37 results on '"Francesca Citron"'
Search Results
2. miR‐9 modulates and predicts the response to radiotherapy and EGFR inhibition in HNSCC
- Author
-
Francesca Citron, Ilenia Segatto, Lorena Musco, Ilenia Pellarin, Gian Luca Rampioni Vinciguerra, Giovanni Franchin, Giuseppe Fanetti, Francesco Miccichè, Vittorio Giacomarra, Valentina Lupato, Andrea Favero, Isabella Concina, Sanjana Srinivasan, Michele Avanzo, Isabella Castiglioni, Luigi Barzan, Sandro Sulfaro, Gianluigi Petrone, Andrea Viale, Giulio F Draetta, Andrea Vecchione, Barbara Belletti, and Gustavo Baldassarre
- Subjects
EGFR inhibitors ,HNSCC ,KLF5 ,radiotherapy ,Sp1 ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Radiotherapy (RT) plus the anti‐EGFR monoclonal antibody Cetuximab (CTX) is an effective combination therapy for a subset of head and neck squamous cell carcinoma (HNSCC) patients. However, predictive markers of efficacy are missing, resulting in many patients treated with disappointing results and unnecessary toxicities. Here, we report that activation of EGFR upregulates miR‐9 expression, which sustains the aggressiveness of HNSCC cells and protects from RT‐induced cell death. Mechanistically, by targeting KLF5, miR‐9 regulates the expression of the transcription factor Sp1 that, in turn, stimulates tumor growth and confers resistance to RT+CTX in vitro and in vivo. Intriguingly, high miR‐9 levels have no effect on the sensitivity of HNSCC cells to cisplatin. In primary HNSCC, miR‐9 expression correlated with Sp1 mRNA levels and high miR‐9 expression predicted poor prognosis in patients treated with RT+CTX. Overall, we have discovered a new signaling axis linking EGFR activation to Sp1 expression that dictates the response to combination treatments in HNSCC. We propose that miR‐9 may represent a valuable biomarker to select which HNSCC patients might benefit from RT+CTX therapy.
- Published
- 2021
- Full Text
- View/download PDF
3. p27kip1 at the crossroad between actin and microtubule dynamics
- Author
-
Gian Luca Rampioni Vinciguerra, Francesca Citron, Ilenia Segatto, Barbara Belletti, Andrea Vecchione, and Gustavo Baldassarre
- Subjects
p27 ,Microtubule ,Actin ,Cytoskeleton ,Stathmin ,Migration ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 ,Cytology ,QH573-671 - Abstract
Abstract The p27kip1 protein, mainly known as a negative regulator of cell proliferation, has also been involved in the control of other cellular processes, including the regulation of cytoskeleton dynamics. Notably, these two functions involve distinct protein domains, residing in the N- and C-terminal halves, respectively. In the last two decades, p27kip1 has been reported to interact with microtubule and acto-myosin cytoskeletons, both in direct and indirect ways, overall drawing a picture in which several factors play their role either in synergy or in contrast one with another. As a result, the role of p27kip1 in cytoskeleton dynamics has been implicated in cell migration, both in physiologic and in neoplastic contexts, modulating cytokinesis, lipid raft trafficking, and neuronal development. Recently, two distinct papers have further reported a central role for p27kip1 in the control of microtubule stability and post-translational modifications, dissecting the interaction between p27kip1 and α-tubulin-acetyl-transferase (α-TAT), an enzyme involved in the stability of microtubules, and protein-regulator of cytokinesis 1 (PRC1), a nuclear regulator of the central spindle during mitosis. In light of these recent evidences, we will comment on the role of p27kip1 on cytoskeleton regulation and its implication for cancer progression.
- Published
- 2019
- Full Text
- View/download PDF
4. Loss of p27kip1 increases genomic instability and induces radio-resistance in luminal breast cancer cells
- Author
-
Stefania Berton, Martina Cusan, Ilenia Segatto, Francesca Citron, Sara D’Andrea, Sara Benevol, Michele Avanzo, Alessandra Dall’Acqua, Monica Schiappacassi, Robert G. Bristow, Barbara Belletti, and Gustavo Baldassarre
- Subjects
Medicine ,Science - Abstract
Abstract Genomic instability represents a typical feature of aggressive cancers. Normal cells have evolved intricate responses to preserve genomic integrity in response to stress, such as DNA damage induced by γ-irradiation. Cyclin-dependent kinases (CDKs) take crucial part to these safeguard mechanisms, but involvement of CDK-inhibitors, such as p27Kip1, is less clear. We generated immortalized fibroblasts from p27kip1 knock-out (KO) mouse embryos and re-expressed p27kip1 WT, or its mutant forms, to identify the function of different domains. We γ-irradiated fibroblasts and observed that loss of p27Kip1 was associated to accumulation of residual DNA damage, increased number of mitotic aberration and, eventually, to survival advantage. Nuclear localization and cyclin/CDK-binding of p27Kip1 were critical to mediate proper response to DNA damage. In human luminal breast cancer (LBC) p27kip1 is frequently down-modulated and CDKN1B, p27Kip1 gene, sporadically mutated. We recapitulated results obtained in mouse fibroblasts in a LBC cell line genetically manipulated to be KO for CDKN1B gene. Following γ-irradiation, we confirmed that p27kip1 expression was necessary to preserve genomic integrity and to recognize and clear-out aberrant cells. Our study provides important insights into mechanisms underlying radio-resistance and unveils the possibility for novel treatment options exploiting DNA repair defects in LBC.
- Published
- 2017
- Full Text
- View/download PDF
5. The role of literary metaphors in aesthetic appreciation.
- Author
-
Francesca Citron, Holly Clarke, Qishan Liao, and Carina Rasse
- Published
- 2020
6. Supplementary Data from Stathmin Is Required for Normal Mouse Mammary Gland Development and Δ16HER2-Driven Tumorigenesis
- Author
-
Barbara Belletti, Gustavo Baldassarre, Andrea Vecchione, Augusto Amici, Cristina Marchini, Monica Schiappacassi, Giorgia Mungo, Stefania Berton, Tiziana Perin, Gian Luca Rampioni Vinciguerra, Sara D'Andrea, Francesca Citron, Mara De Marco Zompit, and Ilenia Segatto
- Abstract
Supplementary Information includes Supplementary Materials and Methods, Supplementary Figures 1-7, with Legends and Supplementary References. Supplementary Figure S1: Loss of stathmin impairs mouse mammary gland development. Supplementary Figure S2: Loss of stathmin decreases the growth of mammary epithelial cells. Supplementary Figure S3: Loss of stathmin alters the mammary gland organization. Supplementary Figure S4: mRNA levels of STAT5 target genes are differently modulated when stathmin is silenced. Supplementary Figure S5: Stathmin expression correlates with poor prognosis in breast cancer patients. Supplementary Figure S6: Î"16HER2-driven mammary neoplastic lesions display luminal features. Supplementary Figure S7: Absence of stathmin in pre-neoplastic mouse mammary glands (13 weeks of age) weakens the activation of Î"16HER2 transformation-related pathways
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Table S1 from An Integrated Approach Identifies Mediators of Local Recurrence in Head and Neck Squamous Carcinoma
- Author
-
Gustavo Baldassarre, Luigi Barzan, Andrea Vecchione, Barbara Belletti, Diego Serraino, Emanuela Vaccher, Riccardo Bomben, Deborah French, Igor Jurisica, Tomas Tokar, Chiara Pastrello, David Otasek, William Klement, Carlo M. Croce, Sandro Sulfaro, Sara D'Andrea, Renato Talamini, Jerry Polesel, Giovanni Franchin, Joshua Armenia, and Francesca Citron
- Abstract
Supplementary Table S1: Molecular and Pathological variables of patients included in the discovery cohort.
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Tables from Downregulation of miR-223 Expression Is an Early Event during Mammary Transformation and Confers Resistance to CDK4/6 Inhibitors in Luminal Breast Cancer
- Author
-
Barbara Belletti, Gustavo Baldassarre, Andrea Vecchione, Augusto Amici, Cristina Marchini, Samuele Massarut, Monica Schiappacassi, Tiziana Perin, Maria Chiara Mattevi, Sara D'Andrea, Giorgia Mungo, Francesca Russo, Lorena Musco, Gian Luca Rampioni Vinciguerra, Ilenia Segatto, and Francesca Citron
- Abstract
Table S1: Clinical Data of BC samples collected at Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy. Table S2: Clinical Data of BC samples collected in University "Sapienza" of Rome, Santo Andrea Hospital, Rome, Italy
- Published
- 2023
- Full Text
- View/download PDF
9. Video S1 from Stathmin Is Required for Normal Mouse Mammary Gland Development and Δ16HER2-Driven Tumorigenesis
- Author
-
Barbara Belletti, Gustavo Baldassarre, Andrea Vecchione, Augusto Amici, Cristina Marchini, Monica Schiappacassi, Giorgia Mungo, Stefania Berton, Tiziana Perin, Gian Luca Rampioni Vinciguerra, Sara D'Andrea, Francesca Citron, Mara De Marco Zompit, and Ilenia Segatto
- Abstract
NMuMG CTR cells
- Published
- 2023
- Full Text
- View/download PDF
10. Data from Stathmin Is Required for Normal Mouse Mammary Gland Development and Δ16HER2-Driven Tumorigenesis
- Author
-
Barbara Belletti, Gustavo Baldassarre, Andrea Vecchione, Augusto Amici, Cristina Marchini, Monica Schiappacassi, Giorgia Mungo, Stefania Berton, Tiziana Perin, Gian Luca Rampioni Vinciguerra, Sara D'Andrea, Francesca Citron, Mara De Marco Zompit, and Ilenia Segatto
- Abstract
Postnatal development of the mammary gland relies on the maintenance of oriented cell division and apicobasal polarity, both of which are often deregulated in cancer. The microtubule (MT) network contributes to control these processes; however, very little is known about the impact of altered MT dynamics in the development of a complex organ and on the role played by MT-interacting proteins such as stathmin. In this study, we report that female stathmin knock-out (STM KO) mice are unable to nurse their litters due to frank impairment of mammary gland development. In mouse mammary epithelial cells, loss of stathmin compromised the trafficking of polarized proteins and the achievement of proper apicobasal polarity. In particular, prolactin receptor internalization and localization was altered in STM KO mammary epithelial cells, leading to decreased protein stability and downmodulation of the Prl/PrlR/STAT5 signaling pathway. Absence of stathmin induced alterations in mitotic spindle orientation, accumulation of mitotic defects, and apoptosis, overall contributing to tissue disorganization and further decreasing the expansion of the mammary epithelial compartment. Loss of stathmin in MMTV-Δ16HER2 transgenic mice decreased the incidence and increased the latency of these very aggressive mammary carcinomas. Collectively, these data identify the essential mammary protein stathmin as protumorigenic and suggest it may serve as a potential therapeutic target in breast cancer.Significance:Stathmin expression is critical to maintain oriented cell division and apicobasal polarity in normal mammary glands and to establish a protumorigenic program that eventually sustains HER2-positive breast cancer formation in mice.
