SAA小鼠模型中T细胞分化及巨噬细胞亚型的研究*

孙静, 方芳, 谭帅, 孙婉玲

中国生物工程杂志 ›› 2023, Vol. 43 ›› Issue (12) : 145-152.

PDF(2947 KB)
PDF(2947 KB)
中国生物工程杂志 ›› 2023, Vol. 43 ›› Issue (12) : 145-152. DOI: 10.13523/j.cb.2310056
研究报告

SAA小鼠模型中T细胞分化及巨噬细胞亚型的研究*

作者信息 +

T Cell Differentiation and Macrophage Polarization in the Aplastic Anemia Mouse Model

Author information +
文章历史 +

摘要

目的:探讨重型再生障碍性贫血(severe aplastic anemia,SAA)小鼠模型中淋巴细胞功能亚群和巨噬细胞亚型的特点。方法:B6D2F1小鼠经过全身照射(total body irradation,TBI)后,接受C57BL/6小鼠的淋巴细胞输注,建立SAA小鼠模型;以仅接受TBI的B6D2F1小鼠为对照组。在造模成功后收集两组小鼠外周血、淋巴结、脾脏和骨髓,通过流式细胞术检测小鼠T淋巴细胞亚群及CD4+和CD8+细胞表型分化情况;流式细胞术和免疫组化观察小鼠骨髓中巨噬细胞比例及其极化状态。结果:与TBI对照组相比,SAA组小鼠在造模的第12~13天出现严重骨髓衰竭,表现为骨髓造血面积明显减少,外周血白细胞、血红蛋白和血小板均显著下降。SAA组小鼠淋巴结、脾脏和外周血中CD8+淋巴细胞比例升高,CD4+/CD8+淋巴细胞比值显著下降。SAA小鼠的CD4+和CD8+淋巴细胞中,CD44+CD62L-效应记忆T细胞增多,而CD44-CD62L+的初始 T细胞和CD44+CD62L+中央记忆T细胞比例显著下降。SAA小鼠骨髓中CD11b+F4/80+巨噬细胞增多,且巨噬细胞以CD86-cCD206+和iNOS-cCD206+的M2型巨噬细胞为主。结论:SAA小鼠模型中,CD4+/CD8+淋巴细胞比值显著下降,淋巴细胞主要为表达CD44+CD62L-的效应记忆T细胞;骨髓巨噬细胞增多,且以M2型巨噬细胞为主。

Abstract

Objective: To explore the changes of T cell subsets and macrophage subtypes in severe aplastic anemia (SAA) mouse models. Methods: To induce the SAA mouse models, the B6D2F1 mice first received a 5 5 Gy total body irradiation (TBI) and then were infused with C57BL/6 lymph node cells at 4×106/mouse via a tail vein injection, while the B6D2F1 mice receiving TBI alone were used as the control group. The mice were euthanized on day 12-13, and their peripheral blood, lymph nodes, spleen, and bone marrow were collected for analysis. T cell subsets and phenotypic differentiation of CD4+ and CD8+ cells were detected by flow cytometry, and the proportion and polarization of macrophages in bone marrow were detected by flow cytometry and immunohistochemistry. Results: Relative to control mice that received 5 Gy TBI without lymph node cells infusion, SAA mouse models exhibited severe bone marrow failure characterized by a significant reduction in the hematopoietic area of the sternum and decreased levels of white blood cells, hemoglobin, and platelets in peripheral blood 12-13 days post-radiation. In the SAA group, the frequencies of CD8+ T cells increased and the ratio of CD4+/CD8+ lymphocytes decreased significantly in lymph nodes, spleen, and peripheral blood. In CD4+ and CD8+ T cells of SAA mouse models, the percentage of CD44+CD62L- effector memory T cells increased, while the proportion of Naïve T cells and CD44+CD62L+ central memory T cells decreased significantly. CD11b+F4/80+ macrophages increased in the bone marrow of SAA mouse models, and most macrophages were CD86- cCD206+ and iNOS- cCD206+ M2 macrophages. Conclusions: In the SAA mouse models, the ratio of CD4+/CD8+ lymphocytes is significantly decreased, and the lymphocytes are mainly CD44+CD62L- effector memory T cells. Bone marrow macrophages increased and were mainly polarized towards M2 macrophages.

关键词

再生障碍性贫血 / 小鼠模型 / 巨噬细胞极化 / T细胞

Key words

Aplastic anemia / Mouse model / Macrophage polarization / T cells

引用本文

导出引用
孙静, 方芳, 谭帅, . SAA小鼠模型中T细胞分化及巨噬细胞亚型的研究*[J]. 中国生物工程杂志, 2023, 43(12): 145-152 https://doi.org/10.13523/j.cb.2310056
Jing SUN, Fang FANG, Shuai TAN, et al. T Cell Differentiation and Macrophage Polarization in the Aplastic Anemia Mouse Model[J]. China Biotechnology, 2023, 43(12): 145-152 https://doi.org/10.13523/j.cb.2310056
中图分类号: Q343   
获得性再生障碍性贫血(aplastic anemia,AA)是一种免疫介导为主要发病机制的骨髓衰竭性疾病,特征性表现为骨髓增生低下和全血细胞减少[1-2],其中异常活化T细胞攻击造血干细胞导致骨髓破坏是造血衰竭的主要原因之一。但关于T细胞异常活化的机制尚有待进一步研究。
巨噬细胞是机体固有免疫的重要组成,可以分为促炎的M1型巨噬细胞和抗炎的M2型巨噬细胞。近年来,许多研究揭示了不同巨噬细胞亚型,即巨噬细胞的不同极化状态对各类疾病的不同影响[3-6]。有研究发现在AA发生发展中巨噬细胞具有重要作用[7],但对于AA中巨噬细胞的极化状态及其作用仍待进一步探索。临床上,AA患者异质性强,且骨髓衰竭状态下难以采集足够数量的细胞标本供研究,因此通过免疫性骨髓衰竭小鼠模型进行相关研究,成为AA研究的重要手段[7]
我们通过小鼠模型,对重型再生障碍性贫血(severe aplastic anemia,SAA)中淋巴细胞的功能亚群及巨噬细胞极化状态进行初步探索,以期进一步了解SAA发病机制。

