Tumor-associated macrophage (TAM)

肿瘤相关巨噬细胞 (TAM)
  • 文章类型: Journal Article
    背景:肝细胞癌(HCC)的预后受其复杂的分子特征和多变的肿瘤微环境(TME)的限制。在这里,我们专注于阐明母体胚胎亮氨酸拉链激酶(MELK)在肿瘤发生中的功能后果,肝癌的进展和转移,探讨MELK对TME中免疫细胞调控的影响,同时明确相应的信令网络。
    方法:生物信息学分析用于验证MELK对HCC的预后价值。鼠异种移植试验和HCC肺转移小鼠模型证实了MELK在HCC肿瘤发生和转移中的作用。荧光素酶测定,RNA测序,免疫纯化-质谱(IP-MS)和免疫共沉淀(CoIP)用于探索上游调节因子,肝癌MELK的下游必需分子及相应机制。
    结果:我们证实MELK是HCC的可靠预后因素,并确定MELK是促进肿瘤发生的有效候选者。programming,和HCC的转移;MELK的作用取决于上游因子miR-505-3p的靶向调节和与STAT3的相互作用,从而诱导STAT3磷酸化并增加其靶基因CCL2在HCC中的表达。此外,我们证实肿瘤细胞固有的MELK抑制有利于刺激M1巨噬细胞极化,阻碍M2巨噬细胞极化和诱导CD8+T细胞募集,依赖于CCL2表达的改变。重要的是,MELK抑制增强了RT相关的免疫效应,从而与RT协同发挥实质性的抗肿瘤作用。OTS167,一种MELK抑制剂,还被证明可以有效地损害HCC的生长和进展,并与放射疗法(RT)结合使用具有出色的抗肿瘤作用。
    结论:总而言之,我们的研究结果强调了MELK作为分子治疗和联合RT治疗改善HCC抗肿瘤效果的一个有前景的靶点的功能作用.
    BACKGROUND: The outcome of hepatocellular carcinoma (HCC) is limited by its complex molecular characteristics and changeable tumor microenvironment (TME). Here we focused on elucidating the functional consequences of Maternal embryonic leucine zipper kinase (MELK) in the tumorigenesis, progression and metastasis of HCC, and exploring the effect of MELK on immune cell regulation in the TME, meanwhile clarifying the corresponding signaling networks.
    METHODS: Bioinformatic analysis was used to validate the prognostic value of MELK for HCC. Murine xenograft assays and HCC lung metastasis mouse model confirmed the role of MELK in tumorigenesis and metastasis in HCC. Luciferase assays, RNA sequencing, immunopurification-mass spectrometry (IP-MS) and coimmunoprecipitation (CoIP) were applied to explore the upstream regulators, downstream essential molecules and corresponding mechanisms of MELK in HCC.
    RESULTS: We confirmed MELK to be a reliable prognostic factor of HCC and identified MELK as an effective candidate in facilitating the tumorigenesis, progression, and metastasis of HCC; the effects of MELK depended on the targeted regulation of the upstream factor miR-505-3p and interaction with STAT3, which induced STAT3 phosphorylation and increased the expression of its target gene CCL2 in HCC. In addition, we confirmed that tumor cell-intrinsic MELK inhibition is beneficial in stimulating M1 macrophage polarization, hindering M2 macrophage polarization and inducing CD8 + T-cell recruitment, which are dependent on the alteration of CCL2 expression. Importantly, MELK inhibition amplified RT-related immune effects, thereby synergizing with RT to exert substantial antitumor effects. OTS167, an inhibitor of MELK, was also proven to effectively impair the growth and progression of HCC and exert a superior antitumor effect in combination with radiotherapy (RT).
