Deyneko IV, Groebe L, Pezoldt J, et al. Development of a unique epigenetic signature during in vivo Th17 differentiation. Nucleic Acids Res. 2015;43:153748.

    • Search Google Scholar
    • Export Citation
  • 27.

    Yang B-H, Hagemann S, Mamareli P, Lauer U, Hoffmann U, Beckstette M, et al. Foxp3+ T cells expressing RORγt represent a stable regulatory T-cell effector lineage with enhanced suppressive capacity during intestinal inflammation. Mucosal Immunol. 2016;9:44457.

    • Search Google Scholar
    • Export Citation
  • 28.

    Kanno T, Nakajima T, Kawashima Y, Yokoyama S, Asou HK, Sasamoto S, et al. Acsbg1-dependent mitochondrial fitness is a metabolic checkpoint for tissue Treg cell homeostasis. Cell Rep. 2021;37.

    • Search Google Scholar
    • Export Citation
  • 29.

    Coleman JLJ, Brennan K, Ngo T, Balaji P, Graham RM, Smith NJ. Rapid knockout and reporter mouse line generation and breeding colony establishment using EUCOMM conditional-ready embryonic stem cells: a case study. Front Endocrinol. 2015;6.

    • Search Google Scholar
    • Export Citation
  • 30.

    Skarnes WC, Rosen B, West AP, Koutsourakis M, Bushell W, Iyer V, et al. A conditional knockout resource for the genome-wide study of mouse gene function. Nature .2011;474:33742.

    • Search Google Scholar
    • Export Citation
  • 31.

    Cossarizza A, Chang H-D, Radbruch A, Abrignani S, Addo R, Akdis M, et al. Guidelines for the use of flow cytometry and cell sorting in immunological studies (third edition). Eur J Immunol. 2021;51:27083145.

    • Search Google Scholar
    • Export Citation
  • 32.

    M M, Ij B, Eh L, Ae W, Eh B, Co E, et al. Differential susceptibility of inbred mouse strains to dextran sulfate sodium-induced colitis. Am J Physiol. 1998;274.

    • Search Google Scholar
    • Export Citation
  • 33.

    Wilson CB, Rowell E, Sekimata M. Epigenetic control of T-helper-cell differentiation. Nat Rev Immunol. 2009;9:91105.

  • 34.

    Schmidl C, Delacher M, Huehn J, Feuerer M. Epigenetic mechanisms regulating T-cell responses. J Allergy Clin Immunol. 2018;142:72843.

    • Search Google Scholar
    • Export Citation
  • 35.

    Huehn J, Beyer M. Epigenetic and transcriptional control of Foxp3+ regulatory T cells. Semin Immunol. 2015;27:108.

  • 36.

    Herppich S, Toker A, Pietzsch B, Kitagawa Y, Ohkura N, Miyao T, et al. Dynamic imprinting of the Treg cell-specific epigenetic signature in developing thymic regulatory T cells. Front Immunol. 2019;10:2382.

    • Search Google Scholar
    • Export Citation
  • 37.

    Ohkura N, Hamaguchi M, Morikawa H, Sugimura K, Tanaka A, Ito Y, et al. T cell receptor stimulation-induced epigenetic changes and Foxp3 expression are independent and complementary events required for Treg cell development. Immunity 2012;37:78599.

    • Search Google Scholar
    • Export Citation
  • 38.

    Kuchroo VK, Awasthi A. Emerging new roles of Th17 cells. Eur J Immunol. 2012;42:22114.

  • 39.

    Wang C, Kang SG, Lee J, Sun Z, Kim CH. The roles of CCR6 in migration of Th17 cells and regulation of effector T-cell balance in the gut. Mucosal Immunol. 2009;2:17383.

    • Search Google Scholar
    • Export Citation
  • 40.

    Yang XO, Pappu BP, Nurieva R, Akimzhanov A, Kang HS, Chung Y, et al. T helper 17 lineage differentiation is programmed by orphan nuclear receptors ROR alpha and ROR gamma. Immunity 2008;28:2939.

    • Search Google Scholar
    • Export Citation
  • 41.

    Zhao X, Wang W, Zhang K, Yang J, Fuchs H, Fan H. Involvement of CD26 in differentiation and functions of TH1 and TH17 subpopulations of T lymphocytes. J Immunol Res. 2021;2021:6671410.

    • Search Google Scholar
    • Export Citation
  • 42.

    Herppich S, Hoenicke L, Kern F, Kruse F, Smout J, Greweling-Pils MC, et al. Zfp362 potentiates murine colonic inflammation by constraining Treg cell function rather than promoting TH17 cell differentiation. Eur J Immunol. 2023;53:e2250270.

    • Search Google Scholar
    • Export Citation
  • 43.

    Hu X, Wang Y, Hao L-Y, Liu X, Lesch CA, Sanchez BM, et al. Sterol metabolism controls TH17 differentiation by generating endogenous RORγ agonists. Nat Chem Biol 2015;11:1417.

