Pub Date : 2025-04-07DOI: 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.
{"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}
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.
{"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}
Pub Date : 2025-04-07DOI: 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}
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.
{"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}
Pub Date : 2025-03-03DOI: 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.
{"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}
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.
{"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}
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.
{"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}
Pub Date : 2025-02-12DOI: 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.
{"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}
Pub Date : 2025-02-10DOI: 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.
{"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}
Pub Date : 2025-02-04DOI: 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}