首页 > 最新文献

Cellular Oncology最新文献

英文 中文
Expression of the zinc-finger transcription factor Osterix (SP7) in invasive breast cancer and its prognostic significance. 锌指转录因子Osterix (SP7)在浸润性乳腺癌中的表达及其预后意义。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-04-07 DOI: 10.1007/s13402-025-01062-9
Behnaz Saidy, Laura Gull, Andrew G Hacker, Emad A Rakha, Andrew R Green, Ian O Ellis, Stewart G Martin, Sarah J Storr

Introduction: Osterix, encoded by SP7, is a transcription factor crucial in osteoblast differentiation and bone formation. While initially characterised in bone development, emerging evidence suggests its involvement in cancer, particularly breast cancer metastasis to bone.

Methods: Osterix protein expression was evaluated in 1340 early-stage invasive breast tumours by immunohistochemistry. Cytoplasmic and nuclear expression levels were assessed and associations with clinicopathological variables and patient survival determined. Additionally, SP7 mRNA expression was examined in the METABRIC cohort of patients. Gene set enrichment analysis (GSEA) was performed to explore the role of osterix in the hallmarks of cancer genesets.

Results: Results revealed significant associations between reduced nuclear osterix protein expression and adverse clinicopathological features, including larger tumour size, higher grade, and poor Nottingham Prognostic Index. Low nuclear osterix protein expression was also linked to shorter breast cancer-specific survival and distant metastasis free survival, particularly in patients with HER2 positive tumours. No associations were found between SP7 mRNA expression and clinicopathological variables or survival outcomes. GSEA identified enrichment of genes involved in KRAS signaling in tumours with high SP7 expression.

Conclusion: These data suggest that reduced nuclear expression of osterix is associated with poor clinical outcome of breast cancer patients and may be of clinical relevance.

Osterix是一种由SP7编码的转录因子,在成骨细胞分化和骨形成中起着至关重要的作用。虽然最初以骨骼发育为特征,但新出现的证据表明它与癌症,特别是乳腺癌骨转移有关。方法:应用免疫组化方法对1340例早期浸润性乳腺肿瘤中Osterix蛋白的表达进行检测。评估细胞质和核表达水平,并确定与临床病理变量和患者生存率的关系。此外,在METABRIC患者队列中检测了SP7 mRNA的表达。进行基因集富集分析(GSEA)以探索osterix在癌症基因集标记中的作用。结果:结果显示核骨蛋白表达减少与不良临床病理特征(包括较大的肿瘤大小、较高的分级和较差的诺丁汉预后指数)之间存在显著关联。低核骨蛋白表达也与较短的乳腺癌特异性生存期和无远处转移生存期有关,特别是在HER2阳性肿瘤患者中。未发现SP7 mRNA表达与临床病理变量或生存结果之间存在关联。GSEA在SP7高表达的肿瘤中发现了KRAS信号通路相关基因的富集。结论:这些数据提示,骨核表达减少与乳腺癌患者临床预后不良有关,可能具有临床意义。
{"title":"Expression of the zinc-finger transcription factor Osterix (SP7) in invasive breast cancer and its prognostic significance.","authors":"Behnaz Saidy, Laura Gull, Andrew G Hacker, Emad A Rakha, Andrew R Green, Ian O Ellis, Stewart G Martin, Sarah J Storr","doi":"10.1007/s13402-025-01062-9","DOIUrl":"https://doi.org/10.1007/s13402-025-01062-9","url":null,"abstract":"<p><strong>Introduction: </strong>Osterix, encoded by SP7, is a transcription factor crucial in osteoblast differentiation and bone formation. While initially characterised in bone development, emerging evidence suggests its involvement in cancer, particularly breast cancer metastasis to bone.</p><p><strong>Methods: </strong>Osterix protein expression was evaluated in 1340 early-stage invasive breast tumours by immunohistochemistry. Cytoplasmic and nuclear expression levels were assessed and associations with clinicopathological variables and patient survival determined. Additionally, SP7 mRNA expression was examined in the METABRIC cohort of patients. Gene set enrichment analysis (GSEA) was performed to explore the role of osterix in the hallmarks of cancer genesets.</p><p><strong>Results: </strong>Results revealed significant associations between reduced nuclear osterix protein expression and adverse clinicopathological features, including larger tumour size, higher grade, and poor Nottingham Prognostic Index. Low nuclear osterix protein expression was also linked to shorter breast cancer-specific survival and distant metastasis free survival, particularly in patients with HER2 positive tumours. No associations were found between SP7 mRNA expression and clinicopathological variables or survival outcomes. GSEA identified enrichment of genes involved in KRAS signaling in tumours with high SP7 expression.</p><p><strong>Conclusion: </strong>These data suggest that reduced nuclear expression of osterix is associated with poor clinical outcome of breast cancer patients and may be of clinical relevance.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":""},"PeriodicalIF":4.9,"publicationDate":"2025-04-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143796844","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Sirtuin 3-mediated delactylation of malic enzyme 2 disrupts redox balance and inhibits colorectal cancer growth. Sirtuin 3介导的苹果酸酶2去乙酰化破坏氧化还原平衡并抑制结直肠癌的生长。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-04-07 DOI: 10.1007/s13402-025-01058-5
Chaoqun Li, Cun Ge, Qingwen Wang, Peng Teng, Heyuan Jia, Surui Yao, Zhaohui Huang

Purpose: Post-translational modifications, such as lactylation, are emerging as critical regulators of metabolic enzymes in cancer progression. Mitochondrial malic enzyme 2 (ME2), a key enzyme in the TCA cycle, plays a pivotal role in maintaining redox homeostasis and supporting tumor metabolism. However, the functional significance of ME2 lactylation and its regulatory mechanisms remain unclear. This study investigates the role of ME2 K352 lactylation in modulating enzymatic activity, redox balance, and tumor progression.

Methods: Immunoprecipitation and western blotting were used to assess ME2 lactylation and its interaction with Sirtuin 3 (SIRT3). Mass spectrometry identified the lactylation site on ME2. Enzymatic activity was measured using NADH production assays. The functional effects of ME2 K352 lactylation were analyzed by measuring ROS levels, NADP⁺/NADPH ratios, metabolic intermediates, and mitochondrial respiration parameters. Cell proliferation was evaluated via CCK-8 and colony formation assays. Xenograft tumor models and Ki-67 immunohistochemical staining were used to assess tumor growth and proliferation in vivo.

Results: Mass spectrometry identified K352 as the primary lactylation site on ME2. Sodium lactate treatment enhanced ME2 lactylation and enzymatic activity, while SIRT3-mediated delactylation at K352 reduced ME2 activity, disrupting redox homeostasis. Cells expressing the K352R mutant exhibited elevated ROS levels, higher NADP⁺/NADPH ratios, and altered levels of metabolic intermediates, including increased malate and lactate with reduced pyruvate. Additionally, re-expression of ME2 K352R in HCT116 cells significantly impaired proliferation and colony formation. In vivo, xenograft models demonstrated that ME2 K352R expression suppressed tumor growth, as evidenced by reduced tumor volume, weight, and Ki-67 staining.

