Molecular OncologyPub Date : 2024-10-01Epub Date: 2024-05-21DOI: 10.1002/1878-0261.13663
Terezia Kurucova, Kamila Reblova, Pavlina Janovska, Jakub Pawel Porc, Veronika Navrkalova, Sarka Pavlova, Jitka Malcikova, Karla Plevova, Boris Tichy, Michael Doubek, Vitezslav Bryja, Jana Kotaskova, Sarka Pospisilova
{"title":"Unveiling the dynamics and molecular landscape of a rare chronic lymphocytic leukemia subpopulation driving refractoriness: insights from single-cell RNA sequencing.","authors":"Terezia Kurucova, Kamila Reblova, Pavlina Janovska, Jakub Pawel Porc, Veronika Navrkalova, Sarka Pavlova, Jitka Malcikova, Karla Plevova, Boris Tichy, Michael Doubek, Vitezslav Bryja, Jana Kotaskova, Sarka Pospisilova","doi":"10.1002/1878-0261.13663","DOIUrl":"10.1002/1878-0261.13663","url":null,"abstract":"<p><p>Early identification of resistant cancer cells is currently a major challenge, as their expansion leads to refractoriness. To capture the dynamics of these cells, we made a comprehensive analysis of disease progression and treatment response in a chronic lymphocytic leukemia (CLL) patient using a combination of single-cell and bulk genomic methods. At diagnosis, the patient presented with unfavorable genetic markers, including notch receptor 1 (NOTCH1) mutation and loss(11q). The initial and subsequent treatment lines did not lead to a durable response and the patient developed refractory disease. Refractory CLL cells featured substantial dysregulation in B-cell phenotypic markers such as human leukocyte antigen (HLA) genes, immunoglobulin (IG) genes, CD19 molecule (CD19), membrane spanning 4-domains A1 (MS4A1; previously known as CD20), CD79a molecule (CD79A) and paired box 5 (PAX5), indicating B-cell de-differentiation and disease transformation. We described the clonal evolution and characterized in detail two cell populations that emerged during the refractory disease phase, differing in the presence of high genomic complexity. In addition, we successfully tracked the cells with high genomic complexity back to the time before treatment, where they formed a rare subpopulation. We have confirmed that single-cell RNA sequencing enables the characterization of refractory cells and the monitoring of their development over time.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":"2541-2553"},"PeriodicalIF":6.6,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11459043/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141071526","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Molecular OncologyPub Date : 2024-10-01Epub Date: 2024-06-16DOI: 10.1002/1878-0261.13672
Ulduz Sobhiafshar, Betül Çakici, Erdem Yilmaz, Nalan Yildiz Ayhan, Laila Hedaya, Mustafa Can Ayhan, Cansu Yerinde, Yasemin Begüm Alankuş, H Kübra Gürkaşlar, Elif Nur Firat-Karalar, N C Tolga Emre
{"title":"Interferon regulatory factor 4 modulates epigenetic silencing and cancer-critical pathways in melanoma cells.","authors":"Ulduz Sobhiafshar, Betül Çakici, Erdem Yilmaz, Nalan Yildiz Ayhan, Laila Hedaya, Mustafa Can Ayhan, Cansu Yerinde, Yasemin Begüm Alankuş, H Kübra Gürkaşlar, Elif Nur Firat-Karalar, N C Tolga Emre","doi":"10.1002/1878-0261.13672","DOIUrl":"10.1002/1878-0261.13672","url":null,"abstract":"<p><p>Interferon regulatory factor 4 (IRF4) was initially identified as a key controller in lymphocyte differentiation and function, and subsequently as a dependency factor and therapy target in lymphocyte-derived cancers. In melanocytes, IRF4 takes part in pigmentation. Although genetic studies have implicated IRF4 in melanoma, how IRF4 functions in melanoma cells has remained largely elusive. Here, we confirmed prevalent IRF4 expression in melanoma and showed that high expression is linked to dependency in cells and mortality in patients. Analysis of genes activated by IRF4 uncovered, as a novel target category, epigenetic silencing factors involved in DNA methylation (DNMT1, DNMT3B, UHRF1) and histone H3K27 methylation (EZH2). Consequently, we show that IRF4 controls the expression of tumour suppressor genes known to be silenced by these epigenetic modifications, for instance cyclin-dependent kinase inhibitors CDKN1A and CDKN1B, the PI3-AKT pathway regulator PTEN, and primary cilium components. Furthermore, IRF4 modulates activity of key downstream oncogenic pathways, such as WNT/β-catenin and AKT, impacting cell proliferation and survival. Accordingly, IRF4 modifies the effectiveness of pertinent epigenetic drugs on melanoma cells, a finding that encourages further studies towards therapeutic targeting of IRF4 in melanoma.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":"2423-2448"},"PeriodicalIF":6.6,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11459048/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141331358","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Molecular OncologyPub Date : 2024-10-01Epub Date: 2024-05-16DOI: 10.1002/1878-0261.13655