1 材料与方法

1.1 实验试剂与仪器

红细胞裂解液购自美国eBioscience公司。D-hanks购自北京索莱宝科技有限公司。FcR阻断剂CD16/32、CD3-APC、CD19-APC、CD49b-APC、Ly6G-APC、F4/80-PE/DazzleTM594、CD11b-FITC、CD86-PE/Cy7、CD206-PE、iNOS-PE、CD206-PE/Cy7、7AAD-PerCP、CD4-PE/DazzleTM594、CD8-PE/Cy7、CD44-FITC、CD62L-PE等流式抗体购自美国Biolegend公司。CD45-PerCP、固定/破膜试剂盒、FACS流式细胞仪购自美国BD公司。免疫组化抗体CD68、F4/80、CD86、CD206、CD163、HRP标记的山羊抗兔IgG购自武汉赛维尔生物科技有限公司。

1.2 实验动物

杂交一代B6D2F1小鼠由C57BL/6N和DBA/2小鼠杂交,雄性,8~12周,作为SAA小鼠模型受者。C57BL/6N小鼠,雄性,8~12周,作为淋巴细胞供者。所有小鼠由北京维通利华实验动物技术有限公司提供,均饲养于清洁级标准环境中。本研究经首都医科大学宣武医院实验动物福利伦理委员会批准(审批号:XW-20210531-1)。

1.3 SAA小鼠模型的建立

将受者B6D2F1小鼠随机分为全身辐照(total body irradiation, TBI)组和SAA组。采集C57BL/6N小鼠颈部、腋下、肠系膜及腹股沟淋巴结,通过研磨、70 μm细胞滤器过滤,制成淋巴细胞悬液。TBI组和SAA组B6D2F1小鼠均接受5.0 Gy TBI,SAA组小鼠在TBI 4 h后经尾静脉注射4×106个C57BL/6N小鼠的淋巴细胞。第12~13天收集两组小鼠标本。

1.4 外周血常规检测

取TBI组和SAA组小鼠尾静脉血,乙二胺四乙酸抗凝,通过全自动血细胞计数器检测白细胞(white blood cell, WBC)、血红蛋白(hemoglobin, HGB)和血小板(platelet, PLT)水平。

1.5 流式标本采集

小鼠麻醉后经腹主动脉取外周血,乙二胺四乙酸抗凝管收集。颈椎离断处死小鼠,取双侧胫骨、股骨,用预冷的D-hanks缓冲液冲洗骨髓腔直至骨头发白,将收集的缓冲液吹匀、过滤、离心制备成骨髓单细胞悬液。取小鼠脾脏和淋巴结,经研磨过滤制备成单细胞悬液。其中外周血、骨髓和脾脏需经红细胞裂解液处理。最终调整细胞浓度为107个/ mL,待行流式细胞术检测。

1.6 流式细胞术

取100 μL外周血、淋巴结、脾脏细胞悬液,加入CD16/32抗体进行Fc受体阻断后,加入荧光标记抗CD3、CD4、CD8、CD44、CD62L抗体,振荡混匀于4℃避光孵育30 min;D-hanks缓冲液洗2遍,重悬细胞;加入7AAD,流式细胞仪上机检测淋巴细胞功能状态。取100 μL骨髓细胞悬液,加入CD16/32抗体进行Fc受体阻断后,加入荧光标记抗CD45、CD3、CD19、CD49b、Ly6G、CD11b、F4/80、CD86抗体;振荡混匀,4℃避光孵育30 min;加入100 μL固定/破膜试剂A液4℃避光处理5 min固定细胞,D-hanks缓冲液洗一遍;加入50 μL破膜剂B液及抗iNOS和CD206抗体;振荡混匀,4℃避光孵育30 min,D-hanks缓冲液洗2遍,重悬细胞,流式细胞仪上机检测骨髓巨噬细胞亚型。

1.7 胸骨免疫组化和病理染色

分离小鼠胸骨,置于4%多聚甲醛的离心管中固定保存。胸骨脱钙后石蜡包埋,切片脱蜡至水、抗原修复后,3%牛血清白蛋白室温封闭30 min,滴加CD68、F4/80、CD86、CD206、CD163一抗,切片平放于湿盒内4℃孵育过夜,磷酸盐缓冲液洗涤3遍后,二抗室温孵育50 min,二氨基联苯胺显色,苏木素复染细胞核后脱水封片;病理染色用苏木精-伊红(hematoxylin-eosin, HE)。光学显微镜下观察两组骨髓组织学特点。

1.8 统计学方法

采用GraphPad Prism 8.0 软件进行绘图与统计学分析,所有数据以均数±标准差(mean±SD)表示,采用Unpaired t-test或One-way ANOVA分析两组数据的显著性。P<0.05为差异有统计学意义,P值用标准缩写格式表示,即* P<0.05、** P<0.01、*** P<0.001。