Conclusions: This study reveals that ME2 K352 lactylation is a critical regulatory mechanism that modulates enzymatic activity, mitochondrial function, and tumor progression. SIRT3-mediated delactylation of ME2 K352 disrupts redox homeostasis and inhibits tumor growth. These findings highlight the potential of targeting ME2 lactylation as a therapeutic strategy in cancer treatment.

目的:乳化等翻译后修饰正在成为癌症进展过程中代谢酶的关键调节因子。线粒体苹果酸酶 2(ME2)是 TCA 循环中的一种关键酶,在维持氧化还原平衡和支持肿瘤代谢方面发挥着关键作用。然而,ME2乳化的功能意义及其调控机制仍不清楚。本研究探讨了ME2 K352乳化在调节酶活性、氧化还原平衡和肿瘤进展中的作用:方法:采用免疫沉淀和免疫印迹法评估 ME2 乳化及其与 Sirtuin 3(SIRT3)的相互作用。质谱法确定了 ME2 上的乳化位点。使用 NADH 生成试验测量了酶活性。通过测量 ROS 水平、NADP⁺/NADPH 比率、代谢中间产物和线粒体呼吸参数,分析了 ME2 K352 乳酰化的功能影响。细胞增殖通过 CCK-8 和集落形成试验进行评估。异种移植肿瘤模型和 Ki-67 免疫组化染色用于评估体内肿瘤的生长和增殖:结果:质谱鉴定出K352是ME2上的主要乳化位点。乳酸钠处理增强了ME2的乳化作用和酶活性,而SIRT3介导的K352脱乳作用降低了ME2的活性,破坏了氧化还原平衡。表达 K352R 突变体的细胞表现出 ROS 水平升高、NADP⁺/NADPH 比率升高以及代谢中间产物水平改变,包括苹果酸和乳酸增加而丙酮酸减少。此外,在 HCT116 细胞中重新表达 ME2 K352R 会显著影响细胞增殖和集落形成。在体内,异种移植模型显示 ME2 K352R 的表达抑制了肿瘤的生长,肿瘤体积、重量和 Ki-67 染色的减少都证明了这一点:这项研究揭示了 ME2 K352 乳化是一种关键的调控机制,可调节酶活性、线粒体功能和肿瘤进展。SIRT3 介导的 ME2 K352 脱乳作用会破坏氧化还原平衡并抑制肿瘤生长。这些发现凸显了靶向 ME2 乳化作为癌症治疗策略的潜力。
{"title":"Sirtuin 3-mediated delactylation of malic enzyme 2 disrupts redox balance and inhibits colorectal cancer growth.","authors":"Chaoqun Li, Cun Ge, Qingwen Wang, Peng Teng, Heyuan Jia, Surui Yao, Zhaohui Huang","doi":"10.1007/s13402-025-01058-5","DOIUrl":"https://doi.org/10.1007/s13402-025-01058-5","url":null,"abstract":"<p><strong>Purpose: </strong>Post-translational modifications, such as lactylation, are emerging as critical regulators of metabolic enzymes in cancer progression. Mitochondrial malic enzyme 2 (ME2), a key enzyme in the TCA cycle, plays a pivotal role in maintaining redox homeostasis and supporting tumor metabolism. However, the functional significance of ME2 lactylation and its regulatory mechanisms remain unclear. This study investigates the role of ME2 K352 lactylation in modulating enzymatic activity, redox balance, and tumor progression.</p><p><strong>Methods: </strong>Immunoprecipitation and western blotting were used to assess ME2 lactylation and its interaction with Sirtuin 3 (SIRT3). Mass spectrometry identified the lactylation site on ME2. Enzymatic activity was measured using NADH production assays. The functional effects of ME2 K352 lactylation were analyzed by measuring ROS levels, NADP⁺/NADPH ratios, metabolic intermediates, and mitochondrial respiration parameters. Cell proliferation was evaluated via CCK-8 and colony formation assays. Xenograft tumor models and Ki-67 immunohistochemical staining were used to assess tumor growth and proliferation in vivo.</p><p><strong>Results: </strong>Mass spectrometry identified K352 as the primary lactylation site on ME2. Sodium lactate treatment enhanced ME2 lactylation and enzymatic activity, while SIRT3-mediated delactylation at K352 reduced ME2 activity, disrupting redox homeostasis. Cells expressing the K352R mutant exhibited elevated ROS levels, higher NADP⁺/NADPH ratios, and altered levels of metabolic intermediates, including increased malate and lactate with reduced pyruvate. Additionally, re-expression of ME2 K352R in HCT116 cells significantly impaired proliferation and colony formation. In vivo, xenograft models demonstrated that ME2 K352R expression suppressed tumor growth, as evidenced by reduced tumor volume, weight, and Ki-67 staining.</p><p><strong>Conclusions: </strong>This study reveals that ME2 K352 lactylation is a critical regulatory mechanism that modulates enzymatic activity, mitochondrial function, and tumor progression. SIRT3-mediated delactylation of ME2 K352 disrupts redox homeostasis and inhibits tumor growth. These findings highlight the potential of targeting ME2 lactylation as a therapeutic strategy in cancer treatment.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":""},"PeriodicalIF":4.9,"publicationDate":"2025-04-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143796852","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Tumor-specific CXCR6 positive precursor CD8+ T cells mediate tumor control in metastatic melanoma. 肿瘤特异性CXCR6阳性前体CD8+ T细胞介导转移性黑色素瘤的肿瘤控制。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-04-07 DOI: 10.1007/s13402-025-01040-1
Yang Song, Ji Chen, Yaqin Zhang, Ning Wu, Yongjun Zhu, Gang Chen, Feng Miao, Zhiming Chen, Yiqing Wang

Background: Adoptive cell therapy (ACT) mediates durable and complete regression of various cancers. However, its efficacy is limited by the long-term persistence of cytotoxic T lymphocytes, given their irreversible dysfunction within the tumor microenvironment. Herein, we aimed to establish an artificial lung metastasis model to examine T-lymphocyte subsets, in order to identify potential effective cell subsets for ACT.

Methods: A metastatic lung melanoma mouse model was established using OVA-expressing melanoma B16 cells. Flow cytometry analysis was conducted to examine the surface markers, transcription factors, and secreted cytokines of tumor-specific CD8+ T cells within metastatic tissues. The infiltrated cells were sorted by flow cytometry for in vitro tumor cell killing assays or in vivo cell infusion therapy combined with chemotherapeutic drugs and immune checkpoint blockade antibodies.

Results: Exhausted CD8+ T cells (Tex) exhibited high heterogeneity in metastatic tissues. Among Tex cells, the CXCR6- precursor cell showed certain memory characteristics, including phenotype, transcription factors, and maintenance, whereas the CXCR6+ subpopulation partially lost these traits. Moreover, CXCR6+ precursor cells effectively replenished effector-like Tex cells in metastatic tissues and exerted direct cytotoxicity against tumor cells. Notably, transferring these tumor-specific CXCR6+ precursor-exhausted T (Texp) cells into recipients induced a substantial regression of metastasis. In addition, these cells could respond to immune checkpoint blockade, which could better control tumor metastasis.