Lilly Anne Torland, Xiaoran Lai, Surendra Kumar, Margit H Riis, Jürgen Geisler, Torben Lüders, Xavier Tekpli, Vessela Kristensen, Kristine Sahlberg, Andliena Tahiri
{"title":"Benign breast tumors may arise on different immunological backgrounds.","authors":"Lilly Anne Torland, Xiaoran Lai, Surendra Kumar, Margit H Riis, Jürgen Geisler, Torben Lüders, Xavier Tekpli, Vessela Kristensen, Kristine Sahlberg, Andliena Tahiri","doi":"10.1002/1878-0261.13655","DOIUrl":"10.1002/1878-0261.13655","url":null,"abstract":"<p><p>Benign breast tumors are a nonthreatening condition defined as abnormal cell growth within the breast without the ability to invade nearby tissue. However, benign lesions hold valuable biological information that can lead us toward better understanding of tumor biology. In this study, we have used two pathway analysis algorithms, Pathifier and gene set variation analysis (GSVA), to identify biological differences between normal breast tissue, benign tumors and malignant tumors in our clinical dataset. Our results revealed that one-third of all pathways that were significantly different between benign and malignant tumors were immune-related pathways, and 227 of them were validated by both methods and in the METABRIC dataset. Furthermore, five of these pathways (all including genes involved in cytokine and interferon signaling) were related to overall survival in cancer patients in both datasets. The cellular moieties that contribute to immune differences in malignant and benign tumors were analyzed using the deconvolution tool, CIBERSORT. The results showed that levels of some immune cells were specifically higher in benign than in malignant tumors, and this was especially the case for resting dendritic cells and follicular T-helper cells. Understanding the distinct immune profiles of benign and malignant breast tumors may aid in developing noninvasive diagnostic methods to differentiate between them in the future.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":"2495-2509"},"PeriodicalIF":6.6,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11459044/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140957919","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Molecular OncologyPub Date : 2024-10-01Epub Date: 2024-05-28DOI: 10.1002/1878-0261.13660
Jonas Wohlfarth, Corinna Kosnopfel, Dominic Faber, Marion Berthold, Claudia Siedel, Melissa Bernhardt, Andreas Schlosser, Tyler Aprati, David Liu, David Schrama, Roland Houben, Dirk Schadendorf, Matthias Goebeler, Svenja Meierjohann, Bastian Schilling
{"title":"Loss of p14 diminishes immunogenicity in melanoma via non-canonical Wnt signaling by reducing the peptide surface density.","authors":"Jonas Wohlfarth, Corinna Kosnopfel, Dominic Faber, Marion Berthold, Claudia Siedel, Melissa Bernhardt, Andreas Schlosser, Tyler Aprati, David Liu, David Schrama, Roland Houben, Dirk Schadendorf, Matthias Goebeler, Svenja Meierjohann, Bastian Schilling","doi":"10.1002/1878-0261.13660","DOIUrl":"10.1002/1878-0261.13660","url":null,"abstract":"<p><p>Immunotherapy has achieved tremendous success in melanoma. However, only around 50% of advanced melanoma patients benefit from immunotherapy. Cyclin-dependent kinase inhibitor 2A (CDKN2A), encoding the two tumor-suppressor proteins p14<sup>ARF</sup> and p16<sup>INK4a</sup>, belongs to the most frequently inactivated gene loci in melanoma and leads to decreased T cell infiltration. While the role of p16<sup>INK4a</sup> has been extensively investigated, knowledge about p14<sup>ARF</sup> in melanoma is scarce. In this study, we elucidate the impact of reduced p14<sup>ARF</sup> expression on melanoma immunogenicity. Knockdown of p14<sup>ARF</sup> in melanoma cell lines diminished their recognition and killing by melanoma differentiation antigen (MDA)-specific T cells. Resistance was caused by a reduction of the peptide surface density of presented MDAs. Immunopeptidomic analyses revealed that antigen presentation via human leukocyte antigen class I (HLA-I) molecules was enhanced upon p14<sup>ARF</sup> downregulation in general, but absolute and relative expression of cognate peptides was decreased. However, this phenotype is associated with a favorable outcome for melanoma patients. Limiting Wnt5a signaling reverted this phenotype, suggesting an involvement of non-canonical Wnt signaling. Taken together, our data indicate a new mechanism limiting MDA-specific T cell responses by decreasing both absolute and relative MDA-peptide presentation in melanoma.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":"2449-2470"},"PeriodicalIF":6.6,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11459041/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141161997","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Molecular OncologyPub Date : 2024-10-01Epub Date: 2024-05-22DOI: 10.1002/1878-0261.13666