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Data from An Integrated Approach Identifies Mediators of Local Recurrence in Head and Neck Squamous Carcinoma
- Author
-
Gustavo Baldassarre, Luigi Barzan, Andrea Vecchione, Barbara Belletti, Diego Serraino, Emanuela Vaccher, Riccardo Bomben, Deborah French, Igor Jurisica, Tomas Tokar, Chiara Pastrello, David Otasek, William Klement, Carlo M. Croce, Sandro Sulfaro, Sara D'Andrea, Renato Talamini, Jerry Polesel, Giovanni Franchin, Joshua Armenia, and Francesca Citron
- Abstract
Supplementary Figure S1. Validation of differentially expressed miRs in the discovery dataset. Supplementary Figure S2. Design of a recurrence prediction model in HNSCC based on miRs expression using a bioinformatic approach. Supplementary Figure S3. In vitro validation of miR-9 putative targets Supplementary Figure S4. Validation of putative miRs seed sites in the 3'-UTR of SASH1 and SP1 genes Supplementary Figure S5. The combined expression of miRs-1, 133a and -150 is necessary to downregulate SP1 mRNA and protein levels. Supplementary Figure S6. Correlation analyses of miRs putative targets in primary HNSCC collected in our Institute and described in Supplementary table S2 as validation set. Supplementary Figure S7. Prognostic significance of 4 miRs-targets in the TCGA HNSCC dataset. Supplementary Table S3. Median miRs' expression (qRT-PCR) according to selected characteristics Supplementary Experimental Procedures
- Published
- 2023
- Full Text
- View/download PDF
12. Data from Downregulation of miR-223 Expression Is an Early Event during Mammary Transformation and Confers Resistance to CDK4/6 Inhibitors in Luminal Breast Cancer
- Author
-
Barbara Belletti, Gustavo Baldassarre, Andrea Vecchione, Augusto Amici, Cristina Marchini, Samuele Massarut, Monica Schiappacassi, Tiziana Perin, Maria Chiara Mattevi, Sara D'Andrea, Giorgia Mungo, Francesca Russo, Lorena Musco, Gian Luca Rampioni Vinciguerra, Ilenia Segatto, and Francesca Citron
- Abstract
miR-223 is an anti-inflammatory miRNA that in cancer acts either as an oncosuppressor or oncopromoter, in a context-dependent manner. In breast cancer, we demonstrated that it dampens the activation of the EGF pathway. However, little is known on the role of miR-223 during breast cancer onset and progression. miR-223 expression was decreased in breast cancer of luminal and HER2 subtypes and inversely correlated with patients' prognosis. In normal luminal mammary epithelial cells, miR-223 acted cell autonomously in the control of their growth and morphology in three-dimensional context. In the MMTV-Δ16HER2 transgenic mouse model, oncogene transformation resulted in a timely abrogation of miR-223 expression, likely due to activation of E2F1, a known repressor of miR-223 transcription. Accordingly, treatment with CDK4/6 inhibitors, which eventually results in restraining E2F1 activity, restored miR-223 expression and miR-223 ablation induced luminal breast cancer resistance to CDK4/6 inhibition, both in vitro and in vivo. Notably, miR-223 expression was lost in microdissected ductal carcinoma in situ (DCIS) from patients with luminal and HER2-positive breast cancer. Altogether, these results identify downmodulation of miR-223 as an early step in luminal breast cancer onset and suggest that it could be used to identify aggressive DCIS and predict the response to targeted therapy.Significance:miR-223 may represent a predictive biomarker of response to CDK4/6 inhibitors and its loss could identify DCIS lesions that are likely to progress into invasive breast cancer.
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Figures 1-6, Supplementary Materials and Methods from Downregulation of miR-223 Expression Is an Early Event during Mammary Transformation and Confers Resistance to CDK4/6 Inhibitors in Luminal Breast Cancer
- Author
-
Barbara Belletti, Gustavo Baldassarre, Andrea Vecchione, Augusto Amici, Cristina Marchini, Samuele Massarut, Monica Schiappacassi, Tiziana Perin, Maria Chiara Mattevi, Sara D'Andrea, Giorgia Mungo, Francesca Russo, Lorena Musco, Gian Luca Rampioni Vinciguerra, Ilenia Segatto, and Francesca Citron
- Abstract
Supplementary Information, Methods and Figures. Supplementary Figure 1. Loss of miR-223 does not alter the murine mammary gland architecture; Supplementary Figure 2. miR-223 KO mice do not display any proliferative advantage during â^†16HER2-driven mammary tumorigenesis; Supplementary Figure 3. miR-223 WT and KO â^†16HER2 tumor-derived mammary epithelial cells (mMECs) display comparable tumorigenic potential; Supplementary Figure 4. miR-223 partially counteracts HER2-driven BC cell proliferation and 3D-organization; Supplementary Figure 5. miR-223 is rapidly down-modulated during cell transformation; Supplementary Figure 6. miR-223 is upregulated in response to anti-proliferative stimuli
- Published
- 2023
- Full Text
- View/download PDF
14. Data from An Integrated Approach Identifies Mediators of Local Recurrence in Head and Neck Squamous Carcinoma
- Author
-
Gustavo Baldassarre, Luigi Barzan, Andrea Vecchione, Barbara Belletti, Diego Serraino, Emanuela Vaccher, Riccardo Bomben, Deborah French, Igor Jurisica, Tomas Tokar, Chiara Pastrello, David Otasek, William Klement, Carlo M. Croce, Sandro Sulfaro, Sara D'Andrea, Renato Talamini, Jerry Polesel, Giovanni Franchin, Joshua Armenia, and Francesca Citron
- Abstract
Purpose: Head and neck squamous cell carcinomas (HNSCCs) cause more than 300,000 deaths worldwide each year. Locoregional and distant recurrences represent worse prognostic events and accepted surrogate markers of patients' overall survival. No valid biomarker and salvage therapy exist to identify and treat patients at high-risk of recurrence. We aimed to verify if selected miRNAs could be used as biomarkers of recurrence in HNSCC.Experimental Design: A NanoString array was used to identify miRNAs associated with locoregional recurrence in 44 patients with HNSCC. Bioinformatic approaches validated the signature and identified potential miRNA targets. Validation experiments were performed using an independent cohort of primary HNSCC samples and a panel of HNSCC cell lines. In vivo experiments validated the in vitro results.Results: Our data identified a four-miRNA signature that classified HNSCC patients at high- or low-risk of recurrence. These miRNAs collectively impinge on the epithelial–mesenchymal transition process. In silico and wet lab approaches showed that miR-9, expressed at high levels in recurrent HNSCC, targets SASH1 and KRT13, whereas miR-1, miR-133, and miR-150, expressed at low levels in recurrent HNSCC, collectively target SP1 and TGFβ pathways. A six-gene signature comprising these targets identified patients at high risk of recurrences, as well. Combined pharmacological inhibition of SP1 and TGFβ pathways induced HNSCC cell death and, when timely administered, prevented recurrence formation in a preclinical model of HNSCC recurrence.Conclusions: By integrating different experimental approaches and competences, we identified critical mediators of recurrence formation in HNSCC that may merit to be considered for future clinical development. Clin Cancer Res; 23(14); 3769–80. ©2017 AACR.
- Published
- 2023
- Full Text
- View/download PDF
15. Abstract 5868: Uncovering key microenvironmental features for immunotherapy response at the subclonal level in pancreatic cancer
- Author
-
Er-Yen Yen, I-Lin Ho, Chieh-Yuan Li, Charles Dyke, Shan Jiang, Francesca Citron, Sergio Attanasio, Rutvi Shah, Ko-Chien Chen, Giulio Draetta, and Andrea Viale
- Subjects
Cancer Research ,Oncology - Abstract
The lack of suitable experimental approaches to investigate and model functional heterogeneity in vivo has had a profound negative impact on our understanding of how heterogeneity affects the response to immunotherapy. To bridge this technological gap, we leverage a new platform that visualizes the spatial architecture of subclones and microenvironments. To study heterogeneous populations of cells and their clonal dynamics in vivo, we performed barcode lineage tracing with next-generation sequencing analysis. It allows a quantitative evaluation of the spatial distribution and temporal clonal dynamic in vivo with multiple pharmacological perturbations, such as immune checkpoint blockade (ICB). We found that treatment with a PD1 inhibitor, despite a limited effect on tumor volume, induced dramatic changes in tumor clonal architecture. Furthermore, we revealed that subclones from well-defined geographical domains in vivo share differential behavior upon immune checkpoint blockade (ICB). By visualizing 40 different markers with multiplexed staining technology, we further identified that the microenvironment also exhibits a high level of spatial heterogeneity. Therefore, we coupled spatial barcode sequencing with high-content imaging and revealed the differential microenvironment feature of sensitive and resistant clones. We will further explore the mechanisms of immune evasion at the subclonal level. Through isolation and deep characterization of clonal lineages endowed with a distinct ability to engage the immune response, we hope to identify new vulnerabilities to overcome resistance to ICB. Citation Format: Er-Yen Yen, I-Lin Ho, Chieh-Yuan Li, Charles Dyke, Shan Jiang, Francesca Citron, Sergio Attanasio, Rutvi Shah, Ko-Chien Chen, Giulio Draetta, Andrea Viale. Uncovering key microenvironmental features for immunotherapy response at the subclonal level in pancreatic cancer. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 5868.