Conclusions: In our study, a subset of antigen-specific CXCR6-expressing Texp cells was observed within the metastatic tissue. The cells served as a crucial source of effector-like Tex cells and exerted direct cytotoxic effects on tumor cells. Adoptive transfer of CXCR6+ Texp cells effectively mitigated lung metastasis in mice. This study helps elucidate the role of Texp cells in metastasis, thereby offering novel insights into enhancing the efficacy and durability of immunotherapy.

背景:采用细胞疗法(ACT)可使各种癌症得到持久、彻底的治疗。然而,由于细胞毒性 T 淋巴细胞在肿瘤微环境中不可逆转的功能障碍,其疗效受限于细胞毒性 T 淋巴细胞的长期存在。在此,我们旨在建立一个人工肺转移模型来研究T淋巴细胞亚群,以确定ACT潜在的有效细胞亚群:方法:使用表达 OVA 的黑色素瘤 B16 细胞建立转移性肺黑色素瘤小鼠模型。流式细胞术分析检测了转移组织中肿瘤特异性 CD8+ T 细胞的表面标志物、转录因子和分泌的细胞因子。通过流式细胞术对浸润细胞进行分拣,用于体外肿瘤细胞杀伤试验或体内细胞输注疗法,并结合化疗药物和免疫检查点阻断抗体:结果:在转移组织中,衰竭的 CD8+ T 细胞(Tex)表现出高度异质性。在Tex细胞中,CXCR6-前体细胞表现出一定的记忆特征,包括表型、转录因子和维持能力,而CXCR6+亚群则部分丧失了这些特征。此外,CXCR6+前体细胞能有效补充转移组织中的效应样Tex细胞,并对肿瘤细胞产生直接的细胞毒性。值得注意的是,将这些肿瘤特异性 CXCR6+ 前体细胞排泄的 T(Texp)细胞转移到受者体内可诱导转移灶的大幅消退。此外,这些细胞还能对免疫检查点阻断产生反应,从而更好地控制肿瘤转移:在我们的研究中,在转移组织中观察到了表达抗原特异性 CXCR6 的 Texp 细胞亚群。这些细胞是效应样 Tex 细胞的重要来源,对肿瘤细胞具有直接的细胞毒性作用。CXCR6+Texp细胞的采纳性转移能有效减轻小鼠的肺转移。这项研究有助于阐明 Texp 细胞在转移中的作用,从而为提高免疫疗法的疗效和持久性提供新的见解。
{"title":"Tumor-specific CXCR6 positive precursor CD8<sup>+</sup> T cells mediate tumor control in metastatic melanoma.","authors":"Yang Song, Ji Chen, Yaqin Zhang, Ning Wu, Yongjun Zhu, Gang Chen, Feng Miao, Zhiming Chen, Yiqing Wang","doi":"10.1007/s13402-025-01040-1","DOIUrl":"https://doi.org/10.1007/s13402-025-01040-1","url":null,"abstract":"<p><strong>Background: </strong>Adoptive cell therapy (ACT) mediates durable and complete regression of various cancers. However, its efficacy is limited by the long-term persistence of cytotoxic T lymphocytes, given their irreversible dysfunction within the tumor microenvironment. Herein, we aimed to establish an artificial lung metastasis model to examine T-lymphocyte subsets, in order to identify potential effective cell subsets for ACT.</p><p><strong>Methods: </strong>A metastatic lung melanoma mouse model was established using OVA-expressing melanoma B16 cells. Flow cytometry analysis was conducted to examine the surface markers, transcription factors, and secreted cytokines of tumor-specific CD8<sup>+</sup> T cells within metastatic tissues. The infiltrated cells were sorted by flow cytometry for in vitro tumor cell killing assays or in vivo cell infusion therapy combined with chemotherapeutic drugs and immune checkpoint blockade antibodies.</p><p><strong>Results: </strong>Exhausted CD8<sup>+</sup> T cells (Tex) exhibited high heterogeneity in metastatic tissues. Among Tex cells, the CXCR6<sup>-</sup> precursor cell showed certain memory characteristics, including phenotype, transcription factors, and maintenance, whereas the CXCR6<sup>+</sup> subpopulation partially lost these traits. Moreover, CXCR6<sup>+</sup> precursor cells effectively replenished effector-like Tex cells in metastatic tissues and exerted direct cytotoxicity against tumor cells. Notably, transferring these tumor-specific CXCR6<sup>+</sup> precursor-exhausted T (Texp) cells into recipients induced a substantial regression of metastasis. In addition, these cells could respond to immune checkpoint blockade, which could better control tumor metastasis.</p><p><strong>Conclusions: </strong>In our study, a subset of antigen-specific CXCR6-expressing Texp cells was observed within the metastatic tissue. The cells served as a crucial source of effector-like Tex cells and exerted direct cytotoxic effects on tumor cells. Adoptive transfer of CXCR6<sup>+</sup> Texp cells effectively mitigated lung metastasis in mice. This study helps elucidate the role of Texp cells in metastasis, thereby offering novel insights into enhancing the efficacy and durability of immunotherapy.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":""},"PeriodicalIF":4.9,"publicationDate":"2025-04-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143796855","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Allelic expression imbalance of CDKN2A variants in childhood acute lymphoblastic leukemia. CDKN2A变异在儿童急性淋巴细胞白血病中的等位基因表达失衡。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-03-04 DOI: 10.1007/s13402-025-01049-6
Zhexuan Tang, Kunlin Pei, Haoyu Xu, Yongzhi Zheng, Shuquan Zhuang, Kaizhi Weng, Yingyi He, Jing Wu, Hui Zhang

Introduction: Germline CDKN2A variant predisposes to childhood acute lymphoblastic leukemia (ALL) through allelic expression imbalance (AEI). It is unknown, therefore, how these germline variations work and whether they all confer B-ALL susceptibility through AEI.

Methods and results: Using allele-specific Taqman PCR assays, we demonstrated that preferentially expressed of those functional inherited coding variants in leukemic cells compared to hematopoietic cells. In an inherent p 16Ink4a-defective Ba/F3 cell model overexpressing functional p16INK4A variants showed enhanced susceptibility to transformation by BCR-ABL1-, NRASG12D-, and JAK2R683G + CRLF2-. Notably, the variant p16INK4A exhibited higher transcription level than wild-type allele in co-expression studies. While CDK4/6 inhibitor partially suppressed NRASG12D-, and JAK2R683G + CRLF2-induced transformation, it proved ineffective against BCR-ABL1-induced leukemic transformation. Differential gene expression analysis revealed upregulation of m6A-related gene PRRC2A, whose knockout partially restored wild-type p16INK4A expression.

Conclusion: These findings illuminate how inherited CDKN2A genetic variations of coding region influence ALL development through AEI mechanisms.