    CONCLUSIONS: Altogether, our findings highlight the functional role of MELK as a promising target in molecular therapy and in the combination of RT therapy to improve antitumor effect for HCC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    泛素化和去泛素化是控制蛋白质稳态的翻译后修饰的重要形式。去泛素化酶(DUB),一个由100多个成员组成的蛋白质超家族,从客户蛋白中去偶联泛素链以调节细胞内稳态。然而,据报道,DUB的失调与几种疾病有关,包括癌症.肿瘤微环境(TME)是一个高度复杂的实体,包含不同的非癌细胞(例如,免疫细胞和基质细胞)和细胞外基质(ECM)。由于TME异质性与肿瘤发生和免疫逃避密切相关,靶向TME成分最近被认为是恢复抗肿瘤免疫力的有吸引力的治疗策略.新兴的研究表明,DUB参与了TME内的免疫调节,包括免疫检查点和免疫细胞浸润和功能的调节,这使得DUB有望用于有效的癌症免疫疗法。然而,DUB在肿瘤及其周围成分之间的串扰中的作用尚未得到全面审查。在这次审查中,我们讨论了DUB参与肿瘤之间的动态相互作用,免疫细胞,和基质细胞,并说明失调的DUB如何促进免疫逃避和促进肿瘤进展。我们还总结了靶向DUB以减轻免疫抑制和抑制肿瘤发生的潜在小分子。最后,我们讨论了DUB靶向治疗癌症的前景和挑战,以及需要进一步研究的几个紧迫问题.
    Ubiquitination and deubiquitination are important forms of posttranslational modification that govern protein homeostasis. Deubiquitinating enzymes (DUBs), a protein superfamily consisting of more than 100 members, deconjugate ubiquitin chains from client proteins to regulate cellular homeostasis. However, the dysregulation of DUBs is reportedly associated with several diseases, including cancer. The tumor microenvironment (TME) is a highly complex entity comprising diverse noncancerous cells (e.g., immune cells and stromal cells) and the extracellular matrix (ECM). Since TME heterogeneity is closely related to tumorigenesis and immune evasion, targeting TME components has recently been considered an attractive therapeutic strategy for restoring antitumor immunity. Emerging studies have revealed the involvement of DUBs in immune modulation within the TME, including the regulation of immune checkpoints and immunocyte infiltration and function, which renders DUBs promising for potent cancer immunotherapy. Nevertheless, the roles of DUBs in the crosstalk between tumors and their surrounding components have not been comprehensively reviewed. In this review, we discuss the involvement of DUBs in the dynamic interplay between tumors, immune cells, and stromal cells and illustrate how dysregulated DUBs facilitate immune evasion and promote tumor progression. We also summarize potential small molecules that target DUBs to alleviate immunosuppression and suppress tumorigenesis. Finally, we discuss the prospects and challenges regarding the targeting of DUBs in cancer immunotherapeutics and several urgent problems that warrant further investigation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:乳酸盐已成为肿瘤微环境中的关键调节因子,包括神经胶质瘤.然而,乳酸如何影响肿瘤细胞和肿瘤相关巨噬细胞(TAMs)之间的通讯的确切机制,神经胶质瘤中最丰富的免疫细胞,仍然知之甚少。本研究旨在阐明肿瘤源性乳酸对TAM的影响,并研究控制TAM介导的神经胶质瘤肿瘤促进的调节途径。
    方法:使用来自TCGA和CGGA的数据集进行生物信息学分析。通过使用UCSC细胞浏览器和单细胞门户分析单细胞RNA-seq数据集。通过CCK8,集落形成评估细胞增殖和迁移率,伤口愈合,和transwell分析。应用蛋白质印迹和免疫荧光染色评估蛋白质表达和细胞分布。RT-PCR和ELISA用于鉴定潜在的分泌因子。通过蛋白质印迹法探索了机制途径,ELISA,shRNA敲除,和特异性抑制剂和活化剂。在体内使用皮下和颅内异种移植肿瘤模型进一步评估途径阻断的效果。
    结果:胶质瘤中LDHA和MCT1的表达升高,并与M2型TAM浸润呈正相关。来自神经胶质瘤细胞的乳酸诱导TAMs向M2亚型极化,随后促进神经胶质瘤细胞增殖,迁移,入侵,和间质转化。GPR65在TAMs上高表达,感觉到TME中的乳酸刺激,通过分泌HMGB1促进胶质瘤细胞恶性进展。TAM上的GPR65通过cAMP/PKA/CREB信号通路响应乳酸刺激而触发HMGB1释放。通过GPR65敲低或HMGB1抑制破坏该反馈回路减轻了体内神经胶质瘤的进展。
    结论:这些发现揭示了TAMs和由乳酸和HMGB1介导的肿瘤细胞之间复杂的相互作用,驱动神经胶质瘤的肿瘤进展。GPR65在胶质瘤中的TAMs上选择性高表达,通过cAMP/PKA/CREB信号通路感知乳酸刺激并促进HMGB1分泌。阻断该反馈回路为GBM提供了有希望的治疗策略。
    BACKGROUND: Lactate has emerged as a critical regulator within the tumor microenvironment, including glioma. However, the precise mechanisms underlying how lactate influences the communication between tumor cells and tumor-associated macrophages (TAMs), the most abundant immune cells in glioma, remain poorly understood. This study aims to elucidate the impact of tumor-derived lactate on TAMs and investigate the regulatory pathways governing TAM-mediated tumor-promotion in glioma.