2 结果

2.1 SAA小鼠模型的建立与验证

淋巴细胞输注后第12~13天,SAA组小鼠出现体重减轻、毛发变灰、弓背、活动减少,甚至死亡等表现;而TBI组小鼠体重无明显变化,生存状态良好(图1A)。小鼠胸骨骨髓组织学显示,TBI组小鼠造血基本恢复,而SAA组小鼠的骨髓造血面积明显减少(图1B),符合SAA小鼠骨髓衰竭的组织病理改变特点。小鼠全血细胞计数结果显示:与TBI组相比,SAA组小鼠外周血中的WBC、HGB和PLT均显著下降(图1C)。上述结果显示SAA小鼠造模成功。
图1 SAA小鼠模型的体重与骨髓造血变化

Fig.1 The weight and bone marrow hematopoietic changes in SAA mouse models

A.Weights of SAA mouse models at 0 day and 12-13 days B. Morphological changes of bone marrow (200× magnification) C. Peripheral blood cell count in mice

Full size|PPT slide

2.2 SAA小鼠T淋巴细胞亚群

造模成功后,取小鼠淋巴结、脾脏和外周血,通过流式细胞术检测T淋巴细胞亚群(图2A)。结果显示,与TBI组小鼠相比,SAA组小鼠淋巴结(图2B)、脾脏(图2C)、外周血(图2D)中CD4+/CD8+淋巴细胞比值均显著下降。即SAA小鼠模型中,CD4+淋巴细胞比例下降,CD8+淋巴细胞比例增加,该结果符合AA患者的临床特征。
图2 SAA小鼠模型中CD4+/CD8+淋巴细胞亚群的变化

Fig.2 Changes of CD4+/CD8+ lymphocyte subsets in SAA mouse models

A. Mice were induced to develop SAA, and then their lymph nodes, spleens, and peripheral blood were examined at 12-13 days post-radiation. Expression of CD4 and CD8 in CD3+ T cells of SAA mice models were analyzed by flow cytometry B. The ratio of CD4+ T cells to CD8+ T cells subsets in lymph nodes of SAA mouse models C. The ratio of CD4+ T cells to CD8+ T cells subsets in spleen of SAA mouse models D. The ratio of CD4+ T cells to CD8+ T cells subsets in peripheral blood of SAA mouse models

Full size|PPT slide

2.3 SAA小鼠T细胞功能亚群变化

通过小鼠T细胞表达CD44和CD62L的不同,可将CD4+和CD8+淋巴细胞划分为不同的功能亚群,即CD44-CD62L+初始T细胞(naïve T cell,Tn)、CD44+CD62L+中央记忆T细胞(central memory T cell, Tcm)、CD44+CD62L-效应记忆T细胞(effector memory T cell, Tem)和CD44-CD62L-终末分化效应记忆T细胞(effector memory RA T cell, Temra)[8-9],从而反映T细胞的活化状态。
与TBI组小鼠相比,SAA组小鼠的淋巴结、脾脏和外周血中,无论CD4+T细胞亚群(图3A、C)还是CD8+T细胞亚群(图3B、D),CD44-CD62L+的Tn细胞比例和CD44+CD62L+ 的Tcm细胞比例均显著下降,而CD44+CD62L-的Tem细胞显著增多。
图3 SAA小鼠模型CD4+和CD8+T细胞中CD44、CD62L的表达

Fig.3 Expression of CD44 and CD62L on CD4+ and CD8+ T lymphocytes in SAA mouse models

A. Flow cytometry plots representing CD44 and CD62L strategy for CD4+ T cells in lymph nodes, spleen, and peripheral blood B. Flow cytometry plots representing CD44 and CD62L strategy for CD8+ T cells. Numbers on plots represent the percent of cells within the gated region C. Bar graphs summarize the data of the CD4+ lymphocyte subset D. Bar graphs summarize the data of the CD8+ lymphocyte subset

Full size|PPT slide

2.4 SAA小鼠模型骨髓中巨噬细胞增多

造模成功后,采用流式细胞术检测骨髓巨噬细胞比例。流式设门策略为:首先设出CD45+细胞群,然后设出CD3-CD19-CD49b-Ly6G-细胞群,从而去除了T、B、NK细胞及中性粒细胞,最后设出F4/80+CD11b+的细胞即为巨噬细胞(图4A)。结果显示,与TBI组相比,SAA组小鼠骨髓巨噬细胞在总骨髓细胞中的比例显著增加(图4B)。
图4 SAA小鼠模型骨髓中巨噬细胞比例

Fig.4 Percentage of macrophages in the bone marrow of SAA mouse models

A. The gating strategy to identify macrophages in bone marrow via flow cytometry B. Bar graphs summarize the data C. The immunohistochemistry staining assay of CD68 and F4/80 in the bone marrow of SAA mouse models (200× magnification)

Full size|PPT slide

小鼠胸骨骨髓免疫组化的结果显示,SAA组小鼠胸骨骨髓中表达巨噬细胞标志物CD68和F4/80的细胞增多(图4C),与上述流式细胞术检测结果一致。

2.5 SAA小鼠模型中骨髓巨噬细胞以M2型为主

为了进一步检测骨髓巨噬细胞的极化状态,分别采用CD86联合cCD206、iNOS联合cCD206两种组合,使用流式细胞术检测巨噬细胞表型。结果显示:SAA组小鼠骨髓巨噬细胞主要以CD86-cCD206+和iNOS-cCD206+的M2型为主,而表达CD86+cCD206-和iNOS+cCD206-的M1型巨噬细胞比例在TBI和SAA两组间无显著差异(图5A、B)。
图5 SAA小鼠模型骨髓中巨噬细胞极化状态

Fig.5 The distribution of macrophage subtypes in the bone marrow of SAA mouse models

A. Macrophage subtypes in CD11b+F4/80+ population were detected by flow cytometry. CD86 and iNOS were used as M1 macrophage markers, and cCD206 was used as M2 macrophage marker B. Bar graphs summarize the data C. The immunohistochemistry staining assay of CD86, CD206, and CD163 in the bone marrow of SAA mouse models (200× magnification)