生殖系CDKN2A变异通过等位基因表达不平衡(AEI)易患儿童急性淋巴细胞白血病(ALL)。因此,尚不清楚这些种系变异是如何起作用的,以及它们是否都通过AEI赋予B-ALL易感性。方法和结果:使用等位基因特异性Taqman PCR检测,我们证明与造血细胞相比,白血病细胞优先表达这些功能遗传编码变体。在固有的p16INK4A缺陷的Ba/F3细胞模型中,过表达功能p16INK4A变异体对BCR-ABL1-、NRASG12D-和JAK2R683G + CRLF2-的转化易感性增强。值得注意的是,在共表达研究中,变异p16INK4A比野生型等位基因表现出更高的转录水平。虽然CDK4/6抑制剂部分抑制NRASG12D-和JAK2R683G + crlf2诱导的白血病转化,但对bcr - abl1诱导的白血病转化无效。差异基因表达分析显示m6a相关基因PRRC2A上调,其敲除部分恢复了野生型p16INK4A的表达。结论:这些发现阐明了CDKN2A编码区的遗传变异如何通过AEI机制影响ALL的发展。
{"title":"Allelic expression imbalance of CDKN2A variants in childhood acute lymphoblastic leukemia.","authors":"Zhexuan Tang, Kunlin Pei, Haoyu Xu, Yongzhi Zheng, Shuquan Zhuang, Kaizhi Weng, Yingyi He, Jing Wu, Hui Zhang","doi":"10.1007/s13402-025-01049-6","DOIUrl":"https://doi.org/10.1007/s13402-025-01049-6","url":null,"abstract":"<p><strong>Introduction: </strong>Germline CDKN2A variant predisposes to childhood acute lymphoblastic leukemia (ALL) through allelic expression imbalance (AEI). It is unknown, therefore, how these germline variations work and whether they all confer B-ALL susceptibility through AEI.</p><p><strong>Methods and results: </strong>Using allele-specific Taqman PCR assays, we demonstrated that preferentially expressed of those functional inherited coding variants in leukemic cells compared to hematopoietic cells. In an inherent p 16<sup>Ink4a</sup>-defective Ba/F3 cell model overexpressing functional p16<sup>INK4A</sup> variants showed enhanced susceptibility to transformation by BCR-ABL1-, NRAS<sup>G12D</sup>-, and JAK2<sup>R683G</sup> + CRLF2-. Notably, the variant p16<sup>INK4A</sup> exhibited higher transcription level than wild-type allele in co-expression studies. While CDK4/6 inhibitor partially suppressed NRAS<sup>G12D</sup>-, and JAK2<sup>R683G</sup> + CRLF2-induced transformation, it proved ineffective against BCR-ABL1-induced leukemic transformation. Differential gene expression analysis revealed upregulation of m6A-related gene PRRC2A, whose knockout partially restored wild-type p16<sup>INK4A</sup> expression.</p><p><strong>Conclusion: </strong>These findings illuminate how inherited CDKN2A genetic variations of coding region influence ALL development through AEI mechanisms.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":""},"PeriodicalIF":4.9,"publicationDate":"2025-03-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143544129","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Spatial distribution of tertiary lymphoid structures in the molecular and clinical context of non-small cell lung cancer. 非小细胞肺癌分子和临床背景下三级淋巴样结构的空间分布。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-03-03 DOI: 10.1007/s13402-025-01052-x
Hedvig Elfving, Hui Yu, Kaleab Kassete Fessehatsion, Hans Brunnström, Johan Botling, Miklos Gulyas, Max Backman, Amanda Lindberg, Carina Strell, Patrick Micke

Introduction: Tertiary lymphoid structures (TLS) are lymphocyte aggregates resembling secondary lymphoid organs and are pivotal in cancer immunity. The ambiguous morphological definition of TLS makes it challenging to ascertain their clinical impact on patient survival and response to immunotherapy.

Objectives: This study aimed to characterize TLS in hematoxylin-eosin tissue sections from lung cancer patients, assessing their occurrence in relation to the local immune environment, mutational background, and patient outcome.

Methods: Two pathologists evaluated one whole tissue section from resection specimens of 680 NSCLC patients. TLS were spatially quantified within the tumor area or periphery and further categorized based on the presence of germinal centers (mature TLS). Metrics were integrated with immune cell counts, genomic and transcriptomic data, and correlated with clinical parameters.

Results: TLS were present in 86% of 536 evaluable cases, predominantly in the tumor periphery, with a median of eight TLS per case. Mature TLS were found in 24% of cases. TLS presence correlated positively with increased plasma cell (CD138+) and lymphocytic cell (CD3+, CD8+, FOXP3+) infiltration. Tumors with higher tumor mutational burden exhibited higher numbers of peripheral TLS. The overall TLS quantity was independently associated with improved patient survival, irrespective of TLS maturation status. This prognostic association held true for peripheral TLS but not for tumor TLS.

Conclusion: TLS in NSCLC is common and their correlation with a specific immune phenotype suggests biological relevance in the local immune reaction. The prognostic significance of this scoring system on routine hematoxylin-eosin sections has the potential to augment diagnostic algorithms for NSCLC patients.

三级淋巴样结构(TLS)是类似次级淋巴样器官的淋巴细胞聚集体,在肿瘤免疫中起关键作用。TLS的模糊形态学定义使得确定其对患者生存和免疫治疗反应的临床影响具有挑战性。目的:本研究旨在表征肺癌患者苏木精-伊红组织切片中的TLS,评估其发生与局部免疫环境、突变背景和患者预后的关系。方法:两位病理学家对680例非小细胞肺癌患者切除标本的一整块组织切片进行评估。在肿瘤区域或周围对TLS进行空间量化,并根据生发中心(成熟TLS)的存在进一步分类。这些指标与免疫细胞计数、基因组和转录组学数据相结合,并与临床参数相关。结果:在536例可评估的病例中,TLS存在于86%,主要在肿瘤周围,每例中位数为8个TLS。成熟的TLS占24%。TLS的存在与浆细胞(CD138+)和淋巴细胞(CD3+、CD8+、FOXP3+)浸润增加呈正相关。肿瘤突变负荷越大,外周TLS数量越多。TLS的总数量与患者生存率的提高独立相关,而与TLS的成熟状态无关。这种预后相关性适用于外周TLS,但不适用于肿瘤TLS。结论:TLS在非小细胞肺癌中是常见的,其与特定免疫表型的相关性提示其在局部免疫反应中具有生物学相关性。该评分系统对常规苏木精-伊红切片的预后意义有可能增强非小细胞肺癌患者的诊断算法。
{"title":"Spatial distribution of tertiary lymphoid structures in the molecular and clinical context of non-small cell lung cancer.","authors":"Hedvig Elfving, Hui Yu, Kaleab Kassete Fessehatsion, Hans Brunnström, Johan Botling, Miklos Gulyas, Max Backman, Amanda Lindberg, Carina Strell, Patrick Micke","doi":"10.1007/s13402-025-01052-x","DOIUrl":"https://doi.org/10.1007/s13402-025-01052-x","url":null,"abstract":"<p><strong>Introduction: </strong>Tertiary lymphoid structures (TLS) are lymphocyte aggregates resembling secondary lymphoid organs and are pivotal in cancer immunity. The ambiguous morphological definition of TLS makes it challenging to ascertain their clinical impact on patient survival and response to immunotherapy.</p><p><strong>Objectives: </strong>This study aimed to characterize TLS in hematoxylin-eosin tissue sections from lung cancer patients, assessing their occurrence in relation to the local immune environment, mutational background, and patient outcome.</p><p><strong>Methods: </strong>Two pathologists evaluated one whole tissue section from resection specimens of 680 NSCLC patients. TLS were spatially quantified within the tumor area or periphery and further categorized based on the presence of germinal centers (mature TLS). Metrics were integrated with immune cell counts, genomic and transcriptomic data, and correlated with clinical parameters.</p><p><strong>Results: </strong>TLS were present in 86% of 536 evaluable cases, predominantly in the tumor periphery, with a median of eight TLS per case. Mature TLS were found in 24% of cases. TLS presence correlated positively with increased plasma cell (CD138+) and lymphocytic cell (CD3+, CD8+, FOXP3+) infiltration. Tumors with higher tumor mutational burden exhibited higher numbers of peripheral TLS. The overall TLS quantity was independently associated with improved patient survival, irrespective of TLS maturation status. This prognostic association held true for peripheral TLS but not for tumor TLS.</p><p><strong>Conclusion: </strong>TLS in NSCLC is common and their correlation with a specific immune phenotype suggests biological relevance in the local immune reaction. The prognostic significance of this scoring system on routine hematoxylin-eosin sections has the potential to augment diagnostic algorithms for NSCLC patients.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":""},"PeriodicalIF":4.9,"publicationDate":"2025-03-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143544130","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The MEK5/ERK5 pathway promotes the activation of the Hedgehog/GLI signaling in melanoma cells. MEK5/ERK5通路促进黑色素瘤细胞中Hedgehog/GLI信号的激活。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-02-25 DOI: 10.1007/s13402-025-01050-z
Ignazia Tusa, Sinforosa Gagliardi, Alessio Menconi, Luisa Maresca, Alessandro Tubita, Matteo Lulli, Barbara Stecca, Elisabetta Rovida