- Published
- 2023
- Full Text
- View/download PDF
16. Abstract 303: DPY30 loss leads to DNA re-replication and immunoediting in pancreatic ductal adenocarcinoma
- Author
-
Francesca Citron, Luigi Perelli, Yanshuo Chu, Sanjana Srinivasan, I-Lin Ho, Er-Yen Yen, Rutvi Shah, Sergio Attanasio, Luis Castillo Montanez, Mauro Di Pilato, Shan Jiang, Wantong Yao, Sisi Gao, Angela Deem, Giannicola Genovese, Linghua Wang, Virginia Giuliani, Andrea Viale, and Giulio F. Draetta
- Subjects
Cancer Research ,Oncology - Abstract
Introduction: DNA replication is a tightly regulated, multi-step process involving licensing and replication factors that faithfully orchestrate a single duplication of the entire genome exclusively during S phase of the cell cycle. Although the mechanisms that prevent uncoordinated DNA replication are well characterized, it remains unclear how cells counteract its potentially harmful effects when it does occur, such as in cancer cells driven by aberrant oncogenic signaling and loss of cell cycle checkpoints. Here, we identify DPY30, an understudied component of all COMPASS complexes, as a modulator of DNA replication. We found upregulated DPY30 expression in pancreatic ductal adenocarcinoma (PDAC) as well as a correlation between increasing DPY30 expression level and higher tumor grade and poor clinical outcomes. Experimental procedure: Human- and mouse-derived PDAC cells silenced or knockout for DPY30 expression were generated and characterized for their in vivo growth in recipient mice with increasing level of immune pressure. Chromatin immunoprecipitation followed by massive parallel sequencing was used to measure the epigenetic landscape in DPY30 or WDR5 knockdown cells. Unbiased proteomics and transcriptomics analyses were performed to elucidate molecular mechanisms. Results: The canonical function of COMPASS complexes is to regulate histone methylation. Unexpectedly, DPY30 loss did not affect histone methylation patterns compared to WDR5 loss, the core member of the COMPASS-like complex. This prompted unbiased proteomics analysis, which showed that DPY30 interacts with members of the DNA helicase, possibly stabilizing the DNA replication fork progression. We found that DPY30 overexpression, by preventing Geminin ubiquitylation, strongly stabilizes its expression, which in turn suppresses Cdt1 activity in late S-phase, buffering the risk of uncoordinated DNA replication in G2/M phase. Consistently, DPY30 loss resulted in DNA re-replication and chromosomal instability, as manifested by DNA fiber and metaphasic spread. In vivo, whole-exome sequencing analysis of DPY30-knockout tumors revealed that DPY30 loss markedly increased mutational burden in immunodeficient mice. In syngeneic models, DPY30 loss strongly inhibited tumor growth, prolonged survival, and increased T cell infiltration of tumors. Moreover, the immune system selectively cleared tumor cells with complex karyotypes, and we observed improved anti-tumor efficacy with DPY30 knockdown and anti-PD-1 treatment compared to either condition alone. Conclusions: In summary, we have identified a previously undescribed function for DPY30 to stabilize the replisome machinery and prevent excessive DNA replication. Our findings indicate that, in PDAC, DPY30 promotes genome stability, thus providing a rationale for targeting DPY30 or its effector proteins in combination with immune-checkpoint inhibitors. Citation Format: Francesca Citron, Luigi Perelli, Yanshuo Chu, Sanjana Srinivasan, I-Lin Ho, Er-Yen Yen, Rutvi Shah, Sergio Attanasio, Luis Castillo Montanez, Mauro Di Pilato, Shan Jiang, Wantong Yao, Sisi Gao, Angela Deem, Giannicola Genovese, Linghua Wang, Virginia Giuliani, Andrea Viale, Giulio F. Draetta. DPY30 loss leads to DNA re-replication and immunoediting in pancreatic ductal adenocarcinoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 303.
- Published
- 2023
- Full Text
- View/download PDF
17. Abstract 6053: Sulfation is required for prostate cancer xenograft tumor formation but is dispensable for cell viabilityin vitro
- Author
-
Ko-Chien Chen, Yonhong Liu, Chenchu Lin, Er-Yen (Nick) Yen, Francesca Citron, Xingdi Ma, Tan Lin, Philip Lorenzi, Florian Muller, and Ronald DePinho
- Subjects
Cancer Research ,Oncology - Abstract
Sulfation of proteins, carbohydrates, lipids, and xenobiotics is an essential post-translational modification (PTM) process thought to play critical roles in diverse biological processes ranging from detoxification, cell signaling, and extracellular matrix architecture, to immune modulation. Sulfation is accomplished by the universal sulfate donor, PAPS (3'-Phosphoadenosine-5'-phosphosulfate), which is synthesized by bifunctional enzymes PAPSS1 and PAPSS2 (PAPS synthases). The PAPSS2 gene situates near PTEN and is frequently deleted with PTEN across cancer types. Approximately 20% of prostate cancer patients exhibit loss of PTEN, and ~50% of these cases also sustain a loss of PAPSS2. However, the loss of PAPSS2 appears to be tolerated and possibly compensated by its functionally redundant paralogue, PAPSS1, located on chromosome 4q24. The functional redundancy between PAPSS1 and PAPSS2 suggests that these two genes may be collateral lethality pair provided that sulfation is essential for cancer cell viability. Thus, we hypothesize that targeting PAPSS1 in PTEN/PAPSS2-null prostate cancer can generate cancer-specific vulnerabilities while leaving normal cells undisturbed. To assess this possibility, knockdown and knockout of PAPSS1 in cell lines of PAPSS2-null and PAPSS2-wildtype background were generated to characterize cell viability in vitro and tumor formation in vivo. PAPS and APS, an intermediary product of the sulfation pathway, are measured to verify that no alternative pathways for sulfate donors exist and that the co-extinction of PAPSS1/2 eliminates all avenues of generating sulfate donors. Combined extinction of PAPSS1/2 across multiple cancer cell lines was shown to be tolerated in vitro, and recurrent changes in morphology were observed. Loss of sulfation verified by the disappearance of sulfotyrosine and mass spectrometry measurements of PAPS and APS are pending. Our In vitro results surprisingly indicate that a major PTM, like sulfation is entirely dispensable for cancer cell viability under normal culture conditions. However, PAPSS1/2-null cell lines demonstrated a profound delay in tumor formation and prolonged survival, suggesting that sulfation may be required for stromal and innate immune modulation. Citation Format: Ko-Chien Chen, Yonhong Liu, Chenchu Lin, Er-Yen (Nick) Yen, Francesca Citron, Xingdi Ma, Tan Lin, Philip Lorenzi, Florian Muller, Ronald DePinho. Sulfation is required for prostate cancer xenograft tumor formation but is dispensable for cell viabilityin vitro [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 6053.
- Published
- 2023
- Full Text
- View/download PDF
18. DPY30 loss leads to DNA re-replication and immunoediting in pancreatic ductal adenocarcinoma
- Author
-
Francesca Citron, Luigi Perelli, Yanshuo Chu, Sanjana Srinivasan, I-Lin Ho, Er-Yen Yen, Rutvi Shah, Sergio Attanasio, Luis Alejandro Castillo Montanez, Mauro Di Pilato, Shan Jiang, Sisi Gao, Angela K. Deem, Giannicola Genovese, Virginia Giuliani, Linghua Wang, Alessandro Carugo, Andrea Viale, and Giulio Draetta
- Published
- 2022
- Full Text
- View/download PDF
19. Clonal dominance defines metastatic dissemination in pancreatic cancer
- Author
-
I-Lin Ho, Chieh-Yuan Li, Fuchenchu Wang, Li Zhao, Jingjing Liu, Er-Yen Yen, Yanshuo Chu, Francesca Citron, Rutvi Shah, Sergio Attanasio, Melinda Soeung, Ziheng Chen, Jiang Hong, Jiang Shan, Sisi Gao, Angela K. Deem, Andrew Futreal, Haoqiang Ying, Giannicola Genovese, Jianhua Zhang, Anirban Maitra, Timothy P. Heffernan, Linghua Wang, Giulio Draetta, Kim-Anh Do, Alessandro Carugo, Ruitao Liu, and Andrea Viale
- Published
- 2022
- Full Text
- View/download PDF
20. Convergent evolutionary trajectories uncover metastatic drivers in renal cancer
- Author
-
Luigi Perelli, Li Zhang, Courtney N. Le, Francesca Citron, Melinda Soeung, Sebastian Lundgren, Hania Khan, Linghua Wang, Pavlos Msaouel, Giulio Draetta, Ari Hakimi, Alessandro Carugo, and Giannicola Genovese
- Published
- 2022
- Full Text
- View/download PDF
21. Epithelial memory of inflammation limits tissue damage while promoting pancreatic tumorigenesis
- Author
-
Prasenjit Dey, Giulio Draetta, Giovanni Valenti, I-Lin Ho, Denise Corti, Andrea Viale, Linghua Wang, Alessandro Carugo, Francesca Citron, Timothy P. Heffernan, Federica Carbone, Yoku Hayakawa, Rutvi Shah, Er-Yen Yen, Chiara Balestrieri, Andrew D. Rhim, Shaojun Zhang, Wantong Yao, Giuseppe R. Diaferia, Angela K. Deem, Anirban Maitra, Timothy C. Wang, Shan Jiang, Haoqiang Ying, Chieh-Yuan Li, Stefan Rose-John, Giannicola Genovese, Hong Jiang, Edoardo Del Poggetto, Gioacchino Natoli, Luigi Sapio, Sara Loponte, Sisi Gao, Del Poggetto, E., Ho, I. -L., Balestrieri, C., Yen, E. -Y., Zhang, S., Citron, F., Shah, R., Corti, D., Diaferia, G. R., Li, C. -Y., Loponte, S., Carbone, F., Hayakawa, Y., Valenti, G., Jiang, S., Sapio, L., Jiang, H., Dey, P., Gao, S., Deem, A. K., Rose-John, S., Yao, W., Ying, H., Rhim, A. D., Genovese, G., Heffernan, T. P., Maitra, A., Wang, T. C., Wang, L., Draetta, G. F., Carugo, A., Natoli, G., and Viale, A.