Full size|PPT slide

小鼠胸骨骨髓免疫组化的结果显示,SAA组小鼠胸骨骨髓中,M1型巨噬细胞标志物CD86的表达量略高于TBI组; M2型巨噬细胞标志物CD206、CD163的表达量明显高于TBI组(图5C)。
由此可见,M1型和M2型巨噬细胞比例在SAA小鼠模型中均升高,M2型巨噬细胞比例更高,SAA小鼠模型骨髓巨噬细胞主要向M2型极化。

3 讨论

AA是一种骨髓衰竭性综合征,特征性表现为骨髓增生低下和全血细胞减少,临床表现为贫血、出血和感染[10]。越来越多的研究显示,获得性再生障碍性贫血的发病机制主要是由自身活化T细胞介导的造血干细胞破坏而引起的造血衰竭,体内多种免疫细胞及细胞因子均参与了自身T细胞的活化过程及造血干细胞的损伤过程[1]。巨噬细胞在不同微环境刺激下可以极化为不同的亚型:M1型,即经典活化的巨噬细胞;M2型,即旁路活化巨噬细胞[11]。本研究通过建立免疫介导的SAA小鼠模型,探索SAA小鼠模型中T细胞功能亚群及巨噬细胞亚型的构成变化,了解T细胞与巨噬细胞在AA发病中的病理作用。
既往研究显示,SAA小鼠模型会表现出弓背、体重减轻、活动减少等状态,出现外周血三系减少和骨髓造血功能障碍[12],本实验结果与既往研究一致,表明成功建立出稳定的SAA小鼠模型。在SAA发病机制中,T细胞是重要一员,T细胞中有两种重要的亚群,CD3+CD4+辅助性T细胞和CD3+CD8+细胞毒性T细胞。本实验结果显示SAA小鼠模型外周血、淋巴结和脾脏中CD4+/CD8+淋巴细胞值下降,该结果与既往研究中SAA患者CD8+T细胞比例升高,CD4+T细胞比例下降[13]的结果一致。进一步证明TBI联合淋巴细胞输注的SAA小鼠模型能够很好模拟临床SAA的发生。
CD44是一种糖蛋白,通过与细胞-细胞和细胞-基质的相互作用,参与细胞增殖、黏附、迁移、造血和淋巴细胞活化等重要生理过程[14]。CD62L,也称为L-选择素,是一种重要的细胞黏附分子,在淋巴细胞向淋巴结运输、归巢中起重要作用[15]。T细胞可以根据CD44和CD62L划分为不同功能的细胞亚群,Tn细胞表达高水平的CD62L,CD44表达较低。Tn细胞接受抗原刺激活化后,可以增殖分化为效应细胞,逐渐高表达CD44并失去CD62L[16]。既往有研究发现SAA患者中CD8+Tem(CD3+CD45RA-CCR7-)、Tcm(CD3+CD45RA-CCR7+)比例升高[17]。本研究通过流式细胞术分析了淋巴结、脾脏和外周血中CD4+和CD8+T细胞亚群中的功能亚群,结果显示在SAA小鼠模型中,无论是CD4+T细胞还是CD8+T细胞亚群,T细胞都以CD44-CD62L+的Tem细胞为主。因此,我们推测淋巴细胞功能亚群的分布失衡、增多的Tem细胞可能就是导致SAA骨髓衰竭的原因。
已有研究提示巨噬细胞在AA发病机制中具有重要作用,清除小鼠体内的巨噬细胞能够延缓SAA的发生发展[7,18-19]。本实验发现在SAA小鼠模型骨髓中巨噬细胞比例显著增多,我们推测骨髓中巨噬细胞增多可能与淋巴细胞异常活化导致骨髓衰竭的发病过程密切相关。
巨噬细胞有不同的亚型,M1型巨噬细胞一般由脂多糖和γ干扰素(interferon-γ,IFN-γ)诱导产生,可以通过分泌促炎因子如肿瘤坏死因子-α、白细胞介素-1β(interleukin-1β, IL-1β)、IL-6、IL-12和IL-23等参与炎症反应;M2型巨噬细胞由IL-4和IL-13等诱导产生,可以通过分泌IL-10等抗炎因子发挥抑制炎症作用,还具有促进组织修复的功能[3]。本研究发现在骨髓中,巨噬细胞都主要向cCD206+M2型巨噬细胞极化。在SAA患者中IFN-γ和肿瘤坏死因子-α细胞因子水平升高[20-21],我们原本推测在SAA小鼠模型中,巨噬细胞会被IFN-γ诱导为M1型巨噬细胞,但本实验结果却显示完全相反。人体是一个精密复杂的系统,各细胞之间及细胞因子的作用错综复杂,我们大胆猜想M2型巨噬细胞是否作为一种负反馈而存在,SAA小鼠模型体内为了应对异常活化的T细胞导致免疫激活状态,增加M2型巨噬细胞从而抑制炎症反应;或M2型巨噬细胞也许通过其他某种机制发挥致病作用。
综上所述,SAA小鼠模型的外周血、淋巴结和脾脏中,T细胞主要是Tem细胞;骨髓中巨噬细胞增多,且以M2型巨噬细胞为主。该结果尚需在SAA患者中加以验证,同时M2型巨噬细胞在SAA发病机制中的具体作用有待进一步研究。