Purpose: Malignant melanoma is the deadliest skin cancer, with a poor prognosis in advanced stages. We reported that both Hedgehog-GLI (HH/GLI) and Mitogen-activated protein Kinase (MAPK) extracellular signal-regulated kinase 5 (ERK5) pathways promote melanoma growth, and that ERK5 activation is required for HH/GLI-dependent melanoma cell proliferation. Here, we explored whether ERK5 regulates HH/GLI signaling.

Methods: Both genetic (using ERK5-specific shRNA) and pharmacologic (using the ERK5 inhibitors JWG-071 and AX15836, and the MAPK/ERK kinase 5, MEK5 inhibitors GW284543 and BIX02189) targeting approaches were used. Luciferase assay using the GLI-binding site luciferase reporter was performed to evaluate GLI transcriptional activity. A constitutively active form of MEK5 (MEK5DD) was used to induce ERK5 activation. 3D spheroid assays were performed in melanoma cells.

Results: Genetic and pharmacologic ERK5 inhibition reduces GLI1 and GLI2 protein levels and transcriptional activity of endogenous HH/GLI pathway induced by the agonist SAG in NIH/3T3 cells. In these cells, MEK5DD overexpression potentiates transcriptional activity of endogenous HH/GLI pathway induced by SAG, whereas ERK5 silencing prevents this effect. Consistently, MEK5DD overexpression increases GLI1 and GLI2 protein levels. In melanoma cells, ERK5 silencing reduces GLI1 and GLI2 mRNA and protein levels and inhibits GLI transcriptional activity. MEK5DD further increases the transcriptional activity of the HH/GLI pathway and GLI1 protein levels. Combination of GLI and MEK5 inhibitors is more effective than single treatments in reducing melanoma spheroid growth.

Conclusions: MEK5-ERK5 is an activator of GLI transcription factors, and combined targeting of these pathways warrants further preclinical investigation as a potential innovative therapeutic approach for melanoma.

目的:恶性黑色素瘤是最致命的皮肤癌,晚期预后较差。我们报道了Hedgehog-GLI (HH/GLI)和丝裂原活化蛋白激酶(MAPK)胞外信号调节激酶5 (ERK5)通路均促进黑色素瘤生长,并且ERK5激活是HH/GLI依赖性黑色素瘤细胞增殖所必需的。在这里,我们探讨了ERK5是否调节HH/GLI信号。方法:采用基因(ERK5特异性shRNA)和药理学(ERK5抑制剂JWG-071和AX15836, MAPK/ERK激酶5、MEK5抑制剂GW284543和BIX02189)靶向方法。利用GLI结合位点荧光素酶报告基因进行荧光素酶测定,以评估GLI的转录活性。MEK5的组成活性形式(MEK5DD)被用来诱导ERK5的激活。在黑色素瘤细胞中进行三维球体检测。结果:遗传和药理学抑制ERK5可降低NIH/3T3细胞中GLI1和GLI2蛋白水平以及激动剂SAG诱导的内源性HH/GLI通路的转录活性。在这些细胞中,MEK5DD过表达增强了SAG诱导的内源性HH/GLI通路的转录活性,而ERK5沉默则阻止了这种作用。一致地,MEK5DD过表达增加GLI1和GLI2蛋白水平。在黑色素瘤细胞中,ERK5沉默降低GLI1和GLI2 mRNA和蛋白水平,抑制GLI转录活性。MEK5DD进一步增加HH/GLI通路的转录活性和GLI1蛋白水平。GLI和MEK5抑制剂联合使用在减少黑色素瘤球形生长方面比单一治疗更有效。结论:MEK5-ERK5是GLI转录因子的激活因子,联合靶向这些途径作为黑色素瘤的潜在创新治疗方法值得进一步的临床前研究。
{"title":"The MEK5/ERK5 pathway promotes the activation of the Hedgehog/GLI signaling in melanoma cells.","authors":"Ignazia Tusa, Sinforosa Gagliardi, Alessio Menconi, Luisa Maresca, Alessandro Tubita, Matteo Lulli, Barbara Stecca, Elisabetta Rovida","doi":"10.1007/s13402-025-01050-z","DOIUrl":"https://doi.org/10.1007/s13402-025-01050-z","url":null,"abstract":"<p><strong>Purpose: </strong>Malignant melanoma is the deadliest skin cancer, with a poor prognosis in advanced stages. We reported that both Hedgehog-GLI (HH/GLI) and Mitogen-activated protein Kinase (MAPK) extracellular signal-regulated kinase 5 (ERK5) pathways promote melanoma growth, and that ERK5 activation is required for HH/GLI-dependent melanoma cell proliferation. Here, we explored whether ERK5 regulates HH/GLI signaling.</p><p><strong>Methods: </strong>Both genetic (using ERK5-specific shRNA) and pharmacologic (using the ERK5 inhibitors JWG-071 and AX15836, and the MAPK/ERK kinase 5, MEK5 inhibitors GW284543 and BIX02189) targeting approaches were used. Luciferase assay using the GLI-binding site luciferase reporter was performed to evaluate GLI transcriptional activity. A constitutively active form of MEK5 (MEK5DD) was used to induce ERK5 activation. 3D spheroid assays were performed in melanoma cells.</p><p><strong>Results: </strong>Genetic and pharmacologic ERK5 inhibition reduces GLI1 and GLI2 protein levels and transcriptional activity of endogenous HH/GLI pathway induced by the agonist SAG in NIH/3T3 cells. In these cells, MEK5DD overexpression potentiates transcriptional activity of endogenous HH/GLI pathway induced by SAG, whereas ERK5 silencing prevents this effect. Consistently, MEK5DD overexpression increases GLI1 and GLI2 protein levels. In melanoma cells, ERK5 silencing reduces GLI1 and GLI2 mRNA and protein levels and inhibits GLI transcriptional activity. MEK5DD further increases the transcriptional activity of the HH/GLI pathway and GLI1 protein levels. Combination of GLI and MEK5 inhibitors is more effective than single treatments in reducing melanoma spheroid growth.</p><p><strong>Conclusions: </strong>MEK5-ERK5 is an activator of GLI transcription factors, and combined targeting of these pathways warrants further preclinical investigation as a potential innovative therapeutic approach for melanoma.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":""},"PeriodicalIF":4.9,"publicationDate":"2025-02-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143494382","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Tumor-infiltrating myeloid cells; mechanisms, functional significance, and targeting in cancer therapy. 肿瘤浸润性骨髓细胞;肿瘤治疗的机制、功能意义和靶向性。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-02-25 DOI: 10.1007/s13402-025-01051-y
Fatemeh Sadat Toghraie, Maryam Bayat, Mahsa Sadat Hosseini, Amin Ramezani