    METHODS: Bioinformatic analysis was conducted using datasets from TCGA and CGGA. Single-cell RNA-seq datasets were analyzed by using UCSC Cell Browser and Single Cell Portal. Cell proliferation and mobility were evaluated through CCK8, colony formation, wound healing, and transwell assays. Western blot and immunofluorescence staining were applied to assess protein expression and cell distribution. RT-PCR and ELISA were employed to identify the potential secretory factors. Mechanistic pathways were explored by western blotting, ELISA, shRNA knockdown, and specific inhibitors and activators. The effects of pathway blockades were further assessed using subcutaneous and intracranial xenograft tumor models in vivo.
    RESULTS: Elevated expressions of LDHA and MCT1 were observed in glioma and exhibited a positive correlation with M2-type TAM infiltration. Lactate derived from glioma cells induced TAMs towards M2-subtype polarization, subsequently promoting glioma cells proliferation, migration, invasion, and mesenchymal transition. GPR65, highly expressed on TAMs, sensed lactate-stimulation in the TME, fueling glioma cells malignant progression through the secretion of HMGB1. GPR65 on TAMs triggered HMGB1 release in response to lactate stimulation via the cAMP/PKA/CREB signaling pathway. Disrupting this feedback loop by GPR65-knockdown or HMGB1 inhibition mitigated glioma progression in vivo.
    CONCLUSIONS: These findings unveil the intricate interplay between TAMs and tumor cells mediated by lactate and HMGB1, driving tumor progression in glioma. GPR65, selectively highly expressed on TAMs in glioma, sensed lactate stimulation and fostered HMGB1 secretion via the cAMP/PKA/CREB signaling pathway. Blocking this feedback loop presents a promising therapeutic strategy for GBM.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    肿瘤相关巨噬细胞(TAM)是肿瘤微环境(TME)中最丰富的浸润白细胞之一。将TAM从原瘤M2重编程为抗肿瘤M1表型是重塑TME和促进抗肿瘤免疫的有希望的策略;然而,制定有效的战略仍然具有挑战性。这里,构建了一种基因修饰的细菌仿生囊泡(BBV),其表面暴露于纳米组装膜孔结构中的IFN-γ。工程化的IFN-γBBV具有蛋白质和脂质囊泡的纳米级结构,存在丰富的模式相关分子模式(PAMPs),和引入的IFN-γ分子的共刺激。体外,IFN-γBBV将M2巨噬细胞重编程为M1,可能通过NF-κB和JAK-STAT信号通路,释放一氧化氮(NO)和炎症细胞因子IL-1β,IL-6和TNF-α以及增加IL-12和iNOS的表达。在荷瘤小鼠中,IFN-γBBV在肿瘤中表现出靶向富集,并成功地将TAM重编程为M1表型;TME中抗原特异性细胞毒性T淋巴细胞(CTL)的反应得到促进,而免疫抑制性髓源性抑制细胞(MDSC)受到抑制。发现肿瘤生长在TC-1肿瘤和CT26肿瘤中均被显著抑制。结果表明,IFN-γBBV的抗肿瘤作用是巨噬细胞依赖性的。Further,在对单独抗PD-1mAb不敏感的原位4T1乳腺癌模型中,IFN-γBBV对TME的调节与免疫检查点抑制剂对肿瘤生长和转移产生协同作用.总之,IFN-γ修饰的BBV显示出通过重编程TAM有效靶向肿瘤和调节冷肿瘤热的强大能力,为肿瘤免疫治疗提供了有效的方法。