- Subjects
Male ,Carcinogenesis ,MAP Kinase Signaling System ,Context (language use) ,Inflammation ,Acinar Cells ,medicine.disease_cause ,Epigenesis, Genetic ,Mice ,Metaplasia ,Spheroids, Cellular ,Carcinoma ,medicine ,Animals ,Pancreas ,Cells, Cultured ,Early Growth Response Protein 1 ,Mutation ,Enzyme Precursors ,Multidisciplinary ,business.industry ,Epithelial Cells ,medicine.disease ,Cellular Reprogramming ,Chromatin ,Cell Transformation, Neoplastic ,Genes, ras ,Pancreatitis ,Cancer research ,Female ,KRAS ,medicine.symptom ,business ,Transcriptome ,Carcinoma, Pancreatic Ductal - Abstract
Defining tumor cell immune evasion Mouse models used to study cancer often lack a full immune system, allowing implantation of human tumors into the mice. By contrast, naturally evolving tumors must contend with a fully functional immune system and its destruction of some of the cells (see the Perspective by Ho and Wood). Two groups now report studies on mouse models with a fully intact immune system. Martin et al . started with preexisting murine tumor cell lines and examined their continued evolution in vivo, whereas Del Poggetto et al . examined the development of new pancreatic tumors in the context of inflammation, as is often seen in human patients. In each study, the authors found that the immune system exerted a selective pressure on cells that would give rise to tumors, promoting the survival of those that had lost expression of tumor suppressor genes or activated a specific oncogene. The findings suggest a major role for the immune system in driving tumor evolution across multiple types of cancer. —YN
- Published
- 2021
22. Pathologist second opinion significantly alters clinical management of pT1 endoscopically resected colorectal cancer
- Author
-
Giammauro Berardi, Emanuela Pilozzi, Stefano Angeletti, Francesca Citron, Giulio Antonelli, Gian Luca Rampioni Vinciguerra, Emilio Di Giulio, Andrea Vecchione, and Gustavo Baldassarre
- Subjects
Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Lymphovascular invasion ,Colorectal cancer ,Concordance ,Lymph node metastasis ,Risk Assessment ,Pathology and Forensic Medicine ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,Internal medicine ,medicine ,Humans ,Referral and Consultation ,Molecular Biology ,Pathological ,Aged ,Retrospective Studies ,Aged, 80 and over ,business.industry ,Carcinoma ,Second opinion ,Endoscopy ,Cell Biology ,General Medicine ,Middle Aged ,Prognosis ,medicine.disease ,Pathologists ,Risk perception ,030104 developmental biology ,Lymphatic Metastasis ,030220 oncology & carcinogenesis ,Female ,Neoplasm Grading ,Colorectal Neoplasms ,business ,Risk assessment - Abstract
We retrospectively collected a series of 82 endoscopically removed early colorectal cancers. Histological specimens were revised by two gastrointestinal pathologists, performing a re-evaluation of all risk factors for lymph node metastasis. The comparison between second opinion and first pathological report revealed that lymphovascular invasion and tumor grading showed a lower level of concordance than other parameters. Our results demonstrated that second opinion modified risk assessment in about 10% of cases. It was mainly due to a lack in reporting of some parameters at the first diagnosis and a different evaluation in second opinion for updated guidelines. Considering the subgroup of patients with modified risk assessment, clinical data revealed that tumors, re-classified as low risk, did not develop lymph node metastasis that, conversely, occurred in patients identified as high risk by second opinion. In conclusion, second opinion significantly alters risk perception of endoscopically removed early colorectal carcinomas representing a valuable tool for their appropriate clinical management.
- Published
- 2019
- Full Text
- View/download PDF
23. Abstract 1327: Epithelial memory of resolved inflammation cooperates with oncogenic KRAS to limit tissue damage while promoting pancreatic cancer
- Author
-
I-Lin Ho, Edoardo Del Poggetto, Chiara Balestrieri, Er-Yen Yen, Francesca Citron, Rutvi Shah, Shaojun Zhang, Shan Jiang, Wantong Yao, Haoqiang Ying, Giannicola Genovese, Timothy Heffernan, Anirban Maitra, Linghua Wang, Timothy Wang, Giulio Draetta, Alessandro Carugo, Gioacchino Natoli, and Andrea Viale
- Subjects
Cancer Research ,Oncology - Abstract
The association between tumors and inflammation is a long-established clinical observation. Although many studies demonstrated that the inflammatory microenvironment can promote tumor growth through the activation of survival and proliferation programs in cancer cells, the reason why inflammation, an evolutionarily conserved response to damage aimed at reestablishing tissue integrity upon injury, might be integral to tumorigenesis still remains unknown. Pancreatic ductal adenocarcinoma (PDAC), a tumor characterized by poor prognosis, represents a distinctive example of cooperation between inflammation and activated oncogenes. Frequently developed in a context of chronic pancreatitis, PDAC is always associated with an inflammatory microenvironment. As supported by a substantial body of evidence across a multitude of experimental models, when occurring in the context of pancreatitis, mutations of KRAS, the universal oncogenic driver of pancreatic cancer, lead to accelerated tumor development inducing the appearance of neoplastic precursor lesions, such as acinar-to-ductal metaplasia (ADM) and pancreatic intraepithelial neoplasia (PanIN), which can evolve into invasive tumors. Interestingly, preneoplastic pancreatic alterations, specifically ADM, have been previously identified in acute and chronic pancreatitis in the absence of oncogene activation. Investigating the effects of inflammation on normal pancreatic epithelial cells, we discovered that long after complete resolution a transient inflammatory event primes cells to cooperate with oncogenic KRAS. Indeed, upon recovery from a single acute inflammation, epithelial cells display an enduring adaptive response associated with sustained epigenetic and transcriptional reprogramming. Such adaptation facilitates the prompt reactivation of acinar-to-ductal metaplasia upon subsequent inflammatory events, representing a physiological mechanism for limiting tissue damage via rapid decrease of zymogen production. Because metaplastic lesions are mediated by the activation of MAPK signaling, we demonstrated that activating mutations of KRAS, maintaining an irreversible ADM, are protective against tissue damage in a contest of pancreatitis. Uncovering a new physiologic role of somatic oncogenic mutations in preserving tissue integrity during repeated damages, we propose that KRAS mutations represent a nearly universal event in pancreatic cancer because beneficial and under strong positive selection in the context of recurrent pancreatitis. Citation Format: I-Lin Ho, Edoardo Del Poggetto, Chiara Balestrieri, Er-Yen Yen, Francesca Citron, Rutvi Shah, Shaojun Zhang, Shan Jiang, Wantong Yao, Haoqiang Ying, Giannicola Genovese, Timothy Heffernan, Anirban Maitra, Linghua Wang, Timothy Wang, Giulio Draetta, Alessandro Carugo, Gioacchino Natoli, Andrea Viale. Epithelial memory of resolved inflammation cooperates with oncogenic KRAS to limit tissue damage while promoting pancreatic cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 1327.
- Published
- 2022
- Full Text
- View/download PDF
24. Evaluation of angiogenesis-related genes as prognostic biomarkers of bevacizumab treated ovarian cancer patients: Results from the phase iv mito16a/mango ov-2 translational study
- Author
-
Gustavo Baldassarre, Nicoletta Colombo, Eliana Bignotti, Vincenzo Canzonieri, Vittorio Simeon, Paolo Chiodini, Sabrina Chiara Cecere, Gian Luca Rampioni Vinciguerra, Daniela Califano, Maria Carmela Piccirillo, Francesca Citron, Giovanni Scambia, Daniela Russo, Marco Montella, Francesco Perrone, Gian Franco Zannoni, Anna Spina, Alessandra Ciucci, Germana Tognon, Clorinda Schettino, Laura Arenare, Nunzia Simona Losito, Michele Del Sesto, Rossella De Cecio, Sandro Pignata, Gabriella Ferrandina, Piera Gargiulo, Giosuè Scognamiglio, Daniela Gallo, Simona Signoriello, Califano, D, Gallo, D, Vinciguerra, G, De Cecio, R, Arenare, L, Signoriello, S, Russo, D, Ferrandina, G, Citron, F, Losito, N, Gargiulo, P, Simeon, V, Scambia, G, Cecere, S, Montella, M, Colombo, N, Tognon, G, Bignotti, E, Zannoni, G, Canzonieri, V, Ciucci, A, Spina, A, Scognamiglio, G, Del Sesto, M, Schettino, C, Piccirillo, M, Perrone, F, Chiodini, P, Pignata, S, Baldassarre, G, Califano, D., Gallo, D., Vinciguerra, G. L. R., De Cecio, R., Arenare, L., Signoriello, S., Russo, D., Ferrandina, G., Citron, F., Losito, N. S., Gargiulo, P., Simeon, V., Scambia, G., Cecere, S. C., Montella, M., Colombo, N., Tognon, G., Bignotti, E., Zannoni, G. F., Canzonieri, V., Ciucci, A., Spina, A., Scognamiglio, G., Del Sesto, M., Schettino, C., Piccirillo, M. C., Perrone, F., Chiodini, P., Pignata, S., and Baldassarre, G.