Tumor-infiltrating myeloid cells (TIMs), which encompass tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), myeloid-derived suppressor cells (MDSCs), and tumor-associated dendritic cells (TADCs), are of great importance in tumor microenvironment (TME) and are integral to both pro- and anti-tumor immunity. Nevertheless, the phenotypic heterogeneity and functional plasticity of TIMs have posed challenges in fully understanding their complexity roles within the TME. Emerging evidence suggested that the presence of TIMs is frequently linked to prevention of cancer treatment and improvement of patient outcomes and survival. Given their pivotal function in the TME, TIMs have recently been recognized as critical targets for therapeutic approaches aimed at augmenting immunostimulatory myeloid cell populations while depleting or modifying those that are immunosuppressive. This review will explore the important properties of TIMs related to immunity, angiogenesis, and metastasis. We will also document the latest therapeutic strategies targeting TIMs in preclinical and clinical settings. Our objective is to illustrate the potential of TIMs as immunological targets that may improve the outcomes of existing cancer treatments.

肿瘤浸润性髓样细胞(TIMs)包括肿瘤相关巨噬细胞(tam)、肿瘤相关中性粒细胞(tan)、髓源性抑制细胞(MDSCs)和肿瘤相关树突状细胞(TADCs),在肿瘤微环境(TME)中非常重要,是肿瘤前免疫和抗肿瘤免疫的组成部分。然而,TIMs的表型异质性和功能可塑性给充分理解其在TME中的复杂作用带来了挑战。新出现的证据表明,TIMs的存在通常与癌症治疗的预防和患者预后和生存的改善有关。考虑到TIMs在TME中的关键功能,TIMs最近被认为是治疗方法的关键靶点,旨在增加免疫刺激骨髓细胞群,同时消耗或修饰那些免疫抑制细胞群。本文将探讨TIMs在免疫、血管生成和转移等方面的重要特性。我们还将在临床前和临床环境中记录针对TIMs的最新治疗策略。我们的目标是阐明TIMs作为免疫靶点的潜力,它可能改善现有癌症治疗的结果。
{"title":"Tumor-infiltrating myeloid cells; mechanisms, functional significance, and targeting in cancer therapy.","authors":"Fatemeh Sadat Toghraie, Maryam Bayat, Mahsa Sadat Hosseini, Amin Ramezani","doi":"10.1007/s13402-025-01051-y","DOIUrl":"https://doi.org/10.1007/s13402-025-01051-y","url":null,"abstract":"<p><p>Tumor-infiltrating myeloid cells (TIMs), which encompass tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), myeloid-derived suppressor cells (MDSCs), and tumor-associated dendritic cells (TADCs), are of great importance in tumor microenvironment (TME) and are integral to both pro- and anti-tumor immunity. Nevertheless, the phenotypic heterogeneity and functional plasticity of TIMs have posed challenges in fully understanding their complexity roles within the TME. Emerging evidence suggested that the presence of TIMs is frequently linked to prevention of cancer treatment and improvement of patient outcomes and survival. Given their pivotal function in the TME, TIMs have recently been recognized as critical targets for therapeutic approaches aimed at augmenting immunostimulatory myeloid cell populations while depleting or modifying those that are immunosuppressive. This review will explore the important properties of TIMs related to immunity, angiogenesis, and metastasis. We will also document the latest therapeutic strategies targeting TIMs in preclinical and clinical settings. Our objective is to illustrate the potential of TIMs as immunological targets that may improve the outcomes of existing cancer treatments.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":""},"PeriodicalIF":4.9,"publicationDate":"2025-02-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143494448","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Meningeal leukaemic aggregates as foci of cell expansion and chemoresistance in acute lymphoblastic leukaemia metastasis. 急性淋巴细胞白血病转移中脑膜白血病聚集体作为细胞扩增和化疗耐药的病灶。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-02-12 DOI: 10.1007/s13402-025-01043-y
Paula Ortiz-Sánchez, Sara González-Soto, Luz H Villamizar, Jaris Valencia, Eva Jiménez, Rosa Sacedón, Manuel Ramírez, Isabel Fariñas, Alberto Varas, Lidia M Fernández-Sevilla, Ángeles Vicente

Purpose: Central nervous system (CNS) involvement and/or relapse remains one of the most important causes of morbidity/mortality in paediatric B-cell precursor acute lymphoblastic leukaemia (BCP-ALL) patients. To identify novel therapeutic targets and develop less aggressive therapies, a better understanding of the cellular and molecular microenvironment in leptomeningeal metastases is key. Here, we aimed to investigate the formation of metastatic leptomeningeal aggregates and their relevance to the expansion, survival and chemoresistance acquisition of leukaemia cells.

Methods: We used BCP-ALL xenograft mouse models, combined with immunohistofluorescence and flow cytometry, to study the development of CNS metastasis and the contribution of leptomeningeal cells to the organisation of leukaemic aggregates. To in vitro mimic the CNS metastasis, we established co-cultures of three-dimensional (3D) ALL cell spheroids and human leptomeningeal cells (hLMCs) and studied the effects on gene expression, proliferation, cytokine production, and chemoresistance.

Results: In xenografted mice, ALL cells infiltrated the CNS at an early stage and, after crossing an ER-TR7+ fibroblast-like meningeal cell layer, they proliferated extensively and formed large vascularised leukaemic aggregates supported by a network of podoplanin+ leptomeningeal cells. In leukaemia spheroid-hLMC co-cultures, unlike conventional 2D co-cultures, meningeal cells strongly promoted the proliferation of leukaemic cells and generated a pro-inflammatory microenvironment. Furthermore, in 3D cell aggregates, leukaemic cells also developed chemoresistance, at least in part due to ABC transporter up-regulation.

Conclusion: Our results provide evidence for the formation of metastatic ALL-leptomeningeal cell aggregates, their pro-inflammatory profile and their contribution to leukaemic cell expansion, survival and chemoresistance in the CNS.