    Tumor-associated macrophages (TAMs) are among the most abundant infiltrating leukocytes in the tumor microenvironment (TME). Reprogramming TAMs from protumor M2 to antitumor M1 phenotype is a promising strategy for remodeling the TME and promoting antitumor immunity; however, the development of an efficient strategy remains challenging. Here, a genetically modified bacterial biomimetic vesicle (BBV) with IFN-γ exposed on the surface in a nanoassembling membrane pore structure was constructed. The engineered IFN-γ BBV featured a nanoscale structure of protein and lipid vesicle, the existence of rich pattern-associated molecular patterns (PAMPs), and the costimulation of introduced IFN-γ molecules. In vitro, IFN-γ BBV reprogrammed M2 macrophages to M1, possibly through NF-κB and JAK-STAT signaling pathways, releasing nitric oxide (NO) and inflammatory cytokines IL-1β, IL-6, and TNF-α and increasing the expression of IL-12 and iNOS. In tumor-bearing mice, IFN-γ BBV demonstrated a targeted enrichment in tumors and successfully reprogrammed TAMs into the M1 phenotype; notably, the response of antigen-specific cytotoxic T lymphocyte (CTL) in TME was promoted while the immunosuppressive myeloid-derived suppressor cell (MDSC) was suppressed. The tumor growth was found to be significantly inhibited in both a TC-1 tumor and a CT26 tumor. It was indicated that the antitumor effects of IFN-γ BBV were macrophage-dependent. Further, the modulation of TME by IFN-γ BBV produced synergistic effects against tumor growth and metastasis with an immune checkpoint inhibitor in an orthotopic 4T1 breast cancer model which was insensitive to anti-PD-1 mAb alone. In conclusion, IFN-γ-modified BBV demonstrated a strong capability of efficiently targeting tumor and tuning a cold tumor hot through reprogramming TAMs, providing a potent approach for tumor immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    在最近的一项研究中,Kerzel等人。报告了一种新的治疗策略,通过诱导IFNα在这些细胞中的表达来在体内工程化肿瘤相关巨噬细胞(TAMs)。这种方法能够改善抗原呈递和T细胞活化,在多种小鼠肝转移模型中导致肿瘤生长受控。
    In a recent study, Kerzel et al. report a novel therapeutic strategy to engineer tumor-associated macrophages (TAMs) in vivo by inducing the expression of IFNα in these cells. This approach enables improved antigen presentation and T cell activation, leading to controlled tumor growth in multiple murine models of liver metastasis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:肿瘤细胞诱导的血小板聚集(TCIPA)不仅是公认的副肿瘤性血小板增多的机制,而且是恶性黑色素瘤(MM)血行转移中对免疫检查点抑制剂(ICIs)低反应的潜在突破性替代方法。然而,没有TCIPA特定的模型来进一步研究TCIPA之间的关系,肿瘤免疫微环境(TIME),和转移。
    方法:我们开发了一种具有晚期血行转移和增强TCIPA特征的TCIPA转移性黑色素瘤模型。我们还研究了TCIPA在TIME中的通路。
    结果:我们发现TCIPA通过分泌B16细胞诱导的血小板源性趋化因子如CCL2、SDF-1和IL-1β,触发肿瘤相关巨噬细胞(TAM)募集到肺转移。通过B16细胞后处理,TCIPA模型中大量的TAM极化为M2型,它们的表面程序性细胞死亡1配体1(PD-L1)表达上调,最终帮助B16细胞逃避宿主免疫并加速MM血行转移。
    结论:TCIPA通过肿瘤培养的血小板(TEPs)加速MM肺转移,触发TAM招募,促进TAM极化(M2),重塑肺转移的抑制时间。
    BACKGROUND: Tumor cell-induced platelet aggregation (TCIPA) is not only a recognized mechanism for paraneoplastic thrombocytosis but also a potential breakthrough alternative for a low response to immune checkpoint inhibitors (ICIs) in hematogenous metastasis of malignant melanoma (MM). However, there is no TCIPA-specific model for further investigation of the relationship among TCIPA, the tumor immune microenvironment (TIME), and metastasis.