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Bevacizumab ,Angiogenesis ,medicine.medical_treatment ,Article ,law.invention ,Bevacizumab treatment ,MicroRNAs ,Ovarian cancer ,Vessel density ,Randomized controlled trial ,Maintenance therapy ,law ,Internal medicine ,Medicine ,RC254-282 ,Chemotherapy ,business.industry ,Hazard ratio ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,MicroRNA ,medicine.disease ,Angiogenesi ,Settore MED/40 - GINECOLOGIA E OSTETRICIA ,Biomarker (medicine) ,business ,medicine.drug - Abstract
Simple Summary The possibility to identify, with appropriate biomarkers, patients that might mostly benefit from any given treatment is the basis of personalized oncology. Cancer biomarkers should be properly identified and validated on a large number of patients possibly enrolled in dedicated clinical trials. Here, we report the first molecular results of the MITO16A-ManGo-OV2 phase IV trial that was specifically designed to identify prognostic biomarkers of survival in epithelial ovarian cancer patients treated in first line with carboplatin-paclitaxel plus Bevacizumab (NCT01706120), a treatment for which validated predictive or prognostic biomarkers are still lacking. With this work we propose not only novel possible biomarkers for Bevacizumab-treated patients but also a way through which they can be properly collected, analyzed and statistically evaluated in the frame of large multicenter clinical trials. Abstract Background. Epithelial ovarian cancer (EOC) is a rare, highly lethal disease. In a subset of high grade EOC patients, maintenance therapy with the antiangiogenic drug Bevacizumab (BEV) is a valuable option. To date, no validated predictive or prognostic biomarkers exist for selecting EOC patients that might benefit from BEV treatment. Methods. Immunohistochemistry and RT-qPCR evaluated the expression of seven angiogenesis-related proteins and of a twelve microRNAs angio-signature in EOC patients, treated in first line with chemotherapy plus BEV (MITO16A/ManGO OV-2 phase IV trial). Centralized statistical analyses assessed the associations between each biomarker, clinical prognostic factors and survival outcomes. Results. High miR-484 expression was associated with longer progression-free and overall survival. Notably, the combined expression of miR-484 and its target VEGFB identified a subset of patients that might mostly benefit from BEV treatment. No other significant correlations were found between the other analyzed biomarkers and patients’ survival. The application of a shrinkage procedure to adjust for over-fitting hazard ratio estimates reduced the association significance. Conclusions. The analysis of angiogenesis related biomarkers in EOC patients homogenously treated with BEV in first line provides novel insight in their prognostic value and suggests that some of them might merit to be tested as predictive markers of drug activity in dedicated randomized trials.
- Published
- 2021
25. Leukotrienes, a potential target for Covid-19
- Author
-
Francesca Citron, Luigi Perelli, Angela K. Deem, Andrea Viale, and Giannicola Genovese
- Subjects
Cyclopropanes ,Leukotrienes ,2019-20 coronavirus outbreak ,Indoles ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Pneumonia, Viral ,Clinical Biochemistry ,Phenylcarbamates ,Acetates ,Sulfides ,Severity of Illness Index ,Article ,Tosyl Compounds ,Betacoronavirus ,Adrenal Cortex Hormones ,Severity of illness ,Pandemic ,Humans ,Hydroxyurea ,Immunologic Factors ,Medicine ,Pandemics ,Sulfonamides ,Arachidonic Acid ,biology ,SARS-CoV-2 ,business.industry ,Anti-Inflammatory Agents, Non-Steroidal ,COVID-19 ,Cell Biology ,biology.organism_classification ,Virology ,Arachidonic acid metabolism ,Quinolines ,Coronavirus Infections ,Cytokine Release Syndrome ,business - Published
- 2020
- Full Text
- View/download PDF
26. Targeting Epigenetic Dependencies in Solid Tumors: Evolutionary Landscape Beyond Germ Layers Origin
- Author
-
Linda Fabris and Francesca Citron
- Subjects
0301 basic medicine ,Cancer Research ,cancer stem cell ,medicine.medical_treatment ,pancreatic cancer ,Context (language use) ,Computational biology ,Review ,Biology ,medicine.disease_cause ,lcsh:RC254-282 ,Chromatin remodeling ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,Cancer stem cell ,medicine ,epigenetics ,glioblastoma ,Cancer ,Epigenome ,medicine.disease ,targeted therapy ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,030104 developmental biology ,ovarian cancer ,Oncology ,Tumor progression ,030220 oncology & carcinogenesis ,Carcinogenesis - Abstract
Extensive efforts recently witnessed the complexity of cancer biology; however, molecular medicine still lacks the ability to elucidate hidden mechanisms for the maintenance of specific subclasses of rare tumors characterized by the silent onset and a poor prognosis (e.g., ovarian cancer, pancreatic cancer, and glioblastoma). Recent mutational fingerprints of human cancers highlighted genomic alteration occurring on epigenetic modulators. In this scenario, the epigenome dependency of cancer orchestrates a broad range of cellular processes critical for tumorigenesis and tumor progression, possibly mediating escaping mechanisms leading to drug resistance. Indeed, in this review, we discuss the pivotal role of chromatin remodeling in shaping the tumor architecture and modulating tumor fitness in a microenvironment-dependent context. We will also present recent advances in the epigenome targeting, posing a particular emphasis on how this knowledge could be translated into a feasible therapeutic approach to individualize clinical settings and improve patient outcomes.
- Published
- 2020
27. CDKN1B mutation and copy number variation are associated with tumor aggressiveness in luminal breast cancer
- Author
-
Giovanni Franchin, Martina Cusan, Luigi Barzan, Giorgio Giorda, Samuele Massarut, Maura Sonego, Andrea Vecchione, Alessandra Dall'Acqua, Francesca Russo, Lorena Musco, Monica Schiappacassi, Gustavo Baldassarre, Lorenzo Gerratana, Tiziana Perin, Vincenzo Canzonieri, Fabio Puglisi, Roberto Sorio, Francesca Citron, Filippo Vit, Giorgia Mungo, Sandro Sulfaro, Jerry Polesel, Emilio Lucia, Vittorio Giacomarra, Sara D'Andrea, Milena S. Nicoloso, Maria Chiara Mattevi, Davide Viotto, Ilaria Anania, Ilenia Segatto, Barbara Belletti, Gian Luca Rampioni Vinciguerra, Federica Toffolutti, Riccardo Bomben, V. Gattei, Viotto, D., Russo, F., Anania, I., Segatto, I., Rampioni Vinciguerra, G. L., Dall'Acqua, A., Bomben, R., Perin, T., Cusan, M., Schiappacassi, M., Gerratana, L., D'Andrea, S., Citron, F., Vit, F., Musco, L., Mattevi, M. C., Mungo, G., Nicoloso, M. S., Sonego, M., Massarut, S., Sorio, R., Barzan, L., Franchin, G., Giorda, G., Lucia, E., Sulfaro, S., Giacomarra, V., Polesel, J., Toffolutti, F., Canzonieri, V., Puglisi, F., Gattei, V., Vecchione, A., Belletti, B., and Baldassarre, G.
- Subjects
Male ,0301 basic medicine ,DNA Copy Number Variations ,CNV ,Breast Neoplasms ,Neuroendocrine tumors ,medicine.disease_cause ,head and neck squamous cell carcinoma ,Pathology and Forensic Medicine ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,breast cancer ,Intestinal Neoplasms ,medicine ,Humans ,Copy-number variation ,CDKN1B ,copy number variation ,liquid biopsy ,mutation ,ovarian cancer ,p27 ,young breast cancer patients ,Original Paper ,Mutation ,copy number variation, CNV ,business.industry ,Prostatic Neoplasms ,medicine.disease ,Original Papers ,Head and neck squamous-cell carcinoma ,Squamous carcinoma ,Neuroendocrine Tumors ,030104 developmental biology ,030220 oncology & carcinogenesis ,MCF-7 Cells ,Cancer research ,Female ,business ,Ovarian cancer ,Cyclin-Dependent Kinase Inhibitor p27 ,CDK inhibitor - Abstract
The CDKN1B gene, encoding for the CDK inhibitor p27kip1, is mutated in defined human cancer subtypes, including breast, prostate carcinomas and small intestine neuroendocrine tumors. Lessons learned from small intestine neuroendocrine tumors suggest that CDKN1B mutations could be subclonal, raising the question of whether a deeper sequencing approach could lead to the identification of higher numbers of patients with mutations. Here, we addressed this question and analyzed human cancer biopsies from breast (n = 396), ovarian (n = 110) and head and neck squamous carcinoma (n = 202) patients, using an ultra‐deep sequencing approach. Notwithstanding this effort, the mutation rate of CDKN1B remained substantially aligned with values from the literature, showing that essentially only hormone receptor‐positive breast cancer displayed CDKN1B mutations in a relevant number of cases (3%). However, the analysis of copy number variation showed that another fraction of luminal breast cancer displayed loss (8%) or gain (6%) of the CDKN1B gene, further reinforcing the idea that the function of p27kip1 is important in this type of tumor. Intriguingly, an enrichment for CDKN1B alterations was found in samples from premenopausal luminal breast cancer patients (n = 227, 4%) and in circulating cell‐free DNA from metastatic luminal breast cancer patients (n = 59, 8.5%), suggesting that CDKN1B alterations could correlate with tumor aggressiveness and/or occur later during disease progression. Notably, many of the identified somatic mutations resulted in p27kip1 protein truncation, leading to loss of most of the protein or of its C‐terminal domain. Using a gene‐editing approach in a luminal breast cancer cell line, MCF‐7, we observed that the expression of p27kip1 truncating mutants that lose the C‐terminal domains failed to rescue most of the phenotypes induced by CDKN1B gene knockout, indicating that the functions retained by the C‐terminal portion are critical for its role as an oncosuppressor, at least in luminal breast cancer. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
- Published
- 2020
28. Downregulation of miR-223 expression is an early event during mammary transformation and confers resistance to CDK4/6 inhibitors in luminal breast cancer
- Author
-
Tiziana Perin, Ilenia Segatto, Francesca Russo, Samuele Massarut, Giorgia Mungo, Sara D'Andrea, Barbara Belletti, Cristina Marchini, Gian Luca Rampioni Vinciguerra, Monica Schiappacassi, Augusto Amici, Maria Chiara Mattevi, Francesca Citron, Andrea Vecchione, Lorena Musco, and Gustavo Baldassarre
- Subjects
0301 basic medicine ,Cancer Research ,Pyridines ,Receptor, ErbB-2 ,medicine.medical_treatment ,Cell Culture Techniques ,Piperazines ,Malignant transformation ,Targeted therapy ,0302 clinical medicine ,mir-223 ,Breast ,skin and connective tissue diseases ,Aged, 80 and over ,Mice, Knockout ,biology ,Middle Aged ,Prognosis ,Gene Expression Regulation, Neoplastic ,microRNA, breast cancer ,Cell Transformation, Neoplastic ,Oncology ,030220 oncology & carcinogenesis ,Disease Progression ,Female ,Adult ,Down-Regulation ,Antineoplastic Agents ,Breast Neoplasms ,Palbociclib ,03 medical and health sciences ,Breast cancer ,Mammary Glands, Animal ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Aged ,Oncogene ,business.industry ,Cyclin-dependent kinase 4 ,Cyclin-Dependent Kinase 4 ,Epithelial Cells ,Cyclin-Dependent Kinase 6 ,medicine.disease ,Disease Models, Animal ,MicroRNAs ,030104 developmental biology ,Carcinoma, Intraductal, Noninfiltrating ,Drug Resistance, Neoplasm ,biology.protein ,Cancer research ,Cyclin-dependent kinase 6 ,business ,E2F1 Transcription Factor - Abstract
miR-223 is an anti-inflammatory miRNA that in cancer acts either as an oncosuppressor or oncopromoter, in a context-dependent manner. In breast cancer, we demonstrated that it dampens the activation of the EGF pathway. However, little is known on the role of miR-223 during breast cancer onset and progression. miR-223 expression was decreased in breast cancer of luminal and HER2 subtypes and inversely correlated with patients' prognosis. In normal luminal mammary epithelial cells, miR-223 acted cell autonomously in the control of their growth and morphology in three-dimensional context. In the MMTV-Δ16HER2 transgenic mouse model, oncogene transformation resulted in a timely abrogation of miR-223 expression, likely due to activation of E2F1, a known repressor of miR-223 transcription. Accordingly, treatment with CDK4/6 inhibitors, which eventually results in restraining E2F1 activity, restored miR-223 expression and miR-223 ablation induced luminal breast cancer resistance to CDK4/6 inhibition, both in vitro and in vivo. Notably, miR-223 expression was lost in microdissected ductal carcinoma in situ (DCIS) from patients with luminal and HER2-positive breast cancer. Altogether, these results identify downmodulation of miR-223 as an early step in luminal breast cancer onset and suggest that it could be used to identify aggressive DCIS and predict the response to targeted therapy. Significance: miR-223 may represent a predictive biomarker of response to CDK4/6 inhibitors and its loss could identify DCIS lesions that are likely to progress into invasive breast cancer.