目的:中枢神经系统(CNS)受累和/或复发仍然是儿童b细胞前体急性淋巴细胞白血病(BCP-ALL)患者发病/死亡的最重要原因之一。为了确定新的治疗靶点和开发更低侵袭性的治疗方法,更好地了解脑轻脑膜转移的细胞和分子微环境是关键。在这里,我们的目的是研究转移性轻脑膜聚集体的形成及其与白血病细胞扩张、存活和化疗耐药获得的相关性。方法:采用BCP-ALL异种移植小鼠模型,结合免疫组织荧光和流式细胞术,研究中枢神经系统转移的发生及轻脑膜细胞在白血病聚集体组织中的作用。为了体外模拟中枢神经系统转移,我们建立了三维(3D) ALL细胞球体和人瘦脑膜细胞(hLMCs)共培养,研究了它们对基因表达、增殖、细胞因子产生和化疗耐药的影响。结果:在异种移植小鼠中,ALL细胞早期浸润中枢神经系统,在穿过ER-TR7+成纤维细胞样脑膜细胞层后,它们广泛增殖并形成由podoplanin+轻脑膜细胞网络支持的大血管化白血病聚集体。在白血病球体- hlmc共培养中,与传统的二维共培养不同,脑膜细胞强烈促进白血病细胞的增殖并产生促炎微环境。此外,在三维细胞聚集体中,白血病细胞也产生了化疗耐药,至少部分原因是ABC转运蛋白上调。结论:我们的研究结果为转移性all - leptomeneneal细胞聚集体的形成、它们的促炎特征以及它们在中枢神经系统中对白血病细胞扩增、存活和化疗耐药的贡献提供了证据。
{"title":"Meningeal leukaemic aggregates as foci of cell expansion and chemoresistance in acute lymphoblastic leukaemia metastasis.","authors":"Paula Ortiz-Sánchez, Sara González-Soto, Luz H Villamizar, Jaris Valencia, Eva Jiménez, Rosa Sacedón, Manuel Ramírez, Isabel Fariñas, Alberto Varas, Lidia M Fernández-Sevilla, Ángeles Vicente","doi":"10.1007/s13402-025-01043-y","DOIUrl":"https://doi.org/10.1007/s13402-025-01043-y","url":null,"abstract":"<p><strong>Purpose: </strong>Central nervous system (CNS) involvement and/or relapse remains one of the most important causes of morbidity/mortality in paediatric B-cell precursor acute lymphoblastic leukaemia (BCP-ALL) patients. To identify novel therapeutic targets and develop less aggressive therapies, a better understanding of the cellular and molecular microenvironment in leptomeningeal metastases is key. Here, we aimed to investigate the formation of metastatic leptomeningeal aggregates and their relevance to the expansion, survival and chemoresistance acquisition of leukaemia cells.</p><p><strong>Methods: </strong>We used BCP-ALL xenograft mouse models, combined with immunohistofluorescence and flow cytometry, to study the development of CNS metastasis and the contribution of leptomeningeal cells to the organisation of leukaemic aggregates. To in vitro mimic the CNS metastasis, we established co-cultures of three-dimensional (3D) ALL cell spheroids and human leptomeningeal cells (hLMCs) and studied the effects on gene expression, proliferation, cytokine production, and chemoresistance.</p><p><strong>Results: </strong>In xenografted mice, ALL cells infiltrated the CNS at an early stage and, after crossing an ER-TR7<sup>+</sup> fibroblast-like meningeal cell layer, they proliferated extensively and formed large vascularised leukaemic aggregates supported by a network of podoplanin<sup>+</sup> leptomeningeal cells. In leukaemia spheroid-hLMC co-cultures, unlike conventional 2D co-cultures, meningeal cells strongly promoted the proliferation of leukaemic cells and generated a pro-inflammatory microenvironment. Furthermore, in 3D cell aggregates, leukaemic cells also developed chemoresistance, at least in part due to ABC transporter up-regulation.</p><p><strong>Conclusion: </strong>Our results provide evidence for the formation of metastatic ALL-leptomeningeal cell aggregates, their pro-inflammatory profile and their contribution to leukaemic cell expansion, survival and chemoresistance in the CNS.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":""},"PeriodicalIF":4.9,"publicationDate":"2025-02-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143400453","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Pathways and outputs orchestrated in tumor microenvironment cells by hypoxia-induced tumor-derived exosomes in pan-cancer. 缺氧诱导的泛癌肿瘤源性外泌体在肿瘤微环境细胞中调控通路和输出。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-02-10 DOI: 10.1007/s13402-025-01042-z
Ozel Capik, Omer Faruk Karatas

Hypoxia is a critical microenvironmental condition that plays a major role in driving tumorigenesis and cancer progression. Increasing evidence has revealed novel functions of hypoxia in intercellular communication. The hypoxia induced tumor derived exosomes (hiTDExs) released in high quantities by tumor cells under hypoxia are packed with unique cargoes that are essential for cancer cells' interactions within their microenvironment. These hiTDExs facilitate not only immune evasion but also promote cancer cell growth, survival, angiogenesis, EMT, resistance to therapy, and the metastatic spread of the disease. Nevertheless, direct interventions targeting hypoxia signaling in cancer therapy face challenges related to tumor progression and resistance, limiting their clinical effectiveness. Therefore, deepening our understanding of the molecular processes through which hiTDExs remodels tumors and their microenvironment, as well as how tumor cells adjust to hypoxic conditions, remains essential. This knowledge will pave the way for novel approaches in treating hypoxic tumors. In this review, we discuss recent work revealing the hiTDExs mediated interactions between tumor and its microenvironment. We have described key hiTDExs cargos (lncRNA, circRNAs, cytokines, etc.) and their targets in the receipt cells, responsible for various biological effects. Moreover, we emphasized the importance of hiTDExs as versatile elements of cell communication in the tumor microenvironment. Finally, we highlighted the effects of hiTDExs on the molecular changes in target cells by executing molecular cargo transfer between cells and altering signaling pathways. Currently, hiTDExs show promise in the treatment of diseases. Understanding the molecular processes through which hiTDExs influence tumor behavior and their microenvironment, along with how tumor cells adapt to and survive in low-oxygen conditions, remains a central focus in cancer research, paving the way for innovative strategies in treating hypoxic tumors and enhancing immunotherapy.