参考文献

[1]
Wang L, Liu H. Pathogenesis of aplastic anemia. Hematology (Amsterdam, Netherlands), 2019, 24(1): 559-566.
[2]
Shallis R M, Ahmad R, Zeidan A M. Aplastic anemia: Etiology, molecular pathogenesis, and emerging concepts. European Journal of Haematology, 2018, 101(6): 711-720.
Aplastic anemia (AA) is rare disorder of bone marrow failure which if severe and not appropriately treated is highly fatal. AA is characterized by morphologic marrow features, namely hypocellularity, and resultant peripheral cytopenias. The molecular pathogenesis of AA is not fully understood, and a uniform process may not be the culprit across all cases. An antigen-driven and likely autoimmune dysregulated T-cell homeostasis is implicated in the hematopoietic stem cell injury which ultimately founds the pathologic features of the disease. Defective telomerase function and repair may also play a role in some cases as evidenced by recurring mutations in related telomerase complex genes such as TERT and TERC. In addition, recurring mutations in BCOR/BCORL, PIGA, DNMT3A, and ASXL1 as well as cytogenetic abnormalities, namely monosomy 7, trisomy 8, and uniparental disomy of the 6p arm seem to be intimately related to AA pathogenesis. The increased incidence of late clonal disease has also provided clues to accurately describe plausible predispositions to the development of AA. The emergence of newer genomic sequencing and other techniques is incrementally improving the understanding of the pathogenic mechanisms of AA, the detection of the disease, and ultimately offers the potential to improve patient outcomes. In this comprehensive review, we discuss the current understanding of the immunobiology, molecular pathogenesis, and future directions of such for AA.© 2018 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.
[3]
Funes S C, Rios M, Escobar-Vera J, et al. Implications of macrophage polarization in autoimmunity. Immunology, 2018, 154(2): 186-195.
Macrophages are extremely heterogeneous and plastic cells with an important role not only in physiological conditions, but also during inflammation (both for initiation and resolution). In the early 1990s, two different phenotypes of macrophages were described: one of them called classically activated (or inflammatory) macrophages (M1) and the other alternatively activated (or wound-healing) macrophages (M2). Currently, it is known that functional polarization of macrophages into only two groups is an over-simplified description of macrophage heterogeneity and plasticity; indeed, it is necessary to consider a continuum of functional states. Overall, the current available data indicate that macrophage polarization is a multifactorial process in which a huge number of factors can be involved producing different activation scenarios. Once a macrophage adopts a phenotype, it still retains the ability to continue changing in response to new environmental influences. The reversibility of polarization has a critical therapeutic value, especially in diseases in which an M1/M2 imbalance plays a pathogenic role. In this review, we assess the high plasticity of macrophages and their potential to be exploited to reduce chronic/detrimental inflammation. On the whole, the evidence detailed in this review underscores macrophage polarization as a target of interest for immunotherapy.© 2018 John Wiley & Sons Ltd.
[4]
Solinas G, Germano G, Mantovani A, et al. Tumor-associated macrophages (TAM) as major players of the cancer-related inflammation. Journal of Leukocyte Biology, 2009, 86(5): 1065-1073.
The microenvironment of solid tumors is characterized by a reactive stroma with an abundance of inflammatory mediators and leukocytes, dysregulated vessels and proteolytic enzymes. TAM, major players in the connection between inflammation and cancer, summarize a number of functions (e.g., promotion of tumor cell proliferation and angiogenesis, incessant matrix turnover, repression of adaptive immunity), which ultimately have an important impact on disease progression. Thus, together with other myeloid-related cells present at the tumor site (Tie2 macrophages and MDSCs), TAM represent an attractive target of novel biological therapies of tumors.
[5]
Wang C, Ma C, Gong L H, et al. Macrophage polarization and its role in liver disease. Frontiers in Immunology, 2021, 12: 803037.
Macrophages are important immune cells in innate immunity, and have remarkable heterogeneity and polarization. Under pathological conditions, in addition to the resident macrophages, other macrophages are also recruited to the diseased tissues, and polarize to various phenotypes (mainly M1 and M2) under the stimulation of various factors in the microenvironment, thus playing different roles and functions. Liver diseases are hepatic pathological changes caused by a variety of pathogenic factors (viruses, alcohol, drugs, etc.), including acute liver injury, viral hepatitis, alcoholic liver disease, metabolic-associated fatty liver disease, liver fibrosis, and hepatocellular carcinoma. Recent studies have shown that macrophage polarization plays an important role in the initiation and development of liver diseases. However, because both macrophage polarization and the pathogenesis of liver diseases are complex, the role and mechanism of macrophage polarization in liver diseases need to be further clarified. Therefore, the origin of hepatic macrophages, and the phenotypes and mechanisms of macrophage polarization are reviewed first in this paper. It is found that macrophage polarization involves several molecular mechanisms, mainly including TLR4/NF-κB, JAK/STATs, TGF-β/Smads, PPARγ, Notch, and miRNA signaling pathways. In addition, this paper also expounds the role and mechanism of macrophage polarization in various liver diseases, which aims to provide references for further research of macrophage polarization in liver diseases, contributing to the therapeutic strategy of ameliorating liver diseases by modulating macrophage polarization.
[6]
Wang L X, Zhang S X, Wu H J, et al. M2b macrophage polarization and its roles in diseases. Journal of Leukocyte Biology, 2019, 106(2): 345-358.