    METHODS: We developed a TCIPA metastatic melanoma model with advanced hematogenous metastasis and enhanced TCIPA characteristics. We also investigated the pathway for TCIPA in the TIME.
    RESULTS: We found that TCIPA triggers the recruitment of tumor-associated macrophages (TAMs) to lung metastases by secreting B16 cell-educated platelet-derived chemokines such as CCL2, SDF-1, and IL-1β. Larger quantities of TAMs in the TCIPA model were polarized to the M2 type by B16 cell reprocessing, and their surface programmed cell death 1 ligand 1 (PD-L1) expression was upregulated, ultimately assisting B16 cells in escaping host immunity and accelerating MM hematogenous metastasis.
    CONCLUSIONS: TCIPA accelerates MM lung metastasis via tumor-educated platelets (TEPs), triggering TAM recruitment, promoting TAM polarization (M2), and remodeling the suppressive TIME in lung metastases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    肿瘤相关巨噬细胞(TAMs),作为肿瘤微环境(TME)的主要和必要组成部分,在协调胰腺癌(PaC)肿瘤发生从起始到血管生成中起关键作用,增长,和系统传播,以及免疫抑制和对化疗和免疫疗法的抗性;然而,负责TAM重新编程和功能的关键内在因素仍有待确定。通过进行单细胞RNA测序,转化生长因子-β诱导蛋白(TGFBI)在小鼠PaC肿瘤中被鉴定为TAM产生因子。TAMs在人PaC中表达TGFBI,且TGFBI表达量与人PaC增长呈正相干。通过在我们开发的TGFBI-floxed小鼠中,在体外用siRNA和体内用Cre-Lox策略诱导巨噬细胞(MΦ)中的TGFBI功能丧失,我们证明了TGFBI的破坏不仅抑制了MΦ极化为M2表型和MΦ介导的对PaC生长的刺激,而且还显著提高了抗肿瘤免疫力,与纤连蛋白1,Cxcl10和Ccl5的调节相关,使PaC对化疗敏感。我们的研究表明,在MΦ中靶向TGFBI可以为高致死性PaC开发有效的治疗干预措施。
    Tumor-associated macrophages (TAMs), as a major and essential component of tumor microenvironment (TME), play a critical role in orchestrating pancreatic cancer (PaC) tumorigenesis from initiation to angiogenesis, growth, and systemic dissemination, as well as immunosuppression and resistance to chemotherapy and immunotherapy; however, the critical intrinsic factors responsible for TAMs reprograming and function remain to be identified. By performing single-cell RNA sequencing, transforming growth factor-beta-induced protein (TGFBI) was identified as TAM-producing factor in murine PaC tumors. TAMs express TGFBI in human PaC and TGFBI expression is positively related with human PaC growth. By inducing TGFBI loss-of-function in macrophage (MΦs) in vitro with siRNA and in vivo with Cre-Lox strategy in our developed TGFBI-floxed mice, we demonstrated disruption of TGFBI not only inhibited MΦ polarization to M2 phenotype and MΦ-mediated stimulation on PaC growth, but also significantly improved anti-tumor immunity, sensitizing PaC to chemotherapy in association with regulation of fibronectin 1, Cxcl10, and Ccl5. Our studies suggest that targeting TGFBI in MΦ can develop an effective therapeutic intervention for highly lethal PaC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    癌症干细胞(CSC)已经成为肿瘤启动的关键贡献者,增长,和转移。此外,CSCs在诱导免疫逃避中发挥重要作用,从而损害癌症治疗的有效性。观察到CSC与肿瘤微环境(TME)之间的相互通信,TME为CSC生存和自我更新提供了支持性的利基,而CSCs,反过来,影响TME的极化和持久性,促进免疫抑制状态。因此,这些相互作用阻碍了当前癌症治疗的疗效,需要探索新的治疗方法来调节TME和靶向CSC。在这次审查中,我们重点介绍了CSCs用于逃避免疫监视和对治疗产生耐药性的复杂策略.此外,我们研究了CSCs和TME之间的动态相互作用,揭示了这种相互作用如何影响癌症进展。此外,我们提供了专门针对CSCs和TME的先进治疗策略的概述,这对未来癌症治疗的临床和转化研究充满希望。