    • Search Google Scholar
    • Export Citation
  • 44.

    Santori FR, Huang P, van de Pavert SA, Douglass EF, Leaver DJ, Haubrich BA, et al. Identification of natural RORγ ligands that regulate the development of lymphoid cells. Cell Metab. 2015;21:28698.

    • Search Google Scholar
    • Export Citation
  • 45.

    Wang C, Yosef N, Gaublomme J, Wu C, Lee Y, Clish CB, et al. CD5L/AIM regulates lipid biosynthesis and restrains TH17 cell pathogenicity. Cell. 2015;163:141327.

    • Search Google Scholar
    • Export Citation
  • Collapse
  • Expand

Senior editors

Editor(s)-in-Chief: Dunay, Ildiko Rita, Prof. Dr. Pharm, Dr. rer. nat., University of Magdeburg, Germany

Editor(s)-in-Chief: Heimesaat, Markus M., Prof. Dr. med., Charité - University Medicine Berlin, Germany

Editorial Board

  • Berit Bangoura, Dr. DVM. PhD,  University of Wyoming, USA
  • Stefan Bereswill, Prof. Dr. rer. nat., Charité - University Medicine Berlin, Germany
  • Dunja Bruder, Prof. Dr. rer. nat., University of Magdeburg, Germany
  • Jan Buer, Prof. Dr. med., University of Duisburg, Germany
  • Edit Buzas, Prof. Dr. med., Semmelweis University, Hungary
  • Renato Damatta, Prof. PhD, UENF, Brazil
  • Maria Deli, MD, PhD, DSc, Biological Research Center, HAS, Hungary
  • Olgica Djurković-Djaković, Prof. Phd, University of Belgrade, Serbia
  • Jean-Dennis Docquier, Prof. Dr. med., University of Siena, Italy
  • Zsuzsanna Fabry, Prof. Phd, University of Washington, USA
  • Ralf Ignatius, Prof. Dr. med., Charité - University Medicine Berlin, Germany
  • Achim Kaasch, Prof. Dr. med., Otto von Guericke University Magdeburg, Germany
  • Oliver Liesenfeld, Prof. Dr. med., Inflammatix, USA
  • Matyas Sandor, Prof. PhD, University of Wisconsin, USA
  • Ulrich Steinhoff, Prof. PhD, University of Marburg, Germany
  • Michal Toborek, Prof. PhD, University of Miami, USA
  • Susanne A. Wolf, PhD, MDC-Berlin, Germany

 

Dr. Dunay, Ildiko Rita
Magdeburg, Germany
E-mail: ildiko.dunay@med.ovgu.de

Indexing and Abstracting Services:

  • PubMed Central
  • Scopus
  • ESCI
  • CABI
  • CABELLS Journalytics

 

2023  
Web of Science  
Total Cites
WoS
674
Journal Impact Factor 3.3
Rank by Impact Factor

Q2

Impact Factor
without
Journal Self Cites
3.1
5 Year
Impact Factor
3.2
Scimago  
Scimago
H-index
15
Scimago
Journal Rank
0.601
Scimago Quartile Score Microbiology (medical) (Q2)
Microbiology (Q3)
Immunology and Allergy (Q3)
Immunology (Q3)
Scopus  
Scopus
Cite Score
5.0
Scopus
CIte Score Rank
Microbiology (medical) Q2
Scopus
SNIP
0.832

 

European Journal of Microbiology and Immunology
Publication Model Gold Open Access
Online only
Submission Fee none
Article Processing Charge 900 EUR/article
Regional discounts on country of the funding agency World Bank Lower-middle-income economies: 50%
World Bank Low-income economies: 100%
Further Discounts Editorial Board / Advisory Board members: 50%
Corresponding authors, affiliated to an EISZ member institution subscribing to the journal package of Akadémiai Kiadó: 100%
Subscription Information Gold Open Access
Purchase per Title  

European Journal of Microbiology and Immunology
Language English
Size A4
Year of
Foundation
2011
Volumes
per Year
1
Issues
per Year
4
Founder Akadémiai Kiadó
Founder's
Address
H-1117 Budapest, Hungary 1516 Budapest, PO Box 245.
Publisher Akadémiai Kiadó
Publisher's
Address
H-1117 Budapest, Hungary 1516 Budapest, PO Box 245.
Responsible
Publisher
Chief Executive Officer, Akadémiai Kiadó
ISSN 2062-509X (Print)
ISSN 2062-8633 (Online)

Monthly Content Usage

Abstract Views Full Text Views PDF Downloads
Oct 2024 0 0 0
Nov 2024 0 0 0
Dec 2024 0 0 0
Jan 2025 0 0 0
Feb 2025 0 14947 162
Mar 2025 0 15904 86
Apr 2025 0 0 0