Macrophages play an important role in a wide variety of physiologic and pathologic processes. Plasticity and functional polarization are hallmarks of macrophages. Macrophages commonly exist in two distinct subsets: classically activated macrophages (M1) and alternatively activated macrophages (M2). M2b, a subtype of M2 macrophages, has attracted increasing attention over the past decade due to its strong immune-regulated and anti-inflammatory effects. A wide variety of stimuli and multiple factors modulate M2b macrophage polarization in vitro and in vivo. M2b macrophages possess both protective and pathogenic roles in various diseases. Understanding the mechanisms of M2b macrophage activation and the modulation of their polarization might provide a great perspective for the design of novel therapeutic strategies. The purpose of this review is to discuss current knowledge of M2b macrophage polarization, the roles of M2b macrophages in a variety of diseases and the stimuli to modulate M2b macrophage polarization.©2018 The Authors. Society for Leukocyte Biology Published by Wiley Periodicals, Inc.
[7]
Sun W L, Wu Z J, Lin Z H, et al. Macrophage TNF-α licenses donor T cells in murine bone marrow failure and can be implicated in human aplastic anemia. Blood, 2018, 132(26): 2730-2743.
Interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α) have been implicated historically in the immune pathophysiology of aplastic anemia (AA) and other bone marrow (BM) failure syndromes. We recently defined the essential roles of IFN-γ produced by donor T cells and the IFN-γ receptor in the host in murine immune-mediated BM failure models. TNF-α has been assumed to function similarly to IFN-γ. We used our murine models and mice genetically deficient in TNF-α or TNF-α receptors (TNF-αRs) to establish an analogous mechanism. Unexpectedly, infusion of TNF-α donor lymph node (LN) cells into CByB6F1 recipients or injection of FVB LN cells into TNF-αR recipients both induced BM failure, with concurrent marked increases in plasma IFN-γ and TNF-α levels. Surprisingly, in TNF-α recipients, BM damage was attenuated, suggesting that TNF-α of host origin was essential for immune destruction of hematopoiesis. Depletion of host macrophages before LN injection reduced T-cell IFN-γ levels and reduced BM damage, whereas injection of recombinant TNF-α into FVB-LN cell-infused TNF-α recipients increased T-cell IFN-γ expression and accelerated BM damage. Furthermore, infusion of TNF-αR donor LN cells into CByB6F1 recipients reduced BM T-cell infiltration, suppressed T-cell IFN-γ production, and alleviated BM destruction. Thus, TNF-α from host macrophages and TNF-αR expressed on donor effector T cells were critical in the pathogenesis of murine immune-mediated BM failure, acting by modulation of IFN-γ secretion. In AA patients, TNF-α-producing macrophages in the BM were more frequent than in healthy controls, suggesting the involvement of this cytokine and these cells in human disease.
[8]
Larbi A, Fulop T. From “truly Naïve” to “exhausted senescent” T cells: when markers predict functionality. Cytometry Part A, 2014, 85(1): 25-35.
The study of T cell biology has been accelerated by substantial progress at the technological level, particularly through the continuing advancement of flow cytometry. The conventional approach of observing T cells as either T helper or T cytotoxic is overly simplistic and does not allow investigators to clearly identify immune mechanisms or alterations in physiological processes that impact on clinical outcomes. The complexity of T cell sub-populations, as we understand them today, combined with the immunological and functional diversity of these subsets represent significant complications for the study of T cell biology. In this article, we review the use of classical markers in delineating T cell sub-populations, from "truly naïve" T cells (recent thymic emigrants with no proliferative history) to "exhausted senescent" T cells (poorly proliferative cells that display severe functional abnormalities) wherein the different phenotypes of these populations reflect their disparate functionalities. In addition, since persistent infections and chronological aging have been shown to be associated with significant alterations in human T cell distribution and function, we also discuss age-associated and cytomegalovirus-driven alterations in the expression of key subset markers.© 2013 International Society for Advancement of Cytometry.
[9]
Natalini A, Simonetti S, Favaretto G, et al. OMIP-079: cell cycle of CD4+ and CD8+ Naïve/memory T cell subsets, and of Treg cells from mouse spleen. Cytometry Part A, 2021, 99(12): 1171-1175.
A multicolor flow cytometry panel was designed and optimized to define the following nine mouse T cell subsets: Treg (CD3 CD4 CD8 FoxP3 ), CD4 T naïve (CD3 CD4 CD8 FoxP3 CD44 CD62L ), CD4 T central memory (CD3 CD4 CD8 FoxP3 CD44 CD62L ), CD4 T effector memory (CD3 CD4 CD8 FoxP3 CD44 CD62L ), CD4 T EMRA (CD3 CD4 CD8 FoxP3 CD44 CD62L ), CD8 T naïve (CD3 CD8 CD4 CD44 CD62L ), CD8 T central memory (CD3 CD8 CD4 CD44 CD62L ), CD8 T effector memory (CD3 CD8 CD4 CD44 CD62L ), and CD8 T EMRA (CD3 CD8 CD4 CD44 CD62L ). In each T cell subset, a dual staining for Ki-67 expression and DNA content was employed to distinguish the following cell cycle phases: G (Ki67, with 2n DNA), G (Ki67, with 2n DNA), and S-G /M (Ki67, with 2n < DNA ≤ 4n). This panel was established for the analysis of mouse (C57BL/6J) spleen.© 2021 The Authors. Cytometry Part A published by Wiley Periodicals LLC. on behalf of International Society for Advancement of Cytometry.
[10]
Brodsky R A, Jones R J. Aplastic anaemia. The Lancet, 2005, 365(9471): 1647-1656.
[11]
Orecchioni M, Ghosheh Y, Pramod A B, et al. Macrophage polarization: different gene signatures in M1(LPS+) vs. classically and M2(LPS-) vs. alternatively activated macrophages. Frontiers in Immunology, 2019, 10: 1084.
