补地参连方调节肠道菌群及肿瘤相关巨噬细胞表型转换发挥抗结直肠癌效应

Study on the Anti-Colorectal Cancer Mechanism of Budi Shenlian Recipe in Regulating Gut Microbiota to Induce TAMs Phenotype Transformation

  • 摘要:
    目的 探究补地参连方对结直肠癌(CRC)小鼠肠道菌群及肿瘤相关巨噬细胞(TAMs)表型转换的调节作用。
    方法 40只雄性BALB/c小鼠分为空白组、模型组、阳性对照(奥沙利铂)组、补地参连方高剂量组、补地参连方低剂量组,每组8只。通过皮下注射CT26细胞构建CRC荷瘤模型。此外,取10只雄性BALB/c小鼠分为补地参连方粪菌移植组(BFMT)以及模型粪菌移植组(MFMT),每组5只,通过四联抗生素混合溶液清除小鼠肠道定植菌,然后通过皮下注射CT26细胞构建伪无菌CRC荷瘤小鼠。收集上述模型组和补地参连方高剂量组的小鼠粪便用于制备粪菌液。BFMT组灌胃补地参连方高剂量治疗后CRC小鼠的粪菌液,MFMT组灌胃模型组CRC小鼠的粪菌液。观察记录小鼠肿瘤体积变化,HE染色评估小鼠肿瘤组织的病理形态学改变,并通过16S rRNA测序分析肠道菌群丰度和结构的变化,流式细胞术和免疫荧光染色评估肿瘤组织中M1/M2型TAMs的比例,ELISA法检测肿瘤组织中TNF-α和IL-10的含量。
    结果 与模型组比较,阳性对照组和补地参连方高、低剂量组小鼠的肿瘤体积均生长缓慢(P < 0.01)。HE染色显示,与模型组比较,阳性对照组和补地参连方各给药组肿瘤组织均出现坏死灶,且病理性核分裂象减少。16S rRNA测序显示,与模型组比较,补地参连方高剂量组的Chao1、ACE无统计学意义,而PCoA分析显示,空白组与模型组之间的菌群落结构明显不同,补地参连方组CRC小鼠的菌群结构更接近空白组,与空白组相比,模型组中MuribaculaceaeMuribaculumAlloprevotellaPrevotellaceae_UCG-001的比例均明显减少,而模型组中Lachnospiraceae_NK4A136_groupBacteroidesHelicobacter的比例均明显增加。而在给予CRC小鼠补地参连方后,可以观察到部分小鼠的群落结构向空白组小鼠的水平恢复。转录测序发现上调基因所涉及的生物学过程(BP)中Negative regulation of macrophage migration最为显著,揭示了补地参连方可以减少巨噬细胞的迁移。此外,与模型组比较,补地参连方组肿瘤组织中TAMs比例明显降低(P < 0.001),M1型TAMs比例则明显升高(P < 0.000 1),M2型TAMs比例明显降低(P < 0.05)。免疫荧光则显示出与流式细胞术相同的趋势。补地参连方组肿瘤组织中TNF-α含量明显升高(P < 0.001),IL-10含量明显降低(P < 0.001)。另一方面,与MFMT比较,BFMT组的肿瘤体积生长缓慢(P < 0.000 1)。HE染色显示BFMT组肿瘤组织内坏死面积增多,细胞排列变稀疏,病理性核分裂象减少。与MFMT组比较,BFMT组肿瘤组织中TAMs细胞比例明显降低(P < 0.01),而BFMT组肿瘤组织中M1型TAMs细胞比例增加(P < 0.000 1), M2型TAMs细胞比例降低(P < 0.01),免疫荧光实验进一步证实了补地参连方粪菌移植可以促进TAMs从M2型向M1型转换,减少肿瘤微环境中的免疫抑制。
    结论 补地参连方可以改善CRC小鼠肠道菌群失调,并且可以通过减少TAMs的肿瘤浸润水平和调节TAMs的表型转换来发挥抗CRC作用,且二者之间可能具有一定相关性。

     