缺氧是一个关键的微环境条件,在驱动肿瘤发生和癌症进展中起着重要作用。越来越多的证据揭示了缺氧在细胞间通讯中的新功能。肿瘤细胞在缺氧条件下大量释放的缺氧诱导肿瘤衍生外泌体(hiTDExs)装载着独特的货物,这些货物对于癌细胞在其微环境内的相互作用至关重要。这些hitdex不仅促进免疫逃避,而且还促进癌细胞生长、存活、血管生成、EMT、对治疗的抵抗以及疾病的转移性扩散。然而,针对缺氧信号的直接干预在癌症治疗中面临着与肿瘤进展和耐药性相关的挑战,限制了其临床有效性。因此,加深我们对hitdex重塑肿瘤及其微环境的分子过程,以及肿瘤细胞如何适应缺氧条件的理解仍然是必要的。这些知识将为治疗缺氧肿瘤的新方法铺平道路。在这篇综述中,我们讨论了揭示hiTDExs介导的肿瘤及其微环境之间相互作用的最新工作。我们描述了关键的hiTDExs货物(lncRNA, circrna, cytokines等)及其在接收细胞中的靶标,负责各种生物效应。此外,我们强调了hitdex作为肿瘤微环境中细胞通信的通用元件的重要性。最后,我们强调了hiTDExs通过在细胞之间执行分子货物转移和改变信号通路对靶细胞中分子变化的影响。目前,hiTDExs在治疗疾病方面显示出前景。了解hiTDExs影响肿瘤行为及其微环境的分子过程,以及肿瘤细胞如何适应和在低氧条件下生存,仍然是癌症研究的中心焦点,为治疗缺氧肿瘤和增强免疫治疗的创新策略铺平道路。
{"title":"Pathways and outputs orchestrated in tumor microenvironment cells by hypoxia-induced tumor-derived exosomes in pan-cancer.","authors":"Ozel Capik, Omer Faruk Karatas","doi":"10.1007/s13402-025-01042-z","DOIUrl":"https://doi.org/10.1007/s13402-025-01042-z","url":null,"abstract":"<p><p>Hypoxia is a critical microenvironmental condition that plays a major role in driving tumorigenesis and cancer progression. Increasing evidence has revealed novel functions of hypoxia in intercellular communication. The hypoxia induced tumor derived exosomes (hiTDExs) released in high quantities by tumor cells under hypoxia are packed with unique cargoes that are essential for cancer cells' interactions within their microenvironment. These hiTDExs facilitate not only immune evasion but also promote cancer cell growth, survival, angiogenesis, EMT, resistance to therapy, and the metastatic spread of the disease. Nevertheless, direct interventions targeting hypoxia signaling in cancer therapy face challenges related to tumor progression and resistance, limiting their clinical effectiveness. Therefore, deepening our understanding of the molecular processes through which hiTDExs remodels tumors and their microenvironment, as well as how tumor cells adjust to hypoxic conditions, remains essential. This knowledge will pave the way for novel approaches in treating hypoxic tumors. In this review, we discuss recent work revealing the hiTDExs mediated interactions between tumor and its microenvironment. We have described key hiTDExs cargos (lncRNA, circRNAs, cytokines, etc.) and their targets in the receipt cells, responsible for various biological effects. Moreover, we emphasized the importance of hiTDExs as versatile elements of cell communication in the tumor microenvironment. Finally, we highlighted the effects of hiTDExs on the molecular changes in target cells by executing molecular cargo transfer between cells and altering signaling pathways. Currently, hiTDExs show promise in the treatment of diseases. Understanding the molecular processes through which hiTDExs influence tumor behavior and their microenvironment, along with how tumor cells adapt to and survive in low-oxygen conditions, remains a central focus in cancer research, paving the way for innovative strategies in treating hypoxic tumors and enhancing immunotherapy.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":""},"PeriodicalIF":4.9,"publicationDate":"2025-02-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143383891","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Predicting the early therapeutic response to hepatic artery infusion chemotherapy in patients with unresectable HCC using a contrast-enhanced computed tomography-based habitat radiomics model: a multi-center retrospective study. 使用基于对比增强计算机断层扫描的生境放射组学模型预测不可切除的肝癌患者对肝动脉灌注化疗的早期治疗反应:一项多中心回顾性研究。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-02-04 DOI: 10.1007/s13402-025-01041-0
Mingsong Wu, Zenglong Que, Shujie Lai, Guanhui Li, Jie Long, Yuqin He, Shunan Wang, Hao Wu, Nan You, Xiang Lan, Liangzhi Wen

Objective: Predicting the therapeutic response before initiation of hepatic artery infusion chemotherapy (HAIC) with fluorouracil, leucovorin, and oxaliplatin (FOLFOX) remains challenging for patients with unresectable hepatocellular carcinoma (HCC). Herein, we investigated the potential of a contrast-enhanced CT-based habitat radiomics model as a novel approach for predicting the early therapeutic response to HAIC-FOLFOX in patients with unresectable HCC.

Methods: A total of 148 patients with unresectable HCC who received HAIC-FOLFOX combined with targeted therapy or immunotherapy at three tertiary care medical centers were enrolled retrospectively. Tumor habitat features were extracted from subregion radiomics based on CECT at different phases using k-means clustering. Logistic regression was used to construct the model. This CECT-based habitat radiomics model was verified by bootstrapping and compared with a model based on clinical variables. Model performance was evaluated using the area under the curve (AUC) and a calibration curve.

Results: Three intratumoral habitats with high, moderate, and low enhancement were identified to construct a habitat radiomics model for therapeutic response prediction. Patients with a greater proportion of high-enhancement intratumoral habitat showed better therapeutic responses. The AUC of the habitat radiomics model was 0.857 (95% CI: 0.798-0.916), and the bootstrap-corrected concordance index was 0.842 (95% CI: 0.785-0.907), resulting in a better predictive value than the clinical variable-based model, which had an AUC of 0.757 (95% CI: 0.679-0.834).

Conclusion: The CECT-based habitat radiomics model is an effective, visualized, and noninvasive tool for predicting the early therapeutic response of patients with unresectable HCC to HAIC-FOLFOX treatment and could guide clinical management and decision-making.

{"title":"Predicting the early therapeutic response to hepatic artery infusion chemotherapy in patients with unresectable HCC using a contrast-enhanced computed tomography-based habitat radiomics model: a multi-center retrospective study.","authors":"Mingsong Wu, Zenglong Que, Shujie Lai, Guanhui Li, Jie Long, Yuqin He, Shunan Wang, Hao Wu, Nan You, Xiang Lan, Liangzhi Wen","doi":"10.1007/s13402-025-01041-0","DOIUrl":"https://doi.org/10.1007/s13402-025-01041-0","url":null,"abstract":"<p><strong>Objective: </strong>Predicting the therapeutic response before initiation of hepatic artery infusion chemotherapy (HAIC) with fluorouracil, leucovorin, and oxaliplatin (FOLFOX) remains challenging for patients with unresectable hepatocellular carcinoma (HCC). Herein, we investigated the potential of a contrast-enhanced CT-based habitat radiomics model as a novel approach for predicting the early therapeutic response to HAIC-FOLFOX in patients with unresectable HCC.</p><p><strong>Methods: </strong>A total of 148 patients with unresectable HCC who received HAIC-FOLFOX combined with targeted therapy or immunotherapy at three tertiary care medical centers were enrolled retrospectively. Tumor habitat features were extracted from subregion radiomics based on CECT at different phases using k-means clustering. Logistic regression was used to construct the model. This CECT-based habitat radiomics model was verified by bootstrapping and compared with a model based on clinical variables. Model performance was evaluated using the area under the curve (AUC) and a calibration curve.</p><p><strong>Results: </strong>Three intratumoral habitats with high, moderate, and low enhancement were identified to construct a habitat radiomics model for therapeutic response prediction. Patients with a greater proportion of high-enhancement intratumoral habitat showed better therapeutic responses. The AUC of the habitat radiomics model was 0.857 (95% CI: 0.798-0.916), and the bootstrap-corrected concordance index was 0.842 (95% CI: 0.785-0.907), resulting in a better predictive value than the clinical variable-based model, which had an AUC of 0.757 (95% CI: 0.679-0.834).</p><p><strong>Conclusion: </strong>The CECT-based habitat radiomics model is an effective, visualized, and noninvasive tool for predicting the early therapeutic response of patients with unresectable HCC to HAIC-FOLFOX treatment and could guide clinical management and decision-making.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":""},"PeriodicalIF":4.9,"publicationDate":"2025-02-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143191102","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Cellular Oncology
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1