- Published
- 2020
29. TIMP-1 is Overexpressed and Secreted by Platinum Resistant Epithelial Ovarian Cancer Cells
- Author
-
E. Poletto, Roberto Sorio, Milena S. Nicoloso, Rosanna Pellicani, Francesca Citron, Gustavo Baldassarre, Maurizio Mongiat, Eliana Pivetta, Maura Sonego, and Gian Luca Rampioni Vinciguerra
- Subjects
Proteomics ,epithelial ovarian cancer ,0301 basic medicine ,Bevacizumab ,endocrine system diseases ,Angiogenesis ,Carcinoma, Ovarian Epithelial ,Matrix metalloproteinase ,Article ,angiogenesis ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,Cell Line, Tumor ,Tumor Microenvironment ,Overall survival ,Humans ,Medicine ,Tumor type ,Epithelial ovarian cancer ,Cell Proliferation ,Neoplasm Staging ,Platinum ,Platinum resistant ,Tissue Inhibitor of Metalloproteinase-1 ,business.industry ,General Medicine ,Survival Analysis ,platinum resistance ,female genital diseases and pregnancy complications ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Disease Progression ,Cancer research ,Biomarker (medicine) ,Female ,epithelial ovarian cancer, platinum resistance, angiogenesis ,business ,medicine.drug - Abstract
Epithelial Ovarian Cancer (EOC) is the most lethal gynecological cancer in developed countries, and the development of new strategies to overcome chemoresistance is an awaited clinical need. Angiogenesis, the development of new blood vessels from pre-existing vasculature, has been validated as a therapeutic target in this tumor type. The aim of this study is to verify if EOC cells with acquired resistance to platinum (PT) treatment display an altered angiogenic potential. Using a proteomic approach, we identified the tissue inhibitor of metalloproteinases 1 (TIMP-1) as the only secreted factor whose expression was up-regulated in PT-resistant TOV-112D and OVSAHO EOC cells used as study models. We report that TIMP-1 acts as a double-edged sword in the EOC microenvironment, directly affecting the response to PT treatment on tumor cells and indirectly altering migration and proliferation of endothelial cells. Interestingly, we found that high TIMP-1 levels in stage III&ndash, IV EOC patients associate with decreased overall survival, especially if they were treated with PT or bevacizumab. Taken together, these results pinpoint TIMP-1 as a key molecule involved in the regulation of EOC PT-resistance and progression disclosing the possibility that it could be used as a new biomarker of PT-resistance and/or therapeutic target.
- Published
- 2019
- Full Text
- View/download PDF
30. Language that conveys emotion: a commentary on Hinojosa, Moreno and Ferré (2019)
- Author
-
Francesca Citron
- Subjects
ComputingMilieux_THECOMPUTINGPROFESSION ,GeneralLiterature_INTRODUCTORYANDSURVEY ,ComputingMilieux_PERSONALCOMPUTING ,ComputingMilieux_LEGALASPECTSOFCOMPUTING ,bepress|Social and Behavioral Sciences|Psychology|Cognitive Psychology ,bepress|Life Sciences|Neuroscience and Neurobiology ,PsyArXiv|Neuroscience|Cognitive Neuroscience ,PsyArXiv|Social and Behavioral Sciences ,PsyArXiv|Neuroscience ,bepress|Social and Behavioral Sciences ,PsyArXiv|Social and Behavioral Sciences|Cognitive Psychology ,bepress|Life Sciences|Neuroscience and Neurobiology|Cognitive Neuroscience ,PsyArXiv|Social and Behavioral Sciences|Emotion ,bepress|Social and Behavioral Sciences|Linguistics ,PsyArXiv|Social and Behavioral Sciences|Linguistics - Abstract
This is a commentary to a paper and, as such, it has no abstract.
- Published
- 2019
31. Stathmin Is Required for Normal Mouse Mammary Gland Development and Δ16HER2-Driven Tumorigenesis
- Author
-
Barbara Belletti, Giorgia Mungo, Francesca Citron, Tiziana Perin, Monica Schiappacassi, Sara D'Andrea, Andrea Vecchione, Mara De Marco Zompit, Gian Luca Rampioni Vinciguerra, Gustavo Baldassarre, Augusto Amici, Cristina Marchini, Stefania Berton, and Ilenia Segatto
- Subjects
0301 basic medicine ,Genetically modified mouse ,Cancer Research ,Cell division ,Carcinogenesis ,Receptor, ErbB-2 ,Receptors, Prolactin ,Mammary gland ,Stathmin ,Mice, Transgenic ,macromolecular substances ,Biology ,medicine.disease_cause ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Mammary Glands, Animal ,medicine ,STAT5 Transcription Factor ,Animals ,Humans ,Mitosis ,Mice, Knockout ,Prolactin receptor ,Mammary Neoplasms, Experimental ,Cell biology ,Prolactin ,030104 developmental biology ,medicine.anatomical_structure ,HEK293 Cells ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,Female ,Signal transduction ,Signal Transduction - Abstract
Postnatal development of the mammary gland relies on the maintenance of oriented cell division and apicobasal polarity, both of which are often deregulated in cancer. The microtubule (MT) network contributes to control these processes; however, very little is known about the impact of altered MT dynamics in the development of a complex organ and on the role played by MT-interacting proteins such as stathmin. In this study, we report that female stathmin knock-out (STM KO) mice are unable to nurse their litters due to frank impairment of mammary gland development. In mouse mammary epithelial cells, loss of stathmin compromised the trafficking of polarized proteins and the achievement of proper apicobasal polarity. In particular, prolactin receptor internalization and localization was altered in STM KO mammary epithelial cells, leading to decreased protein stability and downmodulation of the Prl/PrlR/STAT5 signaling pathway. Absence of stathmin induced alterations in mitotic spindle orientation, accumulation of mitotic defects, and apoptosis, overall contributing to tissue disorganization and further decreasing the expansion of the mammary epithelial compartment. Loss of stathmin in MMTV-Δ16HER2 transgenic mice decreased the incidence and increased the latency of these very aggressive mammary carcinomas. Collectively, these data identify the essential mammary protein stathmin as protumorigenic and suggest it may serve as a potential therapeutic target in breast cancer. Significance: Stathmin expression is critical to maintain oriented cell division and apicobasal polarity in normal mammary glands and to establish a protumorigenic program that eventually sustains HER2-positive breast cancer formation in mice.