Macrophages are found in tissues, body cavities, and mucosal surfaces. Most tissue macrophages are seeded in the early embryo before definitive hematopoiesis is established. Others are derived from blood monocytes. The macrophage lineage diversification and plasticity are key aspects of their functionality. Macrophages can also be generated from monocytes and undergo classical (LPS+IFN-γ) or alternative (IL-4) activation., macrophages with different polarization and different activation markers coexist in tissues. Certain mouse strains preferentially promote T-helper-1 (Th1) responses and others Th2 responses. Their macrophages preferentially induce iNOS or arginase and have been called M1 and M2, respectively. In many publications, M1 and classically activated and M2 and alternatively activated are used interchangeably. We tested whether this is justified by comparing the gene lists positively [M1(=LPS+)] or negatively [M2(=LPS-)] correlated with the ratio of and in transcriptomes of LPS-treated peritoneal macrophages with classically (LPS, IFN-γ) vs. alternatively activated (IL-4) bone marrow derived macrophages, both from published datasets. Although there is some overlap between M1(=LPS+) and classically activated (LPS+IFN-γ) and M2(=LPS-) and alternatively activated macrophages, many more genes are regulated in opposite or unrelated ways. Thus, M1(=LPS+) macrophages are not equivalent to classically activated, and M2(=LPS-) macrophages are not equivalent to alternatively activated macrophages. This fundamental discrepancy explains why most surface markers identified on generated macrophages do not translate to the situation. Valid M1/M2 surface markers remain to be discovered.
[12]
Ni R F, Fan L W, Zhang L, et al. A mouse model of irradiation and spleen-thymus lymphocyte infusion induced aplastic anemia. Hematology, 2022, 27(1): 932-945.
The immune-induced aplastic anemia (AA) mouse model has been used for the study of AA. However, there were no uniform conditions for establishing a model and no assessment of immunological homeostasis. Our study aimed to identify the conditions of establishing a model and assess the AA model in immunology and pathology.We induced an AA mouse model by the combination between sublethal irradiation and spleen-thymus lymphocyte infusion. The success of establishing the AA model was identified by blood routine tests and pathology of bone marrow. The frequency of Th17 and Treg cells was measured by flow cytometry. The frequency of CD34+ and CD41+ cells was detected by immunohistochemical technique.IL-6, IL-8, IL-17, TNF-α and IFN-γ were evaluated by ELISA.The Cs sublethal irradiation (5 Gy) and spleen-thymus lymphocyte infusion (5  106) induced the AA mouse model successfully. The AA mice had a long lifetime and manifested pancytopenia and bone marrow failure. The percentage of Th17 cells increased and the percentage of Treg cells decreased distinctly in AA mice. The area of hematopoietic tissues and count of CD34+ cells and CD41+ cells were significantly reduced in AA mice.The level of cytokines, IL-6, IL-8, IL-17, TNF-α and IFN-γ, was increased significantly in peripheral blood and bone marrow.Our data suggest that the improved AA mouse model conforms to the diagnosis standard of AA and simulates the immune internal environment of human AA. The AA mouse model has a longer lifetime and unbalances of Th17/Treg cells caused the destruction of CD34+ cells and CD41+ cells, which was immune-mediated pathogenesis to adapt to long-term research.
[13]
Zhang H F, Huang Z D, Wu X R, et al. Comparison of T lymphocyte subsets in aplastic anemia and hypoplastic myelodysplastic syndromes. Life Sciences, 2017, 189: 71-75.
[14]
Jordan A R, Racine R R, Hennig M J P, et al. The role of CD 44 in disease pathophysiology and targeted treatment. Frontiers in Immunology, 2015, 6: 182.
The cell-surface glycoprotein CD44 is involved in a multitude of important physiological functions including cell proliferation, adhesion, migration, hematopoiesis, and lymphocyte activation. The diverse physiological activity of CD44 is manifested in the pathology of a number of diseases including cancer, arthritis, bacterial and viral infections, interstitial lung disease, vascular disease, and wound healing. This diversity in biological activity is conferred by both a variety of distinct CD44 isoforms generated through complex alternative splicing, posttranslational modifications (e.g., N- and O-glycosylation), interactions with a number of different ligands, and the abundance and spatial distribution of CD44 on the cell surface. The extracellular matrix glycosaminoglycan hyaluronic acid (HA) is the principle ligand of CD44. This review focuses both CD44-hyaluronan dependent and independent CD44 signaling and the role of CD44 HA interaction in various pathophysiologies. The review also discusses recent advances in novel treatment strategies that exploit the CD44 HA interaction either for direct targeting or for drug delivery.
[15]
Ivetic A, Hoskins Green H L, Hart S J. L-selectin: a major regulator of leukocyte adhesion, migration and signaling. Frontiers in Immunology, 2019, 10: 1068.
L-selectin (CD62L) is a type-I transmembrane glycoprotein and cell adhesion molecule that is expressed on most circulating leukocytes. Since its identification in 1983, L-selectin has been extensively characterized as a tethering/rolling receptor. There is now mounting evidence in the literature to suggest that L-selectin plays a role in regulating monocyte protrusion during transendothelial migration (TEM). The N-terminal calcium-dependent (C-type) lectin domain of L-selectin interacts with numerous glycans, including sialyl Lewis X (sLe) for tethering/rolling and proteoglycans for TEM. Although the signals downstream of L-selectin-dependent adhesion are poorly understood, they will invariably involve the short 17 amino acid cytoplasmic tail. In this review we will detail the expression of L-selectin in different immune cell subsets, and its influence on cell behavior. We will list some of the diverse glycans known to support L-selectin-dependent adhesion, within luminal and abluminal regions of the vessel wall. We will describe how each domain within L-selectin contributes to adhesion, migration and signal transduction. A significant focus on the L-selectin cytoplasmic tail and its proposed contribution to signaling via the ezrin-radixin-moesin (ERM) family of proteins will be outlined. Finally, we will discuss how ectodomain shedding of L-selectin during monocyte TEM is essential for the establishment of front-back cell polarity, bestowing emigrated cells the capacity to chemotax toward sites of damage.