Jing Liang, Aishwarya Gondane, Harri M Itkonen
{"title":"CDK12-inactivation-induced MYC signaling causes dependency on the splicing kinase SRPK1.","authors":"Jing Liang, Aishwarya Gondane, Harri M Itkonen","doi":"10.1002/1878-0261.13666","DOIUrl":"10.1002/1878-0261.13666","url":null,"abstract":"<p><p>Inactivation of cyclin-dependent kinase 12 (CDK12) characterizes an aggressive sub-group of castration-resistant prostate cancer (CRPC). Hyper-activation of MYC transcription factor is sufficient to confer the CRPC phenotype. Here, we show that loss of CDK12 promotes MYC activity, which renders the cells dependent on the otherwise non-essential splicing regulatory kinase SRSF protein kinase 1 (SRPK1). High MYC expression is associated with increased levels of SRPK1 in patient samples, and overexpression of MYC sensitizes prostate cancer cells to SRPK1 inhibition using pharmacological and genetic strategies. We show that Endovion (SCO-101), a compound currently in clinical trials against pancreatic cancer, phenocopies the effects of the well-characterized SRPK1 inhibitor SRPIN340 on nascent transcription. This is the first study to show that Endovion is an SRPK1 inhibitor. Inhibition of SRPK1 with either of the compounds promotes transcription elongation, and transcriptionally activates the unfolded protein response. In brief, here we discover that CDK12 inactivation promotes MYC signaling in an SRPK1-dependent manner, and show that the clinical grade compound Endovion selectively targets the cells with CDK12 inactivation.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":"2510-2523"},"PeriodicalIF":6.6,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11459032/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141076257","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Molecular OncologyPub Date : 2024-10-01Epub Date: 2024-07-02DOI: 10.1002/1878-0261.13695
Bastian Krenz, Jongkuen Lee, Toshitha Kannan, Martin Eilers
{"title":"Immune evasion: An imperative and consequence of MYC deregulation.","authors":"Bastian Krenz, Jongkuen Lee, Toshitha Kannan, Martin Eilers","doi":"10.1002/1878-0261.13695","DOIUrl":"10.1002/1878-0261.13695","url":null,"abstract":"<p><p>MYC has been implicated in the pathogenesis of a wide range of human tumors and has been described for many years as a transcription factor that regulates genes with pleiotropic functions to promote tumorigenic growth. However, despite extensive efforts to identify specific target genes of MYC that alone could be responsible for promoting tumorigenesis, the field is yet to reach a consensus whether this is the crucial function of MYC. Recent work shifts the view on MYC's function from being a gene-specific transcription factor to an essential stress resilience factor. In highly proliferating cells, MYC preserves cell integrity by promoting DNA repair at core promoters, protecting stalled replication forks, and/or preventing transcription-replication conflicts. Furthermore, an increasing body of evidence demonstrates that MYC not only promotes tumorigenesis by driving cell-autonomous growth, but also enables tumors to evade the host's immune system. In this review, we summarize our current understanding of how MYC impairs antitumor immunity and why this function is evolutionarily hard-wired to the biology of the MYC protein family. We show why the cell-autonomous and immune evasive functions of MYC are mutually dependent and discuss ways to target MYC proteins in cancer therapy.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":"2338-2355"},"PeriodicalIF":6.6,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11459038/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141492641","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Molecular OncologyPub Date : 2024-10-01Epub Date: 2024-08-22DOI: 10.1002/1878-0261.13711