- Published
- 2018
32. Abstract 3128: miR-9 expression regulates and predicts the response to EGFR inhibitors in head & neck squamous cell carcinoma
- Author
-
Francesca Citron, Gian Luca Rampioni Vinciguerra, Giuseppe Fanetti, Ilenia Segatto, Barbara Belletti, Andrea Vecchione, Giovanni Franchin, and Gustavo Baldassarre
- Subjects
Cancer Research ,Oncology - Abstract
Introduction. Most Head & Neck Squamous Cell Carcinoma (HNSCC) patients are diagnosed with a locally advanced disease. Radiotherapy (RT) plus anti-EGFR monoclonal antibodies (Cetuximab - CTX) represents an effective combination therapy for locally advanced HNSCC patients. However, the 5-year overall survival is still 45%, mainly due to the appearance of loco-regional recurrences, suggesting that the identification and validation of predictive biomarkers of CTX activity is urgently needed to identify patients at high-risk of recurrence, who will benefit more from this therapeutic strategy. We recently validated a 4-microRNAs (miRs) signature, related to Epithelial to Mesenchymal Transition (EMT) and able to stratify HNSCC patients at high-risk of recurrence development. Among these 4 miRs, miR-9 was the only up-regulated in primary tumors from patients who developed recurrence within 2-year follow-up. Here we evaluated whether miR-9 expression had a functional role in HNSCC onset, progression and response to therapies. Methods. miR-9 modified (overexpressing and silenced) HNSCC cells were generated, characterized for their growth and response to radio-, chemo- and targeted-therapies (i.e.Cisplatin, Paclitaxel, 5-Fluorouracil and CTX) in vitro and in vivo. Preclinical evidences were confirmed in a cohort of primary HNSCC samples by intercrossing the expression of miR-9 and selected target genes with patients’ clinical variables and response to therapy. Results. Biochemical and biological in vitro and in vivo experiments showed that high miR-9 expression triggers EMT and increases tumor initiating properties in HNSCC cells. Interestingly, miR-9 silenced HNSCC cells displayed a higher sensitivity to RT and CTX, but not to chemotherapy, when compared to controls. Using an in vivo model of HNSCC, we observed that intra-tumor injection of anti-miR-9 improves the efficacy of RT alone and remarkably in combination with CTX. Mechanistically, we demonstrated that in HNSCC cells, EGFR activation triggers miR-9 expression that promotes the transcription of SP1, establishing a positive forward loop between EGFR activation and SP1 transcription. Accordingly, in primary HNSCC miR-9 expression strongly correlates with the one of SP1 and EGFR. More importantly, in a cohort of HNSCC patients treated with RT plus CTX, high miR-9 expression acts as an efficient predictive biomarker of intrinsic resistance. Conclusion. Altogether by integrating wet-lab and clinical data, we provide strong evidences indicating that in HNSCC a subpopulation of miR-9 expressing cells is intrinsically resistant to RT plus CTX therapy, confirming its potential prognostic value. Since we set up an assay to easily and quantitatively evaluate miR-9 expression in diagnostic tumor biopsies, we propose that it could be used to personalize the treatments for this group of patients, avoiding unfaithful toxic therapies. Citation Format: Francesca Citron, Gian Luca Rampioni Vinciguerra, Giuseppe Fanetti, Ilenia Segatto, Barbara Belletti, Andrea Vecchione, Giovanni Franchin, Gustavo Baldassarre. miR-9 expression regulates and predicts the response to EGFR inhibitors in head & neck squamous cell carcinoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 3128.
- Published
- 2019
- Full Text
- View/download PDF
33. An Integrated Approach Identifies Mediators of Local Recurrence in Head and Neck Squamous Carcinoma
- Author
-
Chiara Pastrello, Luigi Barzan, Barbara Belletti, Francesca Citron, Jerry Polesel, David Otasek, Andrea Vecchione, Deborah French, Tomas Tokar, Gustavo Baldassarre, Giovanni Franchin, Renato Talamini, Igor Jurisica, Sara D'Andrea, Diego Serraino, Sandro Sulfaro, Emanuela Vaccher, Carlo M. Croce, William Klement, Riccardo Bomben, and Joshua Armenia
- Subjects
0301 basic medicine ,Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Epithelial-Mesenchymal Transition ,Sp1 Transcription Factor ,In silico ,Cell ,Salvage therapy ,Biology ,Bioinformatics ,Article ,03 medical and health sciences ,Mice ,Transforming Growth Factor beta ,Internal medicine ,Cell Line, Tumor ,microRNA ,medicine ,Carcinoma ,Biomarkers, Tumor ,Animals ,Humans ,Aged ,Squamous Cell Carcinoma of Head and Neck ,Tumor Suppressor Proteins ,Keratin-13 ,Cancer ,Middle Aged ,medicine.disease ,Xenograft Model Antitumor Assays ,Squamous carcinoma ,Gene Expression Regulation, Neoplastic ,head and neck cancer ,Micro RNA ,stomatognathic diseases ,MicroRNAs ,030104 developmental biology ,medicine.anatomical_structure ,Head and Neck Neoplasms ,Valid Biomarker ,Carcinoma, Squamous Cell ,Female ,Neoplasm Recurrence, Local ,Signal Transduction - Abstract
PURPOSE: Head and Neck Squamous Cell Carcinomas (HNSCC) cause more than 300,000 deaths worldwide each year. Locoregional and distant recurrences represent worse prognostic events and accepted surrogate markers of patients’ overall survival. No valid biomarker and salvage therapy exist to identify and treat patients at high-risk of recurrence. We aimed to verify if selected microRNAs (miRs) could be used as biomarkers of recurrence in HNSCC. EXPERIMENTAL DESIGN: A Nanostring array was used to identify miRs associated with locoregional recurrence in 44 HNSCC patients. Bioinformatic approaches validated the signature and identified potential miR targets. Validation experiments were performed using an independent cohort of primary HNSCC samples and a panel of HNSCC cell lines. In vivo experiments validated the in vitro results. RESULTS: Our data identified a four-miR signature that classified HNSCC patients at high- or low-risk of recurrence. These miRs collectively impinge on the epithelial-mesenchymal transition process. In silico and wet lab approaches showed that miR-9, expressed at high levels in recurrent HNSCC, targets SASH1 and KRT13, while miR-1, miR-133 and miR-150, expressed at low levels in recurrent HNSCC, collectively target SP1 and TGFβ pathways. A six-genes signature comprising these targets identified patients at high risk of recurrences, as well. Combined pharmacological inhibition of SP1 and TGFβ pathways induced HNSCC cell death and, when timely administered, prevented recurrence formation in a preclinical model of HNSCC recurrence. CONCLUSIONS: By integrating different experimental approaches and competences, we identified critical mediators of recurrence formation in HNSCC that may merit to be considered for future clinical development. TRANSLATIONAL RELEVANCE. Most of HNSCC patients are diagnosed with a locally advanced disease and are treated with the combination of surgery, radiotherapy, and chemotherapy. This highly toxic approach is curative in about half of the cases but recurrent patients do not have effective salvage therapies. Therefore there is the urgency to identify and validate solid biomarkers able to classify patients at high risk that may benefit for specific targeted approaches. Our work tackled these two unmet clinical needs and identified a microRNA signature of locoregional recurrence in HNSCC patients. Starting from this signature, we identified two druggable pathways (i.e. SP1 and TGFβ) that when timely and concomitantly targeted efficiently prevented recurrence formation in a preclinical model. Both SP1 and TGFβ inhibitors have been already used to treat human patients, thus our work is of potential immediate translational relevance.
- Published
- 2016
34. Radiotherapy-induced miR-223 prevents relapse of breast cancer by targeting the EGF pathway
- Author
-
Monica Schiappacassi, Sara D'Andrea, Mario Roncadin, Gaetano Zafarana, Linda Fabris, Michele Avanzo, Milena S. Nicoloso, Cristina Ivan, Robert G. Bristow, Jayant S. Vaidya, Samuele Massarut, Francesca Citron, Tiziana Perin, Gustavo Baldassarre, George A. Calin, Simona Rossi, Barbara Belletti, Ilenia Segatto, Joshua Armenia, and S. Berton
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,Breast Neoplasms ,Biology ,Mice ,03 medical and health sciences ,Paracrine signalling ,0302 clinical medicine ,Breast cancer ,Growth factor receptor ,Recurrence ,Epidermal growth factor ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,Autocrine signalling ,Molecular Biology ,Cell Proliferation ,Wound Healing ,Epidermal Growth Factor ,Radiotherapy ,Manchester Cancer Research Centre ,ResearchInstitutes_Networks_Beacons/mcrc ,Lumpectomy ,Cancer ,medicine.disease ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,Radiation therapy ,MicroRNAs ,030104 developmental biology ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,Original Article ,Female ,Neoplasm Recurrence, Local ,Signal Transduction - Abstract
In breast cancer (BC) patients, local recurrences often arise in proximity of the surgical scar, suggesting that response to surgery may have a causative role. Radiotherapy (RT) after lumpectomy significantly reduces the risk of recurrence. We investigated the direct effects of surgery and of RT delivered intraoperatively (IORT), by collecting irradiated and non-irradiated breast tissues from BC patients, after tumor removal. These breast tissue specimens have been profiled for their microRNA (miR) expression, in search of differentially expressed miR among patients treated or not with IORT. Our results demonstrate that IORT elicits effects that go beyond the direct killing of residual tumor cells. IORT altered the wound response, inducing the expression of miR-223 in the peri-tumoral breast tissue. miR-223 downregulated the local expression of epidermal growth factor (EGF), leading to decreased activation of EGF receptor (EGFR) on target cells and, eventually, dampening a positive EGF-EGFR autocrine/paracrine stimulation loop induced by the post-surgical wound-healing response. Accordingly, both RT-induced miR-223 and peri-operative inhibition of EGFR efficiently prevented BC cell growth and reduced recurrence formation in mouse models of BC. Our study uncovers unknown effects of RT delivered on a wounded tissue and prompts to the use of anti-EGFR treatments, in a peri-operative treatment schedule, aimed to timely treat BC patients and restrain recurrence formation.