[16]
Del Zotto G, Principi E, Antonini F, et al. Comprehensive phenotyping of peripheral blood T lymphocytes in healthy mice. Cytometry Part A: the Journal of the International Society for Analytical Cytology, 2021, 99(3): 243-250.
[17]
Zheng M Y, Liu C Y, Fu R, et al. Abnormal immunomodulatory ability on memory T cells in humans with severe aplastic anemia. International Journal of Clinical and Experimental Pathology, 2015, 8(4): 3659-3669.
Severe aplastic anemia (SAA) is a bone marrow failure disease induced by hyperfunctional autoimmunic Th1 lymphocytes. Memory T cells (TM) are a component of the adaptive immune system. They ensure the host of more aggressive and faster immune response to efficiently eliminate the specific antigens after re-exposure and thus play a key role in T-cell functions. In this study we investigate the quantities and functions of memory T cells in SAA patients before and after immunosuppressive therapy (IST) to further clarify the mechanism of SAA apoptosis of bone marrow hematopoietic cells. Results showed that the percentage of CD4+ effector T cells in peripheral blood and bone marrow lymphocytes was decreased in SAA patients. The ratio of CD4+ memory T lymphocytes to CD8+ memory T subsets (CD4+/CD8+TM) in SAA patients was also lower. The percentage of CD8+ effector T cells in peripheral blood and CD8+ central memory T cells in the bone marrow lymphocytes was significantly higher in newly diagnosed patients. Furthermore, the median expressions of perforin and granzyme B on memory T cells were higher in SAA patients compared to those in normal controls. After IST, the quantities and functions of memory T cells return to normal level. Therefore, we concluded that the abnormal immunomodulatory ability on memory T cells may contribute to the imbalance of Th1/Th2 subsets and thus lead to over-function of T lymphocytes and hematopoiesis failure in SAA.
[18]
McCabe A, Smith J N P, Costello A, et al. Hematopoietic stem cell loss and hematopoietic failure in severe aplastic anemia is driven by macrophages and aberrant podoplanin expression. Haematologica, 2018, 103(9): 1451-1461.
Severe aplastic anemia (SAA) results from profound hematopoietic stem cell loss. T cells and interferon gamma (IFNγ) have long been associated with SAA, yet the underlying mechanisms driving hematopoietic stem cell loss remain unknown. Using a mouse model of SAA, we demonstrate that IFNγ-dependent hematopoietic stem cell loss required macrophages. IFNγ was necessary for bone marrow macrophage persistence, despite loss of other myeloid cells and hematopoietic stem cells. Depleting macrophages or abrogating IFNγ signaling specifically in macrophages did not impair T-cell activation or IFNγ production in the bone marrow but rescued hematopoietic stem cells and reduced mortality. Thus, macrophages are not required for induction of IFNγ in SAA and rather act as sensors of IFNγ. Macrophage depletion rescued thrombocytopenia, increased bone marrow megakaryocytes, preserved platelet-primed stem cells, and increased the platelet-repopulating capacity of transplanted hematopoietic stem cells. In addition to the hematopoietic effects, SAA induced loss of non-hematopoietic stromal populations, including podoplanin-positive stromal cells. However, a subset of podoplanin-positive macrophages was increased during disease, and blockade of podoplanin in mice was sufficient to rescue disease. Our data further our understanding of disease pathogenesis, demonstrating a novel role for macrophages as sensors of IFNγ, thus illustrating an important role for the microenvironment in the pathogenesis of SAA.Copyright© 2018 Ferrata Storti Foundation.
[19]
Seyfried A N, McCabe A, Smith J N P, et al. CCR5 maintains macrophages in the bone marrow and drives hematopoietic failure in a mouse model of severe aplastic anemia. Leukemia, 2021, 35(11): 3139-3151.
Severe aplastic anemia (SAA) is an acquired, T cell-driven bone marrow (BM) failure disease characterized by elevated interferon gamma (IFNγ), loss of hematopoietic stem cells (HSCs), and altered BM microenvironment, including dysfunctional macrophages (MΦs). T lymphocytes are therapeutic targets for treating SAA, however, the underlying mechanisms driving SAA development and how innate immune cells contribute to disease remain poorly understood. In a murine model of SAA, increased beta-chemokines correlated with disease and were partially dependent on IFNγ. IFNγ was required for increased expression of the chemokine receptor CCR5 on MΦs. CCR5 antagonism in murine SAA improved survival, correlating with increased platelets and significantly increased platelet-biased CD41 HSCs. T cells are key drivers of disease, however, T cell-specific CCR5 expression and T cell-derived CCL5 were not necessary for disease. CCR5 antagonism reduced BM MΦs and diminished their expression of Tnf and Ccl5, correlating with reduced frequencies of IFNγ-secreting BM T cells. Mechanistically, CCR5 was intrinsically required for maintaining BM MΦs during SAA. Ccr5 expression was significantly increased in MΦs from aged mice and humans, relative to young counterparts. Our data identify CCR5 signaling as a key axis promoting the development of IFNγ-dependent BM failure, particularly relevant in aging where Ccr5 expression is elevated.
[20]
Su A Y, Yang X Z, Zhang M H, et al. Analysis of correlation of immune T cell subsets, TNF-α, IFN-γ and sFas levels with severity and prognosis in aplastic anemia patients. Journal of Experimental Hematology, 2018, 26(5): 1459-1464.
[21]
Liu B N, Shao Y Y, Liu Z X, et al. Bone marrow plasma cytokine signature profiles in severe aplastic anemia. BioMed Research International, 2020, 2020: 1-11.

基金

*北京市自然科学基金(Z200022)

PDF(2947 KB)

1086

Accesses

0

Citation

Detail

段落导航
相关文章

/