    Cancer stem cells (CSCs) have emerged as key contributors to tumor initiation, growth, and metastasis. In addition, CSCs play a significant role in inducing immune evasion, thereby compromising the effectiveness of cancer treatments. The reciprocal communication between CSCs and the tumor microenvironment (TME) is observed, with the TME providing a supportive niche for CSC survival and self-renewal, while CSCs, in turn, influence the polarization and persistence of the TME, promoting an immunosuppressive state. Consequently, these interactions hinder the efficacy of current cancer therapies, necessitating the exploration of novel therapeutic approaches to modulate the TME and target CSCs. In this review, we highlight the intricate strategies employed by CSCs to evade immune surveillance and develop resistance to therapies. Furthermore, we examine the dynamic interplay between CSCs and the TME, shedding light on how this interaction impacts cancer progression. Moreover, we provide an overview of advanced therapeutic strategies that specifically target CSCs and the TME, which hold promise for future clinical and translational studies in cancer treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    胶质母细胞瘤(Gb)是最常见的原发性脑恶性肿瘤,预后较差。免疫细胞是影响Gb预后的主要因素,肿瘤相关巨噬细胞(TAMs)是Gb免疫微环境中主要的浸润免疫细胞群。分析磁共振成像(MRI)特征与Gb、利用影像学特征表征肿瘤组织中TAMs的浸润程度,可为临床决策和预后评估提供指标。
    收集2018年1月至2022年4月在兰州大学第二医院通过组织病理学和分子诊断诊断的140例异柠檬酸脱氢酶(IDH)野生型Gb患者的数据。横断面研究。MRI图像检查病灶位置,囊肿,坏死,出血,对比增强T1加权MRI信号强度,平均表观扩散系数(ADCmean),和最小表观扩散系数(ADCmin)。使用抗CD163和抗CD68抗体的免疫组织化学染色用于巨噬细胞检测。用ImageJ软件(美国国立卫生研究院)在9个显微镜视野中以400倍放大每全载玻片图像估计阳性细胞百分比。此外,MRI特征之间的关系,分子,状态,并分析CD68和CD163的阳性表达。
    我们的研究发现,CD68+和CD163+TAM的平均值或中值分别为7.39%和14.98%,分别。CD163+TAMs与CD68+TAMs有明显的相关性(r=0.497;P=0.000)。Gb患者CD68+和CD163+巨噬细胞浸润与诊断年龄相关(CD68+:r=0.230,P=0.006;CD163+:r=0.172,P=0.042)。GbTAM在不同肿瘤部位的浸润程度不同,颞叶CD163+巨噬细胞和CD68+巨噬细胞浸润最高(18.58%和9.46%,分别)。CD163+巨噬细胞浸润与ADCmean呈正相关(r=0.208;P=0.014)。CD68+巨噬细胞浸润在不同程度的肿瘤增强组间差异显著(H=4.228;P=0.017)。野生型和突变型端粒酶逆转录酶(TERT)类型之间的CD68TAM和CD163TAM存在显着差异(分别为P=0.004和P=0.031)。
    年龄,肿瘤的位置,肿瘤增强程度,ADC值,TERT突变状态与巨噬细胞浸润相关。这些发现可能是表征Gb患者肿瘤微环境的有效工具。
    UNASSIGNED: Glioblastoma (Gb) is the most common primary malignant tumor of brain with poor prognosis. Immune cells are the main factors affecting the prognosis of Gb, tumor-associated macrophages (TAMs) are the predominant infiltrating immune cell population in the immune microenvironment of Gb. Analyzing the relationship between magnetic resonance imaging (MRI) features and TAMs of Gb, and using imaging features to characterize the infiltration level of TAMs in tumor tissue may provide indicators for clinical decision-making and prognosis evaluation of Gb.
