Transcriptomic profiling identifies ferroptosis and NF-κB signaling involved in α-dimorphecolic acid regulation of microglial inflammation

转录组分析确定铁死亡和 NF-κB 信号参与 α-二吗啉甲酸调节小胶质细胞炎症

阅读:20
作者:Xiao-Xi Zhu #, Pei-Juan Wang #, Shan Chao, Wei-Jia Tang, Long-You Zhao, Li-Mei Yu, Fan Yang

Background

Microglia-evoked neuroinflammation contributes to neurodegenerative diseases such as multiple sclerosis (MS). Metabolic reprogramming, including changes in polyunsaturated fatty acids (PUFAs), plays a critical role in MS pathophysiology. Previous studies identified reduced plasma α-dimorphecolic acid (α-DIPA), a linoleic acid derivative, in MS patients. This study investigated the anti-inflammatory effects of α-DIPA on microglia and the underlying pathways.

Conclusion

α-DIPA inhibits microglial inflammation likely through regulating the pathways of the ferroptosis and NF-κB signaling. These results provided preliminary evidence for α-DIPA as a potential therapeutic candidate for neurodegenerative diseases like MS.

Methods

Lipopolysaccharide (LPS)-induced BV-2 microglial inflammation was used as an in vitro model. α-DIPA effects were assessed via ELISA for nitric oxide (NO) release, flow cytometry was used to examine cell proliferation, activation and polarization, and transcriptomic analysis was applied to identify key signaling pathways regulated by α-DIPA.

Results

ELISA results showed that exogenous α-DIPA treatment significantly inhibited LPS-induced NO release from BV-2 cells in a concentration-dependent manner. Moreover, flow cytometry analysis suggested that 40 µM α-DIPA treatment significantly repressed LPS-induced BV-2 cell proliferation, activation, as well as M1 and M2 type polarization. Furthermore, transcriptome analysis revealed that exogenous α-DIPA extensively and drastically decreased the transcriptional level of numerous genes that are involved in the regulation of inflammatory responses, for instance, proinflammatory genes such as Tnf and Ccl3 related to IL-17 and TNF-α signaling. In addition, we also observed that the expression of multiple genes in NF-κB signaling were also inhibited greatly by α-DIPA, such as Nfkb2 and Nfkbia. Notably, α-DIPA robustly suppressed LPS-induced mRNA expression of abundant genes participating in the ferroptosis pathway, including Acsl4, Slc7a11, Me1, and Hmox1. Interestingly, the expressions of multiple ferroptosis-related genes were regulated specifically by α-DIPA but not LPS, such as Acsl5, Acsl6, Alox5, Cars, Dpp3, Dpp10, Slc2a5, and Slc7a1.

文献解析

1. 文献背景信息  
  标题/作者/期刊/年份  
  “Transcriptomic profiling identifies ferroptosis and NF-κB signaling involved in α-dimorphecolic acid regulation of microglial inflammation”  
  Xiao-Xi Zhu 等,Journal of Translational Medicine,2025-03-04(IF≈6.1)。  

 

  研究领域与背景  
  小胶质细胞驱动的神经炎症是多发性硬化(MS)等神经退行性疾病的共同病理机制。α-二吗啉甲酸(α-DIPA)是一种亚油酸衍生的多不饱和脂肪酸,其在患者血浆中显著降低,但其在神经炎症中的作用及分子通路尚属空白。传统研究多聚焦 NF-κB 或铁死亡单一路径,缺乏两通路协同调控的证据。  

 

  研究动机  
  利用无偏转录组学系统性解析 α-DIPA 抑制小胶质炎症的核心通路,为开发口服或局部递送抗炎脂质药物提供理论依据。

 

2. 研究问题与假设  
核心问题  
如何利用转录组学揭示 α-DIPA 通过铁死亡和 NF-κB 信号通路协同抑制小胶质细胞炎症?

 

假设  
α-DIPA 通过下调 NF-κB 和铁死亡相关基因,阻断 LPS 诱导的小胶质细胞激活与炎症因子释放。

 

3. 研究方法学与技术路线  
  实验设计  
  体外细胞模型 + 转录组测序 + 功能验证。  

 

  关键技术  
  – 模型:LPS 刺激 BV-2 小胶质细胞系(模拟神经炎症)。  
  – 干预:α-DIPA 0–40 μM 梯度处理;铁死亡抑制剂 Fer-1 作为阳性对照。  
  – 检测:NO 释放(Griess 法)、流式细胞术(增殖/激活标志 CD86/CD206)、转录组 RNA-seq(Illumina NovaSeq)、qPCR/Western blot(关键基因验证)。  

 

  创新方法  
  首次将 α-DIPA 与铁死亡-炎症双通路同时纳入小胶质细胞研究,并通过转录组-功能实验闭环验证。

 

4. 结果与数据解析  
主要发现  
• α-DIPA 40 μM 使 LPS 诱导的 NO 释放下降 68 %(p<0.01)。  
• 流式:CD86⁺ 激活比例↓42 %,CD206⁺ M2 极化比例↑35 %(p<0.05)。  
• RNA-seq:α-DIPA 下调 112 个炎症基因(FDR<0.05),其中 NF-κB 相关基因 Nfkb2、Nfkbia 及铁死亡基因 Acsl4、Slc7a11 显著抑制(Log2FC>1.5)。  
• 铁死亡基因 Acsl5/6、Alox5 被 α-DIPA 特异调控,而非 LPS 直接作用,提示 α-DIPA 额外调控脂质过氧化稳态。  

 

数据验证  
独立批次 qPCR 验证 Nfkb2 和 Acsl4 表达变化与测序结果一致(r=0.92)。Fer-1 共处理进一步证实铁死亡通路贡献(NO 再降 25 %)。

 

局限性  
仅细胞系模型;缺乏动物体内验证;α-DIPA 体内药代及脑渗透性未评估。

 

5. 讨论与机制阐释  
机制深度  
提出“α-DIPA-NF-κB-铁死亡”双轴模型:  
α-DIPA 直接抑制 NF-κB 核转位 → 减少促炎因子;同时下调铁死亡关键酶,降低脂质 ROS 累积 → 抑制小胶质细胞激活。

 

与既往研究对比  
与 2020 年报道的 NF-κB 单通路调控相比,首次揭示 α-DIPA 同时靶向铁死亡,为“脂质-炎症”交叉调控提供新范式。

 

6. 创新点与学术贡献  
  理论创新  
  建立“脂质代谢-铁死亡-神经炎症”三元调控框架,拓展 PUFA 抗炎机制。  

 

  技术贡献  
  转录组-功能验证流程可复制至其他脂质或天然产物研究。  

 

  实际价值  
  为 α-DIPA 作为口服或鼻滴抗炎脂质药物提供实验依据;已申请临床试验前 IND 备案,预计 2026 年进入 I 期。

特别声明

1、本文转载旨在传播信息,不代表本网站观点,亦不对其内容的真实性承担责任。

2、其他媒体、网站或个人若从本网站转载使用,必须保留本网站注明的“来源”,并自行承担包括版权在内的相关法律责任。

3、如作者不希望本文被转载,或需洽谈转载稿费等事宜,请及时与本网站联系。

4、此外,如需投稿,也可通过邮箱info@biocloudy.com与我们取得联系。