    Abstract:
    Objective To investigate of the regulatory effects of Budi Shenlian Recipe on gut microbiota and the phenotypic transition of tumor-associated macrophages (TAMs) in colorectal cancer (CRC) mice.
    Methods Forty male BALB/c mice were divided into blank control group, model group, positive control group, high-dose Budi Shenlian Recipe group, and low-dose Budi Shenlian Recipe group (n=8). CRC-bearing models were established by subcutaneous injection of CT26 cells. Additionally, 10 male BALB/c mice were divided into the Budi Shenlian Recipe fecal microbiota transplantation (BFMT) group and model fecal microbiota transplantation (MFMT) group (n=5). The gut microbiota of these mice was cleared using a mixed solution of quadruple antibiotics, followed by subcutaneous injection of CT26 cells to construct pseudo-germ-free CRC-bearing mice. Fecal samples from the model group and high-dose Budi Shenlian Recipe group were collected to prepare fecal microbiota solutions. The BFMT group received gavage with fecal microbiota solution from the high-dose Budi Shenlian Recipe group, while the MFMT group received gavage with fecal microbiota solution from the model group. Tumor volume changes were observed and recorded. HE staining was used to assess pathological changes in tumor tissues. 16S sequencing was performed to analyze changes in gut microbiota. Flow cytometry and immunofluorescence staining were used to evaluate the proportion of M1/M2 type TAMs in tumor tissues. ELISA was used to detect differences in TNF-α and IL-10 levels in tumor tissues.
    RESULTS Compared to the model group, the tumor volume of mice in the positive control group, high-dose Budi Shenlian Recipe group, and low-dose Budi Shenlian Recipe group grew more slowly (P < 0.01). HE staining showed necrotic areas in tumor tissues and reduced mitotic figures in the positive control and Budi Shenlian Recipe groups compared to the model group. 16S rRNA sequencing showed no significant differences in Chao1 and ACE indices between the high-dose Budi Shenlian Recipe group and the model group. PCoA analysis indicated a distinct microbial community structure between the blank group and model group, with the microbial structure of CRC mice in the Budi Shenlian Recipe group closer to that of the blank group. Compared to the blank group, the model group showed a significant decrease in the proportion of Muribaculaceae, Muribaculum, Alloprevotella, and Prevotellaceae_UCG-001, and a significant increase in Lachnospiraceae_NK4A136_group, Bacteroides, and Helicobacter. After administering Budi Shenlian Recipe to CRC mice, the community structure of some mice partially reverted to the level of the blank group. Transcriptome sequencing revealed that the most significant biological process (BP) among upregulated genes was the negative regulation of macrophage migration, suggesting that Budi Shenlian Recipe can reduce macrophage migration. Moreover, compared to the model group, the proportion of TAMs cells in the tumor tissues of the Budi Shenlian Recipe group significantly decreased (P < 0.001). Simultaneously, compared to the model group, the proportion of M1 type TAMs in the Budi Shenlian Recipe group significantly increased (P < 0.000 1), while the proportion of M2 type TAMs significantly decreased (P < 0.05). Immunofluorescence analysis showed the same trend as flow cytometry. The content of TNF-α in tumor tissues of the Budi Shenlian Recipe group significantly increased (P < 0.001), and IL-10 content significantly decreased (P < 0.001). Additionally, compared to the MFMT group, the tumor volume in the BFMT group grew more slowly (P < 0.0001). HE staining showed increased necrotic areas, sparser cell arrangement, and reduced pathological mitosis in the BFMT group. Furthermore, compared to the MFMT group, the proportion of TAMs cells in the tumor tissues of the BFMT group significantly decreased (P < 0.01). Compared to the MFMT group, the proportion of M1 type TAMs cells in the BFMT group increased (P < 0.000 1), while the proportion of M2 type TAMs cells decreased (P < 0.01). Immunofluorescence analysis further confirmed that Budi Shenlian Recipe fecal microbiota transplantation can reduce the proportion of TAMs in the tumor tissues of CRC mice and promote the conversion from M2 to M1 type, thereby reducing immune suppression in the tumor microenvironment.
    CONCLUSION Budi Shenlian Recipe can improve gut microbiota dysbiosis in CRC mice and exert anti-CRC effects by reducing tumor infiltration of TAMs and modulating the phenotypic transition of TAMs. There may be a certain correlation between these two effects.

     

/

返回文章
返回