{"title":"Expression of Concern: T-LAK cell-originated protein kinase (TOPK): an emerging prognostic biomarker and therapeutic target in osteosarcoma.","authors":"","doi":"10.1002/1878-0261.13711","DOIUrl":"10.1002/1878-0261.13711","url":null,"abstract":"","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":"2601"},"PeriodicalIF":6.6,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11459047/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142018045","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Molecular OncologyPub Date : 2024-10-01Epub Date: 2024-07-19DOI: 10.1002/1878-0261.13698
Melica Nourmoussavi Brodeur, Pier Selenica, Weining Ma, Sara Moufarrij, Christian Dagher, Thais Basili, Nadeem R Abu-Rustum, Carol Aghajanian, Qin Zhou, Alexia Iasonos, Lora H Ellenson, Britta Weigelt, M Herman Chui
{"title":"ERBB2 mutations define a subgroup of endometrial carcinomas associated with high tumor mutational burden and the microsatellite instability-high (MSI-H) molecular subtype.","authors":"Melica Nourmoussavi Brodeur, Pier Selenica, Weining Ma, Sara Moufarrij, Christian Dagher, Thais Basili, Nadeem R Abu-Rustum, Carol Aghajanian, Qin Zhou, Alexia Iasonos, Lora H Ellenson, Britta Weigelt, M Herman Chui","doi":"10.1002/1878-0261.13698","DOIUrl":"10.1002/1878-0261.13698","url":null,"abstract":"<p><p>Anti-HER2 therapy is indicated for erb-b2 receptor tyrosine kinase 2 (ERBB2)-amplified/overexpressing endometrial carcinoma (EC). Mutations constitute another mode of ERBB2 activation, but only rare ERBB2-mutated ECs have been reported. We sought to characterize the clinicopathologic and genetic features of ERBB2-mutated EC. From an institutional cohort of 2638 ECs subjected to clinical tumor-normal panel sequencing, 69 (2.6%) with pathogenic ERBB2 mutation(s) were identified, of which 11 were also ERBB2-amplified. The most frequent ERBB2 hotspot mutations were V842I (38%) and R678Q (25%). ERBB2 mutations were clonal in 87% of evaluable cases. Immunohistochemistry revealed low HER2 protein expression in most ERBB2-mutated ECs (0/1+ in 66%, 2+ in 27%); all 3+ tumors (7.3%) were also ERBB2-amplified. Compared to ERBB2-wildtype ECs (with or without ERBB2 amplification), ERBB2-mutated/non-amplified ECs were enriched for the microsatellite instability-high (MSI-H) and, to a lesser extent, DNA polymerase epsilon, catalytic subunit (POLE) molecular subtypes, and associated with high tumor mutational burden and low chromosomal instability. Survival outcomes were similar between patients with ERBB2-mutated/non-amplified versus wildtype EC, whereas ERBB2 amplification was associated with worse prognosis on univariate, but not multivariate, analyses. In conclusion, ERBB2 mutation defines a rare subgroup of ECs that is pathogenically distinct from ERBB2-wildtype and ERBB2-amplified ECs.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":"2356-2368"},"PeriodicalIF":6.6,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11459037/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141731272","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Molecular OncologyPub Date : 2024-10-01Epub Date: 2024-06-25DOI: 10.1002/1878-0261.13679
Alexandre de Nonneville, Elsa Kalbacher, Francesco Cannone, Arnaud Guille, José Adelaïde, Pascal Finetti, Maria Cappiello, Eric Lambaudie, Giuseppe Ettore, Emmanuelle Charafe, Emilie Mamessier, Magali Provansal, François Bertucci, Renaud Sabatier
{"title":"Endometrioid ovarian carcinoma landscape: pathological and molecular characterization.","authors":"Alexandre de Nonneville, Elsa Kalbacher, Francesco Cannone, Arnaud Guille, José Adelaïde, Pascal Finetti, Maria Cappiello, Eric Lambaudie, Giuseppe Ettore, Emmanuelle Charafe, Emilie Mamessier, Magali Provansal, François Bertucci, Renaud Sabatier","doi":"10.1002/1878-0261.13679","DOIUrl":"10.1002/1878-0261.13679","url":null,"abstract":"<p><p>Endometrioid ovarian cancers (EOvC) are usually managed as serous tumors. In this study, we conducted a comprehensive molecular investigation to uncover the distinct biological characteristics of EOvC. This retrospective multicenter study involved patients from three European centers. We collected clinical data and formalin-fixed paraffin-embedded (FFPE) samples for analysis at the DNA level using panel-based next-generation sequencing and array-comparative genomic hybridization. Additionally, we examined mRNA expression using NanoString nCounter® and protein expression through tissue microarray. We compared EOvC with other ovarian subtypes and uterine endometrioid tumors. Furthermore, we assessed the impact of molecular alterations on patient outcomes, including progression-free survival (PFS) and overall survival (OS). Preliminary analysis of clinical data from 668 patients, including 86 (12.9%) EOvC, revealed more favorable prognosis for EOvC compared with serous ovarian carcinoma (5-year OS of 60% versus 45%; P = 0.001) driven by diagnosis at an earlier stage. Immunohistochemistry and copy number alteration (CNA) profiles of 43 cases with clinical data and FFPE samples available indicated that EOvC protein expression and CNA profiles were more similar to endometrioid endometrial tumors than to serous ovarian carcinomas. EOvC exhibited specific alterations, such as lower rates of PTEN loss, mutations in DNA repair genes, and P53 abnormalities. Survival analysis showed that patients with tumors harboring loss of PTEN expression had worse outcomes (median PFS 19.6 months vs. not reached; P = 0.034). Gene expression profile analysis confirmed that EOvC differed from serous tumors. However, comparison to other rare subtypes of ovarian cancer suggested that the EOvC transcriptomic profile was close to that of ovarian clear cell carcinoma. Downregulation of genes involved in the PI3K pathway and DNA methylation was observed in EOvC. In conclusion, EOvC represents a distinct biological entity and should be regarded as such in the development of specific clinical approaches.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":"2586-2600"},"PeriodicalIF":6.6,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11459045/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141458081","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Molecular OncologyPub Date : 2024-10-01Epub Date: 2024-07-31DOI: 10.1002/1878-0261.13673
Ting Ting Cai, Christophe Desterke, Juan Peng, Jean Agnetti, Peixuan Song, Dalila Ouazib, Alexandre Dos Santos, Catherine Guettier, Didier Samuel, Ama Gassama-Diagne
{"title":"Septin 9 expression regulates 'don't eat me' signals and identifies an immune-epithelial class of intrahepatic cholangiocarcinoma.","authors":"Ting Ting Cai, Christophe Desterke, Juan Peng, Jean Agnetti, Peixuan Song, Dalila Ouazib, Alexandre Dos Santos, Catherine Guettier, Didier Samuel, Ama Gassama-Diagne","doi":"10.1002/1878-0261.13673","DOIUrl":"10.1002/1878-0261.13673","url":null,"abstract":"<p><p>Intrahepatic cholangiocarcinoma (iCCA) is a highly heterogeneous and aggressive liver cancer with limited therapeutic options. Precise classification and immunotherapy are perspectives to improve the treatments. We reported the role of septin 9 in apico-basal polarity and epithelial-to-mesenchymal transition (EMT). Here, we aim to elucidate its role in iCCA. We analyzed single-cell transcriptomes from human iCCA tumor cells based on phenotype and cell state. Knockdown of the septin 9 gene (SEPT9) was done using small interfering RNA (siRNA); interferon-γ (IFN-γ) stimulation was performed using different CCA cells; gene expressions were analyzed by reverse transcription and real-time PCR analysis (RT-qPCR); and immunofluorescence, immunoblotting, and flow cytometry were performed to assess the expression of proteins. The differential distributions of SEPT9 and vimentin (VIM) gene expressions allowed us to define specific cellular trajectories of malignant cells and thus identified distinct clusters of iCCA cells. One cluster was enriched in VIM and extracellular-matrix (ECM) remodeling molecules, and another had high expression of SEPT9 and genes from the 'don't eat me' signal involved in immune escape. This antagonism between SEPT9 and VIM was confirmed by in vitro experiments. Notably, SEPT9 and 'don't eat me' gene expressions were inversely correlated to those of vimentin and the EMT markers. SEPT9 expression was upregulated by IFN-γ and SEPT9 knockdown decreased expression of 'don't eat me' signal genes and increased expression of mesenchymal markers. Cancer Cell Line Encyclopedia (CCLE) transcriptome database analyses confirmed that iCCA cells enriched in septin 9 exhibit epithelial-like features. This study revealed septin 9 as a cytoskeleton element of iCCA epithelial-like cells and a regulator of the immune system response. It also brings new insights into the enigmatic relationship between EMT and immune response. Notably, we decoded a potential mechanism that could sensitize patients to immunotherapies.</p>","PeriodicalId":18764,"journal":{"name":"Molecular Oncology","volume":" ","pages":"2369-2392"},"PeriodicalIF":6.6,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11459040/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141856020","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}