    UNASSIGNED: Data from 140 in patients with isocitrate dehydrogenase (IDH) wild-type Gb diagnosed via histopathology and molecular diagnosis in the Second Hospital of Lanzhou University from January 2018 to April 2022 were collected in this retrospective, cross-sectional study. MRI images were reviewed for lesion location, cyst, necrosis, hemorrhage, contrast-enhanced T1-weighted MRI signal intensity, average apparent diffusion coefficient (ADCmean), and minimum apparent diffusion coefficient (ADCmin). Immunohistochemical staining with anti-CD163 and anti-CD68 antibodies was employed for macrophage detection. The positive cell percentage was estimated in 9 microscopic fields at 400× magnification per whole-slide image with ImageJ software (National Institutes of Health). Additionally, the relationship between MRI features, molecular, states and the positive CD68 and CD163 expression was analyzed.
    UNASSIGNED: Our study discovered that the mean or median values of CD68+ and CD163+ TAMs were 7.39% and 14.98%, respectively. There was an obvious correlation between CD163+ TAMs and CD68+ TAMs (r=0.497; P=0.000). CD68+ and CD163+ macrophage infiltration correlated with age at diagnosis in patients with Gb (CD68+: r=0.230, P=0.006; CD163+: r=0.172, P=0.042). The levels of Gb TAM infiltration in different tumor locations varied, with the temporal lobe having the highest CD163+ macrophage and CD68+ macrophage infiltration (18.58% and 9.46%, respectively). CD163+ macrophage infiltration was positively correlated with ADCmean (r=0.208; P=0.014). The infiltration of CD68+ macrophages differed significantly between groups with varying degrees of tumor enhancement (H =4.228; P=0.017). There was a significant difference in CD68+ TAMs and CD163+ TAMs between the wild-type and mutant-type telomerase reverse transcriptase (TERT) types (P=0.004 and P=0.031, respectively).
    UNASSIGNED: Age, location of the tumor, degree of tumor enhancement, ADC value, and TERT mutation status were associated with macrophage infiltration. These findings may serve as an effective tool for characterizing the tumor microenvironment in patients with Gb.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    肿瘤相关巨噬细胞(TAM)影响肿瘤细胞的内在特性和肿瘤微环境(TME),可以刺激肿瘤细胞增殖,迁移,和遗传不稳定性,巨噬细胞多样性包括具有不同功能特征的肿瘤的多样性。巨噬细胞现在是各种疾病的中心药物靶标,尤其是在TME中,which,作为“肿瘤启动子”和“免疫抑制剂”,在肿瘤发展过程中具有不同的责任,并伴随着各种亚群的显着动态改变。通过改变TAM极化,重塑TME的免疫抑制和促进预先存在的抗肿瘤免疫反应至关重要。这与免疫疗法的疗效有关,发现TAM的确切作用机制并确定其特定靶标对于优化当前的免疫疗法至关重要。因此,这篇综述旨在揭示巨噬细胞与程序性死亡和肿瘤治疗的三重相互作用,并将某些关系整合到癌症治疗中。
    Tumor-associated macrophage (TAM) affects the intrinsic properties of tumor cells and the tumor microenvironment (TME), which can stimulate tumor cell proliferation, migration, and genetic instability, and macrophage diversity includes the diversity of tumors with different functional characteristics. Macrophages are now a central drug target in various diseases, especially in the TME, which, as \"tumor promoters\" and \"immunosuppressors\", have different responsibilities during tumor development and accompany by significant dynamic alterations in various subpopulations. Remodelling immunosuppression of TME and promotion of pre-existing antitumor immune responses is critical by altering TAM polarization, which is relevant to the efficacy of immunotherapy, and uncovering the exact mechanism of action of TAMs and identifying their specific targets is vital to optimizing current immunotherapies. Hence, this review aims to reveal the triadic interactions of macrophages with programmed death and oncotherapy, and to integrate certain relationships in cancer treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号