- Published
- 2016
- Full Text
- View/download PDF
35. p27kip1 expression limits H-Ras-driven transformation and tumorigenesis by both canonical and non-canonical mechanisms
- Author
-
Martina Cusan, Samuele Massarut, Ilenia Pellizzari, Francesca Citron, Stefania Berton, Tiziana Perin, Linda Fabris, Sara Benevol, Gustavo Baldassarre, Vincenzo Canzonieri, Ilenia Segatto, Monica Schiappacassi, Barbara Belletti, and Sara D'Andrea
- Subjects
0301 basic medicine ,stathmin ,Carcinogenesis ,Mice, Nude ,Stathmin ,Breast Neoplasms ,medicine.disease_cause ,Metastasis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,metastasis ,Phosphorylation ,Cell Proliferation ,biology ,cell cycle progression ,Cell growth ,business.industry ,Cancer ,Sarcoma ,H-Ras and K-Ras ,medicine.disease ,Xenograft Model Antitumor Assays ,Cyclin-Dependent Kinases ,Gene Expression Regulation, Neoplastic ,Transformation (genetics) ,030104 developmental biology ,Genes, ras ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,biology.protein ,Female ,business ,p27kip1 ,Cyclin-Dependent Kinase Inhibitor p27 ,Priority Research Paper - Abstract
// Ilenia Pellizzari 1,* , Linda Fabris 1,4,* , Stefania Berton 1,* , Ilenia Segatto 1 , Francesca Citron 1 , Sara D’Andrea 1 , Martina Cusan 1 , Sara Benevol 1 , Tiziana Perin 2 , Samuele Massarut 3 , Vincenzo Canzonieri 2 , Monica Schiappacassi 1 , Barbara Belletti 1 and Gustavo Baldassarre 1 1 Division of Experimental Oncology 2, Department of Translational Research, CRO Aviano, National Cancer Institute, Aviano, Italy 2 Pathology Unit, CRO Aviano, National Cancer Institute, Aviano, Italy 3 Breast Surgery Unit, CRO Aviano, National Cancer Institute, Aviano, Italy 4 Department of Experimental Therapeutics, M.D. Anderson Cancer Center, Houston, TX, USA * These authors have contributed equally to this article Correspondence to: Gustavo Baldassarre, email: // Barbara Belletti, email: // Keywords : p27kip1; stathmin; H-Ras and K-Ras; cell cycle progression; metastasis Received : June 15, 2016 Accepted : July 19, 2016 Published : August 27, 2016 Abstract The tumor suppressor protein p27 Kip1 plays a pivotal role in the control of cell growth and metastasis formation. Several studies pointed to different roles for p27 Kip1 in the control of Ras induced transformation, although no explanation has been provided to elucidate these differences. We recently demonstrated that p27 kip1 regulates H-Ras activity via its interaction with stathmin. Here, using in vitro and in vivo models, we show that p27 kip1 is an important regulator of Ras induced transformation. In H-Ras V12 transformed cells, p27 kip1 suppressed cell proliferation and tumor growth via two distinct mechanisms: 1) inhibition of CDK activity and 2) impairment of MT-destabilizing activity of stathmin. Conversely, in K-Ras4B V12 transformed cells, p27 kip1 acted mainly in a CDK-dependent but stathmin-independent manner. Using human cancer-derived cell lines and primary breast and sarcoma samples, we confirmed in human models what we observed in mice. Overall, we highlight a pathway, conserved from mouse to human, important in the regulation of H-Ras oncogenic activity that could have therapeutic and diagnostic implication in patients that may benefit from anti-H-Ras therapies.
- Published
- 2016
36. Abstract A72: Exploring the role of miR-223 loss in mammary epithelial cell transformation
- Author
-
Francesca Citron, Barbara Belletti, Ilenia Segatto, Gustavo Baldassarre, and Gian Luca Rampioni Vinciguerra
- Subjects
Cancer Research ,Oncogene ,Cell ,Biology ,medicine.disease ,Phenotype ,Primary tumor ,medicine.anatomical_structure ,Oncology ,Stroma ,mir-223 ,Cell culture ,Cancer cell ,Cancer research ,medicine ,Molecular Biology - Abstract
Despite novel treatment strategies and prevention, one of the major clinical challenges in breast cancer (BC) is the development of local and distant recurrences. These are linked not only to genetic and epigenetic alteration in cancer cells, but also to tumor-microenvironment and tumor-host crosstalk. In this context, surgical removal of primary tumor, inducing an inflammatory response and a wound healing process, may stimulate residual cancer cell survival, growth, and invasion. Radiotherapy, representing a standard care after surgery in BC, exerts its function by killing residual tumor cells and altering the local microenvironment. We have recently demonstrated that intraoperative radiotherapy strongly induced miR-223 expression that, by targeting EGF released in the peritumoral context, dampened the activation of EGFR/HER2 pathway, eventually resulting as a potent restrainer of residual cancer cell survival and growth. We also observed that miR-223 is highly expressed in normal mammary tissue, while all BC specimens analyzed expressed remarkably low levels. In line with these results, normal mammary epithelial cells of basal (HMEC) or luminal (BPEC) subtype expressed high levels of miR-223 compared to all tested BC cell lines. These data supported the possibility that loss of miR-223 expression could represent a common and possibly early event during mammary epithelial cell transformation. To explore this possibility, we transduced HMEC and BPEC cells using several oncogenes and tested the expression of miR-223. Transient expression of HER2, vSrc, and KRasG12D resulted in immediate drop of miR-223. Furthermore, to mimic oncogene-driven miR-223 down-modulation, we silenced its expression in HMEC and BPEC cells. Intriguingly, the loss of miR-223 per se induced the acquisition of transformed-like phenotypes, such as an increased proliferation in 2D-culture and a loss of polarized acinar structure with the formation of disorganized colonies in 3D-context. Interrogation of the TCGA BC dataset, comprising a large panel of normal and BC samples, confirmed our data and showed that, among all BC subtypes, miR-223 was particularly decreased in HER2+ and luminal BC. Notably, clustering HER2+ BC for miR-223HIGH/pY1068-EGFRLOW stratified patients with a significantly better overall survival. We thus explored the contribution of miR-223 in HER2-driven cell transformation. We overexpressed miR-223 in normal mammary epithelial cells and transformed them with HER2WT, ∆16HER2 (a particularly aggressive alternative splicing form of HER2), or HER2V659E (constitutively active form of HER2). Overexpression of miR-223 was able to efficiently counteract HER2-driven transformation, both in in vitro and in vivo models, not only in the presence of the WT oncogene, but also in the presence of the two aggressive mutants, the ∆16HER2 and HER2V659E. Altogether, our results indicate that loss or decreased expression of miR-223 may represent an early and common event during mammary epithelial cell transformation, contributing to loss of polarity and altering the maintenance of the differentiate phenotype, features profoundly linked to the tumorigenic process. Discovering the molecular mechanism by which miR-223 (and/or its targets) exerts their functions in cellular transformation and their possible interference in the tumor/stroma crosstalk may lead to identification of novel prognostic markers for early lesions and new promising therapeutic targets. Citation Format: Francesca Citron, Ilenia Segatto, Gian Luca Rampioni Vinciguerra, Gustavo Baldassarre, Barbara Belletti. Exploring the role of miR-223 loss in mammary epithelial cell transformation [abstract]. In: Proceedings of the AACR Special Conference: Advances in Breast Cancer Research; 2017 Oct 7-10; Hollywood, CA. Philadelphia (PA): AACR; Mol Cancer Res 2018;16(8_Suppl):Abstract nr A72.
- Published
- 2018
- Full Text
- View/download PDF
37. Abstract 1460: Stathmin regulates mammary gland morphogenesis and tumorigenesis
- Author
-
Gustavo Baldassarre, Tiziana Perin, Francesca Citron, Mara De Marco Zompit, Sara D'Andrea, Ilenia Segatto, Barbara Belletti, and Gian Luca Rampioni Vinciguerra
- Subjects
Cancer Research ,Oncology ,biology.protein ,medicine ,Stathmin ,Biology ,Carcinogenesis ,medicine.disease_cause ,Mammary gland morphogenesis ,Cell biology - Abstract
Microtubule (MT) dynamics is essential to provide a correct apico-basal polarization of epithelial cells, by regulating the movement of vesicles and proteins within the cell and by ensuring a correct orientation of the mitotic spindle during cell division. Little is known about the role of the MT-dynamics and of MT-regulating proteins during development of the mammary gland, a dynamic organ that displays a complex and finely organized apico-basal architecture. Stathmin, a MT-destabilizing protein, is often overexpressed in breast cancer (BC). By studying knockout (STM KO) mice we observed that STM KO females were unable to nurse their offspring. This phenotype was accompanied by defective development of the mammary gland, particularly evident during pregnancy and post-partum phases. Our results showed that stathmin absence mainly impinged on two key features of the mammary epithelial organization, polarity and proliferation. In vitro, normal mammary epithelial cells silenced for stathmin were not able to form organized acini in 3D-matrices. In vivo, when stathmin was KO, many molecules involved in mammary gland functions were not properly localized, indicating a disruption of the apico-basal polarity. Furthermore, loss of stathmin strongly decreased proliferation and induced disoriented positioning of the mitotic spindle in dividing epithelial cells, preventing the correct orientation of the daughter cells. Loss of polarity and deregulated proliferation are tightly linked to tumorigenesis. We therefore examined the role of stathmin also in the context of HER2-driven tumorigenesis. To this end, we used a transgenic mouse model expressing a constitutively active form of HER2 (Δ16HER2), which is an alternatively spliced, very aggressive form of HER2, and intercrossed them with STM WT and KO mice. We analyzed the role of stathmin in both early and late stages of tumorigenesis. STM KO mice displayed a significantly reduced number of pre-neoplastic foci and this phenotype was maintained also in late stages of tumorigenesis. In WT mice, pre-neoplastic lesions expressed high level of stathmin compared with normal tissue supporting the idea that stathmin played an active role in tumor initiation. Analysis of proliferation in tumors revealed that loss of stathmin strongly decreased the mitotic rate of Δ16HER2-transformed cells. Furthermore, we assessed that stathmin plays a cell autonomous function in tumor initiation. Our study has shed new light in the field of mammary gland development and of breast cancer biology. Collectively, our data indicate that loss of stathmin by altering the dynamics of microtubules, causes a profound disorganization of the normal mammary gland architecture and a delay in initiation of the tumorigenic process. We have uncovered new functions of stathmin that could have important implications in BC, also in consideration that several microtubules acting drugs are currently employed in the therapy of BC patients Citation Format: Ilenia Segatto, Mara De Marco Zompit, Gian Luca Rampioni Vinciguerra, Francesca Citron, Sara D'Andrea, Tiziana Perin, Gustavo Baldassarre, Barbara Belletti. Stathmin regulates mammary gland morphogenesis and tumorigenesis [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 1460.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.